[go: up one dir, main page]

WO2024206439A2 - Antibodies and chimeric antigen receptors specific for cd38 - Google Patents

Antibodies and chimeric antigen receptors specific for cd38 Download PDF

Info

Publication number
WO2024206439A2
WO2024206439A2 PCT/US2024/021673 US2024021673W WO2024206439A2 WO 2024206439 A2 WO2024206439 A2 WO 2024206439A2 US 2024021673 W US2024021673 W US 2024021673W WO 2024206439 A2 WO2024206439 A2 WO 2024206439A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
set forth
sequence set
amino acid
acid sequence
Prior art date
Application number
PCT/US2024/021673
Other languages
French (fr)
Other versions
WO2024206439A3 (en
Inventor
Alexandra Leida Liana LAZETIC
James Barnaby TRAGER
Ivan Chan
Luxuan Guo BUREN
Nitinkumar Lakshmanbhai PATEL
Bing Li
Emily KANG
Original Assignee
Nkarta, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nkarta, Inc. filed Critical Nkarta, Inc.
Publication of WO2024206439A2 publication Critical patent/WO2024206439A2/en
Publication of WO2024206439A3 publication Critical patent/WO2024206439A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K40/00Cellular immunotherapy
    • A61K40/10Cellular immunotherapy characterised by the cell type used
    • A61K40/11T-cells, e.g. tumour infiltrating lymphocytes [TIL] or regulatory T [Treg] cells; Lymphokine-activated killer [LAK] cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K40/00Cellular immunotherapy
    • A61K40/10Cellular immunotherapy characterised by the cell type used
    • A61K40/15Natural-killer [NK] cells; Natural-killer T [NKT] cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K40/00Cellular immunotherapy
    • A61K40/30Cellular immunotherapy characterised by the recombinant expression of specific molecules in the cells of the immune system
    • A61K40/31Chimeric antigen receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K40/00Cellular immunotherapy
    • A61K40/40Cellular immunotherapy characterised by antigens that are targeted or presented by cells of the immune system
    • A61K40/41Vertebrate antigens
    • A61K40/42Cancer antigens
    • A61K40/4202Receptors, cell surface antigens or cell surface determinants
    • A61K40/4222CD38 not IgG
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present disclosure relates to CD38 binding molecules, in particular, to anti-CD38 antibodies, including antibody fragments.
  • the present disclosure further relates to recombinant receptors containing such antibodies, including chimeric antigen receptors (CARs).
  • CARs chimeric antigen receptors
  • the disclosure further relates to genetically engineered cells expressing such antibodies and receptors, and uses thereof, including in cell therapy.
  • CD38 is a type II transmembrane glycoprotein expressed on the surface of a variety of cancer cells, including hematologic malignancies and solid tumors. CD38 expression is associated with immunosuppressive effects, and its abnormal overexpression is associated with cancer progression. While various CD38-binding molecules, including anti-CD38 antibodies and CARs are available, improved CD38-binding molecules and engineered CD38-targeting cells are needed. Provided are embodiments that meet such needs.
  • the anti-CD38 antibody or antigen-binding fragment thereof comprises: (i) a heavy chain variable region (VH) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:25, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:23; and a light chain variable region (VL) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:27, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:28, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:29; (ii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:30, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:31, and
  • the anti-CD38 antibody or antigen binding fragment comprises a heavy chain variable region (VH) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:25, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:23; and a light chain variable region (VL) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:27, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:28, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:29.
  • VH heavy chain variable region
  • VL light chain variable region
  • the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:30, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:31, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:32; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:35.
  • the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:36, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:37, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:38; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:40.
  • the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:41, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:42: and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:43, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:45.
  • the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDRI comprising the amino acid sequence set forth in SEQ ID NO:46, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:48: and a VL comprising a CDRI comprising the amino acid sequence set forth in SEQ ID NO:49, a CDR2 comprising the amino acid sequence set forth in SEQ ID NQ:50, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:51.
  • the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:52, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:53: and a VL comprising a CDRI comprising the amino acid sequence set forth in SEQ ID NO: 33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:54.
  • the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDRI comprising the amino acid sequence set forth in SEQ ID NO:55, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:56, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:57; and a VL comprising a CDRI comprising the amino acid sequence set forth in SEQ ID NO:58, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:59, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:60.
  • the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDRI comprising the amino acid sequence set forth in SEQ ID NO:61, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:62: and a VL comprising a CDRI comprising the amino acid sequence set forth in SEQ ID NO: 39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:63.
  • the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDRI comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:66: and a VL comprising a CDRI comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:68.
  • the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:69; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:70, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:71.
  • the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:72, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:73, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:74; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:75, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:76, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:77.
  • the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:78, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:79, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:80: and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:81.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises: (i) a heavy chain variable region (VH) comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:82, and a light chain variable region (VL) comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:83; (ii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:84, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:85; (iii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:86, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a heavy chain variable region (VH) comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 82, and a light chain variable region (VL) comprising the CDR 1 , the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:83.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a heavy chain variable region (VH) comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 82.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a light chain variable region (VL) comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:83.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:84, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:85.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 84. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 85.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:86, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:87.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:86.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:87. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:88, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:89.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:88. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 89.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:90, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:91.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NQ:90.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:91. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:92, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:93.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:94, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:95.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:94.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:95. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:96, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:97.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:96. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:97.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:98, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:99.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:98.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:99. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 100, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 101.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 100. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 101.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 102, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 103.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 102.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 103. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 104, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 105.
  • the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 104. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 105.
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:82, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:83;
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 84, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 85;
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:86, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:87;
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:88, and the VL comprises an amino acid sequence
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:82, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:83.
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:84, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:85.
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:86
  • the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:87.
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:88
  • the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:89.
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:90
  • the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:91
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:92
  • the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:93.
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:94, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:95. In some embodiments, the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:96, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:97.
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:98
  • the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:99.
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 100
  • the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 101.
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 102
  • the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 103
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 104
  • the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 105.
  • the anti-CD38 antibody or antigen-binding fragment thereof is a chimeric antibody or an antigen-binding fragment thereof. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof is a humanized antibody or an antigenbinding fragment thereof. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof comprises a human variable region framework region. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof is a fully human antibody or antigenbinding fragment thereof.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:82, and the VL comprises the amino acid sequence set forth in SEQ ID NO:83;
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:84, and the VL comprises the amino acid sequence set forth in SEQ ID NO:85;
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:86, and the VL comprises the amino acid sequence set forth in SEQ ID NO:87;
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:88, and the VL comprises the amino acid sequence set forth in SEQ ID NO:89;
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:90, and the VL comprises the amino acid sequence set forth in SEQ ID NO:91;
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:92, and the VL comprises the amino acid sequence set forth in SEQ ID NO:93;
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:82, and the VL comprises the amino acid sequence set forth in SEQ ID NO:83. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:82. In some embodiments, the VL comprises the amino acid sequence set forth in SEQ ID NO:83. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO: 84, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 85. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:84. In some embodiments, the VL comprises the amino acid sequence set forth in SEQ ID NO:85.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:86
  • the VL comprises the amino acid sequence set forth in SEQ ID NO:87.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:86.
  • the VL comprises the amino acid sequence set forth in SEQ ID NO:87.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:88
  • the VL comprises the amino acid sequence set forth in SEQ ID NO:89.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:88.
  • the VL comprises the amino acid sequence set
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:90
  • the VL comprises the amino acid sequence set forth in SEQ ID NO:91.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:90.
  • the VL comprises the amino acid sequence set forth in SEQ ID NO:91.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:92
  • the VL comprises the amino acid sequence set forth in SEQ ID NO:93.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:92.
  • the VL comprises the amino acid sequence set forth in SEQ ID NO:93.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:94, and the VL comprises the amino acid sequence set forth in SEQ ID NO:95. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:94. In some embodiments, the VL comprises the amino acid sequence set forth in SEQ ID NO:95. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:96, and the VL comprises the amino acid sequence set forth in SEQ ID NO:97. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:96. In some embodiments, the VL comprises the amino acid sequence set forth in SEQ ID NO:97.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:98
  • the VL comprises the amino acid sequence set forth in SEQ ID NO:99.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:98.
  • the VL comprises the amino acid sequence set forth in SEQ ID NO:99.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO: 100
  • the VL comprises the amino acid sequence set forth in SEQ ID NO: 101.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO: 100,.
  • the VL comprises the amino acid sequence set forth in SEQ ID NO: 101.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO: 102, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 103. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO: 102. In some embodiments, the VL comprises the amino acid sequence set forth in SEQ ID NO: 103. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO: 104, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 105. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO: 104. In some embodiments, the VL comprises the amino acid sequence set forth in SEQ ID NO: 105.
  • the antigen-binding fragment of the antibody is a
  • the antigen-binding fragment of the antibody is a Fab. In some embodiments, the antigen-binding fragment of the antibody is a Fab’. In some embodiments, the antigen-binding fragment of the antibody is a F(ab’)2. In some embodiments, the antigen- binding fragment of the antibody is a Fv. In some embodiments, the antigen-binding fragment of the antibody is a scFv.
  • the scFv comprises a linker between the VH and VL, the linker comprising the amino acid sequence set forth in SEQ ID NO: 1 or SEQ ID NO:3. In some embodiments, the linker comprises the amino acid sequence set forth in SEQ ID NO: 1. In some embodiments, the linker comprises the amino acid sequence set forth in SEQ ID NO: 3.
  • the scFv comprises the amino acid sequence set forth in SEQ ID NO: 108, SEQ ID NO: 109, SEQ ID NO: 110, SEQ ID NO: 111, SEQ ID NO: 112, SEQ ID NO: 113, SEQ ID NO:114, SEQ ID NO:115, SEQ ID NO:116, SEQ ID NO: 117, SEQ ID NO: 118, or SEQ ID NO:119.
  • the scFv comprises the amino acid sequence set forth in SEQ ID NO: 108.
  • the scFv comprises the amino acid sequence set forth in SEQ ID NO: 109.
  • the scFv comprises the amino acid sequence set forth in SEQ ID NO: 110. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 111. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 112. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 113. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 114. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 115. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 1 16.
  • the scFv comprises the amino acid sequence set forth in SEQ ID NO: 117. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 118. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 119.
  • the anti-CD38 antibody or antigen-binding fragment thereof binds to the extracellular domain (ECD) of human CD38 with a binding affinity (KD) of between about 1 nM and about 500 nM, or between about 4 nM and about 40 nM. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof binds to the extracellular domain (ECD) of human CD38 with a binding affinity (KD) of between about 1 nM and about 500 nM. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof binds to the extracellular domain (ECD) of human CD38 with a binding affinity (KD) of between about 4 nM and about 40 nM.
  • KD binding affinity
  • composition comprising any of the anti-CD38 antibody or antigen-binding fragment thereof provided herein.
  • the composition comprises a pharmaceutically acceptable carrier.
  • an immunoconjugate comprising any of the anti- CD38 antibody or antigen-binding fragment thereof provided herein and a cytotoxic agent.
  • the cytotoxic agent is a drug, a cytotoxin, or a radioactive agent.
  • the cytotoxic agent is a drug.
  • the cytotoxic agent is a cytotoxin.
  • the cytotoxic agent is a radioactive agent.
  • a composition comprising any of the immunoconjugate provided herein and a pharmaceutically acceptable carrier.
  • a bispecific molecule comprising any of the anti- CD38 antibody or antigen-binding fragment thereof provided herein and a second moiety that has a binding specificity for an immune cell.
  • the immune cell is a natural killer (NK) cell or a T cell.
  • the immune cell is a natural killer (NK) cell.
  • the immune cell is a T cell.
  • the second moiety binds to one or more of CD 16, NKp30, NKp46, NKG2D, or a combination thereof.
  • the second moiety binds to CD3.
  • the second moiety binds to CD16.
  • the second moiety binds to NKp30.
  • the second moiety binds to NKp46.
  • the second moiety binds to NKG2D.
  • composition comprising any of the bispecific molecules provided herein and a pharmaceutically acceptable carrier.
  • Also provided herein is a method for detecting CD38 in a cell or tissue, including: (a) contacting the cell or tissue with any of the anti-CD38 antibodies or antigen-binding fragments thereof provided herein; and (b) determining the amount of the antibody or antigenbinding fragment thereof bound to the cell or tissue, wherein the amount of the antibody or antigen-binding fragment thereof bound to the cell or tissue indicates the amount of CD38 in the cell or tissue.
  • any of the anti-CD38 antibody or antigenbinding fragment thereof provided herein in the manufacture of a kit for detecting CD38 in a cell or tissue including: (a) contacting the cell or tissue with the anti-CD38 antibody or antigenbinding fragment thereof; and (b) determining the amount of the antibody or antigen-binding fragment thereof bound to the cell or tissue, wherein the amount of the antibody or antigenbinding fragment thereof bound to the cell or tissue indicates the amount of CD38 in the cell or tissue.
  • anti-CD38 chimeric antigen receptor comprising an extracellular antigen-binding domain, a transmembrane domain, and an intracellular signaling domain.
  • the extracellular antigen-binding domain comprises any of the anti-CD38 antibodies or antigen-binding fragments thereof provided herein.
  • the extracellular antigen-binding domain comprises: (i) a heavy chain variable region (VH) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:25, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:23; and a light chain variable region (VL) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:27, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:28, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:29; (ii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:30, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:31, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:32; and a VL
  • the extracellular antigen-binding domain comprises: (i) a heavy chain variable region (VH) comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:82, and a light chain variable region (VL) comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:83; (ii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:84, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:85; (iii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:86, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:87; (iv)
  • the transmembrane domain is or comprises a transmembrane from CD8, CD3, CD4, or CD28. In some embodiments, the transmembrane domain is or comprises a transmembrane from CD8. In some embodiments, the transmembrane domain is or comprises a transmembrane from CD8alpha. In some embodiments, the transmembrane domain is or comprises a transmembrane from CD3. In some embodiments, the transmembrane domain is or comprises a transmembrane from CD4. In some embodiments, the transmembrane domain is or comprises a transmembrane from CD28. In some embodiments, the transmembrane domain is human.
  • the intracellular signaling domain comprises a CD3zeta domain.
  • the CD3zeta domain is a human CD3zeta domain.
  • the intracellular signaling domain comprises a co- stimulatory signaling region.
  • the co-stimulatory signaling region comprises an intracellular signaling region of 0X40, CD28, 4- IBB, ICOS, or DAP 10.
  • the costimulatory signaling region comprises an intracellular signaling region of 0X40.
  • the co-stimulatory signaling region comprises an intracellular signaling region of CD28.
  • the co-stimulatory signaling region comprises an intracellular signaling region of 4-1B.
  • the co-stimulatory signaling region comprises an intracellular signaling region of ICOS. In some embodiments, the co-stimulatory signaling region comprises an intracellular signaling region of DAP10. In some embodiments, the CAR comprises a CD8alpha transmembrane domain and an intracellular signaling domain comprising a CD3zeta domain and a co-stimulatory region comprising an intracellular signaling region of 0X40.
  • the CAR comprises the amino acid sequence set forth in any one of SEQ ID NOS: 327-338. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 327. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 328. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 329. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 330. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 331. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 332.
  • the CAR comprises the amino acid sequence set forth in SEQ ID NO: 333. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 334. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 335. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 336. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 337. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 338.
  • nucleic acid molecule encoding any of the anti- CD38 antibodies or antigen-binding fragments thereof provided herein.
  • nucleic acid molecule encoding any of the anti- CD38 CARs provided herein.
  • the vector comprising any of the nucleic acid molecules provided herein.
  • the vector is a viral vector.
  • the vector is a retroviral vector.
  • the vector is a lentiviral vector.
  • Also provided herein is a cell comprising any of the anti-CD38 antibodies or antigen-binding fragments thereof provided herein.
  • Also provided herein is a cell comprising any of the anti-CD38 CARs provided herein.
  • Also provided herein is a cell comprising any of the nucleic acid molecules provided herein.
  • Also provided herein is a cell comprising any of the vectors provided herein.
  • the cell is a natural killer (NK) cell or a T cell. In some embodiments, the cell is a NK cell. In some embodiments, the cell is a T cell. In some embodiments, the cell is genetically edited to reduce expression of CD38.
  • a natural killer (NK) cell comprising any of the anti- CD38 antibodies or antigen-binding fragments thereof provided herein.
  • the NK cell is genetically edited to reduce expression of CD38.
  • a T cell comprising any of the anti-CD38 antibodies or antigen-binding fragments thereof provided herein. In some embodiments, the T cell is genetically edited to reduce expression of CD38.
  • a natural killer (NK) cell comprising any of the anti- CD38 CARs provided herein.
  • the NK cell is genetically edited to reduce expression of CD38.
  • a T cell comprising any of the anti-CD38 CARs provided herein.
  • the T cell is genetically edited to reduce expression of CD38.
  • the NK cell is genetically edited to reduce expression of CD38.
  • the T cell is genetically edited to reduce expression of CD38.
  • the genetic editing reduces the expression of CD38 by at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, or at least about 80%. In some embodiments, the genetic editing reduces the expression of CD38 by at least about 20%. In some embodiments, the genetic editing reduces the expression of CD38 by at least about 30%. In some embodiments, the genetic editing reduces the expression of CD38 by at least about 40%. In some embodiments, the genetic editing reduces the expression of CD38 by at least about 50%. In some embodiments, the genetic editing reduces the expression of CD38 by at least about 60%. In some embodiments, the genetic editing reduces the expression of CD38 by at least about 70%. In some embodiments, the genetic editing reduces the expression of CD38 by at least about 80%.
  • the cell comprises a membrane -bound interleukin- 15 (mbIL15).
  • the CAR and the mbIL15 are encoded bicistronically by the same nucleic acid molecule.
  • the cell comprises a nucleic acid molecule encoding the CAR and the mbIL15, wherein the sequences encoding the CAR and the mbIL15 are separated by a T2A-encoding sequence.
  • composition comprising a plurality of any of the cells provided herein. Also provided herein is a composition comprising a plurality of any of the NK cells provided herein. Also provided herein is a composition comprising a plurality of any of the T cells provided herein. In some embodiments, the composition comprises a pharmaceutically acceptable carrier.
  • the composition comprises between about 3 x 10 8 CAR-expressing cells and about 3 x 10 9 CAR-expressing cells, or between about 1 x 10 9 CAR- expressing cells and about 2 x 10 9 CAR-expressing cells. In some embodiments, the composition comprises between about 2 x 10 9 CAR-expressing cells and about 5 x 10 9 CAR-expressing cells. In some embodiments, the composition comprises between about 3 x 10 8 CAR-expressing cells and about 3 x 10 9 CAR-expressing cells. In some embodiments, the composition comprises between about 1 x 10 q and about 2 x 10 9 CAR-expressing cells. In some embodiments, the composition comprises about 3 x 10 8 CAR-expressing cells.
  • the composition comprises about 1 x 10 9 CAR-expressing cells. In some embodiments, the composition comprises about 1.5 x 10 9 CAR-expressing cells. In some embodiments, the composition comprises about 2 x 10 9 CAR-expressing cells. In some embodiments, the composition comprises about 2.5 x 10 9 CAR-expressing cells. In some embodiments, the composition comprises about 3 x 10 9 CAR-expressing cells. In some embodiments, the composition comprises about 3.5 x 10 9 CAR-expressing cells. In some embodiments, the composition comprises about 4 x 10 9 CAR-expressing cells. In some embodiments, the composition comprises about 4.5 x 10 9 CAR-expressing cells. In some embodiments, the composition comprises about 5 x 10 9 CAR-expressing cells.
  • Also provided herein is a method of treatment comprising administering to a subject having a CD38-expressing disease or condition, any of the anti-CD38 antibodies or antigen-binding fragments provided herein. Also provided herein is a method of treatment comprising administering to a subject having a disease or disorder associated with CD38, any of the anti-CD38 antibodies or antigen-binding fragments provided herein.
  • Also provided herein is a method of treatment comprising administering to a subject having a CD38-expressing disease or condition any of the anti-CD38 CARs provided herein. Also provided herein is a method of treatment comprising administering to a subject having a disease or disorder associated with CD38, any of the anti-CD38 CARs provided herein.
  • Also provided herein is a method of treatment comprising administering to a subject having a CD38-expressing disease or condition any of the cells provided herein.
  • the cells are allogeneic to the subject.
  • a method of treatment comprising administering to a subject having a disease or disorder associated with CD38, any of the cells provided herein.
  • the cells are allogeneic to the subject.
  • Also provided herein is a method of treatment comprising administering to a subject having a CD38-expressing disease or condition, any of the compositions provided herein. Also provided herein is a method of treatment comprising administering to a subject having a disease or disorder associated with CD38 (e.g., elevated CD38 expression), any of the compositions provided herein.
  • a disease or disorder associated with CD38 e.g., elevated CD38 expression
  • Also provided herein is a method of treatment comprising administering to a subject having a CD38-expressing disease or condition, any of the immunoconjugates provided herein. Also provided herein is a method of treatment comprising administering to a subject having a disease or disorder associated with CD38, any of the immunoconjugates provided herein. Also provided herein is a method of treatment comprising administering to a subject having a CD38-expressing disease or condition, any of the bispecific molecules provided herein. Also provided herein is a method of treatment comprising administering to a subject having a disease or disorder associated with CD38, any of the bispecific molecules provided herein.
  • the CD38-expressing disease or condition is a cancer or a tumor. In some embodiments, the CD38-expressing disease or condition is a solid tumor.
  • the CD38-expressing disease or condition is a cancer.
  • the cancer is selected from the group consisting of leukemia, lymphoma, myeloma, glioma, thyroid cancer, lung cancer, colorectal cancer, head and neck cancer, stomach cancer, liver cancer, pancreatic cancer, renal cancer, urothelial cancer, prostate cancer, testicular cancer, breast cancer, cervical cancer, endometrial cancer, ovarian cancer, and melanoma.
  • the cancer is lung cancer.
  • the cancer is colorectal cancer.
  • the cancer is head and neck cancer.
  • the cancer is ovarian cancer.
  • the cancer is breast cancer.
  • the cancer is glioma.
  • the cancer is melanoma.
  • the cancer is multiple myeloma.
  • the CD38-expressing disease or condition is an autoimmune disease or disorder.
  • the disease or disorder associated with CD38 is an autoimmune disease or disorder.
  • the CD38-expressing disease or condition is an autoimmune disease or disorder is selected from the group consisting of systemic lupus erythematosus (SLE), lupus nephritis, CNS lupus, myositis, multiple sclerosis (MS), myasthenia gravis (MG), rheumatoid arthritis (RA), scleroderma, thyroid disease, diabetes, vasculitis, or any combination thereof.
  • the CD38-expressing disease or condition is systemic lupus erythematosus (SLE).
  • the CD38-expressing disease or condition is lupus nephritis (LN).
  • the CD38-expressing disease or condition is CNS lupus. In some embodiments, the CD38-expressing disease or condition is myositis. In some embodiments, the CD38-expressing disease or condition is multiple sclerosis (MS). In some embodiments, the CD38-expressing disease or condition is myasthenia gravis (MG). In some embodiments, the CD38-expressing disease or condition is rheumatoid arthritis (RA). In some embodiments, the CD38-expressing disease or condition is scleroderma. In some embodiments, the CD38-expressing disease or condition is thyroid disease. In some embodiments, the CD38- expressing disease or condition is diabetes. In some embodiments, the CD38-expressing disease or condition is vasculitis.
  • the subject has been treated with, or is a candidate for treatment with, an anti-CD38 antibody. In some embodiments, the subject has been previously treated with an anti-CD38 antibody. In some embodiments, the subject is a candidate for treatment with an anti-CD38 antibody. In some embodiments, the treatment further comprises administering an anti-CD38 antibody to the subject.
  • the cancer is a relapsed/refractory cancer.
  • the subject has relapsed following treatment with and/or is refractory to a prior line of therapy for a CD38-expressing cancer.
  • the subject has relapsed following treatment with a prior line of therapy for a CD38-expressing cancer.
  • the subject is refractory to a prior line of therapy for a CD38-expressing cancer.
  • the autoimmune disease or disorder is a relapsed/refractory autoimmune disease.
  • the subject has relapsed following treatment with and/or is refractory to a prior line of therapy for an autoimmune disease or disorder.
  • the subject has relapsed following treatment with a prior line of therapy for an autoimmune disease or disorder.
  • the subject is refractory to a prior line of therapy for an autoimmune disease or disorder.
  • the prior line of therapy is one, two, three, or four prior lines of therapy. In some embodiments, the prior line of therapy is one prior line of therapy. In some embodiments, the prior line of therapy is two prior lines of therapy. In some embodiments, the prior line of therapy is three prior lines of therapy. In some embodiments, the prior line of therapy is four prior lines of therapy.
  • Also provided herein is a method of producing a cell targeting CD38, comprising introducing into the cell any of the nucleic acid molecules provided herein.
  • Also provided herein is a method of producing a cell targeting CD38, comprising introducing into the cell any of the vectors provided herein.
  • the method further comprises genetically editing the cell to reduce expression of a target gene.
  • the target gene comprises CD38.
  • the method further comprises genetically editing the cell to reduce expression of CD38.
  • the genetic editing is carried out using a CRISPR/Cas system.
  • the genetic editing comprises use of a CD38 -targeting guide RNA (gRNA).
  • the genetic editing comprises use of a ribonucleoprotein complex (RNP) comprising a CD38 -targeting gRNA.
  • the cell is an immune cell.
  • the cell is a natural killer (NK) cell or a T cell.
  • the cell is a NK cell.
  • the cell is a T cell.
  • Figure 1 shows the binding of a non-limiting embodiment of an anti-CD38 scFvs to CD38-negative cells (y-axis) and CD38-positive cells (x axis). Binders selected for further analysis are shown in light gray.
  • Figure 2A shows antigen-dependent activation of CD38 KO Jurkat cells expressing non-limiting embodiments of anti-CD38 CARs (as measured by CD69 expression; y- axis) compared to the binding affinity of the anti-CD38 scFvs incorporated by the CARs (x-axis).
  • Figures 2B and 2C show the ratio of antigen-dependent activation to antigenindependent (tonic) activation (y axis) and CAR expression (x-axis) as measured in Jurkat cells expressing non-limiting embodiments of CD38-directed CARs. CARs selected for further analysis are shown in the black box ( Figure 2A) and indicated by clone number ( Figure 2B).
  • Figure 3 shows CAR expression as measured in Jurkat cells expressing CARs containing non-limiting embodiments of anti-CD38 scFvs (y-axis) and the epitope bin identified for the non-limiting embodiments of anti-CD38 scFvs (x-axis). Clones selected for further analysis are indicated by clone number.
  • Figure 4 shows CAR expression as measured in primary natural killer (NK) cells expressing CARs containing non-limiting embodiments of anti-CD38 scFvs (clones indicated by number on x-axis).
  • Figure 5 shows the number of MM. IS target cells 24 hours after CD38 CAR-expressing primary NK cells from two different donors were rechallenged with target cells (clones indicated by number on x-axis).
  • CD38 is known to play a role in hematological malignancies, including chronic lymphocytic leukemia (CLL) and multiple myeloma (MM), and is also implicated in chronic inflammatory diseases, such as rheumatoid arthritis (RA) and asthma.
  • CLL chronic lymphocytic leukemia
  • MM multiple myeloma
  • RA rheumatoid arthritis
  • CD38 expression correlates with a more aggressive disease course; patients having greater than 30% CD38-positive B cells require more rigorous treatment regimens and exhibit shorter survival times.
  • CD38 is expressed at uniformly high levels on MM cells, and the U.S. Food and Drug Administration (FDA) approved anti-CD38 antibody daratumumab has been observed to directly kill myeloma cells and deplete CD38+ immunosuppressive cells (e.g., myeloid derived suppressor cells and Tregs). Still, patient responses with daratumumab are variable, and most eventually develop progressive disease (PD) when treated with daratumumab as a monotherapy. Possible causes for daratumumab resistance include CD38 downregulation by myeloma cells and the elimination of CD38+ NK cells, which are thought to mediate antibody-dependent cellular cytotoxicity (ADCC). See Konen et al., Cells (2020) 9(1):52.
  • ADCC antibody-dependent cellular cytotoxicity
  • CD38 is also implicated in the progression of solid tumors, including melanoma, glioma, esophageal, cervical, and lung cancers, where it is hypothesized to play an immunosuppressive role.
  • CD38 catalyzes the first step towards adenosine formation from nicotinamide adenine dinucleotide (NAD).
  • NAD nicotinamide adenine dinucleotide
  • TAE tumor microenvironment
  • ADPR adenosine diphosphate ribose
  • AMP adenosine monophosphate
  • AMP is in turn converted into adenosine, which is thought to promote immunosuppression - including by inhibiting cytotoxic T cells, NK cells, and dendritic cells - via signaling through adenosine receptor 2A (A2A) or 2B (A2B).
  • A2A adenosine receptor 2A
  • 2B A2B
  • CD38 has been identified as a potential therapeutic target for autoimmune diseases.
  • CD38 is expressed by long-lived plasma cells, which produce the autoantibodies characteristic of most systemic autoimmune diseases (Benfaremo and Gabrielli, Cells (2020) 9(1):77).
  • Daratumumab an anti-CD38 antibody, improved clinical manifestations of patients with refractory SLE and eliminated antibody-producing plasma cells (Ostendorf et al., N Engl J Med (2020) 383:1149-55).
  • CD38 may also mediate autoimmune diseases by regulating cell recruitment, cytokine and chemokine release, cell activation, phagocytosis, and antigen presentation during inflammation (Ye et al., Autoimmunity Rev (2023) 22(4): 103289).
  • the anti-CD38 antibodies and CARs and cells expressing the same as provided herein may be effective to treat any autoimmune disease in which autoantibodyproducing plasma cells are implicated.
  • ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇
  • cells such as engineered or recombinant cells expressing such CD38-binding receptors, e.g., anti-CD38 CARs and/or containing nucleic acids encoding such receptors, and compositions and articles of manufacture and therapeutic doses containing such cells.
  • any of the engineered cells incorporating a CD38- binding molecule as provided herein can be used in combination with an anti-CD38 antibody (e.g., daratumumab).
  • an anti-CD38 antibody e.g., daratumumab
  • editing of CD38 in engineered immune cells may reduce or avoid binding of an anti-CD38 antibody to the engineered immune cells, thereby reducing the “sink” of the antibody due to binding of non-target (e.g., cancer) cells. It is further contemplated that genetically editing CD38 in engineered NK cells may reduce undesired elimination of the engineered NK cells, thereby preserving the ability of engineered NK cells to mediate ADCC and potentially potentiating the effects of an anti-CD38 antibody.
  • cells e.g., engineered cells
  • Adoptive cell therapies including those involving the administration of cells expressing chimeric receptors specific for a disease or disorder of interest, such as chimeric antigen receptors (CARs) and/or other recombinant antigen receptors
  • CARs chimeric antigen receptors
  • Adoptive cell therapies can be effective in the treatment of cancer and other diseases and disorders.
  • available approaches to adoptive cell therapy may not always be entirely satisfactory.
  • CD38- binding molecules having the ability to recognize and bind to a target, e.g., target antigen such as CD38, to traffic, localize to and successfully enter appropriate sites within the subject, tumors, and environments thereof, to become activated, expand, to exert various effector functions, including cytotoxic killing and secretion of various factors such as cytokines, to persist, to exhibit certain phenotypic states to provide robust responses following re-exposure to CD38, and avoid or reduce exhaustion and/or differentiation into a suppressive state, are needed.
  • a target e.g., target antigen such as CD38
  • certain recombinant receptors can exhibit antigenindependent activation (also known as “tonic signaling”), which can lead to undesirable effects, such as increased exhaustion of immune cells that express the recombinant receptor. In some aspects, such effects may limit the immune cell's activity, effect or potency. In some cases, during engineering and ex vivo expansion of the cells for recombinant receptor expression, the cells may exhibit phenotypes indicative of exhaustion, due to tonic signaling through the recombinant receptor. Thus, improved CD38-binding molecules that bind to CD38, exhibit low levels of antigen-independent activation, and/or exhibit a high ratio of antigen-dependent activation to antigen-independent activation are needed.
  • CD38-binding molecules, and related CARs and cells, that meet such needs are provided herein.
  • all terms of art, notations and other technical and scientific terms or terminology used herein are intended to have the same meaning as is commonly understood by one of ordinary skill in the art to which the claimed subject matter pertains. In some cases, terms with commonly understood meanings are defined herein for clarity and/or for ready reference, and the inclusion of such definitions herein should not necessarily be construed to represent a substantial difference over what is generally understood in the art.
  • CD38 binding molecules such as CD38- binding polypeptides.
  • Such binding molecules include antibodies (including antigen-binding fragments thereof) that specifically bind to CD38 proteins, such as human CD38 protein.
  • polypeptides containing such antibodies including single chain cell surface proteins, e.g., recombinant receptors such as CARs containing such antibodies.
  • anti-CD38 antibodies including functional antibody fragments.
  • the antibodies include those that are multi-domain antibodies, such as those containing Vn and Vi. domains.
  • the antibodies include a variable heavy chain and a variable light chain, such as scFvs.
  • the antibodies include antibodies that specifically bind to CD38, e.g., human CD38.
  • human CD38 e.g., human CD38.
  • molecules containing such antibodies e.g., single-chain proteins, fusion proteins, and/or recombinant receptors such as chimeric receptors, including CARs.
  • antibody herein is used in the broadest sense and includes polyclonal and monoclonal antibodies, including intact antibodies and functional (antigenbinding) antibody fragments, including fragment antigen binding (Fab) fragments, Ftab'h fragments, Fab' fragments, Fv fragments, recombinant IgG (rlgG) fragments, variable heavy chain (VH) regions capable of specifically binding the antigen, single chain antibody fragments, including single chain variable fragments (scFv), and single domain antibody (e.g., sdAb, sdFv, nanobody) fragments.
  • Fab fragment antigen binding
  • rlgG fragment antigen binding
  • VH variable heavy chain
  • the term encompasses genetically engineered and/or otherwise modified forms of immunoglobulins, such as intrabodies, peptibodies, chimeric antibodies, fully human antibodies, humanized antibodies, and heteroconjugate antibodies, multispecific, e.g., bispecific, antibodies, diabodies, triabodies, and tetrabodies, tandem di-scFv, tandem tri-scFv.
  • antibody should be understood to encompass functional antibody fragments thereof.
  • the term also encompasses intact or full-length antibodies, including antibodies of any class or sub-class, including IgG and sub-classes thereof, IgM, IgE, IgA, and IgD.
  • CDR complementarity determining region
  • HVR hypervariable region
  • FR-H1, FR-H2, FR-H3, and FR-H4 there are four FRs in each full-length heavy chain variable region (FR-H1, FR-H2, FR-H3, and FR-H4), and four FRs in each full- length light chain variable region (FR-L1, FR-L2, FR-L3, and FR-L4).
  • the boundaries of a given CDR or FR may vary depending on the scheme used for identification.
  • the Kabat scheme is based structural alignments
  • the Chothia scheme is based on structural information. Numbering for both the Kabat and Chothia schemes is based upon the most common antibody region sequence lengths, with insertions accommodated by insertion letters, for example, “30a,” and deletions appearing in some antibodies. The two schemes place certain insertions and deletions (“indels”) at different positions, resulting in differential numbering.
  • the Contact scheme is based on analysis of complex crystal structures and is similar in many respects to the Chothia numbering scheme.
  • Table 1 lists position boundaries of CDR-L1, CDR-L2, CDR-L3 and CDR-H1, CDR-H2, CDR-H3 as identified by Kabat, Chothia, and Contact schemes, respectively.
  • residue numbering is listed using both the Kabat and Chothia numbering schemes.
  • FRs are located between CDRs, for example, with FR-L1 located between CDR-L1 and CDR-L2, and so forth. It is noted that because the shown Kabat numbering scheme places insertions at H35A and H35B, the end of the Chothia CDR-H1 loop when numbered using the shown Kabat numbering convention varies between H32 and H34, depending on the length of the loop.
  • a “CDR” or “complementary determining region,” or individual specified CDRs (e.g., “CDR-H1, CDR-H2), of a given antibody or region thereof, such as a variable region thereof, should be understood to encompass a (or the specific) complementary determining region as defined by any of the aforementioned schemes.
  • a particular CDR e.g., a CDR-H3
  • a CDR-H3 contains the amino acid sequence of a corresponding CDR in a given Vnor VL amino acid sequence
  • such a CDR has a sequence of the corresponding CDR (e.g., CDR-H3) within the variable region, as defined by any of the aforementioned schemes.
  • specific CDR sequences are specified.
  • FR or individual specified FR(s) e.g., FR-H1, FR-H2), of a given antibody or region thereof, such as a variable region thereof, should be understood to encompass a (or the specific) framework region as defined by any of the known schemes.
  • the scheme for identification of a particular CDR, FR, or FRs or CDRs is specified, such as the CDR as defined by the IMGT, Kabat, Chothia, or Contact method.
  • the particular amino acid sequence of a CDR or FR is given.
  • variable region refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen.
  • variable domains of the heavy chain and light chain (Vn nd VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three CDRs.
  • FRs conserved framework regions
  • a single Vuor VLdomain may be sufficient to confer antigenbinding specificity.
  • antibody fragments refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds.
  • antibody fragments include but are not limited to Fv, Fab, Fab', Fab'-SH, F(ab')2; diabodies; linear antibodies; variable heavy chain (VH) regions, single-chain antibody molecules such as scFvs and single-domain VH single antibodies; and multispecific antibodies formed from antibody fragments.
  • the antibodies are single-chain antibody fragments comprising a variable heavy chain region and a variable light chain region, such as scFvs.
  • the antibodies are recombinantly-produced fragments, such as fragments comprising arrangements that do not occur naturally, such as those with two or more antibody regions or chains joined by synthetic linkers, e.g., peptide linkers, and/or that are may not be produced by enzyme digestion of a naturally-occurring intact antibody.
  • the antibody fragments are scFvs.
  • Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody.
  • a single-domain antibody is a human or humanized singledomain antibody.
  • a “humanized” antibody is an antibody in which all or substantially all CDR amino acid residues are derived from non-human CDRs and all or substantially all FR amino acid residues are derived from human FRs.
  • a humanized antibody optionally may include at least a portion of an antibody constant region derived from a human antibody.
  • a “humanized form” of a non-human antibody refers to a variant of the non-human antibody that has undergone humanization, typically to reduce immunogenicity to humans, while retaining the specificity and affinity of the parental non-human antibody.
  • some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g., the antibody from which the CDR residues are derived), e.g., to restore or improve antibody specificity or affinity.
  • a non-human antibody e.g., the antibody from which the CDR residues are derived
  • anti-CD38 antibodies are human antibodies.
  • a “human antibody” is an antibody with an amino acid sequence corresponding to that of an antibody produced by a human or a human cell, or non-human source that utilizes human antibody repertoires or other human antibody-encoding sequences, including human antibody libraries.
  • the term excludes humanized forms of non-human antibodies comprising non-human antigen-binding regions, such as those in which all or substantially all CDRs are non-human.
  • the term includes antigen-binding fragments of human antibodies.
  • monoclonal antibodies including monoclonal antibody fragments.
  • the term “monoclonal antibody” as used herein refers to an antibody obtained from or within a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical, except for possible variants containing naturally occurring mutations or arising during production of a monoclonal antibody preparation, such variants generally being present in minor amounts.
  • each monoclonal antibody of a monoclonal antibody preparation is directed against a single epitope on an antigen. The term is not to be construed as requiring production of the antibody by any particular method.
  • a monoclonal antibody may be made by a variety of techniques, including but not limited to generation from a hybridoma, recombinant DNA methods, phage-display and other antibody display methods.
  • polypeptide and “protein” are used interchangeably to refer to a polymer of amino acid residues and are not limited to a minimum length.
  • Polypeptides including the provided antibodies and antibody chains and other peptides, e.g., linkers and CD38-binding peptides, may include amino acid residues including natural and/or non-natural amino acid residues.
  • the terms also include post-expression modifications of the polypeptide, for example, glycosylation, sialylation, acetylation, phosphorylation, and the like.
  • the polypeptides may contain modifications with respect to a native or natural sequence, as long as the protein maintains the desired activity. i. Non-Limiting Features
  • an anti-CD38 antibody or antigen-binding fragment thereof comprises a heavy chain variable region (VH) and a light chain variable region (VL).
  • VH comprises a CDR1, a CDR2, and a CDR3; and the VL comprises a CDR1, a CDR2, and a CDR3.
  • the VH comprises a CDR1, a CDR2, and a CDR3 as set forth in any VH sequence provided herein, and the VL comprises a CDR1, a CDR2, and a CDR3 as set forth in any VL sequence provided herein.
  • the anti- CD38 antibody comprises a VH having at least about 80%, 85%, 90%, 95%, 97% (or more)sequence identity to any VH sequence provided herein, and a VL having at least about 80%, 85%, 90%, 95%, 97% (or more (sequence identity to any VL sequence provided herein.
  • the anti-CD38 antibody or antigen-binding fragment thereof comprises a VH having any VH sequence provided herein, and a VL having any VL sequence provided herein.
  • the anti-CD38 antibody or antigen-binding fragment thereof comprises a linker between the VH and VL region.
  • the linker comprises the sequence set forth in SEQ ID NO: 1 or SEQ ID NO:3. In some embodiments, the linker comprises the sequence set forth in SEQ ID NO:1. In some embodiments, the linker comprises the sequence set forth in SEQ ID NO:3. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof is an scFv. In some embodiments, the scFv comprises any scFv sequence provided herein.
  • the anti-CD38 antibody or antigen-binding fragment thereof comprises: (i) a heavy chain variable region (VH) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:25, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:23; and a light chain variable region (VL) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:27, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:28, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:29; (ii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:30, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:31, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:32;
  • VH heavy chain variable region
  • the anti-CD38 antibody or antigen-binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:78, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:79, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:80; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:81.
  • the anti-CD38 antibody or antigen- binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:66; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:68.
  • the anti-CD38 antibody or antigen-binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:69; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:70, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:7I.
  • the anti-CD38 antibody or antigen-binding fragment thereof comprises: (i) a heavy chain variable region (VH) comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:82, and a light chain variable region (VL) comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:83; (ii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:84, and a VL comprising the CDR 1 , the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 85; (iii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:86, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:
  • the anti-CD38 antibody or antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 100, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 101.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a heavy chain variable region (VH) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:25, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:23; and a light chain variable region (VL) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:27, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 28, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:29.
  • VH heavy chain variable region
  • VL light chain variable region
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a heavy chain variable region (VH) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 121, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 122, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 123; and a light chain variable region (VL) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 124, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 125, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:29.
  • VH heavy chain variable region
  • VL light chain variable region
  • the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:82. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:83. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:82, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:83.
  • the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:82
  • the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:83.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO: 82
  • the VL comprises the amino acid sequence set forth in SEQ ID NO: 83.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 108.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:30, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:31, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:32; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:35.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 126, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 127, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 128; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 129, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 120, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:35.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:84. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:85. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:84, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:85.
  • the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:84
  • the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:85.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:84
  • the VL comprises the amino acid sequence set forth in SEQ ID NO:85.
  • the anti- CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 109.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:36, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:37, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:38; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:40.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 131, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 132, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 133; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 134, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:40.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:86. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:87. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:86, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:87.
  • the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:86
  • the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:87.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:86
  • the VL comprises the amino acid sequence set forth in SEQ ID NO:87.
  • the anti- CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 1 10.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:41, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:42; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:43, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:45.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 135, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 136, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 137; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 138, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 139, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:45.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:88. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 89. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:88, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:89.
  • the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:88
  • the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:89.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:88
  • the VL comprises the amino acid sequence set forth in SEQ ID NO:89.
  • the anti- CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 111.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:46, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:48; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:49, a CDR2 comprising the amino acid sequence set forth in SEQ ID NQ:50, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:51.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 140, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 141, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 142; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 143, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 144, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:51.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:90.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:91In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:90, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:91.
  • the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:90
  • the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:91.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:90
  • the VL comprises the amino acid sequence set forth in SEQ ID NO:91.
  • the anti- CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 112.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:52, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:53; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:54.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 135, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 145, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 146; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 147, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 148, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:54.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:92. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:93. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:92, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:93.
  • the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:92
  • the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:93.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:92
  • the VL comprises the amino acid sequence set forth in SEQ ID NO:93.
  • the anti- CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 113.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:55, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:56, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:57; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:58, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:59, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:60.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 149, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 150, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 151; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 152, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 153, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:60.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:94. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:95. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:94, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:95.
  • the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:94
  • the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:95.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:94
  • the VL comprises the amino acid sequence set forth in SEQ ID NO:95.
  • the anti- CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 114 .
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:61, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:62; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:63.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 154, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 155, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 156; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 157, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 158, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:63.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:96. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:97. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:96, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:97.
  • the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:96
  • the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:97.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:96
  • the VL comprises the amino acid sequence set forth in SEQ ID NO:97.
  • the anti- CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 115.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:66; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:68.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 159, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 160, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 161; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 162, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 130, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:68.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:98. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:99. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:98, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:99.
  • the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:98
  • the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:99.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO:98
  • the VL comprises the amino acid sequence set forth in SEQ ID NO:99.
  • the anti- CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 116.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:69; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:70, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:71.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 159, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 163, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 164; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 165, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 166, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:71.
  • the anti-CD38 antibody or antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 100. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 101.
  • the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 100, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 101.
  • the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more)sequence identity to the amino acid sequence set forth in SEQ ID NO: 100
  • the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more)sequence identity to the amino acid sequence set forth in SEQ ID NO: 101
  • the VH comprises the amino acid sequence set forth in SEQ ID NO: 100
  • the VL comprises the amino acid sequence set forth in SEQ ID NO: 101.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 117.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:72, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:73, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:74; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:75, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:76, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:77.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 167, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 168, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 169; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 170, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 171, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:77.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 102. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 103.
  • the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 102, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 103.
  • the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or morejsequence identity to the amino acid sequence set forth in SEQ ID NO: 102
  • the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO: 103.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO: 102
  • the VL comprises the amino acid sequence set forth in SEQ ID NO: 103.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 118.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:78, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:79, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:80; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:81.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 172, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 173, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 174; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 162, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 130, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:81.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 104. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 105.
  • the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 104, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 105.
  • the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more)sequence identity to the amino acid sequence set forth in SEQ ID NO: 104
  • the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more)sequence identity to the amino acid sequence set forth in SEQ ID NO: 105.
  • the VH comprises the amino acid sequence set forth in SEQ ID NO: 104
  • the VL comprises the amino acid sequence set forth in SEQ ID NO: 105.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 119.
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence as set forth in any one of SEQ ID NOS: 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197,
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence as set forth in any one of SEQ ID NOS: 91, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220,
  • CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence as set forth in any one of SEQ ID NOS: 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217,
  • VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence as set forth in any one of SEQ ID NOS: 91, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198,
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH sequence having at least 80%, 85%, 90%, 95%, 97% (or more)sequence identity to the sequence set forth in any one of SEQ ID NOS: 175, 177, 179, 181, 183, 185, 187,
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VL sequence having at least 80%, 85%, 90%, 95%, 97% (or morejsequence identity to the sequence set forth in any one of SEQ ID NOS: 91, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196,
  • the anti-CD38 antibody or antigen binding fragment thereof comprises a VH sequence having at least 90% sequence identity to the sequence set forth in any one of SEQ ID NOS: 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197,
  • the VH and VL comprise sequences having at least 80%, 85%, 90%, 95%, 97% (or more)sequence identity to the sequences set forth in SEQ ID NOS: 175 and 176, respectively; 177 and 178, respectively, 179 and 180, respectively, 181 and 182, respectively; 183 and 184, respectively; 185 and 186, respectively; 187 and 188, respectively; 189 and 190, respectively; 191 and 192, respectively; 193 and 194, respectively; 195 and 196, respectively; 197 and 198, respectively; 199 and 200, respectively; 201 and 202, respectively; 203 and 204, respectively; 205 and 206, respectively; 208 and 208, respectively; 209 and 210, respectively; 211 and 212, respectively; 213 and 214, respectively; 215 and 216, respectively; 217 and 218, respectively; 219 and 220, respectively; 221 and 222, respectively; 223 and 224, respectively; 225 and 22
  • the anti-CD38 antibody or antigen binding fragment thereof comprises the VH sequence set forth in any one of SEQ ID NOS: 175, 177, 179, 181, 183,
  • the anti-CD38 antibody or antigen binding fragment thereof comprises the VL sequence set forth in any one of SEQ ID NOS: 91, 176, 178, 180, 182, 184, 186, 188, 190,
  • the anti-CD38 antibody or antigen binding fragment thereof comprises the VH sequence set forth in any one of SEQ ID NOS: 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 228, 230, 232, 233, 235, 237, 239, 241, 243, 245,
  • the VH and VL comprise the sequences set forth in
  • the antibody variable regions are joined by a flexible linker.
  • the anti-CD38 antibody or antigen-binding fragment thereof is an scFv in which the VH and VL are joined by a linker.
  • the linker comprises a sequence having at least about 90%, at least about 95%, or at least about 99% sequence identity to the sequence of SEQ ID NO: 1 or SEQ ID NOG.
  • the linker comprises the sequence of SEQ ID NO: 1 or SEQ ID NO:3.
  • the linker comprises a sequence having at least about 90%, at least about 95%, or at least about 99% sequence identity to the sequence of SEQ ID NO:3.
  • the linker comprises the sequence of SEQ ID NO:3. In some embodiments, the linker contains only glycine and serine residues (“a Gly-Ser linker”). In some embodiments, the antibody comprises any Gly-Ser linker known in the art. In some embodiments, the linker comprises a sequence having at least about 90%, at least about 95%, or at least about 99% sequence identity to the sequence of SEQ ID NO: 1. In some embodiments, the linker comprises the sequence of SEQ ID NO: 1.
  • the scFv comprises the sequence of SEQ ID NO: 1.
  • the antibody is an scFv comprising the sequence of SEQ ID NO: 108, SEQ ID NO: 109, SEQ ID NO: 110, SEQ ID NO: 111, SEQ ID NO: 112, SEQ ID NO: 113, SEQ ID NO: 114, SEQ ID NO: 115, SEQ ID NO: 116, SEQ ID NO: 117, SEQ ID NO:118, or SEQ ID NO: 119.
  • the antibody comprises the sequence of SEQ ID NO: 108.
  • the antibody comprises the sequence of SEQ ID NO: 109.
  • the antibody comprises the sequence of SEQ ID NO: 110. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 111. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 112. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 113. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 114. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 115. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 116. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 117. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 118. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 119.
  • the antibody specifically binds to the CD38 protein.
  • CD38 refers to human CD38.
  • the antibody specifically binds to human CD38, such as an epitope or region of human CD381 , such as human CD38 described in SEQ ID NO:351 (e.g., UniProt P28907), or its allele variant or splice variant.
  • the antibody specifically binds to the extracellular domain of human CD38 (e.g., residues 43-300 of SEQ ID NO:351).
  • the antibody binds to non-human CD38, such as monkey, rabbit, rat, mouse, or other CD38species.
  • the antibody can bind to CD38 with at least a certain affinity, such as human CD38, as measured by any of a variety of known methods.
  • the affinity is represented by the dissociation constant (KD).
  • affinity is represented by EC 50.
  • the KD of the antibody to CD38 is from or from about 0.1 nM to 500 nM, 0.1 nM to 100 nM, 0.1 nM to 50 nM, 0.1 nM to 10 nM, 0.1 nM to 1 nM, 1 nM to 500 nM, 1 nM to 100 nM, 1 nM to 50 nM, 1 nM to 10 nM, 10 nM to 500 nM, 10 nM to 100 nM, 10 nM to 50 nM, 50 nM to 500 nM, 50 nM to 100 nM or 100 nM to 500 nM.
  • the KD of the antibody to CD38 (such as human CD38) is below or about below or below or about below or below: lOOnM, 60nM, 50nM, 40nM, 30nM , 25nM, 20nM, 19nM, 18nM, 17nM, 16nM, 15nM, 14nM, 13nM, 12nM, l lnM, lOnM, 9nM, 8nM, 7nM, 6nM, 5nM, 4nM, 3nM, 2nM or InM.
  • the characteristics or characteristics of the provided antibody are described in relation to the characteristics observed by another antibody (for example, a reference antibody).
  • the reference antibody is a human anti- CD38 antibody.
  • the reference antibody is a human antibody named “A2” (see, for example, PCT/US2016/026545 and PCT/IB2018/058642).
  • the reference antibody has a VH region containing the sequence set forth in SEQ ID NO: 106, or contains CDR1, CDR2, and CDR3 within this sequence; and a VL region containing the sequence set forth in SEQ ID NO: 107, or contains CDR1, CDR2, and CDR3 within this sequence.
  • the reference antibody may include the CDR-H1 sequence of SEQ ID NO:340, the CDR-H2 sequence of SEQ ID NO:341, the CDR-H3 sequence of SEQ ID NO:342, the CDR-L1 sequence of SEQ ID NO:343, the CDR- L2 sequence of SEQ ID NO:344, and the CDR-L3 sequence of SEQ ID NO:345.
  • the reference antibody includes the VH sequence of SEQ ID NO: 106 and the VL sequence of SEQ ID NO: 107.
  • the reference antibody is a scFv comprising the amino acid sequence set forth in SEQ ID NO: 120.
  • the reference antibody is formatted as a CAR.
  • the CAR comprises the sequence of SEQ ID NO:339.
  • the antibody has an affinity, e.g., KD, about the same as or lower than that of the corresponding form of the reference antibody, e.g., no more than about 1.5-fold or no more than about 2-fold greater, no more than 3-fold greater, and/or no more than 10-fold greater, than the KD of the corresponding form of the reference antibody.
  • the antibody has an affinity, e.g., KD, that is greater than or greater than about 5- fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80-fold, 90-fold, 100-fold, 150- fold, 200-fold, 250-fold or greater than the KD of the corresponding form of the reference antibody.
  • the antibodies display a binding preference for CD38- expressing cells as compared to CD38-negative cells.
  • the binding preference is observed where a significantly greater degree of binding is measured to the CD38-expressing, as compared to the non-expressing, cells.
  • the fold change in degree of binding detected for example, as measured by mean fluorescence intensity (MFI) in a flow cytometry-based assay and/or KD or ECso, to the CD38-expressing cells as compared to the non-CD38-expressing cells, is at least at or about 1.5, 2, 3, 4, 5, 6, 8, 10, 12, 14, 16, 18, or 20 or more, and/or is about as great, about the same, at least as great or at least about as great, or greater, than the fold change observed for the corresponding form of the reference antibody.
  • the total degree of observed binding to CD38 or to the CD38-expressing cells is approximately the same, at least as great, or greater than that observed for the corresponding form of the reference antibody.
  • the antibodies display a higher ratio of antigendependent activation to antigen-independent activation as compared to the corresponding form of the reference antibody.
  • the antibodies display a ratio of antigen-dependent activation to antigen-independent activation that is at least about 2-fold, 4-fold, 6-fold, 8-fold, or 10-fold higher than that observed for the corresponding form of the reference antibody.
  • the antibodies display a ratio of antigen-dependent activation to antigen-independent activation that is about 2-fold, 4-fold, 6-fold, 8-fold, or 10-fold higher than that observed for the corresponding form of the reference antibody.
  • the antibody is or is part of an immunoconjugate comprising a therapeutic agent.
  • the antibody is conjugated to one or more heterologous molecule(s), such as, but not limited to, a cytotoxic agent or a detection agent, (e.g., a radioisotope).
  • Cytotoxic agents include, but are not limited to, radioactive isotopes (e.g., At211, 1131, 1125, Y90, Rel86, Rel88, Sml53, Bi212, P32, Pb212 and radioactive isotopes of Lu); chemotherapeutic agents (e.g., methotrexate, adriamicin, vinca alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin or other intercalating agents); growth inhibitory agents; enzymes and fragments thereof such as nucleolytic enzymes; antibiotics; toxins such as small molecule toxins or enzymatically active toxins.
  • radioactive isotopes e.g., At211, 1131, 1125, Y90, Rel86, Rel88, Sml53, Bi212, P32, Pb212 and radioactive isotope
  • the antibody is conjugated to one or more cytotoxic agents, such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
  • cytotoxic agents such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
  • cytotoxic agents such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
  • ADCs antibody-drug conjugates
  • an antibody is conjugated to one or more drugs, including but not limited to an anthracycline such as da
  • the immunoconjugates are those in which the antibody is conjugated to an enzymatically active toxin or fragment thereof, including but not limited to diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP- S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes.
  • an enzymatically active toxin or fragment thereof including but not limited to diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain
  • radioactive isotopes include At 211 , 1131 , 1125 , Y 90 , Re 186 , Re 188 , Sm M , Bi 212 , P 32 , Pb 212 and radioactive isotopes of Lu.
  • Conjugates of an antibody and a therapeutic agent may be made using any of a number of known protein coupling agents, e.g., linkers.
  • the linker may be a "cleavable linker" facilitating release of a cytotoxic drug in the cell, such as acid-labile linkers, peptidase-sensitive linkers, photolahile linkers, dimethyl linkers, and disulfide-containing linkers.
  • the CD38-binding molecules e.g., antibodies or polypeptides such as chimeric receptors containing the same
  • the multispecific binding molecules are multispecific antibodies, including, e.g. bispecific.
  • Multispecific binding partners e.g., antibodies
  • one of the binding specificities is for CD38 and the other is for another antigen.
  • bispecific antibodies may bind to two different epitopes of CD38.
  • Bispecific antibodies may also be used to localize cytotoxic agents to cells which express CD38.
  • Bispecific antibodies can be prepared as full-length antibodies or antibody fragments.
  • multispecific antibodies are multispecific single-chain antibodies, e.g., diabodies, triabodies, and tetrabodies, tandem di-scFvs, and tandem tri-scFvs.
  • multispecific chimeric receptors such as multispecific CARs, containing the antibodies.
  • multispecific cells containing the antibodies or polypeptides including the same such as cells containing a cell surface protein including the anti-CD38 antibody and an additional cell surface protein, such as an additional chimeric receptor, which binds to a different antigen or a different epitope on CD38.
