WO2022098982A1 - Breast cancer specific marrow infiltrating lymphocytes and uses thereof - Google Patents
Breast cancer specific marrow infiltrating lymphocytes and uses thereof Download PDFInfo
- Publication number
- WO2022098982A1 WO2022098982A1 PCT/US2021/058221 US2021058221W WO2022098982A1 WO 2022098982 A1 WO2022098982 A1 WO 2022098982A1 US 2021058221 W US2021058221 W US 2021058221W WO 2022098982 A1 WO2022098982 A1 WO 2022098982A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- breast cancer
- hypoxic
- infiltrating lymphocytes
- subject
- activated
- Prior art date
Links
- 206010006187 Breast cancer Diseases 0.000 title claims abstract description 79
- 208000026310 Breast neoplasm Diseases 0.000 title claims abstract description 79
- 210000004698 lymphocyte Anatomy 0.000 title claims abstract description 79
- 238000000034 method Methods 0.000 claims abstract description 91
- 239000013255 MILs Substances 0.000 claims abstract description 3
- 206010021143 Hypoxia Diseases 0.000 claims description 76
- 230000001146 hypoxic effect Effects 0.000 claims description 75
- 210000004027 cell Anatomy 0.000 claims description 73
- 239000000203 mixture Substances 0.000 claims description 66
- 210000001185 bone marrow Anatomy 0.000 claims description 51
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 claims description 49
- 239000001301 oxygen Substances 0.000 claims description 49
- 229910052760 oxygen Inorganic materials 0.000 claims description 49
- 210000001744 T-lymphocyte Anatomy 0.000 claims description 28
- 108010002350 Interleukin-2 Proteins 0.000 claims description 18
- 239000011324 bead Substances 0.000 claims description 18
- 238000012258 culturing Methods 0.000 claims description 18
- 230000001225 therapeutic effect Effects 0.000 claims description 14
- 210000000481 breast Anatomy 0.000 claims description 10
- 239000008194 pharmaceutical composition Substances 0.000 claims description 9
- 238000011282 treatment Methods 0.000 claims description 8
- 208000003721 Triple Negative Breast Neoplasms Diseases 0.000 claims description 4
- 230000000735 allogeneic effect Effects 0.000 claims description 4
- 238000007918 intramuscular administration Methods 0.000 claims description 4
- 238000007912 intraperitoneal administration Methods 0.000 claims description 4
- 238000007911 parenteral administration Methods 0.000 claims description 4
- 208000022679 triple-negative breast carcinoma Diseases 0.000 claims description 3
- 208000017891 HER2 positive breast carcinoma Diseases 0.000 claims 1
- 208000027706 hormone receptor-positive breast cancer Diseases 0.000 claims 1
- 206010028980 Neoplasm Diseases 0.000 description 51
- 102000017420 CD3 protein, epsilon/gamma/delta subunit Human genes 0.000 description 46
- 201000011510 cancer Diseases 0.000 description 17
- 239000000427 antigen Substances 0.000 description 14
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 14
- 102000036639 antigens Human genes 0.000 description 13
- 108091007433 antigens Proteins 0.000 description 13
- 210000005105 peripheral blood lymphocyte Anatomy 0.000 description 12
- 102100031650 C-X-C chemokine receptor type 4 Human genes 0.000 description 10
- 101000922348 Homo sapiens C-X-C chemokine receptor type 4 Proteins 0.000 description 10
- 101001012157 Homo sapiens Receptor tyrosine-protein kinase erbB-2 Proteins 0.000 description 9
- 102100030086 Receptor tyrosine-protein kinase erbB-2 Human genes 0.000 description 9
- 239000006166 lysate Substances 0.000 description 9
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 8
- 102000004127 Cytokines Human genes 0.000 description 8
- 108090000695 Cytokines Proteins 0.000 description 8
- 239000000872 buffer Substances 0.000 description 8
- 238000002659 cell therapy Methods 0.000 description 8
- 201000010099 disease Diseases 0.000 description 8
- 108091008039 hormone receptors Proteins 0.000 description 8
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 7
- 208000035475 disorder Diseases 0.000 description 6
- 238000000684 flow cytometry Methods 0.000 description 6
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 5
- 206010035226 Plasma cell myeloma Diseases 0.000 description 5
- 210000000612 antigen-presenting cell Anatomy 0.000 description 5
- 229960004397 cyclophosphamide Drugs 0.000 description 5
- 229960000390 fludarabine Drugs 0.000 description 5
- GIUYCYHIANZCFB-FJFJXFQQSA-N fludarabine phosphate Chemical compound C1=NC=2C(N)=NC(F)=NC=2N1[C@@H]1O[C@H](COP(O)(O)=O)[C@@H](O)[C@@H]1O GIUYCYHIANZCFB-FJFJXFQQSA-N 0.000 description 5
- 210000004881 tumor cell Anatomy 0.000 description 5
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 4
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 4
- GLNADSQYFUSGOU-GPTZEZBUSA-J Trypan blue Chemical compound [Na+].[Na+].[Na+].[Na+].C1=C(S([O-])(=O)=O)C=C2C=C(S([O-])(=O)=O)C(/N=N/C3=CC=C(C=C3C)C=3C=C(C(=CC=3)\N=N\C=3C(=CC4=CC(=CC(N)=C4C=3O)S([O-])(=O)=O)S([O-])(=O)=O)C)=C(O)C2=C1N GLNADSQYFUSGOU-GPTZEZBUSA-J 0.000 description 4
- 230000004913 activation Effects 0.000 description 4
- 238000002512 chemotherapy Methods 0.000 description 4
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 4
- 230000014509 gene expression Effects 0.000 description 4
- 238000001990 intravenous administration Methods 0.000 description 4
- 229910052757 nitrogen Inorganic materials 0.000 description 4
- 230000035899 viability Effects 0.000 description 4
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Chemical compound O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 4
- 102000008096 B7-H1 Antigen Human genes 0.000 description 3
- 108010074708 B7-H1 Antigen Proteins 0.000 description 3
- WVDDGKGOMKODPV-UHFFFAOYSA-N Benzyl alcohol Chemical compound OCC1=CC=CC=C1 WVDDGKGOMKODPV-UHFFFAOYSA-N 0.000 description 3
- 108010019670 Chimeric Antigen Receptors Proteins 0.000 description 3
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 3
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 3
- 102100035716 Glycophorin-A Human genes 0.000 description 3
- 108091005250 Glycophorins Proteins 0.000 description 3
- 108090000172 Interleukin-15 Proteins 0.000 description 3
- 208000034578 Multiple myelomas Diseases 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- -1 but not limited to Proteins 0.000 description 3
- 239000007789 gas Substances 0.000 description 3
- 230000004048 modification Effects 0.000 description 3
- 238000012986 modification Methods 0.000 description 3
- 210000005087 mononuclear cell Anatomy 0.000 description 3
- 208000002154 non-small cell lung carcinoma Diseases 0.000 description 3
- 239000011780 sodium chloride Substances 0.000 description 3
- 239000000243 solution Substances 0.000 description 3
- 230000000638 stimulation Effects 0.000 description 3
- 208000029729 tumor suppressor gene on chromosome 11 Diseases 0.000 description 3
- 210000003171 tumor-infiltrating lymphocyte Anatomy 0.000 description 3
- CIWBSHSKHKDKBQ-JLAZNSOCSA-N Ascorbic acid Chemical compound OC[C@H](O)[C@H]1OC(=O)C(O)=C1O CIWBSHSKHKDKBQ-JLAZNSOCSA-N 0.000 description 2
- 102100036301 C-C chemokine receptor type 7 Human genes 0.000 description 2
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 2
- 102100027286 Fanconi anemia group C protein Human genes 0.000 description 2
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 2
- 101000716065 Homo sapiens C-C chemokine receptor type 7 Proteins 0.000 description 2
- 229940076838 Immune checkpoint inhibitor Drugs 0.000 description 2
- 108010002352 Interleukin-1 Proteins 0.000 description 2
- 108010002586 Interleukin-7 Proteins 0.000 description 2
- XOGTZOOQQBDUSI-UHFFFAOYSA-M Mesna Chemical compound [Na+].[O-]S(=O)(=O)CCS XOGTZOOQQBDUSI-UHFFFAOYSA-M 0.000 description 2
- PXIPVTKHYLBLMZ-UHFFFAOYSA-N Sodium azide Chemical compound [Na+].[N-]=[N+]=[N-] PXIPVTKHYLBLMZ-UHFFFAOYSA-N 0.000 description 2
- 108091008874 T cell receptors Proteins 0.000 description 2
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 2
- 239000003963 antioxidant agent Substances 0.000 description 2
- 235000006708 antioxidants Nutrition 0.000 description 2
- 230000009286 beneficial effect Effects 0.000 description 2
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 2
- 210000002798 bone marrow cell Anatomy 0.000 description 2
- 239000006143 cell culture medium Substances 0.000 description 2
- 239000013592 cell lysate Substances 0.000 description 2
- 239000002738 chelating agent Substances 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 239000008121 dextrose Substances 0.000 description 2
- 210000003743 erythrocyte Anatomy 0.000 description 2
- 102000015694 estrogen receptors Human genes 0.000 description 2
- 108010038795 estrogen receptors Proteins 0.000 description 2
- 208000005017 glioblastoma Diseases 0.000 description 2
- 239000012274 immune-checkpoint protein inhibitor Substances 0.000 description 2
- 238000001802 infusion Methods 0.000 description 2
- 238000001361 intraarterial administration Methods 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- LXCFILQKKLGQFO-UHFFFAOYSA-N methylparaben Chemical compound COC(=O)C1=CC=C(O)C=C1 LXCFILQKKLGQFO-UHFFFAOYSA-N 0.000 description 2
- 201000000050 myeloid neoplasm Diseases 0.000 description 2
- 238000011275 oncology therapy Methods 0.000 description 2
- 102000003998 progesterone receptors Human genes 0.000 description 2
- 108090000468 progesterone receptors Proteins 0.000 description 2
- 102000005962 receptors Human genes 0.000 description 2
- 108020003175 receptors Proteins 0.000 description 2
- 239000002904 solvent Substances 0.000 description 2
- 239000000725 suspension Substances 0.000 description 2
- 208000024891 symptom Diseases 0.000 description 2
- 238000002560 therapeutic procedure Methods 0.000 description 2
- 210000001519 tissue Anatomy 0.000 description 2
- 102100034540 Adenomatous polyposis coli protein Human genes 0.000 description 1
- 102000036365 BRCA1 Human genes 0.000 description 1
- 108700040618 BRCA1 Genes Proteins 0.000 description 1
- 108700010154 BRCA2 Genes Proteins 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 206010055113 Breast cancer metastatic Diseases 0.000 description 1
- 210000001266 CD8-positive T-lymphocyte Anatomy 0.000 description 1
- 102000000844 Cell Surface Receptors Human genes 0.000 description 1
- 108010001857 Cell Surface Receptors Proteins 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- WQZGKKKJIJFFOK-QTVWNMPRSA-N D-mannopyranose Chemical compound OC[C@H]1OC(O)[C@@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-QTVWNMPRSA-N 0.000 description 1
- 229920002307 Dextran Polymers 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- 241000283073 Equus caballus Species 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 229920001917 Ficoll Polymers 0.000 description 1
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 1
- 102000001398 Granzyme Human genes 0.000 description 1
- 108060005986 Granzyme Proteins 0.000 description 1
- 208000032843 Hemorrhage Diseases 0.000 description 1
- 102000008070 Interferon-gamma Human genes 0.000 description 1
- 108010074328 Interferon-gamma Proteins 0.000 description 1
- 108010002335 Interleukin-9 Proteins 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 206010061535 Ovarian neoplasm Diseases 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 206010061902 Pancreatic neoplasm Diseases 0.000 description 1
- 241000009328 Perro Species 0.000 description 1
- 206010060862 Prostate cancer Diseases 0.000 description 1
- 208000000236 Prostatic Neoplasms Diseases 0.000 description 1
- 208000000453 Skin Neoplasms Diseases 0.000 description 1
- DWAQJAXMDSEUJJ-UHFFFAOYSA-M Sodium bisulfite Chemical compound [Na+].OS([O-])=O DWAQJAXMDSEUJJ-UHFFFAOYSA-M 0.000 description 1
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- 108700012920 TNF Proteins 0.000 description 1
- 230000001594 aberrant effect Effects 0.000 description 1
- 150000001242 acetic acid derivatives Chemical class 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 230000006978 adaptation Effects 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- WNROFYMDJYEPJX-UHFFFAOYSA-K aluminium hydroxide Chemical compound [OH-].[OH-].[OH-].[Al+3] WNROFYMDJYEPJX-UHFFFAOYSA-K 0.000 description 1
- 150000001413 amino acids Chemical class 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000000259 anti-tumor effect Effects 0.000 description 1
- 238000002617 apheresis Methods 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 235000010323 ascorbic acid Nutrition 0.000 description 1
- 229960005070 ascorbic acid Drugs 0.000 description 1
- 239000011668 ascorbic acid Substances 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 235000019445 benzyl alcohol Nutrition 0.000 description 1
- 230000000740 bleeding effect Effects 0.000 description 1
- 239000007975 buffered saline Substances 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 150000001860 citric acid derivatives Chemical class 0.000 description 1
- 150000001875 compounds Chemical class 0.000 description 1
- 238000002591 computed tomography Methods 0.000 description 1
- 230000001143 conditioned effect Effects 0.000 description 1
- 238000003568 cytokine secretion assay Methods 0.000 description 1
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 1
- 230000007423 decrease Effects 0.000 description 1
- 230000003111 delayed effect Effects 0.000 description 1
- 238000012217 deletion Methods 0.000 description 1
- 230000037430 deletion Effects 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 1
- 230000005750 disease progression Effects 0.000 description 1
- 230000000694 effects Effects 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 238000011010 flushing procedure Methods 0.000 description 1
- 238000009472 formulation Methods 0.000 description 1
- 230000000762 glandular Effects 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- RWSXRVCMGQZWBV-WDSKDSINSA-N glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 description 1
- 235000011187 glycerol Nutrition 0.000 description 1
- 238000003306 harvesting Methods 0.000 description 1
- 201000010536 head and neck cancer Diseases 0.000 description 1
- 208000014829 head and neck neoplasm Diseases 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 238000001794 hormone therapy Methods 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 230000008073 immune recognition Effects 0.000 description 1
- 210000000987 immune system Anatomy 0.000 description 1
- 230000036039 immunity Effects 0.000 description 1
- 230000001506 immunosuppresive effect Effects 0.000 description 1
- 238000009169 immunotherapy Methods 0.000 description 1
- 230000006872 improvement Effects 0.000 description 1
- 238000011534 incubation Methods 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- 230000005764 inhibitory process Effects 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 229960003130 interferon gamma Drugs 0.000 description 1
