1
|
Zhang W, Yuan S, Zhang Z, Fu S, Liu S, Liu J, Ma Q, Xia Z, Gu P, Gao S, Zhang Z, Zhang X, Liu Y, Zhang N. Regulating tumor cells to awaken T cell antitumor function and enhance melanoma immunotherapy. Biomaterials 2025; 316:123034. [PMID: 39709849 DOI: 10.1016/j.biomaterials.2024.123034] [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: 08/24/2024] [Revised: 11/26/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024]
Abstract
Tumor cells transmit various immunosuppressive signals and induce a dysfunctional state in T cells, which essentially leads to immune escape and tumor progression. However, developing effective strategies to counteract the domestication of T cells by tumor cells remains a challenge. Here, we prepared pH-responsive lipid nanoparticles (NL/PLDs) co-loaded with PCSK9 shRNA, lonidamine (LND), and low-dose doxorubicin (DOX). NL/PLDs can awaken domesticated T cells function by sending pro-activation, pro-recognition, and pro-killing signals by increasing tumor immunogenicity, increasing the expression of major histocompatibility complex I (MHC-I) on tumor cells, and alleviating the suppression effect of tumor-secreted lactic acid (LA) on the T cell effector function, respectively. In melanoma-bearing mice, NL/PLDs effectively relieved tumor immunosuppressive microenvironment (TIME) and enhanced the antitumor immunity mediated by CD8+ T cells. Furthermore, when combined with aPD-1, NL/PLDs demonstrated strong antitumor effects and increased immunotherapeutic efficacy. This regulatory strategy provides new insights for enhancing immunotherapy by regulating tumor immunosuppressive signals and shows significant potential for clinical tumor treatment.
Collapse
Affiliation(s)
- Weihan Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Shijun Yuan
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Zipeng Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China
| | - Shunli Fu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Shujun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Jinhu Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Qingping Ma
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Zhenxing Xia
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Panpan Gu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Shuying Gao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Zhiyue Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Xinke Zhang
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| |
Collapse
|
2
|
Harthorn A, Kuo TH, Torres SW, Lobb RR, Hackel BJ. Expression-Dependent Tumor Pretargeting via Engineered Avidity. Mol Pharm 2025; 22:558-572. [PMID: 39704255 DOI: 10.1021/acs.molpharmaceut.4c01177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Selective delivery of therapeutic modalities to tumor cells via binding of tumor-selective cell-surface biomarkers has empowered substantial advances in cancer treatment. Yet, tumor cells generally lack a truly specific biomarker that is present in high density on tumor tissue while being completely absent from healthy tissue. Rather, low but nonzero expression in healthy tissues results in on-target, off-tumor activity with detrimental side effects that constrain the therapeutic window or prevent use altogether. Advanced technologies to enhance the selectivity for tumor targeting are sorely needed. We have engineered a binding platform that is quantitatively dependent upon expression levels, via avidity-driven specificity, rather than binarily reliant on the presence or absence of a biomarker. We systematically varied monomeric binding affinity by engineering affibodies to target carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) and folate receptor 1 (FolR1). Two identical affibody ligands were tethered, with varying polypeptide linker lengths, to a nanobody that binds Alfa peptide to create a bispecific, trivalent protein for use in pretargeted radioligand therapy. Expression-dependent targeting was achieved in both systems: with 110 nM monomeric affinity to CEACAM5 with a two-amino-acid linker or with 250 nM monomeric affinity for FolR1 and a 10 amino acid linker. The latter bispecific, trivalent achieved over 25-fold differentiation between FolR1high and FolR1low cells in a mixed culture. Similar selectivity was achieved in a size-efficient bivalent molecule lacking a central nanobody. Moreover, the avid bivalent affibody molecule exhibited minimal inhibition by soluble antigen, whereas high-affinity bivalent antibody was inhibited by 97 ± 2%, which is indicative of serum inhibition of shed antigen. This work advances design principles for achieving expression-dependent tumor targeting via low-affinity, high-avidity ligands.
Collapse
Affiliation(s)
- Abbigael Harthorn
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Tse-Han Kuo
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Sarah W Torres
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Roy R Lobb
- MRB Biotherapeutics, Newton Center, Massachusetts 02459, United States
| | - Benjamin J Hackel
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States
- MRB Biotherapeutics, Newton Center, Massachusetts 02459, United States
| |
Collapse
|
3
|
Buhlmann A, Rom E, Schweiger G, Schneidawind D, David S. Extracorporeal cytokine adsorption as therapeutic option for immune effector cell-associated neurotoxicity syndrome. Neurol Sci 2025; 46:479-481. [PMID: 39417926 PMCID: PMC11698748 DOI: 10.1007/s10072-024-07812-1] [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: 07/10/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
With the rising number of patients receiving chimeric antigen receptor T-cells, the treatment of this therapy's complications is of growing concern to intensivists and neurologists. We used extracorporeal cytokine adsorption as an add-on therapy in a patient suffering from immune effector cell-associated neurotoxicity syndrome. Interleukin-6 level, which as a readily available parameter is generally used to evaluate course of disease, was rapidly reduced using this method. The patient made a full recovery and is still in hematological remission.
Collapse
Affiliation(s)
- Alix Buhlmann
- Institute of Intensive Care Medicine, University Hospital Zurich, Rämistrasse 100, Zurich, 8091, Switzerland
| | - Emanuel Rom
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Giovanna Schweiger
- Institute of Anesthesiology, University Hospital Zurich, Zurich, Switzerland
| | - Dominik Schneidawind
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Sascha David
- Institute of Intensive Care Medicine, University Hospital Zurich, Rämistrasse 100, Zurich, 8091, Switzerland.
| |
Collapse
|
4
|
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
|
5
|
Avouac J, Scherlinger M. CAR T-Cell Therapy for Rheumatic Diseases: What Does the Future Hold? BioDrugs 2024:10.1007/s40259-024-00692-z. [PMID: 39738985 DOI: 10.1007/s40259-024-00692-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2024] [Indexed: 01/02/2025]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy, initially successful in treating hematological malignancies, is emerging as a potential treatment for autoimmune diseases, including rheumatic conditions. CAR T cells, engineered to target and eliminate autoreactive B cells, offer a novel approach to managing diseases like systemic lupus erythematosus (SLE), systemic sclerosis (SSc), and inflammatory myopathies, where B cells play a pivotal role in disease pathology. Early case reports have demonstrated promising results, with patients achieving significant disease remission, normalization of serological markers, and the ability to discontinue traditional immunosuppressive therapies, which supported the initiation of several clinical trials. However, the application of CAR T-cell therapy in chronic inflammatory rheumatic disorders poses unique challenges, including patient heterogeneity, the risk of adverse effects such as cytokine release syndrome, and the high costs associated with the therapy. Despite these challenges, the potential for CAR T cells to provide long-term remission or even a cure in refractory autoimmune diseases is significant. Ongoing research aims to optimize CAR T-cell constructs and improve safety profiles, paving the way for broader application in rheumatic diseases. If these challenges can be addressed, CAR T-cell therapy could revolutionize the treatment landscape for chronic inflammatory rheumatic disorders, offering new hope for patients with severe, treatment-resistant conditions.
Collapse
Affiliation(s)
- Jérôme Avouac
- Service de Rhumatologie, Hôpital Cochin, AP-HP, CUP, Université Paris Descartes, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France.
- INSERM U1016 and UMR8104, Institut Cochin, 75014, Paris, France.
| | - Marc Scherlinger
- Rheumatology Department, Strasbourg University Hospital, 1 Avenue Molière, 67000, Strasbourg, France
- UMR_S INSERM 1109, Immunorhumatologie moléculaire, 1 place de l'hôpital, 67000, Strasbourg, France
| |
Collapse
|
6
|
Munir M, Sayed A, Addison D, Epperla N. Cardiovascular toxicities associated with novel cellular immune therapies. Blood Adv 2024; 8:6282-6296. [PMID: 39418640 PMCID: PMC11698921 DOI: 10.1182/bloodadvances.2024013849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/26/2024] [Accepted: 10/13/2024] [Indexed: 10/19/2024] Open
Abstract
ABSTRACT Over the past decade, T-cell-directed therapies, including chimeric antigen receptor T-cell (CAR-T) and bispecific T-cell engager (BTE) therapies, have reshaped the treatment of an expanding number of hematologic malignancies, whereas tumor-infiltrating lymphocytes, a recently approved cellular therapy, targets solid tumor malignancies. Emerging data suggest that these therapies may be associated with a high incidence of serious cardiovascular toxicities, including atrial fibrillation, heart failure, ventricular arrhythmias, and other cardiovascular toxicities. The development of these events is a major limitation to long-term survival after these treatments. This review examines the current state of evidence, including reported incidence rates, risk factors, mechanisms, and management strategies of cardiovascular toxicities after treatment with these novel therapies. We specifically focus on CAR-T and BTE therapies and their relation to arrhythmia, heart failure, myocarditis, bleeding, and other major cardiovascular events. Beyond the relationship between cytokine release syndrome and cardiotoxicity, we describe other potential mechanisms and highlight key unanswered questions and future directions of research.
Collapse
Affiliation(s)
- Malak Munir
- Department of Medicine, Ain Shams University Faculty of Medicine, Cairo, Egypt
| | - Ahmed Sayed
- Department of Medicine, Ain Shams University Faculty of Medicine, Cairo, Egypt
- Department of Cardiology, Houston Methodist DeBakey Heart & Vascular Center, Houston, TX
| | - Daniel Addison
- Division of Cancer Prevention and Control, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH
- Division of Epidemiology, College of Public Health, The Ohio State University, Columbus, OH
| | - Narendranath Epperla
- Division of Hematology and Hematologic Malignancies, Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| |
Collapse
|
7
|
Machowicz R. How to make transplanted stem cells feel at home. Blood 2024; 144:2564-2566. [PMID: 39699920 DOI: 10.1182/blood.2024026611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024] Open
|
8
|
Bertin L, Crepaldi M, Zanconato M, Lorenzon G, Maniero D, de Barba C, Bonazzi E, Facchin S, Scarpa M, Ruffolo C, Angriman I, Buda A, Zingone F, Barberio B, Savarino EV. Advancing therapeutic frontiers: a pipeline of novel drugs for luminal and perianal Crohn's disease management. Therap Adv Gastroenterol 2024; 17:17562848241303651. [PMID: 39711916 PMCID: PMC11660281 DOI: 10.1177/17562848241303651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 11/12/2024] [Indexed: 12/24/2024] Open
Abstract
Crohn's disease (CD) is a chronic, complex inflammatory disorder of the gastrointestinal tract that presents significant therapeutic challenges. Despite the availability of a wide range of treatments, many patients experience primary non-response, secondary loss of response, or adverse events, limiting the overall effectiveness of current therapies. Clinical trials often report response rates below 60%, partly due to stringent inclusion criteria. Emerging therapies that target novel pathways offer promise in overcoming these limitations. This review explores the latest investigational drugs in phases I, II, and III clinical trials for treating both luminal and perianal CD. We highlight promising therapies that target known mechanisms, including selective Janus kinase inhibitors, anti-adhesion molecules, tumor necrosis factor inhibitors, and IL-23 selective inhibitors. In addition, we delve into novel therapeutic strategies such as sphingosine-1-phosphate receptor modulators, miR-124 upregulators, anti-fractalkine (CX3CL1), anti-TL1A, peroxisome proliferator-activated receptor gamma agonists, TGFBRI/ALK5 inhibitors, anti-CCR9 agents, and other innovative small molecules, as well as combination therapies. These emerging approaches, by addressing new pathways and mechanisms of action, have the potential to surpass the limitations of existing treatments and significantly improve CD management. However, the path to developing new therapies for inflammatory bowel disease (IBD) is fraught with challenges, including complex trial designs, ethical concerns regarding placebo use, recruitment difficulties, and escalating costs. The landscape of IBD clinical trials is shifting toward greater inclusivity, improved patient diversity, and innovative trial designs, such as adaptive and Bayesian approaches, to address these challenges. By overcoming these obstacles, the drug development pipeline can advance more effective, accessible, and timely treatments for CD.