  • Non-limiting examples of additional antigens include B cell specific antigens, other tumor-specific antigens, such as antigens expressed specifically on or associated with B cell leukemia, lymphoma, B cell chronic lymphocytic leukemia (CLL), AML, acute myeloid leukemia (AML), acute lymphocytic leukemia (ALL), Burkett's Lymphoma, mantle cell lymphoma (MCL), non-small cell lung cancer (NSCLC), neuroblastoma, renal cell carcinoma, colon cancer, colorectal cancer, breast cancer, epithelial squamous cell cancer, melanoma, myeloma, stomach cancer, brain cancer, lung cancer, pancreatic cancer, cervical cancer, ovarian cancer, liver cancer, bladder cancer, prostate cancer, testicular cancer, thyroid cancer, uterine cancer, adrenal cancer and/or head and neck cancer, and antigens expressed on NK cells or T cells.
  • B cell specific antigens such as antigens expressed specifically on or associated with B
  • Non-limiting examples of antigens include BCMA, CD3, CD4, CD8, CD19, CD20, CD21, CD22, CD23, CD33, CD39, CD40, CD40L, CD138, CD203, FcRH5, GPRC5D, and SLAMF7.
  • the example of an additional antigen is a T cell antigen. In some embodiments, the additional example of an antigen is CD3. In some embodiments, the example of an additional antigen is a NK cell antigen. In some embodiments, the additional example of an antigen is CD16, NKp30, NKp46, or NKG2D, or a combination thereof. In some embodiments, the additional example of an antigen is CD 16. In some embodiments, the additional example of an antigen is NKp30. In some embodiments, the additional example of an antigen is NKp46. In some embodiments, the additional example of an antigen is NKG2D.
  • a bispecific molecule comprises any of the anti-CD38 antibodies or antigen-binding fragments thereof provided herein and a second moiety that has a binding specificity for an immune cell.
  • the immune cell is a NK cell.
  • the second moiety binds to CD 16, NKp30, NKp46, or NKG2D.
  • the second moiety binds to CD16.
  • the second moiety binds to NKp30.
  • the second moiety binds to NKp46.
  • the second moiety binds to NKG2D.
  • the immune cell is a T cell.
  • the second moiety binds to CD3. iv. Variants
  • amino acid sequences that correspond to any of the nucleic acids disclosed herein (and/or included in the accompanying sequence listing), while accounting for degeneracy of the nucleic acid code.
  • those sequences that vary from those expressly disclosed herein (and/or included in the accompanying sequence listing), but have functional similarity or equivalency are also contemplated within the scope of the present disclosure.
  • the foregoing includes mutants, truncations, substitutions, codon optimization, or other types of modifications.
  • any of the sequences may be used, or a truncated or mutated form of any of the sequences disclosed herein (and/or included in the accompanying sequence listing) may be used and in any combination.
  • Sequences provided for herein that include an identifier, such as a tag or other detectable sequence (e.g., a Flag tag) are also provided for herein with the absence of such a tag or other detectable sequence (e.g., excluding the Flag tag from the listed sequence).
  • a Sequence Listing in electronic format is submitted herewith. Some of the sequences provided in the Sequence Listing may be designated as Artificial Sequences by virtue of being non-naturally occurring fragments or portions of other sequences, including naturally occurring sequences. Some of the sequences provided in the Sequence Listing may be designated as Artificial Sequences by virtue of being combinations of sequences from different origins, such as humanized antibody sequences.
  • the antibodies include one or more amino acid variations, e.g., substitutions, deletions, insertions, and/or mutations, compared to the sequence of an antibody described herein.
  • variants include those designed to improve the binding affinity and/or other biological properties of the antibody.
  • Amino acid sequence variants of an antibody may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the antibody, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of residues within the amino acid sequences of the antibody.
  • the antibodies include one or more amino acid substitutions, e.g., as compared to an antibody sequence described herein and/or compared to a sequence of a natural repertoire, e.g., human repertoire.
  • Sites of interest for substitutional mutagenesis include the CDRs and FRs.
  • Amino acid substitutions may be introduced into an antibody of interest and the products screened for a desired activity, e.g., retained/improved antigen binding, decreased immunogenicity, improved half-life, and/or improved effector function, such as the ability to promote antibody-dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC).
  • Insertional variants of the antibody molecule include the fusion to the N- or C-terminus of the antibody to an enzyme or a polypeptide which increases the serum half-life of the antibody.
  • the provided binding molecules e.g., CD38-binding molecules
  • the receptors include antigen receptors and other chimeric receptors that specifically bind to CD38, such as receptors containing the provided anti-CD38 antibodies, e.g., antibody fragments.
  • the antigen receptors are chimeric antigen receptors (CARs).
  • CARs chimeric antigen receptors
  • cells expressing the recombinant receptors and uses thereof in treatment of diseases and conditions associated with CD38 expression The chimeric receptors, such as CARs, generally include an extracellular antigen-binding domain that includes, is, or is comprised within, one of the provided anti-CD38 antibodies.
  • the chimeric receptors typically include in their extracellular portion one or more CD38-binding molecules, such as one or more antigen-binding fragment as described herein.
  • the CAR includes a heavy chain variable region (VH) and a light chain variable region (VL) region of the antibody, e.g., an scFv antibody fragment.
  • the antigen-binding domain may be or comprise any of those as described herein, e.g., in Section LA.
  • the antigen-binding domain is an scFv comprising a VH and a VL joined by a linker (e.g., SEQ ID NO:1 or SEQ ID NO:3).
  • the antigen-binding domain is an scFv comprising the linker set forth in SEQ ID NO:1.
  • the antigen-binding domain comprises the amino acid sequence set forth in any one of SEQ ID NOS: 108-119.
  • the antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 108.
  • the antigenbinding domain comprises the amino acid sequence of SEQ ID NO: 109. In some embodiments, the antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 110. In some embodiments, the antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 111. In some embodiments, the antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 112. In some embodiments, the antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 113. In some embodiments, the antigen- binding domain comprises the amino acid sequence of SEQ ID NO: 114. In some embodiments, the antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 115.
  • the antigenbinding domain comprises the amino acid sequence of SEQ ID NO: 116. In some embodiments, the antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 117. In some embodiments, the antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 118. In some embodiments, the antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 119.
  • the antigen-binding domain generally is linked to an intracellular signaling domain comprising intracellular signaling components, such as signaling components that mimic activation through an antigen receptor complex, such as a TCR complex, in the case of a CAR.
  • the antigen-binding domain is linked to an intracellular signaling domain by a transmembrane domain.
  • the CD38-binding molecule e.g., antibody
  • the CD38-binding molecule is linked to a transmembrane and intracellular signaling domain.
  • the transmembrane domain is fused to the extracellular domain.
  • the transmembrane domain in some embodiments is derived either from a natural or from a synthetic source. Where the source is natural, the domain in some aspects is derived from any membrane-bound or transmembrane protein.
  • Transmembrane regions include those derived from (e.g., comprising at least the transmembrane region(s) of) CD3, CD4, CD5, CD8, CD9, CD 16, CD22, CD28, CD33, CD37, CD45, CD64, CD80, CD86, CD134, CD137, CD 154, or a combination thereof.
  • the transmembrane domain in some embodiments is synthetic.
  • the transmembrane domain comprises at least a portion of CD8, a transmembrane glycoprotein normally expressed on both T cells and NK cells.
  • the transmembrane domain comprises CD8alpha (CD8a).
  • the transmembrane domain comprises a CD8 (e.g., CD8a) hinge and a CD8 (e.g., CD8a) transmembrane region.
  • the transmembrane domain comprises a hinge, e.g. a CD8a hinge.
  • the sequence encoding the CD8a hinge is truncated or modified.
  • the CD8a hinge is encoded by a nucleic acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the nucleic acid sequence of SEQ ID NO:5.
  • the CD8a hinge comprises the nucleic acid sequence of SEQ ID NO:5.
  • the CD8a hinge is truncated or modified.
  • the CD8a hinge has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the amino acid sequence of SEQ ID NO:6.
  • the hinge of CD8a comprises the amino acid sequence of SEQ ID NO:6.
  • the transmembrane domain comprises a CD8a transmembrane region.
  • the CD8a transmembrane region is truncated or modified.
  • the CD8a transmembrane region is encoded by a nucleic acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the sequence of SEQ ID NO:7.
  • the CD8a transmembrane region is encoded by a nucleic acid sequence of SEQ ID NO:7.
  • the CD8a transmembrane region is truncated or modified.
  • the CD8a transmembrane region has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the sequence of SEQ ID NO:8. In several embodiments, the CD8a transmembrane region comprises the amino acid sequence of SEQ ID NO:8.
  • the CD8 transmembrane domain is truncated or modified and is encoded by a nucleic acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the sequence of SEQ ID NO:9.
  • the CD8 transmembrane domain is encoded by the nucleic acid sequence of SEQ ID NO:9.
  • the CD8 transmembrane domain is truncated or modified and comprises an amino acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the sequence of SEQ ID NO: 10.
  • the transmembrane domain comprises the amino acid sequence of SEQ ID NO: 10.
  • the transmembrane domain comprises a CD28 transmembrane domain or a fragment thereof.
  • the CD28 transmembrane domain is truncated or modified.
  • the CD28 transmembrane domain has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the amino acid sequence of SEQ ID NOT E
  • the CD28 transmembrane domain comprises the amino acid sequence of SEQ ID NO: 11.
  • the receptor e.g., the CAR
  • the receptor generally includes an intracellular signaling domain comprising intracellular signaling components.
  • the receptor includes an intracellular component of a TCR complex, such as a TCR CD3 chain that mediates T-cell activation and cytotoxicity, e.g., CD3 zeta chain.
  • the antigenbinding portion is linked to one or more cell signaling modules.
  • the cytoplasmic domain or intracellular signaling domain of the receptor activates at least one of the normal effector functions or responses of the immune cell, e.g., NK cell engineered to express the CAR.
  • the CAR induces a function of an immune cell (e.g., NK cell) such as cytolytic activity and/or secretion of cytokines or other factors.
  • an immune cell e.g., NK cell
  • a truncated portion of an intracellular signaling domain of an antigen receptor component or costimulatory molecule is used in place of an intact immunostimulatory chain, for example, if it transduces the effector function signal.
  • the intracellular signaling domain includes the cytoplasmic sequences of the T cell receptor (TCR), and in some aspects, also those of co- receptors that in the natural context act in concert with such receptors to initiate signal transduction following antigen receptor engagement.
  • T cell activation is in some aspects described as being mediated by two classes of cytoplasmic signaling sequences: those that initiate antigen-dependent primary activation through the TCR (primary cytoplasmic signaling sequences), and those that act in an antigen independent manner to provide a secondary or co-stimulatory signal (secondary cytoplasmic signaling sequences).
  • primary cytoplasmic signaling sequences those that initiate antigen-dependent primary activation through the TCR
  • secondary cytoplasmic signaling sequences those that act in an antigen independent manner to provide a secondary or co-stimulatory signal.
  • the receptor includes one or both of such signaling components.
  • the receptor includes a primary cytoplasmic signaling sequence that regulates primary activation of the TCR complex.
  • Primary cytoplasmic signaling sequences that act in a stimulatory manner may contain signaling motifs which are known as immunoreceptor tyrosine -based activation motifs or ITAMs.
  • ITAM containing primary cytoplasmic signaling sequences include those derived from TCR zeta, FcR gamma, FcR beta, CD3 gamma, CD3 delta, CD3 epsilon, CDS, CD22, CD79a, CD79b, and CD66d.
  • cytoplasmic signaling molecule(s) in the CAR contain(s) a cytoplasmic signaling domain, portion thereof, or sequence derived from CD3zeta.
  • immune cells engineered according to several embodiments disclosed herein may comprise at least one subunit of the CD3 T cell receptor complex (or a fragment thereof).
  • the signaling domain comprises the CD3 zeta subunit.
  • the CD3zeta can be truncated or modified.
  • the CD3zeta is encoded by a nucleic acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the nucleic acid sequence of SEQ ID NO: 17.
  • the CD3zeta is encoded by the nucleic acid sequence of SEQ ID NO: 17.
  • the CD3zeta is truncated or modified. In some embodiments, the CD3zeta comprises an amino acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 18. In several embodiments, the CD3zeta comprises the amino acid sequence of SEQ ID NO: 18.
  • the intracellular signaling domain comprises a costimulatory signaling region, such as an intracellular signaling region of CD28, 4- IBB, 0X40, DAP10, ICOS, or any combination thereof.
  • the same receptor includes both a CD3zeta and a costimulatory signaling region.
  • the intracellular signaling domain of the recombinant receptor such as CAR, comprises a CD3zeta intracellular domain and a costimulatory signaling region.
  • the intracellular signaling domain comprises an intracellular signaling region of 0X40.
  • the 0X40 intracellular signaling region is encoded by a nucleic acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the nucleic acid sequence of SEQ ID NO: 13. In several embodiments, the 0X40 intracellular signaling region is encoded by the nucleic acid sequence of SEQ ID NO: 13. In several embodiments, the 0X40 intracellular signaling region comprises an amino acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 14.
  • the 0X40 intracellular signaling region comprises the amino acid sequence of SEQ ID NO: 14.
  • 0X40 is used as the sole intracellular signaling component in the construct, however, in several embodiments, 0X40 can be used with one or more other components.
  • combinations of 0X40 and CD3zeta are used in some embodiments.
  • the intracellular signaling domain comprises an 0X40 costimulatory signaling region linked to CD3zeta.
  • the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:327, SEQ ID NO:328, SEQ ID NO: 329, SEQ ID NO:330, SEQ ID NO:331, SEQ ID NO:332, SEQ ID NO:333, SEQ ID NO:334, SEQ ID NO:335, SEQ ID NO:336, SEQ ID NO:337, or SEQ ID NO:338.
  • the chimeric receptor comprises the amino acid sequence set forth in SEQ ID NO:327.
  • the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:328.
  • the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO: 329. In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:330. In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:33L In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:332. In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:333.
  • the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:334. In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:335. In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO: 336. In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:337. In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:338.
  • CD28, 0X40, 4-1BB and/or CD3zeta are used in some embodiments.
  • the intracellular signaling domain comprises an intracellular signaling region of 4-1BB.
  • the 4-1BB intracellular signaling region is encoded by a nucleic acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the nucleic acid sequence of SEQ ID NO: 15.
  • the 4-1BB intracellular signaling region is encoded by the nucleic acid sequence of SEQ ID NO: 15.
  • the 4-1BB intracellular signaling region comprises an amino acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 16.
  • the 4-1BB intracellular signaling region comprises the amino acid sequence of SEQ ID NO: 16.
  • 4-1BB is used as the sole intracellular signaling component in the construct, however, in several embodiments, 4- IBB can be used with one or more other components.
  • the intracellular signaling domain comprises a 4- IBB costimulatory signaling region linked to CD3zeta.
  • combinations of CD28, 0X40, 4-1BB and/or CD3zeta are used in some embodiments.
  • the intracellular signaling domain comprises an intracellular signaling region of CD28.
  • the CD28 intracellular signaling region comprises an amino acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 12.
  • the CD28 intracellular signaling region comprises the amino acid sequence of SEQ ID NO: 12.
  • CD28 is used as the sole intracellular signaling component in the construct, however, in several embodiments, CD28 can be used with one or more other components. For example, combinations of CD28 and CD3zeta are used in some embodiments.
  • the intracellular signaling domain comprises a CD28 costimulatory signaling region linked to CD3zeta.
  • CD28, 0X40, 4-1BB and/or CD3zeta are used in some embodiments.
  • the nucleic acid encoding the chimeric receptor, or a portion thereof is codon-optimized.
  • the polynucleotides are optimized, or contain certain features designed for optimization, such as for codon usage, to reduce RNA heterogeneity and/or to modify, e.g., increase or render more consistent among cell product lots, expression, such as surface expression, of the encoded receptor.
  • polynucleotides, encoding chimeric receptors are modified as compared to a reference polynucleotide, such as to remove cryptic or hidden splice sites, to reduce RNA heterogeneity.
  • polynucleotides, encoding chimeric receptors are codon optimized, such as for expression in a mammalian, e.g., human, cell such as in a human T cell.
  • the modified polynucleotides result in in improved, e.g., increased or more uniform or more consistent level of, expression, e.g., surface expression, when expressed in a cell.
  • the genetic engineering involves introduction of a nucleic acid encoding the genetically engineered component or other component for introduction into the cell, such as a component encoding a gene-disrupting protein or nucleic acid.
  • genes for introduction are those to improve the efficacy of therapy, such as by promoting viability and/or function of transferred cells; genes to provide a genetic marker for selection and/or evaluation of the cells, such as to assess in vivo survival or localization; genes to improve safety, for example, by making the cell susceptible to negative selection in vivo.
  • binding molecules e.g., anti-CD38 binding molecules
  • recombinant receptors e.g., CARs
  • one or more binding molecules, including recombinant receptors can be genetically engineered into cells or a plurality of cells.
  • the genetic engineering generally involves introduction of a nucleic acid encoding the recombinant or engineered component into the cell, such as by retroviral transduction, transfection, or transformation.
  • polynucleotides encoding the antibodies and chimeric antigen receptors and/or portions, e.g., chains, thereof.
  • the provided polynucleotides are those encoding the anti-CD38 chimeric antigen receptors (e.g., antigen-binding fragment) described herein.
  • polynucleotides encoding one or more antibodies and/or portions thereof e.g., those encoding one or more of the anti-CD38 antibodies (e.g., antigenbinding fragment) described herein and/or other antibodies and/or portions thereof, e.g., antibodies and/or portions thereof that binds other target antigens.
  • the polynucleotides may include those encompassing natural and/or non-naturally occurring nucleotides and bases, e.g., including those with backbone modifications.
  • the terms “nucleic acid molecule”, “nucleic acid” and “polynucleotide” may be used interchangeably, and refer to a polymer of nucleotides. Such polymers of nucleotides may contain natural and/or non-natural nucleotides, and include, but are not limited to, DNA, RNA, and PNA.
  • Nucleic acid sequence refers to the linear sequence of nucleotides that comprise the nucleic acid molecule or polynucleotide. Also provided are polynucleotides that have been optimized for codon usage.
  • vectors containing the polynucleotides such as any of the polynucleotides described herein, and cells containing the vectors, e.g., for producing the antibodies or antigen-binding fragments thereof.
  • the vector is a viral vector.
  • the vector is a retroviral vector.
  • the vector is a lentiviral vector.
  • the nucleic acid may encode an amino acid sequence comprising the VL region and/or an amino acid sequence comprising the VH region of the antibody (e.g., the light and/or heavy chains of the antibody).
  • the nucleic acid may encode one or more amino acid sequence comprising the VL region and/or an amino acid sequence comprising the VH region of the antibody (e.g., the light and/or heavy chains of the antibody).
  • one or more vectors e.g., expression vectors
  • a host cell comprising such polynucleotides is provided.
  • a host cell comprises (e.g., has been transformed with) (1) a vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL region of the antibody and an amino acid sequence comprising the VH region of the antibody, or (2) a first vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL region of the antibody and a second vector comprising a nucleic acid that encodes an amino acid sequence comprising the VH region of the antibody.
  • a host cell comprises (e.g., has been transformed with) one or more vectors comprising one or more nucleic acid that encodes one or more an amino acid sequence comprising one or more antibodies and/or portions thereof, e.g., antigen-binding fragments thereof.
  • one or more such host cells are provided.
  • a composition containing one or more such host cells are provided.
  • the one or more host cells can express different antibodies, or the same antibody.
  • each of the host cells can express more than one antibody.
  • nucleic acid sequence encoding a chimeric receptor antibody may be isolated and inserted into one or more vectors for further cloning and/or expression in a host cell.
  • nucleic acid sequences may be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody).
  • a method of making the anti-CD38 chimeric antigen receptor comprises culturing a host cell comprising a nucleic acid sequence encoding the antibody, as provided above, under conditions suitable for expression of the receptor.
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for antibody-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been modified to mimic or approximate those in human cells, resulting in the production of an antibody with a partially or fully human glycosylation pattern.
  • eukaryotic cells that may be used to express polypeptides, including isolated or secreted polypeptides, include, but are not limited to, COS cells, including COS-7 cells; 293 cells, including 293-6E cells; CHO cells, including CHO-S, DG44.
  • the antibody heavy chains and/or light chains may be expressed in yeast. See, e.g., US Publication No. US 2006/0270045 Al.
  • a particular eukaryotic host cell is selected based on its ability to make desired post-translational modifications to the heavy chains and/or light chains (e.g., VH region and/or VL region).
  • CHO cells produce polypeptides that have a higher level of sialylation than the same polypeptide produced in 293 cells.
  • immune cells such as human immune cells are used to express the provided polypeptides encoding chimeric antigen receptors.
  • the immune cells are NK cells including primary NK cells, T cells (e.g., CD4+ and/or CD8+ immune cells) including primary T cells, or any combination thereof.
  • gene transfer is accomplished by first stimulating the cell, such as by combining it with a stimulus that induces a response such as proliferation, survival, and/or activation, e.g., as measured by expression of a cytokine or activation marker, followed by transduction of the activated cells, and expansion in culture to numbers sufficient for clinical applications.
  • a stimulus such as proliferation, survival, and/or activation, e.g., as measured by expression of a cytokine or activation marker
  • the cells further are engineered to promote expression of cytokines or other factors.
  • cytokines e.g., antigen receptors, e.g., CARs
  • exemplary methods include those for transfer of polynucleotides encoding the receptors, including via viral, e.g., retroviral or lentiviral, transduction, transposons, and electroporation.
  • recombinant polynucleotides are transferred into immune cells (e.g., NK or T cells) using recombinant infectious virus particles, such as, e.g., vectors derived from simian virus 40 (SV40), adenoviruses, adeno-associated virus (AAV).
  • recombinant polynucleotides are transferred into immune cells using recombinant lentiviral vectors or retroviral vectors, such as gamma-retroviral vectors.
  • the retroviral vector has a long terminal repeat sequence (LTR), e.g., a retroviral vector derived from the Moloney murine leukemia virus (MoMLV), myeloproliferative sarcoma virus (MPSV), murine embryonic stem cell virus (MESV), murine stem cell virus (MSCV), spleen focus forming virus (SFFV), or human immunodeficiency virus type 1 (HIV-1).
  • LTR long terminal repeat sequence
  • MoMLV Moloney murine leukemia virus
  • MPSV myeloproliferative sarcoma virus
  • MMV murine embryonic stem cell virus
  • MSCV murine stem cell virus
  • SFFV spleen focus forming virus
  • HSV human immunodeficiency virus type 1
  • retroviral vectors are derived from murine retroviruses.
  • the retroviruses include those derived from any avian or mammalian cell source.
  • the retroviruses typically are amphotropic, meaning
  • recombinant polynucleotides are transferred into immune cells (e.g., NK or T cells) via electroporation. In some embodiments, recombinant polynucleotides are transferred into immune cells (e.g., NK or T cells) via transposition.
  • immune cells e.g., NK or T cells
  • Other methods of introducing and expressing genetic material in immune cells include calcium phosphate transfection, protoplast fusion, cationic liposome-mediated transfection; tungsten particle-facilitated microparticle bombardment, and strontium phosphate DNA co-precipitation.
  • genes for introduction are those to improve the efficacy of therapy, such as by promoting viability and/or function of transferred cells; genes to provide a genetic marker for selection and/or evaluation of the cells, such as to assess in vivo survival or localization; genes to improve safety, for example, by making the cell susceptible to negative selection in vivo.
  • one or more binding molecules can be genetically engineered to be expressed in cells or a plurality of cells.
  • a first recombinant receptor and a second binding molecule e.g., recombinant receptor
  • additional binding molecules are engineered to be expressed in cells or a plurality of cells.
  • the polynucleotide containing nucleic acid sequences encoding the CD38-binding receptor contains a signal sequence that encodes a signal peptide.
  • the signal sequence may encode a signal peptide derived from a native polypeptide.
  • the signal sequence may encode a heterologous or non-native signal peptide.
  • a non-limiting example of a signal peptide comprises a CD8 alpha (CD8a) signal peptide set forth in SEQ ID NO:4.
  • the vector or construct can contain promoter and/or enhancer or regulatory elements to regulate expression of the encoded recombinant receptor.
  • the promoter and/or enhancer or regulatory elements can be condition-dependent promoters, enhancers, and/or regulatory elements. In some examples these elements drive expression of the transgene.
  • the vector or construct can contain a single promoter that drives the expression of one or more nucleic acid molecules.
  • nucleic acid molecules e.g., transcripts
  • transcription units can be engineered as a bicistronic unit containing an IRES (internal ribosome entry site), which allows coexpression of gene products (e.g. encoding a chimeric receptor and membrane- bound interleukin- 15) by a message from a single promoter.
  • IRES internal ribosome entry site
  • a single promoter may direct expression of an RNA that contains, in a single open reading frame (ORF), two or three genes (e.g. encoding a chimeric receptor and membrane -bound interleukin- 15) separated from one another by sequences encoding a self-cleavage peptide (e.g., 2A cleavage sequences) or a protease recognition site.
  • ORF open reading frame
  • the ORF thus encodes a single polypeptide, which, either during (in the case of T2A) or after translation, is cleaved into the individual proteins.
  • the peptide such as T2A
  • Many 2A elements are known. Examples of 2A sequences that can be used in the methods and polynucleotides disclosed herein, without limitation, 2A sequences from the foot- and-mouth disease virus (F2A), equine rhinitis A virus (E2A), Thosea asigna virus (T2A, e.g. SEQ ID NO:20, encoded by SEQ ID NO: 19), and porcine teschovirus- 1 (P2A).
  • F2A foot- and-mouth disease virus
  • E2A equine rhinitis A virus
  • T2A e.g. SEQ ID NO:20, encoded by SEQ ID NO: 19
  • porcine teschovirus- 1 P2A
  • the one or more different or separate promoters drive the expression of a nucleic acid molecule encoding a binding molecule, e.g., recombinant receptor and a nucleic acid encoding membrane-bound interleukin- 15.
  • binding molecules e.g., antibodies and/or recombinant receptors provided herein, e.g., CD38-binding molecules and/or the additional recombinant receptors
  • polynucleotides containing one or more nucleic acid molecules encoding the receptors in any combinations or arrangements.
  • one, two, three or more polynucleotides can encode one, two, three or more different receptors or domains.
  • one vector or construct contains nucleic acid molecules encoding one or more binding molecules, e.g., antibody and/or recombinant receptor, and a separate vector or construct contains nucleic acid molecules encoding an additional binding molecule, e.g., antibody and/or recombinant receptor.
  • compositions containing one or more of the nucleic acid molecules, vectors or constructs such as any described above.
  • the nucleic acid molecules, vectors, constructs or compositions can be used to engineer immune cells, such as NK or T cells, to express any of the binding molecules, e.g., antibody or recombinant receptor, and/or the additional binding molecules.
  • a variety of cell types can be engineered to express a CD38-binding molecule or a chimeric receptor incorporating the same. Further, as elaborated on in more detail below, and as described in the Examples, genetic modifications (e.g., to CD38) can be made to these cells in order to enhance one or more aspects of their efficacy (e.g., cytotoxicity) and/or persistence (e.g., in vivo life span).
  • immune cells e.g., NK cells
  • gene editing of the immune cell can advantageously impart to the edited immune cell the ability to resist and/or overcome various inhibitory signals that are generated in the tumor microenvironment (TME).
  • TME tumor microenvironment
  • TME tumor microenvironment
  • gene editing of the immune cell limits this TME suppressive effect on the immune cells, or any edited/engineered immune cell provided for herein.
  • gene editing is employed to reduce or knockout expression of target proteins, for example by disrupting the underlying gene encoding the protein.
  • genetic editing of a gene as disclosed herein is accomplished through targeted introduction of DNA breakage, and a subsequent DNA repair mechanism.
  • double strand breaks of DNA are repaired by non- homologous end joining (NHEJ), wherein enzymes are used to directly join the DNA ends to one another to repair the break.
  • NHEJ non- homologous end joining
  • enzymes are used to directly join the DNA ends to one another to repair the break.
  • NHEJ is an error-prone process.
  • the NHEJ process re-ligates the ends of the cleaved DNA strands, which frequently results in nucleotide deletions and insertions at the cleavage site.
  • double strand breaks are repaired by homology directed repair (HDR), which is advantageously more accurate, thereby allowing sequence specific breaks and repair.
  • HDR uses a homologous sequence as a template for regeneration of missing DNA sequences at the break point, such as a vector with the desired genetic elements (e.g., an insertion element to disrupt the coding sequence of a gene) within a sequence that is homologous to the flanking sequences of a double strand break. This will result in the desired change (e.g., insertion) being inserted at the site of the DSB.
  • the HDR pathway can occur by way of the canonical HDR pathway or the alternative HDR pathway.
  • the term “HDR” or “homology-directed repair” as used herein encompasses both canonical HDR and alternative HDR.
  • Canonical HDR or “canonical homology-directed repair” or cHDR,” are used interchangeably, and refers to the process of repairing DNA damage using a homologous nucleic acid (e.g., an endogenous homologous sequence, such as a sister chromatid; or an exogenous nucleic acid, such as a donor template).
  • a homologous nucleic acid e.g., an endogenous homologous sequence, such as a sister chromatid; or an exogenous nucleic acid, such as a donor template.
  • Canonical HDR typically acts when there has been a significant resection at the DSB, forming at least one single-stranded portion of DNA.
  • canonical HDR typically involves a series of steps such as recognition of the break, stabilization of the break, resection, stabilization of single-stranded DNA, formation of a DNA crossover intermediate, resolution of the crossover intermediate, and ligation.
  • the canonical HDR process requires RAD51 and BRCA2, and the homologous nucleic acid, e.g., repair template, is typically double-stranded.
  • a double-stranded polynucleotide e.g., a doublestranded repair template
  • a double-stranded polynucleotide which comprises a sequence that is homologous to the targeting sequence, and which will either be directly integrated into the targeting sequence or will be used as a template to insert the sequence, or a portion the sequence, of the repair template into the target gene.
  • repair can progress by different pathways, e.g., by the double Holliday junction model (also referred to as the double strand break repair, or DSBR, pathway), or by the synthesis-dependent strand annealing (SDSA) pathway.
  • the double Holliday junction model also referred to as the double strand break repair, or DSBR, pathway
  • SDSA synthesis-dependent strand annealing
  • strand invasion occurs by the two single stranded overhangs of the targeting sequence to the homologous sequences in the doublestranded polynucleotide, e.g., double stranded donor template, which results in the formation of an intermediate with two Holliday junctions.
  • the junctions migrate as new DNA is synthesized from the ends of the invading strand to fill the gap resulting from the resection.
  • the end of the newly synthesized DNA is ligated to the resected end, and the junctions are resolved, resulting in the insertion at the targeting sequence, or a portion of the targeting sequence that includes the gene variant.
  • Crossover with the polynucleotide, e.g., repair template may occur upon resolution of the junctions.
  • Alternative HDR or “alternative homology-directed repair,” or “alternative HDR,” are used interchangeably, and refers, in some embodiments, to the process of repairing DNA damage using a homologous nucleic acid (e.g., an endogenous homologous sequence, such as a sister chromatid; or an exogenous nucleic acid, such as a repair template).
  • a homologous nucleic acid e.g., an endogenous homologous sequence, such as a sister chromatid; or an exogenous nucleic acid, such as a repair template.
  • Alternative HDR is distinct from canonical HDR in that the process utilizes different pathways from canonical HDR, and can be inhibited by the canonical HDR mediators, RAD51 and BRCA2.
  • alternative HDR is also distinguished by the involvement of a single-stranded or nicked homologous nucleic acid template, e.g., repair template
  • canonical HDR generally involves a double-stranded homologous template.
  • a single strand template polynucleotide e.g., repair template
  • a nick, single strand break, or DSB at the cleavage site, for altering a desired target site, e.g., a gene variant in a target gene is mediated by a nuclease molecule, and resection at the break occurs to reveal single stranded overhangs.
  • HDR is carried out by introducing, into a cell, one or more agent(s) capable of inducing a DSB, and a repair template, e.g., a single-stranded oligonucleotide.
  • the introducing can be carried out by any suitable delivery.
  • the conditions under which HDR is allowed to occur can be any conditions suitable for carrying out HDR in a cell.
  • gene editing is accomplished by one or more of a variety of engineered nucleases.
  • restriction enzymes are used, particularly when double strand breaks are desired at multiple regions.
  • a bioengineered nuclease is used.
  • ZFN Zinc Finger Nuclease
  • TALEN transcription-activator like effector nuclease
  • CRISPR/Cas9 clustered regularly interspaced short palindromic repeats
  • Meganucleases are characterized by their capacity to recognize and cut large DNA sequences (from 14 to 40 base pairs).
  • a meganuclease from the LAGLIDADG family is used, and is subjected to mutagenesis and screening to generate a meganuclease variant that recognizes a unique sequence(s), such as a specific site in a gene (e.g., CD38).
  • two or more meganucleases, or functions fragments thereof are fused to create a hybrid enzyme that recognize a desired target sequence within a gene (e.g., CD38).
  • ZFNs and TALEN function based on a nonspecific DNA cutting catalytic domain which is linked to specific DNA sequence recognizing peptides such as zinc fingers or transcription activator-like effectors (TALEs).
  • TALEs transcription activator-like effectors
  • the ZFNs and TALENs thus allow sequence-independent cleavage of DNA, with a high degree of sequence-specificity in target recognition.
  • Zinc finger motifs naturally function in transcription factors to recognize specific DNA sequences for transcription. The C-terminal part of each finger is responsible for the specific recognition of the DNA sequence.
  • ZFNs While the sequences recognized by ZFNs are relatively short, (e.g., ⁇ 3 base pairs), in several embodiments, combinations of 2, 3, 4, 5, 6, 7, 8, 9, 10 or more zinc fingers whose recognition sites have been characterized are used, thereby allowing targeting of specific sequences, such as a portion of CD38.
  • the combined ZFNs are then fused with the catalytic domain(s) of an endonuclease, such as FokI (optionally a FokI heterodimer), in order to induce a targeted DNA break.
  • FokI an endonuclease
  • TALENs Transcription activator-like effector nucleases
  • ZFNs Transcription activator-like effector nucleases
  • TALENs are specific DNA- binding proteins that feature an array of 33 or 34-amino acid repeats.
  • TALENs are a fusion of a DNA cutting domain of a nuclease to TALE domains, which allow for sequenceindependent introduction of double stranded DNA breaks with highly precise target site recognition.
  • TALENs can create double strand breaks at the target site that can be repaired by error-prone non-homologous end-joining (NHEJ), resulting in gene disruptions through the introduction of small insertions or deletions.
  • NHEJ error-prone non-homologous end-joining
  • TALENs are used in several embodiments, at least in part due to their higher specificity in DNA binding, reduced off-target effects, and ease in construction of the DNA-binding domain.
  • CRISPRs Clustered Regularly Interspaced Short Palindromic Repeats
  • the repeats are short sequences that originate from viral genomes and have been incorporated into the bacterial genome.
  • Cas CRISPR associated proteins
  • CRISPR is used to manipulate a gene (e.g., CD38).
  • a Class 1 or Class 2 Cas is used.
  • a Class 1 Cas is used and the Cas type is selected from the following types: I, IA, IB, IC, ID, IE, IF, IU, III, IIIA, IIIB, IIIC, HID, IV IVA, IVB, and combinations thereof.
  • the Cas is selected from the group consisting of Cas3, Cas8a, Cas5, Cas8b, Cas8c, CaslOd, Csel, Cse2, Csyl, Csy2, Csy3, GSU0054, CaslO, Csm2, Cmr5, CaslO, Csxl l, CsxlO, Csfl, and combinations thereof.
  • a Class 2 Cas is used and the Cas type is selected from the following types: n, HA, IIB, IIC, V, VI, and combinations thereof.
  • the Cas is selected from the group consisting of Cas9, Csn2, Cas4, Casl2a (previously known as Cpfl), C2cl, C2c3, Cas 13a (previously known as C2c2), Cas 13b, Cas 13c, CasX, CasY and combinations thereof.
  • the Cas is Cas9.
  • class 2 CasX is used, wherein CasX is capable of forming a complex with a guide nucleic acid and wherein the complex can bind to a target DNA, and wherein the target DNA comprises a non-target strand and a target strand.
  • class 2 CasY is used, wherein CasY is capable of binding and modifying a target nucleic acid and/or a polypeptide associated with target nucleic acid.
  • CD38 plays a role in the maturation cycle of immune cells, and hematological cancers and solid tumors can often present upregulated CD38. Loss of CD38 expression on constituent NK cells allows for greater cytotoxicity due to decreased fratricide (Nagai et al., Blood (2019) 134 (suppl. l):870). Wild-type NK cells self-express CD38, leading to downstream self-targeting effects in wild-type NK cells. For T cells, loss of CD38 expression for constituent T cells leads to increased cytotoxicity.
  • any of the engineered cells provided herein can be used (e.g., administered) in combination with an anti-CD38 antibody (e.g., daratumumab).
  • an anti-CD38 antibody e.g., daratumumab
  • decreasing or eliminating expression of CD38 by any of the engineered cells provided herein may reduce or avoid targeting of the engineered cells by the anti-CD38 antibody.
  • editing (e.g., knocking out) CD38 in engineered immune cells may not only increase the amount of the engineered cells that persist in vivo, but loss of the anti-CD38 antibody to the “sink” that is CD38 -expressing engineered cells may be mitigated.
  • editing e.g., knocking out
  • CD38-expressing target cells e.g., cancer cells
  • An additional advantage of editing (e.g., knocking out) CD38 in engineered NK cells is the preservation of NK cell-mediated antibody -dependent cellular cytotoxicity (ADCC).
  • ADCC is a mechanism of cell-mediated immune defense against opsonized target cells wherein the Fc-fragment of an antibody binds to CD16a (FcyRIIIa) on immune effectors such as NK cells, triggering their activation and targeted killing.
  • CD38 By genetically editing (e.g., knocking out) CD38 in the engineered NK cells, they are more likely to persist in vivo rather than being targeted by an anti-CD38 antibody, and therefore will be present in higher numbers to mediate ADCC, thereby potentiating the effect of the anti-CD38 antibody.
  • gene editing CD38 increases the cytotoxicity, persistence, immune avoidance or otherwise enhances the efficacy of engineered NK, T, or other cell as disclosed herein.
  • any other target protein, or any combination of target proteins can be decreased or eliminated, such as by disrupting the gene(s) encoding the target protein/ s).
  • the expression of a target gene is reduced and/or eliminated in any cells provided herein.
  • Target genes can include any gene such as CD38, CISH, Cbl-b, MED12, and TGFbR2.
  • CD38 encoded by CD38
  • CD38 is disrupted and/or knocked out using one or more of the gene editing methods disclosed herein.
  • CD38 is disrupted and/or knocked out using a Crispr-Cas mediated approach (e.g., using a Cas9 nuclease) as disclosed elsewhere herein, with the Cas nuclease guided by a CD38-specific guide RNA (gRNA).
  • gRNA CD38-specific guide RNA
  • Methods for Crispr-Cas-based disruption of CD38, as well as examples of CD38 -targeting gRNA sequences are known in the art and include any of those as described in Clara et al., J Immunother Cancer (2022) 10(2):e003804 and Kararoudi et al., Blood (2020) 136(21):2416-27.
  • transcription activatorlike effector nucleases TALENs
  • ZFNs zinc finger nucleases
  • gene editing reduces transcription of CD38 by about 30%, about 40%, about 50%, about 60%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 97%, about 98%, about 99%, or more (including any amount between those listed).
  • gene editing reduces transcription of CD38 by at least about 30%.
  • gene editing reduces transcription of CD38 by at least about 40%.
  • gene editing reduces transcription of CD38 by at least about 50%.
  • gene editing reduces transcription of CD38 by at least about 60%.
  • gene editing reduces transcription of CD38 by at least about 70%.
  • gene editing reduces transcription of CD38 by at least about 80%.
  • gene editing reduces transcription of CD38 by at least about 90%.
  • gene editing reduces expression of CD38 by about 30%, about 40%, about 50%, about 60%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 97%, about 98%, about 99%, or more (including any amount between those listed).
  • gene editing reduces expression of CD38 by at least about 30%.
  • gene editing reduces expression of CD38 by at least about 40%.
  • gene editing reduces expression of CD38 by at least about 50%.
  • gene editing reduces expression of CD38 by at least about 60%.
  • gene editing reduces expression of CD38 by at least about 70%.
  • gene editing reduces expression of CD38 by at least about 80%.
  • gene editing reduces expression of CD38 by at least about 90%.
  • any of immune cells as provided herein are engineered to express a membrane-bound interleukin 15 (mbIL15).
  • mbIL15 expression on the immune cell e.g., NK cell
  • the IL15 is expressed from a separate cassette on the construct comprising any one of the CARs disclosed herein.
  • the IL15 is expressed from the same cassette as any one of the CARs disclosed herein.
  • the chimeric receptor and IL15 are separated by a nucleic acid sequence encoding a cleavage site, for example, a proteolytic cleavage site or a T2A, P2A, E2A, or F2A self-cleaving peptide cleavage site.
  • the chimeric receptor and IL15 are separated by a T2A sequence (e.g., SEQ ID NO:20, encoded by SEQ ID NO: 19).
  • the IL15 is a membrane -bound IL15 (mbIL15).
  • the mbIL15 comprises a native IL15 sequence, such as a human native IL15 sequence (e.g., SEQ ID NO:22, encoded by SEQ ID NO:21), and at least one transmembrane domain (e.g., CD8a).
  • IL15 is encoded by the nucleic acid sequence of SEQ ID NO: 21.
  • IL15 can be truncated or modified, such that it is encoded by a nucleic acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the nucleic acid sequence of SEQ ID NO: 21.
  • the IL15 comprises the amino acid sequence of SEQ ID NO: 22. In several embodiments, the IL15 is truncated or modified, such that it has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 22.
  • any of the CARs as described herein are encoded by the same nucleic acid sequence as a mbIL15.
  • a nucleic acid sequence encoding the CAR and a nucleic acid sequence encoding the mbIL15 are separated by a T2A-encoding sequence (e.g., SEQ ID NO: 19).
  • any of the engineered cells as described herein express a CD38-targeting recombinant receptor (e.g., CAR) and a mbIL15.
  • the mbIL15 is membrane -bound by virtue of the fusion of IL15 to a transmembrane domain.
  • mbIL15 comprises a transmembrane domain.
  • the transmembrane domain comprises a CD8a transmembrane domain.
  • the transmembrane domain comprises a hinge and/or a transmembrane region.
  • the transmembrane domain comprises a hinge and a transmembrane region.
  • the hinge is a CD8a hinge sequence (e.g., SEQ ID NO:6).
  • the transmembrane region is a CD8a transmembrane region (e.g., SEQ ID NO:8).
  • the mbIL15 comprises a native IL15 sequence, such as a human native IL15 sequence, and at least one transmembrane domain (e.g., CD8a transmembrane domain).
  • the CD8a transmembrane domain comprises the sequence of SEQ ID NO: 10.
  • the mbIL15 is truncated or modified such that it comprises an amino acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequency identity to the amino acid sequence of SEQ ID NO:23.
  • the mbIL15 comprises the amino acid sequence of SEQ ID NO:23.
  • Membrane -bound IL15 sequences are described in PCT publications WO 2018/183385 and WO 2020/056045, each of which is hereby expressly incorporated by reference in its entirety.
  • iv. Cell Types [00197] Some embodiments of the methods and compositions provided herein relate to a cell such as an immune cell.
  • an immune cell is engineered to express a chimeric receptor that binds to an antigen (e.g., an antigen expressed by a cancer cell).
  • Targeted therapy is a cancer treatment that employs certain drugs that target specific genes or proteins found in cancer cells or cells supporting cancer growth, (like blood vessel cells) to reduce or arrest cancer cell growth.
  • genetic engineering has enabled approaches to be developed that harness certain aspects of the immune system to fight cancers.
  • a patient’s own immune cells are modified to specifically eradicate that patient’s type of cancer.
  • Various types of immune cells can be used, such as T cells, Natural Killer (NK cells), or combinations thereof, as described in more detail below.
  • CAR chimeric antigen receptors
  • some embodiments include a polynucleotide, polypeptide, or vector that encodes, for example a chimeric antigen receptor directed against CD38, to facilitate targeting of an immune cell to a cancer and exerting cytotoxic effects on the cancer cell.
  • Methods of treating cancer and other uses of such cells for cancer immunotherapy are also provided for herein.
  • engineered immune cells e.g., T cells or NK cells expressing such chimeric receptors.
  • cells of the immune system are engineered to have enhanced cytotoxic effects against target cells, such as tumor cells.
  • a cell of the immune system may be engineered to include a CD38-directed CAR as described herein.
  • white blood cells or leukocytes are used, since their native function is to defend the body against growth of abnormal cells and infectious disease.
  • white bloods cells include granulocytes and agranulocytes (presence or absence of granules in the cytoplasm, respectively).
  • Granulocytes include basophils, eosinophils, neutrophils, and mast cells.
  • Agranulocytes include lymphocytes and monocytes.
  • Cells such as those that follow or are otherwise described herein may be engineered to include a CD38-directed CAR, or a nucleic acid encoding the CAR.
  • the immune cells are engineered to co-express a membrane-bound interleukin 15 (mbIL15) co-stimulatory domain.
  • the immune cells engineered to express a CAR are engineered to bicistronically express a mbIL15 domain.
  • the immune cells comprise monocytes.
  • Monocytes are a subtype of leukocyte. Monocytes can differentiate into macrophages and myeloid lineage dendritic cells. Monocytes are associated with the adaptive immune system and serve the main functions of phagocytosis, antigen presentation, and cytokine production. Phagocytosis is the process of uptake of cellular material, or entire cells, followed by digestion and destruction of the engulfed cellular material.
  • a monocyte is positive for cell surface expression of a marker selected from among the group consisting of CCR2, CCR5, CDllc, CD14, CD16, CD62L, CD68+, CX3CR1, HLA-DR, or any combination thereof.
  • a monocyte is positive for cell surface expression of CD14.
  • a monocyte is positive for cell surface expression of CCR2.
  • a monocyte is positive for cell surface expression of CCR5.
  • a monocyte is positive for cell surface expression of CD62L.
  • monocytes are used in connection with one or more additional engineered cells as disclosed herein. Some embodiments of the methods and compositions described herein relate to a monocyte that expresses a CAR that binds to CD38, or a nucleic acid encoding the CAR.
  • the monocytes are engineered to express a membranebound interleukin 15 (mbIL15) domain.
  • the monocytes engineered to express a CAR are engineered to also express (e.g., bicistronically express) a membrane -bound interleukin 15 (mbIL15) domain.
  • the monocytes are engineered to bicistronically express the CAR and mbIL15.
  • the monocytes are allogeneic cells. In some embodiments, the monocytes are obtained from a donor who does not have cancer.
  • Lymphocytes the other primary sub-type of leukocyte include T cells (cell-mediated, cytotoxic adaptive immunity), natural killer cells (cell-mediated, cytotoxic innate immunity), and B cells (humoral, antibody-driven adaptive immunity). While B cells are engineered according to several embodiments, disclosed herein, several embodiments also relate to engineered T cells or engineered NK cells (mixtures of T cells and NK cells are used in some embodiments, either from the same donor, or different donors).
  • the immune cells comprise T cells.
  • the immune cells comprise NK cells.
  • the immune cells comprise T cells and NK cells.
  • the immune cells comprise B cells.
  • lymphocytes are used in connection with one or more additional engineered cells as disclosed herein. Some embodiments of the methods and compositions described herein relate to a lymphocyte that express a CAR that binds to CD38, or a nucleic acid encoding the CAR.
  • the lymphocytes are engineered to express a membrane -bound interleukin 15 (mbIL15) domain.
  • the lymphocytes engineered to express a CAR are engineered to also express (e.g., bicistronically express) a membrane-bound interleukin 15 (mbIL15) domain.
  • lymphocytes are engineered to bicistronically express the CAR and mbIL15.
  • the lymphocytes are allogeneic cells. In some embodiments, the lymphocytes are obtained from a donor who does not have cancer. c. T Cells
  • the immune cells comprise T cells.
  • T cells are distinguishable from other lymphocytes sub-types (e.g., B cells or NK cells) based on the presence of a T-cell receptor on the cell surface.
  • T cells can be divided into various different subtypes, including effector T cells, helper T cells, cytotoxic T cells, memory T cells, regulatory T cells, natural killer T cell, mucosal associated invariant T cells and gamma delta T cells.
  • a specific subtype of T cell is engineered.
  • a T cell is positive for cell surface expression of a marker selected from among the group consisting of CD3, CD4, and/or CD8.
  • a T cell is positive for cell surface expression of CD3.
  • a T cell is positive or cell surface expression of CD4.
  • a T cell is positive or cell surface expression of CD8.
  • CD3+ T cells are engineered.
  • CD4+ T cells are engineered.
  • CD8+ T cells are engineered.
  • regulatory T cells are engineered.
  • gamma delta T cells are engineered.
  • a mixed pool of T cell subtypes is engineered.
  • CD4+ and CD8+ T cells are engineered.
  • specific techniques such as use of cytokine stimulation are used to enhance expansion/collection of T cells with a specific marker profile.
  • activation of certain human T cells e.g. CD4+ T cells, CD8+ T cells is achieved through use of CD3 and/or CD28 as stimulatory molecules.
  • a method of treating or preventing cancer or an infectious disease comprising administering a therapeutically effective amount of T cells expressing the cytotoxic receptor complex and/or a homing moiety as described herein.
  • a method of treating or preventing cancer or an infectious disease comprising administering T cells expressing a cytotoxic receptor complex as described herein.
  • the engineered T cells are autologous cells, while in some embodiments, the T cells are allogeneic cells. In some embodiments, the T cells are allogeneic cells. In some embodiments, the T cells are obtained from a donor who does not have cancer .
  • T cells engineered to express a CAR that binds to CD38 are engineered to express a membrane-bound interleukin 15 (mbIL15) domain.
  • the T cells engineered to express a CAR are engineered to also express (e.g., bicistronically express) a membrane -bound interleukin 15 (mbIL15) domain.
  • the T cells are engineered to bicistronically express the CAR and mbIL15.
  • the immune cells comprise T cells and NK cells (either from the same donor or from different donors d. NK Cells
  • the immune cells comprise natural killer (NK) cells.
  • NK natural killer
  • a method of treating or preventing a disease or condition comprising administering natural killer (NK) cells expressing a CD38-targeting CAR as described herein.
  • the engineered NK cells are autologous cells, while in some embodiments, the NK cells are allogeneic cells.
  • the NK cells are autologous cells. In some embodiments, the NK cells are allogeneic cells.
  • NK cells are preferred because the natural cytotoxic potential of NK cells is relatively high.
  • a NK cell is positive for cell surface expression of a marker selected from among the group consisting of CCR7, CD16, CD56, CD57, CD11, CX3CR 1 , a Killer Ig-like receptor (KIR), NKp30, NKp44, NKp46, or any combination thereof.
  • a NK cell is positive for cell surface expression of CD16.
  • a NK cell is positive for cell surface expression of CD56.
  • a NK cell is positive for cell surface expression of a Killer Ig-like receptor.
  • NK cells engineered to express a CAR that binds to CD38 are engineered to a membrane-bound interleukin 15 (mbIL15) domain.
  • the NK cells engineered to express a CAR are engineered to also express (e.g., bicistronically express) a membrane-bound interleukin 15 (mbIL15) domain.
  • the NK cells are engineered to bicistronically express the CAR and mbIL15.
  • the NK cells are derived from cell line NK-92.
  • NK-92 cells are derived from NK cells, but lack major inhibitory receptors displayed by normal NK cells, while retaining the majority of activating receptors.
  • the NK cells are used in combination with T cells.
  • the immune cells comprise T cells and NK cells (either from the same donor or from different donors).
  • primary NK cells are used in combination with primary T cells as disclosed herein. e. Hematopoietic Stem Cells
  • the immune cells comprise hematopoietic stem cells (HSCs).
  • HSCs are used in the methods disclosed herein.
  • the cells are engineered to express a CAR that binds to CD38.
  • HSCs are used, in several embodiments, to leverage their ability to engraft for long-term blood cell production, which could result in a sustained source of targeted anti-cancer effector cells, for example to combat cancer remissions. In several embodiments, this ongoing production helps to offset anergy or exhaustion of other cell types, for example due to the tumor microenvironment.
  • a HSC is positive for cell surface expression of a marker selected from among the group consisting of CD34, CD59, and CD90. In some embodiments, a HSC is positive for cell surface expression of CD34. In some embodiments, a HSC is positive for cell surface expression of CD59. In some embodiments, a HSC is positive for cell surface expression of CD90.
  • allogeneic HSCs are used, while in some embodiments, autologous HSCs are used.
  • HSCs are used in combination with one or more additional engineered cell type disclosed herein.
  • Some embodiments of the methods and compositions described herein relate to a stem cell, such as a HSC engineered to express a CAR that binds to CD38, or a nucleic acid encoding the CAR.
  • the HSCs are engineered to express a membranebound interleukin 15 (mbIL15) domain.
  • the HSCs engineered to express a CAR are engineered to also express (e.g., bicistronically express) a membrane-bound interleukin 15 (mbIL15) domain.
  • HSCs are engineered to bicistronically express the CAR and mbIL15.
  • the HSCs are allogeneic cells. In some embodiments, the HSCs are obtained from a donor who does not have cancer. f. Induced Pluripotent Stem Cells
  • immune cells are derived (differentiated) from pluripotent stem cells (PSCs).
  • PSCs pluripotent stem cells
  • immune cells e.g., NK and/or T cells
  • iPSCs induced pluripotent stem cells
  • NK cells are derived from iPSCs.
  • iPSCs induced pluripotent stem cells
  • iPSCs are used, in several embodiments, to leverage their ability to differentiate and derive into non-pluripotent cells, including, but not limited to, CD34 cells, hemogenic endothelium cells, HSCs (hematopoietic stem and progenitor cells), hematopoietic multipotcnt progenitor cells, T cell progenitors, NK cell progenitors, T cells, NKT cells, NK cells, and B cells comprising one or several genetic modifications at selected sites through differentiating iPSCs or less differentiated cells comprising the same genetic modifications at the same selected sites.
  • the iPSCs are used to generate iPSC-derived NK or T cells.
  • the iPSCs are used to generate iPSC-derived NK cells.
  • the iPSCs are used to generate iPSC-derived T cells.
  • iPSCs engineered to express a CAR are engineered to also express (e.g., bicistronically express) a membrane- bound interleukin 15 (mbIL15).
  • mbIL15 membrane- bound interleukin 15
  • the engineered iPSCs are differentiated into NK, T, or other immune cells, such as for use in a composition or method provided herein.
  • the engineered iPSCs are differentiated into NK cells.
  • the engineered iPSCs are differentiated into T cells.
  • the engineered iPSCs are differentiated into NK and T cells.
  • preparation of the engineered cells includes one or more culture and/or preparation steps.
  • the cells for introduction of the recombinant receptor e.g., CAR
  • the sample is an apheresis (e.g.., leukapheresis) sample.
  • the subject from which the cells are isolated is one not having the disease or condition in need of a cell therapy or not to which a cell therapy will be administered.
  • the cells are isolated from a subject that is different than the subject in need of a cell therapy or to which a cell therapy will be administered.
  • the cells are allogeneic to the subject to whom they are administered.
  • the subject from which the cells are isolated is one having the disease or condition or in need of a cell therapy or to which a cell therapy will be administered.
  • the cells are isolated from the subject to which a cell therapy will be administered.
  • the cells are autologous to the subject to whom they are administered.
  • the samples include tissue, fluid, and other samples taken directly from the subject, as well as samples resulting from one or more processing steps, such as separation, centrifugation, genetic engineering (e.g., transduction with viral vector), washing, and/or incubation.
  • the biological sample can be a sample obtained directly from a biological source or a sample that is processed.
  • Biological samples include, but are not limited to, body fluids, such as blood, plasma, serum, cerebrospinal fluid, synovial fluid, urine and sweat, tissue and organ samples, including processed samples derived therefrom.
  • the sample from which the cells are derived or isolated is blood or a blood-derived sample, or is or is derived from an apheresis (e.g., a leukapheresis) product.
  • samples include whole blood, peripheral blood mononuclear cells (PBMCs), leukocytes, bone marrow, thymus, tissue biopsy, tumor, leukemia, lymphoma, lymph node, gut associated lymphoid tissue, mucosa associated lymphoid tissue, spleen, other lymphoid tissues, liver, lung, stomach, intestine, colon, kidney, pancreas, breast, bone, prostate, cervix, testes, ovaries, tonsil, or other organ, and/or cells derived therefrom.
  • Samples include, in the context of cell therapy, e.g., adoptive cell therapy, samples from autologous and allogeneic sources.
  • the cells in some embodiments are primary cells, e.g., primary human cells.
  • the cells are immune cells, e.g. primary NK cells or primary T cells.
  • isolation of the cells includes one or more preparation and/or non affinity-based cell separation steps.
  • cells are washed, centrifuged, and/or incubated in the presence of one or more reagents, for example, to remove unwanted components, enrich for desired components, lyse or remove cells sensitive to particular reagents.
  • cells are separated based on one or more property, such as density, adherent properties, size, sensitivity and/or resistance to particular components.
  • apheresis e.g., leukapheresis
  • the cells are isolated from an apheresis (e.g., leukapheresis) sample.
  • the samples contain lymphocytes, including NK cells, T cells, monocytes, granulocytes, B cells, other nucleated white blood cells, red blood cells, and/or platelets, and in some aspects contain cells other than red blood cells and platelets.
  • Non-limiting samples include whole blood, peripheral blood mononuclear cells (PBMCs), leukocytes, bone marrow, thymus, tissue biopsy, tumor, leukemia, lymphoma, lymph node, gut associated lymphoid tissue, mucosa associated lymphoid tissue, spleen, other lymphoid tissues, liver, lung, stomach, intestine, colon, kidney, pancreas, breast, bone, prostate, cervix, testes, ovaries, tonsil, or other organ, and/or cells derived therefrom.
  • the cells are derived from PBMCs.
  • Samples include, in the context of cell therapy, e.g., adoptive cell therapy, samples from autologous and allogeneic sources.
  • the isolation methods include the separation of different cell types based on the expression or presence in the cell of one or more specific molecules, such as surface markers, e.g., surface proteins, intracellular markers, or nucleic acid. In some embodiments, any known method for separation based on such markers may be used. In some embodiments, the separation is affinity- or immunoaffinity-based separation.
  • the isolation in some aspects includes separation of cells and cell populations based on the cells’ expression or expression level of one or more markers, typically cell surface markers, for example, by incubation with an antibody or binding partner that specifically binds to such markers, followed generally by washing steps and separation of cells having bound the antibody or binding partner, from those cells having not bound to the antibody or binding partner.
  • Such separation steps can be based on positive selection, in which the cells having bound the reagents are retained for further use, and/or negative selection, in which the cells having not bound to the antibody or binding partner are retained. In some examples, both fractions are retained for further use. In some aspects, negative selection can be particularly useful where no antibody is available that specifically identifies a cell type in a heterogeneous population, such that separation is best carried out based on markers expressed by cells other than the desired population.
  • the separation need not result in 100% enrichment or removal of a particular cell population or cells expressing a particular marker.
  • positive selection of or enrichment for cells of a particular type refers to increasing the number or percentage of such cells, but need not result in a complete absence of cells not expressing the marker.
  • negative selection, removal, or depletion of cells of a particular type refers to decreasing the number or percentage of such cells, but need not result in a complete removal of all such cells.
  • multiple rounds of separation steps are carried out, where the positively or negatively selected fraction from one step is subjected to another separation step, such as a subsequent positive or negative selection.
  • a single separation step can deplete cells expressing multiple markers simultaneously, such as by incubating cells with a plurality of antibodies or binding partners, each specific for a marker targeted for negative selection.
  • multiple cell types can simultaneously be positively selected by incubating cells with a plurality of antibodies or binding partners expressed on the various cell types.
  • NK cells or specific subpopulations thereof such as cells positive or expressing high levels of one or more surface markers, e.g., CD56+, CCR7+, CD16+, CD57+, CD11+, CX3CR1+, a Killer Ig-like receptor (KIR) +, NKp30+, NKp44+, or NKp46+ NK cells, are isolated by positive or negative selection techniques.
  • NK cells are isolated by positive selection for CD56.
  • CD56+ NK cells can be positively selected using anti-CD56 conjugated magnetic beads.
  • T cells are separated from a PBMC sample by negative selection of markers expressed on non-T cells, such as B cells, monocytes, or other white blood cells, such as CD14.
  • a CD4+ or CD8+ selection step is used to separate CD4+ helper and CD8+ cytotoxic T cells.
  • Such CD4+ and CD8+ populations can be further sorted into sub-populations by positive or negative selection for markers expressed or expressed to a relatively higher degree on one or more naive, memory, and/or effector T cell subpopulations.
  • the cells are expanded in culture prior to, during, and/or following genetic engineering. In some embodiments, the cells are expanded in culture prior to genetic engineering. In some embodiments, the cells are expanded in culture following genetic engineering. In some embodiments, the cells are expanded in culture prior to and following genetic engineering. Methods for expanding cells are known in the art and include any of those described in US Patent Nos. 7,435,596 and 8,026,097; and Patent Application Nos. PCT/SG2018/050138; PCT/US2020/044033; PCT/US2021/071330; and PCT/US2022/074164.
  • expanding the cells in culture comprises co-culturing the cells with feeder cells.
  • the cells are expanded in culture prior to genetic engineering by co-culturing the cells with feeder cells.
  • the feeder cells express IL15 (e.g., membrane -bound IL15) and 4-1BBL.
  • the feeder cells express membrane-bound interleukin 15 (mbIL15) and 4-1BBL.
  • the feeder cells do not express MHCI molecules.
  • the feeder cells do not express MHCII molecules.
  • the feeder cells are immune cells.
  • the feeder cells are K562 cells.
  • Engineered feeder cells are disclosed in, for example, International Patent Application PCT/SG2018/050138.
  • the cells are allowed to further expand in culture.
  • expanding the cells in culture comprising culturing the cells in the presence of IL2, IL12, and/or IL18.
  • the cells are cultured in the presence of IL2.
  • the cells are cultured in the presence of IL12 and IL18.
  • the cells are cultured in the presence of IL2, IL12, and IL18.
  • the preparation methods include steps for freezing, e.g., cryopreserving, the cells, either before or after isolation, engineering, and/or expansion.
  • the cells are cryopreserved after engineering.
  • the cells are cryopreserved after the further expansion.
  • the cells are suspended in a freezing solution. Any of a variety of known freezing solutions and parameters in some aspects may be used.
  • compositions including the CD38-binding molecules, immunoconjugates, recombinant receptors, and engineered cells including pharmaceutical compositions and formulations.
  • compositions comprising a CD38-binding molecule (e.g., antibody), an immunoconjugate, a recombinant receptor (e.g., chimeric antigen receptor), engineered cells expressing said molecules (e.g., antibody or recombinant receptor), a plurality of engineered cells expressing said molecules (e.g., recombinant receptor) and/or additional agents for combination treatment or therapy.
  • a CD38-binding molecule e.g., antibody
  • an immunoconjugate e.g., a recombinant receptor (e.g., chimeric antigen receptor)
  • engineered cells expressing said molecules e.g., antibody or recombinant receptor
  • a plurality of engineered cells expressing said molecules e.g., recombinant receptor
  • additional agents for combination treatment or therapy e.g., additional agents for combination treatment or therapy.
  • the pharmaceutical compositions and formulations generally include one or more optional pharmaceutically acceptable carrier or excipient.
  • pharmaceutical formulation refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
  • a “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject.
  • a pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
  • the choice of carrier is determined in part by the particular cell, binding molecule, and/or antibody, and/or by the method of administration. Accordingly, there are a variety of suitable formulations.
  • the pharmaceutical composition can contain preservatives. Suitable preservatives may include, for example, methylparaben, propylparaben, sodium benzoate, and benzalkonium chloride. In some aspects, a mixture of two or more preservatives is used. The preservative or mixtures thereof are typically present in an amount of about 0.0001% to about 2% by weight of the total composition.
  • Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arg
  • a buffer is included in the composition.
  • Suitable buffering agents include, for example, citric acid, sodium citrate, phosphoric acid, potassium phosphate, and various other acids and salts.
  • a mixture of two or more buffers is used.
  • the buffering agent or mixtures thereof are typically present in an amount of from about 0.001% to about 4% by weight of the total composition. Methods for preparing administrable pharmaceutical compositions are known.
  • Formulations of the antibodies described herein can include lyophilized formulations and aqueous solutions.
  • the formulation or composition may also contain more than one active ingredient useful for the particular indication, disease, or condition being treated with the binding molecules or cells, preferably those with activities complementary to the binding molecule or cell, where the respective activities do not adversely affect one another.
  • active ingredients are suitably present in combination in amounts that are effective for the purpose intended.
  • the pharmaceutical composition further includes other pharmaceutically active agents or drugs, such as chemotherapeutic agents, e.g., asparaginase, busulfan, carboplatin, cisplatin, daunorubicin, doxorubicin, fluorouracil, gemcitabine, hydroxyurea, methotrexate, paclitaxel, rituximab, vinblastine, vincristine, etc.
  • chemotherapeutic agents e.g., asparaginase, busulfan, carboplatin, cisplatin, daunorubicin, doxorubicin, fluorouracil, gemcitabine, hydroxyurea, methotrexate, paclitaxel, rituximab, vinblastine, vincristine, etc.
  • the cells or antibodies are administered in the form of a salt, e.g., a pharmaceutically acceptable salt.
  • Suitable pharmaceutically acceptable acid addition salts include those derived from mineral acids, such as hydrochloric, hydrobromic, phosphoric, metaphosphoric, nitric, and sulphuric acids, and organic acids, such as tartaric, acetic, citric, malic, lactic, fumaric, benzoic, glycolic, gluconic, succinic, and arylsulphonic acids, for example, p-toluenesulphonic acid.
  • mineral acids such as hydrochloric, hydrobromic, phosphoric, metaphosphoric, nitric, and sulphuric acids
  • organic acids such as tartaric, acetic, citric, malic, lactic, fumaric, benzoic, glycolic, gluconic, succinic, and arylsulphonic acids, for example, p-toluenesulphonic acid.
  • the pharmaceutical composition in some embodiments contains the binding molecules and/or cells in amounts effective to treat or prevent the disease or condition, such as a therapeutically effective or prophylactically effective amount.
  • Therapeutic or prophylactic efficacy in some embodiments is monitored by periodic assessment of treated subjects. For repeated administrations over several days or longer, depending on the condition, the treatment is repeated until a desired suppression of disease symptoms occurs.
  • other dosage regimens may be useful and can be determined.
  • the desired dosage can be delivered by a single bolus administration of the composition, by multiple bolus administrations of the composition, or by continuous infusion administration of the composition.
  • Formulations include those for oral, intravenous, intraperitoneal, subcutaneous, pulmonary, transdermal, intramuscular, intranasal, buccal, sublingual, or suppository administration.
  • the agent or cell population is administered to the subject by intravenous, intraperitoneal, or subcutaneous injection using peripheral systemic delivery.
  • the compositions are provided as sterile liquid formulations (e.g., isotonic aqueous solutions, suspensions, emulsions, dispersions, or viscous compositions), which in some aspects may be buffered to a selected pH.
  • sterile liquid formulations e.g., isotonic aqueous solutions, suspensions, emulsions, dispersions, or viscous compositions
  • Liquid formulations are generally easier to prepare than gels, other viscous compositions, and solid compositions.
  • liquid compositions are somewhat more convenient to administer, particularly by injection.
  • the liquid composition can comprise a carrier, which can be a solvent or dispersion medium containing, for example, water, saline, phosphate buffered saline, polyols (e.g., glycerol, propylene glycol, liquid polyethylene glycol), and suitable mixtures thereof.
  • a carrier which can be a solvent or dispersion medium containing, for example, water, saline, phosphate buffered saline, polyols (e.g., glycerol, propylene glycol, liquid polyethylene glycol), and suitable mixtures thereof.
  • Sterile injectable solutions can be prepared by incorporating the agent or cell into a solvent, such as an admixture with a suitable carrier, diluent, or excipient (e.g., sterile water, saline, glucose, dextrose, and the like).
  • a suitable carrier e.g., sterile water, saline, glucose, dextrose, and the like.
  • Formulations for in vivo administration are typically sterile. Sterility can be readily achieved, for example, by filtration through sterile filtration membranes.
  • the dose of engineered cells administered is in a cryopreserved composition. In some aspects, the composition is administered after thawing the cryopreserved composition.
  • compositions for combination therapy are also provided.
  • Any of the additional agents for combination therapy described herein, such as agents described in Section III.B, can be prepared and administered as one or more pharmaceutical compositions, with the CD38-binding molecule (e.g., antibody), immunoconjugate, recombinant receptor (e.g., chimeric antigen receptor) and/or engineered cells expressing said molecules (e.g., recombinant receptor) described herein.
  • the combination therapy can be administered in one or more pharmaceutical compositions, e.g., where the binding molecules, recombinant receptors and/or cells are in the same pharmaceutical composition as the additional agent, or in separate pharmaceutical compositions.
  • the additional agent is an additional engineered cell, e.g., cell engineered to express a different recombinant receptor that targets a different antigen (e.g., BCMA) or a different epitope on CD38, and is administered in the same composition or in a separate composition.
  • each of the pharmaceutical composition is formulated in a suitable formulation according to the particular binding molecule, recombinant receptor, cell, e.g., engineered cell, and/or additional agent, and the particular dosage regimen and/or method of delivery.
  • CD38-binding molecules such as in the treatment of diseases, conditions, and disorders in which CD38 is expressed, and/or in detection, diagnostic, and prognostic methods.
  • methods of treatment, and uses are those that involve administering to a subject engineered cells, such as a plurality of engineered cells, expressing the provided anti-CD38 recombinant receptors (e.g. CARs).
  • methods of combination therapy and/or treatment are also provided.
  • CD38-binding molecules including the anti-CD38 recombinant receptors (e.g., CARs), engineered cells expressing the recombinant receptors (e.g., CARs), plurality of engineered cells expressing the receptors, and/or compositions comprising the same.
  • the anti-CD38 recombinant receptors e.g., CARs
  • engineered cells expressing the recombinant receptors e.g., CARs
  • plurality of engineered cells expressing the receptors e.g., plurality of engineered cells expressing the receptors
  • Such methods and uses include therapeutic methods and uses, for example, involving administration of the molecules (e.g., recombinant receptors), cells (e.g., engineered cells), or compositions containing the same, to a subject having a disease, condition, or disorder associated with CD38 such as a disease, condition, or disorder associated with CD38 expression, and/or in which cells or tissues express, e.g., specifically express, CD38.
  • the binding molecule, cell, and/or composition is/are administered in an effective amount to effect treatment of the disease or disorder.
  • the methods are carried out by administering the binding molecules or cells, or compositions comprising the same, to the subject having, having had, or suspected of having the disease or condition. In some embodiments, the methods thereby treat the disease or condition or disorder in the subject. Also provided herein are of use of any of the compositions, such as pharmaceutical compositions provided herein, for the treatment of a disease or disorder associated with CD38, such as use in a treatment regimen.
  • treatment refers to complete or partial amelioration or reduction of a disease or condition or disorder, or a symptom, adverse effect or outcome, or phenotype associated therewith. Desirable effects of treatment include, but are not limited to, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis. The terms do not imply complete curing of a disease or complete elimination of any symptom or effect(s) on all symptoms or outcomes.
  • “delaying development of a disease” means to defer, hinder, slow, retard, stabilize, suppress and/or postpone development of the disease (such as cancer). This delay can be of varying lengths of time, depending on the history of the disease and/or subject being treated.
  • the provided molecules and compositions are used to delay development of a disease or to slow the progression of a disease. A sufficient or significant delay can, in effect, encompass prevention, in that the subject does not develop the disease. For example, a late-stage cancer, such as development of metastasis, may be delayed.
  • Preventing includes providing prophylaxis with respect to the occurrence or recurrence of a disease in a subject that may be predisposed to the disease but has not yet been diagnosed with the disease.
  • a function or activity is to reduce the function or activity when compared to otherwise same conditions except for a condition or parameter of interest, or alternatively, as compared to another condition.
  • an antibody or composition or cell which suppresses tumor growth reduces the rate of growth of the tumor compared to the rate of growth of the tumor in the absence of the antibody or composition or cell.
  • an “effective amount” of an agent e.g., a pharmaceutical formulation, binding molecule, antibody, cells, or composition, in the context of administration, refers to an amount effective, at dosages/amounts and for periods of time necessary, to achieve a desired result, such as a therapeutic or prophylactic result.
