Lung cancer is the leading cause of cancer death in the United States.1 In the last decade, there have been significant gains in survival for patients with non–small cell lung cancer (NSCLC).2 Despite these improvements, survival in NSCLC is still poor—the 2-year relative survival was 42% for patients diagnosed in 2016—possibly due to the high tumor mutational burden and the heterogeneity seen with the disease.1,3

Despite gains in survival, the 2-year relative survival for patients diagnosed with NSCLC in 2016 was 42%. There is a substantial unmet need for patients with NSCLC who do not respond or have progressed on immune checkpoint inhibitors.

Checkpoint blockade inhibitors have dominated the treatment landscape, markedly improving clinical outcomes for some patients with NSCLC and other solid tumors. However, there is still a substantial unmet need for patients who do not respond or have progressed on immune checkpoint inhibitors.4 A key reason for immune checkpoint failure is extrinsic suppression of infiltrating immune cells by the tumor microenvironment. Therefore, new strategies are needed for those who experience treatment failure with checkpoint inhibitors.

Adoptive Cell Therapies

There has been growing interest in another class of immunotherapy—adoptive cell therapy (ACT)—that exploits the body’s ability to recognize and eliminate cancer cells. In ACT, a patient’s own T cells are either isolated based on their tumor-specific antigen recognition or are genetically modified to target certain molecules present on cancer cells.5 These cells are then expanded ex vivo and infused back into the patient.

ACT has several advantages compared with other immunotherapy options that rely on the active in vivo development of antitumor T cells to mediate cancer regression.6 With ACT, large numbers of antitumor lymphocytes with high recognition of the tumor can be grown in vitro. This allows for the manipulation of the host before cell transfer to provide a favorable microenvironment that better supports antitumor immunity through lymphodepletion.

Chimeric antigen receptor (CAR) T-cell therapy is the only ACT thus far to be approved by the U.S. Food and Drug Administration (FDA), and its use is currently limited to hematologic malignancies. Nonetheless, many other indications and cell types are currently under investigation.7 These include tumor-infiltrating lymphocytes (TIL) and T-cell receptor (TCR) therapy.

Response Rates to Adoptive Cell Therapies

CAR T-Cell Therapy

In CAR T-cell therapy, autologous T cells are genetically redirected and reprogrammed with CARs that target molecules expressed on malignant cells.8 CARs contain an extracellular antigen-identifying domain, which is constructed by fragments of monoclonal antibodies identifying a particular protein on the cell membrane of the cancerous tumor—for example, epidermal growth factor receptor (EGFR) on solid tumor cells or CD19 on B cells—and an intracellular stimulating domain that provides the TCR signaling to trigger CAR T-cell activation and function. Although the first CAR T cells were fairly simplistic, further generations have added costimulatory domains that improve CAR T-cell expansion, function, persistence, and antitumor activity.8

In patients with certain types of leukemia and lymphoma, CAR T-cell therapy has demonstrated durable disease remission—or even cure—by targeting the protein CD19, an antigen found on the surface of B cells. This has shifted the treatment landscape for lymphoid malignancies, especially diffuse large B‐cell lymphoma and acute lymphoblastic leukemia.9 Currently, there are several FDA-approved CAR T-cell products; three of these target CD19, and the FDA recently approved the first CAR T-cell product targeting B-cell maturation antigen for the treatment of multiple myeloma.10–14

CAR-T relies on a protein to be stuck on the surface of the tumor cell. …Most of the time, we don't have a protein on the surface of a tumor cell that we can just wipe out with impunity; otherwise, the patient risks off-tumor, on-target toxicity.Ben Creelan, MD

Despite the success of CAR T-cell therapy in hematologic malignancies, its use in solid tumors is still in its infancy.15 So far, no cell surface antigen with comparable properties as CD19 has yet been identified within solid tumors, although many tumor-associated antigens have been extensively investigated, including HER2, EGFR, WT-1, and mesothelin.16,17

An ideal molecule for CAR targeting should be overexpressed on the cancer cell surface of many patients, with zero or low expression in normal tissues. “CAR-T relies on a protein to be stuck on the surface of the tumor cell,” explained Ben Creelan, MD, a thoracic oncologist at the Moffitt Cancer Center, Tampa, Florida. “Most of the time, we don’t have a protein on the surface of a tumor cell that we can just wipe out with impunity; otherwise, the patient risks off-tumor, on-target toxicity.”

