[go: up one dir, main page]

WO2024025845A1 - Folate-conjugated drugs and uses thereof - Google Patents

Folate-conjugated drugs and uses thereof Download PDF

Info

Publication number
WO2024025845A1
WO2024025845A1 PCT/US2023/028509 US2023028509W WO2024025845A1 WO 2024025845 A1 WO2024025845 A1 WO 2024025845A1 US 2023028509 W US2023028509 W US 2023028509W WO 2024025845 A1 WO2024025845 A1 WO 2024025845A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pharmaceutically acceptable
acceptable salt
substituted
cancer
Prior art date
Application number
PCT/US2023/028509
Other languages
French (fr)
Inventor
Yanwen Fu
Vince Shang Ching YEH
Original Assignee
Sorrento Therapeutics, 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 Sorrento Therapeutics, Inc. filed Critical Sorrento Therapeutics, Inc.
Publication of WO2024025845A1 publication Critical patent/WO2024025845A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/55Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • A61K47/551Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds one of the codrug's components being a vitamin, e.g. niacinamide, vitamin B3, cobalamin, vitamin B12, folate, vitamin A or retinoic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/545Heterocyclic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/554Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being a steroid plant sterol, glycyrrhetic acid, enoxolone or bile acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • SMDCs small molecule-drug conjugates
  • ADCs Antibody-drug conjugates
  • ADCs allow for the targeted delivery of a drug moiety to a tumor, and, in some embodiments intracellular accumulation therein, where systemic administration of unconjugated drugs may result in unacceptable levels of toxicity to normal cells (Polakis P. (2005) Current Opinion in Pharmacology 5:382-387).
  • SMDCs Small molecule-drug conjugates
  • ADCs Small molecule-drug conjugates
  • SMDCs Small molecule-drug conjugates
  • the antibody component is replaced by a targeting ligand that can be a peptide or a small molecule
  • SMDCs have several strengths compared to ADCs. SMDCs are frequently easier to synthesize than biological agents. Most are nonimmunogenic, making them unlikely to provoke an autoimmune response (Min, H.K. et al. (2016) Korean J.
  • SMDCs are more rapidly removed from the blood through glomerular filtration in the kidneys than are ADCs. This results in a better toxicity profile; however, it also has the potential to reduce the effective time on the tumor target (Vlashi, E. et al. (2013) ACS Nano 7:8573-8582).
  • the present disclosure provides SMDCs comprising a folic acid conjugated to the drug moiety (payload) through linker moieties.
  • the folic acid binds to folate receptor-expressing cancer cells and allows for selective uptake of the SMDC into the cancer cells.
  • the SMDCs provided herein selectively deliver an effective amount of drug moiety to tumor tissue and reduce the non-specific toxicity associated with related SMDCs.
  • the SMDC compounds described herein include those with anticancer activity.
  • the folate receptor (FR) is a high-affinity membrane-associated protein, which exhibits limited expression on healthy cells, but is frequently overexpressed on a wide variety of specific cell types, such as epithelial tumor cells (e.g. ovarian, endometrial, breast, colorectal, kidney, lung, nasopharyngeal) and activated (but not resting) macrophages, which are involved in inflammation and autoimmune diseases.
  • hFR ⁇ is overexpressed in a broad variety of cancers, among them adenocarcinomas of uterus, ovary, breast, cervix, kidney and colon and testicular choriocarcinoma, ependymal brain tumors, malignant pleural mesothelioma, and nonfunctioning pituitary adenocarcinoma, while hFR ⁇ in leukemias and activated macrophages (Wibowo, A. et al. (2013) Proc. Natl. Acad. Sci. U.S.A. 110:15180-15188; Low, P. et al. (2007) Acc. Chem. Res. 41:120-129).
  • FR ⁇ Folate Receptor-alpha
  • FLR1 Folate Receptor 1
  • GPI glycosylphosphatidylinositol
  • FR ⁇ has important functions relating to cell proliferation and survival (Kelemen L.E. (2006) Int. J. Cancer 119(2):2430250), and it mediates delivery of the physiological folate, 5-methyltetrahydrofolate, to the interior of cells.
  • FR ⁇ is restricted to the apical membrane of epithelial cells in the kidney proximal tubules, alveolar pneumocytes of the lung, bladder, testes, choroid plexus, and thyroid (Weitman S.D. et al. (1992) Cancer Res. 52:3396-3401; Antony A.C. (1996) Ann. Rev. Nutr. 16:501 -521; Kalli K.R. et al. (2008) Gynecol. Oncol. 108:619-626).
  • FR ⁇ is overexpressed in epithelial-derived tumors including ovarian, uterine, breast, endometrial, pancreatic, renal, lung, colorectal, and brain tumors.
  • FR ⁇ This expression pattern of FR ⁇ makes it a desirable target for FR ⁇ -directed cancer therapy.
  • Folate plays important roles in nucleotide biosynthesis and cell division, intracellular activities which occur in both malignant and certain normal cells. Upon binding the folate receptor, the folate impacts the cell cycle in dividing cells. This led to the use of folic acid and its analogues and derivatives as a targeting agent for the delivery of therapeutic and/or diagnostic agents to these specific cell populations to achieve a selective concentration of pharmaceutical and/or diagnostic agents in these specific cells relative to normal cells (Leamon and Low (2001) Drug Discov. Today 6:44-51; Leamon and Reddy (2004) Adv. Drug Deliv. Rev.
  • the linker in SMDCs usually consists of a spacer and a cleavable bridge. Linkers are designed to preserve the activity of post-cleavage species and to optimize the drug release, pharmacokinetics, and pharmacodynamics of the targeting ligand and payload (Srinivasarao, M. et al. (2015) Nat. Rev. Drug Discov. 14: 203–219; Vlahov, I.R. and Leamon, C.P (2012) Bioconj. Chem. 23:1357-1369). Another function of the spacer is to improve the hydrophilicity of SMDC.
  • the cleavable bridge retains stability during the SMDC transportation from the vasculature to the tumor, and is typically cleaved by one of two triggering methods.
  • the first mechanism is cleavage in the endosomes of the target cells due to low pH.
  • Such a cleavage bridge comprises acetals and hydrazones (Yang, J. et al. (2007) . J. Pharm. Exp. 321:462-468).
  • the second mechanism is through use of a disulfide-based linker, which undergoes cleavage due to an intracellular excess of glutathione (GSH), thioredoxin, peroxiredoxins, and nicotinamide adenine dinucleotides (NADH and NADPH) (Srinivasarao, M. et al. (2015) Nat. Rev. Drug Discov. 14: 203–219).
  • GSH glutathione
  • thioredoxin thioredoxin
  • peroxiredoxins nicotinamide adenine dinucleotides
  • NADH and NADPH nicotinamide adenine dinucleotides
  • multivalent ligands comprising several payloads linked to the targeting compound. Payloads that target mitosis, DNA replication, and protein translation are currently investigated.
  • folate receptors such as folate receptor alpha (FR ⁇ )
  • FR ⁇ folate receptor alpha
  • FR ⁇ folate receptor alpha
  • Folate receptor binds folic acid and analogues and derivatives thereof with very high affinity. Once the folate is bound to the folate receptor it impacts the cell cycle in dividing cells.
  • SMDCs small molecule-drug conjugates
  • L is a multivalent linker bound at least to a folic acid and a drug moiety
  • HL is a half-life extender
  • D is a drug moiety.
  • a method of treating a FR-expressing cancer such as an FR ⁇ -expressing cancer in a subject in need thereof, said method including administering the compound or pharmaceutically acceptable salt thereof as described herein (including in an aspect embodiment table example or claim) to the subject
  • a pharmaceutical composition comprising the compound or pharmaceutically acceptable salt thereof as described herein, and a pharmaceutically acceptable excipient.
  • Embodiment 1 is a compound of the Formula (I) or (II): or a pharmaceutically acceptable salt thereof, wherein: L is a multivalent linker bound at least to a folic acid and a drug moiety; HL is a half-life extender; and D is a drug moiety.
  • Embodiment 2 is the compound or a pharmaceutically acceptable salt thereof of embodiment 1, wherein L is a bond, -C(O)-, -NH-, Amino Acid Unit, Peptoid, – (CH2CH2O)n–, –(CH2)n–, –(4-aminobenzyloxycarbonyl)–, –(C(O)CH2CH2C(O))–, –(C(O)CH2CH2NH)–,
  • Embodiment 3 is the compound or a pharmaceutically acceptable salt thereof of embodiment 1 or 2, wherein L is -C(O)-, -NH-,–(CH2CH2O)n–, –(CH2)n–, –(4- aminobenzyloxycarbonyl)–, -Cys-, -Asp-, -Arg-, -Val-, -Glu-, -citrulline- (-Cit-), -Lys-, combinations thereof.
  • Embodiment 4 is the compound or a pharmaceutically acceptable salt thereof of embodiment 3, wherein L is -C(O)-, -NH-, –(CH2CH2O)n–, –(CH2)n–, –SCH2CH2O–, – (C(O)CH2CH2C(O))–, -Val-, -Cit-, –(4-aminobenzyloxycarbonyl)–, -Arg-, -Asp-, -Lys-, - combinations thereof.
  • Embodiment 5 is the compound or a pharmaceutically acceptable salt thereof of embodiment 4, wherein L is
  • Embodiment 6 is the compound or a pharmaceutically acceptable salt thereof of any one of embodiments 1-5, wherein HL is a cholesterol-like half-life extender or albumin binder half-life extender.
  • Embodiment 7 is the compound or a pharmaceutically acceptable salt thereof of embodiment 6, wherein HL is , ,
  • Embodiment 8 is the compound or a pharmaceutically acceptable salt thereof of embodiment 7, wherein .
  • Embodiment 9 is the compound or a pharmaceutically acceptable salt thereof of any one of embodiments 1-8, wherein D is a tubulin inhibitor or disruptor, kinase inhibitor, DNA damaging agent, transcription inhibitors, or proteolysis-targeting chimera (PROTAC).
  • Embodiment 10 is the compound or a pharmaceutically acceptable salt thereof of embodiment 9, wherein D is a tubulin inhibitor.
  • Embodiment 11 is the compound or a pharmaceutically acceptable salt thereof of any one of embodiments 1-9, wherein D is a pyrrolobenzodiazepine, duocarmycin, anthracycline, maytansinoid, auristatin, calicheamicin, camptothecin, RNA polymerase II inhibitor, topoisomerase I inhibitor, tyrosine kinase inhibitor, EG5 inhibitor, or MEK inhibitor.
  • Embodiment 12 is the compound or a pharmaceutically acceptable salt thereof of embodiment 11, wherein D is an auristatin.
  • Embodiment 13 is the compound or a pharmaceutically acceptable salt thereof of embodiment 11, wherein D is MMAE, MMAF, Duo5, PNU, SN-38, irinotecan, amatoxin, maytansine, exatecan, trametinib, abemaciclib, palbociclib, or examorpholine.
  • Embodiment 14 is the compound or a pharmaceutically acceptable salt thereof of embodiment 13, wherein D is Duo5.
  • Embodiment 15 is the compound or a pharmaceutically acceptable salt thereof of embodiment 13, wherein D is MMAE.
  • Embodiment 16 is the compound or a pharmaceutically acceptable salt thereof of embodiment 13, wherein D is examorpholine.
  • Embodiment 17 is the compound or pharmaceutically acceptable salt thereof of any one of embodiments 1-16, wherein the compound is: ⁇ ⁇ or a pharmaceutically acceptable salt thereof.
  • Embodiment 18 is the compound or pharmaceutically acceptable salt thereof of any one of embodiments 1-17, for use in therapy.
  • Embodiment 19 is the compound or pharmaceutically acceptable salt thereof of embodiment 18, for use in treating a FR-expressing cancer, optionally wherein the FR- expressing cancer is non-small cell lung carcinoma (NSCLC), lung cancer, mesothelioma, or ovarian cancer.
  • NSCLC non-small cell lung carcinoma
  • Embodiment 20 is a method of treating a FR-expressing cancer in a subject, comprising administering the compound or pharmaceutically acceptable salt thereof of any one of embodiments 1-17 to a subject in need thereof.
  • Embodiment 21 is use of the compound or pharmaceutically acceptable salt thereof of any one of embodiments 1-17 for the manufacture of a medicament.
  • Embodiment 22 is use of the compound or pharmaceutically acceptable salt thereof of any one of embodiments 1-17 for the manufacture of a medicament for treating a FR- expressing cancer, optionally wherein the FR-expressing cancer is non-small cell lung carcinoma (NSCLC), lung cancer, mesothelioma, or ovarian cancer.
  • NSCLC non-small cell lung carcinoma
  • Embodiment 23 is the compound or pharmaceutically acceptable salt thereof for use, use, or method of any one of embodiments 19, 20, or 22, wherein the FR-expressing cancer is an epithelial-derived tumor.
  • Embodiment 24 is the compound or pharmaceutically acceptable salt thereof for use, use, or method of embodiment 23, wherein the epithelial-derived tumors are ovarian, uterine, breast, endometrial, pancreatic, renal, lung, colorectal, or brain tumors.
  • Embodiment 25 is the compound or pharmaceutically acceptable salt thereof for use, use, or method of any one of embodiments 19, 20, or 22, wherein the FR-expressing cancer is non-small cell lung carcinoma (NSCLC) or ovarian cancer.
  • NSCLC non-small cell lung carcinoma
  • Embodiment 26 is the compound or pharmaceutically acceptable salt thereof for use, use, or method of any one of embodiments 19, 20, or 22-25, wherein the FR-expressing cancer is in a mammal, optionally wherein the mammal is a human.
  • Embodiment 27 is a method of inhibiting proliferation of a FR-expressing cell, comprising contacting the FR-expressing cell with the compound or pharmaceutically acceptable salt thereof of any one of embodiments 1-17.
  • Embodiment 28 is the use of embodiment 21, wherein the medicament is for inhibiting proliferation of a FR-expressing cell.
  • Embodiment 29 is the compound or pharmaceutically acceptable salt thereof of any one of embodiments 1-17, for use in inhibiting proliferation of a FR-expressing cell.
  • Embodiment 30 is the method, use, or compound or pharmaceutically acceptable salt thereof for use of any one of embodiments 27-29, wherein the FR-expressing cell is a FR-expressing cancer cell, optionally wherein the FR-expressing cancer is non-small cell lung carcinoma (NSCLC), lung cancer, mesothelioma, or ovarian cancer.
  • NSCLC non-small cell lung carcinoma
  • Embodiment 31 is the method, use, or compound or pharmaceutically acceptable salt thereof for use of any one of embodiments 27-29, wherein the FR-expressing cell is a FR-expressing non-small cell lung carcinoma (NSCLC) cell or FR-expressing ovarian cell.
  • Embodiment 32 is a pharmaceutical composition comprising the compound or pharmaceutically acceptable salt thereof of any one of embodiments 1-17, and a pharmaceutically acceptable excipient.
  • FIGS. 1A-B show results of an in vitro efficacy study of Duo5 and MMAE using: KB (FR+) cells and A549 (FR-) cells.
  • FIG. 1A shows results of an in vitro efficacy study of Duo5.
  • FIG. 1B shows results of an in vitro efficacy study of MMAE.
  • the log-molar concentration of the indicated compound is on the horizontal axis.
  • FIGS. 2A-B show results of an in vitro efficacy and stability study of SMDCs (FR- PEG-Duo5, FR-VC- Duo5, FR-IODO- Duo5, FR-VC-PAB-MMAE, and FR-PL-MMAE) using: A549 (FR-) cells.
  • FIG. 2A shows results of a 2-hour pulse assay.
  • FIGS. 3A-B show results of an in vitro efficacy, specificity, and stability study of SMDC FR-PEG- Duo5 (with and without pretreatment with folic acid) using: KB (FR+) cells.
  • FIG. 3A shows results of a 2-hour pulse assay.
  • FIG. 3B shows results of a 72-hour assay.
  • FIGS. 4A-B show results of an in vitro efficacy, specificity, and stability study of SMDC FR-VC- Duo5 (with and without pretreatment with folic acid) using: KB (FR+) cells.
  • FIGS. 5A-B show results of an in vitro efficacy, specificity, and stability study of SMDC FR-IODO- Duo5 (with and without pretreatment with folic acid) using: KB (FR+) cells.
  • FIG. 5A shows results of a 2-hour pulse assay.
  • FIG. 5B shows results of a 72-hour assay.
  • FIGS. 6A-B show results of an in vitro efficacy, specificity, and stability study of SMDC FR-VC-PAB-MMAE (with and without pretreatment with folic acid) using: KB (FR+) cells.
  • FIG. 5A-B shows results of an in vitro efficacy, specificity, and stability study of SMDC FR-VC-PAB-MMAE (with and without pretreatment with folic acid) using: KB (FR+) cells.
  • FIG. 6A shows results of a 2-hour pulse assay.
  • FIG. 6B shows results of a 72- hour assay.
  • FIGS. 7A-B show results of an in vitro efficacy, specificity, and stability study of SMDC FR-PL-MMAE (with and without pretreatment with folic acid) using: KB (FR+) cells.
  • FIG. 7A shows results of a 2-hour pulse assay.
  • FIG. 7B shows results of a 72-hour assay.
  • FIG. 8 shows results of an in vitro efficacy, specificity, and stability study of FR- IODO-Examorpholine using KB (FR+) cells with 120-hour assay.
  • FIG. 8 shows results of an in vitro efficacy, specificity, and stability study of FR- IODO-Examorpholine using KB (FR+) cells with 120-hour assay.
  • the term “and/or” used herein is to be taken mean specific disclosure of each of the specified features or components with or without the other.
  • the term “and/or” as used in a phrase such as “A and/or B” herein is intended to include “A and B,” “A or B,” “A” (alone), and “B” (alone).
  • the term “and/or” as used in a phrase such as “A, B, and/or C” is intended to encompass each of the following aspects: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone).
  • the term “about” refers to a value or composition that is within an acceptable error range for the particular value or composition as determined by one of ordinary skill in the art, which will depend in part on how the value or composition is measured or determined, i.e., the limitations of the measurement system.
  • “about” or “approximately” can mean within one or more than one standard deviation per the practice in the art.
  • “about” or “approximately” can mean a range of up to 10% (i.e., ⁇ 10%) or more depending on the limitations of the measurement system.
  • about 5 mg can include any number between 4.5 mg and 5.5 mg.
  • the terms can mean up to an order of magnitude or up to 5-fold of a value.
  • the meaning of “about” or “approximately” should be assumed to be within an acceptable error range for that particular value or composition. In embodiments, about includes the specified value.
  • “comprises,” “comprising,” “containing” and “having” and the like can have the meaning ascribed to them in U.S. Patent law and can mean “includes,” “including,” and the like.
  • Consisting essentially of or “consists essentially” likewise has the meaning ascribed in U.S.
  • polypeptide polypeptide
  • peptide and “protein” and other related terms used herein are used interchangeably to refer to a polymer of amino acid residues, wherein the polymer may in embodiments be conjugated to a moiety that does not consist of amino acids.
  • the terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymers.
  • a "fusion protein” refers to a chimeric protein encoding two or more separate protein sequences that are recombinantly expressed as a single moiety.
  • Polypeptides include mature molecules that have undergone cleavage. These terms encompass native and artificial proteins, protein fragments and polypeptide analogs (such as muteins, variants, chimeric proteins and fusion proteins) of a protein sequence as well as post-translationally, or otherwise covalently or non-covalently, modified proteins.
  • Two or more polypeptides e.g., 3 polypeptide chains
  • a polypeptide complex can be dimeric, trimeric, tetrameric, or higher order complexes depending on the number of polypeptide chains that form the complex.
  • cancer cancer
  • neoplasm and “tumor” are used interchangeably and, in either the singular or plural form, refer to cells that have undergone a malignant transformation that makes them pathological to the host organism.
  • Primary cancer cells can be readily distinguished from non-cancerous cells by well-established techniques, particularly histological examination.
  • the definition of a cancer cell includes not only a primary cancer cell, but any cell derived from a cancer cell ancestor.
  • a “clinically detectable” tumor is one that is detectable on the basis of tumor mass; e.g., by procedures such as computed tomography (CT) scan, magnetic resonance imaging (MRI), X-ray, ultrasound or palpation on physical examination, and/or which is detectable because of the expression of one or more cancer-specific antigens in a sample obtainable from a patient.
  • CT computed tomography
  • MRI magnetic resonance imaging
  • X-ray X-ray
  • Tumors may be a hematopoietic (or hematologic or hematological or blood-related) cancer, for example, cancers derived from blood cells or immune cells, which may be referred to as “liquid tumors.”
  • liquid tumors include leukemias such as chronic myelocytic leukemia, acute myelocytic leukemia, chronic lymphocytic leukemia and acute lymphocytic leukemia; plasma cell malignancies such as multiple myeloma, MGUS and Waldenstrom's macroglobulinemia; lymphomas such as non- Hodgkin's lymphoma, Hodgkin's lymphoma; and the like.
  • the cancer may be any cancer in which an abnormal number of blast cells or unwanted cell proliferation is present or that is diagnosed as a hematological cancer, including both lymphoid and myeloid malignancies.
  • Myeloid malignancies include, but are not limited to, acute myeloid (or myelocytic or myelogenous or myeloblastic) leukemia (undifferentiated or differentiated), acute promyeloid (or promyelocytic or promyelogenous or promyeloblastic) leukemia, acute myelomonocytic (or myelomonoblastic) leukemia, acute monocytic (or monoblastic) leukemia, erythroleukemia and megakaryocytic (or megakaryoblastic) leukemia.
  • leukemias may be referred together as acute myeloid (or myelocytic or myelogenous) leukemia (AML).
  • Myeloid malignancies also include myeloproliferative disorders (MPD) which include, but are not limited to, chronic myelogenous (or myeloid) leukemia (CML), chronic myelomonocytic leukemia (CMML), essential thrombocythemia (or thrombocytosis), and polcythemia vera (PCV).
  • CML chronic myelogenous leukemia
  • CMML chronic myelomonocytic leukemia
  • PCV polcythemia vera
  • Myeloid malignancies also include myelodysplasia (or myelodysplastic syndrome or MDS), which may be referred to as refractory anemia (RA), refractory anemia with excess blasts (RAEB), and refractory anemia with excess blasts in transformation (RAEBT); as well as myelofibrosis (MFS) with or without agnogenic myeloid metaplasia.
  • RA refractory anemia
  • RAEB refractory anemia with excess blasts
  • RES myelofibrosis
  • Hematopoietic cancers also include lymphoid malignancies, which may affect the lymph nodes, spleens, bone marrow, peripheral blood, and/or extranodal sites.
  • B-cell malignancies include, but are not limited to, B-cell non- Hodgkin's lymphomas (B-NHLs).
  • B-NHLs may be indolent (or low-grade), intermediate- grade (or aggressive) or high-grade (very aggressive).
  • Indolent Bcell lymphomas include follicular lymphoma (FL); small lymphocytic lymphoma (SLL); marginal zone lymphoma (MZL) including nodal MZL, extranodal MZL, splenic MZL and splenic MZL with villous lymphocytes; lymphoplasmacytic lymphoma (LPL); and mucosa-associated-lymphoid tissue (MALT or extranodal marginal zone) lymphoma.
  • FL follicular lymphoma
  • SLL small lymphocytic lymphoma
  • MZL marginal zone lymphoma
  • LPL lymphoplasmacytic lymphoma
  • MALT mucosa-associated-lymphoid tissue
  • Intermediate-grade B-NHLs include mantle cell lymphoma (MCL) with or without leukemic involvement, diffuse large cell lymphoma (DLBCL), follicular large cell (or grade 3 or grade 3B) lymphoma, and primary mediastinal lymphoma (PML).
  • MCL mantle cell lymphoma
  • DLBCL diffuse large cell lymphoma
  • follicular large cell or grade 3 or grade 3B lymphoma
  • PML primary mediastinal lymphoma
  • High-grade B-NHLs include Burkitt's lymphoma (BL), Burkitt-like lymphoma, small non-cleaved cell lymphoma (SNCCL) and lymphoblastic lymphoma.
  • B-NHLs include immunoblastic lymphoma (or immunocytoma), primary effusion lymphoma, HIV associated (or AIDS related) lymphomas, and post-transplant lymphoproliferative disorder (PTLD) or lymphoma.
  • B-cell malignancies also include, but are not limited to, chronic lymphocytic leukemia (CLL), prolymphocytic leukemia (PLL), Waldenstrom's macroglobulinemia (WM), hairy cell leukemia (HCL), large granular lymphocyte (LGL) leukemia, acute lymphoid (or lymphocytic or lymphoblastic) leukemia, and Castleman's disease.
  • CLL chronic lymphocytic leukemia
  • PLL prolymphocytic leukemia
  • WM Waldenstrom's macroglobulinemia
  • HCL hairy cell leukemia
  • LGL large granular lymphocyte
  • LAman's disease Castleman's disease.
  • NHL may also include T-cell non-Hodgkin's lymphoma s (T-NHLs), which include, but are not limited to T-cell non-Hodgkin's lymphoma not otherwise specified (NOS), peripheral T-cell lymphoma (PTCL), anaplastic large cell lymphoma (ALCL), angioimmunoblastic lymphoid disorder (AILD), nasal natural killer (NK) cell/T-cell lymphoma, gamma/delta lymphoma, cutaneous T cell lymphoma, mycosis fungoides, and Sezary syndrome.
  • T-NHLs T-cell non-Hodgkin's lymphoma s
  • Hematopoietic cancers also include Hodgkin's lymphoma (or disease) including classical Hodgkin's lymphoma, nodular sclerosing Hodgkin's lymphoma, mixed cellularity Hodgkin's lymphoma, lymphocyte predominant (LP) Hodgkin's lymphoma, nodular LP Hodgkin's lymphoma, and lymphocyte depleted Hodgkin's lymphoma.
  • Hematopoietic cancers also include plasma cell diseases or cancers such as multiple myeloma (MM) including smoldering MM, monoclonal gammopathy of undetermined (or unknown or unclear) significance (MGUS), plasmacytoma (bone, extramedullary), lymphoplasmacytic lymphoma (LPL), Waldenstrom's Macroglobulinemia, plasma cell leukemia, and primary amyloidosis (AL).
  • MM multiple myeloma
  • MGUS monoclonal gammopathy of undetermined (or unknown or unclear) significance
  • MGUS monoclonal gammopathy of undetermined (or unknown or unclear) significance
  • plasmacytoma bone, extramedullary
  • LPL lymphoplasmacytic lymphoma
  • Waldenstrom's Macroglobulinemia plasma cell leukemia
  • plasma cell leukemia and primary amyloidosis
  • AL primary amyloidosis
  • Hematopoietic cancers may also
  • Tissues which include hematopoietic cells referred herein to as “hematopoietic cell tissues” include bone marrow; peripheral blood; thymus; and peripheral lymphoid tissues, such as spleen, lymph nodes, lymphoid tissues associated with mucosa (such as the gut-associated lymphoid tissues), tonsils, Peyer's patches and appendix, and lymphoid tissues associated with other mucosa, for example, the bronchial linings.
  • Exemplary cancers that may be treated with a compound or method provided herein include brain cancer, glioma, glioblastoma, neuroblastoma, prostate cancer, colorectal cancer, pancreatic cancer, Medulloblastoma, melanoma, cervical cancer, gastric cancer, ovarian cancer, lung cancer, cancer of the head, Hodgkin's Disease, and Non-Hodgkin's Lymphomas.
  • Exemplary cancers that may be treated with a compound or method provided herein include cancer of the thyroid, endocrine system, brain, breast, cervix, colon, head & neck, liver, kidney, lung, ovary, pancreas, rectum, stomach, and uterus.
  • thyroid carcinoma cholangiocarcinoma, pancreatic adenocarcinoma, skin cutaneous melanoma, colon adenocarcinoma, rectum adenocarcinoma, stomach adenocarcinoma, esophageal carcinoma, head and neck squamous cell carcinoma, breast invasive carcinoma, lung adenocarcinoma, lung squamous cell carcinoma, non-small cell lung carcinoma, mesothelioma, multiple myeloma, neuroblastoma, glioma, glioblastoma multiforme, ovarian cancer, rhabdomyosarcoma, primary thrombocytosis, primary macroglobulinemia, primary brain tumors, nasopharyngeal tumors, spinal cord tumors, malignant pancreatic insulanoma, malignant carcinoid, urinary bladder cancer, premalignant skin lesions, testicular cancer, thyroid cancer, neuroblastoma
  • the cancers that may be treated with a compound or method provided herein include epithelial-derived tumors including ovarian, uterine, breast, endometrial, pancreatic, nasopharyngeal, kidney, lung, colorectal, testicular, spinal cord, and brain tumors.
  • the cancers that may be treated with a compound or a method provided herein include serous and endometrioid epithelial ovarian cancer, renal cancer, endometrial adenocarcinoma, non-small cell lung carcinoma (NSCLC) of the adenocarcinoma subtype, mesotheliomas, and triple-negative breast cancer (TNBC).
  • epithelial-derived tumors including ovarian, uterine, breast, endometrial, pancreatic, nasopharyngeal, kidney, lung, colorectal, testicular, spinal cord, and brain tumors.
  • the cancers that may be treated with a compound or a method provided herein
  • An "advanced” cancer is one which has spread outside the site or organ of origin, either by local invasion or metastasis.
  • the term “advanced” cancer includes both locally advanced and metastatic disease.
  • Metal-static cancer refers to cancer that has spread from one part of the body to another part of the body.
  • a "refractory” cancer is one that progresses even though an anti-tumor treatment, such as a chemotherapy, is administered to the cancer patient.
  • An example of a refractory cancer is one which is platinum refractory.
  • a "recurrent" cancer is one that has regrown, either at the initial site or at a distant site, after a response to initial therapy.
  • Binding affinity refers to intrinsic binding affinity which reflects a 1:1 interaction between members of a binding pair (e.g., ligand and receptor).
  • the affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (K D ). Affinity can be measured by common methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described in the following.
  • a dissociation constant can be measured using a BIACORE surface plasmon resonance (SPR) assay.
  • SPR surface plasmon resonance refers to an optical phenomenon that allows for the analysis of real-time interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIACORE system (Biacore Life Sciences division of GE Healthcare, Piscataway, NJ).
  • FR ⁇ or “FOLR1,” as used herein, refers to any native FR ⁇ from any vertebrate source, including mammals such as primates (e.g. humans, cynomolgus monkey (cyno)) and rodents (e.g., mice and rats), unless otherwise indicated.
  • FR ⁇ is also referred to as "human folate receptor 1," and "FOLR 1".
  • FR ⁇ is a single chain membrane protein capable of binding to folic acid and its analogs or derivatives.
  • the term encompasses “full- length,” unprocessed FR ⁇ as well as any form of FR ⁇ that results from processing in the cell.
  • the term also encompasses naturally occurring variants of FR ⁇ , e.g., splice variants, allelic variants, and isoforms.
  • Human FR ⁇ sequences are known and include, for example, the sequences publicly available at UniProtKB Accession No. P 15328 (including isoforms).
  • the term “FR-expressing cancer” refers to a cancer comprising cells that express FR on their surface.
  • FR ⁇ -expressing cancer refers to a cancer comprising cells that express FR ⁇ on their surface.
  • FR ⁇ a FR, such as FR ⁇ polypeptide or a nucleic acid encoding such a polypeptide
  • Such increased expression or overexpression can be caused, for example, by mutation, gene amplification, increased transcription, increased translation, or increased protein stability.
  • cytotoxic agent refers to a substance that inhibits or prevents a cellular function and/or causes cell death or destruction.
  • Cytotoxic agents include, but are not limited to, radioactive isotopes (e.g., 211 At, 131 I, 125 I, 90 Y, 186 Re, 188 Re, 153 Sm, 212 Bi, 32 P, 212 Pb and radioactive isotopes of Lu); chemotherapeutic agents or drugs (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 of
  • a “chemotherapeutic agent” is a chemical compound useful in the treatment of a cancer.
  • chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN®); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); delta-9-tetrahydrocannabinol (dronabinol, MARINOL®); beta-lapachone; lapachol; colchicines; betulinic acid; a camptothecin (including the synthetic analogue topotecan
  • calicheamicin especially calicheamicin gamma1I and calicheamicin omegaI1 (see, e.g., Agnew, Chem Intl. Ed. Engl., 33: 183-186 (1994)); dynemicin, including dynemicin A; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including morpholino-doxorubicin, cyanomorpholino-dox
  • a “small molecule-drug conjugate” or “SMDC” is a targeting ligand conjugated to one or more heterologous molecule(s), including but not limited to a cytotoxic agent.
  • the targeting ligand can be, for example, folic acid as described herein, or its analogs or derivatives (which target the folate receptor).
  • the cytotoxic agent can be any cytotoxic agent described herein.
  • the targeting ligand can be directly linked to the cytotoxic agent via a covalent bond, or the targeting ligand can be linked to the cytotoxic agent indirectly via a linker. Typically, the linker is covalently bonded to the targeting ligand and also covalently bonded to the cytotoxic agent.
  • Such a linker may be a cleavable linker, for example, cleavable under certain pH condition (pH sensitive linker such as acetals or hydrazones), cleavable by a protease (protease sensitive linker such as peptide linkers), or cleavable in the presence of glutathione (glutathione sensitive linker such as disulfide linkers).
  • the linker comprises a protease cleavage site, which may contain 2-5 amino acid residues that are recognizable and/or cleavable by a suitable protease.
  • Such a peptide may comprise naturally-occurring amino acid residues, non-naturally occurring amino acid residues, modified amino acid residues, or a combination thereof.
  • the peptide linker can be a dipeptide linker. Examples include a valine-citrulline (val-cit or VC) linker, a phenylalanine-lysine (phe-lys) linker, or maleimidocapronic-valine-citruline-p- aminobenzyloxycarbonyl (MC-VC-PAB) linker.
  • the linker may be non- cleavable, e.g., a linker comprising optionally substituted alkane or thioether.
  • the linker may comprise a functional group that can form a covalent bond with the targeting ligand.
  • exemplary functional groups include, but are not limited to, a maleimide group, an iodoacetamide group, a vinyl sulfone group, an acrylate group, an acrylamide group, an acrylonitrile group, or a methacrylate group.
  • the term “small molecule-drug conjugate” or “SMDC” refers to a conjugate wherein at least one cytotoxic agent is a therapeutic moiety such as a drug (“D”). As used herein, “D” refers to drug moiety and includes analogs or derivatives thereof.
  • folic acid, or the analog or the derivative thereof is covalently bound to the linker (L), and the drug, or the analog or the derivative thereof, is also covalently bound to the linker (L).
  • the linker (L) can comprise multiple linkers.
  • the linker (L) can comprise one or more components selected from spacer linkers, releasable linkers, and heteroatom linkers, and any combinations thereof, in any order.
  • conjugated when referring to two moieties means the two moieties are bonded, wherein the bond or bonds connecting the two moieties may be covalent or non-covalent.
  • the two moieties are covalently bonded to each other (e.g.
  • an “individual” or “subject” is a mammal. Mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats). In certain embodiments, the individual or subject is a human.
  • the subject is an adult, an adolescent, a child, or an infant.
  • the terms “individual” or “patient” are used and are intended to be interchangeable with “subject”.
  • pharmaceutically acceptable salts is meant to include salts of the active compounds that are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein.
  • base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent.
  • Examples of pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt.
  • acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent.
  • pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p- tolylsulfonic, citric, tartaric, oxalic, methanesulfonic, and the like.
  • salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, for example, Berge et al., “Pharmaceutical Salts”, Journal of Pharmaceutical Science, 1977, 66, 1-19).
  • Certain specific compounds of the present disclosure contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts.
  • the compounds of the present disclosure may exist as salts, such as with pharmaceutically acceptable acids.
  • the present disclosure includes such salts.
  • Non-limiting examples of such salts include hydrochlorides, hydrobromides, phosphates, sulfates, methanesulfonates, nitrates, maleates, acetates, citrates, fumarates, proprionates, tartrates (e.g., (+)-tartrates, (-)-tartrates, or mixtures thereof including racemic mixtures), succinates, benzoates, and salts with amino acids such as glutamic acid, and quaternary ammonium salts (e.g. methyl iodide, ethyl iodide, and the like). These salts may be prepared by methods known to those skilled in the art.
  • the neutral forms of the compounds are preferably regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner.
  • the parent form of the compound may differ from the various salt forms in certain physical properties, such as solubility in polar solvents.
  • the present disclosure provides compounds, which are in a prodrug form.
  • Prodrugs of the compounds described herein are those compounds that readily undergo chemical changes under physiological conditions to provide the compounds of the present disclosure.
  • Prodrugs of the compounds described herein may be converted in vivo after administration.
  • prodrugs can be converted to the compounds of the present disclosure by chemical or biochemical methods in an ex vivo environment, such as, for example, when contacted with a suitable enzyme or chemical reagent.
  • Certain compounds of the present disclosure can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are encompassed within the scope of the present disclosure. Certain compounds of the present disclosure may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present disclosure and are intended to be within the scope of the present disclosure.
  • “Pharmaceutically acceptable excipient” and “pharmaceutically acceptable carrier” refer to a substance that aids the administration of an active agent to and absorption by a subject and can be included in the compositions of the present disclosure without causing a significant adverse toxicological effect on the patient.
  • Non-limiting examples of pharmaceutically acceptable excipients include water, NaCl, normal saline solutions, lactated Ringer’s, normal sucrose, normal glucose, binders, fillers, disintegrants, lubricants, coatings, sweeteners, flavors, salt solutions (such as Ringer’s solution), alcohols, oils, gelatins, carbohydrates such as lactose, amylose or starch, fatty acid esters, hydroxymethycellulose, polyvinyl pyrrolidine, and colors, and the like.
  • compositions can be sterilized and, if desired, mixed with auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the disclosure.
  • auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the disclosure.
  • auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the disclosure.
  • pharmaceutical excipients are useful in the present disclosure.
  • pharmaceutical formulation refers to a preparation which
  • administering refers to the physical introduction of an agent to a subject, using any of the various methods and delivery systems known to those skilled in the art.
  • exemplary routes of administration for the formulations disclosed herein include intravenous, intramuscular, subcutaneous, intraperitoneal, spinal or other parenteral routes of administration, for example by injection or infusion.
  • parenteral administration means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intralymphatic, intralesional, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion, as well as in vivo electroporation.
  • the formulation is administered via a non-parenteral route, e.g., orally.
  • Non-parenteral routes include a topical, epidermal or mucosal route of administration, for example, intranasally, vaginally, rectally, sublingually or topically.
  • Administering can also be performed, for example, once, a plurality of times, and/or over one or more extended periods.
  • Administration “in combination with” one or more further therapeutic agents includes simultaneous (concurrent) or consecutive administration in any order.
  • the combination therapy can provide “synergy” and prove “synergistic”, i.e., the effect achieved when the active ingredients used together is greater than the sum of the effects that results from using the compounds separately.
  • a synergistic effect can be attained when the active ingredients are: (1) co-formulated and administered or delivered simultaneously in a combined, unit dosage formulation; (2) delivered serially, by alternation, or in parallel as separate formulations; or (3) by some other regimen.
  • a synergistic effect can be attained when the compounds are administered or delivered sequentially, e.g., by different injections in separate syringes.
  • a synergistic combination produces effects that are greater than the additive effects of the individual components of the combination.
  • An “effective amount” of an agent e.g., a pharmaceutical formulation, refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
  • saccharide means carbohydrate (or sugar). In embodiments, the saccharide is a monosaccharide. In embodiments, the saccharide is a polysaccharide. The most basic unit of saccharide is a monomer of carbohydrate. The general formula is C n H 2n O n .
  • saccharide derivative means sugar molecules that have been modified with substituents other than hydroxyl groups. Examples include glycosylamines, sugar phosphates, and sugar esters.
  • a Charged Group means a chemical group bearing a positive or a negative charge, such as for example phosphate, phosphonate, sulfate, sulfonate, nitrate, carboxylate, carbonate, and the like.
  • a Charged Group is at least 50% ionized in aqueous solution at least one pH in the range of 5-9.
  • a Charged Group is an anionic Charged Group.
  • Linker or “linker reagent” are used interchangeably and refer to a chemical moiety comprising a covalent bond or a chain of atoms that covalently attaches a targeting ligand to a drug moiety.