  • a “therapeutically effective amount” of an agent refers to an amount effective, at dosages and for periods of time necessary, to achieve a desired therapeutic result, such as for treatment of a disease, condition, or disorder, and/or pharmacokinetic or pharmacodynamic effect of the treatment.
  • the therapeutically effective amount may vary according to factors such as the disease state, age, sex, and weight of the subject, and the populations of cells administered.
  • the provided methods involve administering the molecules, antibodies, cells, and/or compositions at effective amounts, e.g., therapeutically effective amounts.
  • a “prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, but not necessarily, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
  • a “subject” or an “individual” is a mammal.
  • a “mammal” includes humans, non-human primates, domestic and farm animals, and zoo, sports, or pet animals, such as dogs, horses, rabbits, cattle, pigs, hamsters, gerbils, mice, ferrets, rats, cats, monkeys, etc.
  • the subject is human.
  • CD38-associated disease or disorder any disease or disorder associated with CD38 or any disease or disorder in which CD38 is specifically expressed and/or in which CD38 has been targeted for treatment (also referred to herein interchangeably as a “CD38-associated disease or disorder”).
  • Cancers associated with CD38 expression include solid tumors, hematologic malignancies such as multiple myeloma, Waldenstrom macroglobulinemia, as well as both Hodgkin's and non-Hodgkin's lymphoma. See Konen et al., Cells (2020) 9(1 ):52; proteinatlas.org/ENSG00000004468-CD38/pathology).
  • CD38 Since CD38 is highly and uniformly expressed on plasma cells, it is a potential target for cancer therapy. CD38 is also expressed by long-lived plasma cells, which produce autoantibodies associated with autoimmune diseases. Anti-CD38 antibodies have been previously described. See van de Donk, Blood (2016) 131(1): 13- 29.
  • the disease or disorder associated with CD38 is a B cell-related disorder.
  • the disease or disorder associated with CD38 is one or more diseases or conditions from among glioblastoma, lymphomatoid granulomatosis, posttransplant lymphoproliferative disorder, an immunoregulatory disorder, heavy-chain disease, primary or immunocyte-associated amyloidosis, or monoclonal gammopathy of undetermined significance.
  • the disease or disorder associated with CD38 is an autoimmune disease or disorder.
  • autoimmune diseases or disorder include, but are not limited to, systemic lupus erythematosus (SLE), lupus nephritis, CNS lupus, inflammatory bowel disease (IBD, e.g, Crohn’s disease or ulcerative colitis), rheumatoid arthritis (RA; e.g., juvenile rheumatoid arthritis), ANCA associated vasculitis, idiopathic thrombocytopenia purpura (ITP), thrombotic thrombocytopenia purpura (TTP), autoimmune thrombocytopenia, Chagas' disease, Grave's disease, Wegener's granulomatosis, polyarteritis nodosa, Sjogren's syndrome, pemphigus vulgaris, scleroderma, multiple sclerosis(MS), psoriasis, Ig
  • a subject may exhibit symptoms of more than one autoimmune disease.
  • the autoimmune disease is systemic lupus erythematosus (SLE).
  • the autoimmune disease is SLE with renal involvement (e.g., lupus nephritis (LN)).
  • the autoimmune disease is lupus nephritis (LN).
  • the autoimmune disease is CNS lupus.
  • the autoimmune disease is IBD.
  • the autoimmune disease is Crohn’s disease.
  • the autoimmune disease is ulcerative colitis.
  • the autoimmune disease is vasculitis.
  • the autoimmune disease is ANCA vasculitis (AAV). In some embodiments, the autoimmune disease is autoimmune encephalitis (AE). In some embodiments, the autoimmune disease is ITP. In some embodiments, the autoimmune disease is TTP. In some embodiments, the autoimmune disease is autoimmune thrombocytopenia. In some embodiments, the autoimmune disease is Chagas' disease. In some embodiments, the autoimmune disease is Graves’ disease. In some embodiments, the autoimmune disease is Wegener's granulomatosis. In some embodiments, the autoimmune disease is polyarteritis nodosa. In some embodiments, the autoimmune disease is Sjogren's syndrome.
  • the autoimmune disease is pemphigus vulgaris. In some embodiments, the autoimmune disease is psoriasis. In some embodiments, the autoimmune disease is IgA nephropathy. In some embodiments, the autoimmune disease is membranous nephropathy (MN). In some embodiments, the autoimmune disease is IgM polyneuropathies. In some embodiments, the autoimmune disease is vasculitis. In some embodiments, the autoimmune disease is diabetes mellitus. In some embodiments, the autoimmune disease is Reynaud's syndrome. In some embodiments, the autoimmune disease is anti-phospholipid syndrome. In some embodiments, the autoimmune disease is Goodpasture's disease.
  • the autoimmune disease is Kawasaki disease. In some embodiments, the autoimmune disease is autoimmune hemolytic anemia. In some embodiments, the autoimmune disease is myasthenia gravis (MG). In some embodiments, the autoimmune disease is progressive glomerulonephritis. In some embodiments, the autoimmune disease is acquired immunodeficiency syndrome (AIDS). In some embodiments, the autoimmune disease is Addison’s disease. In some embodiments, the autoimmune disease is alopecia areata. In some embodiments, the autoimmune disease is celiac disease. In some embodiments, the autoimmune disease is chronic inflammatory demyelinating polyneuropathy (CIDP). In some embodiments, the autoimmune disease is Guillain-Barre syndrome.
  • CIDP chronic inflammatory demyelinating polyneuropathy
  • the autoimmune disease is Hashimoto thyroiditis. In some embodiments, the autoimmune disease is pernicious anemia. In some embodiments, the autoimmune disease is psoriasis. In some embodiments, the autoimmune disease is psoriatic arthritis. In some embodiments, the autoimmune disease is reactive arthritis. In some embodiments, the autoimmune disease is rheumatoid arthritis (RA). In some embodiments, the autoimmune disease is refractory RA. In some embodiments, RA is refractory to a TNF inhibitor. In some embodiments, the autoimmune disease is multiple sclerosis (MS). In some embodiments, MS is primary progressive MS (PPMS). In some embodiments, MS is secondary-progressive MS (SPMS). In some embodiments, MS is relapsing-remitting MS (RRMS).
  • the autoimmune disease comprises scleroderma. In some embodiments, the autoimmune disease is scleroderma. In some embodiments, the autoimmune disease comprises systemic sclerosis (also known as systemic scleroderma). In some embodiments, the autoimmune disease is systemic sclerosis (also known as systemic scleroderma). In some embodiments, the autoimmune disease comprises localized scleroderma. In some embodiments, the autoimmune disease is localized scleroderma.
  • the autoimmune disease comprises myositis (also known as IIM).
  • the autoimmune disease is myositis (also known as UM).
  • the autoimmune disease is selected from the group consisting of antisynthetase syndrome (ASSD), overlap myopathy (OM), dermatomyositis (DM), clinically amyopathic dermatomyositis, juvenile myositis (JM), necrotizing myopathy (NM; e.g., necrotizing autoimmune myopathy (or immune-mediated necrotizing myopathy), polymyositis (PM), and sporadic inclusion body myositis (sIBM).
  • ASSD antisynthetase syndrome
  • OM overlap myopathy
  • DM dermatomyositis
  • JM juvenile myositis
  • NM necrotizing myopathy
  • PM polymyositis
  • sIBM sporadic inclusion body myositis
  • the autoimmune disease is ASSD.
  • the autoimmune disease is OM. In some embodiments, the autoimmune disease is DM. In some embodiments, the autoimmune disease is JM. In some embodiments, the autoimmune disease is NM In some embodiments, the autoimmune disease is PM. In some embodiments, the autoimmune disease is sIBM.
  • the autoimmune disease comprises vasculitis. In some embodiments, the autoimmune disease is vasculitis. In some embodiments, the vasculitis is large- vessel vasculitis. In some embodiments, the vasculitis is medium-vessel vasculitis. In some embodiments, the vasculitis is small-vessel vasculitis. In some embodiments, the vasculitis is anti- neutrophilic cytoplasmic autoantibody (ANCA) vasculitis. In some embodiments the ANCA vasculitis is granulomatosis with polyangiitis (GPA). In some embodiments the ANCA vasculitis is microscopic polyangiitis (MPA). In some embodiments the ANCA vasculitis is eosinophilic granulomatosis with polyangiitis (EGPA).
  • EGPA polyangiitis
  • the autoimmune disease comprises myasthenia gravis (MG). In some embodiments, the autoimmune disease is MG. In some embodiments, MG is ocular MG. In some embodiments, MG is early-onset generalized MG. In some embodiments, MG is late-onset MG.
  • the autoimmune disease is autoimmune encephalitis (AE).
  • AE comprises an antibody to an intracellular antigen (e.g., anti-Hu or anti-GAD65).
  • AE comprises an autoantibody to an extracellular epitope of an ion channel, receptor, and/or other associated protein (e.g., anti-NMDA receptor).
  • the disease or disorder associated with CD38 is an inflammatory disease or disorder.
  • inflammatory diseases or disorders include, but are not limited to, respiratory synctial virus (RSV), chronic obstructive pulmonary disease (COPD), rheumatoid arthritis (RA), and collagen-induced arthritis (CIA). See Konen et al., Cells (2020) 9(1):52.
  • CD38 is expressed on malignant cells and cancers.
  • the cancer e.g., a CD38-expressing cancer
  • the cancer is a B cell malignancy.
  • the cancer e.g., a CD38-expressing cancer
  • Lymphomas contemplated herein include, but are not limited to, Burkitt lymphoma (e.g., endemic Burkitt's lymphoma or sporadic Burkitt's lymphoma), non-Hodgkin's lymphoma (NHL), Hodgkin's lymphoma, Waldenstrom macroglobulinemia, follicular lymphoma, small non-cleaved cell lymphoma, mucosa-associated lymphatic tissue lymphoma (MALT), marginal zone lymphoma, splenic lymphoma, nodal monocytoid B cell lymphoma, immunoblastic lymphoma, large cell lymphoma, diffuse mixed cell lymphoma, pulmonary B cell angiocentric lymphoma, small lymphocytic lymphoma, primary mediastinal B cell lymphoma, lymphoplasmacytic lymphoma (LPL), or mantle cell lymphoma (MCL).
  • Leukemias contemplated here include, but are not limited to, chronic lymphocytic leukemia (CLL), plasma cell leukemia or acute lymphocytic leukemia (ALL). Also contemplated herein are plasma cell malignancies including, but not limited to, multiple myeloma (e.g., non-secretory multiple myeloma, smoldering multiple myeloma) or plasmacytoma. In some embodiments the disease or condition is multiple myeloma (MM), such as relapsed and/or refractory multiple myeloma (R/R MM).
  • CLL chronic lymphocytic leukemia
  • ALL acute lymphocytic leukemia
  • plasma cell malignancies including, but not limited to, multiple myeloma (e.g., non-secretory multiple myeloma, smoldering multiple myeloma) or plasmacytoma.
  • the disease or condition is multiple myelom
  • disorders or conditions associated with CD38 include, but are not limited to, melanoma, neuroblastoma, renal cell carcinoma, colon cancer, colorectal cancer, breast cancer, epithelial squamous cell cancer, melanoma, myeloma (e.g., multiple myeloma), stomach cancer, brain cancer, lung cancer, pancreatic cancer, cervical cancer, ovarian cancer, liver cancer, bladder cancer, prostate cancer, testicular cancer, thyroid cancer, uterine cancer, adrenal cancer, and head and neck cancer.
  • the disease or disorder is melanoma, glioma, esophageal, cervical, or lung cancer.
  • the methods may identify a subject who has, is suspected to have, or is at risk for developing a CD38-associated disease or disorder.
  • a CD38-binding recombinant receptor e.g., CAR
  • engineered cells expressing the recombinant receptor.
  • a subject may be screened for the presence of a disease or disorder associated with elevated CD38 expression, such as a CD38-expressing cancer.
  • a subject may be screened for the presence of a disease or disorder associated with elevated CD38 expression, such as an autoimmune disease.
  • the methods include screening for or detecting the presence of a CD38-associated disease, e.g. a tumor.
  • the methods include screening for or detecting the presence of a CD38- associated disease, e.g. an autoimmune disease.
  • a sample may be obtained from a patient suspected of having a disease or disorder associated with elevated CD38 expression and assayed for the expression level of CD38.
  • a subject who tests positive for a CD38-associated disease or disorder may be selected for treatment by the present methods, and may be administered a therapeutically effective amount of a recombinant receptor (e.g., CAR) comprising a CD38-binding molecule, cells containing a recombinant receptor or a pharmaceutical composition thereof as described herein.
  • a recombinant receptor e.g., CAR
  • a method for detecting CD38 in a cell or tissue comprising: (a) contacting the cell or tissue with an anti-CD38 antibody or antigen- binding fragment described herein; and (b) determining the amount of the antibody or antigen-binding fragment thereof bound to the cell or tissue, wherein the amount of antibody or antigen-binding fragment thereof bound to the cell or tissue indicates the amount of CD38 in the cell or tissue.
  • the subject has persistent or relapsed disease, e.g., following treatment with another CD38-specific antibody, cells expressing a CD38 -targeting chimeric receptor, a BCMA-specific antibody, cells expressing a BCMA-targeting chimeric receptor, and/or other therapy, including chemotherapy, radiation, and/or hematopoietic stem cell transplantation (HSCT), e.g., allogeneic HSCT or autologous HSCT.
  • HSCT hematopoietic stem cell transplantation
  • the subject has persistent or relapsed disease, e.g., following treatment with a B cell -targeting agent, such as an anti-CD19 antibody cells expressing a CD19-targeting chimeric receptor, or an anti- CD20 antibody or cells expressing a CD20-targeting chimeric receptor.
  • a B cell -targeting agent such as an anti-CD19 antibody cells expressing a CD19-targeting chimeric receptor, or an anti- CD20 antibody or cells expressing a CD20-targeting chimeric receptor.
  • the administration effectively treats the subject despite the subject having become resistant to a BCMA-targeted therapy and/or another CD38-targeted therapy.
  • the administration effectively treats the subject despite the subject having become resistant to a CD19-targeted therapy and/or a CD20-targeted therapy.
  • the subject has not relapsed but is determined to be at risk for relapse, such as at a high risk of relapse, and thus the compound or composition is administered prophylactically, e.g., to reduce the likelihood of or prevent relapse.
  • the subject prior to the initiation of administration of the engineered cells, the subject has received one or more prior therapies. In some embodiments, the subject has received at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 or more prior therapies. In some embodiments, the subject has received at least 3, 4, 5, 6, 7, 8, 9, 10 or more prior therapies. In some embodiments, the subject has received at least 1 prior therapy. In some embodiments, the subject has received at least 2 prior therapies. In some embodiments, the subject has received at least 3 prior therapies. In some embodiments, the subject has received at least 4 prior therapies. [00285] In some aspects, the subject has relapsed or has been refractory to the one or more prior therapies.
  • the prior therapies include treatment with autologous stem cell transplant (ASCT); an immunomodulatory agent (ImiD); a proteasome inhibitor; cells expressing a BCMA-targeted chimeric receptor, a BCMA-specific antibody, and/or an anti-CD38 antibody.
  • ASCT autologous stem cell transplant
  • ImiD immunomodulatory agent
  • the prior therapies include treatment with an anti-CD19 antibody or cells expressing an anti-CD19 chimeric receptor.
  • the prior therapies include treatment with an anti-CD20 antibody (e.g., rituximab).
  • the immunomodulatory agent comprises thalidomide, lenalidomide or pomalidomide.
  • the proteasome inhibitor comprises bortezomib (VELCADE®), carfilzomib, or ixazomib.
  • the cells expressing a BCMA-targeted chimeric receptor are idecabtagene vicleucel (ABECMA®) or ciltacabtagene autoleucel (CARVYKTITM).
  • the BCMA-specific antibody is a bispecific antibody.
  • the BCMA-specific antibody is or comprises teclistamab-cqyv (TECVAYLITM)-
  • the BCMA-specific antibody is an antibody-drug conjugate (ADC).
  • the BCMA specific antibody is or comprises belantamab mafodotin (BLENREP®).
  • the anti-CD38 antibody is or comprises daratumumab (e.g., DARZALEX® or DARZALEX Faspro®).
  • the prior therapies include treatment with an anti-CD19 antibody or cells expressing an anti-CD19 chimeric receptor.
  • the prior therapies include treatment with an anti-CD20 antibody (e.g., rituximab).
  • the cells are isolated and/or otherwise prepared from the subject who is to receive the cell therapy, or from a sample derived from such a subject.
  • the cells are derived from a subject, e.g., patient, in need of a treatment and the cells, following isolation and processing are administered to the same subject.
  • the cells are autologous to the subject to be treated.
  • the cell therapy e.g., adoptive cell therapy, e.g., adoptive T cell therapy
  • the cells are isolated and/or otherwise prepared from a subject other than a subject who is to receive or who ultimately receives the cell therapy, e.g., a first subject.
  • the cells then are administered to a different subject, e.g., a second subject, of the same species.
  • the cells are allogeneic to the subject to be treated.
  • the subject, to whom the cells, cell populations, or compositions are administered is a primate, such as a human.
  • the subject, to whom the cells, cell populations, or compositions are administered is a non-human primate.
  • the non-human primate is a monkey (e.g., cynomolgus monkey) or an ape.
  • the subject can be male or female and can be any suitable age, including infant, juvenile, adolescent, adult, and geriatric subjects.
  • the subject is a non-primate mammal, such as a rodent (e.g., mouse, rat, etc.).
  • the appropriate dosage of the CD38-binding molecule, recombinant receptor or cell may depend on the type of disease to be treated, the type of binding molecule or recombinant receptor, the severity and course of the disease, whether the binding molecule or recombinant receptor is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the recombinant receptor or cell, and the discretion of the attending physician.
  • the compositions and molecules and cells are in some embodiments suitably administered to the patient at one time or over a series of treatments.
  • the dose and/or frequency of administration is determined based on efficacy and/or response. In some embodiments, the dose and/or frequency of administration is determined based on efficacy and/or response. In some embodiments efficacy and/or response is determined by a disease activity index for the disease. For example, in some embodiments efficacy and/or response is determined by the International Myeloma Working Group (IMWG) Uniform Response Criteria. In some examples, dose and/or frequency of administration is determined by the expansion and persistence of the recombinant receptor or cell in the blood and/or bone marrow. In some embodiments, dose and/or frequency of administration is determined based on the antitumor activity of the recombinant receptor or engineered cell.
  • IMWG International Myeloma Working Group
  • treatment of a subject with a genetically engineered cell(s) described herein achieves one, two, three, four, or more of the following effects, including, for example: (i) reduction or amelioration the severity of disease or symptom associated therewith; (ii) reduction in the duration of a symptom associated with a disease; (iii) protection against the progression of a disease or symptom associated therewith; (iv) regression of a disease or symptom associated therewith; (v) protection against the development or onset of a symptom associated with a disease; (vi) protection against the recurrence of a symptom associated with a disease; (vii) reduction in the hospitalization of a subject; (viii) reduction in the hospitalization length; (ix) an increase in the survival of a subject with a disease; (x) a reduction in the number of symptoms associated with a disease; and (xi) an enhancement, improvement, supplementation, complementation, or augmentation of the prophylactic or therapeutic effect(s)
  • the method comprises administering a dose of the engineered cells or a composition comprising a dose of the engineered cells.
  • the engineered cells or compositions containing engineered cells can be used in a treatment regimen, wherein the treatment regimen comprises administering a dose of the engineered cells or a composition comprising a dose of the engineered cells.
  • the dose can contain, for example, a particular number or range of recombinant receptor-expressing immune cells (e.g., NK cells), such as any number of such cells described herein.
  • a composition containing a dose of the cells can be administered.
  • the number, amount or proportion of CAR-expressing cells in a cell population or a cell composition can be assessed by detection of a surrogate marker, e.g., by flow cytometry or other means, or by detecting binding of a labelled molecule, such as a labelled antigen, that can specifically bind to the binding molecules or receptors provided herein.
  • a surrogate marker e.g., by flow cytometry or other means
  • a labelled molecule such as a labelled antigen
  • Doses of immune cells such as NK and/or T cells can be determined for a given subject based on their body mass, disease type and state, and desired aggressiveness of treatment, but range, depending on the embodiments, from about 10 5 cells per kg to about 10 12 cells per kg (e.g., 10 5 -10 7 , 1O 7 -1O 10 , 10’°-10 12 and overlapping ranges therein). In one embodiment, a dose escalation regimen is used. In several embodiments, a range of immune cells such as NK and/or T cells is administered, for example between about 1 x 10 6 cells/kg to about 1 x 10 8 cells/kg.
  • a range of NK cells is administered, for example between about 1 x 10 6 cells/kg to about 1 x 10 8 cells/kg.
  • a range of T cells is administered, for example between about 1 x 10 6 cells/kg to about 1 x 10 8 cells/kg.
  • a dose of engineered cells comprises between about 300 x 10 6 and 1.5 x 10 9 NK cells. In some embodiments, a dose of engineered cells comprises about 300 x 10 6 NK cells. In some embodiments, a dose of engineered cells comprises about 1 x 10 9 NK cells. In some embodiments, a dose of engineered cells comprises about 1.5 x 10 9 NK cells. In some embodiments, a dose of engineered cells comprises about 2 x 10 9 NK cells. In some embodiments, a dose of engineered cells comprises about 2.5 x 10 9 NK cells. In some embodiments, a dose of engineered cells comprises about 3 x 10 9 NK cells.
  • a dose of engineered cells comprises between about 300 x 10 6 and 3 x 10 9 CAR-expressing NK cells. In some embodiments, a dose of engineered cells comprises about 300 x 10 6 CAR-expressing NK cells. In some embodiments, a dose of engineered cells comprises about 1 x 10 9 CAR-expressing NK cells. In some embodiments, a dose of engineered cells comprises about 1.5 x 10 9 CAR-expressing NK cells. In some embodiments, a dose of engineered cells comprises about 2 x 10 9 CAR-expressing NK cells. In some embodiments, a dose of engineered cells comprises about 2.5 x 10 9 CAR-expressing NK cells. In some embodiments, a dose of engineered cells comprises about 3 x 10 9 CAR-expressing NK cells.
  • a dose of engineered cells comprises between about 300 x 10 6 and 1.5 x 10 9 T cells. In some embodiments, a dose of engineered cells comprises about 300 x 10 6 T cells. In some embodiments, a dose of engineered cells comprises about 1 x 10 9 T cells. In some embodiments, a dose of engineered cells comprises about 1.5 x 10 9 T cells.
  • a dose of engineered cells comprises both NK and T cells. In some embodiments, a dose of engineered cells comprises between about 300 x 10 6 and 1.5 x 10 9 NK cells and between about 300 x 10 6 and 1.5 x 10 9 T cells. In some embodiments, a dose comprises about an equal number of NK cells and T cells. In some embodiments, a dose comprises more NK cells than T cell. In some embodiments, a dose comprises more T cells than NK cells.
  • multiple doses are used, for example, two, three, four, or more doses within a dosing cycle. Such multi-dose cycles can be repeated one or more times, as needed to treat a tumor and/or prevent disease progression.
  • dosing is, for example, 3 doses of about 0.5 x 10 9 NK cells or about 2.5 x 10 9 NK cells administered over about 21 to 28 days.
  • dosing is, for example, 3 doses of about 1.0 x 10 9 NK cells or about 1.5 x 10 9 NK cells administered over about 21 to 28 days.
  • a dosing cycle comprises 3 doses of about 1.0 x 10 9 NK cells administered over about 21 to 28 days.
  • a dosing cycle comprises 3 doses of about 1.5 x 10 9 NK cells administered over about 21 to 28 days. In several embodiments, a dosing cycle comprises 3 doses of about 2.0 x 10 9 NK cells administered over about 21 to 28 days.
  • a lymphodepletion process is performed prior to the first dose.
  • the administration of engineered NK cells is preceded by one or more preparatory treatments.
  • the administration of engineered NK cells is preceded by a lymphodepleting therapy (also referred to as “lymphodepletion”).
  • a combination of chemotherapeutic agents is used for lymphodepletion.
  • a single chemotherapeutic agent is used for lymphodepletion.
  • agents with different mechanisms of actions are optionally used.
  • different classes of agents are optionally used.
  • an antimetabolic agent is used.
  • the antimetabolic agent inhibits and/or prevents cell replication.
  • the antimetabolic agent is an altered nucleotide that disrupts DNA replication, making it effective in targeting rapidly dividing tumor cells.
  • cytosine arabinoside is used.
  • a dose of between about 0.2 - about 10 g/nr Ara-C is administered, including doses of about 0.2 g/m 2 , about 0.5 g/m 2 , about 1.0 g/m 2 , about 1.5 g/nr, about 2.0 g/m 2 , about 2.5 g/m 2 , about 3.0 g/m 2 , about 3.5 g/m 2 , about 4.0 g/m 2 , about 5.0 g/m 2 , about 6.0 g/m 2 , about 7.0 g/m 2 , about 8.0 g/m 2 , about 9.0 g/m 2 , about 10.0 about 1.5 g/m 2 , or any dose between those listed.
  • a dose of about 2 g/m 2 of Ara-C is administered.
  • the dose of Ara-C is given daily for at least about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, or about 7 days.
  • the dose of Ara-C is given daily for about 5 days.
  • the dose can be split and given, for example, twice daily.
  • an additional agent is used in combination with the Ara-C.
  • the additional agent is also an antimetabolite.
  • the additional agent inhibits one or more of DNA polymerase alpha, ribonucleotide reductase and/or DNA primase, thus inhibiting DNA synthesis.
  • the additional agent is fludarabine.
  • a dose of between about 5.0 mg/m 2 - about 200 mg/m 2 fludarabine is administered, including doses of about 5.0 mg/m 2 , about 10.0 mg/m 2 , about 15.0 mg/m 2 , about 20.0 mg/m 2 , about 25.0 mg/m 2 , about 30.0 mg/m 2 , about 35.0 mg/m 2 , about 40.0 mg/m 2 , about 45.0 mg/m 2 , about 50.0 mg/m 2 , about 60.0 mg/m 2 , about 70.0 mg/m 2 , about 80.0 mg/m 2 , about 90.0 mg/m 2 , about 100.0 mg/m 2 , about 125.0 mg/m 2 , about 150.0 mg/m 2 , about 175.0 mg/m 2 , about 200.0 mg/m 2 , or any dose between those listed.
  • a dose of about 30 mg/m 2 of fludarabine is administered.
  • the dose of fludarabine is given daily for at least about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, or about 7 days.
  • the dose of fludarabine is given daily for at about 3 days.
  • the dose of fludarabine is given daily for at about 5 days.
  • the dose can be split and given, for example, twice daily.
  • a combination of fludarabine and Ara-C is used with a daily dose of fludarabine of between about 20 mg/m 2 and 40 mg/m 2 and a daily dose of Ara-C of between about 1.5 g/m 2 and 2.5 g/m 2 .
  • a combination of fludarabine and Ara-C is used with a daily dose of fludarabine of about 30 mg/m 2 and a daily dose of Ara-C of about 2 g/m 2 .
  • the combination of fludarabine and Ara-C is administered for about 5 days, with the administration started about 7 days prior to the first administration of the engineered NK cells (for example day - 7 to day -3).
  • lymphodepletion is started at day -5 prior to administration of engineered NK cells.
  • this combination advantageously functions not only as a lymphodepletion regimen, but as an anti-cancer agent as well (in addition to the engineered NK cells).
  • the lymphodepletion regimen works synergistically with the engineered NK cells to provide effect reduction and/or elimination of cancerous cells.
  • the additional agent is cyclophosphamide.
  • a dose of between about 100 mg/m 2 - about 100 mg/m 2 fludarabine is administered, including doses of about 100.0 mg/m 2 , about 200 mg/m 2 , about 300 mg/m 2 , about 400 mg/m 2 , about 500 mg/m 2 , about 600 mg/m 2 , about 700 mg/m 2 , about 800 mg/m 2 , about 900 mg/m 2 , about 1000 mg/m 2 , or any dose between those listed.
  • a dose of about 300 mg/m 2 of cyclophosphamide is administered.
  • a dose of about 500 mg/m 2 of cyclophosphamide is administered. In several embodiments, a dose of about 600 mg/m 2 of cyclophosphamide is administered. In several embodiments, a dose of about 700 mg/m 2 of cyclophosphamide is administered. In several embodiments, a dose of about 800 mg/m 2 of cyclophosphamide is administered. In several embodiments, a dose of about 900 mg/m 2 of cyclophosphamide is administered. In several embodiments, a dose of about 1000 mg/m 2 of cyclophosphamide is administered. In several embodiments, the dose of cyclophosphamide is given once.
  • the dose of cyclophosphamide is given once between about 5 days and about 2 days prior to administration of the engineered NK cells. In several embodiments, the dose of cyclophosphamide is given once between about 5 days prior to administration of the engineered NK cells. In several embodiments, the dose of cyclophosphamide is given once between about 4 days prior to administration of the engineered NK cells. In several embodiments, the dose of cyclophosphamide is given once between about 3 days prior to administration of the engineered NK cells. In several embodiments, the dose of cyclophosphamide is given once between about 2 days prior to administration of the engineered NK cells.
  • the dose of cyclophosphamide is given daily for at least about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, or about 7 days. In several embodiments, the dose of cyclophosphamide is given daily for about 3 days. In several embodiments, if necessary, the dose can be split and given, for example, twice daily.
  • a combination of fludarabine and cyclophosphamide is used.
  • cyclophosphamide (300 mg/m 2 ) and fludarabine (30mg/m 2 ) are administered daily for 3 days.
  • cyclophosphamide (500 mg/m 2 ) and fludarabine (30mg/m 2 ) are administered daily for 3 days.
  • fludarabine and cyclophosphamide are each administered daily 5 days, 4 days, and 3 days prior to administration of the engineered NK cells.
  • a dose of a genetically engineered cell(s) described herein or composition thereof is administered to a subject every day, every other day, every couple of days, every third day, once a week, twice a week, three times a week, or once every two weeks.
  • two, three or four doses of a genetically engineered cell(s) described herein or composition thereof is administered to a subject every day, every couple of days, every third day, once a week or once every two weeks.
  • a dose(s) of a genetically engineered cell(s) described herein or composition thereof is administered for 2 days, 3 days, 5 days, 7 days, 14 days, or 21 days.
  • a dose of a genetically engineered cell(s) described herein or composition thereof is administered for 1 month, 1.5 months, 2 months, 2.5 months, 3 months, 4 months, 5 months, 6 months or more.
  • a subject is subject to lymphodepletion at least one time prior to administration of genetically engineered cells as disclosed herein.
  • lymphodepletion is performed before one or more additional doses of engineered cells are administered.
  • a dosing cycle is used that comprises lymphodepletion followed by at least two doses of engineered cells as disclosed herein, with the two doses separated by a time interval.
  • the time interval is 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or more days (including intervals falling between the time marking a price interval since the last administration, e.g., 84 hours, or 3.5 days).
  • the dosing cycle itself is approximately 14, 21, 28, 35, 42 or more days.
  • three doses are administered, each about 2-5 days apart from each other. In several embodiments, all three doses are administered within about 5-7 days.
  • a subject receives a first dose on day 0 of the cycle, a second dose on day 3 of the cycle and a third dose on day 7 of the cycle. In several embodiments, three doses are administered, ⁇ 1 week apart from each other. In several embodiments, two doses are administered ⁇ 1 week apart from one another. In several embodiments, a subject receives a first dose on day 0 of the cycle, a second dose on day 7 of the cycle and a third dose on day 14 of the cycle.
  • a 28 day cycle is used with primary outcome measures evaluated at day 28 (see e.g., Figure 3A).
  • a subject receives a first dose on day 0 of the cycle and a second dose on day 7 of the cycle.
  • a 28 day cycle is used with primary outcome measures evaluated at day 28 (See e.g., Figure 3B).
  • lymphodepletion is performed prior to the inception of each dosing cycle, if subsequent dosing cycles are required (e.g., the subject requires further treatment).
  • a subject undergoes lymphodepletion, receives a plurality of doses of engineered cells according to a cycle, is evaluated at the end of the cycle time and, if deemed necessary undergoes a second lymphodepletion followed by a second dosing cycle.
  • a first and a second dosing cycle need not be the same (e.g., a first cycle may have 2 doses, while a second uses three doses).
  • 2, 3, 4, 5, 6, 7, 8, 9, 10, or more dosing cycles are performed.
  • an additional therapeutic agent is administered at least once during the lymphodepletion and/or the dosing cycle.
  • the cells, binding molecules, or recombinant receptors are administered as part of a combination treatment, such as simultaneously with or sequentially with, in any order, another therapeutic intervention, such as another antibody or engineered cell or receptor or agent, such as a cytotoxic or therapeutic agent.
  • the cells, binding molecules and/or recombinant receptors in some embodiments are co- administered with one or more additional therapeutic agents or in connection with another therapeutic intervention, either simultaneously or sequentially in any order.
  • the cells are co-administered with another therapy sufficiently close in time such that the cell populations enhance the effect of one or more additional therapeutic agents, or vice versa.
  • the cells, binding molecules and/or recombinant receptors are administered prior to the one or more additional therapeutic agents.
  • the cells, binding molecules and/or recombinant receptors are administered after to the one or more additional therapeutic agents.
  • Combination Therapy includes administration and uses, such as therapeutic and prophylactic uses, of the CD38-binding recombinant receptors (e.g., CARs), engineered cells expressing the recombinant receptors (e.g., CARs), plurality of engineered cells expressing the receptors, and/or compositions comprising the same.
  • CD38-binding recombinant receptors e.g., CARs
  • engineered cells expressing the recombinant receptors e.g., CARs
  • plurality of engineered cells expressing the receptors e.g., CD38-binding recombinant receptors
  • compositions comprising the same.
  • the CD38-binding recombinant receptor (e.g., chimeric antigen receptor) and/or engineered cells expressing said molecules (e.g., recombinant receptor) described herein are administered as part of a combination treatment or combination therapy, such as simultaneously with, sequentially with or intermittently with, in any order, one or more additional therapeutic intervention.
  • the one or more additional therapeutic intervention includes, for example, an antibody, an engineered cell, a receptor and/or an agent, such as a cell expressing a recombinant receptor, and/or cytotoxic or therapeutic agent, e.g., a chemotherapeutic agent.
  • the combination therapy includes administration of one or more additional agents, therapies and/or treatments, e.g., any of the additional agents, therapy and/or treatments described herein.
  • the combination treatment or combination therapy includes an additional treatment, such as a surgical treatment, transplant, and/or radiation therapy.
  • additional treatment such as a surgical treatment, transplant, and/or radiation therapy.
  • methods of combination treatment or combination therapy that includes CD38-binding recombinant receptors (e.g., CARs), cells and/or compositions described herein and one or more additional therapeutic interventions.
  • the additional therapy, treatment or agent includes chemotherapy, radiation therapy, surgery, transplantation, adoptive cell therapy, antibodies, cytotoxic agents, chemotherapeutic agents, cytokines, growth inhibitory agents, anti-hormonal agents, kinase inhibitors, anti-angiogenic agents, cardioprotectants, immunostimulatory agents, immunosuppressive agents, immune checkpoint inhibitors, antibiotics, angiogenesis inhibitors, metabolic modulators or other therapeutic agents or any combination thereof.
  • the additional agent is a protein, a peptide, a nucleic acid, a small molecule agent, a cell, a toxin, a lipid, a carbohydrate or combinations thereof, or any other type of therapeutic agent, e.g.
  • the additional therapy, agent or treatment includes surgery, chemotherapy, radiation therapy, transplantation, administration of cells expressing a recombinant receptor, e.g., CAR, kinase inhibitor, immune checkpoint inhibitor, mTOR pathway inhibitor, immunosuppressive agents, immunomodulators, antibodies, immunoablative agents, antibodies and/or antigen binding fragments thereof, antibody conjugates, other antibody therapies, cytotoxins, steroids, cytokines, peptide vaccines, hormone therapy, antimetabolites, metabolic modulators, alkylating agents, anthracyclines, vinca alkaloids, proteasome inhibitors, protein kinase inhibitors, and/or other types of immunotherapy.
  • the additional agent or treatment is bone marrow transplantation, T cell ablative therapy using chemotherapy agents such as, fludarabine, external-beam radiation therapy (XRT), cyclophosphamide, and/or antibody therapy.
  • the additional agent is a chemotherapeutic agent.
  • the chemotherapeutic agent comprises an antimetabolite, an alkylating agent, a topoisomerase inhibitor, a mitotic inhibitor, an antibiotic, a protein kinase inhibitor, a proteasome inhibitor, an inhibitor of poly (ADP-ribose) polymerase (PARP), or a combination thereof.
  • PARP poly (ADP-ribose) polymerase
  • the chemotherapeutic agent comprises an antimetabolite.
  • the antimetabolite comprises methotrexate, pemetrexed, cytarabine, 5 -fluorouracil (5-FU), capecitabine, gemcitabine, 6-mercaptopurine (6-MP), azathioprine, fludarabine, cladribine, hydroxyurea, or any combination thereof.
  • the therapeutic agent comprises gemcitabine.
  • the therapeutic agent comprises fluorouracil and leucovorin (FOL).
  • the therapeutic agent comprises capecitabine and ovaliplatin (CAPOX).
  • the chemotherapeutic agent comprises an alkylating agent.
  • the alkylating agent comprises cyclophosphamide, ifosfamide, chlorambucil, melphalan, temozolomide, carmustine, lomustine, streptozocin, busulfan, procarbazine, cisplatin, carboplatin, oxaliplatin, or any combination thereof.
  • the therapeutic agent comprises cisplatin.
  • the therapeutic agent comprises bendamustine.
  • the therapeutic agent comprises cyclophosphamide.
  • the therapeutic agent comprises dacarbazine.
  • the therapeutic agent comprises procarbazine.
  • the therapeutic agent comprises capecitabine and ovaliplatin (CAPOX).
  • the chemotherapeutic agent comprises a topoisomerase inhibitor.
  • the topoisomerase inhibitor comprises irinotecan, topotecan, etoposide, or any combination thereof.
  • the therapeutic agent comprises etoposide.
  • the chemotherapeutic agent comprises a mitotic inhibitor.
  • the mitotic inhibitor comprises vincristine, vinblastine, vinorelbine, docetaxel, paclitaxel, eribulin, izabepilone, epothilone, or any combination thereof.
  • the therapeutic agent comprises vincristine.
  • the therapeutic agent comprises vinblastine.
  • the therapeutic agent comprises paclitaxel.
  • the therapeutic agent comprises docetaxel.
  • the chemotherapeutic agent comprises an antibiotic.
  • the antibiotic comprises bleomycin; actinomycin D; an anthracycline, optionally doxorubicin, daunorubicin, or idarubicin, mitomycin, or any combination thereof.
  • the therapeutic agent comprises bleomycin.
  • the therapeutic agent comprises an anthracy cline.
  • the therapeutic agent comprises doxorubicin.
  • the therapeutic agent comprises daunorubicin.
  • the chemotherapeutic agent comprises a protein kinase inhibitor.
  • the protein kinase inhibitor comprises an inhibitor of BCR-ABL, c-KIT, EGFR, VEGF, ALK, BRAF, MAPK, MEK, mTOR, BTK, JAK, CDK, or any combination thereof.
  • the protein kinase inhibitor comprises an inhibitor of BCR-ABL.
  • the protein kinase inhibitor comprises an inhibitor of BCL2 (e.g., venetoclax).
  • the protein kinase inhibitor comprises an inhibitor of c- Kit.
  • the protein kinase inhibitor comprises an inhibitor of EGFR (e.g., cetuximab). In some embodiments, the protein kinase inhibitor comprises an inhibitor of BRAF (e.g., vemurafenib, dabrafenib or encorafenib). In some embodiments, the protein kinase inhibitor comprises an inhibitor of MAPK (e.g., vemurafenib, dabrafenib, or trametinib). In some embodiments, the protein kinase inhibitor comprises an inhibitor of MEK (e.g., trametinib, binimetinib, selumetinib, or cobimetinib).
  • BRAF e.g., vemurafenib, dabrafenib or encorafenib
  • MAPK e.g., vemurafenib, dabrafenib, or trametinib
  • MEK
  • the protein kinase inhibitor comprises an inhibitor of mTOR (e.g., sirolimus, everolimus, or temsirolimus). In some embodiments, the protein kinase inhibitor comprises an inhibitor of BTK (e.g., ibrutinib). In some embodiments, the protein kinase inhibitor comprises an inhibitor of JAK. In some embodiments, the protein kinase inhibitor comprises an inhibitor of CDK.
  • mTOR e.g., sirolimus, everolimus, or temsirolimus
  • BTK e.g., ibrutinib
  • JAK e.g., JAK
  • CDK protein kinase inhibitor
  • the chemotherapeutic agent comprises a proteasome inhibitor.
  • the proteasome inhibitor comprises bortezomib, carfilzomib, ixazomib, or any combination thereof.
  • the proteasome inhibitor comprises bortezomib.
  • the proteasome inhibitor comprises carfilzomib.
  • the proteasome inhibitor comprises ixazomib.
  • the chemotherapeutic agent comprises a PARP inhibitor.
  • the PARP inhibitor comprises olaparib.
  • the therapeutic agent comprises niraparib.
  • the PARP inhibitor comprises rucaparib.
  • the additional agent is a hormone therapy or a therapeutic agent that disrupts or modifies a hormone pathway.
  • the additional agent comprises tamoxifen.
  • the additional agent comprises hyperthermia.
  • the additional agent is an additional form of immunotherapy.
  • the additional agent comprises photodynamic therapy (PDT).
  • the additional agent comprises radiation therapy.
  • the additional agent comprises transplanted stem cells.
  • the cells, CD38-binding recombinant receptors and/or compositions, e.g., CAR-expressing cells are administered in combination with other engineered cells, e.g., other CAR-expressing cells.
  • the additional therapy or treatment is cell therapy, e.g., adoptive cell therapy.
  • the additional therapy includes administration of engineered cells, e.g., additional CAR-expressing cell.
  • the additional engineered cell is a CAR-expressing cell that expresses the same or different recombinant receptor as the engineered cells provided herein, e.g., anti-CD38 CAR-expressing cells.
  • the recombinant receptor, e.g., CAR, expressed on the additional engineered cell recognizes a different antigen, e.g., BCMA, or a different epitope of CD38.
  • the additional agent is a cell engineered to express a recombinant receptor, e.g., CAR, targeting a second or additional antigen expressed in multiple myeloma.
  • the recombinant receptor, e.g., CAR expressed on the additional engineered cell recognizes BCMA.
  • the recombinant receptor, e.g., CAR, expressed on the additional engineered cell recognizes a different epitope of CD38.
  • the additional engineered cell is administered prior to, concurrently with, or after administration (e.g., infusion) of the CAR-expressing cells described herein.
  • the additional engineered cell is allogeneic to the subject being treated.
  • the additional engineered cell is autologous to the subject being treated.
  • the CAR molecules expressing by the different engineered cells may have the same or a different primary intracellular signaling domain, the same or different costimulatory signaling domains, or the same number or a different number of costimulatory signaling domains.
  • the additional agent includes any of the cells or plurality of cells described herein.
  • the therapeutic agent is an NK cell engager (e.g., a molecule that binds both an antigen expressed by cells of the cancer and an antigen expressed by NK cells).
  • the NK cell engager binds to an activating receptor on an NK cell and an antigen expressed by cells of the cancer.
  • the activating receptor on the NK cell is selected from the group consisting of CD 16, NKp30, NKp46, NKG2D, and any combination thereof.
  • the additional agent includes an immune checkpoint inhibitor.
  • Immune checkpoint inhibitors include any agent that blocks or inhibits in a statistically significant manner, the inhibitory pathways of the immune system. Such inhibitors may include small molecule inhibitors or may include antibodies, or antigen binding fragments thereof, that bind to and block or inhibit immune checkpoint receptors, ligands and/or receptor-ligand interaction. In some embodiments, modulation, enhancement and/or stimulation of particular receptors can overcome immune checkpoint pathway components.
  • Illustrative immune checkpoint molecules that may be targeted for blocking, inhibition, modulation, enhancement and/or stimulation include, but are not limited to, PD-1 (CD279), PD-L1 (CD274, B7-H1), CTLA-4, LAG-3 (CD223), TIM-3, 4-1BB (CD137), 4-1BBL (CD137L), GITR (TNFRSF18, AITR), CD40, 0X40 (CD134, TNFRSF4), B7-H3, B7-H4, B7H3, B7H4, VISTA, KIR, 2B4, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), TIGIT, and LAIR1.
  • PD-1 CD279
  • PD-L1 CD274, B7-H1
  • CTLA-4 LAG-3
  • TIM-3 4-1BB
  • 4-1BBL CD137L
  • GITR TNFRSF18, AITR
  • CD40 0X40
  • the additional agent is a CD19 inhibitor, e.g., an anti- CD19 antibody (e.g., an anti-CD19 mono- or bi-specific antibody) or a fragment thereof, antibody-drug conjugate (ADC), or engineered toxin body (ETB).
  • the CD19 inhibitor is an anti-CD19 antibody.
  • the CD19 inhibitor is an ADC.
  • the CD 19 inhibitor is an ETB.
  • the CD 19 inhibitor is a small molecule.
  • the additional agent is a CD20 inhibitor, e.g., an anti- CD20 antibody (e.g., an anti-CD20 mono- or bi-specific antibody) or a fragment thereof, antibody-drug conjugate (ADC), or engineered toxin body (ETB).
  • the CD20 inhibitor is an anti-CD20 antibody.
  • anti-CD20 antibodies include rituximab (RITUXAN®) and obinutuzumab (GAZYVA®).
  • the additional agent is or includes rituximab.
  • the additional agent is or includes obinutuzumab.
  • the CD20 inhibitor is an ADC.
  • the CD20 inhibitor is an ETB.
  • the CD20 inhibitor is a small molecule.
  • the additional agent is a CD22 inhibitor, e.g., an anti- CD22 antibody (e.g., an anti-CD22 mono- or bi-specific antibody) or a fragment thereof, antibody-drug conjugate (ADC), or engineered toxin body (ETB).
  • the CD22 inhibitor is an ADC.
  • the CD22 inhibitor is an ETB.
  • the CD22 inhibitor is a small molecule.
  • the additional agent is an EGFR inhibitor, e.g., an anti-EGFR antibody (e.g., an anti-EGFR mono- or bi-specific antibody) or a fragment thereof, antibody-drug conjugate (ADC), or engineered toxin body (ETB).
  • the EGFR inhibitor is an anti-EGFR antibody.
  • anti-EGFR antibodies include cetuximab, panitumumab (VECTIBIX®), nimotuzumab, and necitumumab (PORTRAZZATM).
  • the additional agent is or includes cetuximab.
  • the EGFR inhibitor is an ADC.
  • the EGFR inhibitor is an ETB.
  • the EGFR inhibitor is a small molecule.
  • the additional agent a BCMA inhibitor, e.g., an anti- BCMA antibody (e.g., an anti-BCMA mono- or bi-specific antibody) or a fragment thereof, antibody-drug conjugate (ADC), or engineered toxin body (ETB).
  • anti- BCMA antibodies include teclistamab-cqyv (TECVAYLITM), AMG 420 (also known as BI 836909), PF-06863135 (also known as PF-3135), JNJ-957, REGN5458, TNB-383B, and CC- 93269.
  • the BCMA inhibitor is or includes teclistamab-cqyv.
  • the BCMA inhibitor is an ADC.
  • Exemplary ADCs include belantamab mafodotin (BLEFREP®), GSK2857916, HDP-101, and MEDI2228. See Shah et al., Leukemia (2020) 34:985-1005.
  • the BCMA inhibitor is an ETB.
  • the BCMA inhibitor is a small molecule.
  • the additional agent is a SLAMF7 inhibitor, e.g., an anti-SLAMF7 antibody (e.g., an anti-SLAMF7 mono- or bi-specific antibody) or a fragment thereof, antibody-drug conjugate (ADC), or engineered toxin body (ETB).
  • Non-limiting examples of anti-SLAMF7 antibodies include elotuzumab (EMPLICITI®).
  • the SLAMF7 inhibitor is or includes elotuzumab.
  • the SLAMF7 inhibitor is an ADC.
  • the SLAMF7 inhibitor is an ETB.
  • the SLAMF7 inhibitor is a small molecule.
  • the additional agent is a CD38 inhibitor, e.g., an anti- CD38 antibody (e.g., an anti-CD38 mono- or bi-specific antibody) or a fragment thereof, antibody-drug conjugate (ADC), or engineered toxin body (ETB).
  • anti- CD38 antibodies include daratumumab (e.g., DARZALEX® or DARZALEX Faspro®), isatuximab (SARCLISA®), MOR202, GBR 1342 (also known as ISB-1342), TAK-079, and TAK-169. See Gozzetti et al., Hum Vaccin Immunother (2022) 18(5):2052658.
  • the anti-CD38 antibody is or includes daratumumab.
  • daratumumab is administered at a dose of between about 10 mg/kg and about 25 mg/kg or between about 15 mg/kg and about 20 mg/kg (e.g., about 16 mg/kg).
  • a dose of daratumumab is administered about every week, about every two weeks, about every three weeks, or about every four weeks.
  • daratumumab is administered intravenously.
  • daratumumab is administered at a dose of between about 1,500 mg and about 3,000 mg (e.g, about 1,800 mg).
  • the dose of daratumumab is administered in combination with a dose of with hyaluronidase.
  • hyaluronidase is administered at a dose of between about 20,000 units and about 40,000 units (e.g., about 30,000) units.
  • a dose of daratumumab and a dose of hyaluronidase and administered in combination about every week, about every two weeks, about every three weeks, or about every four weeks.
  • daratumumab and hyaluronidase are administered in combination (e.g., as a single formulation or composition) subcutaneously.
  • the anti-CD38 antibody is or includes isatuximab.
  • isatuximab is administered at a dose of between about 5 mg/kg and about 15 mg/kg (e.g., about 10 mg/kg). In some embodiments, a dose of isatuximab is administered about every week or about every two weeks. In some embodiments, isatuximab is administered intravenously. In some embodiments, the CD38 inhibitor is a small molecule.
  • the engineered cells are genetically edited to reduce transcription of the CD38 gene and/or expression of CD38 protein, such as described in Section I.A.ii.
  • the genetically engineered cells are “knocked out” (KO) for CD38, such as by a CRISPR/Cas system (e.g., a CD38-targeting ribonucleoprotein (RNP) complex).
  • the genetic edit is carried out by using a DNA binding protein or DNA-binding nucleic acid, which specifically binds to or hybridizes to the CD38 gene at a region targeted for disruption.
  • the protein or nucleic acid is coupled to or complexed with a nuclease, such as in a chimeric or fusion protein.
  • a nuclease such as a Zinc Finger Nuclease (ZFN) or TAL-effector nuclease (TALEN), or an RNA-guided nuclease such as a clustered regularly interspersed short palindromic nucleic acid (CRISPR)-Cas system, such as CRISPR-Cas9 system, specific for the gene being disrupted.
  • ZFN Zinc Finger Nuclease
  • TALEN TAL-effector nuclease
  • RNA-guided nuclease such as a clustered regularly interspersed short palindromic nucleic acid (CRISPR)-Cas system, such as CRISPR-Cas9 system, specific for the gene being disrupted.
  • CRISPR clustered regularly interspersed short palindromic nucleic acid
  • the additional agent is an immunomodulatory agent (IMiD).
  • the immunomodulatory agent comprises lenalidomide, pomalidomide, thalidomide, or any combination thereof.
  • the immunomodulatory agent comprises lenalidomide.
  • the immunomodulatory agent comprises pomalidomide.
  • the immunomodulatory agent comprises thalidomide.
  • the additional agent is or comprises a steroid.
  • the steroid is a corticosteroid.
  • the steroid is a glucocorticoid.
  • the steroid comprises dexamethasone or prednisone.
  • the steroid is or comprises dexamethasone.
  • the steroid is or comprises prednisone.
  • any of the additional agents described herein can be prepared and administered as combination therapy with the CD38-binding recombinant receptor (e.g., chimeric antigen receptor) and/or engineered cells expressing said molecules (e.g., recombinant receptor) described herein, such as in pharmaceutical compositions comprising one or more agents of the combination therapy and a pharmaceutically acceptable carrier, such as any described herein.
  • the CD38-binding recombinant receptor e.g., chimeric antigen receptor
  • engineered cells expressing said molecules e.g., recombinant receptor
  • the CD38-binding recombinant receptor e.g., chimeric antigen receptor
  • engineered cells expressing said molecules e.g., recombinant receptor
  • plurality of engineered cells expressing said molecules e.g., recombinant receptor
  • the additional agent can be co-administered with the CD38-binding recombinant receptors, cells and/or compositions described herein, for example, as part of the same pharmaceutical composition or using the same method of delivery.
  • the additional agent is administered simultaneously with the CD38-binding recombinant receptors, cells and/or compositions described herein, but in separate compositions.
  • the additional agent is an additional engineered cell, e.g., cell engineered to express a different recombinant receptor, and is administered in the same composition or in a separate composition.
  • the additional agent is incubated with the engineered cell, e.g., CAR-expressing cells, prior to administration of the cells.
  • the additional agent is administered subsequent to or prior to the administration of the CD38-binding recombinant receptors, cells and/or compositions described herein, separated by a selected time period.
  • the time period is 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 1 week, 2 weeks, 3 weeks, 1 month, 2 months, or 3 months.
  • the additional agent is administered multiple times and/or the CD38-binding recombinant receptors, cells and/or compositions described herein, is administered multiple times.
  • the additional agent is administered prior to the CD38- binding recombinant receptors, cells and/or compositions described herein, e.g., two weeks, 12 days, 10 days, 8 days, one week, 6 days, 5 days, 4 days, 3 days, 2 days or 1 day before the administration.
  • the additional agent is administered after the CD38-binding recombinant receptors, cells and/or compositions described herein, e.g., two weeks, 12 days, 10 days, 8 days, one week, 6 days, 5 days, 4 days, 3 days, 2 days or 1 day after the administration.
  • the dose of the additional agent can be any therapeutically effective amount, e.g., any dose amount described herein, and the appropriate dosage of the additional agent may depend on the type of disease to be treated, the type, dose and/or frequency of the recombinant receptor, cell and/or composition administered, the severity and course of the disease, whether the recombinant receptor, cell and/or composition is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the recombinant receptor, cell and/or composition, and the discretion of the attending physician.
  • the recombinant receptor, cell and/or composition and/or the additional agent and/or therapy can be administered to the patient at one time, repeated or administered over a series of treatments.
  • kits containing the provided antibodies, recombinant receptors (e.g., CARs), genetically engineered cells, and/or compositions comprising the same.
  • the articles of manufacture may include a container and a label or package insert on or associated with the container.
  • Suitable containers include, for example, bottles, vials, syringes, test tubes, IV solution bags, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container has a sterile access port.
  • Non-limiting examples of containers include intravenous solution bags and vials, including those with stoppers pierceable by a needle for injection.
  • the article of manufacture or kit may further include a package insert indicating that the composition can be used to treat a particular condition such as a condition described herein (e.g., cancer).
  • the article of manufacture or kit may further include another or the same container comprising a pharmaceutically-acceptable buffer. It may further include other materials such as other buffers, diluents, filters, needles, and/or syringes.
  • the label or package insert may indicate that the composition is used for treating aCD38-expressing disease, disorder, or condition in an individual.
  • the label or a package insert, which is on or associated with the container may indicate directions for reconstitution and/or use of the formulation.
  • the label or package insert may further indicate that the formulation is useful or intended for subcutaneous, intravenous, or other modes of administration for treating or preventing a CD38-expressing disease, disorder, or condition in an individual.
  • the container in some embodiments holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the condition.
  • the article of manufacture or kit may include (a) a first container with a composition contained therein (i.e., first medicament), wherein the composition includes the antibody (e.g., anti-CD38 antibody) or antigen-binding fragment thereof or recombinant receptor (e.g., CAR); and (b) a second container with a composition contained therein (i.e., second medicament), wherein the composition includes a further agent, such as a cytotoxic or otherwise therapeutic agent, and which article or kit further comprises instructions on the label or package insert for treating the subject with the second medicament, in an effective amount.
  • a further agent such as a cytotoxic or otherwise therapeutic agent
  • Fc region herein is used to define a C-terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region.
  • the term includes native sequence Fc regions and variant Fc regions.
  • a human IgG heavy chain Fc region extends from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain.
  • the C-terminal lysine (Lys447) of the Fc region may or may not be present.
  • numbering of amino acid residues in the Fc region or constant region is according to the EU numbering system, also called the EU index, as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD, 1991.
  • full length antibody “intact antibody,” and “whole antibody” are used herein interchangeably to refer to an antibody having a structure substantially similar to a native antibody structure or having heavy chains that contain an Fc region as defined herein.
  • An "isolated" antibody is one which has been separated from a component of its natural environment.
  • an antibody is purified to greater than 95% or 99% purity as determined by, for example, electrophoretic (e.g. , SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatographic (e.g. , ion exchange or reverse phase HPLC).
  • electrophoretic e.g. , SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis
  • chromatographic e.g. , ion exchange or reverse phase HPLC.
  • An isolated nucleic acid includes a nucleic acid molecule contained in cells that ordinarily contain the nucleic acid molecule, but the nucleic acid molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location.
  • isolated nucleic acid encoding an anti-CD38 antibody refers to one or more nucleic acid molecules encoding antibody heavy and light chains (or fragments thereof), including such nucleic acid molecule(s) in a single vector or separate vectors, and such nucleic acid molecule(s) present at one or more locations in a host cell.
  • host cell refers to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells.
  • Host cells include “transformants” and “transformed cells,” which include the primary transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein.
  • polypeptide and protein are used interchangeably to refer to a polymer of amino acid residues, and are not limited to a minimum length.
  • Polypeptides including the antibodies and antibody chains and other peptides, e.g. , linkers and CD38-binding peptides, may include amino acid residues including natural and/or non-natural amino acid residues.
  • the terms also include post-expression modifications of the polypeptide, for example, glycosylation, sialylation, acetylation, phosphorylation, and the like.
  • the polypeptides may contain modifications with respect to a native or natural sequence, as long as the protein maintains the desired activity. These modifications may be deliberate, as through site-directed mutagenesis, or may be accidental, such as through mutations of hosts which produce the proteins or errors due to PCR amplification.
  • percent (%) amino acid sequence identity and “percent identity” and “sequence identity” when used with respect to an amino acid sequence (reference polypeptide sequence) is defined as the percentage of amino acid residues in a candidate sequence (e.g. , the subject antibody or fragment) that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
  • An amino acid substitution may include replacement of one amino acid in a polypeptide with another amino acid.
  • Amino acid substitutions may be introduced into a binding molecule, e.g. , antibody, of interest and the products screened for a desired activity, e.g., retained/improved antigen binding, or decreased immunogenicity.
  • Amino acids generally can be grouped according to the following common side- chain properties:
  • Non-conservative amino acid substitutions will involve exchanging a member of one of these classes for another class.
  • vector refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked.
  • the term includes the vector as a selfreplicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced.
  • Certain vectors are capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as "expression vectors.”
  • package insert is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products.
  • composition refers to any mixture of two or more products, substances, or compounds, including cells. It may be a solution, a suspension, liquid, powder, a paste, aqueous, non-aqueous or any combination thereof.
  • a statement that a cell or population of cells is "positive" for a particular marker refers to the detectable presence on or in the cell of a particular marker, typically a surface marker.
  • a surface marker refers to the presence of surface expression as detected by flow cytometry, for example, by staining with an antibody that specifically binds to the marker and detecting said antibody, wherein the staining is detectable by flow cytometry at a level substantially above the staining detected carrying out the same procedure with an isotype -matched control under otherwise identical conditions and/or at a level substantially similar to that for cell known to be positive for the marker, and/or at a level substantially higher than that for a cell known to be negative for the marker.
  • a statement that a cell or population of cells is "negative" for a particular marker refers to the absence of substantial detectable presence on or in the cell of a particular marker, typically a surface marker.
  • a surface marker refers to the absence of surface expression as detected by flow cytometry, for example, by staining with an antibody that specifically binds to the marker and detecting said antibody, wherein the staining is not detected by flow cytometry at a level substantially above the staining detected carrying out the same procedure with an isotype-matched control under otherwise identical conditions, and/or at a level substantially lower than that for cell known to be positive for the marker, and/or at a level substantially similar as compared to that for a cell known to be negative for the marker.
  • An anti-CD38 antibody or an antigen-binding fragment thereof comprising: (i) a heavy chain variable region (VH) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:25, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:23; and a light chain variable region (VL) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:27, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:28, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:29;
  • VH heavy chain variable region
  • VL light chain variable region
  • a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:30, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:31, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:32; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:35;
  • a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:36, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:37, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:38; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:40;
  • VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:41, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:42; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:43, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:45;
  • a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:46, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:48; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:49, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:50, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:51;
  • a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:61, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:62; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:63;
  • VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:66; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:68;
  • (x) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:69; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:70, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:71;
  • a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:72, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:73, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:74; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:75, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:76, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:77; or
  • a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:78, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:79, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:80; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:81.
  • An anti-CD38 antibody or an antigen- binding fragment thereof comprising: (i) a heavy chain variable region (VH) comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 82, and a light chain variable region (VL) comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:83;
  • VH heavy chain variable region
  • VL light chain variable region
  • VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:88, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:89;
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:82
  • the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:83;
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:84, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:85;
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:86
  • the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:87;
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:88
  • the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:89;
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:90
  • the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:91;
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:92
  • the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:93;
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:94
  • the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:95;
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:96
  • the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:97;
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:98
  • the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:99;
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 100
  • the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 101;
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 102
  • the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 103;
  • the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 104
  • the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 105.
  • anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-3 which is a chimeric antibody or an antigen-binding fragment thereof.
  • anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-3 which comprises a human variable region framework region.
  • VH comprises the amino acid sequence set forth in SEQ ID NO:82
  • VL comprises the amino acid sequence set forth in SEQ ID NO:83
  • VH comprises the amino acid sequence set forth in SEQ ID NO:84
  • VL comprises the amino acid sequence set forth in SEQ ID NO:85
  • the VH comprises the amino acid sequence set forth in SEQ ID NO: 86
  • the VL comprises the amino acid sequence set forth in SEQ ID NO:87;
  • VH comprises the amino acid sequence set forth in SEQ ID NO: 88
  • VL comprises the amino acid sequence set forth in SEQ ID NO:89
  • VH comprises the amino acid sequence set forth in SEQ ID NO:90
  • VL comprises the amino acid sequence set forth in SEQ ID NO:91
  • VH comprises the amino acid sequence set forth in SEQ ID NO: 92
  • VL comprises the amino acid sequence set forth in SEQ ID NO:93;
  • VH comprises the amino acid sequence set forth in SEQ ID NO:94
  • VL comprises the amino acid sequence set forth in SEQ ID NO:95
  • VH comprises the amino acid sequence set forth in SEQ ID NO:96
  • VL comprises the amino acid sequence set forth in SEQ ID NO:97;
  • VH comprises the amino acid sequence set forth in SEQ ID NO:98
  • VL comprises the amino acid sequence set forth in SEQ ID NO:99
  • the VH comprises the amino acid sequence set forth in SEQ ID NO: 100
  • the VL comprises the amino acid sequence set forth in SEQ ID NO: 101;
  • the VH comprises the amino acid sequence set forth in SEQ ID NO: 102
  • the VL comprises the amino acid sequence set forth in SEQ ID NO: 103;
  • the VH comprises the amino acid sequence set forth in SEQ ID NO: 104
  • the VL comprises the amino acid sequence set forth in SEQ ID NO: 105.
  • scFv comprises a linker between the VH and VL, the linker comprising the amino acid sequence set forth in SEQ ID NO: 1 or SEQ ID NO:3.
  • ECD extracellular domain
  • KD binding affinity
  • composition comprising the anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-13.
  • composition of embodiment 14, comprising a pharmaceutically acceptable carrier.
  • An immunoconjugate comprising the anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-13 and a therapeutic agent.
  • a composition comprising the immunoconjugate of embodiment 16 or embodiment 17 and a pharmaceutically acceptable carrier.
  • a bispecific molecule comprising the anti-CD38 antibody or antigen-binding fragment of any one of embodiments 1-13 and a second moiety that has a binding specificity for an immune cell.
  • composition comprising the bispecific molecule of any one of embodiments 19-22 and a pharmaceutically acceptable carrier.
  • a method for detecting CD38 in a cell or tissue comprising:
  • An anti-CD38 chimeric antigen receptor comprising an extracellular antigenbinding domain, a transmembrane domain, and an intracellular signaling domain, wherein the extracellular antigen-binding domain comprises the anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-13.
  • the co-stimulatory signaling region comprises an intracellular signaling region of 0X40, CD28, 4-1BB, ICOS, or DAP10.
  • the anti-CD38 CAR of embodiment comprising a CD8alpha transmembrane domain and an intracellular signaling domain comprising a CD3zeta domain and a co-stimulatory region comprising an intracellular signaling region of 0X40 peptide.
  • the anti-CD38 CAR of any one of embodiments 26-33 wherein the CAR comprises the amino acid sequence set forth in SEQ ID NO:327, SEQ ID NO:328, SEQ ID NO:329, SEQ ID NO:330, SEQ ID NO:331, SEQ ID NO:332, SEQ ID NO:333, SEQ ID NO:334, SEQ ID NO:335, SEQ ID NO:336, SEQ ID NO:337, or SEQ ID NO:338.
  • a vector comprising the nucleic acid molecule of embodiment 35.
  • the vector of embodiment 36 which is a viral vector.
  • invention 38 The vector of embodiment 36 or embodiment 37, which is a retroviral vector.
  • a cell comprising the anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-13, the anti-CD38 CAR of any one of embodiments 26-34, the nucleic acid molecule of embodiment 35, or the vector of any one of embodiments 36-38.
  • the cell of embodiment 39 which is a natural killer (NK) cell or a T cell.
  • NK natural killer
  • a natural killer (NK) cell comprising the anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-13 or the anti-CD38 CAR of any one of embodiments 26-34.
  • composition comprising a plurality of the cell of any one of embodiments 39-43.
  • composition of embodiment 44 comprising a pharmaceutically acceptable carrier.
  • composition of embodiment 44 or embodiment 45 comprising between about 3 x 108 CAR-expressing cells and about 3 x 109 CAR-expressing cells, or between about 1 x 109 and about 2 x 109 CAR-expressing cells.
  • a method of treatment comprising administering to a subject having a CD38-expressing disease or condition an anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-13, an anti-CD38 CAR of any one of embodiments 26-34, a cell of any one of embodiments 39-43, a composition of any one of embodiments 14, 15, 18, 23, and 44-46, an immunoconjugate of embodiment 16 or embodiment 17, or a bispecific molecule of any one of embodiments 19-22.
  • a method of treatment comprising administering to a subject having a CD38-expressing disease or condition a composition comprising immune cells expressing an anti-CD38 chimeric antigen receptor (CAR) comprising an extracellular antigen-binding domain, a transmembrane domain, and an intracellular signaling domain, wherein the extracellular antigen-binding domain comprises:
  • VH heavy chain variable region
  • VL light chain variable region
  • a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:30, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:31 , and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:32; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:35;
  • a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:36, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:37, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:38; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:40;
  • VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:41, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:42; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:43, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:45;
  • a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:46, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:48; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:49, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:50, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:51;
  • VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:52, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:53; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:54;
  • a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:55, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:56, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:57; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:58, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:59, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NQ:60;
  • a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:61, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:62; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:63;
  • VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:66; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:68;
  • (x) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:69; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:70, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:71;
  • a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:72, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:73, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:74; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:75, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:76, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:77; or
  • a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:78, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:79, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:80; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:81.
  • a method of treatment comprising administering to a subject having a CD38-expressing disease or condition a composition comprising immune cells expressing an anti-CD38 chimeric antigen receptor (CAR) comprising an extracellular antigen-binding domain, a transmembrane domain, and an intracellular signaling domain, wherein the extracellular antigen-binding domain comprises:
  • VH heavy chain variable region
  • VL light chain variable region
  • VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:88, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:89;
  • cancer selected from the group consisting of leukemia, lymphoma, myeloma, glioma, thyroid cancer, lung cancer, colorectal cancer, head and neck cancer, stomach cancer, liver cancer, pancreatic cancer, renal cancer, urothelial cancer, prostate cancer, testicular cancer, breast cancer, cervical cancer, endometrial cancer, ovarian cancer, and melanoma.
  • the CD38-expressing disease or condition is an autoimmune disease or disorder.
  • the autoimmune disease or disorder is selected from the group consisting of systemic lupus erythematosus (SLE), lupus nephritis, CNS lupus, myositis, multiple sclerosis (MS), myasthenia gravis (MG), rheumatoid arthritis (RA), scleroderma, thyroid disease, diabetes, vasculitis, or any combination thereof.
  • a method of producing a cell targeting CD38 comprising introducing into the cell the nucleic acid molecule of embodiment 35 or the vector of any one of embodiments 36-38.
  • Non-limiting examples of anti-CD38 antibodies formatted as single-chain variable fragments (scFvs), were generated and assessed for binding to CD38.
  • fully human variable domain anti-CD38 antibodies were generated by immunizing human 1g transgenic mice with adjuvant and his-tagged human CD38 extracellular domain (ECD) protein (Aero Biosystems, Newark, DE).
  • ECD extracellular domain
  • B lymphocytes were isolated from the lymph nodes of the immunized mice and fused with SP2/0 cells, and the resultant hybridoma cells were single cell sorted into 96- well plates.
  • [00366] 94 of the 192 clones were formatted as chimeric antigen receptors (CARs) for further testing, as described in Example 2.
  • CARs chimeric antigen receptors
  • the sequences and CD38 binding affinity for the top 12 clones described in Example 2 are shown below in Table El.
  • a known fully human anti- CD38 scFv “A2” was included as a reference antibody (“Ref’; described in PCT Application Nos. PCT/US2016/026545 and PCT/IB2018/058642).
  • the majority of the non-limiting examples of anti-CD38 scFvs were observed to have a higher binding affinity for human CD38 than the anti- CD38 reference antibody, as evidenced by a lower dissociation constant (KD).
  • CARs chimeric antigen receptors
  • each of the CAR constructs contained, from N- to C- terminus: a human anti-CD38 scFv (e.g., SEQ ID NOS: 108-120), a CD8a hinge (e.g., SEQ ID NO:6), a CD8a transmembrane domain (e.g., SEQ ID NO: 8), and an intracellular signaling region containing an 0X40 co-stimulatory region (e.g., SEQ ID NO: 14) and a CD3 zeta intracellular signaling domain (e.g., SEQ ID NO: 18).
  • a human anti-CD38 scFv e.g., SEQ ID NOS: 108-120
  • CD8a hinge e.g., SEQ ID NO:6
  • a CD8a transmembrane domain e.g., SEQ ID NO: 8
  • cDNA clones encoding the CARs were linked to a downstream T2A ribosomal skip element (e.g., a T2A-encoding sequence SEQ ID NO: 19, encoding SEQ ID NO:20) followed by a membrane-bound interleukin- 15 (mbIL15)- encoding sequence (e.g., a sequence encoding SEQ ID NO:23), and cloned into a retroviral expression vector.
  • T2A ribosomal skip element e.g., a T2A-encoding sequence SEQ ID NO: 19, encoding SEQ ID NO:20
  • mbIL15 membrane-bound interleukin- 15
  • CARs chimeric antigen receptors
  • NK cells primary natural killer cells
  • CAR expression was determined by measuring the surface expression of the FLAG tag by flow cytometry. Antigen-dependent and tonic activation were assessed by incubating the CD38 CAR-expressing Jurkat cells overnight in the presence or absence of CD38- expressing MM.1R target cells, respectively, and subsequently measuring surface expression of CD69 on the Jurkat cells by flow cytometry. Antigen-dependent activation and CAR expression were compared to CD38 binding affinity.
  • CAR expression was also compared to the epitope bin for each of the 44 clones.
  • the clones were observed to bind a variety of different epitopes, which in some cases overlapped with one another ( Figure 3); many of those demonstrating high expression when formatted as a CAR were observed to bind to the same epitope region of CD38 (bin 4).
  • NK cells were isolated by immunoaffinity-based enrichment from leukapheresis samples from two different healthy human donors. Isolated NK cells were knocked out for CD38 expression on Day 0 and transduced on Day 6 with a retroviral vector encoding a CAR as set forth in Table E2 and mbIL15. Transduced cells were allowed to expand in culture until they were analyzed for expression of CD38 and CAR on Days 11 and 14. As controls, NK cells were (1) knocked out for CD38 and subjected to mock transduction (CD38 KO); or (2) subjected to mock electroporation and mock transduction (EP only).
  • NK cells isolated from two different healthy human donors were knocked out for CD38 and engineered to express a CD38-directed CAR incorporating one of the 12 non- limiting examples of anti-CD38 scFvs or the reference antibody (Ref), as described above.
  • the CD38 CAR-expressing NK cells were incubated with CD38-expressing THP-1 or MM.
  • Target cell killing was assessed by IncuCyte® analysis over approximately 14 days, including at 24 hours post-tumor rechallenge with MM.
  • NK cells expressing the non-limiting examples of CD38-directed CARs were observed to consistently exhibit cytotoxicity against CD38-expressing target cells.
  • a summary of the relative cytotoxicity of cells expressing the non-limiting examples of CARs at the indicated ratios is shown in Table