CAR T cells return to the bloodstream and lymphatic system, so they have more contact with blood tumor cells. In solid tumors, however, CAR T cells may not be able to penetrate tumor tissue through the vascular endothelium due to the lack of expression of chemokines involved in the trafficking of T cells into tumor tissues and the presence of dense fibrotic matrix in solid tumors.16 Another obstacle with the use of CAR T-cell therapy in solid tumors is a phenomenon known as “tumor antigen escape.” Tumor cells can escape killing by expressing alternative forms of the target antigen that lack the extracellular epitopes recognized by CAR T cells.3

CAR T-Cell Therapy in NSCLC

Several early clinical trials using CAR T-cell therapy in NSCLC have produced lackluster results, with several terminated or remaining unpublished. The most commonly studied target antigens include PD-LI, EGFR, mesothelin, mucin 1, PSCA, carcinoembryonic antigen, and HER2. 3,18–20 In one phase I trial of patients with EGFR-positive relapsed or refractory NSCLC, EGFR-targeted CAR T cells showed modest efficacy; of 11 patients, 2 had a partial response and 5 had stable disease.21 Other trials are attempting to limit toxicity by confining the infused CAR to the pleural space.22

Challenges With CAR T-Cell Therapy

In addition to efficacy concerns with CAR T-cell therapy in solid tumors, other challenges exist, including cytokine-release syndrome; on-target, off-tumor toxicity; neurologic toxicity; and anaphylaxis. Cytokine-release syndrome, the most frequent adverse event associated with CAR T-cell therapy, is estimated to affect 50% to 90% of patients in major clinical trials.23 It is most common within the first days after product infusion. This systemic inflammatory condition originates from direct activation and expansion of T cells after the interaction with target cells, leading to the production of cytokines such as TNF-alpha and INF-gamma.

The real challenge [with CAR T-cell therapy] in solid tumors is reaching a cell dose capable of response, while managing the off-tumor, on-target toxicity, which can be persistent and severe.Ben Creelan, MD

The severity of cytokine-release syndrome can vary widely from patient to patient, from self-limiting flu-like syndrome to life-threatening multiorgan dysfunction. However, Dr. Creelan noted that clinicians have learned to anticipate and manage cytokine-release syndrome. “At Moffitt, we recently treated our 500th patient with CAR T-cell therapy. We have developed streamlined algorithms and management guidelines for the toxicity due to CARs,” he said. “Most patients are willing to withstand the storm of CAR toxicity when it comes to cytokine-release syndrome. The real challenge in solid tumors is reaching a cell dose capable of response, while managing the off-tumor, on-target toxicity, which can be persistent and severe.”

The cost of therapy—estimated, in total, to be around $450,000—and time to manufacture the product are other legitimate concerns.24 “The time to production for autologous T cells is approximately 4 to 6 weeks to harvest the white blood cells, manufacture the T cells, and infuse them back into the patient,” explained Dr. Creelan. “That timeline can be an issue.”

Role of TIL in Tumor Microenvironment

TIL therapy is another adoptive cell therapy currently under investigation for the treatment of NSCLC. The interest in tumor-infiltrating lymphocytes having therapeutic potential spans decades, with studies beginning in the 1980s.25,26,27

TILs are a collection of heterogenous lymphocytes that have penetrated the stroma of a tumor.28,29 Important in the tumor microenvironment, TILs consist of numerous antitumor effector or regulatory T cells and are key players in the host’s immune response to tumors. These tumor-specific T cells are activated through encounters with tumor-associated antigens that are presented by specialized antigen-presenting cells, including dendritic cells. The most consistently beneficial TILs appear to be cytotoxic T lymphocytes—CD8-positive TILs—that specifically recognize and destroy target cells by recognizing tumor-derived antigenic epitopes.

