TIL Therapy as a Personalized Treatment Strategy for NSCLC – Targeted Oncology

Posted: September 16, 2022 at 2:35 am

Advancements in immune checkpoint inhibitors (ICIs) have revolutionized oncology therapy.1 Several ICIs targeting PD-1 or PD-L1 are available for the treatment of advanced nonsmall cell lung cancer (NSCLC).1,2 However, there remains a need for alternative treatments due to ICI resistance, failure to respond to therapy, or disease relapse.2-5 Even when ICIs are used in combination with chemotherapy, patients may experience cancer progression within 12 months.6 Oncology providers should identify opportunities for clinical trials and investigational strategies that provide options for patients with advanced NSCLC beyond ICIs and biomarker-directed therapies.

Adoptive cell therapy (ACT) is a type of immunotherapy where an individuals immune cells are harvested and expanded to help elicit a tumor-specific, cell-mediated response against cancer cells; it includes chimeric antigen receptor (CAR) T-cell therapy (CAR-T), engineered T cell receptor (TCR)-based T cell (TCR-T) immunotherapy and tumor-infiltrating lymphocytes (TILs).7 The first promising results evaluating the use of autologous (self) TILs in patients with metastatic melanoma were published in 1988, and they sparked further research.7,8

Endogenous TILs

Endogenous TILs are composed of T cells isolated from tumor tissue that can recognize tumor-specific antigens to target and attack cancer cells.9,10 In most cancers, immune infiltrate includes various macrophage subtypes and several different types of T lymphocytes.11 Helper T lymphocytes and cytotoxic T lymphocytes (CTLs) play an important role in identifying cancer cells and arresting their growth.

During tumorigenesis, genetic instability can lead to somatic mutations producing new proteins, or neoantigens, in cancer cells. Neoantigens expressed only in tumor cells are referred to as tumor-specific antigens (TSAs). All T cells, including CTLs, express a unique T-cell receptor (TCR) specific to a single TSA. Major histocompatibility complex molecules present TSAs on the tumor cell surface, which are recognized upon TCR binding. Once tumor cells are recognized as non-self, T-cell activation occurs.12,13 CTLs release cytotoxic granules, which fuse with the target cell membrane. Granulysin and perforin create pores in the cell membrane, allowing granzymes to be released into the cytoplasm. Granzymes then initiate a caspase cascade leading to apoptosis.1,14,15 However, tumor cells can initiate adaptive mechanisms to evade CTL activity, including the production of immunosuppressive cytokines that can impede the antitumor immune response.9,16 Therefore, methods to overcome immune evasion and improve upon TIL-mediated tumor cell destruction have been explored.

Development and Potential Utility of TILs for Treatment of Solid Tumors

As noted above, endogenous TILs possess TCRs with the ability to recognize and destroy tumor cells. Removing TILs from the immunosuppressive tumor environment through tumor excision allows for ex vivo assessment of antitumor activity. Once highly active TILs are identified, they are rapidly expanded to produce billions of activated, tumor-specific T cells, which are then infused back to the host to target and destroy tumor cells (Figure).13 This approach has potential utility for treating a variety of solid tumors, including NSCLC.7,17-19

Addressing Limitations of Current Treatment Strategies

TIL Therapy in Immunologically Cold Tumors

NSCLC tumors are often categorized as immunologically cold, meaning that they have features thought to impede a strong immune response, including the lack of TILs within the tumor microenvironment. This may be due to a lack of tumor antigens, defective recruitment of antigen-presenting cells, lack of T-cell costimulation and activation, and modified production of chemokines and cytokines involved in cell trafficking and activation.6,20 TIL therapy may improve immunological response within the tumor by providing more T cells to mount an attack. Moreover, TIL therapy given in combination with an ICI may help to prevent T-cell inactivation via tumor-mediated mechanisms once they have infiltrated the tumor.4,21

Limitations With Other Adoptive Cell Therapies

ACT methodologies are centered around the manipulation of an individuals own immune cells to generate a tumor-specific, cell-mediated response against cancer.7 However, CAR-T and TCR-T therapies have faced challenges in the treatment of solid tumors, including the lack of stable tumor antigen expression and the need for human leukocyte antigen restriction. Severe and unpredictable toxicities can also occur with CAR-T and TCR-T due to cross-reactivity or trace expression of tumor-associated antigens in healthy cells.22-24 Further, acquired resistance can occur following a clinical response, which may be attributed to deletion or mutation of the target antigen, antigenic heterogeneity, or impaired trafficking.6,24,25