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Provided herein are CD38 binding molecules, in particular, to anti-CD38 antibodies, including antibody fragments and chimeric antigen receptors (CARs) incorporating the CD38 binding molecules. Also provided are pharmaceutical compositions containing the binding molecules, genetically engineered cells expressing the CARs, and methods and uses of the provided binding molecules and engineered cells for treating diseases and conditions such as cancer.

Description

ANTIBODIES AND CHIMERIC ANTIGEN RECEPTORS SPECIFIC FOR CD38
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority to United States Provisional Application No. 63/492714, filed March 28, 2023, the entire contents of which is incorporated by reference herein.
FIELD
[0002] The present disclosure relates to CD38 binding molecules, in particular, to anti-CD38 antibodies, including antibody fragments. The present disclosure further relates to recombinant receptors containing such antibodies, including chimeric antigen receptors (CARs). The disclosure further relates to genetically engineered cells expressing such antibodies and receptors, and uses thereof, including in cell therapy.
BACKGROUND
[0003] CD38 is a type II transmembrane glycoprotein expressed on the surface of a variety of cancer cells, including hematologic malignancies and solid tumors. CD38 expression is associated with immunosuppressive effects, and its abnormal overexpression is associated with cancer progression. While various CD38-binding molecules, including anti-CD38 antibodies and CARs are available, improved CD38-binding molecules and engineered CD38-targeting cells are needed. Provided are embodiments that meet such needs.
INCORPORATION BY REFERENCE OF MATERIAL IN SEQUENCE LISTING FILE
[0004] This application incorporates by reference the material contained in the Sequence Listing XML file being submitted concurrently herewith: File name: NKT.102WO_ST26.xml; created on March 26, 2024 and is 330, 520 bytes in size.
SUMMARY
[0005] Provided herein are anti-CD38 antibodies or antigen-binding fragments thereof. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof comprises: (i) a heavy chain variable region (VH) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:25, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:23; and a light chain variable region (VL) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:27, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:28, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:29; (ii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:30, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:31, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:32; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:35; (iii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:36, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:37, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:38; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:40; (iv) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:41, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:42; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:43, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:45; (v) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:46, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:48; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:49, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:50, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:51; (vi) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:52, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:53; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:54; (vii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:55, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:56, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:57; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:58, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:59, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:60; (viii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:61, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:62; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:63; (ix) a VH comprising a CDRI comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:66; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:68; (x) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:69; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:70, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:71; (xi) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:72, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:73, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:74; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:75, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:76, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:77; or (xii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:78, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:79, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:80: and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:81.
[0006] In some embodiments, the anti-CD38 antibody or antigen binding fragment comprises a heavy chain variable region (VH) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:25, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:23; and a light chain variable region (VL) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:27, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:28, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:29. In some embodiments, the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:30, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:31, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:32; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:35. In some embodiments, the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:36, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:37, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:38; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:40. In some embodiments, the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:41, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:42: and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:43, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:45. In some embodiments, the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDRI comprising the amino acid sequence set forth in SEQ ID NO:46, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:48: and a VL comprising a CDRI comprising the amino acid sequence set forth in SEQ ID NO:49, a CDR2 comprising the amino acid sequence set forth in SEQ ID NQ:50, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:51. In some embodiments, the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:52, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:53: and a VL comprising a CDRI comprising the amino acid sequence set forth in SEQ ID NO: 33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:54. In some embodiments, the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDRI comprising the amino acid sequence set forth in SEQ ID NO:55, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:56, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:57; and a VL comprising a CDRI comprising the amino acid sequence set forth in SEQ ID NO:58, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:59, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:60. In some embodiments, the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDRI comprising the amino acid sequence set forth in SEQ ID NO:61, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:62: and a VL comprising a CDRI comprising the amino acid sequence set forth in SEQ ID NO: 39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:63. In some embodiments, the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDRI comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:66: and a VL comprising a CDRI comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:68. In some embodiments, the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:69; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:70, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:71. In some embodiments, the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:72, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:73, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:74; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:75, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:76, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:77. In some embodiments, the anti-CD38 antibody or antigen binding fragment comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:78, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:79, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:80: and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:81.
[0007] In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises: (i) a heavy chain variable region (VH) comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:82, and a light chain variable region (VL) comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:83; (ii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:84, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:85; (iii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:86, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:87; (iv) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:88, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:89; (v) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:90, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:91; (vi) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:92, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:93; (vii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:94, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:95; (viii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:96, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:97; (ix) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:98, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:99; (x) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 100, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 101; (xi) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 102, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 103; or (xii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 104, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 105.
[0008] In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a heavy chain variable region (VH) comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 82, and a light chain variable region (VL) comprising the CDR 1 , the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:83. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a heavy chain variable region (VH) comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 82. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a light chain variable region (VL) comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:83. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:84, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:85. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 84. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 85. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:86, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:87. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:86. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:87. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:88, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:89. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:88. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 89. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:90, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:91. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NQ:90. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:91. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:92, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:93. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:94, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:95. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:94. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:95. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:96, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:97. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:96. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:97. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:98, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:99. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:98. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:99. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 100, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 101. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 100. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 101. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 102, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 103. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 102. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 103. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 104, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 105. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 104. In some embodiments, the anti-CD38 antibody or an antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 105.
[0009] In some embodiments, (i) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:82, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:83; (ii) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 84, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 85; (iii) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:86, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:87; (iv) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:88, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:89; (v) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:90, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:91; (vi) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:92, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:93; (vii) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:94, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:95; (viii) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:96, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:97; (ix) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:98, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:99; (x) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 100, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 101; (xi) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 102, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 103; or (xii) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 104, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 105.
[0010] In some embodiments, the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:82, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:83. In some embodiments, the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:84, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:85. In some embodiments, the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:86, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:87. In some embodiments, the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:88, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:89. In some embodiments, the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:90, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:91. In some embodiments, the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:92, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:93. In some embodiments, the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:94, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:95. In some embodiments, the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:96, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:97. In some embodiments, the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:98, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:99. In some embodiments, the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 100, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 101. In some embodiments, the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 102, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 103. In some embodiments, the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 104, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 105.
[0011] In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof is a chimeric antibody or an antigen-binding fragment thereof. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof is a humanized antibody or an antigenbinding fragment thereof. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof comprises a human variable region framework region. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof is a fully human antibody or antigenbinding fragment thereof.
[0012] In some embodiments, (i) the VH comprises the amino acid sequence set forth in SEQ ID NO:82, and the VL comprises the amino acid sequence set forth in SEQ ID NO:83; (ii) the VH comprises the amino acid sequence set forth in SEQ ID NO:84, and the VL comprises the amino acid sequence set forth in SEQ ID NO:85; (iii) the VH comprises the amino acid sequence set forth in SEQ ID NO:86, and the VL comprises the amino acid sequence set forth in SEQ ID NO:87; (iv) the VH comprises the amino acid sequence set forth in SEQ ID NO:88, and the VL comprises the amino acid sequence set forth in SEQ ID NO:89; (v) the VH comprises the amino acid sequence set forth in SEQ ID NO:90, and the VL comprises the amino acid sequence set forth in SEQ ID NO:91; (vi) the VH comprises the amino acid sequence set forth in SEQ ID NO:92, and the VL comprises the amino acid sequence set forth in SEQ ID NO:93; (vii) the VH comprises the amino acid sequence set forth in SEQ ID NO:94, and the VL comprises the amino acid sequence set forth in SEQ ID NO:95; (viii) the VH comprises the amino acid sequence set forth in SEQ ID NO:96, and the VL comprises the amino acid sequence set forth in SEQ ID NO:97; (ix) the VH comprises the amino acid sequence set forth in SEQ ID NO:98, and the VL comprises the amino acid sequence set forth in SEQ ID NO:99; (x) the VH comprises the amino acid sequence set forth in SEQ ID NO: 100, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 101; (xi) the VH comprises the amino acid sequence set forth in SEQ ID NO: 102, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 103; or (xii) the VH comprises the amino acid sequence set forth in SEQ ID NO: 104, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 105.
[0013] In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:82, and the VL comprises the amino acid sequence set forth in SEQ ID NO:83. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:82. In some embodiments, the VL comprises the amino acid sequence set forth in SEQ ID NO:83. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO: 84, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 85. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:84. In some embodiments, the VL comprises the amino acid sequence set forth in SEQ ID NO:85. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:86, and the VL comprises the amino acid sequence set forth in SEQ ID NO:87. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:86. In some embodiments, the VL comprises the amino acid sequence set forth in SEQ ID NO:87. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:88, and the VL comprises the amino acid sequence set forth in SEQ ID NO:89. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:88. In some embodiments the VL comprises the amino acid sequence set
-li forth in SEQ ID NO:89. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:90, and the VL comprises the amino acid sequence set forth in SEQ ID NO:91. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:90. In some embodiments the VL comprises the amino acid sequence set forth in SEQ ID NO:91. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:92, and the VL comprises the amino acid sequence set forth in SEQ ID NO:93. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:92. In some embodiments, the VL comprises the amino acid sequence set forth in SEQ ID NO:93. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:94, and the VL comprises the amino acid sequence set forth in SEQ ID NO:95. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:94. In some embodiments, the VL comprises the amino acid sequence set forth in SEQ ID NO:95. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:96, and the VL comprises the amino acid sequence set forth in SEQ ID NO:97. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:96. In some embodiments, the VL comprises the amino acid sequence set forth in SEQ ID NO:97. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:98, and the VL comprises the amino acid sequence set forth in SEQ ID NO:99. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:98. In some embodiments, the VL comprises the amino acid sequence set forth in SEQ ID NO:99. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO: 100, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 101. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO: 100,. In some embodiments, the VL comprises the amino acid sequence set forth in SEQ ID NO: 101. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO: 102, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 103. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO: 102. In some embodiments, the VL comprises the amino acid sequence set forth in SEQ ID NO: 103. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO: 104, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 105. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO: 104. In some embodiments, the VL comprises the amino acid sequence set forth in SEQ ID NO: 105.
[0014] In some embodiments, the antigen-binding fragment of the antibody is a
Fab, a Fab’, a F(ab’)2, a Fv, or a single-chain variable fragment (scFv). In some embodiments, the antigen-binding fragment of the antibody is a Fab. In some embodiments, the antigen-binding fragment of the antibody is a Fab’. In some embodiments, the antigen-binding fragment of the antibody is a F(ab’)2. In some embodiments, the antigen- binding fragment of the antibody is a Fv. In some embodiments, the antigen-binding fragment of the antibody is a scFv. [0015] In some embodiments, the scFv comprises a linker between the VH and VL, the linker comprising the amino acid sequence set forth in SEQ ID NO: 1 or SEQ ID NO:3. In some embodiments, the linker comprises the amino acid sequence set forth in SEQ ID NO: 1. In some embodiments, the linker comprises the amino acid sequence set forth in SEQ ID NO: 3.
[0016] In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 108, SEQ ID NO: 109, SEQ ID NO: 110, SEQ ID NO: 111, SEQ ID NO: 112, SEQ ID NO: 113, SEQ ID NO:114, SEQ ID NO:115, SEQ ID NO:116, SEQ ID NO: 117, SEQ ID NO: 118, or SEQ ID NO:119. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 108. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 109. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 110. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 111. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 112. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 113. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 114. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 115. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 1 16. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 117. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 118. In some embodiments, the scFv comprises the amino acid sequence set forth in SEQ ID NO: 119.
[0017] In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof binds to the extracellular domain (ECD) of human CD38 with a binding affinity (KD) of between about 1 nM and about 500 nM, or between about 4 nM and about 40 nM. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof binds to the extracellular domain (ECD) of human CD38 with a binding affinity (KD) of between about 1 nM and about 500 nM. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof binds to the extracellular domain (ECD) of human CD38 with a binding affinity (KD) of between about 4 nM and about 40 nM.
[0018] Also provided herein is a composition comprising any of the anti-CD38 antibody or antigen-binding fragment thereof provided herein. In some embodiments, the composition comprises a pharmaceutically acceptable carrier.
[0019] Also provided herein is an immunoconjugate comprising any of the anti- CD38 antibody or antigen-binding fragment thereof provided herein and a cytotoxic agent. In some embodiments, the cytotoxic agent is a drug, a cytotoxin, or a radioactive agent. In some embodiments, the cytotoxic agent is a drug. In some embodiments, the cytotoxic agent is a cytotoxin. In some embodiments, the cytotoxic agent is a radioactive agent. [0020] Also provided herein is a composition comprising any of the immunoconjugate provided herein and a pharmaceutically acceptable carrier.
[0021] Also provided herein is a bispecific molecule comprising any of the anti- CD38 antibody or antigen-binding fragment thereof provided herein and a second moiety that has a binding specificity for an immune cell. In some embodiments, the immune cell is a natural killer (NK) cell or a T cell. In some embodiments, the immune cell is a natural killer (NK) cell. In some embodiments, the immune cell is a T cell. In some embodiments, the second moiety binds to one or more of CD 16, NKp30, NKp46, NKG2D, or a combination thereof. In some embodiments, the second moiety binds to CD3. In some embodiments, the second moiety binds to CD16. In some embodiments, the second moiety binds to NKp30. In some embodiments, the second moiety binds to NKp46. In some embodiments, the second moiety binds to NKG2D.
[0022] Also provided herein is a composition comprising any of the bispecific molecules provided herein and a pharmaceutically acceptable carrier.
[0023] Also provided herein is a method for detecting CD38 in a cell or tissue, including: (a) contacting the cell or tissue with any of the anti-CD38 antibodies or antigen-binding fragments thereof provided herein; and (b) determining the amount of the antibody or antigenbinding fragment thereof bound to the cell or tissue, wherein the amount of the antibody or antigen-binding fragment thereof bound to the cell or tissue indicates the amount of CD38 in the cell or tissue.
[0024] Also provided herein is use of any of the anti-CD38 antibody or antigenbinding fragment thereof provided herein in the manufacture of a kit for detecting CD38 in a cell or tissue, including: (a) contacting the cell or tissue with the anti-CD38 antibody or antigenbinding fragment thereof; and (b) determining the amount of the antibody or antigen-binding fragment thereof bound to the cell or tissue, wherein the amount of the antibody or antigenbinding fragment thereof bound to the cell or tissue indicates the amount of CD38 in the cell or tissue.
[0025] Also provided herein is anti-CD38 chimeric antigen receptor (CAR) comprising an extracellular antigen-binding domain, a transmembrane domain, and an intracellular signaling domain. In some embodiments, the extracellular antigen-binding domain comprises any of the anti-CD38 antibodies or antigen-binding fragments thereof provided herein.
[0026] In some embodiments, the extracellular antigen-binding domain comprises: (i) a heavy chain variable region (VH) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:25, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:23; and a light chain variable region (VL) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:27, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:28, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:29; (ii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:30, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:31, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:32; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:35; (iii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 36, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:37, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:38; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:40; (iv) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:41, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:42; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:43, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:45; (v) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:46, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:48; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:49, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:50, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:51;(vi) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:52, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:53; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:54; (vii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:55, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:56, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:57; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:58, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:59, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:60; (viii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:61, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:62: and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:63; (ix) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:66; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:68; (x) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:69; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:70, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:71; (xi) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:72, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:73, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:74; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:75, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:76, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:77; or (xii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:78, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:79, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:80; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:81.
[0027] In some embodiments, the extracellular antigen-binding domain comprises: (i) a heavy chain variable region (VH) comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:82, and a light chain variable region (VL) comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:83; (ii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:84, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:85; (iii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:86, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:87; (iv) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:88, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 89; (v) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:90, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:91; (vi) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:92, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:93; (vii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:94, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:95; (viii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:96, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:97; (ix) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:98, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:99; (x) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 100, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 101; (xi) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 102, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 103; or (xii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 104, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 105.
[0028] In some embodiments, the transmembrane domain is or comprises a transmembrane from CD8, CD3, CD4, or CD28. In some embodiments, the transmembrane domain is or comprises a transmembrane from CD8. In some embodiments, the transmembrane domain is or comprises a transmembrane from CD8alpha. In some embodiments, the transmembrane domain is or comprises a transmembrane from CD3. In some embodiments, the transmembrane domain is or comprises a transmembrane from CD4. In some embodiments, the transmembrane domain is or comprises a transmembrane from CD28. In some embodiments, the transmembrane domain is human.
[0029] In some embodiments, the intracellular signaling domain comprises a CD3zeta domain. In some embodiments, the CD3zeta domain is a human CD3zeta domain. In some embodiments, the intracellular signaling domain comprises a co- stimulatory signaling region. In some embodiments, the co-stimulatory signaling region comprises an intracellular signaling region of 0X40, CD28, 4- IBB, ICOS, or DAP 10. In some embodiments, the costimulatory signaling region comprises an intracellular signaling region of 0X40. In some embodiments, the co-stimulatory signaling region comprises an intracellular signaling region of CD28. In some embodiments, the co-stimulatory signaling region comprises an intracellular signaling region of 4-1B. In some embodiments, the co-stimulatory signaling region comprises an intracellular signaling region of ICOS. In some embodiments, the co-stimulatory signaling region comprises an intracellular signaling region of DAP10. In some embodiments, the CAR comprises a CD8alpha transmembrane domain and an intracellular signaling domain comprising a CD3zeta domain and a co-stimulatory region comprising an intracellular signaling region of 0X40.
[0030] In some embodiments, the CAR comprises the amino acid sequence set forth in any one of SEQ ID NOS: 327-338. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 327. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 328. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 329. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 330. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 331. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 332. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 333. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 334. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 335. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 336. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 337. In some embodiments, the CAR comprises the amino acid sequence set forth in SEQ ID NO: 338.
[00 1] Also provided herein is a nucleic acid molecule encoding any of the anti- CD38 antibodies or antigen-binding fragments thereof provided herein.
[0032] Also provided herein is a nucleic acid molecule encoding any of the anti- CD38 CARs provided herein.
[0033] Also provided herein is a vector comprising any of the nucleic acid molecules provided herein. In some embodiments, the vector is a viral vector. In some embodiments, the vector is a retroviral vector. In some embodiments, the vector is a lentiviral vector.
[0034] Also provided herein is a cell comprising any of the anti-CD38 antibodies or antigen-binding fragments thereof provided herein.
[0035] Also provided herein is a cell comprising any of the anti-CD38 CARs provided herein.
[0036] Also provided herein is a cell comprising any of the nucleic acid molecules provided herein.
[0037] Also provided herein is a cell comprising any of the vectors provided herein.
[0038] In some embodiments, the cell is a natural killer (NK) cell or a T cell. In some embodiments, the cell is a NK cell. In some embodiments, the cell is a T cell. In some embodiments, the cell is genetically edited to reduce expression of CD38.
[0039] Also provided herein is a natural killer (NK) cell comprising any of the anti- CD38 antibodies or antigen-binding fragments thereof provided herein. In some embodiments, the NK cell is genetically edited to reduce expression of CD38. Also provided herein is a T cell comprising any of the anti-CD38 antibodies or antigen-binding fragments thereof provided herein. In some embodiments, the T cell is genetically edited to reduce expression of CD38.
[0040] Also provided herein is a natural killer (NK) cell comprising any of the anti- CD38 CARs provided herein. In some embodiments, the NK cell is genetically edited to reduce expression of CD38. Also provided herein is a T cell comprising any of the anti-CD38 CARs provided herein. In some embodiments, the T cell is genetically edited to reduce expression of CD38. [0041] In some embodiments, the NK cell is genetically edited to reduce expression of CD38. In some embodiments, the T cell is genetically edited to reduce expression of CD38. In some embodiments, the genetic editing reduces the expression of CD38 by at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, or at least about 80%. In some embodiments, the genetic editing reduces the expression of CD38 by at least about 20%. In some embodiments, the genetic editing reduces the expression of CD38 by at least about 30%. In some embodiments, the genetic editing reduces the expression of CD38 by at least about 40%. In some embodiments, the genetic editing reduces the expression of CD38 by at least about 50%. In some embodiments, the genetic editing reduces the expression of CD38 by at least about 60%. In some embodiments, the genetic editing reduces the expression of CD38 by at least about 70%. In some embodiments, the genetic editing reduces the expression of CD38 by at least about 80%.
[0042] In some embodiments, the cell comprises a membrane -bound interleukin- 15 (mbIL15). In some embodiments, the CAR and the mbIL15 are encoded bicistronically by the same nucleic acid molecule. In some embodiments, the cell comprises a nucleic acid molecule encoding the CAR and the mbIL15, wherein the sequences encoding the CAR and the mbIL15 are separated by a T2A-encoding sequence.
[0043] Also provided herein is a composition comprising a plurality of any of the cells provided herein. Also provided herein is a composition comprising a plurality of any of the NK cells provided herein. Also provided herein is a composition comprising a plurality of any of the T cells provided herein. In some embodiments, the composition comprises a pharmaceutically acceptable carrier.
[0044] In some embodiments, the composition comprises between about 3 x 108 CAR-expressing cells and about 3 x 109 CAR-expressing cells, or between about 1 x 109 CAR- expressing cells and about 2 x 109 CAR-expressing cells. In some embodiments, the composition comprises between about 2 x 109 CAR-expressing cells and about 5 x 109 CAR-expressing cells. In some embodiments, the composition comprises between about 3 x 108 CAR-expressing cells and about 3 x 109 CAR-expressing cells. In some embodiments, the composition comprises between about 1 x 10q and about 2 x 109 CAR-expressing cells. In some embodiments, the composition comprises about 3 x 108 CAR-expressing cells. In some embodiments, the composition comprises about 1 x 109 CAR-expressing cells. In some embodiments, the composition comprises about 1.5 x 109 CAR-expressing cells. In some embodiments, the composition comprises about 2 x 109 CAR-expressing cells. In some embodiments, the composition comprises about 2.5 x 109 CAR-expressing cells. In some embodiments, the composition comprises about 3 x 109 CAR-expressing cells. In some embodiments, the composition comprises about 3.5 x 109 CAR-expressing cells. In some embodiments, the composition comprises about 4 x 109 CAR-expressing cells. In some embodiments, the composition comprises about 4.5 x 109 CAR-expressing cells. In some embodiments, the composition comprises about 5 x 109 CAR-expressing cells.
[0045] Also provided herein is a method of treatment comprising administering to a subject having a CD38-expressing disease or condition, any of the anti-CD38 antibodies or antigen-binding fragments provided herein. Also provided herein is a method of treatment comprising administering to a subject having a disease or disorder associated with CD38, any of the anti-CD38 antibodies or antigen-binding fragments provided herein.
[0046] Also provided herein is a method of treatment comprising administering to a subject having a CD38-expressing disease or condition any of the anti-CD38 CARs provided herein. Also provided herein is a method of treatment comprising administering to a subject having a disease or disorder associated with CD38, any of the anti-CD38 CARs provided herein.
[0047] Also provided herein is a method of treatment comprising administering to a subject having a CD38-expressing disease or condition any of the cells provided herein. In some embodiments, the cells are allogeneic to the subject. Also provided herein is a method of treatment comprising administering to a subject having a disease or disorder associated with CD38, any of the cells provided herein. In some embodiments, the cells are allogeneic to the subject.
[0048] Also provided herein is a method of treatment comprising administering to a subject having a CD38-expressing disease or condition, any of the compositions provided herein. Also provided herein is a method of treatment comprising administering to a subject having a disease or disorder associated with CD38 (e.g., elevated CD38 expression), any of the compositions provided herein.
[0049] Also provided herein is a method of treatment comprising administering to a subject having a CD38-expressing disease or condition, any of the immunoconjugates provided herein. Also provided herein is a method of treatment comprising administering to a subject having a disease or disorder associated with CD38, any of the immunoconjugates provided herein. Also provided herein is a method of treatment comprising administering to a subject having a CD38-expressing disease or condition, any of the bispecific molecules provided herein. Also provided herein is a method of treatment comprising administering to a subject having a disease or disorder associated with CD38, any of the bispecific molecules provided herein.
[0050] Also provided herein is use of any of the anti-CD38 antibodies or antigenbinding fragments thereof provided herein, any of the anti-CD38 CARs provided herein, any of the cells provided herein, any of the compositions provided herein, any of the immunoconjugates provided herein, or any of the bispecific molecules provided herein in the treatment of a subject having a CD38-expressing disease or condition. Also provided herein is use of any of the anti- CD38 antibodies or antigen- binding fragments thereof provided herein, any of the anti-CD38 CARs provided herein, any of the cells provided herein, any of the compositions provided herein, de any of the immunoconjugates provided herein, or any of the bispecific molecules provided herein in the treatment of a subject having a disease or disorder associated with CD38.
[0051] Also provided herein is use of any of the anti-CD38 antibodies or antigenbinding fragments thereof provided herein, any of the anti-CD38 CARs provided herein, any of the cells provided herein, any of the compositions provided herein, any of the immunoconjugates provided herein, or any of the bispecific molecules provided herein in the manufacture of a medicament for the treatment of a subject having a CD38-expressing disease or condition. Also provided herein is use of any of the anti-CD38 antibodies or antigen-binding fragments thereof provided herein, any of the anti-CD38 CARs provided herein, any of the cells provided herein, any of the compositions provided herein, any of the immunoconjugates provided herein, or any of the bispecific molecules provided herein in the manufacture of a medicament for the treatment of a subject having a disease or disorder associated with CD38.
[0052] In some embodiments, the CD38-expressing disease or condition is a cancer or a tumor. In some embodiments, the CD38-expressing disease or condition is a solid tumor.
[0053] In some embodiments, the CD38-expressing disease or condition is a cancer. In some embodiments, the cancer is selected from the group consisting of leukemia, lymphoma, myeloma, glioma, thyroid cancer, lung cancer, colorectal cancer, head and neck cancer, stomach cancer, liver cancer, pancreatic cancer, renal cancer, urothelial cancer, prostate cancer, testicular cancer, breast cancer, cervical cancer, endometrial cancer, ovarian cancer, and melanoma. In some embodiments, the cancer is lung cancer. In some embodiments, the cancer is colorectal cancer. In some embodiments, the cancer is head and neck cancer. In some embodiments, the cancer is ovarian cancer. In some embodiments, the cancer is breast cancer. In some embodiments, the cancer is glioma. In some embodiments, the cancer is melanoma. In some embodiments, the cancer is multiple myeloma.
[0054] In some embodiments, the CD38-expressing disease or condition is an autoimmune disease or disorder. In some embodiments, the disease or disorder associated with CD38 is an autoimmune disease or disorder.
[0055] In some embodiments, the CD38-expressing disease or condition is an autoimmune disease or disorder is selected from the group consisting of systemic lupus erythematosus (SLE), lupus nephritis, CNS lupus, myositis, multiple sclerosis (MS), myasthenia gravis (MG), rheumatoid arthritis (RA), scleroderma, thyroid disease, diabetes, vasculitis, or any combination thereof. In some embodiments, the CD38-expressing disease or condition is systemic lupus erythematosus (SLE). In some embodiments, the CD38-expressing disease or condition is lupus nephritis (LN). In some embodiments, the CD38-expressing disease or condition is CNS lupus. In some embodiments, the CD38-expressing disease or condition is myositis. In some embodiments, the CD38-expressing disease or condition is multiple sclerosis (MS). In some embodiments, the CD38-expressing disease or condition is myasthenia gravis (MG). In some embodiments, the CD38-expressing disease or condition is rheumatoid arthritis (RA). In some embodiments, the CD38-expressing disease or condition is scleroderma. In some embodiments, the CD38-expressing disease or condition is thyroid disease. In some embodiments, the CD38- expressing disease or condition is diabetes. In some embodiments, the CD38-expressing disease or condition is vasculitis.
[0056] In some embodiments, the subject has been treated with, or is a candidate for treatment with, an anti-CD38 antibody. In some embodiments, the subject has been previously treated with an anti-CD38 antibody. In some embodiments, the subject is a candidate for treatment with an anti-CD38 antibody. In some embodiments, the treatment further comprises administering an anti-CD38 antibody to the subject.
[0057] In some embodiments, the cancer is a relapsed/refractory cancer. In some embodiments, the subject has relapsed following treatment with and/or is refractory to a prior line of therapy for a CD38-expressing cancer. In some embodiments, the subject has relapsed following treatment with a prior line of therapy for a CD38-expressing cancer. In some embodiments, the subject is refractory to a prior line of therapy for a CD38-expressing cancer.
[0058] In some embodiments, the autoimmune disease or disorder is a relapsed/refractory autoimmune disease. In some embodiments, the subject has relapsed following treatment with and/or is refractory to a prior line of therapy for an autoimmune disease or disorder. In some embodiments, the subject has relapsed following treatment with a prior line of therapy for an autoimmune disease or disorder. In some embodiments, the subject is refractory to a prior line of therapy for an autoimmune disease or disorder.
[0059] In some embodiments, the prior line of therapy is one, two, three, or four prior lines of therapy. In some embodiments, the prior line of therapy is one prior line of therapy. In some embodiments, the prior line of therapy is two prior lines of therapy. In some embodiments, the prior line of therapy is three prior lines of therapy. In some embodiments, the prior line of therapy is four prior lines of therapy.
[0060] Also provided herein is a method of producing a cell targeting CD38, comprising introducing into the cell any of the nucleic acid molecules provided herein.
[0061] Also provided herein is a method of producing a cell targeting CD38, comprising introducing into the cell any of the vectors provided herein.
[0062] In some embodiments, the method further comprises genetically editing the cell to reduce expression of a target gene. In some embodiments, the target gene comprises CD38. In some embodiments, the method further comprises genetically editing the cell to reduce expression of CD38. In some embodiments, the genetic editing is carried out using a CRISPR/Cas system. In some embodiments, the genetic editing comprises use of a CD38 -targeting guide RNA (gRNA). In some embodiments, the genetic editing comprises use of a ribonucleoprotein complex (RNP) comprising a CD38 -targeting gRNA. In some embodiments, the cell is an immune cell. In some embodiments, the cell is a natural killer (NK) cell or a T cell. In some embodiments, the cell is a NK cell. In some embodiments, the cell is a T cell.
BRIEF DESCRIPTION OF THE DRAWINGS
[0063] The descriptions of the figures below are related to experiments and results that represent non-limiting embodiments of the inventions disclosed herein.
[0064] Figure 1 shows the binding of a non-limiting embodiment of an anti-CD38 scFvs to CD38-negative cells (y-axis) and CD38-positive cells (x axis). Binders selected for further analysis are shown in light gray.
[0065] Figure 2A shows antigen-dependent activation of CD38 KO Jurkat cells expressing non-limiting embodiments of anti-CD38 CARs (as measured by CD69 expression; y- axis) compared to the binding affinity of the anti-CD38 scFvs incorporated by the CARs (x-axis).
[0066] Figures 2B and 2C show the ratio of antigen-dependent activation to antigenindependent (tonic) activation (y axis) and CAR expression (x-axis) as measured in Jurkat cells expressing non-limiting embodiments of CD38-directed CARs. CARs selected for further analysis are shown in the black box (Figure 2A) and indicated by clone number (Figure 2B).
[0067] Figure 3 shows CAR expression as measured in Jurkat cells expressing CARs containing non-limiting embodiments of anti-CD38 scFvs (y-axis) and the epitope bin identified for the non-limiting embodiments of anti-CD38 scFvs (x-axis). Clones selected for further analysis are indicated by clone number.
[0068] Figure 4 shows CAR expression as measured in primary natural killer (NK) cells expressing CARs containing non-limiting embodiments of anti-CD38 scFvs (clones indicated by number on x-axis).
[0069] Figure 5 shows the number of MM. IS target cells 24 hours after CD38 CAR-expressing primary NK cells from two different donors were rechallenged with target cells (clones indicated by number on x-axis).
DETAILED DESCRIPTION
[0070] Among the provided embodiments are antibodies, recombinant receptors (e.g., CARs), and compositions, bispecific molecules, immunoconjugates, engineered cells, methods and uses related to the same, including those targeting or directed to CD38 and CD38- expressing cells and diseases. CD38 is known to play a role in hematological malignancies, including chronic lymphocytic leukemia (CLL) and multiple myeloma (MM), and is also implicated in chronic inflammatory diseases, such as rheumatoid arthritis (RA) and asthma. In CLL, CD38 expression correlates with a more aggressive disease course; patients having greater than 30% CD38-positive B cells require more rigorous treatment regimens and exhibit shorter survival times. CD38 is expressed at uniformly high levels on MM cells, and the U.S. Food and Drug Administration (FDA) approved anti-CD38 antibody daratumumab has been observed to directly kill myeloma cells and deplete CD38+ immunosuppressive cells (e.g., myeloid derived suppressor cells and Tregs). Still, patient responses with daratumumab are variable, and most eventually develop progressive disease (PD) when treated with daratumumab as a monotherapy. Possible causes for daratumumab resistance include CD38 downregulation by myeloma cells and the elimination of CD38+ NK cells, which are thought to mediate antibody-dependent cellular cytotoxicity (ADCC). See Konen et al., Cells (2020) 9(1):52.
[0071] CD38 is also implicated in the progression of solid tumors, including melanoma, glioma, esophageal, cervical, and lung cancers, where it is hypothesized to play an immunosuppressive role. In particular, CD38 catalyzes the first step towards adenosine formation from nicotinamide adenine dinucleotide (NAD). NAD present within the tumor microenvironment (TME) is catalyzed into adenosine diphosphate ribose (ADPR), from which adenosine monophosphate (AMP) can be generated. AMP is in turn converted into adenosine, which is thought to promote immunosuppression - including by inhibiting cytotoxic T cells, NK cells, and dendritic cells - via signaling through adenosine receptor 2A (A2A) or 2B (A2B). Konen et al., Cells (2020) 9(1 ):52.
[0072] More recently, CD38 has been identified as a potential therapeutic target for autoimmune diseases. To this end, CD38 is expressed by long-lived plasma cells, which produce the autoantibodies characteristic of most systemic autoimmune diseases (Benfaremo and Gabrielli, Cells (2020) 9(1):77). Daratumumab, an anti-CD38 antibody, improved clinical manifestations of patients with refractory SLE and eliminated antibody-producing plasma cells (Ostendorf et al., N Engl J Med (2020) 383:1149-55). CD38 may also mediate autoimmune diseases by regulating cell recruitment, cytokine and chemokine release, cell activation, phagocytosis, and antigen presentation during inflammation (Ye et al., Autoimmunity Rev (2023) 22(4): 103289). Thus, the anti-CD38 antibodies and CARs and cells expressing the same as provided herein may be effective to treat any autoimmune disease in which autoantibodyproducing plasma cells are implicated.
[0073] Among the provided embodiments are approaches useful in the treatment of such diseases and conditions and/or for targeting such cell types, including anti-CD38 antibodies, CD38-binding receptors (e.g., CARs), nucleic acids encoding the same, and compositions comprising the same. The receptors generally can contain antibodies (including antigen-binding antibody fragments, such as heavy chain variable (VH) regions, single domain antibody fragments and single chain fragments, including scFvs) specific for CD38. Also provided are cells, such as engineered or recombinant cells expressing such CD38-binding receptors, e.g., anti-CD38 CARs and/or containing nucleic acids encoding such receptors, and compositions and articles of manufacture and therapeutic doses containing such cells. [0074] It is contemplated that any of the engineered cells incorporating a CD38- binding molecule as provided herein can be used in combination with an anti-CD38 antibody (e.g., daratumumab). Without wishing to be bound by theory, it may be desirable in such instances to genetically edit (e.g., knock out) the engineered cells to reduce expression of CD38. It is contemplated that editing of CD38 in engineered immune cells may reduce or avoid binding of an anti-CD38 antibody to the engineered immune cells, thereby reducing the “sink” of the antibody due to binding of non-target (e.g., cancer) cells. It is further contemplated that genetically editing CD38 in engineered NK cells may reduce undesired elimination of the engineered NK cells, thereby preserving the ability of engineered NK cells to mediate ADCC and potentially potentiating the effects of an anti-CD38 antibody.
[0075] Also provided are methods of determining CD38 expression, including using anti-CD38 antibodies or antigen-binding fragments thereof provided herein to assess expression of CD38 in a cell or tissue. Also provided are methods of making and using (such as in the treatment or amelioration of CD38-expressing diseases and conditions) cells (e.g., engineered cells) expressing or containing the recombinant CD38-binding receptors compositions containing such cells.
[0076] Adoptive cell therapies (including those involving the administration of cells expressing chimeric receptors specific for a disease or disorder of interest, such as chimeric antigen receptors (CARs) and/or other recombinant antigen receptors) can be effective in the treatment of cancer and other diseases and disorders. In certain contexts, available approaches to adoptive cell therapy may not always be entirely satisfactory. For example, CD38- binding molecules having the ability to recognize and bind to a target, e.g., target antigen such as CD38, to traffic, localize to and successfully enter appropriate sites within the subject, tumors, and environments thereof, to become activated, expand, to exert various effector functions, including cytotoxic killing and secretion of various factors such as cytokines, to persist, to exhibit certain phenotypic states to provide robust responses following re-exposure to CD38, and avoid or reduce exhaustion and/or differentiation into a suppressive state, are needed.
[0077] Also, in some contexts, certain recombinant receptors can exhibit antigenindependent activation (also known as “tonic signaling”), which can lead to undesirable effects, such as increased exhaustion of immune cells that express the recombinant receptor. In some aspects, such effects may limit the immune cell's activity, effect or potency. In some cases, during engineering and ex vivo expansion of the cells for recombinant receptor expression, the cells may exhibit phenotypes indicative of exhaustion, due to tonic signaling through the recombinant receptor. Thus, improved CD38-binding molecules that bind to CD38, exhibit low levels of antigen-independent activation, and/or exhibit a high ratio of antigen-dependent activation to antigen-independent activation are needed. CD38-binding molecules, and related CARs and cells, that meet such needs are provided herein. [0078] Unless defined otherwise, all terms of art, notations and other technical and scientific terms or terminology used herein are intended to have the same meaning as is commonly understood by one of ordinary skill in the art to which the claimed subject matter pertains. In some cases, terms with commonly understood meanings are defined herein for clarity and/or for ready reference, and the inclusion of such definitions herein should not necessarily be construed to represent a substantial difference over what is generally understood in the art.
[0079] All publications, including patent documents, scientific articles and databases, referred to in this application are incorporated by reference in their entirety for all purposes to the same extent as if each individual publication were individually incorporated by reference. If a definition set forth herein is contrary to or otherwise inconsistent with a definition set forth in the patents, applications, published applications and other publications that are herein incorporated by reference, the definition set forth herein prevails over the definition that is incorporated herein by reference.
[0080] The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described.
I. CD38 Binding Molecules
[0081] Provided in some aspects are CD38 binding molecules, such as CD38- binding polypeptides. Such binding molecules include antibodies (including antigen-binding fragments thereof) that specifically bind to CD38 proteins, such as human CD38 protein. Also among the binding molecules are polypeptides containing such antibodies, including single chain cell surface proteins, e.g., recombinant receptors such as CARs containing such antibodies.
A. CD38 Antibodies
[0082] Provided are anti-CD38 antibodies, including functional antibody fragments. In some embodiments, the antibodies include those that are multi-domain antibodies, such as those containing Vn and Vi. domains. In some embodiments, the antibodies include a variable heavy chain and a variable light chain, such as scFvs. The antibodies include antibodies that specifically bind to CD38, e.g., human CD38. Among the provided anti-CD38 antibodies are human antibodies. Also provided are molecules containing such antibodies, e.g., single-chain proteins, fusion proteins, and/or recombinant receptors such as chimeric receptors, including CARs.
[0083] The term “antibody” herein is used in the broadest sense and includes polyclonal and monoclonal antibodies, including intact antibodies and functional (antigenbinding) antibody fragments, including fragment antigen binding (Fab) fragments, Ftab'h fragments, Fab' fragments, Fv fragments, recombinant IgG (rlgG) fragments, variable heavy chain (VH) regions capable of specifically binding the antigen, single chain antibody fragments, including single chain variable fragments (scFv), and single domain antibody (e.g., sdAb, sdFv, nanobody) fragments. The term encompasses genetically engineered and/or otherwise modified forms of immunoglobulins, such as intrabodies, peptibodies, chimeric antibodies, fully human antibodies, humanized antibodies, and heteroconjugate antibodies, multispecific, e.g., bispecific, antibodies, diabodies, triabodies, and tetrabodies, tandem di-scFv, tandem tri-scFv. Unless otherwise stated, the term “antibody” should be understood to encompass functional antibody fragments thereof. The term also encompasses intact or full-length antibodies, including antibodies of any class or sub-class, including IgG and sub-classes thereof, IgM, IgE, IgA, and IgD.
[0084] The terms “complementarity determining region,” and “CDR,” synonymous with “hypervariable region” or “HVR,” are known in the art to refer to non-contiguous sequences of amino acids within antibody variable regions, which confer antigen specificity and/or binding affinity. In general, there are three CDRs in each heavy chain variable region (CDR-H1, CDR-H2, CDR-H3) and three CDRs in each light chain variable region (CDR-L1, CDR-L2, CDR-L3). “Framework regions” and “FR” are known in the art to refer to the non-CDR portions of the variable regions of the heavy and light chains. In general, there are four FRs in each full-length heavy chain variable region (FR-H1, FR-H2, FR-H3, and FR-H4), and four FRs in each full- length light chain variable region (FR-L1, FR-L2, FR-L3, and FR-L4).
[0085] The precise amino acid sequence boundaries of a given CDR or FR can be readily determined using any of a number of well-known schemes, including the Kabat numbering scheme (Sequences of Proteins of Immunological Interest, 1987 and 1991, NTH, Bethesda, MD), the Chothia numbering scheme (Chothia & Lesk, 1987, J. Mol. Biol. 196:901- 917; Chothia et al., 1989, Nature 342:878-883), the Contact numbering scheme (MacCallum et al., J. Mol. Biol. 262:732-745 (1996), “Antibody-antigen interactions: Contact analysis and binding site topography,” J. Mol. Biol. 262, 732-745), the AbM numbering scheme (Martin et al., Proc. Natl. Acad. Sci., 86:9268-9272; 1989), the IMGT numbering scheme (the international ImMunoGeneTics information system; Lefranc et al, Dev. Comp. Immunol. 29:185-203; 2005), and the Aho numbering scheme (Honegger and Pluckthun, J. Mol. Biol. 309(3):657-670; 2001).
[0086] The boundaries of a given CDR or FR may vary depending on the scheme used for identification. For example, the Kabat scheme is based structural alignments, while the Chothia scheme is based on structural information. Numbering for both the Kabat and Chothia schemes is based upon the most common antibody region sequence lengths, with insertions accommodated by insertion letters, for example, “30a,” and deletions appearing in some antibodies. The two schemes place certain insertions and deletions (“indels”) at different positions, resulting in differential numbering. The Contact scheme is based on analysis of complex crystal structures and is similar in many respects to the Chothia numbering scheme.
[0087] Table 1, below, lists position boundaries of CDR-L1, CDR-L2, CDR-L3 and CDR-H1, CDR-H2, CDR-H3 as identified by Kabat, Chothia, and Contact schemes, respectively. For CDR-H1, residue numbering is listed using both the Kabat and Chothia numbering schemes. FRs are located between CDRs, for example, with FR-L1 located between CDR-L1 and CDR-L2, and so forth. It is noted that because the shown Kabat numbering scheme places insertions at H35A and H35B, the end of the Chothia CDR-H1 loop when numbered using the shown Kabat numbering convention varies between H32 and H34, depending on the length of the loop.
Figure imgf000030_0001
! Kabat et al., "Sequences of Proteins of Immunological Interest," ( 1991 ) 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD.
2 Al-Lazikani et al., J. Mol. Biol. (1997) 273(4):927-48.
[0088] Thus, unless otherwise specified, a “CDR” or “complementary determining region,” or individual specified CDRs (e.g., “CDR-H1, CDR-H2), of a given antibody or region thereof, such as a variable region thereof, should be understood to encompass a (or the specific) complementary determining region as defined by any of the aforementioned schemes. For example, where it is stated that a particular CDR (e.g., a CDR-H3) contains the amino acid sequence of a corresponding CDR in a given Vnor VL amino acid sequence, it is understood that such a CDR has a sequence of the corresponding CDR (e.g., CDR-H3) within the variable region, as defined by any of the aforementioned schemes. In some embodiments, specific CDR sequences are specified.
[0089] Likewise, unless otherwise specified, a FR or individual specified FR(s) (e.g., FR-H1, FR-H2), of a given antibody or region thereof, such as a variable region thereof, should be understood to encompass a (or the specific) framework region as defined by any of the known schemes. In some instances, the scheme for identification of a particular CDR, FR, or FRs or CDRs is specified, such as the CDR as defined by the IMGT, Kabat, Chothia, or Contact method. In other cases, the particular amino acid sequence of a CDR or FR is given.
[0090] The term “variable region” or “variable domain” refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen. The variable domains of the heavy chain and light chain (Vn nd VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three CDRs. A single Vuor VLdomain may be sufficient to confer antigenbinding specificity.
[0091] Among the provided antibodies are antibody fragments. An “antibody fragment” refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds. Examples of antibody fragments include but are not limited to Fv, Fab, Fab', Fab'-SH, F(ab')2; diabodies; linear antibodies; variable heavy chain (VH) regions, single-chain antibody molecules such as scFvs and single-domain VH single antibodies; and multispecific antibodies formed from antibody fragments. In particular embodiments, the antibodies are single-chain antibody fragments comprising a variable heavy chain region and a variable light chain region, such as scFvs.
[0092] In some embodiments, the antibodies are recombinantly-produced fragments, such as fragments comprising arrangements that do not occur naturally, such as those with two or more antibody regions or chains joined by synthetic linkers, e.g., peptide linkers, and/or that are may not be produced by enzyme digestion of a naturally-occurring intact antibody. In some aspects, the antibody fragments are scFvs.
[0093] Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody. In certain embodiments, a single-domain antibody is a human or humanized singledomain antibody.
[0094] A “humanized” antibody is an antibody in which all or substantially all CDR amino acid residues are derived from non-human CDRs and all or substantially all FR amino acid residues are derived from human FRs. A humanized antibody optionally may include at least a portion of an antibody constant region derived from a human antibody. A “humanized form” of a non-human antibody, refers to a variant of the non-human antibody that has undergone humanization, typically to reduce immunogenicity to humans, while retaining the specificity and affinity of the parental non-human antibody. In some embodiments, some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g., the antibody from which the CDR residues are derived), e.g., to restore or improve antibody specificity or affinity.
[0095] Among the provided anti-CD38 antibodies are human antibodies. A “human antibody” is an antibody with an amino acid sequence corresponding to that of an antibody produced by a human or a human cell, or non-human source that utilizes human antibody repertoires or other human antibody-encoding sequences, including human antibody libraries. The term excludes humanized forms of non-human antibodies comprising non-human antigen-binding regions, such as those in which all or substantially all CDRs are non-human. The term includes antigen-binding fragments of human antibodies. [0096] Among the provided antibodies are monoclonal antibodies, including monoclonal antibody fragments. The term “monoclonal antibody” as used herein refers to an antibody obtained from or within a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical, except for possible variants containing naturally occurring mutations or arising during production of a monoclonal antibody preparation, such variants generally being present in minor amounts. In contrast to polyclonal antibody preparations, which typically include different antibodies directed against different epitopes, each monoclonal antibody of a monoclonal antibody preparation is directed against a single epitope on an antigen. The term is not to be construed as requiring production of the antibody by any particular method. A monoclonal antibody may be made by a variety of techniques, including but not limited to generation from a hybridoma, recombinant DNA methods, phage-display and other antibody display methods.
[0097] The terms “polypeptide” and “protein” are used interchangeably to refer to a polymer of amino acid residues and are not limited to a minimum length. Polypeptides, including the provided antibodies and antibody chains and other peptides, e.g., linkers and CD38-binding peptides, may include amino acid residues including natural and/or non-natural amino acid residues. The terms also include post-expression modifications of the polypeptide, for example, glycosylation, sialylation, acetylation, phosphorylation, and the like. In some aspects, the polypeptides may contain modifications with respect to a native or natural sequence, as long as the protein maintains the desired activity. i. Non-Limiting Features
[0098] In some embodiments, an anti-CD38 antibody or antigen-binding fragment thereof comprises a heavy chain variable region (VH) and a light chain variable region (VL). In some embodiments, the VH comprises a CDR1, a CDR2, and a CDR3; and the VL comprises a CDR1, a CDR2, and a CDR3. In some embodiments, the VH comprises a CDR1, a CDR2, and a CDR3 as set forth in any VH sequence provided herein, and the VL comprises a CDR1, a CDR2, and a CDR3 as set forth in any VL sequence provided herein. In some embodiments, the anti- CD38 antibody comprises a VH having at least about 80%, 85%, 90%, 95%, 97% (or more)sequence identity to any VH sequence provided herein, and a VL having at least about 80%, 85%, 90%, 95%, 97% (or more (sequence identity to any VL sequence provided herein. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof comprises a VH having any VH sequence provided herein, and a VL having any VL sequence provided herein. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof comprises a linker between the VH and VL region. In some embodiments, the linker comprises the sequence set forth in SEQ ID NO: 1 or SEQ ID NO:3. In some embodiments, the linker comprises the sequence set forth in SEQ ID NO:1. In some embodiments, the linker comprises the sequence set forth in SEQ ID NO:3. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof is an scFv. In some embodiments, the scFv comprises any scFv sequence provided herein.
[0099] In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof comprises: (i) a heavy chain variable region (VH) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:25, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:23; and a light chain variable region (VL) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:27, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:28, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:29; (ii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:30, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:31, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:32; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 35; (iii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:36, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:37, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:38; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:40; (iv) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:41, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:42; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:43, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:45; (v) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:46, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 48; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:49, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:50, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:51; (vi) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:52, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:53; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:54; (vii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:55, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:56, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:57; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:58, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:59, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:60; (viii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:61, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:62; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:63; (ix) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:66; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:68; (x) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:69; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:70, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:71; (xi) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:72, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:73, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:74; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:75, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:76, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:77; or (xii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:78, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:79, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:80; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 81. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:78, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:79, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:80; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:81. . In some embodiments, the anti-CD38 antibody or antigen- binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:66; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:68. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:69; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:70, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:7I.
[00100] In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof comprises: (i) a heavy chain variable region (VH) comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:82, and a light chain variable region (VL) comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:83; (ii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:84, and a VL comprising the CDR 1 , the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 85; (iii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:86, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:87; (iv) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 88, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:89; (v) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:90, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:91; (vi) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:92, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:93; (vii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:94, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:95; (viii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:96, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:97; (ix) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:98, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:99; (x) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 100, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 101; (xi) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 102, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 103; or (xii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 104, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 105. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 100, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 101.
[00101] In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a heavy chain variable region (VH) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:25, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:23; and a light chain variable region (VL) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:27, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 28, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:29. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a heavy chain variable region (VH) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 121, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 122, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 123; and a light chain variable region (VL) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 124, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 125, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:29. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:82. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:83. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:82, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:83. In some embodiments, the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:82, and the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:83. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO: 82, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 83. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 108.
[00102] In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:30, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:31, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:32; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:35. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 126, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 127, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 128; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 129, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 120, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:35. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:84. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:85. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:84, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:85. In some embodiments, the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:84, and the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:85. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:84, and the VL comprises the amino acid sequence set forth in SEQ ID NO:85. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 109.
