WO2022074537A1 - Biomarkers of tumor response to cdk4/6 inhibitors - Google Patents
Biomarkers of tumor response to cdk4/6 inhibitors Download PDFInfo
- Publication number
- WO2022074537A1 WO2022074537A1 PCT/IB2021/059086 IB2021059086W WO2022074537A1 WO 2022074537 A1 WO2022074537 A1 WO 2022074537A1 IB 2021059086 W IB2021059086 W IB 2021059086W WO 2022074537 A1 WO2022074537 A1 WO 2022074537A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- subject
- gene
- ccne1
- tmprss2
- erg
- Prior art date
Links
- 229940124297 CDK 4/6 inhibitor Drugs 0.000 title claims abstract description 37
- 206010028980 Neoplasm Diseases 0.000 title claims description 162
- 239000000090 biomarker Substances 0.000 title abstract description 13
- 230000004044 response Effects 0.000 title description 21
- 206010006187 Breast cancer Diseases 0.000 claims abstract description 102
- 230000004547 gene signature Effects 0.000 claims abstract description 98
- 229960004390 palbociclib Drugs 0.000 claims abstract description 94
- AHJRHEGDXFFMBM-UHFFFAOYSA-N palbociclib Chemical compound N1=C2N(C3CCCC3)C(=O)C(C(=O)C)=C(C)C2=CN=C1NC(N=C1)=CC=C1N1CCNCC1 AHJRHEGDXFFMBM-UHFFFAOYSA-N 0.000 claims abstract description 94
- 238000011282 treatment Methods 0.000 claims abstract description 90
- 208000026310 Breast neoplasm Diseases 0.000 claims abstract description 83
- 238000000034 method Methods 0.000 claims abstract description 67
- 230000014509 gene expression Effects 0.000 claims description 135
- 108090000623 proteins and genes Proteins 0.000 claims description 119
- 102100037858 G1/S-specific cyclin-E1 Human genes 0.000 claims description 86
- 101000738568 Homo sapiens G1/S-specific cyclin-E1 Proteins 0.000 claims description 86
- 102100038312 Transcription factor Dp-2 Human genes 0.000 claims description 80
- 239000002131 composite material Substances 0.000 claims description 80
- 101001010792 Homo sapiens Transcriptional regulator ERG Proteins 0.000 claims description 79
- 101000638154 Homo sapiens Transmembrane protease serine 2 Proteins 0.000 claims description 79
- 101001077417 Gallus gallus Potassium voltage-gated channel subfamily H member 6 Proteins 0.000 claims description 78
- 102100022807 Potassium voltage-gated channel subfamily H member 2 Human genes 0.000 claims description 78
- 102100031989 Transmembrane protease serine 2 Human genes 0.000 claims description 78
- 102100030510 Stanniocalcin-2 Human genes 0.000 claims description 77
- 101001043594 Homo sapiens Low-density lipoprotein receptor-related protein 5 Proteins 0.000 claims description 76
- 101000701446 Homo sapiens Stanniocalcin-2 Proteins 0.000 claims description 76
- 102100021926 Low-density lipoprotein receptor-related protein 5 Human genes 0.000 claims description 76
- 108010075654 MAP Kinase Kinase Kinase 1 Proteins 0.000 claims description 76
- 102100033115 Mitogen-activated protein kinase kinase kinase 1 Human genes 0.000 claims description 76
- 239000000523 sample Substances 0.000 claims description 72
- 108091008039 hormone receptors Proteins 0.000 claims description 67
- 108091006920 SLC38A2 Proteins 0.000 claims description 58
- 102100033774 Sodium-coupled neutral amino acid transporter 2 Human genes 0.000 claims description 58
- 101000766332 Homo sapiens Tribbles homolog 1 Proteins 0.000 claims description 57
- 102100026387 Tribbles homolog 1 Human genes 0.000 claims description 57
- 102100027943 Carnitine O-palmitoyltransferase 1, liver isoform Human genes 0.000 claims description 56
- 101000859570 Homo sapiens Carnitine O-palmitoyltransferase 1, liver isoform Proteins 0.000 claims description 56
- 201000011510 cancer Diseases 0.000 claims description 52
- 229940126588 endocrine therapeutic agent Drugs 0.000 claims description 45
- VWUXBMIQPBEWFH-WCCTWKNTSA-N Fulvestrant Chemical compound OC1=CC=C2[C@H]3CC[C@](C)([C@H](CC4)O)[C@@H]4[C@@H]3[C@H](CCCCCCCCCS(=O)CCCC(F)(F)C(F)(F)F)CC2=C1 VWUXBMIQPBEWFH-WCCTWKNTSA-N 0.000 claims description 44
- 229960002258 fulvestrant Drugs 0.000 claims description 44
- 108010009361 Cyclin-Dependent Kinase Inhibitor p19 Proteins 0.000 claims description 37
- 102000009506 Cyclin-Dependent Kinase Inhibitor p19 Human genes 0.000 claims description 37
- 102100024153 Cadherin-15 Human genes 0.000 claims description 36
- 101000762242 Homo sapiens Cadherin-15 Proteins 0.000 claims description 36
- 101001013799 Homo sapiens Metallothionein-1X Proteins 0.000 claims description 36
- 108010093662 Member 11 Subfamily B ATP Binding Cassette Transporter Proteins 0.000 claims description 36
- 102000001479 Member 11 Subfamily B ATP Binding Cassette Transporter Human genes 0.000 claims description 36
- 102100031781 Metallothionein-1X Human genes 0.000 claims description 36
- 206010055113 Breast cancer metastatic Diseases 0.000 claims description 33
- 101000896657 Homo sapiens Mitotic checkpoint serine/threonine-protein kinase BUB1 Proteins 0.000 claims description 32
- 102100021691 Mitotic checkpoint serine/threonine-protein kinase BUB1 Human genes 0.000 claims description 32
- 229960003881 letrozole Drugs 0.000 claims description 32
- HPJKCIUCZWXJDR-UHFFFAOYSA-N letrozole Chemical compound C1=CC(C#N)=CC=C1C(N1N=CN=C1)C1=CC=C(C#N)C=C1 HPJKCIUCZWXJDR-UHFFFAOYSA-N 0.000 claims description 32
- 102100033825 Collagen alpha-1(XI) chain Human genes 0.000 claims description 21
- 101000710623 Homo sapiens Collagen alpha-1(XI) chain Proteins 0.000 claims description 21
- 101000851181 Homo sapiens Epidermal growth factor receptor Proteins 0.000 claims description 19
- 238000009261 endocrine therapy Methods 0.000 claims description 12
- 229940034984 endocrine therapy antineoplastic and immunomodulating agent Drugs 0.000 claims description 12
- 108010025464 Cyclin-Dependent Kinase 4 Proteins 0.000 claims description 11
- 108010025468 Cyclin-Dependent Kinase 6 Proteins 0.000 claims description 11
- 102100036252 Cyclin-dependent kinase 4 Human genes 0.000 claims description 11
- 102100026804 Cyclin-dependent kinase 6 Human genes 0.000 claims description 11
- 229940088597 hormone Drugs 0.000 claims description 11
- 239000005556 hormone Substances 0.000 claims description 11
- 229940125944 selective estrogen receptor degrader Drugs 0.000 claims description 11
- 229940095743 selective estrogen receptor modulator Drugs 0.000 claims description 11
- 239000000333 selective estrogen receptor modulator Substances 0.000 claims description 11
- 239000003112 inhibitor Substances 0.000 claims description 10
- 101100328883 Arabidopsis thaliana COL1 gene Proteins 0.000 claims description 9
- 101100005789 Caenorhabditis elegans cdk-4 gene Proteins 0.000 claims description 9
- 239000003886 aromatase inhibitor Substances 0.000 claims description 9
- 229940122815 Aromatase inhibitor Drugs 0.000 claims description 8
- 101100496845 Arabidopsis thaliana COL11 gene Proteins 0.000 claims description 6
- BFYIZQONLCFLEV-DAELLWKTSA-N Aromasine Chemical compound O=C1C=C[C@]2(C)[C@H]3CC[C@](C)(C(CC4)=O)[C@@H]4[C@@H]3CC(=C)C2=C1 BFYIZQONLCFLEV-DAELLWKTSA-N 0.000 claims description 5
- 229960002932 anastrozole Drugs 0.000 claims description 5
- YBBLVLTVTVSKRW-UHFFFAOYSA-N anastrozole Chemical compound N#CC(C)(C)C1=CC(C(C)(C#N)C)=CC(CN2N=CN=C2)=C1 YBBLVLTVTVSKRW-UHFFFAOYSA-N 0.000 claims description 5
- 229960000255 exemestane Drugs 0.000 claims description 5
- 150000003839 salts Chemical class 0.000 claims description 4
- -1 BLIB1 B Proteins 0.000 claims description 3
- 238000010606 normalization Methods 0.000 claims description 2
- 101000666385 Homo sapiens Transcription factor Dp-2 Proteins 0.000 claims 23
- 230000008901 benefit Effects 0.000 abstract description 16
- 101710102998 Transcription factor Dp-2 Proteins 0.000 description 57
- 229940068196 placebo Drugs 0.000 description 37
- 239000000902 placebo Substances 0.000 description 37
- 206010061289 metastatic neoplasm Diseases 0.000 description 27
- 230000001394 metastastic effect Effects 0.000 description 25
- 101150000874 11 gene Proteins 0.000 description 24
- 238000004458 analytical method Methods 0.000 description 21
- 210000004185 liver Anatomy 0.000 description 21
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 20
- 210000001519 tissue Anatomy 0.000 description 19
- 101150025032 13 gene Proteins 0.000 description 18
- 230000003993 interaction Effects 0.000 description 18
- 101150106774 9 gene Proteins 0.000 description 17
- 210000004027 cell Anatomy 0.000 description 17
- 201000010099 disease Diseases 0.000 description 17
- 239000007787 solid Substances 0.000 description 14
- 239000003814 drug Substances 0.000 description 12
- 230000002124 endocrine Effects 0.000 description 10
- 239000011159 matrix material Substances 0.000 description 10
- 239000013610 patient sample Substances 0.000 description 10
- 230000004083 survival effect Effects 0.000 description 10
- 238000002560 therapeutic procedure Methods 0.000 description 10
- 238000013459 approach Methods 0.000 description 8
- 229940079593 drug Drugs 0.000 description 8
- 230000000694 effects Effects 0.000 description 7
- 108020004999 messenger RNA Proteins 0.000 description 7
- 230000035945 sensitivity Effects 0.000 description 7
- 102100038595 Estrogen receptor Human genes 0.000 description 6
- 208000024891 symptom Diseases 0.000 description 6
- 230000001225 therapeutic effect Effects 0.000 description 6
- 241000282414 Homo sapiens Species 0.000 description 5
- 230000000295 complement effect Effects 0.000 description 5
- 150000001875 compounds Chemical class 0.000 description 5
- 230000002401 inhibitory effect Effects 0.000 description 5
- 230000005764 inhibitory process Effects 0.000 description 5
- 238000009092 lines of therapy Methods 0.000 description 5
- 230000003211 malignant effect Effects 0.000 description 5
- 239000000092 prognostic biomarker Substances 0.000 description 5
- 101000882584 Homo sapiens Estrogen receptor Proteins 0.000 description 4
- 101001012157 Homo sapiens Receptor tyrosine-protein kinase erbB-2 Proteins 0.000 description 4
- 102100030086 Receptor tyrosine-protein kinase erbB-2 Human genes 0.000 description 4
- NKANXQFJJICGDU-QPLCGJKRSA-N Tamoxifen Chemical compound C=1C=CC=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=CC=C1 NKANXQFJJICGDU-QPLCGJKRSA-N 0.000 description 4
- 238000001574 biopsy Methods 0.000 description 4
- 238000002790 cross-validation Methods 0.000 description 4
- 230000003247 decreasing effect Effects 0.000 description 4
- 238000001914 filtration Methods 0.000 description 4
- 238000010199 gene set enrichment analysis Methods 0.000 description 4
- 238000010801 machine learning Methods 0.000 description 4
- 230000035772 mutation Effects 0.000 description 4
- 230000017074 necrotic cell death Effects 0.000 description 4
- 238000011227 neoadjuvant chemotherapy Methods 0.000 description 4
- 239000008194 pharmaceutical composition Substances 0.000 description 4
- 230000002829 reductive effect Effects 0.000 description 4
- 238000012360 testing method Methods 0.000 description 4
- 102100032187 Androgen receptor Human genes 0.000 description 3
- 206010027476 Metastases Diseases 0.000 description 3
- 206010039491 Sarcoma Diseases 0.000 description 3
- 102000013814 Wnt Human genes 0.000 description 3
- 108050003627 Wnt Proteins 0.000 description 3
- 108010080146 androgen receptors Proteins 0.000 description 3
- 230000003388 anti-hormonal effect Effects 0.000 description 3
- 230000031018 biological processes and functions Effects 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- 239000003153 chemical reaction reagent Substances 0.000 description 3
- 230000034994 death Effects 0.000 description 3
- 208000035475 disorder Diseases 0.000 description 3
- 238000001794 hormone therapy Methods 0.000 description 3
- 239000000203 mixture Substances 0.000 description 3
- 210000000056 organ Anatomy 0.000 description 3
- 230000001575 pathological effect Effects 0.000 description 3
- 238000000611 regression analysis Methods 0.000 description 3
- 230000008261 resistance mechanism Effects 0.000 description 3
- 238000012163 sequencing technique Methods 0.000 description 3
- 230000011664 signaling Effects 0.000 description 3
- 229940124597 therapeutic agent Drugs 0.000 description 3
- 230000004614 tumor growth Effects 0.000 description 3
- SIFNOOUKXBRGGB-AREMUKBSSA-N (6r)-6-[2-[ethyl-[[4-[2-(ethylamino)ethyl]phenyl]methyl]amino]-4-methoxyphenyl]-5,6,7,8-tetrahydronaphthalen-2-ol Chemical compound C1=CC(CCNCC)=CC=C1CN(CC)C1=CC(OC)=CC=C1[C@H]1CC2=CC=C(O)C=C2CC1 SIFNOOUKXBRGGB-AREMUKBSSA-N 0.000 description 2
- 108060000903 Beta-catenin Proteins 0.000 description 2
- 102000015735 Beta-catenin Human genes 0.000 description 2
- 101000715943 Caenorhabditis elegans Cyclin-dependent kinase 4 homolog Proteins 0.000 description 2
- 201000009030 Carcinoma Diseases 0.000 description 2
- 108010024986 Cyclin-Dependent Kinase 2 Proteins 0.000 description 2
- 102100036239 Cyclin-dependent kinase 2 Human genes 0.000 description 2
- 206010061818 Disease progression Diseases 0.000 description 2
- 108700012941 GNRH1 Proteins 0.000 description 2
- 239000000579 Gonadotropin-Releasing Hormone Substances 0.000 description 2
- WZUVPPKBWHMQCE-UHFFFAOYSA-N Haematoxylin Chemical compound C12=CC(O)=C(O)C=C2CC2(O)C1C1=CC=C(O)C(O)=C1OC2 WZUVPPKBWHMQCE-UHFFFAOYSA-N 0.000 description 2
- 206010025323 Lymphomas Diseases 0.000 description 2
- 206010027457 Metastases to liver Diseases 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- 206010060862 Prostate cancer Diseases 0.000 description 2
- 208000000236 Prostatic Neoplasms Diseases 0.000 description 2
- 238000003559 RNA-seq method Methods 0.000 description 2
- 230000002159 abnormal effect Effects 0.000 description 2
- 230000003213 activating effect Effects 0.000 description 2
- 230000004913 activation Effects 0.000 description 2
- 239000002671 adjuvant Substances 0.000 description 2
- 239000005557 antagonist Substances 0.000 description 2
- 230000000259 anti-tumor effect Effects 0.000 description 2
- 230000009286 beneficial effect Effects 0.000 description 2
- 239000013060 biological fluid Substances 0.000 description 2
- 210000001185 bone marrow Anatomy 0.000 description 2
- 238000007469 bone scintigraphy Methods 0.000 description 2
- 230000022131 cell cycle Effects 0.000 description 2
- 230000010261 cell growth Effects 0.000 description 2
- 238000012512 characterization method Methods 0.000 description 2
- 238000002591 computed tomography Methods 0.000 description 2
- 230000005750 disease progression Effects 0.000 description 2
- 108010038795 estrogen receptors Proteins 0.000 description 2
- 239000012530 fluid Substances 0.000 description 2
- 230000004927 fusion Effects 0.000 description 2
- 230000012010 growth Effects 0.000 description 2
- 238000009396 hybridization Methods 0.000 description 2
- 238000003384 imaging method Methods 0.000 description 2
- 208000032839 leukemia Diseases 0.000 description 2
- 108020001756 ligand binding domains Proteins 0.000 description 2
- 238000002595 magnetic resonance imaging Methods 0.000 description 2
- 238000002483 medication Methods 0.000 description 2
- 230000009401 metastasis Effects 0.000 description 2
- 230000002018 overexpression Effects 0.000 description 2
- 230000037361 pathway Effects 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- 230000035755 proliferation Effects 0.000 description 2
- 229960004622 raloxifene Drugs 0.000 description 2
- GZUITABIAKMVPG-UHFFFAOYSA-N raloxifene Chemical compound C1=CC(O)=CC=C1C1=C(C(=O)C=2C=CC(OCCN3CCCCC3)=CC=2)C2=CC=C(O)C=C2S1 GZUITABIAKMVPG-UHFFFAOYSA-N 0.000 description 2
- 229960001603 tamoxifen Drugs 0.000 description 2
- 230000002103 transcriptional effect Effects 0.000 description 2
- 238000011222 transcriptome analysis Methods 0.000 description 2
- 238000011269 treatment regimen Methods 0.000 description 2
- 238000002604 ultrasonography Methods 0.000 description 2
- KOAITBOFZOEDOC-BJMVGYQFSA-N (E)-3-[4-[[2-(4-fluoro-2,6-dimethylbenzoyl)-6-hydroxy-1-benzothiophen-3-yl]oxy]phenyl]prop-2-enoic acid Chemical compound FC1=CC(=C(C(=O)C2=C(C3=C(S2)C=C(C=C3)O)OC2=CC=C(C=C2)/C=C/C(=O)O)C(=C1)C)C KOAITBOFZOEDOC-BJMVGYQFSA-N 0.000 description 1
- SJXNPGGVGZXKKI-NYYWCZLTSA-N (E)-3-[4-[[2-[2-(1,1-difluoroethyl)-4-fluorophenyl]-6-hydroxy-1-benzothiophen-3-yl]oxy]phenyl]prop-2-enoic acid Chemical compound FC(C)(F)C1=C(C=CC(=C1)F)C1=C(C2=C(S1)C=C(C=C2)O)OC1=CC=C(C=C1)/C=C/C(=O)O SJXNPGGVGZXKKI-NYYWCZLTSA-N 0.000 description 1
- DFBDRVGWBHBJNR-BBNFHIFMSA-N (e)-3-[3,5-difluoro-4-[(1r,3r)-2-(2-fluoro-2-methylpropyl)-3-methyl-1,3,4,9-tetrahydropyrido[3,4-b]indol-1-yl]phenyl]prop-2-enoic acid Chemical compound C1([C@@H]2C3=C(C4=CC=CC=C4N3)C[C@H](N2CC(C)(C)F)C)=C(F)C=C(\C=C\C(O)=O)C=C1F DFBDRVGWBHBJNR-BBNFHIFMSA-N 0.000 description 1
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 1
- KISZAGQTIXIVAR-VWLOTQADSA-N 6-(2,4-dichlorophenyl)-5-[4-[(3S)-1-(3-fluoropropyl)pyrrolidin-3-yl]oxyphenyl]-8,9-dihydro-7H-benzo[7]annulene-2-carboxylic acid Chemical compound ClC1=C(C=CC(=C1)Cl)C1=C(C2=C(CCC1)C=C(C=C2)C(=O)O)C1=CC=C(C=C1)O[C@@H]1CN(CC1)CCCF KISZAGQTIXIVAR-VWLOTQADSA-N 0.000 description 1
- RHXHGRAEPCAFML-UHFFFAOYSA-N 7-cyclopentyl-n,n-dimethyl-2-[(5-piperazin-1-ylpyridin-2-yl)amino]pyrrolo[2,3-d]pyrimidine-6-carboxamide Chemical compound N1=C2N(C3CCCC3)C(C(=O)N(C)C)=CC2=CN=C1NC(N=C1)=CC=C1N1CCNCC1 RHXHGRAEPCAFML-UHFFFAOYSA-N 0.000 description 1
- 229940125815 AZD9833 Drugs 0.000 description 1
- 206010069754 Acquired gene mutation Diseases 0.000 description 1
- 108010031677 Anaphase-Promoting Complex-Cyclosome Proteins 0.000 description 1
- 102000005446 Anaphase-Promoting Complex-Cyclosome Human genes 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 208000036493 Contralateral breast cancer Diseases 0.000 description 1
- 230000005778 DNA damage Effects 0.000 description 1
- 231100000277 DNA damage Toxicity 0.000 description 1
- 230000006820 DNA synthesis Effects 0.000 description 1
- 108010093502 E2F Transcription Factors Proteins 0.000 description 1
- 102000001388 E2F Transcription Factors Human genes 0.000 description 1
- 238000002965 ELISA Methods 0.000 description 1
- 101150064205 ESR1 gene Proteins 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 108091008794 FGF receptors Proteins 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 229940126088 GDC-9545 Drugs 0.000 description 1
- 108010069236 Goserelin Proteins 0.000 description 1
- BLCLNMBMMGCOAS-URPVMXJPSA-N Goserelin Chemical compound C([C@@H](C(=O)N[C@H](COC(C)(C)C)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCN=C(N)N)C(=O)N1[C@@H](CCC1)C(=O)NNC(N)=O)NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H]1NC(=O)CC1)C1=CC=C(O)C=C1 BLCLNMBMMGCOAS-URPVMXJPSA-N 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000624625 Homo sapiens M-phase inducer phosphatase 1 Proteins 0.000 description 1
- 206010021143 Hypoxia Diseases 0.000 description 1
- 238000012313 Kruskal-Wallis test Methods 0.000 description 1
- 102100023326 M-phase inducer phosphatase 1 Human genes 0.000 description 1
- 108091054455 MAP kinase family Proteins 0.000 description 1
- 102000043136 MAP kinase family Human genes 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 102000029749 Microtubule Human genes 0.000 description 1
- 108091022875 Microtubule Proteins 0.000 description 1
- 101150097381 Mtor gene Proteins 0.000 description 1
- WDHOIABIERMLGY-CMJOXMDJSA-N N-[1-(3-fluoropropyl)azetidin-3-yl]-6-[(6S,8R)-8-methyl-7-(2,2,2-trifluoroethyl)-3,6,8,9-tetrahydropyrazolo[4,3-f]isoquinolin-6-yl]pyridin-3-amine Chemical compound FCCCN1CC(C1)NC=1C=NC(=CC=1)[C@H]1N([C@@H](CC2=C3C(=CC=C12)NN=C3)C)CC(F)(F)F WDHOIABIERMLGY-CMJOXMDJSA-N 0.000 description 1