- 108010074108 interleukin-21 Proteins 0.000 description 1
- 206010073095 invasive ductal breast carcinoma Diseases 0.000 description 1
- 201000010985 invasive ductal carcinoma Diseases 0.000 description 1
- 206010073096 invasive lobular breast carcinoma Diseases 0.000 description 1
- 230000003902 lesion Effects 0.000 description 1
- 210000000265 leukocyte Anatomy 0.000 description 1
- 210000004324 lymphatic system Anatomy 0.000 description 1
- 238000002595 magnetic resonance imaging Methods 0.000 description 1
- 238000009607 mammography Methods 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 239000002609 medium Substances 0.000 description 1
- 210000003071 memory t lymphocyte Anatomy 0.000 description 1
- 230000009245 menopause Effects 0.000 description 1
- 230000005906 menstruation Effects 0.000 description 1
- 208000037819 metastatic cancer Diseases 0.000 description 1
- 208000011575 metastatic malignant neoplasm Diseases 0.000 description 1
- 206010061289 metastatic neoplasm Diseases 0.000 description 1
- 239000004292 methyl p-hydroxybenzoate Substances 0.000 description 1
- 235000010270 methyl p-hydroxybenzoate Nutrition 0.000 description 1
- 229960002216 methylparaben Drugs 0.000 description 1
- 235000013336 milk Nutrition 0.000 description 1
- 239000008267 milk Substances 0.000 description 1
- 210000004080 milk Anatomy 0.000 description 1
- 239000003607 modifier Substances 0.000 description 1
- 230000035772 mutation Effects 0.000 description 1
- 230000001400 myeloablative effect Effects 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 230000007935 neutral effect Effects 0.000 description 1
- 230000000683 nonmetastatic effect Effects 0.000 description 1
- 239000000346 nonvolatile oil Substances 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 238000007427 paired t-test Methods 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 229960002621 pembrolizumab Drugs 0.000 description 1
- 239000000825 pharmaceutical preparation Substances 0.000 description 1
- 235000021317 phosphate Nutrition 0.000 description 1
- 239000002953 phosphate buffered saline Substances 0.000 description 1
- 150000003013 phosphoric acid derivatives Chemical class 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- 239000002504 physiological saline solution Substances 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 229920001184 polypeptide Polymers 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 102000004196 processed proteins & peptides Human genes 0.000 description 1
- 108090000765 processed proteins & peptides Proteins 0.000 description 1
- 210000002307 prostate Anatomy 0.000 description 1
- 102000004169 proteins and genes Human genes 0.000 description 1
- 108090000623 proteins and genes Proteins 0.000 description 1
- 230000005855 radiation Effects 0.000 description 1
- 238000001959 radiotherapy Methods 0.000 description 1
- 239000013643 reference control Substances 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 239000004017 serum-free culture medium Substances 0.000 description 1
- 201000000849 skin cancer Diseases 0.000 description 1
- 235000010267 sodium hydrogen sulphite Nutrition 0.000 description 1
- 238000010186 staining Methods 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 238000002626 targeted therapy Methods 0.000 description 1
- 238000010257 thawing Methods 0.000 description 1
- 239000003104 tissue culture media Substances 0.000 description 1
- 230000004222 uncontrolled growth Effects 0.000 description 1
- 230000003827 upregulation Effects 0.000 description 1
- 239000012808 vapor phase Substances 0.000 description 1
- 230000009385 viral infection Effects 0.000 description 1
- 239000008215 water for injection Substances 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K39/46
- A61K2239/46—Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
- A61K2239/49—Breast
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/461—Cellular immunotherapy characterised by the cell type used
- A61K39/4611—T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/464—Cellular immunotherapy characterised by the antigen targeted or presented
- A61K39/4643—Vertebrate antigens
- A61K39/4644—Cancer antigens
- A61K39/464499—Undefined tumor antigens, e.g. tumor lysate or antigens targeted by cells isolated from tumor
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/04—Antibacterial agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2803—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
- C07K16/2809—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2803—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
- C07K16/2818—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/74—Inducing cell proliferation
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/75—Agonist effect on antigen
Definitions
- the disclosure generally refers to marrow infiltrating lymphocytes (MILs) specific for treating breast cancer and methods of use thereof.
- MILs marrow infiltrating lymphocytes
- Breast cancer is the second most common cancer in women after skin cancer. Brest cancer can be divided into 3 clinical subtypes: hormone receptor positive, human epidermal growth factor receptor 2 (HER2) positive, and triple-negative breast cancer. Each subtype responds differently to various types of treatment. Hormone receptor positive can be treated with hormone therapies and HER2 positive can be treated with therapies that target HER2. Triple negative cancers are the hardest to treat because they lack both hormone receptors and HER2 overexpression, and so chemotherapy is the typical course of treatment. Mammography and early detection remain the best ways of improving outcomes.
- hormone receptor positive can be treated with hormone therapies
- HER2 positive can be treated with therapies that target HER2.
- Triple negative cancers are the hardest to treat because they lack both hormone receptors and HER2 overexpression, and so chemotherapy is the typical course of treatment. Mammography and early detection remain the best ways of improving outcomes.
- Treatment plans for patients diagnosed with breast cancers depend on a number of factors, including the exact location of the tumor, the stage of the cancer, and the person’s age and general health. Treatment can include surgery, radiation therapy, chemotherapy, targeted therapy, or a combination of these.
- Risk factors for breast cancer include a combination of factors, including being over 50, genetic mutations in the BRCA1 and BRCA2 genes, early menstruation or delayed menopause, having dense breasts, and a family history of breast cancer.
- MILs R Marrow infiltrating lymphocytes
- the bone marrow is a specialized niche in the immune system which is enriched for antigen experienced, central memory T cells.
- MILs have been shown to confer immunologically measurable clinical benefits in patients with multiple myeloma (See US Patent 9,687,510).
- the bone marrow microenvironment has also been shown to harbor tumor-antigen specific T cells in patients with solid tumors such as breast, pancreatic and ovarian cancers (Schmitz-Winnenthal F.H. et al., Cancer Res. 2005 (Nov 1);65(21): 10079-1008). Therefore, development of breast cancer specific MILs for use in cancer therapy, possibly in conjunction with traditional chemotherapy/radiation, represents an exciting development.
- a method for treating a subject having breast cancer with marrow infiltrating lymphocytes comprising the steps of: (a) culturing a bone marrow sample obtained from the subject having breast cancer with an anti-CD3 antibody and an anti-CD28 antibody in a hypoxic environment to produce hypoxic-activated marrow infiltrating lymphocytes; (b) culturing the hypoxic-activated marrow infiltrating lymphocytes in a normoxic environment to produce the therapeutic activated marrow infiltrating lymphocytes; and (c) administering the therapeutic activated marrow infiltrating lymphocytes to the subject having breast cancer.
- hypoxic environment has an oxygen content of about 0% to about 5% oxygen.
- lymphocytes are cultured in the presence of IL-2.
- lymphocytes are cultured in the presence of IL-2.
- culturing the hypoxic-activated marrow infiltrating lymphocytes in a normoxic environment is performed in the presence of IL-2.
- bone marrow sample is cultured in the hypoxic environment for about 24 hours.
- Also disclosed herein is an embodiment of the method as described above, wherein the bone marrow sample is cultured in the hypoxic environment for about 2 days.
- Also disclosed herein is an embodiment of the method as described above, wherein the bone marrow sample is cultured in the hypoxic environment for about 3 days.
- Also disclosed herein is an embodiment of the method as described above, wherein the bone marrow sample is cultured in the hypoxic environment for about 2 to about 5 days.
- hypoxic environment is about 1% to about 2% oxygen.
- hypoxic-activated marrow infiltrating lymphocytes are cultured in the normoxic environment for about 2 to about 14 days.
- hypoxic-activated marrow infiltrating lymphocytes are cultured in the normoxic environment for about 6 days.
- hypoxic-activated marrow infiltrating lymphocytes are cultured in the normoxic environment for about 9 days.
- step (a) Also disclosed herein is an embodiment of the method as described above, further comprising the step of removing a bone marrow sample from a subject having cancer prior to step (a).
- step (a) Also disclosed herein is an embodiment of the method as described above, wherein the anti-CD3 antibody and the anti-CD28 antibody are bound on a bead.
- breast cancer is hormone receptor positive.
- hormone receptor is an estrogen receptor and/or a progesterone receptor.
- breast cancer is HER2 positive.
- breast cancer is triple negative breast cancer.
- Also disclosed herein is a method for treating a subject having breast cancer with therapeutic activated marrow infiltrating lymphocytes, the method comprising the steps of:(a) culturing a bone marrow sample obtained from the subject having breast cancer with anti- CD3/anti-CD28 beads in a hypoxic environment of about 1% to about 2% oxygen for about 2 to about 5 days to produce hypoxic-activated marrow infiltrating lymphocytes; (b) culturing the hypoxic-activated marrow infiltrating lymphocytes in a normoxic environment of about 21% oxygen for about 2 to about 12 days in the presence of IL-2 to produce the therapeutic activated marrow infiltrating lymphocytes; and (c) administering the therapeutic activated marrow infiltrating lymphocytes to the subject having breast cancer.
- Also disclosed herein is a method of treating breast cancer in a subject, the method comprising administering a pharmaceutical composition comprising breast cancer specific marrow infiltrating lymphocyte to the subject.
- breast cancer specific marrow infiltrating lymphocyte is obtained from a subject having breast cancer.
- breast cancer specific marrow infiltrating lymphocyte is autologous to the subject being treated.
- breast cancer specific marrow infiltrating lymphocyte is allogeneic to the subject being treated.
- Also disclosed herein is an embodiment of the method as described above, wherein the marrow infiltrating lymphocyte is hypoxic activated.
- marrow infiltrating lymphocyte is hypoxic activated and normoxic activated.
- composition is administered by parenteral administration, intraperitoneal or intramuscular administration.
- Also disclosed herein is an embodiment of the method as described above, wherein about 80% to about 100% of the marrow infiltrating lymphocytes administered to the subject express CD3.
- Also disclosed herein is an embodiment of the method as described above, wherein about 85% to about 100% of the marrow infiltrating lymphocytes administered to the subject express CD3. [0038] Also disclosed herein is an embodiment of the method as described above, wherein about 90% to about 100% of the marrow infiltrating lymphocytes administered to the subject express CD3.
- Also disclosed herein is an embodiment of the method as described above, wherein the ratio of CD4 + :CD8 + T cells present in the composition or MILs administered to the subject is about 2:1.
- composition comprising a population of hypoxic-activated marrow infiltrating lymphocytes isolated from a patient with breast cancer, wherein about 75% to about 100% of the population of the hypoxic activated marrow infiltrating lymphocytes expresses CD3.
- compositions as described above wherein about 80% to about 100% of the population of the hypoxic activated marrow infiltrating lymphocytes expresses CD3.
- compositions as described above wherein about 85% to about 100% of the population of the hypoxic activated marrow infiltrating lymphocytes expresses CD3.
- compositions as described above wherein about 90% to about 100% of the population of the hypoxic activated marrow infiltrating lymphocytes expresses CD3.
- composition as described above, wherein the ratio of CD4 + :CD8 + T cells present in the composition is about 2: 1.
- compositions as described above wherein the cell population is obtainable from a bone marrow sample obtained from a subjecting having breast cancer by: (a) culturing the bone marrow sample with an anti-CD3 antibody and an anti-CD28 antibody in a hypoxic environment of about 1% to about 3% oxygen to produce activated marrow infiltrating lymphocytes; and (b) culturing the activated marrow infiltrating lymphocytes in a normoxic environment in the presence of IL-2 to produce the composition.
- the marrow infiltrating lymphocytes are breast cancer specific.
- Also disclosed herein is an embodiment of the method as described above, wherein the subject has been subjected to treatment with anti-PDl prior to obtaining the bone marrow sample.
- Figure 1 shows the fold expansion (harvested cell number/starting cell number) for each patient’s bone marrow specimen.
- Mean fold expansion ranged from 134 (prostate) to 169.8 (NSCLC) and was not significantly different across tumor types.
- Mean fold expansion for the four reference myeloma samples was 136.8.
- Figure 2 shows the mean percentage of CD3+ T cells in pre-expansion bone marrow compared to post-expansion MILs for each tumor type. Pre- and post-expansion T cells levels were not different across tumor types. Expansion significantly increased the purity of T cells from 8.8 to 59.8% (mean 29.8%) in pre-expansion bone marrow to 92 to 99.3% (mean 96.3%) in post-expansion MILs (paired t-test p ⁇ IxlO 11 ).
- the terms “comprising” (and any form of comprising, such as “comprise”, “comprises”, and “comprised”), “having” (and any form of having, such as “have” and “has”), “including” (and any form of including, such as “includes” and “include”), or “containing” (and any form of containing, such as “contains” and “contain”), are inclusive or open-ended and do not exclude additional, unrecited elements or method steps.
