Genetically engineered T cell and tumour-infiltrating lymphocyte therapies

DOI: https://doi.org/https://doi.org/10.57187/s.4279

Andreas Holbroabc, Heinz Läublibde

Division of Haematology, University Hospital Basel, Basel, Switzerland

Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland

Regional Blood Transfusion Service, Swiss Red Cross, Basel, Switzerland

Department of Biomedicine, University of Basel, Basel, Switzerland

Division of Oncology, University Hospital Basel, Basel, Switzerland

Summary

Haemato-oncology has made significant progress in recent years, particularly through the development of innovative immunotherapeutic approaches such as CAR T cell (chimeric antigen receptor T cell) and tumour-infiltrating lymphocyte therapies. Both methods use the patient’s own immune system to treat cancer, but in different ways. CAR T cell therapy is a form of immunotherapy in which the patient’s own T cells are genetically modified. CAR T cell therapies have proven to be particularly effective in haematological B-cell neoplasms, such as B-cell acute lymphoblastic leukaemia (B-ALL) and B-cell lymphomas, as well as in multiple myeloma. Tumour-infiltrating lymphocyte therapy, on the other hand, exploits the natural ability of T cells to recognise tumour-associated antigens of tumour cells with the T cell receptor. Tumour tissue is taken from the patient then tumour-infiltrating lymphocytes are isolated from it. These tumour-infiltrating lymphocytes are expanded ex vivo to increase their number and activity. This review discusses the principles of these innovative therapies. Both therapies represent significant advances in personalised cancer treatment and offer new hope for our cancer patients.

Abbreviations

B-ALL:

acute B-cell lymphoblastic leukaemia

CAR:

chimeric antigen receptor

DLBCL:

diffuse large B-cell lymphoma

scFv:

single-chain variable fragment

TIL:

tumour-infiltrating lymphocyte

Introduction

Cancer immunotherapy has significantly improved the outcome of patients. In particular, the introduction of immune checkpoint inhibitors has transformed the treatment of patients with solid cancers [1, 2]. However, only a limited number of patients benefit from immune checkpoint inhibition [3]. Cell therapies including genetically engineered T cells expressing a chimeric antigen receptor (CAR) have been used to treat cancer patients [4]. While such CAR T cell therapies have been successfully used mainly to treat patients with haematological B cell neoplasia, naturally occurring tumour-infiltrating lymphocytes (TILs) have been used for the treatment of solid cancers resistant to immune checkpoint inhibitors [5]. Here, we give an overview of these two emerging cell therapy approaches (figure 1) and an outlook on current developments.

Figure 1Illustration of tumour-infiltrating lymphocyte (TIL) therapy (left) and CAR T cell therapy. In tumour-infiltrating lymphocyte therapy, T cells are isolated from the tumour and expanded to >50 × 109 cells with IL-2, CD3 stimulation and allogeneic feeders. After lymphodepletion with cyclophosphamide and fludarabine, the CD3-positive T cells are administered to the patient together with IL-2. CAR T cell therapy uses T cells obtained by apheresis. The T cells are then genetically manipulated (usually with a lenti- or retroviral vector) so that they stably express a CAR. They are then returned to the patient who has undergone lymphodepletion.

CAR T cell therapy

Cellular cancer therapies have been used to treat cancer since the introduction of allogeneic stem cell transplantation by Don Thomas in the 1970s [6]. In the late 1980s, T cells were genetically modified for the first time, equipping them with a synthetic T cell receptor containing an intracellular activation domain, the CD3ζ of the T cell receptor, and an extracellular binding domain, usually a single-chain variable fragment (scFv) of an antibody directed against a surface molecule on the cancer cell [7] (figure 2). These so-called chimeric antigen receptors (CARs) were further developed, showing a good effect in mouse models but also in patients with B-cell neoplasia [4]. Further development led to the so-called second-generation CARs, which also contained a co-stimulatory component in the intracellular domain [4]. The intracellular domain of CD28 or 4-1BB (CD137) was primarily used for this purpose (figure 2). This enabled not only recognition of the target antigen but also proliferation and persistence of these cells in vivo. More recent developments include other co-stimulatory domains or synthetic proteins that lead to an increase in specificity or efficiency as well as the possibility of better control of their proliferation. In contrast to antibodies targeting a tumour surface antigen such as the anti-CD20 antibody rituximab, CAR T cells can persist and lead to long-term control by the immune system.

Figure 2Demonstration of the genetic modification of T cells during CAR T cell production. Chimeric antigen receptors (CARs) are synthetic proteins consisting of an intracellular part with different signalling domains and an antigen-binding domain that is commonly derived from an antibody and expressed as a short-chain variable fragment (ScFv). CAR constructs are delivered to T cells after a leukapheresis and retransfused in the lymphodepleted patient. TCR: T cell receptor. Created with BioRender.com.

Haematological diseases

The treatment of B cell malignancies with cellular immunotherapies has become the standard of care for several indications [4, 8]. Treatment is currently approved when initial treatment has failed or in refractory cases (table 1). In particular, CD19-positive B cell malignancies have been successfully treated with CD19-specific CAR T cells [4, 8]. CAR T cell therapy was able to achieve durable remissions in patients after multiple prior lines of therapy and in some cases highly chemotherapy-refractory disease. Recently, several studies have also shown an advantage of CAR T cell therapy in patients with relapsed/refractory diffuse large B-cell lymphoma compared to the standard of care with chemotherapy followed by autologous stem cell transplantation (diffuse large B-cell lymphoma) [4, 8]. For example, in the ZUMA-7 study, axicabtagene-ciloleucel (Axi-cel) was tested in 359 patients compared to standard treatment [9]. In addition to the CD19-targeted CAR T cell therapies, CAR T cells against the BCMA antigen have also been established for the treatment of multiple myeloma [10, 11]. Other targets on myeloma cells such as SLAMF-7 or G protein-coupled receptor, class C, group 5, member D (GPRC5D) are currently being investigated [12].

Table 1EMA-approved CAR T therapies for B cell malignancies and multiple myeloma (as of September 2024).

Product Kymriah Yescarta Tecartus Breyanzi Abecma Carvykti
Active substance Tisagenlecleucel Axicabtagene-ciloleucel Brexucabtagene autoleucel Lisocabtagene maraleucel Idecabtagene-vicleucel Ciltacabtagenum autoleucelum
Manufacturer Novartis Kyte/Gilead Kyte/Gilead BMS/Celgene BMS/Celgene Janssen
Approval (EMA) 2018 2018 2020 2022 2021 2022
Target CD19 CD19 CD19 CD19 BCMA BCMA
Costimulatory signal 4-1BB CD28 CD28 4-1BB 4-1BB 4-1BB
Indication(s) r/r B-ALL (age ≤25, 3rd line); r/r DLBC (3rd line); r/r FL (3rd line) r/r DLBCL, HGBCL (2nd line*); PMBCL (3rd line); r/r FL (4th line) r/r MCL (3rd line; previous lines included BTK inhibitor); r/r B-ALL (age ≥26, 3rd line) r/r DLBCL, PMBCL, HGBCL (2nd line*); FL grade 3B (2nd line*) r/r MM (3rd line**) r/r MM (2nd line***)

B-ALL: acute B-cell lymphoblastic leukaemia; DLBCL: diffuse large B-cell lymphoma; FL: follicular lymphoma; HGBCL: high-grade B-cell lymphoma; MCL: mantle cell lymphoma; MM: multiple myeloma; PMBCL: primary mediastinal B-cell lymphoma; r/r: relapsed/refractory.

* 2nd line if refractory or early relapse (within 12 months after first-line chemoimmunotherapy); 3rd line if later.

**After at least 2 lines including an immunomodulator, proteasome inhibitor and anti-CD38 antibody.

*** After 1 line including an immunomodulator, proteasome inhibitor and refractory to lenalidomide.

Solid cancers

Several targets for solid tumours are currently under investigation. Early trials for HER2-positive cancers were terminated due to severe toxicity on target tissues outside the tumour (so-called on-target/off-tumour effects) [4, 8]. Major hurdles for CAR T cell therapy in solid malignancies include defining the correct tumour antigen with specific, high-level expression in the tumour, difficulties in trafficking T cells into the tumour microenvironment and depletion of CAR T cells due to an immunosuppressive tumour microenvironment [4, 8]. Nevertheless, some successful studies have already been conducted. For example, a promising response to claudin 18.2-targeted CAR T cells has been shown in patients with advanced gastric or pancreatic cancer [13]. An interesting approach was pursued by Mackensen and colleagues. In addition to the use of claudin 6-directed CAR T cells, a combination with an RNA vaccine, which induces additional activation of the CAR T cells with enhancement of a memory function, was pursued. The study showed promising results in gastrointestinal and gynaecological malignancies [14]. In recent months, several studies on the treatment of glioblastoma with CAR T cells have also been published [15, 16]. One by Bagley et al. used intrathecally administered CAR T cells able to recognise two antigens on glioblastoma cells, namely EGFR and IL13Ralpha2 [16]. The other by Marcela Maus’ group used locally administered  CAR T cells that recognise both the tumour-specific variant of EGFRvIII and the wild-type variant of EGFR. Promising results were also achieved with anti-GD2 CAR T cells in paediatric gliomas, with complete remissions being induced [17]. So far, however, the long-term results are very sobering; they show that the principle also works in solid tumours but that further work is needed to achieve longer-term tumour control or eradication. Recently, the first genetically engineered cell product for solid tumours has been approved by the FDA; however it is not a CAR T cell therapy but a T cell receptor T cell therapy targeting the tumour antigen MAGE-A4. Afamitresgene autoleucel was successfully tested in HLA-A*02:01-, 02-, 03- and 06-positive patients with metastatic synovial sarcoma [18].

Various additional measures can potentially increase the efficiency of CAR T cells against solid tumours. For example, to increase the specificity of the CAR construct, genetic systems have been developed that can integrate two or more tumour-transmitted signals [4, 8]. The synNOTCH system, for example, uses the Notch signalling system to mediate a two-step activation of CAR T cells [19]. The binding of a first antigen induces the expression of the activating CAR, which leads to the activation of the immune cell. This two-step CAR T cell activation can significantly increase the specificity of such a CAR T cell product. The local release of cytokines that promote anti-tumour immunity could improve the activation and efficacy of CAR T cells [8]. For example, secretion of IL-12 could enhance CD19-targeted CAR T cell therapy [20].

CAR T cell therapy for patients with autoimmune disease

Recently, several reports have been published describing a role for CAR T cell therapy in patients with autoimmune disease [21–27]. For example, patients with treatment-refractory systemic lupus erythematosus (SLE) treated with CD19-directed CAR T cells experienced significant improvement [21–24]. Also, CD19-targeted CAR T cell therapies were used to treat patients with treatment-refractory anti-synthetase syndrome [25, 26, 28] or patients with multiple sclerosis and myasthenia gravis [27, 29].

