Adoptive cell transfer (ACT) of autologous tumor-infiltrating lymphocytes (TILs) to treat malignant melanoma: the dawn of a chimeric antigen receptor T (CAR-T) cell therapy from autologous donor
Corresponding Author
Luca Roncati MD, PhD
Department of Medical and Surgical Sciences, University Hospital of Modena, Modena (MO), Italy
Correspondence
Luca Roncati, md, phd
Department of Medical and Surgical Sciences
Institute of Pathology
University Hospital of Modena
Policlinico, I-41124 Modena (MO)
Italy
E-mail: [email protected];[email protected]
Search for more papers by this authorBeniamino Palmieri MD, PhD
Department of Medical and Surgical Sciences, University Hospital of Modena, Modena (MO), Italy
Search for more papers by this authorCorresponding Author
Luca Roncati MD, PhD
Department of Medical and Surgical Sciences, University Hospital of Modena, Modena (MO), Italy
Correspondence
Luca Roncati, md, phd
Department of Medical and Surgical Sciences
Institute of Pathology
University Hospital of Modena
Policlinico, I-41124 Modena (MO)
Italy
E-mail: [email protected];[email protected]
Search for more papers by this authorBeniamino Palmieri MD, PhD
Department of Medical and Surgical Sciences, University Hospital of Modena, Modena (MO), Italy
Search for more papers by this authorAbstract
Background
Tumor-infiltrating lymphocytes (TILs) are B, T-helper, and T-cytotoxic lymphocytes migrated from the blood or lymph stream toward a tumor with the aim to infiltrate and destroy it. They can be histologically graded as brisk, nonbrisk, or absent. Malignant melanoma has been the first malignancy found to be correlated with TILs status, being brisk TILs associated with better clinical outcomes. By the terminology of “adoptive cell transfer” (ACT), the medical oncology refers to the transfer of cells in a tumor-bearing patient from the same recipient or a healthy donor.
Methods
A PubMed literature search on the topic has been performed. Additional documents known to the authors and identified from the reference list of cited publications have been included.
Results
In the past, autologous TILs ACT was successfully tested for the treatment of malignant melanoma and, today, it is a standardized procedure in several centers around the world. It represents the first research step toward the bioengineered chimeric antigen receptor T (CAR-T) cell therapy from autologous donor.
Conclusions
Both autologous TILs ACT and CAR-T cell therapy from autologous donor exploit the anticancer power of targeted self-lymphocytes, but CAR-T cell technology also virtually allows treatment of those melanomas devoid of TILs or with so few cytotoxic TILs that are difficult to identify.
References
- 1Roncati L, Manenti A, Piscioli F. Upgrading the diagnostic assessment in primary pulmonary carcinoids: the tumor-infiltrating lymphocytes (TILs). Am J Clin Oncol 2018; 41: 1288–1289.
- 2Gentles AJ, Newman AM, Liu CL, et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat Med 2015; 21: 938–945.
- 3Roncati L, Barbolini G, Piacentini F, et al. Prognostic factors for breast cancer: an immunomorphological update. Pathol Oncol Res 2016; 22: 449–452.
- 4Roncati L, Manenti A, Barbolini G, Maiorana A. Deep inside of gastric signet-ring cell carcinoma. Neoplasma 2018; 65: 579–584.
- 5Bonaventura P, Shekarian T, Alcazer V, et al. Cold tumors: a therapeutic challenge for immunotherapy. Front Immunol 2019; 10: 168.
- 6Roncati L, Manenti A, Piscioli F, et al. Spontaneous regression of bronchial carcinoid is linked to a "brisk" tumor lymphocytic infiltrate. J Bronchology Interv Pulmonol 2017; 24: e26–e27.
- 7Roncati L, Manenti A, Farinetti A, Pusiol T. The association between tumor-infiltrating lymphocytes (TILs) and metastatic course in neuroendocrine neoplasms. Surgery 2016; 160: 1709.
- 8Roncati L, Manenti A, Piscioli F, et al. The immune score as a further prognostic indicator in carcinoid tumors. Chest 2017; 151: 1186.
- 9Roncati L, Manenti A, Piscioli F, et al. Immunoscoring the lymphocytic infiltration in carcinoid tumours. Histopathology 2017; 70: 1175–1177.
- 10Galon J, Mlecnik B, Bindea G, et al. Towards the introduction of the 'Immunoscore' in the classification of malignant tumours. J Pathol 2014; 232: 199–209.