- beneficial or desired clinical results include, but are not limited to, alleviation of symptoms; diminishment of extent of condition, disorder or disease; stabilized (i.e., not worsening) state of condition, disorder or disease; delay in onset or slowing of condition, disorder or disease progression; amelioration of the condition, disorder or disease state or remission (whether partial or total), whether detectable or undetectable; an amelioration of at least one measurable physical parameter, not necessarily discernible by the patient; or enhancement or improvement of condition, disorder or disease.
- treatment of cancer means an activity that alleviates or ameliorates any of the primary phenomena or secondary symptoms associated with the cancer or any other condition described herein.
- the cancer that is being treated is one of the cancers recited herein.
- the term “subject” can be used interchangeably with the term “patient”.
- the subject can be a mammal, such as a dog, cat, monkey, horse, or cow, for example.
- the subject is a human.
- the subject has been diagnosed with breast cancer.
- the subject is believed to have breast cancer.
- the subject is suspected of having breast cancer.
- the term “express” as it refers to a cell surface receptor, such as, but not limited to, CD3, CD4, and CD8, can also be referred to as the cell being positive for that marker.
- a cell that expresses CD3 can also be referred to as CD3 positive (CD3 + ).
- breast cancer as used herein is defined as disease characterized by the rapid and uncontrolled growth of aberrant cells. Cancer cells can spread locally or through the bloodstream and lymphatic system to other parts of the body.
- breast cancer as used herein is defined as cancer originating from an area within the breast, or cancer on or within any area of the breast, often forming a lump or mass.
- breast cancer originates in the milk-producing ducts and is called invasive ductal carcinoma.
- breast cancer originates in the glandular tissue (lobules) and is called invasive lobular carcinoma.
- breast cancer can originate in other cells or tissue within the breast.
- the breast cancer can be hormone receptor positive.
- the hormone receptor can be an estrogen receptor or a progesterone receptor.
- the breast cancer can be positive for human epidermal growth factor receptor 2 (HER2).
- the breast cancer can contain neither hormone receptors nor HER2. These are called triple-negative breast cancers.
- Effective amount or “therapeutically effective amount” are used interchangeably herein, and refer to an amount of a compound, formulation, material, or composition, as described herein effective to achieve a particular biological result. Such results may include, but are not limited to, the inhibition of virus infection as determined by any means suitable in the art.
- MILs are a subpopulation of immune cells and are described for example in, U.S. Patent No. 9,687,510, which is hereby incorporated by reference in its entirety. MILs significantly differ from peripheral blood lymphocytes (PBLs). For example, MILs are more easily expanded, upregulate activation markers to a greater extent than PBLs, maintain more of a skewed V0 repertoire, traffic to the bone marrow, and most importantly, possess significantly greater tumor specificity.
- PBLs peripheral blood lymphocytes
- MILs can be activated, for example, by incubating them with anti-CD3/anti-CD- 28 beads and under hypoxic conditions, as described herein.
- growing MILs under hypoxic conditions is also described in U.S. Patent No. 9,687,510, and International Application No. W02016/037054, both of which are incorporated by reference herein in their entirety.
- MILs are distinct from two other forms of adoptive cellular therapy. Table 5 compares key characteristics of MILs to chimeric antigen receptor (CAR)-T and genetically engineered T cell receptor (eTCR) cell therapies.
- CAR chimeric antigen receptor
- eTCR genetically engineered T cell receptor
- CAR-T and eTCR cell therapies are dependent upon engagement of the cognate antigen on the tumor cells; selective editing or deletion of that antigen by the tumor will render CAR-T or eTCR therapies non-effective.
- polyclonal recognition of MILs minimizes the risk of generating antigen escape loss tumor variants as a mechanism of disease relapse.
- MILs marrow infiltrating lymphocytes
- CAR-T chimeric antigen receptor
- eTCR engineered T cell receptor
- HLA human leukocyte antigen
- methods to prepare MILs may include removing cells from the bone marrow, lymphocytes, and/or marrow infiltrating lymphocytes from the subject; incubating the cells in a hypoxic environment, thereby producing activated MILs.
- the subject has cancer.
- the cells can also be activated in the presence of anti- CD3/anti-CD28 antibodies and cytokines as described herein.
- Bone marrow may be collected from a patient having breast cancer that has been previously treated with a check point inhibitor.
- the checkpoint inhibitor can be an anti-PD-1 antibody, an anti-PD-Ll antibody, or a combination of these.
- the patient may have non- metastatic or metastatic disease at the time of the bone marrow removal.
- the patient may have a breast cancer in any area as previously described herein.
- the patient may have been previously treated with chemotherapy or not.
- the collected bone marrow may be frozen or immediately used, for example, to create tumor specific MILs. If the bone marrow is frozen, it is preferably thawed before incubation.
- the bone marrow may be treated to purify MILs through methods known to one of ordinary skill in the art.
- the MILs may be activated, for example, with beads, e.g., anti- CD4/CD28 beads.
- the ratio of beads to cells in the solution may vary; in some embodiments, the ratio is 3 to 1.
- the MILs may be expanded in the presence of one or more antibodies, antigens, and/or cytokines, e.g., in the absence of anti-CD3/CD28 beads.
- the cell count for the collected bone marrow may be determined, for example, to adjust the amount of beads, antibodies, antigens, and/or cytokines to be added to the MILs.
- MILs are captured using beads specifically designed to collect the cells.
- the collected MILs can be grown in a hypoxic environment for a first period of time.
- the hypoxic environment may include less than about 7% oxygen, such as less than about 7%, 6%, 5%, 4%, 3%, 2%, or 1% oxygen.
- the hypoxic environment may include about 0% oxygen to about 7% oxygen, 0% oxygen to about 6% oxygen, such as about 0% oxygen to about 5% oxygen, about 0% oxygen to about 4% oxygen, about 0% oxygen to about 3% oxygen, about 0% oxygen to about 2% oxygen, about 0% oxygen to about 1% oxygen.
- the hypoxic environment includes about 1% to about 5% oxygen.
- the hypoxic environment is about 1% to about 2% oxygen.
- the hypoxic environment is about 0.5% to about 1.5% oxygen. In some embodiments, the hypoxic environment is about 0.5% to about 2% oxygen.
- the hypoxic environment may include about 7%, 6%, 5%, 4%, 3%, 2%, 1%, or about 0% oxygen, and all fractions thereof in between these amounts.
- Incubating MILs in a hypoxic environment may include incubating the MILs, e.g., in tissue culture medium, for at least about 1 hour, such as at least about 12 hours, 18 hours, 24 hours, 30 hours, 36 hours, 42 hours, 48 hours, 60 hours, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, or even at least about 14 days.
- Incubating may include incubating the MILs for about 1 hour to about 30 days, such as about 1 day to about 20 days, about 1 day to about 14 days, or about 1 day to about 12 days.
- incubating MILs in a hypoxic environment includes incubating the MILs in a hypoxic environment for about 2 days to about 5 days.
- the method may include incubating MILs in a hypoxic environment for about 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 day, 9 days, 10 days, 11 days, 12 days, 13 days, or 14 days.
- the method includes incubating the MILs in a hypoxic environment for about 3 days.
- the method includes incubating the MILs in a hypoxic environment for about 2 days to about 4 days.
- the method includes incubating the MILs in a hypoxic environment for about 3 days to about 4 days.
- hypoxic-activated MILs are then cultured in a normoxic environment to produce the therapeutically activated marrow infiltrating lymphocytes.
- the normoxic environment may include at least about 7% oxygen.
- the normoxic environment may include about, such as about 8% oxygen to about 30% oxygen, 10% oxygen to about 30% oxygen, about 15% oxygen to about 25% oxygen, about 18% oxygen to about 24% oxygen, about 19% oxygen to about 23% oxygen, or about 20% oxygen to about 22% oxygen.
- the normoxic environment includes about 21% oxygen.
- the MILs are cultured in the presence of IL-2 or other cytokines. In some embodiments, the MILs are cultured in normoxic conditions in the presence of IL-2. In some embodiments, the other cytokines can be IL-7, IL-15, IL-9, IL-21, or any combination thereof. In some embodiments, the MILs can be cultured in cell culture medium that includes one or more cytokines, e.g., such as IL-2, IL-7, and/or IL- 15, or any suitable combination thereof.
- Illustrative examples of suitable concentrations of each cytokine or the total concentration of cytokines includes, but is not limited to, about 25 lU/mL, about 50 lU/mL, about 75 lU/mL, about 100 lU/mL, about 125 lU/mL, about 150 lU/mL, about 175 lU/mL, about 200 lU/mL, about 250 lU/mL, about 300 lU/mL, about 350 lU/mL, about 400 lU/mL, about 450 lU/mL, or about 500 lU/mL or any intervening amount.
- the cells are cultured in about 100 lU/mL of each of, or in total of, IL-2, IL-1, and/or IL-15, or any combination thereof.
- the cell culture medium includes about 250 lU/mL of each of, or in total of, IL-2, IL-1, and/or IL-15, or any combination thereof.
- Incubating MILs in a normoxic environment may include incubating the MILs, for at least about 1 hour, such as at least about 12 hours, 18 hours, 24 hours, 30 hours, 36 hours, 42 hours, 48 hours, 60 hours, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 1 1 days, 12 days, 13 days, or even at least about 14 days.
- Incubating may include incubating the MILs for about 1 hour to about 30 days, such as about 1 day to about 20 days, about 1 day to about 14 days, about 1 day to about 12 days, or about 2 days to about 12 days.
- the MILs are obtained by extracting a bone marrow sample from a subject and culturing/incubating the cells as described herein.
- the bone marrow sample is centrifuged to remove red blood cells.
- the bone marrow sample is not subject to apheresis.
- the bone marrow sample does not include PBLs or the bone marrow sample is substantially free of PBLs.
- TILs tumor infiltrating lymphocytes
- the bone marrow sample contains less than 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or less than 1% PBLs as compared to the total of MILs. In some embodiments, the sample is free of PBLs.
- the cells are also activated by culturing with antibodies to CD3 and CD28. This can be performed, for example by incubating the cells with anti-CD3/anti- CD28 beads that are commercially available or that can be made by one of skill in the art.
- the cells can then be plated in a plate, flask, or bag. Hypoxic conditions can be achieved by flushing either the hypoxic chamber or cell culture bag for 3 minutes with a 95% Nitrogen and 5% CO2 gas mixture. This can lead to, for example, 1-2% or less O2 gas in the receptacle. Examples of such beads and methods of stimulation can be found, for example, in U.S. Patent Nos.
- beads include engineered cells, such as K562 cells, that can be used to stimulate the MILs.
- engineered cells such as K562 cells
- K562 cells that can be used to stimulate the MILs.
- Such methods can be found in, for example, U.S. Patent Nos. 8,637,307 and 7,638,325, each of which is incorporated by reference in its entirety.
- Cells can also be stimulated using other methods, such as those described in U.S. Patent No. 8,383,099, which is incorporated by reference in its entirety.
- activated MILs and/or therapeutic activated MILs are administered to a subject having, or suspected of having, breast cancer.
- hypoxic-activated MILs and/or therapeutic activated MILs are produced from a bone marrow sample from a subject having or suspected of having breast cancer, then administering to the same subject to treat breast cancer.
- the MILs are allogeneic to the subject.
- the MILs can be administered in a pharmaceutical preparation or pharmaceutical composition.
- Pharmaceutical compositions including the breast cancer specific MILs may further include buffers such as neutral buffered saline, phosphate buffered saline and the like; carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelating agents such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and preservatives.
- Compositions can be formulated for parenteral administration, e.g., intravascular (intravenous or intraarterial), intraperitoneal or intramuscular administration.
- the MILs and/or compositions are administered by parenteral administration, e.g., intravascular (intravenous or intraarterial), intraperitoneal or intramuscular administration.
- parenteral administration e.g., intravascular (intravenous or intraarterial), intraperitoneal or intramuscular administration.
- the compositions can also be administered directly into the breast or into the tumor.
- the compositions are administered intravenously.
- compositions may include one or more of the following: DMSO, sterile diluents such as water for injection, saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride, fixed oils such as synthetic mono or diglycerides which may serve as the solvent or suspending medium, polyethylene glycols, glycerin, propylene glycol or other solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose.
- DMSO sterile diluents
- fixed oils such as synthetic mono or diglycerides which may serve as the solvent or suspending medium, polyethylene glycols, glycerin, propylene glycol or other solvents
- antibacterial agents such
- the subject can be pre-conditioned with cyclophosphamide with or without fludarabine.
- cyclophosphamide with or without fludarabine.
- fludarabine (30 mg/m 2 intravenous daily for 3 days)
- cyclophosphamide 300 mg/m 2 intravenous daily for 3 days starting with the first dose of fludarabine.
- the MILsTM can be administered after, e.g., 2 to 14 days after, completion of the fludarabine and cyclophosphamide.
- the cyclophosphamide is administered or 2-3 days at a dose of about 300 to about 600 mg/m 2 .
- the pharmaceutical composition that is administered includes breast cancer-specific MILs as provided for herein.
- a composition of such MILs is also provided for herein.
- the breast cancer specific MILs are hypoxic activated.
- the breast cancer-specific MILs are hypoxic activated/normoxic activated MILs.
- a breast cancer-specific MIL is a MIL that can specifically target breast cancer in a subject.
- the composition includes a population of breast cancer specific MILs that are CD3 positive.
- at least about, or at least, 40% of the MILs are CD3 positive.
- about, or at least, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 86%, 87%, 88%, or 89% of MILs are CD3 positive.
- at least, or about, 80% of the MILs are CD3 positive.
- about 40% to about 100% of the MILs are CD3 positive.
- the composition includes either a population of MILs that do not express CD3, or a population of MILs that expresses low levels of CD3, for example, relative to the expression level of MILs from the population of MILs that express CD3.
- the composition includes a population of MILs that expresses interferon gamma (“IFNy”), i.e., wherein each cell in the population of MILs that expresses IFNy is a marrow infiltrating lymphocyte that expresses IFNy, e.g., as detected by flow cytometry.
- IFNy interferon gamma
- at least about 2% of the cells in the composition may be MILs that express IFNy, or at least about 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, or even at least about 18% of the MILs express IFNy.
- about 2% to about 100% of the MILs express IFNy, such as about 2% to about 100%, about 3% to about 100%, about 4% to about 100%, about 5% to about 100%, about 6% to about 100%, about 7% to about 100%, about 8% to about 100%, about 9% to about 100%, about 10% to about 100%, about 11% to about 100%, about 12% to about 100%, about 13% to about 100%, about 14% to about 100%, about 15% to about 100%, about 16% to about 100%, about 17% to about 100%, or even about 18% to about 100% of the MILs.
- the composition includes either a population of MILs that do not express IFNy, e.g., as detected by flow cytometry, or a population of MILs that expresses low levels of IFNy, i.e., relative to the expression level of MILs from the population of MILs that express IFNy.
- the composition includes a population of MILs that expresses CXCR4.
- the MILs express CXCR4, such as at least about 98.1%, 98.2%, 98.3%, 98.4%, 98.5%, 98.6%, 98.7%, 98.8%, 98.9%, 99.0%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, or even at least about 99.7% of the MILs.
- about 98% to about 100% may be MILs that express CXCR4, such as at least about 98.1% to about 100%, about 98.2% to about 100%, about 98.3% to about 100%, about 98.4% to about 100%, about 98.5% to about 100%, about 98.6% to about 100%, about 98.7% to about 100%, about 98.8% to about 100%, about 98.9% to about 100%, about 99.0% to about 100%, about 99.1% to about 100%, about 99.2% to about 100%, about 99.3% to about 100%, about 99.4% to about 100%, about 99.5% to about 100%, about 99.6% to about 100%, or even about 99.7% to about 100% of the MILs in the composition.
- the composition includes either a population of MILs that do not express CXCR4, e.g., as detected by flow cytometry, or a population of MILs that expresses low levels of CXCR4, i.e., relative to the expression level of MILs from the population of MILs that express CXCR4.
- the composition includes a population of MILs that expresses CD4.
- the population of MILs that expresses CD4 may include a plurality of MILs that expresses CXCR4.
- the population of MILs that expresses CD4 may include a plurality of MILs that expresses 4-1BB.
- at least about 21% of the cells in the composition may be MILs from the plurality of MILs that expresses 4-1BB, such as at least about 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, or even at least about 43% of the cells in the composition.
- about 21% to about 100% of the cells in the composition may be MILs from the plurality of MILs that expresses 4- IBB, such as about 22% to about 100%, about 23% to about 100%, about 24% to about 100%, about 25% to about 100%, about 26% to about 100%, about 27% to about 100%, about 28% to about 100%, about 29% to about 100%, about 30% to about 100%, about 31% to about 100%, about 32% to about 100%, about 33% to about 100%, about 34% to about 100%, about 35% to about 100%, about 36% to about 100%, about 37% to about 100%, about 38% to about 100%, about 39% to about 100%, about 40% to about 100%, about 41% to about 100%, about 42% to about 100%, or even about 43% to about 100% of the cells in the composition.
- the composition may include a population of MILs that expresses CD8.
- the population of MILs that expresses CD8 may include a plurality of MILs that expresses CXCR4.
- the population of MILs that expresses CD8 may include a plurality of MILs that expresses 4-1BB.
- at least about 21% of the cells in the composition may be MILs from the plurality of MILs that expresses 4-1BB, such as at least about 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20% or even at least about 21% of the cells in the composition.
- about 2% to about 100% of the cells in the composition may be MILs from the plurality of MILs that expresses 4-1BB, such as about 8% to about 100%, about 9% to about 100%, about 10% to about 100%, about 11% to about 100%, about 12% to about 100%, about 13% to about 100%, about 14% to about 100%, about 15% to about 100%, about 16% to about 100%, about 17% to about 100%, about 18% to about 100%, about 19% to about 100%, about 20% to about 100%, or even about 21% to about 100% of the cells in the composition.
- MILs from the plurality of MILs that expresses 4-1BB such as about 8% to about 100%, about 9% to about 100%, about 10% to about 100%, about 11% to about 100%, about 12% to about 100%, about 13% to about 100%, about 14% to about 100%, about 15% to about 100%, about 16% to about 100%, about 17% to about 100%, about 18% to about 100%, about 19% to about 100%, about 20% to about 100%, or even about 21% to
- the composition includes a population of MILs that expresses 4-1BB.
- at least about 21% of the cells in the composition may be MILs from the population of MILs that expresses 4-1BB, such as at least about 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, or even at least about 43% of the cells in the composition.
- about 21% to 100% of the cells in the composition may be MILs from the population of MILs that expresses 4- 1BB, such as about 22% to about 100%, about 23% to about 100%, about 24% to about 100%, about 25% to about 100%, about 26% to about 100%, about 27% to about 100%, about 28% to about 100%, about 29% to about 100%, about 30% to about 100%, about 31% to about 100%, about 32% to about 100%, about 33% to about 100%, about 34% to about 100%, about 35% to about 100%, about 36% to about 100%, about 37% to about 100%, about 38% to about 100%, about 39% to about 100%, about 40% to about 100%, about 41% to about 100%, about 42% to about 100%, or even about 43% to about 100% of the cells in the composition.
- the composition includes either a population of MILs that do not express 4-1BB, e.g., as detected by flow cytometry, or a population of MILs that expresses low levels of 4- IBB, i.e., relative to the expression level of MILs from the population of MILs that express 4-1BB.
- the composition includes MILs that express CD4. In some embodiments, the composition includes MILs that express CD8. In some embodiments, the ratio of CD4 + :CD8 + MILs present in the composition is about 2:1.
- the composition may include a population of MILs that expresses CD8.
- the population of MILs that expresses CD8 may include a plurality of MILs that expresses CXCR4.
- the composition includes a population of MILs that expresses CD4.
- the population of MILs that expresses CD4 may include a plurality of MILs that expresses CXCR4.
- the MILs may express the different factors or surface receptors as described herein alone or in combination with one another.
- a MIL can be CD3+, CD4+, and/or CD8+.
- Such cells can also express IFNy.
- the cells can also be positive or negative for the various factors or receptors provided for herein.
- the methods for preventing or treating breast cancer in a subject include administering to a subject one of the compositions described herein, such as, but not limited to, breast-specific MILs as provided herein. In some embodiments, the compositions are administered as provided for herein. In some embodiments, the method includes administering to the subject a therapeutically effective amount of any one of the compositions described herein. In some embodiments, the method includes administering to the subject a therapeutically-effective amount of the breast cancer specific MILs. In some embodiments, the MILs are activated. In some embodiments, the MILs are hypoxic-activated as described herein and referenced herein.
- the MILs are cultured under hypoxic conditions followed by normoxic conditions as described and referenced herein.
- MILs are obtained or extracted from a bone marrow sample obtained from a subject having breast cancer.
- the MILs are allogeneic to the subject being treated.
- the methods include (a) culturing a bone marrow sample from a subject with an anti-CD3 antibody and an anti-CD28 antibody in a hypoxic environment of about 1% to about 3% oxygen to produce activated marrow infiltrating lymphocytes; and (b) culturing the activated marrow infiltrating lymphocytes in a normoxic environment in the presence of IL-2 to produce the composition.
- the composition can be then be administered to the subject with breast cancer.
- Example 1 Producing tumor specific MILs from subjects with solid tumors, including breast cancer.
- Bone marrow samples were collected from the iliac crest under IRB approved informed consent from patients with advanced and metastatic cancers. Samples were collected from four patients with non-small cell lung cancer (NSCLC), prostate cancer, head and neck cancer (H&N), glioblastoma (GBM) and five patients with breast cancer. Samples from four patients with multiple myeloma were used as a reference control
- Red blood cells were removed from the bone marrow using a ficoll gradient. Bone marrow mononuclear cells were frozen at a 10X10 6 cells/ml in liquid nitrogen until the time of activation and expansion.
- Previously frozen bone marrow mononuclear cells were slow thawed; and enumerated for both number and viability using a hemocytometer and trypan blue. Approximately 1 million cells were obtained for flow cytometry to determine the percentage of CD3. Briefly thawed bone marrow cells and PBLs from corresponding patients were washed 2X in FACS buffer (1XHBSS containing 2%FBS and sodium azide). Cells were extracellularly stained with Live/Dead Violet, Glycophorin A (GlyA), CD3, CD4 and CD8 for 10 minutes at room temperature. The cells were washed 2X with FACS buffer and resuspended in lOOul of FACs buffer. The cells were run on a Beckman Coulter Navios flow cytometer and analyzed with Kaluza software. The percentage of CD3 were determined as live/GlyA negative/CD3 positive.
- This percentage of CD3 was used to determine the number of anti-CD3/anti-CD28 beads to add to the culture. For each tumor type, the optimal bead:T cell ratio was determined. Beads were added to the thawed mononuclear bone marrow cells at the optimized ratio. IL2 was added at lOOU/ml and cells were plated in 96 well round bottom plates, 200pl/well, IxlO 6 total cells/ml for a total of 2X10 5 cells/well. [0096] Plates were placed in a hypoxic chamber. Oxygen was flushed out of the chamber for 2 minutes by having one valve of the chamber open and one valve connected to 95% nitrogen and 5% CO2.
- the open valve was closed and the chamber was filled with the gas mixture for 30 seconds.
- the second valve was closed and the chamber was placed in a 37°C/5%CO2 incubator for 3 days.
- the chamber was open and media containing lOOU/ml of IL-2 was added.
- Cells were maintained for the remainder of the culture in 37°C/5%CO2. Cells were evaluated at all consecutive days of culture to determine if the culture needs to be split. When the cells were split, IL-2 was replenished for a total of lOOU/ml IL2. Cells were grown from 5-10 days as needed to have enough cells for experimentation. Cells in these experiments were grown for 10 days.
- the percentage of CD3 was determined as described above. Fold expansion of CD3 was determined as (total cells x %CD3)/total number of CD3 cells at start of expansion.
- CM central memory
- Tumor specificity was assayed utilizing a cytokine-secretion assay.
- Cancer cell line lysates were used as antigen sources. Bone marrow that was autologous to the MILs product was thawed and enumerated with a hemacytometer and Trypan Blue. Cells were resuspended at lX10 6 /ml in media alone (no antigen control), media supplemented with lOOug/ml of myeloma cell line lysate (negative antigen control) or media supplemented withlOOug/ml of breast cancer cell line lysates.
- lOOul of cells were plated in a 96 well flat bottom plate and were incubated for a minimum of 30 minutes 37°C/5%CO2 to adhere antigen presenting cells (APC) to the bottom of the well and to pulse the APC with the lysates.
- APC antigen presenting cells
- MILs autologously matched to the APC were thawed and enumerated for number and viability utilizing a hemactytometer and trypan blue. Cells were then resuspended at lX10 6 /ml in serum free media. lOOpl of this suspension is added to each well of the plate containing APC pulsed with media alone, control lysate and experimental lysate.
- adoptive T cell therapy with MILs as described herein is a surprising and viable novel therapeutic approach for patients with breast cancer, which until the embodiments provided for herein was not expected to be achievable using adoptive T cell therapy.
- the results from the breast-specific MILs were surprising and unexpected.
- Example 2 Administration of MILs to breast cancer patients
- MESNA 2-mercaptoethane sulfonate sodium
- Patients may also receive pembrolizumab (200 mg) administered on Day 1 (approximately 24 hours after MILs administration) and again every 3 weeks.
- Patients may be administered MILs alone. These subjects will be followed closely for 7 days post MILs administration for safety observation.
- the MILs will be administered via a central catheter, which could either be a peripherally inserted central catheter (PICC) line or central line.
- PICC peripherally inserted central catheter
- the subject Prior to administering the activated MILs, the subject will be hydrated with 5% dextrose in water and 50% normal saline (DSW’ANS) at a rate of approximately 200 mL per hour for at least one hour.
- MILsTM will be thawed at the bedside in a 37°C ( ⁇ 2°C) water bath for approximately 90 seconds ( ⁇ 30 seconds) per bag prior to being administered on Day 0 ( ⁇ 1 day).
- Each bag will be removed from the vapor phase liquid nitrogen shipper, one at a time, placed in the waterbath and massaged until there are some small chunks of ice present.
- Each bag of MILs will be infused at a rate of approximately 10 mL per minute and rinsed with saline prior to administering the next bag of MILsTM.
- the patients will be hydrated with D5W1 NS at a rate of approximately 200 mL per hour for 2 hours.
- Administration information including, but not limited to, date and time of thawing, time of administration, and infusion time, will be recorded for each bag. Dose modification is not applicable as the entire MILsTM product will be administered on at least one day, Day 0 (+1 day).
- Patients will be evaluated for both measurable and non-measurable lesions via CT or MRI scans.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Immunology (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Organic Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Oncology (AREA)
- Epidemiology (AREA)
- Microbiology (AREA)
- Cell Biology (AREA)
- Mycology (AREA)
- Communicable Diseases (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Biochemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Molecular Biology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Genetics & Genomics (AREA)
- General Chemical & Material Sciences (AREA)
- Biophysics (AREA)
- Hospice & Palliative Care (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
Abstract
Breast cancer specific marrow infiltrating lymphocytes ("MILs") and methods for making and using the same are described.
Description
BREAST CANCER SPECIFIC MARROW INFILTRATING LYMPHOCYTES AND USES THEREOF
[0001] This application claims priority under 35 U.S.C. §119(e) to U.S. Provisional Patent Application 63/110078, filed November 5, 2020, which is hereby incorporated by reference in its entirety.
GENERAL FIELD
[0002] The disclosure generally refers to marrow infiltrating lymphocytes (MILs) specific for treating breast cancer and methods of use thereof.
BACKGROUND
[0003] Breast cancer is the second most common cancer in women after skin cancer. Brest cancer can be divided into 3 clinical subtypes: hormone receptor positive, human epidermal growth factor receptor 2 (HER2) positive, and triple-negative breast cancer. Each subtype responds differently to various types of treatment. Hormone receptor positive can be treated with hormone therapies and HER2 positive can be treated with therapies that target HER2. Triple negative cancers are the hardest to treat because they lack both hormone receptors and HER2 overexpression, and so chemotherapy is the typical course of treatment. Mammography and early detection remain the best ways of improving outcomes.
[0004] Treatment plans for patients diagnosed with breast cancers depend on a number of factors, including the exact location of the tumor, the stage of the cancer, and the person’s age and general health. Treatment can include surgery, radiation therapy, chemotherapy, targeted therapy, or a combination of these.
[0005] Risk factors for breast cancer include a combination of factors, including being over 50, genetic mutations in the BRCA1 and BRCA2 genes, early menstruation or delayed menopause, having dense breasts, and a family history of breast cancer.
[0006] Autologous cellular immunotherapy is an exciting area of research in cancer therapy, including metastatic breast cancer. The ability to eradicate measurable disease with adoptive T
cell therapy requires T cells to be appropriately activated and present in sufficient numbers, possess appreciable anti-tumor activity, home to the tumor site, effectively kill the tumor upon encounter, and persist over time.
[0007] Marrow infiltrating lymphocytes (MILsR) are the product of activating and expanding bone marrow T cells (see Noonan et al., Cancer Res. (2005) 65(5): 2026-2034). The bone marrow is a specialized niche in the immune system which is enriched for antigen experienced, central memory T cells. MILs have been shown to confer immunologically measurable clinical benefits in patients with multiple myeloma (See US Patent 9,687,510). The bone marrow microenvironment has also been shown to harbor tumor-antigen specific T cells in patients with solid tumors such as breast, pancreatic and ovarian cancers (Schmitz-Winnenthal F.H. et al., Cancer Res. 2005 (Nov 1);65(21): 10079-1008). Therefore, development of breast cancer specific MILs for use in cancer therapy, possibly in conjunction with traditional chemotherapy/radiation, represents an exciting development.
SUMMARY
[0008] Disclosed herein is a method for treating a subject having breast cancer with marrow infiltrating lymphocytes, the method comprising the steps of: (a) culturing a bone marrow sample obtained from the subject having breast cancer with an anti-CD3 antibody and an anti-CD28 antibody in a hypoxic environment to produce hypoxic-activated marrow infiltrating lymphocytes; (b) culturing the hypoxic-activated marrow infiltrating lymphocytes in a normoxic environment to produce the therapeutic activated marrow infiltrating lymphocytes; and (c) administering the therapeutic activated marrow infiltrating lymphocytes to the subject having breast cancer.
[0009] Also disclosed herein is an embodiment of the method as described above, wherein the hypoxic environment has an oxygen content of about 0% to about 5% oxygen.
[0010] Also disclosed herein is an embodiment of the method as described above, wherein the lymphocytes are cultured in the presence of IL-2.
[0011] Also disclosed herein is an embodiment of to provide the method as described above, wherein the culturing the hypoxic-activated marrow infiltrating lymphocytes in a normoxic environment is performed in the presence of IL-2.
[0012] Also disclosed herein is an embodiment of the method as described above, wherein the bone marrow sample is cultured in the hypoxic environment for about 24 hours.
[0013] Also disclosed herein is an embodiment of the method as described above, wherein the bone marrow sample is cultured in the hypoxic environment for about 2 days.
[0014] Also disclosed herein is an embodiment of the method as described above, wherein the bone marrow sample is cultured in the hypoxic environment for about 3 days.
[0015] Also disclosed herein is an embodiment of the method as described above, wherein the bone marrow sample is cultured in the hypoxic environment for about 2 to about 5 days.
[0016] Also disclosed herein is an embodiment of the method as described above, wherein the hypoxic environment is about 1% to about 2% oxygen.
[0017] Also disclosed herein is an embodiment of the method as described above, wherein the hypoxic-activated marrow infiltrating lymphocytes are cultured in the normoxic environment for about 2 to about 14 days.
[0018] Also disclosed herein is an embodiment of the method as described above, wherein the hypoxic-activated marrow infiltrating lymphocytes are cultured in the normoxic environment for about 6 days.
[0019] Also disclosed herein is an embodiment of the method as described above, wherein the hypoxic-activated marrow infiltrating lymphocytes are cultured in the normoxic environment for about 9 days.
[0020] Also disclosed herein is an embodiment of the method as described above, further comprising the step of removing a bone marrow sample from a subject having cancer prior to step (a).
[0021] Also disclosed herein is an embodiment of the method as described above, wherein the anti-CD3 antibody and the anti-CD28 antibody are bound on a bead.
[0022] Also disclosed herein is an embodiment of the method as described above, wherein the breast cancer is hormone receptor positive.
[0023] Also disclosed herein is an embodiment of the method as described above, wherein the hormone receptor is an estrogen receptor and/or a progesterone receptor.
[0024] Also disclosed herein is an embodiment of the method as described above, wherein the breast cancer is HER2 positive.
[0025] Also disclosed herein is an embodiment of the method as described above, wherein the breast cancer is triple negative breast cancer.
[0026] Also disclosed herein is a method for treating a subject having breast cancer with therapeutic activated marrow infiltrating lymphocytes, the method comprising the steps of:(a) culturing a bone marrow sample obtained from the subject having breast cancer with anti- CD3/anti-CD28 beads in a hypoxic environment of about 1% to about 2% oxygen for about 2 to about 5 days to produce hypoxic-activated marrow infiltrating lymphocytes; (b) culturing the hypoxic-activated marrow infiltrating lymphocytes in a normoxic environment of about 21% oxygen for about 2 to about 12 days in the presence of IL-2 to produce the therapeutic activated marrow infiltrating lymphocytes; and (c) administering the therapeutic activated marrow infiltrating lymphocytes to the subject having breast cancer.
[0027] Also disclosed herein is a method of treating breast cancer in a subject, the method comprising administering a pharmaceutical composition comprising breast cancer specific marrow infiltrating lymphocyte to the subject.
[0028] Also disclosed herein is an embodiment of the method as described above, wherein the breast cancer specific marrow infiltrating lymphocyte is obtained from a subject having breast cancer.
[0029] Also disclosed herein is an embodiment of the method as described above, wherein the breast cancer specific marrow infiltrating lymphocyte is autologous to the subject being treated.
[0030] Also disclosed herein is an embodiment of the method as described above, wherein the breast cancer specific marrow infiltrating lymphocyte is allogeneic to the subject being treated.
[0031] Also disclosed herein is an embodiment of the method as described above, wherein the marrow infiltrating lymphocyte is hypoxic activated.
[0032] Also disclosed herein is an embodiment of the method as described above, wherein the marrow infiltrating lymphocyte is hypoxic activated and normoxic activated.
[0033] Also disclosed herein is an embodiment of the method as described above, wherein the pharmaceutical composition is administered by parenteral administration, intraperitoneal or intramuscular administration.
[0034] Also disclosed herein is an embodiment of the method as described above, wherein the pharmaceutical composition is administered directly into the breast of the subject.
[0035] Also disclosed herein is an embodiment of the method as described above, wherein about 75% to about 100% of the marrow infiltrating lymphocytes administered to the subject express CD3.
[0036] Also disclosed herein is an embodiment of the method as described above, wherein about 80% to about 100% of the marrow infiltrating lymphocytes administered to the subject express CD3.
[0037] Also disclosed herein is an embodiment of the method as described above, wherein about 85% to about 100% of the marrow infiltrating lymphocytes administered to the subject express CD3.
[0038] Also disclosed herein is an embodiment of the method as described above, wherein about 90% to about 100% of the marrow infiltrating lymphocytes administered to the subject express CD3.
[0039] Also disclosed herein is an embodiment of the method as described above, wherein the ratio of CD4+:CD8+ T cells present in the composition or MILs administered to the subject is about 2:1.
[0040] Also disclosed herein is a composition comprising a population of hypoxic-activated marrow infiltrating lymphocytes isolated from a patient with breast cancer, wherein about 75% to about 100% of the population of the hypoxic activated marrow infiltrating lymphocytes expresses CD3.
[0041] Also disclosed herein is an embodiment of the composition as described above, wherein about 80% to about 100% of the population of the hypoxic activated marrow infiltrating lymphocytes expresses CD3.
[0042] Also disclosed herein is an embodiment of the composition as described above, wherein about 85% to about 100% of the population of the hypoxic activated marrow infiltrating lymphocytes expresses CD3.
[0043] Also disclosed herein is an embodiment of the composition as described above, wherein about 90% to about 100% of the population of the hypoxic activated marrow infiltrating lymphocytes expresses CD3.
[0044] Also disclosed herein is an embodiment of the composition as described above, wherein the ratio of CD4+:CD8+ T cells present in the composition is about 2: 1.
[0045] Also disclosed herein is an embodiment of the composition as described above, wherein the cell population is obtainable from a bone marrow sample obtained from a subjecting having breast cancer by: (a) culturing the bone marrow sample with an anti-CD3 antibody and an anti-CD28 antibody in a hypoxic environment of about 1% to about 3% oxygen to produce activated marrow infiltrating lymphocytes; and (b) culturing the activated marrow infiltrating lymphocytes in a normoxic environment in the presence of IL-2 to produce the composition.
[0046] Also disclosed herein is an embodiment of the composition as described above, wherein the marrow infiltrating lymphocytes are breast cancer specific.
[0047] Also disclosed herein is an embodiment of the method as described above, wherein the subject has been subjected to treatment with anti-PDl prior to obtaining the bone marrow sample.
BRIEF DESCRIPTION OF THE DRAWINGS
[0048] Figure 1 shows the fold expansion (harvested cell number/starting cell number) for each patient’s bone marrow specimen. Mean fold expansion ranged from 134 (prostate) to 169.8 (NSCLC) and was not significantly different across tumor types. Mean fold expansion for the four reference myeloma samples was 136.8.
[0049] Figure 2 shows the mean percentage of CD3+ T cells in pre-expansion bone marrow compared to post-expansion MILs for each tumor type. Pre- and post-expansion T cells levels were not different across tumor types. Expansion significantly increased the purity of T cells from 8.8 to 59.8% (mean 29.8%) in pre-expansion bone marrow to 92 to 99.3% (mean 96.3%) in post-expansion MILs (paired t-test p < IxlO 11).
[0050] Figure 3 shows the percentage of IFNv-producing CD3+CD4+ (left) and CD3+CD8+ (right) T cells measured following stimulation with autologous APCs pulsed with no lysates (Media), mis-matched negative control tumor cell lysates (Mis-matched Lysates) or matched tumor cell lysates (Matched Lysates) for each of the expanded MILs (red circles) and PBLs (blue squares). Tumor antigen-specific T cells were detected in all the expanded MILs (n=25) but none of the expanded PBLs. On average, 16.0% (1.58 - 42.9) of CD4+ T cells and 26.8% (5.89 - 57.1) of CD8+ T cells in MILs produced IFNy following stimulation with matched tumor lysate- derived antigens.
DETAILED DESCRIPTION OF EXEMPLARY EMBODIMENTS
[0051] As used herein and unless otherwise indicated, the term “about” is intended to mean ± 5% of the value it modifies. Thus, "about 100" means 95 to 105. Additionally, the term “about” modifies a term in a series of terms, such as “about 1, 2, 3, 4, or 5” it should be understood that the term “about” modifies each of the members of the list, such that “about 1, 2, 3, 4, or 5” can be understood to mean “about 1, about 2, about 3, about 4, or about 5.” The same is true for a list that is modified by the term “at least” or other quantifying modifier, such as, but not limited to, “less than,” “greater than,” and the like.
[0052] As used herein and in the appended claims, the singular forms “a”, “an” and “the” include plural reference unless the context clearly dictates otherwise.
[0053] As used herein, the terms “comprising” (and any form of comprising, such as “comprise”, “comprises”, and “comprised”), “having” (and any form of having, such as “have” and “has”), “including” (and any form of including, such as “includes” and “include”), or “containing” (and any form of containing, such as “contains” and “contain”), are inclusive or open-ended and do not exclude additional, unrecited elements or method steps.
[0054] As used herein, the terms “treat,” “treated,” or “treating” mean both therapeutic treatments wherein the object is to slow down (lessen) an undesired physiological condition, disorder or disease, or obtain beneficial or desired clinical results. For purposes of the embodiments described herein, beneficial or desired clinical results include, but are not limited to, alleviation of symptoms; diminishment of extent of condition, disorder or disease; stabilized (i.e., not worsening) state of condition, disorder or disease; delay in onset or slowing of condition, disorder or disease progression; amelioration of the condition, disorder or disease state or remission (whether partial or total), whether detectable or undetectable; an amelioration of at least one measurable physical parameter, not necessarily discernible by the patient; or enhancement or improvement of condition, disorder or disease. Thus, “treatment of cancer” or “treating cancer” means an activity that alleviates or ameliorates any of the primary phenomena or secondary symptoms associated with the cancer or any other condition described herein. In some embodiments, the cancer that is being treated is one of the cancers recited herein.
[0055] As used herein, the term “subject” can be used interchangeably with the term “patient”. The subject can be a mammal, such as a dog, cat, monkey, horse, or cow, for example.
In some embodiments, the subject is a human. In some embodiments, the subject has been diagnosed with breast cancer. In some embodiments, the subject is believed to have breast cancer. In some embodiments, the subject is suspected of having breast cancer.
