Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Nov 28.
Published in final edited form as: Cancer Lett. 2024 Oct 5;605:217281. doi: 10.1016/j.canlet.2024.217281

Therapeutic potential of tumor-infiltrating lymphocytes in non-small cell lung cancer

Daniel R Plaugher 1,#, Avery R Childress 1, Christian M Gosser 1, Dave-Preston Esoe 1, Kassandra J Naughton 1, Zhonglin Hao 2,3, Christine F Brainson 1,3,#
PMCID: PMC11560632  NIHMSID: NIHMS2029185  PMID: 39369769

Abstract

Lung cancer is the leading cause of cancer-related death worldwide, with poor outcomes even for those diagnosed at early stages. Current standard-of-care for most non-small cell lung cancer (NSCLC) patients involves an array of chemotherapy, radiotherapy, immunotherapy, targeted therapy, and surgical resection depending on the stage and location of the cancer. While patient outcomes have certainly improved, advances in highly personalized care remain limited. However, there is growing excitement around harnessing the power of tumor-infiltrating lymphocytes (TILs) through the use of adoptive cell transfer (ACT) therapy. These TILs are naturally occurring, may already recognize tumor-specific antigens, and can have direct anti-cancer effect. In this review, we highlight comparisons of various ACTs, including a brief TIL history, show current advances and successes of TIL therapy in NSCLC, discuss the potential roles for epigenetics in T cell expansion, and highlight challenges and future directions of the field to combat NSCLC in a personalized manner.

Keywords: Tumor-infiltrating lymphocytes, non-small cell lung cancer, adoptive cell transfer

1. Introduction to T Cell Therapies

Despite advances in the spectrum of therapies, the 5-year survival rate of lung cancer in the U.S. remains among the lowest of all cancer types at 23% (other sites: pancreas 12%, esophagus 21%, liver 21%) and is the leading cause of cancer related death among both men and women 1. Lung cancer in general is immensely heterogeneous, first being subcategorized as small cell lung cancer (SCLC), a highly aggressive neuroendocrine tumor, and NSCLC, which includes all other lung carcinomas. In this review, we focus on NSCLC, which accounts for 85% of overall cases and can be further divided into three main subtypes: lung adenocarcinoma (LADC), lung squamous cell carcinoma (LSCC), and mixed/poorly differentiated. The LADC subtype accounts for approximately 45% of diagnoses and is typically found in the distal lung/bronchus, whereas LSCC comprises 18% of cases and often presents proximally2,3. Adding further to NSCLC heterogeneity is the high mutation rate, with hundreds of somatic mutations per genome - largely attributed to smoking and other environmental exposure4,5. This magnifies the difficulty for clinicians to identify effective targeted therapies but can also be a vulnerability of cancer due to the high rate of neo-antigen presentation for T cell recognition6,7.

Metastatic NSCLC harboring actionable driver mutations are currently treated with small molecule tyrosine kinase inhibitors (TKIs) in the first line due to their exceptional response rate (70-80% ORR) and low toxicities8. These include tumors that contain EGFR, ALK, ROS, RET, MET, RAF and NTRK mutants, most of which are LADC 912. Many tumors bearing these particular driver mutations are immunologically suppressive and respond poorly to immune checkpoint inhibitors (ICIs)13,14. Unfortunately, these immunologically cold tumors also often progress within 3 months of treatment with TKI15, and TKI used together (or sequentially) have been shown to induce high rate of pneumonitis16,17. Alternatively, tumors harboring no actionable driver are treated with single agent ICI if tumor expresses PD-L1 in 50% or more cells18,19 whereas tumors expressing less than 50% of PD-L1 are treated with combination chemotherapy and immunotherapy. Patients who have failed targeted therapy are treated with chemotherapy, whereas those who have failed immunotherapy and chemotherapy are often considered for clinical trials. However, salvage docetaxel with or without ramucirumab, which is an antibody blocking vascular endothelial growth factor receptor 2 (VEGFR-2) protein, are often given when clinical trials are not available. The approval of ICI (e.g. anti-CTLA-4, anti-PD-1, anti-PD-L1…etc.) for metastatic melanoma in 201120, then later for NSCLC in 201621, dramatically improved patient outcomes22,23. However, the continued low rate of durable response to therapies in NSCLC underscores the major unmet need for improved treatment options. Moreover, it has been shown that patients with certain mutations (e.g. EGFR, KRAS, ALK, STK11) can have even worse rates of response for reasons still unknown2426. Investigation revealed that these tumors have low CD8+ T cells and low PD-1/PD-L1 expression 24,27. While many concepts are being tested such as combination ICIs, these strategies may prove futile since NSCLC can be immunologically cold in certain cases28. Further, TKI and ICI combination therapy has shown a high rate of severe side effects16.

The novel immunotherapy techniques of ACT have gained momentum over the last decade and continue to show promise moving forward. The three main types of ACT include chimeric antigen receptor T cell (CAR-T), modified T cell receptor (TCR), and TIL therapies (Figure 1). Both CAR and TCR therapies can pull T cells from peripheral blood, however, TIL therapy requires solid tumor infiltrates. Counter to standard therapeutics, ACT is a “living” treatment where re-infused lymphocytes can expand more than 1000-fold after administration and can remain in circulation for several years (or more)6. TIL as second line therapy (or above) in advanced stage cancers after failure of PD1/L1 checkpoint inhibitors appears to be feasible and effective29,30. In melanoma, patients previously treated with BRAF/MEK inhibitors who had progression on these therapies were also included in the initial trial, and neither BRAF status nor PD-L1 status correlated with response to TIL therapy 30. Therefore, combination use TIL therapy after progression on TKI may be an avenue of therapy to overcome TKI resistance, and offer longer term disease control. However, for NSCLC, tumor genotypes and phenotypes that will respond best to TIL therapy, and situations where TIL therapy and ICI therapy together will be effective, are currently unknown. Numerous trials combining ICIs and TIL therapy are underway, and will be discussed more in subsequent sections.

Figure 1: Adoptive cell therapy subtypes.

Figure 1:

(A) CAR-T cells are now up to 5 generations, all containing CD3ζ chains and mixtures of co-stimulatory molecules (CM) or transgenes (TG). (B) Modified-TCR cells include altered variable alpha and beta chains of the TCR to recognize specific tumor (neo)antigens. (C) TILs are isolated from solid tumors, are polyclonal (indicated here by various colored TCRs), and can be cultured in bulk, further selected for specificity, or cultured for a shorter duration (young).

CAR-T Therapy

CARs (Figure 1A) are recombinant receptors that have been synthetically engineered into T cells, usually via retroviral infection, and provide targeted antigen binding as well as T cell activation22,31. A similar process can also be completed with NK cells (CAR-NK therapy)32. To date, there are five generations (Gen) of CARs: 1st Gen contain only a CD3ζ chain in their signaling domain with no co-stimulatory molecules (CM); 2nd Gen have CD3ζ and one CM, allowing dual signaling pathways; 3rd Gen have CD3ζ and several CMs; 4th Gen resemble 2nd Gen but incorporate an additional NFAT-responsive cassette that expresses anti-cancer cytokines (i.e. . triple signals - primary CD3ζ, CM, and expressed transgenic (TG) proteins); lastly, 5th Gen are similar to 2nd and 4th Gen but add membrane receptors IL-2Rβ that provide a binding site for STAT3 and activate the JAK-STAT signaling domain33. The CMs used often include CD28, CD27, CD134 (OX40), and CD137 (4-1BB)34. While CAR-T therapies have shown significant durability in hematologic cancers such as B cell lymphomas, acute lymphocytic leukemia, and multiple myeloma3537, they have not been successful in solid tumors because of their requirement of antigen specificity22.