- 11Tran KQ, Zhou J, Durflinger KH, et al. Minimally cultured tumor-infiltrating lymphocytes display optimal characteristics for adoptive cell therapy. J Immunother 2008; 31: 742–751.
- 12Rosenberg SA, Yang JC, Sherry RM, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res 2011; 17: 4550–4557.
- 13Piscioli F, Pusiol T, Roncati L. Thin melanoma subtyping fits well with the American Joint Committee on Cancer staging system. Melanoma Res 2016; 26: 636.
- 14Roncati L, Piscioli F, Pusiol T. Surgical outcomes reflect the histological types of cutaneous malignant melanoma. J Eur Acad Dermatol Venereol 2017; 31: e279–e280.
- 15Piscioli F, Pusiol T, Roncati L. Higher predictive value of sentinel lymph node biopsy in patients with histological subcategorization of thin melanoma. Int J Dermatol 2017; 56: e93–e94.
- 16Roncati L, Piscioli F, Pusiol T. Current controversies on sentinel node biopsy in thin and thick cutaneous melanoma. Eur J Surg Oncol 2017; 43: 506–507.
- 17Besser MJ, Shapira-Frommer R, Treves AJ, et al. Clinical responses in a phase II study using adoptive transfer of short-term cultured tumor infiltration lymphocytes in metastatic melanoma patients. Clin Cancer Res 2010; 16: 2646–2655.
- 18Radvanyi LG, Bernatchez C, Zhang M, et al. Specific lymphocyte subsets predict response to adoptive cell therapy using expanded autologous tumor-infiltrating lymphocytes in metastatic melanoma patients. Clin Cancer Res 2012; 18: 6758–6770.
- 19Ellebaek E, Iversen TZ, Junker N, et al. Adoptive cell therapy with autologous tumor infiltrating lymphocytes and low-dose interleukin-2 in metastatic melanoma patients. J Transl Med 2012; 10: 169.
- 20Lizée G, Overwijk WW, Radvanyi L, et al. Harnessing the power of the immune system to target cancer. Annu Rev Med 2013; 64: 71–90.
- 21Rosenberg S, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 2015; 348: 62–68.
- 22Gattinoni L, Finkelstein SE, Klebanoff CA, et al. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Med 2005; 202: 907–912.
- 23 US National Institute of Health, National Cancer Institute. (2012) Immunotherapy using tumor infiltrating lymphocytes for patients with metastatic human papillomavirus-associated cancers [WWW document]. https://www.clinicaltrials.gov/ct2/show/NCT01585428 [accessed on 7 March 2018].
- 24 US National Institute of Health, National Cancer Institute. (2010) Immunotherapy using tumor infiltrating lymphocytes for patients with metastatic cancer [WWW document]. https://www.clinicaltrials.gov/ct2/show/NCT01174121 [accessed on 15 May 2019].
- 25Iacopetta B, Grieu F, Amanuel B. Microsatellite instability in colorectal cancer. Asia Pac J Clin Oncol 2010; 6: 260–269.
- 26Roncati L. Microsatellite instability predicts response to anti-PD1 immunotherapy in metastatic melanoma. Acta Dermatovenerol Croat 2018; 26: 341–343.
- 27Syn NL, Teng MWL, Mok TSK, Soo RA. De-novo and acquired resistance to immune checkpoint targeting. Lancet Oncol 2017; 18: e731–e741.
- 28Morgan RA, Dudley ME, Wunderlich JR, et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 2006; 314: 126–129.
- 29Rosenbaum L. Tragedy, perseverance, and chance - the story of CAR-T therapy. N Engl J Med 2017; 377: 1313–1315.
- 30Srivastava S, Riddell SR. Engineering CAR-T cells: design concepts. Trends Immunol 2015; 36: 494–502.
- 31Gross G, Gorochov G, Waks T, Eshhar Z. Generation of effector T cells expressing chimeric T cell receptor with antibody type-specificity. Transplant Proc 1989; 21: 127–130.
- 32Sadelain M, Brentjens R, Rivière I. The basic principles of chimeric antigen receptor design. Cancer Discov 2013; 3: 388–398.
- 33Chandran SS, Klebanoff CA. T cell receptor-based cancer immunotherapy: emerging efficacy and pathways of resistance. Immunol Rev 2019; 290: 127–147.
- 34Jin C, Yu D, Hillerdal V, et al. Allogeneic lymphocyte-licensed DCs expand T cells with improved antitumor activity and resistance to oxidative stress and immunosuppressive factors. Mol Ther Methods Clin Dev 2014; 1: 14001.
