WO2023081537A1 - Cancer biomarkers for immune checkpoint inhibitors - Google Patents
Cancer biomarkers for immune checkpoint inhibitors Download PDFInfo
- Publication number
- WO2023081537A1 WO2023081537A1 PCT/US2022/049332 US2022049332W WO2023081537A1 WO 2023081537 A1 WO2023081537 A1 WO 2023081537A1 US 2022049332 W US2022049332 W US 2022049332W WO 2023081537 A1 WO2023081537 A1 WO 2023081537A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- transformed
- irs
- normalized
- level
- expression
- Prior art date
Links
- 229940076838 Immune checkpoint inhibitor Drugs 0.000 title claims abstract description 427
- 239000012274 immune-checkpoint protein inhibitor Substances 0.000 title claims abstract description 426
- 108091008026 Inhibitory immune checkpoint proteins Proteins 0.000 title abstract description 4
- 102000037984 Inhibitory immune checkpoint proteins Human genes 0.000 title abstract description 4
- 239000000107 tumor biomarker Substances 0.000 title description 2
- 238000002560 therapeutic procedure Methods 0.000 claims abstract description 417
- 206010028980 Neoplasm Diseases 0.000 claims abstract description 281
- 238000000034 method Methods 0.000 claims abstract description 142
- 230000008901 benefit Effects 0.000 claims abstract description 39
- 201000011510 cancer Diseases 0.000 claims abstract description 39
- 230000014509 gene expression Effects 0.000 claims description 822
- 230000004044 response Effects 0.000 claims description 352
- 108091032973 (ribonucleotides)n+m Proteins 0.000 claims description 325
- 101100407308 Mus musculus Pdcd1lg2 gene Proteins 0.000 claims description 315
- 108700030875 Programmed Cell Death 1 Ligand 2 Proteins 0.000 claims description 315
- 102100024213 Programmed cell death 1 ligand 2 Human genes 0.000 claims description 315
- 108091007507 ADAM12 Proteins 0.000 claims description 301
- 102000036663 ADAM12 Human genes 0.000 claims description 301
- 230000009286 beneficial effect Effects 0.000 claims description 294
- 230000002596 correlated effect Effects 0.000 claims description 237
- 238000005259 measurement Methods 0.000 claims description 223
- 102100036011 T-cell surface glycoprotein CD4 Human genes 0.000 claims description 207
- 108010074708 B7-H1 Antigen Proteins 0.000 claims description 203
- 102100024216 Programmed cell death 1 ligand 1 Human genes 0.000 claims description 203
- 101000716102 Homo sapiens T-cell surface glycoprotein CD4 Proteins 0.000 claims description 196
- 108090000623 proteins and genes Proteins 0.000 claims description 120
- 102100033587 DNA topoisomerase 2-alpha Human genes 0.000 claims description 97
- 108010046308 Type II DNA Topoisomerases Proteins 0.000 claims description 97
- 229960002621 pembrolizumab Drugs 0.000 claims description 82
- 239000012472 biological sample Substances 0.000 claims description 62
- 230000035772 mutation Effects 0.000 claims description 61
- 230000001131 transforming effect Effects 0.000 claims description 60
- 238000009169 immunotherapy Methods 0.000 claims description 59
- 238000009097 single-agent therapy Methods 0.000 claims description 54
- 238000011282 treatment Methods 0.000 claims description 46
- 101000955999 Homo sapiens V-set domain-containing T-cell activation inhibitor 1 Proteins 0.000 claims description 44
- 102100038929 V-set domain-containing T-cell activation inhibitor 1 Human genes 0.000 claims description 44
- 108010082945 Eukaryotic Initiation Factor-2B Proteins 0.000 claims description 32
- 102000002639 Eukaryotic Initiation Factor-2B Human genes 0.000 claims description 32
- 101000914051 Homo sapiens Probable cytosolic iron-sulfur protein assembly protein CIAO1 Proteins 0.000 claims description 32
- 102100034391 Porphobilinogen deaminase Human genes 0.000 claims description 32
- 101710189720 Porphobilinogen deaminase Proteins 0.000 claims description 32
- 101710170827 Porphobilinogen deaminase, chloroplastic Proteins 0.000 claims description 32
- 102100026405 Probable cytosolic iron-sulfur protein assembly protein CIAO1 Human genes 0.000 claims description 32
- 101710100896 Probable porphobilinogen deaminase Proteins 0.000 claims description 32
- CXVGEDCSTKKODG-UHFFFAOYSA-N sulisobenzone Chemical compound C1=C(S(O)(=O)=O)C(OC)=CC(O)=C1C(=O)C1=CC=CC=C1 CXVGEDCSTKKODG-UHFFFAOYSA-N 0.000 claims description 32
- 208000002154 non-small cell lung carcinoma Diseases 0.000 claims description 26
- 208000029729 tumor suppressor gene on chromosome 11 Diseases 0.000 claims description 26
- 206010006187 Breast cancer Diseases 0.000 claims description 24
- 208000026310 Breast neoplasm Diseases 0.000 claims description 24
- 108010014064 CCCTC-Binding Factor Proteins 0.000 claims description 20
- 102100024409 Gametogenetin-binding protein 2 Human genes 0.000 claims description 20
- 101000833430 Homo sapiens Gametogenetin-binding protein 2 Proteins 0.000 claims description 20
- 101001015037 Homo sapiens Integrin beta-7 Proteins 0.000 claims description 20
- 101000981952 Homo sapiens Kanadaptin Proteins 0.000 claims description 20
- 101001030211 Homo sapiens Myc proto-oncogene protein Proteins 0.000 claims description 20
- 102100033016 Integrin beta-7 Human genes 0.000 claims description 20
- 102100026797 Kanadaptin Human genes 0.000 claims description 20
- 102100038895 Myc proto-oncogene protein Human genes 0.000 claims description 20
- 230000000869 mutational effect Effects 0.000 claims description 20
- 238000007481 next generation sequencing Methods 0.000 claims description 15
- 229960003301 nivolumab Drugs 0.000 claims description 15
- 206010009944 Colon cancer Diseases 0.000 claims description 14
- 229960003852 atezolizumab Drugs 0.000 claims description 13
- 206010014733 Endometrial cancer Diseases 0.000 claims description 12
- 206010014759 Endometrial neoplasm Diseases 0.000 claims description 12
- 208000032818 Microsatellite Instability Diseases 0.000 claims description 12
- 206010033128 Ovarian cancer Diseases 0.000 claims description 12
- 206010061535 Ovarian neoplasm Diseases 0.000 claims description 12
- 206010061902 Pancreatic neoplasm Diseases 0.000 claims description 12
- 206010060862 Prostate cancer Diseases 0.000 claims description 12
- 208000000236 Prostatic Neoplasms Diseases 0.000 claims description 12
- 206010039491 Sarcoma Diseases 0.000 claims description 12
- 208000014829 head and neck neoplasm Diseases 0.000 claims description 12
- 208000015486 malignant pancreatic neoplasm Diseases 0.000 claims description 12
- 201000002528 pancreatic cancer Diseases 0.000 claims description 12
- 208000008443 pancreatic carcinoma Diseases 0.000 claims description 12
- 229950002916 avelumab Drugs 0.000 claims description 11
- 229950009791 durvalumab Drugs 0.000 claims description 11
- 229950010773 pidilizumab Drugs 0.000 claims description 11
- 229950007213 spartalizumab Drugs 0.000 claims description 11
- 229940045513 CTLA4 antagonist Drugs 0.000 claims description 10
- 108091092878 Microsatellite Proteins 0.000 claims description 10
- 201000008261 skin carcinoma Diseases 0.000 claims description 10
- 208000001333 Colorectal Neoplasms Diseases 0.000 claims description 8
- 206010051066 Gastrointestinal stromal tumour Diseases 0.000 claims description 8
- 239000002246 antineoplastic agent Substances 0.000 claims description 8
- 201000011243 gastrointestinal stromal tumor Diseases 0.000 claims description 8
- 229960005386 ipilimumab Drugs 0.000 claims description 8
- 201000001441 melanoma Diseases 0.000 claims description 8
- 206010005003 Bladder cancer Diseases 0.000 claims description 7
- 206010025323 Lymphomas Diseases 0.000 claims description 7
- 208000007097 Urinary Bladder Neoplasms Diseases 0.000 claims description 7
- 229940127089 cytotoxic agent Drugs 0.000 claims description 7
- 201000005112 urinary bladder cancer Diseases 0.000 claims description 7
- 102100026146 39S ribosomal protein L13, mitochondrial Human genes 0.000 claims description 6
- 206010004593 Bile duct cancer Diseases 0.000 claims description 6
- 208000003174 Brain Neoplasms Diseases 0.000 claims description 6
- 206010008342 Cervix carcinoma Diseases 0.000 claims description 6
- 208000000461 Esophageal Neoplasms Diseases 0.000 claims description 6
- 208000032612 Glial tumor Diseases 0.000 claims description 6
- 206010018338 Glioma Diseases 0.000 claims description 6
- 206010073073 Hepatobiliary cancer Diseases 0.000 claims description 6
- 101000691550 Homo sapiens 39S ribosomal protein L13, mitochondrial Proteins 0.000 claims description 6
- 101000896414 Homo sapiens Nuclear nucleic acid-binding protein C1D Proteins 0.000 claims description 6
- 101001043564 Homo sapiens Prolow-density lipoprotein receptor-related protein 1 Proteins 0.000 claims description 6
- 208000008839 Kidney Neoplasms Diseases 0.000 claims description 6
- 206010058467 Lung neoplasm malignant Diseases 0.000 claims description 6
- 206010030155 Oesophageal carcinoma Diseases 0.000 claims description 6
- 102100021923 Prolow-density lipoprotein receptor-related protein 1 Human genes 0.000 claims description 6
- 101710156592 Putative TATA-binding protein pB263R Proteins 0.000 claims description 6
- 208000015634 Rectal Neoplasms Diseases 0.000 claims description 6
- 206010038389 Renal cancer Diseases 0.000 claims description 6
- 208000004337 Salivary Gland Neoplasms Diseases 0.000 claims description 6
- 206010061934 Salivary gland cancer Diseases 0.000 claims description 6
- 208000005718 Stomach Neoplasms Diseases 0.000 claims description 6
- 102100040296 TATA-box-binding protein Human genes 0.000 claims description 6
- 101710145783 TATA-box-binding protein Proteins 0.000 claims description 6
- 208000024770 Thyroid neoplasm Diseases 0.000 claims description 6
- 208000006105 Uterine Cervical Neoplasms Diseases 0.000 claims description 6
- 201000005188 adrenal gland cancer Diseases 0.000 claims description 6
- 208000024447 adrenal gland neoplasm Diseases 0.000 claims description 6
- 229940121420 cemiplimab Drugs 0.000 claims description 6
- 201000007455 central nervous system cancer Diseases 0.000 claims description 6
- 201000010881 cervical cancer Diseases 0.000 claims description 6
- 208000029742 colonic neoplasm Diseases 0.000 claims description 6
- 201000004101 esophageal cancer Diseases 0.000 claims description 6
- 206010017758 gastric cancer Diseases 0.000 claims description 6
- 201000010982 kidney cancer Diseases 0.000 claims description 6
- 201000007270 liver cancer Diseases 0.000 claims description 6
- 208000014018 liver neoplasm Diseases 0.000 claims description 6
- 201000005202 lung cancer Diseases 0.000 claims description 6
- 208000020816 lung neoplasm Diseases 0.000 claims description 6
- 208000026037 malignant tumor of neck Diseases 0.000 claims description 6
- 201000002120 neuroendocrine carcinoma Diseases 0.000 claims description 6
- 208000029255 peripheral nervous system cancer Diseases 0.000 claims description 6
- 206010038038 rectal cancer Diseases 0.000 claims description 6
- 201000001275 rectum cancer Diseases 0.000 claims description 6
- 201000002314 small intestine cancer Diseases 0.000 claims description 6
- 201000011549 stomach cancer Diseases 0.000 claims description 6
- 201000002510 thyroid cancer Diseases 0.000 claims description 6
- 238000011269 treatment regimen Methods 0.000 claims description 6
- 210000002418 meninge Anatomy 0.000 claims description 3
- 102100023990 60S ribosomal protein L17 Human genes 0.000 claims 60
- 101710089372 Programmed cell death protein 1 Proteins 0.000 claims 60
- 102000016897 CCCTC-Binding Factor Human genes 0.000 claims 6
- 101000611936 Homo sapiens Programmed cell death protein 1 Proteins 0.000 description 364
- 102100040678 Programmed cell death protein 1 Human genes 0.000 description 364
- 238000003364 immunohistochemistry Methods 0.000 description 37
- 230000004083 survival effect Effects 0.000 description 32
- 238000012360 testing method Methods 0.000 description 29
- 238000010200 validation analysis Methods 0.000 description 29
- 238000010824 Kaplan-Meier survival analysis Methods 0.000 description 25
- 101001012157 Homo sapiens Receptor tyrosine-protein kinase erbB-2 Proteins 0.000 description 24
- 102100030086 Receptor tyrosine-protein kinase erbB-2 Human genes 0.000 description 24
- 239000000090 biomarker Substances 0.000 description 20
- 238000009121 systemic therapy Methods 0.000 description 15
- 102100027671 Transcriptional repressor CTCF Human genes 0.000 description 14
- 239000000523 sample Substances 0.000 description 13
- 238000002512 chemotherapy Methods 0.000 description 12
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 10
- 238000012549 training Methods 0.000 description 10
- 108091093088 Amplicon Proteins 0.000 description 9
- 238000004458 analytical method Methods 0.000 description 9
- 238000011161 development Methods 0.000 description 8
- 230000007170 pathology Effects 0.000 description 8
- 210000001519 tissue Anatomy 0.000 description 8
- 238000002648 combination therapy Methods 0.000 description 7
- 238000002790 cross-validation Methods 0.000 description 7
- 238000012217 deletion Methods 0.000 description 7
- 230000037430 deletion Effects 0.000 description 7
- 238000012085 transcriptional profiling Methods 0.000 description 7
- 210000004881 tumor cell Anatomy 0.000 description 7
- 108010009392 Cyclin-Dependent Kinase Inhibitor p16 Proteins 0.000 description 6
- 102100024458 Cyclin-dependent kinase inhibitor 2A Human genes 0.000 description 6
- 108700039887 Essential Genes Proteins 0.000 description 6
- 238000009826 distribution Methods 0.000 description 6
- 108010021064 CTLA-4 Antigen Proteins 0.000 description 5
- 102000008203 CTLA-4 Antigen Human genes 0.000 description 5
- 101000605639 Homo sapiens Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha isoform Proteins 0.000 description 5
- 102100038332 Phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha isoform Human genes 0.000 description 5
- 239000012188 paraffin wax Substances 0.000 description 5
- 230000009885 systemic effect Effects 0.000 description 5
- STUWGJZDJHPWGZ-LBPRGKRZSA-N (2S)-N1-[4-methyl-5-[2-(1,1,1-trifluoro-2-methylpropan-2-yl)-4-pyridinyl]-2-thiazolyl]pyrrolidine-1,2-dicarboxamide Chemical compound S1C(C=2C=C(N=CC=2)C(C)(C)C(F)(F)F)=C(C)N=C1NC(=O)N1CCC[C@H]1C(N)=O STUWGJZDJHPWGZ-LBPRGKRZSA-N 0.000 description 4
- 208000006265 Renal cell carcinoma Diseases 0.000 description 4
- 206010041067 Small cell lung cancer Diseases 0.000 description 4
- 229950010482 alpelisib Drugs 0.000 description 4
- 230000003321 amplification Effects 0.000 description 4
- 238000003199 nucleic acid amplification method Methods 0.000 description 4
- 238000003908 quality control method Methods 0.000 description 4
- 208000000587 small cell lung carcinoma Diseases 0.000 description 4
- 229940121358 tyrosine kinase inhibitor Drugs 0.000 description 4
- 239000005483 tyrosine kinase inhibitor Substances 0.000 description 4
- 208000037844 advanced solid tumor Diseases 0.000 description 3
- 230000015556 catabolic process Effects 0.000 description 3
- 238000010586 diagram Methods 0.000 description 3
- 108091008039 hormone receptors Proteins 0.000 description 3
- 239000003112 inhibitor Substances 0.000 description 3
- 230000035755 proliferation Effects 0.000 description 3
- 238000003757 reverse transcription PCR Methods 0.000 description 3
- 238000012163 sequencing technique Methods 0.000 description 3
- 150000004917 tyrosine kinase inhibitor derivatives Chemical class 0.000 description 3
- 108020004414 DNA Proteins 0.000 description 2
- 240000008168 Ficus benjamina Species 0.000 description 2
- 238000003657 Likelihood-ratio test Methods 0.000 description 2
- 108700020796 Oncogene Proteins 0.000 description 2
- 239000002671 adjuvant Substances 0.000 description 2
- 238000009098 adjuvant therapy Methods 0.000 description 2
- 210000004027 cell Anatomy 0.000 description 2
- 210000003169 central nervous system Anatomy 0.000 description 2
- 239000003795 chemical substances by application Substances 0.000 description 2
- 238000012790 confirmation Methods 0.000 description 2
- 238000011985 exploratory data analysis Methods 0.000 description 2
- 238000007901 in situ hybridization Methods 0.000 description 2
- 238000009092 lines of therapy Methods 0.000 description 2
- 238000001325 log-rank test Methods 0.000 description 2
- 210000004072 lung Anatomy 0.000 description 2
- JMANVNJQNLATNU-UHFFFAOYSA-N oxalonitrile Chemical compound N#CC#N JMANVNJQNLATNU-UHFFFAOYSA-N 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- 102000003998 progesterone receptors Human genes 0.000 description 2
- 108090000468 progesterone receptors Proteins 0.000 description 2
- 230000035945 sensitivity Effects 0.000 description 2
- 238000013517 stratification Methods 0.000 description 2
- VEEGZPWAAPPXRB-BJMVGYQFSA-N (3e)-3-(1h-imidazol-5-ylmethylidene)-1h-indol-2-one Chemical compound O=C1NC2=CC=CC=C2\C1=C/C1=CN=CN1 VEEGZPWAAPPXRB-BJMVGYQFSA-N 0.000 description 1
- 102100033793 ALK tyrosine kinase receptor Human genes 0.000 description 1
- 206010069754 Acquired gene mutation Diseases 0.000 description 1
- 208000009304 Acute Kidney Injury Diseases 0.000 description 1
- 239000000275 Adrenocorticotropic Hormone Substances 0.000 description 1
- 101100004280 Caenorhabditis elegans best-2 gene Proteins 0.000 description 1
- 101800000414 Corticotropin Proteins 0.000 description 1
- 230000009946 DNA mutation Effects 0.000 description 1
- 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 description 1
- 102100031181 Glyceraldehyde-3-phosphate dehydrogenase Human genes 0.000 description 1
- 101000779641 Homo sapiens ALK tyrosine kinase receptor Proteins 0.000 description 1
- 101000686031 Homo sapiens Proto-oncogene tyrosine-protein kinase ROS Proteins 0.000 description 1
- 239000005411 L01XE02 - Gefitinib Substances 0.000 description 1
- 239000005551 L01XE03 - Erlotinib Substances 0.000 description 1
- 239000002146 L01XE16 - Crizotinib Substances 0.000 description 1
- 206010050513 Metastatic renal cell carcinoma Diseases 0.000 description 1
- 208000009525 Myocarditis Diseases 0.000 description 1
- 102100027467 Pro-opiomelanocortin Human genes 0.000 description 1
- 102100023347 Proto-oncogene tyrosine-protein kinase ROS Human genes 0.000 description 1
- 239000013616 RNA primer Substances 0.000 description 1
- 238000011529 RT qPCR Methods 0.000 description 1
- 208000033626 Renal failure acute Diseases 0.000 description 1
- 206010067584 Type 1 diabetes mellitus Diseases 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 201000011040 acute kidney failure Diseases 0.000 description 1
- 201000005180 acute myocarditis Diseases 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 230000000996 additive effect Effects 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- 239000004037 angiogenesis inhibitor Substances 0.000 description 1
- 229940121369 angiogenesis inhibitor Drugs 0.000 description 1
- 239000000611 antibody drug conjugate Substances 0.000 description 1
- 229940049595 antibody-drug conjugate Drugs 0.000 description 1
- 229940034982 antineoplastic agent Drugs 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 238000003556 assay Methods 0.000 description 1
- 210000000988 bone and bone Anatomy 0.000 description 1
- 238000007475 c-index Methods 0.000 description 1
- 238000004422 calculation algorithm Methods 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 206010009887 colitis Diseases 0.000 description 1
- 238000009096 combination chemotherapy Methods 0.000 description 1
- IDLFZVILOHSSID-OVLDLUHVSA-N corticotropin Chemical compound C([C@@H](C(=O)N[C@@H](CO)C(=O)N[C@@H](CCSC)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC=1NC=NC=1)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)NCC(=O)N[C@@H](CCCCN)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](C(C)C)C(=O)NCC(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CCCCN)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CC(N)=O)C(=O)NCC(=O)N[C@@H](C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CO)C(=O)N[C@@H](C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC=1C=CC=CC=1)C(O)=O)NC(=O)[C@@H](N)CO)C1=CC=C(O)C=C1 IDLFZVILOHSSID-OVLDLUHVSA-N 0.000 description 1
- 229960000258 corticotropin Drugs 0.000 description 1
- 229960005061 crizotinib Drugs 0.000 description 1
- KTEIFNKAUNYNJU-GFCCVEGCSA-N crizotinib Chemical compound O([C@H](C)C=1C(=C(F)C=CC=1Cl)Cl)C(C(=NC=1)N)=CC=1C(=C1)C=NN1C1CCNCC1 KTEIFNKAUNYNJU-GFCCVEGCSA-N 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 238000012774 diagnostic algorithm Methods 0.000 description 1
- 239000000104 diagnostic biomarker Substances 0.000 description 1
- 238000001983 electron spin resonance imaging Methods 0.000 description 1
- 102000052116 epidermal growth factor receptor activity proteins Human genes 0.000 description 1
- 108700015053 epidermal growth factor receptor activity proteins Proteins 0.000 description 1
- 229960001433 erlotinib Drugs 0.000 description 1
- AAKJLRGGTJKAMG-UHFFFAOYSA-N erlotinib Chemical compound C=12C=C(OCCOC)C(OCCOC)=CC2=NC=NC=1NC1=CC=CC(C#C)=C1 AAKJLRGGTJKAMG-UHFFFAOYSA-N 0.000 description 1
- 210000003238 esophagus Anatomy 0.000 description 1
- 102000015694 estrogen receptors Human genes 0.000 description 1
- 108010038795 estrogen receptors Proteins 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 230000007717 exclusion Effects 0.000 description 1
- 238000010195 expression analysis Methods 0.000 description 1
- 229960002258 fulvestrant Drugs 0.000 description 1
- 230000006870 function Effects 0.000 description 1
- 230000004927 fusion Effects 0.000 description 1
- 229960002584 gefitinib Drugs 0.000 description 1
- XGALLCVXEZPNRQ-UHFFFAOYSA-N gefitinib Chemical compound C=12C=C(OCCCN3CCOCC3)C(OC)=CC2=NC=NC=1NC1=CC=C(F)C(Cl)=C1 XGALLCVXEZPNRQ-UHFFFAOYSA-N 0.000 description 1
- 108020004445 glyceraldehyde-3-phosphate dehydrogenase Proteins 0.000 description 1
- 208000006454 hepatitis Diseases 0.000 description 1
- 231100000283 hepatitis Toxicity 0.000 description 1
- 230000007062 hydrolysis Effects 0.000 description 1
- 238000006460 hydrolysis reaction Methods 0.000 description 1
- 208000003532 hypothyroidism Diseases 0.000 description 1
- 230000002989 hypothyroidism Effects 0.000 description 1
- 210000000987 immune system Anatomy 0.000 description 1
- 230000010354 integration Effects 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 210000004185 liver Anatomy 0.000 description 1
- 238000007403 mPCR Methods 0.000 description 1
- 238000010197 meta-analysis Methods 0.000 description 1
- 230000033607 mismatch repair Effects 0.000 description 1
- YOHYSYJDKVYCJI-UHFFFAOYSA-N n-[3-[[6-[3-(trifluoromethyl)anilino]pyrimidin-4-yl]amino]phenyl]cyclopropanecarboxamide Chemical compound FC(F)(F)C1=CC=CC(NC=2N=CN=C(NC=3C=C(NC(=O)C4CC4)C=CC=3)C=2)=C1 YOHYSYJDKVYCJI-UHFFFAOYSA-N 0.000 description 1
- 102000039446 nucleic acids Human genes 0.000 description 1
- 108020004707 nucleic acids Proteins 0.000 description 1
- 150000007523 nucleic acids Chemical class 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 210000001428 peripheral nervous system Anatomy 0.000 description 1
- 239000013641 positive control Substances 0.000 description 1
- 230000011514 reflex Effects 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 230000037439 somatic mutation Effects 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 210000002784 stomach Anatomy 0.000 description 1
- 230000002195 synergetic effect Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 238000013520 translational research Methods 0.000 description 1
- 229940049679 trastuzumab deruxtecan Drugs 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
-
- G—PHYSICS
- G16—INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
- G16B—BIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
- G16B25/00—ICT specially adapted for hybridisation; ICT specially adapted for gene or protein expression
- G16B25/10—Gene or protein expression profiling; Expression-ratio estimation or normalisation
-
- G—PHYSICS
- G16—INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
- G16B—BIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
- G16B20/00—ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations
-
- 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
- C12Q2531/00—Reactions of nucleic acids characterised by
- C12Q2531/10—Reactions of nucleic acids characterised by the purpose being amplify/increase the copy number of target nucleic acid
- C12Q2531/113—PCR
-
- 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
- C12Q2535/00—Reactions characterised by the assay type for determining the identity of a nucleotide base or a sequence of oligonucleotides
- C12Q2535/122—Massive parallel sequencing
-
- 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
- C12Q2600/106—Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
-
- 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
- CPIs checkpoint inhibitors
- this focused application reduce unnecessary activation of the immune system via checkpoint inhibitors in subjects that are unlikely to respond to the therapy, thus reducing the number of adverse events in these subjects, which may include colitis, hepatitis, adrenocorticotropic hormone insufficiency, hypothyroidism, type 1 diabetes, acute kidney injury and myocarditis.