[00103] In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:36, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:37, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:38; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:40. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 131, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 132, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 133; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 134, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:40. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:86. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:87. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:86, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:87. In some embodiments, the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:86, and the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:87. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:86, and the VL comprises the amino acid sequence set forth in SEQ ID NO:87. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 1 10.
[00104] In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:41, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:42; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:43, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:45. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 135, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 136, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 137; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 138, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 139, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:45. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:88. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 89. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:88, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:89. In some embodiments, the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:88, and the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:89. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:88, and the VL comprises the amino acid sequence set forth in SEQ ID NO:89. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 111.
[00105] In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:46, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:48; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:49, a CDR2 comprising the amino acid sequence set forth in SEQ ID NQ:50, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:51. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 140, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 141, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 142; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 143, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 144, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:51. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:90. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:91In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:90, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:91. In some embodiments, the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:90, and the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:91. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:90, and the VL comprises the amino acid sequence set forth in SEQ ID NO:91. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 112. [00106] In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:52, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:53; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:54. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 135, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 145, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 146; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 147, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 148, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:54. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:92. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:93. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:92, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:93. In some embodiments, the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:92, and the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:93. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:92, and the VL comprises the amino acid sequence set forth in SEQ ID NO:93. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 113.
[00107] In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:55, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:56, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:57; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:58, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:59, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:60. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 149, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 150, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 151; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 152, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 153, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:60. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:94. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:95. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:94, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:95. In some embodiments, the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:94, and the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:95. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:94, and the VL comprises the amino acid sequence set forth in SEQ ID NO:95. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 114 .
[00108] In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:61, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:62; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:63. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 154, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 155, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 156; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 157, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 158, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:63. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:96. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:97. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:96, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:97. In some embodiments, the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:96, and the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:97. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:96, and the VL comprises the amino acid sequence set forth in SEQ ID NO:97. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 115.
[00109] In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:66; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:68. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 159, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 160, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 161; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 162, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 130, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:68. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:98. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:99. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:98, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:99. In some embodiments, the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:98, and the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO:99. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO:98, and the VL comprises the amino acid sequence set forth in SEQ ID NO:99. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 116.
[00110] In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:69; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:70, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:71. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 159, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 163, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 164; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 165, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 166, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:71. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 100. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 101. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 100, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 101. In some embodiments, the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more)sequence identity to the amino acid sequence set forth in SEQ ID NO: 100, and the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more)sequence identity to the amino acid sequence set forth in SEQ ID NO: 101. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO: 100, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 101. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 117.
[00111] In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:72, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:73, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:74; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:75, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:76, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:77. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 167, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 168, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 169; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 170, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 171, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:77. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 102. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 103. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 102, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 103. In some embodiments, the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or morejsequence identity to the amino acid sequence set forth in SEQ ID NO: 102, and the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more) sequence identity to the amino acid sequence set forth in SEQ ID NO: 103. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO: 102, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 103. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 118.
[00112] In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:78, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:79, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:80; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:81. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 172, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 173, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 174; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 162, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO: 130, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:81. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 104. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 105. In some embodiments, the anti- CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 104, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 105. In some embodiments, the VH comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more)sequence identity to the amino acid sequence set forth in SEQ ID NO: 104, and the VL comprises an amino acid sequence that has at least about 80%, 85%, 90%, 95%, 97% (or more)sequence identity to the amino acid sequence set forth in SEQ ID NO: 105. In some embodiments, the VH comprises the amino acid sequence set forth in SEQ ID NO: 104, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 105. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises the amino acid sequence set forth in SEQ ID NO: 119.
[00113] In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence as set forth in any one of SEQ ID NOS: 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197,
199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 228, 230, 232, 233,
235, 237, 239, 241 , 243, 245, 247, 248, 250, 252, 254, 256, 258, 260, 262, 264, 266, 268, 270,
272, 274, 276, 278, 280, 282, 284, 285, 286, 288, 290, 292, 294, 296, 297, 299, 301, 303, 305,
307, 309, 311, 313, 314, 316, 318, 320, 322, 324, and 326. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence as set forth in any one of SEQ ID NOS: 91, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220,
222, 224, 226, 229, 231, 234, 246, 238, 240, 242, 244, 246, 249, 252, 253, 255, 257, 259, 261,
263, 265, 267, 269, 271, 273, 275, 277, 279, 281, 283, 285, 287, 289, 291, 293, 295, 298, 300,
302, 304, 306, 308, 310, 312, 315, 317, 319, 321, 323, and 325. In some embodiments, the anti-
CD38 antibody or antigen binding fragment thereof comprises a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence as set forth in any one of SEQ ID NOS: 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217,
219, 221, 223, 225, 227, 228, 230, 232, 233, 235, 237, 239, 241, 243, 245, 247, 248, 250, 252,
254, 256, 258, 260, 262, 264, 266, 268, 270, 272, 274, 276, 278, 280, 282, 284, 285, 286, 288,
290, 292, 294, 296, 297, 299, 301, 303, 305, 307, 309, 311, 313, 314, 316, 318, 320, 322, 324, and 326; and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence as set forth in any one of SEQ ID NOS: 91, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198,
200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 229, 231, 234, 246, 238,
240, 242, 244, 246, 249, 252, 253, 255, 257, 259, 261, 263, 265, 267, 269, 271, 273, 275, 277,
279, 281, 283, 285, 287, 289, 291, 293, 295, 298, 300, 302, 304, 306, 308, 310, 312, 315, 317,
319, 321, 323, and 325. [00114] In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH sequence having at least 80%, 85%, 90%, 95%, 97% (or more)sequence identity to the sequence set forth in any one of SEQ ID NOS: 175, 177, 179, 181, 183, 185, 187,
189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225,
227, 228, 230, 232, 233, 235, 237, 239, 241, 243, 245, 247, 248, 250, 252, 254, 256, 258, 260,
262, 264, 266, 268, 270, 272, 274, 276, 278, 280, 282, 284, 285, 286, 288, 290, 292, 294, 296,
297, 299, 301, 303, 305, 307, 309, 311, 313, 314, 316, 318, 320, 322, 324, and 326. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VL sequence having at least 80%, 85%, 90%, 95%, 97% (or morejsequence identity to the sequence set forth in any one of SEQ ID NOS: 91, 176, 178, 180, 182, 184, 186, 188, 190, 192, 194, 196,
198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 229, 231, 234, 246,
238, 240, 242, 244, 246, 249, 252, 253, 255, 257, 259, 261, 263, 265, 267, 269, 271, 273, 275,
277, 279, 281, 283, 285, 287, 289, 291, 293, 295, 298, 300, 302, 304, 306, 308, 310, 312, 315,
317, 319, 321, 323, and 325. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises a VH sequence having at least 90% sequence identity to the sequence set forth in any one of SEQ ID NOS: 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197,
199, 201 , 203, 205, 207, 209, 21 1 , 213, 215, 217, 219, 221 , 223, 225, 227, 228, 230, 232, 233,
235, 237, 239, 241, 243, 245, 247, 248, 250, 252, 254, 256, 258, 260, 262, 264, 266, 268, 270,
272, 274, 276, 278, 280, 282, 284, 285, 286, 288, 290, 292, 294, 296, 297, 299, 301, 303, 305,
307, 309, 311, 313, 314, 316, 318, 320, 322, 324, and 326; and a VL sequence having at least 90% sequence identity to the sequence set forth in any one of SEQ ID NOS: 91, 176, 178, 180,
182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218,
220, 222, 224, 226, 229, 231, 234, 246, 238, 240, 242, 244, 246, 249, 252, 253, 255, 257, 259,
261, 263, 265, 267, 269, 271, 273, 275, 277, 279, 281, 283, 285, 287, 289, 291, 293, 295, 298,
300, 302, 304, 306, 308, 310, 312, 315, 317, 319, 321, 323, and 325.
[00115] In some embodiments, the VH and VL comprise sequences having at least 80%, 85%, 90%, 95%, 97% (or more)sequence identity to the sequences set forth in SEQ ID NOS: 175 and 176, respectively; 177 and 178, respectively, 179 and 180, respectively, 181 and 182, respectively; 183 and 184, respectively; 185 and 186, respectively; 187 and 188, respectively; 189 and 190, respectively; 191 and 192, respectively; 193 and 194, respectively; 195 and 196, respectively; 197 and 198, respectively; 199 and 200, respectively; 201 and 202, respectively; 203 and 204, respectively; 205 and 206, respectively; 208 and 208, respectively; 209 and 210, respectively; 211 and 212, respectively; 213 and 214, respectively; 215 and 216, respectively; 217 and 218, respectively; 219 and 220, respectively; 221 and 222, respectively; 223 and 224, respectively; 225 and 226, respectively; 227 and 224, respectively; 228 and 229, respectively; 230 and 231, respectively; 232 and 222, respectively; 233 and 234, respectively; 235 and 236, respectively; 237 and 238, respectively; 239 and 240, respectively; 241 and 242, respectively; 243 and 244, respectively; 245 and 246, respectively; 247 and 222, respectively; 248 and 249, respectively; 250 and 251, respectively; 252 and 253, respectively; 254 and 255, respectively; 256 and 257, respectively; 258 and 259, respectively; 260 and 261, respectively; 262 and 263, respectively; 264 and 265, respectively; 266 and 267, respectively; 268 and 269, respectively; 270 and 271, respectively; 272 and 273, respectively; 274 and 275, respectively; 276 and 277, respectively; 278 and 279, respectively; 280 and 281, respectively; 282 and 283, respectively; 284 and 283, respectively; 285 and 283, respectively; 286 and 287, respectively; 288 and 289, respectively; 290 and 291, respectively; 292 and 293, respectively; 294 and 295, respectively; 296 and 295, respectively; 297 and 298, respectively; 299 and 300, respectively; 301 and 302, respectively; 303 and 304, respectively; 305 and 306, respectively; 307 and 308, respectively; 309 and 310, respectively; 311 and 312, respectively; 313 and 222, respectively; 314 and 315, respectively; 316 and 317, respectively; 318 and 319, respectively; 320 and 321, respectively; 322 and 323, respectively; 324 and 325, respectively; or 326 and 91, respectively.
[00116] In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises the VH sequence set forth in any one of SEQ ID NOS: 175, 177, 179, 181, 183,
185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221,
223, 225, 227, 228, 230, 232, 233, 235, 237, 239, 241 , 243, 245, 247, 248, 250, 252, 254, 256,
258, 260, 262, 264, 266, 268, 270, 272, 274, 276, 278, 280, 282, 284, 285, 286, 288, 290, 292,
294, 296, 297, 299, 301, 303, 305, 307, 309, 311, 313, 314, 316, 318, 320, 322, 324, and 326. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises the VL sequence set forth in any one of SEQ ID NOS: 91, 176, 178, 180, 182, 184, 186, 188, 190,
192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 229,
231, 234, 246, 238, 240, 242, 244, 246, 249, 252, 253, 255, 257, 259, 261, 263, 265, 267, 269,
271, 273, 275, 277, 279, 281, 283, 285, 287, 289, 291, 293, 295, 298, 300, 302, 304, 306, 308,
310, 312, 315, 317, 319, 321, 323, and 325. In some embodiments, the anti-CD38 antibody or antigen binding fragment thereof comprises the VH sequence set forth in any one of SEQ ID NOS: 175, 177, 179, 181, 183, 185, 187, 189, 191, 193, 195, 197, 199, 201, 203, 205, 207, 209, 211, 213, 215, 217, 219, 221, 223, 225, 227, 228, 230, 232, 233, 235, 237, 239, 241, 243, 245,
247, 248, 250, 252, 254, 256, 258, 260, 262, 264, 266, 268, 270, 272, 274, 276, 278, 280, 282,
284, 285, 286, 288, 290, 292, 294, 296, 297, 299, 301, 303, 305, 307, 309, 311, 313, 314, 316,
318, 320, 322, 324, and 326; and the VL sequence set forth in any one of SEQ ID NOS: 91, 176,
178, 180, 182, 184, 186, 188, 190, 192, 194, 196, 198, 200, 202, 204, 206, 208, 210, 212, 214,
216, 218, 220, 222, 224, 226, 229, 231, 234, 246, 238, 240, 242, 244, 246, 249, 252, 253, 255,
257, 259, 261, 263, 265, 267, 269, 271, 273, 275, 277, 279, 281, 283, 285, 287, 289, 291, 293,
295, 298, 300, 302, 304, 306, 308, 310, 312, 315, 317, 319, 321, 323, and 325.
[00117] In some embodiments, the VH and VL comprise the sequences set forth in
SEQ ID NOS: 175 and 176, respectively; 177 and 178, respectively, 179 and 180, respectively, 181 and 182, respectively; 183 and 184, respectively; 185 and 186, respectively; 187 and 188, respectively; 189 and 190, respectively; 191 and 192, respectively; 193 and 194, respectively; 195 and 196, respectively; 197 and 198, respectively; 199 and 200, respectively; 201 and 202, respectively; 203 and 204, respectively; 205 and 206, respectively; 208 and 208, respectively; 209 and 210, respectively; 211 and 212, respectively; 213 and 214, respectively; 215 and 216, respectively; 217 and 218, respectively; 219 and 220, respectively; 221 and 222, respectively; 223 and 224, respectively; 225 and 226, respectively; 227 and 224, respectively; 228 and 229, respectively; 230 and 231, respectively; 232 and 222, respectively; 233 and 234, respectively; 235 and 236, respectively; 237 and 238, respectively; 239 and 240, respectively; 241 and 242, respectively; 243 and 244, respectively; 245 and 246, respectively; 247 and 222, respectively; 248 and 249, respectively; 250 and 251, respectively; 252 and 253, respectively; 254 and 255, respectively; 256 and 257, respectively; 258 and 259, respectively; 260 and 261, respectively; 262 and 263, respectively; 264 and 265, respectively; 266 and 267, respectively; 268 and 269, respectively; 270 and 271, respectively; 272 and 273, respectively; 274 and 275, respectively; 276 and 277, respectively; 278 and 279, respectively; 280 and 281, respectively; 282 and 283, respectively; 284 and 283, respectively; 285 and 283, respectively; 286 and 287, respectively; 288 and 289, respectively; 290 and 291 , respectively; 292 and 293, respectively; 294 and 295, respectively; 296 and 295, respectively; 297 and 298, respectively; 299 and 300, respectively; 301 and 302, respectively; 303 and 304, respectively; 305 and 306, respectively; 307 and 308, respectively; 309 and 310, respectively; 311 and 312, respectively; 313 and 222, respectively; 314 and 315, respectively; 316 and 317, respectively; 318 and 319, respectively; 320 and 321, respectively; 322 and 323, respectively; 324 and 325, respectively; or 326 and 91, respectively.
[00118] In some embodiments, the antibody variable regions are joined by a flexible linker. In some embodiments, the anti-CD38 antibody or antigen-binding fragment thereof is an scFv in which the VH and VL are joined by a linker. In some embodiments, the linker comprises a sequence having at least about 90%, at least about 95%, or at least about 99% sequence identity to the sequence of SEQ ID NO: 1 or SEQ ID NOG. In some embodiments, the linker comprises the sequence of SEQ ID NO: 1 or SEQ ID NO:3. In some embodiments, the linker comprises a sequence having at least about 90%, at least about 95%, or at least about 99% sequence identity to the sequence of SEQ ID NO:3. In some embodiments, the linker comprises the sequence of SEQ ID NO:3. In some embodiments, the linker contains only glycine and serine residues (“a Gly-Ser linker”). In some embodiments, the antibody comprises any Gly-Ser linker known in the art. In some embodiments, the linker comprises a sequence having at least about 90%, at least about 95%, or at least about 99% sequence identity to the sequence of SEQ ID NO: 1. In some embodiments, the linker comprises the sequence of SEQ ID NO: 1.
[00119] In some embodiments, the scFv comprises the sequence of SEQ ID NO: 1. In some embodiments, the antibody is an scFv comprising the sequence of SEQ ID NO: 108, SEQ ID NO: 109, SEQ ID NO: 110, SEQ ID NO: 111, SEQ ID NO: 112, SEQ ID NO: 113, SEQ ID NO: 114, SEQ ID NO: 115, SEQ ID NO: 116, SEQ ID NO: 117, SEQ ID NO:118, or SEQ ID NO: 119. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 108. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 109. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 110. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 111. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 112. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 113. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 114. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 115. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 116. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 117. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 118. In some embodiments, the antibody comprises the sequence of SEQ ID NO: 119.
[00120] In some embodiments, the antibody specifically binds to the CD38 protein. In some embodiments of any of the embodiments herein, CD38 refers to human CD38. In some embodiments, the antibody specifically binds to human CD38, such as an epitope or region of human CD381 , such as human CD38 described in SEQ ID NO:351 (e.g., UniProt P28907), or its allele variant or splice variant. In some embodiments, the antibody specifically binds to the extracellular domain of human CD38 (e.g., residues 43-300 of SEQ ID NO:351). In some embodiments, the antibody binds to non-human CD38, such as monkey, rabbit, rat, mouse, or other CD38species.
[00121] In some embodiments, the antibody can bind to CD38 with at least a certain affinity, such as human CD38, as measured by any of a variety of known methods. In some embodiments, the affinity is represented by the dissociation constant (KD). In some embodiments, affinity is represented by EC 50. In some embodiments, the KD of the antibody to CD38 (such as human CD38) is from or from about 0.1 nM to 500 nM, 0.1 nM to 100 nM, 0.1 nM to 50 nM, 0.1 nM to 10 nM, 0.1 nM to 1 nM, 1 nM to 500 nM, 1 nM to 100 nM, 1 nM to 50 nM, 1 nM to 10 nM, 10 nM to 500 nM, 10 nM to 100 nM, 10 nM to 50 nM, 50 nM to 500 nM, 50 nM to 100 nM or 100 nM to 500 nM. In certain embodiments, the KD of the antibody to CD38 (such as human CD38) is below or about below or below or about below or below: lOOnM, 60nM, 50nM, 40nM, 30nM , 25nM, 20nM, 19nM, 18nM, 17nM, 16nM, 15nM, 14nM, 13nM, 12nM, l lnM, lOnM, 9nM, 8nM, 7nM, 6nM, 5nM, 4nM, 3nM, 2nM or InM.
[00122] In some embodiments, the characteristics or characteristics of the provided antibody are described in relation to the characteristics observed by another antibody (for example, a reference antibody). In some embodiments, the reference antibody is a human anti- CD38 antibody. In some aspects, the reference antibody is a human antibody named “A2” (see, for example, PCT/US2016/026545 and PCT/IB2018/058642). [00123] For example, in some embodiments, the reference antibody has a VH region containing the sequence set forth in SEQ ID NO: 106, or contains CDR1, CDR2, and CDR3 within this sequence; and a VL region containing the sequence set forth in SEQ ID NO: 107, or contains CDR1, CDR2, and CDR3 within this sequence. For example, the reference antibody may include the CDR-H1 sequence of SEQ ID NO:340, the CDR-H2 sequence of SEQ ID NO:341, the CDR-H3 sequence of SEQ ID NO:342, the CDR-L1 sequence of SEQ ID NO:343, the CDR- L2 sequence of SEQ ID NO:344, and the CDR-L3 sequence of SEQ ID NO:345. IN some embodiments, the reference antibody includes the VH sequence of SEQ ID NO: 106 and the VL sequence of SEQ ID NO: 107. In some embodiments, the reference antibody is a scFv comprising the amino acid sequence set forth in SEQ ID NO: 120. In some embodiments, the reference antibody is formatted as a CAR. In some embodiments, the CAR comprises the sequence of SEQ ID NO:339.
[00124] In some embodiments, the antibody has an affinity, e.g., KD, about the same as or lower than that of the corresponding form of the reference antibody, e.g., no more than about 1.5-fold or no more than about 2-fold greater, no more than 3-fold greater, and/or no more than 10-fold greater, than the KD of the corresponding form of the reference antibody. In some embodiments, the antibody has an affinity, e.g., KD, that is greater than or greater than about 5- fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80-fold, 90-fold, 100-fold, 150- fold, 200-fold, 250-fold or greater than the KD of the corresponding form of the reference antibody.
[00125] In some embodiments, the antibodies display a binding preference for CD38- expressing cells as compared to CD38-negative cells. In some embodiments, the binding preference is observed where a significantly greater degree of binding is measured to the CD38-expressing, as compared to the non-expressing, cells. In some embodiments, the fold change in degree of binding detected, for example, as measured by mean fluorescence intensity (MFI) in a flow cytometry-based assay and/or KD or ECso, to the CD38-expressing cells as compared to the non-CD38-expressing cells, is at least at or about 1.5, 2, 3, 4, 5, 6, 8, 10, 12, 14, 16, 18, or 20 or more, and/or is about as great, about the same, at least as great or at least about as great, or greater, than the fold change observed for the corresponding form of the reference antibody. In some cases, the total degree of observed binding to CD38 or to the CD38-expressing cells is approximately the same, at least as great, or greater than that observed for the corresponding form of the reference antibody.
[00126] In some embodiments, the antibodies display a higher ratio of antigendependent activation to antigen-independent activation as compared to the corresponding form of the reference antibody. For example, in some embodiments, the antibodies display a ratio of antigen-dependent activation to antigen-independent activation that is at least about 2-fold, 4-fold, 6-fold, 8-fold, or 10-fold higher than that observed for the corresponding form of the reference antibody. In some embodiments, the antibodies display a ratio of antigen-dependent activation to antigen-independent activation that is about 2-fold, 4-fold, 6-fold, 8-fold, or 10-fold higher than that observed for the corresponding form of the reference antibody. ii. Imrnunoconi ligates
[00127] In some embodiments, the antibody is or is part of an immunoconjugate comprising a therapeutic agent. In some embodiments, the antibody is conjugated to one or more heterologous molecule(s), such as, but not limited to, a cytotoxic agent or a detection agent, (e.g., a radioisotope). Cytotoxic agents include, but are not limited to, radioactive isotopes (e.g., At211, 1131, 1125, Y90, Rel86, Rel88, Sml53, Bi212, P32, Pb212 and radioactive isotopes of Lu); chemotherapeutic agents (e.g., methotrexate, adriamicin, vinca alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin or other intercalating agents); growth inhibitory agents; enzymes and fragments thereof such as nucleolytic enzymes; antibiotics; toxins such as small molecule toxins or enzymatically active toxins. In some embodiments, the antibody is conjugated to one or more cytotoxic agents, such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes. Among the immunoconjugates are antibody-drug conjugates (ADCs), in which an antibody is conjugated to one or more drugs, including but not limited to an anthracycline such as daunomycin or doxorubicin, methotrexate; vindesine; or a taxane such as docetaxel, paclitaxel, larotaxel, tesetaxel, and ortataxeL
[00128] Also among the immunoconjugates are those in which the antibody is conjugated to an enzymatically active toxin or fragment thereof, including but not limited to diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP- S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes.
[00129] Also among the immunoconjugates are those in which the antibody is conjugated to a radioactive atom to form a radioconjugate. Non-limiting examples of radioactive isotopes include At 211 , 1131 , 1125 , Y90, Re186, Re188, SmM, Bi212, P32, Pb212 and radioactive isotopes of Lu.
[00130] Conjugates of an antibody and a therapeutic agent (e.g., a cytotoxic agent) may be made using any of a number of known protein coupling agents, e.g., linkers. The linker may be a "cleavable linker" facilitating release of a cytotoxic drug in the cell, such as acid-labile linkers, peptidase-sensitive linkers, photolahile linkers, dimethyl linkers, and disulfide-containing linkers. iii. Multispccific Molecules
[00131] In certain embodiments, the CD38-binding molecules, e.g., antibodies or polypeptides such as chimeric receptors containing the same, are multispecific. Among the multispecific binding molecules are multispecific antibodies, including, e.g. bispecific. Multispecific binding partners, e.g., antibodies, have binding specificities for at least two different sites, which may be in the same or different antigens. In certain embodiments, one of the binding specificities is for CD38 and the other is for another antigen. In certain embodiments, bispecific antibodies may bind to two different epitopes of CD38. Bispecific antibodies may also be used to localize cytotoxic agents to cells which express CD38. Bispecific antibodies can be prepared as full-length antibodies or antibody fragments. Among the multispecific antibodies are multispecific single-chain antibodies, e.g., diabodies, triabodies, and tetrabodies, tandem di-scFvs, and tandem tri-scFvs. Also provided are multispecific chimeric receptors, such as multispecific CARs, containing the antibodies. Also provided are multispecific cells containing the antibodies or polypeptides including the same, such as cells containing a cell surface protein including the anti-CD38 antibody and an additional cell surface protein, such as an additional chimeric receptor, which binds to a different antigen or a different epitope on CD38.
[00132] Non-limiting examples of additional antigens include B cell specific antigens, other tumor-specific antigens, such as antigens expressed specifically on or associated with B cell leukemia, lymphoma, B cell chronic lymphocytic leukemia (CLL), AML, acute myeloid leukemia (AML), acute lymphocytic leukemia (ALL), Burkett's Lymphoma, mantle cell lymphoma (MCL), non-small cell lung cancer (NSCLC), neuroblastoma, renal cell carcinoma, colon cancer, colorectal cancer, breast cancer, epithelial squamous cell cancer, melanoma, myeloma, stomach cancer, brain cancer, lung cancer, pancreatic cancer, cervical cancer, ovarian cancer, liver cancer, bladder cancer, prostate cancer, testicular cancer, thyroid cancer, uterine cancer, adrenal cancer and/or head and neck cancer, and antigens expressed on NK cells or T cells. Non-limiting examples of antigens include BCMA, CD3, CD4, CD8, CD19, CD20, CD21, CD22, CD23, CD33, CD39, CD40, CD40L, CD138, CD203, FcRH5, GPRC5D, and SLAMF7.
[00133] In some embodiments, the example of an additional antigen is a T cell antigen. In some embodiments, the additional example of an antigen is CD3. In some embodiments, the example of an additional antigen is a NK cell antigen. In some embodiments, the additional example of an antigen is CD16, NKp30, NKp46, or NKG2D, or a combination thereof. In some embodiments, the additional example of an antigen is CD 16. In some embodiments, the additional example of an antigen is NKp30. In some embodiments, the additional example of an antigen is NKp46. In some embodiments, the additional example of an antigen is NKG2D.
[00134] In some embodiments, a bispecific molecule comprises any of the anti-CD38 antibodies or antigen-binding fragments thereof provided herein and a second moiety that has a binding specificity for an immune cell. In some embodiments, the immune cell is a NK cell. In some embodiments, the second moiety binds to CD 16, NKp30, NKp46, or NKG2D. In some embodiments, the second moiety binds to CD16. In some embodiments, the second moiety binds to NKp30. In some embodiments, the second moiety binds to NKp46. In some embodiments, the second moiety binds to NKG2D. In some embodiments, the immune cell is a T cell. In some embodiments, the second moiety binds to CD3. iv. Variants
[00135] In several embodiments, there are provided amino acid sequences that correspond to any of the nucleic acids disclosed herein (and/or included in the accompanying sequence listing), while accounting for degeneracy of the nucleic acid code. Furthermore, those sequences (whether nucleic acid or amino acid) that vary from those expressly disclosed herein (and/or included in the accompanying sequence listing), but have functional similarity or equivalency are also contemplated within the scope of the present disclosure. The foregoing includes mutants, truncations, substitutions, codon optimization, or other types of modifications.
[00136] In accordance with some embodiments described herein, any of the sequences may be used, or a truncated or mutated form of any of the sequences disclosed herein (and/or included in the accompanying sequence listing) may be used and in any combination. Sequences provided for herein that include an identifier, such as a tag or other detectable sequence (e.g., a Flag tag) are also provided for herein with the absence of such a tag or other detectable sequence (e.g., excluding the Flag tag from the listed sequence). A Sequence Listing in electronic format is submitted herewith. Some of the sequences provided in the Sequence Listing may be designated as Artificial Sequences by virtue of being non-naturally occurring fragments or portions of other sequences, including naturally occurring sequences. Some of the sequences provided in the Sequence Listing may be designated as Artificial Sequences by virtue of being combinations of sequences from different origins, such as humanized antibody sequences.
[00137] In certain embodiments, the antibodies include one or more amino acid variations, e.g., substitutions, deletions, insertions, and/or mutations, compared to the sequence of an antibody described herein. Non-limiting examples of variants include those designed to improve the binding affinity and/or other biological properties of the antibody. Amino acid sequence variants of an antibody may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the antibody, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of residues within the amino acid sequences of the antibody. Any combination of deletion, insertion, and substitution can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., antigen-binding. [00138] In certain embodiments, the antibodies include one or more amino acid substitutions, e.g., as compared to an antibody sequence described herein and/or compared to a sequence of a natural repertoire, e.g., human repertoire. Sites of interest for substitutional mutagenesis include the CDRs and FRs. Amino acid substitutions may be introduced into an antibody of interest and the products screened for a desired activity, e.g., retained/improved antigen binding, decreased immunogenicity, improved half-life, and/or improved effector function, such as the ability to promote antibody-dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC). Insertional variants of the antibody molecule include the fusion to the N- or C-terminus of the antibody to an enzyme or a polypeptide which increases the serum half-life of the antibody.
B. Recombinant Receptors
[00139] Among the provided binding molecules, e.g., CD38-binding molecules, are recombinant receptors, such as those that include one of the provided antibodies. The receptors include antigen receptors and other chimeric receptors that specifically bind to CD38, such as receptors containing the provided anti-CD38 antibodies, e.g., antibody fragments. Among the antigen receptors are chimeric antigen receptors (CARs). Also provided are cells expressing the recombinant receptors and uses thereof in treatment of diseases and conditions associated with CD38 expression. The chimeric receptors, such as CARs, generally include an extracellular antigen-binding domain that includes, is, or is comprised within, one of the provided anti-CD38 antibodies. Thus, the chimeric receptors, e.g., CARs, typically include in their extracellular portion one or more CD38-binding molecules, such as one or more antigen-binding fragment as described herein. In some embodiments, the CAR includes a heavy chain variable region (VH) and a light chain variable region (VL) region of the antibody, e.g., an scFv antibody fragment.
[00140] The antigen-binding domain may be or comprise any of those as described herein, e.g., in Section LA. For example, in some embodiments, the antigen-binding domain is an scFv comprising a VH and a VL joined by a linker (e.g., SEQ ID NO:1 or SEQ ID NO:3). In some embodiments, the antigen-binding domain is an scFv comprising the linker set forth in SEQ ID NO:1. In some embodiments, the antigen-binding domain comprises the amino acid sequence set forth in any one of SEQ ID NOS: 108-119. In some embodiments, the antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 108. In some embodiments, the antigenbinding domain comprises the amino acid sequence of SEQ ID NO: 109. In some embodiments, the antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 110. In some embodiments, the antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 111. In some embodiments, the antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 112. In some embodiments, the antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 113. In some embodiments, the antigen- binding domain comprises the amino acid sequence of SEQ ID NO: 114. In some embodiments, the antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 115. In some embodiments, the antigenbinding domain comprises the amino acid sequence of SEQ ID NO: 116. In some embodiments, the antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 117. In some embodiments, the antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 118. In some embodiments, the antigen-binding domain comprises the amino acid sequence of SEQ ID NO: 119.
[00141] The antigen-binding domain generally is linked to an intracellular signaling domain comprising intracellular signaling components, such as signaling components that mimic activation through an antigen receptor complex, such as a TCR complex, in the case of a CAR. In some embodiments, the antigen-binding domain is linked to an intracellular signaling domain by a transmembrane domain. Thus, in some embodiments, the CD38-binding molecule (e.g., antibody) is linked to a transmembrane and intracellular signaling domain.
[00142] In some embodiments, the transmembrane domain is fused to the extracellular domain. The transmembrane domain in some embodiments is derived either from a natural or from a synthetic source. Where the source is natural, the domain in some aspects is derived from any membrane-bound or transmembrane protein. Transmembrane regions include those derived from (e.g., comprising at least the transmembrane region(s) of) CD3, CD4, CD5, CD8, CD9, CD 16, CD22, CD28, CD33, CD37, CD45, CD64, CD80, CD86, CD134, CD137, CD 154, or a combination thereof. Alternatively the transmembrane domain in some embodiments is synthetic.
[00143] In several embodiments, the transmembrane domain comprises at least a portion of CD8, a transmembrane glycoprotein normally expressed on both T cells and NK cells. In several embodiments, the transmembrane domain comprises CD8alpha (CD8a). In several embodiments, the transmembrane domain comprises a CD8 (e.g., CD8a) hinge and a CD8 (e.g., CD8a) transmembrane region.
[00144] In several embodiments, the transmembrane domain comprises a hinge, e.g. a CD8a hinge. In several embodiments, the sequence encoding the CD8a hinge is truncated or modified. In some embodiments, the CD8a hinge is encoded by a nucleic acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the nucleic acid sequence of SEQ ID NO:5. In several embodiments, the CD8a hinge comprises the nucleic acid sequence of SEQ ID NO:5. In several embodiments, the CD8a hinge is truncated or modified. In some embodiments, the CD8a hinge has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the amino acid sequence of SEQ ID NO:6. In several embodiments, the hinge of CD8a comprises the amino acid sequence of SEQ ID NO:6.
[00145] In several embodiments, the transmembrane domain comprises a CD8a transmembrane region. In several embodiments, the CD8a transmembrane region is truncated or modified. In some embodiments, the CD8a transmembrane region is encoded by a nucleic acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the sequence of SEQ ID NO:7. In several embodiments, the CD8a transmembrane region is encoded by a nucleic acid sequence of SEQ ID NO:7. In several embodiments, the CD8a transmembrane region is truncated or modified. In some embodiments, the CD8a transmembrane region has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the sequence of SEQ ID NO:8. In several embodiments, the CD8a transmembrane region comprises the amino acid sequence of SEQ ID NO:8.
[00146] Thus, in several embodiments, the CD8 transmembrane domain is truncated or modified and is encoded by a nucleic acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the sequence of SEQ ID NO:9. In several embodiments, the CD8 transmembrane domain is encoded by the nucleic acid sequence of SEQ ID NO:9. In some embodiments, the CD8 transmembrane domain is truncated or modified and comprises an amino acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the sequence of SEQ ID NO: 10. In several embodiments, the transmembrane domain comprises the amino acid sequence of SEQ ID NO: 10.
[00147] In some embodiments, the transmembrane domain comprises a CD28 transmembrane domain or a fragment thereof. In several embodiments, the CD28 transmembrane domain is truncated or modified. In some embodiments, the CD28 transmembrane domain has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the amino acid sequence of SEQ ID NOT E In several embodiments, the CD28 transmembrane domain comprises the amino acid sequence of SEQ ID NO: 11.
[00148] The receptor, e.g., the CAR, generally includes an intracellular signaling domain comprising intracellular signaling components. In some embodiments, the receptor includes an intracellular component of a TCR complex, such as a TCR CD3 chain that mediates T-cell activation and cytotoxicity, e.g., CD3 zeta chain. Thus, in some aspects, the antigenbinding portion is linked to one or more cell signaling modules. In some embodiments, upon ligation of the CAR or other chimeric receptor, the cytoplasmic domain or intracellular signaling domain of the receptor activates at least one of the normal effector functions or responses of the immune cell, e.g., NK cell engineered to express the CAR. For example, in some contexts, the CAR induces a function of an immune cell (e.g., NK cell) such as cytolytic activity and/or secretion of cytokines or other factors. In some embodiments, a truncated portion of an intracellular signaling domain of an antigen receptor component or costimulatory molecule is used in place of an intact immunostimulatory chain, for example, if it transduces the effector function signal. In some embodiments, the intracellular signaling domain includes the cytoplasmic sequences of the T cell receptor (TCR), and in some aspects, also those of co- receptors that in the natural context act in concert with such receptors to initiate signal transduction following antigen receptor engagement.
[00149] In the context of a natural TCR, full activation generally requires not only signaling through the TCR, but also a costimulatory signal. Thus, in some embodiments, to promote full activation, a component for generating secondary or co- stimulatory signal is also included in the receptor. T cell activation is in some aspects described as being mediated by two classes of cytoplasmic signaling sequences: those that initiate antigen-dependent primary activation through the TCR (primary cytoplasmic signaling sequences), and those that act in an antigen independent manner to provide a secondary or co-stimulatory signal (secondary cytoplasmic signaling sequences). In some aspects, the receptor includes one or both of such signaling components.
[00150] In some aspects, the receptor includes a primary cytoplasmic signaling sequence that regulates primary activation of the TCR complex. Primary cytoplasmic signaling sequences that act in a stimulatory manner may contain signaling motifs which are known as immunoreceptor tyrosine -based activation motifs or ITAMs. Examples of ITAM containing primary cytoplasmic signaling sequences include those derived from TCR zeta, FcR gamma, FcR beta, CD3 gamma, CD3 delta, CD3 epsilon, CDS, CD22, CD79a, CD79b, and CD66d. In some embodiments, cytoplasmic signaling molecule(s) in the CAR contain(s) a cytoplasmic signaling domain, portion thereof, or sequence derived from CD3zeta.
[00151] For example, immune cells engineered according to several embodiments disclosed herein may comprise at least one subunit of the CD3 T cell receptor complex (or a fragment thereof). In several embodiments, the signaling domain comprises the CD3 zeta subunit. In several embodiments, the CD3zeta can be truncated or modified. In some embodiments, the CD3zeta is encoded by a nucleic acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the nucleic acid sequence of SEQ ID NO: 17. In several embodiments, the CD3zeta is encoded by the nucleic acid sequence of SEQ ID NO: 17. In several embodiments, the CD3zeta is truncated or modified. In some embodiments, the CD3zeta comprises an amino acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 18. In several embodiments, the CD3zeta comprises the amino acid sequence of SEQ ID NO: 18.
[00152] In some embodiments, the intracellular signaling domain comprises a costimulatory signaling region, such as an intracellular signaling region of CD28, 4- IBB, 0X40, DAP10, ICOS, or any combination thereof. In some aspects, the same receptor includes both a CD3zeta and a costimulatory signaling region. Thus, in some embodiments, the intracellular signaling domain of the recombinant receptor, such as CAR, comprises a CD3zeta intracellular domain and a costimulatory signaling region. [00153] In several embodiments, the intracellular signaling domain comprises an intracellular signaling region of 0X40. In several embodiments, the 0X40 intracellular signaling region is encoded by a nucleic acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the nucleic acid sequence of SEQ ID NO: 13. In several embodiments, the 0X40 intracellular signaling region is encoded by the nucleic acid sequence of SEQ ID NO: 13. In several embodiments, the 0X40 intracellular signaling region comprises an amino acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 14. In several embodiments, the 0X40 intracellular signaling region comprises the amino acid sequence of SEQ ID NO: 14. In several embodiments, 0X40 is used as the sole intracellular signaling component in the construct, however, in several embodiments, 0X40 can be used with one or more other components. For example, combinations of 0X40 and CD3zeta are used in some embodiments. In some embodiments, the intracellular signaling domain comprises an 0X40 costimulatory signaling region linked to CD3zeta. In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:327, SEQ ID NO:328, SEQ ID NO: 329, SEQ ID NO:330, SEQ ID NO:331, SEQ ID NO:332, SEQ ID NO:333, SEQ ID NO:334, SEQ ID NO:335, SEQ ID NO:336, SEQ ID NO:337, or SEQ ID NO:338. In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:327. In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:328. In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO: 329. In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:330. In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:33L In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:332. In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:333. In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:334. In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:335. In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO: 336. In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:337. In some embodiments, the chimeric receptor (e.g., CAR) comprises the amino acid sequence set forth in SEQ ID NO:338.
[00154] By way of further example, combinations of CD28, 0X40, 4-1BB and/or CD3zeta are used in some embodiments.
[00155] In several embodiments, the intracellular signaling domain comprises an intracellular signaling region of 4-1BB. In several embodiments, the 4-1BB intracellular signaling region is encoded by a nucleic acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the nucleic acid sequence of SEQ ID NO: 15. In several embodiments, the 4-1BB intracellular signaling region is encoded by the nucleic acid sequence of SEQ ID NO: 15. In several embodiments, the 4-1BB intracellular signaling region comprises an amino acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 16. In several embodiments, the 4-1BB intracellular signaling region comprises the amino acid sequence of SEQ ID NO: 16. In several embodiments, 4-1BB is used as the sole intracellular signaling component in the construct, however, in several embodiments, 4- IBB can be used with one or more other components. For example, combinations of 4-1BB and CD3zeta are used in some embodiments. In some embodiments, the intracellular signaling domain comprises a 4- IBB costimulatory signaling region linked to CD3zeta. By way of further example, combinations of CD28, 0X40, 4-1BB and/or CD3zeta are used in some embodiments.
[00156] In several embodiments, the intracellular signaling domain comprises an intracellular signaling region of CD28. In several embodiments, the CD28 intracellular signaling region comprises an amino acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 12. In several embodiments, the CD28 intracellular signaling region comprises the amino acid sequence of SEQ ID NO: 12. In several embodiments, CD28 is used as the sole intracellular signaling component in the construct, however, in several embodiments, CD28 can be used with one or more other components. For example, combinations of CD28 and CD3zeta are used in some embodiments. In some embodiments, the intracellular signaling domain comprises a CD28 costimulatory signaling region linked to CD3zeta. By way of further example, combinations of CD28, 0X40, 4-1BB and/or CD3zeta are used in some embodiments.
[00157] In any of the provided embodiments, the nucleic acid encoding the chimeric receptor, or a portion thereof, is codon-optimized. In some embodiments, the polynucleotides are optimized, or contain certain features designed for optimization, such as for codon usage, to reduce RNA heterogeneity and/or to modify, e.g., increase or render more consistent among cell product lots, expression, such as surface expression, of the encoded receptor. In some embodiments, polynucleotides, encoding chimeric receptors, are modified as compared to a reference polynucleotide, such as to remove cryptic or hidden splice sites, to reduce RNA heterogeneity. In some embodiments, polynucleotides, encoding chimeric receptors, are codon optimized, such as for expression in a mammalian, e.g., human, cell such as in a human T cell. In some aspects, the modified polynucleotides result in in improved, e.g., increased or more uniform or more consistent level of, expression, e.g., surface expression, when expressed in a cell.
C. Engineered Cells [00158] Also provided are methods, nucleic acids, compositions, and kits, for producing the genetically engineered cells. In some aspects, the genetic engineering involves introduction of a nucleic acid encoding the genetically engineered component or other component for introduction into the cell, such as a component encoding a gene-disrupting protein or nucleic acid. Among additional nucleic acids, e.g., genes for introduction are those to improve the efficacy of therapy, such as by promoting viability and/or function of transferred cells; genes to provide a genetic marker for selection and/or evaluation of the cells, such as to assess in vivo survival or localization; genes to improve safety, for example, by making the cell susceptible to negative selection in vivo. i. Vectors and Methods for Genetic Engineering
[00159] Also provided are methods, polynucleotides, compositions, and kits, for expressing the binding molecules (e.g., anti-CD38 binding molecules), including recombinant receptors (e.g., CARs) comprising the binding molecules, and for producing the genetically engineered cells expressing such binding molecules. In some embodiments, one or more binding molecules, including recombinant receptors (e.g., CARs) can be genetically engineered into cells or a plurality of cells. The genetic engineering generally involves introduction of a nucleic acid encoding the recombinant or engineered component into the cell, such as by retroviral transduction, transfection, or transformation.
[00160] Also provided are polynucleotides encoding the antibodies and chimeric antigen receptors and/or portions, e.g., chains, thereof. Among the provided polynucleotides are those encoding the anti-CD38 chimeric antigen receptors (e.g., antigen-binding fragment) described herein. Also provided are polynucleotides encoding one or more antibodies and/or portions thereof, e.g., those encoding one or more of the anti-CD38 antibodies (e.g., antigenbinding fragment) described herein and/or other antibodies and/or portions thereof, e.g., antibodies and/or portions thereof that binds other target antigens. The polynucleotides may include those encompassing natural and/or non-naturally occurring nucleotides and bases, e.g., including those with backbone modifications. The terms “nucleic acid molecule”, “nucleic acid” and “polynucleotide” may be used interchangeably, and refer to a polymer of nucleotides. Such polymers of nucleotides may contain natural and/or non-natural nucleotides, and include, but are not limited to, DNA, RNA, and PNA. “Nucleic acid sequence” refers to the linear sequence of nucleotides that comprise the nucleic acid molecule or polynucleotide. Also provided are polynucleotides that have been optimized for codon usage.
[00161] Also provided are vectors containing the polynucleotides, such as any of the polynucleotides described herein, and cells containing the vectors, e.g., for producing the antibodies or antigen-binding fragments thereof. In some embodiments, the vector is a viral vector. In some embodiments, the vector is a retroviral vector. In some embodiments, the vector is a lentiviral vector. Also provided are methods for producing the antibodies or antigen-binding fragments thereof. The nucleic acid may encode an amino acid sequence comprising the VL region and/or an amino acid sequence comprising the VH region of the antibody (e.g., the light and/or heavy chains of the antibody). The nucleic acid may encode one or more amino acid sequence comprising the VL region and/or an amino acid sequence comprising the VH region of the antibody (e.g., the light and/or heavy chains of the antibody). In a further embodiment, one or more vectors (e.g., expression vectors) comprising such polynucleotides are provided. In a further embodiment, a host cell comprising such polynucleotides is provided. In another such embodiment, a host cell comprises (e.g., has been transformed with) (1) a vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL region of the antibody and an amino acid sequence comprising the VH region of the antibody, or (2) a first vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL region of the antibody and a second vector comprising a nucleic acid that encodes an amino acid sequence comprising the VH region of the antibody. In some embodiments, a host cell comprises (e.g., has been transformed with) one or more vectors comprising one or more nucleic acid that encodes one or more an amino acid sequence comprising one or more antibodies and/or portions thereof, e.g., antigen-binding fragments thereof. In some embodiments, one or more such host cells are provided. In some embodiments, a composition containing one or more such host cells are provided. In some embodiments, the one or more host cells can express different antibodies, or the same antibody. In some embodiments, each of the host cells can express more than one antibody.
[00162] Also provided are methods of making the anti-CD38 chimeric antigen receptors. For recombinant production of the chimeric receptors, a nucleic acid sequence encoding a chimeric receptor antibody, e.g., as described herein, may be isolated and inserted into one or more vectors for further cloning and/or expression in a host cell. Such nucleic acid sequences may be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody). In some embodiments, a method of making the anti-CD38 chimeric antigen receptor is provided, wherein the method comprises culturing a host cell comprising a nucleic acid sequence encoding the antibody, as provided above, under conditions suitable for expression of the receptor.
[00163] In some aspects, for production of isolated or secreted polypeptides, in addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for antibody-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been modified to mimic or approximate those in human cells, resulting in the production of an antibody with a partially or fully human glycosylation pattern. Non-limiting examples of eukaryotic cells that may be used to express polypeptides, including isolated or secreted polypeptides, include, but are not limited to, COS cells, including COS-7 cells; 293 cells, including 293-6E cells; CHO cells, including CHO-S, DG44. Lecl3 CHO cells, and FUT8 CHO cells; PER.C6® cells; and NSO cells. In some embodiments, the antibody heavy chains and/or light chains (e.g., VH region and/or VL region) may be expressed in yeast. See, e.g., US Publication No. US 2006/0270045 Al. In some embodiments, a particular eukaryotic host cell is selected based on its ability to make desired post-translational modifications to the heavy chains and/or light chains (e.g., VH region and/or VL region). For example, in some embodiments, CHO cells produce polypeptides that have a higher level of sialylation than the same polypeptide produced in 293 cells.
[00164] In particular examples immune cells, such as human immune cells are used to express the provided polypeptides encoding chimeric antigen receptors. In some examples, the immune cells are NK cells including primary NK cells, T cells (e.g., CD4+ and/or CD8+ immune cells) including primary T cells, or any combination thereof.
[00165] In some embodiments, gene transfer is accomplished by first stimulating the cell, such as by combining it with a stimulus that induces a response such as proliferation, survival, and/or activation, e.g., as measured by expression of a cytokine or activation marker, followed by transduction of the activated cells, and expansion in culture to numbers sufficient for clinical applications.
[00166] In some aspects, the cells further are engineered to promote expression of cytokines or other factors. Various methods for the introduction of genetically engineered components, e.g., antigen receptors, e.g., CARs, are well known and may be used with the provided methods and compositions. Exemplary methods include those for transfer of polynucleotides encoding the receptors, including via viral, e.g., retroviral or lentiviral, transduction, transposons, and electroporation.
[00167] In some embodiments, recombinant polynucleotides are transferred into immune cells (e.g., NK or T cells) using recombinant infectious virus particles, such as, e.g., vectors derived from simian virus 40 (SV40), adenoviruses, adeno-associated virus (AAV). In some embodiments, recombinant polynucleotides are transferred into immune cells using recombinant lentiviral vectors or retroviral vectors, such as gamma-retroviral vectors. In some embodiments, the retroviral vector has a long terminal repeat sequence (LTR), e.g., a retroviral vector derived from the Moloney murine leukemia virus (MoMLV), myeloproliferative sarcoma virus (MPSV), murine embryonic stem cell virus (MESV), murine stem cell virus (MSCV), spleen focus forming virus (SFFV), or human immunodeficiency virus type 1 (HIV-1). Most retroviral vectors are derived from murine retroviruses. In some embodiments, the retroviruses include those derived from any avian or mammalian cell source. The retroviruses typically are amphotropic, meaning that they are capable of infecting host cells of several species, including humans. In one embodiment, the gene to be expressed replaces the retroviral gag, pol and/or env sequences. A number of illustrative retroviral systems have been described. Methods of lentiviral transduction are known and described in the art.
[00168] In some embodiments, recombinant polynucleotides are transferred into immune cells (e.g., NK or T cells) via electroporation. In some embodiments, recombinant polynucleotides are transferred into immune cells (e.g., NK or T cells) via transposition. Other methods of introducing and expressing genetic material in immune cells include calcium phosphate transfection, protoplast fusion, cationic liposome-mediated transfection; tungsten particle-facilitated microparticle bombardment, and strontium phosphate DNA co-precipitation.
[00169] Among additional polynucleotides, e.g., genes for introduction are those to improve the efficacy of therapy, such as by promoting viability and/or function of transferred cells; genes to provide a genetic marker for selection and/or evaluation of the cells, such as to assess in vivo survival or localization; genes to improve safety, for example, by making the cell susceptible to negative selection in vivo.
[00170] In some embodiments, one or more binding molecules, including antibodies and/or recombinant receptors (e.g., CARs), can be genetically engineered to be expressed in cells or a plurality of cells. In some embodiments, a first recombinant receptor and a second binding molecule, e.g., recombinant receptor, are encoded by the same or separate nucleic acid molecules. In some embodiments, additional binding molecules are engineered to be expressed in cells or a plurality of cells.
[00171] In some cases, the polynucleotide containing nucleic acid sequences encoding the CD38-binding receptor, e.g., chimeric antigen receptor (CAR), contains a signal sequence that encodes a signal peptide. In some aspects, the signal sequence may encode a signal peptide derived from a native polypeptide. In other aspects, the signal sequence may encode a heterologous or non-native signal peptide. In some aspects, a non-limiting example of a signal peptide comprises a CD8 alpha (CD8a) signal peptide set forth in SEQ ID NO:4.
[00172] In some embodiments the vector or construct can contain promoter and/or enhancer or regulatory elements to regulate expression of the encoded recombinant receptor. In some examples the promoter and/or enhancer or regulatory elements can be condition-dependent promoters, enhancers, and/or regulatory elements. In some examples these elements drive expression of the transgene.
[00173] In some embodiments, the vector or construct can contain a single promoter that drives the expression of one or more nucleic acid molecules. In some embodiments, such nucleic acid molecules, e.g., transcripts, can be multicistronic (bicistronic or tricistronic). For example, in some embodiments, transcription units can be engineered as a bicistronic unit containing an IRES (internal ribosome entry site), which allows coexpression of gene products (e.g. encoding a chimeric receptor and membrane- bound interleukin- 15) by a message from a single promoter. Alternatively, in some cases, a single promoter may direct expression of an RNA that contains, in a single open reading frame (ORF), two or three genes (e.g. encoding a chimeric receptor and membrane -bound interleukin- 15) separated from one another by sequences encoding a self-cleavage peptide (e.g., 2A cleavage sequences) or a protease recognition site. The ORF thus encodes a single polypeptide, which, either during (in the case of T2A) or after translation, is cleaved into the individual proteins. In some cases, the peptide, such as T2A, can cause the ribosome to skip (ribosome skipping) synthesis of a peptide bond at the C-terminus of a 2A element, leading to separation between the end of the 2A sequence and the next peptide downstream. Many 2A elements are known. Examples of 2A sequences that can be used in the methods and polynucleotides disclosed herein, without limitation, 2A sequences from the foot- and-mouth disease virus (F2A), equine rhinitis A virus (E2A), Thosea asigna virus (T2A, e.g. SEQ ID NO:20, encoded by SEQ ID NO: 19), and porcine teschovirus- 1 (P2A). In some embodiments, the one or more different or separate promoters drive the expression of a nucleic acid molecule encoding a binding molecule, e.g., recombinant receptor and a nucleic acid encoding membrane-bound interleukin- 15.
[00174] Any of the binding molecules, e.g., antibodies and/or recombinant receptors provided herein, e.g., CD38-binding molecules and/or the additional recombinant receptors, can be encoded by polynucleotides containing one or more nucleic acid molecules encoding the receptors, in any combinations or arrangements. For example, one, two, three or more polynucleotides can encode one, two, three or more different receptors or domains. In some embodiments, one vector or construct contains nucleic acid molecules encoding one or more binding molecules, e.g., antibody and/or recombinant receptor, and a separate vector or construct contains nucleic acid molecules encoding an additional binding molecule, e.g., antibody and/or recombinant receptor.
[00175] Also provided are compositions containing one or more of the nucleic acid molecules, vectors or constructs, such as any described above. In some embodiments, the nucleic acid molecules, vectors, constructs or compositions can be used to engineer immune cells, such as NK or T cells, to express any of the binding molecules, e.g., antibody or recombinant receptor, and/or the additional binding molecules. ii. Genetic Editing
[00176] As discussed below in Section I.A.iv, a variety of cell types can be engineered to express a CD38-binding molecule or a chimeric receptor incorporating the same. Further, as elaborated on in more detail below, and as described in the Examples, genetic modifications (e.g., to CD38) can be made to these cells in order to enhance one or more aspects of their efficacy (e.g., cytotoxicity) and/or persistence (e.g., in vivo life span). As discussed herein, in several embodiments immune cells (e.g., NK cells) are used for treatment of a subject having a disease or condition. In several embodiments provided for herein, gene editing of the immune cell (e.g., NK cell) can advantageously impart to the edited immune cell the ability to resist and/or overcome various inhibitory signals that are generated in the tumor microenvironment (TME). It is known that tumors generate a variety of signaling molecules that are intended to reduce the anti-tumor effects of immune cells. As discussed in more detail below, in several embodiments, gene editing of the immune cell limits this TME suppressive effect on the immune cells, or any edited/engineered immune cell provided for herein. As discussed below, gene editing is employed to reduce or knockout expression of target proteins, for example by disrupting the underlying gene encoding the protein.
[00177] In several embodiments, genetic editing of a gene as disclosed herein, is accomplished through targeted introduction of DNA breakage, and a subsequent DNA repair mechanism. In several embodiments, double strand breaks of DNA are repaired by non- homologous end joining (NHEJ), wherein enzymes are used to directly join the DNA ends to one another to repair the break. NHEJ is an error-prone process. In general, in the absence of a repair template, the NHEJ process re-ligates the ends of the cleaved DNA strands, which frequently results in nucleotide deletions and insertions at the cleavage site. In several embodiments, however, double strand breaks (DSB) are repaired by homology directed repair (HDR), which is advantageously more accurate, thereby allowing sequence specific breaks and repair. HDR uses a homologous sequence as a template for regeneration of missing DNA sequences at the break point, such as a vector with the desired genetic elements (e.g., an insertion element to disrupt the coding sequence of a gene) within a sequence that is homologous to the flanking sequences of a double strand break. This will result in the desired change (e.g., insertion) being inserted at the site of the DSB. The HDR pathway can occur by way of the canonical HDR pathway or the alternative HDR pathway. Unless otherwise indicated, the term “HDR” or “homology-directed repair” as used herein encompasses both canonical HDR and alternative HDR.
[00178] Canonical HDR or “canonical homology-directed repair” or cHDR,” are used interchangeably, and refers to the process of repairing DNA damage using a homologous nucleic acid (e.g., an endogenous homologous sequence, such as a sister chromatid; or an exogenous nucleic acid, such as a donor template). Canonical HDR typically acts when there has been a significant resection at the DSB, forming at least one single-stranded portion of DNA. In a normal cell, canonical HDR typically involves a series of steps such as recognition of the break, stabilization of the break, resection, stabilization of single-stranded DNA, formation of a DNA crossover intermediate, resolution of the crossover intermediate, and ligation. The canonical HDR process requires RAD51 and BRCA2, and the homologous nucleic acid, e.g., repair template, is typically double-stranded. In canonical HDR, a double-stranded polynucleotide, e.g., a doublestranded repair template, is introduced, which comprises a sequence that is homologous to the targeting sequence, and which will either be directly integrated into the targeting sequence or will be used as a template to insert the sequence, or a portion the sequence, of the repair template into the target gene. After resection at the break, repair can progress by different pathways, e.g., by the double Holliday junction model (also referred to as the double strand break repair, or DSBR, pathway), or by the synthesis-dependent strand annealing (SDSA) pathway.
[00179] In the double Holliday junction model, strand invasion occurs by the two single stranded overhangs of the targeting sequence to the homologous sequences in the doublestranded polynucleotide, e.g., double stranded donor template, which results in the formation of an intermediate with two Holliday junctions. The junctions migrate as new DNA is synthesized from the ends of the invading strand to fill the gap resulting from the resection. The end of the newly synthesized DNA is ligated to the resected end, and the junctions are resolved, resulting in the insertion at the targeting sequence, or a portion of the targeting sequence that includes the gene variant. Crossover with the polynucleotide, e.g., repair template, may occur upon resolution of the junctions.
[00180] In the SDSA pathway, only one single stranded overhang invades the polynucleotide, e.g., donor template, and new DNA is synthesized from the end of the invading strand to fill the gap resulting from resection. The newly synthesized DNA then anneals to the remaining single stranded overhang, new DNA is synthesized to fill in the gap, and the strands are ligated to produce the modified DNA duplex.
[00181] Alternative HDR, or “alternative homology-directed repair,” or “alternative HDR,” are used interchangeably, and refers, in some embodiments, to the process of repairing DNA damage using a homologous nucleic acid (e.g., an endogenous homologous sequence, such as a sister chromatid; or an exogenous nucleic acid, such as a repair template). Alternative HDR is distinct from canonical HDR in that the process utilizes different pathways from canonical HDR, and can be inhibited by the canonical HDR mediators, RAD51 and BRCA2. Moreover, alternative HDR is also distinguished by the involvement of a single-stranded or nicked homologous nucleic acid template, e.g., repair template, whereas canonical HDR generally involves a double-stranded homologous template. In the alternative HDR pathway, a single strand template polynucleotide, e.g., repair template, is introduced. A nick, single strand break, or DSB at the cleavage site, for altering a desired target site, e.g., a gene variant in a target gene, is mediated by a nuclease molecule, and resection at the break occurs to reveal single stranded overhangs. Incorporation of the sequence of the template polynucleotide, e.g., repair template, to alter the target site of the DNA typically occurs by the SDSA pathway, as described herein. In some embodiments, HDR is carried out by introducing, into a cell, one or more agent(s) capable of inducing a DSB, and a repair template, e.g., a single-stranded oligonucleotide. The introducing can be carried out by any suitable delivery. The conditions under which HDR is allowed to occur can be any conditions suitable for carrying out HDR in a cell.
[00182] In several embodiments, gene editing is accomplished by one or more of a variety of engineered nucleases. In several embodiments, restriction enzymes are used, particularly when double strand breaks are desired at multiple regions. In several embodiments, a bioengineered nuclease is used. Depending on the embodiment, one or more of a Zinc Finger Nuclease (ZFN), transcription-activator like effector nuclease (TALEN), meganuclease and/or clustered regularly interspaced short palindromic repeats (CRISPR/Cas9) system are used to specifically edit the genes encoding one or more of the TCR subunits.
[00183] Meganucleases are characterized by their capacity to recognize and cut large DNA sequences (from 14 to 40 base pairs). In several embodiments, a meganuclease from the LAGLIDADG family is used, and is subjected to mutagenesis and screening to generate a meganuclease variant that recognizes a unique sequence(s), such as a specific site in a gene (e.g., CD38). In several embodiments, two or more meganucleases, or functions fragments thereof, are fused to create a hybrid enzyme that recognize a desired target sequence within a gene (e.g., CD38).
[00184] In contrast to meganucleases, ZFNs and TALEN function based on a nonspecific DNA cutting catalytic domain which is linked to specific DNA sequence recognizing peptides such as zinc fingers or transcription activator-like effectors (TALEs). Advantageously, the ZFNs and TALENs thus allow sequence-independent cleavage of DNA, with a high degree of sequence-specificity in target recognition. Zinc finger motifs naturally function in transcription factors to recognize specific DNA sequences for transcription. The C-terminal part of each finger is responsible for the specific recognition of the DNA sequence. While the sequences recognized by ZFNs are relatively short, (e.g., ~3 base pairs), in several embodiments, combinations of 2, 3, 4, 5, 6, 7, 8, 9, 10 or more zinc fingers whose recognition sites have been characterized are used, thereby allowing targeting of specific sequences, such as a portion of CD38. The combined ZFNs are then fused with the catalytic domain(s) of an endonuclease, such as FokI (optionally a FokI heterodimer), in order to induce a targeted DNA break.
[00185] Transcription activator-like effector nucleases (TALENs) are specific DNA- binding proteins that feature an array of 33 or 34-amino acid repeats. Like ZFNs, TALENs are a fusion of a DNA cutting domain of a nuclease to TALE domains, which allow for sequenceindependent introduction of double stranded DNA breaks with highly precise target site recognition. TALENs can create double strand breaks at the target site that can be repaired by error-prone non-homologous end-joining (NHEJ), resulting in gene disruptions through the introduction of small insertions or deletions. Advantageously, TALENs are used in several embodiments, at least in part due to their higher specificity in DNA binding, reduced off-target effects, and ease in construction of the DNA-binding domain.