- 206010061309 Neoplasm progression Diseases 0.000 description 1
- 238000000636 Northern blotting Methods 0.000 description 1
- 102000038030 PI3Ks Human genes 0.000 description 1
- 108091007960 PI3Ks Proteins 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- 108010018070 Proto-Oncogene Proteins c-ets Proteins 0.000 description 1
- 238000011529 RT qPCR Methods 0.000 description 1
- 238000012952 Resampling Methods 0.000 description 1
- 230000018199 S phase Effects 0.000 description 1
- 229940125943 SAR439859 Drugs 0.000 description 1
- 208000003837 Second Primary Neoplasms Diseases 0.000 description 1
- 102100023085 Serine/threonine-protein kinase mTOR Human genes 0.000 description 1
- 101710142154 Stanniocalcin-2 Proteins 0.000 description 1
- 238000000692 Student's t-test Methods 0.000 description 1
- 102000040945 Transcription factor Human genes 0.000 description 1
- 108091023040 Transcription factor Proteins 0.000 description 1
- 229940126003 ZN-c5 Drugs 0.000 description 1
- 229950001573 abemaciclib Drugs 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 230000000996 additive effect Effects 0.000 description 1
- 238000009098 adjuvant therapy Methods 0.000 description 1
- 229950003105 afimoxifene Drugs 0.000 description 1
- TXUZVZSFRXZGTL-QPLCGJKRSA-N afimoxifene Chemical compound C=1C=CC=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=C(O)C=C1 TXUZVZSFRXZGTL-QPLCGJKRSA-N 0.000 description 1
- 239000000556 agonist Substances 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- 230000001093 anti-cancer Effects 0.000 description 1
- 239000002246 antineoplastic agent Substances 0.000 description 1
- 229940046844 aromatase inhibitors Drugs 0.000 description 1
- 229960000817 bazedoxifene Drugs 0.000 description 1
- UCJGJABZCDBEDK-UHFFFAOYSA-N bazedoxifene Chemical compound C=1C=C(OCCN2CCCCCC2)C=CC=1CN1C2=CC=C(O)C=C2C(C)=C1C1=CC=C(O)C=C1 UCJGJABZCDBEDK-UHFFFAOYSA-N 0.000 description 1
- 208000013404 behavioral symptom Diseases 0.000 description 1
- 230000003542 behavioural effect Effects 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- 210000000481 breast Anatomy 0.000 description 1
- 230000004611 cancer cell death Effects 0.000 description 1
- 230000008235 cell cycle pathway Effects 0.000 description 1
- 230000006369 cell cycle progression Effects 0.000 description 1
- 230000032823 cell division Effects 0.000 description 1
- 238000002648 combination therapy Methods 0.000 description 1
- 238000011284 combination treatment Methods 0.000 description 1
- 238000011109 contamination Methods 0.000 description 1
- 208000031513 cyst Diseases 0.000 description 1
- 229940127089 cytotoxic agent Drugs 0.000 description 1
- 230000003111 delayed effect Effects 0.000 description 1
- 238000009795 derivation Methods 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 239000000032 diagnostic agent Substances 0.000 description 1
- 229940039227 diagnostic agent Drugs 0.000 description 1
- 229910003460 diamond Inorganic materials 0.000 description 1
- 239000010432 diamond Substances 0.000 description 1
- 229950005473 elacestrant Drugs 0.000 description 1
- 230000003511 endothelial effect Effects 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 230000029578 entry into host Effects 0.000 description 1
- YQGOJNYOYNNSMM-UHFFFAOYSA-N eosin Chemical compound [Na+].OC(=O)C1=CC=CC=C1C1=C2C=C(Br)C(=O)C(Br)=C2OC2=C(Br)C(O)=C(Br)C=C21 YQGOJNYOYNNSMM-UHFFFAOYSA-N 0.000 description 1
- 229940011871 estrogen Drugs 0.000 description 1
- 239000000262 estrogen Substances 0.000 description 1
- 239000000328 estrogen antagonist Substances 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 238000010195 expression analysis Methods 0.000 description 1
- 102000052178 fibroblast growth factor receptor activity proteins Human genes 0.000 description 1
- 238000009093 first-line therapy Methods 0.000 description 1
- 229960002913 goserelin Drugs 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 206010073071 hepatocellular carcinoma Diseases 0.000 description 1
- 231100000844 hepatocellular carcinoma Toxicity 0.000 description 1
- 239000000833 heterodimer Substances 0.000 description 1
- 208000018819 hormone-resistant breast carcinoma Diseases 0.000 description 1
- 230000007954 hypoxia Effects 0.000 description 1
- 229940126389 imlunestrant Drugs 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 238000000338 in vitro Methods 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 230000008595 infiltration Effects 0.000 description 1
- 238000001764 infiltration Methods 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 230000002452 interceptive effect Effects 0.000 description 1
- 229960002367 lasofoxifene Drugs 0.000 description 1
- GXESHMAMLJKROZ-IAPPQJPRSA-N lasofoxifene Chemical compound C1([C@@H]2[C@@H](C3=CC=C(C=C3CC2)O)C=2C=CC(OCCN3CCCC3)=CC=2)=CC=CC=C1 GXESHMAMLJKROZ-IAPPQJPRSA-N 0.000 description 1
- 239000003446 ligand Substances 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 210000005229 liver cell Anatomy 0.000 description 1
- 210000005228 liver tissue Anatomy 0.000 description 1
- 210000001165 lymph node Anatomy 0.000 description 1
- 210000004324 lymphatic system Anatomy 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 208000037819 metastatic cancer Diseases 0.000 description 1
- 208000011575 metastatic malignant neoplasm Diseases 0.000 description 1
- 210000004688 microtubule Anatomy 0.000 description 1
- 239000003607 modifier Substances 0.000 description 1
- 201000000050 myeloid neoplasm Diseases 0.000 description 1
- UZWDCWONPYILKI-UHFFFAOYSA-N n-[5-[(4-ethylpiperazin-1-yl)methyl]pyridin-2-yl]-5-fluoro-4-(7-fluoro-2-methyl-3-propan-2-ylbenzimidazol-5-yl)pyrimidin-2-amine Chemical compound C1CN(CC)CCN1CC(C=N1)=CC=C1NC1=NC=C(F)C(C=2C=C3N(C(C)C)C(C)=NC3=C(F)C=2)=N1 UZWDCWONPYILKI-UHFFFAOYSA-N 0.000 description 1
- 230000001338 necrotic effect Effects 0.000 description 1
- 238000009099 neoadjuvant therapy Methods 0.000 description 1
- 210000005170 neoplastic cell Anatomy 0.000 description 1
- 230000001613 neoplastic effect Effects 0.000 description 1
- 230000002611 ovarian Effects 0.000 description 1
- 201000008129 pancreatic ductal adenocarcinoma Diseases 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 230000002093 peripheral effect Effects 0.000 description 1
- 239000008177 pharmaceutical agent Substances 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- 238000002360 preparation method Methods 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 230000000069 prophylactic effect Effects 0.000 description 1
- 230000000306 recurrent effect Effects 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 238000003757 reverse transcription PCR Methods 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- 229950003687 ribociclib Drugs 0.000 description 1
- 229940073462 rintodestrant Drugs 0.000 description 1
- 208000011581 secondary neoplasm Diseases 0.000 description 1
- 238000004904 shortening Methods 0.000 description 1
- 230000037439 somatic mutation Effects 0.000 description 1
- 238000000528 statistical test Methods 0.000 description 1
- 238000012353 t test Methods 0.000 description 1
- 238000002626 targeted therapy Methods 0.000 description 1
- XFCLJVABOIYOMF-QPLCGJKRSA-N toremifene Chemical compound C1=CC(OCCN(C)C)=CC=C1C(\C=1C=CC=CC=1)=C(\CCCl)C1=CC=CC=C1 XFCLJVABOIYOMF-QPLCGJKRSA-N 0.000 description 1
- 229960005026 toremifene Drugs 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 230000005751 tumor progression Effects 0.000 description 1
- 238000007473 univariate analysis Methods 0.000 description 1
- 230000003827 upregulation Effects 0.000 description 1
- 230000002792 vascular Effects 0.000 description 1
- 238000001262 western blot Methods 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q1/00—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
- C12Q1/68—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
- C12Q1/6876—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
- C12Q1/6883—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
- C12Q1/6886—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q2600/00—Oligonucleotides characterized by their use
- C12Q2600/112—Disease subtyping, staging or classification
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q2600/00—Oligonucleotides characterized by their use
- C12Q2600/118—Prognosis of disease development
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q2600/00—Oligonucleotides characterized by their use
- C12Q2600/158—Expression markers
Definitions
- the invention relates to biomarkers of tumor response, gene signatures, and methods for the selection and treatment of subjects having hormone receptor positive (HR+), human epidermal growth factor receptor 2 negative (HER2-) breast cancer who are likely to benefit from administration of a CDK4/6 inhibitor.
- HR+ hormone receptor positive
- HER2- human epidermal growth factor receptor 2 negative
- the cell cycle mechanism has been exploited by therapeutic agents that impair disparate aspects of cell division, including DNA synthesis, microtubule assembly, and DNA damage. More recently, targeted approaches, including CDK4/6 inhibitors such as palbociclib, have been developed. In clinical trials, palbociclib was found to significantly improve progression free survival (PFS) in hormone receptor positive (HR+), human epidermal growth factor receptor 2 negative (HER2-) advanced or metastatic breast cancer when combined with endocrine therapy.
- PFS progression free survival
- HR+ hormone receptor positive
- HER2- human epidermal growth factor receptor 2 negative
- Palbociclib, ribociclib and abemaciclib have been approved for treatment of hormone receptor (HR)-positive, human epidermal growth factor receptor 2 negative (HER2-) advanced or metastatic breast cancer in combination with aromatase inhibitors, such as letrozole, in a first line setting and with fulvestrant in second or later lines of therapy in certain patients.
- HR hormone receptor
- HER2- human epidermal growth factor receptor 2 negative
- aromatase inhibitors such as letrozole
- CCNE1 mRNA expression level alone could not significantly stratify patient response from the PALOMA-2 trial (ClinicalTrials.gov identifier: NCT01740427), in patients with estrogen receptor positive (ER+), HER2- advanced breast cancer who had not received prior endocrine therapy for their advanced disease. These first-line patients were randomly assigned to receive palbociclib plus letrozole or placebo plus letrozole. See Finn et al., Biomarker Analyses of Response to Cyclin-Dependent Kinase 4/6 Inhibition and Endocrine Therapy in Women with Treatment-Naive Metastatic Breast Cancer. Clin Cancer Res 26: 110-121 , 2020.
- This invention relates to biomarkers of tumor response, gene signatures, and methods for the selection and treatment of subjects having hormone receptor positive (HR+), human epidermal growth factor receptor 2 negative (HER2-) breast cancer who are likely to benefit from administration of a CDK4/6 inhibitor.
- the invention provides a method of treating hormone receptorpositive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer in a subject in need thereof, comprising:
- the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
- the invention provides a method of selecting a subject having hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer for treatment with a CDK4/6 inhibitor and an endocrine therapeutic agent, comprising:
- the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
- the invention provides a method of predicting whether a subject having hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer will respond to treatment with a CDK4/6 inhibitor and an endocrine therapeutic agent, comprising: (a) determining the expression level for each gene in a gene signature in a tumor sample from the subject;
- the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
- the CDK4/6 inhibitor is palbociclib, or a pharmaceutically acceptable salt thereof.
- the endocrine therapeutic agent is selected from the group consisting of an aromatase inhibitor, a selective estrogen receptor degrader (SERD), and a selective estrogen receptor modulator (SERM).
- the endocrine therapeutic agent is selected from the group consisting of letrozole, anastrozole, exemestane and fulvestrant.
- FIG. 1 shows an 11 -gene exploratory signature from PALOMA-3 using the elastic net algorithm.
- Bar length (top x-axis) reflects the proportion of 100 bootstrap models containing the gene, while diamond points (bottom x-axis) reflect the average non-zero coefficient across bootstrap models.
- FIG. 2 shows a hierarchical clustering heatmap of the 11 -gene exploratory signature and progression-free survival (PFS). Shown are patients treated with palbociclib + fulvestrant (A) or placebo + fulvestrant (B), in each case sorted by PFS excluding those with a follow-up time of ⁇ 12 months and no event. Clustering was conducted using 1 -Pearson correlation as distance metric and average linkage.
- FIG. 3 shows the association between the 11 -gene exploratory signature and clinical benefit (PFS) in metastatic samples from PALOMA-3 (A) and in the full data set from PALOMA-3 (B). Patients were dichotomized by median signature score into low (dashed line) and high (solid line) score groups. Also shown are hazard ratios for each gene in the 11 -gene exploratory signature using the full data set from PALOMA-3 (C).
- PFS clinical benefit
- FIG. 4 shows the association between the 11 -gene exploratory signature and clinical benefit (PFS) from the addition of palbociclib in an independent metastatic breast cancer cohort from PALOMA-2. Patients were dichotomized by median signature score into low (dashed line) and high (solid line) score groups.
- FIG. 5 shows a 9-gene exploratory signature from PALOMA-3 using the elastic net algorithm.
- Bar length (top x-axis) reflects the proportion of 1 ,000 bootstrap models containing the gene, while circular points (bottom x-axis) reflect the average non-zero coefficient across bootstrap models.
- FIG. 6 shows the association between the 9-gene exploratory signature and clinical benefit (PFS) in the full data set from PALOMA-3 (A) and in an independent metastatic breast cancer cohort from PALOMA-2 (B). Patients were dichotomized by median signature score into low (dashed line) and high (solid line) score groups.
- FIG. 7 shows a 13-gene exploratory signature from PALOMA-3 using the elastic net algorithm.
- Bar length (top x-axis) reflects the proportion of 1 ,000 bootstrap models containing the gene, while circular points (bottom x-axis) reflect the average non-zero coefficient across bootstrap models.
- FIG. 8 shows the association between the 13-gene exploratory signature and clinical benefit (PFS) in the full data set from PALOMA-3 after filtering out outliers from NMF liver factor (A), and in an independent metastatic breast cancer cohort from PALOMA-2 (B). Patients were dichotomized by median signature score into low (dashed line) and high (solid line) score groups.
- FIG. 9 shows identification of samples with significant liver component in PALOMA-3 data set before (A) and after filtering out outliers from NMF liver factor (B).
- administration refers to contact of an exogenous pharmaceutical, therapeutic or diagnostic agent, or composition, to the animal, human, experimental subject, cell, tissue, organ or biological fluid.
- Treatment of a cell encompasses contact of a reagent to the cell, as well as contact of a reagent to a fluid, where the fluid is in contact with the cell.
- administration may also relate to in vitro and ex vivo treatment, e.g., of a cell, by a reagent, diagnostic, binding compound, or by another cell.
- cancer refers to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth.
- cancer refers to any malignant and/or invasive growth or tumor caused by abnormal cell growth.
- cancer refers to solid tumors named for the type of cells that form them, cancer of blood, bone marrow, or the lymphatic system. Examples of solid tumors include but not limited to sarcomas and carcinomas. Examples of cancers of the blood include but not limited to leukemias, lymphomas and myeloma.
- cancer includes but is not limited to a primary cancer that originates at a specific site in the body, a metastatic cancer that has spread from the place in which it started to other parts of the body, a recurrence from the original primary cancer after remission, and a second primary cancer that is a new primary cancer in a person with a history of previous cancer of a different type from latter one.
- a subject may be identified as having de novo metastatic disease or after progression from an earlier-identified cancer.
- patient refers to any single subject for which therapy is desired or that is participating in a clinical trial, epidemiological study or used as a control, including humans and mammalian veterinary patients such as cattle, horses, dogs and cats.
- the subject is a human.
- the subject is a postmenopausal woman or a man.
- the subject is a pre- or perimenopausal woman treated with a luteinizing hormone releasing hormone (LNRH) agonist, such as goserelin, so that their ovarian function is suppressed.
- LNRH luteinizing hormone releasing hormone
- Subjects may be treatment naive (i.e., the subject has not received prior treatment for advanced disease if metastatic, or for early disease if diagnosed with early breast cancer) or may have received one or more prior lines of treatment (i.e., in second or later line settings), such as one or more endocrine therapeutic agents or chemotherapeutic agents.
- the subject is treated with an aromatase inhibitor as their initial endocrine therapeutic agent (i.e., as initial endocrine based therapy).
- the subject has disease progression on or after treatment with an endocrine therapeutic agent in an adjuvant or metastatic setting.
- treat or “treating” a cancer as used herein means to administer a combination therapy comprising a CDK4/6 inhibitor and an endocrine therapeutic agent, according to the present invention to a subject having cancer, or diagnosed with cancer, to achieve at least one positive therapeutic effect, such as, for example, reduced number of cancer cells, reduced tumor size, reduced rate of cancer cell infiltration into peripheral organs, or reduced rate of tumor metastases or tumor growth, reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition.
- treatment refers to the act of treating as "treating” is defined immediately above.
- the term “treating” also includes adjuvant and neo-adjuvant treatment of a subject.
- a “reference breast cancer population” refers to a population of individuals having HR+, HER2- breast cancer of which the subject is a member.
- the reference breast cancer population could refer to a cohort of patients in a clinical trial enrolling the same or similar stage(s) of breast cancer as the subject.
- the reference breast cancer population could refer to a relevant population of individuals who have been successfully treated with a CDK4/6 inhibitor and endocrine therapy.
- the reference breast cancer population could refer to the cohorts from the PALOMA-2 or PALOMA-3 studies for advanced or metastatic breast cancer, or to a clinical trial cohorts from a relevant early breast cancer trial.
- a “control population” refers to a population of individuals who do not have cancer but are otherwise matched to the subject. The skilled person will be able to select an appropriate control population to provide the requisite reference value.
