NZ737717B2 - Method for inducing early t memory response with short peptides anti-tumor vaccine - Google Patents
Method for inducing early t memory response with short peptides anti-tumor vaccine Download PDFInfo
- Publication number
- NZ737717B2 NZ737717B2 NZ737717A NZ73771715A NZ737717B2 NZ 737717 B2 NZ737717 B2 NZ 737717B2 NZ 737717 A NZ737717 A NZ 737717A NZ 73771715 A NZ73771715 A NZ 73771715A NZ 737717 B2 NZ737717 B2 NZ 737717B2
- Authority
- NZ
- New Zealand
- Prior art keywords
- cancer
- use according
- seq
- peptide
- therapeutic peptide
- Prior art date
Links
- 230000004044 response Effects 0.000 title claims abstract description 104
- 108090000765 processed proteins & peptides Proteins 0.000 title claims description 36
- 102000004196 processed proteins & peptides Human genes 0.000 title claims description 36
- 230000001939 inductive effect Effects 0.000 title abstract description 15
- 229960005486 vaccines Drugs 0.000 title description 42
- 230000015654 memory Effects 0.000 title description 36
- 230000000259 anti-tumor Effects 0.000 title description 3
- 210000003071 memory T lymphocyte Anatomy 0.000 claims abstract description 68
- 201000011510 cancer Diseases 0.000 claims abstract description 65
- 230000001225 therapeutic Effects 0.000 claims abstract description 61
- 108010071384 Peptide T Proteins 0.000 claims abstract description 60
- IWHCAJPPWOMXNW-LYKMMFCUSA-N Peptide T Chemical compound C[C@H](N)C(=O)N[C@@H](CO)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@H](C(=O)N[C@@H]([C@H](O)C)C(O)=O)CC1=CC=C(O)C=C1 IWHCAJPPWOMXNW-LYKMMFCUSA-N 0.000 claims abstract description 60
- 238000002560 therapeutic procedure Methods 0.000 claims abstract description 41
- 230000035492 administration Effects 0.000 claims abstract description 32
- 239000000427 antigen Substances 0.000 claims abstract description 24
- 102000038129 antigens Human genes 0.000 claims abstract description 24
- 108091007172 antigens Proteins 0.000 claims abstract description 24
- 210000000265 Leukocytes Anatomy 0.000 claims abstract description 8
- 210000004027 cells Anatomy 0.000 claims description 53
- 108009000071 Non-small cell lung cancer Proteins 0.000 claims description 40
- 208000002154 Non-Small-Cell Lung Carcinoma Diseases 0.000 claims description 38
- 239000000203 mixture Substances 0.000 claims description 35
- 238000009169 immunotherapy Methods 0.000 claims description 32
- 210000003283 T-Lymphocytes, Helper-Inducer Anatomy 0.000 claims description 25
- 210000002443 helper T lymphocyte Anatomy 0.000 claims description 23
- 206010028980 Neoplasm Diseases 0.000 claims description 19
- 239000007924 injection Substances 0.000 claims description 17
- 206010009944 Colon cancer Diseases 0.000 claims description 16
- 239000002671 adjuvant Substances 0.000 claims description 16
- 230000000240 adjuvant Effects 0.000 claims description 16
- 206010026673 Malignant pleural effusion Diseases 0.000 claims description 13
- 238000002347 injection Methods 0.000 claims description 8
- 206010006187 Breast cancer Diseases 0.000 claims description 7
- 239000012274 immune-checkpoint protein inhibitor Substances 0.000 claims description 7
- 229940121650 immune-checkpoint protein inhibitors Drugs 0.000 claims description 7
- QNAYBMKLOCPYGJ-UWTATZPHSA-N D-alanine Chemical compound C[C@@H](N)C(O)=O QNAYBMKLOCPYGJ-UWTATZPHSA-N 0.000 claims description 6
- 206010033128 Ovarian cancer Diseases 0.000 claims description 6
- 239000003814 drug Substances 0.000 claims description 6
- BVAUMRCGVHUWOZ-ZETCQYMHSA-N (2S)-2-(cyclohexylazaniumyl)propanoate Chemical compound OC(=O)[C@H](C)NC1CCCCC1 BVAUMRCGVHUWOZ-ZETCQYMHSA-N 0.000 claims description 5
- 206010058467 Lung neoplasm malignant Diseases 0.000 claims description 5
- 206010025323 Lymphomas Diseases 0.000 claims description 5
- 239000002246 antineoplastic agent Substances 0.000 claims description 5
- 201000005202 lung cancer Diseases 0.000 claims description 5
- 239000012275 CTLA-4 inhibitor Substances 0.000 claims description 4
- 229940121652 CTLA-4 inhibitors Drugs 0.000 claims description 4
- 210000004072 Lung Anatomy 0.000 claims description 4
- 206010025650 Malignant melanoma Diseases 0.000 claims description 4
- 206010059282 Metastases to central nervous system Diseases 0.000 claims description 4
- 206010027476 Metastasis Diseases 0.000 claims description 4
- 108010019706 Nivolumab Proteins 0.000 claims description 4
- 206010025310 Other lymphomas Diseases 0.000 claims description 4
- 201000001441 melanoma Diseases 0.000 claims description 4
- 229960003301 nivolumab Drugs 0.000 claims description 4
- 238000001959 radiotherapy Methods 0.000 claims description 4
- FUOOLUPWFVMBKG-UHFFFAOYSA-N α-aminoisobutanoic acid Chemical compound CC(C)(N)C(O)=O FUOOLUPWFVMBKG-UHFFFAOYSA-N 0.000 claims description 4
- 229950002916 Avelumab Drugs 0.000 claims description 3
- 108010090685 BMS-936559 Proteins 0.000 claims description 3
- 206010005003 Bladder cancer Diseases 0.000 claims description 3
- 206010008342 Cervix carcinoma Diseases 0.000 claims description 3
- 206010024324 Leukaemias Diseases 0.000 claims description 3
- 229950010773 Pidilizumab Drugs 0.000 claims description 3
- 206010038389 Renal cancer Diseases 0.000 claims description 3
- 206010039491 Sarcoma Diseases 0.000 claims description 3
- 206010057644 Testis cancer Diseases 0.000 claims description 3
- 206010046766 Uterine cancer Diseases 0.000 claims description 3
- 108010010826 avelumab Proteins 0.000 claims description 3
- 230000002496 gastric Effects 0.000 claims description 3
- 210000004602 germ cell Anatomy 0.000 claims description 3
- 201000010982 kidney cancer Diseases 0.000 claims description 3
- 238000004519 manufacturing process Methods 0.000 claims description 3
- 230000002611 ovarian Effects 0.000 claims description 3
- 108010051812 pidilizumab Proteins 0.000 claims description 3
- 201000003120 testicular cancer Diseases 0.000 claims description 3
- 201000005112 urinary bladder cancer Diseases 0.000 claims description 3
- 210000004556 Brain Anatomy 0.000 claims description 2
- 102100005310 CTLA4 Human genes 0.000 claims description 2
- 101700054183 CTLA4 Proteins 0.000 claims description 2
- 210000003679 Cervix Uteri Anatomy 0.000 claims description 2
- 201000011231 colorectal cancer Diseases 0.000 claims description 2
- 201000010536 head and neck cancer Diseases 0.000 claims description 2
- 108010026276 pembrolizumab Proteins 0.000 claims description 2
- 229960002621 pembrolizumab Drugs 0.000 claims description 2
- 238000007920 subcutaneous administration Methods 0.000 claims description 2
- 235000003197 Byrsonima crassifolia Nutrition 0.000 claims 1
- 240000001546 Byrsonima crassifolia Species 0.000 claims 1
- 102100007290 CD274 Human genes 0.000 claims 1
- 101710012053 CD274 Proteins 0.000 claims 1
- 239000012269 PD-1/PD-L1 inhibitor Substances 0.000 claims 1
- 229940121653 PD-1/PD-L1 inhibitors Drugs 0.000 claims 1
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 48
- 210000000400 T-Lymphocytes, Cytotoxic Anatomy 0.000 description 46
- 239000012071 phase Substances 0.000 description 22
- 238000003114 enzyme-linked immunosorbent spot assay Methods 0.000 description 20
- 210000003819 Peripheral blood mononuclear cell Anatomy 0.000 description 18
- 230000028993 immune response Effects 0.000 description 15
- 238000002619 cancer immunotherapy Methods 0.000 description 14
- 230000004083 survival Effects 0.000 description 14
- 238000004166 bioassay Methods 0.000 description 13
- 201000010099 disease Diseases 0.000 description 12
- 238000000338 in vitro Methods 0.000 description 11
- 210000001744 T-Lymphocytes Anatomy 0.000 description 10
- 101710038532 rpl44e Proteins 0.000 description 10
- 208000002151 Pleural Effusion Diseases 0.000 description 8
- 239000003112 inhibitor Substances 0.000 description 8
- 230000002401 inhibitory effect Effects 0.000 description 8
- 230000000638 stimulation Effects 0.000 description 8
- 230000000875 corresponding Effects 0.000 description 7
- 150000002500 ions Chemical class 0.000 description 7
- 238000002203 pretreatment Methods 0.000 description 7
- 238000002512 chemotherapy Methods 0.000 description 6
- 230000002163 immunogen Effects 0.000 description 6
- 101700025368 ERBB2 Proteins 0.000 description 5
- 101700081293 LMX1A Proteins 0.000 description 5
- 102100019764 PDCD1 Human genes 0.000 description 5
- 102100019730 TP53 Human genes 0.000 description 5
- 101710026335 TP53 Proteins 0.000 description 5
- 238000009566 cancer vaccine Methods 0.000 description 5
- 230000000694 effects Effects 0.000 description 5
- 239000002480 mineral oil Substances 0.000 description 5
- 235000010446 mineral oil Nutrition 0.000 description 5
- 239000008194 pharmaceutical composition Substances 0.000 description 5
- 238000002255 vaccination Methods 0.000 description 5
- 229960004562 Carboplatin Drugs 0.000 description 4
- OLESAACUTLOWQZ-UHFFFAOYSA-L Carboplatin Chemical compound O=C1O[Pt]([N]([H])([H])[H])([N]([H])([H])[H])OC(=O)C11CCC1 OLESAACUTLOWQZ-UHFFFAOYSA-L 0.000 description 4
- 210000001072 Colon Anatomy 0.000 description 4
- 229960001433 Erlotinib Drugs 0.000 description 4
- AAKJLRGGTJKAMG-UHFFFAOYSA-N Erlotinib Chemical compound C=12C=C(OCCOC)C(OCCOC)=CC2=NC=NC=1NC1=CC=CC(C#C)=C1 AAKJLRGGTJKAMG-UHFFFAOYSA-N 0.000 description 4
- 239000005551 L01XE03 - Erlotinib Substances 0.000 description 4
- 206010061289 Metastatic neoplasm Diseases 0.000 description 4
- 229940035295 Ting Drugs 0.000 description 4
- 210000000038 chest Anatomy 0.000 description 4
- 230000018109 developmental process Effects 0.000 description 4
- 210000003162 effector T lymphocyte Anatomy 0.000 description 4
- 230000001394 metastastic Effects 0.000 description 4
- 229960005079 pemetrexed Drugs 0.000 description 4
- WBXPDJSOTKVWSJ-ZDUSSCGKSA-N pemetrexed Chemical compound C=1NC=2NC(N)=NC(=O)C=2C=1CCC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 WBXPDJSOTKVWSJ-ZDUSSCGKSA-N 0.000 description 4
- 239000000825 pharmaceutical preparation Substances 0.000 description 4
- 231100000486 side effect Toxicity 0.000 description 4
- 230000020382 suppression by virus of host antigen processing and presentation of peptide antigen via MHC class I Effects 0.000 description 4
- 210000000481 Breast Anatomy 0.000 description 3
- 102100016662 ERBB2 Human genes 0.000 description 3
- RCINICONZNJXQF-MZXODVADSA-N Intaxel Chemical compound O([C@@H]1[C@@]2(C[C@@H](C(C)=C(C2(C)C)[C@H](C([C@]2(C)[C@@H](O)C[C@H]3OC[C@]3([C@H]21)OC(C)=O)=O)OC(=O)C)OC(=O)[C@H](O)[C@@H](NC(=O)C=1C=CC=CC=1)C=1C=CC=CC=1)O)C(=O)C1=CC=CC=C1 RCINICONZNJXQF-MZXODVADSA-N 0.000 description 3
- 102100003019 MAGEA2 Human genes 0.000 description 3
- 101710027663 MAGEA2B Proteins 0.000 description 3
- 102100003017 MAGEA3 Human genes 0.000 description 3
- 101710027664 MAGEA3 Proteins 0.000 description 3
- 206010027406 Mesothelioma Diseases 0.000 description 3
- 229960001592 Paclitaxel Drugs 0.000 description 3
- 238000004458 analytical method Methods 0.000 description 3
- 108090001123 antibodies Proteins 0.000 description 3
- 102000004965 antibodies Human genes 0.000 description 3
- 229940079593 drugs Drugs 0.000 description 3
- 230000004904 long-term response Effects 0.000 description 3
- 201000004792 malaria Diseases 0.000 description 3
- 238000005259 measurement Methods 0.000 description 3
- 239000002609 media Substances 0.000 description 3
- 238000000034 method Methods 0.000 description 3
- 238000009021 pre-vaccination Methods 0.000 description 3
- 230000001681 protective Effects 0.000 description 3
- 229930003347 taxol Natural products 0.000 description 3