- 35Jin C, Fotaki G, Ramachandran M, et al. Safe engineering of CAR T cells for adoptive cell therapy of cancer using long-term episomal gene transfer. EMBO Mol Med 2016; 8: 702–711.
- 36Jensen TI, Axelgaard E, Bak RO. Therapeutic gene editing in haematological disorders with CRISPR/Cas9. Br J Haematol 2019; 185: 821–835.
- 37Makita S, Yoshimura K, Tobinai K. Clinical development of anti-CD19 chimeric antigen receptor T-cell therapy for B-cell non-Hodgkin lymphoma. Cancer Sci 2017; 108: 1109–1118.
- 38Hartmann J, Schüßler-Lenz M, Bondanza A, Buchholz CJ. Clinical development of CAR T cells-challenges and opportunities in translating innovative treatment concepts. EMBO Mol Med 2017; 9: 1183–1197.
- 39Quintarelli C, Vera JF, Savoldo B, et al. Co-expression of cytokine and suicide genes to enhance the activity and safety of tumor-specific cytotoxic T lymphocytes. Blood 2007; 110: 2793–2802.
- 40Wilkie S, van Schalkwyk MC, Hobbs S, et al. Dual targeting of ErbB2 and MUC1 in breast cancer using chimeric antigen receptors engineered to provide complementary signaling. J Clin Immunol 2012; 32: 1059–1070.
- 41Wu CY, Roybal KT, Puchner EM, et al. Remote control of therapeutic T cells through a small molecule-gated chimeric receptor. Science 2015; 350: aab4077.
- 42Kim CH, Axup JY, Lawson BR, et al. Bispecific small molecule-antibody conjugate targeting prostate cancer. Proc Natl Acad Sci U S A 2013; 110: 17796–17801.
- 43Bonifant CL, Jackson HJ, Brentjens RJ, Curran KJ. Toxicity and management in CAR T-cell therapy. Mol Ther Oncolytics 2016; 3: 16011.
- 44Kim DW, Cho JY. Recent advances in allogeneic CAR-T cells. Biomolecules 2020; 10: 263. https://doi.org/10.3390/biom10020263
- 45Chen H, Wang F, Zhang P, et al. Management of cytokine release syndrome related to CAR-T cell therapy. Front Med 2019; 13: 610–617.
- 46Brudno JN, Kochenderfer JN. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood 2016; 127: 3321–3330.
- 47Turtle CJ, Hanafi LA, Berger C, et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J Clin Invest 2016; 126: 2123–2138.
- 48Maus MV, Haas AR, Beatty GL, et al. T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol Res 2013; 1: 26–31.
- 49 US Food & Drug Administration. (2017) FDA approval brings first gene therapy to the United States [WWW document]. https://www.fda.gov/news-events/press-announcements/fda-approval-brings-first-gene-therapy-united-states [accessed on 26 March 2018].
- 50 US Food & Drug Administration. (2017) FDA approves CAR-T cell therapy to treat adults with certain types of large B-cell lymphoma [WWW document]. https://www.fda.gov/news-events/press-announcements/fda-approves-car-t-cell-therapy-treat-adults-certain-types-large-b-cell-lymphoma [accessed on 21 March 2018].
- 51Schultz L, Mackall C. Driving CAR T cell translation forward. Sci Transl Med 2019; 11: 481.
- 52Lim WA, June CH. The principles of engineering immune cells to treat cancer. Cell 2017; 168: 724–740.
- 53Tahmasebi S, Elahi R, Esmaeilzadeh A. Solid tumors challenges and new insights of CAR T cell engineering. Stem Cell Rev Rep 2019; 15: 619–636.
- 54Xia AL, Wang XC, Lu YJ, et al. Chimeric-antigen receptor T (CAR-T) cell therapy for solid tumors: challenges and opportunities. Oncotarget 2017; 8: 90521–90531.
- 55Titov A, Valiullina A, Zmievskaya E, et al. T-cell therapy for solid tumors: lessons learned from lymphoma treatment. Cancers (Basel) 2020; 12: 125. https://doi.org/10.3390/cancers12010125
- 56Roncati L, Gasparri P, Gallo G, et al. Appendix tumor microenvironment. Adv Exp Med Biol 2020; 1226: 87–95.
- 57Li J, Li W, Huang K, et al. Chimeric antigen receptor T cell (CAR-T) immunotherapy for solid tumors: lessons learned and strategies for moving forward. J Hematol Oncol 2018; 11: 22.
