1
|
Yao X, Wang H, Lei X, Yao S, Wang W, Yang J. Pre-infusion 18F-FDG PET/CT for Prognostic and Toxicity Prediction in B-cell Non-Hodgkin Lymphoma Patients Undergoing Chimeric Antigen Receptor T-cell Therapy. Clin Nucl Med 2025:00003072-990000000-01639. [PMID: 40197422 DOI: 10.1097/rlu.0000000000005888] [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: 12/01/2024] [Accepted: 03/05/2025] [Indexed: 04/10/2025]
Abstract
PURPOSE The aim of this study was to evaluate the value of 18F-FDG PET/CT in predicting outcomes and toxicity for patients with B-cell non-Hodgkin lymphoma (B-NHL) who underwent chimeric antigen receptor T (CAR-T) cell therapy. METHODS This retrospective study included B-NHL patients who underwent CAR-T therapy and had pre-infusion 18F-FDG PET/CT images. We recorded SUVmax, metabolic tumor volume (MTV), total lesion glycolysis (TLG), and various clinical and laboratory indexes. The primary endpoints were progression-free survival (PFS) and overall survival (OS). PFS and OS were estimated using the Kaplan-Meier method. In addition, we reported the correlation between PET/CT parameters and the objective response (OR), as well as cytokine release syndrome (CRS). RESULTS A total of 133 patients were enrolled in this study. The median follow-up duration was 20.8 months. SUVmax (with a cutoff value of 15.65) emerged as an independent metabolic parameter associated with PFS, OS, and OR. Patients with SUVmax ≥15.65 had a median PFS of 9.13 months (95% CI: 0.11-18.16), while the PFS for those with SUVmax<15.65 was not reached (P=0.006). Furthermore, patients with SUVmax ≥15.65 exhibited significantly shorter average OS compared with those with SUVmax<15.65 (26.89 mo vs. 45.14 mo, P=0.010). In addition, the odds ratio for achieving an OR in patients with SUVmax ≥15.65 was found to be lower at 0.173 (95% CI: 0.056-0.539). Other factors associated with PFS included ECOG-PS, B symptoms, bulky mass, and extranodal sites, whereas IPI and LDH were associated with OS. Furthermore, SUVmax and Deauville scores showed a weak positive correlation with the occurrence of CRS. CONCLUSIONS The pretreatment PET/CT parameter SUVmax appears to be a promising predictive factor for efficacy and prognosis, as well as being associated with the occurrence of CRS. Consequently, we can conclude that this metabolic parameter from pretreatment PET/CT scans may serve as a valuable tool in guiding patient selection for CAR-T therapy and predicting potential side effects.
Collapse
Affiliation(s)
- Xilan Yao
- Department of Nuclear Medicine, Beijing Friendship Hospital of Capital Medical University
| | - Hongrong Wang
- Department of Nuclear Medicine, Beijing Boren Hospital
| | - Xiao Lei
- Department of Nuclear Medicine, Beijing Boren Hospital
| | - Shuang Yao
- Department of Nuclear Medicine, Beijing Fengtai You'anmen Hospital, Beijing, China
| | - Wei Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital of Capital Medical University
| | - Jigang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital of Capital Medical University
| |
Collapse
|
2
|
Hu MM, Zhao Y, Zhang N, Gong FY, Zhang W, Dong CS, Dai JF, Wang J. Tumor Microenvironment: Obstacles and Opportunities for T Cell-Based Tumor Immunotherapies. Mol Cancer Res 2025; 23:277-287. [PMID: 39898773 DOI: 10.1158/1541-7786.mcr-24-0747] [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: 08/09/2024] [Revised: 11/20/2024] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
The complex composition and dynamic change of the tumor microenvironment (TME), mainly consisting of tumor cells, immune cells, stromal cells, and extracellular components, significantly impede the effector function of cytotoxic T lymphocytes (CTL), thus representing a major obstacle for tumor immunotherapies. In this review, we summarize and discuss the impacts and underlying mechanisms of major elements in the TME (different cell types, extracellular matrix, nutrients and metabolites, etc.) on the infiltration, survival, and effector functions of T cells, mainly CD8+ CTLs. Moreover, we also highlight recent advances that may potentiate endogenous antitumor immunity and improve the efficacy of T cell-based immunotherapies in patients with cancer by manipulating components inside/outside of the TME. A deeper understanding of the effects and action mechanisms of TME components on the tumor-eradicating ability of CTLs may pave the way for discovering new targets to augment endogenous antitumor immunity and for designing combinational therapeutic regimens to enhance the efficacy of tumor immunotherapies in the clinic.
Collapse
Affiliation(s)
- Miao-Miao Hu
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Ying Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Nan Zhang
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Fang-Yuan Gong
- Department of Immunology, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Wei Zhang
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Chun-Sheng Dong
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jian-Feng Dai
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jun Wang
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, China
| |
Collapse
|
3
|
Liu S, Guan T, Yang F, Zhang H, Yao Y, Huang J, Zhao P, Huang W, Wu T, Lin H, Fu T, Chen S, Dai P, Ding Z, Chen D, Li W, Zeng H, Zhang X. Targeting CD276: a promising strategy for CAR-NK cell immunotherapy in human oral tongue squamous cell carcinoma. BMC Cancer 2025; 25:593. [PMID: 40169956 PMCID: PMC11963283 DOI: 10.1186/s12885-025-13973-9] [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: 12/12/2024] [Accepted: 03/19/2025] [Indexed: 04/03/2025] Open
Abstract
Human oral tongue squamous cell carcinoma (OTSCC) is a prevalent form of head and neck squamous cell carcinoma (HNSCC), often presenting at an advanced stage with a grim prognosis. Traditional therapeutic approaches such as surgery, adjuvant radiotherapy, and chemoradiotherapy have shown limited efficacy in treating advanced OTSCC, underscoring the urgent need for innovative treatment strategies. Our bioinformatics analysis identified CD276 as a significant biomarker in OTSCC, with high protein expression levels correlating to a markedly reduced survival rate in late-stage patients. This discovery has led us to develop chimeric antigen receptor-natural killer (CAR-NK) cells derived from umbilical cord blood cells (UCBCs), specifically targeting CD276. Our aim is to investigate this novel therapeutic approach for its potential to combat OTSCC under pre-clinical conditions. Our in vitro and ex vivo studies have demonstrated that CD276-targeted CAR-NK cells exhibit remarkable efficacy in lysing OTSCC cell lines and primary cells, as well as in eliminating OTSCC organoids. These promising results underscore the pivotal role of CD276 in OTSCC pathogenesis and highlight the potential of CAR-NK cell therapy as a groundbreaking treatment option for advanced-stage OTSCC, offering new hope for translational medicine in the field of stomatology.
Collapse
Affiliation(s)
- Shiyuan Liu
- Department of Oral and Maxillofacial Surgery, Xuchang Central Hospital, Xuchang, 461000, China
| | - Tian Guan
- Department of Oral and Maxillofacial Surgery, Xuchang Central Hospital, Xuchang, 461000, China
| | - Futian Yang
- Department of Oral and Maxillofacial Surgery, Xuchang Central Hospital, Xuchang, 461000, China
| | - Honglei Zhang
- Department of Oral and Maxillofacial Surgery, Xuchang Central Hospital, Xuchang, 461000, China
| | - Yao Yao
- Guangdong Procapzoom Biosciences, Inc, Guangdong, Guangzhou, China
| | - Jiman Huang
- Guangdong Procapzoom Biosciences, Inc, Guangdong, Guangzhou, China
| | - Pengfei Zhao
- Department of Oral and Maxillofacial Surgery, Xuchang Central Hospital, Xuchang, 461000, China
| | - Wei Huang
- Guangdong Procapzoom Biosciences, Inc, Guangdong, Guangzhou, China
| | - Tailiang Wu
- Department of Oral and Maxillofacial Surgery, Xuchang Central Hospital, Xuchang, 461000, China
| | - Hui Lin
- Guangdong Procapzoom Biosciences, Inc, Guangdong, Guangzhou, China
| | - Ting Fu
- Guangdong Procapzoom Biosciences, Inc, Guangdong, Guangzhou, China
| | - Shaoyu Chen
- Guangdong Procapzoom Biosciences, Inc, Guangdong, Guangzhou, China
| | - Pengzhan Dai
- Department of Oral and Maxillofacial Surgery, Xuchang Central Hospital, Xuchang, 461000, China
| | - Zhechun Ding
- Guangdong Procapzoom Biosciences, Inc, Guangdong, Guangzhou, China
| | - Dongmei Chen
- Guangdong Procapzoom Biosciences, Inc, Guangdong, Guangzhou, China
| | - Wei Li
- Department of Oral and Maxillofacial Surgery, Xuchang Central Hospital, Xuchang, 461000, China.
- Oral Medicine Center, Xuchang Central Hospital, Henan, China.
| | - Haoyu Zeng
- Guangdong Procapzoom Biosciences, Inc, Guangdong, Guangzhou, China.
- Key Laboratory of Molecular Target & Clinical Pharmacology, State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangdong, Guangzhou, China.
- Department of Cancer Research, Guangdong Procapzoom Biosciences Inc, 11 Guangpuzhong Rd, Guangzhou, 510700, China.
| | - Xianjun Zhang
- Department of Oral and Maxillofacial Surgery, Xuchang Central Hospital, Xuchang, 461000, China.
- Oral Medicine Center, Xuchang Central Hospital, Henan, China.
- Oral Medicine Center, Xuchang Central Hospital, Xuchang, 461000, China.
| |
Collapse
|
4
|
DiAndreth B, Nesterenko PA, Winters AG, Flynn AD, Jette CA, Suryawanshi V, Shafaattalab S, Martire S, Daris M, Moore E, Elshimali R, Gill T, Riley TP, Miller S, Netirojjanakul C, Hamburger AE, Kamb A. Multi-targeted, NOT gated CAR-T cells as a strategy to protect normal lineages for blood cancer therapy. Front Immunol 2025; 16:1493329. [PMID: 40191207 PMCID: PMC11968376 DOI: 10.3389/fimmu.2025.1493329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 02/21/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Despite advances in treatment of blood cancers, several-including acute myeloid leukemia (AML)-continue to be recalcitrant. Cell therapies based on chimeric antigen receptors (CARs) have emerged as promising approaches for blood cancers. However, current CAR-T treatments suffer from on-target, off-tumor toxicity, because most familiar blood cancer targets are also expressed in normal lineages. In addition, they face the common problem of relapse due to target-antigen loss. Cell therapeutics engineered to integrate more than one signal, often called logic-gated cells, can in principle achieve greater selectivity for tumors. Methods We applied such a technology, a NOT gated system called Tmod™ that is being developed to treat solid-tumor patients, to the problem of therapeutic selectivity for blood cancer cells. Results Here we show that Tmod cells can be designed to target 2-4 antigens to provide different practical and conceptual options for a blood cancer therapy: (i) mono- and bispecific activating receptors that target CD33, a well-known AML antigen expressed on the majority of AML tumors (as well as healthy myeloid cells) and CD43 (SPN), an antigen expressed on many hematopoietic cancers (and normal blood lineages); and (ii) mono- and bispecific inhibitory receptors that target CD16b (FCGR3B) and CLEC9A, antigens expressed on key normal blood cells but not on most blood cancers. Discussion These results further demonstrate the robust modularity of the Tmod system and generalize the Tmod approach beyond solid tumors.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Animals
- Hematologic Neoplasms/therapy
- Hematologic Neoplasms/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- T-Lymphocytes/metabolism
- Mice
- Antigens, Neoplasm/immunology
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Cell Line, Tumor
- Cell Lineage
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Alexander Kamb
- A2 Biotherapeutics Discovery Research, Agoura Hills, CA, United States
| |
Collapse
|
5
|
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
|
6
|
Xu J, Zhang H, Zhao Y, Zhang X, Guo S, Shi X, Xiao X, Lyu H, Zhang Y, He X, Zhao M. Infectious complications distribution following CLL1 CAR-T cell therapy for acute myeloid leukemiass. Cancer Immunol Immunother 2025; 74:149. [PMID: 40088283 PMCID: PMC11910464 DOI: 10.1007/s00262-025-03998-1] [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: 04/06/2024] [Accepted: 02/24/2025] [Indexed: 03/17/2025]
Abstract
The CLL1-targeted chimeric antigen receptor T (CAR-T) cell therapy offers a novel therapeutic approach for refractory or relapsed acute myeloid leukemia (AML).The targeted elimination of tumor cells by CLL1 CAR-T therapy also induces cytotoxic effects on neutrophils, leading to a severe granulocytopenia, thereby significantly increasing the risk of infectious complications during CAR-T therapy. However, the infectious complications associated with this strategy have not been comprehensively investigated. The objective of this study was to evaluate the incidence rate of infectious complications within a 28-day period in a cohort of 51 patients who underwent CLL1 CAR-T cell infusion. Meanwhile, the univariate and multivariate analyses were employed to access the risk factors of infectious complications during CLL1 CAR-T therapy. The study observed a total of 46 infection events in 32 out of 51 patients (63%), with the median onset of infection occurring at 9 days following CAR-T cell infusion. The cumulative incidence of infection events within 28 days was 56.9% (95%CI: 50.4-61.3%), with bacterial and fungal infections being the most prevalent early infection events. The results of multivariate analysis revealed that a lower neutrophil counts prior to lymphodepletion chemotherapy (OR = 3.875, P = 0.041) and more severe complications of cytokine release syndrome (OR = 4.141, P = 0.037) were identified as independent risk factors associated with an increased likelihood of early infection events. This study examined the distribution of early infection events and identified potential risk factors, with the goal of offering guidance to physicians on implementing more effective intervention strategies to decrease treatment-related mortality rates and improve patient prognosis. This study has been registered in the Chinese Clinical Trial Registry (Trial registration number: ChiCTR2000041054).
Collapse
Affiliation(s)
- Jianmei Xu
- The First Central Clinical College of Tianjin Medical University, Tianjin, 300380, China
| | - Huan Zhang
- The First Central Clinical College of Tianjin Medical University, Tianjin, 300380, China
| | - Yifan Zhao
- The First Central Clinical College of Tianjin Medical University, Tianjin, 300380, China
| | - Xiaomei Zhang
- Nankai University School of Medicine, Tianjin, 300380, China
| | - Shujing Guo
- The First Central Clinical College of Tianjin Medical University, Tianjin, 300380, China
| | - Xiaoxue Shi
- The First Central Clinical College of Tianjin Medical University, Tianjin, 300380, China
| | - Xia Xiao
- Department of Hematology, Tianjin First Central Hospital, No.2 Baoshanxi Rd, Xiqing District,, 300380, Tianjin, China
| | - Hairong Lyu
- Department of Hematology, Tianjin First Central Hospital, No.2 Baoshanxi Rd, Xiqing District,, 300380, Tianjin, China
| | - Yu Zhang
- Department of Hematology, Tianjin First Central Hospital, No.2 Baoshanxi Rd, Xiqing District,, 300380, Tianjin, China
| | - Xiaoyuan He
- Department of Hematology, Tianjin First Central Hospital, No.2 Baoshanxi Rd, Xiqing District,, 300380, Tianjin, China
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, No.2 Baoshanxi Rd, Xiqing District,, 300380, Tianjin, China.
| |
Collapse
|
7
|
Peter J, Toppeta F, Trubert A, Danhof S, Hudecek M, Däullary T. Multi-Targeting CAR-T Cell Strategies to Overcome Immune Evasion in Lymphoid and Myeloid Malignancies. Oncol Res Treat 2025:1-15. [PMID: 40090318 DOI: 10.1159/000543806] [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: 10/02/2024] [Accepted: 01/15/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-T cell therapy has become a groundbreaking treatment for hematological malignancies, particularly lymphomas and multiple myeloma, with high remission rates in refractory and relapsed patients. However, most CAR-T therapies target a single antigen, such as CD19, which can result in immune evasion through antigen escape. This mechanism describes the downregulation or complete loss of the targeted antigen by the tumor cells, eventually leading to relapse. To address this issue, multi-targeting strategies like logic-gated CARs, adapter CARs, or combination therapies can increase the potency of CAR-T cells. These approaches aim to minimize immune evasion by targeting multiple antigens simultaneously, thereby increasing treatment durability. Additionally, advanced tools such as next-generation sequencing (NGS), direct stochastic optical reconstruction microscopy (dSTORM), or multiparametric flow cytometry are helping to identify novel tumor-specific targets and improve therapy designs. SUMMARY This review explores the current landscape of CAR-T cell therapies in lymphoid and myeloid malignancies, highlights ongoing clinical trials, and discusses the future of these innovative multi-targeting approaches to improve patient outcome. KEY MESSAGES Antigen escape limits CAR-T cell therapy success, but multi-targeting strategies like logic gates and adapter CARs offer solutions. Optimizing antigen selection and CAR design, along with larger clinical trials, is essential for improving patient outcomes. Personalization using advanced technologies like CRISPR screening and single-cell RNA sequencing can enhance durability and effectiveness of treatments for heavily pretreated patients.
Collapse
Affiliation(s)
- Jessica Peter
- Chair in Cellular Immunotherapy, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
- NCT WERA, National Center for Tumor Diseases (Würzburg, Erlangen, Regensburg and Augsburg), Würzburg, Germany
- BZKF, Bavarian Center for Cancer Research, Erlangen, Germany
| | - Fabio Toppeta
- Chair in Cellular Immunotherapy, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
- NCT WERA, National Center for Tumor Diseases (Würzburg, Erlangen, Regensburg and Augsburg), Würzburg, Germany
- BZKF, Bavarian Center for Cancer Research, Erlangen, Germany
| | - Alexandre Trubert
- Chair in Cellular Immunotherapy, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
- NCT WERA, National Center for Tumor Diseases (Würzburg, Erlangen, Regensburg and Augsburg), Würzburg, Germany
- BZKF, Bavarian Center for Cancer Research, Erlangen, Germany
| | - Sophia Danhof
- Chair in Cellular Immunotherapy, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
- NCT WERA, National Center for Tumor Diseases (Würzburg, Erlangen, Regensburg and Augsburg), Würzburg, Germany
- BZKF, Bavarian Center for Cancer Research, Erlangen, Germany
| | - Michael Hudecek
- Chair in Cellular Immunotherapy, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
- NCT WERA, National Center for Tumor Diseases (Würzburg, Erlangen, Regensburg and Augsburg), Würzburg, Germany
- BZKF, Bavarian Center for Cancer Research, Erlangen, Germany
| | - Thomas Däullary
- Chair in Cellular Immunotherapy, Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
- NCT WERA, National Center for Tumor Diseases (Würzburg, Erlangen, Regensburg and Augsburg), Würzburg, Germany
- BZKF, Bavarian Center for Cancer Research, Erlangen, Germany
| |
Collapse
|
8
|
Guo S, Xi X. Nanobody-enhanced chimeric antigen receptor T-cell therapy: overcoming barriers in solid tumors with VHH and VNAR-based constructs. Biomark Res 2025; 13:41. [PMID: 40069884 PMCID: PMC11899093 DOI: 10.1186/s40364-025-00755-5] [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: 12/26/2024] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
CAR-T cells are genetically modified T lymphocytes that express chimeric antigen receptors (CAR) on their surfaces. These receptors enable T lymphocytes to recognize specific antigens on target cells, triggering a response that leads to targeted cytotoxicity. While CAR-T therapy has effectively treated various blood cancers, it faces significant challenges in addressing solid tumors. These challenges include identifying precise tumor antigens, overcoming antigen evasion, and enhancing the function of CAR-T cells within the tumor microenvironment. Single domain antibody, versatile tools with low immunogenicity, high stability, and strong affinity, show promise for improving the efficacy of CAR-T cells against solid tumors. By addressing these challenges, single domain antibody has the potential to overcome the limitations associated with ScFv antibody-based CAR-T therapies. This review highlights the benefits of utilizing single domain antibody in CAR-T therapy, particularly in targeting tumor antigens, and explores development strategies that could advance the field.
Collapse
Affiliation(s)
- Shasha Guo
- Shandong Women's University, Jinan, 250300, People's Republic of China.
| | - Xiaozhi Xi
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, People's Republic of China.
- Oncology Department, Shandong Second Provincial General Hospital, Jinan, 250023, People's Republic of China.
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ministry of Education, Ocean University of China, Qingdao, 266003, People's Republic of China.
| |
Collapse
|
9
|
Cai Z, Zou D, Ma Q, Sun W, Guo Y. Combined autologous hematopoietic stem cell transplantation and CD19 CAR T-cell therapy for relapsed/refractory diffuse large B-cell lymphoma with TP53 mutation: A case report. SAGE Open Med Case Rep 2025; 13:2050313X241306236. [PMID: 40078172 PMCID: PMC11898238 DOI: 10.1177/2050313x241306236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/25/2024] [Indexed: 03/14/2025] Open
Abstract
Despite advancements in the treatment of diffuse large B-cell lymphoma, including CAR T-cell therapy, TP53 mutations remain a significant negative prognostic factor in patients with relapsed/refractory diffuse large B-cell lymphoma. The combination of autologous stem cell transplantation and CAR T-cell therapy may enhance long-term prognosis and reduce adverse effects, including severe cytokine release syndrome. This case report presents a 41-year-old man with relapsed/refractory diffuse large B-cell lymphoma harboring TP53 mutations who underwent autologous stem cell transplantation combined with CD19 CAR T-cell therapy. Two years posttreatment, the patient remains in sustained complete remission, highlighting the potential efficacy of this combination approach for relapsed/refractory diffuse large B-cell lymphoma with TP53 mutation.
Collapse
Affiliation(s)
- Ziyi Cai
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing, P. R. China
| | - Dongmei Zou
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing, P. R. China
| | - Qiang Ma
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing, P. R. China
| | - Wanling Sun
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing, P. R. China
| | - Yixian Guo
- Department of Hematology, Xuanwu Hospital, Capital Medical University, Beijing, P. R. China
| |
Collapse
|
10
|
Ji X, Sun Y, Xie Y, Gao J, Zhang J. Advance in chimeric antigen receptor T therapy in autoimmune diseases. Front Immunol 2025; 16:1533254. [PMID: 40103816 PMCID: PMC11913860 DOI: 10.3389/fimmu.2025.1533254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
Autoimmune diseases are a group of diseases in which the body's immune system misrecognizes its own antigens resulting in an abnormal immune response, which can lead to pathological damage to or abnormal functioning of its own tissues. Current treatments are mainly hormones and broad-spectrum immunosuppressants, but these can lead to a decline in the patient's immunity. Chimeric antigen receptor T (CAR-T) Cell therapy has emerged, and now the structure of CAR has changed from first generation to fourth generation of CAR. The significant achievement of CAR-T therapy to B-cell leukemia has also inspired the treatment of autoimmune diseases, and by investigating the mechanisms of different autoimmune diseases, different designs of CAR-T can be used to specifically treat autoimmune diseases. In this review, we will discuss the therapeutic strategies of CAR-T cells in different autoimmune diseases and the limitations of the treatment.
Collapse
Affiliation(s)
- Xiaolan Ji
- Department of Ophthalmology, The Second Affiliated Hospital of Suzhou University, Suzhou, China
| | - Yunfan Sun
- The First Clinical College of Nanjing Medical University, Nanjing, China
| | - Yuyang Xie
- The First Clinical Medicine School, Suzhou Medical College, Soochow University, Suzhou, China
| | - Jianling Gao
- Department of Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Ji Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Suzhou University, Suzhou, China
| |
Collapse
|
11
|
Almawash S. Revolutionary Cancer Therapy for Personalization and Improved Efficacy: Strategies to Overcome Resistance to Immune Checkpoint Inhibitor Therapy. Cancers (Basel) 2025; 17:880. [PMID: 40075727 PMCID: PMC11899125 DOI: 10.3390/cancers17050880] [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: 01/06/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a significant public health issue worldwide, standing as a primary contributor to global mortality, accounting for approximately 10 million fatalities in 2020 [...].
Collapse
Affiliation(s)
- Saud Almawash
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
12
|
Song Z, Tao Y, You J. The potential applications of peptide-loading complex in cancer treatment. Front Immunol 2025; 16:1526137. [PMID: 40098955 PMCID: PMC11911339 DOI: 10.3389/fimmu.2025.1526137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/05/2025] [Indexed: 03/19/2025] Open
Abstract
Immunotherapy for cancer has made significant strides in the last several years. The prognosis for cancer patients has significantly improved as a result, particularly in hematological diseases. However, it was discovered that translating these achievements to solid tumors proved challenging. The peptide-loading complex (PLC), a temporary multisubunit membrane assembly in the endoplasmic reticulum (ER), is crucial for initiating a hierarchical immune response. Chaperones calreticulin and tapasin make up the PLC, unique to class I glycoproteins, thiooxido-reductase ERp57, and a transporter associated with antigen processing. The loading and editing of major histocompatibility complex class I (MHC-I) molecules with peptide translocation into the ER are synchronized by the PLC. One of the immune escape strategies revealed for tumors so far is changes in the expression of MHC molecules. This is because MHC antigens are crucial in presenting antigens to T-lymphocytes and controlling NK cell activity. Furthermore, decreased MHC-I expression has been linked to malignancies resistant to T-cell-based cancer immunotherapies (adoptive transfer of antitumor CD8 T-cells or checkpoint inhibition). The PLC is essential for T-cell priming, differentiation, and tumor growth control because it can bind to a wide range of MHC-I allomorphs. In this review, we have looked into PLC's function and effects in all forms of cancer to improve cancer therapy techniques.
Collapse
Affiliation(s)
- Zhidu Song
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Ying Tao
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jiaxin You
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
Lv P, Zhang Y, Wu W, Jiang X, Xiang W. Pan-cancer analysis identifies ADAM12 as a prognostic biomarker and indicator of immune infiltration in glioma. Sci Rep 2025; 15:6314. [PMID: 39984619 PMCID: PMC11845722 DOI: 10.1038/s41598-025-90121-0] [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: 11/20/2024] [Accepted: 02/11/2025] [Indexed: 02/23/2025] Open
Abstract
ADAM12, part of the adisintegrin and metalloproteases (ADAMs) family, has been widely reported in recent years to be associated with various malignant tumor behaviors, including migration, invasion, and treatment resistance. However, its role at the pan-cancer level remains insufficiently characterized. In this study, pan-cancer data were utilized to elucidate the expression patterns, prognostic significance, and potential roles of ADAM12 within the tumor immune microenvironment. An in-depth analysis of ADAM12 in gliomas was also conducted. Our findings revealed that ADAM12 expression is markedly overexpressed in glioma tissues, enhances glioma cell malignancy, and is associated with a worse prognosis. These results suggest that ADAM12 may serve as a biomarker for predicting glioma malignancy and patient prognosis, as well as a potential therapeutic target in glioma treatment.
Collapse
Affiliation(s)
- Peng Lv
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue NO.1277, Wuhan, 430022, Hubei Province, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yanbin Zhang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue NO.1277, Wuhan, 430022, Hubei Province, China
| | - Wenjie Wu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue NO.1277, Wuhan, 430022, Hubei Province, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue NO.1277, Wuhan, 430022, Hubei Province, China.
| | - Wei Xiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue NO.1277, Wuhan, 430022, Hubei Province, China.
| |
Collapse
|
14
|
Feigelson SW, Dadosh T, Levi N, Sapoznikov A, Weinstein-Marom H, Blokon-Kogan D, Avraham Y, Unger T, Gross G, Dahan R, Alon R. CD32B1, a versatile non-signaling antibody-binding scaffold for enhanced T cell adhesion to tumor stromal cognate antigens. Front Immunol 2025; 16:1398757. [PMID: 39995660 PMCID: PMC11847833 DOI: 10.3389/fimmu.2025.1398757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 01/15/2025] [Indexed: 02/26/2025] Open
Abstract
Targeting cytotoxic T lymphocytes (CTLs), as chimeric antigen T cells (CAR-T), T cell receptor-engineered (TCR)-T cells or adoptive cell transfer of tumor infiltrating T cells (TILs) to solid tumors is a major therapeutic challenge. We describe a new strategy to confer these lymphocytes with de novo adhesiveness to surface proteins enriched in the tumor microenvironment. This approach is based on decorating CTLs with monoclonal antibodies (mAbs) specific to any surface protein of interest within the stroma and the extracelullar matrix of solid tumors. For efficient mAb decoration, we have introduced a mAb binding Fc receptor (FcR) scaffold, FcγRIIB1 (CD32B1), which we found to be enriched on B lymphocyte microvilli (MV). This isoform contains an inhibitory ITIM motif within a cytoplasmic tail anchored to the cortical cytoskeleton. We thus generated a non-signaling CD32B1 mutant lacking the ITIM motif (termed ITIM-less CD32B1, or ILCD32B1) and successfully expressed it in human T cells which normally do not express this FcR. The ILCD32B1 expressing lymphocytes bound multiple IgG1 mAbs whose Fc domain was engineered with a 5-residue substitution to reach a nM range of Fc-FcγCR dissociation constants. The mAb decorated ILCD32B1 expressing T cells could readily adhere to a surface-bound cognate antigen. To broaden the utility of this scaffold, we have also generated a new fusion protein in which the entire Fc binding domain was truncated (tILCD32B1) and replaced with a monomeric streptavidin variant, mSA2, via a CD8 hinge. The molecule, termed mSA2-CD8h-tILCD32B1, was also successfully expressed in T cells, readily and stably bound biotinylated IgG mAbs in vitro and once decorated with the biotin labeled mAbs, conferred the T cells with high adhesiveness to multiple surface-coated antigens. mSA2-CD8h-tILCD32B1 expressing human T cells decorated ex vivo with a biotin-labeled mAb retained the antibody for hours after accumulation inside breast tumors implanted in immunodeficient recipient mice. Our results collectively suggest that a non-signaling CD32B1 can be used as a versatile scaffold for mAb decoration of T cells. Our mAb decoration approach can confer new cell adhesive reactivities to improve tumor CTL (CAR-T and TIL) accumulation and retention inside solid tumors.
Collapse
Affiliation(s)
- Sara W. Feigelson
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Tali Dadosh
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Nehora Levi
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Anita Sapoznikov
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hadas Weinstein-Marom
- Laboratory of Immunology, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel
- Department of Biotechnology, Tel-Hai College, Upper Galilee, Israel
| | - Dayana Blokon-Kogan
- Laboratory of Immunology, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel
- Department of Biotechnology, Tel-Hai College, Upper Galilee, Israel
| | - Yahel Avraham
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Tamar Unger
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Gideon Gross
- Laboratory of Immunology, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel
- Department of Biotechnology, Tel-Hai College, Upper Galilee, Israel
| | - Rony Dahan
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Ronen Alon
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
15
|
Wang J, Wang H, Ding Y, Cao N, Nan F, Wu F, Li C, Liang X, Xiao M, Guo J, Gao Z, Yan L, Zhou T, Li Y, Zhai Z. Gp350-targeted CAR-T therapy in EBV-positive Burkitt lymphoma: pre-clinical development of gp350 CAR-T. J Transl Med 2025; 23:171. [PMID: 39930509 PMCID: PMC11809011 DOI: 10.1186/s12967-025-06188-w] [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: 12/11/2024] [Accepted: 01/28/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Epstein-Barr virus (EBV) is an oncovirus belonging to the herpesvirus family, associated with the pathogenesis of multiple malignancies, particularly Burkitt lymphoma (BL). The virus remains latent in host cells and plays a critical role in tumor progression through various mechanisms. A key glycoprotein, gp350, expressed during the lytic phase of EBV, is instrumental in viral entry into B cells and presents a unique antigenic target, making it a promising candidate for immunotherapeutic approaches, such as chimeric antigen receptor T-cell (CAR-T) therapy. METHODS In this study, we engineered CAR-T cells targeted against the gp350 glycoprotein and assessed their therapeutic potential through a series of in vitro and in vivo experiments. The efficacy of the gp350-CAR-T cells was evaluated by comparing their cytotoxic effects against both EBV-positive and -negative tumor cell lines. We utilized a xenograft model of Burkitt lymphoma to monitor the impact of gp350-CAR-T cell administration on tumor progression and overall survival. RESULTS The engineered gp350-CAR-T cells demonstrated potent cytotoxicity specifically against EBV-positive tumor cell lines. In our in vivo xenograft model, administration of gp350-CAR-T cells resulted in significant inhibition of tumor growth, highlighting their capability to effectively target and eliminate EBV-positive lymphomas. This selectivity underscores the potential of utilizing gp350 as a specific target for immunotherapy. CONCLUSION Our findings advocate for the clinical application of gp350-directed CAR-T therapy as a prospective treatment strategy for patients with relapsed or refractory EBV-positive tumors. Given the encouraging preclinical results, further research is warranted to optimize CAR-T cell production processes and extend the potential of this therapy to other EBV-associated malignancies, paving the way for improved outcomes in affected patient populations.
Collapse
Affiliation(s)
- Jiajia Wang
- Department of Hematology/Hematologic Diseases Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
- Department of Hematology, Tongling People's Hospital, Tongling, 244000, Anhui, China
| | - Huiping Wang
- Department of Hematology/Hematologic Diseases Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Yangyang Ding
- Department of Hematology/Hematologic Diseases Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Nengneng Cao
- Department of Hematology/Hematologic Diseases Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Fengya Nan
- Department of Pathology, Department of Pathology, Anhui Medical University, The First Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Fan Wu
- Department of Hematology/Hematologic Diseases Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Cong Li
- Department of Hematology/Hematologic Diseases Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Xue Liang
- Department of Hematology/Hematologic Diseases Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Meng Xiao
- Department of Hematology/Hematologic Diseases Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
- Department of Hematology, Jining NO. 1 People's Hospital, Jining, 272000, Shandong, China
| | - Jinjing Guo
- Department of Hematology/Hematologic Diseases Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
- Department of Laboratory, Fuyang People's Hospital, Fuyang, 236000, Anhui, China
| | - Zhimai Gao
- ZENO Biotechnology (Shenzhen) Co, Shenzhen, 518000, Guangdong, China
| | - Li Yan
- Department of Hematology/Hematologic Diseases Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Tielin Zhou
- Zeno Therapeutics Pte. Ltd., 600 North Bridge Road, Singapore, 188778, Singapore
- Eximmium Pte. Ltd., 600 North Bridge Road, Singapore, 188778, Singapore
| | - Yanli Li
- Department of Hematology/Hematologic Diseases Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China.
- Department of Pathology, Department of Pathology, Anhui Medical University, The First Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China.
| | - Zhimin Zhai
- Department of Hematology/Hematologic Diseases Research Center, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China.
| |
Collapse
|
16
|
Sabir S, León-Triana O, Serrano S, Barrio R, Pérez-García VM. Mathematical Model of CAR T-Cell Therapy for a B-Cell Lymphoma Lymph Node. Bull Math Biol 2025; 87:40. [PMID: 39918662 PMCID: PMC11805830 DOI: 10.1007/s11538-025-01417-1] [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: 07/31/2024] [Accepted: 01/16/2025] [Indexed: 02/09/2025]
Abstract
CAR T-cell therapies have demonstrated significant success in treating B-cell leukemia in children and young adults. However, their effectiveness in treating B-cell lymphomas has been limited in comparison to leukemia. In this paper we present a mathematical model that elucidates the dynamics of diffuse large B-cell lymphoma and CAR T-cells in a lymph node. The mathematical model aids in understanding the complex interplay between the cell populations involved and proposes ways to identify potential underlying dynamical causes of treatment failure. We also study the phenomenon of immunosuppression induced by tumor cells and theoretically demonstrate its impact on cell dynamics. Through the examination of various response scenarios, we underscore the significance of product characteristics in treatment outcomes.
Collapse
Affiliation(s)
- Soukaina Sabir
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), Universidad de Castilla-La Mancha, Ciudad Real, Spain.
| | - Odelaisy León-Triana
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), Universidad de Castilla-La Mancha, Ciudad Real, Spain
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, Hospital La Paz Institute for Health Research-IdiPAZ, Madrid, Spain
| | - Sergio Serrano
- Department of Applied Mathematics, Computational Dynamics Group (CoDy), Universidad de Zaragoza, Zaragoza, Spain
| | - Roberto Barrio
- Department of Applied Mathematics, Computational Dynamics Group (CoDy), Universidad de Zaragoza, Zaragoza, Spain
| | - Victor M Pérez-García
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), Universidad de Castilla-La Mancha, Ciudad Real, Spain
| |
Collapse
|
17
|
Sun L, Huang J, Wang X, Huang P, Dong B, Liang Z, Wu J, Wang J. Enucleated bone marrow-derived mesenchymal stromal cells regulate immune microenvironment and promote testosterone production through efferocytosis. Reprod Biol Endocrinol 2025; 23:21. [PMID: 39915874 PMCID: PMC11800438 DOI: 10.1186/s12958-025-01352-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 01/27/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND Testosterone deficiency (TD) occurs most frequently in older men and can cause many health problems. Testosterone replacement therapy (TRT) is widely used to treat TD, but this regimen can lead to a series of side effects. Stem cell therapy has been wildly studied in vitro. However, due to the multidirectional differentiation potential and heterogeneity of stem cells, it is difficult to achieve the good efficiency and reproducibility in basic research and clinical applications. This study aims to identify a new strategy for the treatment of TD. METHODS Bone marrow-derived mesenchymal stromal cells (BMSCs) were enucleated by Ficoll density gradient centrifugation. The organelles and cellular functions of enucleated BMSCs were analyzed by immunofluorescence staining and flow cytometry. Extracellular vesicles (EVs) were isolated by ultracentrifugation and characterized. For the animal studies, enucleated BMSCs were labelled with Mitotracker and injected into ethane dimethanesulfone (EDS)-treated rats. Testosterone production and spermatogenesis were detected at different time points through various tests. To determine the mechanism of efferocytosis, we analysed the number of macrophages by immunofluorescence staining and quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS The injection of enucleated BMSCs (Cargocytes) into the testes of EDS-treated rats restored the levels of serum testosterone, increased the number of Leydig cells (LCs), and improved spermatogenesis. We found that enucleated BMSCs underwent apoptosis earlier than BMSCs did. Subsequently, testicular interstitial macrophages phagocytosed apoptotic enucleated BMSCs through efferocytosis. Efferocytosis promoted macrophage polarization from the M1 to the M2 phenotype, reduced the expression of proinflammatory cytokines, and decreased the levels of inflammation and oxidative stress. CONCLUSIONS In summary, this study pioneered the application of stromal cell enucleation technology to repair tissue damage in the reproductive system, explored the potential of cell burial in the treatment of reproductive system diseases and provided a new approach for the clinical treatment of male infertility.
Collapse
Affiliation(s)
- Lu Sun
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Jiayu Huang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
- Department of Urology, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
| | - Xuezi Wang
- School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Peng Huang
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Baolin Dong
- School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Zehang Liang
- School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Jiahong Wu
- School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Jiancheng Wang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
18
|
Liu Z, Shi Z, Jiang W, Shen Z, Chen W, Shen K, Sun Y, Tang Z, Wang X. Circulating tumor DNA analysis for prediction of prognosis and molecular insights in patients with resectable gastric cancer: results from a prospective study. MedComm (Beijing) 2025; 6:e70065. [PMID: 39830022 PMCID: PMC11742430 DOI: 10.1002/mco2.70065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 12/03/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
This study aimed to evaluate the prognostic value of plasma circulating tumor DNA (ctDNA) level in patients with resectable gastric cancer (GC). A total of 59 patients were prospectively enrolled, with their ctDNA detected and paired tumor tissue collected at various peri-operative time points. Patients with higher 1-month post-operative ctDNA levels demonstrated shorter overall survival status (hazard ratio [HR] = 5.30, p = 0.0022) and a higher risk of recurrence (HR = 3.85, p = 0.011). The model combining ctDNA with conventional serum tumor markers for GC, including carcinoembryonic antigen, carbohydrate antigen 19-9, and CA72-4, shows high predictive effectiveness for GC prognosis with an area under the curve of 0.940 (p = 0.002), which is higher than net ctDNA and other models without ctDNA. Patients with lower ctDNA levels were more likely to have positive stromal programmed cell death ligand 1 expression (p = 0.046). Additionally, DCAF4L2 mutation was identified as the crucial gene mutation in ctDNA suggesting poor prognosis of patients with GC. Overall, this study highlights that post-operative ctDNA can serve as an effective biomarker for prognostic prediction and recurrence surveillance in resectable GC.
Collapse
Affiliation(s)
- Zheng Liu
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Zhongyi Shi
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Wenchao Jiang
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Zhenbin Shen
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Weidong Chen
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Kuntang Shen
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Yihong Sun
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
- Department of General SurgeryZhongshan Hospital (Xiamen Branch)Fudan UniversityShanghaiChina
| | - Zhaoqing Tang
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
- Department of General SurgeryZhongshan Hospital (Xiamen Branch)Fudan UniversityShanghaiChina
| | - Xuefei Wang
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
- Department of General SurgeryZhongshan Hospital (Xiamen Branch)Fudan UniversityShanghaiChina
| |
Collapse
|
19
|
Park H, Mugundu GM, Singh AP. Mechanistic Evaluation of Anti-CD19 CAR-T Cell Therapy Repurposed in Systemic Lupus Erythematosus Using a Quantitative Systems Pharmacology Model. Clin Transl Sci 2025; 18:e70146. [PMID: 39936636 DOI: 10.1111/cts.70146] [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: 10/24/2024] [Revised: 01/05/2025] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
CAR-T cell therapy, renowned for its success in oncology, is now venturing into the realm of B cell-mediated autoimmune diseases. Recent observations have revealed significant pharmacological effects of CD19 CAR-T cells in patients with systemic lupus erythematosus (SLE), suggesting promising applications in other autoimmune conditions. Consequently, as of December 2024, there are 116 different clinical trials evaluating CAR-T cells against autoimmune conditions. While the field is starting to understand the overall pharmacological actions of CAR-T cells in autoimmune diseases, the dose-exposure-response relationship remains inadequately characterized due to limited clinical data. To address these uncertainties, we have developed a Quantitative Systems Pharmacology (QSP) model using short-term limited clinical data of anti-CD19 CAR-Ts in autoimmune disease patients (n = 5), followed by a model qualification step utilizing an external dataset (n = 13). The developed QSP model integrated and effectively characterized the (1) cellular kinetics of different immunophenotypic population of CAR-T cells, (2) impact of lymphodepletion chemotherapy on host immune cells, (3) CAR-mediated elimination of CD19+ B-cells and (4) dynamic changes in disease surrogate biomarkers and its relationship with clinical score. The key pharmacological biomarkers which were incorporated within the QSP model included anti double stranded DNA (anti-dsDNA) antibodies, proteinuria, C3 protein and IFN-alpha. Later, a linear regression analysis-based relationship was developed between continuous disease biomarkers and the categorical SLE disease activity index (SLE-DAI) determined by the investigators offering a predictive framework for disease progression in SLE patients. This proposed QSP model holds potential to elucidate quantitative pharmacology and expedite clinical advancement of autologous and allogeneic cell therapies in autoimmune diseases.
Collapse
Affiliation(s)
- Hyunseo Park
- Cell Therapy Clinical Pharmacology and Modeling, Precision and Translational Medicine Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Sciences Center, Memphis, Tennessee, USA
| | - Ganesh M Mugundu
- Cell Therapy Clinical Pharmacology and Modeling, Precision and Translational Medicine Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Aman P Singh
- Cell Therapy Clinical Pharmacology and Modeling, Precision and Translational Medicine Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| |
Collapse
|
20
|
Panuccio G, Correale P, d'Apolito M, Mutti L, Giannicola R, Pirtoli L, Giordano A, Labate D, Macheda S, Carabetta N, Abdelwahed YS, Landmesser U, Tassone P, Tagliaferri P, De Rosa S, Torella D. Immuno-related cardio-vascular adverse events associated with immuno-oncological treatments: an under-estimated threat for cancer patients. Basic Res Cardiol 2025; 120:153-169. [PMID: 39225869 PMCID: PMC11790807 DOI: 10.1007/s00395-024-01077-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/16/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
Immunotherapy represents an emergent and heterogeneous group of anticancer treatments harnessing the human immune-surveillance system, including immune-checkpoint inhibitor monoclonal antibodies (mAbs), Chimeric Antigen Receptor T Cells (CAR-T) therapy, cancer vaccines and lymphocyte activation gene-3 (LAG-3) therapy. While remarkably effective against several malignancies, these therapies, often in combination with other cancer treatments, have showed unforeseen toxicity, including cardiovascular complications. The occurrence of immuno-mediated adverse (irAEs) events has been progressively reported in the last 10 years. These irAEs present an extended range of severity, from self-limiting to life-threatening conditions. Although recent guidelines in CardioOncology have provided important evidence in managing cancer treatments, they often encompass general approaches. However, a specific focus is required due to the particular etiology, unique risk factors, and associated side effects of immunotherapy. This review aims to deepen the understanding of the prevalence and nature of cardiovascular issues in patients undergoing immunotherapy, offering insights into strategies for risk stratification and management.
Collapse
Affiliation(s)
- Giuseppe Panuccio
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200, Berlin, Germany.
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| | - Pierpaolo Correale
- Medical Oncology Unit, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
| | - Maria d'Apolito
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
- Medical Oncology Unit, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Luciano Mutti
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
- Department of Applied Sciences and Biotechnology, Università dell'Aquila, L'Aquila, Italy
| | - Rocco Giannicola
- Medical Oncology Unit, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Luigi Pirtoli
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
- Department of Medical Biotechnology, University of Siena, 53100, Siena, Italy
| | - Demetrio Labate
- Unit of Intensive Care Medicine and Anesthesia, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Sebastiano Macheda
- Unit of Intensive Care Medicine and Anesthesia, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Nicole Carabetta
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Youssef S Abdelwahed
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), 10785, Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), 10785, Berlin, Germany
- Berlin Institute of Health (BIH), 10178, Berlin, Germany
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| |
Collapse
|
21
|
Jaing TH, Hsiao YW, Wang YL. Chimeric Antigen Receptor Cell Therapy: Empowering Treatment Strategies for Solid Tumors. Curr Issues Mol Biol 2025; 47:90. [PMID: 39996811 PMCID: PMC11854309 DOI: 10.3390/cimb47020090] [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: 12/30/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/26/2025] Open
Abstract
Chimeric antigen receptor-T (CAR-T) cell therapy has demonstrated impressive efficacy in the treatment of blood cancers; however, its effectiveness against solid tumors has been significantly limited. The differences arise from a range of difficulties linked to solid tumors, including an unfriendly tumor microenvironment, variability within the tumors, and barriers to CAR-T cell infiltration and longevity at the tumor location. Research shows that the reasons for the decreased effectiveness of CAR-T cells in treating solid tumors are not well understood, highlighting the ongoing need for strategies to address these challenges. Current strategies frequently incorporate combinatorial therapies designed to boost CAR-T cell functionality and enhance their capacity to effectively target solid tumors. However, these strategies remain in the testing phase and necessitate additional validation to assess their potential benefits. CAR-NK (natural killer), CAR-iNKT (invariant natural killer T), and CAR-M (macrophage) cell therapies are emerging as promising strategies for the treatment of solid tumors. Recent studies highlight the construction and optimization of CAR-NK cells, emphasizing their potential to overcome the unique challenges posed by the solid tumor microenvironment, such as hypoxia and metabolic barriers. This review focuses on CAR cell therapy in the treatment of solid tumors.
Collapse
Affiliation(s)
- Tang-Her Jaing
- Division of Hematology and Oncology, Department of Pediatrics, Chang Gung Memorial Hospital, 5 Fu-Shin Street, Kwei-Shan, Taoyuan 33315, Taiwan;
| | - Yi-Wen Hsiao
- Division of Nursing, Chang Gung Memorial Hospital, 5 Fu-Shin Street, Kwei-Shan, Taoyuan 33315, Taiwan;
| | - Yi-Lun Wang
- Division of Hematology and Oncology, Department of Pediatrics, Chang Gung Memorial Hospital, 5 Fu-Shin Street, Kwei-Shan, Taoyuan 33315, Taiwan;
| |
Collapse
|
22
|
Summer M, Riaz S, Ali S, Noor Q, Ashraf R, Khan RRM. Understanding the Dual Role of Macrophages in Tumor Growth and Therapy: A Mechanistic Review. Chem Biodivers 2025:e202402976. [PMID: 39869825 DOI: 10.1002/cbdv.202402976] [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: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/29/2025]
Abstract
Macrophages are heterogeneous cells that are the mediators of tissue homeostasis. These immune cells originated from monocytes and are classified into two basic categories, M1 and M2 macrophages. M1 macrophages exhibit anti-tumorous inflammatory reactions due to the behavior of phagocytosis. M2 macrophages or tumor-associated macrophages (TAMs) are the most abundant immune cells in the tumor microenvironment (TME) and have a basic role in tumor progression by interacting with other immune cells in TME. By the expression of various cytokines, chemokines, and growth factors, TAMs lead to strengthening tumor cell proliferation, angiogenesis, and suppression of the immune system which further support invasion and metastasis. This review discusses recent and updated mechanisms regarding tumor progression by M2 macrophages. Moreover, the current therapeutic approaches targeting TAMs, their advantages, and limitations are also summarized, and further treatment approaches are outlined along with an elaboration of the tumor regression role of macrophages. This comprehensive review article possibly helps to understand the mechanisms underlying the tumor progression and regression role of macrophages in a comparative way from a basic level to the advanced one.
Collapse
Affiliation(s)
- Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Saima Riaz
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Shaukat Ali
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Qudsia Noor
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Rimsha Ashraf
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Rana Rashad Mahmood Khan
- Faculty of Chemistry and Life Sciences, Department of Chemistry, Government College University Lahore, Lahore, Pakistan
| |
Collapse
|
23
|
Chen C, Song H, Xu H, Chen M, Liang Z, Zhang M. Fall risk factors and mitigation strategies for hematological malignancy patients: insights from a qualitative study using the reason model. Support Care Cancer 2025; 33:118. [PMID: 39849285 DOI: 10.1007/s00520-025-09170-w] [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: 08/20/2024] [Accepted: 01/11/2025] [Indexed: 01/25/2025]
Abstract
PURPOSE Our study aim was to understand the (human and organizational) factors influencing fall risk among people with hematological malignancies using the Reason model as a framework, providing insights that can inform the development of safe and effective fall management strategies. METHODS Purposive sampling was employed to conduct semi-structured interviews with 13 people with hematological malignancies and 12 nurses from the hematology department of a tertiary grade A hospital in Guangzhou from December 2023 to February 2024. The topic analysis method was utilized to analyze the interview data. RESULTS Factors influencing fall risk among people with hematological malignancies were categorized into four themes: (1) precondition of unsafe arts (lack of work experience in junior nurses, poor patient compliance, adverse drug reactions, inadequate ward facilities); (2) unsafe supervision (inadequate inspection management, inadequate accompanying capacity); (3) unsafe arts (variability in subjective assessment, lack of bidirectional education); and (4) organizational influences (limited nursing human resources, lack of organizational process management models). CONCLUSION The specific fall risk factors among people with hematological malignancies, as summarized based on the Reason model framework, provide a theoretical basis and direction for the construction of specialized fall risk assessment tools, aiming to improve the quality of fall management for inpatients and reduce the incidence of falls.
Collapse
Affiliation(s)
- Cuishan Chen
- Department of Nursing, Nanfang Hosptial of Southern Medical University, Guangzhou, 510515, People's Republic of China
- School of Nursing, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Huijuan Song
- Department of Nursing, Nanfang Hosptial of Southern Medical University, Guangzhou, 510515, People's Republic of China.
| | - Huijuan Xu
- Department of Hematology, Nanfang Hosptial of Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Min Chen
- Department of Hematology, Nanfang Hosptial of Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Zilu Liang
- Department of Nursing, Nanfang Hosptial of Southern Medical University, Guangzhou, 510515, People's Republic of China
- School of Nursing, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Muchen Zhang
- Department of Nursing, Nanfang Hosptial of Southern Medical University, Guangzhou, 510515, People's Republic of China
- School of Nursing, Southern Medical University, Guangzhou, 510515, People's Republic of China
| |
Collapse
|
24
|
O'Shea PJ, Johnson PC, El-Jawahri A, Leblanc TW. Unmet needs and lived experience of patients receiving CAR T-cell therapy. Leuk Lymphoma 2025:1-11. [PMID: 39838695 DOI: 10.1080/10428194.2025.2455488] [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: 09/03/2024] [Revised: 12/16/2024] [Accepted: 01/14/2025] [Indexed: 01/23/2025]
Abstract
Chimeric Antigen Receptor T-Cell (CAR-T) therapy is an effective therapy and promising frontier in the treatment of hematologic malignancies. However, this revolutionary treatment has led to new challenges for patients, caregivers, and the healthcare system. In this review article, we discuss the various difficulties patients face both in the acute and long-term period following CAR-T infusion. We highlight the various ways these difficulties are addressed, as well as further areas of research and support needed to improve patient experience. Additionally, we consider the difficulties and burdens placed on caregivers and healthcare systems, as well as barriers to accessing CAR-T therapy. Finally, we address future directions of research and intervention development to meet patient and caregiver needs and improve equitable access. We pose early integration of specialty palliative care for individuals and their caregivers undergoing CAR-T therapy as one promising strategy to help improve patient experience and meet their needs.
Collapse
Affiliation(s)
- Patrick J O'Shea
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Patrick Connor Johnson
- Division of Hematology & Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Areej El-Jawahri
- Division of Hematology & Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Thomas W Leblanc
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Cancer Institute, Durham, NC, USA
| |
Collapse
|
25
|
Kuipers MT, Kersten MJ. CD19-directed chimeric antigen receptor T-cell therapy: what can we learn from the haematologist? Lupus Sci Med 2025; 12:e001157. [PMID: 39832905 PMCID: PMC11751780 DOI: 10.1136/lupus-2024-001157] [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: 10/21/2024] [Accepted: 12/14/2024] [Indexed: 01/22/2025]
Abstract
CD19-directed chimeric antigen receptor (CAR) T-cell therapy, originally developed for haematological malignancies, has recently emerged as a promising therapy for patients with autoimmune diseases. By selectively depleting CD19-positive B-cells, this therapy brings a new approach in resetting immune dysregulation and potentially providing long-term remission for patients with a refractory disease. Recent reports have highlighted its effectiveness in conditions such as SLE, systemic sclerosis and myositis. However, while these early results are encouraging, questions remain regarding strategies for optimal patient selection and minimising toxicity on the short and long term. The experiences with CD19 CAR T-cell therapy in haematology may offer valuable insights for immunologists and rheumatologists. This article reviews the key principles learnt in haematology, the results and the mechanisms behind its efficacy, toxicities, and the challenges that need to be addressed for its broader application in clinical practice.
Collapse
MESH Headings
- Humans
- Antigens, CD19/immunology
- Antigens, CD19/therapeutic use
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/therapeutic use
- Receptors, Antigen, T-Cell/immunology
- Autoimmune Diseases/therapy
- Autoimmune Diseases/immunology
- T-Lymphocytes/immunology
- B-Lymphocytes/immunology
- Hematologic Neoplasms/therapy
- Hematologic Neoplasms/immunology
- Lupus Erythematosus, Systemic/therapy
- Lupus Erythematosus, Systemic/immunology
Collapse
Affiliation(s)
| | - Marie José Kersten
- Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- LYMMCARE Lymphoma and Myeloma Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
26
|
Guo Z, He M, Liu N, Yang Y, Sun R, Wang J, Wang Q. In vitro functional validation of anti-CD19 chimeric antigen receptor T cells expressing lysine-specific demethylase 1 short hairpin RNA for the treatment of diffuse large B cell lymphoma. Front Immunol 2025; 15:1521778. [PMID: 39872520 PMCID: PMC11770093 DOI: 10.3389/fimmu.2024.1521778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 12/24/2024] [Indexed: 01/30/2025] Open
Abstract
Background Chimeric antigen receptor T (CAR-T) cell therapy is more effective in relapsed or refractory diffuse large B cell lymphoma (DLBCL) than other therapies, but a high proportion of patients relapse after CAR-T cell therapy owing to antigen escape, limited persistence of CAR-T cells, and immunosuppression in the tumor microenvironment. CAR-T cell exhaustion is a major cause of relapse. Epigenetic modifications can regulate T cell activation, maturation and depletion; they can be applied to reduce T cell depletion, improve infiltration, and promote memory phenotype formation to reduce relapse after CAR-T cell therapy. Purpose We propose to develop and validate in vitro the function of novel CAR-T cells for the treatment of DLBCL, which simultaneously express an anti-CD19 CAR with lysine-specific demethylase 1 (LSD1) short hairpin (sh)RNA to prevent depletion and prolong the survival of CAR-T cells. Methods We designed an shRNA sequence targeting LSD1 mRNA, and created a vector with the following elements: the U6 promoter driving expression of the LSD1 shRNA sequence, the EF1a promoter driving a second-generation anti-CD19 CAR sequence encoding an anti-CD19 single-chain variable fragment (FMC63), the CD8 hinge and transmembrane structural domains, the CD28 co-stimulatory structural domain, and the CD3ζ-activating structural domain. The MFG-LSD1 shRNA anti-CD19 CAR plasmid was first constructed, then packaged in retroviral vectors and transduced into human primary peripheral blood mononuclear cell-derived T cells to generate the corresponding CAR-T cells. We examined by flow cytometry the efficiency of two CAR-T cells in killing U-2932 cells (a human DLBCL line) upon co-culture with RNAU6 anti-CD19 CAR-T cells or LSD1 shRNA anti-CD19 CAR-T cells. We analyzed Ki-67 staining of the CAR-T cells by flow cytometry on days 0, 5, and 10, and counted the cells to assess expansion. We also used flow cytometry to detect the central memory T cell (TCM) proportion. Results We detected the expression of the CAR in the CAR-T cells by flow cytometry, and observed transduction rates of 31.5% for RNAU6 anti-CD19 CAR-T cells and 60.7% for LSD1 shRNA anti-CD19 CAR-T cells. The killing efficiency of LSD1 shRNA anti-CD19 CAR-T cells was significantly higher than that of RNAU6 anti-CD19 CAR-T cells at the low effector target ratio. We further found that LSD1 shRNA anti-CD19 CAR-T cells secreted more IFN-γ and granzyme B than RNAU6 anti-CD19 CAR-T cells. CAR-T cells proliferated after U-2932 cell stimulation and were able to sustain proliferation. After stimulation via U-2932 cell co-culture, both RNAU6 anti-CD19 CAR-T and LSD1 shRNA anti-CD19 CAR-T populations had increased proportions of cells with the TCM phenotype, with a higher percentage among LSD1 shRNA anti-CD19 CAR-T cells. Conclusion We developed a novel, feasible CD19-LSD1 shRNA CAR-T cell strategy for the treatment of DLBCL. Our in vitro assay results showed that LSD1 shRNA anti-CD19 CAR-T cells more effectively killed target cells than RNAU6 anti-CD19 CAR-T cells, and developed a higher proportion of TCM phenotype cells. LSD1 shRNA anti-CD19 CAR-T cells may represent a potential treatment for DLBCL.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/genetics
- Antigens, CD19/immunology
- Antigens, CD19/genetics
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Immunotherapy, Adoptive/methods
- RNA, Small Interfering/genetics
- Cell Line, Tumor
- Histone Demethylases/genetics
- Histone Demethylases/metabolism
- Histone Demethylases/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Zhi Guo
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
- Department of Hematology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Mingxin He
- Department of Hematology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Ning Liu
- Department of Hematology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Yiqing Yang
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, China
| | - Rui Sun
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, China
| | - Jianxun Wang
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, China
| | - Qiang Wang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
Xie D, Lu G, Mai G, Guo Q, Xu G. Tissue-resident memory T cells in diseases and therapeutic strategies. MedComm (Beijing) 2025; 6:e70053. [PMID: 39802636 PMCID: PMC11725047 DOI: 10.1002/mco2.70053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Tissue-resident memory T (TRM) cells are crucial components of the immune system that provide rapid, localized responses to recurrent pathogens at mucosal and epithelial barriers. Unlike circulating memory T cells, TRM cells are located within peripheral tissues, and they play vital roles in antiviral, antibacterial, and antitumor immunity. Their unique retention and activation mechanisms, including interactions with local epithelial cells and the expression of adhesion molecules, enable their persistence and immediate functionality in diverse tissues. Recent advances have revealed their important roles in chronic inflammation, autoimmunity, and cancer, illuminating both their protective and their pathogenic potential. This review synthesizes current knowledge on TRM cells' molecular signatures, maintenance pathways, and functional dynamics across different tissues. We also explore the interactions of TRM cells with other immune cells, such as B cells, macrophages, and dendritic cells, highlighting the complex network that underpins the efficacy of TRM cells in immune surveillance and response. Understanding the nuanced regulation of TRM cells is essential for developing targeted therapeutic strategies, including vaccines and immunotherapies, to enhance their protective roles while mitigating adverse effects. Insights into TRM cells' biology hold promise for innovative treatments for infectious diseases, cancer, and autoimmune conditions.
Collapse
Affiliation(s)
- Daoyuan Xie
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Guanting Lu
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Gang Mai
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Qiuyan Guo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di Herbs, Artemisinin Research Center, Institute of Chinese Materia MedicaAcademy of Chinese Medical SciencesBeijingChina
| | - Guofeng Xu
- Inflammation & Allergic Diseases Research UnitThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
28
|
Navab R, Futela P, Kumari V, Valecha J, Gupta RB, Jain R. Advancing Multiple Myeloma Immunotherapy: A Review of Chimeric Antigen Receptor T-Cell and Bispecific T-Cell Engagers Cell Therapies in Revolutionizing Treatment. IRANIAN JOURNAL OF MEDICAL SCIENCES 2025; 50:1-10. [PMID: 39957814 PMCID: PMC11829063 DOI: 10.30476/ijms.2024.101739.3446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 05/16/2024] [Accepted: 07/07/2024] [Indexed: 02/18/2025]
Abstract
Multiple Myeloma (MM) is a hematologic malignancy characterized by clonal plasma cell development, leading to serious complications. Despite traditional treatments, MM remains incurable, necessitating innovative therapeutic approaches. Chimeric Antigen Receptor (CAR) T-cell therapy and Bispecific T-cell engagers (BiTEs) are emerging immunotherapies showing promise in MM treatment. CAR T-cell therapy involves modifying patient T-cells to target specific antigens, primarily B Cell Maturation Antigen (BCMA). BiTEs, on the other hand, are non-IgG-like bispecific antibodies designed to engage both CD3 and tumor-associated antigens. These therapies exhibit impressive efficacy in clinical trials, leading to FDA approvals for specific MM patient populations. Despite their successes, these therapies come with unique challenges and adverse effects, such as cytokine release syndrome (CRS) and neurotoxicity. This narrative review explores the mechanisms, efficacy, challenges, and potential benefits of CAR T-cell and BiTE therapies for MM patients, shedding light on their roles in addressing this complex disease.
Collapse
MESH Headings
- Humans
- Multiple Myeloma/therapy
- Multiple Myeloma/immunology
- Receptors, Chimeric Antigen/therapeutic use
- Receptors, Chimeric Antigen/immunology
- Antibodies, Bispecific/therapeutic use
- Antibodies, Bispecific/pharmacology
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/trends
- T-Lymphocytes/immunology
- Immunotherapy/methods
- Immunotherapy/trends
- Receptors, Antigen, T-Cell/therapeutic use
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
- Rahul Navab
- PES Institute of Medical Sciences and Research, Kuppam, India
| | | | - Verkha Kumari
- Liaquat National Hospital and Medical College, Karachi, Pakistan
| | | | | | - Rohit Jain
- Penn State Health Milton S. Hershey Medical Center, Pennsylvania, United States of America
| |
Collapse
|
29
|
Palecki J, Bhasin A, Bernstein A, Mille PJ, Tester WJ, Kelly WK, Zarrabi KK. T-Cell redirecting bispecific antibodies: a review of a novel class of immuno-oncology for advanced prostate cancer. Cancer Biol Ther 2024; 25:2356820. [PMID: 38801069 PMCID: PMC11135853 DOI: 10.1080/15384047.2024.2356820] [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: 01/07/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024] Open
Abstract
Novel T-cell immunotherapies such as bispecific T-cell engagers (BiTEs) are emerging as promising therapeutic strategies for prostate cancer. BiTEs are engineered bispecific antibodies containing two distinct binding domains that allow for concurrent binding to tumor-associated antigens (TAAs) as well as immune effector cells, thus promoting an immune response against cancer cells. Prostate cancer is rich in tumor associated antigens such as, but not limited to, PSMA, PSCA, hK2, and STEAP1 and there is strong biologic rationale for employment of T-cell redirecting BiTEs within the prostate cancer disease space. Early generation BiTE constructs employed in clinical study have demonstrated meaningful antitumor activity, but challenges related to drug delivery, immunogenicity, and treatment-associated adverse effects limited their success. The ongoing development of novel BiTE constructs continues to address these barriers and to yield promising results in terms of efficacy and safety. This review will highlight some of most recent developments of BiTE therapies for patients with advanced prostate cancer and the evolving data surrounding BiTE constructs undergoing clinical evaluation.
Collapse
Affiliation(s)
- Julia Palecki
- Department of Internal Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Amman Bhasin
- Department of Internal Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Andrew Bernstein
- Department of Internal Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Patrick J. Mille
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - William J. Tester
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Wm. Kevin Kelly
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Kevin K. Zarrabi
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| |
Collapse
|
30
|
Huang Q, Li H, Zhang Y. A bibliometric and knowledge-map study on the treatment of hematological malignancies with CAR-T cells from 2012 to 2023. Hum Vaccin Immunother 2024; 20:2371664. [PMID: 38961667 PMCID: PMC11225924 DOI: 10.1080/21645515.2024.2371664] [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: 04/16/2024] [Accepted: 06/20/2024] [Indexed: 07/05/2024] Open
Abstract
Recently, CAR-T cell therapy in hematological malignancies has received extensive attention. The objective of this study is to gain a comprehensive understanding of the current research status, development trends, research hotspots, and emerging topics pertaining to CAR-T cells in the treatment of hematological malignancies. Articles pertaining to CAR-T cell therapy for hematological malignancies from the years 2012 to 2023 were obtained and assessed from the Web of Science Core Collection (WoSCC). A bibliometric approach was employed to conduct a scientific, comprehensive, and objective quantitative analysis, as well as a visual analysis, of this particular research domain. A comprehensive analysis was conducted on a corpus of 3643 articles, which were collaboratively authored by 72 countries and various research institutions. CAR-T cell research in treating hematological malignancies shows an increasing trend each year. Notably, the study identified the countries and institutions displaying the highest level of activity, the journals with the most citations and output, as well as the authors who garnered the highest frequency of citations and co-citations. Furthermore, the analysis successfully identified the research hotspots and highlighted six emerging topics within this domain. This study conducted a comprehensive exploration and analysis of the research status, development trends, research hotspots, and emerging topics about CAR-T cells in the treatment of hematological malignancies from 2012 to 2023. The findings of this study will serve as a valuable reference and guide for researchers seeking to delve deeper into this field and determine the future direction of their research.
Collapse
Affiliation(s)
- Qing Huang
- Department of Hematology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Huimin Li
- Department of Hematology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Yuan Zhang
- Department of Hematology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
31
|
Amer H, Kampan NC, Itsiopoulos C, Flanagan KL, Scott CL, Kartikasari AER, Plebanski M. Interleukin-6 Modulation in Ovarian Cancer Necessitates a Targeted Strategy: From the Approved to Emerging Therapies. Cancers (Basel) 2024; 16:4187. [PMID: 39766086 PMCID: PMC11674514 DOI: 10.3390/cancers16244187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
Despite significant advances in treatments, ovarian cancer (OC) remains one of the most prevalent and lethal gynecological cancers in women. The frequent detection at the advanced stages has contributed to low survival rates, resistance to various treatments, and disease recurrence. Thus, a more effective approach is warranted to combat OC. The cytokine Interleukin-6 (IL6) has been implicated in various stages of OC development. High IL6 levels are also correlated with a lower survival rate in OC patients. In this current review, we summarized the pivotal roles of IL6 in OC, including the initiation, development, invasion, metastasis, and drug resistance mechanisms. This article systematically highlights how targeting IL6 improves OC outcomes by altering various cancer processes and reports the ongoing clinical trials that would further shape the IL6-based targeted therapies. This review also suggests how combining IL6-targeted therapies with other therapeutic strategies could further enhance their efficacy to combat OC.
Collapse
Affiliation(s)
- Hina Amer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia; (H.A.); (A.E.R.K.)
| | - Nirmala C. Kampan
- Department of Obstetrics and Gynecology, Faculty of Medicine, University Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Catherine Itsiopoulos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia; (H.A.); (A.E.R.K.)
| | - Katie L. Flanagan
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia; (H.A.); (A.E.R.K.)
- School of Medicine and School of Health Sciences, University of Tasmania, Launceston, TAS 7250, Australia
- Tasmanian Vaccine Trial Centre, Clifford Craig Foundation, Launceston General Hospital, Launceston, TAS 7250, Australia
| | - Clare L. Scott
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Faculty of Medicine, Dentistry, and Health Sciences, The University of Melbourne, Parkville, VIC 3052, Australia
- The Royal Women’s Hospital, Parkville, VIC 3052, Australia
| | - Apriliana E. R. Kartikasari
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia; (H.A.); (A.E.R.K.)
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3082, Australia; (H.A.); (A.E.R.K.)
| |
Collapse
|
32
|
Mohammad A, Yurina A, Simonyan T, Chistyakov D, Salman R, Zornikova K, Minina E, Bogolyubova A. Modular (universal) CAR-T platforms in vivo: a comprehensive systematic review. Front Immunol 2024; 15:1409665. [PMID: 39712013 PMCID: PMC11659234 DOI: 10.3389/fimmu.2024.1409665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 11/14/2024] [Indexed: 12/24/2024] Open
Abstract
Background Modular (universal) CAR T-platforms were developed to combat the limitations of traditional CAR-T therapy, allowing for multiple targeting of tumor-associated antigens and the ability to control CAR-T cell activity. The modular CAR-T platform consists of a universal receptor (signaling module) that recognizes an adapter molecule on the soluble module, which is responsible for antigen recognition. Multiple platforms have been developed over the last 12 years, and some of them have entered the clinical trial phase. This systematic review seeks to evaluate the different parameters of modular CAR-T platforms performance in animal models. Methods A systematic search of literature in the PubMed database and in Google Scholar and BASE (Bielefeld Academic Search Engine) search engines was performed according to predefined eligibility criteria. All studies conducted on xenograft mouse models with any variant of modular CAR-T platforms were included. Forest plots were generated for visual presentation of the extracted quantitative findings (standardized mean difference (SMD) and median survival rate (MSR)). Results A total of 33 studies employing 15 different modular CAR-T platforms were included. The platforms varied in terms of CAR-T cells, soluble module doses, and their frequency of administration. The studies showed a reduction in tumor burden and in tumor volume compared to the combined negative group. In comparison with the positive control group, there was no significant change in tumor burden or volume. In all the included studies the experimental group had a higher survival probability compared to the combined negative group at the study endpoint, with no significant difference in survival rate compared to the positive control group. Conclusion The modular CAR-T platforms are generally effective and are a valuable addition to the arsenal of CAR therapy. Systematic Review Registration https://www.crd.york.ac.uk/prospero/ PROSPERO, identifier CRD42023443984.
Collapse
|
33
|
Sun Y, Liu J, Zhan D, Wei J, XianShi L, Zhang R, Duan C, Zhang D, Tang X, Lin T, Li L, Lai X. Depletion of Tregs from CD4 + CAR-T cells enhances the tumoricidal effect of CD8 + CAR-T cells in anti-CD19 CAR-T therapy. FEBS J 2024. [PMID: 39632397 DOI: 10.1111/febs.17326] [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: 11/18/2023] [Revised: 07/18/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024]
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy, which targets CD19 for hematological malignancies, represents a breakthrough in cancer immunotherapy. However, some patients may develop resistance to CAR-T treatment, underscoring the importance of optimizing CAR-T design to enhance responsiveness. Here, we investigated the impact of different subpopulations in anti-CD19 CAR-T cells on the tumoricidal effect. Different populations of anti-CD19 CAR-T cells were isolated by magnetic-activated cell sorting (MACS). Their lytic activities on the acute lymphocytic leukemia cell line SUP-B15 and diffuse large B-cell lymphoma EB-3 cell line were examined in a co-culture system. The anti-tumorigenic outcome of different CAR-T cell compositions was evaluated in a xenograft mouse model of EB-3 cells. CD8+CAR-T cells exhibited the most potent tumoricidal activity against SUP-B15 and EB-3 cells. Additionally, CD4+ T helper cells enhanced the lytic effects of CD8+ CAR-T cells by increasing the availability of interleukin-2 (IL-2). Depleting CD25+Treg (T regulatory) cells from CD4+CAR-T population further augmented the tumoricidal activity of CD8+CAR-T cells by preventing IL-2 deprivation. Consistently, in vivo experiments demonstrated that the CD4+CD25+ Treg population dampened the antitumor activity of CD8+CAR-T cells, while depletion of Tregs from CD4+CAR-T cells enhanced the tumoricidal effect. These findings emphasize the potential role of CAR Treg cells in therapeutic resistance, suggesting that the depletion of Tregs in the anti-CD19 CAR-T population may serve as a strategy to augment the anticancer effect of CD8+CAR-T cells.
Collapse
Affiliation(s)
- Yunyan Sun
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Jinyan Liu
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Dong Zhan
- Department of Human Anatomy and Histology & Embrology, School of Basic Medical Sciences, Kunming Medical University, China
| | - Jia Wei
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Li XianShi
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Rui Zhang
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Ci Duan
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Disi Zhang
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Xiaorong Tang
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Tuo Lin
- Yunnan College of Business Management, Kunming, China
| | - Limei Li
- Yunnan College of Business Management, Kunming, China
| | - Xun Lai
- Department of Hematology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| |
Collapse
|
34
|
Sainatham C, Yadav D, Dilli Babu A, Tallapalli JR, Kanagala SG, Filippov E, Murillo Chavez F, Ahmed N, Lutfi F. The current socioeconomic and regulatory landscape of immune effector cell therapies. Front Med (Lausanne) 2024; 11:1462307. [PMID: 39697210 PMCID: PMC11652178 DOI: 10.3389/fmed.2024.1462307] [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: 07/09/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
Immune cell effector therapies, including chimeric antigen receptor (CAR)-T cells, T-cell receptor (TCR) T cells, natural killer (NK) cells, and macrophage-based therapies, represent a transformative approach to cancer treatment, harnessing the immune system to target and eradicate malignant cells. CAR-T cell therapy, the most established among these, involves engineering T cells to express CARs specific to cancer cell antigens, showing remarkable efficacy in hematologic malignancies like leukemias, B-cell lymphomas, and multiple myeloma. Similarly, TCR-modified therapies, which reprogram T cells to recognize intracellular tumor antigens presented by major histocompatibility complex (MHC) molecules, offer promise for a range of solid tumors. NK-cell therapies leverage NK cells' innate cytotoxicity, providing an allogeneic approach that avoids some of the immune-related complications associated with T-cell-based therapies. Macrophage-based therapies, still in early stages of the development, focus on reprogramming macrophages to stimulate an immune response against cancer cells in the tumor microenvironment. Despite their promise, socioeconomic and regulatory challenges hinder the accessibility and scalability of immune cell effector therapies. These treatments are costly, with CAR-T therapies currently exceeding $400,000 per patient, creating significant disparities in access based on socioeconomic status and geographic location. The high manufacturing costs stem from the personalized, labor-intensive processes of harvesting, modifying, and expanding patients' cells. Moreover, complex logistics for manufacturing and delivering these therapies limit their reach, particularly in low-resource settings. Regulatory pathways further complicate the landscape. In the United States., the Food and Drug Administrations' (FDA) accelerated approval processes for cell-based therapies facilitate innovation but do not address cost-related barriers. In Europe, the European Medicines Agency (EMA) offers adaptive pathways, yet decentralized reimbursement systems create uneven access across member states. Additionally, differing regulatory standards for manufacturing and quality control worldwide pose hurdles for global harmonization and access. To expand the reach of immune effector cell therapies, a multipronged approach is needed-streamlined regulatory frameworks, policies to reduce treatment costs, and international collaborations to standardize manufacturing. Addressing these socioeconomic and regulatory obstacles is essential to make these life-saving therapies accessible to a broader patient population worldwide. We present a literature review on the current landscape of immune effector cell therapies and barriers of access to currently approved standard of care therapy at various levels.
Collapse
Affiliation(s)
- Chiranjeevi Sainatham
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Devvrat Yadav
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Aravind Dilli Babu
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Jayanth Reddy Tallapalli
- Division of Infectious Diseases, Department of Internal Medicine, University of South Florida, Tampa, FL, United States
| | - Sai Gautham Kanagala
- Department of Internal Medicine, New York Medical College/Metropolitan Hospital Center, New York, NY, United States
| | - Evgenii Filippov
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Franco Murillo Chavez
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Nausheen Ahmed
- Department of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Forat Lutfi
- Department of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
35
|
Ahuja S, Zaheer S. The evolution of cancer immunotherapy: a comprehensive review of its history and current perspectives. KOREAN JOURNAL OF CLINICAL ONCOLOGY 2024; 20:51-73. [PMID: 39778508 PMCID: PMC11717579 DOI: 10.14216/kjco.24009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/24/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025]
Abstract
Cancer immunotherapy uses the body's immune system to combat cancer, marking a significant advancement in treatment. This review traces its evolution from the late 19th century to its current status. It began with William Coley's pioneering work using bacterial toxins to stimulate the immune system against cancer cells, establishing the foundational concept of immunotherapy. In the mid-20th century, cytokine therapies like interferons and interleukins emerged, demonstrating that altering the immune response could reduce tumors and highlighting the complex interplay between cancer and the immune system. The discovery of immune checkpoints, regulatory pathways that prevent autoimmunity but are exploited by cancer cells to evade detection, was a pivotal development. Another major breakthrough is CAR-T cell therapy, which involves modifying a patient's T cells to target cancer-specific antigens. This personalized treatment has shown remarkable success in certain blood cancers. Additionally, cancer vaccines aim to trigger immune responses against tumor-specific or associated antigens, and while challenging, ongoing research is improving their efficacy. The historical progression of cancer immunotherapy, from Coley's toxins to modern innovations like checkpoint inhibitors and CAR-T cell therapy, underscores its transformative impact on cancer treatment. As research delves deeper into the immune system's complexities, immunotherapy is poised to become even more crucial in oncology, offering renewed hope to patients globally.
Collapse
Affiliation(s)
- Sana Ahuja
- Department of Pathology, Safdarjung Hospital, Vardhman Mahavir Medical College, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Safdarjung Hospital, Vardhman Mahavir Medical College, New Delhi, India
| |
Collapse
|
36
|
Laface C, Fina E, Ricci AD, Guven DC, Ambrogio F, De Summa S, Vitale E, Massafra R, Brunetti O, Rizzo A. Immunobiology of biliary tract cancer and recent clinical findings in approved and upcoming immune checkpoint inhibitors. Expert Opin Biol Ther 2024; 24:1363-1374. [PMID: 39545466 DOI: 10.1080/14712598.2024.2431088] [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: 08/01/2024] [Revised: 11/14/2024] [Accepted: 11/14/2024] [Indexed: 11/17/2024]
Abstract
INTRODUCTION Recently, immunotherapy has offered new hope for treating biliary tract cancer (BTC). However, several issues are to be considered, including the lack of validated predictive biomarkers that could help to identify patient groups which are most likely to benefit from such therapeutic approaches. AREAS COVERED In the current article, we will provide an overview of recent results and ongoing and future research directions of immunotherapy in BTC, with a special focus on recently published, practice-changing data, and ongoing active and recruiting clinical trials. EXPERT OPINION At this moment, dozens of clinical trials in phases I to III are evaluating the role of cancer immunotherapy in this setting, with the hope of adding more therapeutic options for BTC patients. Future research must focus on the development of novel agents and combinations, but the validation of biomarkers remains an urgent need. As more research results emerge, novel combinatorial strategies are destined to further transform the treatment paradigm for this heterogeneous and aggressive tumor type.
Collapse
Affiliation(s)
- Carmelo Laface
- Azienda Sanitaria Provinciale, Reggio Calabria (RC), Italy
| | - Emanuela Fina
- Thoracic Surgery Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angela Dalia Ricci
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS "S. de Bellis" Research Hospital, Castellana Grotte, Italy
| | - Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
- Medical Oncology Clinic, Elazig City Hospital, Health Sciences University, Elazig, Turkey
| | - Francesca Ambrogio
- Section of Dermatology, Department of Biomedical Science and Human Oncology, University of Bari, Bari, Italy
| | - Simona De Summa
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori, "Giovanni Paolo II", Bari, Italy
| | - Elsa Vitale
- Scientific Directorate, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Raffaella Massafra
- Scientific Directorate, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Oronzo Brunetti
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Alessandro Rizzo
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| |
Collapse
|
37
|
Liu X, Chen H, Tan G, Zhong L, Jiang H, Smith SM, Wang HZ. A comprehensive neuroimaging review of the primary and metastatic brain tumors treated with immunotherapy: current status, and the application of advanced imaging approaches and artificial intelligence. Front Immunol 2024; 15:1496627. [PMID: 39669560 PMCID: PMC11634813 DOI: 10.3389/fimmu.2024.1496627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 10/28/2024] [Indexed: 12/14/2024] Open
Abstract
Cancer immunotherapy has emerged as a novel clinical therapeutic option for a variety of solid tumors over the past decades. The application of immunotherapy in primary and metastatic brain tumors continues to grow despite limitations due to the physiological characteristics of the immune system within the central nervous system (CNS) and distinct pathological barriers of malignant brain tumors. The post-immunotherapy treatment imaging is more complex. In this review, we summarize the clinical application of immunotherapies in solid tumors beyond the CNS. We provide an overview of current immunotherapies used in brain tumors, including immune checkpoint inhibitors (ICIs), oncolytic viruses, vaccines, and CAR T-cell therapies. We focus on the imaging criteria for the assessment of treatment response to immunotherapy, and post-immunotherapy treatment imaging patterns. We discuss advanced imaging techniques in the evaluation of treatment response to immunotherapy in brain tumors. The imaging characteristics of immunotherapy treatment-related complications in CNS are described. Lastly, future imaging challenges in this field are explored.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Radiology, The Affiliated Yuebei People’s Hospital of Shantou University Medical College, Shaoguan, Guangdong, China
- Advanced Neuroimaging Laboratory, The Affiliated Yuebei People’s Hospital of Shantou University Medical College, Shaoguan, Guangdong, China
| | - Hongyan Chen
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guirong Tan
- Department of Radiology, The Affiliated Yuebei People’s Hospital of Shantou University Medical College, Shaoguan, Guangdong, China
- Advanced Neuroimaging Laboratory, The Affiliated Yuebei People’s Hospital of Shantou University Medical College, Shaoguan, Guangdong, China
| | - Lijuan Zhong
- Department of Pathology, The Affiliated Yuebei People’s Hospital of Shantou University Medical College, Shaoguan, Guangdong, China
| | - Haihui Jiang
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, China
| | - Stephen M. Smith
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY, United States
| | - Henry Z. Wang
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
38
|
Hu L, Wang X, Song Z, Chen F, Wu B. Leveraging CAR macrophages targeting c-Met for precision immunotherapy in pancreatic cancer: insights from single-cell multi-omics. Mol Med 2024; 30:231. [PMID: 39592929 PMCID: PMC11590533 DOI: 10.1186/s10020-024-00996-4] [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: 08/27/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Pancreatic cancer is known for its poor prognosis and resistance to conventional therapies, largely due to the presence of cancer stem cells (CSCs) and aggressive angiogenesis. Effectively targeting these CSCs and associated angiogenic pathways is crucial for effective treatment. This study leverages single-cell multi-omics to explore a novel therapeutic approach involving Chimeric Antigen Receptor (CAR) macrophages engineered to target the c-Met protein on pancreatic CSCs. METHODS We employed single-cell RNA sequencing to analyze pancreatic cancer tissue, identifying c-Met as a key marker of CSCs. CAR macrophages were engineered using a lentiviral system to express a c-Met-specific receptor. The phagocytic efficiency of these CAR macrophages against pancreatic CSCs was assessed in vitro, along with their ability to inhibit angiogenesis. The in vivo efficacy of CAR macrophages was evaluated in a mouse model of pancreatic cancer. RESULTS CAR macrophages demonstrated high specificity for c-Met + CSCs, significantly enhancing phagocytosis and reducing the secretion of angiogenic factors such as VEGFA, FGF2, and ANGPT. In vivo, these macrophages significantly suppressed tumor growth and angiogenesis, prolonging survival in pancreatic cancer-bearing mice. CONCLUSION CAR macrophages targeting c-Met represent a promising therapeutic strategy for pancreatic cancer, offering targeted elimination of CSCs and disruption of tumor angiogenesis. This study highlights the potential of single-cell multi-omics in guiding the development of precision immunotherapies.
Collapse
Affiliation(s)
- Lingyu Hu
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, No. 1518 North Huancheng Road, Jiaxing, Zhejiang, 314000, People's Republic of China
| | - Xiaoguang Wang
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, No. 1518 North Huancheng Road, Jiaxing, Zhejiang, 314000, People's Republic of China
| | - Zhengwei Song
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, No. 1518 North Huancheng Road, Jiaxing, Zhejiang, 314000, People's Republic of China
| | - Fei Chen
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, No. 1518 North Huancheng Road, Jiaxing, Zhejiang, 314000, People's Republic of China
| | - Bin Wu
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, No. 1518 North Huancheng Road, Jiaxing, Zhejiang, 314000, People's Republic of China.
| |
Collapse
|
39
|
Ribeiro J, Pagès-Geli C, Meglan A, Velarde J, Blandin J, Vaccaro K, Wienclaw T, Fernández-Guzmán P, Hahn CK, Crespo M, Weiskopf K. Unbiased discovery of antibody therapies that stimulate macrophage-mediated destruction of B-cell lymphoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.623229. [PMID: 39605364 PMCID: PMC11601295 DOI: 10.1101/2024.11.13.623229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Macrophages are critical effectors of antibody therapies for lymphoma, but the best targets for this purpose remain unknown. Here, we sought to define a comprehensive repertoire of cell surface antigens that can be targeted to stimulate macrophage-mediated destruction of B-cell lymphoma. We developed a high-throughput assay to screen hundreds of antibodies for their ability to provoke macrophages to attack B-cell lymphoma cells. Across both mouse and human systems, we identified multiple unappreciated targets of opsonization as well as putative immune checkpoints. We used this information to engineer a compendium of 156 bispecific antibodies, and we identified dozens of bispecifics that dramatically stimulate macrophage-mediated cytotoxicity of lymphoma cells. Among these, a bispecific comprising a SIRPα decoy domain and a CD38-targeting arm (WTa2d1×CD38) exhibited maximal efficacy while minimizing the risk of hematologic toxicity. This bispecific stimulated robust anti-tumor responses in multiple xenograft models of aggressive B-cell lymphoma. Our approach can be directly applied to other cancers to rapidly discover bispecific antibodies that leverage anti-tumor responses by macrophages or other innate immune cells.
Collapse
Affiliation(s)
- Juliano Ribeiro
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
| | - Carlota Pagès-Geli
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
- Experimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, C/ Natzaret, 115-117, 08035 Barcelona, Spain
- Department of Medicine, Universitat Aut noma de Barcelona, 08193 Bellaterra, Spain
| | - Anna Meglan
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
| | - Jose Velarde
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
| | - Jasmine Blandin
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
| | - Kyle Vaccaro
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
| | - Thomas Wienclaw
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
| | - Patricia Fernández-Guzmán
- Experimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, C/ Natzaret, 115-117, 08035 Barcelona, Spain
- Department of Medicine, Universitat Aut noma de Barcelona, 08193 Bellaterra, Spain
| | - Cynthia K. Hahn
- Dana-Farber Cancer Institute, Boston, MA 02115
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142
- Harvard Medical School, Boston, MA 02115
| | - Marta Crespo
- Experimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, C/ Natzaret, 115-117, 08035 Barcelona, Spain
- Department of Medicine, Universitat Aut noma de Barcelona, 08193 Bellaterra, Spain
| | - Kipp Weiskopf
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
- Dana-Farber Cancer Institute, Boston, MA 02115
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology; Cambridge, MA 02139, USA
| |
Collapse
|
40
|
Luo Y, Shentu J, Xu H, Xia Y, Fang L, Duan S. Innovative pan-tumor target strategy for CAR-T therapy: cancer-specific exons as novel targets for pediatric solid and brain tumors. J Transl Med 2024; 22:1019. [PMID: 39533264 PMCID: PMC11556178 DOI: 10.1186/s12967-024-05861-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) immunotherapy has achieved remarkable success in treating chemotherapy-refractory hematological malignancies. However, its efficacy in solid and brain tumors remains limited due to challenges such as insufficient target antigens, poor T-cell adaptability, inefficient tumor site trafficking, and the immunosuppressive tumor microenvironment. To address these challenges, Shaw and colleagues proposed an innovative strategy targeting cancer-specific exons (CSEs) in pediatric solid and brain tumors. Using RNA sequencing data from 16 tumor types, the study identified 157 highly tumor-specific targets, including both known and novel proteins. The researchers validated several targets, including FN1 and COL11A1, demonstrating their therapeutic potential in in vitro and in vivo models. The study's approach of integrating exon-level analysis with a broad search for extracellular matrix proteins offers a new frontier for CAR-T therapy, providing valuable insights for improving immunotherapy in pediatric solid tumors. Although promising, the study also highlights the need for further evaluation of tumor recurrence and CAR-T cell exhaustion. The identification of novel pan-tumor targets may revolutionize CAR-T therapy design and expand its application in pediatric cancer treatment.
Collapse
Affiliation(s)
- Yuqing Luo
- Department of Hematology, Yuyao People's Hospital of Zhejiang Province, The Affiliated Yangming Hospital of Ningbo University, Yuyao, 315400, Zhejiang, China
| | - Jianqiao Shentu
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang, China
| | - Hening Xu
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang, China
| | - Yongming Xia
- Department of Hematology, Yuyao People's Hospital of Zhejiang Province, The Affiliated Yangming Hospital of Ningbo University, Yuyao, 315400, Zhejiang, China
| | - Lili Fang
- Department of Hematology, Yuyao People's Hospital of Zhejiang Province, The Affiliated Yangming Hospital of Ningbo University, Yuyao, 315400, Zhejiang, China.
| | - Shiwei Duan
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang, China.
| |
Collapse
|
41
|
Yang Y, Peng H, Wang J, Li F. New insights into CAR T-cell hematological toxicities: manifestations, mechanisms, and effective management strategies. Exp Hematol Oncol 2024; 13:110. [PMID: 39521987 PMCID: PMC11549815 DOI: 10.1186/s40164-024-00573-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/12/2024] [Indexed: 11/16/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy represents a highly efficacious treatment modality demonstrated to enhance outcomes in patients afflicted with malignancies, particularly those enduring relapsed or refractory hematological malignancies. However, the escalating adoption of CAR T-cell therapy has unveiled several life-threatening toxicities, notably cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), infections, and hematological toxicities (HTs), thereby hindering the broad implementation of CAR T-cell therapy. HTs encompass a spectrum of adverse effects, including cytopenias, hemophagocytic lymphohistiocytosis (HLH), coagulopathies, and B-cell aplasia. While our comprehension of the underlying mechanisms governing CRS and ICANS is advancing, the intricate pathophysiology of HTs remains inadequately elucidated. Such knowledge gaps may precipitate suboptimal therapeutic decisions, potentially culminating in substantial medical resource depletion and detriment to patients' quality of life. In this comprehensive review, based on recent updated findings, we delineate various mechanisms contributing to HTs subsequent to CAR T-cell therapy, explicate manifestations of HTs, and proffer strategic interventions to mitigate this relevant clinical challenge.
Collapse
Affiliation(s)
- Yuanyuan Yang
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hongwei Peng
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, National Clinical Research Center for Blood Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Fei Li
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
- Jiangxi Clinical Research Center for Hematologic Disease, Nanchang, Jiangxi, China.
- Institute of Lymphoma and Myeloma, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
42
|
Amiri M, Moaveni AK, Majidi Zolbin M, Shademan B, Nourazarian A. Optimizing cancer treatment: the synergistic potential of CAR-T cell therapy and CRISPR/Cas9. Front Immunol 2024; 15:1462697. [PMID: 39582866 PMCID: PMC11581867 DOI: 10.3389/fimmu.2024.1462697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/23/2024] [Indexed: 11/26/2024] Open
Abstract
Optimizing cancer treatment has become a pivotal goal in modern oncology, with advancements in immunotherapy and genetic engineering offering promising avenues. CAR-T cell therapy, a revolutionary approach that harnesses the body's own immune cells to target and destroy cancer cells, has shown remarkable success, particularly in treating acute lymphoblastic leukemia (ALL), and in treating other hematologic malignancies. While CAR-T cell therapy has shown promise, challenges such as high cost and manufacturing complexity remain. However, its efficacy in solid tumors remains limited. The integration of CRISPR/Cas9 technology, a powerful and precise genome-editing tool, also raises safety concerns regarding unintended edits and off-target effects, offers a synergistic potential to overcome these limitations. CRISPR/Cas9 can enhance CAR-T cell therapy by improving the specificity and persistence of CAR-T cells, reducing off-target effects, and engineering resistance to tumor-induced immunosuppression. This combination can also facilitate the knockout of immune checkpoint inhibitors, boosting the anti-tumor activity of CAR-T cells. Recent studies have demonstrated that CRISPR/Cas9-edited CAR-T cells can target previously untreatable cancer types, offering new hope for patients with refractory cancers. This synergistic approach not only enhances the efficacy of cancer treatment but also paves the way for personalized therapies tailored to individual genetic profiles. This review highlights the ongoing research efforts to refine this approach and explores its potential to revolutionize cancer treatment across a broader range of malignancies. As research progresses, the integration of CAR-T cell therapy and CRISPR/Cas9 holds the promise of transforming cancer treatment, making it more effective and accessible. This review explores the current advancements, challenges, and future prospects of this innovative therapeutic strategy.
Collapse
Affiliation(s)
- Maryam Amiri
- Pediatric Urology and Regenerative Medicine Research Center, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Kian Moaveni
- Pediatric Urology and Regenerative Medicine Research Center, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Majidi Zolbin
- Pediatric Urology and Regenerative Medicine Research Center, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Behrouz Shademan
- Medical Journalism, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| |
Collapse
|
43
|
Yu XJ, Liu C, Hu SZ, Yuan ZY, Ni HY, Sun SJ, Hu CY, Zhan HQ. Application of CAR-T cell therapy in B-cell lymphoma: a meta-analysis of randomized controlled trials. Clin Transl Oncol 2024:10.1007/s12094-024-03774-0. [PMID: 39514165 DOI: 10.1007/s12094-024-03774-0] [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: 06/10/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND This study aims to compare the efficacy and safety of chimeric antigen receptor T-cell (CAR-T) immunotherapy with standard treatment for B-cell lymphoma, providing evidence-based support for the more efficient use of CAR-T cell immunotherapy. METHODS We conducted a comprehensive literature search of high-quality randomized controlled trials (RCTs) on CAR-T therapy for B-cell lymphoma in the following databases: Wanfang, Web of Science, CNKI, VIP database, and PubMed, up to February 2024. The outcome measures included objective remission rate (ORR), complete remission rate (CRR), and incidence of adverse reactions. Subgroup analysis was performed based on the differences in co-stimulatory domains. Meta-analysis was conducted using Review Manager 5.4 and Stata software. RESULTS A total of five RCTs involving 1670 patients were included in this meta-analysis. The results showed that the CAR-T treatment group had significantly higher ORR (RR: 1.47, 95% CI 1.23-1.76, I2 = 80%, p < 0.0001), CRR (RR: 2.19, 95% CI 2.16-3.79, I2 = 93%, p = 0.005), cytokine release syndrome (CRS) incidence (RR: 34.51, 95% CI 2.27-523.78, I2 = 98%, p = 0.01), neurotoxicity (NT) incidence (RR: 6.00, 95% CI 1.82-19.75, I2 = 80%, p = 0.003), neutropenia incidence (RR: 1.39, 95% CI 1.02-1.88, I2 = 93%, p = 0.03), leukopenia incidence (RR: 1.39, 95% CI 1.04-1.87, I2 = 61%, p = 0.03), and headache incidence (RR: 1.56, 95% CI 1.25-1.95, I2 = 34%, p < 0.0001) compared to the standard treatment group. Subgroup analysis based on co-stimulatory domains revealed that the 4-1BB subgroup had higher incidences of CRR, CRS, NT and leukopenia than the CD28 subgroup; however, the CD28 subgroup exhibited higher ORR and neutropenia than the 4-1BB subgroup. CONCLUSION CAR-T cell immunotherapy demonstrates superior efficacy compared to standard therapy in treating B-cell lymphoma. However, CAR-T treatment can lead to adverse reactions such as CRS and NT. Infusion of an appropriate dose of CAR-T cells (e.g., 100 × 106) may be a strategy to mitigate the risk of CRS and NT.
Collapse
Affiliation(s)
- Xiao-Jing Yu
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Chang Liu
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Shi-Zhi Hu
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zhan-Yuan Yuan
- Department of Plastic and Reconstructive Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Hai-Yan Ni
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Sheng-Jia Sun
- Clinical Medical College of Anhui Medical University, Hefei, 230031, China
| | - Cheng-Yang Hu
- Department of Epidemiology and Biostatistics, Anhui Medical University, Hefei, 230032, China.
| | - He-Qin Zhan
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
44
|
Zhou Y, Wei S, Xu M, Wu X, Dou W, Li H, Zhang Z, Zhang S. CAR-T cell therapy for hepatocellular carcinoma: current trends and challenges. Front Immunol 2024; 15:1489649. [PMID: 39569202 PMCID: PMC11576447 DOI: 10.3389/fimmu.2024.1489649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024] Open
Abstract
Hepatocellular carcinoma (HCC) ranks among the most prevalent cancers worldwide, highlighting the urgent need for improved diagnostic and therapeutic methodologies. The standard treatment regimen generally involves surgical intervention followed by systemic therapies; however, the median survival rates for patients remain unsatisfactory. Chimeric antigen receptor (CAR) T-cell therapy has emerged as a pivotal advancement in cancer treatment. Both clinical and preclinical studies emphasize the notable efficacy of CAR T cells in targeting HCC. Various molecules, such as GPC3, c-Met, and NKG2D, show significant promise as potential immunotherapeutic targets in liver cancer. Despite this, employing CAR T cells to treat solid tumors like HCC poses considerable challenges within the discipline. Numerous innovations have significant potential to enhance the efficacy of CAR T-cell therapy for HCC, including improvements in T cell trafficking, strategies to counteract the immunosuppressive tumor microenvironment, and enhanced safety protocols. Ongoing efforts to discover therapeutic targets for CAR T cells highlight the need for the development of more practical manufacturing strategies for CAR-modified cells. This review synthesizes recent findings and clinical advancements in the use of CAR T-cell therapies for HCC treatment. We elucidate the therapeutic benefits of CAR T cells in HCC and identify the primary barriers to their broader application. Our analysis aims to provide a comprehensive overview of the current status and future prospects of CAR T-cell immunotherapy for HCC.
Collapse
Affiliation(s)
- Yexin Zhou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- The General Hospital of Western Theater Command, Chengdu, China
| | - Shanshan Wei
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Menghui Xu
- The General Hospital of Western Theater Command, Chengdu, China
| | - Xinhui Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Wenbo Dou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Huakang Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zhonglin Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Shuo Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
45
|
Russo E, Gambella M, Raiola AM, Beltrametti E, Zanetti V, Chirco G, Viazzi F, Angelucci E, Esposito P. Acute kidney injury in hematological patients treated with CAR-T cells: risk factors, clinical presentation and impact on outcomes. Sci Rep 2024; 14:26886. [PMID: 39506012 PMCID: PMC11542077 DOI: 10.1038/s41598-024-77720-z] [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: 09/02/2024] [Accepted: 10/24/2024] [Indexed: 11/08/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has revolutionized the treatment of hematologic malignancies, yet it carries significant risks, including acute kidney injury (AKI). In this study, we investigated the risk factors and clinical impact of AKI in patients undergoing CAR-T cell therapy. This retrospective study involved hematologic patients treated with CAR-T therapy. Clinical and laboratory data were collected, and clinical outcomes were monitored during follow-up after CAR-T infusion. AKI was defined according to KDIGO criteria. The outcome measures included early mortality, overall survival (OS), and disease-free survival (DFS). Among the 48 patients analyzed, 14 (29%) developed AKI, with a mean onset of 6 days after CAR-T infusion. The risk of AKI was associated with baseline performance status (OR 8.65, IC95% 6.2-12, p = 0.032) and the development of severe cytokine release syndrome post-therapy (OR 16.4 95%CI 1.9-138.5, p = 0.01). Patients with AKI more frequently required intensive care. Furthermore, severe AKI was independently associated with worse clinical outcomes, including reduced OS and DFS (HR 18.2, 95%CI 2.6-27.3, p = 0.003). Additionally, patients who developed AKI post-CAR-T therapy were more likely to progress to chronic kidney disease during follow-up. In conclusion, frail patients undergoing CAR-T therapy are at an increased risk of developing AKI, which can significantly affect both short- and long-term outcomes. Preventive strategies and early recognition of AKI are essential in these patients.
Collapse
Affiliation(s)
- Elisa Russo
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, Genova, Italy
- Unit of Nephrology, Dialysis and Transplantation, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Massimiliano Gambella
- Unit of Hematology and Cellular Therapy, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Anna Maria Raiola
- Unit of Hematology and Cellular Therapy, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Elena Beltrametti
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, Genova, Italy
| | - Valentina Zanetti
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, Genova, Italy
| | - Giuseppe Chirco
- Unit of Nephrology, Dialysis and Transplantation, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesca Viazzi
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, Genova, Italy
- Unit of Nephrology, Dialysis and Transplantation, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Emanuele Angelucci
- Unit of Hematology and Cellular Therapy, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Pasquale Esposito
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genova, Genova, Italy.
- Unit of Nephrology, Dialysis and Transplantation, IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| |
Collapse
|
46
|
Khazamipour N, Oo HZ, Al-Nakouzi N, Marzban M, Khazamipour N, Roberts ME, Farivar N, Moskalev I, Lo J, Ghaidi F, Nelepcu I, Moeen A, Truong S, Dagil R, Choudhary S, Gustavsson T, Zhai B, Heitzender S, Salanti A, Sorensen PH, Daugaard M. Transient CAR T cells with specificity to oncofetal glycosaminoglycans in solid tumors. EMBO Mol Med 2024; 16:2775-2794. [PMID: 39406935 PMCID: PMC11554890 DOI: 10.1038/s44321-024-00153-8] [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: 05/31/2024] [Revised: 09/24/2024] [Accepted: 10/01/2024] [Indexed: 11/13/2024] Open
Abstract
Glycosaminoglycans are often deprioritized as targets for synthetic immunotherapy due to the complexity of glyco-epitopes and limited options for obtaining specific subtype binding. Solid tumors express proteoglycans that are modified with oncofetal chondroitin sulfate (CS), a modification normally restricted to the placenta. Here, we report the design and functionality of transient chimeric antigen receptor (CAR) T cells with selectivity to oncofetal CS. Following expression in T cells, the CAR could be "armed" with recombinant VAR2CSA lectins (rVAR2) to target tumor cells expressing oncofetal CS. While unarmed CAR T cells remained inactive in the presence of target cells, VAR2-armed CAR T cells displayed robust activation and the ability to eliminate diverse tumor cell types in vitro. Cytotoxicity of the CAR T cells was proportional to the concentration of rVAR2 available to the CAR, offering a potential molecular handle to finetune CAR T cell activity. In vivo, armed CAR T cells rapidly targeted bladder tumors and increased the survival of tumor-bearing mice. Thus, our work indicates that cancer-restricted glycosaminoglycans may be exploited as potential targets for CAR T cell therapy.
Collapse
Affiliation(s)
- Nastaran Khazamipour
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
| | - Htoo Zarni Oo
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
- Molecular Pathology & Cell Imaging Laboratory, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
| | - Nader Al-Nakouzi
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
| | - Mona Marzban
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
| | - Nasrin Khazamipour
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
| | - Morgan E Roberts
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
| | - Negin Farivar
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
| | - Igor Moskalev
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
| | - Joey Lo
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
| | - Fariba Ghaidi
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
| | - Irina Nelepcu
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
| | - Alireza Moeen
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
| | - Sarah Truong
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
| | - Robert Dagil
- Centre for Translational Medicine and Parasitology at Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Swati Choudhary
- Centre for Translational Medicine and Parasitology at Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Tobias Gustavsson
- Centre for Translational Medicine and Parasitology at Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Beibei Zhai
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
| | - Sabine Heitzender
- Division of Pediatric Hematology/Oncology/Stem Cell Transplantation and Regenerative Medicine, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Ali Salanti
- Centre for Translational Medicine and Parasitology at Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Poul H Sorensen
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Mads Daugaard
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada.
- Molecular Pathology & Cell Imaging Laboratory, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada.
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark.
| |
Collapse
|
47
|
Sorrentino C, Ciummo SL, Fieni C, Di Carlo E. Nanomedicine for cancer patient-centered care. MedComm (Beijing) 2024; 5:e767. [PMID: 39434967 PMCID: PMC11491554 DOI: 10.1002/mco2.767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/13/2024] [Accepted: 09/13/2024] [Indexed: 10/23/2024] Open
Abstract
Cancer is a leading cause of morbidity and mortality worldwide, and an increase in incidence is estimated in the next future, due to population aging, which requires the development of highly tolerable and low-toxicity cancer treatment strategies. The use of nanotechnology to tailor treatments according to the genetic and immunophenotypic characteristics of a patient's tumor, and to allow its targeted release, can meet this need, improving the efficacy of treatment and minimizing side effects. Nanomedicine-based approach for the diagnosis and treatment of cancer is a rapidly evolving field. Several nanoformulations are currently in clinical trials, and some have been approved and marketed. However, their large-scale production and use are still hindered by an in-depth debate involving ethics, intellectual property, safety and health concerns, technical issues, and costs. Here, we survey the key approaches, with specific reference to organ-on chip technology, and cutting-edge tools, such as CRISPR/Cas9 genome editing, through which nanosystems can meet the needs for personalized diagnostics and therapy in cancer patients. An update is provided on the nanopharmaceuticals approved and marketed for cancer therapy and those currently undergoing clinical trials. Finally, we discuss the emerging avenues in the field and the challenges to be overcome for the transfer of nano-based precision oncology into clinical daily life.
Collapse
Affiliation(s)
- Carlo Sorrentino
- Department of Medicine and Sciences of Aging“G. d'Annunzio” University” of Chieti‐PescaraChietiItaly
- Anatomic Pathology and Immuno‐Oncology Unit, Center for Advanced Studies and Technology (CAST)“G. d'Annunzio” University of Chieti‐PescaraChietiItaly
| | - Stefania Livia Ciummo
- Department of Medicine and Sciences of Aging“G. d'Annunzio” University” of Chieti‐PescaraChietiItaly
- Anatomic Pathology and Immuno‐Oncology Unit, Center for Advanced Studies and Technology (CAST)“G. d'Annunzio” University of Chieti‐PescaraChietiItaly
| | - Cristiano Fieni
- Department of Medicine and Sciences of Aging“G. d'Annunzio” University” of Chieti‐PescaraChietiItaly
- Anatomic Pathology and Immuno‐Oncology Unit, Center for Advanced Studies and Technology (CAST)“G. d'Annunzio” University of Chieti‐PescaraChietiItaly
| | - Emma Di Carlo
- Department of Medicine and Sciences of Aging“G. d'Annunzio” University” of Chieti‐PescaraChietiItaly
- Anatomic Pathology and Immuno‐Oncology Unit, Center for Advanced Studies and Technology (CAST)“G. d'Annunzio” University of Chieti‐PescaraChietiItaly
| |
Collapse
|
48
|
Zhang S, Huang Q, Ji T, Li Q, Hu C. Copper homeostasis and copper-induced cell death in tumor immunity: implications for therapeutic strategies in cancer immunotherapy. Biomark Res 2024; 12:130. [PMID: 39482784 PMCID: PMC11529036 DOI: 10.1186/s40364-024-00677-8] [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: 07/10/2024] [Accepted: 10/23/2024] [Indexed: 11/03/2024] Open
Abstract
Copper is an important trace element for maintaining key biological functions such as cellular respiration, nerve conduction, and antioxidant defense. Maintaining copper homeostasis is critical for human health, and its imbalance has been linked to various diseases, especially cancer. Cuproptosis, a novel mechanism of copper-induced cell death, provides new therapeutic opportunities for metal ion regulation to interact with cell fate. This review provides insights into the complex mechanisms of copper metabolism, the molecular basis of cuproptosis, and its association with cancer development. We assess the role of cuproptosis-related genes (CRGs) associated with tumorigenesis, their importance as prognostic indicators and therapeutic targets, and the impact of copper homeostasis on the tumor microenvironment (TME) and immune response. Ultimately, this review highlights the complex interplay between copper, cuproptosis, and cancer immunotherapy.
Collapse
Affiliation(s)
- Suhang Zhang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430030, China
| | - Qibo Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tuo Ji
- School of Medicine, New York Medical College, Valhalla, NY, 10595, USA
| | - Qilin Li
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430030, China.
| | - Chuanyu Hu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430030, China.
| |
Collapse
|
49
|
Zhu X, Xue J, Jiang H, Xue D. CAR-NK cells for gastrointestinal cancer immunotherapy: from bench to bedside. Mol Cancer 2024; 23:237. [PMID: 39443938 PMCID: PMC11515662 DOI: 10.1186/s12943-024-02151-3] [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: 07/14/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Gastrointestinal (GI) cancers represent a significant health burden worldwide. Their incidence continues to increase, and their management remains a clinical challenge. Chimeric antigen receptor (CAR) natural killer (NK) cells have emerged as a promising alternative to CAR-T cells for immunotherapy of GI cancers. Notably, CAR-NK cells offer several advantages, including reduced risk of graft-versus-host disease, lower cytokine release syndrome, and the ability to target cancer cells through both CAR-dependent and natural cytotoxic mechanisms. MAIN BODY This review comprehensively discusses the development and applications of CAR-NK cells in the treatment of GI cancers. We explored various sources of NK cells, CAR design strategies, and the current state of CAR-NK cell therapy for GI cancers, highlighting recent preclinical and clinical trials. Additionally, we addressed existing challenges and propose potential strategies to enhance the efficacy and safety of CAR-NK cell therapy. CONCLUSIONS Our findings highlight the potential of CAR-NK cells to revolutionize GI cancer treatment and pave the way for future clinical applications.
Collapse
Affiliation(s)
- Xingwang Zhu
- Department of Urinary Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, 110032, P.R. China
| | - Jieyun Xue
- China Medical University, Shenyang, Liaoning Province, 110000, P.R. China
| | - Hongzhou Jiang
- Department of Neurosurgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, 110032, P.R. China
| | - Dongwei Xue
- Department of Urinary Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, 110032, P.R. China.
| |
Collapse
|
50
|
Bangolo A, Amoozgar B, Zhang L, Nagesh VK, Sekhon I, Weissman S, Vesole D, Phull P, Donato M, Biran N, Siegel D, Parmar H. Impact of Allogeneic Stem Cell Transplant on Safety and Outcomes of Chimeric Antigen Receptor T Cell (CAR-T) Therapy in Patients with Multiple Myeloma (MM). J Clin Med 2024; 13:6207. [PMID: 39458157 PMCID: PMC11508289 DOI: 10.3390/jcm13206207] [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: 09/05/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Allogeneic stem cell transplantation (allo-SCT) has seen limited use in treating multiple myeloma (MM), despite its potential to offer long-term survival or even cure through the graft-versus-myeloma effect. Its limited application is largely due to concerns over serious complications like infections and graft-versus-host disease (GVHD). The possibility of GVHD exacerbation when CAR-T cells are administered to patients previously treated with allo-SCT remains a topic of concern. Ciltacabtagene autoleucel (Cilta-cel) and idecabtagene vicleucel (Ide-cel) are CAR-T therapies that have been FDA-approved for relapsed/refractory (R/R) MM. A recent study using data from the CARTITUDE-1 trial has shown promising safety and efficacy of Cilta-Cel in patients with a prior history of allo-SCT. This report outlines our real-world experience with CAR-T treatment in such patients. The objective of this study is to assess the safety and effectiveness of CAR-T therapy in R/R MM patients who have previously undergone allo-SCT. Methods: We conducted a retrospective analysis of adult patients (18-70 years old) with R/R MM treated with CAR-T therapy as part of an institutional IRB-approved protocol. Data were collected on safety and efficacy outcomes from the institution's records. Adverse events (AEs) were evaluated using the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE) version 5.0. Cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) were graded based on American Society for Transplantation and Cellular Therapy (ASTCT) criteria. Efficacy metrics included overall response rate (ORR) and progression-free survival (PFS), analyzed through the Kaplan-Meier method, with PFS defined as the time from CAR-T initiation to disease progression or death. Results: Of the 56 patients treated with CAR-T therapy, 8 (14.3%) had previously undergone allo-SCT. These patients had a median of seven prior therapy lines (LOTs), compared to five LOTs in the non-allo-SCT group (p = 0.04). CAR-T infusion occurred a median of 98.8 months after allo-SCT, with a range from 57.9 months to 178.5 months. CRS occurred in 87.5% of the allo-SCT group versus 77.1% in the non-allo-SCT group (p = 0.48). One patient in the allo-SCT group developed hemophagocytic lymphohistiocytosis (HLH), requiring anakinra. At a median follow-up of 4.8 months, the ORR was 87.5% in the allo-SCT group versus 75% in the non-allo-SCT group (p = 0.4). Median PFS had not been reached for the allo-SCT group at the time of analysis compared to 11.9 months in the non-allo-SCT group (p = 0.5). No treatment-related mortality or acute GVHD was noted in the allo-SCT cohort. Conclusions: The study suggests that prior allo-SCT does not adversely affect the safety or efficacy of CAR-T therapy in patients with R/R MM. These findings highlight the need for further investigations with larger patient samples and longer follow-up to better understand the interaction between allo-SCT and CAR-T therapy.
Collapse
Affiliation(s)
- Ayrton Bangolo
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | - Behzad Amoozgar
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | - Lili Zhang
- Department of Hematology and Oncology, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | - Vignesh K. Nagesh
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, USA
| | - Imranjot Sekhon
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, USA
| | - Simcha Weissman
- Department of Internal Medicine, Palisades Medical Center, North Bergen, NJ 07047, USA
| | - David Vesole
- Division of Myeloma, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | - Pooja Phull
- Division of Myeloma, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | - Michele Donato
- Division of Bone Marrow Transplant and Cellular Therapy, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | - Noa Biran
- Division of Myeloma, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | - David Siegel
- Division of Myeloma, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | - Harsh Parmar
- Division of Myeloma, John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| |
Collapse
|