- UCSJYZPVAKXKNQ-HZYVHMACSA-N 1-[(1S,2R,3R,4S,5R,6R)-3-carbamimidamido-6-{[(2R,3R,4R,5S)-3-{[(2S,3S,4S,5R,6S)-4,5-dihydroxy-6-(hydroxymethyl)-3-(methylamino)oxan-2-yl]oxy}-4-formyl-4-hydroxy-5-methyloxolan-2-yl]oxy}-2,4,5-trihydroxycyclohexyl]guanidine Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- GHASVSINZRGABV-UHFFFAOYSA-N 5-flurouricil Chemical compound FC1=CNC(=O)NC1=O GHASVSINZRGABV-UHFFFAOYSA-N 0.000 description 2
- 210000004369 Blood Anatomy 0.000 description 2
- 210000001266 CD8-Positive T-Lymphocytes Anatomy 0.000 description 2
- -1 CEA Proteins 0.000 description 2
- UJOBWOGCFQCDNV-UHFFFAOYSA-N Carbazole Chemical compound C1=CC=C2C3=CC=CC=C3NC2=C1 UJOBWOGCFQCDNV-UHFFFAOYSA-N 0.000 description 2
- ZDZOTLJHXYCWBA-VCVYQWHSSA-N Docetaxel Chemical compound O([C@H]1[C@H]2[C@@](C([C@H](O)C3=C(C)[C@@H](OC(=O)[C@H](O)[C@@H](NC(=O)OC(C)(C)C)C=4C=CC=CC=4)C[C@]1(O)C3(C)C)=O)(C)[C@@H](O)C[C@H]1OC[C@]12OC(=O)C)C(=O)C1=CC=CC=C1 ZDZOTLJHXYCWBA-VCVYQWHSSA-N 0.000 description 2
- 102000027760 ERBB2 Human genes 0.000 description 2
- 206010063045 Effusion Diseases 0.000 description 2
- 229960002949 Fluorouracil Drugs 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 210000000987 Immune System Anatomy 0.000 description 2
- 108060003951 Immunoglobulins Proteins 0.000 description 2
- 102000018358 Immunoglobulins Human genes 0.000 description 2
- 241001446467 Mama Species 0.000 description 2
- 206010061309 Neoplasm progression Diseases 0.000 description 2
- 210000002966 Serum Anatomy 0.000 description 2
- 208000000587 Small Cell Lung Carcinoma Diseases 0.000 description 2
- 206010041067 Small cell lung cancer Diseases 0.000 description 2
- 238000000692 Student's t-test Methods 0.000 description 2
- 229940120982 Tarceva Drugs 0.000 description 2
- 239000008280 blood Substances 0.000 description 2
- 201000005216 brain cancer Diseases 0.000 description 2
- 239000003795 chemical substances by application Substances 0.000 description 2
- 229960004316 cisplatin Drugs 0.000 description 2
- 230000002596 correlated Effects 0.000 description 2
- 230000001472 cytotoxic Effects 0.000 description 2
- 231100000433 cytotoxic Toxicity 0.000 description 2
- 230000034994 death Effects 0.000 description 2
- 231100000517 death Toxicity 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- 229960003668 docetaxel Drugs 0.000 description 2
- 239000003937 drug carrier Substances 0.000 description 2
- 239000001963 growth media Substances 0.000 description 2
- 230000036541 health Effects 0.000 description 2
- 238000010191 image analysis Methods 0.000 description 2
- 230000036039 immunity Effects 0.000 description 2
- 230000000670 limiting Effects 0.000 description 2
- 230000007787 long-term memory Effects 0.000 description 2
- 230000036210 malignancy Effects 0.000 description 2
- 230000001575 pathological Effects 0.000 description 2
- 238000010837 poor prognosis Methods 0.000 description 2
- 230000002062 proliferating Effects 0.000 description 2
- 102000004169 proteins and genes Human genes 0.000 description 2
- 108090000623 proteins and genes Proteins 0.000 description 2
- 108020003175 receptors Proteins 0.000 description 2
- 102000005962 receptors Human genes 0.000 description 2
- 239000007929 subcutaneous injection Substances 0.000 description 2
- 231100000419 toxicity Toxicity 0.000 description 2
- 230000001988 toxicity Effects 0.000 description 2
- LXZZYRPGZAFOLE-UHFFFAOYSA-L transplatin Chemical compound [H][N]([H])([H])[Pt](Cl)(Cl)[N]([H])([H])[H] LXZZYRPGZAFOLE-UHFFFAOYSA-L 0.000 description 2
- 229960002066 vinorelbine Drugs 0.000 description 2
- NRUKOCRGYNPUPR-QBPJDGROSA-N (5S,5aR,8aR,9R)-5-[[(2R,4aR,6R,7R,8R,8aS)-7,8-dihydroxy-2-thiophen-2-yl-4,4a,6,7,8,8a-hexahydropyrano[3,2-d][1,3]dioxin-6-yl]oxy]-9-(4-hydroxy-3,5-dimethoxyphenyl)-5a,6,8a,9-tetrahydro-5H-[2]benzofuro[6,5-f][1,3]benzodioxol-8-one Chemical compound COC1=C(O)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H]4O[C@@H](OC[C@H]4O3)C=3SC=CC=3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 NRUKOCRGYNPUPR-QBPJDGROSA-N 0.000 description 1
- 229920000160 (ribonucleotides)n+m Polymers 0.000 description 1
- VERWOWGGCGHDQE-UHFFFAOYSA-N 5-chloro-2-N-(5-methyl-4-piperidin-4-yl-2-propan-2-yloxyphenyl)-4-N-(2-propan-2-ylsulfonylphenyl)pyrimidine-2,4-diamine Chemical compound CC=1C=C(NC=2N=C(NC=3C(=CC=CC=3)S(=O)(=O)C(C)C)C(Cl)=CN=2)C(OC(C)C)=CC=1C1CCNCC1 VERWOWGGCGHDQE-UHFFFAOYSA-N 0.000 description 1
- 101710034857 ATIC Proteins 0.000 description 1
- 229960001686 Afatinib Drugs 0.000 description 1
- 229940110282 Alimta Drugs 0.000 description 1
- WNROFYMDJYEPJX-UHFFFAOYSA-K Aluminium hydroxide Chemical compound [OH-].[OH-].[OH-].[Al+3] WNROFYMDJYEPJX-UHFFFAOYSA-K 0.000 description 1
- DIZPMCHEQGEION-UHFFFAOYSA-H Aluminium sulfate Chemical compound [Al+3].[Al+3].[O-]S([O-])(=O)=O.[O-]S([O-])(=O)=O.[O-]S([O-])(=O)=O DIZPMCHEQGEION-UHFFFAOYSA-H 0.000 description 1
- 206010003816 Autoimmune disease Diseases 0.000 description 1
- 206010004146 Basal cell carcinoma Diseases 0.000 description 1
- 108010005144 Bevacizumab Proteins 0.000 description 1
- 210000004204 Blood Vessels Anatomy 0.000 description 1
- 210000000988 Bone and Bones Anatomy 0.000 description 1
- 102100008151 CCR7 Human genes 0.000 description 1
- 210000001666 CD4-positive, alpha-beta memory T lymphocyte Anatomy 0.000 description 1
- KTEIFNKAUNYNJU-GFCCVEGCSA-N Crizotinib Chemical compound O([C@H](C)C=1C(=C(F)C=CC=1Cl)Cl)C(C(=NC=1)N)=CC=1C(=C1)C=NN1C1CCNCC1 KTEIFNKAUNYNJU-GFCCVEGCSA-N 0.000 description 1
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 1
- 229960004397 Cyclophosphamide Drugs 0.000 description 1
- 210000004443 Dendritic Cells Anatomy 0.000 description 1
- 102000016911 Deoxyribonucleases Human genes 0.000 description 1
- 108010053770 Deoxyribonucleases Proteins 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- 208000000059 Dyspnea Diseases 0.000 description 1
- 206010013975 Dyspnoeas Diseases 0.000 description 1
- 229940095399 Enema Drugs 0.000 description 1
- 241000792859 Enema Species 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 229960005420 Etoposide Drugs 0.000 description 1
- VJJPUSNTGOMMGY-MRVIYFEKSA-N Etoposide Chemical compound COC1=C(O)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H]4O[C@H](C)OC[C@H]4O3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 VJJPUSNTGOMMGY-MRVIYFEKSA-N 0.000 description 1
- IRXSLJNXXZKURP-UHFFFAOYSA-N Fluorenylmethyloxycarbonyl chloride Chemical compound C1=CC=C2C(COC(=O)Cl)C3=CC=CC=C3C2=C1 IRXSLJNXXZKURP-UHFFFAOYSA-N 0.000 description 1
- 210000001035 Gastrointestinal Tract Anatomy 0.000 description 1
- XGALLCVXEZPNRQ-UHFFFAOYSA-N Gefitinib Chemical compound C=12C=C(OCCCN3CCOCC3)C(OC)=CC2=NC=NC=1NC1=CC=C(F)C(Cl)=C1 XGALLCVXEZPNRQ-UHFFFAOYSA-N 0.000 description 1
- SDUQYLNIPVEERB-QPPQHZFASA-N Gemcitabine Chemical compound O=C1N=C(N)C=CN1[C@H]1C(F)(F)[C@H](O)[C@@H](CO)O1 SDUQYLNIPVEERB-QPPQHZFASA-N 0.000 description 1
- 229940020967 Gemzar Drugs 0.000 description 1
- 229960002743 Glutamine Drugs 0.000 description 1
- JKMHFZQWWAIEOD-UHFFFAOYSA-N HEPES Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 1
- 239000007995 HEPES buffer Substances 0.000 description 1
- 102000011786 HLA-A Antigens Human genes 0.000 description 1
- 108010075704 HLA-A Antigens Proteins 0.000 description 1
- 102000006354 HLA-DR Antigens Human genes 0.000 description 1
- 108010058597 HLA-DR Antigens Proteins 0.000 description 1
- 108010068250 Herpes Simplex Virus Protein Vmw65 Proteins 0.000 description 1
- 229960001101 Ifosfamide Drugs 0.000 description 1
- HOMGKSMUEGBAAB-UHFFFAOYSA-N Ifosfamide Chemical compound ClCCNP1(=O)OCCCN1CCCl HOMGKSMUEGBAAB-UHFFFAOYSA-N 0.000 description 1
- 108010089187 Ipilimumab Proteins 0.000 description 1
- 241000229754 Iva xanthiifolia Species 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- 239000005411 L01XE02 - Gefitinib Substances 0.000 description 1
- 239000002146 L01XE16 - Crizotinib Substances 0.000 description 1
- 210000004698 Lymphocytes Anatomy 0.000 description 1
- 101700036258 MECOM Proteins 0.000 description 1
- 208000002720 Malnutrition Diseases 0.000 description 1
- 229960004857 Mitomycin Drugs 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 241000699660 Mus musculus Species 0.000 description 1
- NWIBSHFKIJFRCO-WUDYKRTCSA-N Mytomycin Chemical compound C1N2C(C(C(C)=C(N)C3=O)=O)=C3[C@@H](COC(N)=O)[C@@]2(OC)[C@@H]2[C@H]1N2 NWIBSHFKIJFRCO-WUDYKRTCSA-N 0.000 description 1
- 229940086322 Navelbine Drugs 0.000 description 1
- 239000000020 Nitrocellulose Substances 0.000 description 1
- 210000001672 Ovary Anatomy 0.000 description 1
- 229940049954 Penicillin Drugs 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 210000004224 Pleura Anatomy 0.000 description 1
- 210000004910 Pleural fluid Anatomy 0.000 description 1
- 206010035623 Pleuritic pain Diseases 0.000 description 1
- XOFYZVNMUHMLCC-ZPOLXVRWSA-N Prednisone Chemical compound O=C1C=C[C@]2(C)[C@H]3C(=O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 XOFYZVNMUHMLCC-ZPOLXVRWSA-N 0.000 description 1
- 102000004022 Protein-Tyrosine Kinases Human genes 0.000 description 1
- 108090000412 Protein-Tyrosine Kinases Proteins 0.000 description 1
- 101710037934 QRSL1 Proteins 0.000 description 1
- 239000007759 RPMI Media 1640 Substances 0.000 description 1
- 102100001773 SENP7 Human genes 0.000 description 1
- 101700006371 SENP7 Proteins 0.000 description 1
- 101700064309 SP2 Proteins 0.000 description 1
- 210000003491 Skin Anatomy 0.000 description 1
- 206010041823 Squamous cell carcinoma Diseases 0.000 description 1
- 229960005322 Streptomycin Drugs 0.000 description 1
- 108091008153 T cell receptors Proteins 0.000 description 1
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 1
- 101710042353 TRMT1 Proteins 0.000 description 1
- BPEGJWRSRHCHSN-UHFFFAOYSA-N Temodal Chemical compound O=C1N(C)N=NC2=C(C(N)=O)N=CN21 BPEGJWRSRHCHSN-UHFFFAOYSA-N 0.000 description 1
- 229960001278 Teniposide Drugs 0.000 description 1
- UCFGDBYHRUNTLO-QHCPKHFHSA-N Topotecan Chemical compound C1=C(O)C(CN(C)C)=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 UCFGDBYHRUNTLO-QHCPKHFHSA-N 0.000 description 1
- 229950007217 Tremelimumab Drugs 0.000 description 1
- 102000009524 Vascular Endothelial Growth Factor A Human genes 0.000 description 1
- 108010073929 Vascular Endothelial Growth Factor A Proteins 0.000 description 1
- ZCBJDQBSLZREAA-UHFFFAOYSA-N [4-[2-(4-acetyloxyphenyl)-3-oxo-4H-1,4-benzoxazin-2-yl]phenyl] acetate Chemical compound C1=CC(OC(=O)C)=CC=C1C1(C=2C=CC(OC(C)=O)=CC=2)C(=O)NC2=CC=CC=C2O1 ZCBJDQBSLZREAA-UHFFFAOYSA-N 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 230000001154 acute Effects 0.000 description 1
- 230000002730 additional Effects 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 230000000996 additive Effects 0.000 description 1
- 239000000443 aerosol Substances 0.000 description 1
- ULXXDDBFHOBEHA-CWDCEQMOSA-N afatinib Chemical compound N1=CN=C2C=C(O[C@@H]3COCC3)C(NC(=O)/C=C/CN(C)C)=CC2=C1NC1=CC=C(F)C(Cl)=C1 ULXXDDBFHOBEHA-CWDCEQMOSA-N 0.000 description 1
- 230000032683 aging Effects 0.000 description 1
- 125000000539 amino acid group Chemical group 0.000 description 1
- 150000001413 amino acids Chemical group 0.000 description 1
- 230000003466 anti-cipated Effects 0.000 description 1
- 230000000890 antigenic Effects 0.000 description 1
- 239000008365 aqueous carrier Substances 0.000 description 1
- 229960000626 benzylpenicillin Drugs 0.000 description 1
- 229960000397 bevacizumab Drugs 0.000 description 1
- 229960000074 biopharmaceuticals Drugs 0.000 description 1