  • linkers include a divalent radical.
  • linkers can comprise one or more amino acid residues. The linker can be cleavable or non-cleavable.
  • “Amino Acid Unit” has the formula hydrogen, methyl, isopropyl, isobutyl, sec-butyl, benzyl, p-hydroxybenzyl, —CH2OH, —CH(OH)CH3, — CH 2 SH —CH2CH2SCH3, —CH2CONH2, —CH2COOH, —CH2CH2CONH2, —CH2CH2COOH, —(CH2)3NHC( ⁇ NH)NH2, —(CH2)3NH2, —(CH2)3NHCOCH3, —(CH2)3NHCHO, —(CH 2 ) 4 NHC( ⁇ NH)NH 2 , —(CH 2 ) 4 NH 2 , —(CH 2 ) 4 NHCOCH 3 , —(CH 2 ) 4 NHCHO, —(CH 2 ) 3 NHCONH 2 , —(CH 2 ) 4 NHCONH 2 , —CH 2 CH 2 CH(OH)CH 2 NH 2 ,
  • “Amino Acid Unit” has the formula methyl, isopropyl, isobutyl, sec-butyl, benzyl, p-hydroxybenzyl, —CH2OH, —CH(OH)CH3, — CH 2 CH 2 SCH 3 , —CH 2 CONH 2 , —CH 2 COOH, —CH2CH2CONH2, —CH2CH2COOH, —(CH2)3NHC( ⁇ NH)NH2, —(CH2)3NH2, —CH2SH, —(CH2)3NHCOCH3, —(CH2)3NHCHO, —(CH2)4NHC( ⁇ NH)NH2, —(CH2)4NH2, —(CH2)4NHCOCH3, —(CH2)4NHCHO, —(CH2)3NHCONH2, —(CH2)4NHCONH2, —CH2CH2CH(OH)CH2NH2, 2-pyridylmethyl-, 3-pyridylmethyl-, 4-pyridylmethyl-,
  • Amino Acid Unit has the formula .
  • Amino Acid Unit includes not only naturally occurring amino acids but also minor amino acids and non-naturally occurring amino acid analogs, such as for example, citrulline, norleucine, selenomethionine, ⁇ -amino acids (e.g., ⁇ -alanine, ⁇ -aspargine), and the like.
  • the amino acid can be a modified amino acid, such as for example, ⁇ - amino acid amide, oxazole amino acid, thiazole amino acid, triazole amino acid, and the like.
  • the modified amino acid has the formula .
  • an amino acid unit may be referred to by its standard three-letter code for the amino acid (e.g., Ala, Cys, Asp, Glu etc.).
  • “Peptoid” has the formula , here R 0 is methyl, isopropyl, isobutyl, sec-butyl, benzyl, p-hydroxybenzyl, —CH2OH, —CH(OH)CH3, —CH2CH2SCH3, — CH2CONH2, —CH2COOH, —CH2CH2CONH2, —CH2CH2COOH, — (CH 2 ) 3 NHC( ⁇ NH)NH 2 , —(CH 2 ) 3 NH 2 , —(CH 2 ) 3 NHCOCH 3 , —(CH 2 ) 3 NHCHO, — (CH 2 ) 4 NHC( ⁇ NH)NH 2 , —(CH 2 ) 4 NH 2 , —(CH2)4NHCOCH3, —(CH2)4NHCO
  • peptoid has the formula .
  • the term “half-life extender” refers to molecules that extend half-life of biopharmaceuticals.
  • the half-life of SMDCs described herein can be extended by PEGylation (covalently linking a PEG to the SMDC), lipidation (covalently linking a lipid to the SMDC).
  • the half-life of SMDCs described herein can be extended by covalently linking a cholesterol-like compound to the SMDC.
  • the half-life of SMDCs described herein can be extended by covalently linking a small molecule albumin binder to the SMDC.
  • the term “cholesterol-like” half-life extender refers to a compound with a structure closely resembling cholesterol with a linker on the hydroxyl end. In embodiments, “cholesterol-like” half-life extender refers to .
  • the term “albumin binder” half-life extender refers to a small molecule that can be reversibly (non-covalently) bound by serum albumin. Serum albumin can bind a large diversity of small organic molecules such as fatty acids, dicarboxylic acids, bulky heterocycles, and aromatic carboxylic acids with a peripheric negative charge.
  • bioconjugate and “bioconjugate linker” refers to the resulting association between atoms or molecules of “bioconjugate reactive groups” or “bioconjugate reactive moieties”.
  • the association can be direct or indirect.
  • a conjugate between a first bioconjugate reactive group e.g –NH 2 –C(O)OH –N- hydroxysuccinimide, or –maleimide
  • a second bioconjugate reactive group e.g., sulfhydryl, sulfur-containing amino acid, amine, amine sidechain containing amino acid, or carboxylate
  • covalent bond or linker e.g.
  • bioconjugates or bioconjugate linkers are formed using bioconjugate chemistry (i.e.
  • bioconjugate reactive groups including, but are not limited to nucleophilic substitutions (e.g., reactions of amines and alcohols with acyl halides, active esters), electrophilic substitutions (e.g., enamine reactions) and additions to carbon-carbon and carbon-heteroatom multiple bonds (e.g., Michael reaction, Diels-Alder addition).
  • nucleophilic substitutions e.g., reactions of amines and alcohols with acyl halides, active esters
  • electrophilic substitutions e.g., enamine reactions
  • additions to carbon-carbon and carbon-heteroatom multiple bonds e.g., Michael reaction, Diels-Alder addition.
  • the first bioconjugate reactive group e.g., maleimide moiety
  • the second bioconjugate reactive group e.g. a sulfhydryl
  • the first bioconjugate reactive group (e.g., haloacetyl moiety) is covalently attached to the second bioconjugate reactive group (e.g. a sulfhydryl).
  • the first bioconjugate reactive group (e.g., pyridyl moiety) is covalently attached to the second bioconjugate reactive group (e.g. a sulfhydryl).
  • the first bioconjugate reactive group e.g., –N- hydroxysuccinimide moiety
  • is covalently attached to the second bioconjugate reactive group (e.g. an amine).
  • the first bioconjugate reactive group (e.g., fluorophenyl ester moiety) reacts with the second bioconjugate reactive group (e.g. an amine) to form a covalent bond.
  • the first bioconjugate reactive group (e.g., –sulfo–N- hydroxysuccinimide moiety) reacts with the second bioconjugate reactive group (e.g. an amine) to form a covalent bond.
  • bioconjugate reactive moieties used for bioconjugate chemistries herein include, for example: (a) carboxyl groups and various derivatives thereof including, but not limited to, N-hydroxysuccinimide esters, N-hydroxybenztriazole esters, acid halides, acyl imidazoles, thioesters, p-nitrophenyl esters, alkyl, alkenyl, alkynyl and aromatic esters; (b) hydroxyl groups which can be converted to esters, ethers, aldehydes, etc.
  • haloalkyl groups wherein the halide can be later displaced with a nucleophilic group such as, for example, an amine, a carboxylate anion, thiol anion, carbanion, or an alkoxide ion, thereby resulting in the covalent attachment of a new group at the site of the halogen atom;
  • dienophile groups which are capable of participating in Diels-Alder reactions such as, for example, maleimido or maleimide groups;
  • aldehyde or ketone groups such that subsequent derivatization is possible via formation of carbonyl derivatives such as, for example, imines, hydrazones, semicarbazones or oximes, or via such mechanisms as Grignard addition or alkyllithium addition;
  • sulfonyl halide groups for subsequent reaction with amines, for example, to form sulfonamides;
  • thiol groups which can be converted to disulf
  • bioconjugate reactive groups can be chosen such that they do not participate in, or interfere with, the chemical stability of the conjugate described herein. Alternatively, a reactive functional group can be protected from participating in the crosslinking reaction by the presence of a protecting group.
  • the bioconjugate comprises a molecular entity derived from the reaction of an unsaturated bond, such as a maleimide, and a sulfhydryl group.
  • “Derivative” is a compound that is derived from a similar compound by a chemical reaction. In biochemistry, the word is used for compounds that at least theoretically can be formed from the precursor compound. In the past, derivative also meant a compound that can be imagined to arise from another compound, if one atom or group of atoms is replaced with another atom or group of atoms, but modern chemical language now uses the term structural analog for this meaning, thus eliminating ambiguity.
  • Analog or “analogue” is used in accordance with its plain ordinary meaning within Chemistry and Biology and refers to a chemical compound that is structurally similar to another compound (i.e., a so-called “reference” compound) but differs in composition, e.g., in the replacement of one atom by an atom of a different element, or in the presence of a particular functional group, or the replacement of one functional group by another functional group, or the absolute stereochemistry of one or more chiral centers of the reference compound.
  • reference compound i.e., a so-called “reference” compound
  • folic acid and “folate” are often used interchangeably, though more appropriately, “folic acid” refers to the fully oxidized synthetic compound (pteroylmono- glutamic acid) used in dietary supplements and in food fortification, whereas “folate” refers to the various tetrahydrofolate derivatives naturally present in foods. Reduced folates are found as the partially reduced form 7,8-dihydrofolate or the reduced species 5,6,7,8- tetrahydrofolate (THF).
  • THF 5,6,7,8- tetrahydrofolate
  • folic acid and “folate” are used interchangeably herein to refer to the fully oxidized synthetic compound.
  • alkyl by itself or as part of another substituent, means, unless otherwise stated, a straight (i.e., unbranched) or branched carbon chain (or carbon), or combination thereof, which may be fully saturated, mono- or polyunsaturated and can include mono-, di- and multivalent radicals.
  • the alkyl may include a designated number of carbons (e.g., C1-C10 means one to ten carbons).
  • Alkyl is an uncyclized chain.
  • saturated hydrocarbon radicals include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, methyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like.
  • An unsaturated alkyl group is one having one or more double bonds or triple bonds.
  • Examples of unsaturated alkyl groups include, but are not limited to, vinyl, 2-propenyl, crotyl, 2-isopentenyl, 2-(butadienyl), 2,4- pentadienyl, 3-(1,4-pentadienyl), ethynyl, 1- and 3-propynyl, 3-butynyl, and the higher homologs and isomers.
  • An alkoxy is an alkyl attached to the remainder of the molecule via an oxygen linker (-O-).
  • An alkyl moiety may be an alkenyl moiety.
  • An alkyl moiety may be an alkynyl moiety.
  • An alkyl moiety may be fully saturated.
  • alkenyl may include more than one double bond and/or one or more triple bonds in addition to the one or more double bonds.
  • An alkynyl may include more than one triple bond and/or one or more double bonds in addition to the one or more triple bonds.
  • alkylene by itself or as part of another substituent, means, unless otherwise stated, a divalent radical derived from an alkyl, as exemplified, but not limited by, -CH2CH2CH2CH2-.
  • an alkyl (or alkylene) group will have from 1 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being preferred herein.
  • a “lower alkyl” or “lower alkylene” is a shorter chain alkyl or alkylene group, generally having eight or fewer carbon atoms.
  • alkenylene by itself or as part of another substituent, means, unless otherwise stated, a divalent radical derived from an alkene.
  • heteroalkyl by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched chain, or combinations thereof, including at least one carbon atom and at least one heteroatom (e.g., O, N, P, Si, or S), and wherein the nitrogen and sulfur atoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized.
  • heteroatom(s) e.g., O, N, S, Si, or P
  • the heteroatom(s) may be placed at any interior position of the heteroalkyl group or at the position at which the alkyl group is attached to the remainder of the molecule.
  • Heteroalkyl is an uncyclized chain.
  • a heteroalkyl moiety may include one heteroatom (e.g., O, N, S, Si, or P).
  • a heteroalkyl moiety may include two optionally different heteroatoms (e.g., O, N, S, Si, or P).
  • a heteroalkyl moiety may include three optionally different heteroatoms (e.g., O, N, S, Si, or P).
  • a heteroalkyl moiety may include four optionally different heteroatoms (e.g., O, N, S, Si, or P).
  • a heteroalkyl moiety may include five optionally different heteroatoms (e.g., O, N, S, Si, or P).
  • a heteroalkyl moiety may include up to 8 optionally different heteroatoms (e.g., O, N, S, Si, or P).
  • the term “heteroalkenyl,” by itself or in combination with another term, means, unless otherwise stated, a heteroalkyl including at least one double bond.
  • a heteroalkenyl may optionally include more than one double bond and/or one or more triple bonds in addition to the one or more double bonds.
  • heteroalkynyl by itself or in combination with another term, means, unless otherwise stated, a heteroalkyl including at least one triple bond.
  • heteroalkynyl may optionally include more than one triple bond and/or one or more double bonds in addition to the one or more triple bonds.
  • heteroalkylene by itself or as part of another substituent, means, unless otherwise stated, a divalent radical derived from heteroalkyl, as exemplified, but not limited by, -CH2-CH2-S-CH2-CH2- and -CH2-S-CH2-CH2-NH-CH2-.
  • heteroatoms can also occupy either or both of the chain termini (e.g., alkyleneoxy, alkylenedioxy, alkyleneamino, alkylenediamino, and the like).
  • heteroalkyl groups include those groups that are attached to the remainder of the molecule through a heteroatom, such as -C(O)R', -C(O)NR', -NR'R'', -OR', -SR', and/or -SO2R'.
  • heteroalkyl is recited, followed by recitations of specific heteroalkyl groups, such as - NR'R'' or the like, it will be understood that the terms heteroalkyl and -NR'R'' are not redundant or mutually exclusive. Rather, the specific heteroalkyl groups are recited to add clarity. Thus, the term “heteroalkyl” should not be interpreted herein as excluding specific heteroalkyl groups, such as -NR'R'' or the like. [00109]
  • Cycloalkyl and heterocycloalkyl are not aromatic. Additionally, for heterocycloalkyl, a heteroatom can occupy the position at which the heterocycle is attached to the remainder of the molecule.
  • Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like.
  • heterocycloalkyl examples include, but are not limited to, 1- (1,2,5,6-tetrahydropyridyl), 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-morpholinyl, 3- morpholinyl, tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothien-2-yl, tetrahydrothien-3-yl, 1-piperazinyl, 2-piperazinyl, and the like.
  • the term “cycloalkyl” means a monocyclic, bicyclic, or a multicyclic cycloalkyl ring system.
  • monocyclic ring systems are cyclic hydrocarbon groups containing from 3 to 8 carbon atoms, where such groups can be saturated or unsaturated, but not aromatic.
  • cycloalkyl groups are fully saturated.
  • monocyclic cycloalkyls examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, and cyclooctyl.
  • Bicyclic cycloalkyl ring systems are bridged monocyclic rings or fused bicyclic rings.
  • bridged monocyclic rings contain a monocyclic cycloalkyl ring where two non adjacent carbon atoms of the monocyclic ring are linked by an alkylene bridge of between one and three additional carbon atoms (i.e., a bridging group of the form (CH 2 ) w , where w is 1, 2, or 3).
  • alkylene bridge of between one and three additional carbon atoms
  • a bridging group of the form (CH 2 ) w i.e., a bridging group of the form (CH 2 ) w , where w is 1, 2, or 3).
  • bicyclic ring systems include, but are not limited to, bicyclo[3.1.1]heptane, bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane, bicyclo[3.2.2]nonane, bicyclo[3.3.1]nonane, and bicyclo[4.2.1]nonane.
  • fused bicyclic cycloalkyl ring systems contain a monocyclic cycloalkyl ring fused to either a phenyl, a monocyclic cycloalkyl, a monocyclic cycloalkenyl, a monocyclic heterocyclyl, or a monocyclic heteroaryl.
  • the bridged or fused bicyclic cycloalkyl is attached to the parent molecular moiety through any carbon atom contained within the monocyclic cycloalkyl ring.
  • cycloalkyl groups are optionally substituted with one or two groups which are independently oxo or thia.
  • the fused bicyclic cycloalkyl is a 5 or 6 membered monocyclic cycloalkyl ring fused to either a phenyl ring, a 5 or 6 membered monocyclic cycloalkyl, a 5 or 6 membered monocyclic cycloalkenyl, a 5 or 6 membered monocyclic heterocyclyl, or a 5 or 6 membered monocyclic heteroaryl, wherein the fused bicyclic cycloalkyl is optionally substituted by one or two groups which are independently oxo or thia.
  • multicyclic cycloalkyl ring systems are a monocyclic cycloalkyl ring (base ring) fused to either (i) one ring system selected from the group consisting of a bicyclic aryl, a bicyclic heteroaryl, a bicyclic cycloalkyl, a bicyclic cycloalkenyl, and a bicyclic heterocyclyl; or (ii) two other ring systems independently selected from the group consisting of a phenyl, a bicyclic aryl, a monocyclic or bicyclic heteroaryl, a monocyclic or bicyclic cycloalkyl, a monocyclic or bicyclic cycloalkenyl, and a monocyclic or bicyclic heterocyclyl.
  • multicyclic cycloalkyl is attached to the parent molecular moiety through any carbon atom contained within the base ring.
  • multicyclic cycloalkyl ring systems are a monocyclic cycloalkyl ring (base ring) fused to either (i) one ring system selected from the group consisting of a bicyclic aryl, a bicyclic heteroaryl, a bicyclic cycloalkyl, a bicyclic cycloalkenyl, and a bicyclic heterocyclyl; or (ii) two other ring systems independently selected from the group consisting of a phenyl, a monocyclic heteroaryl, a monocyclic cycloalkyl, a monocyclic cycloalkenyl, and a monocyclic heterocyclyl.
  • a cycloalkyl is a cycloalkenyl.
  • the term “cycloalkenyl” is used in accordance with its plain ordinary meaning.
  • a cycloalkenyl is a monocyclic, bicyclic, or a multicyclic cycloalkenyl ring system.
  • monocyclic cycloalkenyl ring systems are cyclic hydrocarbon groups containing from 3 to 8 carbon atoms, where such groups are unsaturated (i.e., containing at least one annular carbon carbon double bond), but not aromatic.
  • monocyclic cycloalkenyl ring systems include cyclopentenyl and cyclohexenyl.
  • bicyclic cycloalkenyl rings are bridged monocyclic rings or a fused bicyclic rings.
  • bridged monocyclic rings contain a monocyclic cycloalkenyl ring where two non adjacent carbon atoms of the monocyclic ring are linked by an alkylene bridge of between one and three additional carbon atoms (i.e., a bridging group of the form (CH 2 ) w , where w is 1, 2, or 3).
  • alkylene bridge of between one and three additional carbon atoms
  • bicyclic cycloalkenyls include, but are not limited to, norbornenyl and bicyclo[2.2.2]oct 2 enyl.
  • fused bicyclic cycloalkenyl ring systems contain a monocyclic cycloalkenyl ring fused to either a phenyl, a monocyclic cycloalkyl, a monocyclic cycloalkenyl, a monocyclic heterocyclyl, or a monocyclic heteroaryl.
  • the bridged or fused bicyclic cycloalkenyl is attached to the parent molecular moiety through any carbon atom contained within the monocyclic cycloalkenyl ring.
  • cycloalkenyl groups are optionally substituted with one or two groups which are independently oxo or thia.
  • multicyclic cycloalkenyl rings contain a monocyclic cycloalkenyl ring (base ring) fused to either (i) one ring system selected from the group consisting of a bicyclic aryl, a bicyclic heteroaryl, a bicyclic cycloalkyl, a bicyclic cycloalkenyl, and a bicyclic heterocyclyl; or (ii) two ring systems independently selected from the group consisting of a phenyl, a bicyclic aryl, a monocyclic or bicyclic heteroaryl, a monocyclic or bicyclic cycloalkyl, a monocyclic or bicyclic cycloalkenyl, and a monocyclic or bicyclic heterocyclyl.
  • multicyclic cycloalkenyl is attached to the parent molecular moiety through any carbon atom contained within the base ring.
  • multicyclic cycloalkenyl rings contain a monocyclic cycloalkenyl ring (base ring) fused to either (i) one ring system selected from the group consisting of a bicyclic aryl, a bicyclic heteroaryl, a bicyclic cycloalkyl, a bicyclic cycloalkenyl, and a bicyclic heterocyclyl; or (ii) two ring systems independently selected from the group consisting of a phenyl, a monocyclic heteroaryl, a monocyclic cycloalkyl, a monocyclic cycloalkenyl, and a monocyclic heterocyclyl.
  • a heterocycloalkyl is a heterocyclyl.
  • heterocyclyl as used herein, means a monocyclic, bicyclic, or multicyclic heterocycle.
  • the heterocyclyl monocyclic heterocycle is a 3, 4, 5, 6 or 7 membered ring containing at least one heteroatom independently selected from the group consisting of O, N, and S where the ring is saturated or unsaturated, but not aromatic.
  • the 3 or 4 membered ring contains 1 heteroatom selected from the group consisting of O, N and S.
  • the 5 membered ring can contain zero or one double bond and one, two or three heteroatoms selected from the group consisting of O, N and S.
  • the 6 or 7 membered ring contains zero, one or two double bonds and one, two or three heteroatoms selected from the group consisting of O, N and S.
  • the heterocyclyl monocyclic heterocycle is connected to the parent molecular moiety through any carbon atom or any nitrogen atom contained within the heterocyclyl monocyclic heterocycle.
  • heterocyclyl monocyclic heterocycles include, but are not limited to, azetidinyl, azepanyl, aziridinyl, diazepanyl, 1,3-dioxanyl, 1,3-dioxolanyl, 1,3-dithiolanyl, 1,3-dithianyl, imidazolinyl, imidazolidinyl, isothiazolinyl, isothiazolidinyl, isoxazolinyl, isoxazolidinyl, morpholinyl, oxadiazolinyl, oxadiazolidinyl, oxazolinyl, oxazolidinyl, piperazinyl, piperidinyl, pyranyl, pyrazolinyl, pyrazolidinyl, pyrrolinyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydrothienyl
  • the heterocyclyl bicyclic heterocycle is a monocyclic heterocycle fused to either a phenyl, a monocyclic cycloalkyl, a monocyclic cycloalkenyl, a monocyclic heterocycle, or a monocyclic heteroaryl.
  • the heterocyclyl bicyclic heterocycle is connected to the parent molecular moiety through any carbon atom or any nitrogen atom contained within the monocyclic heterocycle portion of the bicyclic ring system.
  • bicyclic heterocyclyls include, but are not limited to, 2,3-dihydrobenzofuran-2-yl, 2,3- dihydrobenzofuran-3-yl, indolin-1-yl, indolin-2-yl, indolin-3-yl, 2,3-dihydrobenzothien-2-yl, decahydroquinolinyl, decahydroisoquinolinyl, octahydro-1H-indolyl, and octahydrobenzofuranyl.
  • heterocyclyl groups are optionally substituted with one or two groups which are independently oxo or thia.
  • the bicyclic heterocyclyl is a 5 or 6 membered monocyclic heterocyclyl ring fused to a phenyl ring, a 5 or 6 membered monocyclic cycloalkyl, a 5 or 6 membered monocyclic cycloalkenyl, a 5 or 6 membered monocyclic heterocyclyl, or a 5 or 6 membered monocyclic heteroaryl, wherein the bicyclic heterocyclyl is optionally substituted by one or two groups which are independently oxo or thia.
  • Multicyclic heterocyclyl ring systems are a monocyclic heterocyclyl ring (base ring) fused to either (i) one ring system selected from the group consisting of a bicyclic aryl, a bicyclic heteroaryl, a bicyclic cycloalkyl, a bicyclic cycloalkenyl, and a bicyclic heterocyclyl; or (ii) two other ring systems independently selected from the group consisting of a phenyl, a bicyclic aryl, a monocyclic or bicyclic heteroaryl, a monocyclic or bicyclic cycloalkyl, a monocyclic or bicyclic cycloalkenyl, and a monocyclic or bicyclic heterocyclyl.
  • multicyclic heterocyclyl is attached to the parent molecular moiety through any carbon atom or nitrogen atom contained within the base ring.
  • multicyclic heterocyclyl ring systems are a monocyclic heterocyclyl ring (base ring) fused to either (i) one ring system selected from the group consisting of a bicyclic aryl, a bicyclic heteroaryl, a bicyclic cycloalkyl, a bicyclic cycloalkenyl, and a bicyclic heterocyclyl; or (ii) two other ring systems independently selected from the group consisting of a phenyl, a monocyclic heteroaryl, a monocyclic cycloalkyl, a monocyclic cycloalkenyl, and a monocyclic heterocyclyl.
  • multicyclic heterocyclyl groups include, but are not limited to 10H-phenothiazin-10-yl, 9,10-dihydroacridin-9-yl, 9,10-dihydroacridin-10-yl, 10H-phenoxazin-10-yl, 10,11-dihydro-5H-dibenzo[b,f]azepin-5-yl, 1,2,3,4- tetrahydropyrido[4,3-g]isoquinolin-2-yl, 12H-benzo[b]phenoxazin-12-yl, and dodecahydro- 1H-carbazol-9-yl.
  • halo or “halogen,” by themselves or as part of another substituent, mean, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom. Additionally, terms such as “haloalkyl” are meant to include monohaloalkyl and polyhaloalkyl.
  • halo(C1-C4)alkyl includes, but is not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like.
  • acyl means, unless otherwise stated, -C(O)R where R is a substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
  • aryl means, unless otherwise stated, a polyunsaturated, aromatic, hydrocarbon substituent, which can be a single ring or multiple rings (preferably from 1 to 3 rings) that are fused together (i.e., a fused ring aryl) or linked covalently.
  • a fused ring aryl refers to multiple rings fused together wherein at least one of the fused rings is an aryl ring.
  • heteroaryl refers to aryl groups (or rings) that contain at least one heteroatom such as N, O, or S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quaternized.
  • heteroaryl includes fused ring heteroaryl groups (i.e., multiple rings fused together wherein at least one of the fused rings is a heteroaromatic ring).
  • a 5,6-fused ring heteroarylene refers to two rings fused together, wherein one ring has 5 members and the other ring has 6 members, and wherein at least one ring is a heteroaryl ring.
  • a 6,6-fused ring heteroarylene refers to two rings fused together, wherein one ring has 6 members and the other ring has 6 members, and wherein at least one ring is a heteroaryl ring.
  • a 6,5-fused ring heteroarylene refers to two rings fused together, wherein one ring has 6 members and the other ring has 5 members, and wherein at least one ring is a heteroaryl ring.
  • a heteroaryl group can be attached to the remainder of the molecule through a carbon or heteroatom.
  • Non-limiting examples of aryl and heteroaryl groups include phenyl, naphthyl, pyrrolyl, pyrazolyl, pyridazinyl, triazinyl, pyrimidinyl, imidazolyl, pyrazinyl, purinyl, oxazolyl, isoxazolyl, thiazolyl, furyl, thienyl, pyridyl, pyrimidyl, benzothiazolyl, benzoxazoyl benzimidazolyl, benzofuran, isobenzofuranyl, indolyl, isoindolyl, benzothiophenyl, isoquinolyl, quinoxalinyl, quinolyl, 1-naphthyl, 2- naphthyl, 4-biphenyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4- imidazolyl
  • Substituents for each of the above noted aryl and heteroaryl ring systems are selected from the group of acceptable substituents described below.
  • a heteroaryl group substituent may be -O- bonded to a ring heteroatom nitrogen.
  • a fused ring heterocyloalkyl-aryl is an aryl fused to a heterocycloalkyl.
  • a fused ring heterocycloalkyl-heteroaryl is a heteroaryl fused to a heterocycloalkyl.
  • a fused ring heterocycloalkyl-cycloalkyl is a heterocycloalkyl fused to a cycloalkyl.
  • a fused ring heterocycloalkyl-heterocycloalkyl is a heterocycloalkyl fused to another heterocycloalkyl.
  • Fused ring heterocycloalkyl-aryl, fused ring heterocycloalkyl-heteroaryl, fused ring heterocycloalkyl-cycloalkyl, or fused ring heterocycloalkyl-heterocycloalkyl may each independently be unsubstituted or substituted with one or more of the substitutents described herein.
  • Spirocyclic rings are two or more rings wherein adjacent rings are attached through a single atom.
  • the individual rings within spirocyclic rings may be identical or different.
  • Individual rings in spirocyclic rings may be substituted or unsubstituted and may have different substituents from other individual rings within a set of spirocyclic rings.
  • Possible substituents for individual rings within spirocyclic rings are the possible substituents for the same ring when not part of spirocyclic rings (e.g. substituents for cycloalkyl or heterocycloalkyl rings).
  • Spirocylic rings may be substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloalkylene, substituted or unsubstituted heterocycloalkyl or substituted or unsubstituted heterocycloalkylene and individual rings within a spirocyclic ring group may be any of the immediately previous list, including having all rings of one type (e.g. all rings being substituted heterocycloalkylene wherein each ring may be the same or different substituted heterocycloalkylene).
  • heterocyclic spirocyclic rings means a spirocyclic rings wherein at least one ring is a heterocyclic ring and wherein each ring may be a different ring.
  • substituted spirocyclic rings means that at least one ring is substituted and each substituent may optionally be different.
  • alkylsulfonyl means a moiety having the formula -S(O2)-R', where R' is a substituted or unsubstituted alkyl group as defined above. R' may have a specified number of carbons (e.g., “C1-C4 alkylsulfonyl”).
  • alkylarylene as an arylene moiety covalently bonded to an alkylene moiety (also referred to herein as an alkylene linker). In embodiments, the alkylarylene group has the formula: .
  • An alkylarylene moiety may be substituted (e.g.
  • the alkylarylene is unsubstituted.
  • Each of the above terms e.g., “alkyl,” “heteroalkyl,” “cycloalkyl,” “heterocycloalkyl,” “aryl,” and “heteroaryl” includes both substituted and unsubstituted forms of the indicated radical. Preferred substituents for each type of radical are provided below.
  • R, R', R'', R'', and R''' each preferably independently refer to hydrogen, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl (e.g., aryl substituted with 1-3 halogens), substituted or unsubstituted heteroaryl, substituted or unsubstituted alkyl, alkoxy, or thioalkoxy groups, or arylalkyl groups.
  • aryl e.g., aryl substituted with 1-3 halogens
  • substituted or unsubstituted heteroaryl substituted or unsubstituted alkyl, alkoxy, or thioalkoxy groups, or arylalkyl groups.
  • each of the R groups is independently selected as are each R', R'', R''', and R''' group when more than one of these groups is present.
  • R' and R'' are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a 4-, 5-, 6-, or 7- membered ring.
  • -NR'R'' includes, but is not limited to, 1-pyrrolidinyl and 4- morpholinyl.
  • alkyl is meant to include groups including carbon atoms bound to groups other than hydrogen groups, such as haloalkyl (e.g., -CF 3 and -CH 2 CF 3 ) and acyl (e.g., - C(O)CH3, -C(O)CF3, -C(O)CH2OCH3, and the like).
  • haloalkyl e.g., -CF 3 and -CH 2 CF 3
  • acyl e.g., - C(O)CH3, -C(O)CF3, -C(O)CH2OCH3, and the like.
  • each of the R groups is independently selected as are each R', R'', R'', and R''' groups when more than one of these groups is present.
  • Substituents for rings e.g. cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene
  • substituents on the ring may be depicted as substituents on the ring rather than on a specific atom of a ring (commonly referred to as a floating substituent).
  • the substituent may be attached to any of the ring atoms (obeying the rules of chemical valency) and in the case of fused rings or spirocyclic rings, a substituent depicted as associated with one member of the fused rings or spirocyclic rings (a floating substituent on a single ring), may be a substituent on any of the fused rings or spirocyclic rings (a floating substituent on multiple rings).
  • the multiple substituents may be on the same atom, same ring, different atoms, different fused rings, different spirocyclic rings, and each substituent may optionally be different.
  • a point of attachment of a ring to the remainder of a molecule is not limited to a single atom (a floating substituent)
  • the attachment point may be any atom of the ring and in the case of a fused ring or spirocyclic ring, any atom of any of the fused rings or spirocyclic rings while obeying the rules of chemical valency.
  • a ring, fused rings, or spirocyclic rings contain one or more ring heteroatoms and the ring, fused rings, or spirocyclic rings are shown with one more floating substituents (including, but not limited to, points of attachment to the remainder of the molecule), the floating substituents may be bonded to the heteroatoms.
  • the ring heteroatoms are shown bound to one or more hydrogens (e.g. a ring nitrogen with two bonds to ring atoms and a third bond to a hydrogen) in the structure or formula with the floating substituent, when the heteroatom is bonded to the floating substituent, the substituent will be understood to replace the hydrogen, while obeying the rules of chemical valency.
  • Two or more substituents may optionally be joined to form aryl, heteroaryl, cycloalkyl, or heterocycloalkyl groups.
  • Such so-called ring-forming substituents are typically, though not necessarily, found attached to a cyclic base structure.
  • the ring-forming substituents are attached to adjacent members of the base structure.
  • two ring-forming substituents attached to adjacent members of a cyclic base structure create a fused ring structure.
  • the ring-forming substituents are attached to a single member of the base structure.
  • two ring-forming substituents attached to a single member of a cyclic base structure create a spirocyclic structure.
  • the ring-forming substituents are attached to non- adjacent members of the base structure.
  • Two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally form a ring of the formula -T-C(O)-(CRR')p-U-, wherein T and U are independently -NR-, -O-, -CRR'-, or a single bond, and p is an integer of from 0 to 3.
  • two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula -A-(CH2)r-B-, wherein A and B are independently -CRR'-, -O-, -NR-, -S-, -S(O) -, -S(O) 2 -, -S(O) 2 NR'-, or a single bond, and r is an integer of from 1 to 4.
  • One of the single bonds of the new ring so formed may optionally be replaced with a double bond.
  • two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula - (CRR')s-X'- (C''R''')d-, where s and d are independently integers of from 0 to 3, and X' is - O-, -NR'-, -S-, -S(O)-, -S(O)2-, or -S(O)2NR'-.
  • R, R', R'', and R''' are preferably independently selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl.
  • heteroatom or “ring heteroatom” are meant to include oxygen (O), nitrogen (N), sulfur (S), phosphorus (P), and silicon (Si).
  • a “substituent group,” as used herein, means a group selected from the following moieties: (A) oxo, halogen, -CCl3, -CBr3, -CF3, -CI3, -CH2Cl, -CH2Br, -CH2F, -CH2I, -CHCl2, -CHBr2, -CHF2, -CHI2, -CN, -OH, -NH2, -COOH, -CONH2, -NO2, -SH, -SO3H, -SO 4 H, -SO 2 NH 2 , ⁇ NHNH 2 , ⁇ ONH 2 , ⁇ NHC(O)NHNH 2 , -NHC(O)NH 2 , -NHSO 2 H, -NHC(O)H, -NHC(O)OH, -NHOH, -OCCl 3 , -OCF 3 , -OCBr 3 , -OCI 3
  • a “size-limited substituent” or “ size-limited substituent group,” as used herein, means a group selected from all of the substituents described above for a “substituent group,” wherein each substituted or unsubstituted alkyl is a substituted or unsubstituted C1-C20 alkyl, each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 20 membered heteroalkyl, each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C3-C8 cycloalkyl, each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 3 to 8 membered heterocycloalkyl, each substituted or unsubstituted aryl is a substituted or unsubstituted C 6 -C 10 aryl, and each substituted or unsubstituted heteroaryl is
  • a “lower substituent” or “ lower substituent group,” as used herein, means a group selected from all of the substituents described above for a “substituent group,” wherein each substituted or unsubstituted alkyl is a substituted or unsubstituted C 1 -C 8 alkyl, each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 8 membered heteroalkyl, each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C3- C7 cycloalkyl, each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 3 to 7 membered heterocycloalkyl, each substituted or unsubstituted aryl is a substituted or unsubstituted phenyl, and each substituted or unsubstituted heteroaryl is a substituted or unsubstitute
  • each substituted group described in the compounds herein is substituted with at least one substituent group. More specifically, in some embodiments, each substituted alkyl, substituted heteroalkyl, substituted cycloalkyl, substituted heterocycloalkyl, substituted aryl, substituted heteroaryl, substituted alkylene, substituted heteroalkylene, substituted cycloalkylene, substituted heterocycloalkylene, substituted arylene, and/or substituted heteroarylene described in the compounds herein are substituted with at least one substituent group. In other embodiments, at least one or all of these groups are substituted with at least one size-limited substituent group.
  • each substituted or unsubstituted alkyl may be a substituted or unsubstituted C 1 -C 20 alkyl
  • each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 20 membered heteroalkyl
  • each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C3-C8 cycloalkyl
  • each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 3 to 8 membered heterocycloalkyl
  • each substituted or unsubstituted aryl is a substituted or unsubstituted C 6 - C10 aryl
  • each substituted or unsubstituted heteroaryl is a substituted or unsubstituted or unsubstituted
  • each substituted or unsubstituted alkylene is a substituted or unsubstituted C 1 -C 20 alkylene
  • each substituted or unsubstituted heteroalkylene is a substituted or unsubstituted 2 to 20 membered heteroalkylene
  • each substituted or unsubstituted cycloalkylene is a substituted or unsubstituted C 3 -C 8 cycloalkylene
  • each substituted or unsubstituted heterocycloalkylene is a substituted or unsubstituted 3 to 8 membered heterocycloalkylene
  • each substituted or unsubstituted arylene is a substituted or unsubstituted C6-C10 arylene
  • each substituted or unsubstituted heteroarylene is a substituted or unsubstituted 5 to 10 membered heteroarylene.
  • each substituted or unsubstituted alkyl is a substituted or unsubstituted C 1 -C 8 alkyl
  • each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 8 membered heteroalkyl
  • each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C3-C7 cycloalkyl
  • each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 3 to 7 membered heterocycloalkyl
  • each substituted or unsubstituted aryl is a substituted or unsubstituted C6-C10 aryl
  • each substituted or unsubstituted heteroaryl is a substituted or unsubstituted 5 to 9 membered heteroaryl.
  • each substituted or unsubstituted alkylene is a substituted or unsubstituted C 1 -C 8 alkylene
  • each substituted or unsubstituted heteroalkylene is a substituted or unsubstituted 2 to 8 membered heteroalkylene
  • each substituted or unsubstituted cycloalkylene is a substituted or unsubstituted C 3 -C 7 cycloalkylene
  • each substituted or unsubstituted heterocycloalkylene is a substituted or unsubstituted 3 to 7 membered heterocycloalkylene
  • each substituted or unsubstituted arylene is a substituted or unsubstituted C6-C10 arylene
  • each substituted or unsubstituted heteroarylene is a substituted or unsubstituted 5 to 9 membered heteroarylene.
  • the compound is a chemical species set forth in the Examples section, figures, or tables below.
  • a substituted or unsubstituted moiety e.g., substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, substituted or unsubstituted cycloalkylene, substituted or unsubstituted heterocycloalkylene, substituted or unsubstituted arylene, and/or substituted or unsubstituted heteroarylene) is unsubstituted (e.g., is an unsubstituted alkyl, unsubstituted cycloalkyl, substituted
  • a substituted or unsubstituted moiety e.g., substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, substituted or unsubstituted cycloalkylene, substituted or unsubstituted heterocycloalkylene, substituted or unsubstituted arylene, and/or substituted or unsubstituted heteroarylene) is substituted (e.g., is a substituted alkyl, substituted heteroalkyl, substituted cycloalkyl, substituted heterocycloalkyl, substituted aryl, substituted heteroaryl, substituted alky
  • a substituted moiety e.g., substituted alkyl, substituted heteroalkyl, substituted cycloalkyl, substituted heterocycloalkyl, substituted aryl, substituted heteroaryl, substituted alkylene, substituted heteroalkylene, substituted cycloalkylene, substituted heterocycloalkylene, substituted arylene, and/or substituted heteroarylene
  • is substituted with at least one substituent group wherein if the substituted moiety is substituted with a plurality of substituent groups, each substituent group may optionally be different. In embodiments, if the substituted moiety is substituted with a plurality of substituent groups, each substituent group is different.
  • a substituted moiety e.g., substituted alkyl, substituted heteroalkyl, substituted cycloalkyl, substituted heterocycloalkyl, substituted aryl, substituted heteroaryl, substituted alkylene, substituted heteroalkylene, substituted cycloalkylene, substituted heterocycloalkylene, substituted arylene, and/or substituted heteroarylene
  • is substituted with at least one size-limited substituent group wherein if the substituted moiety is substituted with a plurality of size-limited substituent groups, each size-limited substituent group may optionally be different.
  • each size-limited substituent group is different.
  • a substituted moiety e.g., substituted alkyl, substituted heteroalkyl, substituted cycloalkyl, substituted heterocycloalkyl, substituted aryl, substituted heteroaryl, substituted alkylene, substituted heteroalkylene, substituted cycloalkylene, substituted heterocycloalkylene, substituted arylene, and/or substituted heteroarylene
  • each lower substituent group is different.
  • a substituted moiety e.g., substituted alkyl, substituted heteroalkyl, substituted cycloalkyl, substituted heterocycloalkyl, substituted aryl, substituted heteroaryl, substituted alkylene, substituted heteroalkylene, substituted cycloalkylene, substituted heterocycloalkylene, substituted arylene, and/or substituted heteroarylene
  • substituted moiety is substituted with a plurality of groups selected from substituent groups, size-limited substituent groups, and lower substituent groups; each substituent group, size-limited substituent group, and/or lower substituent group is different.
  • the phrase “or combinations thereof” means that any two or more of the items in the preceding list may be combined, and optionally repeated, in any order and in any orientation.
  • q q Examorpholine morpholine analog of exatecan the structure of which is shown in Example S6, and which is also referred to as compound 50 FA Folic acid Compositions Small Molecule-Drug Conjugates
  • L is a multivalent linker bound at least to a folic acid and a drug moiety
  • HL is a half-life extender
  • D is a drug moiety.
  • L is a multivalent linker bound to a folic acid, a drug moiety, and HL; wherein: HL is a half-life extender; and D is a drug moiety.
  • HL is a half-life extender