Modified TCR Therapy

The TCR is the antigen recognition structure that is expressed by all T cells and connects with antigen presenting cells (APCs) via the major histocompatibility complex (MHC) molecules on their surface. Upon binding, the T cells either recognize the antigen presented as self or foreign, and then they proceed accordingly34. Unlike CARs, TCRs are naturally occurring and are somatically rearranged through V/D/J recombination to confer recognition of epitopes derived from proteins residing within any subcellular compartment38,39. This enables TCRs to detect a broad spectrum of targets and, since TCRs have evolved to efficiently detect and amplify antigenic signals, these receptors are much more sensitive to smaller epitope densities than those required for CAR-signaling34. With modified TCR therapy, a retrovirus is often used to transduce modified alpha and beta chains of the TCR to recognize tumor specific antigen(s) (Figure 1B)32,34. Similar to CAR-T therapy, TCR therapy is restricted by its narrow scope of antigen recognition but has shown good results in melanoma, nasopharyngeal cancer, and cervical carcinoma40.

TIL Therapy

The origins of clinical TIL research are credited to Dr. Steven A. Rosenberg beginning in the late 1980’s at the National Cancer Institute in Bethesda, Maryland4144. While the infiltrating nature of leukocytes was observed as early as the 1860’s32,45, Rosenberg first isolated polyclonal TILs (Figure 1C) from mice in the early 1980’s and showed that pairing TILs with IL-2 could improve colon cancer in mice46. High dose IL-2 (720,000 IU/kg) would go on to be approved for renal cell carcinoma and melanoma in 199247, but the progress for TILs was much slower due to the complex nature of the process, as well as skepticism of patient efficacy and tolerance. It was not until February 2024 that the first FDA approved TIL therapy48, called lifileucel, to be used as second-line therapy in advanced melanoma. Cost remains a major area for improvement, because the newly approved treatment is estimated at over $500,000 USD per patient – more than some CAR-T treatments. A possible solution to this problem may be observed in our European counterparts, where TILs are produced in-house directly at the hospitals rather than outsourcing to companies49,50 – an estimated cost of only $55,000 USD48. However, regardless of the many challenges still ahead, this new groundbreaking approval of lifileucel will now pave the way for advances in other cancer types, like NSCLC. According to clinicaltrials.gov, there are already TIL trials involving cancers of the brain, liver, cervix, breast, pancreas, gastrointestine, colorectal, esophagus, and more.

Success of TIL therapy is dependent on numerous variables and mechanisms. First, perhaps most obvious, there must be tumor-reactive TIL present in the patients surgical or biopsy specimen. For this reason, it is often recommended that tumors must be greater than 1.5cm in diameter to ensure sufficient TIL counts for expansion but smaller masses could possibly be utilized51,52. T cells must also maintain memory status to avoid being terminally differentiated into effector type T cells during the expansion process53. Further, upon re-infusion TILs must navigate from the blood to the site(s) of remaining tumor that was not previously removed. They must then exit through endothelial venules according to a three-step process involving rolling via L-selectin, activation via CCR7, and sticking via LFA-154,55. Lastly, once they have arrived at their destination, they must selectively identify their target and induce effector functions (releasing perforin, granzyme B, IFN-γ, TNF-α,…etc.) to eliminate the cancer39.

2. TIL Expansion and Infusion Protocols

At a surface level, the process of producing TILs appears straightforward. First, a patient’s tumor is resected and dissociated, then TILs are then expanded in culture with IL-2, tested for quality and reactivity, and finally re-infused into the patient (Figure 2). Yet, at each step there are multiple options for procedural variation, manual labor can be time consuming, and the space required to achieve the necessary TIL counts under good manufacturing practices (GMP) can quickly overcome a standard research lab. Additionally, there are various subsets of TILs, which depending on expansion, can accelerate or further complicate the process.

Figure 2: Schematic of simplified TIL process. Here we show a simplified overview of the TIL process in NSCLC.

Figure 2:

Tumors are resected and dissociated, TILs are then expanded in culture with IL-2, tested for quality and reactivity, and finally re-infused into the patient. However, depending on the protocol chosen (bulk, selected, young, or hybrid), there are various caveats along the way that add to the complexity of development.

The initial steps are approximately the same for all TIL subtypes prior to the rapid expansion protocol (pre-REP). First, TILs can be acquired from enzymatic tumor digests OR tumor fragments. Next, cell cultures can be initiated in 24-well plates with 1x106 tumor digest cells per well, one tumor fragment in 2 mL of complete medium (RPMI 1640 with glutamine, supplemented with 10% human AB serum, 25 mM HEPES buffer and 10 μg/mL gentamicin) and 6000 IU/mL of IL-2, OR using gas-permeable flasks56. Then cells are incubated at 37°C in 5% CO2. It typically takes ~1 week for the initial TIL cultures to overtake the tumor cells57.

Bulk TILs vs. Selected TILs vs. Young TILs

At this point, a decision will need to be made regarding expansion in bulk, selected, or young TILs. Each type has unique advantages and disadvantages. In general, bulk TILs offer simplicity and broad responsiveness, selected TILs are more specific, and young TILs have more proliferative potential and persistence (Figure 3). Further, to overcome the possibility of T cells becoming terminally differentiated, it is possible to reprogram cells to a naïve state by: (1) reprogramming into induced pluripotent stem cells, then to naïve T cells, or (2) direct reprogramming from differentiated to less differentiated53,58. Researchers are also exploring hybrid options of each technique23. In general, the final result will primarily consist of tumor antigen reactive polyclonal CD8+ and CD4+ T cells22,32.

Figure 3: TIL subtypes.

Figure 3:

Depending on which rapid expansion procedure (REP) chosen, different TIL products can be achieved. Bulk TILs offer simplicity and broad responsiveness but can become exhausted or terminally differentiated (darker depiction). Selected TILs are more specific but can take longer because of a smaller starting population. Young (minimal) TILs have more proliferative potential and persistence (lighter depiction) but have fewer cell totals at infusion.

Bulk TILs are expanded in totality, regardless of which subset of lymphocyte they are associated with. The heterogeneous population is composed of CD8+, CD4+ (Th (T helper)1, Th2, Th9, Th17, Th22, Treg (regulatory T cells), and Tfh (follicular helper T cells)), natural killer (NK) cells, myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages, B cells, and other elements of the tumor environment59. Importantly, the culturing conditions provide an environment most favorable to T cells, so while other cell types may be present at the final infusions step, they are at very low levels. Bulk would also comprise naïve, activated, and exhausted cells, which leads to bulk TILs requiring large numbers of cells (5 – 500 x 109 cells or more) for infusion to achieve best results. In total, the process for bulk TIL therapy could take 4-6 weeks start-to-finish, but new industrial advancements are improving long wait times29,52,57.