Collapse
Affiliation(s)
- Luisa Bertin
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Martina Crepaldi
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Miriana Zanconato
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Greta Lorenzon
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Daria Maniero
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Caterina de Barba
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Erica Bonazzi
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Sonia Facchin
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Marco Scarpa
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padua, Italy
| | - Cesare Ruffolo
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padua, Italy
| | - Imerio Angriman
- Chirurgia Generale 3 Unit, Azienda Ospedale Università di Padova, Padua, Italy
| | - Andrea Buda
- Gastroenterology Unit, Department of Oncological Gastrointestinal Surgery, Santa Maria del Prato Hospital, Feltre, Italy
| | - Fabiana Zingone
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Brigida Barberio
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Edoardo Vincenzo Savarino
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani, 2, Padua 35128, Italy
| |
Collapse
|
9
|
Zeng L, Li Y, Xiang W, Xiao W, Long Z, Sun L. Advances in chimeric antigen receptor T cell therapy for autoimmune and autoinflammatory diseases and their complications. J Autoimmun 2024; 150:103350. [PMID: 39700677 DOI: 10.1016/j.jaut.2024.103350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 11/27/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Chimeric antigen receptor T (CAR-T) cells are genetically engineered T cells expressing transmembrane chimeric antigen receptors with specific targeting abilities. As an emerging immunotherapy, the use of CAR-T cells has made significant breakthroughs in cancer treatment, particularly for hematological malignancies. The success of CAR-T cell therapy in blood cancers highlights its potential for other conditions in which the clearance of pathological cells is therapeutic, such as liver diseases, infectious diseases, heart failure, and diabetes. Given the limitations of current therapies for autoimmune diseases, researchers have actively explored the potential therapeutic value of CAR-T cells and their derivatives in the field of autoimmune diseases. This review focuses on the research progress and current challenges of CAR-T cells in autoimmune diseases with the aim of providing a theoretical basis for the precise treatment of autoimmune diseases. In the future, CAR-T cells may present new therapeutic modalities and ultimately provide hope for patients with autoimmune diseases.
Collapse
Affiliation(s)
- Liuting Zeng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China.
| | - Yan Li
- People's Hospital of Ningxiang City, Ningxiang City, China
| | - Wang Xiang
- Department of Rheumatology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City), Changde City, China
| | - Wei Xiao
- Department of Rheumatology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City), Changde City, China.
| | - Zhiyong Long
- Department of Physical Medicine and Rehabilitation, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China; Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
10
|
Guo Q, Li J, Wang J, Li L, Wei J, Zhang L. The advent of chimeric antigen receptor T Cell therapy in recalibrating immune balance for rheumatic autoimmune disease treatment. Front Pharmacol 2024; 15:1502298. [PMID: 39734406 PMCID: PMC11672202 DOI: 10.3389/fphar.2024.1502298] [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: 09/26/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024] Open
Abstract
CAR-T cell therapy, a cutting-edge cellular immunotherapy with demonstrated efficacy in treating hematologic malignancies, also exhibits significant promise for addressing autoimmune diseases. This innovative therapeutic approach holds promise for achieving long-term remission in autoimmune diseases, potentially offering significant benefits to affected patients. Current targets under investigation for the treatment of these conditions include CD19, CD20, and BCMA, among others. However, CAR-T therapy faces difficulties such as time-consuming cell manufacturing, complex and expensive process, and the possibility of severe adverse reactions complicating the treatment, etc. This article examines CAR-T therapy across various rheumatic autoimmune diseases, including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), Sjögren's syndrome (SS), systemic sclerosis (SSc), antisynthetase syndrome (ASS), and ANCA-associated vasculitis (AAV), highlighting both therapeutic advancements and ongoing challenges.
Collapse
Affiliation(s)
- Qianyu Guo
- Department of Rheumatology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Jie Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Juanjuan Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Linxin Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Jia Wei
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
11
|
Karamivandishi A, Hatami A, Eslami MM, Soleimani M, Izadi N. Chimeric antigen receptor natural killer cell therapy: A systematic review of preclinical studies for hematologic and solid malignancies. Hum Immunol 2024; 86:111207. [PMID: 39667204 DOI: 10.1016/j.humimm.2024.111207] [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: 09/02/2024] [Revised: 11/08/2024] [Accepted: 11/24/2024] [Indexed: 12/14/2024]
Abstract
Advancements in the field of CAR-T therapy have brought about a revolution in the treatment of numerous types of cancer in the past ten years. However, despite the remarkable success achieved thus far, certain barriers impede the widespread implementation of this therapy such as intricate manufacturing processes and treatment-associated toxicities. As an alternative, chimeric antigen receptor-engineered natural killer cell (CAR-NK) therapy presents a viable opportunity for a simpler and more cost-effective "off-the-shelf" treatment option, which is likely to result in fewer adverse reactions. A total of 71 studies were included in this review. Eligible studies were searched and reviewed from the databases of PubMed, Web of Science and Scopus. Based on data extracted from articles, we concluded that CAR-NK cell efficiency can vary considerably depending on factors such as tumor model, dosage, CAR generation and expansion method. Furthermore, investigating consequences of utilizing various constructs and generations of CAR-NK cells on their anti-tumor activity examined in this review.
Collapse
Affiliation(s)
- Arezoo Karamivandishi
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Hatami
- Hematology and Cell Therapy Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Masoud Eslami
- Hematology and Cell Therapy Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Masoud Soleimani
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Hematology and Cell Therapy Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Neda Izadi
- Research Center for Social Determinants of Health,Research institute for metabolic and obesity disorders, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Kegyes D, Moisoiu V, Constantinescu C, Tanase A, Ghiaur G, Einsele H, Tomuleasa C, Lazarus HM, Gale RP. Neuro-toxicities of chemo- and immune-therapies in haematologic malignancies: from mechanism to management. Blood Rev 2024:101254. [PMID: 39674687 DOI: 10.1016/j.blre.2024.101254] [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: 09/19/2024] [Revised: 11/28/2024] [Accepted: 12/07/2024] [Indexed: 12/16/2024]
Abstract
Chemo- and immune therapies administered to treat haematologic malignancies frequently cause neurologic injury. The adverse events range from mild cognitive impairment and headaches to severe conditions such as seizures, stroke and encephalitis. We performed a comprehensive literature review and report the types, mechanisms, management and prevention of neuro-toxicity resulting from these therapies in subjects who develop these toxic effects. Our paper will not discuss radiation therapy, as it has already been extensively reviewed by many authors. Our focus will be on recently developed anti-cancer drugs.
Collapse
Affiliation(s)
- David Kegyes
- Department of Hematology, Ion Chiricuta Cancer Center, Cluj-Napoca, Romania; Department of Hematology / Department of Personalized Medicine and Rare Diseases - Medfuture Institute for Biomedical Research, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Vlad Moisoiu
- Department of Hematology / Department of Personalized Medicine and Rare Diseases - Medfuture Institute for Biomedical Research, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania; Department of Neurology and Clinical Neuroscience Center, University Hospital Zurich, Zurich, Switzerland
| | - Catalin Constantinescu
- Department of Hematology, Ion Chiricuta Cancer Center, Cluj-Napoca, Romania; Department of Hematology / Department of Personalized Medicine and Rare Diseases - Medfuture Institute for Biomedical Research, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alina Tanase
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Gabriel Ghiaur
- Department of Hematology / Department of Personalized Medicine and Rare Diseases - Medfuture Institute for Biomedical Research, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania; Department of Leukemia, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Hermann Einsele
- Department of Hematology / Department of Personalized Medicine and Rare Diseases - Medfuture Institute for Biomedical Research, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania; Department of Internal Medicine, Julius Maximilians University of Wurzburg, Wurzburg, Germany
| | - Ciprian Tomuleasa
- Department of Hematology, Ion Chiricuta Cancer Center, Cluj-Napoca, Romania; Department of Hematology / Department of Personalized Medicine and Rare Diseases - Medfuture Institute for Biomedical Research, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| | - Hillard M Lazarus
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Robert Peter Gale
- Centre for Haematology, Imperial College of Science, Technology and Medicine, London, UK; Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
13
|
Fan M, Zheng J, Huang Y, Lu M, Shang Z, Du M. Nanoparticle-mediated universal CAR-T therapy. Int J Pharm 2024; 666:124779. [PMID: 39349228 DOI: 10.1016/j.ijpharm.2024.124779] [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: 05/22/2024] [Revised: 09/16/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024]
Abstract
In recent years, chimeric antigen receptor (CAR)-T cell therapy has been highly successful in treating hematological malignancies, leading to significant advancements in the cancer immunotherapy field. However, the typical CAR-T therapy necessitates the enrichment of patients' own leukocytes for ex vivo production of CAR-T cells, this customized pattern requires a complicated and time-consuming manufacturing procedure, making it costly and less accessible. The off-the-shelf universal CAR-T strategy could reduce manufacturing costs and realize timely drug administration, presenting as an ideal substitute for typical CAR-T therapy. Utilizing nanocarriers for targeted gene delivery is one of the approaches for the realization of universal CAR-T therapy, as biocompatible and versatile nanoparticles could deliver CAR genes to generate CAR-T cells in vivo. Nanoparticle-mediated in situ generation of CAR-T cells possesses multiple advantages, including lowered cost, simplified manufacturing procedure, and shortened administration time, this strategy is anticipated to provide a potentially cost-effective alternative to current autologous CAR-T cell manufacturing, thus facilitating the prevalence and improvement of CAR-T therapy.
Collapse
Affiliation(s)
- Mingliang Fan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Jiayu Zheng
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yue Huang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Mingxia Lu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Biomaterials and Tissue Engineering Research Center, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China.
| | - Zhi Shang
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Mingwei Du
- Department of Dermatology, Shanghai Key Laboratory of Medical Mycology, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China.
| |
Collapse
|
14
|
Ji Q, Dong Y, Zhang Y, Wu X, Bai Z, Huang S, Dong X, Wang Y, Pan J, Lu J, Hu S, Wu S. Earlier intrathecal dexamethasone effectively alleviate immune effector cell-associated neurotoxicity syndrome. Int Immunopharmacol 2024; 142:113214. [PMID: 39321707 DOI: 10.1016/j.intimp.2024.113214] [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: 05/11/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024]
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy is effective in treating relapsed/refractory B-cell acute lymphoblastic leukemia (R/R B-ALL). However, the side effects of immune effector cell-associated neurotoxicity syndrome (ICANS) remain a problem. The current frontline therapies for ICANS include steroids and supportive care. For the steroid-refractory and severe ICANS, several studies have reported excellent efficacy of intrathecal (IT) corticosteroids alone or in combination with chemotherapy. However, whether patients can benefit from IT dexamethasone (dex) before grade 3 or refractory ICANS remains unclear. In this study, the patients with ICANS (≥1) after CAR-T cell therapy were assigned to the IT group and non-IT group. Clinical information, laboratory parameters, and serum cytokine levels were analyzed. A significant and rapid reduction in inflammatory cytokines and biomarkers was observed after 24 h of IT dex treatment. With IT dex, 83.3 % (15/18) of patients recovered from neurotoxicity. Moreover, this option significantly shortens the recovery time of ICANS without affecting the efficacy of CAR-T cell therapy. Earlier initiation of IT dex is the optimal management of ICANS resulting from CAR-T cell therapy, but larger sample studies are needed to determine its efficacy in these settings.
Collapse
Affiliation(s)
- Qi Ji
- Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Yi Dong
- Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Yongping Zhang
- Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Xiaochen Wu
- Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Zhenjiang Bai
- Pediatric Intensive Care Unit, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Saihu Huang
- Pediatric Intensive Care Unit, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Xingqiang Dong
- Pediatric Intensive Care Unit, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Yuxuan Wang
- Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, No. 92 Zhongnan Street, SIP, Suzhou 215003, China
| | - Jun Lu
- Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China.
| | - Shaoyan Hu
- Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China.
| | - Shuiyan Wu
- Hematology & Oncology, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China; Pediatric Intensive Care Unit, Children's Hospital of Soochow University, Suzhou 215000, Jiangsu, China.
| |
Collapse
|
15
|
Yao P, Liu YG, Huang G, Hao L, Wang R. The development and application of chimeric antigen receptor natural killer (CAR-NK) cells for cancer therapy: current state, challenges and emerging therapeutic advances. Exp Hematol Oncol 2024; 13:118. [PMID: 39633491 PMCID: PMC11616395 DOI: 10.1186/s40164-024-00583-7] [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: 05/16/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024] Open
Abstract
Immunotherapy has transformed the landscape of cancer treatment, with chimeric antigen receptor (CAR)-engineered T (CAR-T) cell therapy emerging as a front runner in addressing some hematological malignancies. Despite its considerable efficacy, the occurrence of severe adverse effects associated with CAR-T cell therapy has limited their scope and prompted the exploration of alternative therapeutic strategies. Natural killer (NK) cells, characterized by both their innate cytotoxicity and ability to lyse target cells without the constraint of peptide specificity conferred by a major histocompatibility complex (MHC), have similarly garnered attention as a viable immunotherapy. As such, another therapeutic approach has recently emerged that seeks to combine the continued success of CAR-T cell therapy with the flexibility of NK cells. Clinical trials involving CAR-engineered NK (CAR-NK) cell therapy have exhibited promising efficacy with fewer deleterious side effects. This review aims to provide a concise overview of the cellular and molecular basis of NK cell biology, facilitating a better understanding of advancements in CAR design and manufacturing. The focus is on current approaches and strategies employed in CAR-NK cell development, exploring at both preclinical and clinical settings. We will reflect upon the achievements, advantages, and challenges intrinsic to CAR-NK cell therapy. Anticipating the maturation of CAR-NK cell therapy technology, we foresee its encouraging prospects for a broader range of cancer patients and other conditions. It is our belief that this CAR-NK progress will bring us closer to making significant strides in the treatment of refractory and recurrent cancers, as well as other immune-mediated disorders.
Collapse
Affiliation(s)
- Pin Yao
- Department of Health Management, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Ya-Guang Liu
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Gang Huang
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Liangchun Hao
- Department of Pediatrics, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Shenyang, 110004, Liaoning, China
| | - Runan Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
16
|
Bates SM, Evans KV, Delsing L, Wong R, Cornish G, Bahjat M. Immune safety challenges facing the preclinical assessment and clinical progression of cell therapies. Drug Discov Today 2024; 29:104239. [PMID: 39521331 DOI: 10.1016/j.drudis.2024.104239] [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: 04/08/2024] [Revised: 10/15/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
The promise of curative outcomes for life-limiting diseases using cell therapies is starting to become a reality, not only for patients with end-stage cancer, but also increasingly for regenerative therapies, including dentistry, ocular, neurodegenerative, and cardiac diseases. The introduction of often genetically modified cells into a patient can come with an extensive range of safety considerations. From an immune perspective, cell-based therapies carry inherent consequences and consideration of factors, such as the cell source (donor-derived autologous cells versus allogeneic cells), the intrinsic cellular nature of the therapy, and engineering/manufacturing methods, all of which influence the likelihood of inducing unwanted immune responses. Here, we provide an overview of the potential immune safety risks associated with cell therapies and explore possible mitigation approaches.
Collapse
Affiliation(s)
- Stephanie M Bates
- Safety Innovation, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Kelly V Evans
- Safety Innovation, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Louise Delsing
- Cell and Gene Therapy Safety, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ryan Wong
- Cell and Gene Therapy Safety, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Georgina Cornish
- Oncology Safety, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Mahnoush Bahjat
- Safety Innovation, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
17
|
Olivera I, Etxeberria I, Luri-Rey C, Molero-Glez P, Melero I. Regional and intratumoral adoptive T-cell therapy. IMMUNO-ONCOLOGY TECHNOLOGY 2024; 24:100715. [PMID: 39055165 PMCID: PMC11269935 DOI: 10.1016/j.iotech.2024.100715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Adoptive T-cell therapies (ACTs) including tumor-infiltrating lymphocytes and engineered T cells (transgenic T-cell receptor and chimeric antigen receptor T cells), have made an important impact in the field of cancer treatment over the past years. Most of these therapies are typically administered systemically in approaches that facilitate the elimination of hematologic malignancies. Therapeutical efficacy against solid tumors, however, with the exception of tumor-infiltrating lymphocytes against melanoma, remains limited due to several barriers preventing lymphocyte access to the tumor bed. Building upon the experience of regional administration in other immunotherapies, the regional administration of adoptive cell therapies is being assessed to overcome this challenge, granting a first round of access of the transferred T cells to the tumor niche and thereby ensuring their activation and expansion. Intralesional and intracavitary routes of delivery have been tested with promising antitumor objective responses in preclinical and clinical studies. Additionally, several strategies are being developed to further improve T-cell activity after reinfusing them back to the patient such as combinations with other immunotherapy agents or direct engineering of the transferred T cells, achieving long-term immune memory. Clinical trials testing different regional adoptive T-cell therapies are ongoing but some issues related to methodology of administration and correct selection of the target antigen to avoid on-target/off-tumor side-effects need to be further evaluated and improved. Herein, we discuss the current preclinical and clinical landscape of intratumoral and locoregional delivery of adoptive T-cell therapies.
Collapse
Affiliation(s)
- I. Olivera
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - I. Etxeberria
- Human Oncology and Pathogenesis Program (HOPP), Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York
- Parker Institute for Cancer Immunotherapy, New York, USA
| | - C. Luri-Rey
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - P. Molero-Glez
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - I. Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona
- Department of Oncology, Clínica Universidad de Navarra, Madrid
- Centro del Cancer de la Universidad de Navarra (CCUN), Pamplona, Spain
- Nuffield Department of Medicine (NDM), University of Oxford, Oxford, UK
| |
Collapse
|
18
|
Chen Q, Liu Y, Chen Q, Li M, Xu L, Lin B, Tan Y, Liu Z. DNA Nanostructures: Advancing Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2405231. [PMID: 39308253 DOI: 10.1002/smll.202405231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/28/2024] [Indexed: 12/06/2024]
Abstract
Cancer immunotherapy is a groundbreaking medical revolution and a paradigm shift from traditional cancer treatments, harnessing the power of the immune system to target and destroy cancer cells. In recent years, DNA nanostructures have emerged as prominent players in cancer immunotherapy, exhibiting immense potential due to their controllable structure, surface addressability, and biocompatibility. This review provides an overview of the various applications of DNA nanostructures, including scaffolded DNA, DNA hydrogels, tetrahedral DNA nanostructures, DNA origami, spherical nucleic acids, and other DNA-based nanostructures in cancer immunotherapy. These applications explore their roles in vaccine development, immune checkpoint blockade therapies, adoptive cellular therapies, and immune-combination therapies. Through rational design and optimization, DNA nanostructures significantly bolster the immunogenicity of the tumor microenvironment by facilitating antigen presentation, T-cell activation, tumor infiltration, and precise immune-mediated tumor killing. The integration of DNA nanostructures with cancer therapies ushers in a new era of cancer immunotherapy, offering renewed hope and strength in the battle against this formidable foe of human health.
Collapse
Affiliation(s)
- Qianqian Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan Province, 410083, P. R. China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan Province, 410083, P. R. China
| | - Qiwen Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan Province, 410083, P. R. China
| | - Mingfeng Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan Province, 410013, P. R. China
| | - Lishang Xu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan Province, 410013, P. R. China
| | - Bingyu Lin
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan Province, 410083, P. R. China
| | - Yifu Tan
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan Province, 410013, P. R. China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan Province, 410013, P. R. China
| |
Collapse
|
19
|
Pan D, Richter J. Management of Toxicities Associated with BCMA, GPRC5D, and FcRH5-Targeting Bispecific Antibodies in Multiple Myeloma. Curr Hematol Malig Rep 2024; 19:237-245. [PMID: 39145912 DOI: 10.1007/s11899-024-00740-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] [Accepted: 07/31/2024] [Indexed: 08/16/2024]
Abstract
PURPOSE OF REVIEW The introduction of bispecific antibodies is one of the most significant recent advances in the treatment of relapsed/refractory multiple myeloma. This review will summarize the management of the toxicities associated with newly approved T cell-engaging bispecific antibodies and those which may be approved in the near future. RECENT FINDINGS Numerous trials have shown that bispecific antibodies can be both effective and tolerable when adverse events are properly managed. Cytokine release syndrome and increased infections are observed across all bispecific antibodies. Additional adverse events are target-specific, such as the more severe hypogammaglobulinemia and infections of BCMA bispecific antibodies and the dysgeusia, nail dystrophy, and skin changes of GPRC5D bispecific antibodies. Bispecific antibodies will surely become a mainstay of multiple myeloma therapy given their efficacy and accessibility. Their unique toxicities must be carefully considered and managed to ensure they are utilized safely.
Collapse
Affiliation(s)
- Darren Pan
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Joshua Richter
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA.
| |
Collapse
|
20
|
D'Avanzo C, Blaeschke F, Lysandrou M, Ingelfinger F, Zeiser R. Advances in cell therapy: progress and challenges in hematological and solid tumors. Trends Pharmacol Sci 2024; 45:1119-1134. [PMID: 39603960 DOI: 10.1016/j.tips.2024.10.016] [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: 08/12/2024] [Revised: 10/20/2024] [Accepted: 10/20/2024] [Indexed: 11/29/2024]
Abstract
Cell-based therapies harness the endogenous ability of the immune system to fight cancer and have shown promising results in the treatment of hematological malignancies. However, their clinical application beyond B cell malignancies is hampered by numerous hurdles, ranging from relapsed disease to a hostile tumor microenvironment (TME). Recent advances in cell engineering and TME modulation may expand the applicability of these therapies to a wider range of cancers, creating new treatment possibilities. Breakthroughs in advanced gene editing and sophisticated cell engineering, have also provided promising solutions to longstanding challenges. In this review, we examine the challenges and future directions of the most prominent cell-based therapies, including chimeric antigen receptor (CAR)-T cells, tumor-infiltrating lymphocytes (TILs), and natural killer (NK) cells, and emerging modalities. We provide a comprehensive analysis of emerging cell types and combination strategies translated into clinical trials, offering insights into the next generation of cell-based cancer treatments.
Collapse
Affiliation(s)
- Claudia D'Avanzo
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Franziska Blaeschke
- German Cancer Research Center (DKFZ), Heidelberg, Germany; Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany; Heidelberg University Hospital, Heidelberg, Germany
| | - Memnon Lysandrou
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Florian Ingelfinger
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
21
|
Zheng X, Zhang S, Wu H, Xia J, Zheng K, Wang Y, Qin Y. Plasma exchange as an effective treatment for cytokine release syndrome following T cell receptor‑engineered T cell immunotherapy: A case report. Oncol Lett 2024; 28:607. [PMID: 39483965 PMCID: PMC11526442 DOI: 10.3892/ol.2024.14740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/16/2024] [Indexed: 11/03/2024] Open
Abstract
T-cell receptor-engineered T-cell (TCR-T) immunotherapy is a promising approach for the treatment of solid tumors. However, TCR-T therapy can result in severe cytokine release syndrome (CRS), thus limiting its therapeutic application. The present study reported the case of a patient with TCR-T-related CRS, which was treated successfully with plasma exchange (PE). A 35-year-old male patient, who was diagnosed with hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC) with lung metastases, was enrolled in a clinical trial for hepatitis B virus surface antigen-specific TCR-expressing autologous T-cell therapy for HBV-related HCC after failing multiple lines of targeted immunotherapy and local treatments. Therefore, TCR-Ts were infused after peripheral blood mononuclear cell collection, engineering and lymphodepletion chemotherapy. However, following engineered T-cell reinfusion, the patient developed a fever, hypotension, edema, multiple serous effusion and acute kidney injury, and was consequently diagnosed with grade 3 CRS and transferred to the Intensive Care Unit. The patient received three daily PE sessions (3,000 ml of fresh frozen plasma per session), renal replacement therapy, tocilizumab and 1,000 mg pulse methylprednisolone for 3 days. Following treatment, the patient's hemodynamic condition was stabilized and the C-reactive protein, ferritin and IL-6 levels were markedly reduced. During follow-up, a stable disease state was exhibited by the liver cancer and lung metastatic lesions. To the best of our knowledge, this is the first case reporting PE as a treatment approach for managing CRS following TCR-T therapy for solid tumors. The present study demonstrated that blood purification treatments, such as PE, which target inflammatory mediators and restore the balance between pro- and anti-inflammatory cytokines, could be a notable component in managing severe CRS associated with engineered T-cell treatment. However, additional clinical and translational studies are needed to further understand the mechanisms of T-cell immunotherapy to treat patients with solid tumors.
Collapse
Affiliation(s)
- Xixi Zheng
- Department of Nephrology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Shuo Zhang
- Department of Nephrology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Haiting Wu
- Department of Nephrology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Jinghua Xia
- Department of Nephrology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Ke Zheng
- Department of Nephrology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Ying Wang
- Department of Nephrology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Yan Qin
- Department of Nephrology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
22
|
Lei T, Wang Y, Zhang Y, Yang Y, Cao J, Huang J, Chen J, Chen H, Zhang J, Wang L, Xu X, Gale RP, Wang L. Leveraging CRISPR gene editing technology to optimize the efficacy, safety and accessibility of CAR T-cell therapy. Leukemia 2024; 38:2517-2543. [PMID: 39455854 PMCID: PMC11588664 DOI: 10.1038/s41375-024-02444-y] [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: 04/19/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024]
Abstract
Chimeric Antigen Receptor (CAR)-T-cell therapy has revolutionized cancer immune therapy. However, challenges remain including increasing efficacy, reducing adverse events and increasing accessibility. Use of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) technology can effectively perform various functions such as precise integration, multi-gene editing, and genome-wide functional regulation. Additionally, CRISPR screening using large-scale guide RNA (gRNA) genetic perturbation provides an unbiased approach to understanding mechanisms underlying anti-cancer efficacy of CAR T-cells. Several emerging CRISPR tools with high specificity, controllability and efficiency are useful to modify CAR T-cells and identify new targets. In this review we summarize potential uses of the CRISPR system to improve results of CAR T-cells therapy including optimizing efficacy and safety and, developing universal CAR T-cells. We discuss challenges facing CRISPR gene editing and propose solutions highlighting future research directions in CAR T-cell therapy.
Collapse
Affiliation(s)
- Tao Lei
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Yazhuo Wang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuchen Zhang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Yufei Yang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Jiaying Cao
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Jiansong Huang
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Jiali Chen
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Huajing Chen
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Jiayi Zhang
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510145, China
| | - Luzheng Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Robert Peter Gale
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College of Science, Technology and Medicine, London, UK.
| | - Liang Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
23
|
Bhagwat AS, Torres L, Shestova O, Shestov M, Mellors PW, Fisher HR, Farooki SN, Frost BF, Loken MR, Gaymon AL, Frazee D, Rogal W, Frey N, Hexner EO, Luger SM, Loren AW, Martin ME, McCurdy SR, Perl AE, Stadtmauer EA, Brogdon JL, Fraietta JA, Hwang WT, Siegel DL, Plesa G, Aplenc R, Porter DL, June CH, Gill SI. Cytokine-mediated CAR T therapy resistance in AML. Nat Med 2024; 30:3697-3708. [PMID: 39333315 DOI: 10.1038/s41591-024-03271-5] [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: 03/16/2024] [Accepted: 08/27/2024] [Indexed: 09/29/2024]
Abstract
Acute myeloid leukemia (AML) is a rapidly progressive malignancy without effective therapies for refractory disease. So far, chimeric antigen receptor (CAR) T cell therapy in AML has not recapitulated the efficacy seen in B cell malignancies. Here we report a pilot study of autologous anti-CD123 CAR T cells in 12 adults with relapsed or refractory AML. CAR T cells targeting CD123+ cells were successfully manufactured in 90.4% of runs. Cytokine release syndrome was observed in 10 of 12 infused individuals (83.3%, 90% confidence interval 0.5-0.97). Three individuals achieved clinical response (25%, 90% confidence interval 0.07-0.53). We found that myeloid-supporting cytokines are secreted during cell therapy and support AML blast survival via kinase signaling, leading to CAR T cell exhaustion. The prosurvival effect of therapy-induced cytokines presents a unique resistance mechanism in AML that is distinct from any observed in B cell malignancies. Our findings suggest that autologous CART manufacturing is feasible in AML, but treatment is associated with high rates of cytokine release syndrome and relatively poor clinical efficacy. Combining CAR T cell therapies with cytokine signaling inhibitors could enhance immunotherapy efficacy in AML and achieve improved outcomes (ClinicalTrials.gov identifier: NCT03766126 ).
Collapse
Affiliation(s)
- Anand S Bhagwat
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leonel Torres
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Immunology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Olga Shestova
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maksim Shestov
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick W Mellors
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Han R Fisher
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Saamia N Farooki
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Benjamin F Frost
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Avery L Gaymon
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Diane Frazee
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Walter Rogal
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Noelle Frey
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth O Hexner
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Selina M Luger
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alison W Loren
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Mary Ellen Martin
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Shannon R McCurdy
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander E Perl
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward A Stadtmauer
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Joseph A Fraietta
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei-Ting Hwang
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Don L Siegel
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gabriela Plesa
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard Aplenc
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - David L Porter
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Saar I Gill
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Division of Hematology-Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Cell Therapy and Transplant, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
24
|
Lotze MT, Olejniczak SH, Skokos D. CD28 co-stimulation: novel insights and applications in cancer immunotherapy. Nat Rev Immunol 2024; 24:878-895. [PMID: 39054343 PMCID: PMC11598642 DOI: 10.1038/s41577-024-01061-1] [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] [Accepted: 06/18/2024] [Indexed: 07/27/2024]
Abstract
Substantial progress in understanding T cell signalling, particularly with respect to T cell co-receptors such as the co-stimulatory receptor CD28, has been made in recent years. This knowledge has been instrumental in the development of innovative immunotherapies for patients with cancer, including immune checkpoint blockade antibodies, adoptive cell therapies, tumour-targeted immunostimulatory antibodies, and immunostimulatory small-molecule drugs that regulate T cell activation. Following the failed clinical trial of a CD28 superagonist antibody in 2006, targeted CD28 agonism has re-emerged as a technologically viable and clinically promising strategy for cancer immunotherapy. In this Review, we explore recent insights into the molecular functions and regulation of CD28. We describe how CD28 is central to the success of current cancer immunotherapies and examine how new questions arising from studies of CD28 as a clinical target have enhanced our understanding of its biological role and may guide the development of future therapeutic strategies in oncology.
Collapse
Affiliation(s)
- Michael T Lotze
- Department of Surgery, University of Pittsburgh Hillman Cancer Center, Pittsburgh, PA, USA.
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Scott H Olejniczak
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| | | |
Collapse
|
25
|
Wang Z, Han X, Sun G, Yu M, Qin J, Zhang Y, Ding D. Advances in cancer diagnosis and therapy by alginate-based multifunctional hydrogels: A review. Int J Biol Macromol 2024; 283:137707. [PMID: 39566758 DOI: 10.1016/j.ijbiomac.2024.137707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/30/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024]
Abstract
The field of oncology has been changed by the application of hydrogels. These 3D polymeric networks have demonstrated significant promise in the treatment of cancer and can boost the efficacy of conventional therapeutics including chemotherapy and immunotherapy. Noteworthy, the development of biocompatible and effective hydrogels has been of interest. In this case, alginate as a biopolymer and carbohydrate polymer has been used to modify or synthesis multifunctional nanoparticles for the treatment of human diseases, especially cancer. Therefore, highlighting the function of alginate in the development of hydrogels in cancer therapy can provide new insights for improving outcome and survival rate of patients. Alginate hydrogels improve the specific and selective delivery of cargo and therefore, they reduce the systemic toxicity of drugs, while they enhance anti-cancer activity. Alginate hydrogels protect the genes against degradation by enzymes and increase blood circulation time. The alginate hydrogels can respond to the specific stimuli in the tumor microenvironment including pH, redox and light to improve the site-specific release of cargo. The nanoparticles can be incorporated in the structure of alginate hydrogels to augment their anti-cancer activity. In addition, alginate hydrogels can accelerate immunotherapy and phototherapy through delivery of immunomodulators and photosensitizers, respectively.
Collapse
Affiliation(s)
- Ziwen Wang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xu Han
- Department of Emergency, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Guowei Sun
- Interventional Center, Fengcheng Central Hospital, Fengcheng 118199, China
| | - Miao Yu
- Department of Respiratory, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Juan Qin
- Department of Endocrinology and Metabolism, Shenyang Fourth People Hospital, Shenyang 110001, China
| | - Yuting Zhang
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Ding Ding
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
26
|
Schneider S, Aamodt WW, Pruitt AA, Berger JR. Parkinsonism Associated with Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor T-Cell Therapy. Mov Disord Clin Pract 2024; 11:1625-1628. [PMID: 39429213 DOI: 10.1002/mdc3.14239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/23/2024] [Accepted: 10/06/2024] [Indexed: 10/22/2024] Open
Affiliation(s)
- Sabine Schneider
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Whitley W Aamodt
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Amy A Pruitt
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joseph R Berger
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
27
|
Li Y, Xiao J, Li C, Yang M. Memory inflation: Beyond the acute phase of viral infection. Cell Prolif 2024; 57:e13705. [PMID: 38992867 PMCID: PMC11628752 DOI: 10.1111/cpr.13705] [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: 05/16/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 07/13/2024] Open
Abstract
Memory inflation is confirmed as the most commonly dysregulation of host immunity with antigen-independent manner in mammals after viral infection. By generating large numbers of effector/memory and terminal differentiated effector memory CD8+ T cells with diminished naïve subsets, memory inflation is believed to play critical roles in connecting the viral infection and the onset of multiple diseases. Here, we reviewed the current understanding of memory inflated CD8+ T cells in their distinct phenotypic features that different from exhausted subsets; the intrinsic and extrinsic roles in regulating the formation of memory inflation; and the key proteins in maintaining the expansion and proliferation of inflationary populations. More importantly, based on the evidences from both clinic and animal models, we summarized the potential mechanisms of memory inflation to trigger autoimmune neuropathies, such as Guillain-Barré syndrome and multiple sclerosis; the correlations of memory inflation between tumorigenesis and resistance of tumour immunotherapies; as well as the effects of memory inflation to facilitate vascular disease progression. To sum up, better understanding of memory inflation could provide us an opportunity to beyond the acute phase of viral infection, and shed a light on the long-term influences of CD8+ T cell heterogeneity in dampen host immune homeostasis.
Collapse
Affiliation(s)
- Yanfei Li
- School of Basic Medical SciencesChengdu University of Traditional Chinese MedicineChengduChina
| | - Jie Xiao
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Chen Li
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Mu Yang
- School of Basic Medical SciencesChengdu University of Traditional Chinese MedicineChengduChina
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| |
Collapse
|
28
|
Smith EC, Mott BT, Douglas E, Tatter SB, Watabe K. Immunotherapy for leptomeningeal disease from solid tumors: current clinical outcomes and future opportunities. Cancer Metastasis Rev 2024; 44:10. [PMID: 39612029 PMCID: PMC11607011 DOI: 10.1007/s10555-024-10235-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/22/2024] [Indexed: 11/30/2024]
Abstract
Leptomeningeal disease is a debilitating, late-stage form of metastatic cancer disseminated within the cerebrospinal fluid, subarachnoid space, and leptomeninges, leading to significant neurological morbidity and mortality. As systemic cancer treatments improve, rates of leptomeningeal disease have increased, yet prognosis remains exceedingly poor. A wide range of treatment modalities have been trialed; however, no standard of care has been established. Additionally, many clinical trials exclude patients with leptomeningeal disease, limiting available prospective data. In this review, we discuss the efficacy of immunotherapy for leptomeningeal disease from solid tumors including systemic and intrathecal therapies, as well as combined therapy regimens. Our review indicates a continued deficiency in the current prospective literature and highlights ongoing research regarding the leptomeningeal immune microenvironment, which will be critical in directing future study of leptomeningeal disease treatment. Currently, the efficacy of immunotherapies on leptomeningeal disease appears limited, and further prospective research is needed to draw significant conclusions. However, recent advancement in understanding the leptomeningeal microenvironment points to potential efficacy of novel immunotherapies targeting the innate immune system, and further study is warranted to evaluate the efficacy of these treatments in this subpopulation of patients.
Collapse
Affiliation(s)
- Eleanor C Smith
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA.
- Department of Neurological Surgery, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| | - Bryan T Mott
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
- Department of Neurological Surgery, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Emily Douglas
- Department of Internal Medicine, Section on Hematology and Oncology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Stephen B Tatter
- Department of Neurological Surgery, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Kounosuke Watabe
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| |
Collapse
|
29
|
Yang Y, Vedvyas Y, Alcaina Y, Trumper SJ, Babu DS, Min IM, Tremblay JM, Shoemaker CB, Jin MM. Affinity-tuned mesothelin CAR T cells demonstrate enhanced targeting specificity and reduced off-tumor toxicity. JCI Insight 2024; 9:e186268. [PMID: 39576012 PMCID: PMC11601908 DOI: 10.1172/jci.insight.186268] [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: 08/23/2024] [Accepted: 09/26/2024] [Indexed: 11/29/2024] Open
Abstract
The application of chimeric antigen receptor (CAR) T cell therapy in solid tumors is hindered by life-threatening toxicities resulting from on-target, off-tumor killing of nonmalignant cells that express low levels of the target antigen. Mesothelin (MSLN) has been identified as a target antigen for CAR T cell treatment of mesothelioma, lung, ovarian, and other cancers because of its high expression on tumor cells and limited expression on mesothelial cells. However, fatal off-tumor toxicity of high-affinity MSLN-targeting CAR T cells has been reported in multiple clinical trials. In this study, we constructed CARs using mutant variants of a single-domain nanobody that bind both human and mouse MSLN with a wide range of affinities and examined tumor responses and their toxicities from on-target, off-tumor interactions in mouse models. CAR T cells with low nanomolar affinity (equilibrium dissociation constant, KD) exhibited profound systemic expansion with no apparent infiltration into the tumor. With a gradual reduction of CAR affinity toward the micromolar KD, the expansion of CAR T cells became more restricted to tumors. Our preclinical studies demonstrated that high-affinity MSLN CARs were associated with fatal on-target, off-tumor toxicity and that affinity-tuned CARs rendered T cells more selective for MSLN-high tumors.
Collapse
Affiliation(s)
- Yanping Yang
- Department of Radiology, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Yogindra Vedvyas
- Department of Radiology, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Yago Alcaina
- Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Sydney J. Trumper
- Department of Radiology, Houston Methodist Research Institute, Houston, Texas, USA
| | - Diella S. Babu
- Department of Radiology, Houston Methodist Research Institute, Houston, Texas, USA
| | - Irene M. Min
- Department of Radiology, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Jacqueline M. Tremblay
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | - Charles B. Shoemaker
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | - Moonsoo M. Jin
- Department of Radiology, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
30
|
Li L, Jiang R, Yu JF, Li M. A Near-Infrared II Photo-Triggered Multifunctional Plasmonic Hyperthermia Immunomodulator for SERS-Guided Combination Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2409154. [PMID: 39564687 DOI: 10.1002/smll.202409154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Indexed: 11/21/2024]
Abstract
Immunotherapy represents a promising therapeutic strategy for cancer treatment, but its clinical applications are currently hindered by insufficient therapeutic potency, nonspecific delivery, and adverse side effects. Herein, a novel near-infrared II (NIR-II) photo-triggered plasmonic hyperthermia immunomodulator (RP@IR-pcNS@HA nanoparticles (NPs)) for anticancer treatment of both primary and distant cancers is reported. This immunomodulator comprises an IR-1061 dye-encoded NIR-II porous cubic AuAg nanoshell (pcNS) loaded with a Toll-like receptor 7 agonist - R837 in phase change materials (PCMs), further modified with hyaluronic acid (HA). In response to NIR-II photoirradiation, the RP@IR-pcNS@HA NPs controllably deliver and release R837 to tumor sites, subsequently perform plasmonic hyperthermia therapy for direct ablation of primary tumors, and elicit robust anticancer immune responses. It is demonstrated that upon NIR-II irradiation, such a plasmonic hyperthermia immunomodulator combined with anti-programmed death 1 antibody (αPD-1) completely eradicates both primary and distant cancers. In addition, this combination treatment successfully elicits robust immune memory responses for effective suppression of recurrence and distant metastasis of cancer. With the excellent NIR-II surface-enhanced Raman scattering (SERS) detection ability, the RP@IR-pcNS@HA NPs combined with αPD-1 represent an efficient way to develop high-performance theranostic agents for SERS-guided combination cancer photoimmunotherapy.
Collapse
Affiliation(s)
- Linhu Li
- School of Materials Science and Engineering, Central South University, Changsha, Hunan, 410083, China
| | - Renting Jiang
- School of Materials Science and Engineering, Central South University, Changsha, Hunan, 410083, China
| | - Jin-Feng Yu
- School of Materials Science and Engineering, Central South University, Changsha, Hunan, 410083, China
| | - Ming Li
- School of Materials Science and Engineering, Central South University, Changsha, Hunan, 410083, China
| |
Collapse
|
31
|
Vera-Cruz S, Jornet Culubret M, Konetzki V, Alb M, Friedel SR, Hudecek M, Einsele H, Danhof S, Scheller L. Cellular Therapies for Multiple Myeloma: Engineering Hope. Cancers (Basel) 2024; 16:3867. [PMID: 39594822 PMCID: PMC11592760 DOI: 10.3390/cancers16223867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Multiple myeloma (MM) treatment remains challenging due to its relapsed/refractory disease course as well as intra- and inter-patient heterogeneity. Cellular immunotherapies, especially chimeric antigen receptor (CAR)-T cells targeting B cell maturation antigen (BCMA), mark a major breakthrough, achieving long-lasting remissions and instilling hope for a potential cure. While ongoing clinical trials are increasingly driving approved cellular products towards earlier lines of therapy, novel targets as well as advanced approaches employing natural killer (NK) cells or dendritic cell (DC) vaccines are currently under investigation. Treatment resistance, driven by tumor-intrinsic factors such as antigen escape and the intricate dynamics of the tumor microenvironment (TME), along with emerging side effects such as movement and neurocognitive treatment-emergent adverse events (MNTs), are the major limitations of approved cellular therapies. To improve efficacy and overcome resistance, cutting-edge research is exploring strategies to target the microenvironment as well as synergistic combinatorial approaches. Recent advances in CAR-T cell production involve shortened manufacturing protocols and "off-the-shelf" CAR-T cells, aiming at decreasing socioeconomic barriers and thereby increasing patient access to this potential lifesaving therapy. In this review, we provide an extensive overview of the evolving field of cellular therapies for MM, underlining the potential to achieve long-lasting responses.
Collapse
Affiliation(s)
- Sarah Vera-Cruz
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Maria Jornet Culubret
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Verena Konetzki
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Miriam Alb
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Sabrina R. Friedel
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
- Fraunhofer-Institut für Zelltherapie und Immunologie (IZI), Außenstelle Zelluläre Immuntherapie, 97080 Würzburg, Germany
| | - Hermann Einsele
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Sophia Danhof
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
- Mildred Scheel Early Career Center, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Lukas Scheller
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
- Interdisziplinäres Zentrum für Klinische Forschung (IZKF), Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
32
|
Demaria O, Habif G, Vetizou M, Gauthier L, Remark R, Chiossone L, Vagne C, Rebuffet L, Courtois R, Denis C, Le Floch F, Muller M, Girard-Madoux M, Augier S, Lopez J, Carrette B, Maguer A, Vallier JB, Grondin G, Baron W, Galluso J, Yessaad N, Giordano M, Simon L, Chanuc F, Alvarez AB, Perrot I, Bonnafous C, Represa A, Rossi B, Morel A, Morel Y, Paturel C, Vivier E. A tetraspecific engager armed with a non-alpha IL-2 variant harnesses natural killer cells against B cell non-Hodgkin lymphoma. Sci Immunol 2024; 9:eadp3720. [PMID: 39546590 DOI: 10.1126/sciimmunol.adp3720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/07/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024]
Abstract
NK cells offer a promising alternative to T cell therapies in cancer. We evaluated IPH6501, a clinical-stage, tetraspecific NK cell engager (NKCE) armed with a non-alpha IL-2 variant (IL-2v), which targets CD20 and was developed for treating B cell non-Hodgkin lymphoma (B-NHL). CD20-NKCE-IL2v boosts NK cell proliferation and cytotoxicity, showing activity against a range of B-NHL cell lines, including those with low CD20 density. Whereas it presented reduced toxicities compared with those commonly associated with T cell therapies, CD20-NKCE-IL2v showed greater killing efficacy over a T cell engager targeting CD20 in in vitro preclinical models. CD20-NKCE-IL2v also increased the cell surface expression of NK cell-activating receptors, leading to activity against CD20-negative tumor cells. In vivo studies in nonhuman primates and tumor mouse models further validated its efficacy and revealed that CD20-NKCE-IL2v induces peripheral NK cell homing at the tumor site. CD20-NKCE-IL2v emerges as a potential alternative in the treatment landscape of B-NHL.
Collapse
Affiliation(s)
- Olivier Demaria
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Guillaume Habif
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Marie Vetizou
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Laurent Gauthier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Romain Remark
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Laura Chiossone
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Constance Vagne
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Lucas Rebuffet
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Rachel Courtois
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Caroline Denis
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - François Le Floch
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Marianna Muller
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | | | - Séverine Augier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Julie Lopez
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Barbara Carrette
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Aurélie Maguer
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | | | | | - William Baron
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Justine Galluso
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Nadia Yessaad
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Marilyn Giordano
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Léa Simon
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Fabien Chanuc
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | | | - Ivan Perrot
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Cécile Bonnafous
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Agnès Represa
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Benjamin Rossi
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Ariane Morel
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Yannis Morel
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Carine Paturel
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Eric Vivier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
- APHM, Hôpital de la Timone, Marseille-Immunopôle Profiling Platform, Marseille, France
- Paris-Saclay Cancer Cluster, Le Kremlin-Bicêtre, France
- Université Paris-Saclay, Gustave Roussy, INSERM, Prédicteurs moléculaires et nouvelles cibles en oncologie, 94800, Villejuif, France
| |
Collapse
|
33
|
Deschênes-Simard X, Santomasso BD, Dahi PB. Clinical features, pathophysiology, and management of acute myelopathy following CAR T-cell therapy. Blood 2024; 144:2083-2094. [PMID: 39226460 DOI: 10.1182/blood.2024025679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024] Open
Abstract
ABSTRACT Chimeric antigen receptor (CAR) T-cell therapy has revolutionized the treatment of patients with relapsed or refractory hematologic malignancies, but it comes with unique toxicities, notably cytokine release syndrome and ICANS (immune effector cell-associated neurotoxicity syndrome). As experience with CAR T-cell therapy grows, distinct and infrequent neurologic complications are becoming increasingly evident. Recently, reports of acute myelopathy after the administration of CAR T-cell therapies have been accumulating. Despite the establishment of consensus guidelines for managing ICANS, there remains limited guidance on the appropriate investigations and treatments for this rare complication. In this manuscript, we delve into the clinical features, pathophysiology, and strategies for the optimal management of acute myelitis after CAR T-cell therapy and draw insights from reported cases in the literature.
Collapse
Affiliation(s)
- Xavier Deschênes-Simard
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, University of Montreal, Montréal, QC, Canada
| | - Bianca D Santomasso
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Parastoo B Dahi
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| |
Collapse
|
34
|
Ma H, Yan Z, Gu R, Xu Y, Qiu S, Xing H, Tang K, Tian Z, Rao Q, Wang M, Wang J. Loop33 × 123 CAR-T targeting CD33 and CD123 against immune escape in acute myeloid leukemia. Cancer Immunol Immunother 2024; 74:20. [PMID: 39535595 PMCID: PMC11561222 DOI: 10.1007/s00262-024-03847-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 09/24/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Immunotherapy, such as chimeric antigen receptor T (CAR-T) cells targeting CD33 or CD123, has been well developed over the past decade for the treatment of acute myeloid leukemia (AML). However, the inability to sustain tumor-free survival and the possibility of relapse due to antigen loss have raised concerns. A dual targeting of CD33 and CD123 is needed for better outcomes. METHODS Based on our previously constructed CD33 and CD123 monovalent CAR-T, Loop33 × 123 and Loop123 × 33 CAR-T were constructed with molecular cloning techniques. All CAR-T cells were generated by lentivirus transduction of T cells from healthy donors. Phenotype detection was evaluated on day 7 concerning activation, exhaustion, and subtype proportions. Coculture killing assays were conducted using various AML cell lines and primary AML cells. Degranulation and cytokine secretion levels were detected by flow cytometry. Cell-derived xenograft models were established using wild-type Molm 13 cell lines, or a mixture of Molm 13-KO33 and Molm 13-KO123 cells as an ideal model of immune escape. By monitoring body weight and survival of tumor-bearing mice, Loop33 × 123 and Loop123 × 33 CAR-T cells were further assessed for their efficacy in vivo. RESULTS In vitro study, our results demonstrated that Loop33 × 123 CAR-T cells could efficiently eliminate AML cell lines and primary AML cells with elevated degranulation and cytokine secretion levels. Compared with our previously constructed monovalent CD33 or CD123 CAR-T cells, Loop33 × 123 CAR-T cells showed superior advantages in an immune escape model. In vivo studies further confirmed that Loop33 × 123 CAR-T cells could effectively prolong the survival of mice without significant toxicity. However, Loop123 × 33 CAR-T cells failed to show the same effects. Furthermore, Loop33 × 123 CAR-T cells efficiently circumvented potential immune escape, a challenge where monovalent CAR-T cells failed. CONCLUSIONS Loop33 × 123 CAR-T targeting CD33 and CD123 could efficiently eliminate AML cells and prolong survival of tumor-bearing mice, while addressing the issue of immune escape.
Collapse
MESH Headings
- Animals
- Humans
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/therapy
- Interleukin-3 Receptor alpha Subunit/immunology
- Interleukin-3 Receptor alpha Subunit/metabolism
- Sialic Acid Binding Ig-like Lectin 3/immunology
- Sialic Acid Binding Ig-like Lectin 3/metabolism
- Mice
- Immunotherapy, Adoptive/methods
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Tumor Escape/immunology
- Xenograft Model Antitumor Assays
- T-Lymphocytes/immunology
- Cell Line, Tumor
- Mice, Inbred NOD
- Mice, SCID
- Female
Collapse
Affiliation(s)
- Haotian Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
| | - Zhifeng Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
| | - Runxia Gu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
| | - Yingxi Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
| | - Shaowei Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
| | - Haiyan Xing
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
| | - Kejing Tang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
| | - Zheng Tian
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
| | - Qing Rao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301617, China
| | - Min Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301617, China.
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Key Laboratory of Cell Therapy for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301617, China.
| |
Collapse
|
35
|
Palacios-Berraquero ML, Rodriguez-Marquez P, Calleja-Cervantes ME, Berastegui N, Zabaleta A, Burgos L, Alignani D, San Martin-Uriz P, Vilas-Zornoza A, Rodriguez-Diaz S, Inoges S, Lopez-Diaz de Cerio A, Huerga S, Tamariz E, Rifon J, Alfonso-Pierola A, Lasarte JJ, Paiva B, Hernaez M, Rodriguez-Otero P, San-Miguel J, Ezponda T, Rodriguez-Madoz JR, Prosper F. Molecular mechanisms promoting long-term cytopenia after BCMA CAR-T therapy in multiple myeloma. Blood Adv 2024; 8:5479-5492. [PMID: 39058976 PMCID: PMC11532743 DOI: 10.1182/bloodadvances.2023012522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 06/12/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
ABSTRACT Hematologic toxicity is a common side effect of chimeric antigen receptor T-cell (CAR-T) therapies, being particularly severe among patients with relapsed or refractory multiple myeloma (MM). In this study, we characterized 48 patients treated with B-cell maturation antigen (BCMA) CAR-T cells to understand kinetics of cytopenia, identify predictive factors, and determine potential mechanisms underlying these toxicities. We observed that overall incidence of cytopenia was 95.7%, and grade >3 thrombocytopenia and neutropenia, 1 month after infusion, was observed in 57% and 53% of the patients, respectively, being still present after 1 year in 4 and 3 patients, respectively. Baseline cytopenia and high peak inflammatory markers were highly correlated with cytopenia that persisted up to 3 months. To determine potential mechanisms underlying cytopenias, we evaluated the paracrine effect of BCMA CAR-T cells on hematopoietic stem and progenitor cell (HSPC) differentiation using an ex vivo myeloid differentiation model. Phenotypic analysis showed that supernatants from activated CAR-T cells (spCAR) halted HSPC differentiation, promoting more immature phenotypes, which could be prevented with a combination of interferon γ, tumor necrosis factor α/β, transforming growth factor β, interleukin-6 (IL-6) and IL-17 inhibitors. Single-cell RNA sequencing demonstrated upregulation of transcription factors associated with early stages of hematopoietic differentiation in the presence of spCAR (GATA2, RUNX1, CEBPA) and a decrease in the activity of key regulons involved in neutrophil and monocytic maturation (ID2 and MAFB). These results suggest that CAR-T activation induces HSPC maturation arrest through paracrine effects and provides potential treatments to mitigate the severity of this toxicity.
Collapse
Affiliation(s)
- Maria Luisa Palacios-Berraquero
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Paula Rodriguez-Marquez
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Maria Erendira Calleja-Cervantes
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Computational Biology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Nerea Berastegui
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Aintzane Zabaleta
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Leire Burgos
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Diego Alignani
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Patxi San Martin-Uriz
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Amaia Vilas-Zornoza
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Saray Rodriguez-Diaz
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Susana Inoges
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Immunology and Immunotherapy Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Ascensión Lopez-Diaz de Cerio
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Immunology and Immunotherapy Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Sofia Huerga
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Esteban Tamariz
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Jose Rifon
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Ana Alfonso-Pierola
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Juan Jose Lasarte
- Immunology and Immunotherapy Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Bruno Paiva
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Mikel Hernaez
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Computational Biology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Data Science and Artificial Intelligence Institute, Universidad de Navarra, Pamplona, Spain
| | - Paula Rodriguez-Otero
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Jesus San-Miguel
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Teresa Ezponda
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Juan Roberto Rodriguez-Madoz
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Felipe Prosper
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| |
Collapse
|
36
|
Wang T, Lau CH, Wang N, Li J, Wang J, Huang Z, Wu W, Chen X, Li J, Zou M, Zhang W, Li Y, Li J, Ma W, Huang Y, Xu M, Zhu H, Chen G. SynNotch-Programmed Macrophages for Cancerous Cell Detection and Sensing. ACS Sens 2024. [PMID: 39496105 DOI: 10.1021/acssensors.4c01997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2024]
Abstract
Synthetic Notch (synNotch) receptors have enabled mammalian cells to sense extracellular ligands and respond by activating user-prescribed transcriptional programs. Based on the synNotch system, we describe a cell-based in vivo sensor for cancerous cell detection. We attempted to engineer synNotch-programmed macrophages to sense cancer cells via urinary analysis of human chorionic gonadotropin (HCGB5). Principally, when the synNotch receptors of macrophages bind to the ligands of cancer cells, Notch is activated and undergoes intramembrane proteolysis to release the transcriptional activator into the nucleus. The transcriptional activator targets and activates downstream gene expression, such as human chorionic gonadotropin (HCGB5) in macrophages. When HCGB5 is secreted extracellularly into urine, it can be detected with commercial HCGB5 colloidal gold test strips. As a proof of principle, we demonstrated the feasibility of synNotch-programmed macrophages in detecting breast cancer cells engineered with artificial EGFP ligands. We demonstrated that HCGB5 expression was only induced when the cancer cell expressing EGFP ligands is present; thereby, extracellular HCGB5 expression is directly proportional to the number of cancer cells. Further optimizations of the synNotch system can realize the ultimate goal of establishing cell-based in vivo sensors as the paragon of cancer diagnostics for point-of-care testing and home self-test.
Collapse
Affiliation(s)
- Tao Wang
- Department of Biology, College of Science, Shantou University, 515063 Shantou, Guangdong, China
| | - Cia-Hin Lau
- Department of Biology, College of Science, Shantou University, 515063 Shantou, Guangdong, China
| | - Naian Wang
- Department of Biology, College of Science, Shantou University, 515063 Shantou, Guangdong, China
| | - Jiaqi Li
- Department of Biology, College of Science, Shantou University, 515063 Shantou, Guangdong, China
| | - Jianchao Wang
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, 350014 Fuzhou, Fujian, China
| | - Zhihao Huang
- Department of Biology, College of Science, Shantou University, 515063 Shantou, Guangdong, China
| | - Weidong Wu
- Department of Biology, College of Science, Shantou University, 515063 Shantou, Guangdong, China
| | - Xiaoqing Chen
- Department of Biology, College of Science, Shantou University, 515063 Shantou, Guangdong, China
| | - Jiahui Li
- Department of Biology, College of Science, Shantou University, 515063 Shantou, Guangdong, China
| | - Minghai Zou
- Department of Biology, College of Science, Shantou University, 515063 Shantou, Guangdong, China
| | - Wenju Zhang
- Department of Biology, College of Science, Shantou University, 515063 Shantou, Guangdong, China
| | - Yulin Li
- Department of Biology, College of Science, Shantou University, 515063 Shantou, Guangdong, China
| | - Jingrong Li
- Department of Biology, College of Science, Shantou University, 515063 Shantou, Guangdong, China
| | - Wenkai Ma
- Department of Biology, College of Science, Shantou University, 515063 Shantou, Guangdong, China
| | - Yumei Huang
- Department of Biology, College of Science, Shantou University, 515063 Shantou, Guangdong, China
| | - Meijing Xu
- Xiamen Fly Gene Biomedical Technology Co., Ltd., Biomedical Industrial Park, 361000 Xiamen, Fujian, China
| | - Haibao Zhu
- Department of Biology, College of Science, Shantou University, 515063 Shantou, Guangdong, China
| | - Gang Chen
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, 350014 Fuzhou, Fujian, China
| |
Collapse
|
37
|
Zhao D, Wen X, Wu J, Chen F. Photoimmunotherapy for cancer treatment based on organic small molecules: Recent strategies and future directions. Transl Oncol 2024; 49:102086. [PMID: 39181114 PMCID: PMC11387906 DOI: 10.1016/j.tranon.2024.102086] [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: 06/03/2024] [Revised: 07/25/2024] [Accepted: 08/11/2024] [Indexed: 08/27/2024] Open
Abstract
Photodynamic therapy (PDT) is considered as a promising anticancer approach, owning to its high efficiency and spatiotemporal selectivity. Ample evidence indicated that PDT can trigger immunogenic cell death by releasing antigens that activate immune cells to promote anti-tumor immunity. Nevertheless, the inherent nature of tumors and their complex heterogeneity often limits the efficiency of PDT, which can be overcome with a novel strategy of photo-immunotherapy (PIT) strategy. By exploring the principles of PDT induction and ICD enhancement, combined with other therapies such as chemotherapy or immune checkpoint blockade, the tailored solutions can be designed to address specific challenges of drug resistance, hypoxic conditions, and tumor immunosuppressive microenvironments (TIMEs), which enables targeted enhancement of systemic immunity to address most distant and recurrent cancers. The present article summarizes the specific strategies of PIT and discusses recent existing limitations. More importantly, we anticipate that the perspectives presented herein will help address the clinical translation challenges associated with PIT.
Collapse
Affiliation(s)
- Deming Zhao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Xin Wen
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Jiani Wu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Feihong Chen
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China.
| |
Collapse
|
38
|
Locke FL, Neelapu SS, Bartlett NL, Lekakis LJ, Jacobson CA, Braunschweig I, Oluwole OO, Siddiqi T, Lin Y, Timmerman JM, Kersten MJ, Zheng Y, Zhang T, Nater J, Shen R, Miao H, Kim JJ, Miklos DB. Tocilizumab Prophylaxis Following Axicabtagene Ciloleucel in Relapsed or Refractory Large B-Cell Lymphoma. Transplant Cell Ther 2024; 30:1065-1079. [PMID: 39187161 DOI: 10.1016/j.jtct.2024.08.018] [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: 05/17/2024] [Revised: 08/12/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024]
Abstract
Axicabtagene ciloleucel (axi-cel) is an autologous anti-CD19 chimeric antigen receptor (CAR) T-cell therapy approved in patients with relapsed/refractory (R/R) large B-cell lymphoma (LBCL). Most patients treated with axi-cel experience cytokine release syndrome (CRS) and/or adverse neurologic events (NEs). To explore potential approaches for reducing CAR T-cell-related toxicities with axi-cel, several safety expansion cohorts were added to the pivotal ZUMA-1 trial. ZUMA-1 Cohort 3 was an exploratory safety cohort that investigated the use of the IL-6 receptor-blocking antibody tocilizumab and anticonvulsant levetiracetam as prophylaxis against CRS and NEs in patients treated with axi-cel. Patients with R/R LBCL were enrolled in Cohort 3 and received conditioning chemotherapy on d -5 through -3 followed by a single infusion of axi-cel (2 × 106 cells/kg) on d 0. Prophylactic tocilizumab (8 mg/kg) was administered 48 h after axi-cel infusion. Primary endpoints were incidence and severity of CRS and NEs. Key secondary endpoints included the incidence of adverse events, objective response rate (ORR), duration of response, progression-free survival, overall survival (OS), and biomarker analyses (eg, circulating CAR T cells, cytokines, chemokines). Forty-two patients were enrolled in Cohort 3, 38 of whom received axi-cel. In the 24-month analysis, any-grade CRS and NEs occurred in 92% and 87% of patients, and Grade ≥3 CRS and NEs occurred in 3% and 42% of patients, respectively. One Grade 5 NE (cerebral edema) occurred. With 24-mo minimum follow-up, the ORR was 63%, and 39.5% of patients had ongoing response. With 48-month follow-up, median OS was 34.8 mo (95% CI, 5.4-not estimable). CAR T-cell expansion in ZUMA-1 Cohort 3 was comparable with pivotal Cohorts 1 and 2. Consistent with tocilizumab-mediated inhibition of IL-6R, serum IL-6 levels were increased relative to Cohorts 1 and 2. Grade ≥3 NEs were associated with elevated IL-6 levels, proinflammatory cytokines, and myeloid cells in the cerebrospinal fluid. Based on these findings, prophylactic tocilizumab is not recommended to prevent CAR T-cell-related adverse events, and beneficial effects of prophylactic levetiracetam remain uncertain in patients with R/R LBCL.
Collapse
Affiliation(s)
| | | | | | - Lazaros J Lekakis
- University of Miami Health System, Sylvester Comprehensive Cancer Center, Miami, Florida
| | | | - Ira Braunschweig
- Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | | | - Tanya Siddiqi
- City of Hope National Medical Center, Duarte, California
| | - Yi Lin
- Mayo Clinic, Rochester, Minnesota
| | | | - Marie José Kersten
- Amsterdam UMC, University of Amsterdam, Amsterdam, Cancer Center Amsterdam, Netherlands, on behalf of HOVON/LLPC
| | - Yan Zheng
- Kite, a Gilead Company, Santa Monica, California
| | - Teresa Zhang
- Kite, a Gilead Company, Santa Monica, California
| | - Jenny Nater
- Kite, a Gilead Company, Santa Monica, California
| | - Rhine Shen
- Kite, a Gilead Company, Santa Monica, California
| | - Harry Miao
- Kite, a Gilead Company, Santa Monica, California
| | - Jenny J Kim
- Kite, a Gilead Company, Santa Monica, California
| | - David B Miklos
- Stanford University School of Medicine, Stanford, California
| |
Collapse
|
39
|
Ochenduszko S, Landete L, Martinez DC, Feria AG, Francés C, Torregrosa MD, Maiques IM. Cytokine release syndrome and immune effector cell‑associated neurotoxicity syndrome in a melanoma patient treated with adjuvant pembrolizumab. Exp Ther Med 2024; 28:423. [PMID: 39301256 PMCID: PMC11412105 DOI: 10.3892/etm.2024.12712] [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: 04/03/2024] [Accepted: 07/11/2024] [Indexed: 09/22/2024] Open
Abstract
The emergence of immune checkpoint inhibitors (ICIs) has significantly improved the prognosis of patients with solid tumors. However, along with their efficacy, new toxicities related to immune system activation have surfaced, some of which pose life-threatening risks. Cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) are among the serious, albeit rare, immune-related adverse effects (irAEs) observed. Although commonly associated with hematologic malignancies and chimeric antigen receptor T cell therapies, CRS has been reported in patients treated with ICIs, with ICANS being a less documented complication. The present study presents a case report of a 76-year-old patient with resected melanoma who developed clinical symptoms of CRS and ICANS following adjuvant pembrolizumab therapy. The patient presented with neurological symptoms of weakness and encephalopathy with confusion, bradypsychia, dysarthria, tremors and visual hallucinations. Laboratory tests revealed elevated serum levels of tumor necrosis factor-alpha and interleukin-6 along with inflammatory markers, hepatic and renal dysfunction, as well as rapidly progressive normochromic-normocytic anemia. Treatment with corticosteroids led to rapid symptom resolution, albeit with subsequent symptom recurrence after tapering its dose. This case underscores the importance of recognizing and managing irAEs associated with ICIs and highlights the need for vigilant monitoring and individualized therapeutic approaches.
Collapse
Affiliation(s)
| | - Lamberto Landete
- Department of Neurology, Doctor Peset University Hospital, 46017 Valencia, Spain
| | | | - Ana García Feria
- Department of Hematology, Doctor Peset University Hospital, 46017 Valencia, Spain
| | - Carla Francés
- Department of Endocrinology, Doctor Peset University Hospital, 46017 Valencia, Spain
| | | | | |
Collapse
|
40
|
Lin Y, Chen M, Huang S, Chen Y, Ho JH, Lin F, Tan X, Chiang H, Huang C, Tu C, Cho D, Chiu S. Targeting Dual Immune Checkpoints PD-L1 and HLA-G by Trispecific T Cell Engager for Treating Heterogeneous Lung Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309697. [PMID: 39234811 PMCID: PMC11538689 DOI: 10.1002/advs.202309697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/04/2024] [Indexed: 09/06/2024]
Abstract
Immunotherapy targeting immune checkpoints (ICPs), such as programmed death-ligand-1 (PD-L1), is used as a treatment option for advanced or metastatic non-small cell lung cancer (NSCLC). However, overall response rate to anti-PD-L1 treatment is limited due to antigen heterogeneity and the immune-suppressive tumor microenvironment. Human leukocyte antigen-G (HLA-G), an ICP as well as a neoexpressed tumor-associated antigen, is previously demonstrated to be a beneficial target in combination with anti-PD-L1. In this study, a nanobody-based trispecific T cell engager (Nb-TriTE) is developed, capable of simultaneously binding to T cells, macrophages, and cancer cells while redirecting T cells toward tumor cells expressing PD-L1- and/or HLA-G. Nb-TriTE shows broad spectrum anti-tumor effects in vitro by augmenting cytotoxicity mediated by human peripheral blood mononuclear cells (PBMCs). In a humanized immunodeficient murine NSCLC model, Nb-TriTE exhibits superior anti-cancer potency compared to monoclonal antibodies and bispecific T cell engagers. Nb-TriTE, at the dose with pharmacoactivity, does not induce additional enhancement of circulating cytokines secretion from PMBCs. Nb-TriTE effectively prolongs the survival of mice without obvious adverse events. In conclusion, this study introduces an innovative therapeutic approach to address the challenges of immunotherapy and the tumor microenvironment in NSCLC through utilizing the dual ICP-targeting Nb-TriTE.
Collapse
Affiliation(s)
- Yu‐Chuan Lin
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
- Shine‐On BioMedical Co. Ltd.Rm. B, 10F., No. 573, Sec. 2, Taiwan Blvd., West Dist.Taichung City403Taiwan
| | - Mei‐Chih Chen
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
| | - Shi‐Wei Huang
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
- Institute of New Drug DevelopmentChina Medical UniversityTaichung City404Taiwan
- Institute of Biomedical SciencesNational Chung Hsing UniversityTaichung City402Taiwan
| | - Yeh Chen
- Department of Food Science and BiotechnologyNational Chung Hsing UniversityTaichung City402Taiwan
| | - Jennifer Hui‐Chun Ho
- Shine‐On BioMedical Co. Ltd.Rm. B, 10F., No. 573, Sec. 2, Taiwan Blvd., West Dist.Taichung City403Taiwan
- Center for Translational Genomics and Regenerative Medicine ResearchChina Medical University HospitalTaichung City404Taiwan
- Department of OphthalmologyChina Medical University HospitalChina Medical UniversityTaichung City404Taiwan
- Department of Medical ResearchEye CenterChina Medical University HospitalTaichung City404Taiwan
| | - Fang‐Yu Lin
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
| | - Xiao‐Tong Tan
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
| | - Hung‐Che Chiang
- Shine‐On BioMedical Co. Ltd.Rm. B, 10F., No. 573, Sec. 2, Taiwan Blvd., West Dist.Taichung City403Taiwan
- College of MedicineChina Medical UniversityTaichung City404Taiwan
| | - Chiu‐Ching Huang
- Shine‐On BioMedical Co. Ltd.Rm. B, 10F., No. 573, Sec. 2, Taiwan Blvd., West Dist.Taichung City403Taiwan
- Division of Nephrology and the Kidney InstituteDepartment of Internal MedicineChina Medical University HospitalTaichung City404Taiwan
| | - Chih‑Yen Tu
- Division of Pulmonary and Critical CareDepartment of Internal MedicineChina Medical University HospitalTaichung City404Taiwan
- School of MedicineCollege of MedicineChina Medical UniversityTaichung City404Taiwan
| | - Der‐Yang Cho
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
- Institute of New Drug DevelopmentChina Medical UniversityTaichung City404Taiwan
- Drug Development CenterChina Medical UniversityTaichung City404Taiwan
- Department of NeurosurgeryChina Medical University HospitalTaichung City404Taiwan
| | - Shao‐Chih Chiu
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
- Shine‐On BioMedical Co. Ltd.Rm. B, 10F., No. 573, Sec. 2, Taiwan Blvd., West Dist.Taichung City403Taiwan
- Institute of New Drug DevelopmentChina Medical UniversityTaichung City404Taiwan
- Drug Development CenterChina Medical UniversityTaichung City404Taiwan
| |
Collapse
|
41
|
Ursu R, Belin C, Cuzzubbo S, Carpentier AF. CAR T-cell-associated neurotoxicity: A comprehensive review. Rev Neurol (Paris) 2024; 180:989-994. [PMID: 39289138 DOI: 10.1016/j.neurol.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 07/24/2024] [Indexed: 09/19/2024]
Abstract
Chimeric antigen receptor T-cell (CAR T-cell) therapies have emerged as a promising treatment modality for several malignancies, particularly haematological malignancies, by inducing robust antitumour responses. However, CAR T-cell therapies are associated with a spectrum of adverse events, including neurological complications. We here provide a review of neurological adverse events observed in patients undergoing CAR T-cell therapy, focusing on their incidence, clinical manifestations, underlying mechanisms and potential management strategies.
Collapse
Affiliation(s)
- R Ursu
- Service de neurologie, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France
| | - C Belin
- Service de neurologie, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France
| | - S Cuzzubbo
- Service de neurologie, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France; Paris-Diderot, université de Paris, 75010 Paris, France
| | - A F Carpentier
- Service de neurologie, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France; Paris-Diderot, université de Paris, 75010 Paris, France.
| |
Collapse
|
42
|
Lima JFC, Santos FM, de Miranda TB, Ramos GG, Andia DC, Lima AF, Ciotti DL. Inflammatory and adhesion profile of gingival fibroblasts to lithium disilicate ceramic surfaces. Dent Mater 2024; 40:2025-2033. [PMID: 39358190 DOI: 10.1016/j.dental.2024.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/15/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVES Lithium disilicate (LS) ceramic emerges as a compelling option for customized implant abutments. However, ensuring its safety and reliability requires clarification on key aspects, notably its impact on inflammation and potential for cell adhesion. This study delves into these considerations, examining the influence of LS ceramic on cytokine release and the transcriptional profile of human gingival fibroblasts (hGFs) in direct contact with various LS surfaces. METHODS hGFs were cultured on LS disks featuring three distinct surfaces (unpolished, polished, and polished glaze), while titanium disks served as reference material and cells cultured directly on plates as controls. The surface of the disks was analyzed using a scanning electron microscope. The cell metabolism was analyzed by MTT test, cytokine release by MAGPIX and the expression of genes related to cell adhesion was evaluated by qPCR. RESULTS The disks exhibited similar topography with smooth surfaces, except for the unpolished LS disks, which had an irregular surface. Contact with LS surfaces did not substantially reduce cell metabolism. Moreover, it generally decreased cytokine release compared to controls, particularly pro-inflammatory mediators like IL-1β, IL-6, and TNF-α. Significantly increased expression of genes related to cell adhesion to LS was observed, comparable to titanium, the gold standard material for implant abutments. SIGNIFICANCE This study unveils that LS ceramic not only fails to trigger pro-inflammatory cytokine release, but also significantly enhances gene expression associated with cell adhesion. These mechanisms are closely linked to gene pathways such as PTK2, SRC, MAPK1, and transcription factors ELK-1 and MYC. In summary, the findings underscore LS ceramic's potential as a biocompatible material for implant abutments, shedding light on its favorable inflammatory response and enhanced cell adhesion properties.
Collapse
Affiliation(s)
| | - Filipe Milazzo Santos
- Dental Research Division, Paulista University, Rua Doutor Bacelar, 1212, Sao Paulo 04026-002, Brazil
| | - Taís Browne de Miranda
- Dental Research Division, Paulista University, Rua Doutor Bacelar, 1212, Sao Paulo 04026-002, Brazil
| | - Guilherme Gama Ramos
- São Leopoldo Mandic Institute and Dental Research Center, Campinas, São Paulo, Brazil
| | - Denise Carleto Andia
- Dental Research Division, Paulista University, Rua Doutor Bacelar, 1212, Sao Paulo 04026-002, Brazil.
| | - Adriano F Lima
- Dental Research Division, Paulista University, Rua Doutor Bacelar, 1212, Sao Paulo 04026-002, Brazil.
| | - Danilo Lazzari Ciotti
- São Leopoldo Mandic Institute and Dental Research Center, Campinas, São Paulo, Brazil.
| |
Collapse
|
43
|
Pu J, Liu T, Sharma A, Jiang L, Wei F, Ren X, Schmidt-Wolf IGH, Hou J. Advances in adoptive cellular immunotherapy and therapeutic breakthroughs in multiple myeloma. Exp Hematol Oncol 2024; 13:105. [PMID: 39468695 PMCID: PMC11514856 DOI: 10.1186/s40164-024-00576-6] [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: 07/11/2024] [Accepted: 10/14/2024] [Indexed: 10/30/2024] Open
Abstract
The basic idea of modulating the immune system to better recognize and fight tumor cells has led to the successful introduction of adoptive cellular immunotherapy (ACT). ACT-based treatment regimens, in which the patient's own immune cells are isolated and subsequently expanded (ex vivo) and reinfused, have also contributed significantly to the development of a personalized treatment strategy. Complementing this, the unprecedented advances in ACTs as chimeric antigen receptor (CAR)-T cell therapies and their derivatives such as CAR-NK, CAR-macrophages, CAR-γδT and CAR-NKT have further maximized the therapeutic outcomes. Herein, we provide a comprehensive overview of the development of ACTs in multiple myeloma (MM) and outline how they have evolved from an experimental form to a mainstay of standard clinical settings. Besides, we provide insights into cytokine-induced killer cell (CIK) therapy, an alternative form of ACT that (as CIK or CAR-CIK) has enormous potential in the clinical spectrum of MM. We also summarize the results of the major preclinical and clinical studies of adoptive cell therapy in MM and address the current challenges (such as cytokine release syndrome (CRS) and neurotoxicity) that limit its complete success in the cancer landscape.
Collapse
Affiliation(s)
- Jingjing Pu
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, 53127, Bonn, NRW, Germany
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Ting Liu
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, NRW, Germany
| | - Amit Sharma
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, 53127, Bonn, NRW, Germany
| | - Liping Jiang
- Wuxi Maternity and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China
| | - Feng Wei
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300070, China
| | - Xiubao Ren
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300070, China.
| | - Ingo G H Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, 53127, Bonn, NRW, Germany.
| | - Jian Hou
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
44
|
Zhang Y, Xing Y, Zhou H, Ma E, Xu W, Zhang X, Jiang C, Ye S, Deng Y, Wang H, Li J, Zheng S. NIR-activated Janus nanomotors with promoted tumor permeability for synergistic photo-immunotherapy. Acta Biomater 2024:S1742-7061(24)00632-9. [PMID: 39490462 DOI: 10.1016/j.actbio.2024.10.040] [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: 07/02/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Nanoparticle-based photo-immunotherapy has become an attractive strategy to eliminate tumors and activate host immune responses. However, the therapeutic efficacy is heavily restricted by low tumoral penetration and immunosuppressive tumor microenvironment (TME). Herein, near infrared laser (NIR)-propelled Janus nanomotors were presented for deep tumoral penetration, photothermal tumor ablation and photothermal-triggered augmented immunotherapy. The Janus nanomotors (AuNR/PMO@CPG JNMs) were constructed with gold nanorods (AuNR) and periodic mesoporous organo-silica nanospheres (PMO), followed by loading of immune adjuvant (CPG ODNs). Under NIR irradiation, the nanomotors exhibited superior photothermal effect, which produced active motion with a speed of 19.3 µm/s for deep tumor penetration and accumulation in vivo. Moreover, the good photothermal heating also benefited effective photothermal ablation to trigger immunogenic cell death (ICD). Subsequently, the ICD effect promoted the release of tumor-associated antigens (TAAs) and damage associated molecular patterns (DAMPs), and further generated abundant tumor vaccines in situ for reprograming the immunosuppressive TME in combination with CPG ODNs to inhibit tumor growth. As a result, a notable in vivo synergistic therapeutic effect was realized on CT26-bearing mice by combining photothermal therapy-induced ICD with modulation of immunosuppressive TME. Thus, we believe that the synthesized nanomotors can provide a new inspect to boost photothermal therapy-induced ICD in tumor immunotherapy. STATEMENT OF SIGNIFICANCE: Nanoparticle-based synergistic photo-immunotherapy has become a popular strategy to eliminate tumors and activate host immune responses. However, the therapeutic efficacy is heavily restricted by low tumoral penetration and immunosuppressive tumor microenvironment (TME). In this work, near infrared laser (NIR)-propelled Janus nanomotors were presented for deep tumoral penetration, photothermal tumor ablation and photothermal-triggered augmented immunotherapy. Under NIR irradiation, the nanomotors exhibited a superior photothermal effect, which produced active motion for deep tumor penetration and accumulation in vivo. Moreover, good photothermal heating also facilitated effective photothermal ablation to trigger immunogenic cell death (ICD), which promoted the release of tumor-associated antigens and damage-associated molecular patterns (DAMPs), and further generated abundant tumor vaccines in situ for reprograming the immunosuppressive TME to inhibit tumor growth.
Collapse
Affiliation(s)
- Yingying Zhang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yujuan Xing
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, China
| | - Hong Zhou
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, China
| | - Enhui Ma
- School of Chemical Engineering & Technology, China University of Mining and Technology, Xuzhou, 221116, China
| | - Wenbei Xu
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, China
| | - Xinran Zhang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, China
| | - Canran Jiang
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, China
| | - Shuo Ye
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yanjia Deng
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, China
| | - Hong Wang
- School of Chemical Engineering & Technology, China University of Mining and Technology, Xuzhou, 221116, China.
| | - Jingjing Li
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, China; Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, China.
| | - Shaohui Zheng
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, 221004, China; Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, China.
| |
Collapse
|
45
|
Angelillo P, Alarcon Tomas A, Campodonico E, Novak U, Gabellier L, Blau IW, Kwon M, Ram R, Forcade E, Besley C, Vucinic V, Kröger N, Corral LL, Vydra J, Ferrari S, von Tresckow B, Rubio MT, Hernani R, Sica S, Stölzel F, Rovira M, Wagner-Drouet E, Pérez-Simón JA, Castilla-Llorente C, Ferreri AJM, Hoogenboom JD, Sanchez Ortega I, Malard F, Kuball J, Ruggeri A. Severe ICANS after CAR T-cell therapy and assessment of prevention with levetiracetam for seizure prophylaxis following CAR T-cell for DLBCL & PMBCL in Europe: a survey on behalf of the Cellular Therapy & Immunobiology Working Party (CTIWP) of the EBMT. Bone Marrow Transplant 2024:10.1038/s41409-024-02451-8. [PMID: 39455896 DOI: 10.1038/s41409-024-02451-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/07/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Affiliation(s)
| | | | | | - Urban Novak
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Igor Wolfgang Blau
- Medizinische Klinik m. S. Hämatologie, Onkologie und Tumorimmunologie, Berlin, Germany
| | - Mi Kwon
- Department of Hematology, Hospital General Unversitario, Madrid, Spain
- Universidad Complutense de Madrid, Madrid, Spain
| | - Ron Ram
- Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | | | - Caroline Besley
- University Hospitals Bristol and Weston NHSFT, Bristol, United Kingdom
| | | | | | - Lucía López Corral
- Hospital Universitario de Salamanca (Spain), IBSAL, CIBERONC, Salamanca, Spain
| | - Jan Vydra
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | | | - Bastian von Tresckow
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | | | | | | | - Friedrich Stölzel
- University Hospital Schleswig-Holstein, Division of Stem Cell Transplantation and Cellular Immunotherapies, Kiel, Germany
| | | | | | - Jose Antonio Pérez-Simón
- Department of Hematology, University Hospital Virgen Del Rocio, Instituto de Biomedicina de Sevilla (IBIS), CSIC, Universidad de Sevilla, Sevilla, Spain
| | | | | | | | | | - Florent Malard
- Sorbonne Université, Hôpital Saint-Antoine, AP-HP, Service d'Hématologie Clinique et Thérapie Cellulaire, Paris, France
| | - Jürgen Kuball
- University Medical Center Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|
46
|
Minguet S, Maus MV, Schamel WW. From TCR fundamental research to innovative chimeric antigen receptor design. Nat Rev Immunol 2024:10.1038/s41577-024-01093-7. [PMID: 39433885 DOI: 10.1038/s41577-024-01093-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 10/23/2024]
Abstract
Engineered T cells that express chimeric antigen receptors (CARs) have transformed the treatment of haematological cancers. CARs combine the tumour-antigen-binding function of antibodies with the signalling functions of the T cell receptor (TCR) ζ chain and co-stimulatory receptors. The resulting constructs aim to mimic the TCR-based and co-receptor-based activation of T cells. Although these have been successful for some types of cancer, new CAR formats are needed, to limit side effects and broaden their use to solid cancers. Insights into the mechanisms of TCR signalling, including the identification of signalling motifs that are not present in the TCR ζ chain and mechanistic insights in TCR activation, have enabled the development of CAR formats that outcompete the current CARs in preclinical mouse models and clinical trials. In this Perspective, we explore the mechanistic rationale behind new CAR designs.
Collapse
Affiliation(s)
- Susana Minguet
- Signalling Research Centers BIOSS and CIBSS, Freiburg, Germany.
- Department of Synthetic Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany.
- Centre for Chronic Immunodeficiency (CCI), Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Marcela V Maus
- Cellular Immunotherapy Program and Krantz Family Center for Cancer Research, Mass General Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Wolfgang W Schamel
- Signalling Research Centers BIOSS and CIBSS, Freiburg, Germany.
- Centre for Chronic Immunodeficiency (CCI), Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
47
|
Zhou H, Wu L. Reprogramming tumor cells to fight cancer. Science 2024; 386:274-275. [PMID: 39418388 DOI: 10.1126/science.ads6228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Cancer cells reprogrammed into dendritic cells in vivo promote antitumor immunity.
Collapse
Affiliation(s)
- Haibo Zhou
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Wu
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| |
Collapse
|
48
|
Sætersmoen M, Kotchetkov IS, Torralba-Raga L, Mansilla-Soto J, Sohlberg E, Krokeide SZ, Hammer Q, Sadelain M, Malmberg KJ. Targeting HLA-E-overexpressing cancers with a NKG2A/C switch receptor. MED 2024:100521. [PMID: 39423821 DOI: 10.1016/j.medj.2024.09.010] [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: 01/30/2024] [Revised: 06/06/2024] [Accepted: 09/23/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Human leukocyte antigen (HLA)-E is overexpressed by a large proportion of solid tumors, including malignant glioblastoma, and acts as a major checkpoint for NKG2A+ CD8+ T cells and natural killer (NK) cells in the tumor microenvironment and circulation. This axis operates alongside PD-L1 to inhibit effector responses by T and NK cells. METHODS We engineered a chimeric A/C switch receptor, combining the high HLA-E binding affinity of the NKG2A receptor ectodomain with the activating signaling of the NKG2C receptor endodomain. The cytotoxic function of A/C switch-transduced NK and T cells was evaluated against tumor cells with varying levels of HLA-E expression. In vivo efficacy was assessed using a xenograft model of glioblastoma. FINDINGS A/C switch-transduced NK and T cells exhibited superior and specific cytotoxicity against tumor cells with medium to high HLA-E expression. A/C switch-expressing human T cells demonstrated enhanced anti-tumor function in a glioblastoma xenograft model. The activity of the modified T cells was governed by an equilibrium between A/C switch levels and HLA-E expression, creating a therapeutic window to minimize on-target, off-tumor toxicities. Normal cells remained insensitive to A/C switch T cells, even after interferon (IFN)-γ pretreatment to induce HLA-E expression. CONCLUSIONS The A/C switch receptor effectively targets tumor cells expressing high levels of HLA-E, either alone or in combination with other engineered specificities, to overcome the suppressive NKG2A/HLA-E checkpoint. This approach offers a promising therapeutic strategy with a favorable safety profile for targeting HLA-E-overexpressing tumors. FUNDING This work was funded by The Research Council of Norway, the Norwegian Cancer Society, and the National Cancer Institute.
Collapse
Affiliation(s)
- Michelle Sætersmoen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Ivan S Kotchetkov
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Center for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lamberto Torralba-Raga
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Jorge Mansilla-Soto
- Center for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ebba Sohlberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Silje Zandstra Krokeide
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Quirin Hammer
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Michel Sadelain
- Center for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karl-Johan Malmberg
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
49
|
Li Q, Geng S, Luo H, Wang W, Mo YQ, Luo Q, Wang L, Song GB, Sheng JP, Xu B. Signaling pathways involved in colorectal cancer: pathogenesis and targeted therapy. Signal Transduct Target Ther 2024; 9:266. [PMID: 39370455 PMCID: PMC11456611 DOI: 10.1038/s41392-024-01953-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/25/2024] [Accepted: 08/16/2024] [Indexed: 10/08/2024] Open
Abstract
Colorectal cancer (CRC) remains one of the leading causes of cancer-related mortality worldwide. Its complexity is influenced by various signal transduction networks that govern cellular proliferation, survival, differentiation, and apoptosis. The pathogenesis of CRC is a testament to the dysregulation of these signaling cascades, which culminates in the malignant transformation of colonic epithelium. This review aims to dissect the foundational signaling mechanisms implicated in CRC, to elucidate the generalized principles underpinning neoplastic evolution and progression. We discuss the molecular hallmarks of CRC, including the genomic, epigenomic and microbial features of CRC to highlight the role of signal transduction in the orchestration of the tumorigenic process. Concurrently, we review the advent of targeted and immune therapies in CRC, assessing their impact on the current clinical landscape. The development of these therapies has been informed by a deepening understanding of oncogenic signaling, leading to the identification of key nodes within these networks that can be exploited pharmacologically. Furthermore, we explore the potential of integrating AI to enhance the precision of therapeutic targeting and patient stratification, emphasizing their role in personalized medicine. In summary, our review captures the dynamic interplay between aberrant signaling in CRC pathogenesis and the concerted efforts to counteract these changes through targeted therapeutic strategies, ultimately aiming to pave the way for improved prognosis and personalized treatment modalities in colorectal cancer.
Collapse
Affiliation(s)
- Qing Li
- The Shapingba Hospital, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Shan Geng
- Central Laboratory, The Affiliated Dazu Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Wei Wang
- Chongqing Municipal Health and Health Committee, Chongqing, China
| | - Ya-Qi Mo
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Lu Wang
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China
| | - Guan-Bin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.
| | - Jian-Peng Sheng
- College of Artificial Intelligence, Nanjing University of Aeronautics and Astronautics, Nanjing, China.
| | - Bo Xu
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing University Cancer Hospital and School of Medicine, Chongqing University, Chongqing, China.
| |
Collapse
|
50
|
Jayasinghe MK, Lay YS, Liu DXT, Lee CY, Gao C, Yeo BZ, How FYX, Prajogo RC, Hoang DV, Le HA, Pham TT, Peng B, Phung CD, Tenen DG, Le MTN. Extracellular vesicle surface display enhances the therapeutic efficacy and safety profile of cancer immunotherapy. Mol Ther 2024; 32:3558-3579. [PMID: 39033322 PMCID: PMC11489549 DOI: 10.1016/j.ymthe.2024.07.013] [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: 04/14/2024] [Revised: 06/13/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024] Open
Abstract
Immunotherapy has emerged as a mainstay in cancer therapy, yet its efficacy is constrained by the risk of immune-related adverse events. In this study, we present a nanoparticle-based delivery system that enhances the therapeutic efficacy of immunomodulatory ligands while concurrently limiting systemic toxicity. We demonstrate that extracellular vesicles (EVs), lipid bilayer enclosed particles released by cells, can be efficiently engineered via inverse electron demand Diels-Alder (iEDDA)-mediated conjugation to display multiple immunomodulatory ligands on their surface. Display of immunomodulatory ligands on the EV surface conferred substantial enhancements in signaling efficacy, particularly for tumor necrosis factor receptor superfamily (TNFRSF) agonists, where the EV surface display served as an alternative FcγR-independent approach to induce ligand multimerization and efficient receptor crosslinking. EVs displaying a complementary combination of immunotherapeutic ligands were able to shift the tumor immune milieu toward an anti-tumorigenic phenotype and significantly suppress tumor burden and increase survival in multiple models of metastatic cancer to a greater extent than an equivalent dose of free ligands. In summary, we present an EV-based delivery platform for cancer immunotherapeutic ligands that facilitates superior anti-tumor responses at significantly lower doses with fewer side effects than is possible with conventional delivery approaches.
Collapse
Affiliation(s)
- Migara Kavishka Jayasinghe
- Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Yock Sin Lay
- Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Dawn Xiao Tian Liu
- Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Chang Yu Lee
- Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Chang Gao
- Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Brendon Zhijie Yeo
- Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Faith Yuan Xin How
- Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Rebecca Carissa Prajogo
- Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Dong Van Hoang
- Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Hong Anh Le
- Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Thach Tuan Pham
- Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Boya Peng
- Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Cao Dai Phung
- Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Daniel G Tenen
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02138, USA
| | - Minh T N Le
- Institute for Digital Medicine and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Institute of Molecular and Cell Biology, A∗STAR, Singapore 138673, Singapore.
| |
Collapse
|