- beneficial or desired clinical results include, but are not limited to, one or more of the following: reducing the proliferation of (or destroying) neoplastic or cancerous cell; inhibiting metastasis or neoplastic cells; shrinking or decreasing the size of a tumor; remission of the cancer; decreasing symptoms resulting from the cancer; increasing the quality of life of those suffering from the cancer; decreasing the dose of other medications required to treat the cancer; delaying the progression of the cancer; curing the cancer; overcoming one or more resistance mechanisms of the cancer; and/or prolonging survival of patients the cancer.
- T/C tumor growth inhibition
- NCI National Cancer Institute
- the treatment achieved by a combination of the invention is defined by reference to any of the following: partial response (PR), complete response (CR), overall response (OR), progression free survival (PFS), disease free survival (DFS), invasive disease free survival (iDFS), and overall survival (OS).
- PR partial response
- CR complete response
- OR overall response
- PFS progression free survival
- DFS disease free survival
- iDFS invasive disease free survival
- OS overall survival
- PFS also referred to as “Time to Tumor Progression” indicates the length of time during and after treatment that the cancer does not grow and includes the amount of time patients have experienced a CR or PR, as well as the amount of time patients have experienced stable disease (SD).
- SD stable disease
- iDFS is defined according to Hudis (J Clin Oncol 2007) as the time between randomization and first event (e.g., ipsi- or contralateral invasive inbreast or loco-regional recurrence, distant recurrence, death from breast cancer, death from non-breast cancer cause, death from unknown cause, invasive contralateral breast cancer, second primary invasive cancer (non-breast)).
- OS refers to a prolongation in life expectancy as compared to naive or untreated subjects or patients.
- response to a combination of the invention is any of PR, CR, PFS, DFS, OR or OS that is assessed using Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 response criteria.
- the treatment regimen that is effective to treat a cancer patient may vary according to factors such as the disease state, age, and weight of the patient, and the ability of the therapy to elicit an anti-cancer response in the subject. While the aspects of the present invention may not be effective in achieving a positive therapeutic effect in every subject, it should do so in a statistically significant number of subjects as determined by any statistical test known in the art such as the Student’s t-test, the chi2- test the ll-test according to Mann and Whitney, the Kruskal-Wallis test (H-test), Jonckheere-Terpstrat-testy and the Wilcon on-test.
- treatment regimen may be used interchangeably to refer to the dose and timing of administration of each therapeutic agent administered according to the invention herein.
- “Ameliorating” means a lessening or improvement of one or more symptoms upon treatment with a combination described herein, as compared to not administering the combination. “Ameliorating” also includes shortening or reduction in duration of a symptom.
- an “effective dosage” or “effective amount” of drug, compound or pharmaceutical composition is an amount sufficient to affect any one or more beneficial or desired, including biochemical, histological and I or behavioral symptoms, of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
- an effective amount refers to that amount which will relieve to some extent one or more of the symptoms of the disorder being treated.
- a therapeutically effective amount refers to that amount which has the effect of (1 ) reducing the size of the tumor, (2) inhibiting (that is, slowing to some extent, preferably stopping) tumor metastasis, (3) inhibiting to some extent (that is, slowing to some extent, preferably stopping) tumor growth or tumor invasiveness, (4) relieving to some extent (or, preferably, eliminating) one or more signs or symptoms associated with the cancer, (5) decreasing the dose of other medications required to treat the disease, and/or (6) enhancing the effect of another medication, and/or (7) delaying the progression of the disease in a patient.
- an effective dosage can be administered in one or more administrations.
- an effective dosage of drug, compound, or pharmaceutical composition is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly.
- an effective dosage of drug, compound or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound or pharmaceutical composition.
- Tumor as it applies to a subject diagnosed with, or suspected of having, a cancer refers to a malignant or potentially malignant neoplasm or tissue mass of any size and includes primary tumors and secondary neoplasms.
- a solid tumor is an abnormal growth or mass of tissue that usually does not contain cysts or liquid areas. Examples of solid tumors are sarcomas, carcinomas, and lymphomas. Leukaemia’s (cancers of the blood) generally do not form solid tumors (National Cancer Institute, Dictionary of Cancer Terms).
- Tumor burden refers to the total amount of tumorous material distributed throughout the body. Tumor burden refers to the total number of cancer cells or the total size of tumor(s), throughout the body, including lymph nodes and bone marrow. Tumor burden can be determined by a variety of methods known in the art, such as, e.g., using calipers, or while in the body using imaging techniques, e.g., ultrasound, bone scan, computed tomography (CT), or magnetic resonance imaging (MRI) scans.
- imaging techniques e.g., ultrasound, bone scan, computed tomography (CT), or magnetic resonance imaging (MRI) scans.
- tumor size refers to the total size of the tumor which can be measured as the length and width of a tumor. Tumor size may be determined by a variety of methods known in the art, such as, e.g., by measuring the dimensions of tumor(s) upon removal from the subject, e.g., using calipers, or while in the body using imaging techniques, e.g., bone scan, ultrasound, CR or MRI scans.
- imaging techniques e.g., bone scan, ultrasound, CR or MRI scans.
- the PALOMA-3 trial (ClinicalTrials.gov identifier: NCT01942135) was a Phase III double-blind study in women aged 18 years or older with HR+, HER2- metastatic breast cancer, who had progressed after previous endocrine therapy. 521 endocrine-pretreated patients were randomly assigned to receive palbociclib plus fulvestrant or placebo plus fulvestrant (palbociclib arm, 347 patients; placebo arm, 174 patients). The primary endpoint was PFS.
- the PALOMA-2 trial (ClinicalTrials.gov identifier: NCT01740427) was a Phase III double-blind study in post-menopausal women with ER+, HER2- advanced breast cancer, who had not received prior treatment for advanced disease. 666 patients were randomly assigned to receive palbociclib plus letrozole or placebo plus letrozole (palbociclib arm, 444 patients; placebo arm, 222 patients). The primary endpoint was PFS.
- Baseline tumor tissue samples from the PALOMA-3 trial were profiled using the EdgeSeq Oncology Biomarker Panel (OBP) panel (HTG Molecular Diagnostics), with 302 tumor samples evaluable for analysis.
- OBP EdgeSeq Oncology Biomarker Panel
- the number of evaluable samples from each arm of the PALOMA-2 trial are summarized in Table 2 below.
- the tumor samples came from a mixture of primary or metastatic biopsies and the sites of tumor tissue collection were not recorded.
- the elastic net technique was employed to derive exploratory, predictive multigene expression features using patient samples from the PALOMA-3 study, which could then be tested for predictive power using the data set from the PALOMA-2 study.
- the machine-learning based elastic net regularization algorithm was applied.
- Clinical outcome association was performed by Cox PH regression using a number of bootstraps (e.g., 100 or 1 ,000 bootstraps) to provide a list of complementary gene expression features that comprise a model from each round. Genes were selected based on frequency observed in the bootstraps. Typically, at least 100 bootstraps are performed, and frequently 1 ,000 bootstraps are performed. In some methods, bootstraps are performed until the results have stabilized.
- the elastic net method linearly combines the L1 and L2 penalties of the lasso (least absolute shrinkage and selection operator) and ridge regression analysis methods.
- Lasso regression analysis performs variable selection and regularization using the penalty, which imposes sparsity among the coefficients to increase the accuracy and interpretability of the fitted model.
- the ridge regression method uses regularization to limit coefficient vector size.
- the machine-learning based elastic net regularization algorithm was applied, with clinical outcome association by Cox PH regression.
- the elastic net process involves:
- bootstraps typically at least 100 bootstraps are used, and frequently 1 ,000 bootstraps are used. In some methods, additional bootstraps are performed until the results have stabilized.
- the elastic net method was applied to identify multivariate gene signatures capable of predicting palbociclib efficacy in HR+, HER2- breast cancer.
- Gene signatures were derived using expression data obtained from patient samples in the PALOMA-3 and PALOMA-2 trials using the EdgeSeq Oncology Biomarker Panel (OBP) panel (HTG Molecular Diagnostics).
- OBP EdgeSeq Oncology Biomarker Panel
- Three multi-gene signatures were derived, encompassing different combinations of the following genes: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
- Example 1 an 11 -gene signature was discovered that is predictive of palbociclib efficacy in both endocrine-refractory and advanced treatment naive HR+, HER2- advanced or metastatic breast cancer.
- the 11 -gene signature was discovered using 92 metastatic samples from the palbociclib plus fulvestrant treatment arm of the PALOMA-3 trial in HR+, HER2- endocrine pretreated metastatic breast cancer patients.
- Six of the genes (CCNE1 , CPT1A, ERG, LRP5, TMPRSS2, and TRIB1 ) appear to be associated with relative resistance to palbociclib, while the remaining five genes (CDH15, MAP3K1 , SLC38A2, STC2, and TFDP2) appear to be associated with relative sensitivity to palbociclib (FIG. 1 ).
- FIG. 2 shows a hierarchical clustering heatmap of the 11- gene exploratory signature and progression-free survival (PFS). Shown are patients treated with palbociclib + fulvestrant in FIG. 2(A) or placebo + fulvestrant FIG. 2(B), in each case sorted by PFS excluding those with an insufficient follow-up time of ⁇ 12 months and no event.
- PFS progression-free survival
- FIG. 3(A) shows the association between the 11 -gene exploratory signature and clinical benefit (PFS) as a Kaplan-Meier plot for the metastatic samples from PALOMA-3.
- the gene signature was further applied to the full cohort from the PALOMA-3 trial, including 302 primary and metastatic samples from the palbociclib treatment and placebo arms (Table 1 ).
- FIG. 3(B) shows the association between the 11 -gene exploratory signature and clinical benefit (PFS) as a Kaplan-Meier plot for the full PALOMA-3 data set, including both metastatic and primary tumor samples.
- TFDP2 transcription factor DP-2
- E2F transcription factor DP-2
- TMPRSS2 and ERG are known fusion partners found in approximately half of prostate cancer patients, resulting in androgen receptor (AR) induced over-expression (PMID: 16254181 and 18283340).
- FIG. 6(A) shows the association between the 9-gene exploratory signature and clinical benefit (PFS) as a Kaplan-Meier plot for the full PALOMA-3 data set, including 302 primary and metastatic samples from the palbociclib treatment and placebo arms (Table 1 ).
- the 9-gene signature identified some of the same genes as in the 11 -gene signature, including transcription factor DP-2 (TFDP2) and the genes TMPRSS2 and ERG associated with AR induced over-expression.
- TFDP2 transcription factor DP-2
- TMPRSS2 transcription factor DP-2
- ERG ERG associated with AR induced over-expression
- NMF Non-Negative Matrix Factorization
- the virtual microdissection identified fourteen molecular factors that characterize biological processes differentiating baseline tumor samples of this cohort.
- One of the fourteen factors unambiguously represented the liver specific genes. Since a number of samples were from biopsies of liver metastases, a liver-specific factor was used to identify samples with significant liver component as described herein. Using this approach, 30 samples with significant liver content were identified and excluded from further analysis.
- FIG. 7 shows the 13-gene exploratory signature, where bar length (top x-axis) reflects the proportion of 1000 bootstrap models containing the gene while purple points (bottom x-axis) reflect average non-zero coefficient across bootstrap models.
- FIG. 8(A) shows the association between the 13-gene exploratory signature and clinical benefit (PFS) in the full data set from PALOMA-3 after filtering out outliers from NMF liver factor (FIG. 9).
- the 13-gene signature was further validated using independent samples from the PALOMA-2 trial.
- the ability of the gene signature to predict palbociclib efficacy across lines of therapy was demonstrated by application of the 13-gene signature to an independent metastatic breast cancer cohort in the PALOMA-2 trial.
- Association between the 13-gene exploratory signature and clinical benefit from the combination of palbociclib plus letrozole in the PALOMA-2 trial is shown in the Kaplan-Meier plot in FIG. 8(B).
- BUB1 B and CDKN2D are regulators of the cell cycle pathway, with the former inhibiting anaphase-promoting complex/cyclosome (Vleugel et al, 2015) and the latter an inhibitor to the activation of the CDK4/6 complex (Rolland et al, 2014).
- TRIB1 regulates the MAPK pathway (Jamieson et al, 2018).
- Additional genes associated with relative palbociclib resistance in the 13-gene signature include CCNE1 , as well as targets of MYC and WNT signaling. See Birdsey et al., The endothelial transcription factor ERG promotes vascular stability and growth through Wnt/beta-catenin signaling. Dev Cell 32:82-96, 2015; Pacilli et al., Carnitineacyltransferase system inhibition, cancer cell death, and prevention of myc-induced lymphomagenesis. J Natl Cancer Inst 105:489-498, 2013; Stanford et al., Cyclin E1 and cyclin-dependent kinase 2 are critical for initiation, but not for progression of hepatocellular carcinoma.
- EdgeSeq profiling has been used to evaluate expression levels for the gene signatures provided herein, the skilled person would understand that other techniques (e.g., qPCR, RNAseq, or NanoString technology) could be employed to evaluate gene expression could be used to train the EN models.
- the invention provides a method of treating hormone receptorpositive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer in a subject in need thereof, comprising:
- the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
- the subject is selected for treatment when the composite score is less than the first quartile of the reference composite score computed for the gene signature in the reference breast cancer population.
- the invention provides a method of selecting a subject having hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer for treatment with a CDK4/6 inhibitor and an endocrine therapeutic agent, comprising:
- the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
- the subject is selected for treatment when the composite score is less than the first quartile of the reference composite score computed for the gene signature in the reference breast cancer population.
- the invention provides a method of predicting whether a subject having hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer will respond to treatment with a CDK4/6 inhibitor and an endocrine therapeutic agent, comprising:
- the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
- the subject is predicted to respond to treatment when the composite score is less than the first quartile of the reference composite score computed for the gene signature in the reference breast cancer population.
- the invention provides a method of predicting whether a subject having hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer will be resistant to treatment with a CDK4/6 inhibitor and an endocrine therapeutic agent, comprising:
- the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
- the subject is predicted to be resistant to treatment when the composite score is greater than the third quartile of the reference composite score computed for the gene signature in the reference breast cancer population.
- the breast cancer is HR+, HER2- advanced or metastatic breast cancer.
- the HR+, HER2- advanced or metastatic breast cancer had progressed on prior endocrine therapy (i.e., the method relates to a second line or later line of therapy).
- the HR+, HER2- advanced or metastatic breast cancer is treatment naive (i.e. , the method relates to a first line of therapy).
- the breast cancer is HR+, HER2- early breast cancer. In some embodiments of each of the methods herein, the breast cancer is HR+, HER2- high risk early breast cancer. In some embodiments of each of the methods herein, the breast cancer is HR+, HER2- early breast cancer at high risk of relapse after showing less than pathological complete response to neoadjuvant chemotherapy. In some embodiments of each of the methods herein, the breast cancer is HR+, HER2- early breast cancer with residual invasive disease after neoadjuvant chemotherapy.
- the CDK4/6 inhibitor and the endocrine therapeutic agent are administered sequentially, simultaneously or concurrently.
- the endocrine therapeutic agent is selected from the group consisting of an aromatase inhibitor, a selective estrogen receptor degrader (SERD), and a selective estrogen receptor modulator (SERM).
- the endocrine therapeutic agent is selected from the group consisting of letrozole, anastrozole, exemestane and fulvestrant.
- the aromatase inhibitor is selected from the group consisting of letrozole, anastrozole, and exemestane. In some such embodiments, the aromatase inhibitor is letrozole. In some embodiments, the endocrine therapeutic agent is a SERD.
- the SERD is selected from the group consisting of fulvestrant, elacestrant (RAD-1901 , Radius Health), SAR439859 (Sanofi), RG6171 (Roche), AZD9833 (AstraZeneca), AZD9496 (AstraZeneca), rintodestrant (G1 Therapeutics), ZN-c5 (Zentalis), LSZ102 (Novartis), D-0502 (Inventisbio), LY3484356 (Lilly), and SHR9549 (Jiansu Hengrui Medicine).
- the SERD is fulvestrant.
- the endocrine therapeutic agent is a SERM.
- the SERM is selected from the group consisting of tamoxifen, raloxifene, toremifene, lasofoxifene, apeledoxifene and afimoxifene. In some such embodiments, the SERM is tamoxifen or raloxifene.
- the CDK4/6 inhibitor is palbociclib, or a pharmaceutically acceptable salt thereof.
- the gene signature comprises: (a) CCNE1 , CDH15, CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1 ;
- the gene signature consists essentially of:
- the gene signature consists of:
- the invention provides a method of treating hormone receptorpositive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer in a subject in need thereof, comprising:
- the expression levels of CCNE1 , CPT1A, ERG, LRP5, TMPRSS2, and TRIB1 in the tumor sample are downregulated relative to the expression levels in a non-tumor sample from the subject or relative to the median expression levels in a control population; and/or (ii) the expression levels of CDH15, MAP3K1 , SLC38A2, STC2, and TFDP2 in the tumor sample are upregulated relative to the expression levels in a non-tumor sample from the subject or relative to the median expression levels in a control population; and
- the subject is selected for treatment when:
- the expression levels of CCNE1 , CPT1A, ERG, LRP5, TMPRSS2, and TRIB1 in the tumor sample are downregulated relative to the expression levels in a non-tumor sample from the subject or relative to the first quartile expression levels in a control population;
- the expression levels of CDH15, MAP3K1 , SLC38A2, STC2, and TFDP2 in the tumor sample are upregulated relative to the expression levels in a non-tumor sample from the subject or relative to the first quartile expression levels in a control population.
- the invention provides a method of treating hormone receptorpositive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer in a subject in need thereof, comprising:
- the expression levels of CCNE1 , CPT1A, ERG, LRP5, and TMPRSS2 in the tumor sample are downregulated relative to the expression levels in a non-tumor sample from the subject or the median expression levels in a control population;
- the expression levels of MAP3K1 , SLC38A2, STC2, and TFDP2 in the tumor sample are upregulated relative to the expression levels in a non- tumor sample from the subject or the median expression levels in a control population;
- the subject is selected for treatment when: (i) the expression levels of CCNE1 , CPT1A, ERG, LRP5, and TMPRSS2 in the tumor sample are downregulated relative to the expression levels in a non-tumor sample from the subject or the first quartile expression levels in a control population; and/or
- the expression levels of MAP3K1 , SLC38A2, STC2, and TFDP2 in the tumor sample are upregulated relative to the expression levels in a nontumor sample from the subject or the first quartile expression levels in a control population.
- the invention provides a method of treating hormone receptorpositive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer in a subject in need thereof, comprising:
- the expression levels of BUB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, and TRIB1 in the tumor sample are downregulated relative to the expression levels in a non-tumor sample from the subject or the median expression levels in a control population; and/or
- the expression levels of ABCB11 , COL11 A1 , MAP3K1 , STC2 and TFDP2 in the tumor sample are upregulated relative to the expression levels in a non-tumor sample from the subject or the median expression levels in a control population;
- the subject is selected for treatment when:
- the expression levels of BUB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, and TRIB1 in the tumor sample are downregulated relative to the expression levels in a non-tumor sample from the subject or the first quartile expression levels in a control population;
- the expression levels of ABCB11 , COL11 A1 , MAP3K1 , STC2 and TFDP2 in the tumor sample are upregulated relative to the expression levels in a non-tumor sample from the subject or the first quartile expression levels in a control population.
- the invention provides use of a combination comprising a CDK4/6 inhibitor and an endocrine therapeutic agent for treating hormone receptorpositive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer in a subject in need thereof, comprising:
- the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
- the subject is selected for treatment when the composite score is less than the first quartile of the reference composite score computed for the gene signature in the reference breast cancer population.
- the invention provides a combination of a CDK4/6 inhibitor and an endocrine therapeutic agent for use in treating hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer in a subject in need thereof, comprising:
- the subject is selected for treatment with the combination when the composite score is less than the median of a reference composite score computed for the gene signature in a reference breast cancer population; wherein the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
- the subject is selected for treatment when the composite score is less than the first quartile of the reference composite score computed for the gene signature in the reference breast cancer population.
- the breast cancer is HR+, HER2- advanced or metastatic breast cancer.
- the HR+, HER2- advanced or metastatic breast cancer had progressed on prior endocrine therapy (i.e., the method relates to a second line or later line of therapy).
- the HR+, HER2- advanced or metastatic breast cancer is treatment naive (i.e., the method relates to a first line of therapy).
- the breast cancer is HR+, HER2- early breast cancer. In some embodiments of each of the uses and combinations herein, the breast cancer is HR+, HER2- high risk early breast cancer. In some embodiments of each of the uses and combinations herein, the breast cancer is HR+, HER2- early breast cancer at high risk of relapse after showing less than pathological complete response to neoadjuvant chemotherapy. In some embodiments of each of the uses and combinations herein, the breast cancer is HR+, HER2- early breast cancer with residual invasive disease after neoadjuvant chemotherapy.
- the CDK4/6 inhibitor and the endocrine therapeutic agent are administered sequentially, simultaneously or concurrently.
- the endocrine therapeutic agent is selected from the group consisting of an aromatase inhibitor, a selective estrogen receptor degrader (SERD), and a selective estrogen receptor modulator (SERM).
- the endocrine therapeutic agent is selected from the group consisting of letrozole, anastrozole, exemestane and fulvestrant.
- the CDK4/6 inhibitor is palbociclib, or a pharmaceutically acceptable salt thereof.
- the gene signature comprises:
- the gene signature consists essentially of:
- the gene signature consists of:
- the invention provides a kit for assaying a tumor sample to determine a composite score for a gene signature in the tumor sample, comprising a first set of probes for detecting the expression level of each gene in the gene signature, wherein the gene signature comprises:
- the invention provides a kit further comprising a second set of probes for detecting the expression level of a set of normalization genes in the tumor sample.
- a composite signature score was computed by weighting expression of signature genes by their coefficients.
- FIG. 1 shows the 11 -gene exploratory signature, where bar length (top x-axis) reflects the proportion of 100 bootstrap models containing the gene while purple points (bottom x-axis) reflect average non-zero coefficient across bootstrap models.
- Six of the genes (CCNE1 , CPT1A, ERG, LRP5, TMPRSS2, TRIB1 ) appear to be associated with relative resistance, while the remaining five genes (CDH15, MAP3K1 , SLC38A2, STC2, TFDP2) appear to be associated with relative sensitivity (FIG. 1 ).
- a composite signature score was computed by weighting each component gene by its average coefficient across bootstrap models.
- FIG. 2 A hierarchical clustering heatmap of the 11 -gene exploratory signature and progression-free survival (PFS) is provided in FIG. 2.
- Clustering was conducted using 1 -Pearson correlation as distance metric and average linkage.
- Shown in FIG. 2(A) are the metastatic samples across the palbociclib plus fulvestrant treatment arm of PALOMA-3 .
- Shown in FIG. 2(B) are placebo plus fulvestrant control arm. For each arm, patients are sorted by PFS, excluding those with an insufficient follow-up time of less than 12 months and no event.
- the 11 -gene signature was applied to 142 metastatic patient samples from PALOMA-3, comprising both the palbociclib plus fulvestrant and the placebo plus fulvestrant arms (Table 1 ).
- Composite signature scores were computed by weighting each component gene by its average coefficient across bootstrap models.
- Composite scores for patients in the palbociclib arm and the placebo arm were dichotomized by median into high and low scoring groups.
- the 11 -gene signature was further applied to the full PALOMA-3 data set, comprising 302 patient samples, including both primary and metastatic samples (Table 1 ).
- Composite signature scores were computed by weighting each component gene by its average coefficient across bootstrap models.
- Composite scores for patients in the palbociclib arm and the placebo arm were dichotomized by median into high and low scoring groups.
- Interaction P value for statistical interaction between gene expression and treatment for the 11 genes in the signature (CCNE1 , CPT1A, ERG, LRP5, TMPRSS2, TRIB1 , CDH15, MAP3K1 , SLC38A2, STC2, and TFDP2).
- the 11 -gene signature was further validated on an independent metastatic breast cancer cohort from first-line PALOMA-2 trial, where tumor samples came from a mixture of primary or metastatic biopsies and the origin of tissue samples were not recorded (Table 2).
- Composite signature scores were computed by weighting each component gene by its average coefficient across bootstrap models.
- Composite scores for patients in the palbociclib arm and the placebo arm were dichotomized by median into high and low scoring groups.
- FIG. 5 shows the 9-gene exploratory signature, where bar length (top x-axis) reflects the proportion of 1000 bootstrap models containing the gene while purple points (bottom x-axis) reflect average non-zero coefficient across bootstrap models.
- CCNE1 Five of the genes (CCNE1 , CPT1A, ERG, LRP5, and TMPRSS2) appear to be associated with relative resistance to palbociclib, while the remaining four genes (MAP3K1 , SLC38A2, STC2, and TFDP2) appear to be associated with relative sensitivity to palbociclib.
- the 9-gene signature was further applied to the full PALOMA-3 data set, comprising 302 patient samples, including both primary and metastatic samples (Table 1 ).
- Composite signature scores were computed by weighting each component gene by its average coefficient across bootstrap models.
- the exploratory 9-gene signature was then tested on the independent metastatic breast cancer cohort from the PALOMA-2 clinical trial comprising 454 patient samples. Despite marked cross-cohort differences in combination partner (fulvestrant vs. letrozole) and clinical setting (endocrine resistant vs. treatment naive), the 9-gene signature remained predictive of palbociclib efficacy (FIG. 5(C)).
- Non-Negative Matrix Factorization was applied to the 302 primary and metastatic samples from the PALOMA-3 trial.
- the virtual microdissection identified fourteen molecular factors that characterize biological processes differentiating baseline tumor samples of this cohort.
- One of the fourteen factors unambiguously represented the liver specific genes. Since some samples were from biopsies of liver metastases, a liver-specific factor was used to identify samples with significant liver component as described in the methods below. Using this approach, 30 samples with significant liver content were identified and excluded from further analysis.
- Example 2 The Elastic Net method described in Example 2 was then reapplied to the remaining dataset after removing potential outliers for liver factors.
- This analysis provided a refined 13-gene signature with higher bootstrapping frequencies than the previous 9-gene signature.
- a composite signature score was computed by weighting each component gene by its average coefficient across bootstrap models.
- the 13 selected genes included LRP5, TRIB1 , TFDP2, MAP3K1 , ERG, TMPRSS2, BUB1 B, ABCB11 , MT1X, CCNE1 , CDKN2D, STC2 and COL11A1.
- FIG. 7 shows the 13-gene exploratory signature, where bar length (top x- axis) reflects the proportion of 1000 bootstrap models containing the gene while purple points (bottom x-axis) reflect average non-zero coefficient across bootstrap models.
- the 13-gene signature was further applied to the PALOMA-3 data set after removing samples with significant liver content, comprising 272 patient samples, including both primary and metastatic samples (Table 1 ).
- Composite signature scores were computed by weighting each component gene by its average coefficient across bootstrap models.
- NMF was applied to HTG gene expression of 302 patient samples from PALOMA-3.
- the NMF algorithm factorizes the gene expression matrix V of g genes and s samples into two non-negative matrices of k factors: gene factor matrix W of weights of n gene for k factors and sample factor matrix H of weights of m sample for k factors. W represents the expression pattern of the k parts and H represents the respective contribution of k parts in each sample or bulk tumor 1 .
- the expression values for each patient were rank transformed. NMF was then performed on rank transformed gene expression matrix V paloma-3 with the R package “NMF” which used the “brunet” algorithm 2 . 30 runs of NMF were performed and the factorization that achieved the lowest approximation error for subsequent analyses was chosen.
- GSEA Gene Set Enrichment Analysis
- a predictive biomarker refers to an NMF factor whose magnitude significantly correlates with palbociclib treatment outcome, manifest in the p-value associated with the factor x arm interaction term coefficient in the Cox PH model.
- a prognostic biomarker refers to an NMF factor whose magnitude significantly correlates with overall clinical outcome in a cohort, regardless of treatment arms (adjusting for treatment arms in a Cox PH model).
- Predictive biomarkers were obtained from the interactive Cox PH model with PFS as the end-point eq. 1 : h(t
- x) h 0 (t) expCpiX-t + p 2 x 2 + p 3 XiX 2 ) eq. 1 where xi is a dichotomous variable with 1 representing palbociclib plus fulvestrant treatment and 0 representing control treatment and X2 is a dichotomous variable with values 1 or 0 indicating whether the biomarker (NMF factor) is above median value or below. Interaction p-value is the p value associated with p 3 .
- the EdgeSeq Oncology Biomarker Panel (HTG Molecular Diagnostics, Inc) was used for mRNA profiling of 2,534 cancer-related genes.
- the EdgeSeq system uses targeted capture sequencing to quantitate mRNA expression levels of gene targets in FFPE tissues.
- the first section of breast cancer FFPE tissue was stained with hematoxylin and eosin (H&E).
- H&E hematoxylin and eosin
- a board-certified pathologist assessed the tumor cell content and tissue necrosis. Tumor content was estimated based on the number of malignant cells as a percentage of all cells (i.e. , malignant plus normal cells in the tissue section).
- the acceptance criterion for analysis was set at >70% of tumor content. Necrosis was assessed based on the percentage of necrotic tissue within the total tissue area.
- the acceptance criterion for analysis was set at ⁇ 20% necrosis. Macrodissection was performed on the tissue sections if the tumor content was ⁇ 70% or if necrosis was >20%. Sample preparation was conducted following laboratory processes and manufactory protocols. Sequencing was performed on an Illumina NextSeq 500 Sequencer (Illumina, Inc.).
- probe counts were transformed into Iog2 counts per million. Expression values were quantile normalized. HTG Molecular Diagnostics, Inc., was blinded to patient information and clinical outcomes.
- Alternative approaches to determine gene expression levels include may comprise performing RT-PCR, a hybridization, transcriptome analysis, RNAseq, a Northern blot, a Western blot, or an ELISA.
- assaying the expression level of genes in a gene signature can comprise performing an array hybridization.
- transcriptome analysis may comprise obtaining sequence information of expressed RNA molecules.
Landscapes
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Engineering & Computer Science (AREA)
- Immunology (AREA)
- Pathology (AREA)
- Analytical Chemistry (AREA)
- Zoology (AREA)
- Genetics & Genomics (AREA)
- Wood Science & Technology (AREA)
- Physics & Mathematics (AREA)
- Biotechnology (AREA)
- Microbiology (AREA)
- Molecular Biology (AREA)
- Hospice & Palliative Care (AREA)
- Biophysics (AREA)
- Oncology (AREA)
- Biochemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
This invention relates to biomarkers, gene signatures, and methods for the selection and treatment of patients having HR+, HER2- breast cancer who are likely to benefit from administration of a CDK4/6 inhibitor such as palbociclib.
Description
BIOMARKERS OF TUMOR RESPONSE TO CDK4/6 INHIBITORS
BACKGROUND OF THE INVENTION
Field of the Invention
The invention relates to biomarkers of tumor response, gene signatures, and methods for the selection and treatment of subjects having hormone receptor positive (HR+), human epidermal growth factor receptor 2 negative (HER2-) breast cancer who are likely to benefit from administration of a CDK4/6 inhibitor.
Description of Related Art
The cell cycle mechanism has been exploited by therapeutic agents that impair disparate aspects of cell division, including DNA synthesis, microtubule assembly, and DNA damage. More recently, targeted approaches, including CDK4/6 inhibitors such as palbociclib, have been developed. In clinical trials, palbociclib was found to significantly improve progression free survival (PFS) in hormone receptor positive (HR+), human epidermal growth factor receptor 2 negative (HER2-) advanced or metastatic breast cancer when combined with endocrine therapy. Palbociclib, ribociclib and abemaciclib have been approved for treatment of hormone receptor (HR)-positive, human epidermal growth factor receptor 2 negative (HER2-) advanced or metastatic breast cancer in combination with aromatase inhibitors, such as letrozole, in a first line setting and with fulvestrant in second or later lines of therapy in certain patients. (O’Leary et al. Treating cancer with selective CDK4/6 inhibitors. Nature Reviews (2016) 13:417-430).
While disease progression has been found to be significantly delayed by the addition of a CDK4/6 inhibitor to an anti-hormonal therapy, many patients ultimately progress after a period of stable disease/remission or do not respond to the combination treatment, representing acquired and inherent resistance, respectively. Resistance to anti-hormonal therapies have been partially attributed to non-synonymous mutations in the ligand binding domain of the ESR1 gene resulting in ligand independent activity of the mature transcription factor. See Robinson et al., Activating ESR1 mutations in hormone-resistant metastatic breast cancer. Nat Genet 45:1446-1451 , 2013; Toy et al., ESR1 ligand-binding domain mutations in hormone-resistant breast cancer. Nat Genet 45:1439-1445, 2013. Other resistance mechanisms observed for anti-hormonal therapy in breast cancer include upregulation of HER or FGFR family signaling and induction of the PI3K/AKT/MTOR pathway. See Giltnane et al., Genomic profiling of ER(+) breast cancers after short-term estrogen suppression reveals alterations associated with
endocrine resistance. Sci Transl Med 9, 2017; Higgins and Baselga, Targeted therapies for breast cancer. J Clin Invest 121 :3797-3803, 2011.
A univariate analysis of 2,534 cancer-related genes was conducted in archival tumor samples from the PALOMA-3 trial (ClinicalTrials.gov identifier: NCT01942135), using the EdgeSeq Oncology Biomarker Panel (OBP) panel (HTG Molecular Diagnostics) for mRNA profiling. The association of gene expression with the effect of palbociclib on progression-free survival (PFS) was evaluated using Cox proportional hazards (Cox PH) regression analysis, with gene expression as a continuous variable or dichotomized by median. The analysis identified cyclin E1 (CCNE1) mRNA expression as strongly associated with palbociclib efficacy in previously treated HR+, HER2- metastatic breast cancer patients who failed on prior endocrine therapy. Patients with high baseline CCNE1 mRNA expression were less responsive to the combination of palbociclib plus fulvestrant relative to patients with low CCNE1 mRNA expression, implicating CDK2 activity in determining relative resistance to palbociclib in the second- line clinical setting. See Turner et al., Cyclin E1 Expression and Palbociclib Efficacy in Previously Treated Hormone Receptor-Positive Metastatic Breast Cancer. J. Clin. Oncol. 37:1169-1178, 2019.
However, CCNE1 mRNA expression level alone could not significantly stratify patient response from the PALOMA-2 trial (ClinicalTrials.gov identifier: NCT01740427), in patients with estrogen receptor positive (ER+), HER2- advanced breast cancer who had not received prior endocrine therapy for their advanced disease. These first-line patients were randomly assigned to receive palbociclib plus letrozole or placebo plus letrozole. See Finn et al., Biomarker Analyses of Response to Cyclin-Dependent Kinase 4/6 Inhibition and Endocrine Therapy in Women with Treatment-Naive Metastatic Breast Cancer. Clin Cancer Res 26: 110-121 , 2020.
The identification of multivariate gene expression signatures predictive of response to treatment with CDK4/6 inhibitors in HR+, HER2- breast cancer across multiple lines of therapy are needed.
BRIEF SUMMARY OF THE INVENTION
This invention relates to biomarkers of tumor response, gene signatures, and methods for the selection and treatment of subjects having hormone receptor positive (HR+), human epidermal growth factor receptor 2 negative (HER2-) breast cancer who are likely to benefit from administration of a CDK4/6 inhibitor.
In one aspect, the invention provides a method of treating hormone receptorpositive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer in a subject in need thereof, comprising:
(a) determining the expression level for each gene in a gene signature in a tumor sample from the subject;
(b) computing a composite score for the gene signature as a weighted sum of the expression levels for each gene in the gene signature;
(c) selecting the subject for treatment with a cyclin dependent kinase 4/6 (CDK 4/6) inhibitor and an endocrine therapeutic agent when the composite score is less than the median of a reference composite score computed for the gene signature in a reference breast cancer population; and
(d) administering to the selected subject an amount of a CDK4/6 inhibitor and an amount of an endocrine therapeutic agent, wherein the amounts together are effective to treat cancer; wherein the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
In another aspect, the invention provides a method of selecting a subject having hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer for treatment with a CDK4/6 inhibitor and an endocrine therapeutic agent, comprising:
(a) determining the expression level for each gene in a gene signature in a tumor sample from the subject;
(b) computing a composite score for the gene signature as a weighted sum of the expression levels for each gene in the gene signature; and
(c) selecting the subject for treatment when the composite score is less than the median of a reference composite score computed for the gene signature in a reference breast cancer population; wherein the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
In another aspect, the invention provides a method of predicting whether a subject having hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer will respond to treatment with a CDK4/6 inhibitor and an endocrine therapeutic agent, comprising:
(a) determining the expression level for each gene in a gene signature in a tumor sample from the subject;
(b) computing a composite score for the gene signature as a weighted sum of the expression levels for each gene in the gene signature;
(c) comparing the composite score to a reference composite score computed for the gene signature in a reference breast cancer population; and
(d) predicting the subject will respond to treatment when the composite score is less than the median of the reference composite score; wherein the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
In preferred embodiments of each of the aspects and embodiments described herein, the CDK4/6 inhibitor is palbociclib, or a pharmaceutically acceptable salt thereof.
In some embodiments of each of the aspects herein, the endocrine therapeutic agent is selected from the group consisting of an aromatase inhibitor, a selective estrogen receptor degrader (SERD), and a selective estrogen receptor modulator (SERM). In some embodiments, the endocrine therapeutic agent is selected from the group consisting of letrozole, anastrozole, exemestane and fulvestrant.
Each of the aspects and embodiments of the present invention described herein may be combined with one or more other embodiments of the present invention which is not inconsistent with the embodiment(s) with which it is combined.
BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWINGS
FIG. 1 shows an 11 -gene exploratory signature from PALOMA-3 using the elastic net algorithm. Bar length (top x-axis) reflects the proportion of 100 bootstrap models containing the gene, while diamond points (bottom x-axis) reflect the average non-zero coefficient across bootstrap models.
FIG. 2 shows a hierarchical clustering heatmap of the 11 -gene exploratory signature and progression-free survival (PFS). Shown are patients treated with palbociclib + fulvestrant (A) or placebo + fulvestrant (B), in each case sorted by PFS excluding those with a follow-up time of <12 months and no event. Clustering was conducted using 1 -Pearson correlation as distance metric and average linkage.
FIG. 3 shows the association between the 11 -gene exploratory signature and clinical benefit (PFS) in metastatic samples from PALOMA-3 (A) and in the full data set from PALOMA-3 (B). Patients were dichotomized by median signature score into low
(dashed line) and high (solid line) score groups. Also shown are hazard ratios for each gene in the 11 -gene exploratory signature using the full data set from PALOMA-3 (C).
FIG. 4 shows the association between the 11 -gene exploratory signature and clinical benefit (PFS) from the addition of palbociclib in an independent metastatic breast cancer cohort from PALOMA-2. Patients were dichotomized by median signature score into low (dashed line) and high (solid line) score groups.
FIG. 5 shows a 9-gene exploratory signature from PALOMA-3 using the elastic net algorithm. Bar length (top x-axis) reflects the proportion of 1 ,000 bootstrap models containing the gene, while circular points (bottom x-axis) reflect the average non-zero coefficient across bootstrap models.
FIG. 6 shows the association between the 9-gene exploratory signature and clinical benefit (PFS) in the full data set from PALOMA-3 (A) and in an independent metastatic breast cancer cohort from PALOMA-2 (B). Patients were dichotomized by median signature score into low (dashed line) and high (solid line) score groups.
FIG. 7 shows a 13-gene exploratory signature from PALOMA-3 using the elastic net algorithm. Bar length (top x-axis) reflects the proportion of 1 ,000 bootstrap models containing the gene, while circular points (bottom x-axis) reflect the average non-zero coefficient across bootstrap models.
FIG. 8 shows the association between the 13-gene exploratory signature and clinical benefit (PFS) in the full data set from PALOMA-3 after filtering out outliers from NMF liver factor (A), and in an independent metastatic breast cancer cohort from PALOMA-2 (B). Patients were dichotomized by median signature score into low (dashed line) and high (solid line) score groups.
FIG. 9 shows identification of samples with significant liver component in PALOMA-3 data set before (A) and after filtering out outliers from NMF liver factor (B).
DETAILED DESCRIPTION OF THE INVENTION
The present invention may be understood more readily by reference to the following detailed description of the embodiments of the invention and the Examples included herein. It is to be understood that the terminology used herein is for the purpose of describing specific embodiments only and is not intended to be limiting. It is further to be understood that unless specifically defined herein, the terminology used herein is to be given its traditional meaning as known in the relevant art.
The invention described herein may be suitably practiced in the absence of any element(s) not specifically disclosed herein. Thus, for example, in each instance herein
any of the terms "comprising", "consisting essentially of", and "consisting of" may be replaced with either of the other two terms.
As used herein, the singular form "a", "an", and "the" include plural references unless indicated otherwise. For example, "a" substituent includes one or more substituents.
The term "about" means having a value falling within an accepted standard of error of the mean, when considered by one of ordinary skill in the art, typically such as ± 10%.
The term “administration” or “treatment” as it applies to an animal, human, experimental subject, cell, tissue, organ or biological fluid, refers to contact of an exogenous pharmaceutical, therapeutic or diagnostic agent, or composition, to the animal, human, experimental subject, cell, tissue, organ or biological fluid. Treatment of a cell encompasses contact of a reagent to the cell, as well as contact of a reagent to a fluid, where the fluid is in contact with the cell. “Administration” and “treatment” may also relate to in vitro and ex vivo treatment, e.g., of a cell, by a reagent, diagnostic, binding compound, or by another cell.
The term “cancer”, “cancerous”, “malignant” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. As used herein “cancer” refers to any malignant and/or invasive growth or tumor caused by abnormal cell growth. As used herein “cancer” refers to solid tumors named for the type of cells that form them, cancer of blood, bone marrow, or the lymphatic system. Examples of solid tumors include but not limited to sarcomas and carcinomas. Examples of cancers of the blood include but not limited to leukemias, lymphomas and myeloma. The term “cancer” includes but is not limited to a primary cancer that originates at a specific site in the body, a metastatic cancer that has spread from the place in which it started to other parts of the body, a recurrence from the original primary cancer after remission, and a second primary cancer that is a new primary cancer in a person with a history of previous cancer of a different type from latter one. A subject may be identified as having de novo metastatic disease or after progression from an earlier-identified cancer.
The term “patient” or “subject” refer to any single subject for which therapy is desired or that is participating in a clinical trial, epidemiological study or used as a control, including humans and mammalian veterinary patients such as cattle, horses, dogs and cats. In preferred embodiments, the subject is a human.
In some embodiments, the subject is a postmenopausal woman or a man. In other embodiments, the subject is a pre- or perimenopausal woman treated with a luteinizing hormone releasing hormone (LNRH) agonist, such as goserelin, so that their ovarian function is suppressed. Subjects may be treatment naive (i.e., the subject has not received prior treatment for advanced disease if metastatic, or for early disease if diagnosed with early breast cancer) or may have received one or more prior lines of treatment (i.e., in second or later line settings), such as one or more endocrine therapeutic agents or chemotherapeutic agents. In some embodiments, the subject is treated with an aromatase inhibitor as their initial endocrine therapeutic agent (i.e., as initial endocrine based therapy). In other embodiments, the subject has disease progression on or after treatment with an endocrine therapeutic agent in an adjuvant or metastatic setting.
The term “treat” or “treating” a cancer as used herein means to administer a combination therapy comprising a CDK4/6 inhibitor and an endocrine therapeutic agent, according to the present invention to a subject having cancer, or diagnosed with cancer, to achieve at least one positive therapeutic effect, such as, for example, reduced number of cancer cells, reduced tumor size, reduced rate of cancer cell infiltration into peripheral organs, or reduced rate of tumor metastases or tumor growth, reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition. The term "treatment", as used herein, unless otherwise indicated, refers to the act of treating as "treating" is defined immediately above. The term “treating” also includes adjuvant and neo-adjuvant treatment of a subject.
A “reference breast cancer population” refers to a population of individuals having HR+, HER2- breast cancer of which the subject is a member. For example, the reference breast cancer population could refer to a cohort of patients in a clinical trial enrolling the same or similar stage(s) of breast cancer as the subject. Alternatively, the reference breast cancer population could refer to a relevant population of individuals who have been successfully treated with a CDK4/6 inhibitor and endocrine therapy. For example, the reference breast cancer population could refer to the cohorts from the PALOMA-2 or PALOMA-3 studies for advanced or metastatic breast cancer, or to a clinical trial cohorts from a relevant early breast cancer trial.
A “control population” refers to a population of individuals who do not have cancer but are otherwise matched to the subject. The skilled person will be able to select an appropriate control population to provide the requisite reference value.
For the purposes of this invention, beneficial or desired clinical results include, but are not limited to, one or more of the following: reducing the proliferation of (or destroying) neoplastic or cancerous cell; inhibiting metastasis or neoplastic cells; shrinking or decreasing the size of a tumor; remission of the cancer; decreasing symptoms resulting from the cancer; increasing the quality of life of those suffering from the cancer; decreasing the dose of other medications required to treat the cancer; delaying the progression of the cancer; curing the cancer; overcoming one or more resistance mechanisms of the cancer; and/or prolonging survival of patients the cancer. Positive therapeutic effects in cancer can be measured in a number of ways (see, for example, W. A. Weber, Assessing tumor response to therapy, J. Nucl. Med. 50 Suppl. 1 : 1 S-1 OS (2009). For example, with respect to tumor growth inhibition (T/C), according to the National Cancer Institute (NCI) standards, a T/C less than or equal to 42% is the minimum level of anti-tumor activity. A T/C <10% is considered a high anti-tumor activity level, with T/C (%) = median tumor volume of the treated I median tumor volume of the control x 100.
In some embodiments, the treatment achieved by a combination of the invention is defined by reference to any of the following: partial response (PR), complete response (CR), overall response (OR), progression free survival (PFS), disease free survival (DFS), invasive disease free survival (iDFS), and overall survival (OS). PFS, also referred to as “Time to Tumor Progression” indicates the length of time during and after treatment that the cancer does not grow and includes the amount of time patients have experienced a CR or PR, as well as the amount of time patients have experienced stable disease (SD). DFS refers to the length of time during and after treatment that the patient remains free of disease. iDFS is defined according to Hudis (J Clin Oncol 2007) as the time between randomization and first event (e.g., ipsi- or contralateral invasive inbreast or loco-regional recurrence, distant recurrence, death from breast cancer, death from non-breast cancer cause, death from unknown cause, invasive contralateral breast cancer, second primary invasive cancer (non-breast)). OS refers to a prolongation in life expectancy as compared to naive or untreated subjects or patients. In some embodiments, response to a combination of the invention is any of PR, CR, PFS, DFS, OR or OS that is assessed using Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 response criteria.
The treatment regimen that is effective to treat a cancer patient may vary according to factors such as the disease state, age, and weight of the patient, and the ability of the therapy to elicit an anti-cancer response in the subject. While the aspects
of the present invention may not be effective in achieving a positive therapeutic effect in every subject, it should do so in a statistically significant number of subjects as determined by any statistical test known in the art such as the Student’s t-test, the chi2- test the ll-test according to Mann and Whitney, the Kruskal-Wallis test (H-test), Jonckheere-Terpstrat-testy and the Wilcon on-test.
The terms “treatment regimen”, “dosing protocol” and “dosing regimen” may be used interchangeably to refer to the dose and timing of administration of each therapeutic agent administered according to the invention herein.
“Ameliorating” means a lessening or improvement of one or more symptoms upon treatment with a combination described herein, as compared to not administering the combination. “Ameliorating” also includes shortening or reduction in duration of a symptom.
As used herein, an “effective dosage” or “effective amount” of drug, compound or pharmaceutical composition is an amount sufficient to affect any one or more beneficial or desired, including biochemical, histological and I or behavioral symptoms, of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease. For therapeutic use, an effective amount refers to that amount which will relieve to some extent one or more of the symptoms of the disorder being treated. In reference to the treatment of cancer, a therapeutically effective amount refers to that amount which has the effect of (1 ) reducing the size of the tumor, (2) inhibiting (that is, slowing to some extent, preferably stopping) tumor metastasis, (3) inhibiting to some extent (that is, slowing to some extent, preferably stopping) tumor growth or tumor invasiveness, (4) relieving to some extent (or, preferably, eliminating) one or more signs or symptoms associated with the cancer, (5) decreasing the dose of other medications required to treat the disease, and/or (6) enhancing the effect of another medication, and/or (7) delaying the progression of the disease in a patient.
An effective dosage can be administered in one or more administrations. For the purposes of this invention, an effective dosage of drug, compound, or pharmaceutical composition is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly. As is understood in the clinical context, an effective dosage of drug, compound or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound or pharmaceutical composition.
“Tumor” as it applies to a subject diagnosed with, or suspected of having, a cancer refers to a malignant or potentially malignant neoplasm or tissue mass of any size and includes primary tumors and secondary neoplasms. A solid tumor is an
abnormal growth or mass of tissue that usually does not contain cysts or liquid areas. Examples of solid tumors are sarcomas, carcinomas, and lymphomas. Leukaemia’s (cancers of the blood) generally do not form solid tumors (National Cancer Institute, Dictionary of Cancer Terms).
“Tumor burden” or “tumor load’, refers to the total amount of tumorous material distributed throughout the body. Tumor burden refers to the total number of cancer cells or the total size of tumor(s), throughout the body, including lymph nodes and bone marrow. Tumor burden can be determined by a variety of methods known in the art, such as, e.g., using calipers, or while in the body using imaging techniques, e.g., ultrasound, bone scan, computed tomography (CT), or magnetic resonance imaging (MRI) scans.
The term “tumor size” refers to the total size of the tumor which can be measured as the length and width of a tumor. Tumor size may be determined by a variety of methods known in the art, such as, e.g., by measuring the dimensions of tumor(s) upon removal from the subject, e.g., using calipers, or while in the body using imaging techniques, e.g., bone scan, ultrasound, CR or MRI scans.
The PALOMA-3 trial (ClinicalTrials.gov identifier: NCT01942135) was a Phase III double-blind study in women aged 18 years or older with HR+, HER2- metastatic breast cancer, who had progressed after previous endocrine therapy. 521 endocrine-pretreated patients were randomly assigned to receive palbociclib plus fulvestrant or placebo plus fulvestrant (palbociclib arm, 347 patients; placebo arm, 174 patients). The primary endpoint was PFS.
The PALOMA-2 trial (ClinicalTrials.gov identifier: NCT01740427) was a Phase III double-blind study in post-menopausal women with ER+, HER2- advanced breast cancer, who had not received prior treatment for advanced disease. 666 patients were randomly assigned to receive palbociclib plus letrozole or placebo plus letrozole (palbociclib arm, 444 patients; placebo arm, 222 patients). The primary endpoint was PFS.
The patient cohorts from the PALOMA-2 and PALOMA-3 trials, and their associated molecular profiles, were utilized to discover multivariate gene expression signatures capable of predicting strong versus weak response to palbociclib in combination with endocrine therapy, regardless of the line of therapy. An aggregated consideration of the features within such signatures revealed intrinsic resistance mechanisms to CDK4/6 inhibition that are common among HR+, HER2- breast cancer patients.
Baseline tumor tissue samples from the PALOMA-3 trial were profiled using the EdgeSeq Oncology Biomarker Panel (OBP) panel (HTG Molecular Diagnostics), with 302 tumor samples evaluable for analysis. Anonymized gene expression datasets from the PALOMA-3 trial have been deposited in NCBI’s Gene Expression Omnibus (Turner et al., Cyclin E1 Expression and Palbociclib Efficacy in Previously Treated Hormone Receptor-Positive Metastatic Breast Cancer, Journal of Clinical Oncology 37:14, 1169- 1178, 2019) and are accessible through GEO Series accession number GSE128500 (https://www.ncbi. nlm.nih.gov/geo/query/acc.cgi?acc=GSE128500).
The number of evaluable samples from each arm of the PALOMA-3 trial, as well as the sites of tumor tissue collection, are summarized in Table 1 .
Baseline tumor tissue samples from the PALOMA-2 trial were profiled using the EdgeSeq Oncology Biomarker Panel (OBP) panel (HTG Molecular Diagnostics), with 455 tumor samples evaluable for analysis. Anonymized gene expression datasets from the PALOMA-2 trial have been deposited in NCBI’s Gene Expression Omnibus (Finn et al., Biomarker Analyses of Response to Cyclin-Dependent Kinase 4/6 Inhibition and Endocrine Therapy in Women with Treatment-Naive Metastatic Breast Cancer, Clin. Cancer Res. 26:1 , 110-121 , 2020) and are accessible through GEO Series accession number GSE133394 (https://www.ncbi. nlm.nih.gov/geo/query/acc.cgi?acc=GSE133394).
The number of evaluable samples from each arm of the PALOMA-2 trial are summarized in Table 2 below. The tumor samples came from a mixture of primary or metastatic biopsies and the sites of tumor tissue collection were not recorded.
Two distinct techniques, Elastic Net (EN) and Non-Negative Matrix Factorization (NMF), were employed to derive predictive multi-gene expression signatures associated with relative benefit from treatment with palbociclib in combination with endocrine therapy in a first-line or second-line setting.
The elastic net technique was employed to derive exploratory, predictive multigene expression features using patient samples from the PALOMA-3 study, which could then be tested for predictive power using the data set from the PALOMA-2 study. The machine-learning based elastic net regularization algorithm was applied. Clinical outcome association was performed by Cox PH regression using a number of bootstraps (e.g., 100 or 1 ,000 bootstraps) to provide a list of complementary gene expression features that comprise a model from each round. Genes were selected based on frequency observed in the bootstraps. Typically, at least 100 bootstraps are performed, and frequently 1 ,000 bootstraps are performed. In some methods, bootstraps are performed until the results have stabilized. See Goodhue et al., Does PLS have advantages for small sample size or non-normal data? MIS Quarterly, 36(3):981-1001 , 2012; Efron et al., (2004). Resampling methods of estimation. In N.J. Smelser, & P.B. Baltes (Eds.). International Encyclopedia of the Social & Behavioral Sciences (pp. 13216-13220). New York, NY: Elsevier.
The elastic net method linearly combines the L1 and L2 penalties of the lasso (least absolute shrinkage and selection operator) and ridge regression analysis methods. Lasso regression analysis performs variable selection and regularization using the penalty, which imposes sparsity among the coefficients to increase the accuracy and interpretability of the fitted model. The ridge regression method uses regularization to limit coefficient vector size. Elastic net regularization reduces overfitting by adding a complexity penalty that is part L1 and part L2 as a compromise between the ridge regression penalty (a = 0) and the Lasso penalty (a = 1 ):
It was previously demonstrated that the collection of tissue closer to treatment time facilitated the identification of predictive markers (Turner et al., 2019). Therefore,
model derivation efforts were initially focused on metastatic tumor samples collected from patients in the palbociclib treatment arm of the PALOMA-3 cohort.
The machine-learning based elastic net regularization algorithm was applied, with clinical outcome association by Cox PH regression. The elastic net process involves:
(1 ) cross validation to select an optimal alpha (a) and lambda (X) pair with minimum error;
(2) with the best a and X selected, perform a number of bootstraps to obtain a list of complementary gene expression features that comprise a model from each round;
(3) select genes based on frequency observed in the bootstraps; and
(4) compute a composite signature score by weighting expression of signature genes by their coefficients.
Typically, at least 100 bootstraps are used, and frequently 1 ,000 bootstraps are used. In some methods, additional bootstraps are performed until the results have stabilized.
Gene Signatures
The elastic net method was applied to identify multivariate gene signatures capable of predicting palbociclib efficacy in HR+, HER2- breast cancer. Gene signatures were derived using expression data obtained from patient samples in the PALOMA-3 and PALOMA-2 trials using the EdgeSeq Oncology Biomarker Panel (OBP) panel (HTG Molecular Diagnostics).
Three multi-gene signatures were derived, encompassing different combinations of the following genes: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
11-Gene Signature
As described in Example 1 , an 11 -gene signature was discovered that is predictive of palbociclib efficacy in both endocrine-refractory and advanced treatment naive HR+, HER2- advanced or metastatic breast cancer.
The 11 -gene signature was discovered using 92 metastatic samples from the palbociclib plus fulvestrant treatment arm of the PALOMA-3 trial in HR+, HER2- endocrine pretreated metastatic breast cancer patients.
The 11 genes identified, CCNE1 , CDH15, CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1 , were found to have non-zero coefficients in at least 40% of the models (FIG. 1 ). Six of the genes (CCNE1 , CPT1A, ERG, LRP5, TMPRSS2, and TRIB1 ) appear to be associated with relative resistance to palbociclib, while the remaining five genes (CDH15, MAP3K1 , SLC38A2, STC2, and TFDP2) appear to be associated with relative sensitivity to palbociclib (FIG. 1 ).
Hierarchical clustering was conducted using 1 -Pearson correlation as distance metric and average linkage. FIG. 2 shows a hierarchical clustering heatmap of the 11- gene exploratory signature and progression-free survival (PFS). Shown are patients treated with palbociclib + fulvestrant in FIG. 2(A) or placebo + fulvestrant FIG. 2(B), in each case sorted by PFS excluding those with an insufficient follow-up time of <12 months and no event.
The 11 -gene signature was applied to the 142 metastatic patient samples from both palbociclib treatment and placebo arms of the PALOMA-3 trial (Table 1 ). FIG. 3(A) shows the association between the 11 -gene exploratory signature and clinical benefit (PFS) as a Kaplan-Meier plot for the metastatic samples from PALOMA-3. The gene signature was further applied to the full cohort from the PALOMA-3 trial, including 302 primary and metastatic samples from the palbociclib treatment and placebo arms (Table 1 ). FIG. 3(B) shows the association between the 11 -gene exploratory signature and clinical benefit (PFS) as a Kaplan-Meier plot for the full PALOMA-3 data set, including both metastatic and primary tumor samples. Patients were dichotomized by median
signature score into low (dashed line) and high (solid line) score groups. Also shown are hazard ratios for each gene in the 11 -gene exploratory signature using the full data set from PALOMA-3 in FIG. 3(C).
The ability of the gene signature to predict palbociclib efficacy across lines of therapy was demonstrated by application to an independent metastatic breast cancer cohort in the PALOMA-2 trial, which included ER+, HER2- advanced breast cancer patients who were treatment naive. Association between the 11 -gene exploratory signature and clinical benefit from the addition of palbociclib to letrozole in 454 samples from the palbociclib treatment and placebo plus letrozole control arms of the PALOMA-2 trial (Table 2) is shown in the Kaplan-Meier plot in FIG. 4. Patients were dichotomized by median signature score into low (dashed line) and high (solid line) score groups. The signature was also predictive based on continuous variable analysis (interaction P = 0.016 and P = 0.0031 before and after adjusting for baseline clinicopathologic characteristics respectively).
Some of the genes identified in the 11 -gene signature have a clear mechanistic link to the cell cycle, such as transcription factor DP-2 (TFDP2) which forms a heterodimer with the E2F transcription factors in regulating cell cycle progression from G1 to S phase (PMID: 8755520). Others could imply novel and previously unappreciated biology. For example, two of the genes, TMPRSS2 and ERG, are known fusion partners found in approximately half of prostate cancer patients, resulting in androgen receptor (AR) induced over-expression (PMID: 16254181 and 18283340). 9-Gene Signature
As described in Example 2, an 9-gene signature was discovered that is predictive of palbociclib efficacy in both endocrine-pretreated (second or later line) and treatment naive (first line) HR+, HER2- advanced or metastatic breast cancer.
The 9-gene signature was discovered using the 92 metastatic samples from the palbociclib plus fulvestrant treatment arm of the PALOMA-3 trial in HR+, HER2- endocrine pretreated metastatic breast cancer patients. Nine genes were selected using a C-lndex to build an optimal predictive model. Table 4
The 9 genes identified, CCNE1 , CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, and TMPRSS2, were found to have non-zero coefficients in at least 10% of the bootstrap models (FIG. 5). Five of the genes (CCNE1 , CPT1A, ERG, LRP5, and TMPRSS2) appear to be associated with relative resistance to palbociclib. The remaining four genes (MAP3K1 , SLC38A2, STC2, and TFDP2) appear to be associated with relative sensitivity to palbociclib (FIG. 5).
FIG. 6(A) shows the association between the 9-gene exploratory signature and clinical benefit (PFS) as a Kaplan-Meier plot for the full PALOMA-3 data set, including 302 primary and metastatic samples from the palbociclib treatment and placebo arms (Table 1 ).
The ability of the gene signature to predict palbociclib efficacy across lines of therapy was demonstrated by application to and independent metastatic breast cancer cohort in the PALOMA-2 trial, which included ER+, HER2- advanced breast cancer patients who were treatment naive. Association between the 9-gene exploratory signature and clinical benefit from the combination of palbociclib plus letrozole in the PALOMA-2 trial is shown in the Kaplan-Meier plot in FIG. 6(B) ( interaction P=0.036).
Despite marked cross-cohort differences, including both the combination partner (fulvestrant versus letrozole) and the clinical setting (endocrine resistant vs. treatment naive), the 9-gene signature remained predictive of palbociclib efficacy in the PALOMA- 2 patient samples.
The 9-gene signature identified some of the same genes as in the 11 -gene signature, including transcription factor DP-2 (TFDP2) and the genes TMPRSS2 and ERG associated with AR induced over-expression.
13-Gene Signature
As described in Example 3, a 13-gene signature was discovered using the PALOMA-3 samples and then validated using the PALOMA-2 samples.
To further investigate multigene expression features that differentiate among the 302 primary and metastatic samples from the PALOMA-3 trial, a virtual microdissection
approach, Non-Negative Matrix Factorization (NMF), was applied. NMF was conducted as described in Moffitt et al., Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat Genet. 2015;47(10):1168-1178.). The NMF algorithm factorizes the gene expression matrix into two non-negative matrices: a gene factor matrix and a sample factor. Due to its constraints for all matrices to be non-negative, NMF is particularly suited to find biological processes that underlie expression data.
The virtual microdissection identified fourteen molecular factors that characterize biological processes differentiating baseline tumor samples of this cohort. One of the fourteen factors unambiguously represented the liver specific genes. Since a number of samples were from biopsies of liver metastases, a liver-specific factor was used to identify samples with significant liver component as described herein. Using this approach, 30 samples with significant liver content were identified and excluded from further analysis.
The Elastic Net methods described above were then reapplied to the remaining dataset after removing potential outliers for liver factors. This analysis provided a refined 13-gene signature (a = 1 , X = 0.204) with higher bootstrapping frequencies than the previous 9-gene signature. Thirteen genes were selected using a C-lndex to build an optimal predictive model.
Table 5
FIG. 7 shows the 13-gene exploratory signature, where bar length (top x-axis) reflects the proportion of 1000 bootstrap models containing the gene while purple points (bottom x-axis) reflect average non-zero coefficient across bootstrap models. The 13 genes identified, BUB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, TRIB1, ABCB11 , COL11A1 , MAP3K1 , STC2 and TFDP2, were found to have non-zero coefficients in the bootstrap models. Eight of the genes (BLIB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, and TRIB1 ) appear to be associated with relative resistance to palbociclib, while the remaining five genes (ABCB11 , COL11 A1 , MAP3K1 , STC2 and TFDP2) appear to be associated with relative sensitivity to palbociclib (FIG. 7).
The 13-gene signature was applied to the full cohort from the PALOMA-3 trial, including 302 primary and metastatic samples from the palbociclib treatment and placebo arms (Table 1). Data are provided in the Kaplan-Meier plots. FIG. 8(A) shows the association between the 13-gene exploratory signature and clinical benefit (PFS) in the full data set from PALOMA-3 after filtering out outliers from NMF liver factor (FIG. 9).
The 13-gene signature was further validated using independent samples from the PALOMA-2 trial. The ability of the gene signature to predict palbociclib efficacy across lines of therapy was demonstrated by application of the 13-gene signature to an independent metastatic breast cancer cohort in the PALOMA-2 trial. Association between the 13-gene exploratory signature and clinical benefit from the combination of palbociclib plus letrozole in the PALOMA-2 trial is shown in the Kaplan-Meier plot in FIG. 8(B).
Among the genes associated with palbociclib resistance, BUB1 B and CDKN2D are regulators of the cell cycle pathway, with the former inhibiting anaphase-promoting complex/cyclosome (Vleugel et al, 2015) and the latter an inhibitor to the activation of the CDK4/6 complex (Rolland et al, 2014). TRIB1 regulates the MAPK pathway (Jamieson et al, 2018).
Additional genes associated with relative palbociclib resistance in the 13-gene signature include CCNE1 , as well as targets of MYC and WNT signaling. See Birdsey et al., The endothelial transcription factor ERG promotes vascular stability and growth through Wnt/beta-catenin signaling. Dev Cell 32:82-96, 2015; Pacilli et al., Carnitineacyltransferase system inhibition, cancer cell death, and prevention of myc-induced lymphomagenesis. J Natl Cancer Inst 105:489-498, 2013; Sonntag et al., Cyclin E1 and cyclin-dependent kinase 2 are critical for initiation, but not for progression of
hepatocellular carcinoma. Proc Natl Acad Sci II S A 115:9282-9287, 2018; Tomlins et al., Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 310:644-648, 2005; Vijayakumar et al., High-frequency canonical Wnt activation in multiple sarcoma subtypes drives proliferation through a TCF/beta-catenin target gene, CDC25A. Cancer Cell 19:601-612, 2011 ).
Genes associated with relative palbociclib sensitivity in the 13-gene signature have been associated with HR+ breast cancer etiology, either as modifiers of patient response to ESR1 antagonists, susceptibility to the disease or direct target genes. See Bouras et al., Stanniocalcin 2 is an estrogen-responsive gene coexpressed with the estrogen receptor in human breast cancer. Cancer Res 62:1289-1295, 2002; Griffith et al., The prognostic effects of somatic mutations in ER-positive breast cancer. Nat Commun 9: 3476, 2018; Morotti et al., Hypoxia-induced switch in SNAT2/SLC38A2 regulation generates endocrine resistance in breast cancer. Proc Natl Acad Sci II S A 116:12452-12461 , 2019; Subramanian et al., Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci II S A 102:15545-15550, 2005; and Toy et al., Activating ESR1 Mutations Differentially Affect the Efficacy of ER Antagonists. Cancer discovery 7:277-287, 2017.
While EdgeSeq profiling has been used to evaluate expression levels for the gene signatures provided herein, the skilled person would understand that other techniques (e.g., qPCR, RNAseq, or NanoString technology) could be employed to evaluate gene expression could be used to train the EN models.
Therapeutic Methods, Uses and Combinations
In one aspect, the invention provides a method of treating hormone receptorpositive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer in a subject in need thereof, comprising:
(a) determining the expression level for each gene in a gene signature in a tumor sample from the subject;
(b) computing a composite score for the gene signature as a weighted sum of the expression levels for each gene in the gene signature;
(c) selecting the subject for treatment with a cyclin dependent kinase 4/6 (CDK 4/6) inhibitor and an endocrine therapeutic agent when the composite score is less than the median of a reference composite score computed for the gene signature in a reference breast cancer population; and
(d) administering to the selected subject an amount of a CDK4/6 inhibitor and an amount of an endocrine therapeutic agent, wherein the amounts together are effective to treat cancer; wherein the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
In some embodiments of this aspect, the subject is selected for treatment when the composite score is less than the first quartile of the reference composite score computed for the gene signature in the reference breast cancer population.
In another aspect, the invention provides a method of selecting a subject having hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer for treatment with a CDK4/6 inhibitor and an endocrine therapeutic agent, comprising:
(a) determining the expression level for each gene in a gene signature in a tumor sample from the subject;
(b) computing a composite score for the gene signature as a weighted sum of the expression levels for each gene in the gene signature; and
(c) selecting the subject for treatment when the composite score is less than the median of a reference composite score computed for the gene signature in a reference breast cancer population; wherein the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
In some embodiments of this aspect, the subject is selected for treatment when the composite score is less than the first quartile of the reference composite score computed for the gene signature in the reference breast cancer population.
In another aspect, the invention provides a method of predicting whether a subject having hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer will respond to treatment with a CDK4/6 inhibitor and an endocrine therapeutic agent, comprising:
(a) determining the expression level for each gene in a gene signature in a tumor sample from the subject;
(b) computing a composite score for the gene signature as a weighted sum of the expression levels for each gene in the gene signature;
(c) comparing the composite score to a reference composite score computed for the gene signature in a reference breast cancer population; and
(d) predicting the subject will respond to treatment when the composite score is less than the median of the reference composite score; wherein the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
In some embodiments of this aspect, the subject is predicted to respond to treatment when the composite score is less than the first quartile of the reference composite score computed for the gene signature in the reference breast cancer population.
In a further aspect, the invention provides a method of predicting whether a subject having hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer will be resistant to treatment with a CDK4/6 inhibitor and an endocrine therapeutic agent, comprising:
(a) determining the expression level for each gene in a gene signature in a tumor sample from the subject;
(b) computing a composite score for the gene signature as a weighted sum of the expression levels for each gene in the gene signature;
(c) comparing the composite score to a reference composite score computed for the gene signature in a reference breast cancer population; and
(d) predicting the subject will be resistant to treatment when the composite score is greater than the median of the reference composite score; wherein the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
In some embodiments of this aspect, the subject is predicted to be resistant to treatment when the composite score is greater than the third quartile of the reference composite score computed for the gene signature in the reference breast cancer population.
In some embodiments of each of the methods, uses and combinations herein, the breast cancer is HR+, HER2- advanced or metastatic breast cancer. In some such embodiments, the HR+, HER2- advanced or metastatic breast cancer had progressed on prior endocrine therapy (i.e., the method relates to a second line or later line of
therapy). In other such embodiments, the HR+, HER2- advanced or metastatic breast cancer is treatment naive (i.e. , the method relates to a first line of therapy).
In some embodiments of each of the methods herein, the breast cancer is HR+, HER2- early breast cancer. In some embodiments of each of the methods herein, the breast cancer is HR+, HER2- high risk early breast cancer. In some embodiments of each of the methods herein, the breast cancer is HR+, HER2- early breast cancer at high risk of relapse after showing less than pathological complete response to neoadjuvant chemotherapy. In some embodiments of each of the methods herein, the breast cancer is HR+, HER2- early breast cancer with residual invasive disease after neoadjuvant chemotherapy.
In some embodiments of each of the methods herein, the CDK4/6 inhibitor and the endocrine therapeutic agent are administered sequentially, simultaneously or concurrently. In some embodiments of each of the methods herein, the endocrine therapeutic agent is selected from the group consisting of an aromatase inhibitor, a selective estrogen receptor degrader (SERD), and a selective estrogen receptor modulator (SERM). In some such embodiments the endocrine therapeutic agent is selected from the group consisting of letrozole, anastrozole, exemestane and fulvestrant.
In some such embodiments, the aromatase inhibitor is selected from the group consisting of letrozole, anastrozole, and exemestane. In some such embodiments, the aromatase inhibitor is letrozole. In some embodiments, the endocrine therapeutic agent is a SERD. In some such embodiments, the SERD is selected from the group consisting of fulvestrant, elacestrant (RAD-1901 , Radius Health), SAR439859 (Sanofi), RG6171 (Roche), AZD9833 (AstraZeneca), AZD9496 (AstraZeneca), rintodestrant (G1 Therapeutics), ZN-c5 (Zentalis), LSZ102 (Novartis), D-0502 (Inventisbio), LY3484356 (Lilly), and SHR9549 (Jiansu Hengrui Medicine). In some such embodiments, the SERD is fulvestrant. In some embodiments, the endocrine therapeutic agent is a SERM. In some such embodiments, the SERM is selected from the group consisting of tamoxifen, raloxifene, toremifene, lasofoxifene, bazedoxifene and afimoxifene. In some such embodiments, the SERM is tamoxifen or raloxifene.
In preferred embodiments of each of the methods herein, the CDK4/6 inhibitor is palbociclib, or a pharmaceutically acceptable salt thereof.
In some embodiments of each of the methods herein, the gene signature comprises:
(a) CCNE1 , CDH15, CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1 ;
(b) CCNE1 , CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, and TMPRSS2; or
(c) BUB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, TRIB1 , ABCB11 , COL1 1 A1 , MAP3K1 , STC2 and TFDP2.
In some embodiments of each of the methods herein, the gene signature consists essentially of:
(a) CCNE1 , CDH15, CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1 ;
(b) CCNE1 , CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, and TMPRSS2; or
(c) BUB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, TRIB1 , ABCB11 , COL1 1 A1 , MAP3K1 , STC2 and TFDP2.
In some embodiments of each of the methods herein, the gene signature consists of:
(a) CCNE1 , CDH15, CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1 ;
(b) CCNE1 , CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, and TMPRSS2; or
(c) BUB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, TRIB1 , ABCB11 , COL1 1 A1 , MAP3K1 , STC2 and TFDP2.
In another aspect, the invention provides a method of treating hormone receptorpositive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer in a subject in need thereof, comprising:
(a) determining the expression levels of CCNE1 , CDH15, CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1 in a tumor sample from the subject;
(b) selecting the subject for treatment with a cyclin dependent kinase 4/6 (CDK 4/6) inhibitor and an endocrine therapeutic agent when:
(i) the expression levels of CCNE1 , CPT1A, ERG, LRP5, TMPRSS2, and TRIB1 in the tumor sample are downregulated relative to the expression levels in a non-tumor sample from the subject or relative to the median expression levels in a control population; and/or
(ii) the expression levels of CDH15, MAP3K1 , SLC38A2, STC2, and TFDP2 in the tumor sample are upregulated relative to the expression levels in a non-tumor sample from the subject or relative to the median expression levels in a control population; and
(c) administering to the selected subject an amount of a CDK4/6 inhibitor and an amount of an endocrine therapeutic agent, wherein the amounts together are effective to treat cancer.
In some embodiments of this aspect, the subject is selected for treatment when:
(i) the expression levels of CCNE1 , CPT1A, ERG, LRP5, TMPRSS2, and TRIB1 in the tumor sample are downregulated relative to the expression levels in a non-tumor sample from the subject or relative to the first quartile expression levels in a control population; and/or
(ii) the expression levels of CDH15, MAP3K1 , SLC38A2, STC2, and TFDP2 in the tumor sample are upregulated relative to the expression levels in a non-tumor sample from the subject or relative to the first quartile expression levels in a control population.
In another aspect, the invention provides a method of treating hormone receptorpositive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer in a subject in need thereof, comprising:
(a) determining the expression levels of CCNE1 , CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, and TMPRSS2 in a tumor sample from the subject;
(b) selecting the subject for treatment with a cyclin dependent kinase 4/6 (CDK 4/6) inhibitor and an endocrine therapeutic agent when:
(i) the expression levels of CCNE1 , CPT1A, ERG, LRP5, and TMPRSS2 in the tumor sample are downregulated relative to the expression levels in a non-tumor sample from the subject or the median expression levels in a control population; and/or
(ii) the expression levels of MAP3K1 , SLC38A2, STC2, and TFDP2 in the tumor sample are upregulated relative to the expression levels in a non- tumor sample from the subject or the median expression levels in a control population; and
(c) administering to the selected subject an amount of a CDK4/6 inhibitor and an amount of an endocrine therapeutic agent, wherein the amounts together are effective to treat cancer.
In some embodiments of this aspect, the subject is selected for treatment when:
(i) the expression levels of CCNE1 , CPT1A, ERG, LRP5, and TMPRSS2 in the tumor sample are downregulated relative to the expression levels in a non-tumor sample from the subject or the first quartile expression levels in a control population; and/or
(ii) the expression levels of MAP3K1 , SLC38A2, STC2, and TFDP2 in the tumor sample are upregulated relative to the expression levels in a nontumor sample from the subject or the first quartile expression levels in a control population.
In another aspect, the invention provides a method of treating hormone receptorpositive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer in a subject in need thereof, comprising:
(a) determining the expression levels of BLIB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, TRIB1 , ABCB11 , COL11A1 , MAP3K1 , STC2 and TFDP2 in a tumor sample from the subject;
(b) selecting the subject for treatment with a cyclin dependent kinase 4/6 (CDK 4/6) inhibitor and an endocrine therapeutic agent when:
(i) the expression levels of BUB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, and TRIB1 in the tumor sample are downregulated relative to the expression levels in a non-tumor sample from the subject or the median expression levels in a control population; and/or
(ii) the expression levels of ABCB11 , COL11 A1 , MAP3K1 , STC2 and TFDP2 in the tumor sample are upregulated relative to the expression levels in a non-tumor sample from the subject or the median expression levels in a control population; and
(c) administering to the selected subject an amount of a CDK4/6 inhibitor and an amount of an endocrine therapeutic agent, wherein the amounts together are effective to treat cancer.
In some embodiments of this aspect, the subject is selected for treatment when:
(i) the expression levels of BUB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, and TRIB1 in the tumor sample are downregulated relative to the expression levels in a non-tumor sample from the subject or the first quartile expression levels in a control population; and/or
(ii) the expression levels of ABCB11 , COL11 A1 , MAP3K1 , STC2 and TFDP2 in the tumor sample are upregulated relative to the expression levels in a
non-tumor sample from the subject or the first quartile expression levels in a control population.
In another aspect, the invention provides use of a combination comprising a CDK4/6 inhibitor and an endocrine therapeutic agent for treating hormone receptorpositive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer in a subject in need thereof, comprising:
(a) determining the expression level for each gene in a gene signature in a tumor sample from the subject;
(b) computing a composite score for the gene signature as a weighted sum of the expression levels for each gene in the gene signature; and
(c) selecting the subject for treatment with the combination when the composite score is less than the median of a reference composite score computed for the gene signature in a reference breast cancer population; wherein the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
In some embodiments of this aspect, the subject is selected for treatment when the composite score is less than the first quartile of the reference composite score computed for the gene signature in the reference breast cancer population.
In another aspect, the invention provides a combination of a CDK4/6 inhibitor and an endocrine therapeutic agent for use in treating hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer in a subject in need thereof, comprising:
(a) determining the expression level for each gene in a gene signature in a tumor sample from the subject;
(b) computing a composite score for the gene signature as a weighted sum of the expression levels for each gene in the gene signature; and
(c) selecting the subject for treatment with the combination when the composite score is less than the median of a reference composite score computed for the gene signature in a reference breast cancer population; wherein the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
In some embodiments of this aspect, the subject is selected for treatment when the composite score is less than the first quartile of the reference composite score computed for the gene signature in the reference breast cancer population.
In some embodiments of each of the uses and combinations herein, the breast cancer is HR+, HER2- advanced or metastatic breast cancer. In some such embodiments, the HR+, HER2- advanced or metastatic breast cancer had progressed on prior endocrine therapy (i.e., the method relates to a second line or later line of therapy). In other such embodiments, the HR+, HER2- advanced or metastatic breast cancer is treatment naive (i.e., the method relates to a first line of therapy).
In some embodiments of each of the uses and combinations herein, the breast cancer is HR+, HER2- early breast cancer. In some embodiments of each of the uses and combinations herein, the breast cancer is HR+, HER2- high risk early breast cancer. In some embodiments of each of the uses and combinations herein, the breast cancer is HR+, HER2- early breast cancer at high risk of relapse after showing less than pathological complete response to neoadjuvant chemotherapy. In some embodiments of each of the uses and combinations herein, the breast cancer is HR+, HER2- early breast cancer with residual invasive disease after neoadjuvant chemotherapy.
In some embodiments of each of the uses and combinations herein, the CDK4/6 inhibitor and the endocrine therapeutic agent are administered sequentially, simultaneously or concurrently. In some embodiments of each of the methods herein, the endocrine therapeutic agent is selected from the group consisting of an aromatase inhibitor, a selective estrogen receptor degrader (SERD), and a selective estrogen receptor modulator (SERM). In some such embodiments the endocrine therapeutic agent is selected from the group consisting of letrozole, anastrozole, exemestane and fulvestrant.
In some embodiments of each of the uses and combinations herein, the CDK4/6 inhibitor is palbociclib, or a pharmaceutically acceptable salt thereof.
In some embodiments of each of the uses and combinations herein, the gene signature comprises:
(a) CCNE1 , CDH15, CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1 ;
(b) CCNE1 , CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, and TMPRSS2; or
(c) BUB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, TRIB1 , ABCB11 ,
COL1 1 A1 , MAP3K1 , STC2 and TFDP2.
In some embodiments of each of the uses and combinations herein, the gene signature consists essentially of:
(a) CCNE1 , CDH15, CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1 ;
(b) CCNE1 , CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, and TMPRSS2; or
(c) BUB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, TRIB1 , ABCB11 , COL1 1 A1 , MAP3K1 , STC2 and TFDP2.
In some embodiments of each of the uses and combinations herein, the gene signature consists of:
(a) CCNE1 , CDH15, CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1 ;
(b) CCNE1 , CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, and TMPRSS2; or
(c) BUB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, TRIB1 , ABCB11 , COL1 1 A1 , MAP3K1 , STC2 and TFDP2.
In a further aspect, the invention provides a kit for assaying a tumor sample to determine a composite score for a gene signature in the tumor sample, comprising a first set of probes for detecting the expression level of each gene in the gene signature, wherein the gene signature comprises:
(a) five or more genes selected from the group consisting of ABCB11 , BLIB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1 ;
(b) CCNE1 , CDH15, CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1 ;
(c) CCNE1 , CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, and TMPRSS2; or
(d) BUB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, TRIB1 , ABCB11 , COL11 A1 , MAP3K1 , STC2 and TFDP2.
In some embodiments of this aspect, the invention provides a kit further comprising a second set of probes for detecting the expression level of a set of normalization genes in the tumor sample.
These and other aspects of the invention, including the exemplary specific embodiments listed below, will be apparent from the teachings contained herein.
Example 1
Elastic Net Analysis: 11 -Gene Signature
An 11 -gene signature was derived from the PALOMA-3 cohort. Metastatic tissue samples from 92 patients in the palbociclib plus fulvestrant treatment arm were used to generate the signature. A machine-learning based elastic net regularization algorithm with clinical outcome association by Cox PH regression was applied. A 10-fold cross validation was run to select an a = 0.53 and X = 0.31 and then 100 bootstraps were performed to provide a list of complementary expression features comprising a model from each round.
Genes were selected based on frequency observed in the bootstraps. A composite signature score was computed by weighting expression of signature genes by their coefficients.
Eleven genes were found to have non-zero coefficients in at least 40% of the models. FIG. 1 shows the 11 -gene exploratory signature, where bar length (top x-axis) reflects the proportion of 100 bootstrap models containing the gene while purple points (bottom x-axis) reflect average non-zero coefficient across bootstrap models. Six of the genes (CCNE1 , CPT1A, ERG, LRP5, TMPRSS2, TRIB1 ) appear to be associated with relative resistance, while the remaining five genes (CDH15, MAP3K1 , SLC38A2, STC2, TFDP2) appear to be associated with relative sensitivity (FIG. 1 ). A composite signature score was computed by weighting each component gene by its average coefficient across bootstrap models.
A hierarchical clustering heatmap of the 11 -gene exploratory signature and progression-free survival (PFS) is provided in FIG. 2. Clustering was conducted using 1 -Pearson correlation as distance metric and average linkage. Shown in FIG. 2(A) are the metastatic samples across the palbociclib plus fulvestrant treatment arm of PALOMA-3 . Shown in FIG. 2(B) are placebo plus fulvestrant control arm. For each arm, patients are sorted by PFS, excluding those with an insufficient follow-up time of less than 12 months and no event.
The 11 -gene signature was applied to 142 metastatic patient samples from PALOMA-3, comprising both the palbociclib plus fulvestrant and the placebo plus fulvestrant arms (Table 1 ). Composite signature scores were computed by weighting each component gene by its average coefficient across bootstrap models. Composite scores for patients in the palbociclib arm and the placebo arm were dichotomized by median into high and low scoring groups. PFS in months was plotted in the Kaplan- Meier plot in FIG. 3(A) (interaction P = 3.53e-05):
(1 ) palbociclib plus fulvestrant: high (n=47; mPFS = 7.3 mo.) - solid, blue line;
(2) palbociclib plus fulvestrant: low (n=45; mPFS = 16.1 mo.) - dashed, blue line;
(3) placebo plus fulvestrant: high (n=24; mPFS = 3.6 mo.) - solid, red line; and
(4) placebo plus fulvestrant: low (n=26; mPFS = 2.1 mo.) - dashed, red line.
All patient groups benefited from the palbociclib combination (HR<1 ) but to a varying extent (HR = 0.84 and 0.13 for higher and lower by median, respectively).
The 11 -gene signature was further applied to the full PALOMA-3 data set, comprising 302 patient samples, including both primary and metastatic samples (Table 1 ). Composite signature scores were computed by weighting each component gene by its average coefficient across bootstrap models. Composite scores for patients in the palbociclib arm and the placebo arm were dichotomized by median into high and low scoring groups. Median PFS (mPFS) in months was plotted in the Kaplan-Meier plot in FIG. 3(B) (interaction P = 3.31 e-05):
(1 ) palbociclib plus fulvestrant: high (n=102; mPFS = 9.3 mo.) - solid, blue line;
(2) palbociclib plus fulvestrant: low (n=92; mPFS = 16.6 mo.) - dashed, blue line;
(3) placebo plus fulvestrant: high (n=49; mPFS = 5.4 mo.) - solid, red line; and
(4) placebo plus fulvestrant: low (n=59; mPFS = 3.7 mo.) - dashed, red line.
All patient groups benefited from the palbociclib combination (HR<1 ) but to a varying extent (HR = 0.99 and 0.27 for higher and lower by median, respectively).
The association between gene expression level and efficacy for palbociclib treatment was assessed for the 11 component genes individually using the 302 samples from the full PALOMA-3 trial. The result was plotted as a Forest plot in FIG. 3(C). Gene expression levels were dichotomized by median expression, with hazard ratios (HRs) determined for PFS of palbociclib plus fulvestrant (PAL + Fulv) versus placebo plus fulvestrant (PCB + Fulv). HRs were derived from a Cox PH regression model. Interaction P value for statistical interaction between gene expression and treatment for the 11 genes in the signature (CCNE1 , CPT1A, ERG, LRP5, TMPRSS2, TRIB1 , CDH15, MAP3K1 , SLC38A2, STC2, and TFDP2).
The 11 -gene signature was further validated on an independent metastatic breast cancer cohort from first-line PALOMA-2 trial, where tumor samples came from a mixture of primary or metastatic biopsies and the origin of tissue samples were not recorded (Table 2). Composite signature scores were computed by weighting each component gene by its average coefficient across bootstrap models.
Composite scores for patients in the palbociclib arm and the placebo arm were dichotomized by median into high and low scoring groups. Median PFS (mPFS) in months was plotted in the Kaplan-Meier plot in FIG. 4(A) (interaction P = 9.78e-03):
(1 ) palbociclib plus letrozole: high (n=156; mPFS = 19.2 mo.) - solid, blue line;
(2) palbociclib plus letrozole: low (n=146; mPFS = 33.1 mo.) - dashed, blue line;
(3) placebo plus letrozole: high (n=71 ; mPFS = 13.8 mo.) - solid, red line; and
(4) placebo plus letrozole: low (n=81 ; mPFS = 13.8 mo.) - dashed, red line.
Despite marked cross-cohort differences in combination partner (fulvestrant vs letrozole) and clinical setting (endocrine resistant vs treatment naive), the 11 -gene signature remained predictive of palbociclib efficacy (FIG. 4; interaction P=9.78E-3).
Signature-treatment statistical interaction was also apparent from continuous variable analysis (P = 0.016 and P = 0.0031 before and after adjusting for baseline clinicopathologic characteristics respectively). All patient groups benefited from the palbociclib combination (HR<1 ) but to a varying extent (HR = 0.79 and 0.43 for higher and lower by median respectively).
Example 2
Elastic Net Analysis: 9-Gene Signature
A 9-gene signature was derived from the PALOMA-3 cohort. Metastatic tissue samples from 92 patients in the palbociclib plus fulvestrant treatment arm were used to generate the signature. A machine-learning based elastic net regularization algorithm with clinical outcome association by Cox PH regression was applied. A 5-fold cross validation was run to select an a = 1 and X = 0.277 and then 1 ,000 bootstraps were performed to provide a list of complementary expression features comprising a model from each round.
Genes were selected based on frequency observed in the bootstraps and 9 genes were selected using a C-lndex to build optimal predictive model including CCNE1. A composite signature score was computed by weighting each component gene by its average coefficient across bootstrap models. The 9 selected genes included CCNE1 , CPT1A, ERG, LRP5, TMPRSS2, MAP3K1 , SLC38A2, STC2, and TFDP2 (FIG. 5). FIG. 5 shows the 9-gene exploratory signature, where bar length (top x-axis) reflects the proportion of 1000 bootstrap models containing the gene while purple points (bottom x-axis) reflect average non-zero coefficient across bootstrap models. Five of the genes (CCNE1 , CPT1A, ERG, LRP5, and TMPRSS2) appear to be associated with
relative resistance to palbociclib, while the remaining four genes (MAP3K1 , SLC38A2, STC2, and TFDP2) appear to be associated with relative sensitivity to palbociclib.
The 9-gene signature was further applied to the full PALOMA-3 data set, comprising 302 patient samples, including both primary and metastatic samples (Table 1 ). Composite signature scores were computed by weighting each component gene by its average coefficient across bootstrap models.
Composite scores for patients in the palbociclib arm and the placebo arm were dichotomized by median into high and low scoring groups. Median PFS (mPFS) in months was plotted in the Kaplan-Meier plot in FIG. 5(B) (interaction P = 0.00011 ):
(1 ) palbociclib plus fulvestrant: high (n=99; mPFS = 7.62 mo.) - solid, blue line;
(2) palbociclib plus fulvestrant: low (n=95; mPFS = 16.62 mo.) - dashed, blue line;
(3) placebo plus fulvestrant: high (n=52; mPFS = 4.70 mo.) - solid, red line; and
(4) placebo plus fulvestrant: low (n=56; mPFS = 3.71 mo.) - dashed, red line.
The exploratory 9-gene signature was then tested on the independent metastatic breast cancer cohort from the PALOMA-2 clinical trial comprising 454 patient samples. Despite marked cross-cohort differences in combination partner (fulvestrant vs. letrozole) and clinical setting (endocrine resistant vs. treatment naive), the 9-gene signature remained predictive of palbociclib efficacy (FIG. 5(C)).
Composite scores for patients in the palbociclib arm and the placebo arm were dichotomized by median into high and low scoring groups. Median PFS (mPFS) in months was plotted in the Kaplan-Meier plot in FIG. 5(C) (interaction P = 0.036):
(1 ) palbociclib plus letrozole: high (n=154; mPFS = 19.2 mo.) - solid, blue line;
(2) palbociclib plus letrozole: low (n=149; mPFS = 30.3 mo.) - dashed, blue line;
(3) placebo plus letrozole: high (n=73; mPFS = 13.7 mo.) - solid, red line; and
(4) placebo plus letrozole: low (n=78; mPFS = 13.9 mo.) - dashed, red line.
Signature-treatment statistical interaction was also apparent from continuous variable analysis (P = 0.014). All patient groups benefited from the palbociclib combination (HR<1 ) but to a varying extent (HR = 0.77 and 0.46 for higher and lower by median respectively).
Example 3
Elastic Net Analysis: 13-Gene Signature
Non-Negative Matrix Factorization (NMF) was applied to the 302 primary and metastatic samples from the PALOMA-3 trial. The virtual microdissection identified
fourteen molecular factors that characterize biological processes differentiating baseline tumor samples of this cohort. One of the fourteen factors unambiguously represented the liver specific genes. Since some samples were from biopsies of liver metastases, a liver-specific factor was used to identify samples with significant liver component as described in the methods below. Using this approach, 30 samples with significant liver content were identified and excluded from further analysis.
The Elastic Net method described in Example 2 was then reapplied to the remaining dataset after removing potential outliers for liver factors. A 5-fold cross validation was run to select an a = 1 and X = 0.204 and then 1 ,000 bootstraps were performed to provide a list of complementary expression features comprising a model from each round. This analysis provided a refined 13-gene signature with higher bootstrapping frequencies than the previous 9-gene signature. A composite signature score was computed by weighting each component gene by its average coefficient across bootstrap models. The 13 selected genes included LRP5, TRIB1 , TFDP2, MAP3K1 , ERG, TMPRSS2, BUB1 B, ABCB11 , MT1X, CCNE1 , CDKN2D, STC2 and COL11A1. FIG. 7 shows the 13-gene exploratory signature, where bar length (top x- axis) reflects the proportion of 1000 bootstrap models containing the gene while purple points (bottom x-axis) reflect average non-zero coefficient across bootstrap models. Eight of the genes (LRP5, TRIB1 , ERG, TMPRSS2, BUB1 B, MT1X, CCNE1 , and CDKN2D) appear to be associated with relative resistance to palbociclib, while the remaining five genes (TFDP2, MAP3K1 , ABCB11 , STC2 and COL11A1 ) appear to be associated with relative sensitivity to palbociclib (FIG. 7).
The 13-gene signature was further applied to the PALOMA-3 data set after removing samples with significant liver content, comprising 272 patient samples, including both primary and metastatic samples (Table 1 ). Composite signature scores were computed by weighting each component gene by its average coefficient across bootstrap models.
Composite scores for patients in the palbociclib arm and the placebo arm were dichotomized by median into high and low scoring groups. Median PFS (mPFS) in months in PALOMA-3 was plotted in the Kaplan-Meier plot in FIG. 8(A) (interaction P = 0.00078):
(1 ) palbociclib plus fulvestrant: high (n=91 ; mPFS = 9.23 mo.) - solid, blue line;
(2) palbociclib plus fulvestrant: low (n=84; mPFS = 16.13 mo.) - dashed, blue line;
(3) placebo plus fulvestrant: high (n=45; mPFS = 7.23 mo.) - solid, red line; and
(4) placebo plus fulvestrant: low (n=52; mPFS = 3.71 mo.) - dashed, red line.
The transferability of the observed molecular factors’ association with palbociclib response was examined in connection with an independent cohort of ER+, HER2- advanced breast cancer patients from the PALOMA-2 study. This cohort represents a patient population at an earlier stage of HR+ metastatic breast cancer and was treated with palbociclib in combination with the aromatase inhibitor, letrozole, or letrozole plus placebo as a first line therapy. The 14 factors identified in PALOMA-3 cohort were projected onto the PALOMA-2 data using method described in Tamayo et. Al., Metagene projection for cross-platform, cross-species characterization of global transcriptional states. Proc. Natl. Acad. Sci. USA 104, 5959-5964, 2007. In this cohort, 37 samples were identified with significant liver content and excluded from further analyses as for PALOMA-3.
The exploratory 13-gene signature was then tested on the independent metastatic breast cancer cohort of 417 samples from PALOMA-2 after filtering out samples with significant liver content and remained predictive of palbociclib efficacy (FIG. 8(B)). Composite scores for patients in the palbociclib arm and the placebo arm were dichotomized by median into high and low scoring groups. Median PFS (mPFS) in months was plotted in the Kaplan-Meier plot in FIG. 8(B) (interaction P = 0.018):
(1 ) palbociclib plus letrozole: high (n=142; mPFS = 16.6 mo.) - solid, blue line;
(2) palbociclib plus letrozole: low (n=137; mPFS = 35.7 mo.) - dashed, blue line;
(3) placebo plus letrozole: high (n=66; mPFS = 13.7 mo.) - solid, red line; and placebo plus letrozole: low (n=72; mPFS = 16.5 mo.) - dashed, red line.
Signature-treatment statistical interaction was also apparent from continuous variable analysis (P = 0.062). All patient groups benefited from the palbociclib combination (HR<1 ) but to a varying extent (HR = 0.76 and 0.41 for higher and lower by median respectively). Methods Virtual microdissection analysis
NMF was applied to HTG gene expression of 302 patient samples from PALOMA-3. The NMF algorithm factorizes the gene expression matrix V of g genes and s samples into two non-negative matrices of k factors: gene factor matrix W of weights of n gene for k factors and sample factor matrix H of weights of m sample for k factors. W represents the expression pattern of the k parts and H represents the respective contribution of k parts in each sample or bulk tumor1. Before applying NMF, the expression values for each patient were rank transformed. NMF was then performed on
rank transformed gene expression matrix Vpaloma-3with the R package “NMF” which used the “brunet” algorithm2. 30 runs of NMF were performed and the factorization that achieved the lowest approximation error for subsequent analyses was chosen. To determine the optimal k, we computed the cophenetic coefficient and chose k=14 that maximized the coefficient score. See Brunet et al., Metagenes and molecular pattern discovery using matrix factorization. Proc Natl Acad Sci II S A 101 , 4164-4169 (2004).
To extract exemplar genes for each of the k factors, genes with their contribution to the corresponding basis components of gene factor greater than 0.2 were selected. Gene Set Enrichment Analysis (GSEA) (Subramanian, et al., Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci II S A 102(43): 15545-15550, 2005) was performed on the pre-ranked gene lists based on their gene factor weights and the MSigDB gene set collection to annotate the factors.
Factor 14 (F14) was found to be enriched in liver specific genes with top exemplar genes uniquely expressing in liver tissues according to GTEX. To further confirm this, t test was performed on all factors between samples labeled as metastatic samples and samples labeled as primary samples based on clinical annotation. F14 showed significantly higher weights in the metastatic samples (p=1.7e-5).
To reduce the noise contributed by high liver cell contamination in the tumor, samples with high liver content based on F14 were removed. The cutoff=0.478 was chosen as the upper quartile plus 1.5 fold IQR (inter-quartile range). Using this approach, 30 samples were removed from the PALOMA-3 cohort, of which 28 were labeled as metastatic samples in clinical annotation while 2 were labeled as primary samples.
Using the metagene projection methodology, the low-dimensional NMF representation derived from the PALOMA-3 HTG data was projected onto the PALOMA-2 HTG data. See Tamayo, P., Scanfeld, D., Ebert, B.L., Gillette, M.A., Roberts, C.W., and Mesirov, J.P. (2007). Metagene projection for cross-platform, crossspecies characterization of global transcriptional states. Proc. Natl. Acad. Sci. USA 104, 5959-5964.
This projection enabled direct comparison of the two cohorts. The equation: Hpaioma-2 = (Wpaioma-3 )"1 x Vpaloma-2 was obtained using the Moore-Penrose generalized pseudoinverse on the rank transformed HTG data from PALOMA-2. Ben- Israel & Greville (2003) Generalized Inverses: Theory and Applications (Springer, New
York). Based on the projected liver factor F14 in PALOMA-2, the same cutoff=0.478 was applied and 37 samples with high liver content were removed.
Factor association with end-point PFS
The predictive and prognostic effect for each NMF factor was evaluated. A predictive biomarker refers to an NMF factor whose magnitude significantly correlates with palbociclib treatment outcome, manifest in the p-value associated with the factor x arm interaction term coefficient in the Cox PH model. A prognostic biomarker refers to an NMF factor whose magnitude significantly correlates with overall clinical outcome in a cohort, regardless of treatment arms (adjusting for treatment arms in a Cox PH model).
Predictive biomarkers were obtained from the interactive Cox PH model with PFS as the end-point eq. 1 : h(t|x) = h0 (t) expCpiX-t + p2x2 + p3XiX2) eq. 1 where xi is a dichotomous variable with 1 representing palbociclib plus fulvestrant treatment and 0 representing control treatment and X2 is a dichotomous variable with values 1 or 0 indicating whether the biomarker (NMF factor) is above median value or below. Interaction p-value is the p value associated with p3.
To determine prognostic biomarkers, the additive Cox PH regression model was used without an interaction term (eq. 2). The p-value associated with 2 was used to discover prognostic biomarkers. h(t|x) = h0 (t) exp(p2x2) eq. 2
Gene Expression Analysis
The EdgeSeq Oncology Biomarker Panel (HTG Molecular Diagnostics, Inc) was used for mRNA profiling of 2,534 cancer-related genes. The EdgeSeq system uses targeted capture sequencing to quantitate mRNA expression levels of gene targets in FFPE tissues. The first section of breast cancer FFPE tissue was stained with hematoxylin and eosin (H&E). A board-certified pathologist assessed the tumor cell content and tissue necrosis. Tumor content was estimated based on the number of malignant cells as a percentage of all cells (i.e. , malignant plus normal cells in the tissue section). The acceptance criterion for analysis was set at >70% of tumor content. Necrosis was assessed based on the percentage of necrotic tissue within the total tissue area. The acceptance criterion for analysis was set at <20% necrosis. Macrodissection was performed on the tissue sections if the tumor content was <70% or if necrosis was >20%. Sample preparation was conducted following laboratory
processes and manufactory protocols. Sequencing was performed on an Illumina NextSeq 500 Sequencer (Illumina, Inc.).
To normalize the sequencing data, probe counts were transformed into Iog2 counts per million. Expression values were quantile normalized. HTG Molecular Diagnostics, Inc., was blinded to patient information and clinical outcomes.
Alternative approaches to determine gene expression levels include may comprise performing RT-PCR, a hybridization, transcriptome analysis, RNAseq, a Northern blot, a Western blot, or an ELISA. For example, assaying the expression level of genes in a gene signature can comprise performing an array hybridization. In some embodiments, transcriptome analysis may comprise obtaining sequence information of expressed RNA molecules.
Claims
1. A method of treating hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer in a subject in need thereof, comprising:
(a) determining the expression level for each gene in a gene signature in a tumor sample from the subject;
(b) computing a composite score for the gene signature as a weighted sum of the expression levels for each gene in the gene signature;
(c) selecting the subject for treatment with a cyclin dependent kinase 4/6 (CDK 4/6) inhibitor and an endocrine therapeutic agent when the composite score is less than the median of a reference composite score computed for the gene signature in a reference breast cancer population; and
(d) administering to the selected subject an amount of a CDK4/6 inhibitor and an amount of an endocrine therapeutic agent, wherein the amounts together are effective to treat cancer; wherein the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
2. A method of selecting a subject having hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer for treatment with a CDK4/6 inhibitor and an endocrine therapeutic agent, comprising:
(a) determining the expression level for each gene in a gene signature in a tumor sample from the subject;
(b) computing a composite score for the gene signature as a weighted sum of the expression levels for each gene in the gene signature; and
(c) selecting the subject for treatment when the composite score is less than the median of a reference composite score computed for the gene signature in a reference breast cancer population; wherein the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
38
3. A method of predicting whether a subject having hormone receptorpositive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer will respond to treatment with a CDK4/6 inhibitor and an endocrine therapeutic agent, comprising:
(a) determining the expression level for each gene in a gene signature in a tumor sample from the subject;
(b) computing a composite score for the gene signature as a weighted sum of the expression levels for each gene in the gene signature;
(c) comparing the composite score to a reference composite score computed for the gene signature in a reference breast cancer population; and
(d) predicting the subject will respond to treatment when the composite score is less than the median of the reference composite score; wherein the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
4. A method of predicting whether a subject having hormone receptorpositive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer will be resistant to treatment with a CDK4/6 inhibitor and an endocrine therapeutic agent, comprising:
(a) determining the expression level for each gene in a gene signature in a tumor sample from the subject;
(b) computing a composite score for the gene signature as a weighted sum of the expression levels for each gene in the gene signature;
(c) comparing the composite score to a reference composite score computed for the gene signature in a reference breast cancer population; and
(d) predicting the subject will be resistant to treatment when the composite score is greater than the median of the reference composite score; wherein the gene signature comprises five or more genes selected from the group consisting of: ABCB11 , BUB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1.
5. The method of any one of claims 1 to 4, wherein the breast cancer is advanced or metastatic breast cancer.
39
6. The method of claim 5, wherein the advanced or metastatic breast cancer had progressed on prior endocrine therapy.
7. The method of claim 5, wherein the advanced or metastatic breast cancer is treatment naive.
8. The method of any one of claims 1 to 4, wherein the breast cancer is early breast cancer.
9. The method of any one of claims 1 to 8, wherein the CDK4/6 inhibitor and the endocrine therapeutic agent are administered sequentially, simultaneously or concurrently.
10. The method of any one of claims 1 to 9, wherein the endocrine therapeutic agent is selected from the group consisting of an aromatase inhibitor, a selective estrogen receptor degrader (SERD), and a selective estrogen receptor modulator (SERM).
11. The method of claim 10, wherein the endocrine therapeutic agent is selected from the group consisting of letrozole, anastrozole, exemestane and fulvestrant
12. The method of any one of claims 1 to 11 , wherein the CDK4/6 inhibitor is palbociclib, or a pharmaceutically acceptable salt thereof.
13. The method of any one of claims 1 to 12, wherein the gene signature comprises:
(a) CCNE1 , CDH15, CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2,
TMPRSS2, and TRIB1 ;
(b) CCNE1 , CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, and TMPRSS2; or
(c) BUB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, TRIB1 , ABCB11 , COL1 1 A1 , MAP3K1 , STC2 and TFDP2.
14. The method of any one of claims 1 to 12, wherein the gene signature consists essentially of:
(a) CCNE1 , CDH15, CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1 ;
(b) CCNE1 , CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, and TMPRSS2; or
(c) BUB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, TRIB1 , ABCB11 , COL1 1 A1 , MAP3K1 , STC2 and TFDP2.
15. The method of any one of claims 1 to 12, wherein the gene signature consists of:
(a) CCNE1 , CDH15, CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1 ;
(b) CCNE1 , CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, and TMPRSS2; or
(c) BUB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, TRIB1 , ABCB11 , COL1 1 A1 , MAP3K1 , STC2 and TFDP2.
16. A method of treating hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer in a subject in need thereof, comprising:
(a) determining the expression levels of CCNE1 , CDH15, CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1 in a tumor sample from the subject;
(b) selecting the subject for treatment with a cyclin dependent kinase 4/6 (CDK 4/6) inhibitor and an endocrine therapeutic agent when:
(i) the expression levels of CCNE1 , CPT1A, ERG, LRP5, TMPRSS2, and TRIB1 in the tumor sample are downregulated relative to the expression levels in a non-tumor sample from the subject or relative to the median expression levels in a control population; and/or
(ii) the expression levels of CDH15, MAP3K1 , SLC38A2, STC2, and TFDP2 in the tumor sample are upregulated relative to the expression levels in a non-tumor sample from the subject or relative to the median expression levels in a control population; and
(c) administering to the selected subject an amount of a CDK4/6 inhibitor and an amount of an endocrine therapeutic agent, wherein the amounts together are effective to treat cancer.
17. A method of treating hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer in a subject in need thereof, comprising:
(a) determining the expression levels of CCNE1 , CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, and TMPRSS2 in a tumor sample from the subject;
(b) selecting the subject for treatment with a cyclin dependent kinase 4/6 (CDK 4/6) inhibitor and an endocrine therapeutic agent when:
(i) the expression levels of CCNE1 , CPT1A, ERG, LRP5, and TMPRSS2 in the tumor sample are downregulated relative to the expression levels in a non-tumor sample from the subject or the median expression levels in a control population; and/or
(ii) the expression levels of MAP3K1 , SLC38A2, STC2, and TFDP2 in the tumor sample are upregulated relative to the expression levels in a nontumor sample from the subject or the median expression levels in a control population; and
(c) administering to the selected subject an amount of a CDK4/6 inhibitor and an amount of an endocrine therapeutic agent, wherein the amounts together are effective to treat cancer.
18. A method of treating hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2-) breast cancer in a subject in need thereof, comprising:
(a) determining the expression levels of BLIB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, TRIB1 , ABCB11 , COL11A1 , MAP3K1 , STC2 and TFDP2 in a tumor sample from the subject;
(b) selecting the subject for treatment with a cyclin dependent kinase 4/6 (CDK 4/6) inhibitor and an endocrine therapeutic agent when:
(i) the expression levels of BUB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, and TRIB1 in the tumor sample are downregulated relative to the expression levels in a non-tumor sample from the subject or the median expression levels in a control population; and/or
(ii) the expression levels of ABCB11 , COL11 A1 , MAP3K1 , STC2 and TFDP2 in the tumor sample are upregulated relative to the expression levels in a non-tumor sample from the subject or the median expression levels in a control population; and
(c) administering to the selected subject an amount of a CDK4/6 inhibitor and an amount of an endocrine therapeutic agent, wherein the amounts together are effective to treat cancer.
19. A kit for assaying a tumor sample to determine a composite score for a gene signature in the tumor sample, comprising a first set of probes for detecting the expression level of each gene in the gene signature, wherein the gene signature comprises:
(a) five or more genes selected from the group consisting of ABCB11 , BLIB1 B, CCNE1 , CDH15, CDKN2D, COL11A1 , CPT1A, ERG, LRP5, MAP3K1 , MT1X, SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1 ;
(b) CCNE1 , CDH15, CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, TMPRSS2, and TRIB1 ;
(c) CCNE1 , CPT1A, ERG, LRP5, MAP3K1 , SLC38A2, STC2, TFDP2, and TMPRSS2; or
(d) BUB1 B, CCNE1 , CDKN2D, ERG, LRP5, MT1X, TMPRSS2, TRIB1 , ABCB11 , COL11 A1 , MAP3K1 , STC2 and TFDP2.
20. The kit for of claim 19, further comprising a second set of probes for detecting the expression level of a set of normalization genes in the tumor sample.
43
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202063088932P | 2020-10-07 | 2020-10-07 | |
US63/088,932 | 2020-10-07 | ||
US202063089270P | 2020-10-08 | 2020-10-08 | |
US63/089,270 | 2020-10-08 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2022074537A1 true WO2022074537A1 (en) | 2022-04-14 |
Family
ID=78087427
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/IB2021/059086 WO2022074537A1 (en) | 2020-10-07 | 2021-10-04 | Biomarkers of tumor response to cdk4/6 inhibitors |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2022074537A1 (en) |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2015134674A1 (en) * | 2014-03-04 | 2015-09-11 | The Regents Of The University Of California | Biomarkers of response to cyclin d-cdk4/6 targeted therapies in cancer |
WO2017198685A1 (en) * | 2016-05-18 | 2017-11-23 | Université Libre de Bruxelles | Method for determining sensitivity to a cdk4/6 inhibitor |
WO2020157709A1 (en) * | 2019-02-01 | 2020-08-06 | Pfizer Inc. | Combination of a cdk inhibitor and a pim inhibitor |
-
2021
- 2021-10-04 WO PCT/IB2021/059086 patent/WO2022074537A1/en active Application Filing
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2015134674A1 (en) * | 2014-03-04 | 2015-09-11 | The Regents Of The University Of California | Biomarkers of response to cyclin d-cdk4/6 targeted therapies in cancer |
WO2017198685A1 (en) * | 2016-05-18 | 2017-11-23 | Université Libre de Bruxelles | Method for determining sensitivity to a cdk4/6 inhibitor |
WO2020157709A1 (en) * | 2019-02-01 | 2020-08-06 | Pfizer Inc. | Combination of a cdk inhibitor and a pim inhibitor |
Non-Patent Citations (30)
Title |
---|
BIRDSEY ET AL.: "The endothelial transcription factor ERG promotes vascular stability and growth through Wnt/beta-catenin signaling", DEV CELL, vol. 32, 2015, pages 82 - 96 |
BOURAS ET AL.: "Stanniocalcin 2 is an estrogen-responsive gene coexpressed with the estrogen receptor in human breast cancer", CANCER RES, vol. 62, 2002, pages 1289 - 1295 |
BRUNET ET AL.: "Metagenes and molecular pattern discovery using matrix factorization", PROC NATL ACAD SCI U S A, vol. 101, 2004, pages 4164 - 4169, XP009096387, DOI: 10.1073/pnas.0308531101 |
EFRON ET AL.: "International Encyclopedia of the Social & Behavioral Sciences", 2004, ELSEVIER, article "Resampling methods of estimation", pages: 13216 - 13220 |
FINN ET AL.: "Biomarker Analyses of Response to Cyclin-Dependent Kinase 4/6 Inhibition and Endocrine Therapy in Women with Treatment-Naive Metastatic Breast Cancer", CLIN CANCER RES, vol. 26, 2020, pages 110 - 121, XP055814465, DOI: 10.1158/1078-0432.CCR-19-0751 |
FINN ET AL.: "Biomarker Analyses of Response to Cyclin-Dependent Kinase 4/6 Inhibition and Endocrine Therapy in Women with Treatment-Naive Metastatic Breast Cancer", CLIN. CANCER RES., vol. 26, no. 1, 2020, pages 110 - 121, XP055814465, DOI: 10.1158/1078-0432.CCR-19-0751 |
FINN RICHARD S ET AL: "Treatment effect of palbociclib plus endocrine therapy by prognostic and intrinsic subtype and biomarker analysis in patients with bone-only disease: a joint analysis of PALOMA-2 and PALOMA-3 clinical trials", BREAST CANCER RESEARCH AND TREATMENT, SPRINGER US, NEW YORK, vol. 184, no. 1, 11 August 2020 (2020-08-11), pages 23 - 35, XP037272715, ISSN: 0167-6806, [retrieved on 20200811], DOI: 10.1007/S10549-020-05782-4 * |
GILTNANE ET AL.: "Genomic profiling of ER(+) breast cancers after short-term estrogen suppression reveals alterations associated with endocrine resistance", SCI TRANSL MED, vol. 9, 2017 |
GOODHUE ET AL.: "Does PLS have advantages for small sample size or non-normal data?", MIS QUARTERLY, vol. 36, no. 3, 2012, pages 981 - 1001 |
GRIFFITH ET AL.: "The prognostic effects of somatic mutations in ER-positive breast cancer", NAT COMMUN, vol. 9, 2018, pages 3476 |
HIGGINSBASELGA: "Targeted therapies for breast cancer", J CLIN INVEST, vol. 121, 2011, pages 3797 - 3803 |
I ANONYMOUS: "Technical Note. Design and Performance of the GeneChip Human Genome U133 Plus 2.0 and Human Genome U133A 2.0 Arrays", 1 January 2003 (2003-01-01), Santa Clara, CA 95051 USA, pages 1 - 9, XP055381704, Retrieved from the Internet <URL:http://tools.thermofisher.com/content/sfs/brochures/hgu133_p2_technote.pdf> [retrieved on 20170614] * |
MOFFITT ET AL.: "Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma", NAT GENET, vol. 47, no. 10, 2015, pages 1168 - 1178 |
MOROTTI ET AL.: "Hypoxia-induced switch in SNAT2/SLC38A2 regulation generates endocrine resistance in breast cancer", PROC NATL ACAD SCI U S A, vol. 116, 2019, pages 12452 - 12461 |
O'LEARY ET AL.: "Treating cancer with selective CDK4/6 inhibitors", NATURE REVIEWS, vol. 13, 2016, pages 417 - 430, XP055581045, DOI: 10.1038/nrclinonc.2016.26 |
PACILLI ET AL.: "Carnitine-acyltransferase system inhibition, cancer cell death, and prevention of myc-induced lymphomagenesis", J NATL CANCER INST, vol. 105, 2013, pages 489 - 498 |
REID-LAWRENCE SONYA ET AL: "Novel Agents for the Management of Endocrine Resistant Breast Cancer", CURRENT BREAST CANCER REPORTS, SPRINGER US, BOSTON, vol. 10, no. 4, 15 November 2018 (2018-11-15), pages 274 - 281, XP036647225, ISSN: 1943-4588, [retrieved on 20181115], DOI: 10.1007/S12609-018-0298-3 * |
ROBINSON ET AL.: "Activating ESR1 mutations in hormone-resistant metastatic breast cancer", NAT GENET, vol. 45, 2013, pages 1446 - 1451, XP055187988, DOI: 10.1038/ng.2823 |
SONNTAG ET AL.: "Cyclin E1 and cyclin-dependent kinase 2 are critical for initiation, but not for progression of hepatocellular carcinoma", PROC NATL ACAD SCI U S A, vol. 115, 2018, pages 9282 - 9287 |
SUBRAMANIAN ET AL.: "Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles", PROC NATL ACAD SCI U S A, vol. 102, no. 43, 2005, pages 15545 - 15550, XP002464143, DOI: 10.1073/pnas.0506580102 |
TAMAYO, P.SCANFELD, D.EBERT, B.L.GILLETTE, M.A.ROBERTS, C.W.MESIROV, J.P.: "Metagene projection for cross-platform, cross-species characterization of global transcriptional states", PROC. NATL. ACAD. SCI. USA, vol. 104, 2007, pages 5959 - 5964 |
TOMLINS ET AL.: "Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer", SCIENCE, vol. 310, 2005, pages 644 - 648, XP055458560, DOI: 10.1126/science.1117679 |
TOY ET AL.: "Activating ESR1 Mutations Differentially Affect the Efficacy of ER Antagonists", CANCER DISCOVERY, vol. 7, 2017, pages 277 - 287, XP055791815, DOI: 10.1158/2159-8290.CD-15-1523 |
TOY ET AL.: "ESR1 ligand-binding domain mutations in hormone-resistant breast cancer", NAT GENET, vol. 45, 2013, pages 1439 - 1445, XP055791821, DOI: 10.1038/ng.2822 |
TURNER ET AL.: "Cyclin E1 Expression and Palbociclib Efficacy in Previously Treated Hormone Receptor-Positive Metastatic Breast Cancer", J. CLIN. ONCOL., vol. 37, 2019, pages 1169 - 1178, XP055814575 |
TURNER ET AL.: "Cyclin E1 Expression and Palbociclib Efficacy in Previously Treated Hormone Receptor-Positive Metastatic Breast Cancer", JOURNAL OF CLINICAL ONCOLOGY, vol. 37, no. 14, 2019, pages 1169 - 1178, XP055814575 |
VIJAYAKUMAR ET AL.: "High-frequency canonical Wnt activation in multiple sarcoma subtypes drives proliferation through a TCF/beta-catenin target gene, CDC25A", CANCER CELL, vol. 19, 2011, pages 601 - 612 |
VIJAYARAGHAVAN SMRUTHI ET AL: "Inhibiting CDK in Cancer Therapy: Current Evidence and Future Directions", TARGETED ONCOLOGY, SPRINGER INTERNATIONAL PUBLISHING, CHAM, vol. 13, no. 1, 7 December 2017 (2017-12-07), pages 21 - 38, XP036441931, ISSN: 1776-2596, [retrieved on 20171207], DOI: 10.1007/S11523-017-0541-2 * |
W. A. WEBER: "Assessing tumor response to therapy", J. NUCL. MED., vol. 50, 2009, pages 1S - 10S, XP055156561, DOI: 10.2967/jnumed.108.057174 |
XI JING ET AL: "Sequencing Endocrine Therapy for Metastatic Breast Cancer: What Do We Do After Disease Progression on a CDK4/6 Inhibitor?", CURRENT ONCOLOGY REPORTS, CURRENT SCIENCE, GB, vol. 22, no. 6, 16 May 2020 (2020-05-16), XP037170146, ISSN: 1523-3790, [retrieved on 20200516], DOI: 10.1007/S11912-020-00917-8 * |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Hayama et al. | G12V and G12C mutations in the gene KRAS are associated with a poorer prognosis in primary colorectal cancer | |
Doane et al. | An estrogen receptor-negative breast cancer subset characterized by a hormonally regulated transcriptional program and response to androgen | |
Lips et al. | Breast cancer subtyping by immunohistochemistry and histological grade outperforms breast cancer intrinsic subtypes in predicting neoadjuvant chemotherapy response | |
Chen et al. | PPAR signaling pathway may be an important predictor of breast cancer response to neoadjuvant chemotherapy | |
Korde et al. | Gene expression pathway analysis to predict response to neoadjuvant docetaxel and capecitabine for breast cancer | |
Ma et al. | Predicting endocrine therapy responsiveness in breast cancer | |
CN106834462A (en) | One group of application of stomach oncogene | |
JP2011500071A (en) | Gene-based algorithmic cancer prognosis and patient clinical outcome | |
Rotunno et al. | Parity-related molecular signatures and breast cancer subtypes by estrogen receptor status | |
Foy et al. | The dynamics of gene expression changes in a mouse model of oral tumorigenesis may help refine prevention and treatment strategies in patients with oral cancer | |
Fu et al. | Identification of hub genes using co-expression network analysis in breast cancer as a tool to predict different stages | |
Abba et al. | Breast cancer biomarker discovery in the functional genomic age: a systematic review of 42 gene expression signatures | |
Meir et al. | Molecular characteristics of liver metastases from uveal melanoma | |
Xing et al. | Identification of miRNA signature in breast cancer to predict neoadjuvant chemotherapy response | |
Xiao et al. | Expression of MIF and c-erbB-2 in endometrial cancer | |
Shi et al. | Two novel long noncoding RNAs–RP11-296E3. 2 and LEF1-AS1can–separately serve as diagnostic and prognostic bio-markers of metastasis in colorectal cancer | |
Li et al. | A comprehensive evaluation of clinicopathologic characteristics, molecular features and prognosis in lung adenocarcinoma with solid component | |
Al-Zahrani et al. | Assessing the efficacy of androgen receptor and Sox10 as independent markers of the triple-negative breast cancer subtype by transcriptome profiling | |
US8455196B2 (en) | Biomarker for identifying subgroup of early-stage lung adenocarcinoma patients | |
Tuluhong et al. | Prognostic value of differentially expressed LncRNAs in triple-negative breast cancer: a systematic review and meta-analysis | |
Darb-Esfahani et al. | Estrogen receptor 1 mRNA is a prognostic factor in ovarian carcinoma: determination by kinetic PCR in formalin-fixed paraffin-embedded tissue | |
Zhu et al. | Cyclin-dependent kinase 5 acts as a promising biomarker in clear cell renal cell carcinoma | |
Zhao et al. | Multi-omics characterization of esophageal squamous cell carcinoma identifies molecular subtypes and therapeutic targets | |
Liu et al. | Albumin‐to‐alkaline phosphatase ratio: A novel prognostic index for patients with driver mutation‐negative advanced non‐small cell lung cancer | |
Zhang et al. | Evaluation of core serous epithelial ovarian cancer genes as potential prognostic markers and indicators of the underlying molecular mechanisms using an integrated bioinformatics analysis |
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: 21790567 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 21790567 Country of ref document: EP Kind code of ref document: A1 |