View Article

Abstract

The understanding about tumor-infiltrating lymphocytes (TIL) plays a monumental role in the treatment of cancer, creating a hitting spot in immuno-oncology. Apart from being used as a prognostic marker for other chemotherapy, TIL places its own importance in the treatment as well. TIL therapy uses autologous tumor-infiltrating lymphocytes extracted from the tumor following the expansion in an in vitro manner. After completing lymphodepletion, the patient was ready to receive the TIL therapy. Herein, we concisely describe the process of preparing TIL for therapy. Later, we emphasize the role of TIL in various cancers. And discuss the challenges, combination drugs, and future scope of TIL therapy.

Keywords

Tumor infiltrating lymphocytes therapy, tumor microenvironment, immuno-oncology, tumor, cancer.

Introduction

For the past twenty-five decades, the cancer was an unfathomable condition, in which the research is still ongoing to identify an unbeatable treatment to cure it [1]. Treating cancer patients with remission and metastasis is always a challenge in the field of cancer research. The existing treatments, such as surgery, chemotherapy, radiotherapy, and biologics, provide better results in cancer patients. But, they also result in early and late remission. These treatments only focus on the tumor cells, ignoring the complex tumor microenvironment (TME). This is why these treatments are not effective in treating cancer remission and metastasis [2]. Until 1940, cancer immunotherapy was not considered a treatment option by the medical community. After 1980, the immunotherapy era in cancer begins with lots of research [3]. Nowadays, cancer immunotherapy is much more interestingly expanded through the role of immune checkpoint inhibitors like anti-PD-L 1 (programmed cell death receptor 1/programmed cell death receptor ligand 1) and anti-CTLA-4 (cytotoxic T-lymphocyte-associated protein 4) agents, oncolytics virus therapy, cancer vaccines, cytokine therapy, immunomodulatory therapy  and most recently, adoptive cell transfer (ACT) therapy [4]. ACT therapy was classified into three types: tumor-infiltrating therapy (TIL), engineered T cell receptor (TCR)-T cell therapy, and chimeric antigen receptor (CAR)-T cell therapy. In recent days, CAR-T cell therapy has extensively been used for the treatment of hematological cancers such as lymphoma and leukemia. And turns the world treatment towards the development of ACT therapy. Through this, TIL therapy research is widely expanded. Comparatively, TIL therapy has many advantages over CAR-T cell therapy because of its large antigen specificity [5,6]. In ACT therapy, TIL transfer was considered as a treatment option for metastasis patients. In the study in the early 1980s by Rosenberg et al., they performed an excision of tumors from the mice and isolated and expanded their lymphocytes under an aseptic condition that opened the field of adoptive cell therapy (ACT) using tumor-infiltrating lymphocytes (TIL) [7]. After the success of expanding TIL, there is an insufficient number of lymphocytes in that culture. In order to increase the number, interleukin-2 (IL-2) was administered during the expansion period of lymphocytes in the cell suspension [8]. Later on, lymphokine-activated killer (LAK) cells with recombinant IL-2 provide a significant lytic action towards pulmonary metastases in mice [9]. In 1986, Rosenberg et al. concluded that the potent antitumor activity of TIL therapy over the LAK cells with IL-2 treatment against lung and liver metastases in mice [10]. Following the subsequent years, he identified that TIL activity was increased fivefold when co-administered with IL-2 and cyclophosphamide. That increases the overall response rate in advanced metastases melanoma patients [11,12]. And his work in non-myeloablative (NMA) lymphodeletion plays a monumental role in the entire development of TIL therapy [13]. Recently, TIL got FDA approval for treating metastatic melanoma [14]. As well, further developments of TIL are emerging in the prognosis and targeted therapy for solid tumors like breast cancer, lung cancer, colorectal cancer, liver cancer, and cervical cancer. In TIL therapy, tumor immune microenvironment (TIME) provides a significant part. It consists of mainly tumor cells surrounded by the immune cells along with the extracellular matrix. It was considered a biomarker for many cancer treatments. [15] The information about TIME was not completely understood due to its complexity and heterogenicity. From researchs, TIME laid the path for the invention of many immunotherapies. TIME was behind the reason for chemotherapeutic drug resistance through various mechanisms, such as activation of P-glycoprotein through the ATP-binding cassette pathway. It was responsible for the metastasis of cancer too. The presence of specific lymphocytes in the tumor microenvironment (TIME) is called tumor infiltrating lymphocytes [16]. Major components of the TIL architecture was composed of T cells, B cells, and NK cells which essentially contribute to tumor elimination. In T cells, there are various subtypes such as TCR???, TCR??? CD4? CD8? double positive cell, TCR??? CD4? CD8? double negative cell, CD8? TCR?? cell, CD3?  T cell and finally CD4?  TCR?? cells. In CD8? TCR?? subsets, they all together to produce high cytotoxic action towards the tumor. In CD4? TCR?? subsets, the T helper cells exhibit the role of CD8? cell activation and perform rapid lytic action. On the contrary, T regulatory (Treg) cells inhibit the activation of T cells through suppressing the cytokine production. In B cells, there are antigen presenting cells, plasma cells and B regulatory cells to preform the role of production and activation of antibodies [17].  Dysfunction of normal T cells leads to the growth of cancer. Due to the continuous exposure of T cells to tumor cells, this results T cell exhaustion and sensescence. Exhaustion reduces the cytolytic activity of the existing T cell [18]. Some other cells, such as tumor-associated macrophages, marrow-derived suppression cells, and tumor-associated neutrophils, contribute towards immunosuppression [19]. Due to the immunosuppression, normal existing, TIL is unable to produce its action in TME. But in the case of TIL therapy, it overcomes the immunosuppression and produces a lytic action.

Construction And Working Of Til:

In the process of construction of TIL, the primary steps involve excision of the existing tumor and rapid expansion of TIL. In metastasis patients, excision of tumor performed, and that excised tumor made into a slice to create a single cell suspension [20]. Then it was kept in a culture medium preinstalled with the T cell-stimulating growth factor such as IL-2. The selection of TIL for culture was based on the presence of interferon-alpha traces initially in the 1990s. But this became a major limiting step in the TIL expansion. Later on, the selection was cancelled and said that unselected TILs. The unselected TILs, also called young TILs, are those having a prominent lytic activity from the beginning towards the tumor. After 2-4 weeks, the culture medium grows to attain the necessary count of T cells, then it is subjected to rapid expansion [21]. During rapid expansion, the culture medium was immerged with IL-2, anti-CD3 (initial phase only), and irradiated feeder cells. All these were used to enhance the T cell growth quickly. It would take nearly 14 days.  After the success of complete TIL expansion, the secondary steps will begin with non-myeloablative (NMA) lymphodepletion. The execution of lymphodepletion takes the TIL therapy to the next step in its efficacy. It enhances the effect of TIL through removing the Treg cells from the TME and also improves the APC activity of other cells towards the TILs. The patient underwent NMA lymphodepletion through either chemotherapy or total body irradiation. The chemotherapy was done by the use of cyclophosphamide (60 mg/kg/day) and fludarabin (25 mg/m/day) for nearly two and five subsequent days, respectively [22].The completion of NMA lymphodepletion initiates the administration of expanded TIL along with a high dose of IL-2. IL-2 co-administration improves the cytolytic activity of TIL [23]. After the intravenous administration of TIL into the patient body, it travels through the blood to reach its target sites. Due to metastasis, it produced its antitumor effect in various area. By reaching its target site, it invades into the TME and try to recognise the neoantigen those expressed by the tumor itself. The presence of neoantigen, activate the autologous TIL to produce the cytolytic activity towards the tumor. Thereby, it decreases tumor size and produces a complete response in metastasis patients [24]. There is no reported adverse effect from the administration of expanded TIL. But from the administration, high doses of IL-2 produce grade 3 and grade 4 toxicities in clinical trials, and some reports recorded from the chemotherapy too. In order to reduce the toxicity, some studies instructed the subjects to take prophylactive medicine to prevent that effect [25]. The simple overview of constructing TIL was shown in Figure.1

       
            FIGURE-1.png
       

Role Of Til In Cancer:

TIL is being an option for the treatment of all solid tumors, especially metastases melanoma, ovarian cancer, and non small cell lung cancer. [26] There are lots of clinical trials regarding the TIL therapy/TIL combination therapy. Some of them are noted in Table 1. [27]

Metastases melanoma:

Melanoma is the malignancy of the skin in which it involves the melanocytes present in the basal epidermis [26]. Melanoma is the seventeenth leading cancer in the world, with actual incidence and death of 331,647 and 58,645 per year, respectively [28]. In the mid-1970s, the first chemotherapeutic agent used for metastases of melanoma was decabracine, considered a first-line treatment even though its response is low [29]. Later on, lots of treatment options are identified, such as radiation therapy and immunotherapy. Those patients having resistance towards chemotherapy and radiation therapy are subjected to immunotherapy and get a better response due to the tumor immunogenicity [30]. In 2015, immunotherapy became the first line treatment of metastases melanoma through the prominent increase in ORR from the usage of immune checkpoint inhibitors (ICI) such as anti-PD-1 (pembrolizumab and nivolumab) and anti-CTLA-4 (ipilimumab) agents and targeted therapy such as B-Raf serine/threonine kinase inhibitors BRAK/MEK inhibitors as compared with decarbacine [31-33]. Following years, the patient develops resistance to ICB and (BRAK)/ mitogen-activated protein kinase (MEK) inhibitors, which create an unmet medical need for refractory metastases in melanoma. Fortunately, that was fulfilled by the development of ACT using TIL.

The study conducted by Chesney J. et al. involving 153 metastasis melanoma patients who already failed with ICB agents was subjected to TIL therapy using Iifileucel (Ln-144). It produced overall response rate (ORR) of 31.4% with eight complete responses and forty partial responses in the patients. Through this, the efficacy and safety of Ln-144 were elucidated and got FDA approval under the indication for treating metastases in melanoma disease [14]. During the TIL therapy, it increases the peripheral blood count of neoantigens, those specific to the T cell. That increases the antitumor activity though stimulates the immune system in metastases melanoma patients and produces a better survival rate of more than 108 months [24]. Some studies reveal that the length of telomerase and the clonal type of lymphocyte in TIL determine its antitumor activity [34]. Despite all immunotherapy, TIL becomes the foremost treatment for metastasis melanoma patients.


Table 1 : Some ongoing clinical trial related to TIL therapy [27]

 

Cancer Type

Clinicaltrials.Gov Id

Clinical Phase

No.Of.

Subjects

Intervention

Result

 

 

 

 

 

 

 

 

Advanced melanoma

NCT05098184

Early phase-I (Recruiting)

50

Tumor Infiltrating Lymphocytes

N/A

NCT03991741

Phase-I

(Recruiting)

24

Autologous Tumor Infiltrating Lymphocytes and High-Dose Interleukin 2

N/A

NCT06120712

Phase-I/II

(Recruiting)

20

GC101 TIL

N/A

NCT05869539

Phase-I

(Recruiting)

10

Combination of Tumor-infiltrating lymphocyte transfer with ANV419

N/A

NCT02360579

Phase-II

(Active)

178

Lifileucel (LN-144)

N/A

 

 

Ovarian cancer

NCT05152797

Phase-I/II

(Not yet recruiting)

91

Autologous tumor-infiltrating lymphocytes

N/A

NCT03449108

Phase-II

(Active)

30

LN-145/LN-145-S1

N/A

 

 

Non small cell lung cancer

(NSCLC)

NCT03215810

Phase-I

(completed)

20

TIL+ Nivolumab

OR (3)

NCT05366478

Phase-I

(Recruiting)

15

LM103

N/A

NCT05576077

Phase-I

(Recruiting)

60

TBio-4101 and Pembrolizumab

N/A

Head and neck squamous cell carcinoma

NCT03083873

Phase-II

(completed)

64

LN-145/LN-145-S1

PFS (3.9 months), DCR (99.7%)

NCT03645928

Phase-II

(Recruiting)

178

LN-145-S1/ LN-145-S1/ LN-145/ LN-145 + Pembrolizumab

N/A

Breast cancer

NCT04111510

Phase-II

(completed)

6

LN-145

PD(5), SD(1)

Colorectal and breast cancer

NCT05576077

Phase-I

(Recruiting)

60

TBio-4101and pembrolizumab

N/A


N/A- Not Available, OR-Objective Response, PFS-Progression Free Survival, DCR-Disease Control Rate,

PD-Progressive Disease, SD-Stable Disease

Ovarian cancer:

Ovarian cancer (OC) was the most common cancer in women after breast cancer and cervix cancer in the world population. It nearly results in 206,839 deaths per year. [28] Debulking surgery along with platinum-paclitaxel chemotherapy was considered first-line treatment for OC. Due to the platinum resistance or platinum sensitivity, the remission of OC occurred in many patients. For them, the second-line therapy consisting of bevacizumab or a poly-ADP-ribose polymerase (PARP) inhibitor was used. But it does not increase the overall survival rate. [35-37] With the drawbacks of existing therapy results in early remission of OC, create a demand for the newer treatment. [38] Implementation of immunotherapy using ICB (anti-PD-1/PD-L1 or anti-CTLA 4) and TIL is under investigation. [39] R.S. Freedman et al. conducted the TIL therapy with a low dose of IL-2 through interperitoneal administration. In this study, TIL mostly contains either CD3?, CD8?, CD4? or CD3?, CD4?, TCR??? T cell lines. That resulted in tumor regression and increased the overall response rate in the patients. Due to the low dose of IL-2, it reduces the toxicity caused by high dose administration [40]. Sometimes, TIL therapy produces low results due to the activation of immunosuppression by the LAG3/MHC-II and PD-1/PD-L2 pathways. One of the major limitations in treating OC with immunotherapy was its low mutational burden and low neoantigen specificity [41]. In order to increase the antitumor activity of TIL, Amaria R. et al. used CD3 and 4-1BB in the process of harvesting TIL. And the expanded TIL had an increased number of CD8? cells with potent activity. The result had PFS as 2.53 months and overall survival (OS) as 18.86 with a lack of ORR [42]. Later on, combination therapy of TIL with carboplastin plus paclitaxel with or without interferon-alpha (IFN-?) was conducted. The immunosuppression caused by chemotherapy administration increased the activity of TIL and resulted in OR of 83% [43]. Still, there are many TIL combinational therapies under investigation for the treatment of OC.

Non small cell lung cancer:

Non-small cell lung carcinoma (NSCLC) is the leading cancer among all, with the incidence of 2,480,301 and mortality of 1,817,172 people per year [28]. Naturally, NSCLC has a high tumor mutational burden (TMB) and neantigen specificity. Due to these characteristics, immunotherapy provides a better treatment result for NSCLC than other treatments such as chemotherapy and radiation therapy. In immunotherapy, ICB (anti-PD-1/PD-L1) was considered the first-line treatment for it. If the patient gets resistance to the anti-PD-1/PD-L1 agent, then they are instructed to take the combination therapy of anti-PD-1/PD-L1 with platinum doublet chemotherapy. The research about the role of TIL in NSCLC shows its significant response to the refractory and metastasis patients. The recent development of ATL001, a product of clonal neoantigen reactive T cell (cNET) from the autologous TIL, performed its action by increasing the reactiveness of the immune system by overcoming the TIME heterogenicity. [44] Massarelli et al. perform an autologous TIL therapy (Ln-145) in NSCLC patients. It produces an ORR of 25% and signifies the role as well as efficacy of ln-145 for NSCLC. [45] Further, research about combination therapy of TIL with nivolumab results in two complete responses in NSCLC patients. Administration of nivolumab increases the antitumor activity of TIL; through this, it becomes able to recognize many mutations in tumors. [46] Schoenfeld A, Lee S, Paz-Ares L, et al. conducted monotherapy using Ln-145 for NSCLC that resulted in one complete response, five partial responses, and ORR of 21.4%. [47]

Other cancers:

Initially, TIL was used as a prognostic indicator to evaluate the clinical outcomes of treatment in cancers such as breast cancer, head and neck squamous cell carcinoma, and colorectal cancer. [48-50]. In breast cancer, TIL therapy is more significant for triple-negative breast cancer (TNBC) and human epidermal growth factor receptor 2 (HER2)-positive breast cancer. [51] In TNBC patients having a high level of T regulatory cells, those infiltrated into the tumor and resulted in immunosuppression tumor environment. Enriched CD8? cells in TIL overcome that immunosuppression and perform its antitumor activity. [52] Zacharakis et al. demonstrated the use of TIL therapy for metastases in breast cancer. Through his study, it gives a new path for TIL therapy in breast cancer. [53] In heads and neck squamous cell carcinoma, TIL therapy is still in the developmenal stage. The virus proteins on the cell surface of HPV and EPV were considered the target for TIL therapy. [48] A recent clinical trial shows the efficacy of TIL when combined with pembrolizumab for treating metastases. The patient reveals 42.9% ORR. [54] Now, LN-145/145-S1 drug treatment for head and neck squamous cell carcinoma is under phase 2 trial. In colorectal cancer, the CD8? and CD4? in TIL are initially considered for their biomarker roles. Later, usage of TIL for metastases in colorectal cancer shows promising clinical effects. Especially for KRAS mutant metastases patients, CD8-specific T cells target the KRAS G12D mutant [55]. Now, a phase I study of TBio-4101 with pembrolizumab for treating metastasis colorectal is still active and expected to have a better result in treating colorectal cancer patients.

Challenges In Til :

Implementing certain types of novel technologies and new strategies in the health care sector has been a great challenge. However, TILs also have a lot of consequences in their selection, isolation, differentiation, and development. Plesca et al. noted that the therapeutic strategy, using adoptive cellular transfer of TIL through the expansion of immune cells away from their suppressive TME [56], leads to the harvest and isolation of TILs from a resected tumor, expansion in an in vitro manner, and then reinfusion back into the patient. Several animal studies demonstrated the potential of isolated immune cells to mediate tumor regression [57]. Due to the complexity of TIL, it requires only highly educated and trained personnel. And resulted in limited accessibility, which became the major drawback in the implementation of TIL [58]. Unfortunately, these existing TILs are rendered ineffective because of the pre-infiltrated Treg cells in TME [59]. The tumor heterogeneity develops a fault in the recognizing ability of T cells towards tumor antigen. Also some other factors related to tumors are their location, blood flow, and costimulatory cytokine levels being a challenge in TIL therapy in order to provide the same efficacy [60]. Nowadays, much of the intratumoral heterogeneity is a concern in metastatic diseases that have been found and studied in multiple cancer types and may help to explain some forms of resistance mechanisms [61]. This TIL also has disadvantages, like manufacturing challenges. However, their expansion and persistence may not be produced in the sufficient members to achieve an effective anti-tumor response. So, Chensey et al. studied the duration, efficacy, and safety of lifileucel and denoted the manufacturing and infusion methods [14]. But standardizing the expansion technique of TIL is much more complex and difficult. Prolonged exposure to tumor antigens can lead to TIL exhaustion and reduced functionality. Upon persistent antigen stimulation, T cells show a gradual decrease in their effector functions known as T cell exhaustion, which is characterized by a decrease in proliferative and cytolytic capacity and upregulation of multiple inhibitory signals, including PD1, LAG-3, CD160, 2B4, TIM-3, and TIGIT [62]. Although characterizing the exhausted phenotype, these T cells retain their cytolytic and proliferative capacity [19]. There are patient-related challenges such as immune system activity and pre-exposed treatments, especially chemotherapy, being a challenge to get a prominent result from TIL [12]. So at last, the cost of the TIL therapy creates challenges in developing countries like India. TIL therapy is a highly payable therapy because of its own complexity of manufacturing process. Retel et al. compared the cost of TIL therapy and ipilumumab and concluded that the probability of TILs is most costlier than ipilumumab. The treatment was estimated approximately to be around €62,000 in the Netherlands, which was the first tested TIL therapy for melanoma patients, and in Europe it was estimated around €60,000. According to Indian currency, it nearly cost around 55–60 lakhs [63]. By overcoming these types of challenges wisely, TILs can be easily implemented in all types of tumor patients everywhere.

Combination Therapy Of Til:

The development of a healthy immune response, which is marked by certain immunological checkpoints that should stop undesirable and damaging self-directed activities such as autoimmunity. There are many combination therapies that combine with the TILs: ICI, BRAF, and others. Immune checkpoint inhibitors in cancer immunology have made some notable advances in the past few years. Tumor regresssion was acknowledged in cancer patients who underwent CTLA-4 and PD-1 or PD-L1 blockade treatment [64]. CTLA-4 blockade is an attractive immunotherapeutic strategy to significantly enhance the effector T cell-mediated antitumor immunity [65]. The combination of anti-PD-1 and TIL has the potential to harness a potent pool of tumor-reactive T cells and overcome their inhibition in vivo. With this in mind, we combined the anti-PD-1 blocking antibody, Nivolumab, with TIL therapy in anti-PD-1 and TIL treatment naïve patients with metastatic melanoma [66]. Some studies reported the successful combination of anti-PD-1 and TIL transfer in anti-PD-1 refractory metastatic non-small cell lung cancer patients with an initial reduction in tumor burden and a median change in target lesion size [46]. Overall, the combination therapy of ACT with TIL expanded ex vivo using anti-4-1BB along with systemically administered anti-PD-1 was found to be a safe and feasible treatment for metastatic melanoma patients [67]. Presently, the most widely accepted hypothesis is that tumors with more mutations are likely to generate more neoepitopes, which can be recognized by TIL. Checkpoint-blocking antibodies reactivate these T cells in vivo and thus lead to tumor regressions [68]. Also, not only the immune checkpoint inhibitors are the essential combination for TIL. The BRAF gene plays an important role in cell growth and differentiation. In some cancers, BRAF inhibitors change the cascade of ERK/MSPK signals, resulting in increased cell proliferation [69]. The combination of TIL and BRAF inhibitors may provide significant results in upcoming research [70-72]. In a clinical trial of TIL therapy combined with adenovirses in the treatment of metastatic melanoma, 5 patients achieved an objective response, of which 3 achieved a complete response. The virus can combat tumor immunosuppression by producing cytokines that promote the anti-tumor effects of TIL [73]. Preclinical mouse modeling demonstrated the extended persistence of adoptively transferred gp100-reactive T cells when combined with antigen-specific dendritic cell (DC) vaccination, resulting in superior tumor control [74]. To investigate the combination of lactoferrin-derived lytic peptide (LTX-315) and ACT using TIL, the combination of LTX-315 with TIL can induce a tumor-specific immune response in patients with soft tissue sarcoma. Further, the combination of LTX-315 and ACT using TIL is not only feasible and tolerable, but tumor-specific T cells can be expanded in vitro from tumors that have been pretreated by LTX-315 [75]. Nevertheless, LIFILEUCEL produced durable responses and a favorable safety profile across subgroups of heavily pretreated patients with high tumor burden, regardless of age, BRAF mutation status, PD-L1 status, baseline ECOGPS status, and presence of liver or brain lesions at baseline, which supports a potential benefit for a broad population of patients with melanoma [71]. So most of the new cancer treatment therapies are combined, producing some good results and increasing the quality of life of the patients.

CONCLUSION:

All existing therapy provides only a short span of disease-free life in cancer patients. Some of them experience either remission or resistance. This creates unfulfilled demands for future research. The emergence of immunotherapy in oncology has always played a magnificent role in the treatment option. Likewise, in ACT, using TIL showcases its efficiency in treating metastases in melanoma patients and gained FDA approval for it. This was the first approval related to TIL therapy. Being apart from other therapies, TIL therapy results in treating resistance patients from prior chemotherapy and ICI therapy. It comparatively increases the disease-free survival time and has a chance to become either a first-line or second-line treatment for cancers, especially for solid tumors. There are lots of clinical trials related to the TIL and its combination treatment for various types of cancer that are still ongoing and expect promising results. In the future, the development of TIL therapy and further research about TIME role in TIL therapy contribute to valuable insights in immuno-oncology.

Future Scope:

Due to the promising results from an existing clinical trial showing a complete tumor-free survival of advanced metastasis patients from TIL therapy. But this is not for all patients; some of them experience a partial reduction in the tumor size and do not well respond to it. If eliminating the challenges and unsolved question about TIL therapy may provide a foremost treatment for cancer in the future. And having a probability to become second-line treatment for all cancer tumor patients.

Declarations:

Ethics approval and consent to participate - Not applicable

Consent for publication - Not applicable

Availability of data and materials - Not applicable

Competing interests - The authors declare that they have no competing interests

Funding - Not applicable

Authors’ contribution:

TK, AS and DK analyse the recent clinical trial information and gathering information regarding the title of challenges and combination therapy of TIL.