[00186] CRISPRs (Clustered Regularly Interspaced Short Palindromic Repeats) are genetic elements that bacteria use as protection against viruses. The repeats are short sequences that originate from viral genomes and have been incorporated into the bacterial genome. Cas (CRISPR associated proteins) process these sequences and cut matching viral DNA sequences. By introducing plasmids containing Cas genes and specifically constructed CRISPRs into eukaryotic cells, the eukaryotic genome can be cut at any desired position. n several embodiments, CRISPR is used to manipulate a gene (e.g., CD38).
[00187] Depending on the embodiment and which target gene is to be edited, a Class 1 or Class 2 Cas is used. In several embodiments, a Class 1 Cas is used and the Cas type is selected from the following types: I, IA, IB, IC, ID, IE, IF, IU, III, IIIA, IIIB, IIIC, HID, IV IVA, IVB, and combinations thereof. In several embodiments, the Cas is selected from the group consisting of Cas3, Cas8a, Cas5, Cas8b, Cas8c, CaslOd, Csel, Cse2, Csyl, Csy2, Csy3, GSU0054, CaslO, Csm2, Cmr5, CaslO, Csxl l, CsxlO, Csfl, and combinations thereof. In several embodiments, a Class 2 Cas is used and the Cas type is selected from the following types: n, HA, IIB, IIC, V, VI, and combinations thereof. In several embodiments, the Cas is selected from the group consisting of Cas9, Csn2, Cas4, Casl2a (previously known as Cpfl), C2cl, C2c3, Cas 13a (previously known as C2c2), Cas 13b, Cas 13c, CasX, CasY and combinations thereof. In some embodiments, the Cas is Cas9. In some embodiments, class 2 CasX is used, wherein CasX is capable of forming a complex with a guide nucleic acid and wherein the complex can bind to a target DNA, and wherein the target DNA comprises a non-target strand and a target strand. In some embodiments, class 2 CasY is used, wherein CasY is capable of binding and modifying a target nucleic acid and/or a polypeptide associated with target nucleic acid.
[00188] CD38 plays a role in the maturation cycle of immune cells, and hematological cancers and solid tumors can often present upregulated CD38. Loss of CD38 expression on constituent NK cells allows for greater cytotoxicity due to decreased fratricide (Nagai et al., Blood (2019) 134 (suppl. l):870). Wild-type NK cells self-express CD38, leading to downstream self-targeting effects in wild-type NK cells. For T cells, loss of CD38 expression for constituent T cells leads to increased cytotoxicity.
[00189] Further, it is contemplated that any of the engineered cells provided herein can be used (e.g., administered) in combination with an anti-CD38 antibody (e.g., daratumumab). Without wishing to be bound by theory, decreasing or eliminating expression of CD38 by any of the engineered cells provided herein (e.g., NK and/or T cells) may reduce or avoid targeting of the engineered cells by the anti-CD38 antibody. Thus, editing (e.g., knocking out) CD38 in engineered immune cells may not only increase the amount of the engineered cells that persist in vivo, but loss of the anti-CD38 antibody to the “sink” that is CD38 -expressing engineered cells may be mitigated. Accordingly, editing (e.g., knocking out) CD38 in engineered immune cells may increase the amount of anti-CD38 antibody available to bind to CD38-expressing target cells (e.g., cancer cells). An additional advantage of editing (e.g., knocking out) CD38 in engineered NK cells is the preservation of NK cell-mediated antibody -dependent cellular cytotoxicity (ADCC). ADCC is a mechanism of cell-mediated immune defense against opsonized target cells wherein the Fc-fragment of an antibody binds to CD16a (FcyRIIIa) on immune effectors such as NK cells, triggering their activation and targeted killing. By genetically editing (e.g., knocking out) CD38 in the engineered NK cells, they are more likely to persist in vivo rather than being targeted by an anti-CD38 antibody, and therefore will be present in higher numbers to mediate ADCC, thereby potentiating the effect of the anti-CD38 antibody. Thus, according to several embodiments, gene editing CD38 increases the cytotoxicity, persistence, immune avoidance or otherwise enhances the efficacy of engineered NK, T, or other cell as disclosed herein.
[00190] The expression of any other target protein, or any combination of target proteins, can be decreased or eliminated, such as by disrupting the gene(s) encoding the target protein/ s). In several embodiments, the expression of a target gene is reduced and/or eliminated in any cells provided herein. Target genes can include any gene such as CD38, CISH, Cbl-b, MED12, and TGFbR2. In several embodiments, the expression of CD38 (encoded by CD38) is reduced and/or eliminated in any cells provided herein. In several embodiments, CD38 is disrupted and/or knocked out using one or more of the gene editing methods disclosed herein. In several embodiments, CD38 is disrupted and/or knocked out using a Crispr-Cas mediated approach (e.g., using a Cas9 nuclease) as disclosed elsewhere herein, with the Cas nuclease guided by a CD38-specific guide RNA (gRNA). Methods for Crispr-Cas-based disruption of CD38, as well as examples of CD38 -targeting gRNA sequences are known in the art and include any of those as described in Clara et al., J Immunother Cancer (2022) 10(2):e003804 and Kararoudi et al., Blood (2020) 136(21):2416-27. In other embodiments, transcription activatorlike effector nucleases (TALENs) or zinc finger nucleases (ZFNs) are used.
[00191] In several embodiments, gene editing reduces transcription of CD38 by about 30%, about 40%, about 50%, about 60%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 97%, about 98%, about 99%, or more (including any amount between those listed). In several embodiments, gene editing reduces transcription of CD38 by at least about 30%. In several embodiments, gene editing reduces transcription of CD38 by at least about 40%. In several embodiments, gene editing reduces transcription of CD38 by at least about 50%. In several embodiments, gene editing reduces transcription of CD38 by at least about 60%. In several embodiments, gene editing reduces transcription of CD38 by at least about 70%. In several embodiments, gene editing reduces transcription of CD38 by at least about 80%. In several embodiments, gene editing reduces transcription of CD38 by at least about 90%.
[00192] In several embodiments, gene editing reduces expression of CD38 by about 30%, about 40%, about 50%, about 60%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 97%, about 98%, about 99%, or more (including any amount between those listed). In several embodiments, gene editing reduces expression of CD38 by at least about 30%. In several embodiments, gene editing reduces expression of CD38 by at least about 40%. In several embodiments, gene editing reduces expression of CD38 by at least about 50%. In several embodiments, gene editing reduces expression of CD38 by at least about 60%. In several embodiments, gene editing reduces expression of CD38 by at least about 70%. In several embodiments, gene editing reduces expression of CD38 by at least about 80%. In several embodiments, gene editing reduces expression of CD38 by at least about 90%. iii. Membrane-Bound Interleukin- 15
[00193] In several embodiments, any of immune cells as provided herein are engineered to express a membrane-bound interleukin 15 (mbIL15). In such embodiments, mbIL15 expression on the immune cell (e.g., NK cell) enhances the cytotoxic effects of the engineered cell by enhancing the proliferation and/or longevity of the cells. In some embodiments, the IL15 is expressed from a separate cassette on the construct comprising any one of the CARs disclosed herein. In some embodiments, the IL15 is expressed from the same cassette as any one of the CARs disclosed herein.
[00194] In some embodiments, the chimeric receptor and IL15 are separated by a nucleic acid sequence encoding a cleavage site, for example, a proteolytic cleavage site or a T2A, P2A, E2A, or F2A self-cleaving peptide cleavage site. In some embodiments, the chimeric receptor and IL15 are separated by a T2A sequence (e.g., SEQ ID NO:20, encoded by SEQ ID NO: 19). In some embodiments, the IL15 is a membrane -bound IL15 (mbIL15). In some embodiments, the mbIL15 comprises a native IL15 sequence, such as a human native IL15 sequence (e.g., SEQ ID NO:22, encoded by SEQ ID NO:21), and at least one transmembrane domain (e.g., CD8a). In several embodiments, IL15 is encoded by the nucleic acid sequence of SEQ ID NO: 21. In several embodiments, IL15 can be truncated or modified, such that it is encoded by a nucleic acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the nucleic acid sequence of SEQ ID NO: 21. In several embodiments, the IL15 comprises the amino acid sequence of SEQ ID NO: 22. In several embodiments, the IL15 is truncated or modified, such that it has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequence identity to the amino acid sequence of SEQ ID NO: 22.
[00195] Thus, in some embodiments, any of the CARs as described herein are encoded by the same nucleic acid sequence as a mbIL15. In some embodiments, a nucleic acid sequence encoding the CAR and a nucleic acid sequence encoding the mbIL15 are separated by a T2A-encoding sequence (e.g., SEQ ID NO: 19). In some embodiments, any of the engineered cells as described herein express a CD38-targeting recombinant receptor (e.g., CAR) and a mbIL15.
[00196] In some embodiments, the mbIL15 is membrane -bound by virtue of the fusion of IL15 to a transmembrane domain. Thus, in some embodiments, mbIL15 comprises a transmembrane domain. In some embodiments, the transmembrane domain comprises a CD8a transmembrane domain. In some embodiments, the transmembrane domain comprises a hinge and/or a transmembrane region. In some embodiments, the transmembrane domain comprises a hinge and a transmembrane region. In some embodiments, the hinge is a CD8a hinge sequence (e.g., SEQ ID NO:6). In some embodiments, the transmembrane region is a CD8a transmembrane region (e.g., SEQ ID NO:8). In some embodiments, the mbIL15 comprises a native IL15 sequence, such as a human native IL15 sequence, and at least one transmembrane domain (e.g., CD8a transmembrane domain). In some embodiments, the CD8a transmembrane domain comprises the sequence of SEQ ID NO: 10. In several embodiments, the mbIL15 is truncated or modified such that it comprises an amino acid sequence that has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% sequency identity to the amino acid sequence of SEQ ID NO:23. In several embodiments, the mbIL15 comprises the amino acid sequence of SEQ ID NO:23. Membrane -bound IL15 sequences are described in PCT publications WO 2018/183385 and WO 2020/056045, each of which is hereby expressly incorporated by reference in its entirety. iv. Cell Types [00197] Some embodiments of the methods and compositions provided herein relate to a cell such as an immune cell. In some embodiments, an immune cell is engineered to express a chimeric receptor that binds to an antigen (e.g., an antigen expressed by a cancer cell).
[00198] Traditional anti-cancer therapies relied on a surgical approach, radiation therapy, chemotherapy, or combinations of these methods. As research led to a greater understanding of some of the mechanisms of certain cancers, this knowledge was leveraged to develop targeted cancer therapies. Targeted therapy is a cancer treatment that employs certain drugs that target specific genes or proteins found in cancer cells or cells supporting cancer growth, (like blood vessel cells) to reduce or arrest cancer cell growth. More recently, genetic engineering has enabled approaches to be developed that harness certain aspects of the immune system to fight cancers. In some cases, a patient’s own immune cells are modified to specifically eradicate that patient’s type of cancer. Various types of immune cells can be used, such as T cells, Natural Killer (NK cells), or combinations thereof, as described in more detail below.
[00199] To facilitate cancer immunotherapies, there are provided for herein polynucleotides, polypeptides, and vectors that encode chimeric antigen receptors (CAR) that comprise a target binding moiety (e.g., an extracellular binder of a ligand, or a tumor marker- directed chimeric receptor, expressed by a cancer cell) and a cytotoxic signaling complex. For example, some embodiments include a polynucleotide, polypeptide, or vector that encodes, for example a chimeric antigen receptor directed against CD38, to facilitate targeting of an immune cell to a cancer and exerting cytotoxic effects on the cancer cell. Methods of treating cancer and other uses of such cells for cancer immunotherapy are also provided for herein. Also provided are engineered immune cells (e.g., T cells or NK cells) expressing such chimeric receptors.
[00200] In several embodiments, cells of the immune system are engineered to have enhanced cytotoxic effects against target cells, such as tumor cells. For example, a cell of the immune system may be engineered to include a CD38-directed CAR as described herein. In several embodiments, white blood cells or leukocytes, are used, since their native function is to defend the body against growth of abnormal cells and infectious disease. There are a variety of types of white bloods cells that serve specific roles in the human immune system and are therefore a preferred starting point for the engineering of cells disclosed herein. White blood cells include granulocytes and agranulocytes (presence or absence of granules in the cytoplasm, respectively). Granulocytes include basophils, eosinophils, neutrophils, and mast cells. Agranulocytes include lymphocytes and monocytes. Cells such as those that follow or are otherwise described herein may be engineered to include a CD38-directed CAR, or a nucleic acid encoding the CAR. In several embodiments, the immune cells are engineered to co-express a membrane-bound interleukin 15 (mbIL15) co-stimulatory domain. In some embodiments, the immune cells engineered to express a CAR are engineered to bicistronically express a mbIL15 domain. a. Monocytes
[00201] In some embodiments, the immune cells comprise monocytes. Monocytes are a subtype of leukocyte. Monocytes can differentiate into macrophages and myeloid lineage dendritic cells. Monocytes are associated with the adaptive immune system and serve the main functions of phagocytosis, antigen presentation, and cytokine production. Phagocytosis is the process of uptake of cellular material, or entire cells, followed by digestion and destruction of the engulfed cellular material.
[00202] In some embodiments, a monocyte is positive for cell surface expression of a marker selected from among the group consisting of CCR2, CCR5, CDllc, CD14, CD16, CD62L, CD68+, CX3CR1, HLA-DR, or any combination thereof. In some embodiments, a monocyte is positive for cell surface expression of CD14. In some embodiments, a monocyte is positive for cell surface expression of CCR2. In some embodiments, a monocyte is positive for cell surface expression of CCR5. In some embodiments, a monocyte is positive for cell surface expression of CD62L.
[00203] In several embodiments, monocytes are used in connection with one or more additional engineered cells as disclosed herein. Some embodiments of the methods and compositions described herein relate to a monocyte that expresses a CAR that binds to CD38, or a nucleic acid encoding the CAR.
[00204] In some embodiments, the monocytes are engineered to express a membranebound interleukin 15 (mbIL15) domain. In some embodiments, the monocytes engineered to express a CAR are engineered to also express (e.g., bicistronically express) a membrane -bound interleukin 15 (mbIL15) domain. Thus, in some embodiments, the monocytes are engineered to bicistronically express the CAR and mbIL15.
[00205] In some embodiments, the monocytes are allogeneic cells. In some embodiments, the monocytes are obtained from a donor who does not have cancer. b. Lymphocytes [00206] In the immune cells comprise lymphocytes. Lymphocytes, the other primary sub-type of leukocyte include T cells (cell-mediated, cytotoxic adaptive immunity), natural killer cells (cell-mediated, cytotoxic innate immunity), and B cells (humoral, antibody-driven adaptive immunity). While B cells are engineered according to several embodiments, disclosed herein, several embodiments also relate to engineered T cells or engineered NK cells (mixtures of T cells and NK cells are used in some embodiments, either from the same donor, or different donors). Thus, in some embodiments, the immune cells comprise T cells. In some embodiments, the immune cells comprise NK cells. In some embodiments, the immune cells comprise T cells and NK cells. In some embodiments, the immune cells comprise B cells.
[00207] In several embodiments, lymphocytes are used in connection with one or more additional engineered cells as disclosed herein. Some embodiments of the methods and compositions described herein relate to a lymphocyte that express a CAR that binds to CD38, or a nucleic acid encoding the CAR.
[00208] In some embodiments, the lymphocytes are engineered to express a membrane -bound interleukin 15 (mbIL15) domain. In some embodiments, the lymphocytes engineered to express a CAR are engineered to also express (e.g., bicistronically express) a membrane-bound interleukin 15 (mbIL15) domain. Thus, in some embodiments, lymphocytes are engineered to bicistronically express the CAR and mbIL15.
[00209] In some embodiments, the lymphocytes are allogeneic cells. In some embodiments, the lymphocytes are obtained from a donor who does not have cancer. c. T Cells
[00210] In some embodiments, the immune cells comprise T cells. T cells are distinguishable from other lymphocytes sub-types (e.g., B cells or NK cells) based on the presence of a T-cell receptor on the cell surface.
[00211] T cells can be divided into various different subtypes, including effector T cells, helper T cells, cytotoxic T cells, memory T cells, regulatory T cells, natural killer T cell, mucosal associated invariant T cells and gamma delta T cells. In some embodiments, a specific subtype of T cell is engineered. In some embodiments, a T cell is positive for cell surface expression of a marker selected from among the group consisting of CD3, CD4, and/or CD8. In some embodiments, a T cell is positive for cell surface expression of CD3. In some embodiments, a T cell is positive or cell surface expression of CD4. In some embodiments, a T cell is positive or cell surface expression of CD8.
[00212] In some embodiments, CD3+ T cells are engineered. In some embodiments, CD4+ T cells are engineered. In some embodiments, CD8+ T cells are engineered. In some embodiments, regulatory T cells are engineered. In some embodiments, gamma delta T cells are engineered. In some embodiments, a mixed pool of T cell subtypes is engineered. For example, in some embodiments, CD4+ and CD8+ T cells are engineered. In some embodiments, there is no specific selection of a type of T cells to be engineered to express the cytotoxic receptor complexes disclosed herein. In several embodiments, specific techniques, such as use of cytokine stimulation are used to enhance expansion/collection of T cells with a specific marker profile. For example, in several embodiments, activation of certain human T cells, e.g. CD4+ T cells, CD8+ T cells is achieved through use of CD3 and/or CD28 as stimulatory molecules.
[00213] In several embodiments, there is provided a method of treating or preventing cancer or an infectious disease, comprising administering a therapeutically effective amount of T cells expressing the cytotoxic receptor complex and/or a homing moiety as described herein. In several embodiments, there is provided a method of treating or preventing cancer or an infectious disease, comprising administering T cells expressing a cytotoxic receptor complex as described herein. In several embodiments, the engineered T cells are autologous cells, while in some embodiments, the T cells are allogeneic cells. In some embodiments, the T cells are allogeneic cells. In some embodiments, the T cells are obtained from a donor who does not have cancer .
[00214] Several embodiments of the methods and compositions disclosed herein relate to T cells engineered to express a CAR that binds to CD38. In some embodiments, the T cells are engineered to express a membrane-bound interleukin 15 (mbIL15) domain. In some embodiments, the T cells engineered to express a CAR are engineered to also express (e.g., bicistronically express) a membrane -bound interleukin 15 (mbIL15) domain. Thus, in some embodiments, the T cells are engineered to bicistronically express the CAR and mbIL15.
[00215] In some embodiments, the immune cells comprise T cells and NK cells (either from the same donor or from different donors d. NK Cells
[00216] In some embodiments, the immune cells comprise natural killer (NK) cells. In several embodiments, there is provided a method of treating or preventing a disease or condition, comprising administering natural killer (NK) cells expressing a CD38-targeting CAR as described herein. In several embodiments, the engineered NK cells are autologous cells, while in some embodiments, the NK cells are allogeneic cells. In several embodiments, the NK cells are autologous cells. In some embodiments, the NK cells are allogeneic cells.
[00217] In several embodiments, NK cells are preferred because the natural cytotoxic potential of NK cells is relatively high. In several embodiments, it is unexpectedly beneficial that the engineered cells disclosed herein can further upregulate the cytotoxic activity of NK cells, leading to an even more effective activity against target cells (e.g., tumor or other diseased cells).
[00218] In some embodiments, a NK cell is positive for cell surface expression of a marker selected from among the group consisting of CCR7, CD16, CD56, CD57, CD11, CX3CR 1 , a Killer Ig-like receptor (KIR), NKp30, NKp44, NKp46, or any combination thereof. In some embodiments, a NK cell is positive for cell surface expression of CD16. In some embodiments, a NK cell is positive for cell surface expression of CD56. In some embodiments, a NK cell is positive for cell surface expression of a Killer Ig-like receptor.
[00219] Some embodiments of the methods and compositions described herein relate to NK cells engineered to express a CAR that binds to CD38. In some embodiments, the NK cells are engineered to a membrane-bound interleukin 15 (mbIL15) domain. In some embodiments, the NK cells engineered to express a CAR are engineered to also express (e.g., bicistronically express) a membrane-bound interleukin 15 (mbIL15) domain. Thus, in some embodiments, the NK cells are engineered to bicistronically express the CAR and mbIL15.
[00220] In some embodiments, the NK cells are derived from cell line NK-92. NK-92 cells are derived from NK cells, but lack major inhibitory receptors displayed by normal NK cells, while retaining the majority of activating receptors. Some embodiments of NK-92 cells described herein related to NK-92 cell engineered to silence certain additional inhibitory receptors, for example, SMAD3, allowing for upregulation of interferon-y (IFNy), granzyme B, and/or perforin production. Additional information relating to the NK-92 cell line is disclosed in WO 1998/49268 and U.S. Patent Application Publication No. 2002-0068044 and incorporated in their entireties herein by reference.
[00221] In some embodiments, the NK cells are used in combination with T cells. Thus, in some embodiments, the immune cells comprise T cells and NK cells (either from the same donor or from different donors). For example, in one embodiment, primary NK cells are used in combination with primary T cells as disclosed herein. e. Hematopoietic Stem Cells
[00222] In some embodiments, the immune cells comprise hematopoietic stem cells (HSCs). In some embodiments, HSCs are used in the methods disclosed herein. In several embodiments, the cells are engineered to express a CAR that binds to CD38. HSCs are used, in several embodiments, to leverage their ability to engraft for long-term blood cell production, which could result in a sustained source of targeted anti-cancer effector cells, for example to combat cancer remissions. In several embodiments, this ongoing production helps to offset anergy or exhaustion of other cell types, for example due to the tumor microenvironment.
[00223] In some embodiments, a HSC is positive for cell surface expression of a marker selected from among the group consisting of CD34, CD59, and CD90. In some embodiments, a HSC is positive for cell surface expression of CD34. In some embodiments, a HSC is positive for cell surface expression of CD59. In some embodiments, a HSC is positive for cell surface expression of CD90.
[00224] In several embodiments allogeneic HSCs are used, while in some embodiments, autologous HSCs are used. In several embodiments, HSCs are used in combination with one or more additional engineered cell type disclosed herein. Some embodiments of the methods and compositions described herein relate to a stem cell, such as a HSC engineered to express a CAR that binds to CD38, or a nucleic acid encoding the CAR.
[00225] In some embodiments, the HSCs are engineered to express a membranebound interleukin 15 (mbIL15) domain. In some embodiments, the HSCs engineered to express a CAR are engineered to also express (e.g., bicistronically express) a membrane-bound interleukin 15 (mbIL15) domain. Thus, in some embodiments, HSCs are engineered to bicistronically express the CAR and mbIL15.
[00226] In some embodiments, the HSCs are allogeneic cells. In some embodiments, the HSCs are obtained from a donor who does not have cancer. f. Induced Pluripotent Stem Cells
[00227] In some embodiments, immune cells are derived (differentiated) from pluripotent stem cells (PSCs). In some embodiments, immune cells (e.g., NK and/or T cells) derived from induced pluripotent stem cells (iPSCs) are used in the method of immunotherapy disclosed herein. For example, in some embodiments, NK cells are derived from iPSCs. In some embodiments, induced pluripotent stem cells (iPSCs) are used in a method disclosed herein. iPSCs are used, in several embodiments, to leverage their ability to differentiate and derive into non-pluripotent cells, including, but not limited to, CD34 cells, hemogenic endothelium cells, HSCs (hematopoietic stem and progenitor cells), hematopoietic multipotcnt progenitor cells, T cell progenitors, NK cell progenitors, T cells, NKT cells, NK cells, and B cells comprising one or several genetic modifications at selected sites through differentiating iPSCs or less differentiated cells comprising the same genetic modifications at the same selected sites. In several embodiments, the iPSCs are used to generate iPSC-derived NK or T cells. In several embodiments, the iPSCs are used to generate iPSC-derived NK cells. In several embodiments, the iPSCs are used to generate iPSC-derived T cells.
[00228] Several embodiments of the methods and compositions disclosed herein relate to induced pluripotent stem cells engineered to express a CAR that binds to CD38. In some embodiments, the iPSCs engineered to express a CAR are engineered to also express (e.g., bicistronically express) a membrane- bound interleukin 15 (mbIL15).
[00229] In several embodiments, the engineered iPSCs are differentiated into NK, T, or other immune cells, such as for use in a composition or method provided herein. In several embodiments, the engineered iPSCs are differentiated into NK cells. In several embodiments, the engineered iPSCs are differentiated into T cells. In several embodiments, the engineered iPSCs are differentiated into NK and T cells. v. Preparation of Cells for Genetic Engineering
[00230] In some embodiments, preparation of the engineered cells includes one or more culture and/or preparation steps. The cells for introduction of the recombinant receptor (e.g., CAR) may be isolated from a sample, such as a biological sample, e.g., one obtained from or derived from a subject. In some embodiments, the sample is an apheresis (e.g.., leukapheresis) sample.
[00231] In some embodiments, the subject from which the cells are isolated is one not having the disease or condition in need of a cell therapy or not to which a cell therapy will be administered. In some embodiments, the cells are isolated from a subject that is different than the subject in need of a cell therapy or to which a cell therapy will be administered. Thus, in some embodiments, the cells are allogeneic to the subject to whom they are administered.
[00232] In some embodiments, the subject from which the cells are isolated is one having the disease or condition or in need of a cell therapy or to which a cell therapy will be administered. In some embodiments, the cells are isolated from the subject to which a cell therapy will be administered. Thus, in some embodiments, the cells are autologous to the subject to whom they are administered.
[00233] The samples include tissue, fluid, and other samples taken directly from the subject, as well as samples resulting from one or more processing steps, such as separation, centrifugation, genetic engineering (e.g., transduction with viral vector), washing, and/or incubation. The biological sample can be a sample obtained directly from a biological source or a sample that is processed. Biological samples include, but are not limited to, body fluids, such as blood, plasma, serum, cerebrospinal fluid, synovial fluid, urine and sweat, tissue and organ samples, including processed samples derived therefrom.
[00234] In some aspects, the sample from which the cells are derived or isolated is blood or a blood-derived sample, or is or is derived from an apheresis (e.g., a leukapheresis) product. Non-limiting examples of samples include whole blood, peripheral blood mononuclear cells (PBMCs), leukocytes, bone marrow, thymus, tissue biopsy, tumor, leukemia, lymphoma, lymph node, gut associated lymphoid tissue, mucosa associated lymphoid tissue, spleen, other lymphoid tissues, liver, lung, stomach, intestine, colon, kidney, pancreas, breast, bone, prostate, cervix, testes, ovaries, tonsil, or other organ, and/or cells derived therefrom. Samples include, in the context of cell therapy, e.g., adoptive cell therapy, samples from autologous and allogeneic sources.
[00235] The cells in some embodiments are primary cells, e.g., primary human cells. In some embodiments, the cells are immune cells, e.g. primary NK cells or primary T cells.
[00236] In some embodiments, isolation of the cells includes one or more preparation and/or non affinity-based cell separation steps. In some examples, cells are washed, centrifuged, and/or incubated in the presence of one or more reagents, for example, to remove unwanted components, enrich for desired components, lyse or remove cells sensitive to particular reagents. In some examples, cells are separated based on one or more property, such as density, adherent properties, size, sensitivity and/or resistance to particular components. [00237] In some examples, cells from the circulating blood of a subject are obtained, e.g., by apheresis (e.g., leukapheresis). In some embodiments, the cells are isolated from an apheresis (e.g., leukapheresis) sample. The samples, in some aspects, contain lymphocytes, including NK cells, T cells, monocytes, granulocytes, B cells, other nucleated white blood cells, red blood cells, and/or platelets, and in some aspects contain cells other than red blood cells and platelets. Non-limiting samples include whole blood, peripheral blood mononuclear cells (PBMCs), leukocytes, bone marrow, thymus, tissue biopsy, tumor, leukemia, lymphoma, lymph node, gut associated lymphoid tissue, mucosa associated lymphoid tissue, spleen, other lymphoid tissues, liver, lung, stomach, intestine, colon, kidney, pancreas, breast, bone, prostate, cervix, testes, ovaries, tonsil, or other organ, and/or cells derived therefrom. In some embodiments, the cells are derived from PBMCs. Samples include, in the context of cell therapy, e.g., adoptive cell therapy, samples from autologous and allogeneic sources.
[00238] In some embodiments, the isolation methods include the separation of different cell types based on the expression or presence in the cell of one or more specific molecules, such as surface markers, e.g., surface proteins, intracellular markers, or nucleic acid. In some embodiments, any known method for separation based on such markers may be used. In some embodiments, the separation is affinity- or immunoaffinity-based separation. For example, the isolation in some aspects includes separation of cells and cell populations based on the cells’ expression or expression level of one or more markers, typically cell surface markers, for example, by incubation with an antibody or binding partner that specifically binds to such markers, followed generally by washing steps and separation of cells having bound the antibody or binding partner, from those cells having not bound to the antibody or binding partner.
[00239] Such separation steps can be based on positive selection, in which the cells having bound the reagents are retained for further use, and/or negative selection, in which the cells having not bound to the antibody or binding partner are retained. In some examples, both fractions are retained for further use. In some aspects, negative selection can be particularly useful where no antibody is available that specifically identifies a cell type in a heterogeneous population, such that separation is best carried out based on markers expressed by cells other than the desired population.
[00240] The separation need not result in 100% enrichment or removal of a particular cell population or cells expressing a particular marker. For example, positive selection of or enrichment for cells of a particular type, such as those expressing a marker, refers to increasing the number or percentage of such cells, but need not result in a complete absence of cells not expressing the marker. Likewise, negative selection, removal, or depletion of cells of a particular type, such as those expressing a marker, refers to decreasing the number or percentage of such cells, but need not result in a complete removal of all such cells. [00241] In some examples, multiple rounds of separation steps are carried out, where the positively or negatively selected fraction from one step is subjected to another separation step, such as a subsequent positive or negative selection. In some examples, a single separation step can deplete cells expressing multiple markers simultaneously, such as by incubating cells with a plurality of antibodies or binding partners, each specific for a marker targeted for negative selection. Likewise, multiple cell types can simultaneously be positively selected by incubating cells with a plurality of antibodies or binding partners expressed on the various cell types.
[00242] For example, in some aspects, NK cells or specific subpopulations thereof, such as cells positive or expressing high levels of one or more surface markers, e.g., CD56+, CCR7+, CD16+, CD57+, CD11+, CX3CR1+, a Killer Ig-like receptor (KIR) +, NKp30+, NKp44+, or NKp46+ NK cells, are isolated by positive or negative selection techniques. In some aspects, NK cells are isolated by positive selection for CD56. For example, CD56+ NK cells can be positively selected using anti-CD56 conjugated magnetic beads.
[00243] In some embodiments, T cells are separated from a PBMC sample by negative selection of markers expressed on non-T cells, such as B cells, monocytes, or other white blood cells, such as CD14. In some aspects, a CD4+ or CD8+ selection step is used to separate CD4+ helper and CD8+ cytotoxic T cells. Such CD4+ and CD8+ populations can be further sorted into sub-populations by positive or negative selection for markers expressed or expressed to a relatively higher degree on one or more naive, memory, and/or effector T cell subpopulations.
[00244] In some embodiments, the cells (e.g., NK cells) are expanded in culture prior to, during, and/or following genetic engineering. In some embodiments, the cells are expanded in culture prior to genetic engineering. In some embodiments, the cells are expanded in culture following genetic engineering. In some embodiments, the cells are expanded in culture prior to and following genetic engineering. Methods for expanding cells are known in the art and include any of those described in US Patent Nos. 7,435,596 and 8,026,097; and Patent Application Nos. PCT/SG2018/050138; PCT/US2020/044033; PCT/US2021/071330; and PCT/US2022/074164.
[00245] In some embodiments, expanding the cells in culture comprises co-culturing the cells with feeder cells. Thus, in some embodiments, the cells are expanded in culture prior to genetic engineering by co-culturing the cells with feeder cells. In some embodiments, the feeder cells express IL15 (e.g., membrane -bound IL15) and 4-1BBL. In some embodiments, the feeder cells express membrane-bound interleukin 15 (mbIL15) and 4-1BBL. In some embodiments, the feeder cells do not express MHCI molecules. In some embodiments, the feeder cells do not express MHCII molecules. In some embodiments, the feeder cells are immune cells. In some embodiments, the feeder cells are K562 cells. Engineered feeder cells are disclosed in, for example, International Patent Application PCT/SG2018/050138. In some embodiments, following genetic engineering, the cells are allowed to further expand in culture. [00246] In some embodiments, expanding the cells in culture comprising culturing the cells in the presence of IL2, IL12, and/or IL18. In some embodiments, the cells are cultured in the presence of IL2. In some embodiments, the cells are cultured in the presence of IL12 and IL18. In some embodiments, the cells are cultured in the presence of IL2, IL12, and IL18.
[00247] In some embodiments, the preparation methods include steps for freezing, e.g., cryopreserving, the cells, either before or after isolation, engineering, and/or expansion. In some aspects, the cells are cryopreserved after engineering. In some aspects, such as when the cells are further expanded in culture after genetic engineering, the cells are cryopreserved after the further expansion. In some embodiments, the cells are suspended in a freezing solution. Any of a variety of known freezing solutions and parameters in some aspects may be used.
II. Compositions and Formulations
[00248] Also provided are compositions including the CD38-binding molecules, immunoconjugates, recombinant receptors, and engineered cells, including pharmaceutical compositions and formulations. Also provided are compositions comprising engineered cells that express the CD38-binding molecules provided herein, such as recombinant receptors (e.g., CARs), including pharmaceutical compositions and formulations.
[00249] Provided are pharmaceutical formulations comprising a CD38-binding molecule (e.g., antibody), an immunoconjugate, a recombinant receptor (e.g., chimeric antigen receptor), engineered cells expressing said molecules (e.g., antibody or recombinant receptor), a plurality of engineered cells expressing said molecules (e.g., recombinant receptor) and/or additional agents for combination treatment or therapy. The pharmaceutical compositions and formulations generally include one or more optional pharmaceutically acceptable carrier or excipient. In some embodiments, the composition includes at least one additional therapeutic agent.
[00250] The term “pharmaceutical formulation” refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
[00251] A “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject. A pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
[00252] In some aspects, the choice of carrier is determined in part by the particular cell, binding molecule, and/or antibody, and/or by the method of administration. Accordingly, there are a variety of suitable formulations. For example, the pharmaceutical composition can contain preservatives. Suitable preservatives may include, for example, methylparaben, propylparaben, sodium benzoate, and benzalkonium chloride. In some aspects, a mixture of two or more preservatives is used. The preservative or mixtures thereof are typically present in an amount of about 0.0001% to about 2% by weight of the total composition. Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g. Zn-protein complexes); and/or non-ionic surfactants such as polyethylene glycol (PEG).
[00253] In some aspects, a buffer is included in the composition. Suitable buffering agents include, for example, citric acid, sodium citrate, phosphoric acid, potassium phosphate, and various other acids and salts. In some aspects, a mixture of two or more buffers is used. The buffering agent or mixtures thereof are typically present in an amount of from about 0.001% to about 4% by weight of the total composition. Methods for preparing administrable pharmaceutical compositions are known.
[00254] Formulations of the antibodies described herein can include lyophilized formulations and aqueous solutions. The formulation or composition may also contain more than one active ingredient useful for the particular indication, disease, or condition being treated with the binding molecules or cells, preferably those with activities complementary to the binding molecule or cell, where the respective activities do not adversely affect one another. Such active ingredients are suitably present in combination in amounts that are effective for the purpose intended. Thus, in some embodiments, the pharmaceutical composition further includes other pharmaceutically active agents or drugs, such as chemotherapeutic agents, e.g., asparaginase, busulfan, carboplatin, cisplatin, daunorubicin, doxorubicin, fluorouracil, gemcitabine, hydroxyurea, methotrexate, paclitaxel, rituximab, vinblastine, vincristine, etc. In some embodiments, the cells or antibodies are administered in the form of a salt, e.g., a pharmaceutically acceptable salt. Suitable pharmaceutically acceptable acid addition salts include those derived from mineral acids, such as hydrochloric, hydrobromic, phosphoric, metaphosphoric, nitric, and sulphuric acids, and organic acids, such as tartaric, acetic, citric, malic, lactic, fumaric, benzoic, glycolic, gluconic, succinic, and arylsulphonic acids, for example, p-toluenesulphonic acid.
[00255] The pharmaceutical composition in some embodiments contains the binding molecules and/or cells in amounts effective to treat or prevent the disease or condition, such as a therapeutically effective or prophylactically effective amount. Therapeutic or prophylactic efficacy in some embodiments is monitored by periodic assessment of treated subjects. For repeated administrations over several days or longer, depending on the condition, the treatment is repeated until a desired suppression of disease symptoms occurs. However, other dosage regimens may be useful and can be determined. The desired dosage can be delivered by a single bolus administration of the composition, by multiple bolus administrations of the composition, or by continuous infusion administration of the composition.
[00256] Formulations include those for oral, intravenous, intraperitoneal, subcutaneous, pulmonary, transdermal, intramuscular, intranasal, buccal, sublingual, or suppository administration. In some embodiments, the agent or cell population is administered to the subject by intravenous, intraperitoneal, or subcutaneous injection using peripheral systemic delivery.
[00257] In some embodiments, the compositions are provided as sterile liquid formulations (e.g., isotonic aqueous solutions, suspensions, emulsions, dispersions, or viscous compositions), which in some aspects may be buffered to a selected pH. Liquid formulations are generally easier to prepare than gels, other viscous compositions, and solid compositions. In addition, liquid compositions are somewhat more convenient to administer, particularly by injection. The liquid composition can comprise a carrier, which can be a solvent or dispersion medium containing, for example, water, saline, phosphate buffered saline, polyols (e.g., glycerol, propylene glycol, liquid polyethylene glycol), and suitable mixtures thereof.
[00258] Sterile injectable solutions can be prepared by incorporating the agent or cell into a solvent, such as an admixture with a suitable carrier, diluent, or excipient (e.g., sterile water, saline, glucose, dextrose, and the like). Formulations for in vivo administration are typically sterile. Sterility can be readily achieved, for example, by filtration through sterile filtration membranes. In some embodiments, the dose of engineered cells administered is in a cryopreserved composition. In some aspects, the composition is administered after thawing the cryopreserved composition.
[00259] Also provided are pharmaceutical compositions for combination therapy. Any of the additional agents for combination therapy described herein, such as agents described in Section III.B, can be prepared and administered as one or more pharmaceutical compositions, with the CD38-binding molecule (e.g., antibody), immunoconjugate, recombinant receptor (e.g., chimeric antigen receptor) and/or engineered cells expressing said molecules (e.g., recombinant receptor) described herein. The combination therapy can be administered in one or more pharmaceutical compositions, e.g., where the binding molecules, recombinant receptors and/or cells are in the same pharmaceutical composition as the additional agent, or in separate pharmaceutical compositions. For example, in some embodiments, the additional agent is an additional engineered cell, e.g., cell engineered to express a different recombinant receptor that targets a different antigen (e.g., BCMA) or a different epitope on CD38, and is administered in the same composition or in a separate composition. In some embodiments, each of the pharmaceutical composition is formulated in a suitable formulation according to the particular binding molecule, recombinant receptor, cell, e.g., engineered cell, and/or additional agent, and the particular dosage regimen and/or method of delivery.
III. Methods and Uses
[00260] Provided herein are methods of using and uses of the CD38-binding molecules, immunoconjugates, recombinant receptors, engineered cells, and pharmaceutical compositions and formulations thereof, such as in the treatment of diseases, conditions, and disorders in which CD38 is expressed, and/or in detection, diagnostic, and prognostic methods. Among such methods, such as methods of treatment, and uses, are those that involve administering to a subject engineered cells, such as a plurality of engineered cells, expressing the provided anti-CD38 recombinant receptors (e.g. CARs). Also provided are methods of combination therapy and/or treatment.
A. Therapeutic and Prophylactic Methods and Uses
[00261] Also provided are methods of administering and uses, such as therapeutic and prophylactic uses, of the CD38-binding molecules, including the anti-CD38 recombinant receptors (e.g., CARs), engineered cells expressing the recombinant receptors (e.g., CARs), plurality of engineered cells expressing the receptors, and/or compositions comprising the same. Such methods and uses include therapeutic methods and uses, for example, involving administration of the molecules (e.g., recombinant receptors), cells (e.g., engineered cells), or compositions containing the same, to a subject having a disease, condition, or disorder associated with CD38 such as a disease, condition, or disorder associated with CD38 expression, and/or in which cells or tissues express, e.g., specifically express, CD38. In some embodiments, the binding molecule, cell, and/or composition is/are administered in an effective amount to effect treatment of the disease or disorder.
[00262] Provided herein are uses of the recombinant receptors (e.g., CARs), and cells (e.g., engineered cells) in such methods and treatments, and in the preparation of a medicament in order to carry out such therapeutic methods. In some embodiments, the methods are carried out by administering the binding molecules or cells, or compositions comprising the same, to the subject having, having had, or suspected of having the disease or condition. In some embodiments, the methods thereby treat the disease or condition or disorder in the subject. Also provided herein are of use of any of the compositions, such as pharmaceutical compositions provided herein, for the treatment of a disease or disorder associated with CD38, such as use in a treatment regimen.
[00263] As used herein, “treatment” (and grammatical variations thereof such as “treat” or “treating”) refers to complete or partial amelioration or reduction of a disease or condition or disorder, or a symptom, adverse effect or outcome, or phenotype associated therewith. Desirable effects of treatment include, but are not limited to, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis. The terms do not imply complete curing of a disease or complete elimination of any symptom or effect(s) on all symptoms or outcomes.
[00264] As used herein, “delaying development of a disease” means to defer, hinder, slow, retard, stabilize, suppress and/or postpone development of the disease (such as cancer). This delay can be of varying lengths of time, depending on the history of the disease and/or subject being treated. In some embodiments, the provided molecules and compositions are used to delay development of a disease or to slow the progression of a disease. A sufficient or significant delay can, in effect, encompass prevention, in that the subject does not develop the disease. For example, a late-stage cancer, such as development of metastasis, may be delayed.
[00265] “Preventing,” as used herein, includes providing prophylaxis with respect to the occurrence or recurrence of a disease in a subject that may be predisposed to the disease but has not yet been diagnosed with the disease.
[00266] As used herein, to “suppress” a function or activity is to reduce the function or activity when compared to otherwise same conditions except for a condition or parameter of interest, or alternatively, as compared to another condition. For example, an antibody or composition or cell which suppresses tumor growth reduces the rate of growth of the tumor compared to the rate of growth of the tumor in the absence of the antibody or composition or cell.
[00267] An “effective amount” of an agent, e.g., a pharmaceutical formulation, binding molecule, antibody, cells, or composition, in the context of administration, refers to an amount effective, at dosages/amounts and for periods of time necessary, to achieve a desired result, such as a therapeutic or prophylactic result.
[00268] A “therapeutically effective amount” of an agent, e.g., a pharmaceutical formulation, binding molecule, antibody, cells, or composition refers to an amount effective, at dosages and for periods of time necessary, to achieve a desired therapeutic result, such as for treatment of a disease, condition, or disorder, and/or pharmacokinetic or pharmacodynamic effect of the treatment. The therapeutically effective amount may vary according to factors such as the disease state, age, sex, and weight of the subject, and the populations of cells administered. In some embodiments, the provided methods involve administering the molecules, antibodies, cells, and/or compositions at effective amounts, e.g., therapeutically effective amounts. [00269] A “prophylactically effective amount” refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, but not necessarily, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount will be less than the therapeutically effective amount.
[00270] As used herein, a “subject” or an “individual” is a mammal. In some embodiments, a “mammal” includes humans, non-human primates, domestic and farm animals, and zoo, sports, or pet animals, such as dogs, horses, rabbits, cattle, pigs, hamsters, gerbils, mice, ferrets, rats, cats, monkeys, etc. In some embodiments, the subject is human.
[00271] Methods for administration of cells for adoptive cell therapy are known and may be used in connection with the provided methods and compositions.
[00272] Among the diseases to be treated is any disease or disorder associated with CD38 or any disease or disorder in which CD38 is specifically expressed and/or in which CD38 has been targeted for treatment (also referred to herein interchangeably as a “CD38-associated disease or disorder”). Cancers associated with CD38 expression include solid tumors, hematologic malignancies such as multiple myeloma, Waldenstrom macroglobulinemia, as well as both Hodgkin's and non-Hodgkin's lymphoma. See Konen et al., Cells (2020) 9(1 ):52; proteinatlas.org/ENSG00000004468-CD38/pathology). Since CD38 is highly and uniformly expressed on plasma cells, it is a potential target for cancer therapy. CD38 is also expressed by long-lived plasma cells, which produce autoantibodies associated with autoimmune diseases. Anti-CD38 antibodies have been previously described. See van de Donk, Blood (2018) 131(1): 13- 29.
[00273] In some embodiments, the disease or disorder associated with CD38 is a B cell-related disorder. In some embodiments, the disease or disorder associated with CD38 is one or more diseases or conditions from among glioblastoma, lymphomatoid granulomatosis, posttransplant lymphoproliferative disorder, an immunoregulatory disorder, heavy-chain disease, primary or immunocyte-associated amyloidosis, or monoclonal gammopathy of undetermined significance.
[00274] In some embodiments, the disease or disorder associated with CD38 is an autoimmune disease or disorder. Such autoimmune diseases or disorder include, but are not limited to, systemic lupus erythematosus (SLE), lupus nephritis, CNS lupus, inflammatory bowel disease (IBD, e.g, Crohn’s disease or ulcerative colitis), rheumatoid arthritis (RA; e.g., juvenile rheumatoid arthritis), ANCA associated vasculitis, idiopathic thrombocytopenia purpura (ITP), thrombotic thrombocytopenia purpura (TTP), autoimmune thrombocytopenia, Chagas' disease, Grave's disease, Wegener's granulomatosis, polyarteritis nodosa, Sjogren's syndrome, pemphigus vulgaris, scleroderma, multiple sclerosis(MS), psoriasis, IgA nephropathy, IgM polyneuropathies, vasculitis, diabetes mellitus, Reynaud's syndrome, anti-phospholipid syndrome, Goodpasture's disease, Kawasaki disease, autoimmune hemolytic anemia, myasthenia gravis(MG), or progressive glomerulonephritis. In some embodiments, a subject may exhibit symptoms of more than one autoimmune disease. In some embodiments, the autoimmune disease is systemic lupus erythematosus (SLE). In some embodiments, the autoimmune disease is SLE with renal involvement (e.g., lupus nephritis (LN)). In some embodiments, the autoimmune disease is lupus nephritis (LN). In some embodiments, the autoimmune disease is CNS lupus. In some embodiments, the autoimmune disease is IBD. In some embodiments, the autoimmune disease is Crohn’s disease. In some embodiments, the autoimmune disease is ulcerative colitis. In some embodiments, the autoimmune disease is vasculitis. In some embodiments, the autoimmune disease is ANCA vasculitis (AAV). In some embodiments, the autoimmune disease is autoimmune encephalitis (AE). In some embodiments, the autoimmune disease is ITP. In some embodiments, the autoimmune disease is TTP. In some embodiments, the autoimmune disease is autoimmune thrombocytopenia. In some embodiments, the autoimmune disease is Chagas' disease. In some embodiments, the autoimmune disease is Graves’ disease. In some embodiments, the autoimmune disease is Wegener's granulomatosis. In some embodiments, the autoimmune disease is polyarteritis nodosa. In some embodiments, the autoimmune disease is Sjogren's syndrome. In some embodiments, the autoimmune disease is pemphigus vulgaris. In some embodiments, the autoimmune disease is psoriasis. In some embodiments, the autoimmune disease is IgA nephropathy. In some embodiments, the autoimmune disease is membranous nephropathy (MN). In some embodiments, the autoimmune disease is IgM polyneuropathies. In some embodiments, the autoimmune disease is vasculitis. In some embodiments, the autoimmune disease is diabetes mellitus. In some embodiments, the autoimmune disease is Reynaud's syndrome. In some embodiments, the autoimmune disease is anti-phospholipid syndrome. In some embodiments, the autoimmune disease is Goodpasture's disease. In some embodiments, the autoimmune disease is Kawasaki disease. In some embodiments, the autoimmune disease is autoimmune hemolytic anemia. In some embodiments, the autoimmune disease is myasthenia gravis (MG). In some embodiments, the autoimmune disease is progressive glomerulonephritis. In some embodiments, the autoimmune disease is acquired immunodeficiency syndrome (AIDS). In some embodiments, the autoimmune disease is Addison’s disease. In some embodiments, the autoimmune disease is alopecia areata. In some embodiments, the autoimmune disease is celiac disease. In some embodiments, the autoimmune disease is chronic inflammatory demyelinating polyneuropathy (CIDP). In some embodiments, the autoimmune disease is Guillain-Barre syndrome. In some embodiments, the autoimmune disease is Hashimoto thyroiditis. In some embodiments, the autoimmune disease is pernicious anemia. In some embodiments, the autoimmune disease is psoriasis. In some embodiments, the autoimmune disease is psoriatic arthritis. In some embodiments, the autoimmune disease is reactive arthritis. In some embodiments, the autoimmune disease is rheumatoid arthritis (RA). In some embodiments, the autoimmune disease is refractory RA. In some embodiments, RA is refractory to a TNF inhibitor. In some embodiments, the autoimmune disease is multiple sclerosis (MS). In some embodiments, MS is primary progressive MS (PPMS). In some embodiments, MS is secondary-progressive MS (SPMS). In some embodiments, MS is relapsing-remitting MS (RRMS).
[00275] In some embodiments, the autoimmune disease comprises scleroderma. In some embodiments, the autoimmune disease is scleroderma. In some embodiments, the autoimmune disease comprises systemic sclerosis (also known as systemic scleroderma). In some embodiments, the autoimmune disease is systemic sclerosis (also known as systemic scleroderma). In some embodiments, the autoimmune disease comprises localized scleroderma. In some embodiments, the autoimmune disease is localized scleroderma.
[00276] In some embodiments, the autoimmune disease comprises myositis (also known as IIM). In some embodiments, the autoimmune disease is myositis (also known as UM). In some embodiments, the autoimmune disease is selected from the group consisting of antisynthetase syndrome (ASSD), overlap myopathy (OM), dermatomyositis (DM), clinically amyopathic dermatomyositis, juvenile myositis (JM), necrotizing myopathy (NM; e.g., necrotizing autoimmune myopathy (or immune-mediated necrotizing myopathy), polymyositis (PM), and sporadic inclusion body myositis (sIBM). In some embodiments, the autoimmune disease is ASSD. In some embodiments, the autoimmune disease is OM. In some embodiments, the autoimmune disease is DM. In some embodiments, the autoimmune disease is JM. In some embodiments, the autoimmune disease is NM In some embodiments, the autoimmune disease is PM. In some embodiments, the autoimmune disease is sIBM.
[00277] In some embodiments, the autoimmune disease comprises vasculitis. In some embodiments, the autoimmune disease is vasculitis. In some embodiments, the vasculitis is large- vessel vasculitis. In some embodiments, the vasculitis is medium-vessel vasculitis. In some embodiments, the vasculitis is small-vessel vasculitis. In some embodiments, the vasculitis is anti- neutrophilic cytoplasmic autoantibody (ANCA) vasculitis. In some embodiments the ANCA vasculitis is granulomatosis with polyangiitis (GPA). In some embodiments the ANCA vasculitis is microscopic polyangiitis (MPA). In some embodiments the ANCA vasculitis is eosinophilic granulomatosis with polyangiitis (EGPA).
[00278] In some embodiments, the autoimmune disease comprises myasthenia gravis (MG). In some embodiments, the autoimmune disease is MG. In some embodiments, MG is ocular MG. In some embodiments, MG is early-onset generalized MG. In some embodiments, MG is late-onset MG.
[00279] In some embodiments, the autoimmune disease is autoimmune encephalitis (AE). In some embodiments, AE comprises an antibody to an intracellular antigen (e.g., anti-Hu or anti-GAD65). In some embodiments, AE comprises an autoantibody to an extracellular epitope of an ion channel, receptor, and/or other associated protein (e.g., anti-NMDA receptor). In some embodiments, the disease or disorder associated with CD38 is an inflammatory disease or disorder. Such inflammatory diseases or disorders include, but are not limited to, respiratory synctial virus (RSV), chronic obstructive pulmonary disease (COPD), rheumatoid arthritis (RA), and collagen-induced arthritis (CIA). See Konen et al., Cells (2020) 9(1):52.
[00280] In certain diseases and conditions, CD38 is expressed on malignant cells and cancers. In some embodiments, the cancer (e.g., a CD38-expressing cancer) is a B cell malignancy. In some embodiments, the cancer (e.g., a CD38-expressing cancer) is a lymphoma, a leukemia, or a plasma cell malignancy. Lymphomas contemplated herein include, but are not limited to, Burkitt lymphoma (e.g., endemic Burkitt's lymphoma or sporadic Burkitt's lymphoma), non-Hodgkin's lymphoma (NHL), Hodgkin's lymphoma, Waldenstrom macroglobulinemia, follicular lymphoma, small non-cleaved cell lymphoma, mucosa-associated lymphatic tissue lymphoma (MALT), marginal zone lymphoma, splenic lymphoma, nodal monocytoid B cell lymphoma, immunoblastic lymphoma, large cell lymphoma, diffuse mixed cell lymphoma, pulmonary B cell angiocentric lymphoma, small lymphocytic lymphoma, primary mediastinal B cell lymphoma, lymphoplasmacytic lymphoma (LPL), or mantle cell lymphoma (MCL). Leukemias contemplated here, include, but are not limited to, chronic lymphocytic leukemia (CLL), plasma cell leukemia or acute lymphocytic leukemia (ALL). Also contemplated herein are plasma cell malignancies including, but not limited to, multiple myeloma (e.g., non-secretory multiple myeloma, smoldering multiple myeloma) or plasmacytoma. In some embodiments the disease or condition is multiple myeloma (MM), such as relapsed and/or refractory multiple myeloma (R/R MM). Among the diseases, disorders or conditions associated with CD38 (e.g., a CD38-expressing cancer) that can be treated include, but are not limited to, melanoma, neuroblastoma, renal cell carcinoma, colon cancer, colorectal cancer, breast cancer, epithelial squamous cell cancer, melanoma, myeloma (e.g., multiple myeloma), stomach cancer, brain cancer, lung cancer, pancreatic cancer, cervical cancer, ovarian cancer, liver cancer, bladder cancer, prostate cancer, testicular cancer, thyroid cancer, uterine cancer, adrenal cancer, and head and neck cancer. In some embodiments, the disease or disorder is melanoma, glioma, esophageal, cervical, or lung cancer.
[00281] In some embodiments, the methods may identify a subject who has, is suspected to have, or is at risk for developing a CD38-associated disease or disorder. Hence, provided are methods for identifying subjects with diseases or disorders associated with elevated CD38 expression and selecting them for treatment with a provided CD38-binding recombinant receptor (e.g., CAR), and/or engineered cells expressing the recombinant receptor.
[00282] For example, a subject may be screened for the presence of a disease or disorder associated with elevated CD38 expression, such as a CD38-expressing cancer. In some embodiments, a subject may be screened for the presence of a disease or disorder associated with elevated CD38 expression, such as an autoimmune disease. In some embodiments, the methods include screening for or detecting the presence of a CD38-associated disease, e.g. a tumor. In some embodiments, the methods include screening for or detecting the presence of a CD38- associated disease, e.g. an autoimmune disease. Thus, in some aspects, a sample may be obtained from a patient suspected of having a disease or disorder associated with elevated CD38 expression and assayed for the expression level of CD38. In some aspects, a subject who tests positive for a CD38-associated disease or disorder may be selected for treatment by the present methods, and may be administered a therapeutically effective amount of a recombinant receptor (e.g., CAR) comprising a CD38-binding molecule, cells containing a recombinant receptor or a pharmaceutical composition thereof as described herein. In some aspects, provided herein is a method for detecting CD38 in a cell or tissue comprising: (a) contacting the cell or tissue with an anti-CD38 antibody or antigen- binding fragment described herein; and (b) determining the amount of the antibody or antigen-binding fragment thereof bound to the cell or tissue, wherein the amount of antibody or antigen-binding fragment thereof bound to the cell or tissue indicates the amount of CD38 in the cell or tissue.
[00283] In some embodiments, the subject has persistent or relapsed disease, e.g., following treatment with another CD38-specific antibody, cells expressing a CD38 -targeting chimeric receptor, a BCMA-specific antibody, cells expressing a BCMA-targeting chimeric receptor, and/or other therapy, including chemotherapy, radiation, and/or hematopoietic stem cell transplantation (HSCT), e.g., allogeneic HSCT or autologous HSCT. In some embodiments, the subject has persistent or relapsed disease, e.g., following treatment with a B cell -targeting agent, such as an anti-CD19 antibody cells expressing a CD19-targeting chimeric receptor, or an anti- CD20 antibody or cells expressing a CD20-targeting chimeric receptor. In some embodiments, the administration effectively treats the subject despite the subject having become resistant to a BCMA-targeted therapy and/or another CD38-targeted therapy In some embodiments, the administration effectively treats the subject despite the subject having become resistant to a CD19-targeted therapy and/or a CD20-targeted therapy. In some embodiments, the subject has not relapsed but is determined to be at risk for relapse, such as at a high risk of relapse, and thus the compound or composition is administered prophylactically, e.g., to reduce the likelihood of or prevent relapse.
[00284] In some embodiments, prior to the initiation of administration of the engineered cells, the subject has received one or more prior therapies. In some embodiments, the subject has received at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 or more prior therapies. In some embodiments, the subject has received at least 3, 4, 5, 6, 7, 8, 9, 10 or more prior therapies. In some embodiments, the subject has received at least 1 prior therapy. In some embodiments, the subject has received at least 2 prior therapies. In some embodiments, the subject has received at least 3 prior therapies. In some embodiments, the subject has received at least 4 prior therapies. [00285] In some aspects, the subject has relapsed or has been refractory to the one or more prior therapies. In some aspects, the prior therapies include treatment with autologous stem cell transplant (ASCT); an immunomodulatory agent (ImiD); a proteasome inhibitor; cells expressing a BCMA-targeted chimeric receptor, a BCMA-specific antibody, and/or an anti-CD38 antibody. In some aspects, the prior therapies include treatment with an anti-CD19 antibody or cells expressing an anti-CD19 chimeric receptor. In some aspects, the prior therapies include treatment with an anti-CD20 antibody (e.g., rituximab). In some embodiments, the immunomodulatory agent comprises thalidomide, lenalidomide or pomalidomide. In some embodiments, the proteasome inhibitor comprises bortezomib (VELCADE®), carfilzomib, or ixazomib. In some embodiments, the cells expressing a BCMA-targeted chimeric receptor are idecabtagene vicleucel (ABECMA®) or ciltacabtagene autoleucel (CARVYKTI™). In some embodiments, the BCMA-specific antibody is a bispecific antibody. In some embodiments, the BCMA-specific antibody is or comprises teclistamab-cqyv (TECVAYLI™)- In some embodiments, the BCMA-specific antibody is an antibody-drug conjugate (ADC). In some embodiments, the BCMA specific antibody is or comprises belantamab mafodotin (BLENREP®). In some embodiments, the anti-CD38 antibody is or comprises daratumumab (e.g., DARZALEX® or DARZALEX Faspro®). In some aspects, the prior therapies include treatment with an anti-CD19 antibody or cells expressing an anti-CD19 chimeric receptor. In some aspects, the prior therapies include treatment with an anti-CD20 antibody (e.g., rituximab).
[00286] In some embodiments, the cells are isolated and/or otherwise prepared from the subject who is to receive the cell therapy, or from a sample derived from such a subject. Thus, in some aspects, the cells are derived from a subject, e.g., patient, in need of a treatment and the cells, following isolation and processing are administered to the same subject. Thus, in some embodiments, the cells are autologous to the subject to be treated.
[00287] In some embodiments, the cell therapy, e.g., adoptive cell therapy, e.g., adoptive T cell therapy, is carried out by allogeneic transfer, in which the cells are isolated and/or otherwise prepared from a subject other than a subject who is to receive or who ultimately receives the cell therapy, e.g., a first subject. In such embodiments, the cells then are administered to a different subject, e.g., a second subject, of the same species. Thus, in some embodiments, the cells are allogeneic to the subject to be treated.
[00288] In some embodiments, the subject, to whom the cells, cell populations, or compositions are administered, is a primate, such as a human. In some embodiments, the subject, to whom the cells, cell populations, or compositions are administered, is a non-human primate. In some embodiments, the non-human primate is a monkey (e.g., cynomolgus monkey) or an ape. The subject can be male or female and can be any suitable age, including infant, juvenile, adolescent, adult, and geriatric subjects. In some embodiments, the subject is a non-primate mammal, such as a rodent (e.g., mouse, rat, etc.). [00289] For the prevention or treatment of disease, the appropriate dosage of the CD38-binding molecule, recombinant receptor or cell may depend on the type of disease to be treated, the type of binding molecule or recombinant receptor, the severity and course of the disease, whether the binding molecule or recombinant receptor is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the recombinant receptor or cell, and the discretion of the attending physician. The compositions and molecules and cells are in some embodiments suitably administered to the patient at one time or over a series of treatments.
[00290] In some embodiments, the dose and/or frequency of administration is determined based on efficacy and/or response. In some embodiments, the dose and/or frequency of administration is determined based on efficacy and/or response. In some embodiments efficacy and/or response is determined by a disease activity index for the disease. For example, in some embodiments efficacy and/or response is determined by the International Myeloma Working Group (IMWG) Uniform Response Criteria. In some examples, dose and/or frequency of administration is determined by the expansion and persistence of the recombinant receptor or cell in the blood and/or bone marrow. In some embodiments, dose and/or frequency of administration is determined based on the antitumor activity of the recombinant receptor or engineered cell.
[00291] In certain embodiments, treatment of a subject with a genetically engineered cell(s) described herein achieves one, two, three, four, or more of the following effects, including, for example: (i) reduction or amelioration the severity of disease or symptom associated therewith; (ii) reduction in the duration of a symptom associated with a disease; (iii) protection against the progression of a disease or symptom associated therewith; (iv) regression of a disease or symptom associated therewith; (v) protection against the development or onset of a symptom associated with a disease; (vi) protection against the recurrence of a symptom associated with a disease; (vii) reduction in the hospitalization of a subject; (viii) reduction in the hospitalization length; (ix) an increase in the survival of a subject with a disease; (x) a reduction in the number of symptoms associated with a disease; and (xi) an enhancement, improvement, supplementation, complementation, or augmentation of the prophylactic or therapeutic effect(s) of another therapy.
[00292] In some embodiments, the method comprises administering a dose of the engineered cells or a composition comprising a dose of the engineered cells. In some embodiments, the engineered cells or compositions containing engineered cells can be used in a treatment regimen, wherein the treatment regimen comprises administering a dose of the engineered cells or a composition comprising a dose of the engineered cells. In some embodiments, the dose can contain, for example, a particular number or range of recombinant receptor-expressing immune cells (e.g., NK cells), such as any number of such cells described herein. In some embodiments, a composition containing a dose of the cells can be administered. In some aspects, the number, amount or proportion of CAR-expressing cells in a cell population or a cell composition can be assessed by detection of a surrogate marker, e.g., by flow cytometry or other means, or by detecting binding of a labelled molecule, such as a labelled antigen, that can specifically bind to the binding molecules or receptors provided herein.
[00293] Doses of immune cells such as NK and/or T cells can be determined for a given subject based on their body mass, disease type and state, and desired aggressiveness of treatment, but range, depending on the embodiments, from about 105 cells per kg to about 1012 cells per kg (e.g., 105-107, 1O7-1O10, 10’°-1012 and overlapping ranges therein). In one embodiment, a dose escalation regimen is used. In several embodiments, a range of immune cells such as NK and/or T cells is administered, for example between about 1 x 106 cells/kg to about 1 x 108 cells/kg. In several embodiments, a range of NK cells is administered, for example between about 1 x 106 cells/kg to about 1 x 108 cells/kg. In several embodiments, a range of T cells is administered, for example between about 1 x 106 cells/kg to about 1 x 108 cells/kg.
[00294] In some embodiments, a dose of engineered cells comprises between about 300 x 106 and 1.5 x 109 NK cells. In some embodiments, a dose of engineered cells comprises about 300 x 106 NK cells. In some embodiments, a dose of engineered cells comprises about 1 x 109 NK cells. In some embodiments, a dose of engineered cells comprises about 1.5 x 109 NK cells. In some embodiments, a dose of engineered cells comprises about 2 x 109 NK cells. In some embodiments, a dose of engineered cells comprises about 2.5 x 109 NK cells. In some embodiments, a dose of engineered cells comprises about 3 x 109NK cells.
[00295] In some embodiments, a dose of engineered cells comprises between about 300 x 106 and 3 x 109 CAR-expressing NK cells. In some embodiments, a dose of engineered cells comprises about 300 x 106 CAR-expressing NK cells. In some embodiments, a dose of engineered cells comprises about 1 x 109 CAR-expressing NK cells. In some embodiments, a dose of engineered cells comprises about 1.5 x 109 CAR-expressing NK cells. In some embodiments, a dose of engineered cells comprises about 2 x 109 CAR-expressing NK cells. In some embodiments, a dose of engineered cells comprises about 2.5 x 109 CAR-expressing NK cells. In some embodiments, a dose of engineered cells comprises about 3 x 109 CAR-expressing NK cells.
[00296] In some embodiments, a dose of engineered cells comprises between about 300 x 106 and 1.5 x 109 T cells. In some embodiments, a dose of engineered cells comprises about 300 x 106 T cells. In some embodiments, a dose of engineered cells comprises about 1 x 109 T cells. In some embodiments, a dose of engineered cells comprises about 1.5 x 109T cells.
[00297] In some embodiments, a dose of engineered cells comprises both NK and T cells. In some embodiments, a dose of engineered cells comprises between about 300 x 106 and 1.5 x 109 NK cells and between about 300 x 106 and 1.5 x 109 T cells. In some embodiments, a dose comprises about an equal number of NK cells and T cells. In some embodiments, a dose comprises more NK cells than T cell. In some embodiments, a dose comprises more T cells than NK cells.
[00298] In several embodiments, multiple doses are used, for example, two, three, four, or more doses within a dosing cycle. Such multi-dose cycles can be repeated one or more times, as needed to treat a tumor and/or prevent disease progression. In several embodiments, dosing is, for example, 3 doses of about 0.5 x 109 NK cells or about 2.5 x 109 NK cells administered over about 21 to 28 days. In several embodiments, dosing is, for example, 3 doses of about 1.0 x 109 NK cells or about 1.5 x 109 NK cells administered over about 21 to 28 days. In several embodiments, a dosing cycle comprises 3 doses of about 1.0 x 109 NK cells administered over about 21 to 28 days. In several embodiments, a dosing cycle comprises 3 doses of about 1.5 x 109 NK cells administered over about 21 to 28 days. In several embodiments, a dosing cycle comprises 3 doses of about 2.0 x 109 NK cells administered over about 21 to 28 days.
[00299] In several embodiments, a lymphodepletion process is performed prior to the first dose. In several embodiments, the administration of engineered NK cells is preceded by one or more preparatory treatments. In several embodiments, the administration of engineered NK cells is preceded by a lymphodepleting therapy (also referred to as “lymphodepletion”). In several embodiments, a combination of chemotherapeutic agents is used for lymphodepletion. In several embodiments, a single chemotherapeutic agent is used for lymphodepletion. In several embodiments, wherein a combination of chemotherapeutic agents is used, agents with different mechanisms of actions are optionally used. In several embodiments, different classes of agents are optionally used. In several embodiments, an antimetabolic agent is used. In several embodiments, the antimetabolic agent inhibits and/or prevents cell replication. In several embodiments, the antimetabolic agent is an altered nucleotide that disrupts DNA replication, making it effective in targeting rapidly dividing tumor cells.