  • D is a drug moiety.
  • a compound of Formula (I): or a pharmaceutically acceptable salt thereof wherein: L is a multivalent linker bound to a folic acid, a drug moiety, and HL; wherein: HL is a half-life extender; and D is a drug moiety.
  • a compound of Formula (II): or a pharmaceutically acceptable salt thereof wherein: L is a multivalent linker bound at least to a folic acid and a drug moiety; HL is a half-life extender; and D is a drug moiety.
  • a compound of Formula (II): or a pharmaceutically acceptable salt thereof wherein: L is a multivalent linker bound to a folic acid, a drug moiety, and HL wherein HL is a half-life extender; and D is a drug moiety.
  • Formulae (I) and (II) encompass tautomers, a mixture of two or more tautomers, isotopic variants, and/or a pharmaceutically acceptable salt, solvate, or hydrate thereof.
  • D is a tubulin inhibitor or disruptor, apoptosis inducer, RNA splicing inhibitor, kinase inhibitor, DNA damaging agent, nicotinamide phosphoribosyltransferase inhibitor (NAMPT), peptidic proteasome inhibitors, transcription inhibitors, or proteolysis-targeting chimera (PROTAC).
  • D is a tubulin inhibitor or disruptor, kinase inhibitor, DNA damaging agent, transcription inhibitors, or proteolysis-targeting chimera (PROTAC).
  • D is a tubulin inhibitor.
  • D is a tubulin disruptor.
  • D is a kinase inhibitor.
  • D is a DNA damaging agent.
  • D is a transcription inhibitor.
  • D is a proteolysis-targeting chimera (PROTAC).
  • D is a pyrrolobenzodiazepine, indolinobenzodiazepine, duocarmycin, tubulysin, cryptomycin, anthracycline, maytansinoid, auristatin, carmaphycin, calicheamicin, camptothecin, thailanstatin and analogues, RNA polymerase II inhibitor, topoisomerase I inhibitor, tyrosine kinase inhibitor, Bcl-xL Inhibitor, EG5 inhibitor, or MEK inhibitor.
  • D is a pyrrolobenzodiazepine, duocarmycin, anthracycline, maytansinoid, auristatin, calicheamicin, camptothecin, RNA polymerase II inhibitor, topoisomerase I inhibitor, tyrosine kinase inhibitor, EG5 inhibitor, or MEK inhibitor.
  • D is a pyrrolobenzodiazepine.
  • D is a duocarmycin.
  • D is an anthracycline.
  • D is a maytansinoid.
  • D is an auristatin.
  • D is a calicheamicin.
  • D is a camptothecin. In embodiments, D is a topoisomerase I inhibitor. In embodiments, D is an RNA polymerase II inhibitor. In embodiments, D is a tyrosine kinase inhibitor. In embodiments, D is an EG5 inhibitor. In embodiments, D is a MEK inhibitor.
  • D is MMAE.
  • D is MMAF.
  • D is Duo5. In embodiments, D is PNU-159682. In embodiments, D is SN-38. In embodiments, D is irinotecan. In embodiments, D is amatoxin. In embodiments, D is maytansine. In embodiments, D is exatecan. In embodiments, D is trametinib. In embodiments, D is abemaciclib. In embodiments, D is Palbociclib. In some embodiments, D is a morpholine analog of exatecan. In embodiments, D is examorpholine (the morpholine analog of exatecan for which the structure is shown in Example S6, and which is also referred to as compound 50). [00157] In embodiments, wherein the wavy line indicates a bond to the multivalent linker (L). In embodiments, D is wherein the wavy line indicates a bond to the multivalent linker (L). In embodiments, D is wherein the wavy line indicates a bond to the multivalent linker (L). In embodiments,
  • L is a cleavable or a non-cleavable linker as described in US Patents Nos. US 9,884,127, US 9,981,046, US 9,801,951, US 10,117,944, US 10,590,165, and US 10,590,165, and US Patent publications Nos. US 2017/0340750, and US 2018/0360985, all of which are incorporated herein in their entireties.
  • L is a bond, -C(O)-, -NH-, Amino Acid Unit, Peptoid, –(CH 2 CH 2 O) n –, –(CH 2 ) n –, –(4-aminobenzyloxycarbonyl)–, –(C(O)CH 2 CH 2 C(O))–, thereof; wherein n is an integer from 1 to 24; each R 2 and R 3 is independently H or substituted or unsubstituted alkyl.
  • n is an integer from 1 to 24. In embodiments, n is 1. In embodiments, n is 2. In embodiments, n is 3. In embodiments, n is 4.
  • n is 5. In embodiments, n is 6. In embodiments, n is 7. In embodiments, n is 8. In embodiments, n is 9. In embodiments, n is 10. In embodiments, n is 11. In embodiments, n is 12. In embodiments, n is 13. In embodiments, n is 14. In embodiments, n is 15. In embodiments, n is 16. In embodiments, n is 17. In embodiments, n is 18. In embodiments, n is 19. In embodiments, n is 20. In embodiments, n is 21. In embodiments, n is 22. In embodiments, n is 23. In embodiments, n is 24.
  • each R 2 and R 3 is independently H or substituted or unsubstituted alkyl (e.g., C1-C8 alkyl, C1-C6 alkyl, or C1-C4 alkyl). In embodiments, each R 2 and R 3 is independently H. In embodiments, each R 2 and R 3 is independently substituted or unsubstituted alkyl. In embodiments, each R 2 and R 3 is independently substituted or unsubstituted alkyl (e.g., C1-C8 alkyl, C1-C6 alkyl, or C1-C4 alkyl).
  • each R 2 and R 3 is independently unsubstituted alkyl (e.g., C1-C8 alkyl, C1-C6 alkyl, or C1-C4 alkyl). In embodiments, each R 2 and R 3 is independently substituted alkyl (e.g., C 1 -C 8 alkyl, C 1 -C 6 alkyl, or C1-C4 alkyl).
  • each R 2 and R 3 is independently H or substituted (e.g., substituted with at least one substituent group, size-limited substituent group, or lower substituent group) or unsubstituted alkyl (e.g., C 1 -C 8 alkyl, C 1 -C 6 alkyl, or C 1 -C 4 alkyl).
  • each R 2 and R 3 is independently substituted (e.g., substituted with at least one substituent group, size-limited substituent group, or lower substituent group) or unsubstituted alkyl.
  • each R 2 and R 3 is independently substituted (e.g., substituted with at least one substituent group, size-limited substituent group, or lower substituent group) or unsubstituted alkyl (e.g., C1-C8 alkyl, C1-C6 alkyl, or C1-C4 alkyl). In embodiments, each R 2 and R 3 is independently unsubstituted alkyl (e.g., C 1 -C 8 alkyl, C 1 -C 6 alkyl, or C 1 -C 4 alkyl).
  • each R 2 and R 3 is independently substituted (e.g., substituted with at least one substituent group, size-limited substituent group, or lower substituent group) alkyl (e.g., C1- C 8 alkyl, C 1 -C 6 alkyl, or C 1 -C 4 alkyl).
  • each R 2 and R 3 is independently methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, isopentyl, or hexyl.
  • each R 2 and R 3 is independently methyl.
  • each R 2 and R 3 is independently ethyl.
  • each R 2 and R 3 is independently propyl. In embodiments, each R 2 and R 3 is independently butyl.
  • L is -C(O)-, -NH-,–(CH2CH2O)n–, –(CH2)n–, -Cys-, -Asp-, -Arg- , -Val-, –(4-aminobenzyloxycarbonyl)–, -Glu-, -citrulline- (-Cit-), -Lys-, – (C(O)CH 2 CH 2 NH)–, –(C(O)CH2CH2C(O))–, –(C(O)(CH2)nNH)–, –S(CH2)nO–, –(N(R 2 )(CH2)nN(R 3 ))–, thereof.
  • L is -C(O)-, -NH-, –(CH 2 CH 2 O) n –, –(CH 2 ) n –, –SCH 2 CH 2 O–, –(C(O)CH 2 CH 2 C(O))–, -Cys-, -Val-, -Cit-, -Arg-, -Asp-, -Lys-, –(4- aminobenzyloxycarbonyl) , , , , r combinations thereof.
  • L is -C(O)-.
  • L is -NH-.
  • L is –(CH2CH2O)n–.
  • L is –(CH2)n–. In embodiments, L is –SCH2CH2O–. In embodiments, L is –(C(O)CH2CH2C(O))–. In embodiments, L is -Cys-. In embodiments, L is -Val-. In embodiments, L is -Cit-. In embodiments, L is -Arg-. In embodiments, L is -Asp-. In embodiments, L is -Lys-. In embodiments, L is –(4-aminobenzyloxycarbonyl)–. In embodiments, L is . In embodiments, . embodiments, L is . , . , .
  • L is . embodiments . embodiments, L is . [00170]
  • HL is cholesterol-like half-life extender or albumin binder half- life extender.
  • HL is cholesterol-like half-life extender.
  • HL is an albumin binder half-life extender.
  • HL is . In embodiments, HL is
  • compositions, Uses, and Methods of Use [00182]
  • a pharmaceutical composition including a compound of Formula (I) or (II) (an SMDC) as described herein, including embodiment
  • the SMDC as described herein is included in a therapeutically effective amount.
  • a pharmaceutical composition comprising a compound provided herein, including a compound of Formula (I) or (II), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and a pharmaceutically acceptable carrier (or excipient).
  • the pharmaceutical composition may include optical isomers, diastereomers, enantiomers, isoforms, polymorphs, hydrates, solvates or products, or pharmaceutically acceptable salts of the compound described herein.
  • the compound provided herein may be administered alone, or in combination with one or more other compounds.
  • the pharmaceutical compositions that comprise a compound provided herein, e.g., a compound of Formula (I) or (II) can be formulated in various dosage forms for oral, parenteral, and topical administration.
  • compositions can also be formulated as modified release dosage forms, including delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated-, fast-, targeted-, programmed-release, and gastric retention dosage forms.
  • modified release dosage forms including delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated-, fast-, targeted-, programmed-release, and gastric retention dosage forms.
  • These dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art (see, Remington: The Science and Practice of Pharmacy, supra; Modified-Release Drug Delivery Technology, 2 nd Edition, Rathbone et al., Eds., Marcel Dekker, Inc.: New York, NY, 2008).
  • the pharmaceutical composition may be formulated for oral administration, suppository administration, topical administration, intravenous administration, intraperitoneal administration, intramuscular administration, intralesional administration, intrathecal administration, intranasal administration, subcutaneous administration, implantation, transdermal administration, or transmucosal administration as described herein.
  • the pharmaceutical compositions provided herein are formulated in a dosage form for oral administration, which comprise a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more pharmaceutically acceptable excipients or carriers.
  • the pharmaceutical compositions provided herein are formulated as a suspension for oral administration, which comprise a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more pharmaceutically acceptable excipients or carriers.
  • a compound provided herein e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more pharmaceutically acceptable excip
  • the suspension provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more excipients or carriers selected from the group consisting of water, glycerin, sorbitol, sodium saccharin, xanthan gum, flavoring, citric acid, sodium citrate, methylparaben, propylparaben, and potassium sorbate.
  • a compound provided herein e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or
  • the suspension provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and water, glycerin, sorbitol, sodium saccharin, xanthan gum, flavoring, citric acid, sodium citrate, methylparaben, propylparaben, and potassium sorbate.
  • a compound provided herein e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or
  • the pharmaceutical compositions provided herein are formulated in a dosage form for parenteral administration, which comprise a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more pharmaceutically acceptable excipients or carriers.
  • the pharmaceutical compositions provided herein are formulated in a dosage form for intravenous administration.
  • the pharmaceutical compositions provided herein are formulated in a dosage form for intramuscular administration. In yet another embodiment, the pharmaceutical compositions provided herein are formulated in a dosage form for subcutaneous administration. [00190] In yet another embodiment, the pharmaceutical compositions provided herein are formulated in a dosage form for topical administration, which comprise a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more pharmaceutically acceptable excipients or carriers.
  • a compound provided herein e.g., a compound of Formula (I) or (II)
  • an enantiomer e.g., a mixture of enantiomers, a mixture of two or more
  • the pharmaceutical compositions provided herein are formulated as a cream for topical administration, which comprise a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more pharmaceutically acceptable excipients or carriers.
  • the cream provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more excipients or carriers selected from the group consisting of water, octyldodecanol, mineral oil, stearyl alcohol, cocamide DEA, polysorbate 60, myristyl alcohol, sorbitan monostearate, lactic acid, and benzyl alcohol.
  • a compound provided herein e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer
  • the cream provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and water, octyldodecanol, mineral oil, stearyl alcohol, cocamide DEA, polysorbate 60, myristyl alcohol, sorbitan monostearate, lactic acid, and benzyl alcohol.
  • a compound provided herein e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or iso
  • the pharmaceutical compositions provided herein are formulated as a gel for topical administration, which comprise a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more pharmaceutically acceptable excipients or carriers.
  • a compound provided herein e.g., a compound of Formula (I) or (II)
  • an enantiomer e.g., a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof
  • a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof e.
  • the gel provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more excipients or carriers selected from the group consisting of water, isopropyl alcohol, octyldodecanol, dimethicone copolyol 190, carbomer 980, sodium hydroxide, and docusate sodium.
  • a compound provided herein e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more
  • the gel provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and water, isopropyl alcohol, octyldodecanol, dimethicone copolyol 190, carbomer 980, sodium hydroxide, and docusate sodium.
  • a compound provided herein e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutical
  • the pharmaceutical compositions provided herein are formulated as a shampoo for topical administration, which comprise a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more pharmaceutically acceptable excipients or carriers.
  • the shampoo provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more excipients or carriers selected from the group consisting of water, sodium laureth sulfate, disodium laureth sulfosuccinate, sodium chloride, and laureth-2.
  • a compound provided herein e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutical
  • the shampoo provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and water, sodium laureth sulfate, disodium laureth sulfosuccinate, sodium chloride, and laureth-2.
  • a compound provided herein e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof
  • the pharmaceutical compositions provided herein are formulated as a lacquer for topical administration, which comprise a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more pharmaceutically acceptable excipients or carriers.
  • a compound provided herein e.g., a compound of Formula (I) or (II)
  • an enantiomer e.g., a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof
  • a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof
  • the lacquer provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more excipients or carriers selected from the group consisting of ethyl acetate, isopropyl alcohol, and butyl monoester of poly(methylvinyl ether/maleic acid) in isopropyl alcohol.
  • a compound provided herein e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or is
  • the lacquer provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and ethyl acetate, isopropyl alcohol, and butyl monoester of poly(methylvinyl ether/maleic acid) in isopropyl alcohol.
  • a compound provided herein e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solv
  • compositions provided herein can be provided in a unit- dosage form or multiple-dosage form.
  • a unit-dosage form refers to physically discrete a unit suitable for administration to a human and animal subject, and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of an active ingredient(s) sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carriers or excipients. Examples of a unit- dosage form include an ampoule, syringe, and individually packaged tablet and capsule. A unit-dosage form may be administered in fractions or multiples thereof.
  • a multiple-dosage form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dosage form.
  • Examples of a multiple-dosage form include a vial, bottle of tablets or capsules, or bottle of pints or gallons.
  • the pharmaceutical compositions provided herein can be administered at once, or multiple times at intervals of time. It is understood that the precise dosage and duration of treatment may vary with the age, weight, and condition of the patient being treated, and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test or diagnostic data.
  • compositions include those suitable for oral, rectal, nasal, topical (including transdermal, buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration or administration via an implant.
  • the compositions may be prepared by any method well known in the art of pharmacy.
  • Such methods include the step of bringing in association compounds of this disclosure or combinations thereof with any auxiliary agent.
  • auxiliary agent(s) also named accessory ingredient(s)
  • auxiliary agent(s) include those conventional in the art, such as carriers, fillers, binders, diluents, disintegrants, lubricants, colorants, flavouring agents, anti-oxidants, and wetting agents.
  • Pharmaceutical compositions suitable for oral administration may be presented as discrete dosage units such as pills, tablets, cachets, dragées, lozenges, or capsules, or as a powder or granules, or as a solution or suspension.
  • the active ingredient may also be presented as a bolus or paste.
  • the compositions can further be processed into a suppository or enema for rectal administration.
  • compositions include aqueous and non- aqueous sterile injection.
  • the compositions may be presented in unit-dose or multi-dose containers, for example sealed vials and ampoules, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of sterile liquid carrier, for example water, prior to use.
  • sterile liquid carrier for example water
  • transdermal administration e.g. gels, patches or sprays can be contemplated.
  • Compositions or formulations suitable for pulmonary administration e.g. by nasal inhalation include fine dusts or mists which may be generated by means of metered dose pressurized aerosols, nebulisers or insufflators.
  • the exact dose and regimen of administration of the composition will necessarily be dependent upon the therapeutic or nutritional effect to be achieved and may vary with the particular formula, the route of administration, and the age and condition of the individual subject to whom the composition is to be administered.
  • the therapeutically effective amount for each active compound can vary with factors including but not limited to the activity of the compound used, stability of the active compound in the patient’s body, the severity of the conditions to be alleviated, the total weight of the patient treated, the route of administration, the ease of absorption, distribution, and excretion of the active compound by the body, the age and sensitivity of the patient to be treated, and the like, as will be apparent to a skilled artisan.
  • the amount of administration can be adjusted as the various factors change over time.
  • the active compounds can be incorporated into a formulation that includes pharmaceutically acceptable carriers such as binders (e.g., gelatin, cellulose, gum tragacanth), excipients (e.g., starch, lactose), lubricants (e.g., magnesium stearate, silicon dioxide), disintegrating agents (e.g., alginate, Primogel, and corn starch), and sweetening or flavoring agents (e.g., glucose, sucrose, saccharin, methyl salicylate, and peppermint).
  • binders e.g., gelatin, cellulose, gum tragacanth
  • excipients e.g., starch, lactose
  • lubricants e.g., magnesium stearate, silicon dioxide
  • disintegrating agents e.g., alginate, Primogel, and corn starch
  • sweetening or flavoring agents e.g., glucose, sucrose, saccharin, methyl salicylate, and peppermint
  • the capsules and tablets can also be coated with various coatings known in the art to modify the flavors, tastes, colors, and shapes of the capsules and tablets.
  • liquid carriers such as fatty oil can also be included in capsules.
  • Suitable oral formulations can also be in the form of suspension, syrup, chewing gum, wafer, elixir, and the like. If desired, conventional agents for modifying flavors, tastes, colors, and shapes of the special forms can also be included.
  • the active compounds can be dissolved in an acceptable lipophilic vegetable oil vehicle such as olive oil, corn oil and safflower oil.
  • the active compounds can also be administered parenterally in the form of solution or suspension, or in lyophilized form capable of conversion into a solution or suspension form before use.
  • diluents or pharmaceutically acceptable carriers such as sterile water and physiological saline buffer can be used.
  • Other conventional solvents, pH buffers, stabilizers, anti-bacteria agents, surfactants, and antioxidants can all be included.
  • useful components include sodium chloride, acetates, citrates or phosphates buffers, glycerin, dextrose, fixed oils, methyl parabens, polyethylene glycol, propylene glycol, sodium bisulfate, benzyl alcohol, ascorbic acid, and the like.
  • parenteral formulations can be stored in any conventional containers such as vials and ampoules.
  • Routes of topical administration include nasal, buccal, mucosal, rectal, or vaginal applications.
  • the active compounds can be formulated into lotions, creams, ointments, gels, powders, pastes, sprays, suspensions, drops and aerosols.
  • one or more thickening agents, humectants, and stabilizing agents can be included in the formulations. Examples of such agents include, but are not limited to, polyethylene glycol, sorbitol, xanthan gum, petrolatum, beeswax, or mineral oil, lanolin, squalene, and the like.
  • a special form of topical administration is delivery by a transdermal patch.
  • Methods for preparing transdermal patches are disclosed, e.g., in Brown, et al. (1988) Ann. Rev. Med. 39:221-229 which is incorporated herein by reference.
  • Subcutaneous implantation for sustained release of the active compounds may also be a suitable route of administration. This entails surgical procedures for implanting an active compound in any suitable formulation into a subcutaneous space, e.g., beneath the anterior abdominal wall. See, e.g., Wilson et al. (1984) J. Clin. Psych. 45:242-247.
  • Hydrogels can be used as a carrier for the sustained release of the active compounds. Hydrogels are generally known in the art.
  • hydrogels are typically made by crosslinking high molecular weight biocompatible polymers into a network, which swells in water to form a gel like material.
  • hydrogels are biodegradable or biosorbable.
  • hydrogels made of polyethylene glycols, collagen, or poly(glycolic-co-L- lactic acid) may be useful. See, e.g., Phillips et al. (1984) J. Pharmaceut. Sci., 73: 1718- 1720.
  • This disclosure further provides a compound as defined in any one of the embodiments herein above for use in therapy.
  • This disclosure further provides a compound as defined in any one of the embodiments herein above for use as a medicament.
  • the pharmaceutical composition may be formulated for dissolution into a solution for administration by such techniques as, for example, intravenous administration.
  • the SMDCs and pharmaceutical compositions thereof are particularly useful for parenteral administration, i.e., subcutaneously (s.c.), intrathecally, intraperitoneally, intramuscularly (i.m.) or intravenously (i.v.).
  • the SMDCs and pharmaceutical compositions thereof are administered intravenously or subcutaneously.
  • Actual methods for preparing parenterally administrable compositions are well known or will be apparent to those skilled in the art and are described in more detail in, for example, Remington’s Pharmaceutical Science, 15 th ed., Mack Publishing Company, Easton, Pa.
  • compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, etc.
  • concentration of the antigen binding protein of the disclosure in such pharmaceutical formulation can vary widely, i.e., from less than about 0.5%, usually at or at least about 1% to as much as about 15 or 20% by weight and will be selected primarily based on fluid volumes, viscosities, etc., according to the particular mode of administration selected.
  • This disclosure further provides a compound as defined in any one of the embodiments herein above for use in therapy.
  • This disclosure further provides a compound as defined in any one of the embodiments herein above for use as a medicament.
  • a method of treating a disease in a subject in need thereof including administering an effective amount of a small molecule drug conjugate (SMDC) comprising folic acid, a conjugation linker moiety (L) that binds to the carbonyl of the folic acid, and to a drug moiety covalently bound to linker L, and where L is optionally covalently bound to a half-life extender (HL).
  • SMDC small molecule drug conjugate
  • L conjugation linker moiety
  • an SMDC provided herein is used in a method of inhibiting proliferation of a FR-expressing cell, such as an FR-expressing cell, the method comprising contacting the cell with the SMDC, e.g., exposing the cell to the SMDC under conditions permissive for binding of the folic acid of the SMDC on the surface of the cell, thereby inhibiting the proliferation of the cell.
  • the method is an in vitro or an in vivo method.
  • the cell is a cancer cell.
  • the cell is a non-small cell lung carcinoma (NSCLC).
  • the cell is an ovarian cancer cell.
  • the cell may be a mammalian cell, such as a human cell.
  • Inhibition of cell proliferation in vitro may be assayed using the CellTiter-Glo TM Luminescent Cell Viability Assay, which is commercially available from Promega (Madison, WI). That assay determines the number of viable cells in culture based on quantitation of ATP present, which is an indication of metabolically active cells. See Crouch et al. (1993) J. Immunol. Meth. 160:81-88, US Pat. No. 6602677.
  • the assay may be conducted in 96- or 384- well format, making it amenable to automated high-throughput screening (HTS). See Cree et al. (1995) AntiCancer Drugs 6:398-404.
  • the assay procedure involves adding a single reagent (CellTiter-Glo ® Reagent) directly to cultured cells. This results in cell lysis and generation of a luminescent signal produced by a luciferase reaction.
  • the luminescent signal is proportional to the amount of ATP present, which is directly proportional to the number of viable cells present in culture. Data can be recorded by luminometer or CCD camera imaging device.
  • the luminescence output is expressed as relative light units (RLU).
  • a SMDC for use as a medicament is provided.
  • a SMDC for use in a method of treatment is provided.
  • a method of treating a disease in a subject in need thereof said method including administering an effective amount of a pharmaceutical composition of the SMDC as described herein.
  • the disease is cancer.
  • the cancer is associated with overexpression of FR, such as FR ⁇ .
  • the FR-expressing cancer is an epithelial-derived tumor.
  • the FR-expressing cancer is non-small cell lung carcinoma (NSCLC), lung cancer, mesothelioma, or ovarian cancer.
  • the FR-expressing cancer is non- small cell lung carcinoma (NSCLC) or ovarian cancer.
  • the FR-expressing cancer is non-small cell lung carcinoma (NSCLC).
  • the FR-expressing cancer is ovarian cancer. Any of the foregoing FR-expressing cancer types may be FR ⁇ - expressing cancers.
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent.
  • the FR-expressing cancer such as the FR ⁇ -expressing cancer
  • the mammal is human.
  • the present disclosure provides for the use of a SMDC in the manufacture or preparation of a medicament.
  • the medicament is for treatment of an FR-expressing cancer, such as an FR ⁇ -expressing cancer.
  • the medicament is for use in a method of treating an FR-expressing cancer, such as an FR ⁇ -expressing cancer, the method comprising administering to an individual having an FR-expressing cancer, such as an FR ⁇ -expressing cancer, an effective amount of the medicament.
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent.
  • the methods provided herein are for treating cancer in a mammal. In embodiments, the methods provided herein are for treating cancer in a human.
  • the cancers that may be treated with an immunoconjugate or method provided herein include epithelial-derived tumors including ovarian, uterine, breast, endometrial, pancreatic, renal, lung, colorectal, and brain tumors.
  • the cancers that may be treated with an immunoconjugate or a method provided herein include serous and endometrioid epithelial ovarian cancer, endometrial adenocarcinoma, non-small cell lung carcinoma (NSCLC) of the adenocarcinoma subtype, squamous lung cancer, mesotheliomas, and triple-negative breast cancer (TNBC).
  • the cancer is ovarian cancer.
  • the cancer is breast cancer.
  • the cancer is lung cancer.
  • the cancer is triple-negative breast cancer.
  • the cancer is non-small cell lung carcinoma (NSCLC).
  • the cancer is mesothelioma.
  • an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above is provided.
  • the article of manufacture (a kit) comprises a container and a label or package insert on or associated with the container.
  • Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the disorder and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is an SMDC as described herein.
  • the label or package insert indicates that the composition is used for treating the condition of choice.
  • the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises an SMDC as described herein; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent.
  • the article of manufacture in this embodiment of the disclosure may further comprise a package insert indicating that the compositions can be used to treat a particular condition.
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution or dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution or dextrose solution.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution or dextrose solution.
  • BWFI bacteriostatic water for injection
  • Ringer's solution such as bacterio
  • reaction mixture was then filtered and washed with DMF (3 times 10 mL each), and isopropyl alcohol (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 3).
  • the resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), and isopropyl alcohol (3 times 10 mL each).
  • Resin deprotection The resin was then treated with 2% hydrazine in DMF (10 mL). The mixture was allowed to react for 15 minutes. The resin was then washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each) and dichloromethane (3 times 10 mL each).
  • Resin deprotection The resin was treated with peptide deprotection mixture (TFA, water, triisopropyl silane (TIPS), 2,2'-(ethane-1,2-diylbis(oxy))bis(ethane-1-thiol) (SDBS), 94/2/2/2) at 45 o C for 30 minutes before it was filtered.
  • TIPS triisopropyl silane
  • SDBS 2,2'-(ethane-1,2-diylbis(oxy))bis(ethane-1-thiol)
  • reaction mixture was then filtered and washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 15).
  • the resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each).
  • reaction mixture was then filtered and washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 17).
  • the resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each).
  • reaction mixture was then filtered and washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 31).
  • the resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each).
  • reaction mixture was then filtered and washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 33).
  • the resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each).
  • Resin deprotection The resin was treated with peptide deprotection mixture (TFA, water, triisopropyl silane (TIPS), 2,2'-(ethane-1,2-diylbis(oxy))bis(ethane-1-thiol) (SDBS), 94/2/2/2) at 45 o C for 30 minutes before it was filtered. The resulting TFA mixture was diluted with cold ether and the suspension was centrifuged with 5 minutes.
  • TFA peptide deprotection mixture
  • TIPS triisopropyl silane
  • SDBS 2,2'-(ethane-1,2-diylbis(oxy))bis(ethane-1-thiol)
  • reaction mixture was then filtered and washed with DMF (3 times 10 mL each), and isopropyl alcohol (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 38).
  • the resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), and isopropyl alcohol (3 times 10 mL each).
  • reaction mixture was then filtered and washed with DMF (3 times 10 mL each), and isopropyl alcohol (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 42).
  • the resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), and isopropyl alcohol (3 times 10 mL each).
  • Resin deprotection The resin was treated with peptide deprotection mixture (TFA, water, triisopropyl silane (TIPS), 2,2'-(ethane-1,2-diylbis(oxy))bis(ethane-1-thiol) (SDBS), 94/2/2/2) at 45 o C for 30 minutes before it was filtered. The resulting TFA mixture was diluted with cold ether and the suspension was centrifuged with 5 minutes.
  • TFA peptide deprotection mixture
  • TIPS triisopropyl silane
  • SDBS 2,2'-(ethane-1,2-diylbis(oxy))bis(ethane-1-thiol)
  • FR-VC-PAB-MMAE was isolated as a fluffy yellowish powder. MS m/z 2670 (M+H).
  • Example B1 In vitro Efficacy of Duo5 and MMAE.
  • the in vitro efficacies of Duo5 (the synthesis of which has been previously described in US Patent 10,590,165, which is incorporated herein in its entirety and MMAE were evaluated using the following human cancer cell lines: KB (FR+) and A549 (FR-), purchased from the American Type Culture Collection (ATCC; Manassas, VA) and routinely cultured in folic acid free RPMI 1640 medium (Catalog #27016021; Thermo Fisher Scientific; Waltham, MA) and RPMI 1640 medium (ATCC modification) (Catalog #A1049101; Thermo Fisher Scientific; Waltham, MA) supplemented with 10% fetal bovine serum (FBS; Catalog #F4135; Sigma-Aldrich; St.
  • FBS fetal bovine
  • Tumor cells were washed with Dulbecco’s Phosphate Buffered Saline (DPBS; Catalog #PBL01; Caisson Labs; Smithfield, UT) and harvested by detachment with TrypLE Express (Catalog #1204013; Thermo Fisher Scientific; Waltham, MA). Viable cell counts were made by Trypan blue exclusion using a Countess II automated cell counter.
  • DPBS Dulbecco’s Phosphate Buffered Saline
  • TrypLE Express Catalog #1204013; Thermo Fisher Scientific; Waltham, MA.
  • CellTiter-Glo 2.0 assay (Catalog #G9242; Promega; Madison, WI, USA) based on the manufacturer’s instructions.
  • CellTiter Glo reagent reacts with ATP in metabolically active cells to give a luminescent readout that is directly proportional to the number of viable cells. Briefly, plates were removed from the incubator and equilibrated to room temperature before addition of CellTiter Glo reagent. Luminescence was measured using a SpectraMax iD3 microplate reader (Molecular Devices; San Jose, CA).
  • FIGS.1A and 1B Duo5 and MMAE dose-dependently reduced KB and A549 cell viability in 3-day assays.
  • the potencies of Duo5 and MMAE in KB as determined by IC50 were similar with 0.8982 nM and 0.3707 nM, respectively.
  • IC 50 values of Duo5 and MMAE were higher in A549 than in KB, they inhibited cell proliferation across both cell lines in a dose-dependent manner regardless of FR expression level.
  • FIGS. 1A and 1B Cell viability for Duo5 and MMAE are shown in FIGS. 1A and 1B and IC50 values are shown in Table 1.
  • FIGS. 1A and 1B Cell viability for Duo5 and MMAE are shown in FIGS. 1A and 1B and IC50 values are shown in Table 1.
  • FIGS. 1A and 1B Cell viability for Duo5 and MMAE are shown in FIGS. 1A and 1B and IC50 values are shown in Table 1.
  • 1A-B show results of an in vitro efficacy study of A) Duo5 and B) MMAE using: KB (FR+) cells and A549 (FR-) cells.
  • Table 1 IC50 Values (nM) of Duo5 and MMAE in Human Tumor Cells
  • Example B2 In vitro Efficacy, Specificity, and Stability of Small Molecule-Drug Conjugates (SMDCs).
  • SMDCs Small Molecule-Drug Conjugates
  • SMDCs were evaluated using the following human cancer cell lines: FR-positive KB and FR-negative A549, purchased from the American Type Culture Collection (ATCC; Manassas, VA) and routinely cultured in folic acid free RPMI 1640 medium (Catalog #27016021; Thermo Fisher Scientific; Waltham, MA) and RPMI 1640 medium (ATCC modification) (Catalog #A1049101; Thermo Fisher Scientific; Waltham, MA) supplemented with 10% fetal bovine serum (FBS; Catalog #MT35011CV; Corning), respectively, and maintained at 37°C with 5% CO 2 in a humidified environment.
  • ATCC American Type Culture Collection
  • FBS fetal bovine serum
  • Tumor cells were washed with Dulbecco’s Phosphate Buffered Saline (DPBS; Catalog #PBL01; Caisson Labs; Smithfield, UT) and harvested by detachment with TrypLE Express (Catalog #1204013; Thermo Fisher Scientific; Waltham, MA). Viable cell counts were made by Trypan blue exclusion using a Countess or Countess II automated cell counter.
  • DPBS Phosphate Buffered Saline
  • TrypLE Express Catalog #1204013; Thermo Fisher Scientific; Waltham, MA.
  • Cell Viability Assay All cells were harvested and seeded into 384-well white wall flat bottom plates (Catalog #3570; Corning) at a density of 1,000 cells/well (for 120-hour assay) or 3,000 cells/well in folic acid free RPMI 1640 medium or RPMI 1640 medium (ATCC modification) supplemented with 10% fetal bovine (complete growth media). Plates were maintained at 37°C overnight to allow cells to adhere to the plate. The outer wells of plates contained complete growth media only. Thirty minutes prior to the addition of SMDCs, the media in designated wells of KB cells were replaced with complete growth media containing 100 ⁇ M folic acid (a binding site competitor).
  • KB cells in those designated wells were used to determine the targeting specificity of SMDCs for FR.
  • Working solutions of test articles were prepared at 100X final concentrations with 5-fold serial dilutions in DMSO and subsequently diluted at 5X final concentrations with 5-fold serial dilutions in complete growth media (in the presence or absence of 100 ⁇ M folic acid for KB).
  • Cell treatment was performed in triplicates and maintained at 37°C for (1) 2 hours followed by washing 3 times with complete growth media and incubating with complete growth media for another 70 hours (2-hour pulse assay); or (2) 72 hours or 120 hours (for FR-IODO-Examorpholine treatment) (72-hour assay or 120-hour assay).
  • Cells treated with SMDCs using 72-hour assay or 120-hour assay were used to determine the stability of SMDCs in complete growth media. After treatment, cell viability was determined by CellTiter-Glo 2.0 assay (Catalog #G9242; Promega; Madison, WI, USA) based on the manufacturer’s instructions.
  • CellTiter Glo reagent reacts with ATP in metabolically active cells to give a luminescent readout that is directly proportional to the number of viable cells. Briefly, plates were removed from the incubator and equilibrated to room temperature before addition of CellTiter Glo reagent. Luminescence was measured using a SpectraMax iD3 microplate reader (Molecular Devices; San Jose, CA).
  • IC50 Values (nM) of FA-SMDCs in Human Tumor Cells with 2-hour Pulse Assay are presented in Table 2.
  • Table 2 IC50 Values (nM) of FA-SMDCs in Human Tumor Cells with 2-hour Pulse Assay
  • FIGS. 3A-B show results of an in vitro efficacy, specificity, and stability study of FR-PEG-Duo5 using KB (FR+) cells with: A) 2-hour pulse assay and B) 72-hour assay.
  • FIGS. 3A-B show results of an in vitro efficacy, specificity, and stability study of FR-PEG-Duo5 using KB (FR+) cells with: A) 2-hour pulse assay and B) 72-hour assay.
  • FIGS. 4A-B show results of an in vitro efficacy, specificity, and stability study of FR-VC-Duo5 using KB (FR+) cells with: A) 2-hour pulse assay and B) 72-hour assay.
  • FIGS. 5A-B show results of an in vitro efficacy, specificity, and stability study of FR-IODO-Duo5 using KB (FR+) cells with: A) 2-hour pulse assay and B) 72-hour assay.
  • FIGS. 6A-B show results of an in vitro efficacy, specificity, and stability study of FR-VC-PAB-MMAE using KB (FR+) cells with: A) 2-hour pulse assay and B) 72-hour assay.
  • FIGS. 7A-B show results of an in vitro efficacy, specificity, and stability study of FR-PL-MMAE using KB (FR+) cells with: A) 2-hour pulse assay and B) 72-hour assay.
  • FIG. 8 shows results of an in vitro efficacy, specificity, and stability study of FR- IODO-Examorpholine using KB (FR+) cells with 120-hour assay.
  • In vitro stability of the SMDCs described herein was evaluated with 72-hour assay or 120-hour assay against FR-positive KB and FR-negative A549 cancer cell lines using standard cell viability assays. As shown in FIGS.
  • Example B3 Mouse Pharmacokinetics (PK) for FR-VC-IODO-Duo5.
  • FR-VC-IODO-Duo5 and Duo5 are both dissolved as standard solution in DMSO (10 mM).
  • the dosing formulation consists of 1% DMSO and 99% PBS. The dosing level was 1 mg/Kg.
  • 30 male ICR mice was used in the study and the FR-VC-IODO-Duo5 was injected via IV route.
  • the blood was sampled at the following time points: 0.5, 2, 4, 6, 8, 24, 48, 72 and 120 hours post dose.
  • the sampled blood was treated with EDTA-K2 and 1 molar citric acid solution to denature proteases in serum.
  • LCMS analysis was performed to monitor the serum level of FR-VC-IODO-Duo5 and Duo5. The result of the PK study is presented in FIG. 9, Table 4, and Table 5.
  • BLQ Below the lower limit of quantification (LLOQ) If the adjusted rsq (linear regression coefficient of the concentration value on the terminal phase) is less than 0.9, T 1/2 might not be accurately estimated. No abnormal clinical sign was observed during the entire in-life study.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Botany (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Provided, inter alia, are small molecule drug conjugates (SMDCs) which specifically bind Folate Receptor (FR). Further disclosed are pharmaceutical compositions, and methods for treating cancer.