[0056] As used herein, the term “express” as it refers to a cell surface receptor, such as, but not limited to, CD3, CD4, and CD8, can also be referred to as the cell being positive for that marker. For example, a cell that expresses CD3 can also be referred to as CD3 positive (CD3+).
[0057] The term “cancer” as used herein is defined as disease characterized by the rapid and uncontrolled growth of aberrant cells. Cancer cells can spread locally or through the bloodstream and lymphatic system to other parts of the body. The term “breast cancer” as used herein is defined as cancer originating from an area within the breast, or cancer on or within any area of the breast, often forming a lump or mass. In some embodiments, breast cancer originates in the milk-producing ducts and is called invasive ductal carcinoma. In some embodiments, breast cancer originates in the glandular tissue (lobules) and is called invasive lobular carcinoma. In some embodiments, breast cancer can originate in other cells or tissue within the breast.
[0058] The breast cancer can be hormone receptor positive. In some embodiments, the hormone receptor can be an estrogen receptor or a progesterone receptor. In some embodiments, the breast cancer can be positive for human epidermal growth factor receptor 2 (HER2). In some embodiments, the breast cancer can contain neither hormone receptors nor HER2. These are called triple-negative breast cancers.
[0059] “Effective amount” or “therapeutically effective amount” are used interchangeably herein, and refer to an amount of a compound, formulation, material, or composition, as described herein effective to achieve a particular biological result. Such results may include, but are not limited to, the inhibition of virus infection as determined by any means suitable in the art.
[0060] As used herein, “marrow infiltrating lymphocytes” or “MILs” are a subpopulation of immune cells and are described for example in, U.S. Patent No. 9,687,510, which is hereby incorporated by reference in its entirety. MILs significantly differ from peripheral blood lymphocytes (PBLs). For example, MILs are more easily expanded, upregulate activation markers to a greater extent than PBLs, maintain more of a skewed V0 repertoire, traffic to the
bone marrow, and most importantly, possess significantly greater tumor specificity. In some embodiments, MILs can be activated, for example, by incubating them with anti-CD3/anti-CD- 28 beads and under hypoxic conditions, as described herein. In some embodiments, growing MILs under hypoxic conditions is also described in U.S. Patent No. 9,687,510, and International Application No. W02016/037054, both of which are incorporated by reference herein in their entirety.
[0061] Compared to previous methods of using MILs in non-solid tumor type cancers such as multiple myeloma, use of MILs to treat solid tumor cancers require a different and distinct paradigm. Many tumors are known to exploit the PD-1/PD-L1 pathway through upregulation of PD-L1 on tumor cells and other cells to escape T cell-mediated tumor-specific immunity. Inhibiting the interaction between PD-1 and PD-L1 decreases this immunosuppressive signal, allowing tumor-specific cytotoxic T cells to access and kill the tumor cells. MILs are distinct from two other forms of adoptive cellular therapy. Table 5 compares key characteristics of MILs to chimeric antigen receptor (CAR)-T and genetically engineered T cell receptor (eTCR) cell therapies. Most critical is that the efficacy of CAR-T and eTCR cell therapies are dependent upon engagement of the cognate antigen on the tumor cells; selective editing or deletion of that antigen by the tumor will render CAR-T or eTCR therapies non-effective. In contrast, the polyclonal recognition of MILs minimizes the risk of generating antigen escape loss tumor variants as a mechanism of disease relapse.
Abbreviations: MILs = marrow infiltrating lymphocytes; CAR-T = chimeric antigen receptor; eTCR = engineered T cell receptor; HLA = human leukocyte antigen
[0062] In some embodiments, methods to prepare MILs may include removing cells from the bone marrow, lymphocytes, and/or marrow infiltrating lymphocytes from the subject;
incubating the cells in a hypoxic environment, thereby producing activated MILs. In some embodiments, the subject has cancer. The cells can also be activated in the presence of anti- CD3/anti-CD28 antibodies and cytokines as described herein.
[0063] Bone marrow may be collected from a patient having breast cancer that has been previously treated with a check point inhibitor. The checkpoint inhibitor can be an anti-PD-1 antibody, an anti-PD-Ll antibody, or a combination of these. The patient may have non- metastatic or metastatic disease at the time of the bone marrow removal. The patient may have a breast cancer in any area as previously described herein. The patient may have been previously treated with chemotherapy or not.
[0064] The collected bone marrow may be frozen or immediately used, for example, to create tumor specific MILs. If the bone marrow is frozen, it is preferably thawed before incubation. The bone marrow may be treated to purify MILs through methods known to one of ordinary skill in the art. The MILs may be activated, for example, with beads, e.g., anti- CD4/CD28 beads. The ratio of beads to cells in the solution may vary; in some embodiments, the ratio is 3 to 1. Similarly, the MILs may be expanded in the presence of one or more antibodies, antigens, and/or cytokines, e.g., in the absence of anti-CD3/CD28 beads. The cell count for the collected bone marrow may be determined, for example, to adjust the amount of beads, antibodies, antigens, and/or cytokines to be added to the MILs. In some embodiments, MILs are captured using beads specifically designed to collect the cells.
[0065] The collected MILs can be grown in a hypoxic environment for a first period of time. The hypoxic environment may include less than about 7% oxygen, such as less than about 7%, 6%, 5%, 4%, 3%, 2%, or 1% oxygen. For example, the hypoxic environment may include about 0% oxygen to about 7% oxygen, 0% oxygen to about 6% oxygen, such as about 0% oxygen to about 5% oxygen, about 0% oxygen to about 4% oxygen, about 0% oxygen to about 3% oxygen, about 0% oxygen to about 2% oxygen, about 0% oxygen to about 1% oxygen. In some embodiments, the hypoxic environment includes about 1% to about 5% oxygen. In some embodiments, the hypoxic environment is about 1% to about 2% oxygen. In some embodiments, the hypoxic environment is about 0.5% to about 1.5% oxygen. In some embodiments, the hypoxic environment is about 0.5% to about 2% oxygen. The hypoxic environment may include
about 7%, 6%, 5%, 4%, 3%, 2%, 1%, or about 0% oxygen, and all fractions thereof in between these amounts.
[0066] Incubating MILs in a hypoxic environment may include incubating the MILs, e.g., in tissue culture medium, for at least about 1 hour, such as at least about 12 hours, 18 hours, 24 hours, 30 hours, 36 hours, 42 hours, 48 hours, 60 hours, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, or even at least about 14 days. Incubating may include incubating the MILs for about 1 hour to about 30 days, such as about 1 day to about 20 days, about 1 day to about 14 days, or about 1 day to about 12 days. In some embodiments, incubating MILs in a hypoxic environment includes incubating the MILs in a hypoxic environment for about 2 days to about 5 days. The method may include incubating MILs in a hypoxic environment for about 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 day, 9 days, 10 days, 11 days, 12 days, 13 days, or 14 days. In some embodiments, the method includes incubating the MILs in a hypoxic environment for about 3 days. In some embodiments, the method includes incubating the MILs in a hypoxic environment for about 2 days to about 4 days. In some embodiments, the method includes incubating the MILs in a hypoxic environment for about 3 days to about 4 days.
[0067] In some embodiments, hypoxic-activated MILs are then cultured in a normoxic environment to produce the therapeutically activated marrow infiltrating lymphocytes. In some embodiments, the normoxic environment may include at least about 7% oxygen. In some embodiments, the normoxic environment may include about, such as about 8% oxygen to about 30% oxygen, 10% oxygen to about 30% oxygen, about 15% oxygen to about 25% oxygen, about 18% oxygen to about 24% oxygen, about 19% oxygen to about 23% oxygen, or about 20% oxygen to about 22% oxygen. In some embodiments, the normoxic environment includes about 21% oxygen.
[0068] In some embodiments, the MILs are cultured in the presence of IL-2 or other cytokines. In some embodiments, the MILs are cultured in normoxic conditions in the presence of IL-2. In some embodiments, the other cytokines can be IL-7, IL-15, IL-9, IL-21, or any combination thereof. In some embodiments, the MILs can be cultured in cell culture medium that includes one or more cytokines, e.g., such as IL-2, IL-7, and/or IL- 15, or any suitable
combination thereof. Illustrative examples of suitable concentrations of each cytokine or the total concentration of cytokines includes, but is not limited to, about 25 lU/mL, about 50 lU/mL, about 75 lU/mL, about 100 lU/mL, about 125 lU/mL, about 150 lU/mL, about 175 lU/mL, about 200 lU/mL, about 250 lU/mL, about 300 lU/mL, about 350 lU/mL, about 400 lU/mL, about 450 lU/mL, or about 500 lU/mL or any intervening amount. In some embodiments, the cells are cultured in about 100 lU/mL of each of, or in total of, IL-2, IL-1, and/or IL-15, or any combination thereof. In some embodiments, the cell culture medium includes about 250 lU/mL of each of, or in total of, IL-2, IL-1, and/or IL-15, or any combination thereof.
[0069] Incubating MILs in a normoxic environment may include incubating the MILs, for at least about 1 hour, such as at least about 12 hours, 18 hours, 24 hours, 30 hours, 36 hours, 42 hours, 48 hours, 60 hours, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 1 1 days, 12 days, 13 days, or even at least about 14 days. Incubating may include incubating the MILs for about 1 hour to about 30 days, such as about 1 day to about 20 days, about 1 day to about 14 days, about 1 day to about 12 days, or about 2 days to about 12 days.
[0070] In some embodiments, the MILs are obtained by extracting a bone marrow sample from a subject and culturing/incubating the cells as described herein. In some embodiments, the bone marrow sample is centrifuged to remove red blood cells. In some embodiments, the bone marrow sample is not subject to apheresis. In some embodiments, the bone marrow sample does not include PBLs or the bone marrow sample is substantially free of PBLs. These methods select for cells that are not the same as what have become to be known as tumor infiltrating lymphocytes ("TILs"), which have distinct limitations for use in adoptive T cell therapy. Thus, a MIL is not a TIL. In some embodiments, the bone marrow sample contains less than 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or less than 1% PBLs as compared to the total of MILs. In some embodiments, the sample is free of PBLs.
[0071] In some embodiments, the cells are also activated by culturing with antibodies to CD3 and CD28. This can be performed, for example by incubating the cells with anti-CD3/anti- CD28 beads that are commercially available or that can be made by one of skill in the art. The cells can then be plated in a plate, flask, or bag. Hypoxic conditions can be achieved by flushing either the hypoxic chamber or cell culture bag for 3 minutes with a 95% Nitrogen and 5% CO2
gas mixture. This can lead to, for example, 1-2% or less O2 gas in the receptacle. Examples of such beads and methods of stimulation can be found, for example, in U.S. Patent Nos. 6,352,694, 6,534,055, 6,692,964, 6,797,514, 6,867,041, and 6,905,874, each of which is incorporated by reference in its entirety. Alternatives to beads include engineered cells, such as K562 cells, that can be used to stimulate the MILs. Such methods can be found in, for example, U.S. Patent Nos. 8,637,307 and 7,638,325, each of which is incorporated by reference in its entirety. Cells can also be stimulated using other methods, such as those described in U.S. Patent No. 8,383,099, which is incorporated by reference in its entirety.
[0072] In some embodiments, activated MILs and/or therapeutic activated MILs are administered to a subject having, or suspected of having, breast cancer. In some embodiments, hypoxic-activated MILs and/or therapeutic activated MILs are produced from a bone marrow sample from a subject having or suspected of having breast cancer, then administering to the same subject to treat breast cancer. In some embodiments, the MILs are allogeneic to the subject.
[0073] In some embodiments, the MILs can be administered in a pharmaceutical preparation or pharmaceutical composition. Pharmaceutical compositions including the breast cancer specific MILs may further include buffers such as neutral buffered saline, phosphate buffered saline and the like; carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelating agents such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and preservatives. Compositions can be formulated for parenteral administration, e.g., intravascular (intravenous or intraarterial), intraperitoneal or intramuscular administration. In some embodiments, the MILs and/or compositions are administered by parenteral administration, e.g., intravascular (intravenous or intraarterial), intraperitoneal or intramuscular administration. The compositions can also be administered directly into the breast or into the tumor. In some embodiments, the compositions are administered intravenously.
[0074] In some embodiments, compositions, whether they be solutions, suspensions or other like form, may include one or more of the following: DMSO, sterile diluents such as water for injection, saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride, fixed oils such as synthetic mono or diglycerides which may serve as the solvent or
suspending medium, polyethylene glycols, glycerin, propylene glycol or other solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose.
[0075] In some embodiments, the subject can be pre-conditioned with cyclophosphamide with or without fludarabine. One such example is provided for in U.S. Patent No. 9,855,298, which is hereby incorporated by reference. Another non-limiting example is administering fludarabine (30 mg/m2 intravenous daily for 3 days) and cyclophosphamide (300 mg/m2 intravenous daily for 3 days starting with the first dose of fludarabine). After administration, the MILs™ can be administered after, e.g., 2 to 14 days after, completion of the fludarabine and cyclophosphamide. In some embodiments, the cyclophosphamide is administered or 2-3 days at a dose of about 300 to about 600 mg/m2.
[0076] In some embodiments, the pharmaceutical composition that is administered includes breast cancer-specific MILs as provided for herein. A composition of such MILs is also provided for herein. In some embodiments, the breast cancer specific MILs are hypoxic activated. In some embodiments, the breast cancer-specific MILs are hypoxic activated/normoxic activated MILs. A breast cancer-specific MIL is a MIL that can specifically target breast cancer in a subject.
[0077] In some embodiments, the composition includes a population of breast cancer specific MILs that are CD3 positive. In some embodiments, at least about, or at least, 40% of the MILs are CD3 positive. In some embodiments, about, or at least, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 86%, 87%, 88%, or 89% of MILs are CD3 positive. In some embodiments, at least, or about, 80% of the MILs are CD3 positive. In some embodiments, about 40% to about 100% of the MILs are CD3 positive. In some embodiments, about 45% to about 100%, about 50% to about 100%, about 55% to about 100%, about 60% to about 100%, about 65% to about 100%, about 70% to about 100%, about 75% to about 100%, about 80% to about 100%, about 85% to about 100%, about 86% to about 100%, about 87% to about 100%, about 88% to about 100%, or about 90% to about 100% of the MILs are CD3 positive (express CD3).
[0078] In some embodiments, the composition includes either a population of MILs that do not express CD3, or a population of MILs that expresses low levels of CD3, for example, relative to the expression level of MILs from the population of MILs that express CD3.
[0079] In some embodiments, the composition includes a population of MILs that expresses interferon gamma (“IFNy”), i.e., wherein each cell in the population of MILs that expresses IFNy is a marrow infiltrating lymphocyte that expresses IFNy, e.g., as detected by flow cytometry. For example, at least about 2% of the cells in the composition may be MILs that express IFNy, or at least about 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, or even at least about 18% of the MILs express IFNy. In some embodiments, about 2% to about 100% of the MILs express IFNy, such as about 2% to about 100%, about 3% to about 100%, about 4% to about 100%, about 5% to about 100%, about 6% to about 100%, about 7% to about 100%, about 8% to about 100%, about 9% to about 100%, about 10% to about 100%, about 11% to about 100%, about 12% to about 100%, about 13% to about 100%, about 14% to about 100%, about 15% to about 100%, about 16% to about 100%, about 17% to about 100%, or even about 18% to about 100% of the MILs. In some embodiments, the composition includes either a population of MILs that do not express IFNy, e.g., as detected by flow cytometry, or a population of MILs that expresses low levels of IFNy, i.e., relative to the expression level of MILs from the population of MILs that express IFNy.
[0080] In some embodiments, the composition includes a population of MILs that expresses CXCR4. For example, at least about 98% of the MILs express CXCR4, such as at least about 98.1%, 98.2%, 98.3%, 98.4%, 98.5%, 98.6%, 98.7%, 98.8%, 98.9%, 99.0%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, or even at least about 99.7% of the MILs. In some embodiments, about 98% to about 100% may be MILs that express CXCR4, such as at least about 98.1% to about 100%, about 98.2% to about 100%, about 98.3% to about 100%, about 98.4% to about 100%, about 98.5% to about 100%, about 98.6% to about 100%, about 98.7% to about 100%, about 98.8% to about 100%, about 98.9% to about 100%, about 99.0% to about 100%, about 99.1% to about 100%, about 99.2% to about 100%, about 99.3% to about 100%, about 99.4% to about 100%, about 99.5% to about 100%, about 99.6% to about 100%, or even about 99.7% to about 100% of the MILs in the composition. In some embodiments, the composition includes
either a population of MILs that do not express CXCR4, e.g., as detected by flow cytometry, or a population of MILs that expresses low levels of CXCR4, i.e., relative to the expression level of MILs from the population of MILs that express CXCR4.
[0081] In some embodiments, the composition includes a population of MILs that expresses CD4. The population of MILs that expresses CD4 may include a plurality of MILs that expresses CXCR4.
[0082] The population of MILs that expresses CD4 may include a plurality of MILs that expresses 4-1BB. For example, at least about 21% of the cells in the composition may be MILs from the plurality of MILs that expresses 4-1BB, such as at least about 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, or even at least about 43% of the cells in the composition. In some embodiments, about 21% to about 100% of the cells in the composition may be MILs from the plurality of MILs that expresses 4- IBB, such as about 22% to about 100%, about 23% to about 100%, about 24% to about 100%, about 25% to about 100%, about 26% to about 100%, about 27% to about 100%, about 28% to about 100%, about 29% to about 100%, about 30% to about 100%, about 31% to about 100%, about 32% to about 100%, about 33% to about 100%, about 34% to about 100%, about 35% to about 100%, about 36% to about 100%, about 37% to about 100%, about 38% to about 100%, about 39% to about 100%, about 40% to about 100%, about 41% to about 100%, about 42% to about 100%, or even about 43% to about 100% of the cells in the composition.
[0083] The composition may include a population of MILs that expresses CD8. The population of MILs that expresses CD8 may include a plurality of MILs that expresses CXCR4.
[0084] The population of MILs that expresses CD8 may include a plurality of MILs that expresses 4-1BB. For example, at least about 21% of the cells in the composition may be MILs from the plurality of MILs that expresses 4-1BB, such as at least about 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20% or even at least about 21% of the cells in the composition. In some embodiments, about 2% to about 100% of the cells in the composition may be MILs from the plurality of MILs that expresses 4-1BB, such as about 8% to about 100%, about 9% to about 100%, about 10% to about 100%, about 11% to about 100%, about 12% to about 100%, about 13% to about 100%, about 14% to about 100%, about 15% to about 100%,
about 16% to about 100%, about 17% to about 100%, about 18% to about 100%, about 19% to about 100%, about 20% to about 100%, or even about 21% to about 100% of the cells in the composition.
[0085] In some embodiments, the composition includes a population of MILs that expresses 4-1BB. For example, at least about 21% of the cells in the composition may be MILs from the population of MILs that expresses 4-1BB, such as at least about 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, or even at least about 43% of the cells in the composition. In some embodiments, about 21% to 100% of the cells in the composition may be MILs from the population of MILs that expresses 4- 1BB, such as about 22% to about 100%, about 23% to about 100%, about 24% to about 100%, about 25% to about 100%, about 26% to about 100%, about 27% to about 100%, about 28% to about 100%, about 29% to about 100%, about 30% to about 100%, about 31% to about 100%, about 32% to about 100%, about 33% to about 100%, about 34% to about 100%, about 35% to about 100%, about 36% to about 100%, about 37% to about 100%, about 38% to about 100%, about 39% to about 100%, about 40% to about 100%, about 41% to about 100%, about 42% to about 100%, or even about 43% to about 100% of the cells in the composition. In some embodiments, the composition includes either a population of MILs that do not express 4-1BB, e.g., as detected by flow cytometry, or a population of MILs that expresses low levels of 4- IBB, i.e., relative to the expression level of MILs from the population of MILs that express 4-1BB.
[0086] In some embodiments, the composition includes MILs that express CD4. In some embodiments, the composition includes MILs that express CD8. In some embodiments, the ratio of CD4+:CD8+ MILs present in the composition is about 2:1.
[0087] The composition may include a population of MILs that expresses CD8. The population of MILs that expresses CD8 may include a plurality of MILs that expresses CXCR4.
[0088] In some embodiments, the composition includes a population of MILs that expresses CD4. The population of MILs that expresses CD4 may include a plurality of MILs that expresses CXCR4.
[0089] The MILs may express the different factors or surface receptors as described herein alone or in combination with one another. Thus, for example, a MIL can be CD3+, CD4+, and/or CD8+. Such cells can also express IFNy. The cells can also be positive or negative for the various factors or receptors provided for herein.
[0090] In some embodiments, the methods for preventing or treating breast cancer in a subject are provided. In some embodiments, the methods include administering to a subject one of the compositions described herein, such as, but not limited to, breast-specific MILs as provided herein. In some embodiments, the compositions are administered as provided for herein. In some embodiments, the method includes administering to the subject a therapeutically effective amount of any one of the compositions described herein. In some embodiments, the method includes administering to the subject a therapeutically-effective amount of the breast cancer specific MILs. In some embodiments, the MILs are activated. In some embodiments, the MILs are hypoxic-activated as described herein and referenced herein. In some embodiments, the MILs are cultured under hypoxic conditions followed by normoxic conditions as described and referenced herein. In some embodiments, MILs are obtained or extracted from a bone marrow sample obtained from a subject having breast cancer. In some embodiments, the MILs are allogeneic to the subject being treated. In some embodiments, the methods include (a) culturing a bone marrow sample from a subject with an anti-CD3 antibody and an anti-CD28 antibody in a hypoxic environment of about 1% to about 3% oxygen to produce activated marrow infiltrating lymphocytes; and (b) culturing the activated marrow infiltrating lymphocytes in a normoxic environment in the presence of IL-2 to produce the composition. The composition can be then be administered to the subject with breast cancer.
[0091] The following examples are illustrative, but not limiting, of the compositions and methods described herein. Other suitable modifications and adaptations known to those skilled in the art are within the scope of the following embodiments.
EXAMPLES
Example 1 : Producing tumor specific MILs from subjects with solid tumors, including breast cancer.
Bone marrow mononuclear cell separation
[0092] Bone marrow samples were collected from the iliac crest under IRB approved informed consent from patients with advanced and metastatic cancers. Samples were collected from four patients with non-small cell lung cancer (NSCLC), prostate cancer, head and neck cancer (H&N), glioblastoma (GBM) and five patients with breast cancer. Samples from four patients with multiple myeloma were used as a reference control
[0093] Red blood cells were removed from the bone marrow using a ficoll gradient. Bone marrow mononuclear cells were frozen at a 10X106 cells/ml in liquid nitrogen until the time of activation and expansion.
Activation and expansion of bone marrow T cells
[0094] Previously frozen bone marrow mononuclear cells were slow thawed; and enumerated for both number and viability using a hemocytometer and trypan blue. Approximately 1 million cells were obtained for flow cytometry to determine the percentage of CD3. Briefly thawed bone marrow cells and PBLs from corresponding patients were washed 2X in FACS buffer (1XHBSS containing 2%FBS and sodium azide). Cells were extracellularly stained with Live/Dead Violet, Glycophorin A (GlyA), CD3, CD4 and CD8 for 10 minutes at room temperature. The cells were washed 2X with FACS buffer and resuspended in lOOul of FACs buffer. The cells were run on a Beckman Coulter Navios flow cytometer and analyzed with Kaluza software. The percentage of CD3 were determined as live/GlyA negative/CD3 positive.
[0095] This percentage of CD3 was used to determine the number of anti-CD3/anti-CD28 beads to add to the culture. For each tumor type, the optimal bead:T cell ratio was determined. Beads were added to the thawed mononuclear bone marrow cells at the optimized ratio. IL2 was added at lOOU/ml and cells were plated in 96 well round bottom plates, 200pl/well, IxlO6 total cells/ml for a total of 2X105cells/well.
[0096] Plates were placed in a hypoxic chamber. Oxygen was flushed out of the chamber for 2 minutes by having one valve of the chamber open and one valve connected to 95% nitrogen and 5% CO2. After 2 min the open valve was closed and the chamber was filled with the gas mixture for 30 seconds. The second valve was closed and the chamber was placed in a 37°C/5%CO2 incubator for 3 days. On the third day the chamber was open and media containing lOOU/ml of IL-2 was added.
[0097] Cells were maintained for the remainder of the culture in 37°C/5%CO2. Cells were evaluated at all consecutive days of culture to determine if the culture needs to be split. When the cells were split, IL-2 was replenished for a total of lOOU/ml IL2. Cells were grown from 5-10 days as needed to have enough cells for experimentation. Cells in these experiments were grown for 10 days.
[0098] On the final day of culture a magnet was used to remove the anti-CD3/anti-CD28 beads. The cells were washed and enumerated for number and viability using a hemacytometer and Trypan Blue.
[0099] The percentage of CD3 was determined as described above. Fold expansion of CD3 was determined as (total cells x %CD3)/total number of CD3 cells at start of expansion.
Flow cytometry on MILs
[0100] Cells were slow thawed and enumerated for number and viability. Cells were washed 2X with FACS buffer (as before) and stained extra-cellularly with Live/Dead Violet, CD3, CD4, CD8, CD45RO and CCR7 for 10 minutes at room temperature. The cells were washed 2X with FACS buffer and resuspended in lOOul of FACs buffer. The cells were run on a Beckman Coulter Navios flow cytometer and analyzed with Kaluza software. Percentage of CD8 was determined as Live/CD3+/CD8+. Percentage of CD4 was determined as Live/CD3+/CD4+.
Percentage of central memory (CM) was determined as Live/CD3+/CD45RO+/CCR7+.
Tumor specificity
[0101] Tumor specificity was assayed utilizing a cytokine-secretion assay. Cancer cell line lysates were used as antigen sources. Bone marrow that was autologous to the MILs product
was thawed and enumerated with a hemacytometer and Trypan Blue. Cells were resuspended at lX106/ml in media alone (no antigen control), media supplemented with lOOug/ml of myeloma cell line lysate (negative antigen control) or media supplemented withlOOug/ml of breast cancer cell line lysates. lOOul of cells were plated in a 96 well flat bottom plate and were incubated for a minimum of 30 minutes 37°C/5%CO2 to adhere antigen presenting cells (APC) to the bottom of the well and to pulse the APC with the lysates.
[0102] During this time MILs autologously matched to the APC were thawed and enumerated for number and viability utilizing a hemactytometer and trypan blue. Cells were then resuspended at lX106/ml in serum free media. lOOpl of this suspension is added to each well of the plate containing APC pulsed with media alone, control lysate and experimental lysate.
[0103] Cells were cultured for 5 days. Golgi plug was added on day 5 a minimum of 4 hours before harvest. Cells were harvested from the wells and stained extracellularly for CD3, CD4, and CD8 for 10 minutes at room temperature. After extra-cellular staining, cells were permeabilized and stained intracellularly for IFNy, TNFa and Granzyme B for 15 minutes at room temperature. Cells were washed 2X with perm wash and then resuspended in lOOpl of perm wash. The cells were run on a Beckman Coulter Navios flow cytometer and analyzed with Kaluza software. Tumor-specific T cells were defined as the IFNy-producing CD3+CD4+ and IFNy-producing CD3+CD8+ populations (see figure 3).
[0104] MILs were successfully expanded from each patient (n=5) having breast cancer with an average fold expansion of 154.2 (range: 70.0 to 385.7). After activation and expansion, MILs were on average 95.4% CD3+ with an approximate 2: 1 ratio of CD4+:CD8+ T cells [63.4% to 31.5%, respectively],
[0105] Tumor-specific T cells were detected in all of the expanded MILs products (n=5). On average, 25.1% of the total CD8+ T cell repertoire and 16.5% of the CD4+ T cell repertoire were tumor specific for breast cancer antigens in the MILs products. In contrast, matched PBLs expanded and activated from four patients demonstrated no measurable tumor-specific T cells (see figure 3).
[0106] MILs were present and were expanded from all breast cancer bone marrow samples tested. MILs from all patients contained functionally active tumor-specific T cells. In contrast, the corresponding PBLs failed to show any detectable tumor-specific immune recognition. As such, adoptive T cell therapy with MILs as described herein is a surprising and viable novel therapeutic approach for patients with breast cancer, which until the embodiments provided for herein was not expected to be achievable using adoptive T cell therapy. The results from the breast-specific MILs were surprising and unexpected.
Example 2: Administration of MILs to breast cancer patients
[0107] Prior to administration of MILs, patients will receive non-myeloablative lymphodepletion with cyclophosphamide (300 mg/m2/day) and fludarabine (30 mg/m2/day) from Days -5 to -3. Lymphodepletion has been shown to increase the overall efficacy of adoptive T cell therapy. 2-mercaptoethane sulfonate sodium (MESNA) may be used to minimize any bleeding in the bladder as required.
[0108] Patients may also receive pembrolizumab (200 mg) administered on Day 1 (approximately 24 hours after MILs administration) and again every 3 weeks.
[0109] Patients may be administered MILs alone. These subjects will be followed closely for 7 days post MILs administration for safety observation.
[0110] The MILs will be administered via a central catheter, which could either be a peripherally inserted central catheter (PICC) line or central line. Prior to administering the activated MILs, the subject will be hydrated with 5% dextrose in water and 50% normal saline (DSW’ANS) at a rate of approximately 200 mL per hour for at least one hour. MILs™ will be thawed at the bedside in a 37°C (± 2°C) water bath for approximately 90 seconds (± 30 seconds) per bag prior to being administered on Day 0 (±1 day). Each bag will be removed from the vapor phase liquid nitrogen shipper, one at a time, placed in the waterbath and massaged until there are some small chunks of ice present. Each bag of MILs will be infused at a rate of approximately 10 mL per minute and rinsed with saline prior to administering the next bag of MILs™.
[0111] Following MILs infusion, the patients will be hydrated with D5W1 NS at a rate of approximately 200 mL per hour for 2 hours. Administration information including, but not limited to, date and time of thawing, time of administration, and infusion time, will be recorded for each bag. Dose modification is not applicable as the entire MILs™ product will be administered on at least one day, Day 0 (+1 day).
[0112] Patients will be evaluated for both measurable and non-measurable lesions via CT or MRI scans.
[0113] This description is not limited to the particular processes, compositions, or methodologies described, as these may vary. The terminology used in the description is for the purpose of describing the particular versions or embodiments only, and it is not intended to limit the scope of the embodiments described herein. Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art. In some cases, terms with commonly understood meanings are defined herein for clarity and/or for ready reference, and the inclusion of such definitions herein should not necessarily be construed to represent a substantial difference over what is generally understood in the art. However, in case of conflict, the patent specification, including definitions, will prevail.
[0114] From the foregoing, it will be appreciated that various embodiments of the present disclosure have been described herein for purposes of illustration, and that various modification can be made without departing from the scope and spirit of the present disclosure. Accordingly, the various embodiments disclosed herein are not intended to be limiting. All references cited herein are hereby incorporated by reference.
Claims
1. A method for treating a subject having breast cancer with marrow infiltrating lymphocytes, the method comprising the steps of:
(a) culturing a bone marrow sample obtained from the subject having breast cancer with an anti-CD3 antibody and an anti-CD28 antibody in a hypoxic environment to produce hypoxic- activated marrow infiltrating lymphocytes;
(b) culturing the hypoxic-activated marrow infiltrating lymphocytes in a normoxic environment to produce the therapeutic activated marrow infiltrating lymphocytes; and
(c) administering the therapeutic activated marrow infiltrating lymphocytes to the subject having breast cancer.
2. The method of claim 1, wherein the hypoxic environment has an oxygen content of about 0% to about 5% oxygen.
3. The method of claim 1, wherein the lymphocytes are cultured in the presence of IL-2.
4. The method of claim 1, wherein the culturing the hypoxic-activated marrow infiltrating lymphocytes in a normoxic environment is performed in the presence of IL-2.
5. The method of claim 1, wherein the bone marrow sample is cultured in the hypoxic environment for about 24 hours.
6. The method of claim 1, wherein the bone marrow sample is cultured in the hypoxic environment for about 2 days.
7. The method of claim 1, wherein the bone marrow sample is cultured in the hypoxic environment for about 3 days.
8. The method of claim 1, wherein the bone marrow sample is cultured in the hypoxic environment for about 2 to about 5 days.
9. The method of claim 1, wherein the hypoxic environment is about 1% to about 2% oxygen.
10. The method of claim 1, wherein the hypoxic-activated marrow infiltrating lymphocytes are cultured in the normoxic environment for about 2 to about 14 days.
11. The method of claim 1, wherein the hypoxic-activated marrow infiltrating lymphocytes are cultured in the normoxic environment for about 6 days.
12. The method of claim 1, wherein the hypoxic-activated marrow infiltrating lymphocytes are cultured in the normoxic environment for about 9 days.
13. The method of claim 1, further comprising the step of removing said bone marrow sample from said subject having breast cancer prior to step (a).
14. The method of claim 1, wherein the anti-CD3 antibody and the anti-CD28 antibody are bound on a bead.
15. The method of claim 1, wherein the breast cancer is selected from the group consisting of hormone receptor positive breast cancer, HER2 positive breast cancer, triple-negative breast cancer, and combinations thereof.
16. A method for treating a subject having breast cancer with therapeutic activated marrow infiltrating lymphocytes, the method comprising the steps of:
(a) culturing a bone marrow sample obtained from the subject having breast cancer with anti-CD3/anti-CD28 beads in a hypoxic environment of about 1% to about 2% oxygen for about 2 to about 5 days to produce hypoxic-activated marrow infiltrating lymphocytes;
(b) culturing the hypoxic-activated marrow infiltrating lymphocytes in a normoxic environment of about 21% oxygen for about 2 to about 12 days in the presence of IL-2 to produce the therapeutic activated marrow infiltrating lymphocytes; and
(c) administering the therapeutic activated marrow infiltrating lymphocytes to the subject having breast cancer.
17. A method of treating breast cancer in a subject, the method comprising administering a pharmaceutical composition comprising breast cancer specific marrow infiltrating lymphocyte to the subject.
18. The method of claim 17, wherein the breast cancer specific marrow infiltrating lymphocyte is obtained from a subject having breast cancer.
19. The method of claim 17, wherein the breast cancer specific marrow infiltrating lymphocyte is autologous to the subject being treated.
20. The method of claim 17, wherein the breast cancer specific marrow infiltrating lymphocyte is allogeneic to the subject being treated.
21. The method of claim 17, wherein the marrow infiltrating lymphocyte is hypoxic activated.
22. The method of claim 17, wherein the marrow infiltrating lymphocyte is hypoxic activated and normoxic activated.
23. The method of claim 17, wherein the pharmaceutical composition is administered by parenteral administration, intraperitoneal administration, or intramuscular administration.
24. The method of claim 17, wherein the pharmaceutical composition is administered directly into the breast of the subject.
25. The method of any one of claims 1-24, wherein about 75% to about 100% of the marrow infiltrating lymphocytes administered to the subject express CD3.
28
26. The method of any one of claims 1-25, wherein about 80% to about 100% of the marrow infiltrating lymphocytes administered to the subject express CD3.
27. The method of any one of claims 1-26, wherein about 85% to about 100% of the marrow infiltrating lymphocytes administered to the subject express CD3.
28. The method of any one of claims 1-27, wherein about 90% to about 100% of the marrow infiltrating lymphocytes administered to the subject express CD3.
29. The method of any one of claims 1-27, wherein the ratio of CD4+:CD8+ T cells present in the composition or MILs administered to the subject is about 2: 1.
30. A composition comprising a population of hypoxic-activated marrow infiltrating lymphocytes isolated from a patient with breast cancer, wherein about 75% to about 100% of the population of the hypoxic activated marrow infiltrating lymphocytes expresses CD3.
31. The composition of claim 30, wherein about 80% to about 100% of the population of the hypoxic activated marrow infiltrating lymphocytes expresses CD3.
32. The composition of claim 30, wherein about 85% to about 100% of the population of the hypoxic activated marrow infiltrating lymphocytes expresses CD3.
33. The composition of claim 30, wherein about 90% to about 100% of the population of the hypoxic activated marrow infiltrating lymphocytes expresses CD3.
34. The composition of claim 30, wherein the ratio of CD4+:CD8+ T cells present in the composition is about 2: 1.
35. The composition of claim 30, wherein the cell population is obtained from a bone marrow sample obtained from a subject having breast cancer by:
29
(a) culturing the bone marrow sample with an anti-CD3 antibody and an anti-CD28 antibody in a hypoxic environment of about 1% to about 3% oxygen to produce activated marrow infiltrating lymphocytes; and
(b) culturing the activated marrow infiltrating lymphocytes in a normoxic environment in the presence of IL-2 to produce the composition.
36. The composition of any of claims 30-35, wherein the marrow infiltrating lymphocytes are breast cancer specific.
37. The method of claim 1 or 16, wherein the subject had been subjected to treatment with anti-PD-1 prior obtaining the bone marrow sample.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202063110078P | 2020-11-05 | 2020-11-05 | |
US63/110,078 | 2020-11-05 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2022098982A1 true WO2022098982A1 (en) | 2022-05-12 |
Family
ID=81457380
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2021/058221 WO2022098982A1 (en) | 2020-11-05 | 2021-11-05 | Breast cancer specific marrow infiltrating lymphocytes and uses thereof |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2022098982A1 (en) |
Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2020113000A1 (en) * | 2018-11-30 | 2020-06-04 | Windmil Therapeutics, Inc. | MARROW INFILTRATING LYMPHOCYTES (MILs) EXPRESSING CHIMERIC ANTIGEN RECEPTORS (CAR), METHOD OF MANUFACTURING SAME, AND METHOD OF USING IN THERAPY |
-
2021
- 2021-11-05 WO PCT/US2021/058221 patent/WO2022098982A1/en active Application Filing
Patent Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2020113000A1 (en) * | 2018-11-30 | 2020-06-04 | Windmil Therapeutics, Inc. | MARROW INFILTRATING LYMPHOCYTES (MILs) EXPRESSING CHIMERIC ANTIGEN RECEPTORS (CAR), METHOD OF MANUFACTURING SAME, AND METHOD OF USING IN THERAPY |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US20230000919A1 (en) | Activation of marrow infiltrating lymphocytes in hypoxic alternating with normoxic conditions | |
US20210000876A1 (en) | Prostate Cancer Specific Marrow Infiltrating Lymphocytes and Uses Thereof | |
CN113613673A (en) | Pharmaceutical composition for treating pancreatic cancer | |
Wood et al. | A pilot study of autologous cancer cell vaccination and cellular immunotherapy using anti-CD3 stimulated lymphocytes in patients with recurrent grade III/IV astrocytoma | |
Schwinger et al. | Feasibility of high-dose interleukin-2 in heavily pretreated pediatric cancer patients | |
Peng et al. | Treatment of subcutaneous tumor with adoptively transferred T cells | |
WO2013078392A1 (en) | Methods and compositions involving induced senescent cells for cancer treatment | |
US20210353674A1 (en) | Pharmaceutical composition for use in the treatment of pancreatic cancer | |
US20220168350A1 (en) | Lung Cancer Specific Marrow Infiltrating Lymphocytes and Uses Thereof | |
KR20070086663A (en) | Alpha thymosin peptides as cancer vaccine adjuvants | |
Aruga et al. | Induction of autologous tumor‐specific cytotoxic T cells in patients with liver cancer. Characterizations and clinical utilization | |
US20190160099A1 (en) | Pharmaceutical composition and use thereof | |
WO2022098982A1 (en) | Breast cancer specific marrow infiltrating lymphocytes and uses thereof | |
WO2019108750A1 (en) | Methods of mobilizing marrow infiltrating lymphocytes and uses thereof | |
WO2022098985A1 (en) | Glioblastoma specific marrow infiltrating lymphocytes and uses thereof | |
WO2022098977A1 (en) | Head and neck cancer specific marrow infiltrating lymphocytes and uses thereof | |
US6368593B1 (en) | Enhanced immunogenic cell populations prepared using H2 receptor antagonists | |
JP2002502880A (en) | Hapten-modified tumor cell membranes and uses thereof | |
US20240200030A1 (en) | Tumor proximal cell collection | |
Xu et al. | Radiation-based immunogenic vaccine combined with a macrophage “checkpoint inhibitor” for boosting innate and adaptive immunity against metastatic colon cancers | |
US20230100744A1 (en) | Methods for enriching marrow infiltrating lymphocytes ("mils"), compositions containing enriched mils, and methods of using enriched mils | |
JP6993812B2 (en) | A method for culturing lymphocytes with improved cytotoxic activity and a cell immunotherapeutic agent containing lymphocytes with improved cytotoxic activity obtained by the method. | |
Franzolin et al. | PlexinB1 inactivation reprograms immune cells in the tumor microenvironment, inhibiting breast cancer growth and metastatic dissemination | |
RU2645464C1 (en) | Immunotherapy of breast cancer using antigen-activated dendritic cells | |
CN113769096A (en) | Medical application of 6-phosphoglucose dehydrogenase inhibitor |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 21890135 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 21890135 Country of ref document: EP Kind code of ref document: A1 |