What Is TIL Therapy?

While other types of ACT utilize circulating T cells from the blood, TIL therapy harvests neoantigen-directed T cells that are isolated from a tumor biopsy. Once the TILs are isolated, they are cultured and expanded. The patient receives a lymphodepleting conditioning regimen that diminishes immunosuppressive cells, and the TILs are then infused into the patient.

Because they recognize antigens specific to the individual patient’s tumor—somatic tumor-specific mutations—TILs are less likely to cause off-target toxicity compared with CAR T cells.4 Additionally, there is a lower risk for cytokine-release syndrome compared with CAR T-cell therapy, said Dr. Creelan. “The TILs use your own native repertoire of T cells and are not genetically modified like CARs,” he explained. “So, you have much less potential for the cytokine storm that can occur with CAR T, as those genetically modified T cells proliferate quickly.”

Use of TIL Therapy in Other Tumor Types

TIL therapy has demonstrated efficacy and safety in several patient populations with high unmet medical need, including unresectable and metastatic melanoma; relapsed, refractory or persistent cervical cancer; and head and neck squamous cell carcinoma.30–32

The most extensive and robust data with the use of TIL therapy are in the treatment of melanoma.33 In a National Institutes of Health (NIH) study, 93 patients with metastatic melanoma received TIL plus IL-2 following a preparative lymphodepleting regimen. The treatment led to durable complete responses in 22% of patients, and 95% of these complete responses were ongoing beyond 3 years.34 In a prospective, phase II open-label, single-arm multicenter study in 66 patients with advanced melanoma progressing on immune checkpoint inhibitor therapy (and BRAF inhibition with or without MEK inhibition if BRAF V600 mutated), lifileucel showed a response rate of 36.4%.30 Lifileucel, an autologous TIL cell therapy, was produced from harvested tumor specimens using a streamlined 22-day process. At 33.1 months of follow-up, the median duration of response has not been reached for this cohort.54 A phase III study is underway that compares the current standard of care with ipilimumab to TIL in patients with advanced melanoma; the role of TIL as possible first-line therapy in combination with anti–PD-1 is an additional subject of clinical trials.33,35

The success achieved with TIL therapy in melanoma is encouraging for its use in NSCLC due to the similarities between the two malignancies. NSCLC has one of the highest clonal mutation burdens of all cancers through exposure to carcinogens in tobacco smoke—second only to melanoma.4,36 In patients with melanoma, the tumor mutational load and predicted neoantigen load have been shown to correlate with clinical outcomes for TIL therapies, suggesting that TIL efficacy is driven through neoantigen-reactive T cells within the product.37 As Dr. Creelan pointed out, the immune microenvironment within the lung tissue has evolved to be the body’s strongest line of defense to fight foreign proteins. “Because the lungs are the first barrier to fight against pathogens, we see some of the most robust antigen-specific T cells within the lung.”

TIL Therapy in NSCLC

PD-1/PD-L1 inhibitors as first-line therapy, with or without platinum-based chemotherapy, have substantially improved overall survival in patients with NSCLC.38–41 However, 45% to 50% of patients with metastatic NSCLC do not achieve an optimal response with chemotherapy and checkpoint inhibition—and almost three-quarters of patients experience disease progression or die within 12 months of starting treatment.4 For those with resistant disease following PD-1/PD-L1 inhibitor plus platinum doublet chemotherapy as first-line treatment, there is currently no standard second-line treatment.4

In this early lung cancer trial, we saw responses, including complete responses, in patients who have progressed on PD-1 inhibitors and had subsequent tumor growth. The key point is that TIL can work in this population.Ben Creelan, MD

Early studies of TIL in NSCLC began in 1996.42 However, recent improvements in the manufacturing process—including the use of good manufacturing practices to isolate and expand TIL cultures using state-of-the-art protocols—coupled with the incorporation of a preconditioning regimen have increased the feasibility of TIL therapy.36

Recently, a phase I trial investigated the combination of TIL therapy and anti–PD-1 therapy in patients with metastatic NSCLC.43 Patients with no prior exposure to PD-1 inhibitors had their TILs harvested from a resected metastatic lesion before receiving four doses of nivolumab. Those patients with tumor enlargement or new lesions (n = 13) proceeded to lymphodepletion, TIL infusion, and attenuated IL-2; then, nivolumab was resumed for up to 11 doses to augment TIL persistence.