[00300] In several embodiments, cytosine arabinoside (Ara-C) is used. In several embodiments, a dose of between about 0.2 - about 10 g/nr Ara-C is administered, including doses of about 0.2 g/m2, about 0.5 g/m2, about 1.0 g/m2, about 1.5 g/nr, about 2.0 g/m2, about 2.5 g/m2, about 3.0 g/m2, about 3.5 g/m2, about 4.0 g/m2, about 5.0 g/m2, about 6.0 g/m2, about 7.0 g/m2, about 8.0 g/m2, about 9.0 g/m2, about 10.0 about 1.5 g/m2, or any dose between those listed. In several embodiments, a dose of about 2 g/m2 of Ara-C is administered. In several embodiments, the dose of Ara-C is given daily for at least about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, or about 7 days. In several embodiments, the dose of Ara-C is given daily for about 5 days. In several embodiments, if necessary, the dose can be split and given, for example, twice daily. In several embodiments, an additional agent is used in combination with the Ara-C. In several embodiments, the additional agent is also an antimetabolite. In several embodiments, the additional agent inhibits one or more of DNA polymerase alpha, ribonucleotide reductase and/or DNA primase, thus inhibiting DNA synthesis. [00301] In several embodiments, the additional agent is fludarabine. In several embodiments, a dose of between about 5.0 mg/m2 - about 200 mg/m2 fludarabine is administered, including doses of about 5.0 mg/m2, about 10.0 mg/m2, about 15.0 mg/m2, about 20.0 mg/m2, about 25.0 mg/m2, about 30.0 mg/m2, about 35.0 mg/m2, about 40.0 mg/m2, about 45.0 mg/m2, about 50.0 mg/m2, about 60.0 mg/m2, about 70.0 mg/m2, about 80.0 mg/m2, about 90.0 mg/m2, about 100.0 mg/m2, about 125.0 mg/m2, about 150.0 mg/m2, about 175.0 mg/m2, about 200.0 mg/m2, or any dose between those listed. In several embodiments, a dose of about 30 mg/m2 of fludarabine is administered. In several embodiments, the dose of fludarabine is given daily for at least about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, or about 7 days. In several embodiments, the dose of fludarabine is given daily for at about 3 days. In several embodiments, the dose of fludarabine is given daily for at about 5 days. In several embodiments, if necessary, the dose can be split and given, for example, twice daily.
[00302] In several embodiments, a combination of fludarabine and Ara-C is used with a daily dose of fludarabine of between about 20 mg/m2 and 40 mg/m2 and a daily dose of Ara-C of between about 1.5 g/m2 and 2.5 g/m2. In several embodiments, a combination of fludarabine and Ara-C is used with a daily dose of fludarabine of about 30 mg/m2 and a daily dose of Ara-C of about 2 g/m2. In several embodiments, the combination of fludarabine and Ara-C (or any other agent or agents as disclosed herein) is administered for about 5 days, with the administration started about 7 days prior to the first administration of the engineered NK cells (for example day - 7 to day -3). In several embodiments, lymphodepletion is started at day -5 prior to administration of engineered NK cells. In several embodiments, this combination advantageously functions not only as a lymphodepletion regimen, but as an anti-cancer agent as well (in addition to the engineered NK cells). In several embodiments, the lymphodepletion regimen works synergistically with the engineered NK cells to provide effect reduction and/or elimination of cancerous cells.
[00303] In several embodiments, the additional agent is cyclophosphamide. In several embodiments, a dose of between about 100 mg/m2 - about 100 mg/m2 fludarabine is administered, including doses of about 100.0 mg/m2, about 200 mg/m2, about 300 mg/m2, about 400 mg/m2, about 500 mg/m2, about 600 mg/m2, about 700 mg/m2, about 800 mg/m2, about 900 mg/m2, about 1000 mg/m2, or any dose between those listed. In several embodiments, a dose of about 300 mg/m2 of cyclophosphamide is administered. In several embodiments, a dose of about 500 mg/m2 of cyclophosphamide is administered. In several embodiments, a dose of about 600 mg/m2 of cyclophosphamide is administered. In several embodiments, a dose of about 700 mg/m2 of cyclophosphamide is administered. In several embodiments, a dose of about 800 mg/m2 of cyclophosphamide is administered. In several embodiments, a dose of about 900 mg/m2 of cyclophosphamide is administered. In several embodiments, a dose of about 1000 mg/m2 of cyclophosphamide is administered. In several embodiments, the dose of cyclophosphamide is given once. In several embodiments, the dose of cyclophosphamide is given once between about 5 days and about 2 days prior to administration of the engineered NK cells. In several embodiments, the dose of cyclophosphamide is given once between about 5 days prior to administration of the engineered NK cells. In several embodiments, the dose of cyclophosphamide is given once between about 4 days prior to administration of the engineered NK cells. In several embodiments, the dose of cyclophosphamide is given once between about 3 days prior to administration of the engineered NK cells. In several embodiments, the dose of cyclophosphamide is given once between about 2 days prior to administration of the engineered NK cells.
[00304] In several embodiments, the dose of cyclophosphamide is given daily for at least about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, or about 7 days. In several embodiments, the dose of cyclophosphamide is given daily for about 3 days. In several embodiments, if necessary, the dose can be split and given, for example, twice daily.
[00305] In several embodiments, a combination of fludarabine and cyclophosphamide is used. In several embodiments, cyclophosphamide (300 mg/m2) and fludarabine (30mg/m2) are administered daily for 3 days. In several embodiments, cyclophosphamide (500 mg/m2) and fludarabine (30mg/m2) are administered daily for 3 days. In some embodiments, fludarabine and cyclophosphamide are each administered daily 5 days, 4 days, and 3 days prior to administration of the engineered NK cells.
[00306] In certain embodiments, a dose of a genetically engineered cell(s) described herein or composition thereof is administered to a subject every day, every other day, every couple of days, every third day, once a week, twice a week, three times a week, or once every two weeks. In other embodiments, two, three or four doses of a genetically engineered cell(s) described herein or composition thereof is administered to a subject every day, every couple of days, every third day, once a week or once every two weeks. In some embodiments, a dose(s) of a genetically engineered cell(s) described herein or composition thereof is administered for 2 days, 3 days, 5 days, 7 days, 14 days, or 21 days. In certain embodiments, a dose of a genetically engineered cell(s) described herein or composition thereof is administered for 1 month, 1.5 months, 2 months, 2.5 months, 3 months, 4 months, 5 months, 6 months or more.
[00307] In several embodiments, a subject is subject to lymphodepletion at least one time prior to administration of genetically engineered cells as disclosed herein. In several embodiments, lymphodepletion is performed before one or more additional doses of engineered cells are administered. In several embodiments, a dosing cycle is used that comprises lymphodepletion followed by at least two doses of engineered cells as disclosed herein, with the two doses separated by a time interval. In several embodiments, the time interval is 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or more days (including intervals falling between the time marking a price interval since the last administration, e.g., 84 hours, or 3.5 days). In several embodiments, the dosing cycle itself is approximately 14, 21, 28, 35, 42 or more days. In several embodiments, three doses are administered, each about 2-5 days apart from each other. In several embodiments, all three doses are administered within about 5-7 days. In several embodiments, a subject receives a first dose on day 0 of the cycle, a second dose on day 3 of the cycle and a third dose on day 7 of the cycle. In several embodiments, three doses are administered, ~1 week apart from each other. In several embodiments, two doses are administered ~1 week apart from one another. In several embodiments, a subject receives a first dose on day 0 of the cycle, a second dose on day 7 of the cycle and a third dose on day 14 of the cycle. In several such embodiments, a 28 day cycle is used with primary outcome measures evaluated at day 28 (see e.g., Figure 3A). In several embodiments, a subject receives a first dose on day 0 of the cycle and a second dose on day 7 of the cycle. In several such embodiments, a 28 day cycle is used with primary outcome measures evaluated at day 28 (See e.g., Figure 3B). In several embodiments, lymphodepletion is performed prior to the inception of each dosing cycle, if subsequent dosing cycles are required (e.g., the subject requires further treatment). For example, in several embodiments, a subject undergoes lymphodepletion, receives a plurality of doses of engineered cells according to a cycle, is evaluated at the end of the cycle time and, if deemed necessary undergoes a second lymphodepletion followed by a second dosing cycle. In such embodiments where multiple dosing cycles are used, a first and a second dosing cycle need not be the same (e.g., a first cycle may have 2 doses, while a second uses three doses). Depending on the subject 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more dosing cycles are performed.
[00308] In several embodiments, an additional therapeutic agent is administered at least once during the lymphodepletion and/or the dosing cycle. In some embodiments, the cells, binding molecules, or recombinant receptors are administered as part of a combination treatment, such as simultaneously with or sequentially with, in any order, another therapeutic intervention, such as another antibody or engineered cell or receptor or agent, such as a cytotoxic or therapeutic agent.
[00309] The cells, binding molecules and/or recombinant receptors in some embodiments are co- administered with one or more additional therapeutic agents or in connection with another therapeutic intervention, either simultaneously or sequentially in any order. In some contexts, the cells are co-administered with another therapy sufficiently close in time such that the cell populations enhance the effect of one or more additional therapeutic agents, or vice versa. In some embodiments, the cells, binding molecules and/or recombinant receptors are administered prior to the one or more additional therapeutic agents. In some embodiments, the cells, binding molecules and/or recombinant receptors are administered after to the one or more additional therapeutic agents.
B. Combination Therapy [00310] Also provided are methods of combination therapy that includes administration and uses, such as therapeutic and prophylactic uses, of the CD38-binding recombinant receptors (e.g., CARs), engineered cells expressing the recombinant receptors (e.g., CARs), plurality of engineered cells expressing the receptors, and/or compositions comprising the same.
[00311] In some embodiments, the CD38-binding recombinant receptor (e.g., chimeric antigen receptor) and/or engineered cells expressing said molecules (e.g., recombinant receptor) described herein are administered as part of a combination treatment or combination therapy, such as simultaneously with, sequentially with or intermittently with, in any order, one or more additional therapeutic intervention. In some embodiments, the one or more additional therapeutic intervention includes, for example, an antibody, an engineered cell, a receptor and/or an agent, such as a cell expressing a recombinant receptor, and/or cytotoxic or therapeutic agent, e.g., a chemotherapeutic agent. In some embodiments, the combination therapy includes administration of one or more additional agents, therapies and/or treatments, e.g., any of the additional agents, therapy and/or treatments described herein. In some embodiments, the combination treatment or combination therapy includes an additional treatment, such as a surgical treatment, transplant, and/or radiation therapy. Also provided are methods of combination treatment or combination therapy that includes CD38-binding recombinant receptors (e.g., CARs), cells and/or compositions described herein and one or more additional therapeutic interventions.
[00312] In some embodiments, the additional therapy, treatment or agent includes chemotherapy, radiation therapy, surgery, transplantation, adoptive cell therapy, antibodies, cytotoxic agents, chemotherapeutic agents, cytokines, growth inhibitory agents, anti-hormonal agents, kinase inhibitors, anti-angiogenic agents, cardioprotectants, immunostimulatory agents, immunosuppressive agents, immune checkpoint inhibitors, antibiotics, angiogenesis inhibitors, metabolic modulators or other therapeutic agents or any combination thereof. In some embodiments, the additional agent is a protein, a peptide, a nucleic acid, a small molecule agent, a cell, a toxin, a lipid, a carbohydrate or combinations thereof, or any other type of therapeutic agent, e.g. radiation. In some embodiments, the additional therapy, agent or treatment includes surgery, chemotherapy, radiation therapy, transplantation, administration of cells expressing a recombinant receptor, e.g., CAR, kinase inhibitor, immune checkpoint inhibitor, mTOR pathway inhibitor, immunosuppressive agents, immunomodulators, antibodies, immunoablative agents, antibodies and/or antigen binding fragments thereof, antibody conjugates, other antibody therapies, cytotoxins, steroids, cytokines, peptide vaccines, hormone therapy, antimetabolites, metabolic modulators, alkylating agents, anthracyclines, vinca alkaloids, proteasome inhibitors, protein kinase inhibitors, and/or other types of immunotherapy. In some embodiments, the additional agent or treatment is bone marrow transplantation, T cell ablative therapy using chemotherapy agents such as, fludarabine, external-beam radiation therapy (XRT), cyclophosphamide, and/or antibody therapy.
[00313] In some embodiments, the additional agent is a chemotherapeutic agent. In several embodiments, the chemotherapeutic agent comprises an antimetabolite, an alkylating agent, a topoisomerase inhibitor, a mitotic inhibitor, an antibiotic, a protein kinase inhibitor, a proteasome inhibitor, an inhibitor of poly (ADP-ribose) polymerase (PARP), or a combination thereof.
[00314] In some embodiments, the chemotherapeutic agent comprises an antimetabolite. In some embodiments, the antimetabolite comprises methotrexate, pemetrexed, cytarabine, 5 -fluorouracil (5-FU), capecitabine, gemcitabine, 6-mercaptopurine (6-MP), azathioprine, fludarabine, cladribine, hydroxyurea, or any combination thereof. In some embodiments, the therapeutic agent comprises gemcitabine. In some embodiments, the therapeutic agent comprises fluorouracil and leucovorin (FOL). In some embodiments, the therapeutic agent comprises capecitabine and ovaliplatin (CAPOX).
[00315] In some embodiments, the chemotherapeutic agent comprises an alkylating agent. In some embodiments, the alkylating agent comprises cyclophosphamide, ifosfamide, chlorambucil, melphalan, temozolomide, carmustine, lomustine, streptozocin, busulfan, procarbazine, cisplatin, carboplatin, oxaliplatin, or any combination thereof. In some embodiments, the therapeutic agent comprises cisplatin. In some embodiments, the therapeutic agent comprises bendamustine. In some embodiments, the therapeutic agent comprises cyclophosphamide. In some embodiments, the therapeutic agent comprises dacarbazine. In some embodiments, the therapeutic agent comprises procarbazine. In some embodiments, the therapeutic agent comprises capecitabine and ovaliplatin (CAPOX).
[00316] In In some embodiments, the chemotherapeutic agent comprises a topoisomerase inhibitor. In some embodiments, the topoisomerase inhibitor comprises irinotecan, topotecan, etoposide, or any combination thereof. In some embodiments, the therapeutic agent comprises etoposide.
[00317] In some embodiments, the chemotherapeutic agent comprises a mitotic inhibitor. In some embodiments, the mitotic inhibitor comprises vincristine, vinblastine, vinorelbine, docetaxel, paclitaxel, eribulin, izabepilone, epothilone, or any combination thereof. In some embodiments, the therapeutic agent comprises vincristine. In some embodiments, the therapeutic agent comprises vinblastine. In some embodiments, the therapeutic agent comprises paclitaxel. In some embodiments, the therapeutic agent comprises docetaxel.
[00318] In some embodiments, the chemotherapeutic agent comprises an antibiotic. In some embodiments, the antibiotic comprises bleomycin; actinomycin D; an anthracycline, optionally doxorubicin, daunorubicin, or idarubicin, mitomycin, or any combination thereof. In some embodiments, the therapeutic agent comprises bleomycin. In some embodiments, the therapeutic agent comprises an anthracy cline. In some embodiments, the therapeutic agent comprises doxorubicin. In some embodiments, the therapeutic agent comprises daunorubicin.
[00319] In some embodiments, the chemotherapeutic agent comprises a protein kinase inhibitor. In some embodiments, the protein kinase inhibitor comprises an inhibitor of BCR-ABL, c-KIT, EGFR, VEGF, ALK, BRAF, MAPK, MEK, mTOR, BTK, JAK, CDK, or any combination thereof. In some embodiments, the protein kinase inhibitor comprises an inhibitor of BCR-ABL. In some embodiments, the protein kinase inhibitor comprises an inhibitor of BCL2 (e.g., venetoclax). In some embodiments, the protein kinase inhibitor comprises an inhibitor of c- Kit. In some embodiments, the protein kinase inhibitor comprises an inhibitor of EGFR (e.g., cetuximab). In some embodiments, the protein kinase inhibitor comprises an inhibitor of BRAF (e.g., vemurafenib, dabrafenib or encorafenib). In some embodiments, the protein kinase inhibitor comprises an inhibitor of MAPK (e.g., vemurafenib, dabrafenib, or trametinib). In some embodiments, the protein kinase inhibitor comprises an inhibitor of MEK (e.g., trametinib, binimetinib, selumetinib, or cobimetinib). In some embodiments, the protein kinase inhibitor comprises an inhibitor of mTOR (e.g., sirolimus, everolimus, or temsirolimus). In some embodiments, the protein kinase inhibitor comprises an inhibitor of BTK (e.g., ibrutinib). In some embodiments, the protein kinase inhibitor comprises an inhibitor of JAK. In some embodiments, the protein kinase inhibitor comprises an inhibitor of CDK.
[00320] In some embodiments, the chemotherapeutic agent comprises a proteasome inhibitor. In some embodiments, the proteasome inhibitor comprises bortezomib, carfilzomib, ixazomib, or any combination thereof. In some embodiments, the proteasome inhibitor comprises bortezomib. In some embodiments, the proteasome inhibitor comprises carfilzomib. In some embodiments, the proteasome inhibitor comprises ixazomib.
[00321] In some embodiments, the chemotherapeutic agent comprises a PARP inhibitor. In some embodiments, the PARP inhibitor comprises olaparib. In some embodiments, the therapeutic agent comprises niraparib. In some embodiments, the PARP inhibitor comprises rucaparib.
[00322] In several embodiments, the additional agent is a hormone therapy or a therapeutic agent that disrupts or modifies a hormone pathway. In some embodiments, the additional agent comprises tamoxifen. In several embodiments, the additional agent comprises hyperthermia. In several embodiments, the additional agent is an additional form of immunotherapy. In several embodiments, the additional agent comprises photodynamic therapy (PDT). In several embodiments, the additional agent comprises radiation therapy. In several embodiments, the additional agent comprises transplanted stem cells.
[00323] In some embodiments, the cells, CD38-binding recombinant receptors and/or compositions, e.g., CAR-expressing cells, are administered in combination with other engineered cells, e.g., other CAR-expressing cells. In some embodiments, the additional therapy or treatment is cell therapy, e.g., adoptive cell therapy. In some embodiments, the additional therapy includes administration of engineered cells, e.g., additional CAR-expressing cell. In some embodiments, the additional engineered cell is a CAR-expressing cell that expresses the same or different recombinant receptor as the engineered cells provided herein, e.g., anti-CD38 CAR-expressing cells. In some embodiments, the recombinant receptor, e.g., CAR, expressed on the additional engineered cell, recognizes a different antigen, e.g., BCMA, or a different epitope of CD38. In some embodiments, the additional agent is a cell engineered to express a recombinant receptor, e.g., CAR, targeting a second or additional antigen expressed in multiple myeloma. In some embodiments, the recombinant receptor, e.g., CAR, expressed on the additional engineered cell recognizes BCMA. In some embodiments, the recombinant receptor, e.g., CAR, expressed on the additional engineered cell recognizes a different epitope of CD38. In such embodiments, the additional engineered cell is administered prior to, concurrently with, or after administration (e.g., infusion) of the CAR-expressing cells described herein. In some embodiments, the additional engineered cell is allogeneic to the subject being treated. In some embodiments, the additional engineered cell is autologous to the subject being treated. In some embodiments, the CAR molecules expressing by the different engineered cells may have the same or a different primary intracellular signaling domain, the same or different costimulatory signaling domains, or the same number or a different number of costimulatory signaling domains. In some embodiments, the additional agent includes any of the cells or plurality of cells described herein.
[00324] In several embodiments, the therapeutic agent is an NK cell engager (e.g., a molecule that binds both an antigen expressed by cells of the cancer and an antigen expressed by NK cells). In several embodiments, the NK cell engager binds to an activating receptor on an NK cell and an antigen expressed by cells of the cancer. In some embodiments, the activating receptor on the NK cell is selected from the group consisting of CD 16, NKp30, NKp46, NKG2D, and any combination thereof.
[00325] In some embodiments, the additional agent includes an immune checkpoint inhibitor. Immune checkpoint inhibitors include any agent that blocks or inhibits in a statistically significant manner, the inhibitory pathways of the immune system. Such inhibitors may include small molecule inhibitors or may include antibodies, or antigen binding fragments thereof, that bind to and block or inhibit immune checkpoint receptors, ligands and/or receptor-ligand interaction. In some embodiments, modulation, enhancement and/or stimulation of particular receptors can overcome immune checkpoint pathway components. Illustrative immune checkpoint molecules that may be targeted for blocking, inhibition, modulation, enhancement and/or stimulation include, but are not limited to, PD-1 (CD279), PD-L1 (CD274, B7-H1), CTLA-4, LAG-3 (CD223), TIM-3, 4-1BB (CD137), 4-1BBL (CD137L), GITR (TNFRSF18, AITR), CD40, 0X40 (CD134, TNFRSF4), B7-H3, B7-H4, B7H3, B7H4, VISTA, KIR, 2B4, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), TIGIT, and LAIR1. [00326] In some embodiments, the additional agent is a CD19 inhibitor, e.g., an anti- CD19 antibody (e.g., an anti-CD19 mono- or bi-specific antibody) or a fragment thereof, antibody-drug conjugate (ADC), or engineered toxin body (ETB). In some embodiments, the CD19 inhibitor is an anti-CD19 antibody. In some embodiments, the CD19 inhibitor is an ADC. In some embodiments, the CD 19 inhibitor is an ETB. In some embodiments, the CD 19 inhibitor is a small molecule.
[00327] In some embodiments, the additional agent is a CD20 inhibitor, e.g., an anti- CD20 antibody (e.g., an anti-CD20 mono- or bi-specific antibody) or a fragment thereof, antibody-drug conjugate (ADC), or engineered toxin body (ETB). In some embodiments, the CD20 inhibitor is an anti-CD20 antibody. Non-limiting examples of anti-CD20 antibodies include rituximab (RITUXAN®) and obinutuzumab (GAZYVA®). In some embodiments, the additional agent is or includes rituximab. In some embodiments, the additional agent is or includes obinutuzumab. In some embodiments, the CD20 inhibitor is an ADC. In some embodiments, the CD20 inhibitor is an ETB. In some embodiments, the CD20 inhibitor is a small molecule.
[00328] In some embodiments, the additional agent is a CD22 inhibitor, e.g., an anti- CD22 antibody (e.g., an anti-CD22 mono- or bi-specific antibody) or a fragment thereof, antibody-drug conjugate (ADC), or engineered toxin body (ETB). In some embodiments, the CD22 inhibitor is an ADC. In some embodiments, the CD22 inhibitor is an ETB. In some embodiments, the CD22 inhibitor is a small molecule.
[00329] In some embodiments, the additional agent is an EGFR inhibitor, e.g., an anti-EGFR antibody (e.g., an anti-EGFR mono- or bi-specific antibody) or a fragment thereof, antibody-drug conjugate (ADC), or engineered toxin body (ETB). In some embodiments, the EGFR inhibitor is an anti-EGFR antibody. Non-limiting examples of anti-EGFR antibodies include cetuximab, panitumumab (VECTIBIX®), nimotuzumab, and necitumumab (PORTRAZZA™). In some embodiments, the additional agent is or includes cetuximab. In some embodiments, the EGFR inhibitor is an ADC. In some embodiments, the EGFR inhibitor is an ETB. In some embodiments, the EGFR inhibitor is a small molecule.
[00330] In some embodiments, the additional agent a BCMA inhibitor, e.g., an anti- BCMA antibody (e.g., an anti-BCMA mono- or bi-specific antibody) or a fragment thereof, antibody-drug conjugate (ADC), or engineered toxin body (ETB). Non-limiting examples of anti- BCMA antibodies include teclistamab-cqyv (TECVAYLI™), AMG 420 (also known as BI 836909), PF-06863135 (also known as PF-3135), JNJ-957, REGN5458, TNB-383B, and CC- 93269. In some embodiments, the BCMA inhibitor is or includes teclistamab-cqyv. In some embodiments, the BCMA inhibitor is an ADC. Exemplary ADCs include belantamab mafodotin (BLEFREP®), GSK2857916, HDP-101, and MEDI2228. See Shah et al., Leukemia (2020) 34:985-1005. In some embodiments, the BCMA inhibitor is an ETB. In some embodiments, the BCMA inhibitor is a small molecule. [00331] In some embodiments, the additional agent is a SLAMF7 inhibitor, e.g., an anti-SLAMF7 antibody (e.g., an anti-SLAMF7 mono- or bi-specific antibody) or a fragment thereof, antibody-drug conjugate (ADC), or engineered toxin body (ETB). Non-limiting examples of anti-SLAMF7 antibodies include elotuzumab (EMPLICITI®). In some embodiments, the SLAMF7 inhibitor is or includes elotuzumab. In some embodiments, the SLAMF7 inhibitor is an ADC. In some embodiments, the SLAMF7 inhibitor is an ETB. In some embodiments, the SLAMF7 inhibitor is a small molecule.
[00332] In some embodiments, the additional agent is a CD38 inhibitor, e.g., an anti- CD38 antibody (e.g., an anti-CD38 mono- or bi-specific antibody) or a fragment thereof, antibody-drug conjugate (ADC), or engineered toxin body (ETB). Non-limiting examples of anti- CD38 antibodies include daratumumab (e.g., DARZALEX® or DARZALEX Faspro®), isatuximab (SARCLISA®), MOR202, GBR 1342 (also known as ISB-1342), TAK-079, and TAK-169. See Gozzetti et al., Hum Vaccin Immunother (2022) 18(5):2052658. In some embodiments, the anti-CD38 antibody is or includes daratumumab. In some embodiments, daratumumab is administered at a dose of between about 10 mg/kg and about 25 mg/kg or between about 15 mg/kg and about 20 mg/kg (e.g., about 16 mg/kg). In some embodiments, a dose of daratumumab is administered about every week, about every two weeks, about every three weeks, or about every four weeks. In some embodiments, daratumumab is administered intravenously. In some embodiments, daratumumab is administered at a dose of between about 1,500 mg and about 3,000 mg (e.g, about 1,800 mg). In some embodiments, the dose of daratumumab is administered in combination with a dose of with hyaluronidase. In some embodiments, hyaluronidase is administered at a dose of between about 20,000 units and about 40,000 units (e.g., about 30,000) units. In some embodiments, a dose of daratumumab and a dose of hyaluronidase and administered in combination about every week, about every two weeks, about every three weeks, or about every four weeks. In some embodiments, daratumumab and hyaluronidase are administered in combination (e.g., as a single formulation or composition) subcutaneously. In some embodiments, the anti-CD38 antibody is or includes isatuximab. In some embodiments, isatuximab is administered at a dose of between about 5 mg/kg and about 15 mg/kg (e.g., about 10 mg/kg). In some embodiments, a dose of isatuximab is administered about every week or about every two weeks. In some embodiments, isatuximab is administered intravenously. In some embodiments, the CD38 inhibitor is a small molecule.
[00333] In some embodiments, when the additional agent is a CD38 inhibitor, e.g., an anti-CD38 antibody, the engineered cells are genetically edited to reduce transcription of the CD38 gene and/or expression of CD38 protein, such as described in Section I.A.ii. In some embodiments, the genetically engineered cells are “knocked out” (KO) for CD38, such as by a CRISPR/Cas system (e.g., a CD38-targeting ribonucleoprotein (RNP) complex). In some aspects, the genetic edit is carried out by using a DNA binding protein or DNA-binding nucleic acid, which specifically binds to or hybridizes to the CD38 gene at a region targeted for disruption. In some aspects, the protein or nucleic acid is coupled to or complexed with a nuclease, such as in a chimeric or fusion protein. For example, in some embodiments, the disruption is effected using a fusion comprising a DNA-targeting protein and a nuclease, such as a Zinc Finger Nuclease (ZFN) or TAL-effector nuclease (TALEN), or an RNA-guided nuclease such as a clustered regularly interspersed short palindromic nucleic acid (CRISPR)-Cas system, such as CRISPR-Cas9 system, specific for the gene being disrupted.
[00334] In some embodiments, the additional agent is an immunomodulatory agent (IMiD). In some embodiments, the immunomodulatory agent comprises lenalidomide, pomalidomide, thalidomide, or any combination thereof. In some embodiments, the immunomodulatory agent comprises lenalidomide. In some embodiments, the immunomodulatory agent comprises pomalidomide. In some embodiments, the immunomodulatory agent comprises thalidomide.
[00335] In some embodiments, the additional agent is or comprises a steroid. In some embodiments, the steroid is a corticosteroid. In some embodiments, the steroid is a glucocorticoid. In some embodiments, the steroid comprises dexamethasone or prednisone. In some embodiments, the steroid is or comprises dexamethasone. In some embodiments, the steroid is or comprises prednisone.
[00336] Any of the additional agents described herein can be prepared and administered as combination therapy with the CD38-binding recombinant receptor (e.g., chimeric antigen receptor) and/or engineered cells expressing said molecules (e.g., recombinant receptor) described herein, such as in pharmaceutical compositions comprising one or more agents of the combination therapy and a pharmaceutically acceptable carrier, such as any described herein. In some embodiments, the CD38-binding recombinant receptor (e.g., chimeric antigen receptor), engineered cells expressing said molecules (e.g., recombinant receptor), plurality of engineered cells expressing said molecules (e.g., recombinant receptor) can be administered simultaneously, concurrently or sequentially, in any order with the additional agents, therapy or treatment, wherein such administration provides therapeutically effective levels each of the agents in the body of the subject. In some embodiments, the additional agent can be co-administered with the CD38-binding recombinant receptors, cells and/or compositions described herein, for example, as part of the same pharmaceutical composition or using the same method of delivery. In some embodiments, the additional agent is administered simultaneously with the CD38-binding recombinant receptors, cells and/or compositions described herein, but in separate compositions. In some embodiments, the additional agent is an additional engineered cell, e.g., cell engineered to express a different recombinant receptor, and is administered in the same composition or in a separate composition. In some embodiments, the additional agent is incubated with the engineered cell, e.g., CAR-expressing cells, prior to administration of the cells. [00337] In some examples, the additional agent is administered subsequent to or prior to the administration of the CD38-binding recombinant receptors, cells and/or compositions described herein, separated by a selected time period. In some examples, the time period is 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 1 week, 2 weeks, 3 weeks, 1 month, 2 months, or 3 months. In some examples, the additional agent is administered multiple times and/or the CD38-binding recombinant receptors, cells and/or compositions described herein, is administered multiple times. For example, in some embodiments, the additional agent is administered prior to the CD38- binding recombinant receptors, cells and/or compositions described herein, e.g., two weeks, 12 days, 10 days, 8 days, one week, 6 days, 5 days, 4 days, 3 days, 2 days or 1 day before the administration. For example, in some embodiments, the additional agent is administered after the CD38-binding recombinant receptors, cells and/or compositions described herein, e.g., two weeks, 12 days, 10 days, 8 days, one week, 6 days, 5 days, 4 days, 3 days, 2 days or 1 day after the administration.
[00338] The dose of the additional agent can be any therapeutically effective amount, e.g., any dose amount described herein, and the appropriate dosage of the additional agent may depend on the type of disease to be treated, the type, dose and/or frequency of the recombinant receptor, cell and/or composition administered, the severity and course of the disease, whether the recombinant receptor, cell and/or composition is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the recombinant receptor, cell and/or composition, and the discretion of the attending physician. The recombinant receptor, cell and/or composition and/or the additional agent and/or therapy can be administered to the patient at one time, repeated or administered over a series of treatments.
IV. Articles of Manufacture and Kits
[00339] Also provided are articles of manufacture or kits containing the provided antibodies, recombinant receptors (e.g., CARs), genetically engineered cells, and/or compositions comprising the same. The articles of manufacture may include a container and a label or package insert on or associated with the container. Suitable containers include, for example, bottles, vials, syringes, test tubes, IV solution bags, etc. The containers may be formed from a variety of materials such as glass or plastic. In some embodiments, the container has a sterile access port. Non-limiting examples of containers include intravenous solution bags and vials, including those with stoppers pierceable by a needle for injection. The article of manufacture or kit may further include a package insert indicating that the composition can be used to treat a particular condition such as a condition described herein (e.g., cancer). Alternatively, or additionally, the article of manufacture or kit may further include another or the same container comprising a pharmaceutically-acceptable buffer. It may further include other materials such as other buffers, diluents, filters, needles, and/or syringes. [00340] The label or package insert may indicate that the composition is used for treating aCD38-expressing disease, disorder, or condition in an individual. The label or a package insert, which is on or associated with the container, may indicate directions for reconstitution and/or use of the formulation. The label or package insert may further indicate that the formulation is useful or intended for subcutaneous, intravenous, or other modes of administration for treating or preventing a CD38-expressing disease, disorder, or condition in an individual.
[00341] The container in some embodiments holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the condition. The article of manufacture or kit may include (a) a first container with a composition contained therein (i.e., first medicament), wherein the composition includes the antibody (e.g., anti-CD38 antibody) or antigen-binding fragment thereof or recombinant receptor (e.g., CAR); and (b) a second container with a composition contained therein (i.e., second medicament), wherein the composition includes a further agent, such as a cytotoxic or otherwise therapeutic agent, and which article or kit further comprises instructions on the label or package insert for treating the subject with the second medicament, in an effective amount.
DEFINITIONS
[00342] The terms described below, or elsewhere herein, shall be understood to have their ordinary meaning and shall also be understood to have the meanings specifically described herein, unless otherwise specifically indicated.
[00343] The term "Fc region" herein is used to define a C-terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region. The term includes native sequence Fc regions and variant Fc regions. In one embodiment, a human IgG heavy chain Fc region extends from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain. However, the C-terminal lysine (Lys447) of the Fc region may or may not be present. Unless otherwise specified herein, numbering of amino acid residues in the Fc region or constant region is according to the EU numbering system, also called the EU index, as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD, 1991.
[00344] The terms "full length antibody," "intact antibody," and "whole antibody" are used herein interchangeably to refer to an antibody having a structure substantially similar to a native antibody structure or having heavy chains that contain an Fc region as defined herein.
[00345] An "isolated" antibody is one which has been separated from a component of its natural environment. In some embodiments, an antibody is purified to greater than 95% or 99% purity as determined by, for example, electrophoretic (e.g. , SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatographic (e.g. , ion exchange or reverse phase HPLC). [00346] An "isolated" nucleic acid refers to a nucleic acid molecule that has been separated from a component of its natural environment. An isolated nucleic acid includes a nucleic acid molecule contained in cells that ordinarily contain the nucleic acid molecule, but the nucleic acid molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location.
[00347] "Isolated nucleic acid encoding an anti-CD38 antibody" refers to one or more nucleic acid molecules encoding antibody heavy and light chains (or fragments thereof), including such nucleic acid molecule(s) in a single vector or separate vectors, and such nucleic acid molecule(s) present at one or more locations in a host cell.
[00348] The terms "host cell," "host cell line," and "host cell culture" are used interchangeably and refer to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells. Host cells include "transformants" and "transformed cells," which include the primary transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein.
[00349] The terms "polypeptide" and "protein" are used interchangeably to refer to a polymer of amino acid residues, and are not limited to a minimum length. Polypeptides, including the antibodies and antibody chains and other peptides, e.g. , linkers and CD38-binding peptides, may include amino acid residues including natural and/or non-natural amino acid residues. The terms also include post-expression modifications of the polypeptide, for example, glycosylation, sialylation, acetylation, phosphorylation, and the like. In some aspects, the polypeptides may contain modifications with respect to a native or natural sequence, as long as the protein maintains the desired activity. These modifications may be deliberate, as through site-directed mutagenesis, or may be accidental, such as through mutations of hosts which produce the proteins or errors due to PCR amplification.
[00350] As used herein, "percent (%) amino acid sequence identity" and "percent identity" and "sequence identity" when used with respect to an amino acid sequence (reference polypeptide sequence) is defined as the percentage of amino acid residues in a candidate sequence (e.g. , the subject antibody or fragment) that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
[00351] An amino acid substitution may include replacement of one amino acid in a polypeptide with another amino acid. Amino acid substitutions may be introduced into a binding molecule, e.g. , antibody, of interest and the products screened for a desired activity, e.g., retained/improved antigen binding, or decreased immunogenicity.
[00352] Amino acids generally can be grouped according to the following common side- chain properties:
(1) hydrophobic: Norleucine, Met, Ala, Vai, Leu, lie;
(2) neutral hydrophilic: Cys, Ser, Thr, Asn, Gin;
(3) acidic: Asp, Glu;
(4) basic: His, Lys, Arg;
(5) residues that influence chain orientation: Gly, Pro;
(6) aromatic: Trp, Tyr, Phe.
[00353] Non-conservative amino acid substitutions will involve exchanging a member of one of these classes for another class.
[00354] The term "vector," as used herein, refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked. The term includes the vector as a selfreplicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced. Certain vectors are capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as "expression vectors."
[00355] The term "package insert" is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products.
[00356] As used herein, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. For example, "a" or "an" means "at least one" or "one or more." It is understood that aspects, embodiments, and variations described herein include "comprising," "consisting," and/or "consisting essentially of aspects, embodiments and variations.
[00357] Throughout this disclosure, various aspects of the claimed subject matter are presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the claimed subject matter. Accordingly, the description of a range should be considered to have specifically disclosed all the possible sub-ranges as well as individual numerical values within that range. For example, where a range of values is provided, it is understood that each intervening value, between the upper and lower limit of that range and any other stated or intervening value in that stated range is encompassed within the claimed subject matter. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the claimed subject matter, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the claimed subject matter. This applies regardless of the breadth of the range.
[00358] The term "about" as used herein refers to the usual error range for the respective value readily known to the skilled person in this technical field. Reference to "about" a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. For example, description referring to "about X" includes description of "X".
[00359] As used herein, a "composition" refers to any mixture of two or more products, substances, or compounds, including cells. It may be a solution, a suspension, liquid, powder, a paste, aqueous, non-aqueous or any combination thereof.
[00360] As used herein, a statement that a cell or population of cells is "positive" for a particular marker refers to the detectable presence on or in the cell of a particular marker, typically a surface marker. When referring to a surface marker, the term refers to the presence of surface expression as detected by flow cytometry, for example, by staining with an antibody that specifically binds to the marker and detecting said antibody, wherein the staining is detectable by flow cytometry at a level substantially above the staining detected carrying out the same procedure with an isotype -matched control under otherwise identical conditions and/or at a level substantially similar to that for cell known to be positive for the marker, and/or at a level substantially higher than that for a cell known to be negative for the marker.
[00361] As used herein, a statement that a cell or population of cells is "negative" for a particular marker refers to the absence of substantial detectable presence on or in the cell of a particular marker, typically a surface marker. When referring to a surface marker, the term refers to the absence of surface expression as detected by flow cytometry, for example, by staining with an antibody that specifically binds to the marker and detecting said antibody, wherein the staining is not detected by flow cytometry at a level substantially above the staining detected carrying out the same procedure with an isotype-matched control under otherwise identical conditions, and/or at a level substantially lower than that for cell known to be positive for the marker, and/or at a level substantially similar as compared to that for a cell known to be negative for the marker.
NON-LIMITING EMBODIMENTS
[00362] Among the embodiments provided herein are:
1. An anti-CD38 antibody or an antigen-binding fragment thereof, comprising: (i) a heavy chain variable region (VH) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:25, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:23; and a light chain variable region (VL) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:27, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:28, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:29;
(ii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:30, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:31, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:32; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:35;
(iii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:36, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:37, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:38; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:40;
(iv) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:41, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:42; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:43, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:45;
(v) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:46, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:48; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:49, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:50, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:51;
(vi) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:52, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:53; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:54; (vii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:55, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:56, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:57; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:58, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:59, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:60;
(viii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:61, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:62; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:63;
(ix) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:66; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:68;
(x) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:69; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:70, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:71;
(xi) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:72, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:73, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:74; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:75, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:76, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:77; or
(xii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:78, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:79, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:80; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:81.
2. An anti-CD38 antibody or an antigen- binding fragment thereof, comprising: (i) a heavy chain variable region (VH) comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 82, and a light chain variable region (VL) comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:83;
(ii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:84, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:85;
(iii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:86, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:87;
(iv) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:88, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:89;
(v) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NQ:90, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:91 ;
(vi) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:92, and a VL comprising the CDR1 , the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:93;
(vii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:94, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:95;
(viii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:96, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:97;
(ix) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:98, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:99;
(x) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 100, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 101 ;
(xi) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 102, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 103; or
(xii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 104, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 105. 3. The anti-CD38 antibody or antigen-binding fragment thereof of embodiment 1 or embodiment 2, wherein:
(i) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:82, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:83;
(ii) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:84, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:85;
(iii) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:86, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:87;
(iv) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:88, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:89;
(v) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:90, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:91;
(vi) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:92, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:93;
(vii) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:94, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:95;
(viii) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:96, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:97;
(ix) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:98, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:99;
(x) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 100, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 101;
(xi) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 102, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 103; or
(xii) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 104, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 105.
4. The anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-3, which is a chimeric antibody or an antigen-binding fragment thereof.
5. The anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-3, which comprises a human variable region framework region.
6. The anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-3 and 5, which is a fully human antibody or antigen-binding fragment thereof.
7. The anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-3, 5, and 6, wherein:
(i) the VH comprises the amino acid sequence set forth in SEQ ID NO:82, and the VL comprises the amino acid sequence set forth in SEQ ID NO:83;
(ii) the VH comprises the amino acid sequence set forth in SEQ ID NO:84, and the VL comprises the amino acid sequence set forth in SEQ ID NO:85;
(iii) the VH comprises the amino acid sequence set forth in SEQ ID NO: 86, and the VL comprises the amino acid sequence set forth in SEQ ID NO:87;
(iv) the VH comprises the amino acid sequence set forth in SEQ ID NO: 88, and the VL comprises the amino acid sequence set forth in SEQ ID NO:89;
(v) the VH comprises the amino acid sequence set forth in SEQ ID NO:90, and the VL comprises the amino acid sequence set forth in SEQ ID NO:91;
(vi) the VH comprises the amino acid sequence set forth in SEQ ID NO: 92, and the VL comprises the amino acid sequence set forth in SEQ ID NO:93;
(vii) the VH comprises the amino acid sequence set forth in SEQ ID NO:94, and the VL comprises the amino acid sequence set forth in SEQ ID NO:95;
-Il l- (viii) the VH comprises the amino acid sequence set forth in SEQ ID NO:96, and the VL comprises the amino acid sequence set forth in SEQ ID NO:97;
(ix) the VH comprises the amino acid sequence set forth in SEQ ID NO:98, and the VL comprises the amino acid sequence set forth in SEQ ID NO:99;
(x) the VH comprises the amino acid sequence set forth in SEQ ID NO: 100, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 101;
(xi) the VH comprises the amino acid sequence set forth in SEQ ID NO: 102, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 103; or
(xii) the VH comprises the amino acid sequence set forth in SEQ ID NO: 104, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 105.
8. The anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-7, wherein the antigen-binding fragment of the antibody is a Fab, a Fab’, a F(ab’)2, a Fv, or a single-chain variable fragment (scFv).
9. The anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-8, wherein the antigen-binding fragment of the antibody is an scFv.
10. The anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments
1 -9, wherein the scFv comprises a linker between the VH and VL, the linker comprising the amino acid sequence set forth in SEQ ID NO: 1 or SEQ ID NO:3.
11. The anti-CD38 antibody or antigen-binding fragment thereof of embodiment 10, wherein the linker comprises the amino acid sequence set forth in SEQ ID NO:1.
12. The anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 8-11, wherein the scFv comprises the amino acid sequence set forth in SEQ ID NO: 108, SEQ ID NO: 109, SEQ ID NO: 110, SEQ ID NO: 111, SEQ ID NO: 112, SEQ ID NO: 113, SEQ ID NO: 114, SEQ ID NO: 115, SEQ ID NO: 116, SEQ ID NO: 117, SEQ ID NO: 118, or SEQ ID NO: 119.
13. The anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-12, wherein the anti-CD38 antibody or antigen-binding fragment thereof binds to the extracellular domain (ECD) of human CD38 with a binding affinity (KD) of between about 1 nM and about 500 nM, or between about 4 nM and about 40 nM.
14. A composition comprising the anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-13.
15. The composition of embodiment 14, comprising a pharmaceutically acceptable carrier.
16. An immunoconjugate comprising the anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-13 and a therapeutic agent.
17. The immunoconjugate of embodiment 16, wherein the therapeutic agent is a cytotoxic agent.
18. A composition comprising the immunoconjugate of embodiment 16 or embodiment 17 and a pharmaceutically acceptable carrier. 19. A bispecific molecule comprising the anti-CD38 antibody or antigen-binding fragment of any one of embodiments 1-13 and a second moiety that has a binding specificity for an immune cell.
20. The bispecific molecule of embodiment 19, wherein the immune cell is a natural killer (NK) cell or a T cell.
21. The bispecific molecule of embodiment 19 or embodiment 20, wherein the second moiety binds to CD 16, NKp30, NKp46, NKG2D, or a combination thereof.
22. The bispecific molecule of embodiment 19 or embodiment 20, wherein the second moiety binds to CD3.
23. A composition comprising the bispecific molecule of any one of embodiments 19-22 and a pharmaceutically acceptable carrier.
24. A method for detecting CD38 in a cell or tissue, comprising:
(a) contacting the cell or tissue with the anti-CD38 antibody or antigen-binding fragment of any one of embodiments 1-13; and
(b) determining the amount of the antibody or antigen-binding fragment thereof bound to the cell or tissue, wherein the amount of the antibody or antigen-binding fragment thereof bound to the cell or tissue indicates the amount of CD38 in the cell or tissue.
25. Use of the anti-CD38 antibody or antigen-binding fragment of any one of embodiments 1-13 in the manufacture of a kit for detecting CD38 in a cell or tissue, comprising:
(a) contacting the cell or tissue with the anti-CD38 antibody or antigen-binding fragment of any one of embodiments 1-13; and
(b) determining the amount of the antibody or antigen-binding fragment thereof bound to the cell or tissue, wherein the amount of the antibody or antigen-binding fragment thereof bound to the cell or tissue indicates the amount of CD38 in the cell or tissue.
26. An anti-CD38 chimeric antigen receptor (CAR) comprising an extracellular antigenbinding domain, a transmembrane domain, and an intracellular signaling domain, wherein the extracellular antigen-binding domain comprises the anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-13.
27. The anti-CD38 CAR of embodiment 26, wherein the transmembrane domain is or comprises a transmembrane from CD8, CD3, CD4, or CD28.
28. The anti-CD38 CAR of embodiment 26 or embodiment 27, wherein the transmembrane domain is or comprises a transmembrane from CD8, optionally CD8alpha.
29. The anti-CD38 CAR of any one of embodiments 26-28, wherein the intracellular signaling domain comprises a CD3zeta domain.
30. The anti-CD38 CAR of any one of embodiments 26-29, wherein the intracellular signaling domain comprises a co-stimulatory signaling region. 31. The anti-CD38 CAR of embodiment 30, wherein the co-stimulatory signaling region comprises an intracellular signaling region of 0X40, CD28, 4-1BB, ICOS, or DAP10.
32. The anti-CD38 CAR of embodiment 30 or embodiment 31, wherein the co-stimulatory signaling region comprises an intracellular signaling region of 0X40 peptide.
33. The anti-CD38 CAR of embodiment, comprising a CD8alpha transmembrane domain and an intracellular signaling domain comprising a CD3zeta domain and a co-stimulatory region comprising an intracellular signaling region of 0X40 peptide.
34. The anti-CD38 CAR of any one of embodiments 26-33, wherein the CAR comprises the amino acid sequence set forth in SEQ ID NO:327, SEQ ID NO:328, SEQ ID NO:329, SEQ ID NO:330, SEQ ID NO:331, SEQ ID NO:332, SEQ ID NO:333, SEQ ID NO:334, SEQ ID NO:335, SEQ ID NO:336, SEQ ID NO:337, or SEQ ID NO:338.
35. A nucleic acid molecule encoding the anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-13 or the anti-CD38 CAR of any one of embodiments 26-
36. A vector comprising the nucleic acid molecule of embodiment 35.
37. The vector of embodiment 36, which is a viral vector.
38. The vector of embodiment 36 or embodiment 37, which is a retroviral vector.
39. A cell comprising the anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-13, the anti-CD38 CAR of any one of embodiments 26-34, the nucleic acid molecule of embodiment 35, or the vector of any one of embodiments 36-38.
40. The cell of embodiment 39, which is a natural killer (NK) cell or a T cell.
41. A natural killer (NK) cell comprising the anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-13 or the anti-CD38 CAR of any one of embodiments 26-34.
42. The cell of any one of embodiments 39-41, which is genetically edited to reduce expression of CD38, optionally wherein the expression of CD38 is reduced by at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, or at least about 80% .
43. The cell of any one of embodiments 39-42, comprising membrane-bound interleukin- 15 (mbIL15), optionally wherein the CAR and the mbIL15 are encoded bicistronically by the same nucleic acid molecule.
44. A composition comprising a plurality of the cell of any one of embodiments 39-43.
45. The composition of embodiment 44, comprising a pharmaceutically acceptable carrier.
46. The composition of embodiment 44 or embodiment 45, comprising between about 3 x 108 CAR-expressing cells and about 3 x 109 CAR-expressing cells, or between about 1 x 109 and about 2 x 109 CAR-expressing cells.
47. A method of treatment comprising administering to a subject having a CD38-expressing disease or condition an anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-13, an anti-CD38 CAR of any one of embodiments 26-34, a cell of any one of embodiments 39-43, a composition of any one of embodiments 14, 15, 18, 23, and 44-46, an immunoconjugate of embodiment 16 or embodiment 17, or a bispecific molecule of any one of embodiments 19-22.
48. A method of treatment comprising administering to a subject having a CD38-expressing disease or condition a composition comprising immune cells expressing an anti-CD38 chimeric antigen receptor (CAR) comprising an extracellular antigen-binding domain, a transmembrane domain, and an intracellular signaling domain, wherein the extracellular antigen-binding domain comprises:
(i) a heavy chain variable region (VH) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:25, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:23; and a light chain variable region (VL) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:27, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:28, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO: 29;
(ii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:30, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:31 , and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:32; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:35;
(iii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:36, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:37, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:38; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:40;
(iv) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:41, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:42; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:43, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:45;
(v) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:46, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:48; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:49, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:50, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:51;
(vi) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:52, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:53; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:54;
(vii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:55, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:56, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:57; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:58, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:59, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NQ:60;
(viii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:61, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:62; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:63;
(ix) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:66; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:68;
(x) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:69; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:70, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:71;
(xi) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:72, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:73, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:74; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:75, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:76, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:77; or
(xii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:78, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:79, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:80; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:81.
49. A method of treatment comprising administering to a subject having a CD38-expressing disease or condition a composition comprising immune cells expressing an anti-CD38 chimeric antigen receptor (CAR) comprising an extracellular antigen-binding domain, a transmembrane domain, and an intracellular signaling domain, wherein the extracellular antigen-binding domain comprises:
(i) a heavy chain variable region (VH) comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 82, and a light chain variable region (VL) comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:83;
(ii) a VH comprising the CDR1 , the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:84, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:85;
(iii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:86, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:87;
(iv) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:88, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:89;
(v) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 90, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:91;
(vi) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:92, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:93;
(vii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:94, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:95;
(viii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:96, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:97; (ix) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:98, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:99;
(x) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 100, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 101 ;
(xi) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 102, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 103; or
(xii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 104, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 105.
50. Use of an anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-13, an anti-CD38 CAR of any one of embodiments 26-34, a cell of any one of embodiments 39-43, a composition of any one of embodiments 14, 15, 18, 23, and 44-46, an immunoconjugate of embodiment 16 or embodiment 17, or a bispecific molecule of any one of embodiments 19-22 in the treatment of a subject having a CD38-expressing disease or condition.
51. Use of an anti-CD38 antibody or antigen-binding fragment thereof of any one of embodiments 1-13, an anti-CD38 CAR of any one of embodiments 26-34, a cell of any one of embodiments 39- 43, a composition of any one of embodiments 14, 15, 18, 23, and 44-46, an immunoconjugate of embodiment 16 or embodiment 17, or a bispecific molecule of any one of embodiments 19-22 in the manufacture of a medicament for the treatment of a subject having a CD38-expressing disease or condition.
52. The method of any one of embodiments 47-49 or the use of embodiment 50 or embodiment 51, wherein the CD38-expressing disease or condition is a cancer or a tumor.
53. The method of any one of embodiments 47-49 or embodiment 52 or the use of any one of embodiments 50-52, wherein the CD38-expressing disease or condition is a solid tumor.
54. The method or use of embodiment 52, wherein the cancer is selected from the group consisting of leukemia, lymphoma, myeloma, glioma, thyroid cancer, lung cancer, colorectal cancer, head and neck cancer, stomach cancer, liver cancer, pancreatic cancer, renal cancer, urothelial cancer, prostate cancer, testicular cancer, breast cancer, cervical cancer, endometrial cancer, ovarian cancer, and melanoma.
55. The method or use of embodiment 52 or embodiment 54, wherein the cancer is multiple myeloma.
56. The method of any one of claims 47-49 or the use of claim 50 or claim 51, wherein the CD38-expressing disease or condition is an autoimmune disease or disorder. 57. The method or use of claim 56, wherein the autoimmune disease or disorder is selected from the group consisting of systemic lupus erythematosus (SLE), lupus nephritis, CNS lupus, myositis, multiple sclerosis (MS), myasthenia gravis (MG), rheumatoid arthritis (RA), scleroderma, thyroid disease, diabetes, vasculitis, or any combination thereof.
58. The method of any one of embodiments 47-49 and 52-57 or the use of any one of embodiments 50-57, wherein the cells are allogeneic to the subject.
59. The method of any one of embodiments 47-49 and 52-58 or the use of any one of embodiments 50-58, wherein the subject has been treated with, or is a candidate for treatment with, an anti-CD38 antibody.
60. The method of any one of embodiments 47-49 and 52-59 or the use of any one of embodiments 50-59, wherein the treatment further comprises administering an anti-CD38 antibody to the subject.
61. The method of any one of embodiments 47-49 and 52-60 or the use of any one of embodiments 50-60, wherein the subject has relapsed following treatment with and/or is refractory to a prior line of therapy for a CD38-expressing cancer.
62. The method or use of embodiment 61, wherein the prior line of therapy is one, two, three, or four prior lines of therapy.
63. A method of producing a cell targeting CD38, comprising introducing into the cell the nucleic acid molecule of embodiment 35 or the vector of any one of embodiments 36-38.
64. The method of embodiment 63, further comprising genetically editing the cell to reduce expression of CD38.
65. The method of embodiment 64, wherein the genetic editing is carried out using a CRISPR/Cas system.
EXAMPLES
[00363] The following examples are included for illustrative purposes only and are not intended to limit the scope of the invention.
Example 1: Generation and Assessment of Anti-CD38 Antibodies
[00364] Non-limiting examples of anti-CD38 antibodies, formatted as single-chain variable fragments (scFvs), were generated and assessed for binding to CD38. In particular, fully human variable domain anti-CD38 antibodies were generated by immunizing human 1g transgenic mice with adjuvant and his-tagged human CD38 extracellular domain (ECD) protein (Aero Biosystems, Newark, DE). B lymphocytes were isolated from the lymph nodes of the immunized mice and fused with SP2/0 cells, and the resultant hybridoma cells were single cell sorted into 96- well plates. [00365] Supernatants from sorted hybridoma clones were assessed for binding to human CD38-overexpressing Jurkat cells (CD38+) and CD38 knockout (KO) Jurkat cells (CD38- ). 379 clones were identified as having at least 20-fold greater binding to CD38+ cells than to CD38- cells. Of these, the 192 clones demonstrating the best CD38-specific binding were sequenced and further analyzed for binding affinity to his-tagged human CD38 ECD via surface plasmon resonance (SPR) detection (Figure 1; 192 shown in light gray). Heatmap analysis revealed the 192 clones to be diverse in sequence. Epitope binning analysis was also performed generally as described in Abdiche et al., PLoS ONE (2014) 9(3): e92451.
[00366] 94 of the 192 clones were formatted as chimeric antigen receptors (CARs) for further testing, as described in Example 2. The sequences and CD38 binding affinity for the top 12 clones described in Example 2 are shown below in Table El. A known fully human anti- CD38 scFv “A2” was included as a reference antibody (“Ref’; described in PCT Application Nos. PCT/US2016/026545 and PCT/IB2018/058642). The majority of the non-limiting examples of anti-CD38 scFvs were observed to have a higher binding affinity for human CD38 than the anti- CD38 reference antibody, as evidenced by a lower dissociation constant (KD).
Table El. Anti-CD38 scFvs
Figure imgf000122_0001
Example 2: Generation of Chimeric Antigen Receptors (CARs) Against CD38
[00367] Polynucleotides encoding non-limiting examples of chimeric antigen receptors (CARs) were generated, each containing a human anti-CD38 scFv antigen-binding domain as described in Example 1. Specifically, each of the CAR constructs contained, from N- to C- terminus: a human anti-CD38 scFv (e.g., SEQ ID NOS: 108-120), a CD8a hinge (e.g., SEQ ID NO:6), a CD8a transmembrane domain (e.g., SEQ ID NO: 8), and an intracellular signaling region containing an 0X40 co-stimulatory region (e.g., SEQ ID NO: 14) and a CD3 zeta intracellular signaling domain (e.g., SEQ ID NO: 18). cDNA clones encoding the CARs were linked to a downstream T2A ribosomal skip element (e.g., a T2A-encoding sequence SEQ ID NO: 19, encoding SEQ ID NO:20) followed by a membrane-bound interleukin- 15 (mbIL15)- encoding sequence (e.g., a sequence encoding SEQ ID NO:23), and cloned into a retroviral expression vector.
[00368] In total, 94 anti-CD38 scFvs, as well as the reference antibody, were formatted as chimeric antigen receptors (CARs) and expressed in CD38 KO Jurkat or primary natural killer (NK cells) for analysis of CAR expression, antigen-dependent activation, and antigen-independent (tonic) activation.
Assessment of CAR Expression and Function in Jurkat Cells
[00369] To assess expression and activation of CD38-directed CARs, Jurkat cells were knocked out (KO) for CD38 expression via electroporation with a CD38-targeting Cas- gRNA ribonucleoprotein (RNP) complex and subsequently expanded in culture until transduction with a retroviral vector encoding a CD38-targeting CAR containing a non-limiting example of an anti-CD38 scFv (e.g., SEQ ID NOS: 108-120) and a FLAG tag.
[00370] CAR expression was determined by measuring the surface expression of the FLAG tag by flow cytometry. Antigen-dependent and tonic activation were assessed by incubating the CD38 CAR-expressing Jurkat cells overnight in the presence or absence of CD38- expressing MM.1R target cells, respectively, and subsequently measuring surface expression of CD69 on the Jurkat cells by flow cytometry. Antigen-dependent activation and CAR expression were compared to CD38 binding affinity.
[00371] Antigen-dependent activation (as measured by CD69 expression) and CD38 binding affinity were not observed to significantly correlate (Figure 2A). The ratio of antigendependent to tonic activation was plotted against CAR expression, and the top 44 clones were selected for further testing in the CAR format Cells incorporating CARs exhibiting higher expression tended to also exhibit greater CD38-specific activation (Figure 2B; box shows top 44 clones). Of the top 44 clones, 12 clones - each having a higher antigen-dependent:tonic activation ratio than the reference antibody - were selected for expression in primary natural killer (NK) cells and further analysis (Figure 2C: clone number indicated). The components of each of the 12 constructs are provided below in Table E2.
Table E2. CD38-Directed CARs
Figure imgf000123_0001
Figure imgf000124_0001
[00372] CAR expression was also compared to the epitope bin for each of the 44 clones. The clones were observed to bind a variety of different epitopes, which in some cases overlapped with one another (Figure 3); many of those demonstrating high expression when formatted as a CAR were observed to bind to the same epitope region of CD38 (bin 4).
Assessment of CAR Expression and Function in Primary NK Cells
[00373] NK cells were isolated by immunoaffinity-based enrichment from leukapheresis samples from two different healthy human donors. Isolated NK cells were knocked out for CD38 expression on Day 0 and transduced on Day 6 with a retroviral vector encoding a CAR as set forth in Table E2 and mbIL15. Transduced cells were allowed to expand in culture until they were analyzed for expression of CD38 and CAR on Days 11 and 14. As controls, NK cells were (1) knocked out for CD38 and subjected to mock transduction (CD38 KO); or (2) subjected to mock electroporation and mock transduction (EP only).
[00374] Among cells knocked out for CD38 and expressing CARs incorporating any of the 12 non-limiting examples of anti-CD38 scFvs, the percentage of cells expressing CD38 on Days 11 and 14 was observed to be less than 2% (Table E3), and the percentage of cells expressing the non-limiting examples of generated CARs on Days 11 and 14 was observed to be similar to or higher than the percentage of cells expressing a CAR containing the reference anti- CD38 scFv (Table E3). Expression levels of the CARs were measured on Day 11 (Figure 4).
Table E3. CD38 and CAR Expression
Figure imgf000124_0002
Figure imgf000125_0001
[00375] To determine the cytotoxicity of NK cells expressing the non-limiting examples of CD38-directed CARs, NK cells isolated from two different healthy human donors were knocked out for CD38 and engineered to express a CD38-directed CAR incorporating one of the 12 non- limiting examples of anti-CD38 scFvs or the reference antibody (Ref), as described above. The CD38 CAR-expressing NK cells were incubated with CD38-expressing THP-1 or MM. IS target cells lines at effector to target ratios (E:T) of between 1:2 and 1:16, with tumor rechallenge on day 4. Target cell killing was assessed by IncuCyte® analysis over approximately 14 days, including at 24 hours post-tumor rechallenge with MM. IS target cells (Figure 5). NK cells expressing the non-limiting examples of CD38-directed CARs were observed to consistently exhibit cytotoxicity against CD38-expressing target cells. A summary of the relative cytotoxicity of cells expressing the non-limiting examples of CARs at the indicated ratios is shown in Table
E4 below.
Table E4. Cytotoxicity Rankings of Non-limiting Examples of CD38 CAR-Expressing NK Cells
Figure imgf000126_0001
[00376] Together, the data are consistent with a finding that cells expressing CARs incorporating the non-limiting examples of anti-CD38 scFvs exhibit specific binding to CD38, as well as comparable or improved antigen-dependent activatiomtonic activation and cytotoxicity against CD38-expressing cells, as compared to a reference antibody.
[00377] The present invention is not intended to be limited in scope to the particular disclosed embodiments, which are provided, for example, to illustrate various aspects of the invention. Various modifications to the compositions and methods described will become apparent from the description and teachings herein. Such variations may be practiced without departing from the true scope and spirit of the disclosure and are intended to fall within the scope of the present disclosure.
[00378] It is contemplated that various combinations or subcombinations of the specific features and aspects of the embodiments disclosed above may be made and still fall within one or more of the inventions. Further, the disclosure herein of any particular feature, aspect, method, property, characteristic, quality, attribute, element, or the like in connection with an embodiment can be used in all other embodiments set forth herein. Accordingly, it should be understood that various features and aspects of the disclosed embodiments can be combined with or substituted for one another in order to form varying modes of the disclosed inventions. Thus, it is intended that the scope of the present inventions herein disclosed should not be limited by the particular disclosed embodiments described above. Moreover, while the invention is susceptible to various modifications, and alternative forms, specific examples thereof have been shown in the drawings and are herein described in detail. It should be understood, however, that the invention is not to be limited to the particular forms or methods disclosed, but to the contrary, the invention is to cover all modifications, equivalents, and alternatives falling within the spirit and scope of the various embodiments described and the appended claims. Any methods disclosed herein need not be performed in the order recited. The methods disclosed herein include certain actions taken by a practitioner; however, they can also include any third-party instruction of those actions, either expressly or by implication. In addition, where features or aspects of the disclosure are described in terms of Markush groups, those skilled in the art will recognize that the disclosure is also thereby described in terms of any individual member or subgroup of members of the Markush group. SEQUENCES
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001