Acknowledgements - Not applicable

REFERENCES

  1. Milestones in cancer research and discovery. (Viewed on 1st july 2024) https://www.cancer.gov/research/progress/250-years-milestones
  2. Lin B, Du L, Li H, Zhu X, Cui L, Li X. Tumor-infiltrating lymphocytes: Warriors fight against tumors powerfully. Biomed Pharmacother. 2020 Dec;132:110873. doi: 10.1016/j.biopha.2020.110873. Epub 2020 Oct 14. PMID: 33068926.
  3. Eno J. Immunotherapy Through the Years. J Adv Pract Oncol. 2017 Nov-Dec;8(7):747-753. Epub 2017 Nov 1. PMID: 30333937; PMCID: PMC6188092.
  4. Weber EW, Maus MV, Mackall CL. The Emerging Landscape of Immune Cell Therapies. Cell. 2020 Apr 2;181(1):46-62. doi: 10.1016/j.cell.2020.03.001. PMID: 32243795; PMCID: PMC8900215.
  5. Granhøj JS, Witness Præst Jensen A, Presti M, Met Ö, Svane IM, Donia M. Tumor-infiltrating lymphocytes for adoptive cell therapy: recent advances, challenges, and future directions. Expert Opin Biol Ther. 2022 May;22(5):627-641. doi: 10.1080/14712598.2022.2064711. Epub 2022 Apr 20. PMID: 35414331.
  6. Zhang P, Zhang G, Wan X. Challenges and new technologies in adoptive cell therapy. J Hematol Oncol. 2023 Aug 18;16(1):97. doi: 10.1186/s13045-023-01492-8. PMID: 37596653; PMCID: PMC10439661.
  7. Yron I, Wood TA Jr, Spiess PJ, Rosenberg SA. In vitro growth of murine T cells. V. The isolation and growth of lymphoid cells infiltrating syngeneic solid tumors. J Immunol. 1980 Jul;125(1):238-45. PMID: 6966652.
  8. Eberlein TJ, Rosenstein M, Rosenberg SA. Regression of a disseminated syngeneic solid tumor by systemic transfer of lymphoid cells expanded in interleukin 2. J Exp Med. 1982 Aug 1;156(2):385-97. doi: 10.1084/jem.156.2.385. PMID: 6980254; PMCID: PMC2186754.
  9. Mulé JJ, Shu S, Schwarz SL, Rosenberg SA. Adoptive immunotherapy of established pulmonary metastases with LAK cells and recombinant interleukin-2. Science. 1984 Sep 28;225(4669):1487-9. doi: 10.1126/science.6332379. PMID: 6332379
  10. Rosenberg SA, Spiess P, Lafreniere R. A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science. 1986 Sep 19;233(4770):1318-21. doi: 10.1126/science.3489291. PMID: 3489291.
  11. Rosenberg SA, Packard BS, Aebersold PM, Solomon D, Topalian SL, Toy ST, Simon P, Lotze MT, Yang JC, Seipp CA, et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N Engl J Med. 1988 Dec 22;319(25):1676-80. doi: 10.1056/NEJM198812223192527. PMID: 3264384.
  12. Rosenberg SA, Yannelli JR, Yang JC, Topalian SL, Schwartzentruber DJ, Weber JS, Parkinson DR, Seipp CA, Einhorn JH, White DE. Treatment of patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and interleukin 2. J Natl Cancer Inst. 1994 Aug 3;86(15):1159-66. doi: 10.1093/jnci/86.15.1159. PMID: 8028037.
  13. Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, Citrin DE, Restifo NP, Robbins PF, Wunderlich JR, Morton KE, Laurencot CM, Steinberg SM, White DE, Dudley ME. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res. 2011 Jul 1;17(13):4550-7. doi: 10.1158/1078-0432.CCR-11-0116. Epub 2011 Apr 15. PMID: 21498393; PMCID: PMC3131487.
  14. Chesney J, Lewis KD, Kluger H, Hamid O, Whitman E, Thomas S, Wermke M, Cusnir M, Domingo-Musibay E, Phan GQ, Kirkwood JM, Hassel JC, Orloff M, Larkin J, Weber J, Furness AJS, Khushalani NI, Medina T, Egger ME, Graf Finckenstein F, Jagasia M, Hari P, Sulur G, Shi W, Wu X, Sarnaik A. 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. 2022 Dec;10(12):e005755. doi: 10.1136/jitc-2022-005755. PMID: 36600653; PMCID: PMC9748991.
  15. Mao X, Xu J, Wang W, Liang C, Hua J, Liu J, Zhang B, Meng Q, Yu X, Shi S. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: new findings and future perspectives. Mol Cancer. 2021 Oct 11;20(1):131. doi: 10.1038/s41577-018-0044-0. PMID: 30057419.
  16. Khalaf K, Hana D, Chou JT, Singh C, Mackiewicz A, Kaczmarek M. Aspects of the Tumor Microenvironment Involved in Immune Resistance and Drug Resistance. Front Immunol. 2021 May 27;12:656364. doi: 10.3389/fimmu.2021.656364. PMID: 34122412; PMCID: PMC8190405.
  17. Paijens ST, Vledder A, de Bruyn M, Nijman HW. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell Mol Immunol. 2021 Apr;18(4):842-859. doi: 10.1038/s41423-020-00565-9. Epub 2020 Nov 2. PMID: 33139907; PMCID: PMC8115290.
  18. Borst J, Ahrends T, B?ba?a N, Melief CJM, Kastenmüller W. CD4+ T cell help in cancer immunology and immunotherapy. Nat Rev Immunol. 2018 Oct;18(10):635-647. doi: 10.1038/s41577-018-0044-0. PMID: 30057419.
  19. Zhao Y, Shao Q, Peng G. Exhaustion and senescence: two crucial dysfunctional states of T cells in the tumor microenvironment. Cell Mol Immunol. 2020 Jan;17(1):27-35. doi: 10.1038/s41423-019-0344-8. Epub 2019 Dec 18. PMID: 31853000; PMCID: PMC6952436.
  20. Lu C, Rong D, Zhang B, Zheng W, Wang X, Chen Z, Tang W. Current perspectives on the immunosuppressive tumor microenvironment in hepatocellular carcinoma: challenges and opportunities. Mol Cancer. 2019 Aug 29;18(1):130. doi: 10.1186/s12943-019-1047-6. PMID: 31464625; PMCID: PMC6714090.
  21. Kumar A, Watkins R, Vilgelm AE. Cell Therapy With TILs: Training and Taming T Cells to Fight Cancer. Front Immunol. 2021 Jun 1;12:690499. doi: 10.3389/fimmu.2021.690499. PMID: 34140957; PMCID: PMC8204054.
  22. Wickström S, Lövgren T. Expansion of Tumor-Infiltrating Lymphocytes from Melanoma Tumors. Methods Mol Biol. 2019;1913:105-118. doi: 10.1007/978-1-4939-8979-9_7. PMID: 30666601.
  23. Zhao Y, Deng J, Rao S, Guo S, Shen J, Du F, Wu X, Chen Y, Li M, Chen M, Li X, Li W, Gu L, Sun Y, Zhang Z, Wen Q, Xiao Z, Li J. Tumor Infiltrating Lymphocyte (TIL) Therapy for Solid Tumor Treatment: Progressions and Challenges. Cancers (Basel). 2022 Aug 27;14(17):4160. doi: 10.3390/cancers14174160. PMID: 36077696; PMCID: PMC9455018.
  24. Grimm EA, Mazumder A, Zhang HZ, Rosenberg SA. Lymphokine-activated killer cell phenomenon. Lysis of natural killer-resistant fresh solid tumor cells by interleukin 2-activated autologous human peripheral blood lymphocytes. J Exp Med. 1982 Jun 1;155(6):1823-41. doi: 10.1084/jem.155.6.1823. PMID: 6176669; PMCID: PMC2186695.
  25. Van den Berg JH, Heemskerk B, van Rooij N, Gomez-Eerland R, Michels S, van Zon M, de Boer R, Bakker NAM, Jorritsma-Smit A, van Buuren MM, Kvistborg P, Spits H, Schotte R, Mallo H, Karger M, van der Hage JA, Wouters MWJM, Pronk LM, Geukes Foppen MH, Blank CU, Beijnen JH, Nuijen B, Schumacher TN, Haanen JBAG. Tumor infiltrating lymphocytes (TIL) therapy in metastatic melanoma: boosting of neoantigen-specific T cell reactivity and long-term follow-up. J Immunother Cancer. 2020 Aug;8(2):e000848. doi: 10.1136/jitc-2020-000848. PMID: 32753545; PMCID: PMC7406109.
  26. Hirai I, Funakoshi T, Kamijuku H, Fukuda K, Mori M, Sakurai M, Koda Y, Kato J, Mori T, Watanabe N, Noji S, Yaguchi T, Iwata T, Ohta S, Fujita T, Tanosaki R, Handa M, Okamoto S, Amagai M, Kawakami Y. Adoptive cell therapy using tumor-infiltrating lymphocytes for melanoma refractory to immune-checkpoint inhibitors. Cancer Sci. 2021 Aug;112(8):3163-3172. doi: 10.1111/cas.15009. Epub 2021 Jun 30. PMID: 34101300; PMCID: PMC8353905.
  27. Kazemi MH, Sadri M, Najafi A, Rahimi A, Baghernejadan Z, Khorramdelazad H, Falak R. Tumor-infiltrating lymphocytes for treatment of solid tumors: It takes two to tango? Front Immunol. 2022 Oct 28;13:1018962. doi: 10.3389/fimmu.2022.1018962. PMID: 36389779; PMCID: PMC9651159.
  28. Clinical studies around the world. Viewed on 04th july 2024. https://clinicaltrials.gov/
  29. Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, Jemal A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024 May-Jun;74(3):229-263. doi: 10.3322/caac.21834. Epub 2024 Apr 4. PMID: 38572751.
  30. Wilson MA, Schuchter LM. Chemotherapy for Melanoma. Cancer Treat Res. 2016;167:209-29. doi: 10.1007/978-3-319-22539-5_8. PMID: 26601864.
  31. Ralli M, Botticelli A, Visconti IC, Angeletti D, Fiore M, Marchetti P, Lambiase A, de Vincentiis M, Greco A. Immunotherapy in the Treatment of Metastatic Melanoma: Current Knowledge and Future Directions. J Immunol Res. 2020 Jun 28;2020:9235638. doi: 10.1155/2020/9235638. PMID: 32671117; PMCID: PMC7338969.
  32. Lugowska I, Teterycz P, Rutkowski P. Immunotherapy of melanoma. Contemp Oncol (Pozn). 2018 Mar;22(1A):61-67. doi: 10.5114/wo.2018.73889. Epub 2018 Mar 5. PMID: 29628796; PMCID: PMC5885078.
  33. Lipson EJ, Drake CG. Ipilimumab: an anti-CTLA-4 antibody for metastatic melanoma. Clin Cancer Res. 2011 Nov 15;17(22):6958-62. doi: 10.1158/1078-0432.CCR-11-1595. Epub 2011 Sep 7. PMID: 21900389; PMCID: PMC3575079.
  34. Babacan NA, Eroglu Z. Treatment Options for Advanced Melanoma After Anti-PD-1 Therapy. Curr Oncol Rep. 2020 Mar 20;22(4):38. doi: 10.1007/s11912-020-0894-z. PMID: 32198651.
  35. Inozume T. Adoptive cell transfer therapy for melanoma. Exp Dermatol. 2023 Mar;32(3):250-255. doi: 10.1111/exd.14707. Epub 2022 Nov 23. PMID: 36382355.
  36. Morand S, Devanaboyina M, Staats H, Stanbery L, Nemunaitis J. Ovarian Cancer Immunotherapy and Personalized Medicine. Int J Mol Sci. 2021 Jun 18;22(12):6532. doi: 10.3390/ijms22126532. PMID: 34207103; PMCID: PMC8234871.
  37. Foley OW, Rauh-Hain JA, del Carmen MG. Recurrent epithelial ovarian cancer: an update on treatment. Oncology (Williston Park). 2013 Apr;27(4):288-94, 298. PMID: 23781692.
  38. Cortez AJ, Tudrej P, Kujawa KA, Lisowska KM. Advances in ovarian cancer therapy. Cancer Chemother Pharmacol. 2018 Jan;81(1):17-38. doi: 10.1007/s00280-017-3501-8. Epub 2017 Dec 16. PMID: 29249039; PMCID: PMC5754410.
  39. Sarivalasis A, Morotti M, Mulvey A, Imbimbo M, Coukos G. Cell therapies in ovarian cancer. Ther Adv Med Oncol. 2021 Apr 22;13:17588359211008399. doi: 10.1177/17588359211008399. Erratum in: Ther Adv Med Oncol. 2021 Jun 17;13:17588359211028457. doi: 10.1177/17588359211028457. PMID: 33995591; PMCID: PMC8072818.
  40. Borella F, Ghisoni E, Giannone G, Cosma S, Benedetto C, Valabrega G, Katsaros D. Immune Checkpoint Inhibitors in Epithelial Ovarian Cancer: An Overview on Efficacy and Future Perspectives. Diagnostics (Basel). 2020 Mar 7;10(3):146. doi: 10.3390/diagnostics10030146. PMID: 32156035; PMCID: PMC7151145.
  41. Freedman RS, Edwards CL, Kavanagh JJ, Kudelka AP, Katz RL, Carrasco CH, Atkinson EN, Scott W, Tomasovic B, Templin S, et al. Intraperitoneal adoptive immunotherapy of ovarian carcinoma with tumor-infiltrating lymphocytes and low-dose recombinant interleukin-2: a pilot trial. J Immunother Emphasis Tumor Immunol. 1994 Oct;16(3):198-210. doi: 10.1097/00002371-199410000-00004. PMID: 7834119.
  42. Pedersen M, Westergaard MCW, Milne K, Nielsen M, Borch TH, Poulsen LG, Hendel HW, Kennedy M, Briggs G, Ledoux S, Nøttrup TJ, Andersen P, Hasselager T, Met Ö, Nelson BH, Donia M, Svane IM. Adoptive cell therapy with tumor-infiltrating lymphocytes in patients with metastatic ovarian cancer: a pilot study. Oncoimmunology. 2018 Sep 26;7(12):e1502905. doi: 10.1080/2162402X.2018.1502905. PMID: 30524900; PMCID: PMC6279323.
  43. Amaria R, Knisely A, Vining D, et al Efficacy and safety of autologous tumor-infiltrating lymphocytes in recurrent or refractory ovarian cancer, colorectal cancer, and pancreatic ductal adenocarcinoma,Journal for ImmunoTherapy of Cancer 2024;12:e006822. doi: 10.1136/jitc-2023-006822
  44. Kroep, J.R. et al. 729P Adoptive T-cell therapy during chemotherapy with or without peginterferon-? (IFN?) in patients with platinum sensitive recurrent epithelial ovarian cancer (EOC), Annals of Oncology, Volume 32, S732 - S733
  45. Robertson J, Salm M, Dangl M. Adoptive cell therapy with tumour-infiltrating lymphocytes: the emerging importance of clonal neoantigen targets for next-generation products in non-small cell lung cancer. Immunooncol Technol. 2019 Oct 9;3:1-7. doi: 10.1016/j.iotech.2019.09.003. PMID: 35757302; PMCID: PMC9216247.
  46. Massarelli. E. et al, P14.04 A Phase 2 Multicenter Study of Iovance Autologous Tumor Infiltrating Lymphocytes (TIL, LN-145) Cell Therapy in Patients With Metastatic NSCLC, Journal of Thoracic Oncology, Volume 16, Issue 10, S1012.
  47. Creelan BC, Wang C, Teer JK, Toloza EM, Yao J, Kim S, Landin AM, Mullinax JE, Saller JJ, Saltos AN, Noyes DR, Montoya LB, Curry W, Pilon-Thomas SA, Chiappori AA, Tanvetyanon T, Kaye FJ, Thompson ZJ, Yoder SJ, Fang B, Koomen JM, Sarnaik AA, Chen DT, Conejo-Garcia JR, Haura EB, Antonia SJ. Tumor-infiltrating lymphocyte treatment for anti-PD-1-resistant metastatic lung cancer: a phase 1 trial. Nat Med. 2021 Aug;27(8):1410-1418. doi: 10.1038/s41591-021-01462-y. Epub 2021 Aug 12. PMID: 34385708; PMCID: PMC8509078.
  48. Schoenfeld A, Lee S, Paz-Ares L, et al 458 First phase 2 results of autologous tumor-infiltrating lymphocyte (TIL; LN-145) monotherapy in patients with advanced, immune checkpoint inhibitor-treated, non-small cell lung cancer (NSCLC),Journal for ImmunoTherapy of Cancer 2021;9:doi: 10.1136/jitc-2021-SITC2021.458
  49. Loi, S. & Michiels, Stefan & Adams, S. & Loibl, S. & Budczies, Jan & Denkert, Carsten & Salgado, Renato. (2021). The journey of tumor infiltrating lymphocytes (TIL) as a biomarker in breast cancer: clinical utility in an era of checkpoint inhibition. Annals of Oncology. 32. 10.1016/j.annonc.2021.07.007.
  50. Almangush A, De Keukeleire S, Rottey S, Ferdinande L, Vermassen T, Leivo I, Mäkitie AA. Tumor-Infiltrating Lymphocytes in Head and Neck Cancer: Ready for Prime Time? Cancers (Basel). 2022 Mar 18;14(6):1558. doi: 10.3390/cancers14061558. PMID: 35326709; PMCID: PMC8946626.
  51. Zemanek T, Nova Z, Nicodemou A. Tumor-Infiltrating Lymphocytes and Adoptive Cell Therapy: State of the Art in Colorectal, Breast and Lung Cancer. Physiol Res. 2023 Oct 27;72(S3):S209-S224. doi: 10.33549/physiolres.935155. PMID: 37888965; PMCID: PMC10669950.
  52. Kazemi MH, Sadri M, Najafi A, Rahimi A, Baghernejadan Z, Khorramdelazad H, Falak R. Tumor-infiltrating lymphocytes for treatment of solid tumors: It takes two to tango? Front Immunol. 2022 Oct 28;13:1018962. doi: 10.3389/fimmu.2022.1018962. PMID: 36389779; PMCID: PMC9651159.
  53. Li R, Cao L. The role of tumor-infiltrating lymphocytes in triple-negative breast cancer and the research progress of adoptive cell therapy. Front Immunol. 2023 May 18;14:1194020. doi: 10.3389/fimmu.2023.1194020. PMID: 37275874; PMCID: PMC10233026.
  54. Zacharakis N, Huq LM, Seitter SJ, Kim SP, Gartner JJ, Sindiri S, et al. Breast cancers are immunogenic: immunologic analyses and a phase II pilot clinical trial using mutation-reactive autologous lymphocytes. J Clin Oncol (2022) 40(16):1741–54. doi: 10.1200/jco.21.02170
  55. O’Malley D, Lee S, Psyrri A, Sukari A, Thomas S, Wenham R, et al. 492 phase 2 efficacy and safety of autologous tumor-infiltrating lymphocyte (Til) cell therapy in combination with pembrolizumab in immune checkpoint inhibitor-naïve patients with advanced cancers. BMJ Specialist J (2021) 9:523–24.
  56. Tran E, Robbins PF, Lu YC, Prickett TD, Gartner JJ, Jia L, Pasetto A, Zheng Z, Ray S, Groh EM, Kriley IR, Rosenberg SA. T-Cell Transfer Therapy Targeting Mutant KRAS in Cancer. N Engl J Med. 2016 Dec 8;375(23):2255-2262. doi: 10.1056/NEJMoa1609279. PMID: 27959684; PMCID: PMC5178827.
  57. Plesca I, Tunger A, Müller L, Wehner R, Lai X, Grimm MO, Rutella S, Bachmann M, Schmitz M. Characteristics of Tumor-Infiltrating Lymphocytes Prior to and During Immune Checkpoint Inhibitor Therapy. Front Immunol. 2020 Mar 4;11:364. doi: 10.3389/fimmu.2020.00364. PMID: 32194568; PMCID: PMC7064638.
  58. Eberlein TJ, Rosenstein M, Rosenberg SA. Regression of a disseminated syngeneic solid tumor by systemic transfer of lymphoid cells expanded in interleukin 2. J Exp Med. 1982 Aug 1;156(2):385-97. doi: 10.1084/jem.156.2.385. PMID: 6980254; PMCID: PMC2186754.
  59. Brambilla E, Le Teuff G, Marguet S, Lantuejoul S, Dunant A, Graziano S, Pirker R, Douillard JY, Le Chevalier T, Filipits M, Rosell R, Kratzke R, Popper H, Soria JC, Shepherd FA, Seymour L, Tsao MS. Prognostic Effect of Tumor Lymphocytic Infiltration in Resectable Non-Small-Cell Lung Cancer. J Clin Oncol. 2016 Apr 10;34(11):1223-30. doi: 10.1200/JCO.2015.63.0970. Epub 2016 Feb 1. PMID: 26834066; PMCID: PMC4872323.
  60. Whiteside TL. The tumor microenvironment and its role in promoting tumor growth. Oncogene. 2008 Oct 6;27(45):5904-12. doi: 10.1038/onc.2008.271. PMID: 18836471; PMCID: PMC3689267.
  61. Chen N, Li X, Chintala NK, Tano ZE, Adusumilli PS. Driving CARs on the uneven road of antigen heterogeneity in solid tumors. Curr Opin Immunol. 2018 Apr;51:103-110. doi: 10.1016/j.coi.2018.03.002. Epub 2018 Mar 16. PMID: 29554494; PMCID: PMC5943172.
  62. Zhang J, Fujimoto J, Zhang J, Wedge DC, Song X, Zhang J, Seth S, Chow CW, Cao Y, Gumbs C, Gold KA, Kalhor N, Little L, Mahadeshwar H, Moran C, Protopopov A, Sun H, Tang J, Wu X, Ye Y, William WN, Lee JJ, Heymach JV, Hong WK, Swisher S, Wistuba II, Futreal PA. Intratumor heterogeneity in localized lung adenocarcinomas delineated by multiregion sequencing. Science. 2014 Oct 10;346(6206):256-9. doi: 10.1126/science.1256930. PMID: 25301631; PMCID: PMC4354858.
  63. Egelston CA, Avalos C, Tu TY, Simons DL, Jimenez G, Jung JY, Melstrom L, Margolin K, Yim JH, Kruper L, Mortimer J, Lee PP. Human breast tumor-infiltrating CD8+ T cells retain polyfunctionality despite PD-1 expression. Nat Commun. 2018 Oct 16;9(1):4297. doi: 10.1038/s41467-018-06653-9. PMID: 30327458; PMCID: PMC6191461.
  64. Retèl VP, Steuten LMG, Geukes Foppen MH, Mewes JC, Lindenberg MA, Haanen JBAG, van Harten WH. Early cost-effectiveness of tumor infiltrating lymphocytes (TIL) for second line treatment in advanced melanoma: a model-based economic evaluation. BMC Cancer. 2018 Sep 15;18(1):895. doi: 10.1186/s12885-018-4788-5. PMID: 30219040; PMCID: PMC6139174.
  65. Ding W, Xu X, Qian Y, Xue W, Wang Y, Du J, Jin L, Tan Y. Prognostic value of tumor-infiltrating lymphocytes in hepatocellular carcinoma: A meta-analysis. Medicine (Baltimore). 2018 Dec;97(50):e13301. doi: 10.1097/MD.0000000000013301. PMID: 30557978; PMCID: PMC6320107.
  66. Peggs KS, Quezada SA, Chambers CA, Korman AJ, Allison JP. Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. J Exp Med. 2009 Aug 3;206(8):1717-25. doi: 10.1097/MD.0000000000013301. PMID: 30557978; PMCID: PMC6320107.
  67. Mullinax JE, Hall M, Prabhakaran S, Weber J, Khushalani N, Eroglu Z, Brohl AS, Markowitz J, Royster E, Richards A, Stark V, Zager JS, Kelley L, Cox C, Sondak VK, Mulé JJ, Pilon-Thomas S, Sarnaik AA. Combination of Ipilimumab and Adoptive Cell Therapy with Tumor-Infiltrating Lymphocytes for Patients with Metastatic Melanoma. Front Oncol. 2018 Mar 2;8:44. doi: 10.3389/fonc.2018.00044. PMID: 29552542; PMCID: PMC5840208.
  68. Cafri G, Gartner JJ, Zaks T, Hopson K, Levin N, Paria BC, Parkhurst MR, Yossef R, Lowery FJ, Jafferji MS, Prickett TD, Goff SL, McGowan CT, Seitter S, Shindorf ML, Parikh A, Chatani PD, Robbins PF, Rosenberg SA. mRNA vaccine-induced neoantigen-specific T cell immunity in patients with gastrointestinal cancer. J Clin Invest. 2020 Nov 2;130(11):5976-5988. doi: 10.1172/JCI134915. PMID: 33016924; PMCID: PMC7598064.
  69. Klebanoff CA, Gattinoni L, Restifo NP. CD8+ T-cell memory in tumor immunology and immunotherapy. Immunol Rev. 2006 Jun;211:214-24. doi: 10.1111/j.0105-2896.2006.00391.x. PMID: 16824130; PMCID: PMC1501075.
  70. Subbiah V, Baik C, Kirkwood JM. Clinical Development of BRAF plus MEK Inhibitor Combinations. Trends Cancer. 2020 Sep;6(9):797-810. doi: 10.1111/j.0105-2896.2006.00391.x. PMID: 16824130; PMCID: PMC1501075.
  71. Sloot S, Zager JS, Kudchadkar RR, Messina JL, Benedict JJ, Gonzalez RJ, DeConti R, Turner LM, McCardle T, Smalley KS, Weber JS, Sondak VK, Gibney GT. BRAF inhibition for advanced locoregional BRAF V600E mutant melanoma: a potential neoadjuvant strategy. Melanoma Res. 2016 Feb;26(1):83-7. doi: 10.1097/CMR.0000000000000214. PMID: 26731560.
  72. Cooper ZA, Frederick DT, Juneja VR, Sullivan RJ, Lawrence DP, Piris A, Sharpe AH, Fisher DE, Flaherty KT, Wargo JA. BRAF inhibition is associated with increased clonality in tumor-infiltrating lymphocytes. Oncoimmunology. 2013 Oct 1;2(10):e26615. doi: 10.4161/onci.26615. Epub 2013 Oct 15. PMID: 24251082; PMCID: PMC3827093
  73. Sosman JA, Kim KB, Schuchter L, Gonzalez R, Pavlick AC, Weber JS, McArthur GA, Hutson TE, Moschos SJ, Flaherty KT, Hersey P, Kefford R, Lawrence D, Puzanov I, Lewis KD, Amaravadi RK, Chmielowski B, Lawrence HJ, Shyr Y, Ye F, Li J, Nolop KB, Lee RJ, Joe AK, Ribas A. Survival in BRAF V600-mutant advanced melanoma treated with vemurafenib. N Engl J Med. 2012 Feb 23;366(8):707-14. doi: 10.1056/NEJMoa1112302. PMID: 22356324; PMCID: PMC3724515
  74. Khammari A, Nguyen JM, Saint-Jean M, Knol AC, Pandolfino MC, Quereux G, Brocard A, Peuvrel L, Saiagh S, Bataille V, Limacher JM, Dreno B. Adoptive T cell therapy combined with intralesional administrations of TG1042 (adenovirus expressing interferon-?) in metastatic melanoma patients. Cancer Immunol Immunother. 2015 Jul;64(7):805-15. doi: 10.1007/s00262-015-1691-7. Epub 2015 Apr 7. PMID: 25846669; PMCID: PMC11029588.
  75. Lou Y, Wang G, Lizée G, Kim GJ, Finkelstein SE, Feng C, Restifo NP, Hwu P. Dendritic cells strongly boost the antitumor activity of adoptively transferred T cells in vivo. Cancer Res. 2004 Sep 15;64(18):6783-90. doi: 10.1158/0008-5472.CAN-04-1621. PMID: 15374997; PMCID: PMC2241750.
  76. Donia M, Junker N, Ellebaek E, Andersen MH, Straten PT, Svane IM. Characterization and comparison of 'standard' and 'young' tumour-infiltrating lymphocytes for adoptive cell therapy at a Danish translational research institution. Scand J Immunol. 2012 Feb;75(2):157-67. doi: 10.1111/j.1365-3083.2011.02640.x. PMID: 21955245

Reference

  1. Milestones in cancer research and discovery. (Viewed on 1st july 2024) https://www.cancer.gov/research/progress/250-years-milestones
  2. Lin B, Du L, Li H, Zhu X, Cui L, Li X. Tumor-infiltrating lymphocytes: Warriors fight against tumors powerfully. Biomed Pharmacother. 2020 Dec;132:110873. doi: 10.1016/j.biopha.2020.110873. Epub 2020 Oct 14. PMID: 33068926.
  3. Eno J. Immunotherapy Through the Years. J Adv Pract Oncol. 2017 Nov-Dec;8(7):747-753. Epub 2017 Nov 1. PMID: 30333937; PMCID: PMC6188092.
  4. Weber EW, Maus MV, Mackall CL. The Emerging Landscape of Immune Cell Therapies. Cell. 2020 Apr 2;181(1):46-62. doi: 10.1016/j.cell.2020.03.001. PMID: 32243795; PMCID: PMC8900215.
  5. Granhøj JS, Witness Præst Jensen A, Presti M, Met Ö, Svane IM, Donia M. Tumor-infiltrating lymphocytes for adoptive cell therapy: recent advances, challenges, and future directions. Expert Opin Biol Ther. 2022 May;22(5):627-641. doi: 10.1080/14712598.2022.2064711. Epub 2022 Apr 20. PMID: 35414331.
  6. Zhang P, Zhang G, Wan X. Challenges and new technologies in adoptive cell therapy. J Hematol Oncol. 2023 Aug 18;16(1):97. doi: 10.1186/s13045-023-01492-8. PMID: 37596653; PMCID: PMC10439661.
  7. Yron I, Wood TA Jr, Spiess PJ, Rosenberg SA. In vitro growth of murine T cells. V. The isolation and growth of lymphoid cells infiltrating syngeneic solid tumors. J Immunol. 1980 Jul;125(1):238-45. PMID: 6966652.
  8. Eberlein TJ, Rosenstein M, Rosenberg SA. Regression of a disseminated syngeneic solid tumor by systemic transfer of lymphoid cells expanded in interleukin 2. J Exp Med. 1982 Aug 1;156(2):385-97. doi: 10.1084/jem.156.2.385. PMID: 6980254; PMCID: PMC2186754.
  9. Mulé JJ, Shu S, Schwarz SL, Rosenberg SA. Adoptive immunotherapy of established pulmonary metastases with LAK cells and recombinant interleukin-2. Science. 1984 Sep 28;225(4669):1487-9. doi: 10.1126/science.6332379. PMID: 6332379
  10. Rosenberg SA, Spiess P, Lafreniere R. A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science. 1986 Sep 19;233(4770):1318-21. doi: 10.1126/science.3489291. PMID: 3489291.
  11. Rosenberg SA, Packard BS, Aebersold PM, Solomon D, Topalian SL, Toy ST, Simon P, Lotze MT, Yang JC, Seipp CA, et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N Engl J Med. 1988 Dec 22;319(25):1676-80. doi: 10.1056/NEJM198812223192527. PMID: 3264384.
  12. Rosenberg SA, Yannelli JR, Yang JC, Topalian SL, Schwartzentruber DJ, Weber JS, Parkinson DR, Seipp CA, Einhorn JH, White DE. Treatment of patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and interleukin 2. J Natl Cancer Inst. 1994 Aug 3;86(15):1159-66. doi: 10.1093/jnci/86.15.1159. PMID: 8028037.
  13. Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, Citrin DE, Restifo NP, Robbins PF, Wunderlich JR, Morton KE, Laurencot CM, Steinberg SM, White DE, Dudley ME. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res. 2011 Jul 1;17(13):4550-7. doi: 10.1158/1078-0432.CCR-11-0116. Epub 2011 Apr 15. PMID: 21498393; PMCID: PMC3131487.
  14. Chesney J, Lewis KD, Kluger H, Hamid O, Whitman E, Thomas S, Wermke M, Cusnir M, Domingo-Musibay E, Phan GQ, Kirkwood JM, Hassel JC, Orloff M, Larkin J, Weber J, Furness AJS, Khushalani NI, Medina T, Egger ME, Graf Finckenstein F, Jagasia M, Hari P, Sulur G, Shi W, Wu X, Sarnaik A. 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. 2022 Dec;10(12):e005755. doi: 10.1136/jitc-2022-005755. PMID: 36600653; PMCID: PMC9748991.
  15. Mao X, Xu J, Wang W, Liang C, Hua J, Liu J, Zhang B, Meng Q, Yu X, Shi S. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: new findings and future perspectives. Mol Cancer. 2021 Oct 11;20(1):131. doi: 10.1038/s41577-018-0044-0. PMID: 30057419.
  16. Khalaf K, Hana D, Chou JT, Singh C, Mackiewicz A, Kaczmarek M. Aspects of the Tumor Microenvironment Involved in Immune Resistance and Drug Resistance. Front Immunol. 2021 May 27;12:656364. doi: 10.3389/fimmu.2021.656364. PMID: 34122412; PMCID: PMC8190405.
  17. Paijens ST, Vledder A, de Bruyn M, Nijman HW. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell Mol Immunol. 2021 Apr;18(4):842-859. doi: 10.1038/s41423-020-00565-9. Epub 2020 Nov 2. PMID: 33139907; PMCID: PMC8115290.
  18. Borst J, Ahrends T, B?ba?a N, Melief CJM, Kastenmüller W. CD4+ T cell help in cancer immunology and immunotherapy. Nat Rev Immunol. 2018 Oct;18(10):635-647. doi: 10.1038/s41577-018-0044-0. PMID: 30057419.
  19. Zhao Y, Shao Q, Peng G. Exhaustion and senescence: two crucial dysfunctional states of T cells in the tumor microenvironment. Cell Mol Immunol. 2020 Jan;17(1):27-35. doi: 10.1038/s41423-019-0344-8. Epub 2019 Dec 18. PMID: 31853000; PMCID: PMC6952436.
  20. Lu C, Rong D, Zhang B, Zheng W, Wang X, Chen Z, Tang W. Current perspectives on the immunosuppressive tumor microenvironment in hepatocellular carcinoma: challenges and opportunities. Mol Cancer. 2019 Aug 29;18(1):130. doi: 10.1186/s12943-019-1047-6. PMID: 31464625; PMCID: PMC6714090.
  21. Kumar A, Watkins R, Vilgelm AE. Cell Therapy With TILs: Training and Taming T Cells to Fight Cancer. Front Immunol. 2021 Jun 1;12:690499. doi: 10.3389/fimmu.2021.690499. PMID: 34140957; PMCID: PMC8204054.
  22. Wickström S, Lövgren T. Expansion of Tumor-Infiltrating Lymphocytes from Melanoma Tumors. Methods Mol Biol. 2019;1913:105-118. doi: 10.1007/978-1-4939-8979-9_7. PMID: 30666601.
  23. Zhao Y, Deng J, Rao S, Guo S, Shen J, Du F, Wu X, Chen Y, Li M, Chen M, Li X, Li W, Gu L, Sun Y, Zhang Z, Wen Q, Xiao Z, Li J. Tumor Infiltrating Lymphocyte (TIL) Therapy for Solid Tumor Treatment: Progressions and Challenges. Cancers (Basel). 2022 Aug 27;14(17):4160. doi: 10.3390/cancers14174160. PMID: 36077696; PMCID: PMC9455018.
  24. Grimm EA, Mazumder A, Zhang HZ, Rosenberg SA. Lymphokine-activated killer cell phenomenon. Lysis of natural killer-resistant fresh solid tumor cells by interleukin 2-activated autologous human peripheral blood lymphocytes. J Exp Med. 1982 Jun 1;155(6):1823-41. doi: 10.1084/jem.155.6.1823. PMID: 6176669; PMCID: PMC2186695.
  25. Van den Berg JH, Heemskerk B, van Rooij N, Gomez-Eerland R, Michels S, van Zon M, de Boer R, Bakker NAM, Jorritsma-Smit A, van Buuren MM, Kvistborg P, Spits H, Schotte R, Mallo H, Karger M, van der Hage JA, Wouters MWJM, Pronk LM, Geukes Foppen MH, Blank CU, Beijnen JH, Nuijen B, Schumacher TN, Haanen JBAG. Tumor infiltrating lymphocytes (TIL) therapy in metastatic melanoma: boosting of neoantigen-specific T cell reactivity and long-term follow-up. J Immunother Cancer. 2020 Aug;8(2):e000848. doi: 10.1136/jitc-2020-000848. PMID: 32753545; PMCID: PMC7406109.
  26. Hirai I, Funakoshi T, Kamijuku H, Fukuda K, Mori M, Sakurai M, Koda Y, Kato J, Mori T, Watanabe N, Noji S, Yaguchi T, Iwata T, Ohta S, Fujita T, Tanosaki R, Handa M, Okamoto S, Amagai M, Kawakami Y. Adoptive cell therapy using tumor-infiltrating lymphocytes for melanoma refractory to immune-checkpoint inhibitors. Cancer Sci. 2021 Aug;112(8):3163-3172. doi: 10.1111/cas.15009. Epub 2021 Jun 30. PMID: 34101300; PMCID: PMC8353905.
  27. Kazemi MH, Sadri M, Najafi A, Rahimi A, Baghernejadan Z, Khorramdelazad H, Falak R. Tumor-infiltrating lymphocytes for treatment of solid tumors: It takes two to tango? Front Immunol. 2022 Oct 28;13:1018962. doi: 10.3389/fimmu.2022.1018962. PMID: 36389779; PMCID: PMC9651159.
  28. Clinical studies around the world. Viewed on 04th july 2024. https://clinicaltrials.gov/
  29. Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, Jemal A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024 May-Jun;74(3):229-263. doi: 10.3322/caac.21834. Epub 2024 Apr 4. PMID: 38572751.
  30. Wilson MA, Schuchter LM. Chemotherapy for Melanoma. Cancer Treat Res. 2016;167:209-29. doi: 10.1007/978-3-319-22539-5_8. PMID: 26601864.
  31. Ralli M, Botticelli A, Visconti IC, Angeletti D, Fiore M, Marchetti P, Lambiase A, de Vincentiis M, Greco A. Immunotherapy in the Treatment of Metastatic Melanoma: Current Knowledge and Future Directions. J Immunol Res. 2020 Jun 28;2020:9235638. doi: 10.1155/2020/9235638. PMID: 32671117; PMCID: PMC7338969.
  32. Lugowska I, Teterycz P, Rutkowski P. Immunotherapy of melanoma. Contemp Oncol (Pozn). 2018 Mar;22(1A):61-67. doi: 10.5114/wo.2018.73889. Epub 2018 Mar 5. PMID: 29628796; PMCID: PMC5885078.
  33. Lipson EJ, Drake CG. Ipilimumab: an anti-CTLA-4 antibody for metastatic melanoma. Clin Cancer Res. 2011 Nov 15;17(22):6958-62. doi: 10.1158/1078-0432.CCR-11-1595. Epub 2011 Sep 7. PMID: 21900389; PMCID: PMC3575079.
  34. Babacan NA, Eroglu Z. Treatment Options for Advanced Melanoma After Anti-PD-1 Therapy. Curr Oncol Rep. 2020 Mar 20;22(4):38. doi: 10.1007/s11912-020-0894-z. PMID: 32198651.
  35. Inozume T. Adoptive cell transfer therapy for melanoma. Exp Dermatol. 2023 Mar;32(3):250-255. doi: 10.1111/exd.14707. Epub 2022 Nov 23. PMID: 36382355.
  36. Morand S, Devanaboyina M, Staats H, Stanbery L, Nemunaitis J. Ovarian Cancer Immunotherapy and Personalized Medicine. Int J Mol Sci. 2021 Jun 18;22(12):6532. doi: 10.3390/ijms22126532. PMID: 34207103; PMCID: PMC8234871.
  37. Foley OW, Rauh-Hain JA, del Carmen MG. Recurrent epithelial ovarian cancer: an update on treatment. Oncology (Williston Park). 2013 Apr;27(4):288-94, 298. PMID: 23781692.
  38. Cortez AJ, Tudrej P, Kujawa KA, Lisowska KM. Advances in ovarian cancer therapy. Cancer Chemother Pharmacol. 2018 Jan;81(1):17-38. doi: 10.1007/s00280-017-3501-8. Epub 2017 Dec 16. PMID: 29249039; PMCID: PMC5754410.
  39. Sarivalasis A, Morotti M, Mulvey A, Imbimbo M, Coukos G. Cell therapies in ovarian cancer. Ther Adv Med Oncol. 2021 Apr 22;13:17588359211008399. doi: 10.1177/17588359211008399. Erratum in: Ther Adv Med Oncol. 2021 Jun 17;13:17588359211028457. doi: 10.1177/17588359211028457. PMID: 33995591; PMCID: PMC8072818.
  40. Borella F, Ghisoni E, Giannone G, Cosma S, Benedetto C, Valabrega G, Katsaros D. Immune Checkpoint Inhibitors in Epithelial Ovarian Cancer: An Overview on Efficacy and Future Perspectives. Diagnostics (Basel). 2020 Mar 7;10(3):146. doi: 10.3390/diagnostics10030146. PMID: 32156035; PMCID: PMC7151145.
  41. Freedman RS, Edwards CL, Kavanagh JJ, Kudelka AP, Katz RL, Carrasco CH, Atkinson EN, Scott W, Tomasovic B, Templin S, et al. Intraperitoneal adoptive immunotherapy of ovarian carcinoma with tumor-infiltrating lymphocytes and low-dose recombinant interleukin-2: a pilot trial. J Immunother Emphasis Tumor Immunol. 1994 Oct;16(3):198-210. doi: 10.1097/00002371-199410000-00004. PMID: 7834119.
  42. Pedersen M, Westergaard MCW, Milne K, Nielsen M, Borch TH, Poulsen LG, Hendel HW, Kennedy M, Briggs G, Ledoux S, Nøttrup TJ, Andersen P, Hasselager T, Met Ö, Nelson BH, Donia M, Svane IM. Adoptive cell therapy with tumor-infiltrating lymphocytes in patients with metastatic ovarian cancer: a pilot study. Oncoimmunology. 2018 Sep 26;7(12):e1502905. doi: 10.1080/2162402X.2018.1502905. PMID: 30524900; PMCID: PMC6279323.
  43. Amaria R, Knisely A, Vining D, et al Efficacy and safety of autologous tumor-infiltrating lymphocytes in recurrent or refractory ovarian cancer, colorectal cancer, and pancreatic ductal adenocarcinoma,Journal for ImmunoTherapy of Cancer 2024;12:e006822. doi: 10.1136/jitc-2023-006822
  44. Kroep, J.R. et al. 729P Adoptive T-cell therapy during chemotherapy with or without peginterferon-? (IFN?) in patients with platinum sensitive recurrent epithelial ovarian cancer (EOC), Annals of Oncology, Volume 32, S732 - S733
  45. Robertson J, Salm M, Dangl M. Adoptive cell therapy with tumour-infiltrating lymphocytes: the emerging importance of clonal neoantigen targets for next-generation products in non-small cell lung cancer. Immunooncol Technol. 2019 Oct 9;3:1-7. doi: 10.1016/j.iotech.2019.09.003. PMID: 35757302; PMCID: PMC9216247.
  46. Massarelli. E. et al, P14.04 A Phase 2 Multicenter Study of Iovance Autologous Tumor Infiltrating Lymphocytes (TIL, LN-145) Cell Therapy in Patients With Metastatic NSCLC, Journal of Thoracic Oncology, Volume 16, Issue 10, S1012.
  47. Creelan BC, Wang C, Teer JK, Toloza EM, Yao J, Kim S, Landin AM, Mullinax JE, Saller JJ, Saltos AN, Noyes DR, Montoya LB, Curry W, Pilon-Thomas SA, Chiappori AA, Tanvetyanon T, Kaye FJ, Thompson ZJ, Yoder SJ, Fang B, Koomen JM, Sarnaik AA, Chen DT, Conejo-Garcia JR, Haura EB, Antonia SJ. Tumor-infiltrating lymphocyte treatment for anti-PD-1-resistant metastatic lung cancer: a phase 1 trial. Nat Med. 2021 Aug;27(8):1410-1418. doi: 10.1038/s41591-021-01462-y. Epub 2021 Aug 12. PMID: 34385708; PMCID: PMC8509078.
  48. Schoenfeld A, Lee S, Paz-Ares L, et al 458 First phase 2 results of autologous tumor-infiltrating lymphocyte (TIL; LN-145) monotherapy in patients with advanced, immune checkpoint inhibitor-treated, non-small cell lung cancer (NSCLC),Journal for ImmunoTherapy of Cancer 2021;9:doi: 10.1136/jitc-2021-SITC2021.458
  49. Loi, S. & Michiels, Stefan & Adams, S. & Loibl, S. & Budczies, Jan & Denkert, Carsten & Salgado, Renato. (2021). The journey of tumor infiltrating lymphocytes (TIL) as a biomarker in breast cancer: clinical utility in an era of checkpoint inhibition. Annals of Oncology. 32. 10.1016/j.annonc.2021.07.007.
  50. Almangush A, De Keukeleire S, Rottey S, Ferdinande L, Vermassen T, Leivo I, Mäkitie AA. Tumor-Infiltrating Lymphocytes in Head and Neck Cancer: Ready for Prime Time? Cancers (Basel). 2022 Mar 18;14(6):1558. doi: 10.3390/cancers14061558. PMID: 35326709; PMCID: PMC8946626.
  51. Zemanek T, Nova Z, Nicodemou A. Tumor-Infiltrating Lymphocytes and Adoptive Cell Therapy: State of the Art in Colorectal, Breast and Lung Cancer. Physiol Res. 2023 Oct 27;72(S3):S209-S224. doi: 10.33549/physiolres.935155. PMID: 37888965; PMCID: PMC10669950.
  52. Kazemi MH, Sadri M, Najafi A, Rahimi A, Baghernejadan Z, Khorramdelazad H, Falak R. Tumor-infiltrating lymphocytes for treatment of solid tumors: It takes two to tango? Front Immunol. 2022 Oct 28;13:1018962. doi: 10.3389/fimmu.2022.1018962. PMID: 36389779; PMCID: PMC9651159.
  53. Li R, Cao L. The role of tumor-infiltrating lymphocytes in triple-negative breast cancer and the research progress of adoptive cell therapy. Front Immunol. 2023 May 18;14:1194020. doi: 10.3389/fimmu.2023.1194020. PMID: 37275874; PMCID: PMC10233026.
  54. Zacharakis N, Huq LM, Seitter SJ, Kim SP, Gartner JJ, Sindiri S, et al. Breast cancers are immunogenic: immunologic analyses and a phase II pilot clinical trial using mutation-reactive autologous lymphocytes. J Clin Oncol (2022) 40(16):1741–54. doi: 10.1200/jco.21.02170
  55. O’Malley D, Lee S, Psyrri A, Sukari A, Thomas S, Wenham R, et al. 492 phase 2 efficacy and safety of autologous tumor-infiltrating lymphocyte (Til) cell therapy in combination with pembrolizumab in immune checkpoint inhibitor-naïve patients with advanced cancers. BMJ Specialist J (2021) 9:523–24.
  56. Tran E, Robbins PF, Lu YC, Prickett TD, Gartner JJ, Jia L, Pasetto A, Zheng Z, Ray S, Groh EM, Kriley IR, Rosenberg SA. T-Cell Transfer Therapy Targeting Mutant KRAS in Cancer. N Engl J Med. 2016 Dec 8;375(23):2255-2262. doi: 10.1056/NEJMoa1609279. PMID: 27959684; PMCID: PMC5178827.
  57. Plesca I, Tunger A, Müller L, Wehner R, Lai X, Grimm MO, Rutella S, Bachmann M, Schmitz M. Characteristics of Tumor-Infiltrating Lymphocytes Prior to and During Immune Checkpoint Inhibitor Therapy. Front Immunol. 2020 Mar 4;11:364. doi: 10.3389/fimmu.2020.00364. PMID: 32194568; PMCID: PMC7064638.
  58. Eberlein TJ, Rosenstein M, Rosenberg SA. Regression of a disseminated syngeneic solid tumor by systemic transfer of lymphoid cells expanded in interleukin 2. J Exp Med. 1982 Aug 1;156(2):385-97. doi: 10.1084/jem.156.2.385. PMID: 6980254; PMCID: PMC2186754.
  59. Brambilla E, Le Teuff G, Marguet S, Lantuejoul S, Dunant A, Graziano S, Pirker R, Douillard JY, Le Chevalier T, Filipits M, Rosell R, Kratzke R, Popper H, Soria JC, Shepherd FA, Seymour L, Tsao MS. Prognostic Effect of Tumor Lymphocytic Infiltration in Resectable Non-Small-Cell Lung Cancer. J Clin Oncol. 2016 Apr 10;34(11):1223-30. doi: 10.1200/JCO.2015.63.0970. Epub 2016 Feb 1. PMID: 26834066; PMCID: PMC4872323.
  60. Whiteside TL. The tumor microenvironment and its role in promoting tumor growth. Oncogene. 2008 Oct 6;27(45):5904-12. doi: 10.1038/onc.2008.271. PMID: 18836471; PMCID: PMC3689267.
  61. Chen N, Li X, Chintala NK, Tano ZE, Adusumilli PS. Driving CARs on the uneven road of antigen heterogeneity in solid tumors. Curr Opin Immunol. 2018 Apr;51:103-110. doi: 10.1016/j.coi.2018.03.002. Epub 2018 Mar 16. PMID: 29554494; PMCID: PMC5943172.
  62. Zhang J, Fujimoto J, Zhang J, Wedge DC, Song X, Zhang J, Seth S, Chow CW, Cao Y, Gumbs C, Gold KA, Kalhor N, Little L, Mahadeshwar H, Moran C, Protopopov A, Sun H, Tang J, Wu X, Ye Y, William WN, Lee JJ, Heymach JV, Hong WK, Swisher S, Wistuba II, Futreal PA. Intratumor heterogeneity in localized lung adenocarcinomas delineated by multiregion sequencing. Science. 2014 Oct 10;346(6206):256-9. doi: 10.1126/science.1256930. PMID: 25301631; PMCID: PMC4354858.
  63. Egelston CA, Avalos C, Tu TY, Simons DL, Jimenez G, Jung JY, Melstrom L, Margolin K, Yim JH, Kruper L, Mortimer J, Lee PP. Human breast tumor-infiltrating CD8+ T cells retain polyfunctionality despite PD-1 expression. Nat Commun. 2018 Oct 16;9(1):4297. doi: 10.1038/s41467-018-06653-9. PMID: 30327458; PMCID: PMC6191461.
  64. Retèl VP, Steuten LMG, Geukes Foppen MH, Mewes JC, Lindenberg MA, Haanen JBAG, van Harten WH. Early cost-effectiveness of tumor infiltrating lymphocytes (TIL) for second line treatment in advanced melanoma: a model-based economic evaluation. BMC Cancer. 2018 Sep 15;18(1):895. doi: 10.1186/s12885-018-4788-5. PMID: 30219040; PMCID: PMC6139174.
  65. Ding W, Xu X, Qian Y, Xue W, Wang Y, Du J, Jin L, Tan Y. Prognostic value of tumor-infiltrating lymphocytes in hepatocellular carcinoma: A meta-analysis. Medicine (Baltimore). 2018 Dec;97(50):e13301. doi: 10.1097/MD.0000000000013301. PMID: 30557978; PMCID: PMC6320107.
  66. Peggs KS, Quezada SA, Chambers CA, Korman AJ, Allison JP. Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. J Exp Med. 2009 Aug 3;206(8):1717-25. doi: 10.1097/MD.0000000000013301. PMID: 30557978; PMCID: PMC6320107.
  67. Mullinax JE, Hall M, Prabhakaran S, Weber J, Khushalani N, Eroglu Z, Brohl AS, Markowitz J, Royster E, Richards A, Stark V, Zager JS, Kelley L, Cox C, Sondak VK, Mulé JJ, Pilon-Thomas S, Sarnaik AA. Combination of Ipilimumab and Adoptive Cell Therapy with Tumor-Infiltrating Lymphocytes for Patients with Metastatic Melanoma. Front Oncol. 2018 Mar 2;8:44. doi: 10.3389/fonc.2018.00044. PMID: 29552542; PMCID: PMC5840208.
  68. Cafri G, Gartner JJ, Zaks T, Hopson K, Levin N, Paria BC, Parkhurst MR, Yossef R, Lowery FJ, Jafferji MS, Prickett TD, Goff SL, McGowan CT, Seitter S, Shindorf ML, Parikh A, Chatani PD, Robbins PF, Rosenberg SA. mRNA vaccine-induced neoantigen-specific T cell immunity in patients with gastrointestinal cancer. J Clin Invest. 2020 Nov 2;130(11):5976-5988. doi: 10.1172/JCI134915. PMID: 33016924; PMCID: PMC7598064.
  69. Klebanoff CA, Gattinoni L, Restifo NP. CD8+ T-cell memory in tumor immunology and immunotherapy. Immunol Rev. 2006 Jun;211:214-24. doi: 10.1111/j.0105-2896.2006.00391.x. PMID: 16824130; PMCID: PMC1501075.
  70. Subbiah V, Baik C, Kirkwood JM. Clinical Development of BRAF plus MEK Inhibitor Combinations. Trends Cancer. 2020 Sep;6(9):797-810. doi: 10.1111/j.0105-2896.2006.00391.x. PMID: 16824130; PMCID: PMC1501075.
  71. Sloot S, Zager JS, Kudchadkar RR, Messina JL, Benedict JJ, Gonzalez RJ, DeConti R, Turner LM, McCardle T, Smalley KS, Weber JS, Sondak VK, Gibney GT. BRAF inhibition for advanced locoregional BRAF V600E mutant melanoma: a potential neoadjuvant strategy. Melanoma Res. 2016 Feb;26(1):83-7. doi: 10.1097/CMR.0000000000000214. PMID: 26731560.
  72. Cooper ZA, Frederick DT, Juneja VR, Sullivan RJ, Lawrence DP, Piris A, Sharpe AH, Fisher DE, Flaherty KT, Wargo JA. BRAF inhibition is associated with increased clonality in tumor-infiltrating lymphocytes. Oncoimmunology. 2013 Oct 1;2(10):e26615. doi: 10.4161/onci.26615. Epub 2013 Oct 15. PMID: 24251082; PMCID: PMC3827093
  73. Sosman JA, Kim KB, Schuchter L, Gonzalez R, Pavlick AC, Weber JS, McArthur GA, Hutson TE, Moschos SJ, Flaherty KT, Hersey P, Kefford R, Lawrence D, Puzanov I, Lewis KD, Amaravadi RK, Chmielowski B, Lawrence HJ, Shyr Y, Ye F, Li J, Nolop KB, Lee RJ, Joe AK, Ribas A. Survival in BRAF V600-mutant advanced melanoma treated with vemurafenib. N Engl J Med. 2012 Feb 23;366(8):707-14. doi: 10.1056/NEJMoa1112302. PMID: 22356324; PMCID: PMC3724515
  74. Khammari A, Nguyen JM, Saint-Jean M, Knol AC, Pandolfino MC, Quereux G, Brocard A, Peuvrel L, Saiagh S, Bataille V, Limacher JM, Dreno B. Adoptive T cell therapy combined with intralesional administrations of TG1042 (adenovirus expressing interferon-?) in metastatic melanoma patients. Cancer Immunol Immunother. 2015 Jul;64(7):805-15. doi: 10.1007/s00262-015-1691-7. Epub 2015 Apr 7. PMID: 25846669; PMCID: PMC11029588.
  75. Lou Y, Wang G, Lizée G, Kim GJ, Finkelstein SE, Feng C, Restifo NP, Hwu P. Dendritic cells strongly boost the antitumor activity of adoptively transferred T cells in vivo. Cancer Res. 2004 Sep 15;64(18):6783-90. doi: 10.1158/0008-5472.CAN-04-1621. PMID: 15374997; PMCID: PMC2241750.
  76. Donia M, Junker N, Ellebaek E, Andersen MH, Straten PT, Svane IM. Characterization and comparison of 'standard' and 'young' tumour-infiltrating lymphocytes for adoptive cell therapy at a Danish translational research institution. Scand J Immunol. 2012 Feb;75(2):157-67. doi: 10.1111/j.1365-3083.2011.02640.x. PMID: 21955245

Photo
Swathi Madhavan
Corresponding author

Department of pharmacy, Faculty of Engineering and technology, Annamalai university, Chidambaram, 608002.

Photo
T. Keerthana
Co-author

Department of pharmacy, Faculty of Engineering and Technology, Annamalai university, Chidambaram, 608002.

Photo
A. Sowmiya
Co-author

Department of pharmacy, Faculty of Engineering and Technology, Annamalai university, Chidambaram, 608002.

Photo
D. Kilimozhi
Co-author

Department of pharmacy, Faculty of Engineering and Technology, Annamalai university, Chidambaram, 608002

Swathi Madhavan*, T. Keerthana, A. Sowmiya, D. Kilimozhi, T Cell Line Treatment for Cancer Using Tumor-Infiltrating Lymphocytes, Int. J. of Pharm. Sci., 2024, Vol 2, Issue 11, 1653-1668. https://doi.org/10.5281/zenodo.14245431

More related articles
Biological Activities Of Ginseng And Its Applicati...
Tushar Dewangan , Suraj Sahu, Suraj Patel, Miss Divyani Soni, Dr....
An Overview of Thyroxine Drug and Its Application...
vikas sonur, Ayan Sayyad, Digvijay Dalvi, ...
Niosomes: A Review Of Drug Delivery System...
Pradnya H. Kapse, Vrushali S. Gangurde, Khanderao Jadhav, Rishik...
Leveraging Artificial Intelligence for Enhanced Pharmacy Practice: A Comprehensi...
Abhijit S. Dongardive, Salman A. Khan, Sagar Sirsat, Trupti Kokane, Akshay Jagtap, ...
Review on Achyranthes aspera...
Ramakrishna S., Ruqiya bi, ...
Ancient methods and Novel methods of Insulin delivery...
Pallavi Saxena, Mohd Shuaib, Abhinay Gupta, ...
Related Articles
Extraction, Standardization And Evaluation Of Gallic Acid Obtained From Peels An...
Pratham S. Shinde, Jeeshan J. Shaikh, Yash N. Sharma, Poonam R. Shewale, Chaitanya S. Shisode, ...
Review on Cucumis Sativus ...
Pranav Sanap, Meghana Rayjade, Saurabh Khairnar , Dhiraj Maind, Omkar Phopse, ...
Caesalpinia Bonducella: A Pharmacognostic And Pharmacological Overview ...
Lilachand Bhaidas Patil, Khushal Kalyan Chaudhari, Mayur Sharad Patel, Nandini Arun Marathe, ...
DESIGN AND DEVELOPMENT OF BATH BOMB...
Manisha. U. Mishra, Vijaya. K. Parihar, Vedika. A. Kumarwar, Urwashi. C. Baghele, Vaishnavi. D. Th...
Biological Activities Of Ginseng And Its Application To Human Health...
Tushar Dewangan , Suraj Sahu, Suraj Patel, Miss Divyani Soni, Dr. Shruti Rathore, ...
More related articles
Biological Activities Of Ginseng And Its Application To Human Health...
Tushar Dewangan , Suraj Sahu, Suraj Patel, Miss Divyani Soni, Dr. Shruti Rathore, ...
An Overview of Thyroxine Drug and Its Application...
vikas sonur, Ayan Sayyad, Digvijay Dalvi, ...
Niosomes: A Review Of Drug Delivery System...
Pradnya H. Kapse, Vrushali S. Gangurde, Khanderao Jadhav, Rishikesh Bachhav, ...
Biological Activities Of Ginseng And Its Application To Human Health...
Tushar Dewangan , Suraj Sahu, Suraj Patel, Miss Divyani Soni, Dr. Shruti Rathore, ...
An Overview of Thyroxine Drug and Its Application...
vikas sonur, Ayan Sayyad, Digvijay Dalvi, ...
Niosomes: A Review Of Drug Delivery System...
Pradnya H. Kapse, Vrushali S. Gangurde, Khanderao Jadhav, Rishikesh Bachhav, ...