- PD-L1 immunohistochemistry is required for treatment in many tumor types and serves as a companion diagnostic biomarker; although antibodies, staining platforms, PD-L1 expressing cells included in scoring algorithms, and cutoffs vary across tumor types 4 4 .
- TMB-H TMB high
- CGP genomic profiling
- biomarkers enabling the identification of PD-(L)1 monotherapy benefit is of particular importance in tumor types where only combination therapy regimens are approved (or monotherapy is only approved in later lines) as combination regimens have increased clinical and financial toxicity and a recent meta-analysis demonstrating essentially no evidence for additive or synergistic benefit between PD-(L) 1 therapies and other agents in approved combination regimens 33 .
- CGP + qTP quantitative transcriptomic profiling
- NCT03061305 an observational clinical trial evaluating the impact of molecular profiling on patients with advanced solid tumors — the inventors have developed and validated an integrated Immunotherapy Response Score (IRS) that predicts pan-solid tumor PD-(L) 1 benefit by both real- world progression free survival (rwPFS) and overall survival (OS) by an analytically and clinically validated CGP + qTP laboratory developed test (LDT) applicable to minute formalin-fixed paraffin-embedded (FFPE) tissue specimens.
- IRS Immunotherapy Response Score
- Some aspects of the present invention are directed to a method of treatment, comprising: (a)(i) measuring expression levels of RNA transcripts for at least two of PD-1, TOP2A, PD-L1 and ADAM 12 obtained from a tumor specimen from a subject; (b) measuring tumor mutation burden (TMB) in the biological sample and log2 transforming the TMB measurement to provide a transformed TMB measurement; (c) calculating an Immunotherapy Response Score (IRS) from the expression levels of the RNA transcripts of the at least two of PD-1, TOP2A, PD-L1 and ADAM 12 obtained in step (a), and the transformed TMB measurement, wherein the IRS is positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy, and obtaining an IRS having a value indicating a beneficial response to checkpoint inhibitor therapy; and (d) administering the checkpoint inhibitor therapy to the subject.
- TMB tumor mutation burden
- the step of measuring expression levels of RNA transcripts comprises measuring expression levels of RNA transcripts for at least three of PD-1, TOP2A, PD-L1 and ADAM12
- the step of calculating an IRS comprises calculating the IRS from the expression levels of the RNA transcripts of the at least three of PD-1, TOP2A, PD-L1 and ADAM12, and the transformed TMB measurement.
- the step of measuring expression levels of RNA transcripts comprises measuring expression levels of RNA transcripts for all of PD-1, TOP2A, PD-L1 and ADAM12, and the step of calculating an IRS comprises calculating the IRS from the expression levels of the RNA transcripts of all of PD-1, TOP2A, PD-L1 and ADAM12, and the transformed TMB measurement.
- the step of measuring expression levels of RNA transcripts comprises measuring expression levels of RNA transcripts of at least PD-1 and PD- Ll, and the step of calculating an IRS comprises calculating the IRS from the expression levels of the RNA transcripts of at least PD-1 and PD-L1, and the transformed TMB measurement.
- the step of measuring expression levels of RNA transcripts comprises measuring expression levels of RNA transcripts of PD-1, PD-L1, and ADAM 12 and wherein the step of calculating an IRS comprises calculating the IRS from the expression levels of the RNA transcripts of PD-1, PD-L1, and ADAM12, and the transformed TMB measurement.
- step (a) further comprises ii) measuring expression levels of RNA transcripts for at least one reference gene in the biological sample, and iii) normalizing the measured expression levels of the measured RNA transcripts of the at least two, the at least three, or all of PD-1, TOP2A, PD-L1 and ADAM 12 against the level of the RNA transcripts of the at least one reference gene to provide normalized expression levels of the PD-1, TOP2A, PD-L1 and ADAM12 RNA transcripts.
- the expression levels of RNA transcripts used to calculate the IRS comprises normalized expression levels of RNA transcripts.
- step (a) further comprises iv) median centering the measured expression levels of RNA transcripts of the at least two, the at least three, or all of PD-1, TOP2A, PD- L1 and ADAM12, prior or after to normalizing the expression levels of the measured RNA transcripts.
- step (a) further comprises v) log2 transforming the measured expression levels, the median centered expression levels, the normalized expression levels or the median centered normalized expression levels of RNA transcripts of the at least two, the at least three, or all of PD-1, TOP2A, PD-L1 and ADAM12, and wherein the expression levels utilized to calculate the IRS in step c are transformed expression levels, transformed median centered expression levels, transformed normalized expression levels or transformed median centered normalized expression levels.
- the transformed PD-1 level, the transformed PD-L1 level, the transformed ADAM 12 level, the transformed TOP2A level are each one of a transformed expression level, a transformed median centered expression level, a transformed normalized expression level, or a transformed median centered normalized expression level.
- the transformed PD-1 level, the transformed PD-L1 level, the transformed ADAM 12 level, the transformed TOP2A level are each one of a transformed expression level, a transformed median centered expression level, a transformed normalized expression level, or a transformed median centered normalized expression level.
- the IRS value indicating beneficial response to checkpoint inhibitor therapy is approximately 0.87 or more. In some embodiments, the IRS value indicating beneficial response to checkpoint inhibitor therapy is 0.873569 or higher.
- the one or more reference genes comprise three genes selected from CIAO1, EIF2B1, HMBS, CTCF, GGNBP2, ITGB7, MYC and SLC4A1AP. In some embodiments, the one or more reference genes comprise the combination of CIAO1, EIF2B1 and HMBS.
- the tumor specimen is a formalin-fixed paraffin-embedded
- the tumor specimen contains at least 20% tumor content.
- the tumor specimen is breast cancer, central or peripheral nervous system cancer, cancer of unknown primary, colorectal cancer, endometrial cancer, a gastrointestinal stromal tumor, glioma, hepatobiliary cancer, neuroendocrine cancer, ovarian cancer, pancreatic cancer, prostate cancer salivary gland cancer, sarcoma, or thyroid cancer.
- the tumor specimen is assessed as having microsatellite instability-low or microsatellite stability. In some embodiments, the tumor specimen is assessed as having a low tumor mutational burden, wherein low tumor mutational burden is classified as having less than 10 mutations per megabase (muts/Mb). In some embodiments, the expression levels of RNA transcripts are measured using PCR and next-generation sequencing.
- the checkpoint inhibitor is an anti-PD-1 antibody, an anti- CTLA-4 antibody, an anti-PD-Ll antibody, or an anti-PD-L2.
- the checkpoint inhibitor is nivolumab, pembrolizumab, atezolizumab, cemiplimab, durvalumab, pidilizumab, atezolimumab, PDR001, BMS- 936559, avelumab, ipilimumab, or SHR-1210.
- the checkpoint inhibitor therapy is administered as a monotherapy. In some embodiments, the checkpoint inhibitor therapy is administered in combination with one or more other chemotherapeutic agents.
- the tumor specimen shows a TPS score of 1-49%.
- the checkpoint inhibitor is administered as part of a 1 st line treatment regimen. In some embodiments, the checkpoint inhibitor is administered as part of a 2 nd line treatment regimen or higher.
- Some aspects of the present disclosure are directed to a method of identifying a subject that would benefit from checkpoint inhibitor therapy, comprising: (a) measuring expression levels of RNA transcripts for PD-1, TOP2A, PD-L1 and ADAM12, and one or more reference genes in a biological sample obtained from a tumor specimen from the subject, wherein the one or more reference gene comprises three genes selected from CIAO1, EIF2B1, HMBS, CTCF, GGNBP2, ITGB7, MYC and SLC4A1AP; (b) log2 transforming, median centering, and normalizing the measured expression levels of the RNA transcripts against the level of the RNA transcripts of at the least one reference gene to provide normalized levels of the RNA transcripts; (c) measuring tumor mutation burden (TMB) in the biological sample and log2 transforming the TMB measurement to provide a transformed TMB measurement; (d) calculating a Immunotherapy Response Score (IRS) from the transformed normalized levels of the RNA transcripts of PD-1, TOP2A
- the tumor specimen is from a cancer not approved for labeled use of the checkpoint inhibitor therapy.
- the tumor specimen is assessed as having microsatellite instability-low or microsatellite stability.
- the tumor specimen is assessed as having a low tumor mutational burden, wherein low tumor mutational burden is classified as less than 10 mutations per megabase (muts/Mb).
- the tumor specimen shows a TPS score of 1-49%.
- Some aspects of the present disclosure are directed to a method of identifying a subject that would benefit from checkpoint inhibitor therapy, comprising: (a) receiving, by a processor, measured expression levels of RNA transcripts for at least two of PD-1, TOP2A, PD-L1 and ADAM12, and at least one reference gene in a biological sample obtained from a tumor specimen from the subject; (b) log2 transforming, median centering, and normalizing, by a processor, the measured expression levels of the RNA transcripts against the level of the RNA transcripts of at the least one reference gene to provide transformed normalized levels of the RNA transcripts; (c) receiving, by a processor, measured tumor mutation burden (TMB) in the biological sample; (d) log2 transforming, by a processor, the TMB measurement to provide a transformed TMB measurement; (e) calculating, by a processor, an Immunotherapy Response Score (IRS) from the transformed normalized levels of the RNA transcripts of the at least two of PD-1, TOP2A, PD-
- the IRS value indicating beneficial response to checkpoint inhibitor therapy is approximately 0.87 or more. In some embodiments, the IRS value indicating beneficial response to checkpoint inhibitor therapy is 0.873569 or higher.
- the one or more reference genes comprise three genes selected from CIAO1, EIF2B1, HMBS, CTCF, GGNBP2, ITGB7, MYC and SLC4A1AP. In some embodiments, the one or more reference genes comprise the combination of CIAO1, EIF2B1 and HMBS.
- Some aspects of the present disclosure are directed to a method of treatment of a subject in need thereof with a checkpoint inhibitor therapy, comprising administering to said subject the checkpoint inhibitor therapy, wherein said subject in need thereof was previously identified by a method comprising: (a) measuring expression levels of RNA transcripts for at least two of PD-1, TOP2A, PD-L1 and ADAM12, and one or more reference genes in a biological sample obtained from a tumor specimen from the subject, wherein the one or more reference gene comprises three genes selected from CIAO1, EIF2B1, HMBS, CTCF, GGNBP2, ITGB7, MYC and SLC4A1AP; (b) log2 transforming, median centering, and normalizing the measured expression levels of the RNA transcripts against the level of the RNA transcripts of at the least one reference gene to provide transformed normalized levels of the RNA transcripts; (c) measuring tumor mutation burden (TMB) in the biological sample and log2 transforming the TMB measurement to provide a transformed TMB measurement; (d
- the IRS value indicating beneficial response to checkpoint inhibitor therapy is approximately 0.87 or more. In some embodiments, the IRS value indicating beneficial response to checkpoint inhibitor therapy is 0.873569 or higher.
- the one or more reference genes comprise the combination of HMBS, CIAO1 and EIF2B1.
- the tumor specimen is from a cancer not approved for labeled use of the checkpoint inhibitor therapy.
- the tumor specimen is assessed as having microsatellite instability-low or microsatellite stability.
- the tumor specimen is assessed as having a low tumor mutational burden, wherein low tumor mutational burden is classified as less than 10 mutations per megabase (muts/Mb).
- the checkpoint inhibitor therapy is administered as a monotherapy. In some embodiments, the checkpoint inhibitor therapy is administered in combination with one or more other chemotherapeutic agents. In some embodiments, the tumor specimen shows a TPS score of 1-49%.
- the tumor specimen is a formalin-fixed paraffin-embedded (FFPE) tumor specimen.
- the tumor specimen is breast cancer, central or peripheral nervous system cancer, cancer of unknown primary, colorectal cancer, endometrial cancer, a gastrointestinal stromal tumor, glioma, hepatobiliary cancer, neuroendocrine cancer, ovarian cancer, pancreatic cancer, prostate cancer salivary gland cancer, sarcoma, or thyroid cancer.
- the expression levels of RNA transcripts are measured using PCR and next-generation sequencing.
- the checkpoint inhibitor is an anti-PD-1 antibody, an anti- CTLA-4 antibody, an anti-PD-Ll antibody, or an anti-PD-L2.
- the checkpoint inhibitor is nivolumab, pembrolizumab, atezolizumab, cemiplimab, durvalumab, pidilizumab, atezolimumab, PDR001, BMS- 936559, avelumab, ipilimumab, or SHR-1210.
- Some aspects of the present invention are directed to a method of treatment of a subject in need thereof with a checkpoint inhibitor therapy, comprising administering to said subject the checkpoint inhibitor therapy, wherein said subject in need thereof was previously identified by a method comprising: a. measuring expression levels of RNA transcripts for PD-1 and PD-L2 obtained from a tumor specimen from a subject, b. Iog2 transforming, median centering, and normalizing the measured expression levels of the RNA transcripts against the level of the RNA transcripts of the one or more reference genes to provide transformed normalized levels of the RNA transcripts; c. measuring tumor mutation burden (TMB) in the biological sample and log2 transforming the TMB measurement to provide a transformed TMB measurement; and d.
- TMB tumor mutation burden
- IRS Immunotherapy Response Score
- step a. further comprises measuring the expression level of RNA transcripts for ADAM12.
- step a. further comprises measuring the expression level of RNA transcripts for PD-L1.
- step a. further comprises measuring the expression level of RNA transcripts for CD4.
- step a. further comprises measuring the expression level of RNA transcripts for VTCN1.
- step a. further comprises measuring expression levels of RNA transcripts for at least one reference gene in the biological sample
- step b. comprises normalizing the measured expression levels of the other measured RNA transcripts against the level of the RNA transcripts of at the least one reference gene to provide normalized levels of the other RNA transcripts
- step c. comprises calculating the IRS from the normalized levels.
- the IRS value indicating beneficial response to checkpoint inhibitor therapy is 10 or more.
- the one or more reference genes comprise three genes selected from LRP1, MRPL13, TBP, HMBS, ITGB7, MYC, CIAO1, CTCF, EIF2B1, GGNBP2, SLC4A1AP.
- the tumor specimen is a formalin-fixed paraffin-embedded (FFPE) tumor specimen.
- the tumor specimen is adrenal cancer, biliary cancer, bladder cancer, brain cancer, breast cancer, cervical cancer, colon cancer, rectum cancer, endometrial cancer, esophageal cancer, head or neck cancer, kidney cancer, liver cancer, non-small cell lung cancer, lung cancer, lymphoma, melanoma, meninges cancer, non-melanoma skin cancer, ovarian cancer, pancreatic cancer, prostate cancer, sarcoma, small intestine cancer, or stomach cancer.
- FFPE formalin-fixed paraffin-embedded
- the expression levels of RNA transcripts are measured using PCR and next-generation sequencing.
- the checkpoint inhibitor is an anti-PD-1 antibody, an anti- CTLA-4 antibody, an anti-PD-Ll antibody, or an anti-PD-L2.
- the checkpoint inhibitor is nivolumab, pembrolizumab, atezolizumab, durvalumab, pidilizumab, PDR001, BMS- 936559, avelumab, or SHR-1210.
- Some aspects of the present disclosure are directed to a method of identifying a subject that would benefit from checkpoint inhibitor therapy, comprising: a. measuring expression levels of RNA transcripts for PD-1, PD-L2 and, optionally, expression levels of RNA transcripts for one or more of CD4, ADAM12, PD-L1, and VTCN1, and at least one reference gene in a biological sample obtained from a tumor specimen from the subject; b. Iog2 transforming, median centering, and normalizing the measured expression levels of the RNA transcripts against the level of the RNA transcripts of at the least one reference gene to provide normalized levels of the RNA transcripts; c.
- TMB tumor mutation burden
- IRS Immunotherapy Response Score
- the tumor specimen is from a cancer not approved for labeled use of the checkpoint inhibitor therapy.
- the calculated IRS value indicates that the median time-to-next-treatment (TNTT) is 24 months or greater.
- Some aspects of the present disclosure are directed to a method of identifying a subject that would benefit from checkpoint inhibitor therapy, comprising: a. receiving, by a processor, measured expression levels of RNA transcripts for PD-1, PD-L2 and, optionally, expression levels of RNA transcripts for one or more of CD4, ADAM12, PD-L1, and VTCN1, and at least one reference gene in a biological sample obtained from a tumor specimen from the subject; b. Iog2 transforming, median centering, and normalizing, by a processor, the measured expression levels of the RNA transcripts against the level of the RNA transcripts of at the least one reference gene to provide normalized levels of the RNA transcripts; c.
- TMB tumor mutation burden
- Iog2 transforming, by a processor, the TMB measurement to provide a transformed TMB measurement
- e. calculating, by a processor, a Immunotherapy Response Score (IRS) from the normalized levels of the RNA transcripts of PD-1, PD-L2 and, optionally, one or more of CD4, ADAM12, PD-L1, and VTCN1, and the transformed TMB measurement, that is positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy
- f. providing a determination if the subject has a checkpoint inhibitor responsive cancer.
- Some aspects of the present disclosure are directed to a method of treatment of a subject in need thereof with a checkpoint inhibitor therapy, comprising administering to said subject the checkpoint inhibitor therapy, wherein said subject in need thereof was previously identified by a method comprising: a.
- TMB tumor mutation burden
- IRS Immunotherapy Response Score
- the IRS is calculated using the Cox model as 10 times the inverse of the patient hazard ratio as compared to the median hazard rate.
- the IRS value indicating beneficial response to checkpoint inhibitor therapy is 10 or more.
- Some aspects of the present disclosure are directed to a method of treatment, comprising: a. measuring expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and at least one reference gene in a biological sample obtained from a tumor specimen from a subject; b. Iog2 transforming, median centering, and normalizing the measured expression levels of the RNA transcripts of PD-1, PD-L2, CD4, and ADAM12 against the level of the RNA transcripts of at the least one reference gene to provide normalized levels of the RNA transcripts of PD-1, PD-L2, CD4, and ADAM12; c.
- TMB tumor mutation burden
- IRS Immunotherapy Response Score
- the IRS value indicating beneficial response to checkpoint inhibitor therapy is 10 or more.
- the tumor specimen is a formalin-fixed paraffin-embedded (FFPE) tumor specimen.
- the tumor specimen is adrenal cancer, biliary cancer, bladder cancer, brain cancer, breast cancer, cervical cancer, colon cancer, rectum cancer, endometrial cancer, esophageal cancer, head or neck cancer, kidney cancer, liver cancer, non-small cell lung cancer, lung cancer, lymphoma, melanoma, meninges cancer, non-melanoma skin cancer, ovarian cancer, pancreatic cancer, prostate cancer, sarcoma, small intestine cancer, or stomach cancer.
- the expression levels of RNA transcripts are measured using PCR and next-generation sequencing.
- the checkpoint inhibitor is an anti-PD-1 antibody, an anti-CTLA- 4 antibody, an anti-PD-Ll antibody, or an anti-PD-L2.
- the checkpoint inhibitor is nivolumab, pembrolizumab, atezolizumab, durvalumab, pidilizumab, PDR001, BMS- 936559, avelumab, or SHR-1210.
- FIG. 1 provides a flowchart of the process used to obtain the present methods
- Fig. 2 shows expression by 3’ and 5’ amplicons for PD-L1 or PD-1 highly correlate and therefore were averaged together.
- Fig. 3 shows the partial-AIC/BIC for each tested model when trained on all 708 samples in dataset (i.e., the full dataset).
- Fig. 4 shows the median partial-AIC and log-likelihood for each model when trained on a random 2/3 of dataset x 100 iterations. Also shown is the median log-likelihood score of the test sets (the 1/3 left-out).
- Fig. 5 shows the process by which the IRS scores were divided into 3 groups, a low, medium, and high group, wherein the high group has the greatest benefit from ICI treatment.
- Fig. 6 shows pembrolizumab TTNT is correlated to overall survival (OS).
- Fig. 7 shows covariant correlations for the top 10 biomarkers.
- Fig. 8 shows survival for pembrolizumab (pembro) and chemotherapy (chemo) high/medium/low IRS groups, showing that the IRS score determined by the methods disclosed herein are predictive of response to ICI instead of predictive of overall effectiveness of any cancer treatment.
- Fig. 9 shows real world progression-free survival per low/medium/high IRS groups with pembro or chemo treatment.
- Fig. 10 shows real world progression-free survival of NSCLC patients and other cancer patients per low/medium/high IRS groups with pembro.
- Fig. 11 shows IRG rates (i.e., IRS groups) for on-label (Melanoma, Lung - NSCLC, Lung - Other, Head and Neck, Lymphoma, Bladder, Esophagus, Biliary, Stomach, Cervical, Liver, Kidney, Melanoma) and off-label cancers.
- IRG rates i.e., IRS groups
- on-label Melanoma, Lung - NSCLC, Lung - Other, Head and Neck, Lymphoma, Bladder, Esophagus, Biliary, Stomach, Cervical, Liver, Kidney, Melanoma
- Fig. 12 shows IRG rates across Strata Trial cohort by cancer type.
- Fig. 13 shows Pembro monotherapy vs pembro in combination with chemotherapy for IRS groups.
- Fig. 14 shows real world progression-free survival for IRS groups divided into TMB- High (TMB-H) and TMB-Low (TMB-L) groups.
- TMB-High is defined as 10 or more mutations per megabase.
- TMB low is less than 10 mutations per megabase.
- Fig. 15 shows rates of Immunotherapy Response Groups vs. TMB-H/L.
- Fig. 16 shows real world progression-free survival of Pembro treated patients by IRG and sample tumor content.
- Fig. 17 shows real world progression-free survival of Chemo treated patients by IRG and sample tumor content.
- Fig. 18 shows the 2/3-dataset brute force search for the best 2 covariates to add to the bare-bones model (PD-1, PD-L2, and TMB). The model with the lowest AIC was selected to derive train/test statistics for that cut.
- Fig. 19 is the results of the small model search showing the claimed method with ADAM 12+CD4+PD- 1 +PD-L2+TMB .
- Fig. 20 shows the results of backward selection starting with 21 markers and TMB. A multivariant fit was performed and the least significant markers dropped and removed. Partial AIC was compared before and after drop.
- Fig. 21 shows the best model via brute-force training on a full dataset.
- Fig. 22 shows the best model with PD-L1 added via brute-force training on a full dataset.
- Fig. 23 shows the bare-bones model with just PD-1, PD-L2 and TMB trained on the full data set.
- Fig. 24 shows a large model derived from backwards selection
- Fig. 25 shows coefficients from iterations of 2/3 cross-validation. “Brute-force” model without and with PD-L1, Yellow lines are final model coefficients from the model w/o PD-L1.
- Fig. 26 shows coefficients from iterations of 2/3 cross-validation. Small (“Bare- bones”) and Large (“Backward Selection”) models. Yellow lines are brute-force (“bru”) model coefficients from the model w/o PD-L1.
- Fig. 27 shows model cross validation wherein the specimen is collected after pembro start date.
- Fig. 28 shows an alternate biomarker equation with TMB + PD-L2 + PD-1+ ADAM12 + CD4 + PD-L1.
- Fig. 29 shows an alternate biomarker equation with TMB + PD-L2 + PD-1.
- Fig. 30 shows an alternate biomarker equation with TMB + PD-L2 + PD-1+
- Fig. 31 shows an alternate biomarker equation with TMB + PD-L2 + PD-1+ PD-L1.
- Fig. 32 shows an alternate biomarker equation with TMB + PD-L2 + PD-1+ CD4.
- Fig. 33 shows an alternate biomarker equation with TMB + PD-L2 + PD-1+ VTCN1.
- Fig. 34 shows an alternate biomarker equation with TMB + PD-L2 + PD-1+ CD4 +
- FIGS. 35a-c depict development of an integrative immunotherapy response rcore (IRS) model to stratify PD-(L)1 therapy benefit in patients with advanced solid tumors.
- FIG. 35a depicts real-world treatment and molecular profiling data from formalin fixed paraffin embedded (FFPE) tumor tissue from patients enrolled in the StrataTrial (NCT03061305) are collected in the Strata Clinical Molecular Database (SCMD).
- FFPE formalin fixed paraffin embedded
- NCT03061305 StrataTrial
- SCMD Strata Clinical Molecular Database
- Molecular data from both DNA (yellow) and RNA (blue) include both comprehensive genomic profiling (CGP) with both DNA and RNA components, and in-parallel quantitative transcriptional profiling (qTP) comprised of RNA from analytically and clinically validated tests.
- CGP comprehensive genomic profiling
- qTP quantitative transcriptional profiling
- a cohort of 648 patients was identified with available molecular information who were treated with a pembrolizumab (pembro; PD-1) containing systemic therapy line of treatment.
- Lasso- penalized Cox proportional hazards modeling with five-cross validation was used to develop the IRS model for predicting real world progression free survival (rwPFS; by time to next therapy), which includes tumor mutation burden (TMB; from CGP) and expression of PD-1, PD-L1, ADAM 12 and TOP2A (from qTP).
- FIG. 35b depicts that IRS stratifies pembrolizumab rwPFS in the development cohort.
- Pembrolizumab rwPFS in the development cohort stratified by IRS groups is shown by Kaplan Meier analysis with the adjusted hazard ratio (HR) and p value (adjusted by variables shown in FIG. 35c) for IRS-H vs. IRS-L.
- FIG. 35c depicts that IRS is robust to potential confounders in the development cohort. Forest plot of variables included in the adjusted Cox proportional hazards model used to evaluate the ability of IRS to stratify pembrolizumab rwPFS. Adjusted hazard ratios with 95% confidence intervals (Cis) are shown for each variable with statistically significant variables bolded.
- FIGS. 36a-f depict PD-[L]1 monotherapy real-world progression-free survival (rwPFS) and overall survival (OS) by Immunotherapy Response Score (IRS) Status.
- FIG. 36a depicts rwPFS for monotherapy pembrolizumab (pembro; PD-1 therapy) treated patients in the discovery cohort.
- Pembrolizumab monotherapy rwPFS in the development cohort stratified by IRS groups is shown by Kaplan-Meier analysis with the adjusted hazard ratio (HR) and p-value for IRS-High [H] vs. IRS-Low [L] groups.
- the number (n) of patients, events, and median rwPFS (with 95% confidence intervals [CI]) for each group are shown.
- FIG. 36b depicts Kaplan-Meier analysis as in 36a, except for OS.
- FIGS. 36c-d depict Kaplan-Meier analysis as in FIGS 36a-b, except assessing rwPFS (36c) and OS (36d) in the independent validation cohort of patients treated with non- pembrolizumab PD-(L)1 monotherapy.
- FIG. 36e depicts Forest plots of adjusted HRs with 95% CI for IRS and tumor mutation burden (TMB; TMB-High [H] >10 mutations/megabase) in otherwise equivalent models separately adjusted for IRS and TMB (H vs. L for each) in both cohorts for rwPFS and OS.
- TMB tumor mutation burden
- FIG. 36f depicts overlap of IRS-H and TMB-H populations in the 24,463 patients with informative IRS and TMB status (regardless of treatment status) in the Strata Clinical Molecular Database (SCMD).
- FIG. 37a-c depicts confirmation of the predictive nature of the Immunotherapy Response Score (IRS) biomarker.
- IRS Immunotherapy Response Score
- FIG. 37b depicts Kaplan-Meier analysis of pembrolizumab monotherapy rwPFS (purple) vs. prior systemic therapy rwPFS (yellow) in the IRS-Low [L] subset of patients (log-rank p-value shown). The number (n) of patients, events, and median rwPFS (with 95% confidence intervals [CI]) for each group are shown.
- FIG. 37c depicts Kaplan-Meier analysis of pembrolizumab monotherapy rwPFS (purple) vs. prior systemic therapy rwPFS (yellow) in the IRS-H subset of patients (log-rank p-value shown). The likelihood ratio test (LRT) p-value for interaction between pembrolizumab vs. immediately prior treatment line and IRS status (IRS-L vs. IRS-High [H]) is also shown.
- LRT likelihood ratio test
- FIGS. 38a-c depicts Immunotherapy Response Score (IRS) for predicting pembrolizumab monotherapy vs. combination chemotherapy benefit in first line NSCLC.
- FIG. 38a depicts a Kaplan-Meier analysis of pembrolizumab monotherapy rwPFS (orange) vs. pembrolizumab + chemotherapy combination therapy (yellow) in the IRS-Low [L] subset of patients (log-rank p-value shown). The number (n) of patients, events, and median rwPFS (with 95% confidence intervals [CI]) for each group are shown.
- FIG 38b depicts a Kaplan-Meier analysis of pembrolizumab monotherapy rwPFS (orange) vs.
- FIG. 38c depicts the distribution of IRS status in a separate cohort of NSCLC tumor samples with PD- L1 IHC (Fig S8) stratified by clinically relevant tumor proportion score (TPS) bins.
- FIGS. 39a-d depict pan-solid tumor distribution of immunotherapy response score (IRS) groups.
- FIG. 39a depicts that IRS groups were determined for all 24,463 patients in the Strata Clinical Molecular Database (SCMD) with informative tumor mutation burden (TMB) and gene expression data needed to generate IRS. IRS group (Low [L; light blue] vs. High [H; dark blue]) distribution is shown by box plot (numbers indicated percentages);
- FIG. 39b depicts stratification of the 24,463 patients by approved and non-approved PD-(L)1 monotherapy tumor types;
- FIG. 39c depicts a breakdown of FIG. 39b by individual tumor types;
- FIG. 39d depicts Breakdown of FIG. 39b by IRS and TMB (High [H] vs.
- NSCLC non-small cell lung cancer
- RCC renal cell carcinoma
- NMSC non-melanoma skin cancer
- SCLC small cell lung cancer
- CNS and PNS central nervous system and peripheral nervous system
- CUP cancer of unknown primary
- CRC colonrectal cancer
- GIST gastrointestinal stromal tumor
- FIG. 40 depicts Overall study diagram from the Strata Clinical Molecular Database (SCMD) used to develop and validate the Immunotherapy Response Score (IRS). Disposition of patients from the Strata Trial (NCT03061305) used to develop and validate IRS are shown. Included populations are indicated by gray boxes. As patients could contribute to multiple analyses (e.g., a subject treated with first line angiogenesis inhibitor and second line pembrolizumab could be eligible for both the “Non-IO 1st line analysis” and the “Discovery cohort” as long as they met both inclusion/exclusion criteria [including the sample was collected before both lines of therapy]), the number of shared patients is indicated by green arrows at the highest branch point. The overall SCMD population is shown in bolded yellow.
- FIG. 41 depicts Assignment of therapy lines from real world treatment data.
- NCT03061305 subjects with treatment data (treatment start and stop dates)
- standardized assignment of adjuvant/systemic therapy lines was performed accounting for adjuvant/systemic therapy, monotherapy/combination therapy, potential overlap of treatment start/stop dates and repeating lines of therapy (whether monotherapy in combination).
- Assigned treatment lines and an example of real world-progression free survival measurement by time to next therapy (TTNT ; start date of therapy to start date of subsequent therapy) are shown for a patient with metastatic renal cell carcinoma.
- FIG. 42 depicts Time to next therapy (TTNT) of patients in the Strata Clinical Molecular Database (SCMD) by line of therapy.
- TTNT Time to next therapy
- SCMD Strata Clinical Molecular Database
- the number (n) of patients, events, and median rwPFS (with 95% confidence intervals [CI]) for each group are shown, along with the overall log rank p value.
- FIGS 43a-c depict Strata Clinical Molecular Database (SCMD) non-small cell lung cancer (NSCLC) analysis.
- HR hazard ratio
- FIG. 44 depicts Correlation of real-world pembrolizumab progression-free survival (rwPFS) and overall survival (OS). Correlation for pembrolizumab rwPFS by time to next therapy (TTNT) and OS for patients in the discovery cohort with more than one line of systemic therapy. Colored boxes indicate patients discussed in the Supplementary Results.
- FIGS. 45a-d depict PD-(L)1 monotherapy real world progression free survival (rwPFS) and overall survival (OS) by tumor mutation burden (TMB) status.
- FIG. 45a depicts Pembrolizumab monotherapy (PD-1) rwPFS (by time to next therapy) in the discovery cohort stratified by TMB groups (TMB-High [H] >10 mutations/megabase by StrataNGS testing vs. TMB- Low [L]) is shown by Kaplan Meier analysis with the adjusted hazard ratio (HR) and p value for TMB-H vs. -L groups.
- HR adjusted hazard ratio
- FIG. 45b as in FIG. 45a except OS.
- FIGS 45c-d as in FIGS 45a-b, except assessing the independent validation cohort of patients treated with non-pembrolizumab PD-(L)1 monotherapy.
- FIGS. 46a-f depict Housekeeping gene selection and validation, accuracy vs qRT- PCR, and replicate amplicon correlation for the quantitative expression component of the integrated comprehensive genomic profiling and quantitative transcriptional profiling (CGP + qTP) laboratory developed test used to report the Immunotherapy Response Score (IRS). IRS is reported from an integrated CGP + qTP test that combines comprehensive genomic profiling (CGP) from the analytically and clinically validated StrataNGS test with in-parallel quantitative transcriptional profiling (qTP) by multiplex RT-PCR based next generation sequencing.
- CGP + qTP integrated comprehensive genomic profiling and quantitative transcriptional profiling laboratory developed test used to report the Immunotherapy Response Score
- IRS Immunotherapy Response Score
- 46a depicts initial pre- clinical versions of the qTP panel contained 6 “positive control” genes across two RNA primer pools previously used in the RNA fusion component of the Oncomine Focus/Precision Assay (OPA positive).
- OPA positive Oncomine Focus/Precision Assay
- FIGS. 46d-f depicts two separate PD-L1, PD-1 and ADAM12 amplicons are present in the current qTP panel (only one of two ADAM 12 amplicons was also present on all previous panels used to develop and validate IRS).
- FIGS. 47a-d depict Accuracy vs. clinical immunohistochemistry and reproducibility for the quantitative expression component of the integrated comprehensive genomic profiling and quantitative transcriptional profiling (CGP + qTP) laboratory developed test used to report the Immunotherapy Response Score (IRS). IRS is reported from an integrated CGP + qTP test that combines comprehensive genomic profiling (CGP) from the analytically and clinically validated StrataNGS test with in-parallel quantitative transcriptional profiling (qTP) by multiplex RT-PCR based next generation sequencing.
- CGP + qTP integrated comprehensive genomic profiling and quantitative transcriptional profiling laboratory developed test used to report the Immunotherapy Response Score
- IRS Immunotherapy Response Score
- 47a depicts the accuracy of the PD-L1 qTP component of IRS was validated against clinical IHC using a cohort of 276 non-small cell lung cancer (NSCLC) formalin fixed paraffin embedded (FFPE) tumor samples with reportable qTP (including tumor content [TC] > 20%) and PD-L1 IHC expression by the 22C3 clone (using tumor proportion score [TPS]) in accompanying pathology reports.
- NSCLC non-small cell lung cancer
- FFPE formalin fixed paraffin embedded
- FIG. 47b depicts accuracy of the TOP2A qTP component of IRS was validated against clinical IHC using a cohort of 956 FFPE tumor tissue samples (36 tumor types) with reportable qTP (including TC > 20%) with proliferation index (percentage of Ki67 positive tumor cells) in accompanying pathology reports.
- the Pearson correlation coefficient of qTP TOP2A expression vs. clinical proliferation index from the scatter plot is shown with 95% confidence interval [CI] and p-value, with points overlying a density heatmap and the line of best fit indicated by the dashed line.
- FIG. 47b depicts accuracy of the TOP2A qTP component of IRS was validated against clinical IHC using a cohort of 956 FFPE tumor tissue samples (36 tumor types) with reportable qTP (including TC > 20%) with proliferation index (percentage of Ki67 positive tumor cells) in accompanying pathology reports.
- the Pearson correlation coefficient of qTP TOP2A expression vs. clinical proliferation index from the scatter plot is shown with 95% confidence interval [
- FIG. 47c depicts the panel wide qTP reproducibility between operators, lots, and instrumentation was established using separate replicate nucleic acid aliquots isolated from FFPE tumor samples. Twenty-seven unique samples were assessed by two operators on different days using different library preparation instrumentation, different library preparation reagent lots, and different templating and sequencing lots and instruments. For each sample, the maximum and minimum nRPM for each target gene across all replicates was plotted (individual target gene amplicons are shown by color) and the concordance correlation coefficient was determined.
- FIG. 47d as in FIG. 47c, except reproducibility of IRS was determined by plotting the maximum and minimum IRS across all replicates for each sample and the concordance correlation coefficient was determined. Qualitative agreement of IRS status (High vs. Low) from the maximum and minimum IRS score across all replicates was also determined.
- FIG. 48 depict Lasso-penalized Cox proportional hazards regression for Immunotherapy Response Score (IRS) development.
- IRS Immunotherapy Response Score
- TMB tumor mutation burden
- the Lasso penalty term was chosen as the value which maximized the concordance index (top panel; gray line) via 5-fold cross validation, with the coefficients shown for TMB and the 23 candidate expression biomarkers vs. alpha (a) (bottom panel), resulting in a five-term model including TMB, PD-1, PD-L1, ADAM12, and TOP2A.
- FIGS. 49a-b depict Pembrolizumab overall survival (OS) by Immunotherapy Response Score (IRS) status.
- FIG. 49a depicts the Pembrolizumab OS in the discovery cohort stratified by IRS groups is shown by Kaplan Meier analysis with the adjusted hazard ratio (HR) and p value (adjusted by variables shown in b) for IRS-H vs. -L. The number (n) of patients, events, and median rwPFS (with 95% confidence intervals [CI]) for each group are shown.
- FIG. 49b depicts the Forest plot of variables included in the adjusted Cox proportional hazards model used to evaluate the ability of IRS to stratify pembrolizumab OS. Adjusted hazard ratios with 95% confidence intervals (CI) are shown for each variable. Statistically significant variables are bolded.
- FIGS. 50a-d depict Real world progression free survival (rwPFS) and overall survival (OS) by Immunotherapy Response Score (IRS) status in the validation cohort stratified by PD-1 vs. PD-L1 therapy.
- FIG. 50a depicts the rwPFS (by time to next therapy) for the monotherapy PD-L1 treated subset of the validation cohort stratified by IRS groups is shown by Kaplan Meier analysis with the adjusted hazard ratio (HR) and p value for IRS-H vs. -L groups.
- HR hazard ratio
- CI median rwPFS
- FIGS. 51a-d depict PD-(L)1 monotherapy real world progression free survival (rwPFS) and overall survival (OS) by Immunotherapy Response Score (IRS) status and Tumor Mutation Burden (TMB).
- FIG. 51a depicts the Pembrolizumab monotherapy rwPFS in the discovery cohort stratified by IRS (IRS-High [-H] vs.
- TMB-H [>10 mutations/megabase] vs. TMB-L is shown by Kaplan Meier analysis. Benjamini Hochberg (BH) adjusted p-value for pairwise log-rank test between the IRS-H/TMB-H and IRS-H/TMB-L groups is shown. The number (n) of patients, events, and median rwPFS (with 95% confidence intervals [CI]) for the analzyed groups are shown.
- FIG. 51b as in FIG. 51a, expect OS.
- FIGS. 51c&d as in FIGS. 51a&b, except assessing rwPFS (FIG. 51c) and OS (FIG. 5 Id) in the independent validation cohort of patients treated with non-pembrolizumab PD-(L)1 monotherapy.
- FIGS. 52a-d depict CDKN2A deep deletion status does not add to Immunotherapy Response Score (IRS) for predicting PD-(L)1 monotherapy real world progression free survival (rwPFS) or overall survival (OS).
- the number (n) of patients, events, and median rwPFS (with 95% confidence intervals [CI]) for each group are shown.
- the forest plot shows adjusted hazard ratios with 95% confidence intervals (CI) for IRS (IRS-H vs. IRS-L) and CDKN2A deep deletion status (CDKN2A deep deletion present vs. CDKN2A deep deletion not present) in the same adjusted model.
- FIG. 52b as in FIG. 52a, except assessing OS.
- FIGS. 53a-b depict Immunotherapy Response Score (IRS) is robust to pre-PD-(L)l sample collection timing.
- FIG. 53b depicts the PD-(L)1 rwPFS stratified by IRS group in 181 patients who otherwise would have been included in the discovery or validation cohorts but had their samples collected after starting PD-(L)1 therapy is shown by Kaplan Meier analysis with the adjusted hazard ratio (HR) and p value for IRS-H vs. IRS-L. The number (n) of patients, events, and median rwPFS (with 95% confidence intervals [CI]) for each group are shown.
- FIGS. 54a-e depict Immunotherapy Response Score (IRS) is robust to variable tumor content.
- FIG. 54a depicts a continuous tumor content term was included in the adjusted Cox proportional hazards (CPH) model for pembrolizumab real world progression free survival (rwPFS; by time to next therapy) in the overall discovery cohort (including age, gender, most common tumor type [NSCLC] vs. others, therapy type [monotherapy/combination] , and line of therapy). Adjusted hazard ratios with 95% confidence intervals (Cis) are shown for each variable with statistically significant variables bolded.
- CPH Cox proportional hazards
- rwPFS real world progression free survival
- FIG. 54b-d depicts the Pembrolizumab rwPFS binned by tumor content (20-35%, 40-70%, and >70%) and stratified by IRS groups is shown by Kaplan Meier analysis. The number (n) of patients, events, and median rwPFS (with 95% confidence intervals [CI]) for each group are shown.