Claims

WHAT IS CLAIMED:
1. An anti-CD38 antibody or an antigen-binding fragment thereof, comprising:
(i) a heavy chain variable region (VH) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:78, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:79, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:80; and a light chain variable region (VL) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:81;
(ii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:66; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:68;
(iii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:69; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:70, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:71;
(iv) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:25, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:23; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:27, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:28, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:29;
(v) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:30, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:31, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:32; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:35;
(vi) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:36, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:37, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:38; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:40;
(vii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:41, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:42; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:43, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:45;
(viii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:46, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:48; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:49, a CDR2 comprising the amino acid sequence set forth in SEQ ID NQ:50, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:51; fix) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO: 24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:52, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:53; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:54;
(x) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:55, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:56, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:57; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:58, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:59, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:60;
(xi) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:61, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:62; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:63; or
(xii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:72, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:73, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:74; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:75, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:76, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:77.
2. The anti-CD38 antibody or antigen-binding fragment thereof of claim 1, wherein:
(i) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 104, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 105;
(ii) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:98, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:99;
(iii) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 100, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 101;
(iv) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:82, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:83;
(v) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:84, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:85;
(vi) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:86, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:87;
(vii) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:88, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:89;
(viii) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:90, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:91; (ix) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:92, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:93;
(x) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:94, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:95;
(xi) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:96, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO:97; or
(xii) the VH comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 102, and the VL comprises an amino acid sequence that has at least about 90% sequence identity to the amino acid sequence set forth in SEQ ID NO: 103.
3. The anti-CD38 antibody or antigen-binding fragment thereof of claim 1, wherein:
(i) the VH comprises the amino acid sequence set forth in SEQ ID NO: 104, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 105;
(ii) the VH comprises the amino acid sequence set forth in SEQ ID NO:98, and the VL comprises the amino acid sequence set forth in SEQ ID NO:99;
(iii) the VH comprises the amino acid sequence set forth in SEQ ID NO: 100, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 101;
(iv) the VH comprises the amino acid sequence set forth in SEQ ID NO: 82, and the VL comprises the amino acid sequence set forth in SEQ ID NO:83;
(v) the VH comprises the amino acid sequence set forth in SEQ ID NO:84, and the VL comprises the amino acid sequence set forth in SEQ ID NO:85;
(vi) the VH comprises the amino acid sequence set forth in SEQ ID NO: 86, and the VL comprises the amino acid sequence set forth in SEQ ID NO:87;
(vii) the VH comprises the amino acid sequence set forth in SEQ ID NO:88, and the VL comprises the amino acid sequence set forth in SEQ ID NO:89;
(viii) the VH comprises the amino acid sequence set forth in SEQ ID NO:90, and the VL comprises the amino acid sequence set forth in SEQ ID NO:91; fix) the VH comprises the amino acid sequence set forth in SEQ ID NO:92, and the VL comprises the amino acid sequence set forth in SEQ ID NO:93; (x) the VH comprises the amino acid sequence set forth in SEQ ID NO:94, and the VL comprises the amino acid sequence set forth in SEQ ID NO:95;
(xi) the VH comprises the amino acid sequence set forth in SEQ ID NO: 96, and the VL comprises the amino acid sequence set forth in SEQ ID NO:97; or
(xii) the VH comprises the amino acid sequence set forth in SEQ ID NO: 102, and the VL comprises the amino acid sequence set forth in SEQ ID NO: 103.
4. An anti-CD38 antibody or an antigen-binding fragment thereof, comprising:
(i) a heavy chain variable region (VH) comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 104, and a light chain variable region (VL) comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 105;
(ii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:98, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:99;
(iii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 100, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth set forth in SEQ ID NO: 101 ;
(iv) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:82, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:83;
(v) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:84, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:85;
(vi) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:86, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:87;
(vii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:88, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:89;
(viii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:90, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:91;
(ix) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:92, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:93; (x) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:94, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:95;
(xi) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:96, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:97; or
(xii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 102, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 103.
5. The anti-CD38 antibody or antigen- binding fragment thereof of claim 1, wherein the antigen-binding fragment of the antibody is an scFv.
6. The anti-CD38 antibody or antigen-binding fragment thereof of claim 5, wherein the scFv comprises the amino acid sequence set forth in SEQ ID NO: 119, SEQ ID NO: 116, SEQ ID NO: 117, SEQ ID NO: 108, SEQ ID NO: 109, SEQ ID NO: 110, SEQ ID NO: 111 , SEQ ID NO: 112, SEQ ID NO: 1 13, SEQ ID NO: 114, SEQ ID NO: 1 15, or SEQ ID NO: 1 18.
7. The anti-CD38 antibody or antigen-binding fragment thereof of claim 5, wherein the scFv comprises a linker between the VH and VL, the linker comprising the amino acid sequence set forth in SEQ ID NO: 1 or SEQ ID NO: 3.
8. The anti-CD38 antibody or antigen-binding fragment thereof of claim 7, wherein the linker comprises the amino acid sequence set forth in SEQ ID NO: 1.
9. The anti-CD38 antibody or antigen-binding fragment thereof of claim 1, wherein the anti-CD38 antibody or antigen-binding fragment thereof binds to the extracellular domain (ECD) of human CD38 with a binding affinity (KD) of between about 1 nM and about 500 nM, or between about 4 nM and about 40 nM.
10. The anti-CD38 antibody or antigen-binding fragment thereof of any one of claims 1-9, which is a chimeric antibody or an antigen-binding fragment thereof.
11. The anti-CD38 antibody or antigen-binding fragment thereof of any one of claims 1-9, which comprises a human variable region framework region.
12. The anti-CD38 antibody or antigen-binding fragment thereof of any one of claims 1-9, which is a fully human antibody or antigen-binding fragment thereof.
13. The anti-CD38 antibody or antigen-binding fragment thereof of any one of claims 1-4 or claims 9-12, wherein the antigen-binding fragment of the antibody is a Fab, a Fab’, a F(ab’)2, or a Fv.
14. A composition comprising the anti-CD38 antibody or antigen-binding fragment thereof of any one of claims 1-13.
15. The composition of claim 14, comprising a pharmaceutically acceptable carrier.
16. An immunoconjugate comprising the anti-CD38 antibody or antigen-binding fragment thereof of any one of claims 1-13 and a therapeutic agent.
17. The immunoconjugate of claim 16, wherein the therapeutic agent is a cytotoxic agent.
18. A composition comprising the immunoconjugate of claim 16 or claim 17 and a pharmaceutically acceptable carrier.
19. A bispecific molecule comprising the anti-CD38 antibody or antigen-binding fragment thereof of any one of claims 1-13 and a second moiety that has a binding specificity for an immune cell.
20. The bispecific molecule of claim 19, wherein the immune cell is a natural killer (NK) cell or a T cell.
21. The bispecific molecule of claim 19 or claim 20, wherein the second moiety binds to CD 16, NKp30, NKp46, NKG2D, or a combination thereof.
22. The bispecific molecule of claim 19 or claim 20, wherein the second moiety binds to CD3.
23. A composition comprising the bispecific molecule of any one of claims 19-22 and a pharmaceutically acceptable carrier.
24. A method for detecting CD38 in a cell or tissue, comprising:
(a) contacting the cell or tissue with the anti-CD38 antibody or antigen-binding fragment thereof of any one of claims 1-13; and
(b) determining the amount of the antibody or antigen-binding fragment thereof bound to the cell or tissue, wherein the amount of the antibody or antigen-binding fragment thereof bound to the cell or tissue indicates the amount of CD38 in the cell or tissue.
25. Use of the anti-CD38 antibody or antigen-binding fragment of any one of claims 1-13 in the manufacture of a kit for detecting CD38 in a cell or tissue, comprising:
(a) contacting the cell or tissue with the anti-CD38 antibody or antigen-binding fragment thereof of any one of claims 1-13; and
(b) determining the amount of the antibody or antigen-binding fragment thereof bound to the cell or tissue, wherein the amount of the antibody or antigen-binding fragment thereof bound to the cell or tissue indicates the amount of CD38 in the cell or tissue.
26. An anti-CD38 chimeric antigen receptor (CAR) comprising an extracellular anti gen -binding domain, a transmembrane domain, and an intracellular signaling domain, wherein the extracellular antigen-binding domain comprises the anti-CD38 antibody or antigen-binding fragment thereof of any one of claims 1-13.
27. The anti-CD38 CAR of claim 26, wherein the transmembrane domain is or comprises a transmembrane from CD8, CD3, CD4, or CD28.
28. The anti-CD38 CAR of claim 26 or claim 27, wherein the transmembrane domain is or comprises a transmembrane from CD8, optionally CD8alpha.
29. The anti-CD38 CAR of any one of claims 26-28, wherein the intracellular signaling domain comprises a CD3zeta domain.
30. The anti-CD38 CAR of any one of claims 26-29, wherein the intracellular signaling domain comprises a co-stimulatory signaling region.
31. The anti-CD38 CAR of claim 30, wherein the co-stimulatory signaling region comprises an intracellular signaling region of 0X40, CD28, 4-1BB, ICOS, or DAP10.
32. The anti-CD38 CAR of claim 30 or claim 31, wherein the co-stimulatory signaling region comprises an intracellular signaling region of 0X40.
33. The anti-CD38 CAR of any one of claims 26-32, comprising a CD8alpha transmembrane domain and an intracellular signaling domain comprising a CD3zeta domain and a co-stimulatory region comprising an intracellular signaling region of 0X40.
34. The anti-CD38 CAR of any one of claims 26-33, wherein the CAR comprises the amino acid sequence set forth in SEQ ID NO:338, SEQ ID NO:335, SEQ ID NO:336, SEQ ID NO:327, SEQ ID NO:328, SEQ ID NO:329, SEQ ID NO:330, SEQ ID NO:331, SEQ ID NO:332, SEQ ID NO:333, SEQ ID NO:334, or SEQ ID NO:337.
35. A nucleic acid molecule encoding the anti-CD38 antibody or antigen-binding fragment thereof of any one of claims 1-13 or the anti-CD38 CAR of any one of claims 26-34.
36. A vector comprising the nucleic acid molecule of claim 35.
37. The vector of claim 36, which is a viral vector.
38. The vector of claim 36 or claim 37, which is a retroviral vector.
39. A cell comprising the anti-CD38 antibody or antigen-binding fragment thereof of any one of claims 1-13, the anti-CD38 CAR of any one of claims 26-34, the nucleic acid molecule of claim 35, or the vector of any one of claims 36-38.
40. The cell of claim 39, which is a natural killer (NK) cell or a T cell.
41. A natural killer (NK) cell comprising the anti-CD38 antibody or antigen-binding fragment thereof of any one of claims 1-13 or the anti-CD38 CAR of any one of claims 26-34.
42. The cell of any one of claims 39-41, which is genetically edited to reduce expression of CD38, optionally wherein the expression of CD38 is reduced by at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, or at least about 80%.
43. The cell of any one of claims 39-42, comprising membrane-bound interleukin- 15 (mbIL15), optionally wherein the CAR and the mbIL15 are encoded bicistronically by the same nucleic acid molecule.
44. A composition comprising a plurality of the cell of any one of claims 39-43.
45. The composition of claim 44, comprising a pharmaceutically acceptable carrier.
46. The composition of claim 44 or claim 45, comprising between about 3 x 108 CAR-expressing cells and about 3 x 109 CAR-expressing cells, or between about 1 x 109 and about 2 x 109 CAR-expressing cells.
47. A method of treatment comprising administering to a subject having a CD38- expressing disease or condition an anti-CD38 antibody or antigen-binding fragment thereof of any one of claims 1-13, an anti-CD38 CAR of any one of claims 26-34, a cell of any one of claims 39- 43, a composition of any one of claims 14, 15, 18, 23, and 44-46, an immunoconjugate of claim 16 or claim 17, or a bispecific molecule of any one of claims 19-22.
48. A method of treatment comprising administering to a subject having a CD38- expressing disease or condition a composition comprising immune cells expressing an anti-CD38 chimeric antigen receptor (CAR) comprising an extracellular antigen-binding domain, a transmembrane domain, and an intracellular signaling domain, wherein the extracellular antigenbinding domain comprises:
(i) a heavy chain variable region (VH) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:78, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:79, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:80; and a light chain variable region (VL) comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:81;
(ii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:66; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:67, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:68;
(iii) ) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:64, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:65, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:69; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:70, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:71; (iv) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:25, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:23; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:27, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:28, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:29;
(v) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:30, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:31, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:32; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:35;
(vi) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:36, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:37, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:38; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:40;
(vii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:41, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:42; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:43, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:44, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:45;
(viii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:46, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:48; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:49, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:50, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:51;
(ix) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:24, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:52, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:53; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:33, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:54; (x) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:55, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:56, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:57; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:58, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:59, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:60;
(xi) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:61, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:47, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:62; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:39, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:34, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:63; or
(xii) a VH comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:72, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:73, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:74; and a VL comprising a CDR1 comprising the amino acid sequence set forth in SEQ ID NO:75, a CDR2 comprising the amino acid sequence set forth in SEQ ID NO:76, and a CDR3 comprising the amino acid sequence set forth in SEQ ID NO:77.
49. A method of treatment comprising administering to a subject having a CD38- expressing disease or condition a composition comprising immune cells expressing an anti-CD38 chimeric antigen receptor (CAR) comprising an extracellular antigen-binding domain, a transmembrane domain, and an intracellular signaling domain, wherein the extracellular antigenbinding domain comprises:
(i) a heavy chain variable region (VH) comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 104, and a light chain variable region (VL) comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 105;
(ii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:98, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:99;
(iii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 100, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 101;
(iv) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:82, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:83; (v) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:84, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:85;
(vi) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:86, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:87;
(vii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:88, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:89;
(viii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:90, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:91; fix) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:92, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:93;
(x) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:94, and a VL comprising the CDR1 , the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:95;
(xi) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO:96, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO:97; or
(xii) a VH comprising the CDR1, the CDR2, and the CDR3 of the VH sequence set forth in SEQ ID NO: 102, and a VL comprising the CDR1, the CDR2, and the CDR3 of the VL sequence set forth in SEQ ID NO: 103.
50. Use of an anti-CD38 antibody or antigen-binding fragment thereof of any one of claims 1-13, an anti-CD38 CAR of any one of claims 26-34, a cell of any one of claims 39-43, a composition of any one of claims 14, 15, 18, 23, and 44-46, an immunoconjugate of claim 16 or claim 17, or a bispecific molecule of any one of claims 19-22 in the treatment of a subject having a CD38-expressing disease or condition.
51. Use of an anti-CD38 antibody or antigen-binding fragment thereof of any one of claims 1-13, an anti-CD38 CAR of any one of claims 26-34, a cell of any one of claims 39-43, a composition of any one of claims 14, 15, 18, 23, and 44-46, an immunoconjugate of claim 16 or claim 17, or a bispecific molecule of any one of claims 19-22 in the manufacture of a medicament for the treatment of a subject having a CD38-expressing disease or condition.
52. The method of any one of claims 47-49 or the use of claim 50 or claim 51, wherein the CD38-expressing disease or condition is a cancer or a tumor.
53. The method of any one of claims 47-49 or claim 52 or the use of any one of claims 48-50, wherein the CD38-expressing disease or condition is a solid tumor.
54. The method or use of claim 52, wherein the cancer is selected from the group consisting of leukemia, lymphoma, myeloma, glioma, thyroid cancer, lung cancer, colorectal cancer, head and neck cancer, stomach cancer, liver cancer, pancreatic cancer, renal cancer, urothelial cancer, prostate cancer, testicular cancer, breast cancer, cervical cancer, endometrial cancer, ovarian cancer, and melanoma.
55. The method or use of claim 52 or claim 54, wherein the cancer is multiple myeloma.
56. The method of any one of claims 47-49 or the use of claim 50 or claim 51, wherein the CD38-expressing disease or condition is an autoimmune disease or disorder.
57. The method or use of claim 56, wherein the autoimmune disease or disorder is selected from the group consisting of systemic lupus erythematosus (SLE), lupus nephritis (LN), CNS lupus, myositis, multiple sclerosis (MS), myasthenia gravis (MG), rheumatoid arthritis (RA), scleroderma, thyroid disease, diabetes, vasculitis, or any combination thereof.
58. The method of any one of claims 47-49 and 52-57 or the use of any one of claims 50-57, wherein the cells are allogeneic to the subject.
59. The method of any one of claims 47-49 and 52-58 or the use of any one of claims 50-58, wherein the subject has been treated with, or is a candidate for treatment with, an anti- CD38 antibody.
60. The method of any one of claims 47-49and 52-59 or the use of any one of claims 50-59, wherein the treatment further comprises administering an anti-CD38 antibody to the subject.
61. The method of any one of claims 47-49 and 52-60 or the use of any one of claims 50-60, wherein the subject has relapsed following treatment with and/or is refractory to a prior line of therapy for a CD38-expressing cancer.
62. The method or use of claim 61, wherein the prior line of therapy is one, two, three, or four prior lines of therapy.
63. A method of producing a cell targeting CD38, comprising introducing into the cell the nucleic acid molecule of claim 35 or the vector of any one of claims 36-38.
64. The method of claim 63, further comprising genetically editing the cell to reduce expression of CD38.
65. The method of claim 64, wherein the genetic editing is carried out using a CRISPR/Cas system.
PCT/US2024/021673 2023-03-28 2024-03-27 Antibodies and chimeric antigen receptors specific for cd38 WO2024206439A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202363492714P 2023-03-28 2023-03-28
US63/492,714 2023-03-28

Publications (2)

Publication Number Publication Date
WO2024206439A2 true WO2024206439A2 (en) 2024-10-03
WO2024206439A3 WO2024206439A3 (en) 2025-01-16

Family

ID=92907712

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2024/021673 WO2024206439A2 (en) 2023-03-28 2024-03-27 Antibodies and chimeric antigen receptors specific for cd38

Country Status (1)

Country Link
WO (1) WO2024206439A2 (en)

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016187216A1 (en) * 2015-05-18 2016-11-24 Bluebird Bio, Inc. Anti-ror1 chimeric antigen receptors
WO2016201389A2 (en) * 2015-06-12 2016-12-15 Alector Llc Anti-cd33 antibodies and methods of use thereof
SG11202003212PA (en) * 2017-10-10 2020-05-28 Sanofi Sa Anti-cd38 antibodies and combinations with anti-cd3 and anti-cd28 antibodies
CN111587254B (en) * 2017-12-05 2025-02-25 特拉维夫医疗中心医学研究基础设施及卫生服务基金 T-cells containing anti-CD38 and anti-CD138 chimeric antigen receptors and uses thereof

Also Published As

Publication number Publication date
WO2024206439A3 (en) 2025-01-16

Similar Documents

Publication Publication Date Title
US11919970B2 (en) Antibodies and chimeric antigen receptors specific for ROR1
US12268741B2 (en) Antibodies and chimeric antigen receptors specific for receptor tyrosine kinase like orphan receptor 1 (ROR1)
US11827714B2 (en) Antibodies and chimeric antigen receptors specific for CD19
AU2018360800B2 (en) Chimeric antigen receptors specific for B-cell maturation antigen (BCMA)
US11623961B2 (en) Antibodies and chimeric antigen receptors specific for B-cell maturation antigen
US20210393689A1 (en) Chimeric antigen receptors specific for g protein-coupled receptor class c group 5 member d (gprc5d)
CA3117419A1 (en) Methods for treatment using chimeric antigen receptors specific for b-cell maturation antigen
AU2023320447A1 (en) Genetic editing of target genes to enhance natural killer cell function
WO2024206439A2 (en) Antibodies and chimeric antigen receptors specific for cd38
US20240226298A1 (en) Chimeric antigen receptors specific for baff-r and cd19 and methods and uses thereof
WO2025006561A2 (en) Methods for treatment using cd19-directed immunotherapies
WO2024031091A2 (en) Chimeric antigen receptors specific for gprc5d and bcma