Selected TILs consist of antigen-specific TILs or can be sorted according to marker specific collections. These can include CD8+, CD4+, CD28+, CD27+, CD134+ (OX40), CD137+ (4-1BB), CD279+ (PD-1), specific neoantigens (e.g. mutations in TP53, KRAS, …etc.)6062, as well as negative selections (e.g. exclude FOXP3+) and many more59,60,63,64. Following the pre-REP phase, TILs will be tested for reactivity against specific (neo)antigens using ELISPOT, flow cytometry, or tetramer staining59. Similarly, they can be sorted according to surface markers using flow cytometry or magnetic sorting. These filtered sets then undergo REP, but starting at a much smaller total population, which leads to longer end-to-end preparation time to achieve similar cell totals as bulk at infusion61.

Lastly, young TILs are less differentiated and have shorter expansion times compared to bulk and selected TILs50,65,66. This allows for a higher proliferative capacity and longer persistence. Young TILs are typically defined by cultures that had only expanded to confluency of the original 2-mL well and had eliminated their co-cultured tumor cells66. Therefore, pooling all of the wells in a single 24 well plate could achieve approximately 5 x 107 young TIL cells. These cultures usually take 10-18 days after initiation, compared to the multi-week process for bulk and selected TILs. Notably, the alternative long culture times required to perform screening assays showed telomere shortening as well as reduced expression of CD27 and CD28 in the TIL cultures50,65,66.

Rapid Expansion Protocol (REP)

There are now multiple solutions to the complicated TIL process, including automated biotech solutions (e.g. Miltenyi Biotec) and better basic lab tools (e.g. Wilson Wolf56). Rosenberg and Jin et al. detailed the two common options for the REP process57. Traditionally, in T-175 flasks there are 1×106 TILs suspended in 150 mL of complete media. The TILs are then cultured with irradiated allogeneic peripheral blood mononuclear cells (PBMC) as “feeder” cells. Next, the cells are cultured in a 1:1 mixture of complete medium and AIM-V medium (50/50 medium), supplemented with 3000 IU/mL of IL-2 and 30 ng/mL of anti-CD3. The T-175 flasks are incubated at 37°C in 5% CO2, and half of the media is changed on day 5 using 50/50 medium with 3000 IU/mL of IL-2. On day 7, TILs from two T-175 flasks are combined in a 3liter bag in 300 mL of AIM V media supplemented with 5% human AB serum and 3000 IU per mL of IL-2. Every day, TILs are re-counted and a corresponding amount of fresh media is addedso that TIL concentration remains between 0.5 and 2.0×106 cells per mL.

Alternatively, a “simplified” method utilizes gas-permeable flasks57. A major limitation of the traditional method is flask height, which restricts how many TILs can expand in one flask. To combat this problem, one can use gas-permeable flasks that are vertically taller and cylindrical. In 500 mL capacity flasks , 5-10×106 TILs are cultured with irradiated allogeneic PBMC in 400 mL of 50/50 medium, supplemented with 5% human AB serum, 3000 IU/mL of IL-2 and 30 ng/mL of anti-CD3, then incubated. On day 5, 250 mL of supernatant is removed and centrifuged. The TIL pellets are re-suspended with 150 mL of fresh medium with 5% human AB serum, 3000 IU/mL of IL-2, and added back to the original flasks. Then, TILs are expanded serially on day 7 and supplemented with fresh media after 4 days. Finally, the cells are harvested on day 14 of culture.

Post-REP Infusion

After TILs have undergone successful REP, they are ready for infusion back into the patient. Prior to infusion, there is typically a round of lymphodepleting chemotherapy 2-7 days beforehand. This pretreatment includes cyclophosphamide and fludarabine (cy/flu) or total body irradiation (TBI) at levels that are non-myeloablative52,67. It has been shown that treatment with cy/flu increases efficacy of the TIL treatment, because the suppressing impact of regulatory cells is dampened by increasing host homeostatic cytokines (e.g. IL-7 and IL-15) as well as decreasing endogenous lymphocyte and myeloid populations41,51,59. Once the TILs are infused, patients undergo high doses of IL-2 (720,000 IU/kg) to help boost TILs. To manage side-effects of IL-2, protocols often elect to administer less than 6 high doses post infusion or use low/intermediate doses over longer periods29,59.

Epigenetic control of T cell Expansion and Exhaustion

As discussed above, one of the challenges of TIL therapy is to expand effector T cells that will remain functional, while limiting regulatory T cells and T cell exhaustion. All these phenotypes are controlled in part through epigenetic mechanisms, including the methylation of DNA and histones to silence gene expression. For example, multiple studies have shown that trimethylation of histone 3 at lysine 27 (H3K27me3), which is catalyzed by the histone methyltransferase EZH2, favors Tregs in the tumor microenvironment 68,69. In addition, H3K27me3 can suppress transcription of genes associated with memory T cells, including the transcription factors Tcf7 and Id3 53,70. Specifically relevant to TILs, targeting EZH2 with pharmacological inhibitors may help improve the efficacy of the therapy by preventing terminally differentiated and exhausted phenotypes that result from extensive ex vivo expansion 71,72. Similarly, use of ex vivo BET bromodomain inhibitors have been shown to enhance stem like and central memory phenotypes, downregulate factors associated with Th17 differentiation, and allow for improved persistence and anti-tumor activity of re-infused T cells 73. Additionally, inhibition of EZH2 74, as well as other epigenetic targets such as DNA methyltransferases (DNMTi) 75, or histone deacetylases (HDACi) 76, can improve patient response to immune checkpoint inhibitors, and therefore have potential to help improve outcomes of TIL infusions, particularly when combined with immune checkpoint inhibitors 53. Given that deletion of Ezh2 can also restrict memory T cell function, finding the appropriate doses and timing of drugs will be critical 70,77. Although more research is needed to validate targeting epigenetic programs in TIL protocols, data from many groups suggest that epi-drugs will have clinical usefulness.

3. Clinical success and current trials in nsclc

Initial trials (primarily in melanoma) in the 1980s yielded disappointing outcomes. Some trials reported that none of the patients achieved greater than 50% reduction of tumor burden78, and others showed similar results because of inability to expand TILs or non-optimized protocols22,79,80. However, after more initial demonstrations of hopeful outcomes, multiple trials around the globe began confirming results with objective response responses (ORR) consistently between 40-70% 81. Rosenberg and colleagues were able to show in several consecutive trials that ORRs of up to 72% were achievable, 10-20% of patients had complete remission, and 40% had durable clinical responses8183. Importantly, the probability of these responses did not appear to depend to previously failed systemic treatment.

As previously mentioned, lifileucel (LN-145 in Table 1) was the first TIL therapy to earn FDA approval in February 2024 for advanced melanoma based on results from the C-144-01 study29. According to a follow-up analysis 84,85, among all patients (n=153), the median overall survival (OS) was 13.9 months (95% CI, 10.6-17.8), and the 4-year OS rate was 22.2%. Within the responders (n=48), 4-year OS rates by response type were: 48.3% in early responders (95% CI, 31.9%-62.9%, n=39), 41.7% in late responders (95% CI, 10.9%-70.8%, n=9), 37.2% in responders without a deepened response (95% CI, 21.0%-53.5%, n=16), and 68.2% in responders with a deepened response (95% CI, 39.5%-85.4%, n=32). Early and late response were defined as before and after 42 days, respectively, and a deepened response was those patients who had an increased in response over time. The median duration of response (DOR) for all responders was not reached (NR; 95% CI, 8.3-NR). Specifically, median DOR according to response type were: NR in early responders (95% CI, 6.1-NR), 19.8 months (95% CI, 4.1-NR) in late responders, 26.2 months (95% CI, 4.1-NR) in responders without a deepened response, and NR (95% CI, 8.3-NR) in responders with deepened responses 84. All patients in this study had progressed on (or after) anti-PD-1/L1 therapy, and 49.7% of patients had tumors with greater than 1% PD-L1+ tumor proportion score.