- FIG. 54e depicts the PD-(L)1 rwPFS-i stratified by IRS group in 64 patients who otherwise would have been included in the discovery or validation cohorts except the tested sample tumor content was ⁇ 20% is shown by Kaplan Meier analysis with the adjusted hazard ratio (HR) and p value for IRS-H vs. -L groups. The number (n) of patients, events, and median rwPFS (with 95% confidence intervals [CI]) for each group are shown
- FIGS. 55a-d depict Additional analyses supporting the predictive nature of the Immunotherapy Response Score (IRS) biomarker.
- FIGS. 55a&b to establish the predictive nature of the IRS model, we assessed an internal comparator cohort for the pembrolizumab monotherapy cohort, consisting of the 146 patients who had received a previous line of systemic therapy prior to monotherapy pembrolizumab therapy. For each patient, real-world progression free survival (rwPFS) was determined for the line of systemic therapy immediately prior to pembrolizumab and the pembrolizumab monotherapy line, with rwPFS stratified by IRS status (see FIGS. 37a-c).
- rwPFS real-world progression free survival
- 55d depicts the Ipilimumab + nivolumab (ipi+nivo) rwPFS in 70 patients who otherwise would have been eligible for the validation cohort but received combination ipilimumab (CTLA4) + nivolumab (PD-1) therapy stratified by IRS status is shown by Kaplan Meier analysis with the adjusted hazard ratio (HR) and p value for IRS-H vs. -L groups.
- CTL4 combination ipilimumab
- PD-1 nivolumab
- FIGS. 56a-g depict Clinical utility of integrated comprehensive genomic profiling and quantitative transcriptional profiling (CGP + qTP) outside of immunotherapy treatment decision making. IRS is reported from an integrated CGP + qTP test that combines comprehensive genomic profiling (CGP) from the analytically and clinically validated StrataNGS test with in-parallel quantitative transcriptional profiling (qTP) by multiplex RT-PCR based next generation sequencing.
- CGP + qTP comprehensive genomic profiling
- StrataNGS test comprehensive genomic profiling
- qTP in-parallel quantitative transcriptional profiling
- ESRI estrogen receptor
- ER estrogen receptor
- FIG. 56b depicts the accuracy of PGR (progesterone receptor; PR) by qTP validated against clinical IHC using a cohort of 291 breast cancer formalin fixed paraffin embedded (FFPE) tumor samples with reportable qTP (including tumor content [TC] > 20%) and PR IHC expression (by % tumor cells positive) in accompanying pathology reports.
- PR progesterone receptor
- FIG. 56c ⁇ depicts the accuracy of the HER2 (ERBB2) by qTP was validated against clinical IHC using a cohort of 545 breast cancer formalin fixed paraffin embedded (FFPE) tumor samples with reportable qTP (including tumor content [TC] > 20%) and HER2 IHC expression (0, 1+, 2+ or 3+) in accompanying pathology reports.
- FFPE breast cancer formalin fixed paraffin embedded
- the threshold was set to favoring NPA and pre-specified acceptable NPA (versus PR 0% IHC) of greater than 95% was set.
- red amplified
- green not amplified [wildtype; wt]
- gray copy number status not evaluable
- PPA and NPA values 95% confidence intervals
- FIGS. 57a-c depict Exploratory analysis defining an Immunotherapy Response Score (IRS) ultra-low subset.
- IRS Immunotherapy Response Score
- 57a depicts the PD-(L)1 real-world progression free survival (rwPFS) in the combined cohorts stratified by IRS-High [H], IRS-L (I), and IRS-L (U) groups is shown by Kaplan Meier analysis with the Benjamini Hochberg (BH) adjusted p-value for pairwise log-rank test between the IRS-L (I) vs. IRS-L (U) groups and the adjusted hazard ratio (HR) and p value for IRS-L (I) vs. IRS-L (U) groups shown.
- the Cox proportional hazard model was adjusted for age, gender, most common tumor type (NSCLC vs. other), line of therapy, type of therapy (monotherapy vs.
- FIG. 57b as in FIG. 57a, except overall survival (OS).
- FIG. 57c this three group IRS classification was applied to all 24,463 patients in the Strata Clinical Molecular Database (SCMD) with valid tumor mutation burden (TMB) and gene expression data. IRS group distribution is shown by box plot (numbers indicated percentages). Stratification and breakdown of PD-(L)1 monotherapy approved tumor types is shown.
- FIGS 58a-d depict the confirmation of the predictive nature of the Immunotherapy Response Score (IRS) Biomarker when an ultra-low subset is defined.
- IRS Immunotherapy Response Score
- FIG. 58a depicts that for each patient, rwPFS was determined for the line of systemic therapy immediately prior to pembrolizumab and the pembrolizumab monotherapy line, with rwPFS stratified by IRS status.
- FIG. 58a depicts Kaplan-Meier analysis of the immediately prior systemic therapy rwPFS in the IRS-High [H], IRS-L (I), and IRS-L (U) groups (overall log-rank p-value is shown). The number (n) of patients, events, and median rwPFS (with 95% confidence intervals [CI]) for each group are shown.
- FIGS. 58b-d depict Kaplan-Meier analysis of pembrolizumab monotherapy rwPFS (purple) vs.
- Immune checkpoint inhibitors are FDA-approved and provide clinical benefit across a wide range of tumor types. However, in most indicated tumor types, only a minority of patients benefit, and additional patients benefit outside of indicated tumor types. Thus, improved diagnostic tools are required to select patients for immunotherapy treatment. Leveraging real-world pembrolizumab outcome data combined with DNA mutation and RNA expression data from a clinical NGS test for 610 diverse solid tumor patients, the inventors demonstrated that TMB, PD-L1 and PD- L2 were independent predictors of treatment benefit and that a multivariate Immunotherapy Response Score (IRS) predicted pembrolizumab benefit relative to chemotherapy across solid tumors.
- IFS Immunotherapy Response Score
- IRS scores are characterized across nearly 20,000 advanced solid tumors and showed that the proportion of patients in high IRS groups predicted the observed pembrolizumab tumor type response rates.
- CGP + qTP quantitative transcriptomic profiling
- Some aspects of the present invention are directed to a method of treatment, comprising: (a)(i) measuring expression levels of RNA transcripts for at least two of PD-1, TOP2A, PD-L1 and ADAM 12 obtained from a tumor specimen from a subject; (b) measuring tumor mutation burden (TMB) in the biological sample and log2 transforming the TMB measurement to provide a transformed TMB measurement; (c) calculating an Immunotherapy Response Score (IRS) from the expression levels of the RNA transcripts of the at least two of PD-1, TOP2A, PD-L1 and ADAM 12 obtained in step (a), and the transformed TMB measurement, wherein the IRS is positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy, and obtaining an IRS having a value indicating a beneficial response to checkpoint inhibitor therapy; and (d) administering the checkpoint inhibitor therapy to the subject.
- TMB tumor mutation burden
- the step of measuring expression levels of RNA transcripts comprises measuring expression levels of RNA transcripts for at least three of PD-1, TOP2A, PD-L1 and ADAM12
- the step of calculating an IRS comprises calculating the IRS from the expression levels of the RNA transcripts of the at least three of PD-1, TOP2A, PD-L1 and ADAM12, and the transformed TMB measurement.
- the step of measuring expression levels of RNA transcripts comprises measuring expression levels of RNA transcripts for all of PD-1, TOP2A, PD-L1 and ADAM12, and the step of calculating an IRS comprises calculating the IRS from the expression levels of the RNA transcripts of all of PD-1, TOP2A, PD-L1 and ADAM12, and the transformed TMB measurement.
- the step of measuring expression levels of RNA transcripts comprises measuring expression levels of RNA transcripts of at least PD-1 and PD- Ll, and the step of calculating an IRS comprises calculating the IRS from the expression levels of the RNA transcripts of at least PD-1 and PD-L1, and the transformed TMB measurement.
- the step of measuring expression levels of RNA transcripts comprises measuring expression levels of RNA transcripts of PD-1, PD-L1, and ADAM 12 and wherein the step of calculating an IRS comprises calculating the IRS from the expression levels of the RNA transcripts of PD-1, PD-L1, and ADAM12, and the transformed TMB measurement.
- expression levels of both PD-1 and PD-L1 are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- a high transformed level of TMB is positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- expression levels of both PD-1 and PD-L1 are as well as a high transformed level of TMB are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- expression levels of ADAM12 are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibit therapy.
- both of the expression levels of PD-L and PD-L1 as well as a high transformed level of TMB are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy, while expression levels of ADAM12 are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step (a) further comprises ii) measuring expression levels of RNA transcripts for at least one reference gene in the biological sample, and iii) normalizing the measured expression levels of the measured RNA transcripts of the at least two, the at least three, or all of PD-1, TOP2A, PD-L1 and ADAM 12 against the level of the RNA transcripts of the at least one reference gene to provide normalized expression levels of the PD-1, TOP2A, PD-L1 and ADAM 12 RNA transcripts.
- the one or more reference genes comprise one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen or more housekeeping genes.
- the reference genes are selected from CIAO1, EIF2B1, HMBS, CTCF, GGNBP2, ITGB7, MYC and SLC4A1AP.
- the one or more reference genes comprise three or more of CIAO1, EIF2B1, HMBS, CTCF, GGNBP2, ITGB7, MYC and SLC4A1AP.
- the one or more reference genes comprise the combination of CIAO1, EIF2B1 and HMBS.
- the expression levels of RNA transcripts used to calculate the IRS comprises normalized expression levels of RNA transcripts.
- normalized expression levels of both PD-1 and PD-L1 as well as a high transformed level of TMB are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- normalized expression levels of both ADAM 12 and TOP2A are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- both of the normalized expression levels of PD-L and PD-L1 as well as a high transformed level of TMB are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy, while expression levels of ADAM12 and TOP2A are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step (a) further comprises iv) median centering the measured expression levels of RNA transcripts of the at least two, the at least three, or all of PD-1, TOP2A, PD- L1 and ADAM12, prior or after to normalizing the expression levels of the measured RNA transcripts.
- step (a) further comprises v) log2 transforming the measured expression levels, the median centered expression levels, the normalized expression levels or the median centered normalized expression levels of RNA transcripts of the at least two, the at least three, or all of PD-1, TOP2A, PD-L1 and ADAM12, and wherein the expression levels utilized to calculate the IRS in step (c) are transformed expression levels, transformed median centered expression levels, transformed normalized expression levels or transformed median centered normalized expression levels.
- transformed expression levels, transformed median centered expression levels, transformed normalized expression levels or transformed median centered normalized expression levels of both PD-1 and PD-L1 as well as a high transformed level of TMB are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- transformed expression levels, transformed median centered expression levels, transformed normalized expression levels or transformed median centered normalized expression levels of both ADAM 12 and TOP2A are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- transformed expression levels, transformed median centered expression levels, transformed normalized expression levels or transformed median centered normalized expression levels of PD-L and PD-L1 as well as a high transformed level of TMB are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy, while transformed expression levels, transformed median centered expression levels, transformed normalized expression levels or transformed median centered normalized expression levels of ADAM 12 and TOP2A are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- a determination that the IRS is positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy is based upon the IRS exceeding a preset threshold.
- the threshold utilized to determine that the IRS is positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy will vary based upon how the IRS is derived from the Cox Proportional Hazard Model.
- the IRS value indicating beneficial response to checkpoint inhibitor therapy is approximately 0.70, 0.75, 0.80, 0.85, 0.90, 0.95, 1.00, 1.05, 1.10, 1.15, 1.20, 1.25, 1.30 or more. In some embodiments, the IRS value indicating beneficial response to checkpoint inhibitor is 0.80, 0.81, 0.82, 0.83, 0.84, 0.85, 0.86, 0.87, 0.88, 0.89, 0.90. 0.91 or more. In some embodiments, the IRS value indicating beneficial response to checkpoint inhibitor therapy is 0.87 or higher. In some embodiments, the IRS value indicating beneficial response to checkpoint inhibitor therapy is 0.873569 or higher.
- Some aspects of the present invention are directed to a method of treatment, comprising: a. measuring expression levels of RNA transcripts for PD-1 and PD-L2 obtained from a tumor specimen from a subject, b. measuring tumor mutation burden (TMB) in the biological sample and log2 transforming the TMB measurement to provide a transformed TMB measurement; c. calculating a Immunotherapy Response Score (IRS) from the expression levels or normalized levels of the RNA transcripts of PD-1 and PD-L2, and the transformed TMB measurement, wherein the IRS is positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy, and obtaining an IRS having a value indicating a beneficial response to checkpoint inhibitor therapy; and d. administering the checkpoint inhibitor therapy to the subject.
- TMB tumor mutation burden
- each of the transformed TMB expression, PD-1 expression and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for ADAM12.
- each of the transformed TMB expression, PD- 1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and ADAM 12 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized ADAM 12 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for PD-L1.
- each of the transformed TMB expression, PD-1 expression, PD-L1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized PD-L1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for CD4.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and PD-L1 are measured.
- each of the transformed TMB expression, PD-1 expression, PD-L1, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 and ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized PD-L1, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression and normalized ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, and ADAM12 are measured.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 and ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression and normalized ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for VTCN1.
- each of the transformed TMB expression, PD-1 expression, VTCN 1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized VTCN1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and VTCN1 are measured.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the IRS value indicating beneficial response to checkpoint inhibitor therapy is 6 or more, 7 or more, 8 or more, 9 or more, 9.5 or more, 10 or more, 10.5 or more, 11 or more, 12 or more, 13 or more, 14 or more, 15 or more, or 20 or more.
- the IRS value indicating beneficial response to checkpoint inhibitor therapy is 8 or more.
- the IRS value indicating beneficial response to checkpoint inhibitor therapy is 10 or more.
- the IRS value indicating beneficial response to checkpoint inhibitor therapy is 12 or more.
- the IRS is calculated using the Cox model as 8, 10 or 12 times the inverse of the patient hazard ratio as compared to the median hazard rate. In some embodiments of the methods disclosed herein, the IRS is calculated using the Cox model as 10 times the inverse of the patient hazard ratio as compared to the median hazard rate. In some embodiments of the methods disclosed herein, the IRS is calculated using the Cox model as between 8 and 12 times the inverse of the patient hazard ratio as compared to the median hazard rate.
- the tumor specimen is a formalin-fixed paraffin-embedded (FFPE) tumor specimen.
- the tumor specimen contains at least 20% tumor content.
- the tumor specimen is breast cancer, central or peripheral nervous system cancer, cancer of unknown primary, colorectal cancer, endometrial cancer, a gastrointestinal stromal tumor, glioma, hepatobiliary cancer, neuroendocrine cancer, ovarian cancer, pancreatic cancer, prostate cancer salivary gland cancer, sarcoma, or thyroid cancer.
- the tumor specimen is adrenal cancer, biliary cancer, bladder cancer, brain cancer, breast cancer, cervical cancer, colon cancer, rectum cancer, endometrial cancer, esophageal cancer, head or neck cancer, kidney cancer, liver cancer, non-small cell lung cancer, lung cancer, lymphoma, melanoma, meniges cancer, non-melanoma skin cancer, ovarian cancer, pancreatic cancer, prostate cancer, sarcoma, small intestine cancer, or stomach cancer.
- the tumor specimen is assessed as having microsatellite instability-low or microsatellite stability. In some embodiments, the tumor specimen is assessed as having a low tumor mutational burden, wherein low tumor mutational burden is classified as having less than 10 mutations per megabase (muts/Mb). In some embodiments, the expression levels of RNA transcripts are measured using PCR and next-generation sequencing.
- the checkpoint inhibitor is an anti-PD-1 antibody, an anti- CTLA-4 antibody, an anti-PD-Ll antibody, or an anti-PD-L2.
- the checkpoint inhibitor is nivolumab, pembrolizumab, atezolizumab, cemiplimab, durvalumab, pidilizumab, atezolimumab, PDR001, BMS- 936559, avelumab, ipilimumab, or SHR-1210.
- the checkpoint inhibitor therapy is administered as a monotherapy. In some embodiments, the checkpoint inhibitor therapy is administered in combination with one or more other chemotherapeutic agents.
- the tumor specimen shows a TPS score of 1-49%.
- the checkpoint inhibitor is administered as part of a 1 st line treatment regimen. In some embodiments, the checkpoint inhibitor is administered as part of a 2 nd line treatment regimen or higher.
- step a. further comprises measuring expression levels of RNA transcripts for at least one reference gene in the biological sample
- step b. comprises normalizing the measured expression levels of the other measured RNA transcripts against the level of the RNA transcripts of at the least one reference gene to provide normalized levels of the other RNA transcripts
- step c. comprises calculating the IRS from the normalized levels.
- the one or more reference genes comprise three genes selected from LRP1, MRPL13, TBP, HMBS, ITGB7, MYC, CIAO1, CTCF, EIF2B1, GGNBP2, SLC4A1AP.
- the one or more reference genes comprise three genes selected from CIAO1, EIF2B1, HMBS, CTCF, GGNBP2, ITGB7, MYC and SLC4A1AP. In some embodiments, the one or more reference genes comprise the combination of CIAO1, EIF2B1 and HMBS.
- Some aspects of the present disclosure are directed to a method of identifying a subject that would benefit from checkpoint inhibitor therapy, comprising: (a) measuring expression levels of RNA transcripts for PD-1, TOP2A, PD-L1 and ADAM12, and at least one reference gene in a biological sample obtained from a tumor specimen from the subject; (b) log2 transforming, median centering, and normalizing the measured expression levels of the RNA transcripts against the level of the RNA transcripts of at the least one reference gene to provide normalized levels of the RNA transcripts; (c) measuring tumor mutation burden (TMB) in the biological sample and log2 transforming the TMB measurement to provide a transformed TMB measurement; (d) calculating a Immunotherapy Response Score (IRS) from the transformed normalized levels of the RNA transcripts of PD-1, TOP2A, PD-L1 and ADAM12, and the transformed TMB measurement, wherein the IRS is positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy, and obtaining an IRS
- the tumor specimen is from a cancer not approved for labeled use of the checkpoint inhibitor therapy.
- the tumor specimen is assessed as having microsatellite instability-low or microsatellite stability.
- the tumor specimen is assessed as having a low tumor mutational burden, wherein low tumor mutational burden is classified as less than 10 mutations per megabase (muts/Mb).
- the tumor specimen shows a TPS score of 1-49%.
- Some aspects of the present disclosure are directed to a method of identifying a subject that would benefit from checkpoint inhibitor therapy, comprising: (a) receiving, by a processor, measured expression levels of RNA transcripts for at least two of PD-1, TOP2A, PD-L1 and ADAM12, and at least one reference gene in a biological sample obtained from a tumor specimen from the subject; (b) log2 transforming, median centering, and normalizing, by a processor, the measured expression levels of the RNA transcripts against the level of the RNA transcripts of at the least one reference gene to provide transformed normalized levels of the RNA transcripts; (c) receiving, by a processor, measured tumor mutation burden (TMB) in the biological sample; (d) log2 transforming, by a processor, the TMB measurement to provide a transformed TMB measurement; (e) calculating, by a processor, an Immunotherapy Response Score (IRS) from the transformed normalized levels of the RNA transcripts of the at least two of PD-1, TOP2A, PD-
- the IRS value indicating beneficial response to checkpoint inhibitor therapy is approximately 0.87 or more. In some embodiments, the IRS value indicating beneficial response to checkpoint inhibitor therapy is 0.873569 or higher.
- the one or more reference genes comprise three genes selected from CIAO1, EIF2B1, HMBS, CTCF, GGNBP2, ITGB7, MYC and SLC4A1AP. In some embodiments, the one or more reference genes comprise the combination of CIAO1, EIF2B1 and HMBS.
- Some aspects of the present disclosure are directed to a method of treatment of a subject in need thereof with a checkpoint inhibitor therapy, comprising administering to said subject the checkpoint inhibitor therapy, wherein said subject in need thereof was previously identified by a method comprising: (a) measuring expression levels of RNA transcripts for at least two of PD-1, TOP2A, PD-L1 and ADAM12, and at least one reference gene in a biological sample obtained from a tumor specimen from the subject; (b) log2 transforming, median centering, and normalizing the measured expression levels of the RNA transcripts against the level of the RNA transcripts of at the least one reference gene to provide transformed normalized levels of the RNA transcripts; (c) measuring tumor mutation burden (TMB) in the biological sample and log2 transforming the TMB measurement to provide a transformed TMB measurement; (d) calculating a Immunotherapy Response Score (IRS) from the transformed normalized levels of the RNA transcripts of the at least two of PD-1, TOP2A, PD-L1 and ADAM
- the IRS value indicating beneficial response to checkpoint inhibitor therapy is approximately 0.87 or more. In some embodiments, the IRS value indicating beneficial response to checkpoint inhibitor therapy is 0.873569 or higher.
- the one or more reference genes comprise three genes selected from CIAO1, EIF2B1, HMBS, CTCF, GGNBP2, ITGB7, MYC and SLC4A1AP. In some embodiments, the one or more reference genes comprise the combination of HMBS, CIAO1 and EIF2B1.
- the tumor specimen is from a cancer not approved for labeled use of the checkpoint inhibitor therapy. In some embodiments, the tumor specimen is assessed as having microsatellite instability-low or microsatellite stability. In some embodiments, the tumor specimen is assessed as having a low tumor mutational burden, wherein low tumor mutational burden is classified as less than 10 mutations per megabase (muts/Mb).
- the checkpoint inhibitor therapy is administered as a monotherapy. In some embodiments, the checkpoint inhibitor therapy is administered in combination with one or more other chemotherapeutic agents. In some embodiments, the tumor specimen shows a TPS score of 1-49%.
- Some aspects of the present disclosure are directed to a method of treatment, comprising: (a) measuring expression levels of RNA transcripts PD-1, TOP2A, PD-L1 and ADAM12, and at least one reference gene in a biological sample obtained from a tumor specimen from a subject; (b) log2 transforming, median centering, and normalizing the measured expression levels of the RNA transcripts of PD-1, TOP2A, PD-L1 and ADAM 12 against the level of the RNA transcripts of at the least one reference gene to provide transformed normalized levels of the RNA transcripts of PD-1, TOP2A, PD-L1 and ADAM12; (c) measuring tumor mutation burden (TMB) in the biological sample and log2 transforming the TMB measurement to provide a transformed TMB measurement; (d) calculating a Immunotherapy Response Score (IRS) from the transformed normalized levels of the RNA transcripts of PD-1, TOP2A, PD-L1 and ADAM 12 and the transformed TMB measurement, wherein the IRS is positively
- the IRS value indicating beneficial response to checkpoint inhibitor therapy is approximately 0.87 or more. In some embodiments, the IRS value indicating beneficial response to checkpoint inhibitor therapy is 0.873569 or higher.
- the one or more reference genes comprise three genes selected from CIAO1, EIF2B1, HMBS, CTCF, GGNBP2, ITGB7, MYC and SLC4A1AP. In some embodiments, the one or more reference genes comprise the combination of HMBS, CIAO1 and EIF2B1.
- the tumor specimen is a formalin-fixed paraffin-embedded (FFPE) tumor specimen.
- the tumor specimen is breast cancer, central or peripheral nervous system cancer, cancer of unknown primary, colorectal cancer, endometrial cancer, a gastrointestinal stromal tumor, glioma, hepatobiliary cancer, neuroendocrine cancer, ovarian cancer, pancreatic cancer, prostate cancer salivary gland cancer, sarcoma, or thyroid cancer.
- the expression levels of RNA transcripts are measured using PCR and next-generation sequencing.
- the checkpoint inhibitor is an anti-PD-1 antibody, an anti- CTLA-4 antibody, an anti-PD-Ll antibody, or an anti-PD-L2.
- the checkpoint inhibitor is nivolumab, pembrolizumab, atezolizumab, cemiplimab, durvalumab, pidilizumab, atezolimumab, PDR001, BMS- 936559, avelumab, ipilimumab, or SHR-1210.
- Some aspects of the present disclosure are directed to a method of identifying a subject that would benefit from checkpoint inhibitor therapy, comprising: (a) measuring expression levels of RNA transcripts for PD-1, TOP2A, PD-L1 and ADAM12, and at least one reference gene in a biological sample obtained from a tumor specimen from the subject; (b) log2 transforming, median centering, and normalizing the measured expression levels of the RNA transcripts of PD-1, TOP2A, PD-L1 and ADAM 12 against the level of the RNA transcripts of at the least one reference gene to provide transformed normalized levels of the RNA transcripts of PD-1, TOP2A, PD-L1 and ADAM12; (c) measuring tumor mutation burden (TMB) in the biological sample and log2 transforming the TMB measurement to provide a transformed TMB measurement; (d) calculating a Immunotherapy Response Score (IRS) from the transformed normalized levels of the RNA transcripts of PD-1, TOP2A, PD-L1 and ADAM 12 and
- the tumor specimen is from a cancer not approved for labeled use of the checkpoint inhibitor therapy.
- the tumor specimen is assessed as having microsatellite instability-low or microsatellite stability.
- the tumor specimen is assessed as having a low tumor mutational burden, wherein low tumor mutational burden is classified as less than 10 mutations per megabase (muts/Mb).
- the tumor specimen contains at least 20% tumor content.
- the checkpoint inhibitor therapy is administered as a monotherapy. In some embodiments, the checkpoint inhibitor therapy is administered in combination with one or more other chemotherapeutic agents. In some embodiments, the tumor specimen shows a TPS score of 1-49%.
- a subject is identified as benefiting from checkpoint inhibitor therapy (e.g., pembrolizumab) or treated with checkpoint inhibitor therapy (e.g., pembrolizumab) when the tumor type is not approved for use with the checkpoint inhibitor (off-label) and the IRS value is 0.90 or greater.
- a subject is identified as benefiting from checkpoint inhibitor therapy (e.g., pembrolizumab) or treated with checkpoint inhibitor therapy (e.g., pembrolizumab) when the tumor type is not approved for use with the checkpoint inhibitor (off-label) and the IRS value is 0.88 or greater.
- a subject is identified as benefiting from checkpoint inhibitor therapy (e.g., pembrolizumab) or treated with checkpoint inhibitor therapy (e.g., pembrolizumab) when the tumor type is not approved for use with the checkpoint inhibitor (off-label), the TMB is 10 mutations per megabase (MPM) or more and the IRS value is 0.87 or greater.
- the TMB is 10 MPM or more and the IRS value is 0.873 or greater.
- a subject is identified as benefiting from checkpoint inhibitor therapy (e.g., pembrolizumab) or treated with checkpoint inhibitor therapy (e.g., pembrolizumab) when the TMB is 10 MPM or more and the IRS value is 0.8736 or greater.
- a subject is identified as benefiting from checkpoint inhibitor therapy (e.g., pembrolizumab) or treated with checkpoint inhibitor therapy (e.g., pembrolizumab) when the TMB is 10 MPM or more and the IRS value is 0.873569 or greater.
- Some aspects of the present disclosure are directed to a method of identifying a subject that would benefit from checkpoint inhibitor therapy, comprising: a. measuring expression levels of RNA transcripts for PD-1, PD-L2 and, optionally, expression levels of RNA transcripts for one or more of CD4, ADAM12, PD-L1, and VTCN1, and at least one reference gene in a biological sample obtained from a tumor specimen from the subject; b. Iog2 transforming, median centering, and normalizing the measured expression levels of the RNA transcripts against the level of the RNA transcripts of at the least one reference gene to provide normalized levels of the RNA transcripts; c.
- TMB tumor mutation burden
- IRS Immunotherapy Response Score
- the tumor specimen is from a cancer not approved for labeled use of the checkpoint inhibitor therapy.
- the calculated IRS value indicates that the median time-to-next-treatment (TNTT) is 24 months or greater.
- each of the transformed TMB expression, PD-1 expression and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for ADAM12.
- each of the transformed TMB expression, PD- 1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and ADAM 12 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized ADAM 12 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for PD-L1.
- each of the transformed TMB expression, PD-1 expression, PD-L1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized PD-L1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for CD4.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and PD-L1 are measured.
- each of the transformed TMB expression, PD-1 expression, PD-L1, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 and ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized PD-L1, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression and normalized ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, and ADAM12 are measured.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 and ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression and normalized ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for VTCN1.
- each of the transformed TMB expression, PD-1 expression, VTCN 1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized VTCN1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and VTCN1 are measured.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- Some aspects of the present disclosure are directed to a method of identifying a subject that would benefit from checkpoint inhibitor therapy, comprising: a. receiving, by a processor, measured expression levels of RNA transcripts for PD-1, PD-L2 and, optionally, expression levels of RNA transcripts for one or more of CD4, ADAM12, PD-L1, and VTCN1, and at least one reference gene in a biological sample obtained from a tumor specimen from the subject; b. Iog2 transforming, median centering, and normalizing, by a processor, the measured expression levels of the RNA transcripts against the level of the RNA transcripts of at the least one reference gene to provide normalized levels of the RNA transcripts; c.
- TMB tumor mutation burden
- Iog2 transforming, by a processor, the TMB measurement to provide a transformed TMB measurement
- e. calculating, by a processor, a Immunotherapy Response Score (IRS) from the normalized levels of the RNA transcripts of PD-1, PD-L2 and, optionally, one or more of CD4, ADAM12, PD-L1, and VTCN1, and the transformed TMB measurement, that is positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy
- f. providing a determination if the subject has a checkpoint inhibitor responsive cancer.
- each of the transformed TMB expression, PD-1 expression and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for ADAM12.
- each of the transformed TMB expression, PD- 1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and ADAM 12 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized ADAM 12 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for PD-L1.
- each of the transformed TMB expression, PD-1 expression, PD-L1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized PD-L1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for CD4.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and PD-L1 are measured.
- each of the transformed TMB expression, PD-1 expression, PD-L1, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 and ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized PD-L1, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression and normalized ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, and ADAM12 are measured.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 and ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression and normalized ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for VTCN1.
- each of the transformed TMB expression, PD-1 expression, VTCN 1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized VTCN1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and VTCN1 are measured.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- Some aspects of the present disclosure are directed to a method of treatment of a subject in need thereof with a checkpoint inhibitor therapy, comprising administering to said subject the checkpoint inhibitor therapy, wherein said subject in need thereof was previously identified by a method comprising: a. measuring expression levels of RNA transcripts for PD-1, PD-L2 and, optionally, expression levels of RNA transcripts for one or more of CD4, ADAM12, PD-L1, and VTCN 1 , and at least one reference gene in a biological sample obtained from a tumor specimen from the subject; b.
- TMB tumor mutation burden
- IRS Immunotherapy Response Score
- the IRS is calculated using the Cox model as 10 times the inverse of the patient hazard ratio as compared to the median hazard rate.
- each of the transformed TMB expression, PD-1 expression and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for ADAM12.
- each of the transformed TMB expression, PD- 1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and ADAM 12 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized ADAM 12 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for PD-L1.
- each of the transformed TMB expression, PD-1 expression, PD-L1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized PD-L1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for CD4.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and PD-L1 are measured.
- each of the transformed TMB expression, PD-1 expression, PD-L1, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 and ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized PD-L1, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression and normalized ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, and ADAM12 are measured.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 and ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression and normalized ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for VTCN1.
- each of the transformed TMB expression, PD-1 expression, VTCN 1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized VTCN1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and VTCN1 are measured.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the IRS value indicating beneficial response to checkpoint inhibitor therapy is 10 or more.
- Some aspects of the present disclosure are directed to a method of treatment, comprising: a. measuring expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and at least one reference gene in a biological sample obtained from a tumor specimen from a subject; b. Iog2 transforming, median centering, and normalizing the measured expression levels of the RNA transcripts of PD-1, PD-L2, CD4, and ADAM12 against the level of the RNA transcripts of at the least one reference gene to provide normalized levels of the RNA transcripts of PD-1, PD-L2, CD4, and ADAM12; c.
- TMB tumor mutation burden
- IRS Immunotherapy Response Score
- each of the transformed TMB expression, PD-1 expression and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for ADAM12.
- each of the transformed TMB expression, PD- 1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and ADAM 12 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized ADAM 12 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for PD-L1.
- each of the transformed TMB expression, PD-1 expression, PD-L1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized PD-L1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for CD4.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and PD-L1 are measured.
- each of the transformed TMB expression, PD-1 expression, PD-L1, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 and ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized PD-L1, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression and normalized ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, and ADAM12 are measured.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 and ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression and normalized ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for VTCN1.
- each of the transformed TMB expression, PD-1 expression, VTCN 1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized VTCN1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and VTCN1 are measured.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the IRS value indicating beneficial response to checkpoint inhibitor therapy is 10 or more.
- the one or more reference genes comprise three genes selected from LRP1, MRPL13, TBP, HMBS, ITGB7, MYC, CIAO1, CTCF, EIF2B1, GGNBP2, SLC4A1AP.
- the tumor specimen is a formalin-fixed paraffin- embedded (FFPE) tumor specimen.
- the tumor specimen is adrenal cancer, biliary cancer, bladder cancer, brain cancer, breast cancer, cervical cancer, colon cancer, rectum cancer, endometrial cancer, esophageal cancer, head or neck cancer, kidney cancer, liver cancer, nonsmall cell lung cancer, lung cancer, lymphoma, melanoma, meniges cancer, non-melanoma skin cancer, ovarian cancer, pancreatic cancer, prostate cancer, sarcoma, small intestine cancer, or stomach cancer.
- the expression levels of RNA transcripts are measured using PCR and next-generation sequencing.
- the checkpoint inhibitor is an anti-PD-1 antibody, an anti-CTLA-4 antibody, an anti-PD-Ll antibody, or an anti-PD-L2.
- the checkpoint inhibitor is nivolumab, pembrolizumab, atezolizumab, durvalumab, pidilizumab, PDR001, BMS- 936559, avelumab, or SHR-1210.
- Some aspects of the present disclosure are directed to a method of identifying a subject that would benefit from checkpoint inhibitor therapy, comprising: a. measuring expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and at least one reference gene in a biological sample obtained from a tumor specimen from the subject; b. Iog2 transforming, median centering, and normalizing the measured expression levels of the RNA transcripts of PD-1, PD-L2, CD4, and ADAM 12 against the level of the RNA transcripts of at the least one reference gene to provide normalized levels of the RNA transcripts of PD-1, PD-L2, CD4, and ADAM12; c.
- TMB tumor mutation burden
- IRS Immunotherapy Response Score
- the tumor specimen is from a cancer not approved for labeled use of the checkpoint inhibitor therapy.
- the calculated IRS value indicates that the median time-to-next-treatment (TNTT) is 24 months or greater.
- each of the transformed TMB expression, PD-1 expression and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for ADAM12.
- each of the transformed TMB expression, PD- 1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and ADAM 12 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized ADAM 12 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for PD-L1.
- each of the transformed TMB expression, PD-1 expression, PD-L1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized PD-L1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for CD4.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and PD-L1 are measured.
- each of the transformed TMB expression, PD-1 expression, PD-L1, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 and ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized PD-L1, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression and normalized ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, and ADAM12 are measured.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 and ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression and normalized ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for VTCN1.
- each of the transformed TMB expression, PD-1 expression, VTCN 1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized VTCN1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and VTCN1 are measured.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- Some aspects of the present disclosure are directed to a method of identifying a subject that would benefit from checkpoint inhibitor therapy, comprising: a. receiving, by a processor, measured expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and at least one reference gene in a biological sample obtained from a tumor specimen from the subject; b. Iog2 transforming, median centering, and normalizing, by a processor, the measured expression levels of the RNA transcripts of PD-1, PD-L2, CD4, and ADAM12 against the level of the RNA transcripts of at the least one reference gene to provide normalized levels of the RNA transcripts of PD-1, PD-L2, CD4, and ADAM12; c.
- TMB tumor mutation burden
- Iog2 transforming, by a processor, the TMB measurement to provide a transformed TMB measurement
- e. calculating, by a processor, a Immunotherapy Response Score (IRS) from the normalized levels of the RNA transcripts of PD-1, PD-L2, CD4, and ADAM12 and the transformed TMB measurement that is positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy
- f providing a determination if the subject has a checkpoint inhibitor responsive cancer.
- each of the transformed TMB expression, PD-1 expression and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for ADAM12.
- each of the transformed TMB expression, PD- 1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and ADAM 12 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized ADAM 12 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for PD-L1.
- each of the transformed TMB expression, PD-1 expression, PD-L1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized PD-L1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for CD4.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and PD-L1 are measured.
- each of the transformed TMB expression, PD-1 expression, PD-L1, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 and ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized PD-L1, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression and normalized ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, and ADAM12 are measured.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 and ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression and normalized ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for VTCN1.
- each of the transformed TMB expression, PD-1 expression, VTCN 1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized VTCN1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and VTCN1 are measured.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- Some aspects of the present disclosure are directed to a method of treatment of a subject in need thereof with a checkpoint inhibitor therapy, comprising administering to said subject the checkpoint inhibitor therapy, wherein said subject in need thereof was previously identified by a method comprising: a. measuring expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM 12, and at least one reference gene in a biological sample obtained from a tumor specimen from the subject; b.
- TMB tumor mutation burden
- IRS Immunotherapy Response Score
- each of the transformed TMB expression, PD-1 expression and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for ADAM12.
- each of the transformed TMB expression, PD- 1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and ADAM 12 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized ADAM 12 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for PD-L1.
- each of the transformed TMB expression, PD-1 expression, PD-L1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized PD-L1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for CD4.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and PD-L1 are measured.
- each of the transformed TMB expression, PD-1 expression, PD-L1, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 and ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized PD-L1, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression and normalized ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, and ADAM12 are measured.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 and ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression and normalized ADAM 12 expression are negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- step a. further comprises measuring the expression level of RNA transcripts for VTCN1.
- each of the transformed TMB expression, PD-1 expression, VTCN 1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, normalized VTCN1 expression and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- the expression levels of RNA transcripts for PD-1, PD-L2, CD4, ADAM12, and VTCN1 are measured.
- each of the transformed TMB expression, PD-1 expression, and PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- each of the transformed TMB expression, normalized PD-1 expression, and normalized PD-L2 expression are positively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy and normalized CD4 expression is negatively correlated with the likelihood that the patient has a beneficial response to checkpoint inhibitor therapy.
- a subject is identified as benefiting from checkpoint inhibitor therapy (e.g., pembrolizumab) or treated with checkpoint inhibitor therapy (e.g., pembrolizumab) when the tumor type is not approved for use with the checkpoint inhibitor (off-label) and the IRS value is 10 or greater.
- a subject is identified as benefiting from checkpoint inhibitor therapy (e.g., pembrolizumab) or treated with checkpoint inhibitor therapy (e.g., pembrolizumab) when the tumor type is not approved for use with the checkpoint inhibitor (off-label) and the IRS value is 12 or greater.
- a subject is identified as benefiting from checkpoint inhibitor therapy (e.g., pembrolizumab) or treated with checkpoint inhibitor therapy (e.g., pembrolizumab) when the tumor type is not approved for use with the checkpoint inhibitor (off-label), the TMB is 10 mutations per megabase (MPM) or more and the IRS value is 10 or greater.
- the TMB is 10 MPM or more and the IRS value is 12 or greater.
- a subject is identified as benefiting from checkpoint inhibitor therapy (e.g., pembrolizumab) or treated with checkpoint inhibitor therapy (e.g., pembrolizumab) when the TMB is 10 MPM or more and the IRS value is 10 or greater.
- a subject is identified as benefiting from checkpoint inhibitor therapy (e.g., pembrolizumab) or treated with checkpoint inhibitor therapy (e.g., pembrolizumab) when the TMB is 10 MPM or more and the IRS value is 12 or greater.
- determining if the tumor will be or is more likely to be responsive to immune checkpoint therapy comprises collecting or providing a tumor specimen from a subject.
- the tumor specimen is a fresh tumor specimen or a formalin-fixed paraffin-embedded (FFPE) tumor specimen.
- the specimen preparation is not limited and may be any suitable preparation known in the art.
- the methods do not include collecting or providing a tumor. Instead, data (e.g., IRS value) or a qualitative assessment (e.g., a determination that the tumor has a suitable IRS value) is provided.
- the data or qualitative assessment is provided to a physician or other health professional and such person uses such data or assessment to determine whether or not to administer the immune checkpoint therapy.
- the provided data or qualitative assessment can be calculated or determined by any of the methods disclosed herein.
- the tumor may be from any cancer is not limited.
- cancer refers to a malignant neoplasm (Stedman’s Medical Dictionary, 25th ed.; Hensyl ed.; Williams & Wilkins: Philadelphia, 1990).
- Exemplary cancers include, but are not limited to, acoustic neuroma; adenocarcinoma; adrenal gland cancer; anal cancer; angiosarcoma (e.g., lymphangiosarcoma, lymphangioendotheliosarcoma, hemangiosarcoma); appendix cancer; benign monoclonal gammopathy; biliary cancer (e.g., cholangiocarcinoma); bladder cancer; breast cancer (e.g., adenocarcinoma of the breast, papillary carcinoma of the breast, mammary cancer, medullary carcinoma of the breast); brain cancer (e.g., meningioma, glioblastomas, glioma (e.g., astrocytoma, oligodendroglioma), medulloblastoma); bronchus cancer; carcinoid tumor; cervical cancer (e.g., cervical adenocarcinoma); choriocar
- Wilms tumor, renal cell carcinoma); liver cancer (e.g., hepatocellular cancer (HCC), malignant hepatoma); lung cancer (e.g., bronchogenic carcinoma, small cell lung cancer (SCLC), non-small cell lung cancer (NSCLC), adenocarcinoma of the lung); leiomyosarcoma (LMS); mastocytosis (e.g., systemic mastocytosis); muscle cancer; myelodysplastic syndrome (MDS); mesothelioma; myeloproliferative disorder (MPD) (e.g., polycythemia vera (PV), essential thrombocytosis (ET), agnogenic myeloid metaplasia (AMM) a.k.a.
- HCC hepatocellular cancer
- lung cancer e.g., bronchogenic carcinoma, small cell lung cancer (SCLC), non-small cell lung cancer (NSCLC), adenocarcinoma of the lung
- myelofibrosis MF
- chronic idiopathic myelofibrosis chronic myelocytic leukemia (CML), chronic neutrophilic leukemia (CNL), hypereosinophilic syndrome (HES)
- neuroblastoma e.g., neurofibromatosis (NF) type 1 or type 2, schwannomatosis
- neuroendocrine cancer e.g., gastroenteropancreatic neuroendoctrine tumor (GEP-NET), carcinoid tumor
- osteosarcoma e.g., bone cancer
- ovarian cancer e.g., cystadenocarcinoma, ovarian embryonal carcinoma, ovarian adenocarcinoma
- papillary adenocarcinoma pancreatic cancer
- pancreatic cancer e.g., pancreatic andenocarcinoma, intraductal papillary mucinous neoplasm (IPMN), Islet cell tumors
- the cancer is not a blood-borne or hematopoietic cancer. In some embodiments, the cancer is not an MSI-H cancer. In some embodiments, the cancer is not 1, 2, 3, 4, 5, 6 or all 7 of melanoma, lung cancer, kidney cancer, bladder cancer, head and neck cancer, and Hodgkin's lymphoma.
- the cancer is adrenal cancer, biliary cancer, bladder cancer, brain cancer, breast cancer, cervical cancer, colon cancer, rectum cancer, endometrial cancer, esophageal cancer, head or neck cancer, kidney cancer, liver cancer, non-small cell lung cancer, lung cancer, lymphoma, melanoma, meninges cancer, nonmelanoma skin cancer, ovarian cancer, pancreatic cancer, prostate cancer, sarcoma, small intestine cancer, or stomach cancer.
- the cancer is not a TMB-H cancer.
- the cancer is not 1, 2, 3, 4, 5, 6, 7, 8, 9, or all 10 of melanoma, lung cancer, kidney cancer, bladder cancer, head and neck cancer, cervical cancer, esophagogastric cancer, hepatobiliary cancer, nonmelanoma skin cancer, and Hodgkin's lymphoma.
- the cancer is adrenal cancer, biliary cancer, bladder cancer, brain cancer, breast cancer, cervical cancer, colon cancer, rectum cancer, endometrial cancer, esophageal cancer, head or neck cancer, kidney cancer, liver cancer, non-small cell lung cancer, lung cancer, lymphoma, melanoma, meninges cancer, nonmelanoma skin cancer, ovarian cancer, pancreatic cancer, prostate cancer, sarcoma, small intestine cancer, or stomach cancer.
- determining or calculating if the tumor will be or is more likely to be responsive to immune checkpoint therapy comprises calculating, collecting or determining immune-response associated data derived from the tumor (e.g., the IRS value).
- the methods disclosed herein comprise obtaining immune-response associated data (quantitative or qualitative) derived from the tumor from another party and determining if the tumor will be or is more likely to be responsive to immune checkpoint therapy.
- immune-response associated data is collected or determined via NGS and/or multiplexed PCR.
- immune-response associated data is obtained from NGS and/or multiplexed PCR performed by another party.
- PD-1 expression is determined or calculated via NGS of gene expression transcripts using multiplex PCR (amplicon).
- PD-1 expression is obtained from NGS of gene expression transcripts using multiplex PCR (amplicon) data.
- PD-1 expression is validated, confirmed, or combined using multiplex PCR and a second amplicon.
- validation or confirmation of PD-1 requires that the second amplicon's percentile value is 70%, 75%, 80%, 85% or more of the calculated PD-1 percentile value.
- validation or confirmation of PD-1 requires that the second amplicon's percentile value is 80% or more of the calculated PD-1 percentile value.
- PD-L2 expression is determined or calculated via NGS of gene expression transcripts using multiplex PCR (amplicon).
- PD-L2 expression is obtained from NGS of gene expression transcripts using multiplex PCR (amplicon) data.
- PD-L2 expression is validated, confirmed, or combined using multiplex PCR and a second amplicon.
- validation or confirmation of PD-L2 requires that the second amplicon's percentile value is 70%, 75%, 80%, 85% or more of the calculated PD-L2 percentile value.
- validation or confirmation of PD-L2 requires that the second amplicon's percentile value is 80% or more of the calculated PD-L2 percentile value.
- CD4 expression is determined or calculated via NGS of gene expression transcripts using multiplex PCR (amplicon).
- CD4 expression is obtained from NGS of gene expression transcripts using multiplex PCR (amplicon) data.
- CD4 expression is validated, confirmed, or combined using multiplex PCR (amplicon) to measure GZMA, GZMB, GZMK, PRF1, IFNG or CD8B expression.
- CD4 expression is validated, confirmed, or combined using multiplex PCR (amplicon) to measure GZMA expression.
- CD4 and GZMA are both part of the interferon-y gene signature.
- validation, confirmation or combination of CD4 requires that the second amplicon measurement's percentile value is 80% or more of the calculated CD4 percentile value.
- ADAM12 expression is determined or calculated via NGS of gene expression transcripts using multiplex PCR (amplicon). In some embodiments, ADAM12 expression is obtained from NGS of gene expression transcripts using multiplex PCR (amplicon) data. In some embodiments, ADAM12 expression is validated, confirmed, or combined using multiplex PCR and a second amplicon. In some embodiments, validation or confirmation of ADAM12 requires that the second amplicon's percentile value is 70%, 75%, 80%, 85% or more of the calculated ADAM12 percentile value. In some embodiments, validation or confirmation of ADAM 12 requires that the second amplicon's percentile value is 80% or more of the calculated ADAM 12 percentile value.
- PD-L1 expression is determined or calculated via NGS of gene expression transcripts using multiplex PCR (amplicon).
- PD-L1 expression is obtained from NGS of gene expression transcripts using multiplex PCR (amplicon) data.
- PD-L1 expression is validated, confirmed, or combined using multiplex PCR and a second amplicon.
- validation or confirmation of PD-L1 requires that the second amplicon's percentile value is 70%, 75%, 80%, 85% or more of the calculated PD-L1 percentile value.
- validation or confirmation of PD-L1 requires that the second amplicon's percentile value is 80% or more of the calculated PD-L1 percentile value.
- VTCN 1 expression is determined or calculated via NGS of gene expression transcripts using multiplex PCR (amplicon). In some embodiments, VTCN1 expression is obtained from NGS of gene expression transcripts using multiplex PCR (amplicon) data. In some embodiments, VTCN1 expression is validated, confirmed, or combined using multiplex PCR and a second amplicon. In some embodiments, validation or confirmation of VTCN1 requires that the second amplicon's percentile value is 70%, 75%, 80%, 85% or more of the calculated VTCN1 percentile value. In some embodiments, validation or confirmation of VTCN 1 requires that the second amplicon's percentile value is 80% or more of the calculated VTCN1 percentile value.
- TOP2A expression is determined or calculated via NGS of gene expression transcripts using multiplex PCR (amplicon).
- TOP2A expression is obtained from NGS of gene expression transcripts using multiplex PCR (amplicon) data.
- TOP2A expression is validated, confirmed, or combined using multiplex PCR and a second amplicon.
- validation or confirmation of TOP2A requires that the second amplicon's percentile value is 70%, 75%, 80%, 85% or more of the calculated PD-L1 percentile value.
- validation or confirmation of TOP2A requires that the second amplicon's percentile value is 80% or more of the calculated TOP2A percentile value.
- TMB is determined or calculated by NGS of tumor DNA. In some embodiments, TMB is obtained from another party. Methods of detecting mutations (e.g., TMB) are not limited. In some embodiments, mutations are detected, calculated or obtained via NGS. In some embodiments, TMB includes non-coding (at highly characterized genomic loci) and coding, synonymous and non-synonymous, single and multinucleotide (two bases) variants present at >10% variant allele frequency (VAF). In some embodiments, mutations per megabase (Mb) estimates and associated 90% confidence interval are calculated via the total number of positions with sufficient depth of coverage necessary for definitive assessment (maximum possible 1.7Mb).
- the checkpoint inhibitor administered is an antibody against at least one checkpoint protein, e.g., PD-1, CTLA-4, PD- L1 or PD-L2.
- the checkpoint inhibitor administered is an antibody that is effective against two or more of the checkpoint proteins selected from the group of PD-1, CTLA-4, PD-L1 and PD-L2.
- the checkpoint inhibitor administered is a small molecule, non-protein compound that inhibits at least one checkpoint protein.
- the checkpoint inhibitor is a small molecule, non-protein compound that inhibits a checkpoint protein selected from the group consisting of PD-1, CTLA-4, PD-L1 and PD-L2.
- the checkpoint inhibitor administered is nivolumab (Opdivo®, BMS-936558, MDX1106, commercially available from BristolMyers Squibb, Princeton NJ), pembrolizumab (Keytruda® MK-3475, lambrolizumab, commercially available from Merck and Company, Kenilworth NJ), atezolizumab (Tecentriq®, Genentech/Roche, South San Francisco CA), durvalumab (MEDI4736, Medimmune/AstraZeneca), pidilizumab (CT-011, CureTech), PDR001 (Novartis), BMS- 936559 (MDX1105, BristolMyers Squibb), avelumab (MSB0010718C, Merck Serono/Pfizer), or SHR-1210 (Incyte).
- nivolumab Opdivo®, BMS-936558, MDX1106, commercially available from BristolMyers Squibb, Princeton NJ
- Additional antibody PD1 pathway inhibitors for use in the methods described herein include those described in United States Patent No.8,217,149 (Genentech, Inc) issued July 10, 2012; United States Patent No.8,168,757 (Merck Sharp and Dohme Corp.) issued May 1, 2012, United States Patent No.8,008,449 (Medarex) issued August 30, 2011, and United States Patent No.7,943,743 (Medarex, Inc) issued May 17, 2011.
- the disclosed methods include performing one or more normalization processes, such as for enabling sequencing outputs (e.g., associated with any suitable biomarkers described herein, etc.) to be comparable to thresholds and/or across different sequencing runs.
- determining IRS values can include background-subtracting sequence read counts; and normalizing the background-subtracted sequence read counts into normalized reads per million (nRPM).
- nRPM normalized reads per million
- the RPM profile can be determined based on an average RPM (and/or other suitable aggregate RPM metric) of a plurality of replicates of biological samples across different validation sequencing runs.
- Housekeeping genes usable for normalization processes can include any one or more of: LRP1, MRPL13, TBP, HMBS, ITGB7, MYC, CIAO1, CTCF, EIF2B1, GGNBP2, SLC4A1AP and/or other suitable housekeeping genes (and/or any suitable genes).
- two, three, four, five, six, seven, or eight of LRP1, MRPL13, TBP, HMBS, ITGB7, MYC, CIAO1, CTCF, EIF2B1, GGNBP2, SLC4A1AP are used for the normalization process.
- nRPM Background Subtracted Read Count / Normalization Ratio.
- Housekeeping genes usable for normalization processes can include any one or more of: CIAO1, EIF2B1, HMBS, CTCF, GGNBP2, ITGB7, MYC and SLC4A1AP.
- three or more of CIAO1, EIF2B1, HMBS, CTCF, GGNBP2, ITGB7, MYC and SLC4A1AP are used for the normalization process.
- the one or more reference genes comprise the combination of CIAO1, EIF2B1 with HMBS, CTCF, GGNBP2, ITGB7, MYC, SLC4A1AP and/or other suitable housekeeping genes (and/or any suitable genes).
- CIAO1, EIF2B1, HMBS, CTCF, GGNBP2, ITGB7, MYC and SLC4A1AP are used for the normalization process.
- three of CIAO1, EIF2B1, HMBS, CTCF, GGNBP2, ITGB7, MYC and SLC4A1AP are used for the normalization process.
- EIF2B1, HMBS, and CIAO1 are used for the normalization process.
- any suitable backgrounding and/or normalizing processes can be performed (e.g., for comparison of values to thresholds; for comparison of values across sequencing runs; etc.).
- measurement of the housekeeping genes is omitted, and an internal standard is used for normalization.
- an internal standard is used for normalization.
- real time quantitative PCR which measures PCR product accumulation through a dual-labeled fluorigenic probe (i.e., TaqMan® probe).
- Real time PCR is compatible both with quantitative competitive PCR, where internal competitor for each target sequence is used for normalization, and with quantitative comparative PCR using a normalization gene contained within the sample, or a housekeeping gene for RT-PCR.
- normalization is based on the mean or median signal (CT) of all of the assayed genes or a large subset thereof (global normalization approach).
- the methods of the claimed invention can include one or more of: collecting a set of biological samples (e.g., FFPE tumor specimens) from a set of patients (e.g., cancer patients; etc.); generating one or more sequencing libraries (e.g., suitable for generating sequencing outputs indicative of biomarkers associated with patient responsiveness to one or more therapies; etc.) based on processing of the biological samples; determining sets of sequencing reads (e.g., for cDNA sequences derived from cDNA converted from mRNA indicating expression levels for the biomarkers provide herein, and, optionally, at least one reference gene) for the set of patients based on the one or more sequencing libraries; processing the sequencing reads for determining immune response-associated data (e.g., PD-L2 gene expression levels; PD-1 gene expression levels; one or more of CD4, ADAM12, TOP2A, PD-L1, and VTCN1 gene expression levels; cDNA sequence data, such as from cDNA converted from a set of biological samples (e.
- IRS values can be used for clinical trials (e.g., clinical trial enrollment and patient selection; stratification of patient populations, such as based on different combinations of biomarkers; therapy characterization; results analysis; and/or other suitable purposes related to clinical trials; etc.), care provision (e.g., providing treatment response characterizations to care providers for guiding care decisions regarding patients; therapy determination for patients; etc.), and/or other suitable applications.
- care provision e.g., providing treatment response characterizations to care providers for guiding care decisions regarding patients; therapy determination for patients; etc.
- embodiments of the methods and systems disclosed herein can function to conserve valuable biological samples, such as lung cancer tissue biopsies, tumor specimens, and/or suitable types of biological samples.
- immune response-associated data collection can be performed based on RNA sequencing e.g., PD-L1 gene expression levels; PD-1 gene expression levels; one or more of CD4, ADAM12, TOP2A, PD- L2, and VTCN1 gene expression levels; cDNA sequence data, such as from cDNA converted from mRNA; DNA sequence data; TMB-associated data; MSI-associated data; etc.) and/or other suitable processing approaches as an alternative to sample processing approaches that can require a relatively larger usage of biological sample (e.g., immunohistochemistry; etc.).
- RNA sequencing e.g., PD-L1 gene expression levels; PD-1 gene expression levels; one or more of CD4, ADAM12, TOP2A, PD- L2, and VTCN1 gene expression levels
- cDNA sequence data such as from cDNA converted from mRNA
- DNA sequence data DNA sequence data
- TMB-associated data e.g., MSI-associated data; etc.
- MSI-associated data e.g
- Embodiments of the methods disclosed herein preferably apply, include, and/or are otherwise associated with next-generation sequencing (NGS) (e.g., processing biological samples to generate sequence libraries for sequencing with next-generation sequencing systems; etc.).
- NGS next-generation sequencing
- Embodiments of the methods disclosed herein can include, apply, and/or otherwise be associated with semiconductor-based sequencing technologies. Additionally or alternatively, embodiments of the methods disclosed herein can include, apply, and/or otherwise be associated with any suitable sequencing technologies (e.g., sequencing library preparation technologies; sequencing systems; sequencing output analysis technologies; etc.). Sequencing technologies preferably include next- generation sequencing technologies.
- Next-generation sequencing technologies can include any one or more of high-throughput sequencing (e.g., facilitated through high-throughput sequencing technologies; massively parallel signature sequencing, Polony sequencing, 454 pyrosequencing, Illumina sequencing, SOLiD sequencing, Ion Torrent semiconductor sequencing and/or other suitable semiconductor-based sequencing technologies, DNA nanoball sequencing, Heliscope single molecule sequencing, Single molecule real time (SMRT) sequencing, Nanopore DNA sequencing, etc.), any generation number of sequencing technologies (e.g., second-generation sequencing technologies, third-generation sequencing technologies, fourth-generation sequencing technologies, etc.), sequencing-by-synthesis, tunneling currents sequencing, sequencing by hybridization, mass spectrometry sequencing, microscopy-based techniques, and/or any suitable next-generation sequencing technologies.
- high-throughput sequencing e.g., facilitated through high-throughput sequencing technologies; massively parallel signature sequencing, Polony sequencing, 454 pyrosequencing, Illumina sequencing, SOLiD sequencing, Ion Torrent semiconductor sequencing and/or other suitable semiconductor-based sequencing
- embodiments of the methods disclosed herein can include applying next-generation sequencing technologies to sequence libraries prepared for facilitating generation of sequence reads associated with a plurality of biomarkers for responsiveness to one or more immune checkpoint therapies (e.g., PD-1/PD-L1 inhibitors; etc.).
- immune checkpoint therapies e.g., PD-1/PD-L1 inhibitors; etc.
- sequencing technologies can include any one or more of: capillary sequencing, Sanger sequencing (e.g., microfluidic Sanger sequencing, etc.), pyrosequencing, nanopore sequencing (Oxford nanopore sequencing, etc.), and/or any other suitable types of sequencing facilitated by any suitable sequencing technologies.
- Embodiments of the methods disclosed herein can include, apply, perform, and/or otherwise be associated with any one or more of: sequencing operations, alignment operation (e.g., sequencing read alignment; etc.), lysing operations, cutting operations, tagging operations (e.g., with barcodes; etc.), ligation operations, fragmentation operations, amplification operations (e.g., helicasedependent amplification (HDA), loop mediated isothermal amplification (LAMP), self-sustained sequence replication (3SR), nucleic acid sequence based amplification (NASBA), strand displacement amplification (SDA), rolling circle amplification (RCA), ligase chain reaction (LCR), etc.), purification operations, cleaning operations, suitable operations for sequencing library preparation, suitable operations for facilitating sequencing and/or downstream analysis, suitable sample processing operations, and/or any suitable sample- and/or sequence -related operations.
- sample processing operations can be performed for processing biological samples to generate sequencing libraries for facilitating characterization of a plurality of biomark
- data described herein can be associated with any suitable temporal indicators (e.g., seconds, minutes, hours, days, weeks, time periods, time points, timestamps, etc.) including one or more: temporal indicators indicating when the data was collected, determined, transmitted, received, and/or otherwise processed; temporal indicators providing context to content described by the data; changes in temporal indicators (e.g., data over time; change in data; data patterns; data trends; data extrapolation and/or other prediction; etc.); and/or any other suitable indicators related to time.
- treatment response characterizations can be performed over time for one or more patients, to facilitate patient monitoring, therapy effectiveness evaluation, additional treatment provision facilitation, and/or other suitable purposes.
- parameters, metrics, inputs, outputs, and/or other suitable data can be associated with value types including any one or more of: binary values (e.g., binary status determinations of presence or absence of one or more biomarkers associated with positive responsiveness to immune checkpoint therapies and/or other suitable therapies, etc.), scores (e.g., aggregate scores indicative of a probability and/or degree of responsiveness to therapies described herein; etc.), values indicative of presence of, absence of, degree of responsiveness to one or more therapies described herein, classifications (e.g., patient classifications for sensitivity to therapies described herein; patent classifications based on absence or presence of different biomarkers of a set of biomarkers associated with responsiveness to therapies described herein, etc.), identifiers (e.g., sample identifiers; sample labels indicating association with different cancer conditions; patient identifiers; biomarker identifiers; etc.), values along a spectrum, and/or any other suitable types of values.
- binary values e.g., binary status determinations of presence
- Any suitable types of data described herein can be used as inputs (e.g., for different models; for comparison against thresholds), generated as outputs (e.g., of different models; for use in treatment response characterizations; etc.), and/or manipulated in any suitable manner for any suitable components associated with embodiments of the methods disclosed herein.
- One or more instances and/or portions of embodiments of the methods disclosed herein can be performed asynchronously (e.g., sequentially), concurrently (e.g., in parallel; concurrently on different threads for parallel computing to improve system processing ability for immune response-associated data processing and/or treatment response characterization generation; multiplex sample processing; multiplex sequencing such as for a plurality of biomarkers in combination, such as in a minimized number of sequencing runs; etc.), in temporal relation to a trigger event (e.g., performance of a portion of a method disclosed herein), and/or in any other suitable order at any suitable time and frequency by and/or using one or more instances of embodiments of inventions described herein.
- a trigger event e.g., performance of a portion of a method disclosed herein
- Embodiments of a system to perform the methods described herein can include one or more: sample handling systems (e.g., for processing samples; for sequencing library generation; etc.); sequencing systems (e.g., for sequencing one or more sequencing libraries; etc.); computing systems (e.g., for sequencing output analysis; for immune response-associated data collection and/or processing; for treatment response characterization generation; for any suitable computational processes; etc.); treatment systems (e.g., for providing treatment recommendations; for facilitating patient selection for clinical trials; for therapy provision; etc.); and/or any other suitable components.
- sample handling systems e.g., for processing samples; for sequencing library generation; etc.
- sequencing systems e.g., for sequencing one or more sequencing libraries; etc.
- computing systems e.g., for sequencing output analysis; for immune response-associated data collection and/or processing; for treatment response characterization generation; for any suitable computational processes; etc.
- treatment systems e.g., for providing treatment recommendations; for facilitating patient selection for clinical trials; for therapy provision; etc.
- Embodiments of the system and/or portions of embodiments of the system described herein can entirely or partially be executed by, hosted on, communicate with, and/or otherwise include one or more: remote computing systems (e.g., a server, at least one networked computing system, stateless, stateful; etc.), local computing systems, user devices (e.g., mobile phone device, other mobile device, personal computing device, tablet, wearable, head-mounted wearable computing device, wrist-mounted wearable computing device, etc.), databases (e.g., including sample data and/or analyses, sequencing data, user data, data described herein, etc.), application programming interfaces (APIs) (e.g., for accessing data described herein, etc.) and/or any suitable components.
- Communication by and/or between any components of the system and/or other suitable components can include wireless communication (e.g., WiFi, Bluetooth, radiofrequency, Zigbee, Z-wave, etc.), wired communication, and/or any other suitable types of communication.
- Components of embodiments of methods and systems described herein can be physically and/or logically integrated in any manner (e.g., with any suitable distributions of functionality across the components). Portions of embodiments of methods and systems described herein are preferably performed by a first party but can additionally or alternatively be performed by one or more third parties, users, and/or any suitable entities. However, of methods and systems described herein can be configured in any suitable manner.
- Embodiments of the methods disclosed herein can include collecting immune response-associated data derived from one or more biological samples, which can function to collect (e.g., generate, determine, receive, etc.) data associated with immune response functionality, for enabling characterization of one or more patients in relation to responsiveness to immune checkpoint therapy (e.g., calculating IRS values with one or more processors).
- Immune response-associated data preferably includes data indicative of biological phenomena associated with (e.g., influencing, influenced by, related to, part of, including components of, etc.) the immune response and/or immune system; however, immune response-associated data can include any suitable data (e.g., derivable by sample processing techniques, bioinformatic techniques, statistical techniques, sensors, etc.) related to the immune response and/or immune system.
- any of the variants described herein e.g., embodiments, variations, examples, specific examples, figures, etc.
- any portion of the variants described herein can be additionally or alternatively combined, aggregated, excluded, used, performed serially, performed in parallel, and/or otherwise applied.
- Portions of embodiments of the methods and systems can be embodied and/or implemented at least in part as a machine (e.g., processor) configured to receive a computer-readable medium storing computer-readable instructions.
- the instructions can be executed by computer- executable components that can be integrated with embodiments of the systems and methods described herein.
- the computer-readable medium can be stored on any suitable computer-readable media such as RAMs, ROMs, flash memory, EEPROMs, optical devices (CD or DVD), hard drives, floppy drives, or any suitable device.
- the computer-executable component can be a general or application specific processor, but any suitable dedicated hardware or hardware/firmware combination device can alternatively or additionally execute the instructions.
- compositions, methods, and respective component! s) thereof that are essential to the method or composition, yet open to the inclusion of unspecified elements, whether essential or not.
- the term “consisting essentially of’ refers to those elements required for a given embodiment. The term permits the presence of elements that do not materially affect the basic and novel or functional characteristic(s) of that embodiment.
- the invention includes embodiments that relate analogously to any intervening value or range defined by any two values in the series, and that the lowest value may be taken as a minimum and the greatest value may be taken as a maximum.
- Numerical values include values expressed as percentages. For any embodiment of the invention in which a numerical value is prefaced by “about” or “approximately”, the invention includes an embodiment in which the exact value is recited. For any embodiment of the invention in which a numerical value is not prefaced by “about” or “approximately”, the invention includes an embodiment in which the value is prefaced by “about” or “approximately”.
- PCR based comprehensive genomic profiling was performed on formalin-fixed paraffin-embedded solid tumor tissue using StrataNGS (Strata Oncology, Ann Arbor, MI) as previously described (Tomlins et al, Journal of Precision Oncology, 2020).
- StrataNGS Strata Oncology, Ann Arbor, MI
- immune gene expression levels were quantified by an analytically validated investigational assay (Strata Oncology, Ann Arbor, MI). Briefly, exon-spanning PCR amplicons were selected for each target gene and 3 housekeeping genes.
- Ion Torrent-based next-generation sequencing was performed targeting -1,000,000 reads per sample.
- Target gene expression was normalized to housekeeping genes and reads per million (nRPM) as compared to a normal control sample.
- TTNT Real-world time to next treatment
- IRS groups were established by dividing the dataset into 8 equal IRS bins and then combining bins based on overlapping TTNT curves.
- Table 1 Tumor types in the pembrolizumab treatment cohort.
- TTNT Real world time to next treatment
- Table 2 Univariate and multivariate biomarker analysis for predicting pebrolizumab time to next treatment. The multivariate analysis includes only the final variable set. P-value is from the Chi squared test.
- IRS 4.03 * exp(0.287 * [transformed TMB measurement] + 0.147 * [PD-1 normalized level] + 0.143 * [PD-L2 normalized level]- 0.138 * [CD4 normalized level] - 0.073 * [ADAM 12 normalized level]).
- an IRS of 10 is equal to the median hazard rate observed in the dataset, values greater than 10 represent decreased hazard (i.e. more benefit from pembrolizumab) and values less than 10 represent increased hazard (i.e. less benefit from pembrolizumab). Patients were assigned to one of three IRS groups to compare patient outcomes (Table 3).
- TMB antigenicity
- PD-L1 and/or PD-L2 were all independent predictors of benefit, indicating a multiplicative predictive effect across the three biomarkers, with increased antigenicity (TMB) and increased immune checkpoint activity (PD-L1 and/or PD-L2) driving benefit from immune checkpoint blockade. While many past studies have established TMB and PD-L1 as predictive biomarkers, and recent studies have established the PD-L2 is also predictive, this is the first to combine and optimize these three variables into a single model, likely providing a more comprehensive predictor of immunotherapy benefit.
- IRS has potential application for both refining the use of pembrolizumab in tumor types for which immunotherapy is indicated and in selecting patients for immunotherapy outside of indicated tumor types (e.g., off-label). It is shown that in low IRS groups, among tumor types such as NSCLC for which pembrolizumab is approved, pembrolizumab has little to no benefit relative to chemotherapy. Given that immunotherapy is expensive and can pose serious toxicity, its use should be considered more cautiously in low IRS groups. Among tumor types for which immunotherapy is not indicated, patients that fall into high IRS groups should be considered for treatment given the potential for significant benefit relative to chemotherapy.
- Pembrolizumab was recently approved for TMB high tumors (>10 mutations per megabase), independent of tumor type, eliciting a 25% objective response rate.
- the disclosed data suggests that an integrated model combining TMB with immune gene expression provides better prediction, stratifying treatment outcomes within TMB high and TMB low patients.
- NCT03061305 The Strata Trial (NCT03061305), is an observational clinical trial evaluating the impact of molecular profiling on patients with advanced solid tumors. It has been reviewed and approved by Advarra Institutional Review Board (IRB; IRB Pro00019183) prior to study start. At enrolling health care systems, all adult patients with locally advanced (stage III), unresectable or metastatic (stage IV) solid tumors and available FFPE tumor tissue were eligible; the protocol also allowed enrollment of patients with rare early-stage tumors.
- stage III locally advanced
- stage IV unresectable or metastatic
- the Strata Clinical Molecular Database contains deidentified subject, molecular profiling, treatment, and survival data for all enrolled NCT03061305 participants.
- Prior antineoplastic therapy including start and stop dates, were collected for trial participants at the time of study entry.
- Antineoplastic therapy data and survival status were prospectively collected for up to 3 years from the time of enrollment and/or informed consent.
- Post-hoc power analysis was not performed to determine the sample size of this discovery cohort.
- a power analysis was then performed to determine the cohort size needed for an independent validation cohort as described below.
- additional inclusion and exclusion criteria included: 1) treatment with systemic non-pembrolizumab anti-PD- (L) 1 monotherapy, 2) the tested tissue specimen was collected prior to the PD-(L)1 therapy start date, 3) had no prior anti-PD-(L)l or CTLA4 blockade therapy prior to the non-pembrolizumab PD-(L)1 line start date, and 4) patients were not in the discovery cohort. Additional inclusion/exclusion criteria for other analyses are described below and in the overall study diagram (FIG. 40). Except in the analysis specifically assessing IRS performance in samples collected after PD-(L)1 therapy, patients with samples collected after the start date of the analyzed therapy line were excluded from all analyses.
- Source data verification in the Strata Trial was performed for high-risk data fields such as demographics and treatment history per an approved Trial Monitoring Plan. Data completeness, consistency, and quality assurance checks were performed across the Strata electronic data capture (EDC) system per an approved Data Management Plan; 100% source data verification was performed for the discovery cohort. Additional details on the Strata Trial experience and Strata molecular profiling have been described 54 56 .
- EHRs electronic health records
- TTNT time to next therapy
- OS overall survival
- All medications were classified into anti-neoplastic or non-antineoplastic treatments, and all anti-neoplastic treatments were further subclassified (e..g chemotherapy, immune-oncology [IO; PD-(L)1 or CTLA4], oncogene TKI, hormonal, etc); non- antineoplastic treatments were excluded from further consideration.
- Line of therapy assignment was performed in two stages: first, single-dose treatments with consecutive doses administered within 90 days were combined into a course of treatment with a single start and end date; next, non-overlapping lines of treatment were inferred by considering each course of medication sequentially by start date. Subsequent treatment courses that began more than 30 days after the start of a given line of treatment, or whose duration of overlap with the line was less than 50%, were considered to establish a new line of treatment. Any treatment line with more than one anti-neoplastic therapy administered during the line was considered combination therapy. First line chemo and/or hormonal therapies which concluded 180 days or more prior to the start of subsequent therapy were considered as adjuvant.
- an effective end date was defined for each course of treatment as either a) date of last record if treatment is ongoing (censored), b) date of death (event), c) the start date of the subsequent therapy line (event), or d) the latest available end date (censored if no subsequent line of therapy or death).
- rwPFS was calculated as the difference, in months, between the start date and effective end date of the treatment line.
- OS was calculated as the difference, in months, between the start date of the treatment line and date of death (or censoring).
- PCR-CGP multiplex PCR-based comprehensive genomic profiling
- FFPE solid tumor tissue using StrataNGS (Strata Oncology, Ann Arbor, MI).
- the current version of StrataNGS is a 437 gene laboratory-developed test (LDT) for FFPE tumor tissue samples performed on co-isolated DNA and RNA, which has been validated on over 1,900 FFPE tumor samples, and is covered for Medicare beneficiaries 55 . While earlier StrataNGS versions were also used during the study period, all had similar performance for the TMB assessment (and MSI) used herein 56 .
- immune gene expression was determined by analytically and clinically validated multiplex PCR-based qTP via an investigational/supplementary test performed on the same co-isolated RNA as described 54 ; different versions of this quantitative transcriptomic profiling test have been run in parallel with StrataNGS (assessing 26, 46 and currently 103 expression targets), with panel specific scaling validated by concordance analyses performed as needed.
- StrataNGS assessing 26, 46 and currently 103 expression targets
- panel specific scaling validated by concordance analyses performed as needed.
- One or more exon-spanning PCR amplicons were selected for each target gene and multiple housekeeping genes (see Supplementary Methods) were included, with 3 pan-cancer stable housekeeping genes used for clinical testing.
- qTP was performed using Ampliseq after reverse transcription followed by Ion Torrent-based next-generation sequencing.
- Expression target transcripts were measured in normalized reads per million (nRPM), whereby raw expression target read counts were normalized by a factor that results in the median housekeeping gene expression value matching the same gene's standard reads per million in a reference FFPE normal cell line sample (GM24149) run in parallel with all clinically tested samples. 54 Relevant components of the analytical and clinical validation of the current version of the integrated CGP + qTP LDT that includes the IRS model are described in the Supplementary Methods.
- TMB sensitive tumor types MSI-H, POLE mutant , NSCLC, head and neck cancer, or melanoma as TMB sensitive; all other samples as TMB insensitive
- Performance status or surrogates were not available from data collected as part of the Strata Trial. Proportional hazard assumptions were checked for each model and cohort of interest using Schoenfeld residuals.
- TMB and 23 candidate immune and proliferation gene expression biomarkers with pembrolizumab rwPFS was determined using Cox proportional hazards regression in the 648 -patient pembrolizumab (both monotherapy and combination therapy) discovery cohort.
- TMB measurements were log2-transformed and gene expression measurements were log2-transformed and median-centered per laboratory workflow prior to analysis.
- Feature selection was performed via Lasso-penalized Cox proportional hazards regression in this 648-patient discovery cohort, with the Lasso penalty term chosen as the value which maximized the concordance index via 5-fold cross validation.
- Model coefficients for the five features with non-0 coefficients in the Lasso model were finalized via standard Cox regression.
- IRS 0.273758 * TMB + 0.112641 * PD-1 + 0.061904 * PD-L1 - 0.077011 * TOP2A - 0.057991 * ADAM 12
- NCT03061375 is an observational clinical trial evaluating the impact of tumor molecular profiling for patients with advanced solid tumors. De-identified demographic, clinical and molecular data from patients in the Strata Trial is maintained in the Strata Clinical Molecular Database (SCMD).
- SCMD Strata Clinical Molecular Database
- the SCMD contains clinical and molecular data from a total of 57,648 unique patients with advanced solid tumors (from 47 tumor types) from 59 United States health care systems who had routine FFPE tumor tissue molecularly profiled by the StrataNGS CGP test 55,56 , with 9,899 Strata Trial patients from 30 United States health care systems (from 43 tumor types) having treatment data from at least one systemic antineoplastic agent (FIG. 40, and Tables SI & S2).
- TTNT Time to next therapy
- rwPFS real-world progression free survival
- IQR interquartile range
- the median number of total systemic lines of therapy per patient was 1 (IQR 1-2), with 49.2% of systemic lines being monotherapy, and the median number of systemic therapies per line was 2 (IQR 1-2).
- the median number of total systemic lines of therapy per patient after Strata trial enrollment was 1 (IQR 0-1), with 47.2% of systemic lines being monotherapy, and the median number of systemic therapies per line was 2 (IQR 1-2).
- rwPFS was inferred for each patient as the time from starting the pembrolizumab containing therapy line to the time of stopping that line and starting a new therapy line or death; both rwPFS and OS were used for studying treatment outcome based on comparisons of these endpoints (Supplementary Results and FIG. 44).
- IRS Immunotherapy Response Scores
- IRS-H patients had significantly longer pembrolizumab rwPFS (IRS-H vs. IRS-L median rwPFS 16.8 [95% CI 14.9-22.9] vs. 7.2 [95% CI 6.2-8.4] months, adjusted hazard ratio 0.49 [95% CI 0.39-0.63], p ⁇ 0.0001) and OS (IRS-H vs. IRS-L median OS Not Reached [95% CI 29.9-NA] vs.
- IRS-H also showed significant rwPFS and OS benefit when using restricted mean survival time analysis (prespecified periods of 24 months and 36 months, respectively), both in an unadjusted analysis (IRS- H vs. IRS-L average event free rwPFS 15.70 [95% CI 14.53 - 16.88] vs.10.63 [95% CI 9.61 - 11.65]; OS 25.50 [95% CI 23.61 - 27.39] vs.
- results were similar when stratifying patients by PD-1 vs. PD-L1 therapy. Taken together, these results demonstrate the development and validation of an integrative, DNA and RNA based predictor of PD- (L) 1 blockade benefit, with IRS-H patients showing significantly longer rwPFS and OS in an independent validation cohort.
- TMB has been shown to predict both monotherapy PD-1 (pembrolizumab and nivolumab) and PD-L1 (atezolizumab) benefit through both retrospective and prospective studies, although ORRs at the same TMB cutoff vary across agents and TMB cutoffs.
- ORRs at the same TMB cutoff vary across agents and TMB cutoffs.
- both TMB and IRS are reported as binary predictors (given the near requirement of categorical biomarkers for clinical implementation), therefore, to have clinical utility, the IRS model should identify a population of patients at least as large as the TMB-H population with similar PD-(L)1 benefit. As shown in FIG.
- IRS but not TMB, was an independent predictor of PD-(L)1 rwPFS (TMB-H vs.
- IRS status but not CDKN2A deep deletion status, was a significant predictor of rwPFS and OS in both the discovery (rwPFS IRS-H vs.
- CDKN2A wt adjusted hazard ratio 0.99 [95% CI 0.56-1.75], p 0.96) cohorts when CDKN2A deep deletion status was added to the appropriate adjusted Cox proportional hazards model, confirming the limitations of genomic markers alone for predicting PD-(L)1 therapy response.
- IRS identifies a larger proportion of patients than TMB alone with similar benefit from PD-(Ll) therapy, establishing clinical utility of the IRS biomarker and demonstrating the value of integrating quantitative gene expression with TMB for predicting PD-1/PD-L1 monotherapy treatment benefit. Additional analyses supporting the robustness of the IRS model to temporal sample collection (prior to CPI treatment) and variable tumor content are described in the Supplementary Results and Figures 53a-b & 54a-e.
- PD(L)-1 combination regimens are rapidly being developed and moved to earlier lines of therapy, highlighting the need for improved biomarkers that can predict PD-(L)1 monotherapy benefit.
- TPS PD-L1 IHC
- IRS identifies a larger population of patients than TMB but with similar PD-(L) 1 monotherapy benefit, however this analysis is limited by the requirement that patients received PD-1/PD-L1 treatment.
- PD-(L)1 monotherapy approved tumor types 69 had a substantially higher proportion of IRS-H patients (37.6%) than non-PD-(L)l monotherapy approved tumor types (11.7%) (FIG. 39b).
- Tumor types with the highest proportion of IRS-H group patients include several known to be highly responsive to PD-(L)1 therapy, including lymphoma, non-melanoma skin cancer, melanoma, NSCLC, and renal cell carcinoma (which nearly invariably has low TMB) (FIG. 39c).
- IRS integrative Immunotherapy Response Score
- IRS-H as more likely to benefit
- TMB alone was not a significant predictor of PD-(L)1 rwPFS, nor OS, in this cohort.
- the IRS-H population was nearly twice the size of the TMB-H population (20.9 vs. 10.8%).
- IRS-H was more frequent in tumor types known to derive benefit from PD-(L)1 therapy, IRS-H occurred in subsets of nearly every tumor type.
- liquid biopsy based TMB is not directly translatable to tissue TMB, even when both tissue and liquid biopsies are performed using FDA approved CGP devices, as in a recent study of both single agent nivolumab and nivolumab + ipilimumab combination therapy, where blood TMB’s predictive ability was conditional on tissue TMB status, but not vice versa 32 .
- the rwPFS endpoint includes some patients who stopped treatment due to treatment toxicity (not assessable herein) or switching therapy to a more appropriate regimen based on molecular results (as described in the Supplementary Results) and not disease progression, although this likely represents a minority of events, and both rwPFS and OS results were highly similar in both the discovery and validation cohorts.
- IRS had essentially similar predictive ability in both the training and validation cohorts (Table S10) when the tumor type term in our adjusted models (most common tumor type vs. others) was replaced with a term using MSKCC defined TMB sensitive vs.
- insensitive tumor types MSI-H, POLE 1 "" 11 " 111 , NSCLC, head and neck cancer or melanoma as sensitive; all others as insensitive 57 ), supporting the more pan-solid tumor nature of IRS vs. TMB alone.
- CDKN2A copy loss which has been identified in two studies as improving upon TMB status for predicting PD-(L)1 response 60,68 , was not a significant predictor of PD-(L)1 rwPFS or OS in either the discovery or validation cohorts, future studies will be required to determine whether inclusion of other single gene -based DNA biomarkers identified as potentially predictive in one or more tumor types (e.g.
- STK11, PBRM1, or additional immune related genes assessed on the current expanded qTP panel can improve the performance of the IRS model; given the clearly established clinical utility for MSI-H status, this biomarker was not included in IRS model development.
- Limited PD-L1 IHC data was available for subjects in the SCMD with PD-(L)1 treatment outcomes, and hence we are not able to directly compare performance of IRS and PD-L1 IHC (or other immunotherapy response biomarkers beyond TMB), which is particularly relevant for our exploratory analysis of pembrolizumab monotherapy vs. combination therapy in first line NSCLC, however we used propensity score matching by PD-L1 qTP expression to mitigate this limitation.
- IRS-H/TMB-L or where both monotherapy and combination therapy are indicated (e.g. IRS-H in PD-L1 IHC 1-49%).
- IRS-H in PD-L1 IHC 1-49% we identified an ultra-low subset of the IRS-L population that shows particularly poor PD-(L) 1 rwPFS and OS (FIGS. 57a-c & FIGS. 59a-d), suggesting that it may be possible to identify patients more likely to benefit from other therapies in PD-(L) 1 approved tumor types when therapeutic choice is present.
- IRS was developed and validated using a single, clinically validated NGS platform capable of simultaneously performing CGP (required for TMB but also for assessing non-immunotherapy treatment biomarkers) and simultaneous, precise quantification of tumor- and tumor microenvironment (TME-relevant gene expression from minute FFPE tumor specimens.
- IRS-H/TMB-L a population shown herein to have similar or better PD-(L)1 benefit as TMB-H — markedly expanding the benefit of immunotherapy across solid tumors by addressing one of the most important challenges in precision oncology.
- TMB tumor mutational burden
- NCCN National Comprehensive Cancer Network
- first-line targeted oncogene TKI i.e., osimertinib, afatinib, alectinib, brigatinib, capmatinib, entrectinib, dabrafenib + trametinib, entrectinib, lorlatinib, selpercatinib or tepotinib
- oncogene TKI i.e., osimertinib, afatinib, alectinib, brigatinib, capmatinib, entrectinib, dabrafenib + trametinib, entrectinib, lorlatinib, selpercatinib or tepotinib
- CGP + qTP laboratory developed test
- LDT Clinical Laboratory Improvement Amendments
- CNA CNA-like nucleic acid
- MSI MSI status
- TMB in mutations/megabase [Muts/Mb]
- Specimen evaluation, processing, and molecularly informed tumor content determination, nucleic acid isolation, quantification, normalization, library preparation, sequencing, quality control and CGP reporting are essentially as described in the validation of the StrataNGS CGP testl.
- the final molecularly informed tumor content is set after completion of StrataNGS testing, initially incorporating variant allele frequencies (VAF) and copy state of mutations in tumor suppressors known to nearly always be homozygous2, with subsequent refinement integrating these results with the remainder of the mutational and copy number profiles along with b-allele frequencies from high confidence SNPsl.
- VAF variant allele frequencies
- the final MTC was set by a single pathologist (S.A.T.) with automatic updating to all applicable variants/ variant classes.
- the current CGP component of the CGP + qTP is an updated version of the validated StrataNGS CGP test, which is covered for Medicare beneficiaries with advanced solid tumors with performance characteristics characterized across >30,000 consecutively submitted FFPE tumor tissue samplesl,3.
- the current version targets 59 fusion driver genes (vs. 46 previously), resulting in a total of 437 unique genes across the CGP test.
- the updated version leverages an independently trained and validated random forest classifier to detect gene fusions, and an independently trained and validated random forest classifier to detect mutations (with equivalent or better performance to the previous version for all variant classes).
- the TMB component of the CGP+qTP test has been extensively validatedl and has not been updated.
- the TMB assessment for the CGP + qTP LDT includes non-coding (at well characterized genomic loci) and coding, synonymous and non- synonymous, SNV and multi-nucleotide (two bases) variants from a panel with a maximum footprint of 1.7 Mb.
- non-coding at well characterized genomic loci
- coding synonymous and non- synonymous, SNV and multi-nucleotide (two bases) variants from a panel with a maximum footprint of 1.7 Mb.
- VAF > *4 of the final molecularly informed tumor content are included in the TMB estimate (inclusion of only clonal mutations and tumor content correction have both been shown to improve prediction of checkpoint inhibitor response 4,5).
- Additional custom filtering is employed to exclude high likelihood technical artifacts and germline variants and the TMB (Muts/Mb) estimate is calculated via the total number of positions with sufficient depth of coverage necessary for definitive assessment (maximum possible 1.7Mb).
- the qTP component of the CGP + qTP test is completed using the same aliquot of RNA and multiplex PCR panel used in StrataNGS (for gene fusions), with the current version of the panel containing 950 amplicons targeting individual gene fusion isoforms involving 59 driver genes [chimeric amplicons; reported as CGP variants as described above], and 106 non-chimeric amplicons (103 target genes and 3 housekeeping genes) used for quantitative expression profiling; pre-clinical and investigational versions of the quantitative expression profiling component have included 26, 46 and 103 target genes.
- Target gene expression is determined in scaled, log2, median-centered units of normalized reads per million [nRPM]6, representing target gene expression normalized to housekeeping gene expression, then scaled to the distribution observed in a common control (normal FFPE genome in a bottle Ashkenazi father cell line [GIAB; GM24149 from Horizon Discovery]) per panel. For reporting, results are then scaled to a fixed pan-cancer median value of 10 after log2 normalization. Hence, all individually reported targets have a median value of 10.0 across all pansolid tumor samples tested, with each unit increase representing a doubling from the median.
- Linearity was thus determined by the concordance correlation coefficient for each target gene after setting all sub-LOQ values to LOQ, and the dynamic range was defined as the LOQ to the highest expressing value for that gene in replicate #1. No upper LOQ is established as there is essentially no chance of clinical misinterpretation of a value higher than that established in this approach given the observed linearity. Hence, the reportable range for each amplicon is floored at the LOQ but has no upper limit. Linearity LOQs have not been applied to any other validation analysis performed herein (unless specified) to present the full range of qTP generated data.
- IRS produces a quantitative hazard that is proportional to the hazard rate observed in the dataset, so that higher values represent decreased hazard (i.e., more benefit from PD-(L)1 therapy benefit) and smaller values represent increased hazard (i.e., less benefit from PD-(L)1 therapy).
- the individual scaled hazard rate is reported for informational purposes however the IRS result is reported categorically as IRS-High or IRS-Low.
- measurand LOD/LOQ and linearity is not applicable, and the reportable range of the quantitative scaled hazard rate is reported as determined without upper or lower bounds.
- Performance of the IRS model with or without LOQs applied to the individual expression components was compared and concordance correlation of IRS scores and quantification of the % of patients changing IRS groups (IRS-H to IRS-L or vice versa) was determined.
- the accuracy of the qTP component was validated through a multi-part accuracy study leveraging qRT-PCR, comparison to known target gene expression across tumor types, and clinical immunohistochemistry.
- clinical FFPE tumor samples from StrataNGS testing and 3 control RNA samples were subjected to qTP and qRT-PCR on replicate RNA aliquots.
- 2-20ng clinically isolated FFPE RNA per sample underwent reverse transcription using SuperScript IV VILO Master Mix (Invitrogen) and pre-amplification using TaqMan PreAmp Master Mix (Applied Biosystems) using a pooled Taqman primer/hydrolysis probe assays and 14 cycles.
- qPCR was then performed in duplicate on a Quantstudio 3 Real Time PCR system using a 1:20 dilution of amplified product per qPCR reaction and TaqMan Fast Universal PCR Master Mix (Applied Biosystems). Individual amplicon level thresholds and baselines were set during the exponential amplification phase to determine cycle crossing threshold (Ct) values. Samples with duplicate qPCR values > 2 Ct difference were excluded unless both values were >30 or singlicate experiments were performed. All undefined Ct values were considered as having Ct of 40.
- qRT-PCR ACt values were determined as: target amplicon Ct - (median of housekeeping gene amplicon Ct) and were otherwise scaled as for qTP results using the same FFPE reference sample run in all clinical and validation runs. Panel- wide concordance correlation coefficient were determined across all included target genes and samples in the cohort. Acceptable concordance correlation coefficient of >0.7 was pre-specified given the expected range of transcript expression across the amplicons/samples.
- IRS was trained on data from both the current qTP panel and the previous 46 gene version (with appropriate panel-specific scaling), we also compared TCGA and qTP results for the 20 immune and proliferation expression candidates included in IRS development (IFNG was excluded from this analysis as it could not be reliably quantified across all qTP panels) across the 24,463 Strata Trial samples with complete sample information used to assess IRS distribution (see FIG. 39a-d). Spearman correlation was determined between TCGA and qTP profiled tumors for all candidate biomarkers.
- TPS does not include PD-L1 expression in non-tumor cells (as for CPS using 22C3 in other tumor types and routinely included in PD-L1 expression by other PD-L1 IHC clones [e.g., SP142], acceptable accuracy was pre-specified as statistically significant, ordinally increasing differences in median qTP PD-L1 expression between the three clinically relevant TPS groups (Kruskal Wallis test, p ⁇ 0.05; Jonckheere-Terpstra trend test [increasing median from 0%, 1-49%, and >50%, p ⁇ 0.05).
- Tumor content LOD was determined by binning the development and validation cohort samples by tumor content (20-35%, 40-70% and >70%) and visualizing TTNT by Kaplan Meier analysis, given that the established LOD for accurate TMB estimation was determined as 20% tumor content, and included a tumor content term (continuous tumor content) in the overall adjusted CPH model in the IRS development cohort (including age, gender, most common tumor type [NSCLC] vs. others, therapy type [monotherapy/combination], and line of therapy) and validation cohort (including age, gender, most common tumor type [melanoma] vs. others, therapy type [PD-1 vs. PD-L1], and line of therapy).
- a tumor content term continuous tumor content
- the threshold was set to favoring NPA and pre-specified acceptable NPA (versus PR 0% IHC) of greater than 95% was set.
- TTNT before vs. after StrataNGS results, median TTNT 22.8 [95% CI 17.3- 31.8] months vs.
- the second was a patient with metastatic melanoma who was briefly treated with pembrolizumab, then ipilimumab + nivolumab, prior to an extended course with imatinib (the patient harbored two VUS in KIT; FIG. 44 blue box), the third was a patient with metastatic melanoma who was treated with pembrolizumab prior to prolonged treatment with ipilumumab monotherapy (FIG.
- the qTP component of the CGP + qTP test used to report IRS is performed from the same aliquot of RNA and multiplex PCR panel used in StrataNGS (for gene fusions), with the current version targeting 106 non-chimeric amplicons (103 target genes and 3 housekeeping genes) used for quantitative expression profiling.
- Target gene expression is determined in scaled, log2, mediancentered units of normalized reads per million [nRPM]16,17, representing target gene expression normalized to housekeeping gene expression (from HMBS, CIAO1 and EIF2B1; see below), then scaled to the distribution observed in a common FFPE cell line control per panel.
- All 106 amplicons (median insert length 94.5, range 61-120) use primers (median primer length 23, range 10-31) that span exon-exon boundaries and only full length reads ( ⁇ 2 base-level mismatches) are counted, ensuring specificity of all normalized target gene values from each amplicon.
- the qTP panel includes amplicons for quality control (candidate housekeeping genes, separate amplicons targeting different regions the same transcript) and multiple classes of target gene expression biomarkers, including those potentially useful for determining hormone receptor status (see below), those measuring proliferation index, and individual biomarkers that may have utility in predicting response (or clinical trial suitability/enrollment,) to investigational or approved expression based therapies (e.g., antibodies, antibody drug conjugates [ADCs], bispecific antibodies, radiopharmaceuticals, CAR-T cells, TCRs, etc.).
- ADCs antibody drug conjugates
- Initial pre-clinical versions of the qTP panel contained 6 “positive control” genes across two RNA primer pools previously used in the RNA fusion component of the Oncomine Focus/Precision Assay.
- pan-cancer housekeeping genes for quantitative expression profiling and/or identify other candidates.
- Correlation of variation was determined for each gene per tumor type, and candidate housekeeping genes were ranked by the number of tumor types in which they ranked in the top 20 most stable genes (by lowest CV).
- Uniformly processed gene expression data in transcripts per million [TPM]) from the 18 highest ranking housekeeping genes, the 6 OPA “positive control genes”, as well as the commonly used housekeeping gene GAPDH were then downloaded for 20,841 total samples contained in the MiPanda databases, which includes 935 Cancer Cell Line Encyclopedia cell lines (from 20 tumor types), 9,966 normal tissue samples (730 TCGA samples from 20 tissue types and 9,236 GTEX samples from 30 tissue types), and 9,940 TCGA cancer tissue samples (9,496 primary samples from 25 tumor types and 444 metastases from 16 tumor types).
- pan-cancer pan-normal tissue stable genes with the lowest average expression (in TPM) as candidate housekeeping genes for the qTP component of the CGP + qTP test thus selected five genes for inclusion: SLC4A1AP, CTCF, EIF2B1, CIAO1 and GGNBP2.
- TPM pan-normal tissue stable genes with the lowest average expression
- sensitivity/specificity/accuracy and linearity/LOQ are complicated in multiplex RNA sequencing due to the lack of absolute standards that can be assessed in a complex RNA mixture with variable RNA amplifiability, individual amplicon efficiency, and the difficulty in appropriately choosing a diluent (water to reduce input RNA amount, normal DNA for genomic alterations, or “normal” RNA to reduce relative amount of highly expressed tumor-specific transcripts are all inappropriate for a multiplex RNAseq pan-tumor approach targeting tumor and tumor microenvironment targets).
- a diluent water to reduce input RNA amount, normal DNA for genomic alterations, or “normal” RNA to reduce relative amount of highly expressed tumor-specific transcripts are all inappropriate for a multiplex RNAseq pan-tumor approach targeting tumor and tumor microenvironment targets.
- the accuracy of the qTP panel and IRS expression components was validated through a multi-part accuracy study leveraging representational qRT-PCR validation, comparison to known target gene expression across tumor types (via comparison to TCGA
- the accuracy of the qTP component of the integrate CGP + qTP test was first determined by representational validation through evaluating target gene expression concordance with hydrolysis probe based qRT-PCR, the gold standard for gene-expression measurement.18 Clinical FFPE tumor samples from StrataNGS testing and 3 control RNA samples were subjected to qTP and qRT-PCR on replicate RNA aliquots. Comparison of target gene expression by qTP and qRT-PCR was performed for 32 target genes in 3 control samples and 24 clinical FFPE tumor samples (analytical validation) and the 24 FFPE tumor samples only (clinical validation). In the analytical validation, the observed concordance correlation coefficient was 0.837 (95% CI 0.816-856; p ⁇ 0.0001).
- the observed concordance correlation coefficient was 0.842 (95% CI 0.820-862; p ⁇ 0.0001);
- the concordance correlation coefficient in the clinical validation was 0.833 (95% CI 0.760-0.886).
- hydrolysis probe based qRT-PCR is the gold standard for RNA transcript quantification
- IHC is the clinical gold standard for clinically relevant target expression evaluation. Therefore, we used optical character recognition and natural language processing to prioritize accompanying pathology reports received with StrataNGS test requests for abstraction of IHC biomarker results as described in the Supplementary Methods. Accuracy results relevant to IRS are described here, with results relevant to breast cancer biomarkers shown in FIGS. 56a-g.
- IRS status could be generated for all 276 NSCLC samples in the PD-L1 IHC cohort, with 31.0%, 34.2% and 58.0% of TPS 0%, 1-49%, and >50% samples being IRS- H, respectively.
- Accuracy of the TOP2A qTP component of IRS was validated against clinical IHC using a cohort of 956 FFPE tumor tissue samples (36 tumor types) with reportable qTP with proliferation index (percentage of Ki67 positive tumor cells) in accompanying pathology reports. As shown in FIG. 47b, TOP2A by qTP was strongly correlated to Ki67 proliferative index (correlation coefficient 0.753 [95% CI 0.724-0.780], p ⁇ 0.0001). These results further support the accuracy of the gene expression component of IRS.
- Tissue based TMB has recently been shown to be stable for nearly all patients with advanced cancer through whole genome sequencing of sequential tissue samples, 19 however less is known about the stability of an integrative CGP + qTP model predicting pembrolizumab benefit.
- the actual tumor content of a given sample is impacted both by normal cells unrelated to the gene expression component of the IRS model (such as benign epithelial cells), as well as tumor infiltrating lymphocytes (such as those that express PDCD1 and/or PD-L1) and cancer associated fibroblasts that express ADAM 1220-25, with these components (and the actual tumor content) directly relevant to the predictive ability of the IRS algorithm.
- normal cells unrelated to the gene expression component of the IRS model
- tumor infiltrating lymphocytes such as those that express PDCD1 and/or PD-L1
- cancer associated fibroblasts that express ADAM 1220-25
- LOD tumor content limit of detection
- TMB tumor-derived protein
- PD-1 expression PD-L1 expression were each independent predictors of pembrolizumab benefit, indicating a multiplicative predictive effect across these biomarkers representing increased antigenicity (TMB) and the direct targets of both PD-1 and PD-L1 monoclonal antibodies.
- PD-L1 evaluation by IHC is the current FDA-approved biomarker to predict PD-(L)1 benefit either individually or in models 26 28
- expression varies by antibody clone and nearly all studies show at least some responsive PD-L1-IHC low/negative patients 29 33 .
- PD-1 expression in both CD8+ and all lymphocytes has also been shown to be predictive of PD-(L)1 therapy benefit, most notably in Merkel cell carcinoma, where PD-1+ and PD-L1+ cell density, as well as close proximity of PD-1 and PD-L1+ cells, were associated with treatment response, while CD8+ cell density (nor CD8+ and PD-L1+ cell proximity) was not 28 .
- TGF-[3 signaling from CAFs has been shown to drive T cell exclusion, a hallmark of low response to ICI 47 51 .
- these results support additional investigation into a potential mechanistic role for ADAM 12 in ICI resistance, as well as demonstrate the complementary nature of the integrative biomarkers in the IRS model, which integrates measurement of tumor neo-antigenicity (TMB), with quantification of key tumor and TME biomarkers.
- TMB tumor neo-antigenicity
- Niknafs, Y. S. et al. MiPanda A Resource for Analyzing and Visualizing Next-Generation Sequencing Transcriptomics Data. Neoplasia 20, 1144-1149, doi:10.1016/j.neo.2018.09.001 (2016).
- ADAM12 is a costimulatory molecule that determines Thl cell fate and mediates tissue inflammation.
- ADAM12 is a prognostic factor associated with an aggressive molecular subtype of high-grade serous ovarian carcinoma.
- Demographics considered for each cohort included gender, race, ethnicity, and line of therapy.
- pembrolizumab monotherapy and combination therapy containing lines were included.
- PD-(L)1 validation cohort only PD-1 or PD- L1 monotherapy (excluding pembrolizumab) were included.
- TCGA The correlation of expression profiles of 20 candidate genes between 9,223 TCGA tumors and 18,305 quantitative transcriptomic profiling (qTP) assessed tumors (of 24,463 samples, limited to 27 directly comparable tumor types) was determined. The number of samples used in comparison (n), the correlation (Spearman rho, P) and the significance with respect to no correlation (p-value, reported in scientific notation).
- TCGA data was obtained from cBioPortal. Components of the final Immunotherapy Response Score (IRS) model are bolded.
- PD-L1 CD274
- PD-1 two independent target amplicons were assessed for each gene; normalized target gene expression was averaged from the independent amplicons (per gene) to yield a composite result.
- TCGA the Cancer Genome Atlas. As limit of quantification (LOQ) could not be established for IFNG by Strata Multiplex PCR based-NGS profiling, it was excluded from this analysis.
- Table S8 Dependency of rwPFS from immediately prior therapy to pembrolizumab monotherapy after adjustment for various covariates Table S9. Pre-/post-propensity score matching covariates in first-line NSCLC cohort treated with pembrolizumab monotherapy or pembrolizumab + chemotherapy combination therapy
- NN nearest neighbor
- sd standard deviation
- n sample size
- % percent
- NA not applicable
- IRS immunotherapy response score
- TMB tumor mutation burden. * p-value is based on a t-test for difference in means for continuous variables and a Fisher's exact test for categorical variables
- the primary analyses presented herein used most common vs. other (e.g. NSCLC vs. all other tumor types in the discovery cohort) as the tumor type term in the Cox proportional hazards model used to evaluate the predictive ability of IRS for rwPFS and OS in the discovery (monotherapy and combination therapy) and validation cohorts.
- the impact on the adjusted HR (for IRS-High [H] vs. IRS-Low [ L] ) and p- value in the model was determined after replacing that tumor type term with the MSKCC definition of TMB sensitive tumor types (MSI-H, POLF mu ’ an ’, NSCLC, head and neck cancer, or melanoma as TMB sensitive; all other samples as TMB insensitive) 52 .
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Genetics & Genomics (AREA)
- Engineering & Computer Science (AREA)
- Physics & Mathematics (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Biophysics (AREA)
- General Health & Medical Sciences (AREA)
- Molecular Biology (AREA)
- Organic Chemistry (AREA)
- Biotechnology (AREA)
- Analytical Chemistry (AREA)
- Spectroscopy & Molecular Physics (AREA)
- Bioinformatics & Computational Biology (AREA)
- Evolutionary Biology (AREA)
- Theoretical Computer Science (AREA)
- Zoology (AREA)
- Immunology (AREA)
- Wood Science & Technology (AREA)
- Pathology (AREA)
- Medical Informatics (AREA)
- Microbiology (AREA)
- Oncology (AREA)
- Hospice & Palliative Care (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Description
Claims
Priority Applications (7)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
KR1020247018785A KR20240134293A (en) | 2021-11-08 | 2022-11-08 | Cancer Biomarkers for Immune Checkpoint Inhibitors |
JP2024526946A JP2024540391A (en) | 2021-11-08 | 2022-11-08 | Cancer Biomarkers for Immune Checkpoint Inhibitors |
AU2022381055A AU2022381055A1 (en) | 2021-11-08 | 2022-11-08 | Cancer biomarkers for immune checkpoint inhibitors |
CA3237909A CA3237909A1 (en) | 2021-11-08 | 2022-11-08 | Cancer biomarkers for immune checkpoint inhibitors |
MX2024005612A MX2024005612A (en) | 2021-11-08 | 2022-11-08 | Cancer biomarkers for immune checkpoint inhibitors. |
CN202280081429.0A CN118660976A (en) | 2021-11-08 | 2022-11-08 | Cancer biomarkers for immune checkpoint inhibitors |
EP22890932.1A EP4430214A1 (en) | 2021-11-08 | 2022-11-08 | Cancer biomarkers for immune checkpoint inhibitors |
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202163277158P | 2021-11-08 | 2021-11-08 | |
US63/277,158 | 2021-11-08 | ||
US202263407606P | 2022-09-16 | 2022-09-16 | |
US63/407,606 | 2022-09-16 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2023081537A1 true WO2023081537A1 (en) | 2023-05-11 |
Family
ID=86241980
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2022/049332 WO2023081537A1 (en) | 2021-11-08 | 2022-11-08 | Cancer biomarkers for immune checkpoint inhibitors |
Country Status (7)
Country | Link |
---|---|
EP (1) | EP4430214A1 (en) |
JP (1) | JP2024540391A (en) |
KR (1) | KR20240134293A (en) |
AU (1) | AU2022381055A1 (en) |
CA (1) | CA3237909A1 (en) |
MX (1) | MX2024005612A (en) |
WO (1) | WO2023081537A1 (en) |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2025101693A1 (en) * | 2023-11-07 | 2025-05-15 | Altos Labs, Inc. | Photomics analysis of samples |
Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20200263261A1 (en) * | 2017-11-06 | 2020-08-20 | Genentech, Inc. | Diagnostic and therapeutic methods for cancer |
-
2022
- 2022-11-08 JP JP2024526946A patent/JP2024540391A/en active Pending
- 2022-11-08 CA CA3237909A patent/CA3237909A1/en active Pending
- 2022-11-08 KR KR1020247018785A patent/KR20240134293A/en active Pending
- 2022-11-08 EP EP22890932.1A patent/EP4430214A1/en active Pending
- 2022-11-08 AU AU2022381055A patent/AU2022381055A1/en active Pending
- 2022-11-08 WO PCT/US2022/049332 patent/WO2023081537A1/en active Application Filing
- 2022-11-08 MX MX2024005612A patent/MX2024005612A/en unknown
Patent Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20200263261A1 (en) * | 2017-11-06 | 2020-08-20 | Genentech, Inc. | Diagnostic and therapeutic methods for cancer |
Non-Patent Citations (2)
Title |
---|
TOMLINS SCOTT A., KHAZANOV NICKOLAY A., BULEN BENJAMIN J., HOVELSON DANIEL H., SHREVE MELISSA J., LAMB LAURA E., MATRANA MARC R., : "Development and validation of an integrative pan-solid tumor predictor of PD-1/PD-L1 blockade benefit", COMMUNICATIONS MEDICINE, vol. 3, no. 1, XP093065949, DOI: 10.1038/s43856-023-00243-7 * |
VAN WILPE SANDRA, WOSIKA VICTORIA, CIARLONI LAURA, HOSSEINIAN EHRENSBERGER SAHAR, JEITZINER RACHEL, ANGELINO PAOLO, DUIVEMAN-DE BO: "Whole Blood Transcriptome Profiling Identifies DNA Replication and Cell Cycle Regulation as Early Marker of Response to Anti-PD-1 in Patients with Urothelial Cancer", CANCERS, M D P I AG, CH, vol. 13, no. 18, 17 September 2021 (2021-09-17), CH , pages 4660, XP055873318, ISSN: 2072-6694, DOI: 10.3390/cancers13184660 * |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2025101693A1 (en) * | 2023-11-07 | 2025-05-15 | Altos Labs, Inc. | Photomics analysis of samples |
Also Published As
Publication number | Publication date |
---|---|
AU2022381055A1 (en) | 2024-05-23 |
CA3237909A1 (en) | 2023-05-11 |
KR20240134293A (en) | 2024-09-09 |
JP2024540391A (en) | 2024-10-31 |
MX2024005612A (en) | 2024-09-23 |
EP4430214A1 (en) | 2024-09-18 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Sabari et al. | A prospective study of circulating tumor DNA to guide matched targeted therapy in lung cancers | |
Sartore-Bianchi et al. | Circulating tumor DNA to guide rechallenge with panitumumab in metastatic colorectal cancer: the phase 2 CHRONOS trial | |
Berger et al. | The emerging clinical relevance of genomics in cancer medicine | |
Hellmann et al. | Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer | |
Chang et al. | Comprehensive molecular and clinicopathologic analysis of 200 pulmonary invasive mucinous adenocarcinomas identifies distinct characteristics of molecular subtypes | |
Tan et al. | Utility of incorporating next-generation sequencing (NGS) in an Asian non-small cell lung cancer (NSCLC) population: Incremental yield of actionable alterations and cost-effectiveness analysis | |
Voss et al. | Genomically annotated risk model for advanced renal-cell carcinoma: a retrospective cohort study | |
Ou et al. | Liquid biopsy to identify actionable genomic alterations | |
Arora et al. | Genetic ancestry correlates with somatic differences in a real-world clinical cancer sequencing cohort | |
Tsoulos et al. | Tumor molecular profiling of NSCLC patients using next generation sequencing | |
Yao et al. | Prognostic value of novel immune-related genomic biomarkers identified in head and neck squamous cell carcinoma | |
Crowley et al. | Liquid biopsy: monitoring cancer-genetics in the blood | |
Nabhan et al. | Predicting prognosis in chronic lymphocytic leukemia in the contemporary era | |
Luen et al. | Association of somatic driver alterations with prognosis in postmenopausal, hormone receptor–positive, HER2-negative early breast cancer: a secondary analysis of the BIG 1-98 randomized clinical trial | |
Kim et al. | Genomic scoring to determine clinical benefit of immunotherapy by targeted sequencing | |
Tomlins et al. | Development and validation of an integrative pan-solid tumor predictor of PD-1/PD-L1 blockade benefit | |
US20220081724A1 (en) | Methods of detecting and treating subjects with checkpoint inhibitor-responsive cancer | |
Jain et al. | Impact of smoking and brain metastasis on outcomes of advanced EGFR mutation lung adenocarcinoma patients treated with first line epidermal growth factor receptor tyrosine kinase inhibitors | |
Araujo et al. | Applications of circulating tumor DNA in a cohort of phase I solid tumor patients treated with immunotherapy | |
Perdrizet et al. | Integrating comprehensive genomic sequencing of non-small cell lung cancer into a public healthcare system | |
Kansara et al. | Early circulating tumor DNA dynamics as a pan‐tumor biomarker for long‐term clinical outcome in patients treated with durvalumab and tremelimumab | |
WO2023081537A1 (en) | Cancer biomarkers for immune checkpoint inhibitors | |
Del Re et al. | Clinical utility of Next Generation Sequencing of plasma cell-free DNA for the molecular profiling of patients with NSCLC at diagnosis and disease progression | |
Rathi et al. | Clinical validation of the 50 gene AmpliSeq Cancer Panel V2 for use on a next generation sequencing platform using formalin fixed, paraffin embedded and fine needle aspiration tumour specimens | |
Shahsiah et al. | Validation of a next generation sequencing panel for detection of hotspot cancer mutations in a clinical laboratory |
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: 22890932 Country of ref document: EP Kind code of ref document: A1 |
|
ENP | Entry into the national phase |
Ref document number: 2024526946 Country of ref document: JP Kind code of ref document: A |
|
ENP | Entry into the national phase |
Ref document number: 3237909 Country of ref document: CA |
|
REG | Reference to national code |
Ref country code: BR Ref legal event code: B01A Ref document number: 112024009030 Country of ref document: BR |
|
ENP | Entry into the national phase |
Ref document number: 2022381055 Country of ref document: AU Date of ref document: 20221108 Kind code of ref document: A |
|
WWE | Wipo information: entry into national phase |
Ref document number: 1020247018785 Country of ref document: KR |
|
WWE | Wipo information: entry into national phase |
Ref document number: 202280081429.0 Country of ref document: CN |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2022890932 Country of ref document: EP |
|
ENP | Entry into the national phase |
Ref document number: 2022890932 Country of ref document: EP Effective date: 20240610 |
|
WWE | Wipo information: entry into national phase |
Ref document number: 11202403242X Country of ref document: SG |
|
ENP | Entry into the national phase |
Ref document number: 112024009030 Country of ref document: BR Kind code of ref document: A2 Effective date: 20240507 |