Description

FOLATE-CONJUGATED DRUGS AND USES THEREOF CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims priority to United States Provisional Application No. 63/392,066, filed July 25, 2022, the disclosure of which is hereby incorporated by reference in its entirety. [0002] Throughout this application various publications, patents, and/or patent applications are referenced. The disclosures of the publications, patents and/or patent applications are hereby incorporated by reference in their entireties into this application in order to more fully describe the state of the art to which this disclosure pertains. TECHNICAL FIELD [0003] The present disclosure relates to small molecule-drug conjugates (SMDCs) comprising folic acid and methods of making and using the same. INTRODUCTION AND SUMMARY [0004] Antibody-drug conjugates (ADCs) allow for the targeted delivery of a drug moiety to a tumor, and, in some embodiments intracellular accumulation therein, where systemic administration of unconjugated drugs may result in unacceptable levels of toxicity to normal cells (Polakis P. (2005) Current Opinion in Pharmacology 5:382-387). Small molecule-drug conjugates (SMDCs) are designed along similar principles as ADCs for drug delivery and tumor targeting applications, with the difference being that the antibody component is replaced by a targeting ligand that can be a peptide or a small molecule (Casi, G. and Neri, D. (2015) J. Med. Chem. 58: 8751–8761; Srinivasarao, M. et al. (2015) Nat. Rev. Drug Discov. 14: 203–219) [0005] SMDCs have several strengths compared to ADCs. SMDCs are frequently easier to synthesize than biological agents. Most are nonimmunogenic, making them unlikely to provoke an autoimmune response (Min, H.K. et al. (2016) Korean J. Intern. Med. 31:608- 611; Alkhayat, A.L.I. et al. (2018) CHEST 154:439A-440A). Transportation, storage, and administration are easier than with ADCs (Kurzrock, R. et al. (2012) Mol. Cancer Ther. 11:308-316). Molecular weights of SMDCs are much lower than those of ADCs, resulting in better cell permeability (particularly in solid tumors that may be poorly vascularized) (Manzoor, A.A. et al. (2012) Cancer Res. 72:5566-5575). The low molecular weight and other chemical features are also associated with better in vitro and in vivo stability than biological agents including monoclonal antibodies (Jain, R.K. and Stylianopoulos, T. (2010) Nat. Rev. Clin. Oncol. 7:653). Notably, SMDCs are more rapidly removed from the blood through glomerular filtration in the kidneys than are ADCs. This results in a better toxicity profile; however, it also has the potential to reduce the effective time on the tumor target (Vlashi, E. et al. (2013) ACS Nano 7:8573-8582). [0006] The present disclosure provides SMDCs comprising a folic acid conjugated to the drug moiety (payload) through linker moieties. In embodiments, the folic acid binds to folate receptor-expressing cancer cells and allows for selective uptake of the SMDC into the cancer cells. In embodiments, the SMDCs provided herein selectively deliver an effective amount of drug moiety to tumor tissue and reduce the non-specific toxicity associated with related SMDCs. The SMDC compounds described herein include those with anticancer activity. [0007] The folate receptor (FR) is a high-affinity membrane-associated protein, which exhibits limited expression on healthy cells, but is frequently overexpressed on a wide variety of specific cell types, such as epithelial tumor cells (e.g. ovarian, endometrial, breast, colorectal, kidney, lung, nasopharyngeal) and activated (but not resting) macrophages, which are involved in inflammation and autoimmune diseases. This membrane protein binds extracellular folates with very high affinity and through an endocytic process, physically delivers them inside the cell for biological consumption. [0008] In humans, there are three functional isoforms of FR, namely hFRα, hFRβ, and hFRγ (Elnakat, H. and Ratnam, M. (2004) Adv. Drug Deliv. Rev. 56:1067-1084). hFRα is overexpressed in a broad variety of cancers, among them adenocarcinomas of uterus, ovary, breast, cervix, kidney and colon and testicular choriocarcinoma, ependymal brain tumors, malignant pleural mesothelioma, and nonfunctioning pituitary adenocarcinoma, while hFRβ in leukemias and activated macrophages (Wibowo, A. et al. (2013) Proc. Natl. Acad. Sci. U.S.A. 110:15180-15188; Low, P. et al. (2007) Acc. Chem. Res. 41:120-129). [0009] Folate Receptor-alpha (FRα), also known as Folate Receptor 1 (FOLR1), or Folate Binding Protein, is a glycosylphosphatidylinositol (GPI)-anchored glycoprotein with a strong binding affinity for folic acid and reduced folic acid derivatives (Leung et al. (2013) Clin. Biochem. 46:1462- 1468). FRα has important functions relating to cell proliferation and survival (Kelemen L.E. (2006) Int. J. Cancer 119(2):2430250), and it mediates delivery of the physiological folate, 5-methyltetrahydrofolate, to the interior of cells. Expression of FRα on normal tissues is restricted to the apical membrane of epithelial cells in the kidney proximal tubules, alveolar pneumocytes of the lung, bladder, testes, choroid plexus, and thyroid (Weitman S.D. et al. (1992) Cancer Res. 52:3396-3401; Antony A.C. (1996) Ann. Rev. Nutr. 16:501 -521; Kalli K.R. et al. (2008) Gynecol. Oncol. 108:619-626). FRα is overexpressed in epithelial-derived tumors including ovarian, uterine, breast, endometrial, pancreatic, renal, lung, colorectal, and brain tumors. This expression pattern of FRα makes it a desirable target for FRα-directed cancer therapy. [0010] Folate plays important roles in nucleotide biosynthesis and cell division, intracellular activities which occur in both malignant and certain normal cells. Upon binding the folate receptor, the folate impacts the cell cycle in dividing cells. This led to the use of folic acid and its analogues and derivatives as a targeting agent for the delivery of therapeutic and/or diagnostic agents to these specific cell populations to achieve a selective concentration of pharmaceutical and/or diagnostic agents in these specific cells relative to normal cells (Leamon and Low (2001) Drug Discov. Today 6:44-51; Leamon and Reddy (2004) Adv. Drug Deliv. Rev. 56:1127-41; Leamon et al, (2005) Bioconjugate Chem. 16:803-811) [0011] The linker in SMDCs usually consists of a spacer and a cleavable bridge. Linkers are designed to preserve the activity of post-cleavage species and to optimize the drug release, pharmacokinetics, and pharmacodynamics of the targeting ligand and payload (Srinivasarao, M. et al. (2015) Nat. Rev. Drug Discov. 14: 203–219; Vlahov, I.R. and Leamon, C.P (2012) Bioconj. Chem. 23:1357-1369). Another function of the spacer is to improve the hydrophilicity of SMDC. The cleavable bridge retains stability during the SMDC transportation from the vasculature to the tumor, and is typically cleaved by one of two triggering methods. The first mechanism is cleavage in the endosomes of the target cells due to low pH. Such a cleavage bridge comprises acetals and hydrazones (Yang, J. et al. (2007) . J. Pharm. Exp. 321:462-468). The second mechanism is through use of a disulfide-based linker, which undergoes cleavage due to an intracellular excess of glutathione (GSH), thioredoxin, peroxiredoxins, and nicotinamide adenine dinucleotides (NADH and NADPH) (Srinivasarao, M. et al. (2015) Nat. Rev. Drug Discov. 14: 203–219). [0012] As with ADCs, optimal conjugates have high binding affinity for their targets (Srinivasarao, M. et al. (2015) Nat. Rev. Drug Discov. 14: 203–219) and have highly cytotoxic payloads, similar to those used in ADCs. In some cases, to increase the cytotoxic activity of the conjugate, multivalent ligands, comprising several payloads linked to the targeting compound, are employed. Payloads that target mitosis, DNA replication, and protein translation are currently investigated. [0013] There is a need for improved methods of modulating the immune regulation of folate receptors such as folate receptor alpha (FRα) and the downstream signaling processes activated by folate receptors such as folate receptor alpha (FRα). Folate receptor binds folic acid and analogues and derivatives thereof with very high affinity. Once the folate is bound to the folate receptor it impacts the cell cycle in dividing cells. The folate receptor is frequently overexpressed on epithelial tumor cells, in contrast, folate receptor expression in normal tissues is limited, making the folate receptor a good target for SMDCs. Thus, small molecule-drug conjugates (SMDCs) where the drug is conjugated to folic acid, can provide a very targeted and potent anti-tumor activity. [0014] In one aspect, provided herein is a compound of Formula (I) or (II):
Figure imgf000006_0001
or a pharmaceutically acceptable salt thereof, wherein: L is a multivalent linker bound at least to a folic acid and a drug moiety; HL is a half-life extender; and D is a drug moiety. [0015] In an aspect, provided herein is a method of treating a FR-expressing cancer, such as an FRα-expressing cancer in a subject in need thereof, said method including administering the compound or pharmaceutically acceptable salt thereof as described herein (including in an aspect embodiment table example or claim) to the subject [0016] In an aspect, provided herein is a pharmaceutical composition comprising the compound or pharmaceutically acceptable salt thereof as described herein, and a pharmaceutically acceptable excipient. [0017] The embodiments disclosed herein include but are not limited to the following. Embodiment 1 is a compound of the Formula (I) or (II):
Figure imgf000007_0001
or a pharmaceutically acceptable salt thereof, wherein: L is a multivalent linker bound at least to a folic acid and a drug moiety; HL is a half-life extender; and D is a drug moiety. [0018] Embodiment 2 is the compound or a pharmaceutically acceptable salt thereof of embodiment 1, wherein L is a bond, -C(O)-, -NH-, Amino Acid Unit, Peptoid, – (CH2CH2O)n–, –(CH2)n–, –(4-aminobenzyloxycarbonyl)–, –(C(O)CH2CH2C(O))–, –(C(O)CH2CH2NH)–,
Figure imgf000008_0001
integer from 1 to 24; and each R2 and R3 is independently H or substituted or unsubstituted alkyl. [0019] Embodiment 3 is the compound or a pharmaceutically acceptable salt thereof of embodiment 1 or 2, wherein L is -C(O)-, -NH-,–(CH2CH2O)n–, –(CH2)n–, –(4- aminobenzyloxycarbonyl)–, -Cys-, -Asp-, -Arg-, -Val-, -Glu-, -citrulline- (-Cit-), -Lys-,
Figure imgf000008_0002
Figure imgf000009_0001
combinations thereof. [0020] Embodiment 4 is the compound or a pharmaceutically acceptable salt thereof of embodiment 3, wherein L is -C(O)-, -NH-, –(CH2CH2O)n–, –(CH2)n–, –SCH2CH2O–, – (C(O)CH2CH2C(O))–, -Val-, -Cit-, –(4-aminobenzyloxycarbonyl)–, -Arg-, -Asp-, -Lys-, -
Figure imgf000009_0003
combinations thereof. [0021] Embodiment 5 is the compound or a pharmaceutically acceptable salt thereof of embodiment 4, wherein L is
Figure imgf000009_0002
,
Figure imgf000010_0001
,
Figure imgf000011_0001
, wherein the carbonyl is linked to the drug moiety (D), the amine is linked to the folic acid, and the optional third linkage is to the half-life extender (HL). [0022] Embodiment 6 is the compound or a pharmaceutically acceptable salt thereof of any one of embodiments 1-5, wherein HL is a cholesterol-like half-life extender or albumin binder half-life extender. Embodiment 7 is the compound or a pharmaceutically acceptable salt thereof of embodiment 6, wherein HL is
Figure imgf000011_0002
, ,
Figure imgf000012_0001
[0023] Embodiment 8 is the compound or a pharmaceutically acceptable salt thereof of embodiment 7, wherein
Figure imgf000012_0002
. [0024] Embodiment 9 is the compound or a pharmaceutically acceptable salt thereof of any one of embodiments 1-8, wherein D is a tubulin inhibitor or disruptor, kinase inhibitor, DNA damaging agent, transcription inhibitors, or proteolysis-targeting chimera (PROTAC). [0025] Embodiment 10 is the compound or a pharmaceutically acceptable salt thereof of embodiment 9, wherein D is a tubulin inhibitor. [0026] Embodiment 11 is the compound or a pharmaceutically acceptable salt thereof of any one of embodiments 1-9, wherein D is a pyrrolobenzodiazepine, duocarmycin, anthracycline, maytansinoid, auristatin, calicheamicin, camptothecin, RNA polymerase II inhibitor, topoisomerase I inhibitor, tyrosine kinase inhibitor, EG5 inhibitor, or MEK inhibitor. [0027] Embodiment 12 is the compound or a pharmaceutically acceptable salt thereof of embodiment 11, wherein D is an auristatin. [0028] Embodiment 13 is the compound or a pharmaceutically acceptable salt thereof of embodiment 11, wherein D is MMAE, MMAF, Duo5, PNU, SN-38, irinotecan, amatoxin, maytansine, exatecan, trametinib, abemaciclib, palbociclib, or examorpholine. Embodiment 14 is the compound or a pharmaceutically acceptable salt thereof of embodiment 13, wherein D is Duo5. Embodiment 15 is the compound or a pharmaceutically acceptable salt thereof of embodiment 13, wherein D is MMAE. [0029] Embodiment 16 is the compound or a pharmaceutically acceptable salt thereof of embodiment 13, wherein D is examorpholine. [0030] Embodiment 17 is the compound or pharmaceutically acceptable salt thereof of any one of embodiments 1-16, wherein the compound is:
Figure imgf000013_0001
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0001
or a pharmaceutically acceptable salt thereof. [0031] Embodiment 18 is the compound or pharmaceutically acceptable salt thereof of any one of embodiments 1-17, for use in therapy. [0032] Embodiment 19 is the compound or pharmaceutically acceptable salt thereof of embodiment 18, for use in treating a FR-expressing cancer, optionally wherein the FR- expressing cancer is non-small cell lung carcinoma (NSCLC), lung cancer, mesothelioma, or ovarian cancer. [0033] Embodiment 20 is a method of treating a FR-expressing cancer in a subject, comprising administering the compound or pharmaceutically acceptable salt thereof of any one of embodiments 1-17 to a subject in need thereof. [0034] Embodiment 21 is use of the compound or pharmaceutically acceptable salt thereof of any one of embodiments 1-17 for the manufacture of a medicament. [0035] Embodiment 22 is use of the compound or pharmaceutically acceptable salt thereof of any one of embodiments 1-17 for the manufacture of a medicament for treating a FR- expressing cancer, optionally wherein the FR-expressing cancer is non-small cell lung carcinoma (NSCLC), lung cancer, mesothelioma, or ovarian cancer. [0036] Embodiment 23 is the compound or pharmaceutically acceptable salt thereof for use, use, or method of any one of embodiments 19, 20, or 22, wherein the FR-expressing cancer is an epithelial-derived tumor. [0037] Embodiment 24 is the compound or pharmaceutically acceptable salt thereof for use, use, or method of embodiment 23, wherein the epithelial-derived tumors are ovarian, uterine, breast, endometrial, pancreatic, renal, lung, colorectal, or brain tumors. [0038] Embodiment 25 is the compound or pharmaceutically acceptable salt thereof for use, use, or method of any one of embodiments 19, 20, or 22, wherein the FR-expressing cancer is non-small cell lung carcinoma (NSCLC) or ovarian cancer. [0039] Embodiment 26 is the compound or pharmaceutically acceptable salt thereof for use, use, or method of any one of embodiments 19, 20, or 22-25, wherein the FR-expressing cancer is in a mammal, optionally wherein the mammal is a human. [0040] Embodiment 27 is a method of inhibiting proliferation of a FR-expressing cell, comprising contacting the FR-expressing cell with the compound or pharmaceutically acceptable salt thereof of any one of embodiments 1-17. [0041] Embodiment 28 is the use of embodiment 21, wherein the medicament is for inhibiting proliferation of a FR-expressing cell. [0042] Embodiment 29 is the compound or pharmaceutically acceptable salt thereof of any one of embodiments 1-17, for use in inhibiting proliferation of a FR-expressing cell. [0043] Embodiment 30 is the method, use, or compound or pharmaceutically acceptable salt thereof for use of any one of embodiments 27-29, wherein the FR-expressing cell is a FR-expressing cancer cell, optionally wherein the FR-expressing cancer is non-small cell lung carcinoma (NSCLC), lung cancer, mesothelioma, or ovarian cancer. [0044] Embodiment 31 is the method, use, or compound or pharmaceutically acceptable salt thereof for use of any one of embodiments 27-29, wherein the FR-expressing cell is a FR-expressing non-small cell lung carcinoma (NSCLC) cell or FR-expressing ovarian cell. [0045] Embodiment 32 is a pharmaceutical composition comprising the compound or pharmaceutically acceptable salt thereof of any one of embodiments 1-17, and a pharmaceutically acceptable excipient. BRIEF DESCRIPTION OF THE DRAWINGS [0046] FIGS. 1A-B show results of an in vitro efficacy study of Duo5 and MMAE using: KB (FR+) cells and A549 (FR-) cells. FIG. 1A shows results of an in vitro efficacy study of Duo5. FIG. 1B shows results of an in vitro efficacy study of MMAE. Here and in subsequent figures, the log-molar concentration of the indicated compound is on the horizontal axis. [0047] FIGS. 2A-B show results of an in vitro efficacy and stability study of SMDCs (FR- PEG-Duo5, FR-VC- Duo5, FR-IODO- Duo5, FR-VC-PAB-MMAE, and FR-PL-MMAE) using: A549 (FR-) cells. FIG. 2A shows results of a 2-hour pulse assay. FIG. 2B shows results of a 72-hour assay or 120-hour assay (for FR-IODO-Examorpholine Treatment). [0048] FIGS. 3A-B show results of an in vitro efficacy, specificity, and stability study of SMDC FR-PEG- Duo5 (with and without pretreatment with folic acid) using: KB (FR+) cells. FIG. 3A shows results of a 2-hour pulse assay. FIG. 3B shows results of a 72-hour assay. [0049] FIGS. 4A-B show results of an in vitro efficacy, specificity, and stability study of SMDC FR-VC- Duo5 (with and without pretreatment with folic acid) using: KB (FR+) cells. FIG. 4A shows results of a 2-hour pulse assay. FIG. 4B shows results of a 72-hour assay. [0050] FIGS. 5A-B show results of an in vitro efficacy, specificity, and stability study of SMDC FR-IODO- Duo5 (with and without pretreatment with folic acid) using: KB (FR+) cells. FIG. 5A shows results of a 2-hour pulse assay. FIG. 5B shows results of a 72-hour assay. [0051] FIGS. 6A-B show results of an in vitro efficacy, specificity, and stability study of SMDC FR-VC-PAB-MMAE (with and without pretreatment with folic acid) using: KB (FR+) cells. FIG. 6A shows results of a 2-hour pulse assay. FIG. 6B shows results of a 72- hour assay. [0052] FIGS. 7A-B show results of an in vitro efficacy, specificity, and stability study of SMDC FR-PL-MMAE (with and without pretreatment with folic acid) using: KB (FR+) cells. FIG. 7A shows results of a 2-hour pulse assay. FIG. 7B shows results of a 72-hour assay. [0053] FIG. 8 shows results of an in vitro efficacy, specificity, and stability study of FR- IODO-Examorpholine using KB (FR+) cells with 120-hour assay. [0054] FIG. 9 shows the results of the pharmacokinetic studies of FR-VC-IODO-Duo5 and Duo5 in mice. DETAILED DESCRIPTION OF THE DISCLOSURE Definitions: [0055] Unless defined otherwise, technical and scientific terms used herein have meanings that are commonly understood by those of ordinary skill in the art unless defined otherwise. Generally, terminologies pertaining to techniques of cell and tissue culture, molecular biology, immunology, microbiology, genetics, transgenic cell production, protein chemistry and nucleic acid chemistry and hybridization described herein are well known and commonly used in the art. The methods and techniques provided herein are generally performed according to conventional procedures well known in the art and as described in various general and more specific references that are cited and discussed herein unless otherwise indicated. See, e.g., Sambrook et al. Molecular Cloning: A Laboratory Manual, 2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989) and Ausubel et al., Current Protocols in Molecular Biology, Greene Publishing Associates (1992). All of the references cited herein are incorporated herein by reference in their entireties. Enzymatic reactions and enrichment/purification techniques are also well known and are performed according to manufacturer's specifications, as commonly accomplished in the art or as described herein. The terminology used in connection with, and the laboratory procedures and techniques of, analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are well known and commonly used in the art. Standard techniques can be used for chemical syntheses, chemical analyses, pharmaceutical preparation, formulation, and delivery, and treatment of patients. [0056] The headings provided herein are not limitations of the various aspects of the disclosure, which aspects can be understood by reference to the specification as a whole. [0057] Unless otherwise required by context herein, singular terms shall include pluralities and plural terms shall include the singular. Singular forms “a”, “an” and “the”, and singular use of any word, include plural referents unless expressly and unequivocally limited on one referent. [0058] It is understood the use of the alternative (e.g., “or”) herein is taken to mean either one or both or any combination thereof of the alternatives. [0059] The term “and/or” used herein is to be taken mean specific disclosure of each of the specified features or components with or without the other. For example, the term “and/or” as used in a phrase such as “A and/or B” herein is intended to include “A and B,” “A or B,” “A” (alone), and “B” (alone). Likewise, the term “and/or” as used in a phrase such as “A, B, and/or C” is intended to encompass each of the following aspects: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone). [0060] As used herein, the term “about” refers to a value or composition that is within an acceptable error range for the particular value or composition as determined by one of ordinary skill in the art, which will depend in part on how the value or composition is measured or determined, i.e., the limitations of the measurement system. For example, “about” or “approximately” can mean within one or more than one standard deviation per the practice in the art. Alternatively, “about” or “approximately” can mean a range of up to 10% (i.e., ±10%) or more depending on the limitations of the measurement system. For example, about 5 mg can include any number between 4.5 mg and 5.5 mg. Furthermore, particularly with respect to biological systems or processes, the terms can mean up to an order of magnitude or up to 5-fold of a value. When particular values or compositions are provided in the instant disclosure, unless otherwise stated, the meaning of “about” or “approximately” should be assumed to be within an acceptable error range for that particular value or composition. In embodiments, about includes the specified value. [0061] In this disclosure, “comprises,” “comprising,” “containing” and “having” and the like can have the meaning ascribed to them in U.S. Patent law and can mean “includes,” “including,” and the like. “Consisting essentially of or “consists essentially” likewise has the meaning ascribed in U.S. Patent law and the term is open-ended, allowing for the presence of more than that which is recited so long as basic or novel characteristics of that which is recited is not changed by the presence of more than that which is recited, but excludes prior art embodiments. [0062] The terms "polypeptide," "peptide" and "protein" and other related terms used herein are used interchangeably to refer to a polymer of amino acid residues, wherein the polymer may in embodiments be conjugated to a moiety that does not consist of amino acids. The terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymers. A "fusion protein" refers to a chimeric protein encoding two or more separate protein sequences that are recombinantly expressed as a single moiety. Polypeptides include mature molecules that have undergone cleavage. These terms encompass native and artificial proteins, protein fragments and polypeptide analogs (such as muteins, variants, chimeric proteins and fusion proteins) of a protein sequence as well as post-translationally, or otherwise covalently or non-covalently, modified proteins. Two or more polypeptides (e.g., 3 polypeptide chains) can associate with each other, via covalent and/or non-covalent association, to form a multimeric polypeptide complex (e.g., multi-specific antigen binding protein complex). Association of the polypeptide chains can also include peptide folding. Thus, a polypeptide complex can be dimeric, trimeric, tetrameric, or higher order complexes depending on the number of polypeptide chains that form the complex. [0063] As used herein, the terms “cancer,” “neoplasm,” and “tumor” are used interchangeably and, in either the singular or plural form, refer to cells that have undergone a malignant transformation that makes them pathological to the host organism. Primary cancer cells can be readily distinguished from non-cancerous cells by well-established techniques, particularly histological examination. The definition of a cancer cell, as used herein, includes not only a primary cancer cell, but any cell derived from a cancer cell ancestor. This includes metastasized cancer cells, and in vitro cultures and cell lines derived from cancer cells. When referring to a type of cancer that normally manifests as a solid tumor, a “clinically detectable” tumor is one that is detectable on the basis of tumor mass; e.g., by procedures such as computed tomography (CT) scan, magnetic resonance imaging (MRI), X-ray, ultrasound or palpation on physical examination, and/or which is detectable because of the expression of one or more cancer-specific antigens in a sample obtainable from a patient. Tumors may be a hematopoietic (or hematologic or hematological or blood-related) cancer, for example, cancers derived from blood cells or immune cells, which may be referred to as “liquid tumors.” Specific examples of clinical conditions based on hematologic tumors include leukemias such as chronic myelocytic leukemia, acute myelocytic leukemia, chronic lymphocytic leukemia and acute lymphocytic leukemia; plasma cell malignancies such as multiple myeloma, MGUS and Waldenstrom's macroglobulinemia; lymphomas such as non- Hodgkin's lymphoma, Hodgkin's lymphoma; and the like. [0064] The cancer may be any cancer in which an abnormal number of blast cells or unwanted cell proliferation is present or that is diagnosed as a hematological cancer, including both lymphoid and myeloid malignancies. Myeloid malignancies include, but are not limited to, acute myeloid (or myelocytic or myelogenous or myeloblastic) leukemia (undifferentiated or differentiated), acute promyeloid (or promyelocytic or promyelogenous or promyeloblastic) leukemia, acute myelomonocytic (or myelomonoblastic) leukemia, acute monocytic (or monoblastic) leukemia, erythroleukemia and megakaryocytic (or megakaryoblastic) leukemia. These leukemias may be referred together as acute myeloid (or myelocytic or myelogenous) leukemia (AML). Myeloid malignancies also include myeloproliferative disorders (MPD) which include, but are not limited to, chronic myelogenous (or myeloid) leukemia (CML), chronic myelomonocytic leukemia (CMML), essential thrombocythemia (or thrombocytosis), and polcythemia vera (PCV). Myeloid malignancies also include myelodysplasia (or myelodysplastic syndrome or MDS), which may be referred to as refractory anemia (RA), refractory anemia with excess blasts (RAEB), and refractory anemia with excess blasts in transformation (RAEBT); as well as myelofibrosis (MFS) with or without agnogenic myeloid metaplasia. [0065] Hematopoietic cancers also include lymphoid malignancies, which may affect the lymph nodes, spleens, bone marrow, peripheral blood, and/or extranodal sites. Lymphoid cancers include B-cell malignancies, which include, but are not limited to, B-cell non- Hodgkin's lymphomas (B-NHLs). B-NHLs may be indolent (or low-grade), intermediate- grade (or aggressive) or high-grade (very aggressive). Indolent Bcell lymphomas include follicular lymphoma (FL); small lymphocytic lymphoma (SLL); marginal zone lymphoma (MZL) including nodal MZL, extranodal MZL, splenic MZL and splenic MZL with villous lymphocytes; lymphoplasmacytic lymphoma (LPL); and mucosa-associated-lymphoid tissue (MALT or extranodal marginal zone) lymphoma. Intermediate-grade B-NHLs include mantle cell lymphoma (MCL) with or without leukemic involvement, diffuse large cell lymphoma (DLBCL), follicular large cell (or grade 3 or grade 3B) lymphoma, and primary mediastinal lymphoma (PML). High-grade B-NHLs include Burkitt's lymphoma (BL), Burkitt-like lymphoma, small non-cleaved cell lymphoma (SNCCL) and lymphoblastic lymphoma. Other B-NHLs include immunoblastic lymphoma (or immunocytoma), primary effusion lymphoma, HIV associated (or AIDS related) lymphomas, and post-transplant lymphoproliferative disorder (PTLD) or lymphoma. B-cell malignancies also include, but are not limited to, chronic lymphocytic leukemia (CLL), prolymphocytic leukemia (PLL), Waldenstrom's macroglobulinemia (WM), hairy cell leukemia (HCL), large granular lymphocyte (LGL) leukemia, acute lymphoid (or lymphocytic or lymphoblastic) leukemia, and Castleman's disease. NHL may also include T-cell non-Hodgkin's lymphoma s (T-NHLs), which include, but are not limited to T-cell non-Hodgkin's lymphoma not otherwise specified (NOS), peripheral T-cell lymphoma (PTCL), anaplastic large cell lymphoma (ALCL), angioimmunoblastic lymphoid disorder (AILD), nasal natural killer (NK) cell/T-cell lymphoma, gamma/delta lymphoma, cutaneous T cell lymphoma, mycosis fungoides, and Sezary syndrome. [0066] Hematopoietic cancers also include Hodgkin's lymphoma (or disease) including classical Hodgkin's lymphoma, nodular sclerosing Hodgkin's lymphoma, mixed cellularity Hodgkin's lymphoma, lymphocyte predominant (LP) Hodgkin's lymphoma, nodular LP Hodgkin's lymphoma, and lymphocyte depleted Hodgkin's lymphoma. Hematopoietic cancers also include plasma cell diseases or cancers such as multiple myeloma (MM) including smoldering MM, monoclonal gammopathy of undetermined (or unknown or unclear) significance (MGUS), plasmacytoma (bone, extramedullary), lymphoplasmacytic lymphoma (LPL), Waldenstrom's Macroglobulinemia, plasma cell leukemia, and primary amyloidosis (AL). Hematopoietic cancers may also include other cancers of additional hematopoietic cells, including polymorphonuclear leukocytes (or neutrophils), basophils, eosinophils, dendritic cells, platelets, erythrocytes and natural killer cells. Tissues which include hematopoietic cells referred herein to as “hematopoietic cell tissues” include bone marrow; peripheral blood; thymus; and peripheral lymphoid tissues, such as spleen, lymph nodes, lymphoid tissues associated with mucosa (such as the gut-associated lymphoid tissues), tonsils, Peyer's patches and appendix, and lymphoid tissues associated with other mucosa, for example, the bronchial linings. [0067] Exemplary cancers that may be treated with a compound or method provided herein include brain cancer, glioma, glioblastoma, neuroblastoma, prostate cancer, colorectal cancer, pancreatic cancer, Medulloblastoma, melanoma, cervical cancer, gastric cancer, ovarian cancer, lung cancer, cancer of the head, Hodgkin's Disease, and Non-Hodgkin's Lymphomas. Exemplary cancers that may be treated with a compound or method provided herein include cancer of the thyroid, endocrine system, brain, breast, cervix, colon, head & neck, liver, kidney, lung, ovary, pancreas, rectum, stomach, and uterus. Additional examples include, thyroid carcinoma, cholangiocarcinoma, pancreatic adenocarcinoma, skin cutaneous melanoma, colon adenocarcinoma, rectum adenocarcinoma, stomach adenocarcinoma, esophageal carcinoma, head and neck squamous cell carcinoma, breast invasive carcinoma, lung adenocarcinoma, lung squamous cell carcinoma, non-small cell lung carcinoma, mesothelioma, multiple myeloma, neuroblastoma, glioma, glioblastoma multiforme, ovarian cancer, rhabdomyosarcoma, primary thrombocytosis, primary macroglobulinemia, primary brain tumors, nasopharyngeal tumors, spinal cord tumors, malignant pancreatic insulanoma, malignant carcinoid, urinary bladder cancer, premalignant skin lesions, testicular cancer, thyroid cancer, neuroblastoma, esophageal cancer, genitourinary tract cancer, malignant hypercalcemia, endometrial cancer, adrenal cortical cancer, neoplasms of the endocrine or exocrine pancreas, medullary thyroid cancer, medullary thyroid carcinoma, melanoma, colorectal cancer, papillary thyroid cancer, hepatocellular carcinoma, or prostate cancer. [0068] In embodiments, the cancers that may be treated with a compound or method provided herein include epithelial-derived tumors including ovarian, uterine, breast, endometrial, pancreatic, nasopharyngeal, kidney, lung, colorectal, testicular, spinal cord, and brain tumors. In embodiments, the cancers that may be treated with a compound or a method provided herein include serous and endometrioid epithelial ovarian cancer, renal cancer, endometrial adenocarcinoma, non-small cell lung carcinoma (NSCLC) of the adenocarcinoma subtype, mesotheliomas, and triple-negative breast cancer (TNBC). [0069] An "advanced" cancer is one which has spread outside the site or organ of origin, either by local invasion or metastasis. The term "advanced" cancer includes both locally advanced and metastatic disease. "Metastatic" cancer refers to cancer that has spread from one part of the body to another part of the body. A "refractory" cancer is one that progresses even though an anti-tumor treatment, such as a chemotherapy, is administered to the cancer patient. An example of a refractory cancer is one which is platinum refractory. A "recurrent" cancer is one that has regrown, either at the initial site or at a distant site, after a response to initial therapy. [0070] “Affinity” refers to the strength of the sum total of noncovalent interactions between a single binding site of a molecule (e.g., a ligand) and its binding partner (e.g., a receptor). Unless indicated otherwise, as used herein, “binding affinity” refers to intrinsic binding affinity which reflects a 1:1 interaction between members of a binding pair (e.g., ligand and receptor). The affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (KD). Affinity can be measured by common methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described in the following. [0071] In one embodiment, a dissociation constant (KD) can be measured using a BIACORE surface plasmon resonance (SPR) assay. Surface plasmon resonance refers to an optical phenomenon that allows for the analysis of real-time interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIACORE system (Biacore Life Sciences division of GE Healthcare, Piscataway, NJ). [0072] The term “FRα” or “FOLR1,” as used herein, refers to any native FRα from any vertebrate source, including mammals such as primates (e.g. humans, cynomolgus monkey (cyno)) and rodents (e.g., mice and rats), unless otherwise indicated. FRα is also referred to as "human folate receptor 1," and "FOLR 1". FRα is a single chain membrane protein capable of binding to folic acid and its analogs or derivatives. The term encompasses “full- length,” unprocessed FRα as well as any form of FRα that results from processing in the cell. The term also encompasses naturally occurring variants of FRα, e.g., splice variants, allelic variants, and isoforms. Human FRα sequences are known and include, for example, the sequences publicly available at UniProtKB Accession No. P 15328 (including isoforms). [0073] The term “FR-expressing cancer” refers to a cancer comprising cells that express FR on their surface. Similarly, the term “FRα-expressing cancer” refers to a cancer comprising cells that express FRα on their surface. [0074] The term "increased expression" or "overexpression" of FR, such as FRα, in a particular tumor, tissue, or cell sample refers to FR, such as FRα (a FR, such as FRα polypeptide or a nucleic acid encoding such a polypeptide) that is present at a level higher than that which is present in a healthy or non- diseased (native, wild type) tissue or cells of the same type or origin. Such increased expression or overexpression can be caused, for example, by mutation, gene amplification, increased transcription, increased translation, or increased protein stability. [0075] The term “cytotoxic agent,” “payload,” or “drug” as used herein refers to a substance that inhibits or prevents a cellular function and/or causes cell death or destruction. Cytotoxic agents include, but are not limited to, radioactive isotopes (e.g., 211At, 131I, 125I, 90Y, 186Re, 188Re, 153Sm, 212Bi, 32P, 212Pb and radioactive isotopes of Lu); chemotherapeutic agents or drugs (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 of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof; and the various antitumor or anticancer agents disclosed below. [0076] A “chemotherapeutic agent” is a chemical compound useful in the treatment of a cancer. Examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN®); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); delta-9-tetrahydrocannabinol (dronabinol, MARINOL®); beta-lapachone; lapachol; colchicines; betulinic acid; a camptothecin (including the synthetic analogue topotecan (HYCAMTIN®), CPT-11 (irinotecan, CAMPTOSAR®), acetylcamptothecin, scopolectin, and 9-aminocamptothecin); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); podophyllotoxin; podophyllinic acid; teniposide; cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; auristatin; pyrrolobenzodiazepine; anthracycline; duostatin; duocarmycin (including the synthetic analogues, KW-2189 and CB1-TM1); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosoureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e. g., calicheamicin, especially calicheamicin gamma1I and calicheamicin omegaI1 (see, e.g., Agnew, Chem Intl. Ed. Engl., 33: 183-186 (1994)); dynemicin, including dynemicin A; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino- doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, porfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5- fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfornithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; 2- ethylhydrazide; procarbazine; PSK® polysaccharide complex (JHS Natural Products, Eugene, OR); razoxane; rhizoxin; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine (ELDISINE®, FILDESIN®); dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside (“Ara-C”); thiotepa; taxoids, e.g., paclitaxel (TAXOL®; Bristol-Myers Squibb Oncology, Princeton, N.J.), ABRAXANETM Cremophor-free, albumin-engineered nanoparticle formulation of paclitaxel (American Pharmaceutical Partners, Schaumberg, Illinois), and docetaxel (TAXOTERE®; Rhône-Poulenc Rorer, Antony, France); chloranbucil; gemcitabine (GEMZAR®); 6-thioguanine; mercaptopurine; methotrexate; tyrosine kinase inhibitors; MEK inhibitors; platinum analogs such as cisplatin and carboplatin; vinblastine (VELBAN®); platinum; etoposide (VP-16); ifosfamide; mitoxantrone; vincristine (ONCOVIN®); oxaliplatin; leucovovin; vinorelbine (NAVELBINE®); novantrone; edatrexate; daunomycin; aminopterin; ibandronate; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoids such as retinoic acid; capecitabine (XELODA®); pharmaceutically acceptable salts, acids or derivatives of any of the above; as well as combinations of two or more of the above such as CHOP, an abbreviation for a combined therapy of cyclophosphamide, doxorubicin, vincristine, and prednisolone; CVP, an abbreviation for a combined therapy of cyclophosphamide, vincristine, and prednisolone; and FOLFOX, an abbreviation for a treatment regimen with oxaliplatin (ELOXATINTM) combined with 5-FU and leucovorin. [0077] A “small molecule-drug conjugate” or “SMDC” is a targeting ligand conjugated to one or more heterologous molecule(s), including but not limited to a cytotoxic agent. The targeting ligand can be, for example, folic acid as described herein, or its analogs or derivatives (which target the folate receptor). The cytotoxic agent can be any cytotoxic agent described herein. The targeting ligand can be directly linked to the cytotoxic agent via a covalent bond, or the targeting ligand can be linked to the cytotoxic agent indirectly via a linker. Typically, the linker is covalently bonded to the targeting ligand and also covalently bonded to the cytotoxic agent. Such a linker may be a cleavable linker, for example, cleavable under certain pH condition (pH sensitive linker such as acetals or hydrazones), cleavable by a protease (protease sensitive linker such as peptide linkers), or cleavable in the presence of glutathione (glutathione sensitive linker such as disulfide linkers). In some examples, the linker comprises a protease cleavage site, which may contain 2-5 amino acid residues that are recognizable and/or cleavable by a suitable protease. Such a peptide may comprise naturally-occurring amino acid residues, non-naturally occurring amino acid residues, modified amino acid residues, or a combination thereof. In one example, the peptide linker can be a dipeptide linker. Examples include a valine-citrulline (val-cit or VC) linker, a phenylalanine-lysine (phe-lys) linker, or maleimidocapronic-valine-citruline-p- aminobenzyloxycarbonyl (MC-VC-PAB) linker. Alternatively, the linker may be non- cleavable, e.g., a linker comprising optionally substituted alkane or thioether. In some examples, the linker may comprise a functional group that can form a covalent bond with the targeting ligand. Exemplary functional groups include, but are not limited to, a maleimide group, an iodoacetamide group, a vinyl sulfone group, an acrylate group, an acrylamide group, an acrylonitrile group, or a methacrylate group. The term “small molecule-drug conjugate” or “SMDC” refers to a conjugate wherein at least one cytotoxic agent is a therapeutic moiety such as a drug (“D”). As used herein, “D” refers to drug moiety and includes analogs or derivatives thereof. Thus, folic acid, or the analog or the derivative thereof, is covalently bound to the linker (L), and the drug, or the analog or the derivative thereof, is also covalently bound to the linker (L). The linker (L) can comprise multiple linkers. For example, the linker (L) can comprise one or more components selected from spacer linkers, releasable linkers, and heteroatom linkers, and any combinations thereof, in any order. [0078] As used herein, the term “conjugated” when referring to two moieties means the two moieties are bonded, wherein the bond or bonds connecting the two moieties may be covalent or non-covalent. In embodiments, the two moieties are covalently bonded to each other (e.g. directly or through a covalently bonded intermediary). In embodiments, the two moieties are non-covalently bonded (e.g. through ionic bond(s), van der waal’s bond(s)/interactions, hydrogen bond(s), polar bond(s), or combinations or mixtures thereof). [0079] An “individual” or “subject” is a mammal. Mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats). In certain embodiments, the individual or subject is a human. In certain embodiments, the subject is an adult, an adolescent, a child, or an infant. In some embodiments, the terms “individual” or “patient” are used and are intended to be interchangeable with “subject”. [0080] The term “pharmaceutically acceptable salts” is meant to include salts of the active compounds that are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein. When compounds of the present disclosure contain relatively acidic functionalities, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt. When compounds of the present disclosure contain relatively basic functionalities, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p- tolylsulfonic, citric, tartaric, oxalic, methanesulfonic, and the like. Also included are salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, for example, Berge et al., “Pharmaceutical Salts”, Journal of Pharmaceutical Science, 1977, 66, 1-19). Certain specific compounds of the present disclosure contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts. [0081] Thus, the compounds of the present disclosure may exist as salts, such as with pharmaceutically acceptable acids. The present disclosure includes such salts. Non-limiting examples of such salts include hydrochlorides, hydrobromides, phosphates, sulfates, methanesulfonates, nitrates, maleates, acetates, citrates, fumarates, proprionates, tartrates (e.g., (+)-tartrates, (-)-tartrates, or mixtures thereof including racemic mixtures), succinates, benzoates, and salts with amino acids such as glutamic acid, and quaternary ammonium salts (e.g. methyl iodide, ethyl iodide, and the like). These salts may be prepared by methods known to those skilled in the art. [0082] The neutral forms of the compounds are preferably regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner. The parent form of the compound may differ from the various salt forms in certain physical properties, such as solubility in polar solvents. [0083] In addition to salt forms, the present disclosure provides compounds, which are in a prodrug form. Prodrugs of the compounds described herein are those compounds that readily undergo chemical changes under physiological conditions to provide the compounds of the present disclosure. Prodrugs of the compounds described herein may be converted in vivo after administration. Additionally, prodrugs can be converted to the compounds of the present disclosure by chemical or biochemical methods in an ex vivo environment, such as, for example, when contacted with a suitable enzyme or chemical reagent. [0084] Certain compounds of the present disclosure can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are encompassed within the scope of the present disclosure. Certain compounds of the present disclosure may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present disclosure and are intended to be within the scope of the present disclosure. [0085] “Pharmaceutically acceptable excipient” and “pharmaceutically acceptable carrier” refer to a substance that aids the administration of an active agent to and absorption by a subject and can be included in the compositions of the present disclosure without causing a significant adverse toxicological effect on the patient. Non-limiting examples of pharmaceutically acceptable excipients include water, NaCl, normal saline solutions, lactated Ringer’s, normal sucrose, normal glucose, binders, fillers, disintegrants, lubricants, coatings, sweeteners, flavors, salt solutions (such as Ringer’s solution), alcohols, oils, gelatins, carbohydrates such as lactose, amylose or starch, fatty acid esters, hydroxymethycellulose, polyvinyl pyrrolidine, and colors, and the like. Such preparations can be sterilized and, if desired, mixed with auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the disclosure. One of skill in the art will recognize that other pharmaceutical excipients are useful in the present disclosure. [0086] 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. [0087] The term “administering”, “administered” and grammatical variants refers to the physical introduction of an agent to a subject, using any of the various methods and delivery systems known to those skilled in the art. Exemplary routes of administration for the formulations disclosed herein include intravenous, intramuscular, subcutaneous, intraperitoneal, spinal or other parenteral routes of administration, for example by injection or infusion. The phrase “parenteral administration” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intralymphatic, intralesional, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion, as well as in vivo electroporation. In some embodiments, the formulation is administered via a non-parenteral route, e.g., orally. Other non-parenteral routes include a topical, epidermal or mucosal route of administration, for example, intranasally, vaginally, rectally, sublingually or topically. Administering can also be performed, for example, once, a plurality of times, and/or over one or more extended periods. [0088] Administration “in combination with” one or more further therapeutic agents includes simultaneous (concurrent) or consecutive administration in any order. The combination therapy can provide “synergy” and prove “synergistic”, i.e., the effect achieved when the active ingredients used together is greater than the sum of the effects that results from using the compounds separately. A synergistic effect can be attained when the active ingredients are: (1) co-formulated and administered or delivered simultaneously in a combined, unit dosage formulation; (2) delivered serially, by alternation, or in parallel as separate formulations; or (3) by some other regimen. When delivered in alternation therapy, a synergistic effect can be attained when the compounds are administered or delivered sequentially, e.g., by different injections in separate syringes. A synergistic combination produces effects that are greater than the additive effects of the individual components of the combination. [0089] An “effective amount” of an agent, e.g., a pharmaceutical formulation, refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result. [0090] The abbreviations used herein have their conventional meaning within the chemical and biological arts. The chemical structures and formulae set forth herein are constructed according to the standard rules of chemical valency known in the chemical arts. [0091] The term saccharide means carbohydrate (or sugar). In embodiments, the saccharide is a monosaccharide. In embodiments, the saccharide is a polysaccharide. The most basic unit of saccharide is a monomer of carbohydrate. The general formula is CnH2nOn. The term saccharide derivative means sugar molecules that have been modified with substituents other than hydroxyl groups. Examples include glycosylamines, sugar phosphates, and sugar esters. Other saccharide derivatives include for example beta-D-glucuronyl, D-galactosyl, and D-glucosyl. [0092] The term “Charged Group” means a chemical group bearing a positive or a negative charge, such as for example phosphate, phosphonate, sulfate, sulfonate, nitrate, carboxylate, carbonate, and the like. In some embodiments, a Charged Group is at least 50% ionized in aqueous solution at least one pH in the range of 5-9. In some embodiments, a Charged Group is an anionic Charged Group. [0093] “Linker” or “linker reagent” are used interchangeably and refer to a chemical moiety comprising a covalent bond or a chain of atoms that covalently attaches a targeting ligand to a drug moiety. In various embodiments, linkers include a divalent radical. In various embodiments, linkers can comprise one or more amino acid residues. The linker can be cleavable or non-cleavable. [0094] “Amino Acid Unit” has the formula