Table 1: Search of NSCLC TIL trials in the US.

The following table includes a summary of clinical trials with TIL therapy according to clinicaltrials.gov as of July 2024.

NCT Number Study Title Brief Summary From ClinicalTrials.Gov Interventions
NCT03215810 Phase 1 Completed Nivolumab and Tumor Infiltrating Lymphocytes (TIL) in Advanced Non-Small Cell Lung Cancer “Investigators plan to study the safety, side effects, and benefits of tumor-infiltrating lymphocytes (TILs) when they are given with the drug nivolumab. Nivolumab is a type of immunotherapy - a drug that is used to boost the ability of the immune system to fight cancer, infection, and other diseases.” TIL / Nivolumab
NCT02848872 Observational Completed Evaluation of Tumor and Blood Immune Biomarkers in Resected Non-small Cell Lung Cancer “The hypothesis of this study is that functional tumor infiltrating lymphocyte (TIL) isolation from resected lung cancer specimens is feasible, allowing determination of tumor antigen-specific T cell reactivities. The primary objective of this study is to investigate the feasibility of isolating functional tumor infiltrating lymphocytes s(TILs) to determine tumor antigen-specific T cell re-activities in 30 resected lung tumor specimens. Successful isolation of TILs will be defined as collecting 1x10−6 viable, CD45+ mononuclear cells or greater from tumors containing \>/=1 gram of excess tissue. If successful isolation of TILs can be obtained from \>/= 66% of resected tumor specimens, the protocol will be considered feasible. The primary exploratory objective is to identify immunologic signatures that predict clinical outcomes from cytotoxic chemotherapy and/or immunotherapy.” TIL
NCT01820754 Phase 2 Completed Evaluation of Circulating T Cells and Tumor Infiltrating Lymphocytes (TILs) During / After Pre-Surgery Chemotherapy in Non-Small Cell Lung Cancer (NSCLC) “The purpose of this study is to evaluate whether the combination of neoadjuvant chemotherapy (chemotherapy before surgery) plus ipilimumab for lung cancer increases the number of patients with detectable circulating T cells with specificities against tumor associated antigens (TAA) from zero percent (0%) of patients prior to therapy to 20% of patients after neoadjuvant chemotherapy plus ipilimumab.” TILs / Ipilimumab / Paclitaxel / Cisplatin / Carboplatin
NCT05397093 Phase 1 Active - not recruiting ITIL-306 in Advanced Solid Tumors “ITIL-306-201 is a multicenter, clinical trial evaluating the safety and feasibility of ITIL-306 in adult participants with advanced solid tumors whose disease has progressed after standard therapy. ITIL-306 is a cell therapy derived from a participant’s own tumor-infiltrating immune cells (lymphocytes; TILs) and contains a unique molecule designed to increase TIL activity when it encounters folate receptor α (FOLR1) on the tumor.” ITIL-306
NCT02818920 Phase 2 Active - not recruiting Neoadjuvant Pembrolizumab “This multi-institutional clinical trial is studying two doses of pembrolizumab administered prior to surgery (neoadjuvant therapy) and 4 doses administered after surgery (adjuvant therapy) for stage IB, II or IIIA non-small cell lung cancer. Pembrolizumab is a type of immunotherapy that may enhance the ability of the immune system to fight off cancer. The study will investigate the effects of pembrolizumab on the immune system and how certain immune cells, called TILs (tumor infiltrating lymphocytes), respond to pembrolizumab. Previous studies suggest that pembrolizumab could alter the immune cells in a way that the the immune cells identify cancer cells. Pembrolizumab has been approved for the treatment of advanced lung cancer, but is investigational in this setting.” TIL / Pembrolizumab
NCT05681780 Phase 1/2 Recruiting Clinical Trial of CD40L-Augmented TIL for Patients With EGFR, ALK, ROS1 or HER2-Driven NSCLC “To determine the effect of a special preparation of cells, called tumor-infiltrating lymphocytes (TIL) stimulated with CD40L, when given with the drug nivolumab, for patients with EGFR, ALK, ROS1, or HER2-genomically altered lung cancer.” TIL / Nivolumab
NCT04614103 Phase 2 Recruiting Autologous LN-145 in Patients With Metastatic Non-Small-Cell Lung Cancer “This is a prospective, open-label, multi-cohort, non-randomized, multicenter study evaluating LN-145 in patients with metastatic non-small-cell lung cancer.” LN-145 (TIL)
NCT05573035 Phase 1 Recruiting A Study to Investigate LYL845 in Adults With Solid Tumors “This is an open-label, multi-center, dose-escalation study with expansion cohorts, designed to evaluate the safety and anti-tumor activity of LYL845, an epigenetically reprogrammed tumor infiltrating lymphocyte (TIL) therapy, in participants with relapsed or refractory (R/R) metastatic or locally advanced melanoma, non-small cell lung cancer (NSCLC), and colorectal cancer (CRC).” LYL845 (TIL)
NCT05576077 Phase 1 Recruiting A Study of TBio-4101 (TIL) and Pembrolizumab in Patients With Advanced Solid Tumors “A multicenter trial to investigate TBio-4101, an autologous, neoantigen-selected, tumor-reactive TIL product, in patients with advanced solid malignancies.” TBio-4101 (TIL) / Pembrolizumab
NCT03645928 Phase 2 Recruiting Study of Autologous Tumor Infiltrating Lymphocytes in Patients With Solid Tumors “A prospective, open-label, multi-cohort, non-randomized, multicenter study evaluating adoptive cell therapy (ACT) with TIL LN-144 (Lifileucel)/LN-145 in combination with checkpoint inhibitors or TIL LN-144 (Lifileucel)/LN-145/LN-145-S1 as a single agent therapy.” LN-145 (TIL) / Pembrolizumab / Ipilimumab / Nivolumab
NCT02133196 Phase 2 Recruiting T Cell Receptor Immunotherapy for Patients With Metastatic Non-Small Cell Lung Cancer “The purpose of this study is to see if these specifically selected tumor fighting cells can cause non-small cell lung cancer (NSCLC) tumors to shrink and to see if this treatment is safe.” Young TIL
NCT06060613 Phase 1/2 Recruiting Safety and Efficacy of OBX-115 in Advanced Solid Tumors “This is a study to investigate the safety and efficacy of an investigational OBX-115 regimen in adult participants with advanced solid tumors.” OBX-115 (TIL)
NCT05361174 Phase 1/2 Recruiting A Study to Investigate the Efficacy and Safety of an Infusion of IOV-4001 in Adult Participants With Unresectable or Metastatic Melanoma or Stage III or IV Non-small-cell Lung Cancer “This is a study to investigate the efficacy and safety of an infusion of IOV-4001 in adult participants with unresectable or metastatic melanoma or advanced non-small-cell lung cancer (NSCLC).” IOV-4001 (TIL)
NCT05566223 Phase 1/2 Not yet recruiting CISH Inactivated TILs in the Treatment of NSCLC “A clinical trial to assess the safety and efficacy of genetically-engineered Tumor Infiltrating Lymphocytes (TIL) in which the intracellular immune checkpoint CISH has been inhibited using CRISPR gene editing for the treatment of Metastatic Non-small Cell Lung Cancer (NSCLC).” CISH Inactivated TIL / Pembrolizumab

