WO2018146128A1 - Detection of kit polymorphism for predicting the response to checkpoint blockade cancer immunotherapy - Google Patents
Detection of kit polymorphism for predicting the response to checkpoint blockade cancer immunotherapy Download PDFInfo
- Publication number
- WO2018146128A1 WO2018146128A1 PCT/EP2018/053035 EP2018053035W WO2018146128A1 WO 2018146128 A1 WO2018146128 A1 WO 2018146128A1 EP 2018053035 W EP2018053035 W EP 2018053035W WO 2018146128 A1 WO2018146128 A1 WO 2018146128A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- antibodies
- kit
- cancer immunotherapy
- checkpoint blockade
- polymorphism
- Prior art date
Links
- 238000002619 cancer immunotherapy Methods 0.000 title claims abstract description 30
- 230000005746 immune checkpoint blockade Effects 0.000 title claims abstract description 30
- 230000004044 response Effects 0.000 title claims abstract description 10
- 238000001514 detection method Methods 0.000 title description 6
- 206010028980 Neoplasm Diseases 0.000 claims abstract description 49
- 238000000034 method Methods 0.000 claims abstract description 27
- 201000011510 cancer Diseases 0.000 claims abstract description 18
- 102200076825 rs3822214 Human genes 0.000 claims abstract description 8
- 239000012472 biological sample Substances 0.000 claims abstract description 5
- 229940126547 T-cell immunoglobulin mucin-3 Drugs 0.000 claims description 13
- 102100029822 B- and T-lymphocyte attenuator Human genes 0.000 claims description 7
- 239000007787 solid Substances 0.000 claims description 4
- 101000864344 Homo sapiens B- and T-lymphocyte attenuator Proteins 0.000 claims description 2
- 238000000338 in vitro Methods 0.000 claims description 2
- 102000037982 Immune checkpoint proteins Human genes 0.000 abstract description 13
- 108091008036 Immune checkpoint proteins Proteins 0.000 abstract description 13
- 239000000523 sample Substances 0.000 abstract description 13
- 239000003795 chemical substances by application Substances 0.000 abstract description 10
- 101150068332 KIT gene Proteins 0.000 abstract description 9
- 239000002773 nucleotide Substances 0.000 abstract description 8
- 125000003729 nucleotide group Chemical group 0.000 abstract description 8
- 230000008901 benefit Effects 0.000 abstract description 5
- 230000008649 adaptation response Effects 0.000 abstract description 3
- 230000003213 activating effect Effects 0.000 abstract description 2
- 230000005809 anti-tumor immunity Effects 0.000 abstract description 2
- 238000013459 approach Methods 0.000 abstract description 2
- 230000001225 therapeutic effect Effects 0.000 abstract description 2
- 210000001266 CD8-positive T-lymphocyte Anatomy 0.000 description 15
- 108020004414 DNA Proteins 0.000 description 14
- 229940076838 Immune checkpoint inhibitor Drugs 0.000 description 14
- 101100519207 Mus musculus Pdcd1 gene Proteins 0.000 description 14
- 239000012274 immune-checkpoint protein inhibitor Substances 0.000 description 14
- 102000037984 Inhibitory immune checkpoint proteins Human genes 0.000 description 13
- 108091008026 Inhibitory immune checkpoint proteins Proteins 0.000 description 13
- 210000001519 tissue Anatomy 0.000 description 13
- 210000001744 T-lymphocyte Anatomy 0.000 description 12
- 108090000623 proteins and genes Proteins 0.000 description 12
- 229940045513 CTLA4 antagonist Drugs 0.000 description 11
- 102100034458 Hepatitis A virus cellular receptor 2 Human genes 0.000 description 11
- 101710083479 Hepatitis A virus cellular receptor 2 homolog Proteins 0.000 description 11
- 239000003112 inhibitor Substances 0.000 description 11
- 102000004169 proteins and genes Human genes 0.000 description 11
- 210000004027 cell Anatomy 0.000 description 10
- 239000003446 ligand Substances 0.000 description 10
- 102100039498 Cytotoxic T-lymphocyte protein 4 Human genes 0.000 description 9
- 102100024216 Programmed cell death 1 ligand 1 Human genes 0.000 description 9
- 150000002500 ions Chemical class 0.000 description 9
- 238000012163 sequencing technique Methods 0.000 description 9
- 102100034922 T-cell surface glycoprotein CD8 alpha chain Human genes 0.000 description 8
- 230000003321 amplification Effects 0.000 description 7
- 238000003199 nucleic acid amplification method Methods 0.000 description 7
- 108010074708 B7-H1 Antigen Proteins 0.000 description 6
- 101000666896 Homo sapiens V-type immunoglobulin domain-containing suppressor of T-cell activation Proteins 0.000 description 6
- 102100038282 V-type immunoglobulin domain-containing suppressor of T-cell activation Human genes 0.000 description 6
- 238000001574 biopsy Methods 0.000 description 6
- 238000005516 engineering process Methods 0.000 description 6
- 102000006639 indoleamine 2,3-dioxygenase Human genes 0.000 description 6
- 108020004201 indoleamine 2,3-dioxygenase Proteins 0.000 description 6
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 5
- 101710144268 B- and T-lymphocyte attenuator Proteins 0.000 description 5
- 108010021064 CTLA-4 Antigen Proteins 0.000 description 5
- 101001137987 Homo sapiens Lymphocyte activation gene 3 protein Proteins 0.000 description 5
- 108060003951 Immunoglobulin Proteins 0.000 description 5
- 102000002698 KIR Receptors Human genes 0.000 description 5
- 108010043610 KIR Receptors Proteins 0.000 description 5
- OIRDTQYFTABQOQ-KQYNXXCUSA-N adenosine group Chemical group [C@@H]1([C@H](O)[C@H](O)[C@@H](CO)O1)N1C=NC=2C(N)=NC=NC12 OIRDTQYFTABQOQ-KQYNXXCUSA-N 0.000 description 5
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 5
- IJJVMEJXYNJXOJ-UHFFFAOYSA-N fluquinconazole Chemical compound C=1C=C(Cl)C=C(Cl)C=1N1C(=O)C2=CC(F)=CC=C2N=C1N1C=NC=N1 IJJVMEJXYNJXOJ-UHFFFAOYSA-N 0.000 description 5
- 102000018358 immunoglobulin Human genes 0.000 description 5
- 229960003301 nivolumab Drugs 0.000 description 5
- 150000007523 nucleic acids Chemical class 0.000 description 5
- 102100038078 CD276 antigen Human genes 0.000 description 4
- 102000004190 Enzymes Human genes 0.000 description 4
- 108090000790 Enzymes Proteins 0.000 description 4
- 102100031351 Galectin-9 Human genes 0.000 description 4
- 101710121810 Galectin-9 Proteins 0.000 description 4
- 101000889276 Homo sapiens Cytotoxic T-lymphocyte protein 4 Proteins 0.000 description 4
- 102100020862 Lymphocyte activation gene 3 protein Human genes 0.000 description 4
- 102100024834 T-cell immunoreceptor with Ig and ITIM domains Human genes 0.000 description 4
- 102100038929 V-set domain-containing T-cell activation inhibitor 1 Human genes 0.000 description 4
- 239000000427 antigen Substances 0.000 description 4
- 108091007433 antigens Proteins 0.000 description 4
- 102000036639 antigens Human genes 0.000 description 4
- 238000009396 hybridization Methods 0.000 description 4
- 230000035772 mutation Effects 0.000 description 4
- 210000000822 natural killer cell Anatomy 0.000 description 4
- 108020004707 nucleic acids Proteins 0.000 description 4
- 102000039446 nucleic acids Human genes 0.000 description 4
- 230000037361 pathway Effects 0.000 description 4
- 229960002621 pembrolizumab Drugs 0.000 description 4
- 239000013615 primer Substances 0.000 description 4
- 102000005962 receptors Human genes 0.000 description 4
- 108020003175 receptors Proteins 0.000 description 4
- 101150051188 Adora2a gene Proteins 0.000 description 3
- 108091023037 Aptamer Proteins 0.000 description 3
- 101710185679 CD276 antigen Proteins 0.000 description 3
- 102100031788 E3 ubiquitin-protein ligase MYLIP Human genes 0.000 description 3
- 101710190174 E3 ubiquitin-protein ligase MYLIP Proteins 0.000 description 3
- 101000831007 Homo sapiens T-cell immunoreceptor with Ig and ITIM domains Proteins 0.000 description 3
- 101100407308 Mus musculus Pdcd1lg2 gene Proteins 0.000 description 3
- 108700030875 Programmed Cell Death 1 Ligand 2 Proteins 0.000 description 3
- 102100024213 Programmed cell death 1 ligand 2 Human genes 0.000 description 3
- 108010079206 V-Set Domain-Containing T-Cell Activation Inhibitor 1 Proteins 0.000 description 3
- 230000003247 decreasing effect Effects 0.000 description 3
- 230000000694 effects Effects 0.000 description 3
- 230000028993 immune response Effects 0.000 description 3
- 230000002401 inhibitory effect Effects 0.000 description 3
- 230000003993 interaction Effects 0.000 description 3
- 239000000463 material Substances 0.000 description 3
- 210000003289 regulatory T cell Anatomy 0.000 description 3
- 238000010839 reverse transcription Methods 0.000 description 3
- 239000000126 substance Substances 0.000 description 3
- IIQKYWMOMQWBER-VIFPVBQESA-N (2s)-2-amino-3-(1-benzofuran-3-yl)propanoic acid Chemical compound C1=CC=C2C(C[C@H](N)C(O)=O)=COC2=C1 IIQKYWMOMQWBER-VIFPVBQESA-N 0.000 description 2
- AWLWPSSHYJQPCH-VIFPVBQESA-N (2s)-2-amino-3-(6-nitro-1h-indol-3-yl)propanoic acid Chemical compound [O-][N+](=O)C1=CC=C2C(C[C@H](N)C(O)=O)=CNC2=C1 AWLWPSSHYJQPCH-VIFPVBQESA-N 0.000 description 2
- ZADWXFSZEAPBJS-JTQLQIEISA-N 1-methyl-L-tryptophan Chemical compound C1=CC=C2N(C)C=C(C[C@H](N)C(O)=O)C2=C1 ZADWXFSZEAPBJS-JTQLQIEISA-N 0.000 description 2
- VFTRKSBEFQDZKX-UHFFFAOYSA-N 3,3'-diindolylmethane Chemical compound C1=CC=C2C(CC=3C4=CC=CC=C4NC=3)=CNC2=C1 VFTRKSBEFQDZKX-UHFFFAOYSA-N 0.000 description 2
- 239000002126 C01EB10 - Adenosine Substances 0.000 description 2
- WZUVPPKBWHMQCE-UHFFFAOYSA-N Haematoxylin Chemical compound C12=CC(O)=C(O)C=C2CC2(O)C1C1=CC=C(O)C(O)=C1OC2 WZUVPPKBWHMQCE-UHFFFAOYSA-N 0.000 description 2
- 102100020880 Kit ligand Human genes 0.000 description 2
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 2
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 2
- 102000012220 Member 14 Tumor Necrosis Factor Receptors Human genes 0.000 description 2
- 108010061593 Member 14 Tumor Necrosis Factor Receptors Proteins 0.000 description 2
- 108091034117 Oligonucleotide Proteins 0.000 description 2
- 102100040678 Programmed cell death protein 1 Human genes 0.000 description 2
- 102100040653 Tryptophan 2,3-dioxygenase Human genes 0.000 description 2
- 101710136122 Tryptophan 2,3-dioxygenase Proteins 0.000 description 2
- 239000002253 acid Substances 0.000 description 2
- 230000004913 activation Effects 0.000 description 2
- 229960005305 adenosine Drugs 0.000 description 2
- 238000003556 assay Methods 0.000 description 2
- 230000000903 blocking effect Effects 0.000 description 2
- 239000003153 chemical reaction reagent Substances 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 150000001875 compounds Chemical class 0.000 description 2
- 230000002596 correlated effect Effects 0.000 description 2
- 230000009089 cytolysis Effects 0.000 description 2
- 238000003935 denaturing gradient gel electrophoresis Methods 0.000 description 2
- 239000012636 effector Substances 0.000 description 2
- 238000007387 excisional biopsy Methods 0.000 description 2
- 108020001507 fusion proteins Proteins 0.000 description 2
- 102000037865 fusion proteins Human genes 0.000 description 2
- 210000000987 immune system Anatomy 0.000 description 2
- 230000001506 immunosuppresive effect Effects 0.000 description 2
- 238000007386 incisional biopsy Methods 0.000 description 2
- 230000008595 infiltration Effects 0.000 description 2
- 238000001764 infiltration Methods 0.000 description 2
- 229960005386 ipilimumab Drugs 0.000 description 2
- 108020004999 messenger RNA Proteins 0.000 description 2
- 229930182817 methionine Natural products 0.000 description 2
- 125000001360 methionine group Chemical group N[C@@H](CCSC)C(=O)* 0.000 description 2
- 238000002493 microarray Methods 0.000 description 2
- BASFCYQUMIYNBI-UHFFFAOYSA-N platinum Chemical compound [Pt] BASFCYQUMIYNBI-UHFFFAOYSA-N 0.000 description 2
- ZADWXFSZEAPBJS-UHFFFAOYSA-N racemic N-methyl tryptophan Natural products C1=CC=C2N(C)C=C(CC(N)C(O)=O)C2=C1 ZADWXFSZEAPBJS-UHFFFAOYSA-N 0.000 description 2
- 230000009467 reduction Effects 0.000 description 2
- 238000007894 restriction fragment length polymorphism technique Methods 0.000 description 2
- 241000894007 species Species 0.000 description 2
- 238000006467 substitution reaction Methods 0.000 description 2
- WMBWREPUVVBILR-WIYYLYMNSA-N (-)-Epigallocatechin-3-o-gallate Chemical compound O([C@@H]1CC2=C(O)C=C(C=C2O[C@@H]1C=1C=C(O)C(O)=C(O)C=1)O)C(=O)C1=CC(O)=C(O)C(O)=C1 WMBWREPUVVBILR-WIYYLYMNSA-N 0.000 description 1
- XJRIDJAGAYGJCK-UHFFFAOYSA-N (1-acetyl-5-bromoindol-3-yl) acetate Chemical compound C1=C(Br)C=C2C(OC(=O)C)=CN(C(C)=O)C2=C1 XJRIDJAGAYGJCK-UHFFFAOYSA-N 0.000 description 1
- GAUUPDQWKHTCAX-VIFPVBQESA-N (2s)-2-amino-3-(1-benzothiophen-3-yl)propanoic acid Chemical compound C1=CC=C2C(C[C@H](N)C(O)=O)=CSC2=C1 GAUUPDQWKHTCAX-VIFPVBQESA-N 0.000 description 1
- FPJGLSZLQLNZIW-VIFPVBQESA-N (2s)-2-amino-3-(4-methyl-1h-indol-3-yl)propanoic acid Chemical compound CC1=CC=CC2=C1C(C[C@H](N)C(O)=O)=CN2 FPJGLSZLQLNZIW-VIFPVBQESA-N 0.000 description 1
- KZDNJQUJBMDHJW-VIFPVBQESA-N (2s)-2-amino-3-(5-bromo-1h-indol-3-yl)propanoic acid Chemical compound C1=C(Br)C=C2C(C[C@H](N)C(O)=O)=CNC2=C1 KZDNJQUJBMDHJW-VIFPVBQESA-N 0.000 description 1
- GDMRVYIFGPMUCG-JTQLQIEISA-N (2s)-2-azaniumyl-3-(6-methyl-1h-indol-3-yl)propanoate Chemical compound CC1=CC=C2C(C[C@H](N)C(O)=O)=CNC2=C1 GDMRVYIFGPMUCG-JTQLQIEISA-N 0.000 description 1
- UGWULZWUXSCWPX-UHFFFAOYSA-N 2-sulfanylideneimidazolidin-4-one Chemical class O=C1CNC(=S)N1 UGWULZWUXSCWPX-UHFFFAOYSA-N 0.000 description 1
- 235000010045 3,3'-diindolylmethane Nutrition 0.000 description 1
- 229940093768 3,3'-diindolylmethane Drugs 0.000 description 1
- LDCYZAJDBXYCGN-VIFPVBQESA-N 5-hydroxy-L-tryptophan Chemical compound C1=C(O)C=C2C(C[C@H](N)C(O)=O)=CNC2=C1 LDCYZAJDBXYCGN-VIFPVBQESA-N 0.000 description 1
- 229940000681 5-hydroxytryptophan Drugs 0.000 description 1
- KVNPSKDDJARYKK-JTQLQIEISA-N 5-methoxytryptophan Chemical compound COC1=CC=C2NC=C(C[C@H](N)C(O)=O)C2=C1 KVNPSKDDJARYKK-JTQLQIEISA-N 0.000 description 1
- HUNCSWANZMJLPM-UHFFFAOYSA-N 5-methyltryptophan Chemical compound CC1=CC=C2NC=C(CC(N)C(O)=O)C2=C1 HUNCSWANZMJLPM-UHFFFAOYSA-N 0.000 description 1
- XHLKOHSAWQPOFO-UHFFFAOYSA-N 5-phenyl-1h-imidazole Chemical compound N1C=NC=C1C1=CC=CC=C1 XHLKOHSAWQPOFO-UHFFFAOYSA-N 0.000 description 1
- YMEXGEAJNZRQEH-UHFFFAOYSA-N 6-Fluoro-DL-tryptophan Chemical compound FC1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 YMEXGEAJNZRQEH-UHFFFAOYSA-N 0.000 description 1
- 102000007471 Adenosine A2A receptor Human genes 0.000 description 1
- 108010085277 Adenosine A2A receptor Proteins 0.000 description 1
- 108700028369 Alleles Proteins 0.000 description 1
- 108091093088 Amplicon Proteins 0.000 description 1
- NHMBEDDKDVIBQD-UHFFFAOYSA-N Brassilexin Chemical class N1C2=CC=CC=C2C2=C1SN=C2 NHMBEDDKDVIBQD-UHFFFAOYSA-N 0.000 description 1
- 102100027138 Butyrophilin subfamily 3 member A1 Human genes 0.000 description 1
- 210000001239 CD8-positive, alpha-beta cytotoxic T lymphocyte Anatomy 0.000 description 1
- 208000001333 Colorectal Neoplasms Diseases 0.000 description 1
- 108090000695 Cytokines Proteins 0.000 description 1
- 102000004127 Cytokines Human genes 0.000 description 1
- 238000000018 DNA microarray Methods 0.000 description 1
- 239000003155 DNA primer Substances 0.000 description 1
- 241000588724 Escherichia coli Species 0.000 description 1
- WMBWREPUVVBILR-UHFFFAOYSA-N GCG Natural products C=1C(O)=C(O)C(O)=CC=1C1OC2=CC(O)=CC(O)=C2CC1OC(=O)C1=CC(O)=C(O)C(O)=C1 WMBWREPUVVBILR-UHFFFAOYSA-N 0.000 description 1
- 101000984934 Homo sapiens Butyrophilin subfamily 3 member A1 Proteins 0.000 description 1
- 101100005713 Homo sapiens CD4 gene Proteins 0.000 description 1
- 101000611936 Homo sapiens Programmed cell death protein 1 Proteins 0.000 description 1
- 101000955999 Homo sapiens V-set domain-containing T-cell activation inhibitor 1 Proteins 0.000 description 1
- IVYPNXXAYMYVSP-UHFFFAOYSA-N Indole-3-carbinol Natural products C1=CC=C2C(CO)=CNC2=C1 IVYPNXXAYMYVSP-UHFFFAOYSA-N 0.000 description 1
- 102100040061 Indoleamine 2,3-dioxygenase 1 Human genes 0.000 description 1
- 101710120843 Indoleamine 2,3-dioxygenase 1 Proteins 0.000 description 1
- 108010002386 Interleukin-3 Proteins 0.000 description 1
- QIVBCDIJIAJPQS-VIFPVBQESA-N L-tryptophane Chemical compound C1=CC=C2C(C[C@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-VIFPVBQESA-N 0.000 description 1
- 102000017578 LAG3 Human genes 0.000 description 1
- 108091054437 MHC class I family Proteins 0.000 description 1
- 102000043129 MHC class I family Human genes 0.000 description 1
- 108700018351 Major Histocompatibility Complex Proteins 0.000 description 1
- 238000000585 Mann–Whitney U test Methods 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- 108091028043 Nucleic acid sequence Proteins 0.000 description 1
- 108010038807 Oligopeptides Proteins 0.000 description 1
- 102000015636 Oligopeptides Human genes 0.000 description 1
- 108700020796 Oncogene Proteins 0.000 description 1
- 102000043276 Oncogene Human genes 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 108010079855 Peptide Aptamers Proteins 0.000 description 1
- 101710089372 Programmed cell death protein 1 Proteins 0.000 description 1
- 102000052575 Proto-Oncogene Human genes 0.000 description 1
- 108700020978 Proto-Oncogene Proteins 0.000 description 1
- VSWDORGPIHIGNW-UHFFFAOYSA-N Pyrrolidine dithiocarbamic acid Chemical compound SC(=S)N1CCCC1 VSWDORGPIHIGNW-UHFFFAOYSA-N 0.000 description 1
- 238000011529 RT qPCR Methods 0.000 description 1
- 108010039445 Stem Cell Factor Proteins 0.000 description 1
- 238000000692 Student's t-test Methods 0.000 description 1
- 230000006044 T cell activation Effects 0.000 description 1
- 230000006052 T cell proliferation Effects 0.000 description 1
- 101710090983 T-cell immunoreceptor with Ig and ITIM domains Proteins 0.000 description 1
- 102000002933 Thioredoxin Human genes 0.000 description 1
- QIVBCDIJIAJPQS-UHFFFAOYSA-N Tryptophan Natural products C1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-UHFFFAOYSA-N 0.000 description 1
- 102000044209 Tumor Suppressor Genes Human genes 0.000 description 1
- 108700025716 Tumor Suppressor Genes Proteins 0.000 description 1
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 1
- FSQKKOOTNAMONP-UHFFFAOYSA-N acemetacin Chemical compound CC1=C(CC(=O)OCC(O)=O)C2=CC(OC)=CC=C2N1C(=O)C1=CC=C(Cl)C=C1 FSQKKOOTNAMONP-UHFFFAOYSA-N 0.000 description 1
- 229960004892 acemetacin Drugs 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 230000033289 adaptive immune response Effects 0.000 description 1
- 238000007844 allele-specific PCR Methods 0.000 description 1
- 238000004458 analytical method Methods 0.000 description 1
- 230000033115 angiogenesis Effects 0.000 description 1
- 238000000137 annealing Methods 0.000 description 1
- -1 antibodies Proteins 0.000 description 1
- 210000000013 bile duct Anatomy 0.000 description 1
- 102000023732 binding proteins Human genes 0.000 description 1
- 108091008324 binding proteins Proteins 0.000 description 1
- 239000000090 biomarker Substances 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- QYKQWFZDEDFELK-UHFFFAOYSA-N brassinin Chemical class C1=CC=C2C(CNC(=S)SC)=CNC2=C1 QYKQWFZDEDFELK-UHFFFAOYSA-N 0.000 description 1
- 210000000481 breast Anatomy 0.000 description 1
- 230000001925 catabolic effect Effects 0.000 description 1
- 230000030833 cell death Effects 0.000 description 1
- 230000022534 cell killing Effects 0.000 description 1
- 210000003679 cervix uteri Anatomy 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 210000001072 colon Anatomy 0.000 description 1
- 229920000547 conjugated polymer Polymers 0.000 description 1
- OPTASPLRGRRNAP-UHFFFAOYSA-N cytosine Chemical group NC=1C=CNC(=O)N=1 OPTASPLRGRRNAP-UHFFFAOYSA-N 0.000 description 1
- 230000001472 cytotoxic effect Effects 0.000 description 1
- 238000002784 cytotoxicity assay Methods 0.000 description 1
- 231100000263 cytotoxicity test Toxicity 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 238000012137 double-staining Methods 0.000 description 1
- 229940079593 drug Drugs 0.000 description 1
- 239000003814 drug Substances 0.000 description 1
- 229940056913 eftilagimod alfa Drugs 0.000 description 1
- 210000004696 endometrium Anatomy 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- 229940030275 epigallocatechin gallate Drugs 0.000 description 1
- 210000003743 erythrocyte Anatomy 0.000 description 1
- 210000003238 esophagus Anatomy 0.000 description 1
- 210000001508 eye Anatomy 0.000 description 1
- 230000002550 fecal effect Effects 0.000 description 1
- 239000012634 fragment Substances 0.000 description 1
- 239000012520 frozen sample Substances 0.000 description 1
- 238000010353 genetic engineering Methods 0.000 description 1
- 230000007614 genetic variation Effects 0.000 description 1
- 210000004907 gland Anatomy 0.000 description 1
- 210000004209 hair Anatomy 0.000 description 1
- 210000003128 head Anatomy 0.000 description 1
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 1
- 238000004128 high performance liquid chromatography Methods 0.000 description 1
- 238000010191 image analysis Methods 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 230000036737 immune function Effects 0.000 description 1
- 108091008915 immune receptors Proteins 0.000 description 1
- 102000027596 immune receptors Human genes 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 238000003364 immunohistochemistry Methods 0.000 description 1
- 238000009169 immunotherapy Methods 0.000 description 1
- 238000000099 in vitro assay Methods 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 235000002279 indole-3-carbinol Nutrition 0.000 description 1
- RUMVKBSXRDGBGO-UHFFFAOYSA-N indole-3-carbinol Chemical compound C1=CC=C[C]2C(CO)=CN=C21 RUMVKBSXRDGBGO-UHFFFAOYSA-N 0.000 description 1
- 230000005764 inhibitory process Effects 0.000 description 1
- 230000002452 interceptive effect Effects 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 238000011005 laboratory method Methods 0.000 description 1
- 210000000265 leukocyte Anatomy 0.000 description 1
- 210000004185 liver Anatomy 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 230000003211 malignant effect Effects 0.000 description 1
- 238000004949 mass spectrometry Methods 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 201000001441 melanoma Diseases 0.000 description 1
- 230000005012 migration Effects 0.000 description 1
- 238000013508 migration Methods 0.000 description 1
- 239000000203 mixture Substances 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 210000001616 monocyte Anatomy 0.000 description 1
- 229940126619 mouse monoclonal antibody Drugs 0.000 description 1
- 231100000219 mutagenic Toxicity 0.000 description 1
- 230000003505 mutagenic effect Effects 0.000 description 1
- 210000004985 myeloid-derived suppressor cell Anatomy 0.000 description 1
- KKFHAJHLJHVUDM-UHFFFAOYSA-N n-vinylcarbazole Chemical compound C1=CC=C2N(C=C)C3=CC=CC=C3C2=C1 KKFHAJHLJHVUDM-UHFFFAOYSA-N 0.000 description 1
- 210000003739 neck Anatomy 0.000 description 1
- 238000013188 needle biopsy Methods 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 238000011275 oncology therapy Methods 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 210000001672 ovary Anatomy 0.000 description 1
- LDCYZAJDBXYCGN-UHFFFAOYSA-N oxitriptan Natural products C1=C(O)C=C2C(CC(N)C(O)=O)=CNC2=C1 LDCYZAJDBXYCGN-UHFFFAOYSA-N 0.000 description 1
- 210000000496 pancreas Anatomy 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 210000005259 peripheral blood Anatomy 0.000 description 1
- 239000011886 peripheral blood Substances 0.000 description 1
- 230000002093 peripheral effect Effects 0.000 description 1
- 230000002688 persistence Effects 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 229910052697 platinum Inorganic materials 0.000 description 1
- 238000003752 polymerase chain reaction Methods 0.000 description 1
- 239000002987 primer (paints) Substances 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 102000004196 processed proteins & peptides Human genes 0.000 description 1
- 108090000765 processed proteins & peptides Proteins 0.000 description 1
- 229940002612 prodrug Drugs 0.000 description 1
- 239000000651 prodrug Substances 0.000 description 1
- 239000000092 prognostic biomarker Substances 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 239000002096 quantum dot Substances 0.000 description 1
- 230000002285 radioactive effect Effects 0.000 description 1
- 108091008598 receptor tyrosine kinases Proteins 0.000 description 1
- 102000027426 receptor tyrosine kinases Human genes 0.000 description 1
- 210000000664 rectum Anatomy 0.000 description 1
- 230000002829 reductive effect Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 238000002271 resection Methods 0.000 description 1
- 210000003296 saliva Anatomy 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 210000000582 semen Anatomy 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- 230000019491 signal transduction Effects 0.000 description 1
- 210000003491 skin Anatomy 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 230000006641 stabilisation Effects 0.000 description 1
- 238000011105 stabilization Methods 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 210000002784 stomach Anatomy 0.000 description 1
- 230000020382 suppression by virus of host antigen processing and presentation of peptide antigen via MHC class I Effects 0.000 description 1
- 210000004243 sweat Anatomy 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 230000009897 systematic effect Effects 0.000 description 1
- 238000012353 t test Methods 0.000 description 1
- 230000008685 targeting Effects 0.000 description 1
- 210000001138 tear Anatomy 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 108060008226 thioredoxin Proteins 0.000 description 1
- 229940094937 thioredoxin Drugs 0.000 description 1
- 210000001685 thyroid gland Anatomy 0.000 description 1
- 230000000451 tissue damage Effects 0.000 description 1
- 231100000827 tissue damage Toxicity 0.000 description 1
- 239000003053 toxin Substances 0.000 description 1
- 231100000765 toxin Toxicity 0.000 description 1
- 102000027257 transmembrane receptors Human genes 0.000 description 1
- 108091008578 transmembrane receptors Proteins 0.000 description 1
- 229950007217 tremelimumab Drugs 0.000 description 1
- 229960004799 tryptophan Drugs 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 210000004981 tumor-associated macrophage Anatomy 0.000 description 1
- 238000010396 two-hybrid screening Methods 0.000 description 1
- 210000003932 urinary bladder Anatomy 0.000 description 1
- 210000002700 urine Anatomy 0.000 description 1
- 210000004291 uterus Anatomy 0.000 description 1
- AIFRHYZBTHREPW-UHFFFAOYSA-N β-carboline Chemical class N1=CC=C2C3=CC=CC=C3NC2=C1 AIFRHYZBTHREPW-UHFFFAOYSA-N 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q1/00—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
- C12Q1/68—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
- C12Q1/6876—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
- C12Q1/6883—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
- C12Q1/6886—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q2600/00—Oligonucleotides characterized by their use
- C12Q2600/106—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
- C12Q2600/156—Polymorphic or mutational markers
Definitions
- the present invention relates to a method for predicting the response of a patient to checkpoint blockade cancer immunotherapy by detecting a single nucleotide polymorphism (SNP) in the KIT gene.
- SNP single nucleotide polymorphism
- Immune checkpoints refer to a plethora of inhibitory pathways hardwired into the immune system that are crucial for maintaining self- tolerance and modulating the duration and amplitude of physiological immune responses in peripheral tissues in order to minimize collateral tissue damage.
- tumors co- opt certain immune-checkpoint pathways as a major mechanism of immune resistance, particularly against T cells that are specific for tumor antigens. Because many of the immune checkpoints are initiated by ligand-receptor interactions, they can be readily blocked by antibodies or modulated by recombinant forms of ligands or receptors.
- checkpoint blockade cancer immunotherapy vary among individuals. It is therefore necessary to define reliable predictive bio markers in an effort to better identify patients who are most likely to benefit from such a treatment.
- checkpoint blockade cancer immunotherapies aim at releasing a brake on the immune system so as to allow the immune cells to infiltrate and attack the tumor.
- this "brake release" In order for this "brake release" to be efficient, there has to be a pre-existing immune response directed against the tumor. Accordingly, in patients having low intra-tumor immune adaptive response, turning off the intra-tumor immunosuppressive response by using a checkpoint blockade cancer immunotherapy would be useless.
- the KIT gene also known as the proto-oncogene receptor tyrosine kinase, is widely known in the art: its sequence can be e.g. found under the ref NG 007456 in the NCBI gene database. It encodes the KIT protein which is a 3 transmembrane receptor for MGF (mast cell growth factor, also known as stem cell factor).
- the SNP of the kit gene according to the present invention consists in a substitution of an adenosine residue into a cytosine residue at position 1621 of the kit gene (A1621C).
- This polymorphism is accessible under the reference rs3822214 in the Single Nucleotide Polymorphism Database (dbSNP), which is a free public archive for genetic variation within and across different species developed and hosted by the National Center for Biotechnology Information (NCBI) in collaboration with the National Human Genome Research Institute (NHGRI).
- dbSNP Single Nucleotide Polymorphism Database
- NCBI National Center for Biotechnology Information
- NHGRI National Human Genome Research Institute
- the present invention relates to the detection of this particular JQT M541L polymorphism for predicting the response of a patient to a checkpoint blockade cancer immunotherapy.
- present invention relates to a checkpoint blockade cancer immunotherapy agent for use in a method for treating solid cancer in an individual, wherein said individual has been selected as not displaying the KIT polymorphism consisting of M541L (KIT M541L ).
- the detection of the KIT polymorphism according to the present invention is thus particularly suitable for discriminating responders from non-responders.
- the term "responder” refers to a patient that will achieve a response, i.e. a patient where the cancer is eradicated, reduced or stabilized.
- a non-responder or refractory patient includes patients for whom the cancer does not show reduction or stabilization after the immune checkpoint therapy.
- the present invention relates to an in vitro method for predicting the response of a patient suffering from cancer to a checkpoint blockade cancer immunotherapy, said method comprising the step consisting of detecting, in a biological sample obtained from said patient, the presence of the KIT polymorphism consisting of M541L (KIT M541L ), wherein the presence of IT M541L indicates that the patient will not respond to checkpoint blockade cancer immunotherapy.
- the present invention also relates to a method for treating a patient suffering from cancer, wherein said method comprises the steps of:
- immunocheckpoint protein is widely known in the art and refers to a molecule that is expressed by T cells and that either turns up a signal (stimulatory checkpoint molecules) or turns down a signal (inhibitory checkpoint molecules).
- Immune checkpoint constitute immune checkpoint pathways such as the CTLA-4 and PD-1 dependent pathways (see e.g. Pardoll, 2012. Nature Rev Cancer 12:252-264; Mellman et al, 2011. Nature 480:480- 489).
- inhibitory checkpoint molecules examples include A2AR, B7-H3, B7-H4, BTLA, CTLA-4, CD277, IDO, KIR, PD-1, LAG-3, TIM-3 TIGIT and VISTA.
- the Adenosine A2A receptor (A2AR) is considered as an important checkpoint in cancer therapy: the presence of adenosine in the immune microenvironment, leads to an A2a-receptor activation, and induces a negative immune feedback loop and the tumor microenvironment has relatively high concentrations of adenosine.
- B7-H3, also called CD276, was originally understood to be a co-stimulatory molecule but is now regarded as co-inhibitory.
- B7-H4 also called VTCN1
- B and T Lymphocyte Attenuator (BTLA) and also called CD272 has HVEM (Herpesvirus Entry Mediator) as its ligand.
- HVEM Herpesvirus Entry Mediator
- Surface expression of BTLA is gradually downregulated during differentiation of human CD8+ T cells from the naive to effector cell phenotype, however tumor-specific human CD8+ T cells express high levels of BTLA.
- CTLA-4 Cytotoxic T-Lymphocyte-Associated protein 4 and also called CD 152. Expression of CTLA-4 on Treg cells serves to control T cell proliferation.
- IDOl Indoleamine 2,3-dioxygenase 1
- TDO tryptophan catabolic enzyme
- Another important molecule is TDO, tryptophan 2,3-dioxygenase.
- IDOl is known to suppress T and NK cells, generate and activate Tregs and myeloid-derived suppressor cells, and promote tumour angiogenesis.
- KIR Killer-cell Immunoglobulin- like Receptor
- LAG3, Lymphocyte Activation Gene-3 works to suppress an immune response by action to Tregs as well as direct effects on CD8+ T cells.
- PD-1 Programmed Death 1 (PD-1) receptor
- PD-L1 and PD-L2 This checkpoint is the target of Merck & Co.'s melanoma drug Keytruda, which gained FDA approval in September 2014.
- An advantage of targeting PD-1 is that it can restore immune function in the tumor microenvironment.
- TIM-3 short for T-cell Immunoglobulin domain and Mucin domain 3, expresses on activated human CD4+ T cells and regulates Thl and Thl7 cytokines.
- TIM-3 acts as a negative regulator of Thl /Tel function by triggering cell death upon interaction with its ligand, galectin-9. VISTA.
- VISTA Short for V-domain Ig suppressor of T cell activation, VISTA is primarily expressed on hematopoietic cells so that consistent expression of VISTA on leukocytes within tumors may allow VISTA blockade to be effective across a broad range of solid tumors.
- TIGIT also called T cell immunoreceptor with Ig and ITIM domains
- NK Natural Killer Cells
- checkpoint blockade cancer immunotherapy agent or “immune checkpoint inhibitor” has its general meaning in the art and refers to any compound inhibiting the function of an immune inhibitory checkpoint protein. Inhibition includes reduction of function and full blockade.
- Preferred immune checkpoint inhibitors are antibodies that specifically recognize immune checkpoint proteins. A number of immune checkpoint inhibitors are known and in analogy of these known immune checkpoint protein inhibitors, alternative immune checkpoint inhibitors may be developed in the (near) future.
- the immune checkpoint inhibitors include peptides, antibodies, nucleic acid molecules and small molecules.
- CD8+ T cells has its general meaning in the art and refers to a subset of T cells which express CD8 on their surface. They are MHC class I-restricted, and function as cytotoxic T cells. "CD8+ T cells” are also called CD8+ T cells are called cytotoxic T lymphocytes (CTL), T-killer cell, cytolytic T cells, CD8+ T cells or killer T cells.
- CTL cytotoxic T lymphocytes
- T-killer cell cytolytic T cells
- CD8 antigens are members of the immunoglobulin supergene family and are associative recognition elements in major histocompatibility complex class I-restricted interactions.
- the ability of the immune checkpoint inhibitor to enhance T CD8 cell killing activity may be determined by any assay well known in the art.
- said assay is an in vitro assay wherein CD8+ T cells are brought into contact with target cells (e.g. target cells that are recognized and/or lysed by CD8+ T cells).
- the immune checkpoint inhibitor of the present invention can be selected for the ability to increase specific lysis by CD8+ T cells by more than about 20%, preferably with at least about 30%, at least about 40%, at least about 50%, or more of the specific lysis obtained at the same effector: target cell ratio with CD8+ T cells or CD8 T cell lines that are contacted by the immune checkpoint inhibitor of the present invention, Examples of protocols for classical cytotoxicity assays are conventional.
- the checkpoint blockade cancer immunotherapy agent is an agent which blocks an immunosuppressive receptor expressed by activated T lymphocytes, such as cytotoxic T lymphocyte-associated protein 4 (CTLA4) and programmed cell death 1 (PDCD1, best known as PD-1), or by NK cells, like various members of the killer cell immunoglobulin-like receptor (KIR) family, or an agent which blocks the principal ligands of these receptors, such as PD-1 ligand CD274 (best known as PD-L1 or B7-H1).
- CTL4 cytotoxic T lymphocyte-associated protein 4
- PDCD1 programmed cell death 1
- NK cells like various members of the killer cell immunoglobulin-like receptor (KIR) family, or an agent which blocks the principal ligands of these receptors, such as PD-1 ligand CD274 (best known as PD-L1 or B7-H1).
- the checkpoint blockade cancer immunotherapy agent is an antibody.
- the checkpoint blockade cancer immunotherapy agent is an antibody selected from the group consisting of anti-CTLA4 antibodies, anti-PDl antibodies, anti-PDLl antibodies, anti-PDL2 antibodies, anti-TIM-3 antibodies, anti-LAG3 antibodies, anti-IDOl antibodies, anti-TIGIT antibodies, anti-B7H3 antibodies, anti-B7H4 antibodies, anti-BTLA antibodies, and anti-B7H6 antibodies.
- anti-CTLA-4 antibodies examples include anti-CTLA-4 antibodies.
- One anti-CDLA-4 antibody is tremelimumab, (ticilimumab, CP-675,206).
- the anti-CTLA-4 antibody is ipilimumab (also known as 10D1, MDX-D010) a fully human monoclonal IgG antibody that binds to CTLA-4.
- PD-1 and PD-L1 antibodies are described in US Patent Nos. 7,488,802; 7,943,743; 8,008,449; 8,168,757; 8,217,149, and PCT Published Patent Application Nos: WO03042402, WO2008156712, WO2010089411, WO2010036959, WO2011066342, WO2011159877, WO2011082400, and WO2011161699.
- the PD-1 blockers include anti- PD-L1 antibodies.
- the PD-1 blockers include anti-PD-1 antibodies and similar binding proteins such as nivolumab (MDX 1106, BMS 936558, ONO 4538), a fully human IgG4 antibody that binds to and blocks the activation of PD-1 by its ligands PD-L1 and PD-L2; lambrolizumab (MK-3475 or SCH 900475), a humanized monoclonal IgG4 antibody against PD-1 ; CT-011 a humanized antibody that binds PD-1 ; AMP-224 is a fusion protein of B7-DC; an antibody Fc portion; BMS-936559 (MDX- 1105-01) for PD-L1 (B7-H1) blockade.
- nivolumab MDX 1106, BMS 936558, ONO 4538
- a fully human IgG4 antibody that binds to and blocks the activation of PD-1 by its ligands PD-L1 and PD-L2
- immune-checkpoint inhibitors include lymphocyte activation gene-3 (LAG-3) inhibitors, such as IMP321, a soluble Ig fusion protein (Brignone et al, 2007, J. Immunol. 179:4202-4211).
- Other immune-checkpoint inhibitors include B7 inhibitors, such as B7-H3 and B7-H4 inhibitors.
- the anti-B7-H3 antibody MGA271 (Loo et al, 2012, Clin. Cancer Res. July 15 (18) 3834).
- TIM3 T-cell immunoglobulin domain and mucin domain 3 inhibitors
- TIM-3 has its general meaning in the art and refers to T cell immunoglobulin and mucin domain-containing molecule 3.
- the natural ligand of TIM-3 is galectin 9 (Gal9).
- TIM-3 inhibitor refers to a compound, substance or composition that can inhibit the function of TIM-3.
- the inhibitor can inhibit the expression or activity of TIM-3, modulate or block the TIM-3 signaling pathway and/or block the binding of TIM-3 to galectin-9.
- Antibodies having specificity for TIM-3 are well known in the art and typically those described in WO2011155607, WO2013006490 and WO2010117057.
- the immune checkpoint inhibitor is an Indoleamine 2,3- dioxygenase (IDO) inhibitor, preferably an IDOl inhibitor. Examples of IDO inhibitors are described in WO 2014150677.
- IDO inhibitors include without limitation 1-methyl- tryptophan (IMT), ⁇ - (3-benzofuranyl)-alanine, P-(3-benzo(b)thienyl)-alanine), 6-nitro- tryptophan, 6- fluoro -tryptophan, 4-methyl-tryptophan, 5 -methyl tryptophan, 6-methyl- tryptophan, 5-methoxy-tryptophan, 5 -hydroxy-tryptophan, indole 3-carbinol, 3,3'- diindolylmethane, epigallocatechin gallate, 5-Br-4-Cl-indoxyl 1,3-diacetate, 9- vinylcarbazole, acemetacin, 5 -bromo -tryptophan, 5-bromoindoxyl diacetate, 3- Amino-naphtoic acid, pyrrolidine dithiocarbamate, 4-phenylimidazole a brassinin derivative, a thiohyl
- the IDO inhibitor is selected from 1- methyl-tryptophan, ⁇ -(3- benzofuranyl)-alanine, 6-nitro-L-tryptophan, 3-Amino-naphtoic acid and ⁇ -[3- benzo(b)thienyl] -alanine or a derivative or prodrug thereof.
- the immune checkpoint inhibitor is an anti-TIGIT (T cell immunoglobin and ITIM domain) antibody.
- the checkpoint blockade cancer immunotherapy agent is a CTLA4 blocking antibody, such as Ipilimumab, or a PD-1 blocking antibody, such as Nivolumab or Pembrolizumab, or a combination thereof.
- kit nucleic acid molecules include mRNA, genomic DNA and cDNA derived from mRNA. DNA or RNA can be single stranded or double stranded. These may be utilized for detection by amplification and/or hybridization with a probe, for instance.
- the nucleotide sequence can be obtained from a genomic DNA sample isolated from the biological sample.
- genomic DNA e.g. not pure red blood cells
- genomic DNA can be conveniently obtained from blood, semen, saliva, tears, urine, fecal material, sweat, buccal cells, skin, hair or other tissue containing nucleic acid of the individual.
- the sample contains peripheral blood monocytes.
- KIT mutations may be detected in a RNA or DNA sample, preferably after amplification.
- the isolated RNA may be subjected to coupled reverse transcription and amplification, such as reverse transcription and amplification by polymerase chain reaction (RT-PCR), using specific oligonucleotide primers that are specific for a the mutated site or that enable amplification of a the region containing the mutated site.
- RT-PCR polymerase chain reaction
- conditions for primer annealing may be chosen to ensure specific reverse transcription (where appropriate) and amplification; so that the appearance of an amplification product be a diagnostic of the presence of the particular KIT SNP according to the invention.
- RNA may be reverse-transcribed and amplified, or DNA may be amplified, after which the mutated site may be detected in the amplified sequence by hybridization with a suitable probe or by direct sequencing, or any other appropriate method known in the art.
- a cDNA obtained from RNA may be cloned and sequenced to identify the SNP according to the present invention in the KIT sequence.
- RNA sequencing e.g. direct sequencing, restriction fragment length polymorphism (RFLP) analysis; hybridization with allele-specific oligonucleotides (ASO) that are short synthetic probes which hybridize only to a perfectly matched sequence under suitably stringent hybridization conditions; allele-specific PCR; PCR using mutagenic primers; ligase-PCR, HOT cleavage; denaturing gradient gel electrophoresis (DGGE), temperature denaturing gradient gel electrophoresis (TGGE), single- stranded conformational polymorphism (SSCP) and denaturing high performance liquid chromatography (Kuklin et al, 1997).
- RFLP restriction fragment length polymorphism
- ASO allele-specific oligonucleotides
- Direct sequencing may be accomplished by any method, including without limitation chemical sequencing, using the Maxam-Gilbert method ; by enzymatic sequencing, using the Sanger method ; mass spectrometry sequencing ; sequencing using a chip-based technology; and realtime quantitative PCR.
- the KIT SNP according to the present invention results in a mutated mature KIT protein, in which the methionine in position 541 is substituted by a leucine (KIT M541L ).
- the SNP in the KIT gene may be detected at the protein level by detecting the IT M541L form of the KIT protein.
- Suitable samples for detecting the mutated KIT protein according to the present invention are cell or tissue samples.
- the sample can be a tumor sample obtained from the patient.
- the tumor sample of the patient may be obtained by biopsy or resection.
- the biopsy technique applied will depend on the tissue type to be evaluated, the size and type of the tumor, among other factors.
- Representative biopsy techniques include, but are not limited to, excisional biopsy, incisional biopsy, needle biopsy, surgical biopsy, and bone marrow biopsy.
- An "excisional biopsy” refers to the removal of an entire tumor mass with a small margin of normal tissue surrounding it.
- An “incisional biopsy” refers to the removal of a wedge of tissue that includes a cross-sectional diameter of the tumor.
- This mutated form of the KIT protein may be detected according to any appropriate method known in the art.
- a sample such as a tissue biopsy, obtained from a subject may be contacted with antibodies specific of the IT M541L form, i.e. antibodies that are capable of distinguishing between the IT M541L form and the wild-type protein (or any other protein), to determine the presence or absence of KIT M541L specified by the antibody.
- the antibodies may be monoclonal or polyclonal antibodies, single chain or double chain, chimeric antibodies, humanized antibodies, or portions of an immunoglobulin molecule, including those portions known in the art as antigen binding fragments Fab, Fab', F(ab')2 and F(v). They can also be immunoconjugated, e.g. with a toxin, or labelled antibodies.
- polyclonal antibodies may be used, monoclonal antibodies are preferred because they are more reproducible in the long run. Procedures for raising "polyclonal antibodies" are also well known. Polyclonal antibodies can be obtained from serum of an animal immunized against the appropriate antigen, which may be produced by genetic engineering for example according to standard methods well-known by one skilled in the art (see e.g. Harlow et al. (1988)).
- a “monoclonal antibody” in its various grammatical forms refers to a population of antibody molecules that contains only one species of antibody combining site capable of immunoreacting with a particular epitope.
- a monoclonal antibody thus typically displays a single binding affinity for any epitope with which it immunoreacts.
- a monoclonal antibody may therefore contain an antibody molecule having a plurality of antibody combining sites, each immunospecific for a different epitope, e.g. a bispecific monoclonal antibody.
- Laboratory methods for preparing monoclonal antibodies are well known in the art (see, for example, Harlow et al., 1988).
- Aptamers which are a class of molecule that represents an alternative to antibodies in term of molecular recognition, can also be used for detecting the IT M541L form in the context of the present invention.
- Aptamers are oligonucleotide or oligopeptide sequences with the capacity to recognize virtually any class of target molecules with high affinity and specificity.
- Such ligands may be isolated through Systematic Evolution of Ligands by Exponential enrichment (SELEX) of a random sequence library, as described in Tuerk C. and Gold L., 1990.
- the random sequence library is obtainable by combinatorial chemical synthesis of DNA. In this library, each member is a linear oligomer, eventually chemically modified, of a unique sequence.
- Peptide aptamers consists of a conformationally constrained antibody variable region displayed by a platform protein, such as E. coli Thioredoxin A that are selected from combinatorial libraries by two hybrid methods (Colas et al., 1996).
- All the probes, primers, aptamers or antibodies used in the context of the present invention may be labelled with a detectable molecule or substance, such as a fluorescent molecule, a radioactive molecule or any others labels known in the art. Labels are known in the art that generally provide (either directly or indirectly) a signal.
- the patient suffering from cancer is a mammalian, preferably a human.
- the cancer is a solid cancer.
- the cancer is a solid cancer affecting one of the following organs selected from the group consisting of uterus, endometrium, rectum, colon, cervix, esophagus, bladder, stomach, head and neck, liver, lung, bile duct, pancreas, eye, kidney, ovary, brain, breast and the thyroid gland.
- Figure 1 Graph representing the density of infiltrating CD8+ cells at the invasive margin of the tumor in KIT mutated and non-mutated patients.
- Example 1 JQX M541L j s correlated with decreased intra-tumor CD8 + cells infiltration Material, methods for KIT mutation detection
- Genomic DNA from 214 patients has been extracted from frozen tumors using QIAmp DNA mini kit (Qiagen, Courtaboeuf, France) or, if frozen samples were not available, from two 5 ⁇ thick FFPE slides using QIAmp DNA FFPE kit (Qiagen). Quantity of double strand DNA have been evaluated using qubit 2.0 fluorometer (Invitrogen, life Technologies, Saint Aubin, France) and lOng (or 20ng if FFPE) of extracted DNA were amplified using Ion AmpliSeq Cancer HotSpot Panel V2 (Ion Torrent, Life Technologies) according to manufacturer's protocol.
- hotspot regions of 50 oncogenes or tumor suppressor genes, including MET were amplified using a panel of 207 primer pairs in a 17 cycles PCR reaction (20 cycles for FFPE samples). Amplicon were then digested with FuPa Reagent and samples were separately barcoded with Ion Xpress Barcodes. IonAmpliSeq Adapters were then added to each sample. DNA banks were then purified using Agencourt AMPure XP Reagent (Beckman Coulter, Villepinte, France) and purified library obtained were amplified using Platinum PCR supermix High fidelity enzyme and purified again with Agencourt process, following the manufacturer's instructions (Ion AmpliSeq Library kit 2.0, Ion Torrent, Life Technologies).
- TMA Tissue microarray
- IM invasive margin
- CT computed tomography
- IM invasive margin
- Assessment of the invasive margin area was performed on standard paraffin sections and was based on the histomorphological variance of the tissue.
- the invasive margin was defined as a region centered on the border separating the host tissue from malignant glands, with the extend of 1 mm.
- TMA sections were incubated (60 min. at room temperature) with monoclonal antibodies against CD8 (4B11, DAKO).
- Envision+ system enzyme-conjugated polymer backbone coupled to secondary antibodies
- DAB- chromogen were applied (Dako, Glostrup, Denmark).
- Double stainings were revealed with phosphate-conjugated secondary antibodies and FastBlue-chromogen.
- tissue sections were counterstained with Harris hematoxylin (Sigma Aldrich Saint Louis, MO). Isotype-matched mouse monoclonal antibodies were used as negative controls. Slides were analyzed using an image analysis workstation (Spot Browser, Excilone, Elancourt, France). Polychromatic high-resolution spot-images (740x540 pixel, 1.181 ⁇ /pixel resolution) were obtained (x200 fold magnification). The density was recorded as the number of positive cells per unit tissue surface area.
- the t-test and the Wilcoxon-Mann- Whitney test were the parametric and non-parametric tests used to identify markers with a significantly different cell density among patient groups. P-value smaller than 0.05 was considered as significant.
- patients displaying the IT M541L mutation have a decreased number of infiltrating CD8+ cells within the invasive margin of the tumor as compared to non-mutated patients.
Landscapes
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Engineering & Computer Science (AREA)
- Immunology (AREA)
- Pathology (AREA)
- Analytical Chemistry (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Genetics & Genomics (AREA)
- Hospice & Palliative Care (AREA)
- Biochemistry (AREA)
- Microbiology (AREA)
- Molecular Biology (AREA)
- Biophysics (AREA)
- Physics & Mathematics (AREA)
- Oncology (AREA)
- Biotechnology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Peptides Or Proteins (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
Abstract
Blockade of immune checkpoints is one of the most promising approaches for activating therapeutic antitumor immunity. However, the overall benefits of checkpoint blockade cancer immunotherapy vary among individuals. The present inventors demonstrated that a specific single nucleotide polymorphism in the KIT gene is associated with a decrease in intra-tumor CD8 infiltrates. Patients presenting this polymorphism present a low intra-tumor immune adaptive response and treating these patients with a checkpoint blockade cancer immunotherapy would thus be useless. Accordingly the present invention relates to a checkpoint blockade cancer immunotherapy agent for use in a method for treating cancer in an individual who does not display the KIT polymorphism consisting of M541L (KITL541). The present invention further relates to an method for predicting the response of a patient suffering from cancer to a checkpoint blockade cancer immunotherapy by detecting the presence of the M541L KIT polymorphism in a biological sample (such as a tumor sample).
Description
DETECTION OF KIT POLYMORPHISM FOR PREDICTING THE RESPONSE TO CHECKPOINT BLOCKADE CANCER IMMUNOTHERAPY
Field of the invention The present invention relates to a method for predicting the response of a patient to checkpoint blockade cancer immunotherapy by detecting a single nucleotide polymorphism (SNP) in the KIT gene.
Background of the invention
It is well established now that the immune microenvironment of a tumor is critical for determining the likeliness of a patient to be treated with immunotherapies (see e.g. Church et al, Immunity. 2015 Oct 20;43(4):631-3). As explained by Pardoll (Nat Rev Cancer. 2012 Mar 22;12(4):252-64), among the most promising approaches for activating therapeutic antitumor immunity is the blockade of immune checkpoints. Immune checkpoints refer to a plethora of inhibitory pathways hardwired into the immune system that are crucial for maintaining self- tolerance and modulating the duration and amplitude of physiological immune responses in peripheral tissues in order to minimize collateral tissue damage. It is now clear that tumors co- opt certain immune-checkpoint pathways as a major mechanism of immune resistance, particularly against T cells that are specific for tumor antigens. Because many of the immune checkpoints are initiated by ligand-receptor interactions, they can be readily blocked by antibodies or modulated by recombinant forms of ligands or receptors.
However, the overall benefits of checkpoint blockade cancer immunotherapy vary among individuals. It is therefore necessary to define reliable predictive bio markers in an effort to better identify patients who are most likely to benefit from such a treatment.
Detailed description
The present inventors have surprisingly discovered that a specific single nucleotide polymorphism in the KIT gene is associated with a decrease in intra-tumor CD8 infiltrates. Patients presenting this polymorphism thus present a low intra-tumor immune adaptive response. As disclosed above, checkpoint blockade cancer immunotherapies aim at releasing a brake on the immune system so as to allow the immune cells to infiltrate and attack the tumor. In order for this "brake release" to be efficient, there has to be a pre-existing immune response directed
against the tumor. Accordingly, in patients having low intra-tumor immune adaptive response, turning off the intra-tumor immunosuppressive response by using a checkpoint blockade cancer immunotherapy would be useless.
Accordingly, the inventors have demonstrated that patients displaying this specific single nucleotide polymorphism in the KIT gene cannot advantageously respond to checkpoint blockade cancer immunotherapies.
The KIT gene, also known as the proto-oncogene receptor tyrosine kinase, is widely known in the art: its sequence can be e.g. found under the ref NG 007456 in the NCBI gene database. It encodes the KIT protein which is a 3 transmembrane receptor for MGF (mast cell growth factor, also known as stem cell factor). The SNP of the kit gene according to the present invention consists in a substitution of an adenosine residue into a cytosine residue at position 1621 of the kit gene (A1621C). This polymorphism is accessible under the reference rs3822214 in the Single Nucleotide Polymorphism Database (dbSNP), which is a free public archive for genetic variation within and across different species developed and hosted by the National Center for Biotechnology Information (NCBI) in collaboration with the National Human Genome Research Institute (NHGRI). This A1621C polymorphism of the KIT gene encodes for the substitution of a methionine in position 541 by a leucine in the mature KIT protein (also referred to as KITL541 or KITM541L)
In a first aspect, the present invention relates to the detection of this particular JQTM541L polymorphism for predicting the response of a patient to a checkpoint blockade cancer immunotherapy.
Particularly, present invention relates to a checkpoint blockade cancer immunotherapy agent for use in a method for treating solid cancer in an individual, wherein said individual has been selected as not displaying the KIT polymorphism consisting of M541L (KITM541L). The detection of the KIT polymorphism according to the present invention is thus particularly suitable for discriminating responders from non-responders. As used herein, the term "responder" refers to a patient that will achieve a response, i.e. a patient where the cancer is eradicated, reduced or stabilized. A non-responder or refractory patient includes patients for whom the cancer does not show reduction or stabilization after the immune checkpoint therapy. Thus, in a further embodiment, the present invention relates to an in vitro method for predicting the response of a patient suffering from cancer to a checkpoint blockade cancer immunotherapy,
said method comprising the step consisting of detecting, in a biological sample obtained from said patient, the presence of the KIT polymorphism consisting of M541L (KITM541L), wherein the presence of ITM541L indicates that the patient will not respond to checkpoint blockade cancer immunotherapy. The present invention also relates to a method for treating a patient suffering from cancer, wherein said method comprises the steps of:
-determining if the patient displays the KIT polymorphism consisting of M541L; and
-administering to said patient a checkpoint blockade cancer immunotherapy agent if said patient does not display the ITM541L polymorphism. The expression "immune checkpoint protein" is widely known in the art and refers to a molecule that is expressed by T cells and that either turns up a signal (stimulatory checkpoint molecules) or turns down a signal (inhibitory checkpoint molecules). Immune checkpoint constitute immune checkpoint pathways such as the CTLA-4 and PD-1 dependent pathways (see e.g. Pardoll, 2012. Nature Rev Cancer 12:252-264; Mellman et al, 2011. Nature 480:480- 489). Examples of inhibitory checkpoint molecules include A2AR, B7-H3, B7-H4, BTLA, CTLA-4, CD277, IDO, KIR, PD-1, LAG-3, TIM-3 TIGIT and VISTA. The Adenosine A2A receptor (A2AR) is considered as an important checkpoint in cancer therapy: the presence of adenosine in the immune microenvironment, leads to an A2a-receptor activation, and induces a negative immune feedback loop and the tumor microenvironment has relatively high concentrations of adenosine. B7-H3, also called CD276, was originally understood to be a co-stimulatory molecule but is now regarded as co-inhibitory. B7-H4, also called VTCN1, is expressed by tumor cells and tumor- associated macrophages and plays a role in tumour escape. B and T Lymphocyte Attenuator (BTLA) and also called CD272, has HVEM (Herpesvirus Entry Mediator) as its ligand. Surface expression of BTLA is gradually downregulated during differentiation of human CD8+ T cells from the naive to effector cell phenotype, however tumor-specific human CD8+ T cells express high levels of BTLA. CTLA-4, Cytotoxic T-Lymphocyte-Associated protein 4 and also called CD 152. Expression of CTLA-4 on Treg cells serves to control T cell proliferation. IDOl, Indoleamine 2,3-dioxygenase 1, is a tryptophan catabolic enzyme. A related immune-inhibitory enzymes. Another important molecule is TDO, tryptophan 2,3-dioxygenase. IDOl is known to suppress T and NK cells, generate and activate Tregs and myeloid-derived suppressor cells, and promote tumour angiogenesis. KIR, Killer-cell Immunoglobulin- like Receptor, is a receptor for MHC Class I molecules on Natural Killer cells. LAG3, Lymphocyte Activation Gene-3, works
to suppress an immune response by action to Tregs as well as direct effects on CD8+ T cells. PD-1, Programmed Death 1 (PD-1) receptor, has two ligands, PD-L1 and PD-L2. This checkpoint is the target of Merck & Co.'s melanoma drug Keytruda, which gained FDA approval in September 2014. An advantage of targeting PD-1 is that it can restore immune function in the tumor microenvironment. TIM-3, short for T-cell Immunoglobulin domain and Mucin domain 3, expresses on activated human CD4+ T cells and regulates Thl and Thl7 cytokines. TIM-3 acts as a negative regulator of Thl /Tel function by triggering cell death upon interaction with its ligand, galectin-9. VISTA. Short for V-domain Ig suppressor of T cell activation, VISTA is primarily expressed on hematopoietic cells so that consistent expression of VISTA on leukocytes within tumors may allow VISTA blockade to be effective across a broad range of solid tumors. TIGIT (also called T cell immunoreceptor with Ig and ITIM domains) is an immune receptor on some percentage of T cells and Natural Killer Cells(NK). TIGIT inhibits T cell activation in vivo.
As used herein, the expression "checkpoint blockade cancer immunotherapy agent" or "immune checkpoint inhibitor" has its general meaning in the art and refers to any compound inhibiting the function of an immune inhibitory checkpoint protein. Inhibition includes reduction of function and full blockade. Preferred immune checkpoint inhibitors are antibodies that specifically recognize immune checkpoint proteins. A number of immune checkpoint inhibitors are known and in analogy of these known immune checkpoint protein inhibitors, alternative immune checkpoint inhibitors may be developed in the (near) future. The immune checkpoint inhibitors include peptides, antibodies, nucleic acid molecules and small molecules. In particular, the immune checkpoint inhibitor of the present invention is administered for enhancing the proliferation, migration, persistence and/or cytoxic activity of CD8+ T cells in the subject and in particular the tumor-infiltrating of CD8+ T cells of the subject. As used herein "CD8+ T cells" has its general meaning in the art and refers to a subset of T cells which express CD8 on their surface. They are MHC class I-restricted, and function as cytotoxic T cells. "CD8+ T cells" are also called CD8+ T cells are called cytotoxic T lymphocytes (CTL), T-killer cell, cytolytic T cells, CD8+ T cells or killer T cells. CD8 antigens are members of the immunoglobulin supergene family and are associative recognition elements in major histocompatibility complex class I-restricted interactions. The ability of the immune checkpoint inhibitor to enhance T CD8 cell killing activity may be determined by any assay well known in the art. Typically said assay is an in vitro assay wherein CD8+ T cells are brought into contact with target cells (e.g. target cells that are recognized and/or lysed by CD8+ T cells). For example, the immune checkpoint inhibitor of the present invention can be selected for the ability to increase specific lysis by CD8+ T cells by more than about 20%, preferably with at least about 30%, at least about 40%, at
least about 50%, or more of the specific lysis obtained at the same effector: target cell ratio with CD8+ T cells or CD8 T cell lines that are contacted by the immune checkpoint inhibitor of the present invention, Examples of protocols for classical cytotoxicity assays are conventional.
Typically, the checkpoint blockade cancer immunotherapy agent is an agent which blocks an immunosuppressive receptor expressed by activated T lymphocytes, such as cytotoxic T lymphocyte-associated protein 4 (CTLA4) and programmed cell death 1 (PDCD1, best known as PD-1), or by NK cells, like various members of the killer cell immunoglobulin-like receptor (KIR) family, or an agent which blocks the principal ligands of these receptors, such as PD-1 ligand CD274 (best known as PD-L1 or B7-H1). Typically, the checkpoint blockade cancer immunotherapy agent is an antibody.
In some embodiments, the checkpoint blockade cancer immunotherapy agent is an antibody selected from the group consisting of anti-CTLA4 antibodies, anti-PDl antibodies, anti-PDLl antibodies, anti-PDL2 antibodies, anti-TIM-3 antibodies, anti-LAG3 antibodies, anti-IDOl antibodies, anti-TIGIT antibodies, anti-B7H3 antibodies, anti-B7H4 antibodies, anti-BTLA antibodies, and anti-B7H6 antibodies.
Examples of anti-CTLA-4 antibodies are described in US Patent Nos: 5,811,097; 5,811,097; 5,855,887; 6,051,227; 6,207,157; 6,682,736; 6,984,720; and 7,605,238. One anti-CDLA-4 antibody is tremelimumab, (ticilimumab, CP-675,206). In some embodiments, the anti-CTLA-4 antibody is ipilimumab (also known as 10D1, MDX-D010) a fully human monoclonal IgG antibody that binds to CTLA-4.
Examples of PD-1 and PD-L1 antibodies are described in US Patent Nos. 7,488,802; 7,943,743; 8,008,449; 8,168,757; 8,217,149, and PCT Published Patent Application Nos: WO03042402, WO2008156712, WO2010089411, WO2010036959, WO2011066342, WO2011159877, WO2011082400, and WO2011161699. In some embodiments, the PD-1 blockers include anti- PD-L1 antibodies. In certain other embodiments the PD-1 blockers include anti-PD-1 antibodies and similar binding proteins such as nivolumab (MDX 1106, BMS 936558, ONO 4538), a fully human IgG4 antibody that binds to and blocks the activation of PD-1 by its ligands PD-L1 and PD-L2; lambrolizumab (MK-3475 or SCH 900475), a humanized monoclonal IgG4 antibody against PD-1 ; CT-011 a humanized antibody that binds PD-1 ; AMP-224 is a fusion protein of B7-DC; an antibody Fc portion; BMS-936559 (MDX- 1105-01) for PD-L1 (B7-H1) blockade.
Other immune-checkpoint inhibitors include lymphocyte activation gene-3 (LAG-3) inhibitors, such as IMP321, a soluble Ig fusion protein (Brignone et al, 2007, J. Immunol. 179:4202-4211).
Other immune-checkpoint inhibitors include B7 inhibitors, such as B7-H3 and B7-H4 inhibitors. In particular, the anti-B7-H3 antibody MGA271 (Loo et al, 2012, Clin. Cancer Res. July 15 (18) 3834).
Also included are TIM3 (T-cell immunoglobulin domain and mucin domain 3) inhibitors (Fourcade et al, 2010, J. Exp. Med. 207:2175-86 and Sakuishi et al, 2010, J. Exp. Med. 207:2187-94). As used herein, the term "TIM-3" has its general meaning in the art and refers to T cell immunoglobulin and mucin domain-containing molecule 3. The natural ligand of TIM-3 is galectin 9 (Gal9). Accordingly, the term "TIM-3 inhibitor" as used herein refers to a compound, substance or composition that can inhibit the function of TIM-3. For example, the inhibitor can inhibit the expression or activity of TIM-3, modulate or block the TIM-3 signaling pathway and/or block the binding of TIM-3 to galectin-9. Antibodies having specificity for TIM-3 are well known in the art and typically those described in WO2011155607, WO2013006490 and WO2010117057. In some embodiments, the immune checkpoint inhibitor is an Indoleamine 2,3- dioxygenase (IDO) inhibitor, preferably an IDOl inhibitor. Examples of IDO inhibitors are described in WO 2014150677. Examples of IDO inhibitors include without limitation 1-methyl- tryptophan (IMT), β- (3-benzofuranyl)-alanine, P-(3-benzo(b)thienyl)-alanine), 6-nitro- tryptophan, 6- fluoro -tryptophan, 4-methyl-tryptophan, 5 -methyl tryptophan, 6-methyl- tryptophan, 5-methoxy-tryptophan, 5 -hydroxy-tryptophan, indole 3-carbinol, 3,3'- diindolylmethane, epigallocatechin gallate, 5-Br-4-Cl-indoxyl 1,3-diacetate, 9- vinylcarbazole, acemetacin, 5 -bromo -tryptophan, 5-bromoindoxyl diacetate, 3- Amino-naphtoic acid, pyrrolidine dithiocarbamate, 4-phenylimidazole a brassinin derivative, a thiohydantoin derivative, a β- carboline derivative or a brassilexin derivative. Preferably the IDO inhibitor is selected from 1- methyl-tryptophan, β-(3- benzofuranyl)-alanine, 6-nitro-L-tryptophan, 3-Amino-naphtoic acid and β-[3- benzo(b)thienyl] -alanine or a derivative or prodrug thereof. In some embodiments, the immune checkpoint inhibitor is an anti-TIGIT (T cell immunoglobin and ITIM domain) antibody.
In a preferred embodiment, the checkpoint blockade cancer immunotherapy agent is a CTLA4 blocking antibody, such as Ipilimumab, or a PD-1 blocking antibody, such as Nivolumab or Pembrolizumab, or a combination thereof.
The presence of the Single Nucleotide Polymorphism (SNP) according to the present invention may be detected by analyzing a KIT nucleic acid molecule. In the context of the invention, kit nucleic acid molecules include mRNA, genomic DNA and cDNA derived from mRNA. DNA or
RNA can be single stranded or double stranded. These may be utilized for detection by amplification and/or hybridization with a probe, for instance.
The nucleotide sequence can be obtained from a genomic DNA sample isolated from the biological sample. In this case, any biological sample containing genomic DNA (e.g. not pure red blood cells) can be used. For example, and without limitation to, genomic DNA can be conveniently obtained from blood, semen, saliva, tears, urine, fecal material, sweat, buccal cells, skin, hair or other tissue containing nucleic acid of the individual. In a particular embodiment, the sample contains peripheral blood monocytes.
KIT mutations may be detected in a RNA or DNA sample, preferably after amplification. For instance, the isolated RNA may be subjected to coupled reverse transcription and amplification, such as reverse transcription and amplification by polymerase chain reaction (RT-PCR), using specific oligonucleotide primers that are specific for a the mutated site or that enable amplification of a the region containing the mutated site. According to a first alternative, conditions for primer annealing may be chosen to ensure specific reverse transcription (where appropriate) and amplification; so that the appearance of an amplification product be a diagnostic of the presence of the particular KIT SNP according to the invention. Otherwise, RNA may be reverse-transcribed and amplified, or DNA may be amplified, after which the mutated site may be detected in the amplified sequence by hybridization with a suitable probe or by direct sequencing, or any other appropriate method known in the art. For instance, a cDNA obtained from RNA may be cloned and sequenced to identify the SNP according to the present invention in the KIT sequence.
Several methods known by the skilled person allows detecting a single nucleotide polymorphism in a nucleic acid sequence. Such methods comprise e.g. direct sequencing, restriction fragment length polymorphism (RFLP) analysis; hybridization with allele-specific oligonucleotides (ASO) that are short synthetic probes which hybridize only to a perfectly matched sequence under suitably stringent hybridization conditions; allele-specific PCR; PCR using mutagenic primers; ligase-PCR, HOT cleavage; denaturing gradient gel electrophoresis (DGGE), temperature denaturing gradient gel electrophoresis (TGGE), single- stranded conformational polymorphism (SSCP) and denaturing high performance liquid chromatography (Kuklin et al, 1997). Direct sequencing may be accomplished by any method, including without limitation chemical sequencing, using the Maxam-Gilbert method ; by enzymatic sequencing, using the Sanger method ; mass spectrometry sequencing ; sequencing using a chip-based technology; and realtime quantitative PCR.
As disclosed above, the KIT SNP according to the present invention results in a mutated mature KIT protein, in which the methionine in position 541 is substituted by a leucine (KITM541L). Thus, according to a further embodiment, the SNP in the KIT gene may be detected at the protein level by detecting the ITM541L form of the KIT protein. Suitable samples for detecting the mutated KIT protein according to the present invention are cell or tissue samples. For instance, the sample can be a tumor sample obtained from the patient. Typically the tumor sample of the patient may be obtained by biopsy or resection. The biopsy technique applied will depend on the tissue type to be evaluated, the size and type of the tumor, among other factors. Representative biopsy techniques include, but are not limited to, excisional biopsy, incisional biopsy, needle biopsy, surgical biopsy, and bone marrow biopsy. An "excisional biopsy" refers to the removal of an entire tumor mass with a small margin of normal tissue surrounding it. An "incisional biopsy" refers to the removal of a wedge of tissue that includes a cross-sectional diameter of the tumor.
This mutated form of the KIT protein may be detected according to any appropriate method known in the art. In particular a sample, such as a tissue biopsy, obtained from a subject may be contacted with antibodies specific of the ITM541L form, i.e. antibodies that are capable of distinguishing between the ITM541L form and the wild-type protein (or any other protein), to determine the presence or absence of KITM541L specified by the antibody.
The antibodies may be monoclonal or polyclonal antibodies, single chain or double chain, chimeric antibodies, humanized antibodies, or portions of an immunoglobulin molecule, including those portions known in the art as antigen binding fragments Fab, Fab', F(ab')2 and F(v). They can also be immunoconjugated, e.g. with a toxin, or labelled antibodies.
Whereas polyclonal antibodies may be used, monoclonal antibodies are preferred because they are more reproducible in the long run. Procedures for raising "polyclonal antibodies" are also well known. Polyclonal antibodies can be obtained from serum of an animal immunized against the appropriate antigen, which may be produced by genetic engineering for example according to standard methods well-known by one skilled in the art (see e.g. Harlow et al. (1988)).
A "monoclonal antibody" in its various grammatical forms refers to a population of antibody molecules that contains only one species of antibody combining site capable of immunoreacting with a particular epitope. A monoclonal antibody thus typically displays a single binding affinity for any epitope with which it immunoreacts. A monoclonal antibody may therefore contain an
antibody molecule having a plurality of antibody combining sites, each immunospecific for a different epitope, e.g. a bispecific monoclonal antibody. Laboratory methods for preparing monoclonal antibodies are well known in the art (see, for example, Harlow et al., 1988).
Aptamers, which are a class of molecule that represents an alternative to antibodies in term of molecular recognition, can also be used for detecting the ITM541L form in the context of the present invention. Aptamers are oligonucleotide or oligopeptide sequences with the capacity to recognize virtually any class of target molecules with high affinity and specificity. Such ligands may be isolated through Systematic Evolution of Ligands by Exponential enrichment (SELEX) of a random sequence library, as described in Tuerk C. and Gold L., 1990. The random sequence library is obtainable by combinatorial chemical synthesis of DNA. In this library, each member is a linear oligomer, eventually chemically modified, of a unique sequence. Possible modifications, uses and advantages of this class of molecules have been reviewed in Jayasena S.D., 1999. Peptide aptamers consists of a conformationally constrained antibody variable region displayed by a platform protein, such as E. coli Thioredoxin A that are selected from combinatorial libraries by two hybrid methods (Colas et al., 1996).
All the probes, primers, aptamers or antibodies used in the context of the present invention may be labelled with a detectable molecule or substance, such as a fluorescent molecule, a radioactive molecule or any others labels known in the art. Labels are known in the art that generally provide (either directly or indirectly) a signal. Typically, the patient suffering from cancer is a mammalian, preferably a human.
The cancer is a solid cancer.
In a preferred embodiment, the cancer is a solid cancer affecting one of the following organs selected from the group consisting of uterus, endometrium, rectum, colon, cervix, esophagus, bladder, stomach, head and neck, liver, lung, bile duct, pancreas, eye, kidney, ovary, brain, breast and the thyroid gland.
Brief Description of the figures
Figure 1: Graph representing the density of infiltrating CD8+ cells at the invasive margin of the tumor in KIT mutated and non-mutated patients.
EXAMPLES
We demonstrate that the polymorphism JQTM541L is correlated with decreased intra-tumor CD8+ cells infiltration. This weak intra-tumor adaptive immune response prevents the patients from
responding to checkpoint blockade cancer immunotherapies. Thus, these findings identify the KITM541L mutation as a biomarker for the response of a patient to checkpoint blockade cancer immunotherapy.
Example 1: JQXM541L js correlated with decreased intra-tumor CD8+ cells infiltration Material, methods for KIT mutation detection
Genomic DNA from 214 patients has been extracted from frozen tumors using QIAmp DNA mini kit (Qiagen, Courtaboeuf, France) or, if frozen samples were not available, from two 5μιη thick FFPE slides using QIAmp DNA FFPE kit (Qiagen). Quantity of double strand DNA have been evaluated using qubit 2.0 fluorometer (Invitrogen, life Technologies, Saint Aubin, France) and lOng (or 20ng if FFPE) of extracted DNA were amplified using Ion AmpliSeq Cancer HotSpot Panel V2 (Ion Torrent, Life Technologies) according to manufacturer's protocol. Briefly, "hotspot" regions of 50 oncogenes or tumor suppressor genes, including MET were amplified using a panel of 207 primer pairs in a 17 cycles PCR reaction (20 cycles for FFPE samples). Amplicon were then digested with FuPa Reagent and samples were separately barcoded with Ion Xpress Barcodes. IonAmpliSeq Adapters were then added to each sample. DNA banks were then purified using Agencourt AMPure XP Reagent (Beckman Coulter, Villepinte, France) and purified library obtained were amplified using Platinum PCR supermix High fidelity enzyme and purified again with Agencourt process, following the manufacturer's instructions (Ion AmpliSeq Library kit 2.0, Ion Torrent, Life Technologies). Quality and Quantity of each libraries have been evaluated thanks to High Sensitivity DNA chip (Agilent technologies, Courtaboeuf, France). Patients were then mixed and libraries obtained were amplified and enriched using the ion OneTouch 2 system (Ion PGM Template OT2 200, life technologies). Sequencing was performed with the Ion Torrent PGM system using Ion316 or Ion318 chip and the Ion PGM sequencing 200 kit V2 in a 520 cycles run. Runs were aligned using Variant Caller (V4.2.1.0) plugin compared to Hgl9 database, and results were analysed using Alamut 2.4.1 software (interactive biosoftware, Rouen, France).
Material, methods for immune densities: Tissue Microarray immunohistochemistry.
Tissue microarray (TMA) from the center (CT) and invasive margin (IM) of colorectal tumors (n=415) were constructed. Assessment of the invasive margin area was performed on standard paraffin sections and was based on the histomorphological variance of the tissue. The invasive margin was defined as a region centered on the border separating the host tissue from malignant glands, with the extend of 1 mm. TMA sections were incubated (60 min. at room temperature)
with monoclonal antibodies against CD8 (4B11, DAKO). Envision+ system (enzyme-conjugated polymer backbone coupled to secondary antibodies) (Dako, Glostrup, Denmark) and DAB- chromogen were applied (Dako, Glostrup, Denmark). Double stainings were revealed with phosphate-conjugated secondary antibodies and FastBlue-chromogen. For single stainings, tissue sections were counterstained with Harris hematoxylin (Sigma Aldrich Saint Louis, MO). Isotype-matched mouse monoclonal antibodies were used as negative controls. Slides were analyzed using an image analysis workstation (Spot Browser, Excilone, Elancourt, France). Polychromatic high-resolution spot-images (740x540 pixel, 1.181 μιη/pixel resolution) were obtained (x200 fold magnification). The density was recorded as the number of positive cells per unit tissue surface area.
The t-test and the Wilcoxon-Mann- Whitney test were the parametric and non-parametric tests used to identify markers with a significantly different cell density among patient groups. P-value smaller than 0.05 was considered as significant.
As shown in Figure 1, patients displaying the ITM541L mutation have a decreased number of infiltrating CD8+ cells within the invasive margin of the tumor as compared to non-mutated patients.
Claims
1. A checkpoint blockade cancer immunotherapy agent for use in a method for treating solid cancer in an individual, wherein said individual has been selected as not displaying the KIT polymorphism consisting of M541L (KITM451L).
2. A in vitro method for predicting the response of a patient suffering from cancer to a checkpoint blockade cancer immunotherapy, said method comprising the step consisting of detecting, in a biological sample obtained from said patient, the presence of the KIT polymorphism consisting of M541L (KITM541L), wherein the presence of ITM541L indicates that the patient will not respond to checkpoint blockade cancer immunotherapy.
3. The checkpoint blockade cancer immunotherapy agent for use according to claim 1 or the method according to claim 2, wherein said checkpoint blockade cancer immunotherapy agent is an antibody selected from the group consisting of anti-CTLA4 antibodies, anti-PDl antibodies, anti-PDLl antibodies, anti-PDL2 antibodies, anti-TIM-3 antibodies, anti-LAG3 antibodies, anti- IDOl antibodies, anti-TIGIT antibodies, anti-B7H3 antibodies, anti-B7H4 antibodies, anti- BTLA antibodies, and anti-B7H6 antibodies.
4. The checkpoint blockade cancer immunotherapy agent for use according to claim 1 or the method according to claim 2, wherein said checkpoint blockade cancer immunotherapy agent is an antibody selected from anti-CTLA4 antibodies and anti-PDl antibodies.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP17305137.6 | 2017-02-07 | ||
EP17305137 | 2017-02-07 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2018146128A1 true WO2018146128A1 (en) | 2018-08-16 |
Family
ID=58094348
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/EP2018/053035 WO2018146128A1 (en) | 2017-02-07 | 2018-02-07 | Detection of kit polymorphism for predicting the response to checkpoint blockade cancer immunotherapy |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2018146128A1 (en) |
Citations (26)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5811097A (en) | 1995-07-25 | 1998-09-22 | The Regents Of The University Of California | Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling |
US5855887A (en) | 1995-07-25 | 1999-01-05 | The Regents Of The University Of California | Blockade of lymphocyte down-regulation associated with CTLA-4 signaling |
US6051227A (en) | 1995-07-25 | 2000-04-18 | The Regents Of The University Of California, Office Of Technology Transfer | Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling |
US6207157B1 (en) | 1996-04-23 | 2001-03-27 | The United States Of America As Represented By The Department Of Health And Human Services | Conjugate vaccine for nontypeable Haemophilus influenzae |
WO2003042402A2 (en) | 2001-11-13 | 2003-05-22 | Dana-Farber Cancer Institute, Inc. | Agents that modulate immune cell activation and methods of use thereof |
US6682736B1 (en) | 1998-12-23 | 2004-01-27 | Abgenix, Inc. | Human monoclonal antibodies to CTLA-4 |
US6984720B1 (en) | 1999-08-24 | 2006-01-10 | Medarex, Inc. | Human CTLA-4 antibodies |
WO2008156712A1 (en) | 2007-06-18 | 2008-12-24 | N. V. Organon | Antibodies to human programmed death receptor pd-1 |
US7488802B2 (en) | 2002-12-23 | 2009-02-10 | Wyeth | Antibodies against PD-1 |
US7605238B2 (en) | 1999-08-24 | 2009-10-20 | Medarex, Inc. | Human CTLA-4 antibodies and their uses |
WO2010036959A2 (en) | 2008-09-26 | 2010-04-01 | Dana-Farber Cancer Institute | Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor |
WO2010089411A2 (en) | 2009-02-09 | 2010-08-12 | Universite De La Mediterranee | Pd-1 antibodies and pd-l1 antibodies and uses thereof |
WO2010117057A1 (en) | 2009-04-10 | 2010-10-14 | 協和発酵キリン株式会社 | Method for treatment of blood tumor using anti-tim-3 antibody |
US7943743B2 (en) | 2005-07-01 | 2011-05-17 | Medarex, Inc. | Human monoclonal antibodies to programmed death ligand 1 (PD-L1) |
WO2011066342A2 (en) | 2009-11-24 | 2011-06-03 | Amplimmune, Inc. | Simultaneous inhibition of pd-l1/pd-l2 |
WO2011082400A2 (en) | 2010-01-04 | 2011-07-07 | President And Fellows Of Harvard College | Modulators of immunoinhibitory receptor pd-1, and methods of use thereof |
US8008449B2 (en) | 2005-05-09 | 2011-08-30 | Medarex, Inc. | Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics |
WO2011155607A1 (en) | 2010-06-11 | 2011-12-15 | 協和発酵キリン株式会社 | Anti-tim-3 antibody |
WO2011159877A2 (en) | 2010-06-18 | 2011-12-22 | The Brigham And Women's Hospital, Inc. | Bi-specific antibodies against tim-3 and pd-1 for immunotherapy in chronic immune conditions |
WO2011161699A2 (en) | 2010-06-25 | 2011-12-29 | Aurigene Discovery Technologies Limited | Immunosuppression modulating compounds |
US8168757B2 (en) | 2008-03-12 | 2012-05-01 | Merck Sharp & Dohme Corp. | PD-1 binding proteins |
US8217149B2 (en) | 2008-12-09 | 2012-07-10 | Genentech, Inc. | Anti-PD-L1 antibodies, compositions and articles of manufacture |
WO2013006490A2 (en) | 2011-07-01 | 2013-01-10 | Cellerant Therapeutics, Inc. | Antibodies that specifically bind to tim3 |
WO2014150677A1 (en) | 2013-03-15 | 2014-09-25 | Bristol-Myers Squibb Company | Inhibitors of indoleamine 2,3-dioxygenase (ido) |
WO2015116868A2 (en) * | 2014-01-29 | 2015-08-06 | Caris Mpi, Inc. | Molecular profiling of immune modulators |
US20160326597A1 (en) * | 2014-01-02 | 2016-11-10 | Memorial Sloan Kettering Cancer Center | Determinants of cancer response to immunotherapy |
-
2018
- 2018-02-07 WO PCT/EP2018/053035 patent/WO2018146128A1/en active Application Filing
Patent Citations (26)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5855887A (en) | 1995-07-25 | 1999-01-05 | The Regents Of The University Of California | Blockade of lymphocyte down-regulation associated with CTLA-4 signaling |
US6051227A (en) | 1995-07-25 | 2000-04-18 | The Regents Of The University Of California, Office Of Technology Transfer | Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling |
US5811097A (en) | 1995-07-25 | 1998-09-22 | The Regents Of The University Of California | Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling |
US6207157B1 (en) | 1996-04-23 | 2001-03-27 | The United States Of America As Represented By The Department Of Health And Human Services | Conjugate vaccine for nontypeable Haemophilus influenzae |
US6682736B1 (en) | 1998-12-23 | 2004-01-27 | Abgenix, Inc. | Human monoclonal antibodies to CTLA-4 |
US7605238B2 (en) | 1999-08-24 | 2009-10-20 | Medarex, Inc. | Human CTLA-4 antibodies and their uses |
US6984720B1 (en) | 1999-08-24 | 2006-01-10 | Medarex, Inc. | Human CTLA-4 antibodies |
WO2003042402A2 (en) | 2001-11-13 | 2003-05-22 | Dana-Farber Cancer Institute, Inc. | Agents that modulate immune cell activation and methods of use thereof |
US7488802B2 (en) | 2002-12-23 | 2009-02-10 | Wyeth | Antibodies against PD-1 |
US8008449B2 (en) | 2005-05-09 | 2011-08-30 | Medarex, Inc. | Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics |
US7943743B2 (en) | 2005-07-01 | 2011-05-17 | Medarex, Inc. | Human monoclonal antibodies to programmed death ligand 1 (PD-L1) |
WO2008156712A1 (en) | 2007-06-18 | 2008-12-24 | N. V. Organon | Antibodies to human programmed death receptor pd-1 |
US8168757B2 (en) | 2008-03-12 | 2012-05-01 | Merck Sharp & Dohme Corp. | PD-1 binding proteins |
WO2010036959A2 (en) | 2008-09-26 | 2010-04-01 | Dana-Farber Cancer Institute | Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor |
US8217149B2 (en) | 2008-12-09 | 2012-07-10 | Genentech, Inc. | Anti-PD-L1 antibodies, compositions and articles of manufacture |
WO2010089411A2 (en) | 2009-02-09 | 2010-08-12 | Universite De La Mediterranee | Pd-1 antibodies and pd-l1 antibodies and uses thereof |
WO2010117057A1 (en) | 2009-04-10 | 2010-10-14 | 協和発酵キリン株式会社 | Method for treatment of blood tumor using anti-tim-3 antibody |
WO2011066342A2 (en) | 2009-11-24 | 2011-06-03 | Amplimmune, Inc. | Simultaneous inhibition of pd-l1/pd-l2 |
WO2011082400A2 (en) | 2010-01-04 | 2011-07-07 | President And Fellows Of Harvard College | Modulators of immunoinhibitory receptor pd-1, and methods of use thereof |
WO2011155607A1 (en) | 2010-06-11 | 2011-12-15 | 協和発酵キリン株式会社 | Anti-tim-3 antibody |
WO2011159877A2 (en) | 2010-06-18 | 2011-12-22 | The Brigham And Women's Hospital, Inc. | Bi-specific antibodies against tim-3 and pd-1 for immunotherapy in chronic immune conditions |
WO2011161699A2 (en) | 2010-06-25 | 2011-12-29 | Aurigene Discovery Technologies Limited | Immunosuppression modulating compounds |
WO2013006490A2 (en) | 2011-07-01 | 2013-01-10 | Cellerant Therapeutics, Inc. | Antibodies that specifically bind to tim3 |
WO2014150677A1 (en) | 2013-03-15 | 2014-09-25 | Bristol-Myers Squibb Company | Inhibitors of indoleamine 2,3-dioxygenase (ido) |
US20160326597A1 (en) * | 2014-01-02 | 2016-11-10 | Memorial Sloan Kettering Cancer Center | Determinants of cancer response to immunotherapy |
WO2015116868A2 (en) * | 2014-01-29 | 2015-08-06 | Caris Mpi, Inc. | Molecular profiling of immune modulators |
Non-Patent Citations (13)
Title |
---|
A. GABRIELSON ET AL: "Intratumoral CD3 and CD8 T-cell Densities Associated with Relapse-Free Survival in HCC", CANCER IMMUNOLOGY RESEARCH, vol. 4, no. 5, 11 March 2016 (2016-03-11), US, pages 419 - 430, XP055387888, ISSN: 2326-6066, DOI: 10.1158/2326-6066.CIR-15-0110 * |
ALESSANDRA IURLO ET AL: "Identification of kit<sup>M541L</sup> somatic mutation in chronic eosinophilic leukemia, not otherwise specified and its implication in low-dose imatinib response", ONCOTARGET, vol. 5, no. 13, 1 May 2014 (2014-05-01), pages 4665 - 4670, XP055387883, DOI: 10.18632/oncotarget.1941 * |
ARMELLE DUFRESNE ET AL: "Impact of KIT exon 10 M541L allelic variant on the response to imatinib in aggressive fibromatosis: analysis of the desminib series by competitive allele specific Taqman PCR technology", BMC CANCER, BIOMED CENTRAL, LONDON, GB, vol. 14, no. 1, 29 August 2014 (2014-08-29), pages 632, XP021195710, ISSN: 1471-2407, DOI: 10.1186/1471-2407-14-632 * |
BRIGNONE ET AL., J. IMMUNOL., vol. 179, 2007, pages 4202 - 4211 |
CHURCH ET AL., IMMUNITY, vol. 43, no. 4, 20 October 2015 (2015-10-20), pages 631 - 3 |
DREW M. PARDOLL: "The blockade of immune checkpoints in cancer immunotherapy", NATURE REVIEWS. CANCER, vol. 12, no. 4, 22 March 2012 (2012-03-22), GB, pages 252 - 264, XP055339916, ISSN: 1474-175X, DOI: 10.1038/nrc3239 * |
FOURCADE ET AL., J. EXP. MED., vol. 207, 2010, pages 2175 - 86 |
LOO ET AL., CLIN. CANCER RES., vol. 15, no. 18, 2012, pages 3834 |
MEHDI BRAHMI ET AL: "KIT exon 10 variant (c.1621 A?>?C) single nucleotide polymorphism as predictor of GIST patient outcome", BMC CANCER, BIOMED CENTRAL, LONDON, GB, vol. 15, no. 1, 24 October 2015 (2015-10-24), pages 780, XP021230882, ISSN: 1471-2407, DOI: 10.1186/S12885-015-1817-5 * |
MELLMAN ET AL., NATURE, vol. 480, 2011, pages 480 - 489 |
PARDOLL, NAT REV CANCER, vol. 12, no. 4, 22 March 2012 (2012-03-22), pages 252 - 64 |
PARDOLL, NATURE REV CANCER, vol. 12, 2012, pages 252 - 264 |
SAKUISHI ET AL., J. EXP. MED., vol. 207, 2010, pages 2187 - 94 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Kline et al. | The immune landscape and response to immune checkpoint blockade therapy in lymphoma | |
Kalaora et al. | Mechanisms of immune activation and regulation: lessons from melanoma | |
Wu et al. | Intratumor heterogeneity: the hidden barrier to immunotherapy against MSI tumors from the perspective of IFN-γ signaling and tumor-infiltrating lymphocytes | |
RU2739036C2 (en) | Method | |
Mino-Kenudson | Programmed cell death ligand-1 (PD-L1) expression by immunohistochemistry: could it be predictive and/or prognostic in non-small cell lung cancer? | |
Dolcetti et al. | High prevalence of activated intraepithelial cytotoxic T lymphocytes and increased neoplastic cell apoptosis in colorectal carcinomas with microsatellite instability | |
CN113453705A (en) | Methods of identifying activating antigen receptor (aCAR)/Inhibitory Chimeric Antigen Receptor (iCAR) pairs for cancer therapy | |
Queirolo et al. | Association of CTLA-4 polymorphisms with improved overall survival in melanoma patients treated with CTLA-4 blockade: a pilot study | |
JP2019509282A (en) | How to treat cancer | |
US20090317816A1 (en) | Methods for identifying risk of breast cancer and treatments thereof | |
Warrick et al. | FOXA1 repression drives lineage plasticity and immune heterogeneity in bladder cancers with squamous differentiation | |
EP2688907B1 (en) | Dominant negative hsp110 mutant and its use in prognosing and treating cancers | |
KR20100015883A (en) | Method of diagnosing, classifying and treating endometrial cancer and precancer | |
WO2016044207A1 (en) | Biomarkers useful for determining response to pd-1 blockade therapy | |
US20130078244A1 (en) | Methods for detecting and regulating alopecia areata and gene cohorts thereof | |
WO2021150925A1 (en) | Uses of biomarkers for improving immunotherapy | |
Zappasodi et al. | Strategies for predicting response to checkpoint inhibitors | |
JP2021527082A (en) | New immune checkpoint inhibitor | |
TW201713359A (en) | Markers for identifying patients responsive to anti-PD-L1 and/or anti-CTLA4 antibody therapy | |
CN109963572A (en) | For characterizing the reactive composition and method of solid tumor confrontation PD-L1 antibody monotherapy | |
WO2017103086A1 (en) | A method for predicting the response to checkpoint blockade cancer immunotherapy | |
Gentile et al. | p53 and survival in early onset breast cancer: analysis of gene mutations, loss of heterozygosity and protein accumulation | |
Rodriguez et al. | Antitumor Activity of a Novel LAIR1 Antagonist in Combination with Anti-PD1 to Treat Collagen-Rich Solid Tumors | |
US11852631B2 (en) | Biomarkers predictive of anti-immune checkpoint response | |
JP2014020930A (en) | Pancreatic cancer diagnosis and biomarker for treatment effect prediction determination |
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: 18703594 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 18703594 Country of ref document: EP Kind code of ref document: A1 |