Unlike other ACTs, TILs are composed of polyclonal cells capable of simultaneously recognizing multiple tumor antigens.19 TILs are derived from genetically unmodified host cells, which may reduce the risk for complications from immune-mediated responses. TILs are also capable of targeting truncal neoantigens clonally expressed by a cancer cell, which may reduce the risk of resistance due to deficient target antigen expression.7

Durable Remissions With TIL Therapy

TIL therapy has the potential for durable, complete remissions.26 This partially is due to the transdifferentiation potential and lifespan of memory T cells.6 Such responses have been observed in heavily pretreated patients with metastatic melanoma after disease progression following treatment with chemotherapy, IL-2, antiCTLA-4 monoclonal antibodies, or a combination of these.26 Additionally, durable remissions following TIL therapy have been reported in a variety of other solid tumor types, including cholangiocarcinoma and cervical, colorectal, and breast cancers.27-30

Future Directions

Clinical trials in metastatic melanoma have demonstrated complete and durable responses from TIL therapy, even in patients who progressed on multiple prior therapies, including antiPD-1 agents.31,32 These findings suggest that TIL therapy may be a viable option for patients with PD-1 resistance or in cancers with lower immunogenicity. Observed similarities between NSCLC and melanoma suggest a role for TIL therapy in the treatment of NSCLC and warrant further investigation.

For information regarding advancements in TIL therapy, resources and further information are available from TILs Working Group at https://www.tilsinbreastcancer.org/.

References

1. Raskov H, Orhan A, Christensen JP, Gogenur I. Cytotoxic CD8+ T cells in cancer and cancer immunotherapy. Br J Cancer. 2020;124:359-367. doi:10.1038/s41416-020-01048-4

2. Horvath L, Thienpont B, Zhao L, Wolf D, Pircher A. Overcoming immunotherapy resistance in non-small cell lung cancer (NSCLC) - novel approaches and future outlook.Mol Cancer. 2020;19(1):141. doi:10.1186/s12943-020-01260-z

3. Nowicki TS, Hu-Lieskovan S, Ribas A. Mechanisms of resistance to PD-1 and PD-L1 blockade.Cancer J. 2018;24(1):47-53. doi:10.1097/PPO.0000000000000303

4. Adoptive cell therapy plus checkpoint inhibitors show promise in non-small cell lung cancer. New release. Moffitt Cancer Center. August 12, 2021. Accessed July 29, 2022. https://moffitt.org/newsroom/press-release-archive/adoptive-cell-therapy-plus-checkpoint-inhibitors-show-promise-in-non-small-cell-lung-cancer/

5. Pathak R, Pharaon RR, Mohanty A, Villaflor VM, Salgia R, Massarelli E. Acquired resistance to PD-1/PD-L1 blockade in lung cancer: mechanisms and patterns of failure.Cancers (Basel). 2020;12(12):3851. doi:10.3390/cancers12123851

6. Creelan BC, Wang C, Teer JK, et al. Tumor-infiltrating lymphocyte treatment for anti-PD-1-resistant metastatic lung cancer: a phase 1 trial.Nat Med. 2021;27(8):1410-1418. doi:10.1038/s41591-021-01462-y

7. Hulen TM, Chamberlain CA, Svane IM, Met O. ACT up TIL now: the evolution of tumor-infiltrating lymphocytes in adoptive cell therapy for the treatment of solid tumors. Immuno. 2022;1(3):194-211. doi:10.3390/immuno1030012

8. Rosenberg SA, Packard BS, Aebersold PM, et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report.N Engl J Med. 1988;319(25):1676-1680. doi:10.1056/NEJM198812223192527

9. Zur RT, Adler G, Shamalov K, et al. Adoptive T-cell immunotherapy: perfecting self-defenses. In: Klink M, Szulc-Kielbik I, eds. Interaction of Immune and Cancer Cells. Springer International Publishing AG; 2022:253-294.

10. Investigational TIL Therapy. Iovance Biotherapeutics. 2022. Accessed September 8, 2022. https://www.iovance.com/about-til/

11. Linette GP, Carreno BM. Tumor-infiltrating lymphocytes in the checkpoint inhibitor era.Curr Hematol Malig Rep. 2019;14(4):286-291. doi:10.1007/s11899-019-00523-x

12. Zhang Z, Lu M, Qin Y, et al. Neoantigen: a new breakthrough in tumor immunotherapy.Front Immunol. 2021;12:672356. doi:10.3389/fimmu.2021.672356

13. Qin SS, Melucci AD, Chacon AC, Prieto PA. Adoptive T cell therapy for solid tumors: pathway to personalized standard of care.Cells. 2021;10(4):808. doi:10.3390/cells10040808

14. Cullen SP, Brunet M, Martin SJ. Granzymes in cancer and immunity.Cell Death Differ. 2010;17(4):616-623. doi:10.1038/cdd.2009.206