Lifileucel is currently being studied for efficacy and safety in patients with metastatic NSCLC patients who are resistant to ICIs (PD-L1–negative, low tumor mutational burden, STK11 mutation,… etc.) and are treatment refractory (NCT03645928). At phase 2 of this multicenter study, the objective response rate was 21.4%, which is a major improvement for this population of patients86. Other recent studies have demonstrated growing improvement in the strategy and feasibility within NSCLC patients – including those with refractory disease30,87. As of July 2024, there have been (or currently are) 14 US clinical trials related to TIL therapy in NSCLC with a variety of combination studies and genetically altered TILs since 2000 (clincialtrials.gov). Of these trials, 3 have been completed – 2 of which only analyzed properties of TIL rather than used TIL as a treatment mechanism. In Table 1, we summarize each trial with its respective treatment – many include combination treatments with ICIs or TKIs.

The lone completed trial utilizing TILs as therapy (NCT03215810) was conducted as a single-arm open-label phase 1 trial of TILs administered with nivolumab in 20 patients with advanced NSCLC following initial progression on nivolumab monotherapy30. Their primary endpoint was safety (achieved with severe toxicity rate ≤17% (95% confidence interval, 3–29%)) and secondary end points were ORR, DOR and T cell persistence. Among the 13 patients who could be evaluated, 3 showed confirmed responses, 11 experienced a reduction in tumor size (−35% median best change), and 2 patients achieved complete responses (sustained 1.5 years later). Further analysis showed that T cells targeting various cancer mutations were identified after TIL treatment and were more prevalent in patients who responded to the therapy. Thus, Creelan et al. showed that TILs are generally safe, show clinical activity, and are a promising new treatment option for NSCLC30.

Challenges for toxicity and patient efficacy

While the prospect of TIL therapy in NSCLC is very bright, there still remain drawbacks and challenges. Advances in technology have shown drastic improvement the REP process, as well as identifying the subcategories of selected TILs for optimal reactivity, plasticity, stemness, and resilience57,64,66,88,89. With respect to production, average turnaround is 4-6 weeks (possibly longer). Such long wait times may not be feasible for patients with extreme late-stage disease, as seen with the loss of two patients in the Creelan et al. study, where TIL production was approximately 63 days30,90. Furthermore, those with heavy tumor burden, fast progression, and advanced age may find the treatment regimen with IL-2 and cy/flu very hard to tolerate even over a short period, with four patient deaths occurring on the C-144-01 study attributed to these treatments29. The study of 16 TIL treated NSCLC patients in the Creelan et al. study, found main side-effects to be hypoalbuminemia (13 - grade 2; 1 - grade 3), hypophosphatemia (4 - grade 2; 8 - grade 3), nausea (7 - grade 1; 7 - grade 3), hyponatremia (8 - grade 1; 2 - grade 3; 1 - grade 4), and diarrhea (5 - grade 1; 4 - grade 2)30. Interestingly, cytokine release syndrome appeared as a rare side-effect. Again, the majority of TIL-related adverse events were related to lymphodepletion and IL-2, but peaked around 7 days post-infusion and resolved within one month. Notably, the recruitment for the study included: 50% current/former smoker, median age 54 years, median PD-L1 proportion 6%, many patients with targetable mutations, predominant LADC histology, median nonsynonymous TMB of 1.5 mutations per megabase, and mostly bulky disease. Thus, a broader cohort for better analysis is still needed.

A common course of TIL treatment includes cyclophosphamide for two days, then fludarabine for 5 days, followed by TIL infusion and IL-26. Pretreatment with cy/flu will deplete the immune system to reduce endogenous impact on TILs (resulting in leukopenia, lymphopenia, neutropenia…etc.), but the immune system is reconstituted within just over one week6,30. As the field continues to develop, substantial hurdles remain to be crossed with regard to both the TIL expansion and reinfusion process. Researchers are aiming to achieve the most effective TILs, while also minimizing adverse impacts of pre- and co-treatment therapeutic regimens with cy/flu and IL-2. For example, IL-2 can lead to terminal effector state of T cells and, to circumvent the production of predominantly exhausted T cells, some have suggested using homeostatic cytokines like IL-7, IL-15, or IL-2181. Further, it has been shown that an elevated number of IL-2 doses (720,000 IU/kg every 8 hours up to 15 doses) can actually increase Treg counts and increase risk of cytokine release syndrome91.

Concluding remarks

As lung cancer continues to harm hundreds of thousands of patients annually, it remains of utmost importance to find personalized treatments for patients that are curated to their specific tumor microenvironment. Arguably the most patient specific approach currently in development is using a patient’s endogenous TILs, utilizing their own natural mechanisms to fight the cancer. Better protocols, modern technology, epigenetic reprogramming, and improved administration are on the rise, all of which paint a highly optimistic future for better treatment of the world’s most prevalent and deadly cancer.

Highlights.

  • Tumor-infiltrating lymphocytes (TILs) are a promising precision therapy for lung cancer

  • TIL subtypes and age can be influenced by expansion protocols

  • Inhibitors of epigenetic enzymes could improve TIL expansion and therapeutic effect

  • Numerous trials are ongoing for TILs in lung cancer

Acknowledgments

This work was supported in part by NCI T32 CA165990 (DRP), R01 CA237643, P20 GM121327-03, R01 HL170193, R01 CA266004, P30 CA177558, the American Institute for Cancer Research Grant 710410, and the Markey Research Women Strong Distinguished Researcher Award 3048116064 (CFB). Images were created in part by biorender.com.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Competing Interests

The authors declare no Competing Financial or Non-Financial Interests.

Declaration of interests

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

The author is an Editorial Board Member/Editor-in-Chief/Associate Editor/Guest Editor for Cancer Letters and was not involved in the editorial review or the decision to publish this article.