Of the 13 patients who received TIL therapy, 2 achieved durable complete responses and 3 maintained a clinical remission by local ablative therapy of an isolated new lesion post-TIL. “In this early lung cancer trial, we saw responses, including complete responses, in patients who have progressed on PD-1 inhibitors and had subsequent tumor growth,” said Dr. Creelan, the lead study author. “The key point is that TIL can work in this population.” The most common nonhematologic adverse events of the treatment regimen included hypoalbuminemia, hypophosphatemia, nausea, hyponatremia, and diarrhea.

A phase II study is now underway to evaluate the efficacy of TIL in metastatic NSCLC, including patients without an actionable driver mutation following a single line of approved systemic therapy consisting of an immune checkpoint inhibitor, chemotherapy, with or without bevacizumab.44

Considerations With TIL Therapy

TIL therapy is a one-time treatment option, which alleviates the risk of repetitive or persistent adverse events. The personalized immunotherapy can come at a substantial cost, though, because a patient-specific infusion product must be produced in highly specialized cGMP facilities.33 Additionally, the production time of a TIL product has historically been too long for some patients with rapidly progressive disease.33 However, “traditionally, TIL production was 6 to 7 weeks, but now the timeline is down to 22 days or so,” said Dr. Creelan. “Overcoming this hurdle has been an important milestone in making this therapy feasible for patients in regular practice.”

The most common toxicities during TIL therapy are associated the lymphodepleting preparative regimens and the subsequent IL-2 after TIL infusion. Because of the risk of toxicity, TIL therapy usually requires inpatient hospital monitoring during the IL-2 phase. If lymphodepletion chemotherapy is added to this inpatient stay, it can result in a median duration of as long as 20 days.45 Nonmyeloablative lymphodepleting chemotherapy may cause hematologic and nonhematologic toxicities, including neutropenic fever, diarrhea, hyperbilirubinemia, and fludarabine-induced neurotoxicity. While high-dose IL-2 therapy toxicity is usually transient, it requires close monitoring in hospital setting; capillary leak syndrome after IL-2 administration is common. High-grade toxicity attributed to the TIL infusion product itself is “exceedingly uncommon.” Patients may develop transient dyspnea, chills, and fever shortly after infusion of TIL.

We need to dispel the stodgy idea that this population is ineligible for high-risk, high-reward treatments.Ben Creelan, MD

A Path Forward

Despite the adoption of immune checkpoint inhibitors for the treatment of advanced NSCLC, there is still a significant clinical need for additional treatment options. ACT therapies appear to be an optimal candidate for the treatment of NSCLC; however, enrolling patients for clinical trials has been a hurdle for researchers. “As medical oncologists, we are lousy at planning ahead,” conceded Dr. Creelan. “We often pick the ‘lowest-hanging fruit’ as the next treatment option instead of strategically planning. The truth is that TIL and other ACT therapies require a reciprocal discussion with the patient to explain the treatment process and assess their motivation level.”

Additionally, he worries that the historical nihilism about lung cancer as a terminal diagnosis affects how treatment of advanced NSCLC is approached. “We need to dispel the stodgy idea that this population is ineligible for high-risk, high-reward treatments. Patient advocacy groups are working to change physician attitudes.”

The “high reward” of TIL therapy cannot be overlooked; as Dr. Creelan noted, some patients with melanoma who were treated with TIL therapy in the 1980s are still in remission today. “While normal effector T cells only last 7 days, our memory T cells can engraft and persist for 60 to 70 years.” Ultimately, collaboration between researchers, cell therapy experts, and community practitioners will benefit the advancement of study in this facet of immuno-oncology for patients with advanced NSCLC.