- 58Newick K, O'Brien S, Moon E, Albelda SM. CAR T cell therapy for solid tumors. Annu Rev Med 2017; 68: 139–152.
- 59Watanabe K, Kuramitsu S, Posey AD Jr, June CH. Expanding the therapeutic window for CAR T cell therapy in solid tumors: the knowns and unknowns of CAR T cell biology. Front Immunol 2018; 9: 2486.
- 60Yong CSM, Dardalhon V, Devaud C, et al. CAR T-cell therapy of solid tumors. Immunol Cell Biol 2017; 95: 356–363.
- 61Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 2009; 324: 1029–1033.
- 62Springuel L, Lonez C, Alexandre B, et al. Chimeric antigen receptor-T cells for targeting solid tumors: current challenges and existing strategies. BioDrugs 2019; 33: 515–537.
- 63Sharpnack MF, Chen B, Aran D, et al. Global transcriptome analysis of RNA abundance regulation by ADAR in lung adenocarcinoma. EBioMedicine 2018; 27: 167–175.
- 64Heczey A, Louis CU, Savoldo B, et al. CAR T cells administered in combination with lymphodepletion and PD-1 inhibition to patients with neuroblastoma. Mol Ther 2017; 25: 2214–2224.
- 65Brown CE, Alizadeh D, Starr R, et al. Regression of glioblastoma after chimeric antigen receptor T-cell therapy. N Engl J Med 2016; 375: 2561–2569.
- 66Zhang C, Wang Z, Yang Z, et al. Phase I escalating-dose trial of CAR-T therapy targeting CEA+ metastatic colorectal cancers. Mol Ther 2017; 25: 1248–1258.
- 67Katz SC, Burga RA, McCormack E, et al. Phase I hepatic immunotherapy for metastases study of intra-arterial chimeric antigen receptor-modified T-cell therapy for CEA+ liver metastases. Clin Cancer Res 2015; 21: 3149–3159.
- 68Feng K, Guo Y, Dai H, et al. Chimeric antigen receptor-modified T cells for the immunotherapy of patients with EGFR-expressing advanced relapsed/refractory non-small cell lung cancer. Sci China Life Sci 2016; 59: 468–479.
- 69Ahmed N, Brawley VS, Hegde M, et al. Human epidermal growth factor receptor 2 (HER2) -specific chimeric antigen receptor-modified T cells for the immunotherapy of HER2-positive sarcoma. J Clin Oncol 2015; 33: 1688–1696.
- 70Feng K, Liu Y, Guo Y, et al. Phase I study of chimeric antigen receptor modified T cells in treating HER2-positive advanced biliary tract cancers and pancreatic cancers. Protein Cell 2018; 9: 838–847.
- 71Ahmed N, Brawley V, Hegde M, et al. HER2-specific chimeric antigen receptor-modified virus-specific T cells for progressive glioblastoma: a phase 1 dose-escalation trial. JAMA Oncol 2017; 3: 1094–1101.
- 72Simon B, Uslu U. CAR-T cell therapy in melanoma: a future success story? Exp Dermatol 2018; 27: 1315–1321.
- 73Kalialis LV, Drzewiecki KT, Klyver H. Spontaneous regression of metastases from melanoma: review of the literature. Melanoma Res 2009; 19: 275–282.
- 74Printz C. Spontaneous regression of melanoma may offer insight into cancer immunology. J Natl Cancer Inst 2001; 93: 1047–1048.
- 75Roncati L, Piscioli F, Pusiol T. The significance of regression in thin melanoma of the skin. Ir J Med Sci 2018; 187: 95–96.
- 76Boasberg PD, Hoon DS, Piro LD, et al. Enhanced survival associated with vitiligo expression during maintenance biotherapy for metastatic melanoma. J Invest Dermatol 2006; 126: 2658–2663.
- 77Le Gal FA, Avril MF, Bosq J, et al. Direct evidence to support the role of antigen-specific CD8(+) T cells in melanoma-associated vitiligo. J Invest Dermatol 2001; 117: 1464–1470.
- 78Maio M. Melanoma as a model tumour for immuno-oncology. Ann Oncol 2012; 23: 10–14.
- 79Kumar A. Malignant transformation of mature T cells after gammaretrovirus mediated transfer of nucleophosmin-anaplastic lymphoma kinase oncogene. Indian J Pathol Microbiol 2015; 58: 301–306.
- 80Milone MC, O'Doherty U. Clinical use of lentiviral vectors. Leukemia 2018; 32: 1529–1541.