References

  • 1.Siegel RL, Giaquinto AN & Jemal A Cancer statistics, 2024. CA: A Cancer Journal for Clinicians 74, 12–49 (2024). [DOI] [PubMed] [Google Scholar]
  • 2.Brainson CF, Huang B, Chen Q, McLouth LE, He C, Hao Z, et al. Description of a Lung Cancer Hotspot: Disparities in Lung Cancer Histology, Incidence, and Survival in Kentucky and Appalachian Kentucky. Clin Lung Cancer 22, e911–e920 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Chen Z, Fillmore CM, Hammerman PS, Kim CF & Wong K-K Non-small-cell lung cancers: a heterogeneous set of diseases. Nat Rev Cancer 14, 535–546 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Collisson EA, Campbell JD, Brooks AN, Berger AH, Lee W, Chmielecki J, et al. Comprehensive molecular profiling of lung adenocarcinoma. Nature 511, 543–550 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kandoth C, McLellan MD, Vandin E, Ye K, Niu B, Lu C, et al. Mutational landscape and significance across 12 major cancer types. Nature 502, 333–339 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Rosenberg SA & Restifo NP Adoptive cell transfer as personalized immunotherapy for human cancer. Science 348, 62–68 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Rosenberg SA, Parkhurst MR & Robbins PF Adoptive cell transfer immunotherapy for patients with solid epithelial cancers. Cancer Cell 41, 646–648 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Thai AA, Solomon BJ, Sequist LV, Gainor JF & Heist RS Lung cancer. The Lancet 398, 535–554 (2021). [DOI] [PubMed] [Google Scholar]
  • 9.Koivunen JP, Mermel C, Zejnullahu K, Murphy C, Lifshits E, Holmes AJ, EML4-ALK Fusion Gene and Efficacy of an ALK Kinase Inhibitor in Lung Cancer. Clinical Cancer Research 14, 4275–4283 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Engelman JA & Jänne PA Factors Predicting Response to EGFR Tyrosine Kinase Inhibitors. Seminars in respiratory and critical care medicine 26, 314–322 (2005). [DOI] [PubMed] [Google Scholar]
  • 11.Politi K & Herbst RS Lung cancer in the era of precision medicine. Clin Cancer Res 21, 2213–2220 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Soria JC, Ohe Y, Vansteenkiste J, Reungwetwattana T, Chewaskulyong B, Lee KH, et al. Osimertinib in Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. N Engl J Med 378, 113–125 (2018). [DOI] [PubMed] [Google Scholar]
  • 13.Lisberg A, Cummings A, Goldman JW, Bornazyan K, Reese N, Wang T, et al. A Phase II Study of Pembrolizumab in EGFR-Mutant, PD-L1+, Tyrosine Kinase Inhibitor Naïve Patients With Advanced NSCLC. J Thorac Oncol 13, 1138–1145 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Mazieres J, Drilon A, Lusque A, Mhanna L, Cortot AB, Mezquita L, et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Ann Oncol 30, 1321–1328 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Takashima Y, Sakakibara-Konishi J, Hatanaka Y, Hatanaka KC, Ohhara Y, Oizumi S, et al. Clinicopathologic Features and Immune Microenvironment of Non-Small-cell Lung Cancer With Primary Resistance to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors. Clin Lung Cancer 19, 352–359 (2018). [DOI] [PubMed] [Google Scholar]
  • 16.Schoenfeld AJ, Arbour KC, Rizvi H, Iqbal AN, Gadgeel SM, Girshman J, et al. Severe immune-related adverse events are common with sequential PD-(L)1 blockade and osimertinib. Ann Oncol 30, 839–844 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Oxnard GR, Yang JC, Yu H, Kim SW, Saka H, Horn L, et al. TATTON: a multi-arm, phase Ib trial of osimertinib combined with selumetinib, savolitinib, or durvalumab in EGFR-mutant lung cancer. Ann Oncol 31, 507–516 (2020). [DOI] [PubMed] [Google Scholar]
  • 18.Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, Fülöp A, et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med 375, 1823–1833 (2016). [DOI] [PubMed] [Google Scholar]
  • 19.Mok TSK, Wu YL, Kudaba I, Kowalski DM, Cho BC, Turna HZ, et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial. Lancet 393, 1819–1830 (2019). [DOI] [PubMed] [Google Scholar]
  • 20.Ledford H Melanoma drug wins US approval. Nature 471, 561–561 (2011). [DOI] [PubMed] [Google Scholar]
  • 21.Shields MD, Marin-Acevedo JA & Pellini B Immunotherapy for Advanced Non–Small Cell Lung Cancer: A Decade of Progress. American Society of Clinical Oncology Educational Book, e105–e127 (2021). [DOI] [PubMed] [Google Scholar]
  • 22.Katiyar V, Chesney J & Kloecker G Cellular Therapy for Lung Cancer: Focusing on Chimeric Antigen Receptor T (CAR T) Cells and Tumor-Infiltrating Lymphocyte (TIL) Therapy. Cancers (Basel) 15, e3733 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Zhang Y & Zhang Z The history and advances in cancer immunotherapy: understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cellular & molecular immunology 17, 807–821 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Gainor JF, Shaw AT, Sequist LV, Fu X, Azzoli CG, Piotrowska Z, et al. EGFR Mutations and ALK Rearrangements Are Associated with Low Response Rates to PD-1 Pathway Blockade in Non-Small Cell Lung Cancer: A Retrospective Analysis. Clin Cancer Res 22, 4585–4593 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Jeanson A, Tomasini P, Souquet-Bressand M, Brandone N, Boucekine M, Grangeon M, et al. Efficacy of Immune Checkpoint Inhibitors in KRAS-Mutant Non-Small Cell Lung Cancer (NSCLC). Journal of Thoracic Oncology 14, 1095–1101 (2019). [DOI] [PubMed] [Google Scholar]
  • 26.Skoulidis F, Goldberg ME, Greenawalt DM, Hellmann MD, Awad MM, Gainor JF, et al. STK11/LKB1 Mutations and PD-1 Inhibitor Resistance in KRAS-Mutant Lung Adenocarcinoma. Cancer Discovery 8, 822–835 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Jiang B, Hu L, Dong D, Guo Z, Wei W, Wang C, et al. TP53 or CDKN2A/B covariation in ALK/RET/ROS1-rearranged NSCLC is associated with a high TMB, tumor immunosuppressive microenvironment and poor prognosis. J Cancer Res Clin Oncol 149, 10041–10052 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Maleki Vareki S. High and low mutational burden tumors versus immunologically hot and cold tumors and response to immune checkpoint inhibitors. Journal for ImmunoTherapy of Cancer 6, e157 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Chesney J, Lewis KD, Kluger H, Hamid O, Whitman E, Thomas S, et al. Efficacy and safety of lifileucel, a one-time autologous tumor-infiltrating lymphocyte (TIL) cell therapy, in patients with advanced melanoma after progression on immune checkpoint inhibitors and targeted therapies: pooled analysis of consecutive cohorts of the C-144-01 study. J Immunother Cancer 10, e005755 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Creelan BC, Wang C, Teer JK, Toloza EM, Yao J, Kim S, et al. Tumor-infiltrating lymphocyte treatment for anti-PD-1-resistant metastatic lung cancer: a phase 1 trial. Nature Medicine 27, 1410–1418 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Sadelain M, Brentjens R & Rivière I The Basic Principles of Chimeric Antigen Receptor Design. Cancer Discovery 3, 388–398 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kazemi MH, Sadri M, Najafi A, Rahimi A, Baghernejadan Z, Khorramdelazad H, et al. Tumor-infiltrating lymphocytes for treatment of solid tumors: It takes two to tango? Frontiers in immunology 13, e1018962 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Asmamaw Dejenie T, Tiruneh GMM, Dessie Terefe G, Tadele Admasu F, Wale Tesega W & Chekol Abebe E Current updates on generations, approvals, and clinical trials of CAR T-cell therapy. Hum Vaccin Immunother 18, e2114254 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chandran SS & Klebanoff CA T cell receptor-based cancer immunotherapy: Emerging efficacy and pathways of resistance. Immunol Rev 290, 127–147 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Locke FL, Miklos DB, Jacobson CA, Perales M-A, Kersten M-J, Oluwole OO, et al. Axicabtagene Ciloleucel as Second-Line Therapy for Large B-Cell Lymphoma. New England Journal of Medicine 386, 640–654 (2022). [DOI] [PubMed] [Google Scholar]