Figure imgf000033_0001
hydrogen, methyl, isopropyl, isobutyl, sec-butyl, benzyl, p-hydroxybenzyl, —CH2OH, —CH(OH)CH3, — CH2SH —CH2CH2SCH3, —CH2CONH2, —CH2COOH, —CH2CH2CONH2, —CH2CH2COOH, —(CH2)3NHC(═NH)NH2, —(CH2)3NH2, —(CH2)3NHCOCH3, —(CH2)3NHCHO, —(CH2)4NHC(═NH)NH2, —(CH2)4NH2, —(CH2)4NHCOCH3, —(CH2)4NHCHO, —(CH2)3NHCONH2, —(CH2)4NHCONH2, —CH2CH2CH(OH)CH2NH2, 2-pyridylmethyl-, 3-pyridylmethyl-, 4-pyridylmethyl-, phenyl, or cyclohexyl; or the formula
Figure imgf000033_0002
. embodiments, “Amino Acid Unit” has the formula
Figure imgf000033_0003
methyl, isopropyl, isobutyl, sec-butyl, benzyl, p-hydroxybenzyl, —CH2OH, —CH(OH)CH3, — CH2CH2SCH3, —CH2CONH2, —CH2COOH, —CH2CH2CONH2, —CH2CH2COOH, —(CH2)3NHC(═NH)NH2, —(CH2)3NH2, —CH2SH, —(CH2)3NHCOCH3, —(CH2)3NHCHO, —(CH2)4NHC(═NH)NH2, —(CH2)4NH2, —(CH2)4NHCOCH3, —(CH2)4NHCHO, —(CH2)3NHCONH2, —(CH2)4NHCONH2, —CH2CH2CH(OH)CH2NH2, 2-pyridylmethyl-, 3-pyridylmethyl-, 4-pyridylmethyl-, phenyl, or cyclohexyl. In embodiments, “Amino Acid Unit” has the formula
Figure imgf000034_0001
. various embodiments, Amino Acid Unit includes not only naturally occurring amino acids but also minor amino acids and non-naturally occurring amino acid analogs, such as for example, citrulline, norleucine, selenomethionine, β-amino acids (e.g., β-alanine, β-aspargine), and the like. In embodiments, the amino acid can be a modified amino acid, such as for example, α- amino acid amide, oxazole amino acid, thiazole amino acid, triazole amino acid, and the like. In embodiments, the modified amino acid has the formula
Figure imgf000034_0002
Figure imgf000034_0003
. An amino acid unit may be referred to by its standard three-letter code for the amino acid (e.g., Ala, Cys, Asp, Glu etc.). [0095] “Peptoid” has the formula
Figure imgf000034_0004
, here R0 is methyl, isopropyl, isobutyl, sec-butyl, benzyl, p-hydroxybenzyl, —CH2OH, —CH(OH)CH3, —CH2CH2SCH3, — CH2CONH2, —CH2COOH, —CH2CH2CONH2, —CH2CH2COOH, — (CH2)3NHC(═NH)NH2, —(CH2)3NH2, —(CH2)3NHCOCH3, —(CH2)3NHCHO, — (CH2)4NHC(═NH)NH2, —(CH2)4NH2, —(CH2)4NHCOCH3, —(CH2)4NHCHO, —(CH2)3NHCONH2, —(CH2)4NHCONH2, —CH2CH2CH(OH)CH2NH2, 2-pyridylmethyl-, 3-pyridylmethyl-, 4-pyridylmethyl-, phenyl, or cyclohexyl. In embodiments, peptoid has the formula
Figure imgf000034_0005
. [0096] As used herein, the term “half-life extender” refers to molecules that extend half-life of biopharmaceuticals. In embodiments, the half-life of SMDCs described herein can be extended by PEGylation (covalently linking a PEG to the SMDC), lipidation (covalently linking a lipid to the SMDC). In embodiments, the half-life of SMDCs described herein can be extended by covalently linking a cholesterol-like compound to the SMDC. In embodiments, the half-life of SMDCs described herein can be extended by covalently linking a small molecule albumin binder to the SMDC. [0097] As used herein, the term “cholesterol-like” half-life extender refers to a compound with a structure closely resembling cholesterol with a linker on the hydroxyl end. In embodiments, “cholesterol-like” half-life extender refers to
Figure imgf000035_0001
. [0098] As used herein, the term “albumin binder” half-life extender refers to a small molecule that can be reversibly (non-covalently) bound by serum albumin. Serum albumin can bind a large diversity of small organic molecules such as fatty acids, dicarboxylic acids, bulky heterocycles, and aromatic carboxylic acids with a peripheric negative charge. In
Figure imgf000035_0002
[0099] As used herein, the terms “bioconjugate” and “bioconjugate linker” refers to the resulting association between atoms or molecules of “bioconjugate reactive groups” or “bioconjugate reactive moieties”. The association can be direct or indirect. For example, a conjugate between a first bioconjugate reactive group (e g –NH2 –C(O)OH –N- hydroxysuccinimide, or –maleimide) and a second bioconjugate reactive group (e.g., sulfhydryl, sulfur-containing amino acid, amine, amine sidechain containing amino acid, or carboxylate) provided herein can be direct, e.g., by covalent bond or linker (e.g. a first linker of second linker), or indirect, e.g., by non-covalent bond (e.g. electrostatic interactions (e.g. ionic bond, hydrogen bond, halogen bond), van der Waals interactions (e.g. dipole-dipole, dipole-induced dipole, London dispersion), ring stacking (pi effects), hydrophobic interactions and the like). In embodiments, bioconjugates or bioconjugate linkers are formed using bioconjugate chemistry (i.e. the association of two bioconjugate reactive groups) including, but are not limited to nucleophilic substitutions (e.g., reactions of amines and alcohols with acyl halides, active esters), electrophilic substitutions (e.g., enamine reactions) and additions to carbon-carbon and carbon-heteroatom multiple bonds (e.g., Michael reaction, Diels-Alder addition). These and other useful reactions are discussed in, for example, March, ADVANCED ORGANIC CHEMISTRY, 3rd Ed., John Wiley & Sons, New York, 1985; Hermanson, BIOCONJUGATE TECHNIQUES, Academic Press, San Diego, 1996; and Feeney et al., MODIFICATION OF PROTEINS; Advances in Chemistry Series, Vol. 198, American Chemical Society, Washington, D.C., 1982. In embodiments, the first bioconjugate reactive group (e.g., maleimide moiety) is covalently attached to the second bioconjugate reactive group (e.g. a sulfhydryl). In embodiments, the first bioconjugate reactive group (e.g., haloacetyl moiety) is covalently attached to the second bioconjugate reactive group (e.g. a sulfhydryl). In embodiments, the first bioconjugate reactive group (e.g., pyridyl moiety) is covalently attached to the second bioconjugate reactive group (e.g. a sulfhydryl). In embodiments, the first bioconjugate reactive group (e.g., –N- hydroxysuccinimide moiety) is covalently attached to the second bioconjugate reactive group (e.g. an amine). In embodiments, the first bioconjugate reactive group (e.g., fluorophenyl ester moiety) reacts with the second bioconjugate reactive group (e.g. an amine) to form a covalent bond. In embodiments, the first bioconjugate reactive group (e.g., –sulfo–N- hydroxysuccinimide moiety) reacts with the second bioconjugate reactive group (e.g. an amine) to form a covalent bond. [00100] Useful bioconjugate reactive moieties used for bioconjugate chemistries herein include, for example: (a) carboxyl groups and various derivatives thereof including, but not limited to, N-hydroxysuccinimide esters, N-hydroxybenztriazole esters, acid halides, acyl imidazoles, thioesters, p-nitrophenyl esters, alkyl, alkenyl, alkynyl and aromatic esters; (b) hydroxyl groups which can be converted to esters, ethers, aldehydes, etc. (c) haloalkyl groups wherein the halide can be later displaced with a nucleophilic group such as, for example, an amine, a carboxylate anion, thiol anion, carbanion, or an alkoxide ion, thereby resulting in the covalent attachment of a new group at the site of the halogen atom; (d) dienophile groups which are capable of participating in Diels-Alder reactions such as, for example, maleimido or maleimide groups; (e) aldehyde or ketone groups such that subsequent derivatization is possible via formation of carbonyl derivatives such as, for example, imines, hydrazones, semicarbazones or oximes, or via such mechanisms as Grignard addition or alkyllithium addition; (f) sulfonyl halide groups for subsequent reaction with amines, for example, to form sulfonamides; (g) thiol groups, which can be converted to disulfides, reacted with acyl halides, or bonded to metals such as gold, or react with maleimides; (h) amine or sulfhydryl groups (e.g., present in cysteine), which can be, for example, acylated, alkylated or oxidized; (i) alkenes, which can undergo, for example, cycloadditions, acylation, Michael addition, etc; (j) epoxides, which can react with, for example, amines and hydroxyl compounds; (k) phosphoramidites and other standard functional groups useful in nucleic acid synthesis; (l) metal silicon oxide bonding; and (m) metal bonding to reactive phosphorus groups (e.g. phosphines) to form, for example, phosphate diester bonds. (n) azides coupled to alkynes using copper catalyzed cycloaddition click chemistry. (o) biotin conjugate can react with avidin or strepavidin to form a avidin-biotin complex or streptavidin-biotin complex. [00101] The bioconjugate reactive groups can be chosen such that they do not participate in, or interfere with, the chemical stability of the conjugate described herein. Alternatively, a reactive functional group can be protected from participating in the crosslinking reaction by the presence of a protecting group. In embodiments, the bioconjugate comprises a molecular entity derived from the reaction of an unsaturated bond, such as a maleimide, and a sulfhydryl group. [00102] “Derivative” is a compound that is derived from a similar compound by a chemical reaction. In biochemistry, the word is used for compounds that at least theoretically can be formed from the precursor compound. In the past, derivative also meant a compound that can be imagined to arise from another compound, if one atom or group of atoms is replaced with another atom or group of atoms, but modern chemical language now uses the term structural analog for this meaning, thus eliminating ambiguity. [00103] “Analog,” or “analogue” is used in accordance with its plain ordinary meaning within Chemistry and Biology and refers to a chemical compound that is structurally similar to another compound (i.e., a so-called “reference” compound) but differs in composition, e.g., in the replacement of one atom by an atom of a different element, or in the presence of a particular functional group, or the replacement of one functional group by another functional group, or the absolute stereochemistry of one or more chiral centers of the reference compound. [00104] The terms “folic acid” and “folate” are often used interchangeably, though more appropriately, “folic acid” refers to the fully oxidized synthetic compound (pteroylmono- glutamic acid) used in dietary supplements and in food fortification, whereas “folate” refers to the various tetrahydrofolate derivatives naturally present in foods. Reduced folates are found as the partially reduced form 7,8-dihydrofolate or the reduced species 5,6,7,8- tetrahydrofolate (THF). The terms “folic acid” and “folate” are used interchangeably herein to refer to the fully oxidized synthetic compound. [00105] The term “alkyl,” by itself or as part of another substituent, means, unless otherwise stated, a straight (i.e., unbranched) or branched carbon chain (or carbon), or combination thereof, which may be fully saturated, mono- or polyunsaturated and can include mono-, di- and multivalent radicals. The alkyl may include a designated number of carbons (e.g., C1-C10 means one to ten carbons). Alkyl is an uncyclized chain. Examples of saturated hydrocarbon radicals include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, methyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like. An unsaturated alkyl group is one having one or more double bonds or triple bonds. Examples of unsaturated alkyl groups include, but are not limited to, vinyl, 2-propenyl, crotyl, 2-isopentenyl, 2-(butadienyl), 2,4- pentadienyl, 3-(1,4-pentadienyl), ethynyl, 1- and 3-propynyl, 3-butynyl, and the higher homologs and isomers. An alkoxy is an alkyl attached to the remainder of the molecule via an oxygen linker (-O-). An alkyl moiety may be an alkenyl moiety. An alkyl moiety may be an alkynyl moiety. An alkyl moiety may be fully saturated. An alkenyl may include more than one double bond and/or one or more triple bonds in addition to the one or more double bonds. An alkynyl may include more than one triple bond and/or one or more double bonds in addition to the one or more triple bonds. [00106] The term “alkylene,” by itself or as part of another substituent, means, unless otherwise stated, a divalent radical derived from an alkyl, as exemplified, but not limited by, -CH2CH2CH2CH2-. Typically, an alkyl (or alkylene) group will have from 1 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being preferred herein. A “lower alkyl” or “lower alkylene” is a shorter chain alkyl or alkylene group, generally having eight or fewer carbon atoms. The term “alkenylene,” by itself or as part of another substituent, means, unless otherwise stated, a divalent radical derived from an alkene. [00107] The term “heteroalkyl,” by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched chain, or combinations thereof, including at least one carbon atom and at least one heteroatom (e.g., O, N, P, Si, or S), and wherein the nitrogen and sulfur atoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized. The heteroatom(s) (e.g., O, N, S, Si, or P) may be placed at any interior position of the heteroalkyl group or at the position at which the alkyl group is attached to the remainder of the molecule. Heteroalkyl is an uncyclized chain. Examples include, but are not limited to: -CH2-CH2-O-CH3, -CH2-CH2-NH-CH3, -CH2-CH2- N(CH3)-CH3, -CH2-S-CH2-CH3, -CH2-S-CH2, -S(O)-CH3, -CH2-CH2-S(O)2-CH3, -CH=CH- O-CH3, -Si(CH3)3, -CH2-CH=N-OCH3, -CH=CH-N(CH3)-CH3, -O-CH3, -O-CH2-CH3, and - CN. Up to two or three heteroatoms may be consecutive, such as, for example, -CH2-NH- OCH3 and -CH2-O-Si(CH3)3. A heteroalkyl moiety may include one heteroatom (e.g., O, N, S, Si, or P). A heteroalkyl moiety may include two optionally different heteroatoms (e.g., O, N, S, Si, or P). A heteroalkyl moiety may include three optionally different heteroatoms (e.g., O, N, S, Si, or P). A heteroalkyl moiety may include four optionally different heteroatoms (e.g., O, N, S, Si, or P). A heteroalkyl moiety may include five optionally different heteroatoms (e.g., O, N, S, Si, or P). A heteroalkyl moiety may include up to 8 optionally different heteroatoms (e.g., O, N, S, Si, or P). The term “heteroalkenyl,” by itself or in combination with another term, means, unless otherwise stated, a heteroalkyl including at least one double bond. A heteroalkenyl may optionally include more than one double bond and/or one or more triple bonds in addition to the one or more double bonds. The term “heteroalkynyl,” by itself or in combination with another term, means, unless otherwise stated, a heteroalkyl including at least one triple bond. A heteroalkynyl may optionally include more than one triple bond and/or one or more double bonds in addition to the one or more triple bonds. [00108] Similarly, the term “heteroalkylene,” by itself or as part of another substituent, means, unless otherwise stated, a divalent radical derived from heteroalkyl, as exemplified, but not limited by, -CH2-CH2-S-CH2-CH2- and -CH2-S-CH2-CH2-NH-CH2-. For heteroalkylene groups, heteroatoms can also occupy either or both of the chain termini (e.g., alkyleneoxy, alkylenedioxy, alkyleneamino, alkylenediamino, and the like). Still further, for alkylene and heteroalkylene linking groups, no orientation of the linking group is implied by the direction in which the formula of the linking group is written. For example, the formula - C(O)2R'- represents both -C(O)2R'- and -R'C(O)2-. As described above, heteroalkyl groups, as used herein, include those groups that are attached to the remainder of the molecule through a heteroatom, such as -C(O)R', -C(O)NR', -NR'R'', -OR', -SR', and/or -SO2R'. Where “heteroalkyl” is recited, followed by recitations of specific heteroalkyl groups, such as - NR'R'' or the like, it will be understood that the terms heteroalkyl and -NR'R'' are not redundant or mutually exclusive. Rather, the specific heteroalkyl groups are recited to add clarity. Thus, the term “heteroalkyl” should not be interpreted herein as excluding specific heteroalkyl groups, such as -NR'R'' or the like. [00109] The terms “cycloalkyl” and “heterocycloalkyl,” by themselves or in combination with other terms, mean, unless otherwise stated, cyclic versions of “alkyl” and “heteroalkyl,” respectively. Cycloalkyl and heterocycloalkyl are not aromatic. Additionally, for heterocycloalkyl, a heteroatom can occupy the position at which the heterocycle is attached to the remainder of the molecule. Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like. Examples of heterocycloalkyl include, but are not limited to, 1- (1,2,5,6-tetrahydropyridyl), 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-morpholinyl, 3- morpholinyl, tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothien-2-yl, tetrahydrothien-3-yl, 1-piperazinyl, 2-piperazinyl, and the like. A “cycloalkylene” and a “heterocycloalkylene,” alone or as part of another substituent, means a divalent radical derived from a cycloalkyl and heterocycloalkyl, respectively. [00110] In embodiments, the term “cycloalkyl” means a monocyclic, bicyclic, or a multicyclic cycloalkyl ring system. In embodiments, monocyclic ring systems are cyclic hydrocarbon groups containing from 3 to 8 carbon atoms, where such groups can be saturated or unsaturated, but not aromatic. In embodiments, cycloalkyl groups are fully saturated. Examples of monocyclic cycloalkyls include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, and cyclooctyl. Bicyclic cycloalkyl ring systems are bridged monocyclic rings or fused bicyclic rings. In embodiments, bridged monocyclic rings contain a monocyclic cycloalkyl ring where two non adjacent carbon atoms of the monocyclic ring are linked by an alkylene bridge of between one and three additional carbon atoms (i.e., a bridging group of the form (CH2)w , where w is 1, 2, or 3). Representative examples of bicyclic ring systems include, but are not limited to, bicyclo[3.1.1]heptane, bicyclo[2.2.1]heptane, bicyclo[2.2.2]octane, bicyclo[3.2.2]nonane, bicyclo[3.3.1]nonane, and bicyclo[4.2.1]nonane. In embodiments, fused bicyclic cycloalkyl ring systems contain a monocyclic cycloalkyl ring fused to either a phenyl, a monocyclic cycloalkyl, a monocyclic cycloalkenyl, a monocyclic heterocyclyl, or a monocyclic heteroaryl. In embodiments, the bridged or fused bicyclic cycloalkyl is attached to the parent molecular moiety through any carbon atom contained within the monocyclic cycloalkyl ring. In embodiments, cycloalkyl groups are optionally substituted with one or two groups which are independently oxo or thia. In embodiments, the fused bicyclic cycloalkyl is a 5 or 6 membered monocyclic cycloalkyl ring fused to either a phenyl ring, a 5 or 6 membered monocyclic cycloalkyl, a 5 or 6 membered monocyclic cycloalkenyl, a 5 or 6 membered monocyclic heterocyclyl, or a 5 or 6 membered monocyclic heteroaryl, wherein the fused bicyclic cycloalkyl is optionally substituted by one or two groups which are independently oxo or thia. In embodiments, multicyclic cycloalkyl ring systems are a monocyclic cycloalkyl ring (base ring) fused to either (i) one ring system selected from the group consisting of a bicyclic aryl, a bicyclic heteroaryl, a bicyclic cycloalkyl, a bicyclic cycloalkenyl, and a bicyclic heterocyclyl; or (ii) two other ring systems independently selected from the group consisting of a phenyl, a bicyclic aryl, a monocyclic or bicyclic heteroaryl, a monocyclic or bicyclic cycloalkyl, a monocyclic or bicyclic cycloalkenyl, and a monocyclic or bicyclic heterocyclyl. In embodiments, the multicyclic cycloalkyl is attached to the parent molecular moiety through any carbon atom contained within the base ring. In embodiments, multicyclic cycloalkyl ring systems are a monocyclic cycloalkyl ring (base ring) fused to either (i) one ring system selected from the group consisting of a bicyclic aryl, a bicyclic heteroaryl, a bicyclic cycloalkyl, a bicyclic cycloalkenyl, and a bicyclic heterocyclyl; or (ii) two other ring systems independently selected from the group consisting of a phenyl, a monocyclic heteroaryl, a monocyclic cycloalkyl, a monocyclic cycloalkenyl, and a monocyclic heterocyclyl. Examples of multicyclic cycloalkyl groups include, but are not limited to tetradecahydrophenanthrenyl, perhydrophenothiazin-1-yl, and perhydrophenoxazin-1-yl. [00111] In embodiments, a cycloalkyl is a cycloalkenyl. The term “cycloalkenyl” is used in accordance with its plain ordinary meaning. In embodiments, a cycloalkenyl is a monocyclic, bicyclic, or a multicyclic cycloalkenyl ring system. In embodiments, monocyclic cycloalkenyl ring systems are cyclic hydrocarbon groups containing from 3 to 8 carbon atoms, where such groups are unsaturated (i.e., containing at least one annular carbon carbon double bond), but not aromatic. Examples of monocyclic cycloalkenyl ring systems include cyclopentenyl and cyclohexenyl. In embodiments, bicyclic cycloalkenyl rings are bridged monocyclic rings or a fused bicyclic rings. In embodiments, bridged monocyclic rings contain a monocyclic cycloalkenyl ring where two non adjacent carbon atoms of the monocyclic ring are linked by an alkylene bridge of between one and three additional carbon atoms (i.e., a bridging group of the form (CH2)w, where w is 1, 2, or 3). Representative examples of bicyclic cycloalkenyls include, but are not limited to, norbornenyl and bicyclo[2.2.2]oct 2 enyl. In embodiments, fused bicyclic cycloalkenyl ring systems contain a monocyclic cycloalkenyl ring fused to either a phenyl, a monocyclic cycloalkyl, a monocyclic cycloalkenyl, a monocyclic heterocyclyl, or a monocyclic heteroaryl. In embodiments, the bridged or fused bicyclic cycloalkenyl is attached to the parent molecular moiety through any carbon atom contained within the monocyclic cycloalkenyl ring. In embodiments, cycloalkenyl groups are optionally substituted with one or two groups which are independently oxo or thia. In embodiments, multicyclic cycloalkenyl rings contain a monocyclic cycloalkenyl ring (base ring) fused to either (i) one ring system selected from the group consisting of a bicyclic aryl, a bicyclic heteroaryl, a bicyclic cycloalkyl, a bicyclic cycloalkenyl, and a bicyclic heterocyclyl; or (ii) two ring systems independently selected from the group consisting of a phenyl, a bicyclic aryl, a monocyclic or bicyclic heteroaryl, a monocyclic or bicyclic cycloalkyl, a monocyclic or bicyclic cycloalkenyl, and a monocyclic or bicyclic heterocyclyl. In embodiments, the multicyclic cycloalkenyl is attached to the parent molecular moiety through any carbon atom contained within the base ring. In embodiments, multicyclic cycloalkenyl rings contain a monocyclic cycloalkenyl ring (base ring) fused to either (i) one ring system selected from the group consisting of a bicyclic aryl, a bicyclic heteroaryl, a bicyclic cycloalkyl, a bicyclic cycloalkenyl, and a bicyclic heterocyclyl; or (ii) two ring systems independently selected from the group consisting of a phenyl, a monocyclic heteroaryl, a monocyclic cycloalkyl, a monocyclic cycloalkenyl, and a monocyclic heterocyclyl. [00112] In embodiments, a heterocycloalkyl is a heterocyclyl. The term “heterocyclyl” as used herein, means a monocyclic, bicyclic, or multicyclic heterocycle. The heterocyclyl monocyclic heterocycle is a 3, 4, 5, 6 or 7 membered ring containing at least one heteroatom independently selected from the group consisting of O, N, and S where the ring is saturated or unsaturated, but not aromatic. The 3 or 4 membered ring contains 1 heteroatom selected from the group consisting of O, N and S. The 5 membered ring can contain zero or one double bond and one, two or three heteroatoms selected from the group consisting of O, N and S. The 6 or 7 membered ring contains zero, one or two double bonds and one, two or three heteroatoms selected from the group consisting of O, N and S. The heterocyclyl monocyclic heterocycle is connected to the parent molecular moiety through any carbon atom or any nitrogen atom contained within the heterocyclyl monocyclic heterocycle. Representative examples of heterocyclyl monocyclic heterocycles include, but are not limited to, azetidinyl, azepanyl, aziridinyl, diazepanyl, 1,3-dioxanyl, 1,3-dioxolanyl, 1,3-dithiolanyl, 1,3-dithianyl, imidazolinyl, imidazolidinyl, isothiazolinyl, isothiazolidinyl, isoxazolinyl, isoxazolidinyl, morpholinyl, oxadiazolinyl, oxadiazolidinyl, oxazolinyl, oxazolidinyl, piperazinyl, piperidinyl, pyranyl, pyrazolinyl, pyrazolidinyl, pyrrolinyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydrothienyl, thiadiazolinyl, thiadiazolidinyl, thiazolinyl, thiazolidinyl, thiomorpholinyl, 1,1-dioxidothiomorpholinyl (thiomorpholine sulfone), thiopyranyl, and trithianyl. The heterocyclyl bicyclic heterocycle is a monocyclic heterocycle fused to either a phenyl, a monocyclic cycloalkyl, a monocyclic cycloalkenyl, a monocyclic heterocycle, or a monocyclic heteroaryl. The heterocyclyl bicyclic heterocycle is connected to the parent molecular moiety through any carbon atom or any nitrogen atom contained within the monocyclic heterocycle portion of the bicyclic ring system. Representative examples of bicyclic heterocyclyls include, but are not limited to, 2,3-dihydrobenzofuran-2-yl, 2,3- dihydrobenzofuran-3-yl, indolin-1-yl, indolin-2-yl, indolin-3-yl, 2,3-dihydrobenzothien-2-yl, decahydroquinolinyl, decahydroisoquinolinyl, octahydro-1H-indolyl, and octahydrobenzofuranyl. In embodiments, heterocyclyl groups are optionally substituted with one or two groups which are independently oxo or thia. In certain embodiments, the bicyclic heterocyclyl is a 5 or 6 membered monocyclic heterocyclyl ring fused to a phenyl ring, a 5 or 6 membered monocyclic cycloalkyl, a 5 or 6 membered monocyclic cycloalkenyl, a 5 or 6 membered monocyclic heterocyclyl, or a 5 or 6 membered monocyclic heteroaryl, wherein the bicyclic heterocyclyl is optionally substituted by one or two groups which are independently oxo or thia. Multicyclic heterocyclyl ring systems are a monocyclic heterocyclyl ring (base ring) fused to either (i) one ring system selected from the group consisting of a bicyclic aryl, a bicyclic heteroaryl, a bicyclic cycloalkyl, a bicyclic cycloalkenyl, and a bicyclic heterocyclyl; or (ii) two other ring systems independently selected from the group consisting of a phenyl, a bicyclic aryl, a monocyclic or bicyclic heteroaryl, a monocyclic or bicyclic cycloalkyl, a monocyclic or bicyclic cycloalkenyl, and a monocyclic or bicyclic heterocyclyl. The multicyclic heterocyclyl is attached to the parent molecular moiety through any carbon atom or nitrogen atom contained within the base ring. In embodiments, multicyclic heterocyclyl ring systems are a monocyclic heterocyclyl ring (base ring) fused to either (i) one ring system selected from the group consisting of a bicyclic aryl, a bicyclic heteroaryl, a bicyclic cycloalkyl, a bicyclic cycloalkenyl, and a bicyclic heterocyclyl; or (ii) two other ring systems independently selected from the group consisting of a phenyl, a monocyclic heteroaryl, a monocyclic cycloalkyl, a monocyclic cycloalkenyl, and a monocyclic heterocyclyl. Examples of multicyclic heterocyclyl groups include, but are not limited to 10H-phenothiazin-10-yl, 9,10-dihydroacridin-9-yl, 9,10-dihydroacridin-10-yl, 10H-phenoxazin-10-yl, 10,11-dihydro-5H-dibenzo[b,f]azepin-5-yl, 1,2,3,4- tetrahydropyrido[4,3-g]isoquinolin-2-yl, 12H-benzo[b]phenoxazin-12-yl, and dodecahydro- 1H-carbazol-9-yl. [00113] The terms “halo” or “halogen,” by themselves or as part of another substituent, mean, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom. Additionally, terms such as “haloalkyl” are meant to include monohaloalkyl and polyhaloalkyl. For example, the term “halo(C1-C4)alkyl” includes, but is not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like. [00114] The term “acyl” means, unless otherwise stated, -C(O)R where R is a substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl. [00115] The term “aryl” means, unless otherwise stated, a polyunsaturated, aromatic, hydrocarbon substituent, which can be a single ring or multiple rings (preferably from 1 to 3 rings) that are fused together (i.e., a fused ring aryl) or linked covalently. A fused ring aryl refers to multiple rings fused together wherein at least one of the fused rings is an aryl ring. The term “heteroaryl” refers to aryl groups (or rings) that contain at least one heteroatom such as N, O, or S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quaternized. Thus, the term “heteroaryl” includes fused ring heteroaryl groups (i.e., multiple rings fused together wherein at least one of the fused rings is a heteroaromatic ring). A 5,6-fused ring heteroarylene refers to two rings fused together, wherein one ring has 5 members and the other ring has 6 members, and wherein at least one ring is a heteroaryl ring. Likewise, a 6,6-fused ring heteroarylene refers to two rings fused together, wherein one ring has 6 members and the other ring has 6 members, and wherein at least one ring is a heteroaryl ring. And a 6,5-fused ring heteroarylene refers to two rings fused together, wherein one ring has 6 members and the other ring has 5 members, and wherein at least one ring is a heteroaryl ring. A heteroaryl group can be attached to the remainder of the molecule through a carbon or heteroatom. Non-limiting examples of aryl and heteroaryl groups include phenyl, naphthyl, pyrrolyl, pyrazolyl, pyridazinyl, triazinyl, pyrimidinyl, imidazolyl, pyrazinyl, purinyl, oxazolyl, isoxazolyl, thiazolyl, furyl, thienyl, pyridyl, pyrimidyl, benzothiazolyl, benzoxazoyl benzimidazolyl, benzofuran, isobenzofuranyl, indolyl, isoindolyl, benzothiophenyl, isoquinolyl, quinoxalinyl, quinolyl, 1-naphthyl, 2- naphthyl, 4-biphenyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4- imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-furyl, 3-furyl, 2-thienyl, 3- thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-benzothiazolyl, purinyl, 2-benzimidazolyl, 5-indolyl, 1-isoquinolyl, 5-isoquinolyl, 2-quinoxalinyl, 5-quinoxalinyl, 3- quinolyl, and 6-quinolyl. Substituents for each of the above noted aryl and heteroaryl ring systems are selected from the group of acceptable substituents described below. An “arylene” and a “heteroarylene,” alone or as part of another substituent, mean a divalent radical derived from an aryl and heteroaryl, respectively. A heteroaryl group substituent may be -O- bonded to a ring heteroatom nitrogen. [00116] A fused ring heterocyloalkyl-aryl is an aryl fused to a heterocycloalkyl. A fused ring heterocycloalkyl-heteroaryl is a heteroaryl fused to a heterocycloalkyl. A fused ring heterocycloalkyl-cycloalkyl is a heterocycloalkyl fused to a cycloalkyl. A fused ring heterocycloalkyl-heterocycloalkyl is a heterocycloalkyl fused to another heterocycloalkyl. Fused ring heterocycloalkyl-aryl, fused ring heterocycloalkyl-heteroaryl, fused ring heterocycloalkyl-cycloalkyl, or fused ring heterocycloalkyl-heterocycloalkyl may each independently be unsubstituted or substituted with one or more of the substitutents described herein. [00117] Spirocyclic rings are two or more rings wherein adjacent rings are attached through a single atom. The individual rings within spirocyclic rings may be identical or different. Individual rings in spirocyclic rings may be substituted or unsubstituted and may have different substituents from other individual rings within a set of spirocyclic rings. Possible substituents for individual rings within spirocyclic rings are the possible substituents for the same ring when not part of spirocyclic rings (e.g. substituents for cycloalkyl or heterocycloalkyl rings). Spirocylic rings may be substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloalkylene, substituted or unsubstituted heterocycloalkyl or substituted or unsubstituted heterocycloalkylene and individual rings within a spirocyclic ring group may be any of the immediately previous list, including having all rings of one type (e.g. all rings being substituted heterocycloalkylene wherein each ring may be the same or different substituted heterocycloalkylene). When referring to a spirocyclic ring system, heterocyclic spirocyclic rings means a spirocyclic rings wherein at least one ring is a heterocyclic ring and wherein each ring may be a different ring. When referring to a spirocyclic ring system, substituted spirocyclic rings means that at least one ring is substituted and each substituent may optionally be different. [00118] The symbol “ ” (a wavy line) denotes the point of attachment of a chemical moiety to the remainder of a molecule or chemical formula. [00119] The term “oxo,” as used herein, means an oxygen that is double bonded to a carbon atom. [00120] The term “alkylsulfonyl,” as used herein, means a moiety having the formula -S(O2)-R', where R' is a substituted or unsubstituted alkyl group as defined above. R' may have a specified number of carbons (e.g., “C1-C4 alkylsulfonyl”). [00121] The term “alkylarylene” as an arylene moiety covalently bonded to an alkylene moiety (also referred to herein as an alkylene linker). In embodiments, the alkylarylene group has the formula:
Figure imgf000047_0001
. [00122] An alkylarylene moiety may be substituted (e.g. with a substituent group) on the alkylene moiety or the arylene linker (e.g. at carbons 2, 3, 4, or 6) with halogen, oxo, -N3, - CF3, -CCl3, -CBr3, -CI3, -CN, -CHO, -OH, -NH2, -COOH, -CONH2, -NO2, -SH, -SO2CH3 - SO3H, , -OSO3H, -SO2NH2, −NHNH2, −ONH2, −NHC(O)NHNH2, substituted or unsubstituted C1-C5 alkyl or substituted or unsubstituted 2 to 5 membered heteroalkyl). In embodiments, the alkylarylene is unsubstituted. [00123] Each of the above terms (e.g., “alkyl,” “heteroalkyl,” “cycloalkyl,” “heterocycloalkyl,” “aryl,” and “heteroaryl”) includes both substituted and unsubstituted forms of the indicated radical. Preferred substituents for each type of radical are provided below. [00124] Substituents for the alkyl and heteroalkyl radicals (including those groups often referred to as alkylene, alkenyl, heteroalkylene, heteroalkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkenyl, and heterocycloalkenyl) can be one or more of a variety of groups selected from, but not limited to, -OR', =O, =NR', =N-OR', -NR'R'', -SR', -halogen, - SiR'R''R''', -OC(O)R', -C(O)R', -CO2R', -CONR'R'', -OC(O)NR'R'', -NR''C(O)R', -NR'- C(O)NR''R''', -NR''C(O)2R', -NR-C(NR'R''R''')=NR'''', -NR-C(NR'R'')=NR''', -S(O)R', - S(O)2R', -S(O)2NR'R'', -NRSO2R', −NR'NR''R''', −ONR'R'', −NR'C(O)NR''NR'''R'''', -CN, - NO2, -NR'SO2R'', -NR'C(O)R'', -NR'C(O)-OR'', -NR'OR'', in a number ranging from zero to (2m'+1), where m' is the total number of carbon atoms in such radical. R, R', R'', R''', and R'''' each preferably independently refer to hydrogen, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl (e.g., aryl substituted with 1-3 halogens), substituted or unsubstituted heteroaryl, substituted or unsubstituted alkyl, alkoxy, or thioalkoxy groups, or arylalkyl groups. When a compound described herein includes more than one R group, for example, each of the R groups is independently selected as are each R', R'', R''', and R'''' group when more than one of these groups is present. When R' and R'' are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a 4-, 5-, 6-, or 7- membered ring. For example, -NR'R'' includes, but is not limited to, 1-pyrrolidinyl and 4- morpholinyl. From the above discussion of substituents, one of skill in the art will understand that the term “alkyl” is meant to include groups including carbon atoms bound to groups other than hydrogen groups, such as haloalkyl (e.g., -CF3 and -CH2CF3) and acyl (e.g., - C(O)CH3, -C(O)CF3, -C(O)CH2OCH3, and the like). [00125] Similar to the substituents described for the alkyl radical, substituents for the aryl and heteroaryl groups are varied and are selected from, for example: -OR', -NR'R'', -SR', - halogen, -SiR'R''R''', -OC(O)R', -C(O)R', -CO2R', -CONR'R'', -OC(O)NR'R'', -NR''C(O)R', - NR'-C(O)NR''R''', -NR''C(O)2R', -NR-C(NR'R''R''')=NR'''', -NR-C(NR'R'')=NR''', -S(O)R', - S(O)2R', -S(O)2NR'R'', -NRSO2R', −NR'NR''R''', −ONR'R'', −NR'C(O)NR''NR'''R'''', -CN, - NO2, -R', -N3, -CH(Ph)2, fluoro(C1-C4)alkoxy, and fluoro(C1-C4)alkyl, -NR'SO2R'', - NR'C(O)R'', -NR'C(O)-OR'', -NR'OR'', in a number ranging from zero to the total number of open valences on the aromatic ring system; and where R', R'', R''', and R'''' are preferably independently selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl. When a compound described herein includes more than one R group, for example, each of the R groups is independently selected as are each R', R'', R''', and R'''' groups when more than one of these groups is present. [00126] Substituents for rings (e.g. cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkylene, heterocycloalkylene, arylene, or heteroarylene) may be depicted as substituents on the ring rather than on a specific atom of a ring (commonly referred to as a floating substituent). In such a case, the substituent may be attached to any of the ring atoms (obeying the rules of chemical valency) and in the case of fused rings or spirocyclic rings, a substituent depicted as associated with one member of the fused rings or spirocyclic rings (a floating substituent on a single ring), may be a substituent on any of the fused rings or spirocyclic rings (a floating substituent on multiple rings). When a substituent is attached to a ring, but not a specific atom (a floating substituent), and a subscript for the substituent is an integer greater than one, the multiple substituents may be on the same atom, same ring, different atoms, different fused rings, different spirocyclic rings, and each substituent may optionally be different. Where a point of attachment of a ring to the remainder of a molecule is not limited to a single atom (a floating substituent), the attachment point may be any atom of the ring and in the case of a fused ring or spirocyclic ring, any atom of any of the fused rings or spirocyclic rings while obeying the rules of chemical valency. Where a ring, fused rings, or spirocyclic rings contain one or more ring heteroatoms and the ring, fused rings, or spirocyclic rings are shown with one more floating substituents (including, but not limited to, points of attachment to the remainder of the molecule), the floating substituents may be bonded to the heteroatoms. Where the ring heteroatoms are shown bound to one or more hydrogens (e.g. a ring nitrogen with two bonds to ring atoms and a third bond to a hydrogen) in the structure or formula with the floating substituent, when the heteroatom is bonded to the floating substituent, the substituent will be understood to replace the hydrogen, while obeying the rules of chemical valency. [00127] Two or more substituents may optionally be joined to form aryl, heteroaryl, cycloalkyl, or heterocycloalkyl groups. Such so-called ring-forming substituents are typically, though not necessarily, found attached to a cyclic base structure. In one embodiment, the ring-forming substituents are attached to adjacent members of the base structure. For example, two ring-forming substituents attached to adjacent members of a cyclic base structure create a fused ring structure. In another embodiment, the ring-forming substituents are attached to a single member of the base structure. For example, two ring-forming substituents attached to a single member of a cyclic base structure create a spirocyclic structure. In yet another embodiment, the ring-forming substituents are attached to non- adjacent members of the base structure. [00128] Two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally form a ring of the formula -T-C(O)-(CRR')p-U-, wherein T and U are independently -NR-, -O-, -CRR'-, or a single bond, and p is an integer of from 0 to 3. Alternatively, two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula -A-(CH2)r-B-, wherein A and B are independently -CRR'-, -O-, -NR-, -S-, -S(O) -, -S(O)2-, -S(O)2NR'-, or a single bond, and r is an integer of from 1 to 4. One of the single bonds of the new ring so formed may optionally be replaced with a double bond. Alternatively, two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula - (CRR')s-X'- (C''R''R''')d-, where s and d are independently integers of from 0 to 3, and X' is - O-, -NR'-, -S-, -S(O)-, -S(O)2-, or -S(O)2NR'-. The substituents R, R', R'', and R''' are preferably independently selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl. [00129] As used herein, the terms “heteroatom” or “ring heteroatom” are meant to include oxygen (O), nitrogen (N), sulfur (S), phosphorus (P), and silicon (Si). [00130] A “substituent group,” as used herein, means a group selected from the following moieties: (A) oxo, halogen, -CCl3, -CBr3, -CF3, -CI3, -CH2Cl, -CH2Br, -CH2F, -CH2I, -CHCl2, -CHBr2, -CHF2, -CHI2, -CN, -OH, -NH2, -COOH, -CONH2, -NO2, -SH, -SO3H, -SO4H, -SO2NH2, −NHNH2, −ONH2, −NHC(O)NHNH2, -NHC(O)NH2, -NHSO2H, -NHC(O)H, -NHC(O)OH, -NHOH, -OCCl3, -OCF3, -OCBr3, -OCI3,-OCHCl2, -OCHBr2, -OCHI2, -OCHF2, -N3, unsubstituted alkyl (e.g., C1-C8 alkyl, C1-C6 alkyl, or C1-C4 alkyl), unsubstituted heteroalkyl (e.g., 2 to 8 membered heteroalkyl, 2 to 6 membered heteroalkyl, or 2 to 4 membered heteroalkyl), unsubstituted cycloalkyl (e.g., C3-C8 cycloalkyl, C3-C6 cycloalkyl, or C5-C6 cycloalkyl), unsubstituted heterocycloalkyl (e.g., 3 to 8 membered heterocycloalkyl, 3 to 6 membered heterocycloalkyl, or 5 to 6 membered heterocycloalkyl), unsubstituted aryl (e.g., C6-C10 aryl, C10 aryl, or phenyl), or unsubstituted heteroaryl (e.g., 5 to 10 membered heteroaryl, 5 to 9 membered heteroaryl, or 5 to 6 membered heteroaryl), and (B) alkyl (e.g., C1-C8 alkyl, C1-C6 alkyl, or C1-C4 alkyl), heteroalkyl (e.g., 2 to 8 membered heteroalkyl, 2 to 6 membered heteroalkyl, or 2 to 4 membered heteroalkyl), cycloalkyl (e.g., C3-C8 cycloalkyl, C3-C6 cycloalkyl, or C5-C6 cycloalkyl), heterocycloalkyl (e.g., 3 to 8 membered heterocycloalkyl, 3 to 6 membered heterocycloalkyl, or 5 to 6 membered heterocycloalkyl), aryl (e.g., C6-C10 aryl, C10 aryl, or phenyl), heteroaryl (e.g., 5 to 10 membered heteroaryl, 5 to 9 membered heteroaryl, or 5 to 6 membered heteroaryl), substituted with at least one substituent selected from: (i) oxo, halogen, -CCl3, -CBr3, -CF3, -CI3, -CH2Cl, -CH2Br, -CH2F, -CH2I, -CHCl2, -CHBr2, -CHF2, -CHI2, -CN, -OH, -NH2, -COOH, -CONH2, -NO2, -SH, -SO3H, -SO4H, -SO2NH2, −NHNH2, −ONH2, −NHC(O)NHNH2, -NHC(O)NH2, -NHSO2H, -NHC(O)H, -NHC(O)OH, -NHOH, -OCCl3, -OCF3, -OCBr3, -OCI3,-OCHCl2, -OCHBr2, -OCHI2, -OCHF2, -N3, unsubstituted alkyl (e.g., C1-C8 alkyl, C1-C6 alkyl, or C1-C4 alkyl), unsubstituted heteroalkyl (e.g., 2 to 8 membered heteroalkyl, 2 to 6 membered heteroalkyl, or 2 to 4 membered heteroalkyl), unsubstituted cycloalkyl (e.g., C3-C8 cycloalkyl, C3-C6 cycloalkyl, or C5-C6 cycloalkyl), unsubstituted heterocycloalkyl (e.g., 3 to 8 membered heterocycloalkyl, 3 to 6 membered heterocycloalkyl, or 5 to 6 membered heterocycloalkyl), unsubstituted aryl (e.g., C6-C10 aryl, C10 aryl, or phenyl), or unsubstituted heteroaryl (e.g., 5 to 10 membered heteroaryl, 5 to 9 membered heteroaryl, or 5 to 6 membered heteroaryl), and (ii) alkyl (e.g., C1-C8 alkyl, C1-C6 alkyl, or C1-C4 alkyl), heteroalkyl (e.g., 2 to 8 membered heteroalkyl, 2 to 6 membered heteroalkyl, or 2 to 4 membered heteroalkyl), cycloalkyl (e.g., C3-C8 cycloalkyl, C3-C6 cycloalkyl, or C5-C6 cycloalkyl), heterocycloalkyl (e.g., 3 to 8 membered heterocycloalkyl, 3 to 6 membered heterocycloalkyl, or 5 to 6 membered heterocycloalkyl), aryl (e.g., C6-C10 aryl, C10 aryl, or phenyl), heteroaryl (e.g., 5 to 10 membered heteroaryl, 5 to 9 membered heteroaryl, or 5 to 6 membered heteroaryl), substituted with at least one substituent selected from: (a) oxo, halogen, -CCl3, -CBr3, -CF3, -CI3, -CH2Cl, -CH2Br, -CH2F, -CH2I, -CHCl2, -CHBr2, -CHF2, -CHI2, -CN, -OH, -NH2, -COOH, -CONH2, -NO2, -SH, -SO3H, -SO4H, -SO2NH2, −NHNH2, −ONH2, −NHC(O)NHNH2, -NHC(O)NH2, -NHSO2H, -NHC(O)H, -NHC(O)OH, -NHOH, -OCCl3, -OCF3, -OCBr3, -OCI3, -OCHCl2, -OCHBr2, -OCHI2, -OCHF2, -N3, unsubstituted alkyl (e.g., C1-C8 alkyl, C1-C6 alkyl, or C1-C4 alkyl), unsubstituted heteroalkyl (e.g., 2 to 8 membered heteroalkyl, 2 to 6 membered heteroalkyl, or 2 to 4 membered heteroalkyl), unsubstituted cycloalkyl (e.g., C3-C8 cycloalkyl, C3-C6 cycloalkyl, or C5-C6 cycloalkyl), unsubstituted heterocycloalkyl (e.g., 3 to 8 membered heterocycloalkyl, 3 to 6 membered heterocycloalkyl, or 5 to 6 membered heterocycloalkyl), unsubstituted aryl (e.g., C6-C10 aryl, C10 aryl, or phenyl), or unsubstituted heteroaryl (e.g., 5 to 10 membered heteroaryl, 5 to 9 membered heteroaryl, or 5 to 6 membered heteroaryl), and (b) alkyl (e.g., C1-C8 alkyl, C1-C6 alkyl, or C1-C4 alkyl), heteroalkyl (e.g., 2 to 8 membered heteroalkyl, 2 to 6 membered heteroalkyl, or 2 to 4 membered heteroalkyl), cycloalkyl (e.g., C3-C8 cycloalkyl, C3-C6 cycloalkyl, or C5-C6 cycloalkyl), heterocycloalkyl (e.g., 3 to 8 membered heterocycloalkyl, 3 to 6 membered heterocycloalkyl, or 5 to 6 membered heterocycloalkyl), aryl (e.g., C6-C10 aryl, C10 aryl, or phenyl), heteroaryl (e.g., 5 to 10 membered heteroaryl, 5 to 9 membered heteroaryl, or 5 to 6 membered heteroaryl), substituted with at least one substituent selected from: oxo, halogen, -CCl3, -CBr3, -CF3, -CI3, -CH2Cl, -CH2Br, -CH2F, -CH2I, -CHCl2, -CHBr2, -CHF2, -CHI2, -CN, -OH, -NH2, -COOH, -CONH2, -NO2, -S H, -SO3H, -SO4H, -SO2NH2, −NHNH2, −ONH2, −NHC(O)NHNH2, -NHC(O)NH2, -NHSO2H, -NHC(O)H, -NHC(O)OH, -NHOH, -OCCl3, -OCF3, -OCBr3, -OCI3,-OCHCl2, -OCHBr2, -OCHI2, -OCHF2, -N3, unsubstituted alkyl (e.g., C1-C8 alkyl, C1-C6 alkyl, or C1-C4 alkyl), unsubstituted heteroalkyl (e.g., 2 to 8 membered heteroalkyl, 2 to 6 membered heteroalkyl, or 2 to 4 membered heteroalkyl), unsubstituted cycloalkyl (e.g., C3-C8 cycloalkyl, C3-C6 cycloalkyl, or C5-C6 cycloalkyl), unsubstituted heterocycloalkyl (e.g., 3 to 8 membered heterocycloalkyl, 3 to 6 membered heterocycloalkyl, or 5 to 6 membered heterocycloalkyl), unsubstituted aryl (e.g., C6-C10 aryl, C10 aryl, or phenyl), or unsubstituted heteroaryl (e.g., 5 to 10 membered heteroaryl, 5 to 9 membered heteroaryl, or 5 to 6 membered heteroaryl). [00131] A “size-limited substituent” or “ size-limited substituent group,” as used herein, means a group selected from all of the substituents described above for a “substituent group,” wherein each substituted or unsubstituted alkyl is a substituted or unsubstituted C1-C20 alkyl, each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 20 membered heteroalkyl, each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C3-C8 cycloalkyl, each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 3 to 8 membered heterocycloalkyl, each substituted or unsubstituted aryl is a substituted or unsubstituted C6-C10 aryl, and each substituted or unsubstituted heteroaryl is a substituted or unsubstituted 5 to 10 membered heteroaryl. [00132] A “lower substituent” or “ lower substituent group,” as used herein, means a group selected from all of the substituents described above for a “substituent group,” wherein each substituted or unsubstituted alkyl is a substituted or unsubstituted C1-C8 alkyl, each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 8 membered heteroalkyl, each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C3- C7 cycloalkyl, each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 3 to 7 membered heterocycloalkyl, each substituted or unsubstituted aryl is a substituted or unsubstituted phenyl, and each substituted or unsubstituted heteroaryl is a substituted or unsubstituted 5 to 6 membered heteroaryl. [00133] In some embodiments, each substituted group described in the compounds herein is substituted with at least one substituent group. More specifically, in some embodiments, each substituted alkyl, substituted heteroalkyl, substituted cycloalkyl, substituted heterocycloalkyl, substituted aryl, substituted heteroaryl, substituted alkylene, substituted heteroalkylene, substituted cycloalkylene, substituted heterocycloalkylene, substituted arylene, and/or substituted heteroarylene described in the compounds herein are substituted with at least one substituent group. In other embodiments, at least one or all of these groups are substituted with at least one size-limited substituent group. In other embodiments, at least one or all of these groups are substituted with at least one lower substituent group. [00134] In other embodiments of the compounds herein, each substituted or unsubstituted alkyl may be a substituted or unsubstituted C1-C20 alkyl, each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 20 membered heteroalkyl, each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C3-C8 cycloalkyl, each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 3 to 8 membered heterocycloalkyl, each substituted or unsubstituted aryl is a substituted or unsubstituted C6- C10 aryl, and/or each substituted or unsubstituted heteroaryl is a substituted or unsubstituted 5 to 10 membered heteroaryl. In some embodiments of the compounds herein, each substituted or unsubstituted alkylene is a substituted or unsubstituted C1-C20 alkylene, each substituted or unsubstituted heteroalkylene is a substituted or unsubstituted 2 to 20 membered heteroalkylene, each substituted or unsubstituted cycloalkylene is a substituted or unsubstituted C3-C8 cycloalkylene, each substituted or unsubstituted heterocycloalkylene is a substituted or unsubstituted 3 to 8 membered heterocycloalkylene, each substituted or unsubstituted arylene is a substituted or unsubstituted C6-C10 arylene, and/or each substituted or unsubstituted heteroarylene is a substituted or unsubstituted 5 to 10 membered heteroarylene. [00135] In some embodiments, each substituted or unsubstituted alkyl is a substituted or unsubstituted C1-C8 alkyl, each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 8 membered heteroalkyl, each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C3-C7 cycloalkyl, each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 3 to 7 membered heterocycloalkyl, each substituted or unsubstituted aryl is a substituted or unsubstituted C6-C10 aryl, and/or each substituted or unsubstituted heteroaryl is a substituted or unsubstituted 5 to 9 membered heteroaryl. In some embodiments, each substituted or unsubstituted alkylene is a substituted or unsubstituted C1-C8 alkylene, each substituted or unsubstituted heteroalkylene is a substituted or unsubstituted 2 to 8 membered heteroalkylene, each substituted or unsubstituted cycloalkylene is a substituted or unsubstituted C3-C7 cycloalkylene, each substituted or unsubstituted heterocycloalkylene is a substituted or unsubstituted 3 to 7 membered heterocycloalkylene, each substituted or unsubstituted arylene is a substituted or unsubstituted C6-C10 arylene, and/or each substituted or unsubstituted heteroarylene is a substituted or unsubstituted 5 to 9 membered heteroarylene. In some embodiments, the compound is a chemical species set forth in the Examples section, figures, or tables below. [00136] In embodiments, a substituted or unsubstituted moiety (e.g., substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, substituted or unsubstituted cycloalkylene, substituted or unsubstituted heterocycloalkylene, substituted or unsubstituted arylene, and/or substituted or unsubstituted heteroarylene) is unsubstituted (e.g., is an unsubstituted alkyl, unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl, unsubstituted heteroaryl, unsubstituted alkylene, unsubstituted heteroalkylene, unsubstituted cycloalkylene, unsubstituted heterocycloalkylene, unsubstituted arylene, and/or unsubstituted heteroarylene, respectively). In embodiments, a substituted or unsubstituted moiety (e.g., substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, substituted or unsubstituted cycloalkylene, substituted or unsubstituted heterocycloalkylene, substituted or unsubstituted arylene, and/or substituted or unsubstituted heteroarylene) is substituted (e.g., is a substituted alkyl, substituted heteroalkyl, substituted cycloalkyl, substituted heterocycloalkyl, substituted aryl, substituted heteroaryl, substituted alkylene, substituted heteroalkylene, substituted cycloalkylene, substituted heterocycloalkylene, substituted arylene, and/or substituted heteroarylene, respectively). [00137] In embodiments, a substituted moiety (e.g., substituted alkyl, substituted heteroalkyl, substituted cycloalkyl, substituted heterocycloalkyl, substituted aryl, substituted heteroaryl, substituted alkylene, substituted heteroalkylene, substituted cycloalkylene, substituted heterocycloalkylene, substituted arylene, and/or substituted heteroarylene) is substituted with at least one substituent group, wherein if the substituted moiety is substituted with a plurality of substituent groups, each substituent group may optionally be different. In embodiments, if the substituted moiety is substituted with a plurality of substituent groups, each substituent group is different. [00138] In embodiments, a substituted moiety (e.g., substituted alkyl, substituted heteroalkyl, substituted cycloalkyl, substituted heterocycloalkyl, substituted aryl, substituted heteroaryl, substituted alkylene, substituted heteroalkylene, substituted cycloalkylene, substituted heterocycloalkylene, substituted arylene, and/or substituted heteroarylene) is substituted with at least one size-limited substituent group, wherein if the substituted moiety is substituted with a plurality of size-limited substituent groups, each size-limited substituent group may optionally be different. In embodiments, if the substituted moiety is substituted with a plurality of size-limited substituent groups, each size-limited substituent group is different. [00139] In embodiments, a substituted moiety (e.g., substituted alkyl, substituted heteroalkyl, substituted cycloalkyl, substituted heterocycloalkyl, substituted aryl, substituted heteroaryl, substituted alkylene, substituted heteroalkylene, substituted cycloalkylene, substituted heterocycloalkylene, substituted arylene, and/or substituted heteroarylene) is substituted with at least one lower substituent group, wherein if the substituted moiety is substituted with a plurality of lower substituent groups, each lower substituent group may optionally be different. In embodiments, if the substituted moiety is substituted with a plurality of lower substituent groups, each lower substituent group is different. [00140] In embodiments, a substituted moiety (e.g., substituted alkyl, substituted heteroalkyl, substituted cycloalkyl, substituted heterocycloalkyl, substituted aryl, substituted heteroaryl, substituted alkylene, substituted heteroalkylene, substituted cycloalkylene, substituted heterocycloalkylene, substituted arylene, and/or substituted heteroarylene) is substituted with at least one substituent group, size-limited substituent group, or lower substituent group; wherein if the substituted moiety is substituted with a plurality of groups selected from substituent groups, size-limited substituent groups, and lower substituent groups; each substituent group, size-limited substituent group, and/or lower substituent group may optionally be different. In embodiments, if the substituted moiety is substituted with a plurality of groups selected from substituent groups, size-limited substituent groups, and lower substituent groups; each substituent group, size-limited substituent group, and/or lower substituent group is different. [00141] The phrase “or combinations thereof” means that any two or more of the items in the preceding list may be combined, and optionally repeated, in any order and in any orientation. [00142] As used herein, common organic chemistry abbreviations are defined as follows:
Figure imgf000056_0001
q q Examorpholine morpholine analog of exatecan, the structure of which is shown in Example S6, and which is also referred to as compound 50 FA Folic acid
Figure imgf000057_0001
Compositions Small Molecule-Drug Conjugates [00143] In one aspect, provided herein is a compound of Formula (I) or (II):
Figure imgf000058_0001
or a pharmaceutically acceptable salt thereof, wherein: L is a multivalent linker bound at least to a folic acid and a drug moiety; HL is a half-life extender; and D is a drug moiety. [00144] In one aspect, provided herein is a compound of Formula (I) or (II):
Figure imgf000059_0001
or a pharmaceutically acceptable salt thereof, wherein: L is a multivalent linker bound to a folic acid, a drug moiety, and HL; wherein: HL is a half-life extender; and D is a drug moiety. [00145] In embodiments, provided herein is a compound of Formula (I):
Figure imgf000059_0002
or a pharmaceutically acceptable salt thereof, wherein: L is a multivalent linker bound at least to a folic acid and a drug moiety; and D is a drug moiety. [00146] In embodiments, provided herein is a compound of Formula (I):
Figure imgf000060_0001
or a pharmaceutically acceptable salt thereof, wherein: L is a multivalent linker bound to a folic acid, a drug moiety, and HL; wherein: HL is a half-life extender; and D is a drug moiety. [00147] In embodiments, provided herein is a compound of Formula (II):
Figure imgf000060_0002
or a pharmaceutically acceptable salt thereof, wherein: L is a multivalent linker bound at least to a folic acid and a drug moiety; HL is a half-life extender; and D is a drug moiety. [00148] In embodiments, provided herein is a compound of Formula (II):
Figure imgf000061_0001
or a pharmaceutically acceptable salt thereof, wherein: L is a multivalent linker bound to a folic acid, a drug moiety, and HL wherein HL is a half-life extender; and D is a drug moiety. [00149] In general, Formulae (I) and (II) encompass tautomers, a mixture of two or more tautomers, isotopic variants, and/or a pharmaceutically acceptable salt, solvate, or hydrate thereof. [00150] In embodiments, D is a tubulin inhibitor or disruptor, apoptosis inducer, RNA splicing inhibitor, kinase inhibitor, DNA damaging agent, nicotinamide phosphoribosyltransferase inhibitor (NAMPT), peptidic proteasome inhibitors, transcription inhibitors, or proteolysis-targeting chimera (PROTAC). [00151] In embodiments, D is a tubulin inhibitor or disruptor, kinase inhibitor, DNA damaging agent, transcription inhibitors, or proteolysis-targeting chimera (PROTAC). [00152] In embodiments, D is a tubulin inhibitor. In embodiments, D is a tubulin disruptor. In embodiments, D is a kinase inhibitor. In embodiments, D is a DNA damaging agent. In embodiments, D is a transcription inhibitor. In embodiments, D is a proteolysis-targeting chimera (PROTAC). [00153] In embodiments, D is a pyrrolobenzodiazepine, indolinobenzodiazepine, duocarmycin, tubulysin, cryptomycin, anthracycline, maytansinoid, auristatin, carmaphycin, calicheamicin, camptothecin, thailanstatin and analogues, RNA polymerase II inhibitor, topoisomerase I inhibitor, tyrosine kinase inhibitor, Bcl-xL Inhibitor, EG5 inhibitor, or MEK inhibitor. [00154] In embodiments, D is a pyrrolobenzodiazepine, duocarmycin, anthracycline, maytansinoid, auristatin, calicheamicin, camptothecin, RNA polymerase II inhibitor, topoisomerase I inhibitor, tyrosine kinase inhibitor, EG5 inhibitor, or MEK inhibitor. [00155] In embodiments, D is a pyrrolobenzodiazepine. In embodiments, D is a duocarmycin. In embodiments, D is an anthracycline. In embodiments, D is a maytansinoid. In embodiments, D is an auristatin. In embodiments, D is a calicheamicin. In embodiments, D is a camptothecin. In embodiments, D is a topoisomerase I inhibitor. In embodiments, D is an RNA polymerase II inhibitor. In embodiments, D is a tyrosine kinase inhibitor. In embodiments, D is an EG5 inhibitor. In embodiments, D is a MEK inhibitor. [00156] In embodiments, D is monomethyl auristatin E (MMAE), monomethyl auristatin F (MMAF), duostatin 5 (Duo5), PNU-159682, 7-ethyl-10-hydroxycamptothecin (SN-38), irinotecan, amatoxin, maytansine, exatecan, trametinib, abemaciclib, palbociclib, =an exatecan derivative or analog, a morpholine analog of exatecan, or examorpholine (morpholine analog of exatecan, the structure of which is shown in Example S6, and which is also referred to as compound 50). In embodiments, D is MMAE. In embodiments, D is MMAF. In embodiments, D is Duo5. In embodiments, D is PNU-159682. In embodiments, D is SN-38. In embodiments, D is irinotecan. In embodiments, D is amatoxin. In embodiments, D is maytansine. In embodiments, D is exatecan. In embodiments, D is trametinib. In embodiments, D is abemaciclib. In embodiments, D is Palbociclib. In some embodiments, D is a morpholine analog of exatecan. In embodiments, D is examorpholine (the morpholine analog of exatecan for which the structure is shown in Example S6, and which is also referred to as compound 50). [00157] In embodiments,
Figure imgf000062_0001
, wherein the wavy line indicates a bond to the multivalent linker (L). In embodiments, D is
Figure imgf000062_0002
wherein the wavy line indicates a bond to the
Figure imgf000063_0001
wavy line indicates a bond to the multivalent linker (L). [00158] In embodiments, L is a cleavable or a non-cleavable linker as described in US Patents Nos. US 9,884,127, US 9,981,046, US 9,801,951, US 10,117,944, US 10,590,165, and US 10,590,165, and US Patent publications Nos. US 2017/0340750, and US 2018/0360985, all of which are incorporated herein in their entireties. [00159] In embodiments, L is a bond, -C(O)-, -NH-, Amino Acid Unit, Peptoid, –(CH2CH2O)n–, –(CH2)n–, –(4-aminobenzyloxycarbonyl)–, –(C(O)CH2CH2C(O))–,
Figure imgf000063_0002
thereof; wherein n is an integer from 1 to 24; each R2 and R3 is independently H or substituted or unsubstituted alkyl. [00160] In embodiments, n is an integer from 1 to 24. In embodiments, n is 1. In embodiments, n is 2. In embodiments, n is 3. In embodiments, n is 4. In embodiments, n is 5. In embodiments, n is 6. In embodiments, n is 7. In embodiments, n is 8. In embodiments, n is 9. In embodiments, n is 10. In embodiments, n is 11. In embodiments, n is 12. In embodiments, n is 13. In embodiments, n is 14. In embodiments, n is 15. In embodiments, n is 16. In embodiments, n is 17. In embodiments, n is 18. In embodiments, n is 19. In embodiments, n is 20. In embodiments, n is 21. In embodiments, n is 22. In embodiments, n is 23. In embodiments, n is 24. [00161] In embodiments, each R2 and R3 is independently H or substituted or unsubstituted alkyl (e.g., C1-C8 alkyl, C1-C6 alkyl, or C1-C4 alkyl). In embodiments, each R2 and R3 is independently H. In embodiments, each R2 and R3 is independently substituted or unsubstituted alkyl. In embodiments, each R2 and R3 is independently substituted or unsubstituted alkyl (e.g., C1-C8 alkyl, C1-C6 alkyl, or C1-C4 alkyl). In embodiments, each R2 and R3 is independently unsubstituted alkyl (e.g., C1-C8 alkyl, C1-C6 alkyl, or C1-C4 alkyl). In embodiments, each R2 and R3 is independently substituted alkyl (e.g., C1-C8 alkyl, C1-C6 alkyl, or C1-C4 alkyl). [00162] In embodiments, each R2 and R3 is independently H or substituted (e.g., substituted with at least one substituent group, size-limited substituent group, or lower substituent group) or unsubstituted alkyl (e.g., C1-C8 alkyl, C1-C6 alkyl, or C1-C4 alkyl). In embodiments, each R2 and R3 is independently substituted (e.g., substituted with at least one substituent group, size-limited substituent group, or lower substituent group) or unsubstituted alkyl. In embodiments, each R2 and R3 is independently substituted (e.g., substituted with at least one substituent group, size-limited substituent group, or lower substituent group) or unsubstituted alkyl (e.g., C1-C8 alkyl, C1-C6 alkyl, or C1-C4 alkyl). In embodiments, each R2 and R3 is independently unsubstituted alkyl (e.g., C1-C8 alkyl, C1-C6 alkyl, or C1-C4 alkyl). In embodiments, each R2 and R3 is independently substituted (e.g., substituted with at least one substituent group, size-limited substituent group, or lower substituent group) alkyl (e.g., C1- C8 alkyl, C1-C6 alkyl, or C1-C4 alkyl). [00163] In embodiments, each R2 and R3 is independently methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, isopentyl, or hexyl. In embodiments, each R2 and R3 is independently methyl. In embodiments, each R2 and R3 is independently ethyl. In embodiments, each R2 and R3 is independently propyl. In embodiments, each R2 and R3 is independently butyl. [00164] In embodiments, L is -C(O)-, -NH-,–(CH2CH2O)n–, –(CH2)n–, -Cys-, -Asp-, -Arg- , -Val-, –(4-aminobenzyloxycarbonyl)–, -Glu-, -citrulline- (-Cit-), -Lys-, – (C(O)CH2CH2NH)–, –(C(O)CH2CH2C(O))–, –(C(O)(CH2)nNH)–, –S(CH2)nO–, –(N(R2)(CH2)nN(R3))–,
Figure imgf000065_0001
thereof. [00166] In embodiments, L is -C(O)-, -NH-, –(CH2CH2O)n–, –(CH2)n–, –SCH2CH2O–, –(C(O)CH2CH2C(O))–, -Cys-, -Val-, -Cit-, -Arg-, -Asp-, -Lys-, –(4- aminobenzyloxycarbonyl)
Figure imgf000066_0001
, , , ,
Figure imgf000066_0002
r combinations thereof. [00167] In embodiments, L is -C(O)-. In embodiments, L is -NH-. In embodiments, L is –(CH2CH2O)n–. In embodiments, L is –(CH2)n–. In embodiments, L is –SCH2CH2O–. In embodiments, L is –(C(O)CH2CH2C(O))–. In embodiments, L is -Cys-. In embodiments, L is -Val-. In embodiments, L is -Cit-. In embodiments, L is -Arg-. In embodiments, L is -Asp-. In embodiments, L is -Lys-. In embodiments, L is –(4-aminobenzyloxycarbonyl)–. In embodiments, L is
Figure imgf000066_0005
. In embodiments,
Figure imgf000066_0003
. embodiments, L is
Figure imgf000066_0004
. , . , .
Figure imgf000067_0001
Figure imgf000068_0001
, wherein the carbonyl is linked to the drug moiety (D), the amine is linked to the folic acid, and the optional third linkage is to the half-life extender (HL).
Figure imgf000068_0002
.
In embodiments, L is
Figure imgf000069_0001
. embodiments
Figure imgf000069_0002
. embodiments, L is
Figure imgf000069_0003
. [00170] In embodiments, HL is cholesterol-like half-life extender or albumin binder half- life extender. In embodiments, HL is cholesterol-like half-life extender. In embodiments, HL is an albumin binder half-life extender.
Figure imgf000070_0001
[00172] In embodiments, HL is
Figure imgf000070_0002
. In embodiments, HL is
Figure imgf000070_0003
Figure imgf000071_0001
T C , P 0 0-3 2 1 0 , -3 2 2 1 0.oN t e k co D y e n r ott A
Figure imgf000072_0001
T C P 0 0-3 2 1 0-3 2 2 1 0.oN t e k co D y e n r ott A
Figure imgf000073_0001
17
Figure imgf000074_0001
Figure imgf000075_0001
T C P 0 0-3 2 1 0-3 2 2 1 0.oN t e k co D y e n r ott A si dnu op m oc e h t ni er eh w,)I( al u mro Ffo sdnu op m oc . e f r o a e r e n i t e h r tl e h as d e el d i b v a o tp r p ec , c s t a n e yl l m a i c d it o u b e c m a e m n I ra hp ] a 6 7 10
Figure imgf000076_0001
T C P 0 0-3 2 1 0-3 2 2 1 0.o N t e k co D y e n r ott A si dnu op m oc e h t ni er eh w,)I( 5 al 7 u mro Ffo sdnu op m oc . e f r o a e r e n i t e h r tl e h as d e el d i b v a o tp r p ec , c s t a n e yl l m a i c d it o u b e c m a e m n I ra hp ] a 9 7 ro 1 0 0
Figure imgf000077_0001
[ T C P 0 0-3 2 1 0-3 2 2 1 0.o N t e k co D y e n r ott A si dnu op m oc e h t ni er e h w,) II( 6 al 7 u mro Ffo dnu op m oc . a f s o i e r e n i e h t r tl e h as d e el d i b v a o tp r p ec , c s t a n e yl l m a i c d it o u b e c m a e m n I ra hp ] a 0 8 10
Figure imgf000078_0001
T C P 0 0-3 2 1 0-3 2 2 1 0.o N t e k co D y e n r ott A 77
Figure imgf000079_0001
Figure imgf000080_0001
Pharmaceutical Compositions, Uses, and Methods of Use [00182] In an aspect, provided herein is a pharmaceutical composition including a compound of Formula (I) or (II) (an SMDC) as described herein, including embodiments, and a pharmaceutically acceptable carrier. In embodiments, the SMDC as described herein is included in a therapeutically effective amount. [00183] In embodiments, provided herein is a pharmaceutical composition comprising a compound provided herein, including a compound of Formula (I) or (II), or an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and a pharmaceutically acceptable carrier (or excipient). [00184] In embodiments, the pharmaceutical composition may include optical isomers, diastereomers, enantiomers, isoforms, polymorphs, hydrates, solvates or products, or pharmaceutically acceptable salts of the compound described herein. [00185] The compound provided herein may be administered alone, or in combination with one or more other compounds. The pharmaceutical compositions that comprise a compound provided herein, e.g., a compound of Formula (I) or (II), can be formulated in various dosage forms for oral, parenteral, and topical administration. The pharmaceutical compositions can also be formulated as modified release dosage forms, including delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated-, fast-, targeted-, programmed-release, and gastric retention dosage forms. These dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art (see, Remington: The Science and Practice of Pharmacy, supra; Modified-Release Drug Delivery Technology, 2nd Edition, Rathbone et al., Eds., Marcel Dekker, Inc.: New York, NY, 2008). [00186] The pharmaceutical composition may be formulated for oral administration, suppository administration, topical administration, intravenous administration, intraperitoneal administration, intramuscular administration, intralesional administration, intrathecal administration, intranasal administration, subcutaneous administration, implantation, transdermal administration, or transmucosal administration as described herein. [00187] In embodiments, the pharmaceutical compositions provided herein are formulated in a dosage form for oral administration, which comprise a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more pharmaceutically acceptable excipients or carriers. [00188] In embodiments, the pharmaceutical compositions provided herein are formulated as a suspension for oral administration, which comprise a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more pharmaceutically acceptable excipients or carriers. In embodiments, the suspension provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more excipients or carriers selected from the group consisting of water, glycerin, sorbitol, sodium saccharin, xanthan gum, flavoring, citric acid, sodium citrate, methylparaben, propylparaben, and potassium sorbate. In another embodiment, the suspension provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and water, glycerin, sorbitol, sodium saccharin, xanthan gum, flavoring, citric acid, sodium citrate, methylparaben, propylparaben, and potassium sorbate. [00189] In another embodiment, the pharmaceutical compositions provided herein are formulated in a dosage form for parenteral administration, which comprise a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more pharmaceutically acceptable excipients or carriers. In embodiments, the pharmaceutical compositions provided herein are formulated in a dosage form for intravenous administration. In another embodiment, the pharmaceutical compositions provided herein are formulated in a dosage form for intramuscular administration. In yet another embodiment, the pharmaceutical compositions provided herein are formulated in a dosage form for subcutaneous administration. [00190] In yet another embodiment, the pharmaceutical compositions provided herein are formulated in a dosage form for topical administration, which comprise a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more pharmaceutically acceptable excipients or carriers. [00191] In embodiments, the pharmaceutical compositions provided herein are formulated as a cream for topical administration, which comprise a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more pharmaceutically acceptable excipients or carriers. In embodiments, the cream provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more excipients or carriers selected from the group consisting of water, octyldodecanol, mineral oil, stearyl alcohol, cocamide DEA, polysorbate 60, myristyl alcohol, sorbitan monostearate, lactic acid, and benzyl alcohol. In another embodiment, the cream provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and water, octyldodecanol, mineral oil, stearyl alcohol, cocamide DEA, polysorbate 60, myristyl alcohol, sorbitan monostearate, lactic acid, and benzyl alcohol. [00192] In embodiments, the pharmaceutical compositions provided herein are formulated as a gel for topical administration, which comprise a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more pharmaceutically acceptable excipients or carriers. In embodiments, the gel provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more excipients or carriers selected from the group consisting of water, isopropyl alcohol, octyldodecanol, dimethicone copolyol 190, carbomer 980, sodium hydroxide, and docusate sodium. In embodiments, the gel provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and water, isopropyl alcohol, octyldodecanol, dimethicone copolyol 190, carbomer 980, sodium hydroxide, and docusate sodium. [00193] In embodiments, the pharmaceutical compositions provided herein are formulated as a shampoo for topical administration, which comprise a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more pharmaceutically acceptable excipients or carriers. In embodiments, the shampoo provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more excipients or carriers selected from the group consisting of water, sodium laureth sulfate, disodium laureth sulfosuccinate, sodium chloride, and laureth-2. In embodiments, the shampoo provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and water, sodium laureth sulfate, disodium laureth sulfosuccinate, sodium chloride, and laureth-2. [00194] In embodiments, the pharmaceutical compositions provided herein are formulated as a lacquer for topical administration, which comprise a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more pharmaceutically acceptable excipients or carriers. In embodiments, the lacquer provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and one or more excipients or carriers selected from the group consisting of ethyl acetate, isopropyl alcohol, and butyl monoester of poly(methylvinyl ether/maleic acid) in isopropyl alcohol. In embodiments, the lacquer provided herein comprises a compound provided herein, e.g., a compound of Formula (I) or (II), including an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or isotopic variants thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof; and ethyl acetate, isopropyl alcohol, and butyl monoester of poly(methylvinyl ether/maleic acid) in isopropyl alcohol. [00195] The pharmaceutical compositions provided herein can be provided in a unit- dosage form or multiple-dosage form. A unit-dosage form, as used herein, refers to physically discrete a unit suitable for administration to a human and animal subject, and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of an active ingredient(s) sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carriers or excipients. Examples of a unit- dosage form include an ampoule, syringe, and individually packaged tablet and capsule. A unit-dosage form may be administered in fractions or multiples thereof. A multiple-dosage form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dosage form. Examples of a multiple-dosage form include a vial, bottle of tablets or capsules, or bottle of pints or gallons. [00196] The pharmaceutical compositions provided herein can be administered at once, or multiple times at intervals of time. It is understood that the precise dosage and duration of treatment may vary with the age, weight, and condition of the patient being treated, and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test or diagnostic data. It is further understood that for any particular individual, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the formulations. [00197] Pharmaceutical compositions include those suitable for oral, rectal, nasal, topical (including transdermal, buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration or administration via an implant. The compositions may be prepared by any method well known in the art of pharmacy. [00198] Such methods include the step of bringing in association compounds of this disclosure or combinations thereof with any auxiliary agent. The auxiliary agent(s), also named accessory ingredient(s), include those conventional in the art, such as carriers, fillers, binders, diluents, disintegrants, lubricants, colorants, flavouring agents, anti-oxidants, and wetting agents. [00199] Pharmaceutical compositions suitable for oral administration may be presented as discrete dosage units such as pills, tablets, cachets, dragées, lozenges, or capsules, or as a powder or granules, or as a solution or suspension. The active ingredient may also be presented as a bolus or paste. The compositions can further be processed into a suppository or enema for rectal administration. [00200] For parenteral administration, suitable compositions include aqueous and non- aqueous sterile injection. The compositions may be presented in unit-dose or multi-dose containers, for example sealed vials and ampoules, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of sterile liquid carrier, for example water, prior to use. For transdermal administration, e.g. gels, patches or sprays can be contemplated. Compositions or formulations suitable for pulmonary administration e.g. by nasal inhalation include fine dusts or mists which may be generated by means of metered dose pressurized aerosols, nebulisers or insufflators. [00201] The exact dose and regimen of administration of the composition will necessarily be dependent upon the therapeutic or nutritional effect to be achieved and may vary with the particular formula, the route of administration, and the age and condition of the individual subject to whom the composition is to be administered. [00202] The therapeutically effective amount for each active compound can vary with factors including but not limited to the activity of the compound used, stability of the active compound in the patient’s body, the severity of the conditions to be alleviated, the total weight of the patient treated, the route of administration, the ease of absorption, distribution, and excretion of the active compound by the body, the age and sensitivity of the patient to be treated, and the like, as will be apparent to a skilled artisan. The amount of administration can be adjusted as the various factors change over time. [00203] For oral delivery, the active compounds can be incorporated into a formulation that includes pharmaceutically acceptable carriers such as binders (e.g., gelatin, cellulose, gum tragacanth), excipients (e.g., starch, lactose), lubricants (e.g., magnesium stearate, silicon dioxide), disintegrating agents (e.g., alginate, Primogel, and corn starch), and sweetening or flavoring agents (e.g., glucose, sucrose, saccharin, methyl salicylate, and peppermint). The formulation can be orally delivered in the form of enclosed gelatin capsules or compressed tablets. Capsules and tablets can be prepared in any conventional techniques. The capsules and tablets can also be coated with various coatings known in the art to modify the flavors, tastes, colors, and shapes of the capsules and tablets. In addition, liquid carriers such as fatty oil can also be included in capsules. [00204] Suitable oral formulations can also be in the form of suspension, syrup, chewing gum, wafer, elixir, and the like. If desired, conventional agents for modifying flavors, tastes, colors, and shapes of the special forms can also be included. In addition, for convenient administration by enteral feeding tube in patients unable to swallow, the active compounds can be dissolved in an acceptable lipophilic vegetable oil vehicle such as olive oil, corn oil and safflower oil. [00205] The active compounds can also be administered parenterally in the form of solution or suspension, or in lyophilized form capable of conversion into a solution or suspension form before use. In such formulations, diluents or pharmaceutically acceptable carriers such as sterile water and physiological saline buffer can be used. Other conventional solvents, pH buffers, stabilizers, anti-bacteria agents, surfactants, and antioxidants can all be included. For example, useful components include sodium chloride, acetates, citrates or phosphates buffers, glycerin, dextrose, fixed oils, methyl parabens, polyethylene glycol, propylene glycol, sodium bisulfate, benzyl alcohol, ascorbic acid, and the like. The parenteral formulations can be stored in any conventional containers such as vials and ampoules. [00206] Routes of topical administration include nasal, buccal, mucosal, rectal, or vaginal applications. For topical administration, the active compounds can be formulated into lotions, creams, ointments, gels, powders, pastes, sprays, suspensions, drops and aerosols. Thus, one or more thickening agents, humectants, and stabilizing agents can be included in the formulations. Examples of such agents include, but are not limited to, polyethylene glycol, sorbitol, xanthan gum, petrolatum, beeswax, or mineral oil, lanolin, squalene, and the like. A special form of topical administration is delivery by a transdermal patch. Methods for preparing transdermal patches are disclosed, e.g., in Brown, et al. (1988) Ann. Rev. Med. 39:221-229 which is incorporated herein by reference. [00207] Subcutaneous implantation for sustained release of the active compounds may also be a suitable route of administration. This entails surgical procedures for implanting an active compound in any suitable formulation into a subcutaneous space, e.g., beneath the anterior abdominal wall. See, e.g., Wilson et al. (1984) J. Clin. Psych. 45:242-247. Hydrogels can be used as a carrier for the sustained release of the active compounds. Hydrogels are generally known in the art. They are typically made by crosslinking high molecular weight biocompatible polymers into a network, which swells in water to form a gel like material. In some instances, hydrogels are biodegradable or biosorbable. For purposes of this disclosure, hydrogels made of polyethylene glycols, collagen, or poly(glycolic-co-L- lactic acid) may be useful. See, e.g., Phillips et al. (1984) J. Pharmaceut. Sci., 73: 1718- 1720. [00208] This disclosure further provides a compound as defined in any one of the embodiments herein above for use in therapy. This disclosure further provides a compound as defined in any one of the embodiments herein above for use as a medicament. [00209] The pharmaceutical composition may be formulated for dissolution into a solution for administration by such techniques as, for example, intravenous administration. The SMDCs and pharmaceutical compositions thereof are particularly useful for parenteral administration, i.e., subcutaneously (s.c.), intrathecally, intraperitoneally, intramuscularly (i.m.) or intravenously (i.v.). In embodiment, the SMDCs and pharmaceutical compositions thereof are administered intravenously or subcutaneously. [00210] Actual methods for preparing parenterally administrable compositions are well known or will be apparent to those skilled in the art and are described in more detail in, for example, Remington’s Pharmaceutical Science, 15th ed., Mack Publishing Company, Easton, Pa. For the preparation of intravenously administrable formulations of the disclosure see Akers, M. J. “Excipient-Drug interactions in Parenteral Formulations”, J. Pharm Sci 91 (2002) 2283-2300; the entire contents of which are incorporated herein by reference and to which the reader is specifically referred. [00211] The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, etc. The concentration of the antigen binding protein of the disclosure in such pharmaceutical formulation can vary widely, i.e., from less than about 0.5%, usually at or at least about 1% to as much as about 15 or 20% by weight and will be selected primarily based on fluid volumes, viscosities, etc., according to the particular mode of administration selected. [00212] This disclosure further provides a compound as defined in any one of the embodiments herein above for use in therapy. This disclosure further provides a compound as defined in any one of the embodiments herein above for use as a medicament. [00213] In an aspect, provided herein is a method of treating a disease in a subject in need thereof, said method including administering an effective amount of a small molecule drug conjugate (SMDC) comprising folic acid, a conjugation linker moiety (L) that binds to the carbonyl of the folic acid, and to a drug moiety covalently bound to linker L, and where L is optionally covalently bound to a half-life extender (HL). In embodiments, the folic acid binds to FR, such as FRα. [00214] In one aspect, an SMDC provided herein is used in a method of inhibiting proliferation of a FR-expressing cell, such as an FR-expressing cell, the method comprising contacting the cell with the SMDC, e.g., exposing the cell to the SMDC under conditions permissive for binding of the folic acid of the SMDC on the surface of the cell, thereby inhibiting the proliferation of the cell. In embodiments, the method is an in vitro or an in vivo method. In embodiments, the cell is a cancer cell. In embodiments, the cell is a non-small cell lung carcinoma (NSCLC). In embodiments, the cell is an ovarian cancer cell. In any of these embodiments, the cell may be a mammalian cell, such as a human cell. [00215] Inhibition of cell proliferation in vitro may be assayed using the CellTiter-GloTM Luminescent Cell Viability Assay, which is commercially available from Promega (Madison, WI). That assay determines the number of viable cells in culture based on quantitation of ATP present, which is an indication of metabolically active cells. See Crouch et al. (1993) J. Immunol. Meth. 160:81-88, US Pat. No. 6602677. The assay may be conducted in 96- or 384- well format, making it amenable to automated high-throughput screening (HTS). See Cree et al. (1995) AntiCancer Drugs 6:398-404. The assay procedure involves adding a single reagent (CellTiter-Glo® Reagent) directly to cultured cells. This results in cell lysis and generation of a luminescent signal produced by a luciferase reaction. The luminescent signal is proportional to the amount of ATP present, which is directly proportional to the number of viable cells present in culture. Data can be recorded by luminometer or CCD camera imaging device. The luminescence output is expressed as relative light units (RLU). [00216] In another aspect, a SMDC for use as a medicament is provided. In further aspects, a SMDC for use in a method of treatment is provided. In another aspect, provided herein is a method of treating a disease in a subject in need thereof, said method including administering an effective amount of a pharmaceutical composition of the SMDC as described herein. [00217] In embodiments, the disease is cancer. In embodiments, the cancer is associated with overexpression of FR, such as FRα. In embodiments, provided herein is SMDC for use in a method of treating an individual having a FR-expressing cancer, such as a FRα- expressing cancer, the method comprising administering to the individual an effective amount of the SMDC. In embodiments, the FR-expressing cancer is an epithelial-derived tumor. In embodiments, the FR-expressing cancer is non-small cell lung carcinoma (NSCLC), lung cancer, mesothelioma, or ovarian cancer. In embodiments, the FR-expressing cancer is non- small cell lung carcinoma (NSCLC) or ovarian cancer. In embodiments, the FR-expressing cancer is non-small cell lung carcinoma (NSCLC). In embodiments, the FR-expressing cancer is ovarian cancer. Any of the foregoing FR-expressing cancer types may be FRα- expressing cancers. In embodiments, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent. [00218] In embodiments, the FR-expressing cancer, such as the FRα-expressing cancer, is in a mammal. In embodiments, the mammal is human. [00219] In a further aspect, the present disclosure provides for the use of a SMDC in the manufacture or preparation of a medicament. In embodiment, the medicament is for treatment of an FR-expressing cancer, such as an FRα-expressing cancer. In a further embodiment, the medicament is for use in a method of treating an FR-expressing cancer, such as an FRα-expressing cancer, the method comprising administering to an individual having an FR-expressing cancer, such as an FRα-expressing cancer, an effective amount of the medicament. In embodiments, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent. [00220] In embodiments, the methods provided herein are for treating cancer in a mammal. In embodiments, the methods provided herein are for treating cancer in a human. [00221] In embodiments, the cancers that may be treated with an immunoconjugate or method provided herein include epithelial-derived tumors including ovarian, uterine, breast, endometrial, pancreatic, renal, lung, colorectal, and brain tumors. In embodiments, the cancers that may be treated with an immunoconjugate or a method provided herein include serous and endometrioid epithelial ovarian cancer, endometrial adenocarcinoma, non-small cell lung carcinoma (NSCLC) of the adenocarcinoma subtype, squamous lung cancer, mesotheliomas, and triple-negative breast cancer (TNBC). [00222] In embodiments, the cancer is ovarian cancer. In embodiments, the cancer is breast cancer. In embodiments, the cancer is lung cancer. In embodiments, the cancer is triple-negative breast cancer. In embodiments, the cancer is non-small cell lung carcinoma (NSCLC). In embodiments, the cancer is mesothelioma. Articles of Manufacture [00223] In a further aspect, provided herein is an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above is provided. The article of manufacture (a kit) comprises a container and a label or package insert on or associated with the container. Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc. The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the disorder and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). At least one active agent in the composition is an SMDC as described herein. The label or package insert indicates that the composition is used for treating the condition of choice. Moreover, the article of manufacture (a kit) may comprise (a) a first container with a composition contained therein, wherein the composition comprises an SMDC as described herein; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent. The article of manufacture in this embodiment of the disclosure may further comprise a package insert indicating that the compositions can be used to treat a particular condition. Alternatively, or additionally, the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution or dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes. EXAMPLES [00224] The following examples are meant to be illustrative and can be used to further understand embodiments of the present disclosure and should not be construed as limiting the scope of the present teachings in any way. [00225] The chemical reactions described in the Examples can be readily adapted to prepare a number of other compounds of the present disclosure, and alternative methods for preparing the compounds of this disclosure are deemed to be within the scope of this disclosure. For example, the synthesis of non-exemplified compounds according to the present disclosure can be successfully performed by modifications apparent to those skilled in the art, e.g., by utilizing other suitable reagents known in the art other than those described, or by making routing modifications of reaction conditions, reagents, and starting materials. Alternatively, other reactions disclosed herein or known in the art will be recognized as having applicability for preparing other compounds of the present disclosure. Synthetic Examples Example S1: Synthesis of Compound FR-PEG-Duo5.
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
[00226] S-Trityl-L-cysteine-2-chlorotrityl resin Chem Impex, compound 1, (0.3 - 1.1 meq/g, 200 - 400 mesh, 2 g, 2.2 mmol) was suspended in DMF (10 mL) in a peptide synthesis vessel. To a solution of compound 2 (2.11 g, 5.5 mmol) and DIPEA (1.14 g, 8.8 mmol) in 10 mL DMF, was added HATU (2.25 g, 5.94 mmol), and the resulting mixture was stirred for 5 minutes before it was added to the peptide vessel (with compound 1) and nitrogen was bubbled through the resulting mixture for two hours. The reaction mixture was then filtered and washed with DMF (3 times 10 mL each), and isopropyl alcohol (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 3). The resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), and isopropyl alcohol (3 times 10 mL each). [00227] To a solution of compound 4 (2.33 g, 5.5 mmol) and DIPEA (1.14 g, 8.8 mmol) in 10 mL DMF, was added HATU (2.25 g, 5.94 mmol), and the resulting mixture was stirred for 5 minutes before it was added to the peptide vessel (which contained compound 3), and nitrogen was bubbled through the resulting mixture for two hours. The reaction mixture was then filtered and washed with DMF (3 times 10 mL each), and isopropyl alcohol (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 5). The resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), and isopropyl alcohol (3 times 10 mL each). [00228] To a solution of compound 6 (0.98 g, 2.42 mmol) and DIPEA (1.14 g, 8.8 mmol) in 10 mL DMF, was added HATU (2.25 g, 5.94 mmol), and the resulting mixture was stirred for 5 minutes before it was added to the peptide vessel (which contained compound 5), and nitrogen was bubbled through the resulting mixture for two hours. The reaction mixture was then filtered and washed with DMF (3 times 10 mL each), and isopropyl alcohol (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 7). The resin was then treated with 2% hydrazine in DMF (10 mL). The mixture was allowed to react for 15 minutes. The resin was then washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each) and dichloromethane (3 times 10 mL each). [00229] Resin deprotection: The resin was treated with peptide deprotection mixture (TFA, water, triisopropyl silane (TIPS), 2,2'-(ethane-1,2-diylbis(oxy))bis(ethane-1-thiol) (SDBS), 94/2/2/2) at 45oC for 30 minutes before it was filtered. The resulting TFA mixture was diluted with cold ether and the suspension was centrifuged with 5 minutes. The ether solution was discarded and the solid was redissolved in minimal amount of acetonitrile/water (6/4) mixture. The crude product was purified directly on reverse phase HPLC and desired product peak was combined and freeze dried. Compound 7 was isolated as a fluffy yellowish powder. MS m/z 690.7 (M+H). [00230] Compound 9 (110 mg, 0.31 mmol) was added to a DMF solution of compound 8 (Duo5, the synthesis of which has been previously described in US Patent 10,590,165, which is incorporated herein in its entirety) (200 mg, 0.26 mmol), DIPEA (100 mg, 0.77 mmol), HOBt (81 mg, 0.6 mmol), and DMAP (50 mg) at room temperature. The resulting mixture was stirred at 50oC for 5 hours and LCMS showed a major desired product peak. The crude reaction mixture was purified directly on reverse phase HPLC and the desired product peak was combined and freeze dried. Compound 10 was isolated as a fluffy white powder. MS m/z 985.5 (M+H). [00231] To a solution of compound 7 (20 mg, 0.027 mmol) in nitrogen degassed acetonitrile/water mixture (6:4, 0.5 mL) were added solution of compound 10 (27 mg, 0.027 mmol) in nitrogen degassed acetonitrile/water mixture (6:4, 0.5 mL) and DIPEA (20 mg, 0.15mmol). The reaction mixture was stirred at room temperature for 1 hour. LCMS showed the desired peak mass. The crude mixture was purified directly on reverse phase HPLC. The desired peak was combined and freeze dried. FR-PEG-Duo5 was isolated as a fluffy yellowish powder. MS m/z 1563.7 (M+H). Example S2: Synthesis of Compound FR-VC-Duo5.
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
[00232] Rink amide-AM resin Chem Impex, compound 11, (1 g, 0.6-1.0 mmol/g, 200-400 mesh) was suspended in 10 mL DMF for 20 minutes in a peptide synthesis vessel. The resin was then treated with 10% piperidine in DMF (10 mL). The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each) and then DMF (3 times 10 mL each). To a solution of compound 12 (1.17 g, 2.5 mmol) were added DIPEA (1.14 g, 8.8 mmol) in 10 mL DMF, and HATU (1.1 g, 2.8 mmol), and the resulting mixture was stirred for 5 minutes before it was added to the peptide vessel (with compound 11) and nitrogen was bubbled through the resulting mixture for two hours. The reaction mixture was then filtered and washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and again with DMF (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 13). The resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). [00233] To a solution of compound 14 (1.02 g, 2.5 mmol) in 10 mL DMF, were added DIPEA (0.52 g, 4.0 mmol) and HATU (1.1 g, 2.8 mmol), and the resulting mixture was stirred for 5 minutes before it was added to the peptide vessel (which contained compound 13), and nitrogen was bubbled through the resulting mixture for two hours. The reaction mixture was then filtered and washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 15). The resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). [00234] To a solution of compound 2 (0.96 g, 2.5 mmol) in 10 mL DMF, were added DIPEA (0.52 g, 4.0 mmol) and HATU (1.1 g, 2.8 mmol), and the resulting mixture was stirred for 5 minutes before it was added to the peptide vessel (which contained compound 15), and nitrogen was bubbled through the resulting mixture for two hours. The reaction mixture was then filtered and washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 16). The resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). [00235] To a solution of compound 4 (1.1 g, 2.5 mmol) in 10 mL DMF, were added DIPEA (0.52 g, 4.0 mmol) and HATU (1.1 g, 2.8 mmol), and the resulting mixture was stirred for 5 minutes before it was added to the peptide vessel (which contained compound 16), and nitrogen was bubbled through the resulting mixture for two hours. The reaction mixture was then filtered and washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 17). The resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). [00236] To a solution of compound 6 (0.5 g, 1.22 mmol) in 10 mL DMF, were added DIPEA (0.52 g, 4.0 mmol) and HATU (1.1 g, 2.8 mmol), and the resulting mixture was stirred for 5 minutes before it was added to the peptide vessel (which contained compound 17), and nitrogen was bubbled through the resulting mixture for two hours. The reaction mixture was then filtered and washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 18). The resin was then treated with 2% hydrazine in DMF (10 mL). The mixture was allowed to react for 15 minutes. The resin was then washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each) and dichloromethane (3 times 10 mL each). [00237] Resin deprotection: The resin was treated with peptide deprotection mixture (TFA, water, triisopropyl silane (TIPS), 2,2'-(ethane-1,2-diylbis(oxy))bis(ethane-1-thiol) (SDBS), 94/2/2/2) at 45oC for 30 minutes before it was filtered. The resulting TFA mixture was diluted with cold ether and the suspension was centrifuged with 5 minutes. The ether solution was discarded and the solid was redissolved in minimal amount of acetonitrile/water (6/4) mixture. The crude product was purified directly on reverse phase HPLC and desired product peak was combined and freeze dried. Compound 18 was isolated as a fluffy
yellowish powder. MS m/z 829.3 (M+H).
Figure imgf000102_0001
Figure imgf000103_0001
[00238] Compounds 8 (Duo5, 5g, 5.65 mmol, 1.0 eq), 19 (2.56 g, 1.0 eq), and HOAt (2.3 g, 3.0 eq) were combined and then 200 mL of DCM and DIPEA (5.9 mL, 6 eq) were added, until the mixture forms a clear solution. Then EDC (3.23 g, 3eq) was added, and the mixture was stirred for 1-2 hours, and checked for completion with LCMS. After reaction was complete, the mixture was washed with water 2x50 mL, then brine (50 mL), and then evaporated to give 764 g of compound 20 [00239] Compound 20 was dissolved in 70 mL of acetonitrile, and 4 ml piperidine was added. The mixture was stirred for 20 minutes and checked for completion with LCMS. After completion reaction mixture was evaporated under vacuum. The residue was redissolved in 50 mL of acetonitrile and evaporated again, then coevaporated with 50 mL of acetonitrile for the second time, dissolved in DCM and washed twice with water. The product was extracted from DCM with 0.5 M HCl (aqueous, about 2.5-3 equivalents of HCl to the product required, the pH of aqueous extract must be 4 or lower). Aqueous solution was washed with DCM, and basified with sodium bicarbonate to pH 9. The resulting mixture was extracted with DCM, washed twice with water, and evaporated to yield 4.8 g of compound 21. [00240] Compounds 21 (1.0 eq), 22 (1.0 eq), and HOBt (3.0 eq) were combined and then 200 mL of DCM and DIPEA (4 eq) were added, until the mixture forms a clear solution. Then EDC (3eq) was added, and the mixture was stirred for 1-2 hours, and checked for completion with LCMS. After reaction was complete, the mixture was washed with water 2x50 mL, then brine (50 mL), and then evaporated to give 6.4 g of compound 23. [00241] Compound 23 (6.4 g) was dissolved in 15 mL of MeOH, then 50 mL of 4M HCl in dioxane was added while stirring. Reaction was checked for completion with LCMS after 20 minutes. After completion, reaction mixture was evaporated under vacuum. The residue was redissolved in 50 mL of acetonitrile and evaporated again, then coevaporated with 50 mL of acetonitrile for the second time, and dried under vacuum for at least 1 hour to yield compound 24. [00242] Compound 24 was dissolved in 25 mL of EtOH and 25 mL of water. DIPEA was added to reach pH of 5-6 (if too much DIPEA added the pH can be adjusted back with AcOH), followed by addition of NaOCN (3 eq) and stirred for 16 hours. The reaction was checked for completion with LCMS (If not completed check pH (pH should be between 5 and 7, if not adjust with AcOH), continue stirring for 24 h). Compound 25 was used directly in the next step. [00243] NaOH (10 eq) dissolved in 3 mL of water was added to the reaction mixture of compound 25 (from previous step). Reaction mixture was warmed to 50 °C while stirring. After 1 hour the reaction was checked for completion using LCMS. After completion reaction mixture was adjusted to pH to 5 with AcOH, purified by HPLC, and lyophilized. 4.17 g of compound 26 was isolated. [00244] TFA salt of compound 26 (4.17 g, 3.36 mmol) was dissolved in 100 mL DCM, followed by addition of compound 27 (5 eq) and add good quality EDC (3 eq). Reaction mixture was stirred for 20 minutes and checked for completion using LCMS. If not completed, more EDC was added and checked again in 10-20 min. After completion, the mixture was washed with 80 mL of water. DCM solution was dried over Na2SO4 for about 2- 5 min, then evaporated the solvent under vacuum at room temperature. The crude residue then dissolved in acetonitrile/water (7/3) and purified by HPLC in 10-15 runs injecting each time about 0.5-0.7 g of mixture. Only fractions containing desired diastereomer (major diastereomer) with acceptable purity were combined to give 3.8 g of compound 28 as a white powder. The yield (from compound 8) was 44%. MS m/z 1405.5 (M+H).
Figure imgf000106_0001
[00245] To a solution of compound 18 (20 mg, 0.024 mmol) in nitrogen degassed acetonitrile/water mixture (6:4, 0.5 mL) were added solution of compound 28 (27 mg, 0.014 mmol) in nitrogen degassed acetonitrile/water mixture (6:4, 0.5 mL) and DIPEA (20 mg, 0.15 mmol). The reaction mixture was stirred at room temperature for 1 hour. LCMS showed the desired peak mass. The crude mixture was purified directly on reverse phase HPLC. The desired peak was combined and freeze dried. FR-VC-Duo5 was isolated as a fluffy yellowish powder. MS m/z 2050.1 (M+H). Example S3: Synthesis of Compound FR-VC-IODO-Duo5.
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
[00246] To a solution of compound 29 (1.85 g, 5.50 mmol) in 10 mL DMF, were added DIPEA (1.20 g, 8.80 mmol) and HATU (2.30 g, 6.05 mmol), and the resulting mixture was stirred for 5 minutes before it was added to the peptide vessel (which contained compound 13), and nitrogen was bubbled through the resulting mixture for two hours. The reaction mixture was then filtered and washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 30). The resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). [00247] To a solution of compound 14 (2.30 g, 5.50 mmol) in 10 mL DMF, were added DIPEA (1.20 g, 8.80 mmol) and HATU (2.30 g, 6.05 mmol), and the resulting mixture was stirred for 5 minutes before it was added to the peptide vessel (which contained compound 30), and nitrogen was bubbled through the resulting mixture for two hours. The reaction mixture was then filtered and washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 31). The resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). [00248] To a solution of compound 2 (2.20 g, 5.50 mmol) in 10 mL DMF, were added DIPEA (1.20 g, 8.80 mmol) and HATU (2.30 g, 6.05 mmol), and the resulting mixture was stirred for 5 minutes before it was added to the peptide vessel (which contained compound 31), and nitrogen was bubbled through the resulting mixture for two hours. The reaction mixture was then filtered and washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 32). The resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). [00249] To a solution of compound 4 (3.40 g, 5.50 mmol) in 10 mL DMF, were added DIPEA (1.20 g, 8.80 mmol) and HATU (2.30 g, 6.05 mmol), and the resulting mixture was stirred for 5 minutes before it was added to the peptide vessel (which contained compound 32), and nitrogen was bubbled through the resulting mixture for two hours. The reaction mixture was then filtered and washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 33). The resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). [00250] To a solution of compound 6 (1.16 g, 2.86 mmol) in 10 mL DMF, were added DIPEA (1.20 g, 8.80 mmol) and HATU (2.30 g, 6.05 mmol), and the resulting mixture was stirred for 5 minutes before it was added to the peptide vessel (which contained compound 33), and nitrogen was bubbled through the resulting mixture for two hours. The reaction mixture was then filtered and washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 34). The resin was then treated with 2% hydrazine in DMF (10 mL). The mixture was allowed to react for 15 minutes. The resin was then washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each) and dichloromethane (3 times 10 mL each).
T C P 0 0-3 2 1 0-3 2 2 1 0.o N t e k co D y e n r ott A 011
Figure imgf000112_0001
[00251] A water solution (1 mL) of ascorbic acid (0.17 g, 1.00 mmol) was added to a DMSO/water (1:1, 1 mL) solution of CuSO4 (0.037 g, 0.15 mmol) and compound 36 (0.13 g, 0.25 mmol) and stirred for 5 minutes. After 5 minutes, the solution was added to a stirred suspension of compound 34 (from reaction above) and compound 35 (0.67 g, 1.25 mmol) in 10 mL DMF. The reaction mixture was stirred for 1 day, the resin was then washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each) and dichloromethane (3 times 10 mL each). [00252] Resin deprotection: The resin was treated with peptide deprotection mixture (TFA, water, triisopropyl silane (TIPS), 2,2'-(ethane-1,2-diylbis(oxy))bis(ethane-1-thiol) (SDBS), 94/2/2/2) at 45oC for 30 minutes before it was filtered. The resulting TFA mixture was diluted with cold ether and the suspension was centrifuged with 5 minutes. The ether solution was discarded and the solid was redissolved in minimal amount of acetonitrile/water (6/4) mixture. The crude product was purified directly on reverse phase HPLC and desired product peak was combined and freeze dried. Compound 37 was isolated as a fluffy yellowish powder. MS m/z 1458.5 (M+H).
Figure imgf000114_0001
[00253] To a solution of compound 37 (20 mg, 0.014 mmol) in nitrogen degassed acetonitrile/water mixture (6:4, 0.5 mL) were added solution of 28 (20 mg, 0.014 mmol) in nitrogen degassed acetonitrile/water mixture (6:4, 0.5 mL) and DIPEA (20 mg, 0.15 mmol). The reaction mixture was stirred at room temperature for 1 hour. LCMS showed the desired peak mass. The crude mixture was purified directly on reverse phase HPLC. The desired peak was combined and freeze dried. FR-VC-IODO-Duo5 was isolated as a fluffy yellowish powder. MS m/z 2679.1 (M+H). Example S4: Synthesis of Compound FR-PL-MMAE.
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
[00254] S-Trityl-L-cysteine-2-chlorotrityl resin Chem Impex, compound 1, (0.3 - 1.1 meq/g, 200 - 400 mesh, 2 g, 2.2 mmol) was suspended in DMF ( 10 mL) in a peptide synthesis vessel. To a solution of compound 14 (1.80 g, 4.4 mmol) in DMF (10 mL) and DIPEA (1.14 g, 8.8 mmol) was added HATU (2.25 g, 5.94 mmol) and the resulting mixture was stirred for 5 minutes before it was added to the peptide vessel (with compound 1) and nitrogen was bubbled through the resulting mixture for 2 hours. The reaction mixture was then filtered and washed with DMF (3 times 10 mL each), and isopropyl alcohol (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 38). The resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), and isopropyl alcohol (3 times 10 mL each). [00255] To a solution of compound 39 (4.30 g, 6.6 mmol) in 10 mL DMF were added DIPEA (1.14 g, 8.8 mmol) and HATU (3.21 g, 6.60 mmol) and the resulting mixture was stirred for 5 minutes before it was added to the peptide vessel (which contained compound 38), and nitrogen was bubbled through the resulting mixture for two hours. The reaction mixture was then filtered and washed with DMF (3 times 10 mL each), and isopropyl alcohol (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 40). The resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), and isopropyl alcohol (3 times 10 mL each). [00256] To a solution of compound 14 (1.80 g, 4.4 mmol) in 10 mL DMF were added DIPEA (1.14 g, 8.8 mmol) and HATU (2.25 g, 5.94 mmol) and the resulting mixture was stirred for 5 minutes before it was added to the peptide vessel (which contained compound 40). The resulting mixture was bubbled with nitrogen for 2 hours. The reaction mixture was then filtered and washed with DMF (3 times 10 mL each), and isopropyl alcohol (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 41). The resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), and isopropyl alcohol (3 times 10 mL each). [00257] To a solution of compound 4 (2.0 g, 4.4 mmol) in 10 mL DMF were added DIPEA (1.14 g, 8.8 mmol) and (2.25 g, 5.94 mmol) and the resulting mixture was stirred for 5 minutes before it was added to the peptide vessel (which contained compound 41), and nitrogen was bubbled through the resulting mixture for two hours. The reaction mixture was then filtered and washed with DMF (3 times 10 mL each), and isopropyl alcohol (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 42). The resin was then treated with 10% piperidine in DMF. The mixture was allowed to react for 1 hour. The resin was then washed with DMF (3 times 10 mL each), and isopropyl alcohol (3 times 10 mL each). [00258] To a solution of compound 6 (0.98 g, 2.42 mmol) in 10 mL DMF were added DIPEA (1.20 g, 8.80 mmol) and HATU (2.25 g, 5.94 mmol), and the resulting mixture was stirred for 5 minutes before it was added to the peptide vessel (which contained compound 42), and nitrogen was bubbled through the resulting mixture for two hours. The reaction mixture was then filtered and washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each), and then DMF (3 times 10 mL each). The completion of the reaction was determined by Kaiser test (formation of compound 43). The resin was then treated with 2% hydrazine in DMF (10 mL). The mixture was allowed to react for 15 minutes. The resin was then washed with DMF (3 times 10 mL each), isopropyl alcohol (3 times 10 mL each) and dichloromethane (3 times 10 mL each). [00259] Resin deprotection: The resin was treated with peptide deprotection mixture (TFA, water, triisopropyl silane (TIPS), 2,2'-(ethane-1,2-diylbis(oxy))bis(ethane-1-thiol) (SDBS), 94/2/2/2) at 45oC for 30 minutes before it was filtered. The resulting TFA mixture was diluted with cold ether and the suspension was centrifuged with 5 minutes. The ether solution was discarded and the solid was redissolved in minimal amount of acetonitrile/water (6/4) mixture. The crude product was purified directly on reverse phase HPLC and desired product peak was combined and freeze dried. Compound 43 was isolated as a fluffy yellowish powder. MS m/z 931.9 (M+H).
Figure imgf000119_0001
[00260] Compound 9 (29 mg, 0.084 mmol) was added to a DMF solution of MMAE compound 44 (50 mg, 0.070 mmol), DIPEA (100 mg, 0.4 mmol), HOBT (81 mg, 0.6 mmol), and DMAP (50 mg) at room temperature. The resulting mixture was stirred at 50oC for 5 hours and LCMS showed a major desired product peak. The crude reaction mixture was purified directly on reverse phase HPLC and the desired product peak was combined and freeze dried. Compound 45 was isolated as a fluffy white powder. MS m/z 931.5 (M+H).
T C P 0 0-3 2 1 0-3 2 2 1 0.o N t e k co D y e n r ott A 811
Figure imgf000120_0001
[00261] To a solution of compound 43 (28 mg, 0.036 mmol) in nitrogen degassed acetonitrile/water mixture (6:4, 0.5 mL) were added solution of compound 45 (22 mg, 0.031 mmol) in nitrogen degassed acetonitrile/water mixture (6:4, 0.5 mL) and DIPEA (20 mg, 0.15 mmol). The reaction mixture was stirred at room temperature for 1 hour. LCMS showed the desired peak mass. The crude mixture was purified directly on reverse phase HPLC. The desired peak was combined and freeze dried. FR-PL-MMAE was isolated as a fluffy yellowish powder. MS m/z 1750.8 (M+H). Example S5: Synthesis of Compound FR-VC-PAB-MMAE.
T C P 0 0-3 2 1 0-3 2 2 1 0.oN t e k co D y e n r ott A 021
Figure imgf000122_0001
[00262] To a solution of compound 7 (16 mg, 0.023 mmol) in nitrogen degassed acetonitrile/water mixture (6:4, 0.5 mL) were added solution of compound 46 (20 mg, 0.015 mmol) (Catalog #SET0201, Levena Biopharma, Suzhou, China) in nitrogen degassed acetonitrile/water mixture (6:4, 0.5 mL) and DIPEA (20 mg, 0.15 mmol). The reaction mixture was stirred at room temperature for 1 hour. LCMS showed the desired peak mass. The crude mixture was purified directly on reverse phase HPLC. The desired peak was combined and freeze dried. FR-PL-MMAE was isolated as a fluffy yellowish powder. MS m/z 2006.0 (M+H).
T C P 0 0-3 2 1 0-3 2 2 1 0.o N t e k co D y e n r ott A 221
Figure imgf000124_0001
[00263] HATU (84 mg, 0.22 mmol) was added to a stirred solution of (s)-4-(tert- butoxycarbonyl)morpholine-3-carboxylic acid (48 mg, 0.20 mmol) and DIPEA (85 mg, 0.66 mmol) in DMF (3 mL). After 2 minutes, exatecan mesylate (100 mg, 0.18 mmol) was added to the reaction solution. The resulting mixture was stirred for 1 hour and the LCMS showed the desired product mass. [00264] The reaction mixture was poured into a separatory funnel with EtOAc and saturated NH4Cl solution. After mixture and separation, the organic phase was washed with brine, dried with Na2SO4, filtered, and evaporated to give crude product as oil. [00265] The crude acylated exatecan product was dissolved in CH2Cl2 (3 mL) and TFA (3 mL) was added dropwise. After 1 hour at room temperature, LCMS showed the desired Boc- deprotected product. The solution was evaporated and azeotrope-distilled with toluene to give the Boc-deprotected amine as TFA salt which was used in the following step without further purification. [00266] The above amine was dissolved in DMF and (9H-fluoren-9-yl)methyl ((S)-3- methyl-1-(((S)-1-((4-((((4-nitrophenoxy)carbonyl)oxy)methyl)phenyl)amino)-1-oxo-5- ureidopentan-2-yl)amino)-1-oxobutan-2-yl)carbamate (98 mg, 0.18 mmol) and DIPEA (85 mg, 0.66 mmol) were added to the solution. After 3 hours at room temperature, LCMS showed full conversion to the desired product. Diethyl amine (200 mg) was added to deprotect the FMOC group. After 3 hours, the solution was purified by reverse phase LCMS using acetonitrile and water as eluent. The desired fractions were combined and freeze dried to obtain a white fluffy powder. [00267] bis(2,5-dioxopyrrolidin-1-yl) 4,7,10,13-tetraoxahexadecanedioate (98 mg, 0.20 mmol) in DMF was added to the above exatecan intermediate and DIPEA (85 mg) in DMF. After 3 hours at room temperature, LCMS showed complete conversion to the desired acylated product. The mixture was purified by reverse phase LCMS using acetonitrile and water as eluent. The desired fractions were combined and freeze dried to obtain a white fluffy powder. MS m/z 1327.5 (M+H).
Figure imgf000126_0001
Figure imgf000127_0001
[00268] To a solution of compound 37 (35 mg, 0.024 mmol) in nitrogen-degassed acetonitrile/water mixture (6:4, 0.5 mL) were added solution of compound 54 (32 mg, 0.024 mmol) in nitrogen-degassed acetonitrile/water mixture (6:4, 0.5 mL) and DIPEA (20 mg, 0.15 mmol). The reaction mixture was stirred at room temperature for 1 hour. LCMS showed the desired peak mass. The crude mixture was purified directly on reverse phase HPLC. The desired peak was combined and freeze dried. FR-VC-PAB-MMAE was isolated as a fluffy yellowish powder. MS m/z 2670 (M+H). Example B1: In vitro Efficacy of Duo5 and MMAE. [00269] The in vitro efficacies of Duo5 (the synthesis of which has been previously described in US Patent 10,590,165, which is incorporated herein in its entirety and MMAE were evaluated using the following human cancer cell lines: KB (FR+) and A549 (FR-), purchased from the American Type Culture Collection (ATCC; Manassas, VA) and routinely cultured in folic acid free RPMI 1640 medium (Catalog #27016021; Thermo Fisher Scientific; Waltham, MA) and RPMI 1640 medium (ATCC modification) (Catalog #A1049101; Thermo Fisher Scientific; Waltham, MA) supplemented with 10% fetal bovine serum (FBS; Catalog #F4135; Sigma-Aldrich; St. Louis, MO), respectively, and maintained at 37°C with 5% CO2 in a humidified environment. [00270] Tumor cells were washed with Dulbecco’s Phosphate Buffered Saline (DPBS; Catalog #PBL01; Caisson Labs; Smithfield, UT) and harvested by detachment with TrypLE Express (Catalog #1204013; Thermo Fisher Scientific; Waltham, MA). Viable cell counts were made by Trypan blue exclusion using a Countess II automated cell counter. Cell Viability Assay: All cells were harvested and seeded into 384-well white wall flat bottom plates (Catalog #3570; Corning) at a density of 3,000 cells/well in folic acid free RPMI 1640 medium or RPMI 1640 medium (ATCC modification) supplemented with 10% fetal bovine serum (complete growth media). Plates were maintained at 37°C overnight to allow cells to adhere to the plate. The outer wells of plates contained complete growth media only. Working solutions of test articles were prepared at 100X final concentrations with 10-fold serial dilutions in DMSO and subsequently diluted at 5X final concentrations with 10-fold serial dilutions in complete growth media. Cell treatment was performed in triplicates and maintained at 37°C for 72-hour assay. After treatment, cell viability was determined by CellTiter-Glo 2.0 assay (Catalog #G9242; Promega; Madison, WI, USA) based on the manufacturer’s instructions. CellTiter Glo reagent reacts with ATP in metabolically active cells to give a luminescent readout that is directly proportional to the number of viable cells. Briefly, plates were removed from the incubator and equilibrated to room temperature before addition of CellTiter Glo reagent. Luminescence was measured using a SpectraMax iD3 microplate reader (Molecular Devices; San Jose, CA). [00271] For Cytotoxicity assays, raw luminescence data was background subtracted with average luminescence from the wells containing medium only using Excel (Microsoft; Albuquerque, NM) and normalized to untreated controls using GraphPad Prism 9.0. Dose- response relationships and IC50 values were determined based on non-linear regression analysis of normalized data fit to a four-parameter logistic equation using GraphPad Prism 9.0. [00272] In vitro cytotoxic activities of the Duo5 and MMAE described herein were evaluated against FR-expressing KB and FR-negative A549 cancer cell lines using standard cell viability assays. As shown in FIGS.1A and 1B, Duo5 and MMAE dose-dependently reduced KB and A549 cell viability in 3-day assays. The potencies of Duo5 and MMAE in KB as determined by IC50 were similar with 0.8982 nM and 0.3707 nM, respectively. Although IC50 values of Duo5 and MMAE were higher in A549 than in KB, they inhibited cell proliferation across both cell lines in a dose-dependent manner regardless of FR expression level. [00273] Cell viability for Duo5 and MMAE are shown in FIGS. 1A and 1B and IC50 values are shown in Table 1. [00274] FIGS. 1A-B show results of an in vitro efficacy study of A) Duo5 and B) MMAE using: KB (FR+) cells and A549 (FR-) cells. Table 1: IC50 Values (nM) of Duo5 and MMAE in Human Tumor Cells
Figure imgf000129_0001
Example B2: In vitro Efficacy, Specificity, and Stability of Small Molecule-Drug Conjugates (SMDCs). [00275] SMDCs were evaluated using the following human cancer cell lines: FR-positive KB and FR-negative A549, purchased from the American Type Culture Collection (ATCC; Manassas, VA) and routinely cultured in folic acid free RPMI 1640 medium (Catalog #27016021; Thermo Fisher Scientific; Waltham, MA) and RPMI 1640 medium (ATCC modification) (Catalog #A1049101; Thermo Fisher Scientific; Waltham, MA) supplemented with 10% fetal bovine serum (FBS; Catalog #MT35011CV; Corning), respectively, and maintained at 37°C with 5% CO2 in a humidified environment. [00276] Tumor cells were washed with Dulbecco’s Phosphate Buffered Saline (DPBS; Catalog #PBL01; Caisson Labs; Smithfield, UT) and harvested by detachment with TrypLE Express (Catalog #1204013; Thermo Fisher Scientific; Waltham, MA). Viable cell counts were made by Trypan blue exclusion using a Countess or Countess II automated cell counter. Cell Viability Assay: All cells were harvested and seeded into 384-well white wall flat bottom plates (Catalog #3570; Corning) at a density of 1,000 cells/well (for 120-hour assay) or 3,000 cells/well in folic acid free RPMI 1640 medium or RPMI 1640 medium (ATCC modification) supplemented with 10% fetal bovine (complete growth media). Plates were maintained at 37°C overnight to allow cells to adhere to the plate. The outer wells of plates contained complete growth media only. Thirty minutes prior to the addition of SMDCs, the media in designated wells of KB cells were replaced with complete growth media containing 100 µM folic acid (a binding site competitor). KB cells in those designated wells were used to determine the targeting specificity of SMDCs for FR. Working solutions of test articles were prepared at 100X final concentrations with 5-fold serial dilutions in DMSO and subsequently diluted at 5X final concentrations with 5-fold serial dilutions in complete growth media (in the presence or absence of 100 µM folic acid for KB). Cell treatment was performed in triplicates and maintained at 37°C for (1) 2 hours followed by washing 3 times with complete growth media and incubating with complete growth media for another 70 hours (2-hour pulse assay); or (2) 72 hours or 120 hours (for FR-IODO-Examorpholine treatment) (72-hour assay or 120-hour assay). Cells treated with SMDCs using 72-hour assay or 120-hour assay were used to determine the stability of SMDCs in complete growth media. After treatment, cell viability was determined by CellTiter-Glo 2.0 assay (Catalog #G9242; Promega; Madison, WI, USA) based on the manufacturer’s instructions. CellTiter Glo reagent reacts with ATP in metabolically active cells to give a luminescent readout that is directly proportional to the number of viable cells. Briefly, plates were removed from the incubator and equilibrated to room temperature before addition of CellTiter Glo reagent. Luminescence was measured using a SpectraMax iD3 microplate reader (Molecular Devices; San Jose, CA). [00277] For Cytotoxicity assays, raw luminescence data was background subtracted with average luminescence from the wells containing medium only using Excel (Microsoft; Albuquerque, NM) and normalized to untreated controls using GraphPad Prism 9.0. Dose- response relationships and IC50 values were determined based on non-linear regression analysis of normalized data fit to a four-parameter logistic equation using GraphPad Prism 9.0. [00278] Cytotoxicity, specificity, and stability of SMDCs are shown in FIGS. 2A, 2B, 3A, 3B, 4A, 4B, 5A, 5B, 6A, 6B, 7A, 7B, and 8, and Tables 2 and 3. [00279] In vitro cytotoxic activities and targeting specificity of the SMDCs described herein were evaluated against FR-positive KB and FR-negative A549 cancer cell lines using standard cell viability assays. The specificity of SMDCs for FR was determined in two ways. First, as shown in FIGS. 2A, 3A (black circles), 4A (black circles), 5A (black circles), 6A (black circles), and 7A (black circles), treatment with SMDCs dose-dependently reduced KB cell viability and showed no or lower potent activity (>100X lower for FR-PEG-Duo5; >1000X lower for FR-VC-Duo5 and FR-VC-PAB-MMAE; >10X lower for FR-IODO-Duo5; and about 100X lower for FR-PL-MMAE) against A549 cells in 2-hour pulse assays. Secondly, as shown in FIGS. 3A, 4A, 5A, 6A, and 7A, treatment with SMDCs in the presence of 100 µM FA showed no or lower potent activity (>70X lower for FR-PEG-Duo5; >1000X lower for FR-VC-Duo5, FR-VC-PAB-MMAE, and FR-PL-MMAE; and >2X lower for FR-IODO-Duo5) against KB cells. Together, these data suggest that SMDCs target cells in a FR-dependent fashion since no potent activity or lower potent activity is observed when FR is absent or “blocked”. The potencies of SMDCs as determined by IC50 with 2-hour pulse assays ranging from 1.545 nM to 303.9 nM against FR-positive KB cells were observed (Table 2). [00280] IC50 Values (nM) of FA-SMDCs in Human Tumor Cells with 2-hour Pulse Assay are presented in Table 2. Table 2: IC50 Values (nM) of FA-SMDCs in Human Tumor Cells with 2-hour Pulse Assay
Figure imgf000132_0001
[00281] FIGS. 2A-B show results of an in vitro efficacy and stability study of SMDCs using A549 (FR-) cells with: A) 2-hour pulse assay and B) 72-hour assay or 120-hour assay (for FR-IODO-Examorpholine Treatment). [00282] FIGS. 3A-B show results of an in vitro efficacy, specificity, and stability study of FR-PEG-Duo5 using KB (FR+) cells with: A) 2-hour pulse assay and B) 72-hour assay. [00283] FIGS. 4A-B show results of an in vitro efficacy, specificity, and stability study of FR-VC-Duo5 using KB (FR+) cells with: A) 2-hour pulse assay and B) 72-hour assay. [00284] FIGS. 5A-B show results of an in vitro efficacy, specificity, and stability study of FR-IODO-Duo5 using KB (FR+) cells with: A) 2-hour pulse assay and B) 72-hour assay. [00285] FIGS. 6A-B show results of an in vitro efficacy, specificity, and stability study of FR-VC-PAB-MMAE using KB (FR+) cells with: A) 2-hour pulse assay and B) 72-hour assay. [00286] FIGS. 7A-B show results of an in vitro efficacy, specificity, and stability study of FR-PL-MMAE using KB (FR+) cells with: A) 2-hour pulse assay and B) 72-hour assay. [00287] FIG. 8 shows results of an in vitro efficacy, specificity, and stability study of FR- IODO-Examorpholine using KB (FR+) cells with 120-hour assay. [00288] In vitro stability of the SMDCs described herein was evaluated with 72-hour assay or 120-hour assay against FR-positive KB and FR-negative A549 cancer cell lines using standard cell viability assays. As shown in FIGS. 2B, 3B, 4B, 5B, 6B, 7B, and 8, treatment with FR-VC-Duo5, FR-IODO-Duo5, FR-VC-PAB-MMAE, and FR-IODO-Examorpholine showed FR-dependent cytotoxicity with higher potencies (>1000X higher for FR-VC-Duo5, >60X higher for FR-IODO-Duo5, >18X higher for FR-VC-PAB-MMAE, and >26X higher for FR-IODO-Examorpholine) in the presence of FR or in the absence of 100 µM FA, while treatment with FR-PEG-Duo5 and FR-PL-MMAE showed cytotoxicity regardless of FR expression level or the presence of free FA. These data indicate that FR-VC-Duo5, FR- IODO-Duo5, FR-VC-PAB-MMAE, and FR-IODO-Examorpholine are more stable in complete growth media than FR-PEG-Duo5 or FR-PL-MMAE. The potencies of SMDCs as determined by IC50 with 72-hour assays or 120-hour assay ranging from 0.5403 nM to 31.42 nM against FR-positive KB cells were observed (Table 3). [00289] IC50 Values (nM) of FA-SMDCs in Human Tumor Cells with 72-hour Assay are presented in Table 3. Table 3: IC50 Values (nM) of FA-SMDCs in Human Tumor Cells with 72-hour Assay or 120-hour Assay (for FR-IODO-Examorpholine Treatment)
Figure imgf000133_0001
Example B3: Mouse Pharmacokinetics (PK) for FR-VC-IODO-Duo5. [00290] FR-VC-IODO-Duo5 and Duo5 are both dissolved as standard solution in DMSO (10 mM). The dosing formulation consists of 1% DMSO and 99% PBS. The dosing level was 1 mg/Kg. [00291] 30 male ICR mice was used in the study and the FR-VC-IODO-Duo5 was injected via IV route. The blood was sampled at the following time points: 0.5, 2, 4, 6, 8, 24, 48, 72 and 120 hours post dose. The sampled blood was treated with EDTA-K2 and 1 molar citric acid solution to denature proteases in serum. [00292] LCMS analysis was performed to monitor the serum level of FR-VC-IODO-Duo5 and Duo5. The result of the PK study is presented in FIG. 9, Table 4, and Table 5. Table 4: Individual and mean plasma concentration-time data of FR-VC-IODO-Duo5 after an IV dose of FR-VC-IODO-Duo5 at 1 mg/kg to male ICR mouse (N=30/group)
Figure imgf000134_0001
Figure imgf000135_0001
LLOQ is 0.2 ng/mL for Duo5 or FR-VC-IODO-Duo5 in mouse plasma. NA: Not available. BLQ = Below the lower limit of quantification (LLOQ) If the adjusted rsq (linear regression coefficient of the concentration value on the terminal phase) is less than 0.9, T1/2 might not be accurately estimated. No abnormal clinical sign was observed during the entire in-life study. Table 5: Individual and mean plasma concentration-time data of Duo5 after an IV dose of Duo5 at 1 mg/kg to male ICR mouse (N=30/group)
Figure imgf000135_0002
[00293] Although the foregoing disclosure has been described in some detail by way of illustration and example for purposes of clarity of understanding, the descriptions and examples should not be construed as limiting the scope of the disclosure. Section headings are provided for the convenience of the reader and do not limit the scope of the disclosure. The disclosures of all patent and scientific literature cited herein are expressly incorporated herein in their entirety by reference. To the extent that any material incorporated by reference is inconsistent with the express content of this disclosure, the express content controls.

Claims

WHAT IS CLAIMED IS: 1. A compound of the Formula (I) or (II):
Figure imgf000137_0001
or a pharmaceutically acceptable salt thereof, wherein: L is a multivalent linker bound at least to a folic acid and a drug moiety; HL is a half-life extender; and D is a drug moiety.
2. The compound or a pharmaceutically acceptable salt thereof of claim 1, wherein L is a bond, -C(O)-, -NH-, Amino Acid Unit, Peptoid, –(CH2CH2O)n–, –(CH2)n–, –(4-aminobenzyloxycarbonyl)–, –(C(O)CH2CH2C(O))–, –(C(O)CH2CH2NH)–,
Figure imgf000138_0002
thereof; wherein n is an integer from 1 to 24; and each R2 and R3 is independently H or substituted or unsubstituted alkyl.
3. The compound or a pharmaceutically acceptable salt thereof of claim 1 or 2, wherein L is -C(O)-, -NH-,–(CH2CH2O)n–, –(CH2)n–, –(4-aminobenzyloxycarbonyl)–, -Cys-, -Asp-,
Figure imgf000138_0001
, , ,
Figure imgf000139_0001
, , r combinations thereof.
4. The compound or a pharmaceutically acceptable salt thereof of claim 3, wherein L is -C(O)-, -NH-, –(CH2CH2O)n–, –(CH2)n–, –SCH2CH2O–, –(C(O)CH2CH2C(O))–, -Val-, -Cit-, –(4-aminobenzyloxycarbonyl)–, -Arg-, -Asp-, -Lys-, -Cys-,
Figure imgf000139_0002
combinations thereof.
5. The compound or a pharmaceutically acceptable salt thereof of claim 4, wherein L is
Figure imgf000139_0003
,
Figure imgf000140_0001
Figure imgf000141_0001
, wherein the carbonyl is linked to the drug moiety (D), the amine is linked to the folic acid, and the optional third linkage is to the half-life extender (HL).
6. The compound or a pharmaceutically acceptable salt thereof of any one of claims 1-5, wherein HL is a cholesterol-like half-life extender or albumin binder half-life extender.
7. The compound or a pharmaceutically acceptable salt thereof of claim 6, wherein HL is
Figure imgf000141_0002
,
Figure imgf000142_0001
8. The compound or a pharmaceutically acceptable salt thereof of claim 7, wherein HL is
Figure imgf000142_0002
.
9. The compound or a pharmaceutically acceptable salt thereof of any one of claims 1-8, wherein D is a tubulin inhibitor or disruptor, kinase inhibitor, DNA damaging agent, transcription inhibitors, or proteolysis-targeting chimera (PROTAC).
10. The compound or a pharmaceutically acceptable salt thereof of claim 9, wherein D is a tubulin inhibitor.
11. The compound or a pharmaceutically acceptable salt thereof of any one of claims 1-9, wherein D is a pyrrolobenzodiazepine, duocarmycin, anthracycline, maytansinoid, auristatin, calicheamicin, camptothecin, RNA polymerase II inhibitor, topoisomerase I inhibitor, tyrosine kinase inhibitor, EG5 inhibitor, or MEK inhibitor.
12. The compound or a pharmaceutically acceptable salt thereof of claim 11, wherein D is an auristatin.
13. The compound or a pharmaceutically acceptable salt thereof of claim 11, wherein D is MMAE, MMAF, Duo5, PNU, SN-38, irinotecan, amatoxin, maytansine, exatecan, trametinib, abemaciclib, palbociclib, or examorpholine.
14. The compound or a pharmaceutically acceptable salt thereof of claim 13, wherein D is Duo5.
15. The compound or a pharmaceutically acceptable salt thereof of claim 13, wherein D is MMAE.
16. The compound or a pharmaceutically acceptable salt thereof of claim 13, wherein D is examorpholine.
17. The compound or pharmaceutically acceptable salt thereof of any one of claims 1-16, wherein the compound is:
T C P 0 0-3 2 1 0-3 2 2 1 0.oN t e k co D y e n r ott A 241
Figure imgf000144_0001
T C P 0 0-3 2 1 0-3 2 2 1 0.o N t e k co D y e n r ott A 341
Figure imgf000145_0001
441
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
or a pharmaceutically acceptable salt thereof.
18. The compound or pharmaceutically acceptable salt thereof of any one of claims 1-17, for use in therapy.
19. The compound or pharmaceutically acceptable salt thereof of claim 18, for use in treating a FR-expressing cancer, optionally wherein the FR-expressing cancer is non-small cell lung carcinoma (NSCLC), lung cancer, mesothelioma, or ovarian cancer.
20. A method of treating a FR-expressing cancer in a subject, comprising administering the compound or pharmaceutically acceptable salt thereof of any one of claims 1-17 to a subject in need thereof.
21. Use of the compound or pharmaceutically acceptable salt thereof of any one of claims 1- 17 for the manufacture of a medicament.
22. Use of the compound or pharmaceutically acceptable salt thereof of any one of claims 1- 17 for the manufacture of a medicament for treating a FR-expressing cancer, optionally wherein the FR-expressing cancer is non-small cell lung carcinoma (NSCLC), lung cancer, mesothelioma, or ovarian cancer.
23. The compound or pharmaceutically acceptable salt thereof for use, use, or method of any one of claims 19, 20, or 22, wherein the FR-expressing cancer is an epithelial-derived tumor.
24. The compound or pharmaceutically acceptable salt thereof for use, use, or method of claim 23, wherein the epithelial-derived tumors are ovarian, uterine, breast, endometrial, pancreatic, renal, lung, colorectal, or brain tumors.
25. The compound or pharmaceutically acceptable salt thereof for use, use, or method of any one of claims 19, 20, or 22, wherein the FR-expressing cancer is non-small cell lung carcinoma (NSCLC) or ovarian cancer.
26. The compound or pharmaceutically acceptable salt thereof for use, use, or method of any one of claims 19, 20, or 22-25, wherein the FR-expressing cancer is in a mammal, optionally wherein the mammal is a human.
27. A method of inhibiting proliferation of a FR-expressing cell, comprising contacting the FR-expressing cell with the compound or pharmaceutically acceptable salt thereof of any one of claims 1-17.
28. The use of claim 21, wherein the medicament is for inhibiting proliferation of a FR- expressing cell.
29. The compound or pharmaceutically acceptable salt thereof of any one of claims 1-17, for use in inhibiting proliferation of a FR-expressing cell.
30. The method, use, or compound or pharmaceutically acceptable salt thereof for use of any one of claims 27-29, wherein the FR-expressing cell is a FR-expressing cancer cell, optionally wherein the FR-expressing cancer is non-small cell lung carcinoma (NSCLC), lung cancer, mesothelioma, or ovarian cancer.
31. The method, use, or compound or pharmaceutically acceptable salt thereof for use of any one of claims 27-29, wherein the FR-expressing cell is a FR-expressing non-small cell lung carcinoma (NSCLC) cell or FR-expressing ovarian cell.
32. A pharmaceutical composition comprising the compound or pharmaceutically acceptable salt thereof of any one of claims 1-17, and a pharmaceutically acceptable excipient.
PCT/US2023/028509 2022-07-25 2023-07-24 Folate-conjugated drugs and uses thereof WO2024025845A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263392066P 2022-07-25 2022-07-25
US63/392,066 2022-07-25

Publications (1)

Publication Number Publication Date
WO2024025845A1 true WO2024025845A1 (en) 2024-02-01

Family

ID=87929276

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/028509 WO2024025845A1 (en) 2022-07-25 2023-07-24 Folate-conjugated drugs and uses thereof

Country Status (1)

Country Link
WO (1) WO2024025845A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013024035A1 (en) * 2011-08-17 2013-02-21 Merck & Cie Folate conjugates of albumin-binding entities
WO2014062697A2 (en) * 2012-10-16 2014-04-24 Endocyte, Inc. Drug delivery conjugates containing unnatural amino acids and methods for using
WO2021142145A1 (en) * 2020-01-07 2021-07-15 Gardeen Spencer S Targeted steroid conjugates

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013024035A1 (en) * 2011-08-17 2013-02-21 Merck & Cie Folate conjugates of albumin-binding entities
WO2014062697A2 (en) * 2012-10-16 2014-04-24 Endocyte, Inc. Drug delivery conjugates containing unnatural amino acids and methods for using
WO2021142145A1 (en) * 2020-01-07 2021-07-15 Gardeen Spencer S Targeted steroid conjugates

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
HALLER STEPHANIE ET AL: "Folate receptor-targeted radionuclide therapy: preclinical investigation of anti-tumor effects and potential radionephropathy", NUCLEAR MEDICINE AND BIOLOGY, vol. 42, no. 10, 1 October 2015 (2015-10-01), US, pages 770 - 779, XP093102698, ISSN: 0969-8051, DOI: 10.1016/j.nucmedbio.2015.06.006 *
NAKHAEI ELNAZ ET AL: "Design of a ligand for cancer imaging with long blood circulation and an enhanced accumulation ability in tumors", MEDCHEMCOMM, vol. 8, no. 6, 1 January 2017 (2017-01-01), United Kingdom, pages 1190 - 1195, XP093102375, ISSN: 2040-2503, DOI: 10.1039/C7MD00102A *
YIWU ZHENG ET AL: "Stabilizing p-Dithiobenzyl Urethane Linkers without Rate-Limiting Self-Immolation for Traceless Drug Release", CHEMMEDCHEM COMMUNICATIONS, WILEY-VCH, DE, vol. 14, no. 12, 16 May 2019 (2019-05-16), pages 1196 - 1203, XP072418567, ISSN: 1860-7179, DOI: 10.1002/CMDC.201900248 *

Similar Documents

Publication Publication Date Title
JP6703632B2 (en) Cytotoxic benzodiazepine derivative
CA3051737C (en) Low molecular weight drug conjugates for binding to carbonic anhydrase ix
CN110960537B (en) Combination of Bcl-2/Bcl-xL inhibitor and chemotherapeutic drug and application thereof
US20250205350A1 (en) Antibody-Drug Conjugates and Uses Thereof
US20240181073A1 (en) Antibody-Drug Conjugates Comprising an Anti-BCMA Antibody
ES2922898T3 (en) Multifunctional inhibitors of the MEK/PI3K and mTOR/MEK/PI3K biological pathways and therapeutic methods using the same
WO2023144379A1 (en) High-affinity ligands of fibroblast activation protein for targeted delivery applications
CA2922542A1 (en) Arylquinoline and analog compounds and use thereof to treat cancer
ES2313365T3 (en) T CELL LYMPHOMA TREATMENT USING 10-PROPARGIL-10-DEAZAAMINOPTERINA.
EP3773544B1 (en) Phospholipid-flavagline conjugates and methods of using the same for targeted cancer therapy
WO2024025845A1 (en) Folate-conjugated drugs and uses thereof
WO2024211236A2 (en) Antibody-drug conjugates and uses thereof
JP7223502B2 (en) Pharmaceutical composition
Vlková et al. Synthesis and biological activity evaluation of novel 2, 6, 9-trisubstituted purine conjugates as potential protein kinases inhibitors
WO2024211234A1 (en) Antibody-drug conjugates and uses thereof
WO2023217133A1 (en) Antibody-drug conjugates comprising an anti-folr1 antibody
CA3133798A1 (en) Non-hydrolyzable, non-cleavable, stable linkers for precision therapeutics and uses thereof
WO2024211235A1 (en) Antibody-drug conjugates and uses thereof
TW202302154A (en) Antibody-drug conjugates comprising an anti-bcma antibody
HK40016287B (en) Combination of bcl-2/bcl-xl inhibitors and chemotherapeutic agent and use thereof
HK40016287A (en) Combination of bcl-2/bcl-xl inhibitors and chemotherapeutic agent and use thereof
WO2025024681A1 (en) Ire1alpha inhibitors and uses thereof
US20250129068A1 (en) 4-(2-pyrazolo[3,4-b]pyridine-5-yl)ethynyl-2-pyridine derivatives useful as gcn2 inhibitors
JP2022145661A (en) PD-1 and/or PD-L1 inhibitors
WO2017079003A1 (en) Deuterated compounds for treating blood cancers, and compositions and methods thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23764742

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE