1
|
Valvano L, Vilella R, D’Auria F, D’Arena G, Libonati R, Soda M, Telesca A, Pietrantuono G, Mansueto GR, Villani O, D’Agostino S, Calice G, Statuto T. Prognostic relevance of bone marrow immune cell fractions in newly diagnosed B-cell non-Hodgkin lymphoma patients. Ann Med 2025; 57:2490825. [PMID: 40232295 PMCID: PMC12001853 DOI: 10.1080/07853890.2025.2490825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/16/2025] Open
Abstract
INTRODUCTION Non-Hodgkin lymphomas (NHLs) are the most common hematological malignancies worldwide. Among these, B-cell lymphomas (B-NHLs) are the second leading cause of death in hematologic neoplasms. MATERIAL AND METHODS In this study, a detailed immunophenotypic analysis of lymphocytes in the bone marrow aspirate (BMA) of 75 patients with four different subtypes of B-NHLs was performed at diagnosis. The samples were analyzed by flow cytometry (FC) using a stain-lyse-no wash technique and a comprehensive six-color antibody panel. RESULTS Our data showed a different trend in the percentage values of the distinct lymphocyte subsets, which did not seem to correlate with a worse prognosis, except for B cells in diffuse large B-cell lymphoma (DLBCL), which were significantly higher in stage IV than in stages II and III. ROC curve analysis showed that the B-cell percentage value could be used to predict the stage of the disease. Total lymphocytes and B cells were greater in lymphomas that presented a lower percentage of disease progression, specifically mantle cell lymphoma (MCL) and marginal zone lymphoma (MZL). In contrast, natural killer (NK) and T cells showed higher values in DLBCL and follicular lymphoma (FL), which progressed more frequently. Interestingly, in DLBCL patients with higher percentage values of double positive (DPT) and helper T cells (Th), we observed a good prognosis. Specifically, univariate Cox regression analyses indicated that a higher value of Th cells at diagnosis was a better prognostic predictor in patients with DLBCL. CONCLUSIONS These preliminary findings encourage us to further investigate the role of lymphocyte subpopulations in B-cell NHL.
Collapse
MESH Headings
- Humans
- Male
- Female
- Prognosis
- Middle Aged
- Aged
- Adult
- Flow Cytometry
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/diagnosis
- Lymphoma, B-Cell/pathology
- Aged, 80 and over
- Immunophenotyping
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Bone Marrow/pathology
- Bone Marrow/immunology
- Bone Marrow Cells/immunology
- B-Lymphocytes/immunology
- Disease Progression
- Lymphoma, Mantle-Cell/immunology
- Lymphoma, Mantle-Cell/pathology
- Lymphoma, Mantle-Cell/diagnosis
Collapse
Affiliation(s)
- Luciana Valvano
- Laboratory of Clinical Research and Advanced Diagnostics, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, Italy
| | - Rocchina Vilella
- Laboratory of Clinical Research and Advanced Diagnostics, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, Italy
| | - Fiorella D’Auria
- Laboratory of Clinical Pathology, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, Italy
| | | | - Rossana Libonati
- Laboratory of Clinical Research and Advanced Diagnostics, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, Italy
| | - Michela Soda
- Laboratory of Clinical Research and Advanced Diagnostics, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, Italy
| | - Alessia Telesca
- Laboratory of Clinical Research and Advanced Diagnostics, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, Italy
| | - Giuseppe Pietrantuono
- Hematology and Stem Cell Transplantation Unit, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, Italy
| | - Giovanna Rosaria Mansueto
- Hematology and Stem Cell Transplantation Unit, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, Italy
| | - Oreste Villani
- Hematology and Stem Cell Transplantation Unit, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, Italy
| | - Simona D’Agostino
- Hematology and Stem Cell Transplantation Unit, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, Italy
| | - Giovanni Calice
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, Italy
| | - Teodora Statuto
- Laboratory of Clinical Research and Advanced Diagnostics, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, Italy
| |
Collapse
|
2
|
Wang Y, Fan S, Wang W. Knowledge mapping and visualized analysis of research progress in onconephrology: a bibliometric analysis. Ren Fail 2025; 47:2477302. [PMID: 40101926 PMCID: PMC11921167 DOI: 10.1080/0886022x.2025.2477302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 02/24/2025] [Accepted: 03/01/2025] [Indexed: 03/20/2025] Open
Abstract
OBJECTIVES Onconephrology is an expanding subspecialty focused on the management of cancer patients with renal injury. This study used a comprehensive bibliometric analysis to emphasize the need for cooperation between oncologists and nephrologists, exploring current trends and future research areas in onconephrology. METHODS Relevant literature on onconephrology published between 1 January 2000 and 27 April 2024 was retrieved from the Science Citation Index Expanded of the Web of Science Core Collection, followed by manual screening. Bibliometric analyses were performed using CiteSpace, VOSviewer, and Bibliometrix software. RESULTS A total of 1,853 publications, including 1,647 articles and 206 reviews, by 11,606 authors from 2,757 institutions in 73 countries, were analyzed. Annual publications generally follow a steadily increasing trend, ranging from 25 to 161 documents. The United States (n = 464), The University of Texas MD Anderson Cancer Center (n = 39), Meletios A. Dimopoulos (n = 21), and Nephrology Dialysis Transplantation (n = 35) were the most productive country, institution, author, and journal, respectively. Immune checkpoint inhibitors, glomerular filtration rate, and cisplatin were clusters of highly cited references after 2015. Oxaliplatin, calcium, open-label, and thrombotic microangiopathy were trending topics after 2020. Outcome, acute kidney injury, immunotherapy, and chronic kidney disease were keyword bursts that persisted through 2024. CONCLUSION Current research of onconephrology is focusing on chemotherapeutic nephrotoxicity, kidney function assessment, dosing of chemotherapeutic agents in chronic kidney disease, glomerular disease in cancer, immunotherapy, and electrolyte disturbances. Future directions in this field include clinical trials and thrombotic microangiopathy.
Collapse
Affiliation(s)
- Yiwei Wang
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Shuling Fan
- Department of Nephrology, Shanghai Tenth People's Hospital, Shanghai, China
| | - Wei Wang
- Department of Nephrology, Shanghai Tenth People's Hospital, Shanghai, China
| |
Collapse
|
3
|
Palani HK, Arunachalam AK, Kulkarni U, Yasar M, Venkatraman A, Palanikumar S, Radhakrishnan RN, Solomon M, Rajasekaran A, Bankar A, Datari PVR, Selvarajan S, Korula A, Dash P, Schneider D, Wirthlin L, Abraham A, George B, Mathews V. Safety, efficacy and total cost of point-of-care manufactured anti-CD19 CAR-T cell therapy in India: VELCART trial. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200977. [PMID: 40248244 PMCID: PMC12005290 DOI: 10.1016/j.omton.2025.200977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/11/2025] [Accepted: 03/21/2025] [Indexed: 04/19/2025]
Abstract
Decentralized or point-of-care (POC) manufacture of CAR-T cells is a potential strategy to improve accessibility and reduce cost and logistic challenges. A total of 10 relapsed/refractory patients (B cell acute lymphoblastic leukemia [B-ALL] N = 6, diffuse large B cell lymphoma [DLBCL] N = 4) were enrolled in this POC phase 1 study. Chimeric antigen receptor (CAR)-T cells were manufactured using the fully automated CliniMACS Prodigy system. The CAR-T cell products had a median 15-fold expansion with a median transduction rate of 38%. The immunophenotypic characterization indicates a significant increase in central memory and effector T cells. All the patients were infused with fresh CAR-T cells. Complete remission rates were 100% for B-ALL and 50% for DLBCL. At a median follow-up of 15 months, 8 of 10 patients remain without disease progression. Adverse events reported were cytokine release syndrome grade 2 or higher in 2 of 10 patients. None of the patients developed immune effector cell-associated neurotoxicity syndrome. Late hematological toxicity of grade 2 or higher was noted only in one patient. Evaluation of health care resource utilization demonstrates that the median cost was US$12,724, while the manufacturing cost was US$35,107. Our data highlight the safety, efficacy, low cost, and potential to enhance the accessibility of CAR-T cell therapy in low- and middle-income countries through a fully automated and closed manufacturing platform.
Collapse
Affiliation(s)
- Hamenth Kumar Palani
- Department of Haematology, Christian Medical College, Ranipet Campus, Vellore 632517, Tamil Nadu, India
| | - Arun Kumar Arunachalam
- Department of Haematology, Christian Medical College, Ranipet Campus, Vellore 632517, Tamil Nadu, India
| | - Uday Kulkarni
- Department of Haematology, Christian Medical College, Ranipet Campus, Vellore 632517, Tamil Nadu, India
| | - Mohammed Yasar
- Department of Haematology, Christian Medical College, Ranipet Campus, Vellore 632517, Tamil Nadu, India
| | - Arvind Venkatraman
- Department of Haematology, Christian Medical College, Ranipet Campus, Vellore 632517, Tamil Nadu, India
| | - Swathy Palanikumar
- Department of Haematology, Christian Medical College, Ranipet Campus, Vellore 632517, Tamil Nadu, India
| | | | - Majeela Solomon
- Department of Haematology, Christian Medical College, Ranipet Campus, Vellore 632517, Tamil Nadu, India
| | - Abirami Rajasekaran
- Department of Haematology, Christian Medical College, Ranipet Campus, Vellore 632517, Tamil Nadu, India
| | - Aniket Bankar
- Princess Margaret Cancer Center, University Avenue, Toronto, ON M5G2C1, Canada
| | | | - Sushil Selvarajan
- Department of Haematology, Christian Medical College, Ranipet Campus, Vellore 632517, Tamil Nadu, India
| | - Anu Korula
- Department of Haematology, Christian Medical College, Ranipet Campus, Vellore 632517, Tamil Nadu, India
| | - Pradyot Dash
- Lentigen Technology Inc., A Miltenyi Biotec Company, Gaithersburg, MD 20878, USA
| | - Dina Schneider
- Lentigen Technology Inc., A Miltenyi Biotec Company, Gaithersburg, MD 20878, USA
| | - Louisa Wirthlin
- Lentigen Technology Inc., A Miltenyi Biotec Company, Gaithersburg, MD 20878, USA
| | - Aby Abraham
- Department of Haematology, Christian Medical College, Ranipet Campus, Vellore 632517, Tamil Nadu, India
| | - Biju George
- Department of Haematology, Christian Medical College, Ranipet Campus, Vellore 632517, Tamil Nadu, India
| | - Vikram Mathews
- Department of Haematology, Christian Medical College, Ranipet Campus, Vellore 632517, Tamil Nadu, India
| |
Collapse
|
4
|
Tazuru K, Sone M, Akamine H, Kogue Y, Sims MJ, Ward G, Smyth T, Matsuyama H, Sudo T. The IAP antagonist tolinapant enhances the anti-tumor activity of cell therapies. Eur J Pharmacol 2025; 995:177400. [PMID: 39984012 DOI: 10.1016/j.ejphar.2025.177400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Various gene-modified cell therapies have been investigated in clinical trials, among which chimeric antigen receptor (CAR)-T cell therapy has been approved for the treatment of B cell tumors and has shown remarkable therapeutic effects. However, challenges, such as, cancer recurrence and manufacturing issues remain. To overcome such limitations, we investigated whether combining CAR-T cells with tolinapant, an inhibitor of apoptosis proteins (IAP) antagonist with immunomodulatory activity, could enhance the anti-tumor effect. Tolinapant induced cancer cell death in the presence of TNF-α. Tumor killing by CAR-T, TCR-T or CÊNK cells was enhanced by tolinapant in vitro in a TNF-α-dependent manner. TNF-α secreted from CAR-T cells, in the presence of tolinapant, also induced cell death of antigen-negative cancer cells not in cell-cell contact with CAR-T cells. Addition of tolinapant potentiated efficacy of not only two different CAR-T, but also TCR-T and CAR-NK cells in vivo. Tolinapant treatment led to faster expansion of stimulated CAR-T cells in vitro and in vivo. Our study suggests that the combination of tolinapant improves the efficacy of cell-based cancer therapies by inducing both cancer cell death and CAR-T cell proliferation. This combination therapy may overcome the current limitations of cell-based therapies and enhance their anti-cancer effect.
Collapse
Affiliation(s)
- Keisuke Tazuru
- Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Osaka, Japan.
| | - Masayuki Sone
- Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Osaka, Japan.
| | - Hiroki Akamine
- Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Osaka, Japan.
| | - Yosuke Kogue
- Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Osaka, Japan.
| | | | | | | | - Hironori Matsuyama
- Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Osaka, Japan.
| | - Toshiki Sudo
- Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Osaka, Japan.
| |
Collapse
|
5
|
Hu H, Fan Y, Wang J, Zhang J, Lyu Y, Hou X, Cui J, Zhang Y, Gao J, Zhang T, Nan K. Single-cell technology for cell-based drug delivery and pharmaceutical research. J Control Release 2025; 381:113587. [PMID: 40032008 DOI: 10.1016/j.jconrel.2025.113587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
Leveraging the capacity to precisely manipulate and analyze individual cells, single-cell technology has rapidly become an indispensable tool in the advancement of cell-based drug delivery systems and innovative cell therapies. This technology offers powerful means to address cellular heterogeneity and significantly enhance therapeutic efficacy. Recent breakthroughs in techniques such as single-cell electroporation, mechanical perforation, and encapsulation, particularly when integrated with microfluidics and bioelectronics, have led to remarkable improvements in drug delivery efficiency, reductions in cytotoxicity, and more precise targeting of therapeutic effects. Moreover, single-cell analyses, including advanced sequencing and high-resolution sensing, offer profound insights into complex disease mechanisms, the development of drug resistance, and the intricate processes of stem cell differentiation. This review summarizes the most significant applications of these single-cell technologies, highlighting their impact on the landscape of modern biomedicine. Furthermore, it provides a forward-looking perspective on future research directions aimed at further optimizing drug delivery strategies and enhancing therapeutic outcomes in the treatment of various diseases.
Collapse
Affiliation(s)
- Huihui Hu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Yunlong Fan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China; MicroTech Medical (Hangzhou) Co., Hangzhou 311100, China
| | - Jiawen Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Jialu Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Yidan Lyu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Xiaoqi Hou
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Jizhai Cui
- Department of Materials Science, Fudan University, Shanghai 200438, China; International Institute of Intelligent Nanorobots and Nanosystems, Fudan University, Shanghai 200438, China
| | - Yamin Zhang
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 117585, Singapore
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China.
| | - Kewang Nan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China.
| |
Collapse
|
6
|
Wang Y, Hu X, Du J, Liu B. CAR-T cell therapy for patients with extramedullary multiple myeloma:Opportunities and challenges. Eur J Cancer 2025; 220:115374. [PMID: 40187093 DOI: 10.1016/j.ejca.2025.115374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/26/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025]
Abstract
Multiple myeloma (MM) is a hematological malignancy characterized by abnormal proliferation of clonal plasma cells, which is usually confined to the bone marrow (BM). But some malignant plasma cells grow independently of the BM, called extramedullary disease (EMD). With the clinical application of proteasome inhibitors, immunomodulators, monoclonal antibodies, and hematopoietic stem cell transplantation, the overall survival of MM patients has been significantly improved, but the survival of patients with EMD is still worse than that of non-EMD patients. There are currently no specific treatment options for EMD. chimeric antigen receptor T (CAR-T) cell therapy has brought a new era of immunotherapy. The application of CAR-T has significantly benefited many MM patients, and CAR-T may be a new hope for patients with EMD in the future. This review retrospectively summarizes the mechanism and prognosis of EMD, focusing on the application and potential of CAR-T in the treatment of EMD. It is hoped that this review can provide ideas for the treatment of EMD with CAR-T in the future.
Collapse
Affiliation(s)
- Yin Wang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xiaoli Hu
- Department of Hematology, Myeloma & Lymphoma Center, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Juan Du
- Department of Hematology, Myeloma & Lymphoma Center, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China.
| | - Bei Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou, China; Department of Hematology, The First Affiliated Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
7
|
Guo R, Wang P. Tumor-derived extracellular vesicles: Hijacking T cell function through exhaustion. Pathol Res Pract 2025; 269:155948. [PMID: 40168777 DOI: 10.1016/j.prp.2025.155948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/17/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025]
Abstract
Extracellular vesicles (EVs) play a vital role in intercellular communication within the tumor microenvironment (TME). These vesicles, secreted by tumor cells, contain proteins, lipids, and nucleic acids that significantly influence immune responses, particularly impacting T-cell function. In cancer, T cell dysfunction and exhaustion-marked by reduced proliferation, diminished cytokine production, and impaired cytotoxic activity-are key barriers to effective immune responses. Tumor-derived extracellular vesicles (TEVs) contribute to this dysfunction by carrying immunosuppressive molecules, such as transforming growth factor-beta (TGF-β) and various microRNAs (miRNAs). These TEV-mediated mechanisms promote T cell exhaustion and foster a broader immunosuppressive environment, enabling tumor progression and immune evasion. Furthermore, TEVs have been implicated in resistance to cancer immunotherapies, including immune checkpoint inhibitors and T cell therapies. Understanding the molecular pathways and cargoes within TEVs that drive T cell dysfunction is crucial for developing novel therapeutic strategies aimed at reinvigorating exhausted T cells, enhancing anti-tumor immunity, and improving cancer treatment outcomes.
Collapse
Affiliation(s)
- RuiJuan Guo
- Department of Oncology, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, Shandong 264003, China
| | - Ping Wang
- Department of Oncology, Yantaishan Hospital Affiliated to Binzhou Medical University, Yantai, Shandong 264003, China.
| |
Collapse
|
8
|
Qi S, Li J, Gu X, Zhang Y, Zhou W, Wang F, Wang W. Impacts of ageing on the efficacy of CAR-T cell therapy. Ageing Res Rev 2025; 107:102715. [PMID: 40058461 DOI: 10.1016/j.arr.2025.102715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/19/2025] [Accepted: 03/02/2025] [Indexed: 03/17/2025]
Abstract
Chimeric antigen receptor T cells recognizing CD19 (19CAR-T) cell therapy has achieved robust clinical efficacy when treating some hematological malignancies, but which patient subgroups benefit mostly remains elusive. Here we summarized the data of 541 patients from 30 clinical trials who underwent 19 CAR-T therapy and analyzed the different clinical responses between young (<44 years), middle-aged (45-59 years) and elderly (>60 years) patients and found that the young patients showed a higher level of complete response (CR) rate. Therefore, we then summarize the advances of studies focusing on the effects of age on anti-tumor efficacy of CAR-T therapy and analyze the reasons for the low CR rate after CAR-T cell therapy in elderly patients with tumors, aiming to provide hints for oncologists to select the most suitable candidate for this cancer immunotherapy.
Collapse
Affiliation(s)
- Shimao Qi
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Jiaqian Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Xinyu Gu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Yalan Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Weilin Zhou
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Fengling Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China
| | - Wei Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
9
|
Skrabalak I, Rajtak A, Malachowska B, Skrzypczak N, Skalina KA, Guha C, Kotarski J, Okla K. Therapy resistance: Modulating evolutionarily conserved heat shock protein machinery in cancer. Cancer Lett 2025; 616:217571. [PMID: 39986370 DOI: 10.1016/j.canlet.2025.217571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025]
Abstract
Therapy resistance is a major barrier to achieving a cure in cancer patients, often resulting in relapses and mortality. Heat shock proteins (HSPs) are a group of evolutionarily conserved proteins that play a prominent role in the progression of cancer and drug resistance. HSP synthesis is upregulated in cancer cells, facilitating adaptation to various tumor microenvironment (TME) stressors, including nutrient deprivation, exposure to DNA-damaging agents, hypoxia, and immune responses. In this review, we present background information about HSP-mediated cancer therapy resistance. Within this context, we emphasize recent progress in the understanding of HSP machinery, exploring the therapeutic potential of HSPs in cancer treatment.
Collapse
Affiliation(s)
- Ilona Skrabalak
- The First Department of Oncologic Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
| | - Alicja Rajtak
- The First Department of Oncologic Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland; IOA, 3 Lotnicza St, 20-322 Lublin, Poland
| | - Beata Malachowska
- Department of Radiation Oncology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Natalia Skrzypczak
- Department of Pathology and Clinical Laboratories, University of Michigan, Ann Arbor, MI, USA
| | - Karin A Skalina
- Department of Radiation Oncology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Chandan Guha
- Department of Radiation Oncology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Jan Kotarski
- The First Department of Oncologic Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
| | - Karolina Okla
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA; IOA, 3 Lotnicza St, 20-322 Lublin, Poland.
| |
Collapse
|
10
|
Mougiakakos D, Meyer EH, Schett G. CAR T cells in autoimmunity: game changer or stepping stone? Blood 2025; 145:1841-1849. [PMID: 39700499 DOI: 10.1182/blood.2024025413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/19/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024] Open
Abstract
ABSTRACT The advent of chimeric antigen receptor (CAR) T cells has revolutionized the treatment landscape for hematologic malignancies, and emerging evidence suggests their potential in autoimmune diseases (AIDs). This article evaluates the early successes and future implications of B-cell-targeting CAR T-cell therapy in AIDs. Initial applications, particularly in refractory systemic lupus erythematosus, have demonstrated significant and durable clinical remissions, with accompanying evaluation of the immune system suggesting a so-called "reset" of innate inflammation and adaptive autoimmunity. This has generated widespread interest in expanding this therapeutic approach. CAR T cells offer unique advantages over other treatment modalities, including very deep B-cell depletion and unique therapeutic activity within inflamed tissues and associated lymphoid structures. However, the field must address key concerns, including long-term toxicity, particularly the risk of secondary malignancies, and future accessibility given the higher prevalence of AIDs compared with malignancies. Technological advances in cell therapy, such as next-generation CAR T cells, allogeneic off-the-shelf products, and alternative cell types, such as regulatory CAR T cells, are being explored in AIDs to improve efficacy and safety. In addition, bispecific antibodies are emerging as potential alternatives or complements to CAR T cells, potentially offering comparable efficacy without the need for complex logistics, lymphodepletion, and the risk of insertional mutagenesis. As the field evolves, cellular therapists will play a critical role in the multidisciplinary teams managing these complex cases. The transformative potential of CAR T cells in AIDs is undeniable, but careful consideration of safety, efficacy, and implementation is essential as this novel therapeutic approach moves forward.
Collapse
Affiliation(s)
- Dimitrios Mougiakakos
- Department of Hematology, Oncology, and Cell Therapy, Otto von Guericke University, Magdeburg, Germany
| | - Everett H Meyer
- Cellular Immune Tolerance Program, Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Georg Schett
- Department of Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University, Erlangen, Germany
| |
Collapse
|
11
|
Ielo C, Breccia M. Extracellular vesicles as source of biomarkers in hematological malignancies: looking towards clinical applications. Expert Rev Mol Diagn 2025:1-12. [PMID: 40178353 DOI: 10.1080/14737159.2025.2488919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 04/01/2025] [Indexed: 04/05/2025]
Abstract
INTRODUCTION Extracellular vesicles are membranous particles released by cells in physiological and pathological conditions. Their cargo is heterogeneous since it includes different biomolecules such as nucleic acids and proteins. Oncogenic alterations affect the composition of extracellular vesicles and model their content during cancer evolution. AREAS COVERED This review provides an overview of the studies focused on extracellular vesicles as source of biomarkers in hematological malignancies. A special insight into extracellular vesicles-derived biomarkers as tools for evaluating the prognosis of hematological malignancies and their response to treatment is given. EXPERT OPINION Extracellular vesicles are a valuable source of biomarkers in hematological malignancies. However, the translation from the bench to the bedside is challenged by the lack of standardization of the preanalytical variables of the experimental workflow. The release of standard operating procedures and the validation of the extracellular vesicles-derived biomarkers in large cohort of patients will help in exploiting the potential of extracellular vesicles in the clinical setting.
Collapse
Affiliation(s)
- Claudia Ielo
- Department of Translational and Precision Medicine, Sapienza University of Rome - Azienda Policlinico Umberto I, Rome, Italy
| | - Massimo Breccia
- Department of Translational and Precision Medicine, Sapienza University of Rome - Azienda Policlinico Umberto I, Rome, Italy
| |
Collapse
|
12
|
Tual M, Bellemare-Pelletier A, Moore S, Guipouy D, Farzam-Kia N, Jafarzadeh L, Quenneville J, Barrette B, Saba-El-Leil MK, Delisle JS, Gagnon E. MARC, a novel modular chimeric antigen receptor, improves T cell-based cancer immunotherapies by preventing early T cell exhaustion and enhancing persistence. J Immunother Cancer 2025; 13:e011829. [PMID: 40254394 PMCID: PMC12010287 DOI: 10.1136/jitc-2025-011829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 03/26/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Chimeric antigen receptor T cell (CAR-T)-based immunotherapies have reshaped the therapeutic landscape of cancer treatment, in particular for patients afflicted with leukemia. However, defects in CAR behaviors and clinical complications have hindered their widespread application across diverse cancer types. Chief among these defects is high tonic signaling, absent in native activating immune receptors, which accelerates T cell exhaustion and undermines treatment efficacy. We hypothesized that these limitations arise because current CAR architectures fail to replicate the modular design of native activating immune receptors, which integrate distinct receptor and signaling modules. This modular assembly is crucial for maintaining proper receptor regulation and function. METHODS Therefore, we set forth to develop a modular chimeric antigen receptor leveraging the same assembly principles found in native activating immune receptors to reestablish the intrinsic safeguards in receptor expression and signaling. RESULTS The resulting Modular Actuation Receptor Complex (MARC) displayed surface expression levels akin to its native immune receptor counterpart, the NK cell receptor KIR2DS3, while eliminating tonic signaling. In a clinically relevant mouse leukemia model, MARC-T cells exhibited remarkable long-term persistence and a less exhausted phenotype compared with conventional CAR-T cells. CONCLUSIONS With its modular architecture, the MARC offers unparalleled opportunities for optimization and broad applicability across different cell types, paving the way for transformative advancements in cell-based therapies. This innovation holds immense promise as a next-generation therapeutic tool in clinical settings.
Collapse
Affiliation(s)
- Margaux Tual
- Département de microbiologie, Université de Montréal, Montreal, Quebec, Canada
- Université de Montréal Institut de Recherche en Immunologie et en Cancérologie, Montréal, Québec, Canada
| | | | - Susan Moore
- Université de Montréal Institut de Recherche en Immunologie et en Cancérologie, Montréal, Québec, Canada
| | | | | | - Leila Jafarzadeh
- Médicine, Maisonneuve-Rosemont Hospital Research Centre, Montréal, Québec, Canada
| | - Jordan Quenneville
- Université de Montréal Institut de Recherche en Immunologie et en Cancérologie, Montréal, Québec, Canada
- Département de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Benoit Barrette
- Département de biologie et pathologie cellulaire, Université de Montréal, Montreal, Quebec, Canada
| | - Marc K Saba-El-Leil
- Université de Montréal Institut de Recherche en Immunologie et en Cancérologie, Montréal, Québec, Canada
| | | | - Etienne Gagnon
- Université de Montréal Institut de Recherche en Immunologie et en Cancérologie, Montréal, Québec, Canada
- Département de microbiobologie, infectriologie et immunologie, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
13
|
Lei W, Liu H, Deng W, Chen W, Liang Y, Gao W, Yuan X, Guo S, Li P, Wang J, Tong X, Sun YE, Liang A, Qian W. Safety and feasibility of 4-1BB co-stimulated CD19-specific CAR-NK cell therapy in refractory/relapsed large B cell lymphoma: a phase 1 trial. NATURE CANCER 2025:10.1038/s43018-025-00940-3. [PMID: 40251398 DOI: 10.1038/s43018-025-00940-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 03/05/2025] [Indexed: 04/20/2025]
Abstract
Chimeric antigen receptor (CAR)-modified NK (CAR-NK) cells are candidates for next-generation cancer immunotherapies. Here we generated CD19-specific CAR-NK cells with 4-1BB and CD3ζ signaling endo-domains (CD19-BBz CAR-NK) by transduction of cord blood-derived NK cells using baboon envelope pseudotyped lentiviral vectors and demonstrated their antitumor activity in preclinical B cell lymphoma models in female mice. We next conducted a phase 1 dose-escalation trial involving repetitive administration of CAR-NK cells in 8 patients with relapsed/refractory large B cell lymphoma (NCT05472558). Primary end points were safety, maximum tolerated dose, and overall response rate. Secondary end points included duration of response, overall survival, and progression-free survival. No dose-limiting toxicities occurred, and the maximum tolerated dose was not reached. No cases of cytokine release syndrome, neurotoxicity, or graft-versus-host disease were observed. Results showed an overall response rate of 62.5% at day 30, with 4 patients (50%) achieving complete response. The median progression-free survival was 9.5 months, and the median overall survival was not reached. A post hoc exploratory single-cell RNA sequencing analysis revealed molecular features of CAR-NK cells associated with therapeutic efficacy and efficacy-related immune cell interaction networks. This study met the pre-specified end points. In conclusion, CD19-BBz CAR-NK cells were feasible and therapeutically safe, capable of inducing durable response in patients with B cell lymphoma.
Collapse
Affiliation(s)
- Wen Lei
- Department of Hematology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education; Biotherapy Research Center, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Hui Liu
- Department of Hematology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Wenhai Deng
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wei Chen
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Yun Liang
- Department of Hematology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Wenxia Gao
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Xianggui Yuan
- Department of Hematology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Shanshan Guo
- Department of Hematology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ping Li
- Department of Hematology, Tongji Hospital of Tongji University, Shanghai, China
| | - Jinyong Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute for Stem Cell and Regeneration, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xiangmin Tong
- Department of Hematology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China.
| | - Yi Eve Sun
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University, School of Medicine, Shanghai, China.
| | - Aibin Liang
- Department of Hematology, Tongji Hospital of Tongji University, Shanghai, China.
| | - Wenbin Qian
- Department of Hematology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education; Biotherapy Research Center, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
14
|
Tin E, Khatri I, Fang K, Na Y, Nawata M, Arteaga J, Minden MD, Rutella S, Lee J, Zhang L. Single-cell RNA sequencing of human double-negative T cells reveals a favorable cellular signature for cancer therapy. J Immunother Cancer 2025; 13:e010684. [PMID: 40246580 PMCID: PMC12007110 DOI: 10.1136/jitc-2024-010684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 04/02/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Allogeneic double-negative T-cell (DNT) therapy has emerged as a novel, off-the-shelf cellular treatment with clinical feasibility, safety, and promising efficacy against leukemia. However, the biology of DNTs is less well characterized, and how DNT therapy distinguishes from conventional γδ T-cell therapy remains unclear. Collectively, this hinders our ability to bolster DNT functionalities in cancer therapy. Here, we performed single-cell RNA sequencing with in vitro and in vivo functional analysis on DNTs. As a significant proportion of DNTs express Vγ9Vδ2 (Vδ2) TCR chain, we compared DNTs with donor-matched conventional Vδ2 T cells expanded with zoledronic acid. METHODS Healthy donor-derived allogeneic DNTs and Vδ2 T cells were expanded ex vivo. Single-cell RNA sequencing analysis was performed on both cellular products to identify the transcriptional landscape and inferred cellular interactions within DNTs, followed by comparisons with donor-matched Vδ2 T cells. Unique cellular subsets found only in DNTs were depleted to identify their contributions to the overall efficacy of DNTs against acute myeloid leukemia. The anti-leukemic activity and in vivo persistence of DNTs and Vδ2 T-cells were explored using flow cytometry-based cytotoxicity assays, memory phenotyping, and xenograft models. RESULTS Despite a shared Vδ2 expression between cellular products, we identified unique cellular compositions in DNTs that contribute to distinct transcriptional and cellular communication patterns relative to the donor-matched Vδ2 T cells, including higher expression of genes identified in chimeric antigen receptor T cells that persist in patients with durable cancer-remission. Vδ2- DNTs exhibited strong persistence characteristics, and their presence promoted the cytotoxic capabilities of Vδ2+ DNTs in repeated stimulation assays. This unique genetic signature and diverse cellular composition of DNTs resulted in better overall ex vivo expansion, prolonged persistence, and superior anti-leukemic activity compared with Vδ2 T cells in vitro and in vivo. CONCLUSIONS These results highlight the unique transcriptional, cellular, and functional profile of human DNTs and support the continued clinical investigation of allogeneic DNT therapy. The data also provide a reference gene signature that may help improve the efficacy of other types of allogeneic adoptive cellular therapies.
Collapse
Affiliation(s)
- Enoch Tin
- University Health Network, Toronto, Ontario, Canada
- Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Ismat Khatri
- University Health Network, Toronto, Ontario, Canada
| | - Karen Fang
- University Health Network, Toronto, Ontario, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Yoosu Na
- University Health Network, Toronto, Ontario, Canada
| | - Michele Nawata
- Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute, Calgary, Alberta, Canada
| | - Juan Arteaga
- Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute, Calgary, Alberta, Canada
| | | | - Sergio Rutella
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK
| | - Jongbok Lee
- Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute, Calgary, Alberta, Canada
| | - Li Zhang
- University Health Network, Toronto, Ontario, Canada
- Immunology, University of Toronto, Toronto, Ontario, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
15
|
Garelick D, Isenberg DA. Baby you can drive my CAR-T cells. Lupus 2025:9612033251335798. [PMID: 40241341 DOI: 10.1177/09612033251335798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
ObjectiveTo evaluate the potential of chimeric antigen receptor T-cell (CAR-T) therapy in revolutionizing the treatment of systemic lupus erythematosus (SLE) and to outline necessary future steps for its implementation.MethodsA careful literature search was conducted for relevant English language papers on pubmed.ResultsPreliminary data suggest that CAR-T therapy could significantly improve SLE outcomes. Demonstrating remarkable clinical and serologic improvements in SLE patients, with all treated patients achieving remission and discontinuing conventional steroids and immunosuppressive drugs. To realize this potential, it is imperative to advance our understanding and application of CAR-T therapy. Rigorous research is necessary to validate current findings, and clinical trials must be conducted to assess both the short- and long-term efficacy and safety across diverse populations. Identifying appropriate patient populations is crucial, as CAR-T may also address compliance issues. Despite its current high cost, the financial burden is comparable to the long-term costs of severe SLE treatment. Strategies to reduce costs, including production efficiencies and outpatient treatment options, are under exploration. Early intervention could enhance its feasibility and impact on long-term prognosis.ConclusionCAR-T therapy holds promise for altering the prognosis of SLE and potentially offering a cure. However, substantial efforts are required to validate its efficacy, ensure safety, identify suitable patient cohorts, and reduce financial barriers. This development represents an exciting advancement in SLE treatment, necessitating urgent and focused research and clinical application.
Collapse
Affiliation(s)
- Daniela Garelick
- Centre for Ageing, Rheumatology & Regenerative Medicine Department, UCL, London, UK
| | - David A Isenberg
- Centre for Ageing, Rheumatology & Regenerative Medicine Department, UCL, London, UK
| |
Collapse
|
16
|
Tan EH, Aljurf M, Hussain F, Chabannon C, Worel N, Weisdorf D, Yakoub-Agha I, Galeano S, Sanchez-Guijo F, Garderet L, Atsuta Y, Ruggeri A, Hamad N, Hashmi S, Frutos C, Kodera Y, Seber A, Bonfim C, Niederwieser D, Rondelli D, Greinix H, Koh MB. Perspectives on the use and availability of chimeric antigen receptor T cells (CAR-T) and cell therapies: A worldwide cross-sectional survey by the worldwide network for blood and marrow transplantation (WBMT). Curr Res Transl Med 2025; 73:103515. [PMID: 40253930 DOI: 10.1016/j.retram.2025.103515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/04/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025]
Abstract
Chimeric antigen receptor T cell therapy (CAR-T) cells represent a new generation of autologous, allogeneic and personalised cell-based therapies that have revolutionised the treatment of B cell haematological malignancies. Despite their significant effectiveness in treating challenging relapsed and refractory diseases, access to this cutting-edge treatment remains a critical issue globally, even in high income countries. To gain insights into these challenges, the Worldwide Network for Blood & Marrow Transplantation (WBMT) initiated a survey focused on the state of CAR-T and cellular therapy availability worldwide. The survey aimed to identify the accessibility, manufacturing capabilities, apheresis, accreditation, reimbursement, presence of regulatory frameworks and legal oversight of these cell-based therapies. The survey included questions on demographics, the respondent's centre, CAR-T availability, details about haematopoietic stem cell transplant programs, supply and indications for CAR-T, quality assurance, and information about other cell and gene therapy products beside CAR-T. Conducted online over three months in 2023, the survey garnered 181 complete responses from various geographical regions, from North America, Asia, Europe, South and Central America, Australia and New Zealand, and Africa. Our findings suggested a promising level of awareness and interest in CAR-T therapy globally, even in lower-income regions. However, survey respondents cited cost as the primary barrier to access, alongside infrastructure and governmental support issues. The survey also highlighted the varying reimbursement strategies across regions, with costs in Europe and North America being relatively similar while Asia showed more variability. There was also variability in the regulatory and accreditation frameworks associated with delivery of these novel therapies As CAR-T therapy continues to grow, innovative solutions such as global partnerships, in-house production, and the establishment of cellular therapy centres in developing countries are essential. Addressing the challenges of access requires a comprehensive approach that combines efforts to lower costs, enhance healthcare infrastructure, and foster international collaborations, ensuring that CAR-T therapy becomes available to all who need it.
Collapse
Affiliation(s)
- Eddie Hp Tan
- Cell and Gene Therapy Facility, Health Sciences Authority Singapore, HSA, Singapore
| | - Mahmoud Aljurf
- King Faisal Specialist Hospital and Research Centre, KSA, Riyadh, Saudi Arabia
| | - Fazal Hussain
- Department of Medicine, University of Texas Health Science Center San Antonio, TX, USA
| | - Christian Chabannon
- Institut Paoli-Calmettes, Inserm CBT-1409 & Aix-Marseille Université, Marseille, France
| | - Nina Worel
- Department of Transfusion Medicine and Cell Therapy, Medical University of Vienna, Vienna, Austria
| | - Daniel Weisdorf
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Ibrahim Yakoub-Agha
- Centre Hospitalier Universitaire de Lille, Université de Lille, INSERM U1286, Lille, France
| | | | - Fermin Sanchez-Guijo
- Hematology Department, IBSAL-University Hospital of Salamanca and University of Salamanca, Salamanca, Spain
| | - Laurent Garderet
- Sorbonne Université, Hôpital Pitié Salpêtrière, Service d'hématologie, APHP, Paris, France
| | - Yoshiko Atsuta
- Center for Hematopoietic Stem Cell Transplantation, Aichi Medical University Hospital, Nagakute, Japan
| | | | - Nada Hamad
- Australasian Bone Marrow Transplant Recipient Registry (ABMTRR), St. Vincent´s Hospital Sydney, Australia
| | - Sharukh Hashmi
- Department of Medicine, Khalifa University, SSMC, Abu Dhabi, and College of Medical and Health Sciences, Abu Dhabi, United Arab Emirates
| | | | - Yoshihisa Kodera
- Department of Promotion for Blood and Marrow Transplantation, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Adriana Seber
- Department of Medicine, Universidad de Federal de Sao Paulo Escola Paulista de Medicina, Sao Paulo, Brazil
| | - Carmem Bonfim
- Pele Pequeno Principe, Research Institute/Pediatric Blood and Marrow Transplantation Program Hospital Pequeno Principe, Curitiba, Brazil
| | - Dietger Niederwieser
- University of Leipzig, Leipzig, Germany; Aichi Medical University School of Medicine, Nagakute, Japan; Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | | | - Mickey Bc Koh
- Department of Haematology, St George's University Hospital and Infection and Immunity CAG, City St George's, University of London, United Kingdom; Cell Therapy Facility, Blood Services Group, Health Sciences Authority, Singapore.
| |
Collapse
|
17
|
Du B, Qin J, Lin B, Zhang J, Li D, Liu M. CAR-T therapy in solid tumors. Cancer Cell 2025; 43:665-679. [PMID: 40233718 DOI: 10.1016/j.ccell.2025.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/17/2025] [Accepted: 03/12/2025] [Indexed: 04/17/2025]
Abstract
While chimeric antigen receptor (CAR) T cell therapy has shown great success in hematologic malignancies, the effectiveness in solid tumors has been limited by several factors, including antigenic heterogeneity and the immunosuppressive nature of the tumor microenvironment (TME). In this review, we discuss the advancements made in clinical studies and challenges faced by CAR-T therapy for solid tumors. To enhance CAR-T cell efficacy in solid tumors, we explore strategies such as enhancing T cell persistence and cytotoxicity, targeting multiple antigens, and utilizing innovative allogeneic CAR-T cell manufacturing. Additionally, we highlight the potential benefits of combining CAR-T therapies with immune checkpoint inhibitors and other treatment modalities to overcome TME limitations. We remain optimistic about the future of CAR-T cell therapy in solid tumors, emphasizing the need for continued research to refine therapeutic approaches and address the clinical needs of patients with cancer.
Collapse
Affiliation(s)
- Bing Du
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China.
| | - Juliang Qin
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Boxu Lin
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jiqin Zhang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Dali Li
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Mingyao Liu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
18
|
Zhao L, Yan F, Tang D, Li W, An N, Ren C, Wang Y, Xu K, Zhao K. The transition between M1 and M2 macrophage phenotypes is associated with the disease status following CD19 CAR-T therapy for B cell lymphoma/leukemia. Cell Death Dis 2025; 16:275. [PMID: 40216772 PMCID: PMC11992075 DOI: 10.1038/s41419-025-07610-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/19/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025]
Abstract
Although anti-CD19 chimeric antigen receptor (CAR-T) cells demonstrate high response rates in relapsed/refractory B-cell lymphomas, a considerable proportion of patients eventually encounter disease progression or relapse. The short-term and long-term outcomes of CAR-T treatment are intricately linked to the tumor microenvironment (TME), wherein macrophages with polarized characteristics can exhibit either anti-tumorigenic or pro-tumorigenic roles. Despite evidence implicating the crucial involvement of macrophages in CAR-T cell-treated lymphoma, their dynamic distribution and immune function related to lymphoma progression remain poorly understood. Immunocompetent mice were utilized to establish syngeneic A20 lymphoma/leukemia models. The distribution and polarization of macrophages were detected using immunohistochemistry (IHC) and flow cytometry techniques. We observed that CD19 CAR-T therapy exhibited significant efficacy in protecting mice against lymphoma, leading to increased infiltration of macrophages into the tumor tissue. Notably, during remission stages, M1-like macrophages (CD11b+F4/80+C206-CD80+) were predominant, whereas in relapsed mice, there was a shift towards M2-like phenotypes (CD11b+F4/80+C206+CD80+). The transition from remissive to relapsed status was accompanied by a reduction in the M1/M2 ratio and a decrease in pro-inflammatory cytokines. Furthermore, quantitative real-time polymerase chain reaction (qRT-PCR) analysis confirmed differential expression levels of CD206 and CD163 between remissive and relapsed mice, while signaling pathways involving PI3K and STAT3 may contribute to the skewing towards M2 polarization. In summary, our findings highlight the dynamic transformation of macrophage polarization during different stages of lymphoma progression and underscore its potential implications for immunotherapeutic interventions.
Collapse
Affiliation(s)
- Li Zhao
- Department of hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fen Yan
- Department of hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Donghai Tang
- Department of hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wenwen Li
- Department of hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Na An
- Department of hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chunxiao Ren
- Department of hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ying Wang
- Department of hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kailin Xu
- Department of hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Blood diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Kai Zhao
- Department of hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Blood diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
19
|
Parveen S, Konde DV, Paikray SK, Tripathy NS, Sahoo L, Samal HB, Dilnawaz F. Nanoimmunotherapy: the smart trooper for cancer therapy. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002308. [PMID: 40230883 PMCID: PMC11996242 DOI: 10.37349/etat.2025.1002308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 03/20/2025] [Indexed: 04/16/2025] Open
Abstract
Immunotherapy has gathered significant attention and is now a widely used cancer treatment that uses the body's immune system to fight cancer. Despite initial successes, its broader clinical application is hindered by limitations such as heterogeneity in patient response and challenges associated with the tumor immune microenvironment. Recent advancements in nanotechnology have offered innovative solutions to these barriers, providing significant enhancements to cancer immunotherapy. Nanotechnology-based approaches exhibit multifaceted mechanisms, including effective anti-tumor immune responses during tumorigenesis and overcoming immune suppression mechanisms to improve immune defense capacity. Nanomedicines, including nanoparticle-based vaccines, liposomes, immune modulators, and gene delivery systems, have demonstrated the ability to activate immune responses, modulate tumor microenvironments, and target specific immune cells. Success metrics in preclinical and early clinical studies, such as improved survival rates, enhanced tumor regression, and elevated immune activation indices, highlight the promise of these technologies. Despite these achievements, several challenges remain, including scaling up manufacturing, addressing off-target effects, and navigating regulatory complexities. The review emphasizes the need for interdisciplinary approaches to address these barriers, ensuring broader clinical adoption. It also provides insights into interdisciplinary approaches, advancements, and the transformative potential of nano-immunotherapy and promising results in checkpoint inhibitor delivery, nanoparticle-mediated photothermal therapy, immunomodulation as well as inhibition by nanoparticles and cancer vaccines.
Collapse
Affiliation(s)
- Suphiya Parveen
- Department of Biotechnology and Genetics, School of Sciences, Jain (Deemed-to-be-University), Bengaluru 560027, Karnataka, India
| | - Dhanshree Vikrant Konde
- Department of Biotechnology and Genetics, School of Sciences, Jain (Deemed-to-be-University), Bengaluru 560027, Karnataka, India
| | - Safal Kumar Paikray
- School of Biotechnology, Centurion University of Technology and Management, Jatni 752050, Odisha, India
| | - Nigam Sekhar Tripathy
- School of Biotechnology, Centurion University of Technology and Management, Jatni 752050, Odisha, India
| | - Liza Sahoo
- School of Biotechnology, Centurion University of Technology and Management, Jatni 752050, Odisha, India
| | - Himansu Bhusan Samal
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Jatni 752050, Odisha, India
| | - Fahima Dilnawaz
- School of Biotechnology, Centurion University of Technology and Management, Jatni 752050, Odisha, India
| |
Collapse
|
20
|
Brody JD, Jørgensen J, Belada D, Costello R, Trněný M, Vitolo U, Lewis DJ, Karimi YH, Sureda A, André M, Wahlin BE, Lugtenburg PJ, Jiang T, Karagoz K, Steele AJ, Abbas A, Wang L, Risum M, Cordoba R. Epcoritamab plus GemOx in transplant-ineligible relapsed/refractory DLBCL: results from the EPCORE NHL-2 trial. Blood 2025; 145:1621-1631. [PMID: 39792928 PMCID: PMC12000653 DOI: 10.1182/blood.2024026830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/13/2024] [Accepted: 12/13/2024] [Indexed: 01/12/2025] Open
Abstract
ABSTRACT Patients with relapsed or refractory (R/R) diffuse large B-cell lymphoma (DLBCL) have poor outcomes (complete response [CR] rates with standard salvage therapy gemcitabine plus oxaliplatin [GemOx], ∼30%; median overall survival [OS], 10 to 13 months). Patients with refractory disease fare worse (CR rate with salvage therapy, 7%; median OS, 6 months). Epcoritamab, a CD3×CD20 bispecific antibody approved for R/R DLBCL after ≥2 therapy lines, has shown promising safety and efficacy in various combinations. We report results from the phase 1b/2 EPCORE NHL-2 trial evaluating epcoritamab plus GemOx in autologous stem cell transplant (ASCT)-ineligible R/R DLBCL. Patients received 48 mg subcutaneous epcoritamab after 2 step-up doses until progression or unacceptable toxicity; GemOx was given once every 2 weeks for 8 doses. The primary end point was overall response rate (ORR). As of 15 December 2023, 103 patients were enrolled (median follow-up, 13.2 months; median age, 72 years). Patients had challenging-to-treat disease: ≥2 prior therapy lines, 62%; prior chimeric antigen receptor T-cell therapy, 28%; primary refractory disease, 52%; refractory to last therapy, 70%. ORR and CR rate were 85% and 61%, respectively. Median duration of CR and OS were 23.6 and 21.6 months, respectively. Common treatment-emergent adverse events were cytopenias and cytokine release syndrome (CRS). CRS events had predictable timing, were primarily low grade (52% overall, 1% grade 3), and resolved without leading to discontinuation. Epcoritamab plus GemOx yielded deep, durable responses and favorable long-term outcomes in ASCT-ineligible R/R DLBCL. This trial was registered at www.clinicaltrials.gov as #NCT04663347.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Female
- Male
- Middle Aged
- Aged
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Adult
- Antibodies, Bispecific/administration & dosage
- Antibodies, Bispecific/adverse effects
- Antibodies, Bispecific/therapeutic use
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/administration & dosage
- Deoxycytidine/adverse effects
- Deoxycytidine/therapeutic use
- Aged, 80 and over
- Salvage Therapy
- Oxaliplatin/administration & dosage
- Oxaliplatin/adverse effects
Collapse
Affiliation(s)
- Joshua D. Brody
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Judit Jørgensen
- Department of Haematology, Aarhus University Hospital, Aarhus, Denmark
| | - David Belada
- 4th Department of Internal Medicine, Hematology, University Hospital and Faculty of Medicine in Hradec Králové, Hradec Králové, Czech Republic
| | - Régis Costello
- Service d'Hematologie et Thérapie Cellulaire, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Marek Trněný
- First Department of Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Umberto Vitolo
- Department of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
| | - David John Lewis
- Department of Haematology, Derriford Hospital, University Hospitals Plymouth National Health Service Trust, Plymouth, United Kingdom
| | - Yasmin H. Karimi
- University of Michigan Division of Hematology & Oncology, Ann Arbor, MI
| | - Anna Sureda
- Clinical Hematology Department, Institut Català d’Oncologia, L’Hospitalet, IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | - Marc André
- Service d'Hématologie, Centre Hospitalier Universitaire Université Catholique de Louvain Namur, Yvoir, Belgium
| | - Björn E. Wahlin
- Division of Haematology, Department of Medicine at Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Pieternella J. Lugtenburg
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Lunenburg Lymphoma Phase I/II Consortium-HOVON/LLPC
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Haematology, Aarhus University Hospital, Aarhus, Denmark
- 4th Department of Internal Medicine, Hematology, University Hospital and Faculty of Medicine in Hradec Králové, Hradec Králové, Czech Republic
- Service d'Hematologie et Thérapie Cellulaire, Assistance Publique Hôpitaux de Marseille, Marseille, France
- First Department of Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
- Department of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
- Department of Haematology, Derriford Hospital, University Hospitals Plymouth National Health Service Trust, Plymouth, United Kingdom
- University of Michigan Division of Hematology & Oncology, Ann Arbor, MI
- Clinical Hematology Department, Institut Català d’Oncologia, L’Hospitalet, IDIBELL, Universitat de Barcelona, Barcelona, Spain
- Service d'Hématologie, Centre Hospitalier Universitaire Université Catholique de Louvain Namur, Yvoir, Belgium
- Division of Haematology, Department of Medicine at Huddinge, Karolinska Institutet, Stockholm, Sweden
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
- Oncology Clinical Development, AbbVie, North Chicago, IL
- Translational Data Science, Genmab, Plainsboro, NJ
- Translational Medicine, Genmab, Plainsboro, NJ
- Clinical Science, Genmab, Plainsboro, NJ
- Biostatistics, Genmab, Plainsboro, NJ
- Medical, Genmab, Copenhagen, Denmark
- Department of Hematology, Fundación Jiménez Diaz University Hospital, Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain
| | - Tony Jiang
- Oncology Clinical Development, AbbVie, North Chicago, IL
| | | | | | | | | | | | - Raul Cordoba
- Department of Hematology, Fundación Jiménez Diaz University Hospital, Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain
| |
Collapse
|
21
|
Perez CR, Garmilla A, Nilsson A, Baghdassarian HM, Gordon KS, Lima LG, Smith BE, Maus MV, Lauffenburger DA, Birnbaum ME. Library-based single-cell analysis of CAR signaling reveals drivers of in vivo persistence. Cell Syst 2025:101260. [PMID: 40215972 DOI: 10.1016/j.cels.2025.101260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/27/2024] [Accepted: 03/17/2025] [Indexed: 04/25/2025]
Abstract
The anti-tumor function of engineered T cells expressing chimeric antigen receptors (CARs) is dependent on signals transduced through intracellular signaling domains (ICDs). Different ICDs are known to drive distinct phenotypes, but systematic investigations into how ICD architectures direct T cell function-particularly at the molecular level-are lacking. Here, we use single-cell sequencing to map diverse signaling inputs to transcriptional outputs, focusing on a defined library of clinically relevant ICD architectures. Informed by these observations, we functionally characterize transcriptionally distinct ICD variants across various contexts to build comprehensive maps from ICD composition to phenotypic output. We identify a unique tonic signaling signature associated with a subset of ICD architectures that drives durable in vivo persistence and efficacy in liquid, but not solid, tumors. Our findings work toward decoding CAR signaling design principles, with implications for the rational design of next-generation ICD architectures optimized for in vivo function.
Collapse
Affiliation(s)
- Caleb R Perez
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Singapore-MIT Alliance for Research and Technology Centre, Singapore, Singapore
| | - Andrea Garmilla
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA; Singapore-MIT Alliance for Research and Technology Centre, Singapore, Singapore; Program in Immunology, Harvard Medical School, Boston, MA, USA; Kranz Family Center for Cancer Research and Cellular Immunotherapy Program, Massachusetts General Hospital, Charlestown, MA, USA
| | - Avlant Nilsson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Hratch M Baghdassarian
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Khloe S Gordon
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Singapore-MIT Alliance for Research and Technology Centre, Singapore, Singapore
| | - Louise G Lima
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Blake E Smith
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA; Program in Immunology, Harvard Medical School, Boston, MA, USA
| | - Marcela V Maus
- Kranz Family Center for Cancer Research and Cellular Immunotherapy Program, Massachusetts General Hospital, Charlestown, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Ragon Institute of MIT, MGH and Harvard, Cambridge, MA, USA
| | - Michael E Birnbaum
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Singapore-MIT Alliance for Research and Technology Centre, Singapore, Singapore; Ragon Institute of MIT, MGH and Harvard, Cambridge, MA, USA.
| |
Collapse
|
22
|
Wermke M, Araurjo DM, Chatterjee M, Tsimberidou AM, Holderried TAW, Jazaeri AA, Reshef R, Bokemeyer C, Alsdorf W, Wetzko K, Brossart P, Aslan K, Backert L, Bunk S, Fritsche J, Gulde S, Hengler S, Hilf N, Hossain MB, Hukelmann J, Kalra M, Krishna D, Kursunel MA, Maurer D, Mayer-Mokler A, Mendrzyk R, Mohamed A, Pozo K, Satelli A, Letizia M, Schuster H, Schoor O, Wagner C, Rammensee HG, Reinhardt C, Singh-Jasuja H, Walter S, Weinschenk T, Luke JJ, Britten CM. Autologous T cell therapy for PRAME + advanced solid tumors in HLA-A*02 + patients: a phase 1 trial. Nat Med 2025:10.1038/s41591-025-03650-6. [PMID: 40205198 DOI: 10.1038/s41591-025-03650-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 03/10/2025] [Indexed: 04/11/2025]
Abstract
In contrast to chimeric antigen receptor T cells, T cell receptor (TCR)-engineered T cells can target intracellular tumor-associated antigens crucial for treating solid tumors. However, most trials published so far show limited clinical activity. Here we report interim data from a first-in-human, multicenter, open-label, 3 + 3 dose-escalation/de-escalation phase 1 trial studying IMA203, an autologous preferentially expressed antigen in melanoma (PRAME)-directed TCR T cell therapy in HLA-A*02+ patients with PRAME+ recurrent and/or refractory solid tumors, including melanoma and sarcoma. Primary objectives include the evaluation of safety and tolerability and the determination of the maximum tolerated dose (MTD) and/or recommended dose for extension. Secondary objectives include the evaluation of IMA203 TCR-engineered T cell persistence in peripheral blood, tumor response as well as duration of response. A total of 27 patients were enrolled in the phase 1a dose escalation and 13 patients in the phase 1b dose extension. IMA203 T cells were safe, and the MTD was not reached. Of the 41 patients receiving treatment (that is, who started lymphodepletion), severe cytokine release syndrome was observed in 4.9% (2/41), and severe neurotoxicity did not occur. In the 40 patients treated with IMA203, an overall response rate consisting of patients with unconfirmed or confirmed response (u/cORR) of 52.5% (21/40) and a cORR of 28.9% (11/38) was observed with a median duration of response of 4.4 months (range, 2.4-23.0, 95% confidence interval: 2.6-not reached) across multiple indications. Rapid T cell engraftment and long-term persistence of IMA203 T cells were observed. IMA203 T cells trafficked to all organs, and confirmed responses were more frequent in patients with higher dose. T cell exhaustion was not observed in the periphery; deep responses were enriched at higher PRAME expression; and higher T cell infiltration resulted in longer progression-free survival. Overall, IMA203 showed promising anti-tumor activity in multiple solid tumors, including refractory melanoma. ClinicalTrials.gov identifier: NCT03686124 .
Collapse
Affiliation(s)
- Martin Wermke
- Department of Medicine I, University Hospital Carl Gustav Carus TU Dresden, Dresden, Germany
- National Center for Tumor Diseases, Dresden, Germany
| | - Dejka M Araurjo
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Manik Chatterjee
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany
| | - Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tobias A W Holderried
- Department of Hematology, Oncology, Immunooncology, Stem Cell Transplantation, and Rheumatology, University Hospital Bonn, Bonn, Germany
| | - Amir A Jazaeri
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ran Reshef
- Columbia University Medical Center, New York, NY, USA
| | - Carsten Bokemeyer
- Department of Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Winfried Alsdorf
- Department of Oncology, Hematology, and Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katrin Wetzko
- Department of Medicine I, University Hospital Carl Gustav Carus TU Dresden, Dresden, Germany
| | - Peter Brossart
- Department of Hematology, Oncology, Immunooncology, Stem Cell Transplantation, and Rheumatology, University Hospital Bonn, Bonn, Germany
| | - Katrin Aslan
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | | | | | | | - Swapna Gulde
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | | | - Norbert Hilf
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jason J Luke
- Cancer Immunotherapeutics Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | | |
Collapse
|
23
|
Teng X, Li S, Zhang C, Ding H, Tian Z, Zhu Y, Liu T, Zhang G, Sun K, Xie H, Tu J, Lu Z. NKG2D/CD28 chimeric receptor boosts cytotoxicity and durability of CAR-T cells for solid and hematological tumors. Exp Hematol Oncol 2025; 14:52. [PMID: 40181405 PMCID: PMC11967049 DOI: 10.1186/s40164-025-00646-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 03/21/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND CAR-T cell therapy faces challenges in solid tumor treatment and hematologic malignancy relapse, among which the limited persistence of CAR-T cells and target antigen downregulation are prominent factors. Therefore, we engineered an NKG2D/CD28 chimeric co-stimulatory receptor (CCR), leveraging its broad ligand expression on tumors to enhance the antitumor activity of MSLN CAR and CD19 CAR-T cells. METHODS We generated MSLN CAR-T and CD19 CAR-T cells co-expressing the NKG2D/CD28 CCR and assessed their antitumor efficacy in vitro and in vivo. CAR-T cell activation, differentiation, and exhaustion were analyzed over time following tumor antigen stimulation. Furthermore, a chronic antigen stimulation model was established using tumor cells with low antigen density to simulate the sustained antigenic pressure encountered in vivo treatment conditions. RESULTS Our study shows that NKG2D/CD28&CAR-T cells exhibit enhanced cytotoxicity against tumor cells, especially those with low antigen density, both in vitro and in vivo. Compared to conventional second-generation MSLN CAR or CD19 CAR-T cells, these dual-targeted NKG2D/CD28&CAR-T cells demonstrate superior sensitivity in recognizing and lysing low-density antigen-expressing lung cancer and leukemia cells, and they are capable of eradicating tumors with low-density antigen expression in vivo. Furthermore, the complementary co-stimulation provided by the 4-1BB and CD28 intracellular domains in the CAR and NKG2D/CD28 promotes cytokine secretion, reduces CAR-T cell exhaustion, and enhances the in vivo persistence of CAR-T cells, significantly improving their antitumor efficacy. CONCLUSION The combination of CAR and NKG2D/CD28 offers a potent strategy to enhance the cytotoxicity and durability of CAR-T cells. This approach is promising for improving therapeutic outcomes in solid and hematological tumors and preventing recurrence in tumors with low target antigen density.
Collapse
Affiliation(s)
- Xia Teng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Shance Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Chaoting Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Huirong Ding
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Zhihua Tian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yuge Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Ting Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Guanyu Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Kang Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Huimin Xie
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jiaxin Tu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Zheming Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
24
|
Xiong Z, Sneiderman CT, Kuminkoski CR, Reinheimer J, Schwegman L, Sever RE, Habib A, Hu B, Agnihotri S, Rajasundaram D, Zinn PO, Forsthuber TG, Pollack IF, Li X, Raphael I, Kohanbash G. Transcript-targeted antigen mapping reveals the potential of POSTN splicing junction epitopes in glioblastoma immunotherapy. Genes Immun 2025:10.1038/s41435-025-00326-6. [PMID: 40181162 DOI: 10.1038/s41435-025-00326-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 03/13/2025] [Accepted: 03/21/2025] [Indexed: 04/05/2025]
Abstract
Tumor antigens are crucial for T-cell mediated immunotherapy, but identified antigens for gliomas remain limited. Aberrant splicing variants are commonly expressed in tumors, resulting in unique tumor isoforms with potential antigenic properties. Herein, we analyzed multi-omics data from 587 glioma patients and assembled a library of putative tumor-enriched isoform antigens (TIA) and corresponding peptides presented on each HLA-I allele. We constructed an individual-specific TIA peptide candidate repertoire for each patient based on their TIA expression and HLA-I haplotypes. TIAs were highly expressed, enriched with glioma malignancy, and demonstrated strong HLA-binding affinity. We focused on periostin isoform-203 (POSTN-203), which was associated with poor survival of patients and contained multiple predicted HLA-restricted peptide epitopes. A selected HLA-A11-restricted peptide from POSTN-203 (POSTN-203A11) induced antigen-specific T-cell responses against both peptide-pulsed and POSTN-203-expressing glioma cells in an HLA-specific manner. Our findings highlight TIAs as a promising source of immunogenic antigens and POSTN-203 as a potential promising target for glioma immunotherapy.
Collapse
Affiliation(s)
- Zujian Xiong
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Chaim T Sneiderman
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chloe R Kuminkoski
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jared Reinheimer
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lance Schwegman
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX, USA
| | - ReidAnn E Sever
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ahmed Habib
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Baoli Hu
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sameer Agnihotri
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Pascal O Zinn
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Thomas G Forsthuber
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX, USA
| | - Ian F Pollack
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Itay Raphael
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gary Kohanbash
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
25
|
Xie YQ, Fussenegger M. Plasmid-based electroporation for efficient genetic engineering in immortalized T lymphocytes. Metab Eng 2025; 91:77-90. [PMID: 40185196 DOI: 10.1016/j.ymben.2025.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 03/07/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
The recent clinical success of genetically modified T-cell therapies underscores the urgent need to accelerate fundamental studies and functional screening methods in T lymphocytes. However, a facile and cost-effective method for efficient genetic engineering of T-cells remains elusive. Current approaches often rely on viral transduction, which is labor-intensive and requires stringent biosafety measures. Plasmid-based electroporation presents an affordable alternative, but remains underexplored in T-cells. Moreover, the availability of prototypical T-cell lines is limited. Here, we address these challenges by focusing on two immortalized murine T-cell lines, HT-2 and CTLL-2, which recapitulate key characteristics of primary T-cells, including cytotoxicity and cytokine-dependent proliferation. Alongside the widely used Jurkat T-cell line, HT-2 and CTLL-2 were successfully transfected with single or multiple genes with high efficiencies by means of optimized electroporation in a cuvette-based system. Notably, optimization of plasmid constructs enabled the delivery of large gene-of-interest (GOI) cargos of up to 6.5 kilobase pairs, as well as stable integration of a GOI into the genome via the Sleeping Beauty transposon system. We also developed advanced methodologies for CRISPR/Cas9-mediated gene editing in immortalized T lymphocytes, achieving knockout efficiencies of up to 97 % and homology-directed repair (HDR)-based targeted knock-in efficiencies of up to 70 %. We believe this optimized plasmid-based electroporation approach will contribute to advances in basic research on lymphocyte biology, as well as providing a practical, cost-effective tool for preclinical studies of T-cell therapies.
Collapse
Affiliation(s)
- Yu-Qing Xie
- Department of Biosystems Science and Engineering, Eidgenössiche Technische Hochschule Zurich, Schanzenstrasse 48, CH-4056, Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, Eidgenössiche Technische Hochschule Zurich, Schanzenstrasse 48, CH-4056, Basel, Switzerland; Faculty of Science, University of Basel, Schanzenstrasse 48, CH-4056, Basel, Switzerland.
| |
Collapse
|
26
|
Uslu U, June CH. Beyond the blood: expanding CAR T cell therapy to solid tumors. Nat Biotechnol 2025; 43:506-515. [PMID: 39533105 DOI: 10.1038/s41587-024-02446-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024]
Abstract
Chimeric antigen receptor (CAR) T cell therapy stands as a transformative advancement in immunotherapy, triumphing against hematological malignancies and, increasingly, autoimmune disorders. After a decade of relatively modest results for solid tumors, recent clinical trials and patient reports have also started to yield promising outcomes in glioblastoma and other challenging solid tumor entities. This Perspective seeks to explore the reasons behind these latest achievements and discusses how they can be sustained and expanded through different strategies involving CAR engineering and synthetic biology. Furthermore, we critically analyze how these breakthroughs can be leveraged to maintain momentum and broaden the therapeutic impact of CAR T cells across a variety of solid tumor landscapes.
Collapse
Affiliation(s)
- Ugur Uslu
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
- Parker Institute for Cancer Immunotherapy at the University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
27
|
Kunitskaya A, Piret JM. Impacts of transient exposure of human T cells to low oxygen, temperature, pH and nutrient levels relevant to bioprocessing for cell therapy applications. Cytotherapy 2025; 27:522-533. [PMID: 39891634 DOI: 10.1016/j.jcyt.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND T-cell therapy advances have stimulated the development of bioprocesses to address the specialized needs of cell therapy manufacturing. During concentrated cell washing, the cells are frequently exposed to transiently reduced oxygen, temperature, pH, and nutrient levels. Longer durations of these conditions can be caused by process deviations or, if they are not harmful, be used to ease the scheduling of process stages during experiments as well as manufacturing. METHODS To avoid unpredictable impacts on T-cell quality during bioprocessing, we measured the influences of such environmental exposures generated by settling 250 million activated human T cells per mL, for up to 6 h at temperatures from 4 to 37°C. RESULTS The measured glucose concentration decreased to as low as 0.5 mM and the pH to 6, while lactate increased up to 55 mM. The concentrated cell conditions at 37°C resulted in by far the greatest losses in viable cell numbers with, on average, only 58% and 41% of the cells recovered after 3 and 6 h, respectively. Likewise, their subsequent cell expansion cultures were substantially reduced even after only 3 h of exposure, and with decreased percentages of central memory T cells and increased percentages of effector memory and effector T cells. Although under similar environmental conditions at room temperatures, the negative impacts of high cell concentrations were greatly diminished for up to 3 h. At 4°C the transient conditions were less extreme, and the cells well maintained for 6 h. CONCLUSIONS Overall, when developing processes and devices for T-cell therapy manufacturing that involve concentrated cells, the results of this study indicate that more practically feasible room temperatures can be used for up to 3 h to obtain high viable cell recoveries whereas lower temperatures such as 4°C should be used if there is a need for more prolonged concentrated T-cell conditions.
Collapse
Affiliation(s)
- Alina Kunitskaya
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; The School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - James M Piret
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; The School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada; Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
28
|
Wang JS, Schellenberg SJ, Demeros A, Lin AY. Exosomes in review: A new frontier in CAR-T cell therapies. Neoplasia 2025; 62:101147. [PMID: 40037165 PMCID: PMC11923832 DOI: 10.1016/j.neo.2025.101147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/20/2025] [Accepted: 02/20/2025] [Indexed: 03/06/2025]
Abstract
Exosomes are extracellular vehicles that facilitate intra-cellular communication via transport of critical proteins and genetic material. Every exosome is intrinsically reflective of the cell from which it was derived and can even mimic effector functions of their parent cells. In recent years, with the success of CAR-T therapies, there has been growing interest in characterizing exosomes derived from CAR-T cells. CAR exosomes contain the same cytotoxic granules as their parent cells and have demonstrated significant anti-tumor activity in vitro and in animal models. Moreover, infusion of CAR exosomes in animal models did not generate cytokine release syndrome. Conversely, there are also novel bispecific antibodies which target tumor-derived exosomes in hopes of derailing immunosuppressive pathways mediated by exosomes produced from malignant cells. The two most promising examples include (a) BsE CD73 x EpCAM which binds and inhibits exosomal CD73 to suppress production of immunosuppressant adenosine and (b) BsE CD3 x PD-L1 which targets exosomal PD-L1 within the tumor microenvironment to guide cytotoxic T-cells towards tumor cells. As our understanding of exosome biology continues to evolve, opportunities for advances in cellular therapies will grow in tandem.
Collapse
Affiliation(s)
- John S Wang
- Northwestern University, Feinberg School of Medicine, Department of Medicine, Chicago, IL, USA
| | - Samuel J Schellenberg
- Northwestern University, Feinberg School of Medicine, Department of Medicine, Chicago, IL, USA
| | | | - Adam Y Lin
- Northwestern University, Feinberg School of Medicine, Department of Medicine, Division of Oncology, Chicago, IL, USA; Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.
| |
Collapse
|
29
|
Watts TH, Yeung KKM, Yu T, Lee S, Eshraghisamani R. TNF/TNFR Superfamily Members in Costimulation of T Cell Responses-Revisited. Annu Rev Immunol 2025; 43:113-142. [PMID: 39745933 DOI: 10.1146/annurev-immunol-082423-040557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Prosurvival tumor necrosis factor receptor (TNFR) superfamily (TNFRSF) members on T cells, including 4-1BB, CD27, GITR, and OX40, support T cell accumulation during clonal expansion, contributing to T cell memory. During viral infection, tumor necrosis factor superfamily (TNFSF) members on inflammatory monocyte-derived antigen-presenting cells (APCs) provide a postpriming signal (signal 4) for T cell accumulation, particularly in the tissues. Patients with loss-of-function mutations in TNFR/TNFSF members reveal a critical role for 4-1BB and CD27 in CD8 T cell control of Epstein-Barr virus and other childhood infections and of OX40 in CD4 T cell responses. Here, on the 20th anniversary of a previous Annual Review of Immunology article about TNFRSF signaling in T cells, we discuss the effects of endogenous TNFRSF signals in T cells upon recognition of TNFSF members on APCs; the role of TNFRSF members, including TNFR2, on regulatory T cells; and recent advances in the incorporation of TNFRSF signaling in T cells into immunotherapeutic strategies for cancer.
Collapse
Affiliation(s)
- Tania H Watts
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada;
| | - Karen K M Yeung
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada;
| | - Tianning Yu
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada;
| | - Seungwoo Lee
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada;
| | | |
Collapse
|
30
|
Bi S, Shen J, Zhu Y, Fan L, Ju H, Liu Y. DNA scaffold-framed natural killer cell with programmed drug release for chemo-adoptive cell therapy. J Control Release 2025; 382:113679. [PMID: 40180252 DOI: 10.1016/j.jconrel.2025.113679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Choosing appropriate delivery system for chemotherapeutic drugs as well as arranging the time spots for adoptive cells administrations is the key to achieve efficient combined chemo-adoptive cell therapy. Tumor-homing character makes adoptive immune cells appropriate targeting delivery carriers, but they are rarely used for chemtoxic payloads considering payloads internalization during administration which impairs adoptive cells. Herein, we frame adoptive NK cells using DNA scaffold with chemotherapeutic payloads fastened exterior, and achieves time-programmed drugs release and NK cell decapsulation to minimize side effects and enhance therapeutic effect. IL-21 nanoparticles are prepared by conjugating cytokine IL-21 with a GSH cleavable linker and act as anchor points for DNA scaffold assembly. Chemotherapeutic payloads are prepared by loading DOX/verapamil drugs to PLGA nanoparticles (PLGAdrugs NPs), and connected to the exterior of DNA scaffold with a ROS cleavable linker. Porous DNA scaffold protects NK cells functions from impairing by chemotherapeutic payloads, while guarantees efficient communication of NK cells with exterior environment to keep tumor homing capability. Reactive oxygen species (ROS) in tumor microenvironment releases PLGAdrugs NPs to perform chemotherapy, which subsequently generates a reductive environment to detach DNA scaffold for NK cell and IL-21 release to achieve combined chemo-adoptive cell therapy with enhanced therapeutic efficiency.
Collapse
Affiliation(s)
- Shiyi Bi
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jieyu Shen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yu Zhu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Lei Fan
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing 210023, China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Ying Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China; Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
31
|
Gorin NC. Not just a combination: a fully-integrated strategy of cellular therapy for acute myeloid leukemia expressing CD7, a heretofore elusive target. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1202-1204. [PMID: 39724395 DOI: 10.1007/s11427-024-2711-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 07/23/2024] [Indexed: 12/28/2024]
Affiliation(s)
- Norbert-Claude Gorin
- Department of Hematology and Cell Therapy, EBMT Global Committee and Paris Office, Hôpital Saint Antoine APHP, Paris, 75012, France.
- Paris Sorbonne University, Paris, 75006, France.
| |
Collapse
|
32
|
Salazar-Riojas R, Alvarado-Navarro DM, Chávez-Estrada YO, Hernández-Navarro AK, Ake-Uc MB, Moncada-Saucedo NK, Jaime-Pérez JC, Quezada-Ramírez SI, Rodriguez-Zuñiga AC, Gómez-Almaguer D, Gómez-De León A. Decentralized Point-of-Care Manufacturing of CD19 Chimeric Antigen Receptor T Cells in Mexico. JCO Glob Oncol 2025; 11:e2400581. [PMID: 40249888 DOI: 10.1200/go-24-00581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/22/2025] [Accepted: 02/21/2025] [Indexed: 04/20/2025] Open
Abstract
PURPOSE To validate and replicate an automated decentralized CD19 chimeric antigen receptor T (CAR-T) cell manufacturing process from healthy adult volunteers in an academic institution in a middle-income country. METHODS Healthy volunteers were recruited and underwent leukapheresis with the continuous mononuclear cell (MNC) collection protocol. Clinical-grade CAR-T cell manufacturing was performed in a closed system using a second-generation CD19 vector with 41BB costimulatory domain. Quality control was assessed at different points in the production process with prespecified release criteria including product's aspect, sterility, cell viability, impurity, and quantity. The target dose formulation was 1 × 106/kg viable CAR-T cells per volunteer. RESULTS Five healthy volunteers were recruited, all donated adequate MNC units, and successfully underwent the manufacturing process. After T-cell culture harvest, the products contained a median CAR-T cell concentration of 16.5 × 106/mL (range, 7.7-22.2 × 106/mL), with a median transduction percentage of 44.7% (range, 39.2%-60.5%) and a median CD3+ cell viability of 97.7% (range, 90.4%-98.7%). Sterility was maintained throughout the manufacturing process. The quantity of cells harvested per kilogram of body weight was 24.6 MB-CART19.1 cells × 106/kg (range, 9.3-33.1 × 106/mL). The quality was similar in both fresh and cryopreserved units. Dose formulations were 1.1 CAR-T cells × 106/kg (range, 1.0-1.2 CAR-T cells × 106/kg). CONCLUSION Our study demonstrates an effective methodology with satisfactory and comparable performance to international reports. Point-of-care manufacturing is a feasible alternative to increase access to CAR-T cells in academic centers.
Collapse
Affiliation(s)
- Rosario Salazar-Riojas
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Dalila M Alvarado-Navarro
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Yair O Chávez-Estrada
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Ana K Hernández-Navarro
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Martha B Ake-Uc
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Nidia K Moncada-Saucedo
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - José C Jaime-Pérez
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Sofía I Quezada-Ramírez
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Anna C Rodriguez-Zuñiga
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - David Gómez-Almaguer
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Andrés Gómez-De León
- Hematology Service, Hospital Universitario "Dr José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| |
Collapse
|
33
|
Valentín-Quiroga J, Zarauza-Santoveña A, López-Collazo E, Ferreira LMR. Chimeric anti-HLA antibody receptor engineered human regulatory T cells suppress alloantigen-specific B cells from pre-sensitized transplant recipients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.27.645777. [PMID: 40236118 PMCID: PMC11996358 DOI: 10.1101/2025.03.27.645777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Organ transplantation is a lifesaving procedure, with 50,000 transplants happening every year in the United States. However, many patients harbor antibodies and B cells directed against allogeneic human leukocyte antigen (HLA) molecules, notably HLA-A2, greatly decreasing their likelihood of receiving a compatible organ. Moreover, antibody-mediated rejection is a significant contributor to chronic transplant rejection. Current strategies to desensitize patients non- specifically target circulating antibodies and B cells, resulting in poor efficacy and complications. Regulatory T cells (Tregs) are immune cells dedicated to suppressing specific immune responses by interacting with both innate and adaptive immune cells. Here, we genetically modified human Tregs with a chimeric anti-HLA antibody receptor (CHAR) consisting of an extracellular HLA-A2 protein fused to a CD28-CD3zeta intracellular signaling domain, driving Treg activation upon recognition of anti-HLA-A2 antibodies on the surface of alloreactive B cells. We find that HLA-A2 CHAR Tregs get activated specifically by anti-HLA-A2 antibody-producing cells. Of note, HLA-A2 CHAR activation does not negatively affect Treg stability, as measured by expression of the Treg lineage transcription factors FOXP3 and HELIOS. Interestingly, HLA-A2 CHAR Tregs are not cytotoxic towards anti-HLA-A2 antibody-producing cells, unlike HLA-A2 CHAR modified conventional CD4 + T cells. Importantly, HLA-A2 CHAR Tregs recognize and significantly suppress high affinity IgG antibody production by B cells from HLA-A2 sensitized patients. Altogether, our results provide proof-of-concept of a new strategy to specifically inhibit alloreactive B cells to desensitize transplant recipients.
Collapse
|
34
|
Davila M, Lee SB, Kang YP, Boucher J, Mandula J, Roselli E, Chang D, Jimenez R, Kotani H, Reid K, Vazquez-Martinez J, Beatty N, Goala P, Sierra-Mondragon R, Liu M, Koomen J, Nguyen J, Hussaini M, Shaw T, Wang X, Faramand R, Jain M, Locke F, Rodriguez P, Sailer C, McSain S, Hamid S, Tariq M, Wang J, Abraham-Miranda J. CAR T cell-driven induction of iNOS in tumor-associated macrophages promotes CAR T cell resistance in B cell lymphoma. RESEARCH SQUARE 2025:rs.3.rs-3481746. [PMID: 40235478 PMCID: PMC11998770 DOI: 10.21203/rs.3.rs-3481746/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Chimeric antigen receptor (CAR) T cell therapies have revolutionized B cell malignancy treatment, but subsets of patients with large B cell lymphoma (LBCL) experience primary resistance or relapse after CAR T cell treatment. To uncover tumor microenvironment (TME)-induced resistance mechanisms, we examined patients' intratumoral immune infiltrates and observed that elevated levels of immunoregulatory macrophages in pre-infusion tumor biopsies are correlated with poor clinical responses. CAR T cell-produced interferon-gamma (IFN-γ) promotes the expression of inducible nitric oxide synthase (iNOS, NOS2) in immunoregulatory macrophages, impairing CAR T cell function. Mechanistically, iNOS-expressing macrophages upregulated the p53 pathway, mediating apoptosis and cell cycle arrest in CAR T cells, while downregulating the MYC pathway involved in ribosome biogenesis and protein synthesis. Furthermore, CAR T cell metabolism is compromised by depletion of glycolytic intermediates and rewiring of the TCA cycle. Pharmacological inhibition of iNOS enhances the CAR T cell treatment efficacy in B cell tumor-bearing mice. Notably, elevated levels of iNOS+CD14+ monocytes were observed in leukaphereses of patients with non-durable response to CAR T cell therapy. These findings suggest that mitigating iNOS in tumor-associated macrophages (TAMs) by blocking IFN-γ secretion from CAR T cells will improve outcomes for LBCL patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Xuefeng Wang
- H. Lee Moffitt Cancer Center & Research Institute
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Canelo-Vilaseca M, Sabbah M, Di Blasi R, Cristinelli C, Sureda A, Caillat-Zucman S, Thieblemont C. Lymphodepletion chemotherapy in chimeric antigen receptor-engineered T (CAR-T) cell therapy in lymphoma. Bone Marrow Transplant 2025:10.1038/s41409-025-02539-9. [PMID: 40148484 DOI: 10.1038/s41409-025-02539-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/29/2024] [Accepted: 02/17/2025] [Indexed: 03/29/2025]
Abstract
The development of chimeric antigen receptor (CAR) T-cells, engineered from peripheral T-lymphocytes of a patient with lymphoma, in order to specifically target tumor cells, has been a revolution in adoptive cell therapy (ACT). As outlined in this review, ACT was initiated by hematopoietic cell transplantation (HSCT) and re-injection of interleukin-boosted tumor-infiltrating lymphocytes (TIL). The innovative venture of genetically modifying autologous peripheral T-cells to target them to cell-surface tumoral antigens through an antibody-derived structure (i.e. independent of major histocompatibility antigen presentation, physiologically necessary for T-cell activation), and intracytoplasmic T-cell costimulatory peptides, via a novel membrane CAR, has been an outstanding breakthrough. Here, focusing on B-cell hematological malignancies and mostly non-Hodgkin lymphoma, attention is brought to the importance of providing an optimal microenvironment for such therapeutic cells to proliferate and positively develop anti-tumoral cytotoxicity. This, perhaps paradoxically, implies a pre-infusion step of deep lymphopenia and deregulation of immunosuppressive mechanisms enhanced by tumoral cells. Fludarabine and cyclophosphamide appear to be the most efficient lymphodepletive drugs in this context, dosage being of importance, as will be illustrated by a thorough literature review.
Collapse
Affiliation(s)
- Marta Canelo-Vilaseca
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint-Louis, Hémato-oncologie, Paris, France
| | - Mohamad Sabbah
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint-Louis, Hémato-oncologie, Paris, France
- Université Paris Cité, Paris, France
| | - Roberta Di Blasi
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint-Louis, Hémato-oncologie, Paris, France
| | - Caterina Cristinelli
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint-Louis, Hémato-oncologie, Paris, France
| | - Anna Sureda
- Clinical Hematology Department, Institut Català d'Oncologia-L'Hospitalet, IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | - Sophie Caillat-Zucman
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint-Louis, Laboratoire d'Immunologie, Paris, France
| | - Catherine Thieblemont
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint-Louis, Hémato-oncologie, Paris, France.
- Université Paris Cité, Paris, France.
- Inserm U1153, Hôpital Saint Louis, Paris, France.
| |
Collapse
|
36
|
Jørgensen LV, Christensen EB, Barnkob MB, Barington T. The clinical landscape of CAR NK cells. Exp Hematol Oncol 2025; 14:46. [PMID: 40149002 PMCID: PMC11951618 DOI: 10.1186/s40164-025-00633-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
Chimeric antigen receptor (CAR) NK cell therapy has emerged as a promising alternative to CAR T cell therapy, offering significant advantages in terms of safety and versatility. Here we explore the current clinical landscape of CAR NK cells, and their application in hematologic malignancies and solid cancers, as well as their potential for treating autoimmune disorders. Our analysis draws from data collected from 120 clinical trials focused on CAR NK cells, and presents insights into the demographics and characteristics of these studies. We further outline the specific targets and diseases under investigation, along with the major cell sources, genetic modifications, combination strategies, preconditioning- and dosing regimens, and manufacturing strategies being utilized. Initial results from 16 of these clinical trials demonstrate promising efficacy of CAR NK cells, particularly in B cell malignancies, where response rates are comparable to those seen with CAR T cells but with lower rates of severe adverse effects, such as cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and graft-versus-host disease (GvHD). However, challenges remain in solid tumor applications, where only modest efficacy has been observed to date. Our analysis reveals that research is increasingly focused on enhancing CAR NK cell persistence, broadening their therapeutic targets, and refining manufacturing processes to improve accessibility and scalability. With recent advancements in NK cell engineering and their increased clinical applications, CAR NK cells are predicted to become an integral component of next-generation immunotherapies, not only for cancer but potentially for immune-mediated diseases as well.
Collapse
Affiliation(s)
- Lasse Vedel Jørgensen
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense, Denmark
| | - Emil Birch Christensen
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense, Denmark
| | - Mike Bogetofte Barnkob
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense, Denmark
| | - Torben Barington
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark.
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense, Denmark.
| |
Collapse
|
37
|
Bours B, Masouridi-Levrat S. Is There (Still) a Place for Sequential Conditioning? Curr Oncol 2025; 32:196. [PMID: 40277753 DOI: 10.3390/curroncol32040196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
There is still an unmet need for the treatment of high-risk hematological malignancies. To date, allogeneic stem cell transplantation remains the only chance of cure. Most patients suffering from high-risk hematological malignancies are of an older age and often present with comorbidities. Moreover, patients achieving remission often suffer from early relapse. Amongst the different treatment options, sequential conditioning has yet to prove its value against other conditioning regimens. Sequential conditioning relies on a short course of intensive chemotherapy that is quickly followed by immunosuppressive conditioning before allogeneic stem cell transplantation. Here, we will try to determine which patients can benefit from sequential conditioning. Amongst the different sequential regimens, we will also try to assess if one regimen is better than all the others. Despite the several studies conducted on sequential conditioning, very few are prospective work and head-to-head comparisons are almost inexistant. Sequential conditioning also relies on the use of prophylactic donor lymphocyte infusion post-transplantation. Hence, limiting non-relapse complications is of primary importance to the allow administration of post-transplant treatment. In the era of new targeting therapies, is there still a place for sequential conditioning? Can patients benefit from an association of new therapeutic agents and sequential conditioning?
Collapse
Affiliation(s)
- Boris Bours
- Division of Hematology, Department of Oncology, Geneva University Hospitals, 1205 Geneva, Switzerland
| | | |
Collapse
|
38
|
Saxena K, Hung SH, Ryu E, Singh S, Zhang Tatarata Q, Zeng Z, Wang Z, Konopleva MY, Yee C. BH3 mimetics augment cytotoxic T cell killing of acute myeloid leukemia via mitochondrial apoptotic mechanism. Cell Death Discov 2025; 11:120. [PMID: 40140361 PMCID: PMC11947210 DOI: 10.1038/s41420-025-02375-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/29/2025] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
Adoptive cell therapy (ACT) can address an unmet clinical need for patients with relapsed/refractory acute myeloid leukemia (AML), but its effect is often modest in the setting of high tumor burden. In this study, we postulated that strategies to lower the AML apoptotic threshold will augment T cell killing of AML cells. BH3 mimetics, such as venetoclax, are a clinically approved class of compounds that predispose cells to intrinsic apoptosis by inhibiting anti-apoptotic mitochondrial proteins. We explored the anti-leukemic efficacy of BH3 mimetics combined with WT1-specific CD8+ T cells on AML cell lines and primary samples from patients with a diverse array of disease characteristics to evaluate if lowering the cellular apoptotic threshold via inhibition of anti-apoptotic mitochondrial proteins can increase leukemic cell sensitivity to T cell therapy. We found that the combination approach of BH3 mimetic and CD8+ T cells led to significantly increased killing of established AML lines as well as of adverse-risk primary AML leukemic blast cells. In contrast to the hypothesis that enhanced killing would be due to combined activation of the intrinsic and extrinsic apoptotic pathways, our data suggests that CTL-mediated killing of AML cells was accomplished primarily through activation of the intrinsic/mitochondrial apoptotic pathway. This highly effective combinatorial activity due to convergence on the mitochondrial apoptotic pathway was conserved across multiple AML cell lines and primary samples, suggesting that mitochondrial priming may represent a novel mechanism of optimizing adoptive cell therapy for AML patients.
Collapse
Affiliation(s)
- Kapil Saxena
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Gilead Sciences Inc., Foster City, CA, USA
| | - Shao-Hsi Hung
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Esther Ryu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shailbala Singh
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qi Zhang Tatarata
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA
| | - Zhihong Zeng
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhe Wang
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Marina Y Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Oncology and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Cassian Yee
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
39
|
Chan LL, Chan SL. Future perspectives on immunotherapy for hepatocellular carcinoma. Ther Adv Med Oncol 2025; 17:17588359251323199. [PMID: 40144682 PMCID: PMC11938898 DOI: 10.1177/17588359251323199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 02/05/2025] [Indexed: 03/28/2025] Open
Abstract
In recent years, several global phase III trials have shown that combinations of immune checkpoint inhibitors (ICIs) offer superior efficacy and survival compared to multi-kinase inhibitors, establishing them as the gold standard for treating patients with advanced hepatocellular carcinoma (HCC). This success has led to investigations into expanding the use of immunotherapy into various other settings and populations, including neoadjuvant and adjuvant therapies, patients with decompensated liver function and those awaiting liver transplantation. Despite its proven efficacy, a significant number of patients still develop resistance to immunotherapy, highlighting the need for innovative strategies to address this challenge. Approaches aimed at enhancing tumour immunogenicity, such as combining immunotherapy with transarterial chemoembolization or radiation therapies, show significant promise. Additionally, novel immunotherapeutics - such as triplet therapy, bispecific antibodies, adoptive T-cell therapy and cancer vaccines - are in early development for HCC. These agents have demonstrated potential for synergistic effects with existing ICIs, with initial studies yielding positive outcomes. In this review, we offer our future perspective on immunotherapy, emphasizing emerging indications, novel combination strategies and the development of new immunotherapeutic agents. Overall, the future of immunotherapy in HCC is brimming with extraordinary potential, set to transform the treatment landscape and redefine the possibilities for managing this challenging disease.
Collapse
Affiliation(s)
- Landon L. Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir YK Pao Centre for Cancer, SIRT, Hong Kong Cancer Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Stephen L. Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir YK Pao Centre for Cancer, SIRT, Hong Kong Cancer Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Shatin, Hong Kong, China
| |
Collapse
|
40
|
Kim YM, Akana RV, Sun C, Laveroni O, Jerby L. Redirecting cytotoxic lymphocytes to breast cancer tumors via metabolite-sensing receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.21.644686. [PMID: 40196673 PMCID: PMC11974742 DOI: 10.1101/2025.03.21.644686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Insufficient infiltration of cytotoxic lymphocytes to solid tumors limits the efficacy of immunotherapies and cell therapies. Here, we report a programmable mechanism to mobilize Natural Killer (NK) and T cells to breast cancer tumors by engineering these cells to express orphan and metabolite-sensing G protein-coupled receptors (GPCRs). First, in vivo and in vitro CRISPR activation screens in NK-92 cells identified GPR183, GPR84, GPR34, GPR18, FPR3, and LPAR2 as top enhancers of both tumor infiltration and chemotaxis to breast cancer. These genes equip NK and T cells with the ability to sense and migrate to chemoattracting metabolites such as 7α,25-dihydroxycholesterol and other factors released from breast cancer. Based on Perturb-seq and functional investigations, GPR183 also enhances effector functions, such that engineering NK and CAR NK cells to express GPR183 enhances their ability to migrate to, infiltrate, and control breast cancer tumors. Our study uncovered metabolite-based tumor immune recruitment mechanisms, opening avenues for spatially targeted cell therapies.
Collapse
Affiliation(s)
- Young-Min Kim
- Department of Genetics, Stanford University School of Medicine; Stanford, CA, USA
| | - Reece V Akana
- Department of Genetics, Stanford University School of Medicine; Stanford, CA, USA
- Cancer Biology Program, Stanford University; Stanford, CA, USA
| | - Chang Sun
- Department of Genetics, Stanford University School of Medicine; Stanford, CA, USA
| | - Olivia Laveroni
- Department of Genetics, Stanford University School of Medicine; Stanford, CA, USA
| | - Livnat Jerby
- Department of Genetics, Stanford University School of Medicine; Stanford, CA, USA
- Cancer Biology Program, Stanford University; Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine; Stanford, CA, USA
- Chan Zuckerberg Biohub; San Francisco, CA, USA
| |
Collapse
|
41
|
Short NJ, Aldoss I, DeAngelo DJ, Konopleva M, Leonard J, Logan AC, Park J, Shah B, Stock W, Jabbour E. Clinical use of measurable residual disease in adult ALL: recommendations from a panel of US experts. Blood Adv 2025; 9:1442-1451. [PMID: 39853316 PMCID: PMC11960638 DOI: 10.1182/bloodadvances.2024015441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/09/2025] [Accepted: 01/09/2025] [Indexed: 01/26/2025] Open
Abstract
ABSTRACT Measurable residual disease (MRD) is a powerful predictor of clinical outcomes in acute lymphoblastic leukemia (ALL). In addition to its clear prognostic importance, MRD information is increasingly used in clinical decision algorithms to guide therapeutic interventions. Although it is well established that achievement of MRD-negative remission is an important end point of ALL therapy, the prognostic and therapeutic implications of MRD in an individual patient are influenced by both disease-related factors (eg, cytomolecular risk) and assay-related factors (eg, sensitivity, specimen source, and timing of assessment), which add complexity to MRD-guided treatment decisions. In this review, we discuss the data supporting the use of MRD assessment in adult ALL and how this information can rationally inform clinical decisions, including selection of patients for MRD-directed therapies or allogeneic hematopoietic stem cell transplantation. We also discuss important interpretative challenges related to novel high sensitivity next-generation sequencing-based MRD assays, which are becoming increasingly used in clinical practice.
Collapse
Affiliation(s)
- Nicholas J. Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ibrahim Aldoss
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA
| | - Daniel J. DeAngelo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Marina Konopleva
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY
| | - Jessica Leonard
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Aaron C. Logan
- Division of Hematology, Blood and Marrow Transplantation, and Cellular Therapy, Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Jae Park
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Bijal Shah
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | - Wendy Stock
- Department of Medicine Section of Hematology-Oncology, University of Chicago, Chicago, IL
| | - Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
42
|
Knelangen N, Bader U, Maniaki E, Langan PS, Engert F, Drees B, Schwarzer J, Kotter B, Kiefer L, Gattinoni L, Engels B, Mittelstaet J, Webster B. CAR T cells re-directed by a rationally designed human peptide tag demonstrate efficacy in preclinical models. Cytotherapy 2025:S1465-3249(25)00592-4. [PMID: 40257412 DOI: 10.1016/j.jcyt.2025.03.506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025]
Abstract
Currently, only a few chimeric antigen receptor (CAR) T cell therapies have been approved by the Food and Drug Administration and European Medicines Agency for the treatment of B-cell malignancies. To enable broader application of the CAR T cell technology in other indications, improved control and flexible targeting of multiple tumor antigens are required. Here, we developed a novel adapter CAR (AdCAR) T cell platform for flexible targeting of multiple tumor antigens. This platform is based on a short peptide tag derived from an interdomain region of fibroblast growth factor receptor 2 (FGFR2), commonly mutated in cancer. To select AdCARs specific for mutated FGFR2-derived peptide tags, a multistep pooled screening approach in primary T cells was employed, incorporating MACS separation and next-generation sequencing. The resulting AdCAR was highly specific for the FGFR2-derived peptide tag. Using different in vitro and in vivo model systems, the activity of AdCAR T cells was shown to be strictly dependent on the presence of the adapter and corresponding target antigen. Moreover, AdCAR T cells could be redirected to different target antigens by the addition of respective adapter molecules (AM). Finally, in situ expression of functional AM in primary T cells under control of a drug-inducible promoter system was demonstrated, highlighting the potential for controlling the activity of AdCAR T cells by cellular micropharmacies.
Collapse
Affiliation(s)
- Nele Knelangen
- Research and Development, Miltenyi Biotec, Bergisch Gladbach, Germany; Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany.
| | - Ulrika Bader
- Research and Development, Miltenyi Biotec, Bergisch Gladbach, Germany.
| | - Evangelia Maniaki
- Research and Development, Miltenyi Biotec, Bergisch Gladbach, Germany
| | - Patricia S Langan
- Research and Development, Lentigen Technology Inc., A Miltenyi Biotec Company, Gaithersburg, Maryland, USA
| | - Fabian Engert
- Research and Development, Miltenyi Biotec, Bergisch Gladbach, Germany
| | - Britta Drees
- Research and Development, Miltenyi Biotec, Bergisch Gladbach, Germany
| | - Juliane Schwarzer
- Research and Development, Miltenyi Biotec, Bergisch Gladbach, Germany
| | - Bettina Kotter
- Research and Development, Miltenyi Biotec, Bergisch Gladbach, Germany
| | - Lukas Kiefer
- Research and Development, Miltenyi Biotec, Bergisch Gladbach, Germany
| | - Luca Gattinoni
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy, Regensburg, Germany; University of Regensburg, Regensburg, Germany
| | - Boris Engels
- Research and Development, Miltenyi Biotec, Bergisch Gladbach, Germany
| | - Joerg Mittelstaet
- Faculty of Life Sciences, Reutlingen University, Reutlingen, Germany
| | | |
Collapse
|
43
|
Bartolini R, Trueb L, Daoudlarian D, Joo V, Noto A, Stadelmann R, Gentner B, Fenwick C, Perreau M, Coukos G, Pantaleo G, Arber C, Obeid M. Enrichment of CD7 +CXCR3 + CAR T cells in infusion products is associated with durable remission in relapsed or refractory diffuse large B-cell lymphoma. Ann Oncol 2025:S0923-7534(25)00122-X. [PMID: 40132760 DOI: 10.1016/j.annonc.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T-cell therapy is the standard of care for relapsed or refractory (R/R) diffuse large B-cell lymphoma (DLBCL). However, more than half of patients fail to achieve durable remission. Identifying predictive biomarkers within the CAR T-cell infusion product (IP) may guide strategies to improve clinical outcomes. PATIENTS AND METHODS This single-center observational study conducted at Lausanne University Hospital (CHUV), Switzerland, analyzed IPs from 13 patients with R/R DLBCL who underwent standard-of-care CAR T-cell therapy. A 39-marker mass cytometry panel was used to compare phenotypic and functional markers between long-term responders (R) and non-responders (NR). Unsupervised and supervised analytic approaches were applied to IP data, and longitudinal peripheral blood samples were collected over 30 days post-infusion to track CAR T-cell subpopulation dynamics. RESULTS At a median follow-up of 13·5 months, median progression-free survival (PFS) was 13·3 months (95% CI 9·7-24·3) in R (n=8) versus 3·5 months (95% CI 0·5-5·4) in NR (n=5) (hazard ratio 56·67 [95% CI 7·3-439·3]; p=0·0001). A CD3+CXCR3+CD7+ CAR T-cell subpopulation-found in both CD4+ and CD8+ compartments-was significantly enriched in R. These cells showed increased expression of perforin, granzyme B, and NKG2D (restricted to CD8+ cells). In contrast, NR had a higher frequency of CXCR3+CD7+LAG3+ CAR T-cells. Surface expression of CD3, CD7, CXCR3, and NKG2D were higher in R, whereas LAG3, Ki67, and CD71 were elevated in NR. A predictive cut-off ratio of CD3+CXCR3+CD7+LAG3+CAR+ T-cells <0·83 and CD3+CXCR3+CD7+NKG2D+CAR+ T-cells >1·034 yielded a predictive accuracy of 0·92. Serum CXCL9 and CXCL10 concentrations did not differ between groups. CONCLUSION Enrichment of CD7+CXCR3+ CAR T-cells alongside elevated NKG2D expression in R, in contrast to higher LAG3 and CD71 in NR, emerged as potentially robust correlates of therapeutic outcome. Although derived from a small, hypothesis-generating cohort, these findings suggest that targeted analysis of IP composition may inform the development of biomarker-driven strategies to optimize CAR T-cell products and improve the likelihood of durable remission in R/R DLBCL.
Collapse
Affiliation(s)
- R Bartolini
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - L Trueb
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Oncology, Immuno-Oncology Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - D Daoudlarian
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - V Joo
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - A Noto
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - R Stadelmann
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Departments of Oncology and Laboratory Medicine, Hematology Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - B Gentner
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Oncology, Immuno-Oncology Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland; Swiss Cancer Center Léman, Lausanne, Switzerland; AGORA Cancer Research Center, Lausanne, Switzerland
| | - C Fenwick
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - M Perreau
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - G Coukos
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Oncology, Immuno-Oncology Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland; Swiss Cancer Center Léman, Lausanne, Switzerland; AGORA Cancer Research Center, Lausanne, Switzerland
| | - G Pantaleo
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland
| | - C Arber
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Oncology, Immuno-Oncology Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland; Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Departments of Oncology and Laboratory Medicine, Hematology Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland; Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland; Swiss Cancer Center Léman, Lausanne, Switzerland; AGORA Cancer Research Center, Lausanne, Switzerland
| | - M Obeid
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Service, Rue du Bugnon 46, CH-1011 Lausanne, Switzerland.
| |
Collapse
|
44
|
Rassek K, Misiak J, Ołdak T, Rozwadowska N, Basak G, Kolanowski T. New player in CAR-T manufacture field: comparison of umbilical cord to peripheral blood strategies. Front Immunol 2025; 16:1561174. [PMID: 40191201 PMCID: PMC11968755 DOI: 10.3389/fimmu.2025.1561174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 02/28/2025] [Indexed: 04/09/2025] Open
Abstract
One of the most successful treatments in hematologic cancer is chimeric antigen receptor (CAR)-T cell-based immunotherapy. However, CAR-T therapy is not without challenges like the costly manufacturing process required to personalize each treatment for individual patients or graft-versus-host disease. Umbilical cord blood (UCB) has been most commonly used for hematopoietic cell transplant as it offers several advantages, including its rich source of hematopoietic stem cells, lower risk of graft-versus-host disease, and easier matching for recipients due to less stringent HLA requirements compared to bone marrow or peripheral blood stem cells. In this review, we have discussed the advantages and disadvantages of different CAR-T cell manufacturing strategies with the use of allogeneic and autologous peripheral blood cells. We compare them to the UCB approach and discuss ongoing pre-clinical and clinical trials in the field. Finally, we propose a cord blood bank as a readily available source of CAR-T cells.
Collapse
Affiliation(s)
- Karolina Rassek
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | | | - Tomasz Ołdak
- FamicordTx, Warsaw, Poland
- Polish Stem Cell Bank (PBKM), Warsaw, Poland
| | - Natalia Rozwadowska
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
- FamicordTx, Warsaw, Poland
| | - Grzegorz Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Tomasz Kolanowski
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
- FamicordTx, Warsaw, Poland
| |
Collapse
|
45
|
Guo S, Liu J, Wang B, Zhang X, Zhao Y, Xu J, Cao X, Zhao M, Xiao X, Zhao M. A viral infection prediction model for patients with r/r B-cell malignancies after CAR-T therapy: a retrospective analysis. Front Oncol 2025; 15:1549809. [PMID: 40190552 PMCID: PMC11968754 DOI: 10.3389/fonc.2025.1549809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 02/24/2025] [Indexed: 04/09/2025] Open
Abstract
Background Chimeric antigen receptor T cell (CAR-T) therapy for relapsed/refractory (r/r) B cell acute lymphoblastic leukemia (B-ALL) and B cell non-Hodgkin lymphoma (B-NHL) patients has shown promising effects, but side effects such as viral infections have been observed. Methods A total of 45 patients with r/r B-ALL and r/r B-NHL were included in this retrospective study. Patient demographics were recorded, with the primary endpoint being viral infection within 3 months post CAR-T treatment. Univariate and multivariate logistic regression analyses and least absolute shrinkage and selection operator (LASSO) regression analysis were used to analyze independent factors. The patients were divided into a training cohort of 28 and a validation cohort of 17 to construct a prediction model based on determined independent factors. The model's discrimination and calibration were assessed using the receiver operating characteristic curve (ROC), calibration plot, and decision curve analysis (DCA curve). Results The univariate and multivariate logistic regression analyses of the 43 patients showed that low baseline lymphocyte ratio was an independent risk factor and using granulocyte colony-stimulating factor (G-CSF) early was a protective factor for viral infection after CAR-T therapy in patients with B-ALL and B-NHL. Based on that, the area under the ROC curve (AUC) of the training cohort and validation cohort was 0.935 (95% CI 0.837-1.000) and 0.869 (95%CI 0.696-1.000), respectively, showing excellent predictive value. Conclusions We established a nomogram to predict the factors' influence on viral infection after CAR-T therapy and found that the ratio of baseline lymphocytes and using G-CSF early or lately were able to predict viral infection after CAR-T therapy in r/r B-ALL and B-NHL.
Collapse
Affiliation(s)
- Shujing Guo
- First Center Clinical College, Tianjin Medical University, Tianjin, China
| | - Jile Liu
- First Center Clinical College, Tianjin Medical University, Tianjin, China
| | - Bing Wang
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Xiaomei Zhang
- School of Medicine, Nankai University, Tianjin, China
| | - Yifan Zhao
- First Center Clinical College, Tianjin Medical University, Tianjin, China
| | - Jianmei Xu
- Department of Hematology, Hebei University Affiliated Hospital, Baoding, Hebei, China
| | - Xinping Cao
- First Center Clinical College, Tianjin Medical University, Tianjin, China
| | - Mohan Zhao
- First Center Clinical College, Tianjin Medical University, Tianjin, China
| | - Xia Xiao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
46
|
Bubb QR, Balood M, Seir GE, Swartzrock L, Haslett E, Ho K, Xu P, Wiltz SG, Sotillo E, Gruber TA, Richards RM, Mackall CL, Czechowicz A. Development of multivalent CAR T cells as dual immunotherapy and conditioning agents. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200944. [PMID: 40034967 PMCID: PMC11872492 DOI: 10.1016/j.omton.2025.200944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/18/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025]
Abstract
Hematopoietic stem cell transplantation (HSCT) is the only definitive cure for pediatric acute myeloid leukemia (AML). Despite adjustments in HSCT protocols and improvements in supportive care, 30% of high-risk patients who receive HSCT as part of their therapy still experience disease relapse with high transplant-related mortality. Relapsed AML has a dismal prognosis, and novel therapies are needed. To improve upon the status quo, HSCT would more effectively eliminate relapse-initiating leukemic cells and be delivered with safer, non-genotoxic conditioning. Here, we investigate hematopoietic cytokine receptors (HCRs) and identify that KIT, MPL, and FLT3 are collectively highly expressed in virtually all pediatric AML samples studied. Further, we establish proof-of-concept of a first-in-class chimeric antigen receptor (CAR) T cell that enables simultaneous targeting of KIT, MPL, and FLT3 through a single receptor, which we term the extracellularly linked concatemeric trivalent cytokine (ELECTRIC) CAR. ELECTRIC CARs exhibit potent cytotoxicity against normal and malignant hematopoietic cells in vitro and display anti-HCR activity in a murine xenograft model. We propose that the ELECTRIC system can be the foundation to developing a non-genotoxic, anti-leukemic conditioning regimen to enable safer, more durable efficacy with minimal toxicity.
Collapse
Affiliation(s)
- Quenton Rashawn Bubb
- Stem Cell Biology and Regenerative Medicine Graduate Program, Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mohammad Balood
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gabe Eduardo Seir
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Leah Swartzrock
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ethan Haslett
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Katie Ho
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Peng Xu
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Saida G. Wiltz
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Elena Sotillo
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tanja A. Gruber
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rebecca M. Richards
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Crystal L. Mackall
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Blood and Stem Cell Transplantation and Cell Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Agnieszka Czechowicz
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
47
|
Renninger J, Kurz L, Stein H. Mitigation and Management of Common Toxicities Associated with the Administration of CAR-T Therapies in Oncology Patients. Drug Saf 2025:10.1007/s40264-025-01538-5. [PMID: 40108072 DOI: 10.1007/s40264-025-01538-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2025] [Indexed: 03/22/2025]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapies are one of the main approaches among targeted cellular therapies. Despite the potential benefit and durable responses observed in some patients receiving CAR-T therapies, serious and potentially fatal toxicities remain a major challenge. The most common CAR-T-associated toxicities include cytokine release syndrome (CRS), neurotoxicity, cytopenias, and infections. While CRS and neurotoxicity are generally managed with tocilizumab and corticosteroids, respectively, high-grade toxicities can be life-threatening. Close postinfusion monitoring and assessment of clinical laboratory parameters, patient-related and clinical risk factors (e.g., age, tumor burden, comorbidities, baseline laboratory parameters, and underlying abnormalities), and therapy-related risk factors (e.g., CAR-T type, dose, and CAR-T-induced toxicity) are effective strategies to mitigate the toxicities. Clinical laboratory parameters, including various cytokines, have been identified for CRS (interleukin [IL]-1, IL-2, IL-5, IL-6, IL-8, IL-10, C-reactive protein [CRP], interferon [IFN]-γ, ferritin, granulocyte-macrophage colony-stimulating factor [GM-CSF], and monocyte chemoattractant protein-1), neurotoxicity (IL-1, IL-2, IL-6, IL-15, tumor necrosis factor [TNF]-α, GM-CSF, and IFN-γ), cytopenias (IL-2, IL-4, IL-6, IL-10, IFN-γ, ferritin, and CRP), and infections (IL-8, IL-1β, CRP, IFN-γ, and procalcitonin). CAR-T-associated toxicities can be monitored and treated to mitigate the risk to patients. Assessment of alterations in clinical laboratory parameter values that are correlated with CAR-T-associated toxicities may predict development and/or severity of a given toxicity, which can improve patient management strategies and ultimately enable the patients to better tolerate these therapies.
Collapse
Affiliation(s)
- Jonathan Renninger
- GSK Safety Evaluation and Risk Management, Global Safety, Philadelphia, PA, USA.
| | - Lisa Kurz
- GSK Safety Evaluation and Risk Management, Global Safety, Upper Providence, PA, USA
| | - Heather Stein
- GSK Safety Evaluation and Risk Management, Global Safety, Cambridge, MA, USA
| |
Collapse
|
48
|
Umyarova E, Pei C, Pellegrino W, Zhao Q, Sharma N, Benson D, Cottini F, Bezerra E, Brammer J, Bumma N, Choe H, Denlinger N, Devarakonda S, Khan A, Penza S, Rosko A, Vasu S, Wall S, Alinari L, Baiocchi R, Bond DA, Christian B, Hanel W, Maddocks K, Reneau J, Sawalha Y, Habib A, Sigmund A, Voorhees TJ, de Lima M, Epperla N. Second primary malignancies following CAR T-cell therapy in patients with hematologic malignancies. J Hematol Oncol 2025; 18:30. [PMID: 40098061 PMCID: PMC11916994 DOI: 10.1186/s13045-025-01676-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 02/16/2025] [Indexed: 03/19/2025] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has transformed the management of patients with relapsed/refractory (R/R) hematologic malignancies, including B-cell lymphomas and multiple myeloma (MM). While data pertaining to the efficacy and toxicity associated with CAR-T have been widely reported, there are limited data on long-term complications. We retrospectively analyzed 246 patients treated with CAR-T for R/R B-cell lymphoma (n = 228) and MM (n = 18) at Ohio State University from 2016 to 2022, with a minimum of two years of follow-up. The median age was 66 years, and the median number of prior treatments was four. With a median follow-up of 38 months (range 11-66), 21 patients (8.5%) developed a second primary malignancy (SPM), with non-melanoma skin cancer being the most common (52%), followed by hematologic malignancies (33%) and non-skin solid tumors (14%). Squamous cell carcinoma accounted for 38% of skin cancers, while myelodysplastic syndrome and acute myeloid leukemia were the predominant hematologic malignancies. Solid tumors included bladder, prostate, and breast cancer. The distinct pattern of SPMs suggests potential CAR-T-related risks, warranting vigilant post-treatment surveillance. Further studies are necessary to elucidate underlying mechanism and predictive factors and guide long-term management of SPM risk in CAR-T survivors.
Collapse
Affiliation(s)
- Elvira Umyarova
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| | - Charles Pei
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - William Pellegrino
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Qiuhong Zhao
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Nidhi Sharma
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Don Benson
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Francesca Cottini
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Evandro Bezerra
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Jonathan Brammer
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Naresh Bumma
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Hannah Choe
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Nathan Denlinger
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Srinivas Devarakonda
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Abdullah Khan
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Sam Penza
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Ashley Rosko
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Sumithira Vasu
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Sarah Wall
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Lapo Alinari
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Robert Baiocchi
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - David A Bond
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Beth Christian
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Walter Hanel
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Kami Maddocks
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - John Reneau
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Yazeed Sawalha
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Alma Habib
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Audrey Sigmund
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Timothy J Voorhees
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Marcos de Lima
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Narendranath Epperla
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
- Division of Hematology and Hematologic Malignancies, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84103, USA.
| |
Collapse
|
49
|
Beck M, Blumenberg V, Bücklein VL, Bundschuh RA, Harrer DC, Hirschbühl K, Jung J, Kunz WG, Menhart K, Winkelmann M, Yakushev I, Illert AL, Eckstein M, Völkl S, Claus R, Hansmann L, Hecker JS, Kuwert T, Mackensen A, Subklewe M, Hellwig D, Müller F. Liver-FDG-uptake augments early PET/CT prognostic value for CD19-targeted CAR-T cell therapy in diffuse large B cell lymphoma. EJNMMI Res 2025; 15:25. [PMID: 40095158 PMCID: PMC11914545 DOI: 10.1186/s13550-025-01201-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/19/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Despite revolutionary efficacy of CD19-CAR-T cell therapy (CAR-T) in aggressive B cell lymphoma, many patients still relapse mostly early. In early failure, distinct drugs support CAR-T which makes reliable and early prediction of imminent relapse/refractoriness critical. A complete metabolic remission (CR) on Fluor-18-Deoxyglucose (FDG) Positron-Emission-Computed Tomography (PET) 30 days after CAR-T (PET30) strongly predicts progression-free survival (PFS), but still fails in a relevant proportion of patients. We aimed to identify additional routine parameters in PET evaluation to enhance CAR-T response prediction. RESULTS Thirty patients with aggressive B cell lymphoma treated with CAR-T were retrospectively analyzed. Pre-CAR-T, LDH was the strongest PFS-predictor also by multivariate analysis. Post-CAR-T, 10 out of 14 patients (71.4%) with PET30-CR remained in disease remission, while 12 out of 16 patients (75%) with incomplete metabolic remission (PET30-nCR) relapsed after CAR-T. 28.6% of patients with PET30-CR ultimately progressed. Change of liver FDG-uptake from baseline to day30 (Delta-Liver-SUVmean) was identified as an independent biomarker for response. PET30-nCR and a decrease of Delta-Liver-SUVmean were associated with a high risk of tumor progression (HR 4.79 and 3.99, respectively). The combination of PET30 and Delta-Liver-SUVmean identified patients at very low, at intermediate and at very high risk of relapse (PFS not reached, 7.5 months, 1.5 months, respectively). CONCLUSION Additionally to PET30 metabolic remission, longitudinal metabolic changes in Delta-Liver-SUVmean predicted CAR-T efficiency. Our results may guide early intervention studies aiming to enhance CAR-T particularly in the very high-risk patients.
Collapse
Affiliation(s)
- Michael Beck
- Department of Nuclear Medicine, University Hospital of Erlangen, Friedrich-Alexander-Universität- Erlangen Nürnberg, Erlangen, Germany.
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany.
| | - Viktoria Blumenberg
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Veit L Bücklein
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, LMU Munich, Munich, Germany
| | - Ralph A Bundschuh
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Nuclear Medicine, Faculty of Medicine, University Hospital of Augsburg, Augsburg, Germany
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus at the TU Dresden, Dresden, Germany
| | - Dennis C Harrer
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Internal Medicine III, Hematology and Medical Oncology, University Hospital of Regensburg, Regensburg, Germany
| | - Klaus Hirschbühl
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Hematology and Oncology, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Johannes Jung
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Medicine III, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Wolfgang G Kunz
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Karin Menhart
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Nuclear Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Michael Winkelmann
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Igor Yakushev
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Nuclear Medicine, School of Medicine, TUM University Hospital, Technical University of Munich, Munich, Germany
| | - Anna Lena Illert
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Medicine III, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Markus Eckstein
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Pathology, University Hospital of Erlangen, Friedrich-Alexander-Universität- Erlangen Nürnberg, Erlangen, Germany
| | - Simon Völkl
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität-Erlangen Nürnberg, Erlangen, Germany
| | - Rainer Claus
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Hematology and Oncology, Medical Faculty, University of Augsburg, Augsburg, Germany
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Leo Hansmann
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Internal Medicine III, Hematology and Medical Oncology, University Hospital of Regensburg, Regensburg, Germany
| | - Judith S Hecker
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Medicine III, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
- Center for Translational Cancer Research, Technical University of Munich (TUM), TranslaTUM, Munich, Germany
| | - Torsten Kuwert
- Department of Nuclear Medicine, University Hospital of Erlangen, Friedrich-Alexander-Universität- Erlangen Nürnberg, Erlangen, Germany
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
| | - Andreas Mackensen
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität-Erlangen Nürnberg, Erlangen, Germany
| | - Marion Subklewe
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Dirk Hellwig
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany
- Department of Nuclear Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Fabian Müller
- Bavarian Cancer Research Center, Resp. Site (Augsburg, LMU Munich, TUM Munich, Erlangen, Regensburg), Germany.
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität-Erlangen Nürnberg, Erlangen, Germany.
| |
Collapse
|
50
|
Lahouty M, Soleymanzadeh A, Kazemi S, Saadati-Maleki H, Masoudi S, Ghasemi A, Kazemi T, Mehranfar S, Fadaee M. Cell-based immunotherapy in oesophageal cancer. J Drug Target 2025:1-11. [PMID: 40063049 DOI: 10.1080/1061186x.2025.2477077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 03/18/2025]
Abstract
Oesophageal cancer (EC) is among the most common illnesses globally, and its prognosis is unfavourable. Surgery, radiotherapy and chemotherapy are the primary therapy options for EC. Despite the occasional efficacy of these traditional treatment modalities, individuals with EC remain at a significant risk for local recurrence and metastasis. Consequently, innovative and efficacious medicines are required. In recent decades, clinical practice has effectively implemented cell therapy, which includes both stem cell and non-stem cell-based approaches, as an innovative tumour treatment, offering renewed hope to patients with oesophageal squamous cell carcinoma (ESCC). This paper examines the theoretical framework and contemporary advancements in cell treatment for individuals with EC. We further described current clinical studies and summarised essential data related to survival and safety assessments.
Collapse
Affiliation(s)
- Masoud Lahouty
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Sama Kazemi
- Faculty of Medicine, Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Haniyeh Saadati-Maleki
- Faculty of Medicine, Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Sanaz Masoudi
- Faculty of Medicine, Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Arash Ghasemi
- Faculty of Medicine, Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Mehranfar
- Department of Genetics and Immunology, Urmia University of Medical Sciences, Urmia, Iran
| | - Manouchehr Fadaee
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|