  • 36.Martin T, Usmani SZ, Berdeja JG, Agha M, Cohen AD, Hari P, et al. Ciltacabtagene Autoleucel, an Anti-B-cell Maturation Antigen Chimeric Antigen Receptor T-Cell Therapy, for Relapsed/Refractory Multiple Myeloma: CARTITUDE-1 2-Year Follow-Up. J Clin Oncol 41, 1265–1274 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Park JH, Rivière I, Gonen M, Wang X, Sénéchal B, Curran KJ, et al. Long-Term Follow-up of CD19 CAR Therapy in Acute Lymphoblastic Leukemia. New England Journal of Medicine 378, 449–459 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Rudolph MG, Stanfield RL & Wilson IA How TCRs bind MHCs, peptides, and coreceptors. Annu Rev Immunol 24, 419–466 (2006). [DOI] [PubMed] [Google Scholar]
  • 39.Parham P & Janeway C The immune system, (Garland Science, Taylor & Francis Group, New York, NY, 2015). [Google Scholar]
  • 40.Tsimberidou A-M, Van Morris K, Vo HH, Eck S, Lin Y-E, Rivas JM, et al. T-cell receptor-based therapy: an innovative therapeutic approach for solid tumors. Journal of Hematology & Oncology 14, e102 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Rosenberg SA, Spiess P & Lafreniere R A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science 233, 1318–1321 (1986). [DOI] [PubMed] [Google Scholar]
  • 42.Spiess PJ, Yang JC & Rosenberg SA In vivo antitumor activity of tumor-infiltrating lymphocytes expanded in recombinant interleukin-2. Journal of the National Cancer Institute 79, 1067–1075 (1987). [PubMed] [Google Scholar]
  • 43.Topalian SL, Muul LM, Solomon D & Rosenberg SA Expansion of human tumor infiltrating lymphocytes for use in immunotherapy trials. J Immunol Methods 102, 127–141 (1987). [DOI] [PubMed] [Google Scholar]
  • 44.Topalian SL, Solomon D & Rosenberg SA Tumor-specific cytolysis by lymphocytes infiltrating human melanomas. J Immunol 142, 3714–3725 (1989). [PubMed] [Google Scholar]
  • 45.Virchow R. Cellular pathology. As based upon physiological and pathological histology. Lecture XVI--Atheromatous affection of arteries. 1858. Nutr Rev 47, 23–25 (1989). [DOI] [PubMed] [Google Scholar]
  • 46.Eberlein TJ, Rosenstein M & Rosenberg SA Regression of a disseminated syngeneic solid tumor by systemic transfer of lymphoid cells expanded in interleukin 2. The Journal of experimental medicine 156, 385–397 (1982). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Fyfe G, Fisher RI, Rosenberg SA, Sznol M, Parkinson DR & Louie AC Results of treatment of 255 patients with metastatic renal cell carcinoma who received high-dose recombinant interleukin-2 therapy. J Clin Oncol 13, 688–696 (1995). [DOI] [PubMed] [Google Scholar]
  • 48.Reardon S. First cell therapy for solid tumours heads to the clinic: what it means for cancer treatment. Nature Epub ahead of print., 10.1038/d41586-41024-00673-w (2024). [DOI] [PubMed] [Google Scholar]
  • 49.Rohaan MW, Borch TH, Berg J.H.v.d., Met Ö, Kessels R, Foppen MHG, et al. Tumor-Infiltrating Lymphocyte Therapy or Ipilimumab in Advanced Melanoma. New England Journal of Medicine 387, 2113–2125 (2022). [DOI] [PubMed] [Google Scholar]
  • 50.Andersen R, Donia M, Ellebaek E, Borch TH, Kongsted P, Iversen TZ, et al. Long-Lasting Complete Responses in Patients with Metastatic Melanoma after Adoptive Cell Therapy with Tumor-Infiltrating Lymphocytes and an Attenuated IL2 Regimen. Clinical Cancer Research 22, 3734–3745 (2016). [DOI] [PubMed] [Google Scholar]
  • 51.Rosenberg SA, Restifo NP, Yang JC, Morgan RA & Dudley ME Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev Cancer 8, 299–308 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Betof Warner A, Hamid O, Komanduri K, Amaria R, Butler MO, Haanen J, et al. Expert consensus guidelines on management and best practices for tumor-infiltrating lymphocyte cell therapy. J Immunother Cancer 12, e008735 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Henning AN, Roychoudhuri R & Restifo NP Epigenetic control of CD8+ T cell differentiation. Nature Reviews Immunology 18, 340–356 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Mora JR & von Andrian UH T-cell homing specificity and plasticity: new concepts and future challenges. Trends Immunol 27, 235–243 (2006). [DOI] [PubMed] [Google Scholar]
  • 55.Ager A, Watson HA, Wehenkel SC & Mohammed RN Homing to solid cancers: a vascular checkpoint in adoptive cell therapy using CAR T-cells. Biochem Soc Trans 44, 377–385 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Forget MA, Haymaker C, Dennison JB, Toth C, Maiti S, Fulbright OJ, et al. The beneficial effects of a gas-permeable flask for expansion of Tumor-Infiltrating lymphocytes as reflected in their mitochondrial function and respiration capacity. Oncoimmunology 5, e1057386 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Jin J, Sabatino M, Somerville R, Wilson JR, Dudley ME, Stroncek DF, et al. Simplified method of the growth of human tumor infiltrating lymphocytes in gas-permeable flasks to numbers needed for patient treatment. J Immunother 35, 283–292 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Crompton JG, Clever D, Vizcardo R, Rao M & Restifo NP Reprogramming antitumor immunity. Trends Immunol 35, 178–185 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Zhao Y, Deng J, Rao S, Guo S, Shen J, Du F, et al. Tumor Infiltrating Lymphocyte (TIL) Therapy for Solid Tumor Treatment: Progressions and Challenges. Cancers (Basel) 14, e4160 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hanada K.-i., Zhao C, Gil-Hoyos R, Gartner JJ, Chow-Parmer C, Lowery FJ, et al. A phenotypic signature that identifies neoantigen-reactive T cells in fresh human lung cancers. Cancer Cell 40, 479–493 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kim SP, Vale NR, Zacharakis N, Krishna S, Yu Z, Gasmi B, et al. Adoptive Cellular Therapy with Autologous Tumor-Infiltrating Lymphocytes and T-cell Receptor–Engineered T Cells Targeting Common p53 Neoantigens in Human Solid Tumors. Cancer Immunology Research 10, 932–946 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Parikh AY, Masi R, Gasmi B, Hanada K.-i., Parkhurst M, Gartner J, et al. Using patient-derived tumor organoids from common epithelial cancers to analyze personalized T-cell responses to neoantigens. Cancer Immunology Immunotherapy 72, 3149–3162 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Radvanyi LG, Bernatchez C, Zhang M, Fox PS, Miller P, Chacon J, et al. Specific lymphocyte subsets predict response to adoptive cell therapy using expanded autologous tumor-infiltrating lymphocytes in metastatic melanoma patients. Clin Cancer Res 18, 6758–6770 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Ye Q, Song D-G, Poussin M, Yamamoto T, Best A, Li C, et al. CD137 Accurately Identifies and Enriches for Naturally Occurring Tumor-Reactive T Cells in Tumor. Clinical Cancer Research 20, 44–55 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Besser MJ, Shapira-Frommer R, Treves AJ, Zippel D, Itzhaki O, Schallmach E, et al. Minimally cultured or selected autologous tumor-infiltrating lymphocytes after a lympho-depleting chemotherapy regimen in metastatic melanoma patients. J Immunother 32, 415–423 (2009). [DOI] [PubMed] [Google Scholar]