- 230000015572 biosynthetic process Effects 0.000 description 1
- UIIMBOGNXHQVGW-UHFFFAOYSA-M buffer Substances [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 1
- 201000009030 carcinoma Diseases 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 229960001602 ceritinib Drugs 0.000 description 1
- 201000010881 cervical cancer Diseases 0.000 description 1
- 230000001684 chronic Effects 0.000 description 1
- 230000001427 coherent Effects 0.000 description 1
- 238000007906 compression Methods 0.000 description 1
- 238000004590 computer program Methods 0.000 description 1
- 230000001276 controlling effect Effects 0.000 description 1
- 229960005061 crizotinib Drugs 0.000 description 1
- 238000010192 crystallographic characterization Methods 0.000 description 1
- 230000002354 daily Effects 0.000 description 1
- 238000007405 data analysis Methods 0.000 description 1
- 230000003247 decreasing Effects 0.000 description 1
- 230000002939 deleterious Effects 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 201000009910 diseases by infectious agent Diseases 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 239000002158 endotoxin Substances 0.000 description 1
- 239000007920 enema Substances 0.000 description 1
- 230000029578 entry into host Effects 0.000 description 1
- 102000017256 epidermal growth factor-activated receptor activity proteins Human genes 0.000 description 1
- 108040009258 epidermal growth factor-activated receptor activity proteins Proteins 0.000 description 1
- 239000003797 essential amino acid Substances 0.000 description 1
- 235000020776 essential amino acid Nutrition 0.000 description 1
- 238000009093 first-line therapy Methods 0.000 description 1
- 238000007710 freezing Methods 0.000 description 1
- 229960002584 gefitinib Drugs 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- 108010065889 glycyl-leucyl-cysteinyl-threonyl-leucyl-valyl-alanyl-methionyl-leucine Proteins 0.000 description 1
- 230000035929 gnawing Effects 0.000 description 1
- 229920000591 gum Polymers 0.000 description 1
- 238000000265 homogenisation Methods 0.000 description 1
- 230000002519 immonomodulatory Effects 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 238000011065 in-situ storage Methods 0.000 description 1
- 230000002458 infectious Effects 0.000 description 1
- 230000036512 infertility Effects 0.000 description 1
- 230000000977 initiatory Effects 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007913 intrathecal administration Methods 0.000 description 1
- 229960005386 ipilimumab Drugs 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 230000003211 malignant Effects 0.000 description 1
- 230000001071 malnutrition Effects 0.000 description 1
- 235000000824 malnutrition Nutrition 0.000 description 1
- 238000009126 molecular therapy Methods 0.000 description 1
- 108010045030 monoclonal antibodies Proteins 0.000 description 1
- 102000005614 monoclonal antibodies Human genes 0.000 description 1
- 230000000926 neurological Effects 0.000 description 1
- 229920001220 nitrocellulos Polymers 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 210000000056 organs Anatomy 0.000 description 1
- 230000000803 paradoxical Effects 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 230000001717 pathogenic Effects 0.000 description 1
- 239000008188 pellet Substances 0.000 description 1
- 239000000546 pharmaceutic aid Substances 0.000 description 1
- 238000009520 phase I clinical trial Methods 0.000 description 1
- 229960004618 prednisone Drugs 0.000 description 1
- 238000002360 preparation method Methods 0.000 description 1
- 230000003449 preventive Effects 0.000 description 1
- 238000004393 prognosis Methods 0.000 description 1
- 230000002250 progressing Effects 0.000 description 1
- 230000000750 progressive Effects 0.000 description 1
- 108010026911 ramucirumab Proteins 0.000 description 1
- 229960002633 ramucirumab Drugs 0.000 description 1
- 230000000306 recurrent Effects 0.000 description 1
- 230000002829 reduced Effects 0.000 description 1
- 239000011347 resin Substances 0.000 description 1
- 229920005989 resin Polymers 0.000 description 1
- 230000003248 secreting Effects 0.000 description 1
- 230000028327 secretion Effects 0.000 description 1
- 230000000276 sedentary Effects 0.000 description 1
- 230000000405 serological Effects 0.000 description 1
- KEAYESYHFKHZAL-UHFFFAOYSA-N sodium Chemical compound [Na] KEAYESYHFKHZAL-UHFFFAOYSA-N 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 239000007790 solid phase Substances 0.000 description 1
- 239000000243 solution Substances 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 231100000803 sterility Toxicity 0.000 description 1
- 230000004936 stimulating Effects 0.000 description 1
- 238000010254 subcutaneous injection Methods 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 230000002459 sustained Effects 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 230000002194 synthesizing Effects 0.000 description 1
- 229960004964 temozolomide Drugs 0.000 description 1
- 125000005931 tert-butyloxycarbonyl group Chemical group [H]C([H])([H])C(OC(*)=O)(C([H])([H])[H])C([H])([H])[H] 0.000 description 1
- 210000001519 tissues Anatomy 0.000 description 1
- 229960000303 topotecan Drugs 0.000 description 1
- 108010072993 tremelimumab Proteins 0.000 description 1
- 238000004450 types of analysis Methods 0.000 description 1
- 239000005483 tyrosine kinase inhibitor Substances 0.000 description 1
- 229940121358 tyrosine kinase inhibitors Drugs 0.000 description 1
- 210000000689 upper leg Anatomy 0.000 description 1
- 238000009423 ventilation Methods 0.000 description 1
- GBABOYUKABKIAF-IELIFDKJSA-N vinorelbine Chemical compound C1N(CC=2C3=CC=CC=C3NC=22)CC(CC)=C[C@H]1C[C@]2(C(=O)OC)C1=CC([C@]23[C@H]([C@@]([C@H](OC(C)=O)[C@]4(CC)C=CCN([C@H]34)CC2)(O)C(=O)OC)N2C)=C2C=C1OC GBABOYUKABKIAF-IELIFDKJSA-N 0.000 description 1
- CILBMBUYJCWATM-IJDPFCGHSA-N vinorelbine L-tartrate Chemical compound OC(=O)[C@H](O)[C@@H](O)C(O)=O.OC(=O)[C@H](O)[C@@H](O)C(O)=O.C1N(CC=2C3=CC=CC=C3NC=22)CC(CC)=C[C@H]1C[C@]2(C(=O)OC)C1=CC([C@]23[C@H]([C@]([C@H](OC(C)=O)[C@]4(CC)C=CCN([C@H]34)CC2)(O)C(=O)OC)N2C)=C2C=C1OC CILBMBUYJCWATM-IJDPFCGHSA-N 0.000 description 1
- 230000003612 virological Effects 0.000 description 1
- 238000010626 work up procedure Methods 0.000 description 1
Abstract
The present invention relates to a therapeutic peptide T specific immune therapy for use in the treatment of a cancer of an HLA-A2 (Human Leukocyte Antigen A2) positive patient, said treatment comprises a priming period consisting in two to three administrations of said therapeutic peptide T specific immune therapy, thereby inducing a memory T cell response. c immune therapy, thereby inducing a memory T cell response.
Description
(12) Granted patent specificaon (19) NZ (11) 737717 (13) B2
(47) Publicaon date: 2021.12.24
(54) METHOD FOR NG EARLY T MEMORY RESPONSE WITH SHORT PEPTIDES ANTI-TUMOR
VACCINE
(51) Internaonal Patent Classificaon(s):
A61K 39/00
(22) Filing date: (73) Owner(s):
6.29 OSE IMMUNOTHERAPEUTICS
(23) Complete specificaon filing date: (74) Contact:
2015.06.29 AJ PARK
(86) Internaonal Applicaon No.: (72) Inventor(s):
COSTANTINI, Dominique
(87) Internaonal Publicaon number:
WO/2017/000983
(57) Abstract:
The present invenon relates to a euc pepde T specific immune therapy for use in the
treatment of a cancer of an HLA-A2 (Human Leukocyte Angen A2) posive paent, said treatment
comprises a g period consisng in two to three straons of said therapeuc pepde T
specific immune therapy, thereby inducing a memory T cell response.
NZ 737717 B2
Method for inducing early T memory response with short peptides anti-
tumor vaccine.
Field ofthe ion
The present invention relates to the field of medicine, in ular of oncology, and more
particularly the present invention relates to the treatment of cancer.
Background ofthe Invention
Memory T cells are a subset of antigen—specific T cells that persist erm after having
tered and responded to their cognate antigen. They quickly expand to large numbers of
effector T cells upon re—exposure to their cognate antigen, thus providing the immune system
with "memory" against past infections as well as cancer cells. At the second ter with the
invader or cancer cells, memory T cells can initiate a faster and stronger immune response than
the first time. Memory T cells se two subtypes: central memory T cells (TCM cells) and
effector memory T cells (TEM cells). Memory cells may be either CD4+ or CD8+.
It has recently been established, for several human cancers, that high densities of or
memory CD8+ cytotoxic T cells are associated with a longer overall survival (Fridman W. et al,
2012, Nat Rev Cancer, 12(4), 298—306). Central memory T cells appears however to have an
even greater capacity to persist in vivo and are also more efficient in mediating protective
immunity because of their increased proliferative capacity (Calarota SA et al, 2013, al
and Developmental logy, Article ID 637649).
In the literature, data indicate that an initial proper activation of the CD8+ T cell response by
peptide vaccines in mineral oil adjuvant does not ensure long—term effectiveness of these CD8+
T cells (Bijker MS et a1, 2007, J Immunol, 179:5033—5040), although, such a long-term
effectiveness would be of huge interest in the field of chronic diseases such as cancer.
Bijker et al bed a specific strategy linked to the use of long peptides to overcome the poor
long term effectiveness of cancer vaccines. However, this strategy was shown able by
Karkada et a1. (Karkada M et a1, 2014, Biologics: Target and Therapy, 8: 27-38). Indeed, the
immune response with such long peptides was not constant at each time point and was
decreasing after the first injection.
Thus, there is still a strong need, nowadays, to develop a method of ent with peptide
cancer vaccine that would be able to ate memory T cells as early as possible, especially
central memory T cells.
The classical administration regimen of short peptide cancer vaccines rely on at least 5-6
administrations of the vaccine. Holmes et al. (defines the optimal scheme of administration of
a specific short peptide cancer vaccine as 6 injections of e for each month of treatment
s JP et al, 2008, Cancer , 113 , 1666–1675). In a phase two trial, subjects were
considering to be effectively part of the ation protocol if they had received at least 6
administrations of the vaccine r S et al, 2012, Nature Medicine, 18 , 1254–126). Seven
to ten short peptide vaccine administration were considered by others for an effective treatment
(Kantoff PW et al, 2010, Cancer J Clin Oncol, 28 :1099-1105; Schwartzentruber DJ et al, 2011,
N Engl J Med, 364 27).
These short peptide vaccine treatments can be painful for the ts and are not devoid of side
effects and toxicity. In addition, controlling health care costs and optimizing cancer treatment
are key issues in eration of the rise of costs. There is thus a strong need to find new
methods allowing to modify the scheme of administration of the vaccine so as to reduce the
number of injections while obtaining a memory T response as early as possible.
Summary of the Invention
The object of the present invention relates to a new administration regimen of a short peptide
cancer vaccine ng an early memory T cells se. In this invention, the inventors
singly showed that no more than three administrations of the short peptide ation
were sufficient to obtain a T specific Immune response through memory T Cells and this
immune response was linked with a longer time to progression (TTP).
The present invention relates to a therapeutic peptide T specific immune therapy for use in the
treatment of a cancer of an HLA-A2 (Human Leukocyte Antigen A2) ve patient, wherein
the therapeutic peptide T specific immune y comprises the peptide aKXVAAWTLKAAa
(SEQ ID No 10, with X and a respectively indicating cyclohexylalanine and d-alanine) and at
least 4, 5, 6, 7, 8 or 9 peptides selected from the group consisting of RLLQETELV (SEQ ID
No 1), YLQLVFGIEV (SEQ ID No 2), LLTFWNPPV (SEQ ID No 3), KVFGSLAFV (SEQ
ID No 4), KLBPVQLWV (SEQ ID No 5, with B indicating α-aminoisobutyric acid),
SMPPPGTRV (SEQ ID No 6), IMIGHLVGV (SEQ ID No 7), KVAEIVHFL (SEQ ID No 8),
and YLSGADLNL (SEQ ID No 9), and wherein said treatment comprises a priming period
consisting in two to three administrations of said therapeutic peptide T specific immune therapy,
thereby inducing a central memory T cell response.
In a particular , the present invention provides a use of a therapeutic peptide T specific
[FOLLOWED BY PAGE 2a]
immune therapy in the manufacture of a medicament for the treatment of a cancer of an HLAA2
(Human Leukocyte Antigen A2) positive patient, wherein the therapeutic peptide T
specific immune y comprises the peptide aKXVAAWTLKAAa (SEQ ID No 10, with X
and a respectively indicating cyclohexylalanine and d-alanine) and the peptides RLLQETELV
(SEQ ID No 1), YLQLVFGIEV (SEQ ID No 2), LLTFWNPPV (SEQ ID No 3),
KVFGSLAFV (SEQ ID No 4), KLBPVQLWV (SEQ ID No 5, with B indicating α-
aminoisobutyric acid), SMPPPGTRV (SEQ ID No 6), IMIGHLVGV (SEQ ID No 7),
KVAEIVHFL (SEQ ID No 8), and LNL (SEQ ID No 9), and wherein said
treatment comprises a priming period consisting of three strations of said therapeutic
peptide T specific immune therapy, the eutic peptide T specific immune therapy being
administered at one dose every three weeks during which a central memory T cell se is
induced against one or several peptides of the therapeutic e T specific immunotherapy,
and wherein the priming period is followed by a maintenance period during which the
administration of said therapeutic e T specific immune therapy occurs every two to
eight months.
In a preferred embodiment, the therapeutic peptide T specific immune therapy is the
combination of 10 peptides (called herein 01) comprising (or consisting in) the
[FOLLOWED BY PAGE 3]
following peptides RLLQETELV (SEQ ID No l), YLQLVFGIEV (SEQ ID No 2),
LLTFWNPPV (SEQ ID No 3), KVFGSLAFV (SEQ ID No 4), KLBPVQLWV (SEQ ID No 5,
with B indicating a—aminoisobutyric acid), SMPPPGTRV (SEQ ID No 6), IMIGHLVGV (SEQ
ID No 7), KVAEIVHFL (SEQ ID No 8), YLSGADLNL (SEQ ID No 9), aKXVAAWTLKAAa
(SEQ ID No 10, with X and a respectively indicating cyclohexylalanine and d—alanine.
More specifically, said treatment comprises a g period consisting in two to three
administrations of the peptide T specific cancer immunotherapy, thereby inducing a central
memory T cell response. More preferably, the treatment comprises a priming period consisting
in three administrations of the therapeutic peptide T specific immune therapy. In a particular
embodiment, in the priming period, the therapeutic peptide T specific immune therapy is
administered every one-four weeks, preferably every ree weeks, more preferably every
3 weeks.
ally, in the treatment, the priming period is followed by a maintenance period during
which the administration of the therapeutic peptide T specific immune therapy occurs every
two—eight months, preferably every two-three , more ably every two months
through one year and then every three months through one year.
Preferably, the therapeutic peptide T specific immune y is administered parentally,
preferably subcutaneously.
Preferably, the peptides are emulsified in incomplete Freund’s nt or the like, preferably
Montanide ISA-51.
Preferably, the doses of peptide are ranging from 0.1 to 10 mg of peptide per injection dose.
Optionally, the total peptide dose for each injection is 5.0 mg.
Preferably, the cancer is a cancer selected from the group consisting of lung cancer such as
NSCLC (non-small cell lung cancer) and small cell lung cancer, melanoma, mesothelioma,
breast cancers, primary brain cancers, ovarian cancer, uterine carcinoma, especially uterine
corpus and/or uterine cervix oma, head and neck cancer, colon or ctal cancer,
gastro—intestinal cancer, renal cancer, sarcoma, germ cell tumors, ia, lymphoma,
testicular cancers and bladder cancers, ably ed from the group consisting of NSCLC,
colon cancer, breast cancer, ovarian cancer, and a cancer of the head and/or neck, more
ably NSCLC.
The t is patient HLA—A2 positive patient. Optionally, the patient suffers from an advanced
or late-stage cancer. Optionally, the patient suffers from metastases, ally brain
metastases. Optionally, the patient has a malignant pleural effusion. Optionally, the patient has
W0 2017/000983
already received several lines of treatment prior to the treatment with the therapeutic peptide T
specific immune therapy.
Optionally, the treatment is used in combination with a treatment with another antitumor drug
and/or with radiotherapy, especially with a checkpoint inhibitor, preferably CTLA—4 and/or PD—
l/PD-Ll inhibitor such as pembrolizumab, nivolumab, pidilizumab, BMS936559, MEDI4736,
AMP-224, AMP—514, MPDL3280A and avelumab, more ularly in order to transform
“non immunogenic T memory” patients into “immunogenic T memory” patients.
Detailed description ofthe Invention
Previously, only CTL effector cells and HTL effector cells responses were identified following
original combination of short peptides epitopes (EPl620456 describing chemical optimization
on the binding to the key receptors of T response and original combination of different nature
of epitopes) as T cell specific immunotherapy in oncology. Both CTL (Cytotoxic T
cytes) and HTL (Helper T cytes) immune responses versus all es were
based on effectors T cells after injection of such combination of epitopes from different nature
(fixed anchors, heteroclitic and wild type epitope). EPl620456 patent describing the immune
response with such original combination was using a standard Elispot measuring effector T
cells assay without in vitro expansion in HLA-A2 transgenic models. This rd Elispot
assay allows the measurement of T cells capable of immediate secretion of IFN-y upon antigen/
e stimulation. These cells mainly represent effector T cells.
The categorization of memory T cells into different subsets (effector memory (TEM) and
central memory (TCM)) was studied in several studies exanimating which type of memory cell
is capable of providing optimal protection (Jennifer D t et a1, 2012, Molecular Therapy,
20, 860—869).
In order to standardize these measures, two t assays allow to separate this 2 main
populations: the standard ELISPOT assay quantifies or memory T cells whereas the
ed ELISPOT assay quantifies expandable memory T cells, representing l memory
T cells. Evidence indicates that a different population of T cells, most likely central memory T
cells that differentiate into effector T cells during the e period, are measured by the
cultured T assay, as compared with the measurement of circulating effector memory T
cells that are quantified by the standard ELISPOT (Calarotra S.A. et al . This ed
ELISPOT assay is performed by ing lymphocytes with specific antigens for 10 days
allowing T cells to expand in response to the antigen. Then, a standard ELISPOT procedure is
applied in response to the corresponding ns used for the 10-day stimulation period
measuring effector function.
Central memory T cells require antigenic re-stimulation to develop effector function. The
cultured ELISPOT assay mainly comprises these central memory T cells because the depletion
of central-memory T cell population ablated completely responses in cultured Elispot (S. M.
Todryk, et al., 2009, Immunology, 128, 83—91). Todryk et al explains the predominant role of
central memory T cells in the cultured ELISPOT providing CD4+ and CD8+ specific response.
Central memory T cells have a r capacity than effector memory T cells to persist in vivo
and are more efficient in mediating protective immunity because of their sed proliferative
capacity.
The invention described below l an unmet need in oncology, providing an optimized
administration regimen allowing an early memory T cell response in cancer treatment,
preferably an early central memory T cell response, and more particularly an early long term
memory T cell response.
More particularly, the present ion relates to a new administration regimen of a short
peptide cancer therapy based on multi—epitope T specific cancer immunotherapy. Surprisingly,
the inventors have demonstrated that the administration of a short peptide multi—epitopes
combination leads to an cted strong immune response involving long term memory T
cells, in particular central memory T cells. Furthermore, the inventors have also discovered that
a very short administration regimen with only three injections is enough to induce this strong
and long term response.
Accordingly, the present ion relates to a therapeutic peptide T specific immune therapy
as defined herein for use in the ent of a cancer of an HLA-A2 (Human Leukocyte Antigen
A2) ve patient, wherein said treatment ses a g period consisting in two to
three administrations of said therapeutic peptide T specific immune therapy. It also s to a
method of treatment of a cancer of an HLA-A2 (Human Leukocyte Antigen A2) ve patient
in need thereof comprising a priming period ting in two to three administrations of said
therapeutic peptide T specific immune therapy OSE2101 as defined herein. Finally, the present
invention relates to the use of a therapeutic peptide T specific immune therapy 1 as
defined herein for the manufacture of a cancer treatment, wherein the therapeutic peptide T
specific immune therapy is to be administered two to three times during the g period.
WO 00983
Definitions
An "epitope" is the collective features of a molecule, such as primary, secondary and tertiary
peptide structure, and charge, that together form a site recognized by an immunoglobulin, T
cell receptor or HLA molecule. Alternatively, an epitope can be defined as a set of amino acid
residues which is involved in recognition by a particular immunoglobulin, or in the context of
T cells, those residues necessary for recognition by T cell or proteins and/or Major
Histocompatibility x (MHC) receptors. Epitopes are present in nature, and can be
isolated, purified or otherwise prepared or derived by humans. For example, epitopes can be
prepared by isolation from a natural source, or they can be synthesized in accordance with
standard protocols in the art. Throughout this disclosure, epitopes may be referred in some cases
as peptides or peptide epitopes.
"Human Leukocyte Antigen" or "HLA" is a human class I or class 11 Major Histocompatibility
Complex (MHC) protein (see, e.g., Stites, et all, IMMUNOLOGY, 8TH ED., Lange Publishing,
Los Altos, CA (1994). HLA molecules are d on the basis of shared peptide—binding
icities. For example, HLA—A2 is a particular type of HLA molecules which share similar
binding affinity for peptides bearing certain amino acid motifs. The methods for ining
the HLA—A2 status in a patient are well—known and easy to obtain (i.e; serological samples) by
the one skilled in the art.
A "peptide epitope" is a peptide that comprises an allele-specific motif or supermotif such that
the peptide will bind an HLA molecule and induce a CTL and/or HTL response. Thus, peptide
epitopes of the invention are capable of binding to an appropriate HLA—A2 molecule and
thereafter inducing a cytotoxic T lymphocyte (CTL) response, or a helper T lymphocyte (HTL)
response, to the peptide.
A "PanDR peptide" or "PADRE®" peptide is a member of a family of molecules that binds
more than one HLA class II molecule. The pattern that defines the PADRE® family of
molecules can be referred to as an HLA Class II otif. A PADRE® molecule binds to
HLA class II molecules and stimulates in vitro and in viva human HTL responses. PADRE
peptides are described in the patent EP735893.
A "CTL and/or an HTL se" is a protective or therapeutic immune response to an n
derived from a pathogenic antigen (e‘ g‘ an antigen from an infectious agent or a tumor n),
which in some way prevents or at least partially s disease symptoms, side effects or
progression. The immune response may also e an antibody response which has been
facilitated by the stimulation of helper T cells.
As used herein, the term "memory T cell" is intended to e both the CCR7— (effector
memory T cells) and CCR7+ al memory T cells) subpopulations of T cells. This definition
also includes both class II-restricted CD4 memory T cells and class I—restricted CD8 memory
T cells.
The staging of a cancer describes the severity of a ’s cancer based on the size and/or
extent (reach) of the original (primary) tumor and whether or not cancer has spread in the body
tasis). NSCLC stages are numbered from 0 to IV. Stages IIIb and IV are the most
advanced stages.
"ECOG (Eastern Cooperative Oncology Group) Performance Status" are used by doctors and
researchers to assess how a patient's disease is progressing and assess how the e affects
the daily living abilities of the patient. ECOG mance Status are numbered from 0 to 5.
A performance status of 0 match to patients who are fully active and able to carry on all pre—
disease performance without restriction. A performance status of 1 match to patients who are
restricted in physically strenuous activity but ambulatory and able to carry out work of a light
or sedentary nature, e. g., light house work, office work.
The term "overall survival" (OS) refers to the length of time from the date of the start of
treatment that patients are still alive. In a clinical trial, measuring the overall al is one
way to see how well a new treatment works.
Therapeutic peptide T specific immunotherapy or peptide vaccine
The Multi—epitopes T ic cancer immunotherapy of the invention is able to help the
immune system to p immune memory that can have asting, tumor—specific effects.
An effective peptide T specific cancer immunotherapy requires induction of a wide breadth of
CTL specificities. This can be best achieved with optimized epitopes targeting multiple Tumor
Associated Antigens (TAAs) as a multi—epitopes combination ing at least 5 tumor antigens
and based on epitopes combination. Preferably, the at least 5 tumor ns include or are
selected among HER2/neu, CEA, MAGEZ, MAGE3 and p53.
The Multi-epitopes T specific cancer immunotherapy comprises a combination of epitopes that
can be wild—type epitopes and modified epitopes (heteroclitic and fixed anchors epitopes).
Preferably, the epitopes T specific cancer immunotherapy comprises at least 5 epitopes.
In a preferred embodiment, the combination of epitopes comprises at least 5 epitopes chosen
among those disclosed in Table 6 of the US application US2014/01474790 (incorporated herein
by reference). More preferably, the Multi—epitopes T ic cancer immunotherapy comprises
at least 5 epitopes allowing to target the combination of the following 5 tumor antigens:
HER2/neu, CEA, MAGE2, MAGE3 and p53. For instance, Multi—epitopes T specific cancer
therapy may comprise at least 5, 6, 7, 8 or 9 epitopes chosen among those disclosed in
Table 6 of the US application US2014/01474790.
The original combination used here (OSE-2lOl) is made by wild-type epitopes and modified
epitopes (heteroclitic and fixed anchors epitopes). More detailed information on heteroclitic
and fixed s epitopes can be found for instance in the patent EPl620456.
OSE—2101 is a multi-epitope T specific cancer immunotherapy composed of 10 synthetic
peptides. Nine of the peptides have been designed to induce a CTL response against TAAs.
More particularly, the T specific immune y is ed for administration to patients for
the induction of CTL directed against oembryonic antigen (CEA), p53, human epidermal
receptor—2/neurological (HER-2/neu) and melanoma antigen 2 and 3 (MAGE—2/3). These TAAs
have been chosen based on epidemiology because they are frequently over—expressed in various
advanced cancers as colon cancers, ovarian cancers, breast cancers and NSCLC. Each CTL
epitope is restricted by HLA—A2 superfamily of major histocompatibility complex class I
molecules, thereby providing coverage of approximately 45 % of the general population. The
tenth synthetic peptide is the pan-DR epitope (PADRE), a rationally designed helper T—
lymphocyte (HTL) epitope included only to increase the magnitude of CTL responses.
OSE-2lOl composition comprises or consists of the following peptides:
RLLQETELV SEQ ID No l
GIEV SEQ ID No 2
LLTFWNPPV SEQ ID No 3
KVFGSLAFV SEQ ID No 4
KLBPVQLWV SEQ ID No 5, with B indicating u—aminoisobutyric acid
SMPPPGTRV SEQ ID No 6
IMIGHLVGV SEQ ID No 7
KVAEIVHFL SEQ ID No 8
YLSGADLNL SEQ ID No 9
WTLKAAa SEQ ID No 10, with X and a tively indicating
exylalanine and d-alanine.
Therefore, the therapeutic peptide T specific immune therapy comprises the peptide
aKXVAAWTLKAAa (SEQ ID No 10, with X and a respectively indicating cyclohexylalanine
and d-alanine) and at least 4, 5, 6, 7, 8 or 9 es selected from the group ting of
RLLQETELV (SEQ ID No l), GIEV (SEQ ID No 2), LLTFWNPPV (SEQ ID No 3),
KVFGSLAFV (SEQ ID No 4), KLBPVQLWV (SEQ ID No 5, with B indicating 0t-
aminoisobutyric acid), SMPPPGTRV (SEQ ID No 6), IMIGHLVGV (SEQ ID No 7),
KVAEIVHFL (SEQ ID No 8), and YLSGADLNL (SEQ ID No 9).
The peptides can be sized using standard Boc or Fmoc chemistry for solid phase e
synthesis starting with the appropriate resin, and purified by standard methods. Alternatively,
the peptide may be produced by c engineering with recombinant cells or by RNA, for
instance by in vitro translation system.
The Multi-epitope T specific cancer immunotherapy composition may comprise a
pharmaceutically acceptable carrier or excipient. More preferably, the pharmaceutically
acceptable carrier is an aqueous carrier, especially a buffer. In particular, it may comprise one
or several adjuvants. For instance, adjuvants can be incomplete Freund’s adjuvant, mineral oil
adjuvant, aluminum hydroxide, or alum, . Other suitable adjuvants are well-known in
the art.
In one embodiment, the Multi-epitope T specific cancer immunotherapy may comprise e
pulsed antigen presentating cells, such as dendritic cells.
Preferably, in the Multi-epitope T specific cancer immunotherapy, the peptides are emulsified
in incomplete Freund’s adjuvant or the like. In a preferred embodiment, the adjuvant is a
mineral oil nt, similar to Incomplete Freund's Adjuvant, manufactured and supplied by
Seppic SA, Paris, . In a most preferred embodiment, the nt is Montanide® ISA
5 l .
Each e of the composition can be present at a concentration of 0.1 mg/ml to 1 mg/ml,
preferably 0.5 mg/ml. ably, all the es are present in the ition at the same
tration.
Preferably, Multi-epitope T specific cancer immunotherapy composition is a sterile,
preservative—free emulsion of the 10 peptides at a concentration of 0.5 mg/ml each, formulated
in Montanide® ISA 51 adjuvant at a ratio of 1:1 (w:w) and filled into rubber—stoppered glass
vials, and refrigerated at 2° to 8°C.
OSE-ZlOl is manufactured under aseptic conditions. Peptides are dissolved in three different
solvents, sterile filtered, pooled and then fied in adjuvant via homogenization under
controlled conditions. Product release testing included appearance, endotoxin, sterility,
viscosity, particle size, peptide concentration of each peptide, , pH and potency.
ation of OSE—2101 composition is detailed in W02004/094454, Fig 3A and pages 105—
106, the disclosure of which being incorporated herein by reference.
Optionally, in addition to the 10 peptides of OSE —2101, the peptide composition of the t
invention may further comprise additional peptides, in particular peptide epitopes used for
inducing of cytotoxic hocyte (CTL) responses and targeting TAAs. For instance, the
peptide composition of the present invention may further comprise a peptide as disclosed in
W02009/143843 (the disclosure of which being incorporated herein by reference), and more
ularly ID05 (SEQ ID No ll).
Administration regimen of the therapeutic peptide T specific immunotherapy:
The administration regimen of a vaccine comprises a priming period and optionally a
maintenance period.
The term “priming period” refers to the period of the vaccination process during which a central
memory T cell response is induced against one or several peptides of the therapeutic peptide T
specific immunotherapy.
The term “maintenance period” or “boosting period” refers to the period of the vaccination
process following the priming period during which the same therapeutic peptide T specific
immunotherapy is administered and the T memory immune response is sustained or enhanced.
According to a red aspect of the present invention, the priming period of a subject under
treatment with therapeutic peptide T specific therapy consists in one to three
strations of the eutic peptide T ic immunotherapy or vaccine.
In an even more preferred embodiment, the priming period consists in three administrations of
the therapeutic peptide T specific immunotherapy or vaccine.
During this priming period, the therapeutic peptide T ic immune therapy is administered
every one-four weeks, preferably every two-three weeks, more preferably every 3 weeks.
The priming period is nt for inducing a central memory T cell response against one or
l es of the therapeutic peptide T specific immunotherapy, preferably against at least
2, 3, 4 or 5 peptides of the therapeutic peptide T specific immunotherapy.
During the optional maintenance period, which follows the priming period, one or several
strations of the therapeutic peptide T specific therapy or vaccine are realized.
During this maintenance , the administration of the vaccine occurs every two-eight
3O months, preferably every two-three months, for instance every two month through one or two
years and then every three months through one or two years.
In an alternative ment, the priming period is not followed by a maintenance period.
The priming period and the maintenance period can be separated by a rest period which does
not include any the therapeutic peptide T specific immunotherapy administration. This rest
period can last four to twelve weeks.
In another alternative embodiment, the g period is followed by a treatment with a
checkpoint inhibitor as the T memory cells induced by the therapeutic peptide T specific
immunotherapy are rendered immunogenic the tumor environment.
The present invention further relates to a method for treating cancer in an HLA—A2 positive
t, comprising administering a therapeutic effective amount of therapeutic peptide T
specific immunotherapy as disclosed , wherein said treatment comprise a priming period
during which one to three administrations of said peptide vaccine are performed. The method
may further comprise a preliminary step of ining the HLA status of the t, selecting
the HLA-A2 positive patients and administering a therapeutic ive amount of the
therapeutic peptide T specific immunotherapy as disclosed herein to the HLA—A2 positive
patients. The method may further comprise a maintenance period as defined above. Preferably,
the therapeutic peptide T specific immunotherapy is 1.
Dosage
Within the context of the invention, the term “treatment” or “treating” denotes curative,
symptomatic, and preventive treatment. Pharmaceutical compositions and preparations of the
invention can be used in humans with existing cancer or tumor, preferably at late stages of
progression of the cancer. The pharmaceutical compositions and preparations of the invention
will not necessarily cure the patient who has the cancer but will delay or slow the progression
or prevent further ssion of the disease, ameliorating thereby the patient’s condition. In
ular, the pharmaceutical compositions and preparations of the invention reduce the
development of tumors, and/or prevent metastasis occurrence or development and cancer
e. In treating the cancer, the pharmaceutical composition of the invention is administered
in a eutically effective amount.
By "effective amount" it is meant the quantity of the pharmaceutical composition of the
invention which prevents, removes or s the deleterious effects of brain metastases. It is
understood that the administered dose may be adapted by those skilled in the art ing to
the patient, the pathology, the mode of administration, etc. The dosage and regimen depends on
the stage and severity of the disease to be treated, the weight and l state of health of the
patient and the judgment of the prescribing physician. More ularly, by “therapeutically
efficient amount of the therapeutic peptide T specific immunotherapy as disclosed herein” is
intended the amount which is sufficient to increase the overall al of a patient. In
particular, by “therapeutically efficient amount of the therapeutic peptide T specific
immunotherapy as sed herein” is intended the amount which is sufficient to induce a
central memory T cell response against one or several peptides of the therapeutic peptide T
ic immunotherapy.
Previous cancer trials have tested ting doses of peptide, ranging from 0.1 to 10 mg of
peptide per injection dose, emulsified in incomplete Freund’s adjuvant. At all doses tested, the
peptide/incomplete ’s adjuvant treatment was deemed to be safe and well tolerated, with
no severe dose-related ic toxicities being reported.
therapeutic peptide T specific immunotherapy as disclosed herein can be administered by any
appropriate route, in ular by eral route such as subcutaneous, intradermal or
intramuscular route or by aerosol, transmucosal, intrapleural, or intrathecal routes. In a most
preferred embodiment, the es composition is stered subcutaneously. Preferably,
the therapeutic peptide T specific immunotherapy as disclosed herein is designed for
subcutaneous injection.
Preferably, the doses of peptide are ranging from 0.1 to 10 mg of peptide per injection dose. In
a preferred embodiment, the total peptide dose for each ion or stration will be 5.0
mg (1 mL of drug product containing 0.5 mg of each peptide).
Cancer
According to a preferred aspect of the present invention, subjects of vaccination with OSE—
2101 composition are patients with a cancer. In a preferred ment, patient’s cancer are
due to one of the following cancers: lung cancer such as NSCLC (non—small cell lung cancer)
and small cell lung cancer, melanoma, mesothelioma, breast cancers, primary brain cancers,
ovarian, uterine carcinoma, especially uterine corpus and/or e cervix carcinoma, head and
neck, colon, gastro—intestinal, renal cancers, sarcoma, germ cell tumors, leukemia, lymphoma,
testicular cancers and bladder cancers, preferably NSCLC, colon cancer, breast cancer, ovarian
cancer, and a cancer of the head and/or neck, more preferably NSCLC.
In another preferred embodiment, patient’s cancer are ed cancer. The term “advanced
” refers to a cancer at an advanced stage of development, i.e. a cancer that has spread in
the body (metastasis). Preferably, the patient suffers from brain metastases.
Alternatively, the patient has a malignant pleural effusion, preferably a metastatic pleural
effusion, in particular associated with lung cancer, breast , lymphoma or leukemia.
Optionally, the patient has already ed several lines of treatment prior to the vaccination
by the peptide composition. In a particular embodiment, the patient has a positive HTL
I'CSpOl’lS6.
Combination with another antitumor drug.
Optionally, the treatment is used in combination with a ent with another antitumor drug,
in particular a chemotherapy, hormonotherapy and/or immunotherapy.
For instance, the chemotherapy can be selected among cisplatin, carboplatin,
cyclophosphamide, etoposide, teniposide, mitomycin, ecan, Vinorelbine, ide,
ifosfamide, temozolomide, fluorouracil (5FU), docetaxel, pemetrexed, navelbine, drugs that
target tumor blood vessel growth (VEGF) such as bevacizumab, ramucirumab; prednisone;
tyrosine kinase inhibitors targeting EGFR such as gefitinib, erlotinib, afatinib ; ALK inhibitors
such as crizotinib; ceritinib and any combination thereof.
In a preferred embodiment, the vaccine treatment of the present invention is used in
combination with a checkpoint inhibitor, especially a CTLA—4 inhibitor and/or a PD—l or PD—
Ll inhibitor; IDO inhibitors. The treatment with the checkpoint inhibitor can be performed
before, aneously or after the ent with the therapeutic peptide T specific
immunotherapy as disclosed , in particular the priming period of the treatment.
The present invention relates to a kit or product sing (a) the eutic effective amount
of eutic peptide T specific immunotherapy as disclosed herein; and (b) a check point
inhibitor, preferably a CTLA—4 inhibitor and/or PD-l or PD—Ll inhibitor, as a combined
preparation for simultaneous, separate or sequential use, in particular in the treatment of cancer.
Preferably, the therapeutic peptide T specific immunotherapy is OSE—2101.
In a preferred embodiment, the treatment with a checkpoint inhibitor is performed after the
priming period of the treatment with the eutic effective amount of therapeutic peptide T
ic immunotherapy as sed herein.
Several PD—l/PD—Ll inhibitors are already available or under clinical development. For
instance, the PD—l/PD—Ll inhibitors can be chosen among the non-exhaustive list including
lizumab (Merk), nivolumab (Bristol Myers Squibb), pidilizumab (Cure Tech),
BMS936559 (Bristol Myers Squibb), MEDI4736 (Astra Zeneca), AMP-224 (Astra Zeneca),
AMP-514 (Astra Zeneca), 80A (Roche), avelumab (also known as 0718C
from Merck KgA Serono /Pfizer) . For instance, the PD—l/PD—Ll inhibitors can be chosen
among those disclosed in W02013/079174,
For instance, the CTLA—4 inhibitors can be chosen among the non—exhaustive list including
Tremelimumab (Pfizer Medimmune ) and ipilimumab (BMS).
Further aspects and advantages of this invention will be disclosed in the following experimental
section, which should be regarded as illustrative and not limiting the scope of this application.
Description of the Figures
Figure 1: Phase 1/2 W9 W18 magnitude of central memory T cell responses induced by
OSE2101.
figm: Phase 2 Central memory T cells responses induced by OSE2101 for l epitopes
through the number of doses.
Figure 3: ation between OSE—2lOl-induced PADRE HTL ses and CTL / central
memory T cell responses.
Figure 4: Kaplan—Meier estimate of TTP.
Examples
Example 1: early immune response though central memory T cells involvement
In the Phase 1 /2 clinical studies, the safety and immunogenicity of a lO-peptide enhanced
epitope combination, OSE-2101, rationally designed to induce broad multi-epitope CTL
responses in early stage colon and NSCLC patients. In addition to multi-epitope coverage, OSE—
2101 also targeted es from five Tumor antigens which are widely expressed on breast,
colon and non—small cell lung, ovarian, tumors (CEA, HER—2/neu, p53, MAGE2 and MAGE3)
making this product suitable for therapy against different cancer tions. As a source of T—
cell helper for CTL ion, the universal HTL epitope Pan DR e was also included
and a mineral oil adjuvant was used for the final ation of this T specific cancer
immunotherapy.
Patient eligibility criteria. OSE—2101 was tested in two disease-specific phase I clinical trials
enrolling + patients with histologically confirmed stage IA NSCLC or stage III
colon cancer. All patients were diagnosed as NED within the past 6 months after undergoing
standard therapy. Patients met normal laboratory parameters for blood chemistry and white cell
counts and had an Eastern Cooperative Oncology Group performance status of 0 or 1. They
were excluded if treated with immunomodulatory agents within one month of study entry or
with other cancer immunotherapies. Patients with a history of other cancers, except basal or
squamous cell carcinoma of the skin or in situ cervical cancer, were also excluded as were
patients with concurrent acute l ions or specified autoimmune diseases.
Peptides. The immunogenicity of all immune monitoring epitopes was confirmed by
stimulating CTL induced by treatment of HLA—A2.1/Kb transgenic mice with a pre—clinical lot
of OSE-2101 (standard Elispot .
Clinical study design. Both trials were designed as phase 1/2, open label, center, single
dose, multiple stration studies to evaluate the safety and immunogenicity of the OSE—
2101 immunotherapy. Patients were treated with 1 ml OSE-2101 every 3 weeks for a total of
six doses. Each dose was administered subcutaneously in the same vicinity in the deltoid or
upper thigh region, or at a contralateral site if local side—effects were observed. The study
duration for each completed patient was 18 weeks. Clinical responses or patient survival were
not measured in either phase 1/ 2 s.
Clinical sample processing. Blood or heresis product was ed from patients at the
pre-treatment, the Week 9 and Week 18 time points, 3 weeks after the third and sixth dose of
OSE—2101 respectively. Peripheral blood mononuclear cells (PBMC) were isolated from
s within 24 hours of collection using a Ficoll-Paque density gradient and cryopreserved
in containers designed to ensure optimal freezing (Mr. Frosty, e). For use, cells were
rapidly thawed at 37°C then transferred to human AB serum-containing medium for work—up.
Measurement of central T memory cells responses and T effector memory cells. The CTL
responses in 16 total patients (10 colon and six NSCLC) were monitored for CTL responses
t each epitope and t the wild—type epitope of epitopes analogs. For each patient, the
pre-treatment, Week 9 and Week 18 time points were batch-tested in the same experiment to
allow pre— versus post- treatment response comparison in the same experiment. Central T
memory cells responses were measured using an IFN-y enzyme-linked spot (cultured
ELISPOT) assay following in vitro stimulation of PBMC for 10 days with each vaccine e.
Briefly, PBMC from each time point were stimulated on day 0 with 10 ug/ml of each epitope
individually in replicate in 48—well culture plates (2 x 106 PBMC/well). All cultures were fed
with rIL—2 (Endogen, Wobum, MA; 10 U/ml final concentration) on days 1, 4 and 7. Ten days
after initiation of e, cells were harvested and tested for activity against specific vaccine
peptides and ponding wild—type peptides of vaccine analogs in the T memory ELISPOT
assay. Cells were tested in Millipore IP 96—well plates pre—coated with mouse anti—human IFN—
7 antibody (Mabtech USA, Cincinnati, OH). Five x 104 cells and 1.25 x 104 cells from each
culture were plated in triplicate wells er with 105 irradiated autologous PBMC and 10
ug/ml peptide. As a control, CTL were also tested against an irrelevant HLA-A2.1—binding
HBV peptide. After 20 hours incubation, T memory ELISPOT plates were developed by
performing sequential incubations with biotinylated anti—human IFN—y antibody (Mabtech
USA), Avidin—Peroxidase Complex (APC, Vector Laboratories, game, CA), and 3—
amino—9—ethy1 carbazole (AEC) substrate (Sigma Aldrich, St. Louis, MO). Spot-forming cells
(SFC) were enumerated using a computer-assisted image analysis system (Zeiss KS ELISPOT
Reader, Carl Zeiss Microlmaging, Thomwood, NJ). Data is reported as the net SFC per 5 x 104
cells after subtracting spots induced with the irrelevant HBV peptide.
The central T memory cultured ELISPOT assay was ied using HLA-A2+ PBMC from
patients who demonstrated a recall T effector memory response against an EBV BMLFl CTL
epitope (sequence, GLCTLVAML) under cal stimulation conditions used for testing
clinical samples. Specific response generated from these donors were titrated at limiting cell
doses and a linear response was ed (correlation coefficient = 0.99). The lower detection
limit of the assay was ined to be 5 SFC/well and the upper limit was 600 SFC. The inter—
experiment reproducibility was evaluated by repeated testing of cryopreserved pre—vaccination
PBMC samples from positive donors on separate days, and the coefficient of variation (CV)
ranged from 7—12% in these assays. To assess inter—operator reproducibility, two operators
tested pre—treatment PBMC from the same patient on different days and the CV was 13—16% at
the optimal cell dose. The CV values were considered to be within an acceptable range of
variability and strongly supported the use of our assay for analyses of clinical immune
responses.
Response criteria. ve response criteria were established prospectively after analyzing the
variability of pre-treatment ses t each epitope and the irrelevant HBV control
epitope. The mean SFC response in a post-treatment sample was ered a positive epitope—
induced T memory cell response if it met all of the following: 1) was greater than 5 SFC above
the irrelevant epitope response, 2) was greater than the mean SFC of the irrelevant epitope
response, plus 2 SD, and 3) was ld greater than the SFC response in the pre—treatment
sample from the same patient, plus 2 SD. The inclusion of “plus two standard deviations” was
used to accommodate the assay variability and served to make the criteria more stringent.
RESULTS
Patient characterization. Fourteen patients with stage III colon cancer enrolled in the trial and
ten patients completed the study after receiving six doses of OSE—2101. In the NSCLC trial ten
ts with stage IIB/IIIA disease were enrolled and six patients completed the study.
OSE—2101 was deemed safe and tolerated by patients in the two clinical trials with l side—
effects common to peptides prepared in mineral oil adjuvant being observed.
Immunogenicity of OSE-2101 A total of 16 patients (10 colon cancer and 6 NSCLC) receiving
the full course of six OSE-2101 doses were evaluated for the frequency, breadth and magnitude
of TAA-specific central T memory cell responses d by the specific immunotherapy. To
improve detection of central memory T cell responses PBMC were stimulated in vitro for 10
days with each epitope. The effector activity of the in vitro expanded PBMC was measured
with an IFN—y ELISPOT assay t the respective epitope and an irrelevant HLA—A*0201—
binding HBV epitope. If the epitope was an analog the corresponding wild-type epitope was
also tested.
Results from immune monitoring of the 16 ts indicated that OSE-ZlOl was capable of
ng a wide breadth of central memory T cell responses in ts. In the colon cancer
trial, eight of the 10 ts surprisingly generated CTL responses against four or more vaccine
epitopes at the Week 9 and/or Week 18 time points (Figure l). The average magnitude of the
ed ELISPOT was in the same range between Week 9 and Week 18.
t 607 demonstrated the widest breadth and highest magnitude of central T cell memory
responses, displaying responses between 60 — 200 SFC per 5 x 104 cells against seven epitopes
at the Week 9 time point. More importantly, five of the seven induced T memory responses in
this patient were directed against wild—type epitopes. At the Week 18 time point, patient 607
generated central memory T cell responses, some exceeding 1000 SFC, against six of the same
es at Week 9.
Equally noteworthy were the multi-epitope responses observed in colon cancer patients 601,
603, 604 and 606 to five or more epitopes surprisingly at the Week 9 and/or also at Week 18
time .
Multi—epitope central memory T cell responses were also observed in NSCLC patients,
surprisingly at the Week 9 and/or also at Week 18 time points. All of the data described utilized
a testing protocol where PBMC samples were cultured in vitro for 10 days with individual OSE—
2101 epitopes to expand in viva—primed CTL prior to testing in the ELISPOT assay and the
measure central memory T cell response and the in vitro stimulation step as ed Elispot
was required to see ses to epitopes.
Immunogenicity of different epitope classes. Analysis of the frequency and magnitude of
CTL responses indicated that most of the es and all of the epitope classes ented in
the product were immunogenic in patients. Overall, eight of the nine vaccine epitopes induced
central memory T cell responses in at least one colon cancer t and six vaccine epitopes
were immunogenic in at least one NSCLC patient. Three of the fixed-anchor analogs
(CEA.24V9 (SEQ ID No 3), HER2.369V2V9 (SEQ ID No 4) and p53.139L2B3 (SEQ ID No
)) and two heteroclitic analogs 91H5 (SEQ ID No 7) and CEA.605D6 (SEQ ID No 9))
were particularly genic as 60-80% of colon cancer patients and 40—80% of the HLA—
A*0201—typed NSCLC patients responded to each of the s (Table 1A and 1B week 9 or
week 18 responses). The heightened immunogenicity of the analogs was also indicated by
average response magnitudes ranging from 70 — 180 SEC at Week 9 and from 90 — 315 SEC at
Week 18 in colon cancer patients (Table 1A) and 60 — 138 SEC at Week 18 in NSCLC patients
(Table 1B).
Immunological testing of the 16 colon and NSCLC patients who ted treatment indicated
that OSE—2101 was successful in inducing a wide breadth of CTL and memory T cell responses
in individual patients as soon as after 3 injections, characterized by the aneous induction
of CTL specificities directed t several es. The broad CTL/ though central memory
T cell responses observed in individual patients in both cohorts indicated the potential long term
efficacy for immunotherapy in early and late stage patients. Instead, simultaneous CTL and
central Memory T cell responses is addressing a long term clinical benefit of the multi—epitope
approach to cancer immunotherapy.
Table 1A
$58 Es w. m m. m 0 c a m w m m w a N m w w. a m w
.0 form
z use: m w w. w a, w o m m m a m N m m m w w m v
Enigma m
- - ed - i u, - - We - - we. - - Wm - - “I. - - - - u - - -. u -
w We a, ad Wm
.. .. . _. .. . . .
_ . _ _ .
, ,
nnflmfinfl m
E . . . , t . . .
m .
. . , . . . .
$ , , : .,
, .
05. . .
. . - , , . .
, 5
2335‘s. ,
.. . ,
. i . , . - .
Efiwuaié . . .
. . . t - . .
. .
msflmmwmxu Now NNN he ammo“, Em “3 0.0m fl: so mmm odmm ,. .1. X - NN Hm? Emma. Gum mm New 3 mm»
at 0.3m
33:: 5 53m hdm ad Hm: awn Wm. Dom as, 5%. .
- flaw . . - mm mwmm fimwfi n3. m.
an w
. 9mm NE. mama BE.
n _
I§.m.fiu . ._, . . ,
:23 .
, . ., .
, c . .1
.t . -
3 gnawing 9m mam . .
, . . . -
a: 9mg Dd Wm out meg
mama—ammo.— .
, . .
., . .. .
, n8. mam _ I .
_ :94 new can: mam...“
. .
- 3. mam 9% - ., - ..
, 5mm 3 mam QB 5mm, we Emma ad cam rm?
HHU .
N war
Nw ad SE. n.8, Eu 08 Now ad , Wm Maw Emma. 3.” Emma m.m New SE.
.3 magmm Enema a m Saga a 3 wow} m we oumwém w me m ma m
e, mp m m» m me m mw m 3. m a
bum—33m Mom?» “mm? Mom? amp? Eu Mmmfi i3 m3? $33 Sushi mom; x83 uumwimmm mamas M33 Maggi ammo?» owning ougém Mom? ammo?» mum>$m Mme? Mmmg 8978; Mow? x83 a??? cmog
Emma Em Sm, mom m8 «8 m8 mam. Em. mom m8 532 mum
a) A, CTL were tested t the e analog.
b) WT, CTL were tested against the vaccine wild type epitope corresponding to the vaccine analog.
c) Value indicates the net pre-vaccination SFC per 50,000 cells tested against the indicated epitope.
d) Value indicates the net SFC per 50,000 cells of vaccine positive responses.
e) -, tes SFC response did not meet criteria and vaccine response was negative.
f) Total numberof vaccine positive se against a vaccine wild type epitope or the wild type epitope corresponding
to the vaccine analog.
2015/064746
Table 1B
magmomwwz pts o o w. o N w 0 w, o o : w
. HER»? o o w a m m a v N m w w
NEWE..mE ES m9. I I Nd I I wd I I WE I I 0.0 I I mN I I I I a o
mmw \ my M M m
4 I
I I I I I I I I I I I I I
n K a a.
N m
mmflmmmunna .5). ad m
I I 3.: I I me I I I I I I I I I, I
m md ca a. a.
d ad I I 3.: I I m.» Emw mdm 0.0 I NON mumm afim ms
3 0.0 an 9am .mmm add. m w
ENwwfiMflfiE ICE mm I a a
0? I a. a.
uflfi _
m>me®nNmu DEN 5mm? w w
aging ado Wag v v
E: EQEU Neg mNfl. N m
a I
-_ Smoodmu HE. Wm Qmm mam a w
nun-Em: mam New me m.m I New NON mgmw w m rm
E! a N
m m m m m m M. «m
_ngm 393E mw we ww ww mw we ww mw
m x3? ul
ESE-m Mmmg x83 Mom; 3%th Mam?» Moog 39me goo? x00?) Seiko E? «mm; uum>IwE m3; Em; £8? among “mam; mom?
.fim SN mom Em mam mmm who :MDE cum
.: guacad mflwaocwmm
a) A, CTL were tested against the vaccine analog.
b) WT, CTL were tested against the vaccine wild type epitope corresponding to the vaccine analog.
c) Value indicates the net pre-vaccination SFC per 50,000 cells tested against the indicated epitope.
d) Value indicates the net SFC per 50,000 cells of vaccine positive responses.
6) -, indicates SFC se did not meet criteria and vaccine response was negative.
f) Total number of vaccine positive response against a vaccine wild type epitope or the wild type epitope
corresponding to the vaccine analog.
Example 2: early T memory cell involvement confirmed in phase 2
CTL Immune responses measured though cultured Elispot. The immunogenicity was
measured in Phase 2 clinical trial on cryopreserved PBMC from each test sample were thawed
in 5% human culture medium (RPMI-1640 medium with 25 mM HEPES, supplemented with
% human AB serum, 4 mM L—glutamine, 0.5 mM sodium te, 0.1 mM MEM non—
essential amino acids, 100 ug/ml streptomycin, and 100 U/ml penicillin) containing 30 ug/ml
DNAase. After centrifugation for 5 minutes at 1200 rpm, the cell pellets were resuspended in
% human culture medium and washed 2 times. For the central Memory T cell cultured
ELISPOT assay, 2x106/ml PBMC were placed in ate wells in a 48—well plates and
stimulated for 10 days with 10 ug/ml of each peptide. As a positive control, PBMC were also
stimulated with a pool of recall viral peptides. Human rIL2 (10 U/ml) was added to the peptide—
stimulated cultures at day 1, day 4 and day 7. After 10 days the stimulated cells were harvested
and plated at a tration of 5X104/we11. Assay wells also ed irradiated autologous
PBMC (1x105/well) and 10 ug/ml peptide, either the vaccine peptide, irrelevant peptide or
positive control es. Cells stimulated in vitro with each analog epitope were also tested
against the corresponding wild—type epitope.
HTL Immune responses measured though standard Elispot. For the HTL ELISPOT assay
measuring T helper responses to the PADRE e, 4x106/ml PBMC were placed in a 12
well—plate for overnight culture. Cells were then ted and placed at 2x105/well in flat—
bottom 96—well nitrocellulose plates which had been pre—coated with anti—IFN—y monoclonal
antibody (mAb) (10 ug/ml; clone 1—D1K; Mabtech). Cells plated in 6—well replicates were
stimulated with 10 ug/ml PADRE e or with an irrelevant malaria SSP2 peptide. After 20
hrs incubation at 37°C, the assay plates were washed with PBS/0.05% 20 and 100
ul/well of biotinylated anti—IFN-y mAb (2 ug/ml; clone 7—B6—1; Mabtech) was added to wells.
The plates were incubated for 2 hrs at 37°C then washed 6 times. Finally, spots from IFN—y—
secreting cells were developed by sequentially incubating wells with Vectastain ABC and 3—
amino—9—ethyl ole (AEC) solutions. Spots were counted by a computer-assisted image
analysis reader (Zeiss KS ELISPOT ).
Data analysis and acceptance criteria. Mean and standard deviation (SD) of spots in replicate
wells were calculated in all assays by transferring raw ELISPOT data from each experiment to
an Excel—based computer program. Positive vaccine-induced T-cell responses were determined
according to the criteria described below.
Acceptance Criteria for peptide-Induced specific T—cell response through central memory T
cells: The positive ia for CTL responses used in the phase 2 trial was identical to the
criteria in the phase l/2 trials which was established prospectively after analyzing the variability
of pre—treatment responses in patients against each vaccine epitope and the irrelevant HBV
control epitope. A positive peptide-induced CTL/ T memory response to a given epitope met
all of the following conditions: 1) was greater than 5 SFC above the irrelevant epitope response,
2) was greater than the mean SFC of the irrelevant epitope response, plus 2 SD, and 3) was two-
fold greater than the SFC response in the ccination sample from the same patient, plus 2
SD. The inclusion of “plus two standard deviations” was used to accommodate the assay
variability and served to increase the ia stringency.
The criteria for a positive HTL response to the HTL PADRE epitope, ed without prior
expansion of PBMC, were as follows: 1) a PADRE—specific response >5 net SFC per 2x105
cells after subtracting background; 2) for each sample tested in 6-well ates, a t-test p value
<0.05 when comparing SFC from wells stimulated with the irrelevant HLA—DR binding malaria
peptide versus wells stimulated with the PADRE peptide; and 3) a t—test p value <0.05 when
comparing SFC d by stimulation with the PADRE e in the pre— versus post—
vaccination samples. All three criteria had to be fulfilled before a HTL response was considered
to be induced by OSE-2101 ation.
RESULTS
Patient enrollment and immune monitoring criteria. A total of 64 HLA—A2+ patients with
stage IIIB, IV or recurrent NSCLC in the phase 2 trial were treated with at least one dose of
OSE—2lOl. Thirty three patients completed the initial phase of the study ting of six doses
of OSE-2101 administered at 3—week intervals and were monitored for epitopes-induced T—cell/
T memory cells responses. Results of tests from this t cohort are described below with a
particular attention on early response at W9 (after 3 injections) and W18 (after 6 injections).
The breadth of CTL/central memory T cell responses d in each of the 11 first patients
and the immunogenicity profile of individual e epitopes was very similar to the phase 1/2
trials in early-stage NSCLC and colon cancer patient with less burden disease.
Most of the CTL effector/central memory T cells responses in treated patients were d
during the initial 3 to 6—dose/18—week treatment phase and surprisingly as early as after 3 weeks
(3 doses). Preferably, the CTL/ central memory T cell responses induced during this period,
including those directed to Wild Type epitopes, have to be maintained by continued boosting
with OSE—2101 sub-cutaneous injections at 2—3 month intervals.
For the remaining 22 patients able to have a leukapheresis and an genicity testing, the
overall data indicated that immune monitoring of s against five of the more
immunogenically relevant epitopes in the product (CEA.24V9 (SEQ ID No 3), CEA.605D6
(SEQ ID No9), 69V2V9 (SEQ ID No 4), MAGE3.11215 (SEQ ID No 8) and
MAGE2. 157 (SEQ ID No 2)) was sufficient for determining T cell and central memory T cell
immunogenicity. The clinical timepoints selected for batch testing were the pre—treatment,
Week 9, Week 18 and Week 30 timepoints since most CTL/central memory T cells responses
observed among the first 11 patients who were tested indicated were already induced by Week
9 and Week 18 and some were maintained at Week 30.
The immunogenicity of OSE 2101 was similar in phase 2 advanced patients (NSCLC HLA A2
positive patients in advanced stage invasive or atic and after at least first line therapy
failure) to that observed previously in the two phase 1/ 2 trials in terms of the overall breadth
of CTL+/ central memory T cells responses induced in patients and the level of immunogenicity
of dual epitopes, thus ming the overall potency of the product in different patient
populations. Multi—epitope CTL+/ central memory T cells responses, defined by responses to
at least 3 of 5 representative immunogenic epitopes in the OSE—2101 ent, were observed
in 22 of the 33 patients (67%) who were monitored in the phase 2 trial and were achieved as
early as Week 9.
Table 2
eek 9 week 18 week 30 month 9 month 12
Antigen I
°/o °/o o/° °/o °/o
CEA CEA24-ANA 13/331 39% 10/321 31% 9/231 28% 4/71 57% 2/41 50%
CEA24-WT 12/331 36% 7/321 22% 5/231 22% 2/71 29% 1/41 0%
CEA605-ANA 9/331 27% 10/321 31% 8/231 35% 1/71 14% 1/41 25%
CEA605-WT 10/331 30% 5/321 16% 3/231 35% 1/71 14% 0/41 0%
CEA691-ANA 7/111 64% 10/111 91% 2/61 33% 3/71 43% 2/41 50%
CEA691-WT 0/111 0% 0/111 0% 0/61 0% 0/71 0% 0/41 0%
HER2 HER2.369-ANA 14/331 42% 14/321 44% 13/231 57% 2/71 29% 2/41 50%
HER2.369-WT 11/331 33% 9/321 28% 10/231 43% 2/71 29% 2/41 50%
89-WT 4/11136% 3/11127% 0/61 0% 0/71 0% 0/41 0%
MAGE MAGE2.157-WT 9/331 27% 9/321 28% 10/231 43% 4/71 57% 2/41 50%
MAGE3.112-ANA 10/331 30% 8/321 25% 10/231 43% 0/71 0% 0/41 0%
MAGE3.112-WT 10/331 30% 3/321 9% 51231 22% 0/71 0% 0/41 0%
p53 p53.139-ANA 6/111 55% 4/111 36% 3/61 50% 2/71 29% 2/41 50%
.53.139-WT 2/111 18% 1/111 9% 0/61 0% 0/71 0% 0/41 0%
p53.149-ANA 1/111 9% 0/111 0% 0/61 0% 0/71 0% 0/41 0%
o53.149-WT 0/111 0% 1/111 9% 0/61 0% 0/71 0% 0/4: 0%
ANA: Analog e, WT=Wi|d-type peptide
The first 11 ts were tested with all 9 peptide epitopes and the remaining 22 patients were
tested with 5 shaded peptide epitopes (CEA24 (SEQ ID No 3), CEA605 (SEQ ID No 9),
HER2.369 (SEQ ID No 4), MAGE2.157 (SEQ ID No 2) and MAGE3.112 (SEQ ID N08)).
Results are also shown in Figure 2.
The early W9 response was at the same level as the W18 response and the long term response
was still t at one year for patients able to receive a leukapheresis. The long term response
was ined though additive injections.
HTL responses were measured from PBMCs without an in vitro expansion step by standard
t. PBMCs were thawed, rested overnight in medium, and 2*105 PBMCs/well were
ated with 10 ug/mL Pan DR epitope (HTL) or irrelevant malaria peptide in the eron
gamma T assay.
IFN—y producing helper T-cells against PADRE were ed in 18 of 33 patients tested (55%),
without short-term in vitro expansion of PBMCs with peptide and was in the same range at
Week 9 or Week 18 demonstrating an early HTL response to the epitopes combination.
Then, a correlation between OSE-2lOl-induced PADRE HTL responses and CTL / central
memory T cell ses exists (Figure 3). The correlation coefficient for this analysis is 0.405.