15. Nirmala JG, Lopus M. Cell death mechanisms in eukaryotes.Cell Biol Toxicol. 2020;36(2):145-164. doi:10.1007/s10565-019-09496-2

16. Vinay DS, Ryan EP, Pawelec G, et al. Immune evasion in cancer: mechanistic basis and therapeutic strategies.Semin Cancer Biol. 2015;35(suppl):S185-S198. doi:10.1016/j.semcancer.2015.03.004

17. Sarnaik AA, Hamid O, Khushalani NI, et al. Lifileucel, a tumor-infiltrating lymphocyte therapy, in metastatic melanoma. J Clin Oncol. 2021;39(24):2656-2666. doi:10.1200/JCO.21.00612

18. Restifo NP, Dudley ME, Rosenberg SA. Adoptive immunotherapy for cancer: harnessing the T cell response.Nat Rev Immunol. 2012;12(4):269-281. doi:10.1038/nri3191

19. Wang S, Sun J, Chen K, Ma P, et al. Perspectives of tumor-infiltrating lymphocyte treatment in solid tumors.BMC Med. 2021;19(1):140. doi:10.1186/s12916-021-02006-4

20. Bonaventura P, Shekarian T, Alcazer V, et al. Cold tumors: a therapeutic challenge for immunotherapy.Front Immunol. 2019;10:168. doi:10.3389/fimmu.2019.00168

21. Lanitis E, Dangaj D, Irving M, Coukos G. Mechanisms regulating T-cell infiltration and activity in solid tumors.Ann Oncol. 2017;28(suppl 12):xii18-xii32. doi:10.1093/annonc/mdx238

22. Blumenschein GR, Devarakonda S, Johnson M, et al. Phase I clinical trial evaluating the safety and efficacy of ADP-A2M10 SPEAR T cells in patients with MAGE-A10+advanced non-small cell lung cancer.J Immunother Cancer. 2022;10(1):e003581. doi:10.1136/jitc-2021-003581

23. Duinkerken CW, Rohaan MW, de Weger VA, et al. Sensorineural hearing loss after adoptive cell immunotherapy for melanoma using MART-1 specific T cells: a case report and its pathophysiology.Otol Neurotol. 2019;40(7):e674-e678. doi:10.1097/MAO.0000000000002332

24. Sterner RC, Sterner RM. CAR-T cell therapy: current limitations and potential strategies.Blood Cancer J. 2021;11(4):69. doi:10.1038/s41408-021-00459-7

25. Haas AR, Tanyi JL, O'Hara MH, et al. Phase I study of lentiviral-transduced chimeric antigen receptor-modified T cells recognizing mesothelin in advanced solid cancers.Mol Ther. 2019;27(11):1919-1929. doi:10.1016/j.ymthe.2019.07.015

26. Rosenberg 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(13):4550-4557. doi:10.1158/1078-0432.CCR-11-0116

27. Zacharakis N, Chinnasamy H, Black M, et al. Immune recognition of somatic mutations leading to complete durable regression in metastatic breast cancer.Nat Med. 2018;24(6):724-730. doi:10.1038/s41591-018-0040-8

28. Stevanovi S, Draper LM, Langhan MM, et al. Complete regression of metastatic cervical cancer after treatment with human papillomavirus-targeted tumor-infiltrating T cells.J Clin Oncol. 2015;33(14):1543-1550. doi:10.1200/JCO.2014.58.9093

29. Tran E, Robbins PF, Lu YC, et al. T-cell transfer therapy targeting mutant KRAS in cancer.N Engl J Med. 2016;375(23):2255-2262. doi:10.1056/NEJMoa1609279

30. Tran E, Turcotte S, Gros A, et al. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer.Science. 2014;344(6184):641-645. doi:10.1126/science.1251102

31. Robertson J, Salm M, Dangl M. Adoptive cell therapy with tumour-infiltrating lymphocytes: the emerging importance of clonal neoantigen targets for next-generation products in non-small cell lung cancer.Immunooncol Technol. 2019;3:1-7. doi:10.1016/j.iotech.2019.09.003

32. Dafni U, Michielin O, Lluesma SM, et al. Efficacy of adoptive therapy with tumor-infiltrating lymphocytes and recombinant interleukin-2 in advanced cutaneous melanoma: a systematic review and meta-analysis.Ann Oncol. 2019;30(12):1902-1913. doi:10.1093/annonc/mdz398

Figure. Development Process of TIL Therapy for Solid Tumors13

TIL, tumor-infiltrating lymphocyte.

View original post here:
TIL Therapy as a Personalized Treatment Strategy for NSCLC - Targeted Oncology

Related Post