Disclosures

Dr. Creelan has served in a consulting or advisory role for Abbvie, AstraZeneca/MedImmune, E.R. Squibb Sons LLC, Achilles plc, and KSQ Therapeutics; has participated in a speakers' bureau for AstraZeneca/MedImmune; has received institutional research funding from Bristol-Myers Squibb, Iovance Biotherapeutics, and Prometheus Laboratories; and has been reimbursed for travel, accommodations, or other expenses by AstraZeneca.

References

  1. Siegel RL, Miller KD, Fuchs HE, et al: Cancer Statistics, 2021. CA Cancer J Clin 71:7-33, 2021.
  2. Chen Z, Fillmore CM, Hammerman PS, et al: Non-small-cell lung cancers: A heterogeneous set of diseases. Nat Rev Cancer 14:535-546, 2014.
  3. Qu J, Mei Q, Chen L, et al: Chimeric antigen receptor (CAR)-T-cell therapy in non-small-cell lung cancer (NSCLC): Current status and future perspectives. Cancer Immunol Immunother 70:619-631, 2021.
  4. 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. IOTECH 3:1-7, 2019.
  5. Olsen, K: Tumor-infiltrating Lymphocyte Therapy Explained. American Association for Cancer Research (AACR) Blog. Published November 19, 2018. https://www.aacr.org/professionals/blog/3944-2-til-therapy. Accessed May 5, 2021.
  6. Rosenberg SA, Restifo NP: Adoptive cell transfer as personalized immunotherapy for human cancer. Science 348:62-68, 2015.
  7. U.S. Food and Drug Administration: Approved cellular and gene therapy products. Available at www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy-products/approved-cellular-and-gene-therapy-products. Accessed May 10, 2021.
  8. Weinkove R, George P, Dasyam N, et al: Selecting costimulatory domains for chimeric antigen receptors: functional and clinical considerations. Clin Transl Immunol 8:e1049, 2019.
  9. Sermer D, Brentjens R. CAR T-cell therapy: Full speed ahead. Hematol Oncol 37(S1):95-100, 2019.
  10. Maude SL, Laetsch TW, Buechner J, et al: Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med 378:439-448, 2018.
  11. Locke FL, Ghobadi A, Jacobson CA, et al: Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): A single-arm, multicentre, phase 1–2 trial. Lancet Oncol 20:31-42, 2019.
  12. Schuster SJ, Bishop MR, Tam CS, et al: Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med 380:45-56, 2019.
  13. Abramson JS, Palomba ML, Gordon LI, et al: Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): A multicentre seamless design study. Lancet 396:839-852, 2019.
  14. U.S. Food and Drug Administration: FDA approves first cell-based gene therapy for adult patients with multiple myeloma. Available at www.fda.gov/news-events/press-announcements/fda-approves-first-cell-based-gene-therapy-adult-patients-multiple-myeloma. Accessed May 10, 2021.
  15. Salmon H, Franciszkiewicz K, Damotte D, et al: Matrix architecture defines the preferential localization and migration of T cells into the stroma of human lung tumors. J Clin Invest 122:899-910, 2012.
  16. Li 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 11:22, 2018.
  17. Yu S, Li A, Liu Q, et al: Chimeric antigen receptor T cells: A novel therapy for solid tumors. J Hematol Oncol 10:78, 2017.
  18. Liu M, Wang X, Li W, et al: Targeting PD-L1 in non-small cell lung cancer using CAR T cells. Oncogenesis 9:1-11, 2020.
  19. Li N, Liu S, Sun M, et al: Chimeric antigen receptor-modified T cells redirected to EphA2 for the immunotherapy of non-small cell lung cancer. Transl Oncol 11:11-17, 2017.
  20. CAR-T cell immunotherapy for advanced lung cancer. ClinicalTrials.gov identifier: NCT03330834. Available at https://clinicaltrials.gov. Accessed June 8, 2021.
  21. Feng 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 59:468-479, 2016.
  22. Adusumilli PS, Zauderer MG, Rusch VW, et al: Regional delivery of mesothelin-targeted CAR T cells for pleural cancers: Safety and preliminary efficacy in combination with anti-PD-1 agent. J Clin Oncol 37:2511, 2019.
  23. Cerrano M, Ruella M, Perales MA, et al: The advent of CAR T-cell therapy for lymphoproliferative neoplasms: Integrating research into clinical practice. Front Immunol 11, 2020.
  24. Lyman GH, Nguyen A, Snyder S, et al: Economic evaluation of chimeric antigen receptor T-cell therapy by site of care among patients with relapsed or refractory large B-cell lymphoma. JAMA Netw Open 3:e202072, 2020.
  25. Rosenberg SA, Lotze MT, Muul LM, et al: Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer. N Engl J Med 313:1485-1492, 1985.
  26. Rosenberg SA, Spiess P, Lafreniere R: A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science 233:1318-1321, 1986.
  27. 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 319:1676-1680, 1988.
  28. Wang J, Tian S, Sun J, et al: The presence of tumour-infiltrating lymphocytes (TILs) and the ratios between different subsets serve as prognostic factors in advanced hypopharyngeal squamous cell carcinoma. BMC Cancer 20:731, 2020.
  29. Restifo NP, Dudley ME, Rosenberg SA: Adoptive immunotherapy for cancer: Harnessing the T cell response. Nat Rev Immunol 12:269-281, 2012.
  30. Sarnaik A, Khushalani NI, Chesney JA, et al: Long-term follow up of lifileucel (LN-144) cryopreserved autologous tumor infiltrating lymphocyte therapy in patients with advanced melanoma progressed on multiple prior therapies. J Clin Oncol 38(suppl):10006, 2020.
  31. Jazaeri AA, Zsiros E, Amaria RN, et al: Safety and efficacy of adoptive cell transfer using autologous tumor infiltrating lymphocytes (LN-145) for treatment of recurrent, metastatic, or persistent cervical carcinoma. J Clin Oncol 37(suppl):2538, 2019.
  32. Jimeno A, Papa S, Haigentz M, et al: Safety and efficacy of tumor infiltrating lymphocytes (TIL, LN-145) in combination with pembrolizumab for advanced, recurrent or metastatic HNSCC. J Immunother Cancer 8(suppl):353, 2020.
  33. Rohaan MW, Berg JH van den, Kvistborg P, et al: Adoptive transfer of tumor-infiltrating lymphocytes in melanoma: a viable treatment option. J Immunother Cancer 6:102, 2018.
  34. Andersen R, Donia M, Ellebaek E, et al: Long-lasting complete responses in patients with metastatic melanoma after adoptive cell therapy with tumor-infiltrating lymphocytes and an attenuated IL2 regimen. Clin Cancer Res 22:3734-3745, 2016.
  35. The Netherlands Cancer Institute. (2020). Randomized phase III study comparing a non-myeloablative lymphocyte depleting regimen of chemotherapy followed by infusion of tumor infiltrating lymphocytes and interleukin-2 to standard ipilimumab treatment in metastatic melanoma. ClinicalTrials.gov identifier: NCT02278887. Available at https://clinicaltrials.gov. Accessed June 10, 2021.
  36. Ben-Avi R, Farhi R, Ben-Nun A, et al: Establishment of adoptive cell therapy with tumor infiltrating lymphocytes for non-small cell lung cancer patients. Cancer Immunol Immunother 67:1221-1230, 2018.
  37. Lauss M, Donia M, Harbst K, et al: Mutational and putative neoantigen load predict clinical benefit of adoptive T cell therapy in melanoma. Nature Communications 8:1738, 2017.
  38. Rodriguez-Abreu D, Powell SF, Hochmair M, et al: Final analysis of KEYNOTE-189: Pemetrexed-platinum chemotherapy with or without pembrolizumab in patients with previously untreated metastatic nonsquamous non-small cell lung cancer. J Clin Oncol 38(suppl):9582, 2020.
  39. Paz-Ares L, Vicente D, Tafreshi A, et al: A randomized, placebo-controlled trial of pembrolizumab plus chemotherapy in patients with metastatic squamous NSCLC: Protocol-specified final analysis of KEYNOTE-407. J Thorac Oncol 15:1657-1669, 2020.
  40. Socinski MA, Jotte RM, Cappuzzo F, et al: Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N Engl J Med 378:2288-2301, 2018.
  41. Brahmer JR, Rodriguez-Abreu D, Robinson A: KEYNOTE-024 5-year OS update: First-line pembrolizumab vs platinum-based chemotherapy in patients with metastatic NSCLC and PD-L1 tumour proportion score ≥50%. Ann Oncol 31(suppl):S1142-S1215, 2020.
  42. Ratto GB, Zino P, Mirabelli S, et al: A randomized trial of adoptive immunotherapy with tumor-infiltrating lymphocytes and interleukin-2 versus standard therapy in the postoperative treatment of resected nonsmall cell lung carcinoma. Cancer 78:244-251, 1996.
  43. Creelan B, Wang C, Teer J, et al: Durable complete responses to adoptive cell transfer using tumor infiltrating lymphocytes (TIL) in non-small cell lung cancer (NSCLC): A phase I trial. Cancer Res 80(suppl):CT056, 2020.
  44. Autologous LN-145 in patients with metastatic non-small-cell lung cancer. ClinicalTrials.gov identifier: NCT04614103. Available at https://clinicaltrials.gov. Accessed June 10, 2021.
  45. Wolf B, Zimmermann S, Arber C, et al: Safety and tolerability of adoptive cell therapy in cancer. Drug Saf 42:315-334, 2019. 
  46. Park JH, Rivière I, Gonen M, et al: Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med 378:449-459, 2018.
  47. Turtle CJ, Hay KA, Hanafi L-A, et al: Durable molecular remissions in chronic lymphocytic leukemia treated with CD19-specific chimeric antigen receptor-modified T cells after failure of ibrutinib. J Clin Oncol 35:3010-3020, 2017.
  48. Raje NS, Berdeja JG, Lin Y, et al: bb2121 anti-BCMA CAR T-cell therapy in patients with relapsed/refractory multiple myeloma: Updated results from a multicenter phase I study. J Clin Oncol 36(suppl 15):8007, 2018.
  49. Fry TJ, Shah NN, Orentas RJ, et al: CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy. Nat Med 24:20-28.
  50. Ramos CA, Bilgi M, Gerken CP, et al: CD30-chimeric antigen receptor (CAR) T cells for therapy of hodgkin lymphoma (HL). Blood 132(suppl 1):680, 2018.
  51. Thomas SS, In GK, Doger B, et al: Safety and efficacy of lifileucel (LN-144), an autologous, tumor infiltrating lymphocyte cell therapy in combination with pembrolizumab for immune checkpoint inhibitor naïve patients with advanced melanoma. J Clin Oncol 39(suppl 15):9537, 2021.
  52. Rosenberg S, Yang J, Sherry R. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res 17:4550-4557, 2011.
  53. Creelan B, Wang C, Teer J, et al. Abstract CT056: Durable complete responses to adoptive cell transfer using tumor infiltrating lymphocytes (TIL) in non-small cell lung cancer (NSCLC): A phase I trial. Cancer Res 80(suppl 16):CT056, 2020.
  54. Larkin J, Sarnaik, Chesney JA, et al: Lifileucel, a cryopreserved autologous tumor infiltrating lymphocyte therapy in patients with advanced melanoma: Evaluation of impact of prior anti-PD-1 therapy. 2021 ASCO Annual Meeting. Abstract 9505. Presented June 6, 2021.

Disclaimer

Sponsored content is not written by and does not necessarily reflect the views of ASCO or the The ASCO Post editorial staff. It is authored by Harborside Studio writers or freelance writers approved by Harborside Studio. Harborside Studio's sponsored content is held to editorial standards expected in The ASCO Post with the intent to provide valuable information to The ASCO Post readers. The mention of any company, product, service, or therapy does not constitute an endorsement of any kind by ASCO. ASCO assumes no responsibility for any injury or damage arising out of or related to use of the sponsored content or any errors or omissions. This sponsored content has been produced with funding support from Iovance Biotherapeutics.