Side effects of CAR T cell therapy

CAR T cell therapy is considered a promising cancer treatment but is associated with various side effects, some of which can be life-threatening [30]. One of the most common side effects is cytokine release syndrome (CRS), which can cause fever, low blood pressure and organ dysfunction. In addition, immune effector cell-associated neurotoxicity syndrome (ICANS) can occur, a reversible but potentially life-threatening complication that can manifest with confusion, seizures and other neurological symptoms [31]. In phase III trials of second-line therapy for patients with diffuse large B-cell lymphoma, second-generation CAR T cells showed that up to 90% of patients experienced cytokine release syndrome and 60% neurological side effects [32]. However, most of these side effects were not severe and were manageable. Mild cytokine release syndrome is treated with supportive measures such as antipyretics and hydration. More severe cytokine release syndrome requires the use of IL-6R blocking antibodies and immunosuppressants such as corticosteroids [33]. In severe cases, patients may need to be transferred to the ICU for circulatory monitoring and possibly oxygen or respiratory support. While IL-6 blockade is often effective in cytokine release syndrome, it does not help in immune effector cell-associated neurotoxicity syndrome and may even worsen the clinical picture. In addition to supportive measures, corticosteroids are used here, possibly in combination with anakinra. Improvements in CAR design could potentially reduce these sometimes life-threatening side effects. It is known that CARs with a CD28 co-stimulatory domain (e.g. axi-cel) proliferate faster and release higher cytokine concentrations, which can lead to earlier and more-severe cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome compared to 4-1BB-containing CARs (e.g. tisa-cel) [31]. Initial studies suggest that changes in the signalling domain of the CD3ζ chain may improve the side effect profile. Medium-term complications include cytopenias, infections and disease relapse. In the latter case, the therapeutic options are very limited and the prognosis is very poor. An increased risk of secondary malignancies has been reported but further studies, in particular to better understand the association, are warranted. Further challenges are the numerous resources and logistics that such a therapy requires. As a result, the costs are also very high.

Tumour-infiltrating lymphocytes

The use of cellular therapy with tumour-infiltrating lymphocytes has been practiced for several decades, but only in specialised clinics [34–36]. The first patients were treated in the late 1980s [35]. In tumour-infiltrating lymphocyte therapy, T cells are isolated from a sample of the primary tumour or a metastasis [37]. By using IL-2, T cells are activated and multiplied. The treatment works in some patients because tumour-specific T cell clones are present in the resected lesion. These tumour-specific T cells have a T cell receptor that recognises an antigen expressed by the tumour. After lymphodepletion with cyclophosphamide and fludarabine, the tumour-infiltrating lymphocyte product is administered together with IL-2 [37]. Recently, a randomised phase III study conducted in the Netherlands and Denmark in patients with metastatic melanoma was published [5]. Here it was shown that after failure of a standard immunotherapy with immune checkpoint inhibition against PD-1, patients responded better to tumour-infiltrating lymphocyte therapy than to a second-line immune checkpoint inhibition with antibodies directed against CTLA-4. In addition, a commercial product, lifileucel, was recently approved by the FDA in the USA for the treatment of melanoma patients. We have recently conducted the BaseTIL study, in which we treated 9 patients with advanced and heavily pre-treated melanoma [38]. In addition to melanoma patients, in principle all patients with T cells containing T cell receptors that recognise a tumour antigen (often so-called neoantigens, which arise through new mutations in cancer cells) can be treated. Patients with non-small cell lung cancer (NSCLC) have also been successfully treated with tumour-infiltrating lymphocytes [39]. Other immunogenic tumour entities such as cervical carcinoma can also potentially be treated with tumour-infiltrating lymphocyte therapy. We have recently opened a trial for patients with NSCLC at the University Hospital in Basel (NCT06455917).

Tumour-infiltrating lymphocyte therapies can of course also be improved. For example, the T cells that recognise tumour antigens can be separated from the other T cells in the tumour. Thus, potentially much larger numbers of T cells attacking the tumour can be isolated and amplified [37, 40]. In order to obtain a more functional phenotype of the T cells, other cytokines or stimulating antibodies can also be added for in vitro expansion [40]. Tumour-infiltrating lymphocyte therapy can also be combined with other substances. For example, tumour-infiltrating lymphocyte therapy has already been combined with immune checkpoint inhibitors in various studies. In some cases, a good response to this combination therapy was observed in patients with NSCLC [41]. Other forms of IL-2 can also be used. There are newer IL-2 preparations that can specifically stimulate the cytotoxic T cells rather than the regulatory T cells [42]. We are currently conducting the BaseTIL-03M study in Basel, which is testing a combination of such a new IL-2 preparation together with tumour-infiltrating lymphocyte therapy in melanoma patients (NCT05869539).

Side effects of tumour-infiltrating lymphocyte therapy

Cytopenias following lymphodepleting chemotherapy and thus infections and bleeding complications are the most common side effects of tumour-infiltrating lymphocyte therapy [5]. Interleukin-2 therapy can also cause cytokine release syndrome, depending on the dose and intensity of IL-2 administration. In addition to cytokine release syndrome, IL-2 treatment can also lead to vascular leak syndrome, sometimes with pulmonary oedema [37]. It is therefore important to consider the use of vasoactive substances at an early stage. As tumour-infiltrating lymphocyte therapy is a classic immunotherapy, immune-mediated side effects with various organ toxicities can also occur, as with immune checkpoint inhibitor therapy [40]. These often have to be treated with steroids, depending on which organs are involved.

Outlook

Emerging cellular therapies, such as CAR T cell therapy and tumour-infiltrating lymphocyte therapy, have significantly improved prognosis for certain patients. CAR T cell therapies are now routinely used for haematological cancers, primarily B-cell lymphomas, and efforts are underway to expand CAR T cell treatments to additional tumour types. Moreover, CAR T cell therapies may soon offer new treatment options for selected patients with refractory autoimmune diseases and a multitude of studies are ongoing [14]. Tumour-infiltrating lymphocyte therapy also holds promise for inducing long-term remissions in patients with immunogenic tumours. With the recent approval of a commercial tumour-infiltrating lymphocyte product for treating melanoma in the United States, this treatment has become accessible to a wider patient population. Trials are currently evaluating its effectiveness for other tumour types, and advancements in expanding tumour-specific T cells are expected to enhance its efficacy.

The availability of these cellular therapies will place new demands on treatment centres, as more patients are expected to seek these specialised options. While the costs of these therapies, including associated treatments and potential complications, remain high, future logistical improvements may help manage expenses. For example, some centres are already producing tumour-infiltrating lymphocyte therapies in their own cleanroom facilities, which may reduce production costs over time. Additionally, certain preparatory and therapeutic steps may transition to outpatient settings, making these therapies more accessible.

Notes

All authors have completed and submitted the International Committee of Medical Journal Editors form for disclosure of potential conflicts of interest. HL has a grant from Fond’Action for TIL therapy and drug support for a TIL trial from Anaveon. AH received support for attending meetings and/or travel from Amgen (EHA Congress 2025) and Kite/Gilead (EHA-EBMT 7th European CAR T-cell Meeting) and has stock or stock options from Novartis.

Andreas Holbro, MD

Division of Haematology

University Hospital Basel

Petersgraben 4

CH-4031 Basel

andreas.holbro[at]usb.ch

and

Heinz Läubli

Division of Oncology

University Hospital Basel

Petersgraben 4

CH-4031 Basel

heinz.laeubli[at]unibas.ch

References

1. Bicak M, Cimen Bozkus C, Bhardwaj N. Checkpoint therapy in cancer treatment: progress, challenges, and future directions. J Clin Invest. 2024 Sep;134(18):e184846. doi: https://doi.org/10.1172/JCI184846

2. Sharma P, Goswami S, Raychaudhuri D, Siddiqui BA, Singh P, Nagarajan A, et al. Immune checkpoint therapy-current perspectives and future directions. Cell. 2023 Apr;186(8):1652–69. doi: https://doi.org/10.1016/j.cell.2023.03.006

3. Haslam A, Prasad V. Estimation of the Percentage of US Patients With Cancer Who Are Eligible for and Respond to Checkpoint Inhibitor Immunotherapy Drugs. JAMA Netw Open. 2019 May;2(5):e192535. doi: https://doi.org/10.1001/jamanetworkopen.2019.2535

4. June CH, Sadelain M. Chimeric Antigen Receptor Therapy. N Engl J Med. 2018 Jul;379(1):64–73. 10.1056/NEJMra1706169

5. Rohaan MW, Borch TH, van den Berg JH, Met Ö, Kessels R, Geukes Foppen MH, et al. Tumor-Infiltrating Lymphocyte Therapy or Ipilimumab in Advanced Melanoma. N Engl J Med. 2022 Dec;387(23):2113–25. doi: https://doi.org/10.1056/NEJMoa2210233

6. Thomas ED. Bone marrow transplantation from the personal viewpoint. Int J Hematol. 2005 Feb;81(2):89–93. doi: https://doi.org/10.1532/IJH97.04197

7. Pinthus JH, Waks T, Malina V, Kaufman-Francis K, Harmelin A, Aizenberg I, et al. Adoptive immunotherapy of prostate cancer bone lesions using redirected effector lymphocytes. J Clin Invest. 2004 Dec;114(12):1774–81. doi: https://doi.org/10.1172/JCI200422284

8. Labanieh L, Mackall CL. CAR immune cells: design principles, resistance and the next generation. Nature. 2023 Feb;614(7949):635–48. doi: https://doi.org/10.1038/s41586-023-05707-3

9. Westin JR, Oluwole OO, Kersten MJ, Miklos DB, Perales MA, Ghobadi A, et al.; ZUMA-7 Investigators; Kite Members. Survival with Axicabtagene Ciloleucel in Large B-Cell Lymphoma. N Engl J Med. 2023 Jul;389(2):148–57. doi: https://doi.org/10.1056/NEJMoa2301665

10. Rodriguez-Otero P, Ailawadhi S, Arnulf B, Patel K, Cavo M, Nooka AK, et al. Ide-cel or Standard Regimens in Relapsed and Refractory Multiple Myeloma. N Engl J Med. 2023 Mar;388(11):1002–14. doi: https://doi.org/10.1056/NEJMoa2213614

11. Sheykhhasan M, Ahmadieh-Yazdi A, Vicidomini R, Poondla N, Tanzadehpanah H, Dirbaziyan A, et al. CAR T therapies in multiple myeloma: unleashing the future. Cancer Gene Ther. 2024 May;31(5):667–86. doi: https://doi.org/10.1038/s41417-024-00750-2

12. Mailankody S, Devlin SM, Landa J, Nath K, Diamonte C, Carstens EJ, et al. GPRC5D-Targeted CAR T Cells for Myeloma. N Engl J Med. 2022 Sep;387(13):1196–206. doi: https://doi.org/10.1056/NEJMoa2209900

13. Qi C, Gong J, Li J, Liu D, Qin Y, Ge S, et al. Claudin18.2-specific CAR T cells in gastrointestinal cancers: phase 1 trial interim results. Nat Med. 2022 Jun;28(6):1189–98. doi: https://doi.org/10.1038/s41591-022-01800-8

14. Mackensen A, Haanen JB, Koenecke C, Alsdorf W, Wagner-Drouet E, Borchmann P, et al. CLDN6-specific CAR-T cells plus amplifying RNA vaccine in relapsed or refractory solid tumors: the phase 1 BNT211-01 trial. Nat Med. 2023 Nov;29(11):2844–53. doi: https://doi.org/10.1038/s41591-023-02612-0

15. Choi BD, Gerstner ER, Frigault MJ, Leick MB, Mount CW, Balaj L, et al. Intraventricular CARv3-TEAM-E T Cells in Recurrent Glioblastoma. N Engl J Med. 2024 Apr;390(14):1290–8. doi: https://doi.org/10.1056/NEJMoa2314390

16. Bagley SJ, Logun M, Fraietta JA, Wang X, Desai AS, Bagley LJ, et al. Intrathecal bivalent CAR T cells targeting EGFR and IL13Rα2 in recurrent glioblastoma: phase 1 trial interim results. Nat Med. 2024 May;30(5):1320–9. doi: https://doi.org/10.1038/s41591-024-02893-z

17. Majzner RG, Ramakrishna S, Yeom KW, Patel S, Chinnasamy H, Schultz LM, et al. GD2-CAR T cell therapy for H3K27M-mutated diffuse midline gliomas. Nature. 2022 Mar;603(7903):934–41. doi: https://doi.org/10.1038/s41586-022-04489-4

18. D’Angelo SP, Araujo DM, Abdul Razak AR, Agulnik M, Attia S, Blay JY, et al. Afamitresgene autoleucel for advanced synovial sarcoma and myxoid round cell liposarcoma (SPEARHEAD-1): an international, open-label, phase 2 trial. Lancet. 2024 Apr;403(10435):1460–71. doi: https://doi.org/10.1016/S0140-6736(24)00319-2

19. Choe JH, Watchmaker PB, Simic MS, Gilbert RD, Li AW, Krasnow NA, et al. SynNotch-CAR T cells overcome challenges of specificity, heterogeneity, and persistence in treating glioblastoma. Sci Transl Med. 2021 Apr;13(591):eabe7378. doi: https://doi.org/10.1126/scitranslmed.abe7378

20. Smole A, Benton A, Poussin MA, Eiva MA, Mezzanotte C, Camisa B, et al. Expression of inducible factors reprograms CAR-T cells for enhanced function and safety. Cancer Cell. 2022 Dec;40(12):1470–1487.e7. doi: https://doi.org/10.1016/j.ccell.2022.11.006

21. Schett G, Müller F, Taubmann J, Mackensen A, Wang W, Furie RA, et al. Advancements and challenges in CAR T cell therapy in autoimmune diseases. Nat Rev Rheumatol. 2024 Sep;20(9):531–44. doi: https://doi.org/10.1038/s41584-024-01139-z

22. Mougiakakos D, Krönke G, Völkl S, Kretschmann S, Aigner M, Kharboutli S, et al. CD19-Targeted CAR T Cells in Refractory Systemic Lupus Erythematosus. N Engl J Med. 2021 Aug;385(6):567–9. doi: https://doi.org/10.1056/NEJMc2107725

23. Mackensen A, Müller F, Mougiakakos D, Böltz S, Wilhelm A, Aigner M, et al. Anti-CD19 CAR T cell therapy for refractory systemic lupus erythematosus. Nat Med. 2022 Oct;28(10):2124–32. doi: https://doi.org/10.1038/s41591-022-02017-5

24. Müller F, Taubmann J, Bucci L, Wilhelm A, Bergmann C, Völkl S, et al. CD19 CAR T-Cell Therapy in Autoimmune Disease - A Case Series with Follow-up. N Engl J Med. 2024 Feb;390(8):687–700. doi: https://doi.org/10.1056/NEJMoa2308917

25. Taubmann J, Knitza J, Müller F, Völkl S, Aigner M, Kleyer A, et al. Rescue therapy of antisynthetase syndrome with CD19-targeted CAR-T cells after failure of several B-cell depleting antibodies. Rheumatology (Oxford). 2024 Jan;63(1):e12–4. doi: https://doi.org/10.1093/rheumatology/kead330

26. Pecher AC, Hensen L, Klein R, Schairer R, Lutz K, Atar D, et al. CD19-Targeting CAR T Cells for Myositis and Interstitial Lung Disease Associated With Antisynthetase Syndrome. JAMA. 2023 Jun;329(24):2154–62. doi: https://doi.org/10.1001/jama.2023.8753

27. Granit V, Benatar M, Kurtoglu M, Miljković MD, Chahin N, Sahagian G, et al.; MG-001 Study Team. Safety and clinical activity of autologous RNA chimeric antigen receptor T-cell therapy in myasthenia gravis (MG-001): a prospective, multicentre, open-label, non-randomised phase 1b/2a study. Lancet Neurol. 2023 Jul;22(7):578–90. doi: https://doi.org/10.1016/S1474-4422(23)00194-1

28. Müller F, Boeltz S, Knitza J, Aigner M, Völkl S, Kharboutli S, et al. CD19-targeted CAR T cells in refractory antisynthetase syndrome. Lancet. 2023 Mar;401(10379):815–8. doi: https://doi.org/10.1016/S0140-6736(23)00023-5

29. Fischbach F, Richter J, Pfeffer LK, Fehse B, Berger SC, Reinhardt S, et al. CD19-targeted chimeric antigen receptor T cell therapy in two patients with multiple sclerosis. Med (N Y). 2024 Jun;5(6):550–558.e2. doi: https://doi.org/10.1016/j.medj.2024.03.002

30. Brudno JN, Kochenderfer JN. Current understanding and management of CAR T cell-associated toxicities. Nat Rev Clin Oncol. 2024 Jul;21(7):501–21. doi: https://doi.org/10.1038/s41571-024-00903-0

31. Morris EC, Neelapu SS, Giavridis T, Sadelain M. Cytokine release syndrome and associated neurotoxicity in cancer immunotherapy. Nat Rev Immunol. 2022 Feb;22(2):85–96. doi: https://doi.org/10.1038/s41577-021-00547-6

32. Locke FL, Miklos DB, Jacobson CA, Perales MA, Kersten MJ, Oluwole OO, et al.; All ZUMA-7 Investigators and Contributing Kite Members. Axicabtagene Ciloleucel as Second-Line Therapy for Large B-Cell Lymphoma. N Engl J Med. 2022 Feb;386(7):640–54. doi: https://doi.org/10.1056/NEJMoa2116133

33. Jain MD, Smith M, Shah NN. How I treat refractory CRS and ICANS after CAR T-cell therapy. Blood. 2023 May;141(20):2430–42. 

34. Klobuch S, Seijkens TT, Schumacher TN, Haanen JB. Tumour-infiltrating lymphocyte therapy for patients with advanced-stage melanoma. Nat Rev Clin Oncol. 2024 Mar;21(3):173–84. doi: https://doi.org/10.1038/s41571-023-00848-w

35. Rosenberg SA, Packard BS, Aebersold PM, Solomon D, Topalian SL, Toy ST, 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;319(25):1676–80. doi: https://doi.org/10.1056/NEJM198812223192527

36. Goff SL, Dudley ME, Citrin DE, Somerville RP, Wunderlich JR, Danforth DN, et al. Randomized, Prospective Evaluation Comparing Intensity of Lymphodepletion Before Adoptive Transfer of Tumor-Infiltrating Lymphocytes for Patients With Metastatic Melanoma. J Clin Oncol. 2016 Jul;34(20):2389–97. doi: https://doi.org/10.1200/JCO.2016.66.7220

37. Hinrichs CS, Rosenberg SA. Exploiting the curative potential of adoptive T-cell therapy for cancer. Immunol Rev. 2014 Jan;257(1):56–71. doi: https://doi.org/10.1111/imr.12132

38. König D, Sandholzer MT, Uzun S, Zingg A, Ritschard R, Thut H, et al. Melanoma clonal heterogeneity leads to secondary resistance after adoptive cell therapy with tumor-infiltrating lymphocytes. Cancer Immunol Res. 2024 Jul;12(7):814–21. doi: https://doi.org/10.1158/2326-6066.CIR-23-0757

39. 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 Discov. 2024 Aug;14(8):1389–402. doi: https://doi.org/10.1158/2159-8290.CD-23-1334

40. Rohaan MW, van den Berg JH, Kvistborg P, Haanen JB. Adoptive transfer of tumor-infiltrating lymphocytes in melanoma: a viable treatment option. J Immunother Cancer. 2018 Oct;6(1):102. doi: https://doi.org/10.1186/s40425-018-0391-1

41. 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. Nat Med. 2021 Aug;27(8):1410–8. doi: https://doi.org/10.1038/s41591-021-01462-y

42. Joerger M, Calvo E, Laubli H, Lopez J, Alonso G, Corral de la Fuente E, et al. Phase 1 first-in-human dose-escalation study of ANV419 in patients with relapsed/refractory advanced solid tumors. J Immunother Cancer. 2023 Nov;11(11):e007784. doi: https://doi.org/10.1136/jitc-2023-007784