Example 3: impact on Time-to-Progression (TTP) though early T memory cells
involvement
TTP results of the short e combination in advanced NSCLC after at least first line
failure (OSE 2101 phase 2 data al report). Clinically in oncology, the presence of high
levels of infiltrating memory T cells, evaluated immunohistochemically, correlated with the
absence of signs of early metastatic invasion, a less advanced pathological stage, and sed
survival in 959 specimens of resected colorectal cancer ( Pages, F et al, 2005, N Engl J Med;
353:2654-2666). The TTP is interesting as a coherent surrogate item correlated with or
memory T cells for long term and disease free survival.
Sixty-three (63) patients who were HLA—A2 and received OSE-2101 were included in the
Time-to—Progression (TTP) analysis. Twenty-eight (28) patients had documented disease
progression. Using the Kaplan—Meier estimator, the median time to progression was
determined to be 285 days.
The median of the Time to progression (TTP median) after short peptides combination OSE—
2101 was 285 days or 9.4 months (SD 86 days). This important al item is related to the
effector memory CD8+cytotoxic T cells early stimulation (TTP is defined as the time from
randomization until objective tumor progression; TTP does not include deaths. PFS is defined
as the time from randomization until objective tumor progression or death).
In the same type of advanced NSCLC population (invasive or metastatic) and after at least first
line failure (second line treatment), the Progression-Free Survival (PFS) was described at 2.2
months with a Tyrosine kinase tor as Erlotinib (Shepherd FA, et al,, 2005, N Engl J
Med.;353, 123-132.).
Two chemotherapeutic agents, xel and pemetrexed, and nib are currently approved
for the second line ent of NSCLC patients. The observed TTP in the two clinical phase 3
for docetaxel was between 10.6 and 26 weeks. From pemetrexed phase 3 results, the median
PFS was 2.9 months (M A Bareschino; J Thorac Dis 2011;3z122-l33). The median PFS was
3.5 months in the nivolumab phase 3 study in squamous NSCLC (Julie Brahmer, et al, 2015,
New England Journal ofMedicine May 31).
The unexpected early results of CTL ion through central memory T cell induction by
short epitope sequences from tumor antigens of 01 vaccine, establish the clinical utility
of epitopes combination as T specific immunotherapy against cancer and is of particular interest
for short term T memory immune responses providing long term efficacy in poor prognosis
cancer patients.
Example 4. Results on advanced cancer patients with malignant pleural effusion
(MPE).
Patients present an MPE as a complication of far-advanced cancer or as the l station
of an underlying malignancy. Common cancer types causing MPEs include lymphomas,
mesotheliomas, and carcinomas of the breast, lung, gastrointestinal tract, and ovaries. The
annual incidence of malignant pleural effusions in the United States is estimated to be greater
than 150,000 cases. al curves for more than 8,000 patients with non—small-cell lung
cancer (NSCLC) with pleural effusion (i.e, stage IIIB) from the SEER database showed that
long—terrn al is uncommon in this group. The median survival time is imately 3
months. Pleural effusion restricts ventilation and causes progressive shortness of breath by
compression of lung tissue as well as paradoxical movement of the inverted diaphragm. Pleural
deposits of tumor cause pleuritic pain.
l effusions occur more commonly in patients with advanced—stage tumors, who
frequently have ases to the brain, bone, and other organs; logic deficits;
malnutrition; debilitation; and other comorbidities. Because of these numerous clinical and
pathologic variables, it is difficult to perform prospective trials in patients with pleural
effusions. For the same reason, it is often difficult to predict a potential ent outcome or
anticipated duration of survival for the specific patient with multiple interrelated clinical
problems. Pleural effusion was the first symptom of cancer in 41% of 209 patients with
malignant pleural effusion; lung cancer in men (42%) and ovarian cancer in women (27%) were
most .
The discovery of malignant cells in pleural fluid and/or parietal pleura signifies inated
or advanced disease and a reduced life expectancy in patients with cancer.
In order to understand the role of central memory T cells discovered in phase 2 Cancer patients,
the inventors have analyzed retrospectively a sub group of patients presenting Malignant Pleural
Effusion. The study was designed to te the safety, efficacy (response and survival), and
immunogenicity of OSE—2lOl in ts with advanced NSCLC (stage IIIb and IV) who were
HLA-A2 positive. The multi-epitope combination was administered subcutaneously at a dose
of 5 mg every 3 weeks for the first 15 weeks, then every 2 months through year 1, then rly
through year 2.
A subgroup of 5 patients was presenting pleural on, 2 patients were NSCLC stage IIIB
and 3 patients were NSCLC stage IV metastatic.
According to the literature, patients with ant Pleural effusions (MPEs) present a severe
prognosis with intrathoracic and extrathoracic malignancies. Median survival after diagnosis of
an MPE is between 3— 4 months. These patients were supposed to have the worse survival time
and were thus supposed to be the first to die, y in the study.
These 5 patients with MPE were achieving, after receiving the OSE-2101 T specific
immunotherapy, a long time without ssion and also a very long term survival.
T_able4A: MPE on NSCLC Patients description
166 (site115) 104 (site121) 172 (site116) 144 (site217) te217)
Number NSCLC NSCLC NSCLC NSCLC NSCLC
mm- female female
“ 58Y 73Y
merican
NSCLC
pleural effusion
. . Pleural
plus perIcardIal . Pleural efiUSIon Pleural n Pleural effusIon
efoSIon
effusion
Chest RXth Thoracentesis
pericardiocentesis Thoracentesis (5040 CY) and 3 lines of
Chest Chest RXth Chest and 4 lines Carboplatin +
Radiotherapy (70.2 CGY) Radiotherapy of paclitaxel
Previous (10300 cGY) and 1 line of (5940 GY) chemotherapy +gemzar
treatments and 1 line of chemotherapy and 1 line of latine TRM 1
chemotherapy Carboplatin + chemotherapy +pac|itaxe| investigational
carboplatin + paclitaxel cisplatin + VP16 Tarceva drug;
paclitaxel Topotecan Tarceva
Alimta
Table 4B: Time to progression — Survival OS - number of injections
Patient 166 121 172 144 170
Number NSCLC NSCLC NSCLC NSCLC NSCLC
“20 months 19 months 18 months 6 months 26 months
Stable Disease
Time to
. lost of follow up 19 months 6 months 5 months 3 months
progresslon
at 20 months
CTL +
CTL+/ central CTL+/ central
central CTL+/ central
Memory T memory T memory T
memory T versus Not done Not done
Versus 4 versus 4
cell 4 epitopes
es epitopes
responses
In s
The time to progression achieved in such poor prognosis MPE population is sive with a
median over than 6 months (3 to 20 months). The median survival achieved is 18 months (6 to
26 months). These long term clinical results are related to an initial priming of 3 injections
though the early involvement of l memory T Cells recruiting effector cells CD8+ T cells.
In addition, 3 patients/5 are presenting strong positive responses versus 4 epitopes.
Claims (23)
1. Use of a therapeutic peptide T specific immune therapy in the manufacture of a medicament for the treatment of a cancer of an HLA-A2 (Human Leukocyte Antigen A2) positive patient, wherein the therapeutic peptide T specific immune therapy comprises the peptide aKXVAAWTLKAAa (SEQ ID No 10, with X and a respectively indicating cyclohexylalanine and d-alanine) and the peptides RLLQETELV (SEQ ID No 1), YLQLVFGIEV (SEQ ID No 2), LLTFWNPPV (SEQ ID No 3), KVFGSLAFV (SEQ ID No 4), KLBPVQLWV (SEQ ID No 5, with B indicating α-aminoisobutyric acid), SMPPPGTRV (SEQ ID No 6), IMIGHLVGV (SEQ ID No 7), KVAEIVHFL (SEQ ID No 8), and YLSGADLNL (SEQ ID No 9), and wherein said ent comprises a priming period consisting of three administrations of said therapeutic peptide T specific immune therapy, the therapeutic peptide T specific immune therapy being administered at one dose every three weeks during which a central memory T cell response is induced against one or several peptides of the therapeutic peptide T specific immunotherapy, and wherein the priming period is followed by a nance period during which the administration of said therapeutic e T ic immune therapy occurs every two to eight months.
2. The use according to of claim 1, wherein the administration of said therapeutic peptide T specific immune therapy in the maintenance period occurs every two to three months.
3. The use according to claim 1 or 2, n the administration of said therapeutic peptide T ic immune y in the maintenance period occurs every two months for one year and then every three months for one year.
4. The use according to any one of claims 1 to 3, wherein said cancer is a cancer selected from the group consisting of lung cancer such as NSCLC (non-small cell lung cancer) and small cell lung , melanoma, elioma, breast cancers, primary brain s, ovarian , uterine carcinoma, uterine corpus and/or uterine cervix carcinoma, head and neck cancer, colon or colorectal cancer, gastro-intestinal cancer, renal cancer, sarcoma, germ cell tumors, leukemia, lymphoma, testicular cancers and bladder cancers.
5. The use according to any one of claims 1 to 3, wherein said cancer is selected from the group consisting of NSCLC, colon cancer, breast cancer, ovarian cancer, and a cancer of the head and/or neck.
6. The use according to any one of claims 1 to 3, wherein said cancer is NSCLC.
7. The use according to any one of claims 1 to 6, wherein said patient suffers from an advanced or late-stage cancer.
8. The use according to any one of claims 1 to 7, wherein said t suffers from metastases.
9. The use according to any one of claims 1 to 8, wherein said t suffers from brain metastases.
10. The use according to any one of claims 1 to 9, wherein said patient has a malignant pleural effusion.
11. The use according to any one of the claims 1 to 10, wherein said patient is HTL (Helper T Cell) positive.
12. The use according to any one of the claims 1 to 11, wherein the therapeutic peptide T specific immune therapy is formulated to be d for a parenteral stration.
13. The use according to any one of the claims 1 to 11, wherein the therapeutic peptide T specific immune therapy is formulated to be adapted for a subcutaneous administration.
14. The use according to any one of the claims 1 to 13, wherein the es are emulsified in incomplete Freund’s adjuvant or the like.
15. The use according to any one of the claims 1 to 13, wherein the peptides are emulsified in Montanide ISA-51.
16. The use according to any one of the claims 1 to 15, n the doses of peptide are ranging from 0.1 to 10 mg of peptide per injection dose.
17. The use according to claim 16, wherein the total e dose for each injection is 5.0 mg.
18. The use according to any one of the claims 1 to 17, wherein said patient has already received several lines of ent prior to the treatment with the therapeutic peptide T specific immune therapy.
19. The use according to any one of the claims 1 to 18, wherein said therapeutic peptide T specific immune therapy is to be used in combination with r antitumor drug and/or with radiotherapy.
20. The use according to any one of the claims 1 to 19, wherein said therapeutic peptide T specific immune therapy is to be used in combination with a checkpoint tor.
21. The use according to claim 20, wherein the checkpoint inhibitor is a CTLA-4 and/or a PD- 1/PD-L1 tor.
22. The use according to claim 21, wherein the CTLA-4 and/or PD-1/PD-L1 inhibitor is selected from the group consisting of pembrolizumab, nivolumab, pidilizumab, BMS936559, MEDI4736, AMP-224, AMP-514 8OA and avelumab.
23. The use according to claim 1, substantially as herein described with reference to any one of the Examples and/or
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
PCT/EP2015/064746 WO2017000983A1 (en) | 2015-06-29 | 2015-06-29 | Method for inducing early t memory response with short peptides anti-tumor vaccine |
Publications (2)
Publication Number | Publication Date |
---|---|
NZ737717A NZ737717A (en) | 2021-08-27 |
NZ737717B2 true NZ737717B2 (en) | 2021-11-30 |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Brayer et al. | WT 1 vaccination in AML and MDS: A pilot trial with synthetic analog peptides | |
CN107735492A (en) | Use the method for activating T cell treatment cancer | |
Yanagimoto et al. | Immunological evaluation of personalized peptide vaccination with gemcitabine for pancreatic cancer | |
JP2022001065A (en) | Breast and ovarian cancer vaccines | |
Chism et al. | Immunogenicity of oncofetal antigens: a review | |
AU2015400687B2 (en) | Method for inducing early T memory response with short peptides anti-tumor vaccine | |
US10434157B2 (en) | Therapeutic multi-peptides T specific immune therapy for treatment of brain metastasis | |
NZ737717B2 (en) | Method for inducing early t memory response with short peptides anti-tumor vaccine | |
Dean et al. | The use of NLR, PLR and CA19. 9 as prognostic markers for locally advanced pancreatic cancer | |
Knetki-Wróblewska et al. | Rechallenge of immunotherapy in non-small cell lung cancer patients—navigating indications and evolving perspectives | |
US20240307516A1 (en) | Combination therapy for cancer | |
EA041118B1 (en) | METHOD FOR INDUCING EARLY MEMORY T-CELL RESPONSE BY ANTITUMOR VACCINE FROM SHORT PEPTIDES | |
WO2022056491A1 (en) | Treatment methods | |
WO2023081270A1 (en) | Compositions and methods for treating cancer via ptp1b inhibition | |
JP6329138B2 (en) | Peptide cocktail vaccine | |
CN116710475A (en) | SQSTM1 and its use in cancer therapy | |
Luimstra et al. | The future of cancer immunotherapy: opportunities for small molecules |