  • 66.Tran KQ, Zhou J, Durflinger KH, Langhan MM, Shelton TE, Wunderlich JR, et al. Minimally cultured tumor-infiltrating lymphocytes display optimal characteristics for adoptive cell therapy. J Immunother 31, 742–751 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Restifo NP, Dudley ME & Rosenberg SA Adoptive immunotherapy for cancer: harnessing the T cell response. Nature Reviews Immunology 12, 269–281 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.DuPage M, Chopra G, Quiros J, Rosenthal WL, Morar MM, Holohan D, et al. The Chromatin-Modifying Enzyme Ezh2 Is Critical for the Maintenance of Regulatory T Cell Identity after Activation. Immunity 42, 227–238 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Wang D, Quiros J, Mahuron K, Pai C-C, Ranzani V, Young A, et al. Targeting EZH2 Reprograms Intratumoral Regulatory T Cells to Enhance Cancer Immunity. Cell reports 23, 3262–3274 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Gray SM, Amezquita RA, Guan T, Kleinstein SH & Kaech SM Polycomb Repressive Complex 2-Mediated Chromatin Repression Guides Effector CD8(+) T Cell Terminal Differentiation and Loss of Multipotency. Immunity 46, 596–608 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Xiong D, Zhang L & Sun Z-J Targeting the epigenome to reinvigorate T cells for cancer immunotherapy. Military Medical Research 10, e59 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Philip M, Fairchild L, Sun L, Horste EL, Camara S, Shakiba M, et al. Chromatin states define tumour-specific T cell dysfunction and reprogramming. Nature 545, 452–456 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Kagoya Y, Nakatsugawa M, Yamashita Y, Ochi T, Guo T, Anczurowski M, et al. BET bromodomain inhibition enhances T cell persistence and function in adoptive immunotherapy models. J Clin Invest 126, 3479–3494 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.DuCote TJ, Song X, Naughton KJ, Chen F, Plaugher DR, Childress AR, et al. EZH2 Inhibition Promotes Tumor Immunogenicity in Lung Squamous Cell Carcinomas. Cancer Research Communications 4, 388–403 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Topper MJ, Vaz M, Chiappinelli KB, DeStefano Shields CE, Niknafs N, Yen R-WC, et al. Epigenetic Therapy Ties MYC Depletion to Reversing Immune Evasion and Treating Lung Cancer. Cell 171, 1284–1300 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Hellmann MD, Jänne PA, Opyrchal M, Hafez N, Raez LE, Gabrilovich DI, et al. Entinostat plus Pembrolizumab in Patients with Metastatic NSCLC Previously Treated with Anti–PD-(L)1 Therapy. Clinical Cancer Research 27, 1019–1028 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.He S, Liu Y, Meng L, Sun H, Wang Y, Ji Y, et al. Ezh2 phosphorylation state determines its capacity to maintain CD8(+) T memory precursors for antitumor immunity. Nat Commun 8, 2125 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Kradin RL, Boyle LA, Preffer FI, Callahan RJ, Barlai-Kovach M, Strauss HW, et al. Tumor-derived interleukin-2-dependent lymphocytes in adoptive immunotherapy of lung cancer. Cancer Immunol Immunother 24, 76–85 (1987). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kradin RL, Kurnick JT, Lazarus DS, Preffer FI, Dubinett SM, Pinto CE, et al. Tumour-infiltrating lymphocytes and interleukin-2 in treatment of advanced cancer. Lancet 1, 577–580 (1989). [DOI] [PubMed] [Google Scholar]
  • 80.Ratto GB, Zino P, Mirabelli S, Minuti P, Aquilina R, Fantino G, 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). [DOI] [PubMed] [Google Scholar]
  • 81.Rohaan MW, van den Berg JH, Kvistborg P & Haanen JBAG. Adoptive transfer of tumor-infiltrating lymphocytes in melanoma: a viable treatment option. Journal for ImmunoTherapy of Cancer 6, e102 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res 17, 4550–4557 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Rosenberg SA, Yannelli JR, Yang JC, Topalian SL, Schwartzentruber DJ, Weber JS, et al. Treatment of patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and interleukin 2. Journal of the National Cancer Institute 86, 1159–1166 (1994). [DOI] [PubMed] [Google Scholar]
  • 84.Medina T, Chesney JA, Whitman E, Kluger H, Thomas S, Sarnaik AA, et al. 776 Long-term efficacy and safety of lifileucel tumor-infiltrating lymphocyte (TIL) cell therapy in patients with advanced melanoma: a 4-year analysis of the C-144–01 study. Journal for ImmunoTherapy of Cancer 11, A873–A873 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Wermke M, Chesney JA, Whitman E, Kluger H, Thomas S, Sarnaik A, Kirkwood JM, Larkin J, Weber JS, Hamid O, Graf Finckenstein F, Chou J, Gastman B, Sulur G, Wu X, Shi W, Domingo-Musibay E. Long-term efficacy and patterns of response of lifileucel tumor-infiltrating lymphocyte (TIL) cell therapy in patients with advanced melanoma: A 4-year analysis of the C-144-01 study. Annals of Oncology 20 100589–100589 (2023). [Google Scholar]
  • 86.Schoenfeld AJ, Lee SM, Doger de Spéville B, Gettinger SN, Häfliger S, Sukari A, et al. Lifileucel, an Autologous Tumor-Infiltrating Lymphocyte Monotherapy, in Patients with Advanced Non–Small Cell Lung Cancer Resistant to Immune Checkpoint Inhibitors. Cancer Discovery 14, 1389–1402 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ben-Avi R, Farhi R, Ben-Nun A, Gorodner M, Greenberg E, Markel G, et al. Establishment of adoptive cell therapy with tumor infiltrating lymphocytes for non-small cell lung cancer patients. Cancer Immunology, Immunotherapy 67, 1221–1230 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Fernandez-Poma SM, Salas-Benito D, Lozano T, Casares N, Riezu-Boj J-I, Mancheño U, et al. Expansion of Tumor-Infiltrating CD8+ T cells Expressing PD-1 Improves the Efficacy of Adoptive T-cell Therapy. Cancer Research 77, 3672–3684 (2017). [DOI] [PubMed] [Google Scholar]
  • 89.Kodumudi KN, Siegel J, Weber AM, Scott E, Sarnaik AA & Pilon-Thomas S Immune Checkpoint Blockade to Improve Tumor Infiltrating Lymphocytes for Adoptive Cell Therapy. PLOS ONE 11, e0153053 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Emens LA, Romero PJ, Anderson AC, Bruno TC, Capitini CM, Collyar D, et al. Challenges and opportunities in cancer immunotherapy: a Society for Immunotherapy of Cancer (SITC) strategic vision. Journal for ImmunoTherapy of Cancer 12, e009063 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Yao X, Ahmadzadeh M, Lu Y-C, Liewehr DJ, Dudley ME, Liu E, et al. Levels of peripheral CD4+FoxP3+ regulatory T cells are negatively associated with clinical response to adoptive immunotherapy of human cancer. Blood 119, 5688–5696 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES