1
|
Ross AB, Puri K, Johnson TL, Correa LD, John TD, Friend B, Navai SA, Ahmed N, Hegde MG, Omer B, Heczey A, Salem BM, Tcharmtchi H, Rouce RH, Steffin DH, Krance RA, Rainusso NC, Bhar S. Cardiovascular Complications of Immune Effector Cell Therapies in Pediatric Hematological and Solid Tumors. Pediatr Blood Cancer 2025:e31557. [PMID: 39833487 DOI: 10.1002/pbc.31557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/22/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Immune effector cell (IEC) therapies, including chimeric antigen receptor (CAR)-modified T-cell therapy, have shown efficacy in pediatric B-cell acute lymphoblastic leukemia (B-ALL) and are being investigated for other malignancies. A common toxicity associated with IEC therapy is cytokine release syndrome (CRS), which can lead to cardiovascular decompensation due to systemic inflammation. Data are limited regarding cardiovascular adverse effects in children. This study aims to describe the cardiovascular adverse effect profile of IEC therapies in pediatric patients with hematologic and solid tumor malignancies. METHODS We retrospectively reviewed patients who received IECs directed towards various targets in patients with hematologic, solid, and brain tumor malignancies from January 2014 to June 2023 at Texas Children's Hospital. The primary end point was hypotension requiring vasoactive support and/or heart failure within 30 days of infusion. RESULTS A total of 203 patients met inclusion criteria. Pretreatment echocardiogram was available for 142 (70%) pediatric patients, of whom 140 (96%) had normal baseline systolic function. Hypotension requiring vasoactive support occurred in 26 (13%) patients. Hematologic malignancy indications (p = 0.002), total body irradiation (TBI) (p = 0.002), and allogenic hematopoietic stem cell transplants (HCT) (p = 0.035) were associated with increased risk of hypotension requiring vasoactive support. Follow-up echocardiograms were available for 14 patients who met the primary end point, and all showed return to baseline within 6 months. CONCLUSIONS Significant hemodynamic compromise occurred in a minority of patients treated with IEC therapies. All experiencing cardiac dysfunction had recovery of function, and there was no cardiovascular-related mortality.
Collapse
Affiliation(s)
- Aaron B Ross
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Kriti Puri
- Critical Care Medicine, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Heart Center, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Thomas L Johnson
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Lilliam D Correa
- Critical Care Medicine, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Tami D John
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - Brian Friend
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - Shoba A Navai
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Nabil Ahmed
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - Meenakshi G Hegde
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - Bilal Omer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Andras Heczey
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - Baheyeldin M Salem
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - Hossein Tcharmtchi
- Critical Care Medicine, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Rayne H Rouce
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - David H Steffin
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - Robert A Krance
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - Nino C Rainusso
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| | - Saleh Bhar
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Critical Care Medicine, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
2
|
Zhao L, Gui Y, Cai J, Deng X. Biometallic ions and derivatives: a new direction for cancer immunotherapy. Mol Cancer 2025; 24:17. [PMID: 39815289 PMCID: PMC11734411 DOI: 10.1186/s12943-025-02225-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/01/2025] [Indexed: 01/18/2025] Open
Abstract
Biometallic ions play a crucial role in regulating the immune system. In recent years, cancer immunotherapy has become a breakthrough in cancer treatment, achieving good efficacy in a wide range of cancers with its specificity and durability advantages. However, existing therapies still face challenges, such as immune tolerance and immune escape. Biometallic ions (e.g. zinc, copper, magnesium, manganese, etc.) can assist in enhancing the efficacy of immunotherapy through the activation of immune cells, enhancement of tumor antigen presentation, and improvement of the tumor microenvironment. In addition, biometallic ions and derivatives can directly inhibit tumor cell progression and offer the possibility of effectively overcoming the limitations of current cancer immunotherapy by promoting immune responses and reducing immunosuppressive signals. This review explores the role and potential application prospects of biometallic ions in cancer immunotherapy, providing new ideas for future clinical application of metal ions as part of cancer immunotherapy and helping to guide the development of more effective and safe therapeutic regimens.
Collapse
Affiliation(s)
- Lin Zhao
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 41001l, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China
| | - Yajun Gui
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 41001l, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China
| | - Jing Cai
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 41001l, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China
| | - Xiangying Deng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 41001l, China.
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China.
- Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
3
|
Meng F, Xiang M, Liu Y, Zeng D. Safety and efficacy of anti-CD30 CAR-T cell therapy in relapsed/refractory classic Hodgkin lymphoma: a systematic review and meta-analysis. BMC Cancer 2025; 25:78. [PMID: 39806291 PMCID: PMC11731380 DOI: 10.1186/s12885-024-13400-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 12/26/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Relapsed/refractory classic Hodgkin lymphoma (R/R cHL) remains challenging to treat, and anti-CD30 chimeric antigen receptor T (CAR-T) cell therapy may be effective. This meta-analysis investigates the efficacy and safety of anti-CD30 CAR-T cell therapy for treating R/R cHL. METHODS A systematic literature search of PubMed, Cochrane, Embase, ClinicalTrials.gov, and Web of Science databases was conducted until February 2024. The odds ratio (OR) with a 95% confidence interval (CI) was analysed using Review Manager 5.4. Outcomes including overall response rate (ORR), complete response (CR), partial response (PR), progression-free survival (PFS), overall survival (OS), and adverse events (AEs) were extracted for meta-analysis. We used the Methodological Index for Non-Randomized Studies (MINORS) to evaluate the quality of the included literature. RESULTS A total of 151 participants from 8 records were included. Meta-analysis showed the ORR of CD30 CAR-T cell therapy for R/R cHL was 57% (95%CI 0.36-0.76, P = 0.50), with a CR of 34% (95%CI 0.13-0.64, P = 0.29) and a PR of 32% (95%CI 0.15-0.55, P = 0.12). With the median follow-up range from 9.5 to 71.5 months, the 1-year PFS was 39% (95% CI 0.30-0.49, P = 0.04), and the 1-year OS was 89% (95% CI 0.65-0.97, P = 0.005). The most common hematologic AE was leukopenia (72%, 95% CI: 0.50-0.87), and the most common non-hematological AE was cytokine release syndrome (CRS) (43%, 95% CI: 0.14-0.76). The grade ≥ 3 AEs was 66% (95%CI 0.06-0.98, I2 = 93%, P = 0.70), 34% (95%CI 0.07-0.78, I2 = 85%, P = 0.51) in neutropenia and thrombocytopenia, respectively. All AEs were tolerable and resolved with treatment. CONCLUSION Current evidence suggests that anti-CD30 CAR-T cell therapy is effective and safe in treating R/R cHL and is worth considering as a viable therapeutic option.
Collapse
Affiliation(s)
- Fanqiao Meng
- Department of Hematology, Daping Hospital, Third Military Medical University (Army Medical University), No.10, Daping Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Maoyuan Xiang
- Department of Hematology, Daping Hospital, Third Military Medical University (Army Medical University), No.10, Daping Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Yu Liu
- Department of Hematology, Daping Hospital, Third Military Medical University (Army Medical University), No.10, Daping Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Dongfeng Zeng
- Department of Hematology, Daping Hospital, Third Military Medical University (Army Medical University), No.10, Daping Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China.
| |
Collapse
|
4
|
Song F, Tsahouridis O, Stucchi S, Walhart T, Mendell S, Hardy PB, Axtman M, Guduru SKR, Gilbert TSK, Graves LM, Herring LE, Savoldo B, Ma X, Woodcock M, Milner JJ, Ivanova A, Pearce KH, Xu Y, Dotti G. A multi-kinase inhibitor screen identifies inhibitors preserving stem-cell-like chimeric antigen receptor T cells. Nat Immunol 2025:10.1038/s41590-024-02042-1. [PMID: 39779871 DOI: 10.1038/s41590-024-02042-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025]
Abstract
Chimeric antigen receptor T cells (CAR T cells) with T stem (TSCM) cell-like phenotypic characteristics promote sustained antitumor effects. We performed an unbiased and automated high-throughput screen of a kinase-focused compound set to identify kinase inhibitors (KIs) that preserve human TSCM cell-like CAR T cells. We identified three KIs, UNC10225387B, UNC10225263A and UNC10112761A, that combined in vitro increased the frequency of CD45RA+CCR7+TCF1hi TSCM cell-like CAR T cells from both healthy donors and patients with cancer. KI-treated CAR T cells showed enhanced antitumor effects both in vitro and in vivo in mouse tumor models. The KI cocktail maintains TSCM cell-like phenotype preferentially in CAR T cells originating from naive T cells and causes transcriptomic changes without arresting T cell activation or modulating the chromatin organization. Specific kinases, ITK, ADCK3, MAP3K4 and CDK13, targeted by the KI cocktail in a dose-dependent manner are directly associated with the preservation of TSCM cell-like CAR T cells. Knockdown of these kinases individually or in combination enriches for TSCM cell-like CAR T cells, but only CAR T cells generated in the presence of the KI cocktail show robust expansion and differentiation on stimulation with tumor cells. Overall, transient pharmacological inhibition of strategically targeted kinases maintains stem-like features in CAR T cells and improves their antitumor activity.
Collapse
Affiliation(s)
- Feifei Song
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ourania Tsahouridis
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Simone Stucchi
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tara Walhart
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sophie Mendell
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - P Brian Hardy
- Center for Integrative Chemical Biology and Drug Discovery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Matthew Axtman
- Center for Integrative Chemical Biology and Drug Discovery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shiva K R Guduru
- Center for Integrative Chemical Biology and Drug Discovery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Thomas S K Gilbert
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lee M Graves
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Laura E Herring
- Michael Hooker Proteomics Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xingcong Ma
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mark Woodcock
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Oncology division, department of medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Justin J Milner
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anastasia Ivanova
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kenneth H Pearce
- Center for Integrative Chemical Biology and Drug Discovery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yang Xu
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
5
|
Wei W, Cao Y, Lu X, Wang L, Li J, Deng G, Li D, Xiao L. RBM47 is a novel immunotherapeutic target and prognostic biomarker in gliomas. Sci Rep 2025; 15:854. [PMID: 39757245 DOI: 10.1038/s41598-024-84719-z] [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: 09/27/2024] [Accepted: 12/26/2024] [Indexed: 01/07/2025] Open
Abstract
The role of RBM47, an RNA-binding protein, in shaping the immune landscape of gliomas and tumor immune responses is yet to be fully studied. Therefore, a comprehensive investigation into the immunomodulatory roles of RBM47 in gliomas was conducted, leveraging gene expression data from multi-omic datasets. The prognosis of patients with gliomas considering RBM47 was elucidated using bioinformatics methods and clinical data, with results validated using immunohistochemistry and immunofluorescence analyses. The expression of RBM47 in gliomas was higher than that in normal tissues and was positively correlated with the World Health Organization tumor grade. Increased RBM47 expression is associated with poor prognosis in patients with glioma, serving as an independent predictor of overall survival. The nomogram combining RBM47 expression levels with clinical prognostic factors demonstrated strong predictive accuracy, achieving a C-index of up to 0.863 in both TCGA training and CGGA validation groups. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and Gene Set Variation Analysis indicated that RBM47 is closely related to immunity and inflammation. Single-cell sequencing and immunofluorescence assays confirmed the enrichment of RBM47 in CD163 + macrophages. Therefore, RBM47 plays a vital role in the immune microenvironment of gliomas and may be a potential immunotherapy target.
Collapse
Affiliation(s)
- Wei Wei
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yongwai Street, Nanchang, 330006, Jiangxi, China
| | - Yongfu Cao
- Department of Neurosurgery, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong High Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xin Lu
- Department of Neurosurgery, Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Long Wang
- Department of Neurosurgery, Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Jianbin Li
- Department of Neurosurgery, Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Guojun Deng
- Department of Neurosurgery, Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Donghai Li
- Department of Neurosurgery, Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Limin Xiao
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yongwai Street, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
6
|
Hosseinalizadeh H, Wang LS, Mirzaei H, Amoozgar Z, Tian L, Yu J. Emerging combined CAR-NK cell therapies in cancer treatment: Finding a dancing partner. Mol Ther 2025:S1525-0016(24)00895-5. [PMID: 39754357 DOI: 10.1016/j.ymthe.2024.12.057] [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: 03/21/2024] [Revised: 09/21/2024] [Accepted: 12/31/2024] [Indexed: 01/06/2025] Open
Abstract
In recent decades, immunotherapy with chimeric antigen receptors (CARs) has revolutionized cancer treatment and given hope where other cancer therapies have failed. CAR-natural killer (NK) cells are NK cells that have been engineered ex vivo with a CAR on the cell membrane with high specificity for specific target antigens of tumor cells. The impressive results of several studies suggest that CAR-NK cell therapy has significant potential and successful performance in cancer treatment. Despite its effectiveness, CAR-NK cell therapy can have significant challenges when it comes to treating cancer. These challenges include tumor heterogeneity, antigen escape, an immunosuppressive tumor microenvironment, limited tissue migration from blood, exhaustion of CAR-NK cells, and inhibition by immunosuppressive checkpoint molecule signaling, etc. In CAR-T cell therapy, the use of combined approaches has shown encouraging outcomes for tumor regression and improved cancer treatment compared to single therapies. Therefore, to overcome these significant challenges in CAR-NK cells, innovative combination therapies of CAR-NK cells with other conventional therapies (e.g., chemotherapy and radiotherapy) or other immunotherapies are needed to counteract the above challenges and thereby increase the activity of CAR-NK cells. This review comprehensively discusses various cancer-treatment approaches in combination with CAR-NK cell therapy in the hope of providing valuable insights that may improve cancer treatment in the near future.
Collapse
Affiliation(s)
- Hamed Hosseinalizadeh
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Li-Shu Wang
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zohreh Amoozgar
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lei Tian
- Division of Hematology/Oncology, Department of Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA; Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
| | - Jianhua Yu
- Division of Hematology/Oncology, Department of Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA; Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
7
|
Fleischer A, Kurth S, Duell J, Topp M, Strunz PP, Mersi J, Rasche L, Sanges C, Hudecek M, Einsele H, Maatouk I. Neuropsychiatric manifestations following chimeric antigen receptor T cell therapy for cancer: a systematic review of clinical outcomes and management strategies. J Immunother Cancer 2024; 12:e009174. [PMID: 39794934 PMCID: PMC11667355 DOI: 10.1136/jitc-2024-009174] [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: 03/20/2024] [Accepted: 11/15/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-T cell therapy has emerged as a transformative modality in the treatment of patients with cancer. However, it is increasingly evident that this therapeutic approach is not without its challenges. The unique nature of CAR-T cells as living drugs introduces a distinct set of side effects. As the application of CAR-T cell therapy expands to treat a broader range of diseases, it becomes increasingly important to devise effective strategies for handling the associated toxicities. Challenges in treating patients with CAR-T cells include addressing complications such as cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, and cytopenias. This comprehensive review seeks to systematically identify, categorize and elucidate all previously described neurological and psychological side effects associated with CAR-T cell therapy, shedding light on the pertinent laboratory findings that underscore these phenomena. METHODS PubMed, Springer Link, and ScienceDirect were systematically searched for empirical studies on adult patients with cancer receiving CAR-T cell therapy for hemato-oncological malignancies. Quality assessment was conducted using Version 2 of the Cochrane risk-of-bias tool (RoB 2) for randomized trials and adherence to the STROBE (Strengthening the Reporting of Observational Studies in Epidemiology) checklist for observational studies. The synthesis of findings was conducted via a narrative approach, consolidating the diverse array of data into a coherent framework. RESULTS From an initial pool of 2,276 citations, 546 studies met the inclusion criteria, exhibiting a rich tapestry of heterogeneity in terms of study characteristics and patient samples. The incidence of neuropsychological symptoms varied notably across different CAR-T cell products and hematological malignancies. Among the most frequently reported neuropsychological symptoms were aphasia, attention deficits, impaired consciousness, and disorientation, alongside a constellation of other symptoms including confusion, cognitive impairment, memory loss, writing difficulties, fatigue, headache, agitation, tremor, seizures, and psychomotor retardation. Early intervention strategies, including corticosteroids and tocilizumab, have shown the potential to reduce the intensity of neuropsychological symptoms and prevent their progression to critical complications. CONCLUSION These insights underscore the imperative of extending neuropsychological assessments beyond the conventional Immune Effector Cell-Associated Encephalopathy score framework.
Collapse
Affiliation(s)
- Anna Fleischer
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Sophia Kurth
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Johannes Duell
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Max Topp
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | | | - Julia Mersi
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Leo Rasche
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Carmen Sanges
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Michael Hudecek
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| | - Imad Maatouk
- Department of Internal Medicine II, University Hospital Wurzburg, Wurzburg, Germany
| |
Collapse
|
8
|
Hsu CY, Pallathadka H, Jasim SA, Rizaev J, Olegovich Bokov D, Hjazi A, Mahajan S, Mustafa YF, Husseen B, Jawad MA. Innovations in cancer immunotherapy: A comprehensive overview of recent breakthroughs and future directions. Crit Rev Oncol Hematol 2024; 206:104588. [PMID: 39667718 DOI: 10.1016/j.critrevonc.2024.104588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/14/2024] Open
Abstract
A major advance in cancer treatment has been the development and refinement of cancer immunotherapy. The discovery of immunotherapies for a wide range of cancers has revolutionized cancer treatment paradigms. Despite relapse or refractory disease, immunotherapy approaches can prolong the life expectancy of metastatic cancer patients. Multiple therapeutic approaches and agents are currently being developed to manipulate various aspects of the immune system. Oncolytic viruses, cancer vaccines, adoptive cell therapies, monoclonal antibodies, cytokine therapies, and inhibitors of immune checkpoints have all proven successful in clinical trials. There are several types of immunotherapeutic approaches available for treating cancer, and others are being tested in preclinical and clinical settings. Immunotherapy has proven successful, and many agents and strategies have been developed to improve its effectiveness. The purpose of this article is to present a comprehensive overview of current immunotherapy approaches used to treat cancer. Cancer immunotherapy advancements, emerging patterns, constraints, and potential future breakthroughs are also discussed.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, AZ 85004, USA
| | | | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques department, College of Health and medical technology, University of Al-maarif, Anbar, Iraq.
| | - Jasur Rizaev
- Department of Public health and Healthcare management, Rector, Samarkand State Medical University, Samarkand, Uzbekistan
| | - Dmitry Olegovich Bokov
- Institute of Pharmacy named after A.P. Nelyubin, Sechenov First Moscow State Medical University, Russia; Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, Moscow, Russia
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Shriya Mahajan
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab 140417, India
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| | - Beneen Husseen
- Medical laboratory technique college, the Islamic University, Najaf, Iraq; Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| | | |
Collapse
|
9
|
Ansell SM. Hodgkin lymphoma: 2025 update on diagnosis, risk-stratification, and management. Am J Hematol 2024; 99:2367-2378. [PMID: 39239794 DOI: 10.1002/ajh.27470] [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: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/07/2024]
Abstract
DISEASE OVERVIEW Hodgkin lymphoma (HL) is an uncommon B-cell lymphoid malignancy affecting 8570 new patients annually and representing ~10% of all lymphomas in the United States. DIAGNOSIS HL is composed of two distinct disease entities: classical HL and nodular lymphocyte predominant HL (also called nodular lymphocyte predominant B-cell lymphoma). Nodular sclerosis, mixed cellularity, lymphocyte depletion, and lymphocyte-rich HL are subgroups of classical HL. RISK STRATIFICATION An accurate assessment of the stage of disease in patients with HL is critical for the selection of the appropriate therapy. Prognostic models that identify patients at low or high risk for recurrence, as well as the response to therapy as determined by positron emission tomography (PET) scan, are used to optimize therapy. RISK-ADAPTED THERAPY Initial therapy for HL patients is based on the histology of the disease, the anatomical stage and the presence of poor prognostic features. Patients with early-stage disease are typically treated with combined modality strategies utilizing abbreviated courses of combination chemotherapy followed by involved-field radiation therapy, whereas those with advanced stage disease receive a longer course of chemotherapy often without radiation therapy. However, newer agents including brentuximab vedotin and anti-PD-1 antibodies are now standardly incorporated into frontline therapy. MANAGEMENT OF RELAPSED/REFRACTORY DISEASE High-dose chemotherapy (HDCT) followed by an autologous stem cell transplant (ASCT) is the standard of care for most patients who relapse following initial therapy. For patients who fail HDCT with ASCT, brentuximab vedotin, PD-1 blockade, non-myeloablative allogeneic transplant or participation in a clinical trial should be considered.
Collapse
Affiliation(s)
- Stephen M Ansell
- Dorotha W. and Grant L. Sundquist Professor in Hematologic Malignancies Research Chair, Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
10
|
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
|
11
|
Grady CB, Li Y, Maude SL, Hexner EO, Frey NV, Porter DL, Hwang WT. Inconsistent Reporting and Definitions of Time-to-Event Endpoints in CAR T Clinical Trials: A Review. Transplant Cell Ther 2024:S2666-6367(24)00781-4. [PMID: 39603418 DOI: 10.1016/j.jtct.2024.11.012] [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: 08/21/2024] [Revised: 11/05/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024]
Abstract
Clinical trials evaluating chimeric antigen receptor T-cell therapy (CAR T) commonly report time-to-event (TTE) endpoints. However, definitions are not necessarily comparable across studies and variability can lead to misinterpretation of results or inappropriate comparisons across products and studies. Amid the rapidly increasing number of published CAR T trials-many of which were used for regulatory approval-this study aims to summarize the variation in the use and reporting of TTE endpoints in CAR T trials. We include CAR T trials published January 2008 to January 2023 on PubMed that reported at least one of these TTE endpoints: overall survival (OS), progression-free survival (PFS), duration of response/remission (DOR), disease-free survival, event-free survival (EFS), relapse-free survival (RFS), time to relapse, time to progression, or time to treatment failure. We abstracted and summarized endpoint definitions, including the time origin, events, competing events, and censoring. We assessed the completeness of endpoint reporting, overall and by subgroups such as study phase, publication year, and the journal's impact factor. We included 116 publications in the analysis. The most frequently reported TTEs were OS (83%,), PFS (56%), DOR (55%), and EFS (23%). Complete reporting of endpoints was poor overall: 32%, 24%, 25%, and 56% for OS, PFS, DOR, and EFS respectively. Complete reporting was lower in articles published before 2018, in lower impact factor journals, and in phase I trials. There was also a large variability in TTE definitions among those reported. For example, among 64 studies reporting DOR, 48% used the date of response as the time origin while 20% used the date of infusion, and 31% did not report a time origin. There is substantial heterogeneity and incompleteness of TTE endpoint definitions in CAR T trials that could impact the interpretation of the study results. Improving TTE reporting, by stating the time origin, event(s) of interest, competing event(s) if any, and censoring, is required to ensure valid assessment of clinical benefit and cross-trial comparison.
Collapse
Affiliation(s)
- Connor B Grady
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yimei Li
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania; Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shannon L Maude
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania; Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth O Hexner
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine and the Hospital of the University of Pennsylvania, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Noelle V Frey
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine and the Hospital of the University of Pennsylvania, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David L Porter
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine and the Hospital of the University of Pennsylvania, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Wei-Ting Hwang
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
12
|
Zhou D, Zhu X, Xiao Y. Advances in CAR-T therapy for central nervous system tumors. Biomark Res 2024; 12:132. [PMID: 39506843 PMCID: PMC11539471 DOI: 10.1186/s40364-024-00679-6] [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: 09/18/2024] [Accepted: 10/27/2024] [Indexed: 11/08/2024] Open
Abstract
The application of chimeric antigen receptor T-cell therapy in central nervous system tumors has significantly advanced; however, challenges pertaining to the blood-brain barrier, immunosuppressive microenvironment, and antigenic heterogeneity continue to be encountered, unlike its success in hematological malignancies such as acute lymphoblastic leukemia and diffuse large B-cell lymphomas. This review examined the research progress of chimeric antigen receptor T-cell therapy in gliomas, medulloblastomas, and lymphohematopoietic tumors of the central nervous system, focusing on chimeric antigen receptor T-cells targeting antigens such as EGFRvIII, HER2, B7H3, GD2, and CD19 in preclinical and clinical studies. It synthesized current research findings to offer valuable insights for future chimeric antigen receptor T-cell therapeutic strategies for central nervous system tumors and advance the development and application of this therapeutic modality in this domain.
Collapse
Affiliation(s)
- Delian Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Xiaojian Zhu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Yi Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
13
|
El Hussein S, O'Malley DP. Classic Hodgkin lymphoma: An illustrative review of select diagnostic limitations and immunomorphological challenges. Hum Pathol 2024:105678. [PMID: 39491628 DOI: 10.1016/j.humpath.2024.105678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/24/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
The diagnosis of classic Hodgkin lymphoma (CHL) in clinical practice remains reliant on tissue morphological and immunohistochemical evaluation. In this article, we illustrate specific scenarios that we have encountered in our clinical practice pertaining to diagnostic challenges in CHL. We begin with select presentations of morphologic variants of CHL and then discuss certain immunophenotypic deviations from what is deemed "normal patterns" of antigen expression by HRS cells. Lastly, we discuss mimickers of HRS cells, in lymphomatous and non-lymphomatous conditions.
Collapse
Affiliation(s)
- Siba El Hussein
- Department of Pathology, University of Vermont Larner College of Medicine, Burlington, VT, USA.
| | | |
Collapse
|
14
|
Khaliulin M, Valiullina A, Petukhov A, Yuan Y, Spada S, Bulatov E. Breaking the shield of solid tumors: a combined approach for enhanced efficacy of CAR-T cells. Cancer Immunol Immunother 2024; 74:3. [PMID: 39487875 PMCID: PMC11531461 DOI: 10.1007/s00262-024-03817-z] [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: 03/30/2024] [Accepted: 08/22/2024] [Indexed: 11/04/2024]
Abstract
The use of chimeric antigen receptor (CAR)-T cells has enhanced the range of available therapeutic modalities in the context of cancer treatment. CAR-T cells have demonstrated considerable efficacy in the targeted eradication of blood cancer cells, thereby stimulating substantial interest in the advancement of such therapeutic approaches. However, the efficacy of CAR-T cells against solid tumor cells has been limited due to the presence of various obstacles. Solid tumors exhibit antigenic diversity and an immunosuppressive microenvironment, which presents a challenge for immune cells attempting to penetrate the tumor. CAR-T cells also demonstrate decreased proliferative activity and cytotoxicity. Furthermore, concerns exist regarding tumor antigen loss and therapy-associated toxicity. Currently, scientists are working to enhance the structure of the CAR and improve the survival and efficiency of CAR-T cells in recognizing tumor antigens in solid tumors. Chemotherapy drugs are frequently employed in the treatment of malignant neoplasms and can also be used prior to cell therapy to enhance CAR-T cell engraftment. Recent studies have demonstrated that chemotherapy drugs can mitigate the suppressive impact of TME, eliminate the physical barrier by destroying the tumor stroma, and facilitate greater penetration of immune cells and CAR-T cells into the tumor. This, in turn, increases their survival, persistence, and cytotoxicity, as well as affects the metabolism of immune cells inside the tumor. However, the effectiveness of the combined approach against solid tumors depends on several factors, including the type of tumor, dosage, population of CAR-T cells, and individual characteristics of the body. This review examines the principal obstacles to the utilization of CAR-T cells against solid tumors, proposes solutions to these issues, and assesses the potential advantages of a combined approach to radiation exposure, which has the potential to enhance the sensitivity of the tumor to other agents.
Collapse
Affiliation(s)
- Marat Khaliulin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia, 420008
| | - Aygul Valiullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia, 420008
| | - Alexey Petukhov
- Nazarbaev University, Qabanbay Batyr Ave 53, 010000, Astana, Kazakhstan
| | - Youyong Yuan
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, People's Republic of China
| | - Sheila Spada
- Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Emil Bulatov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia, 420008.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia, 117997.
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia, 119048.
| |
Collapse
|
15
|
Kahn J, Dabaja B, Wu S, Kelly K, Berkahn L, Pavlovsky A, Sureda A, LaCasce A. Classic Hodgkin lymphoma. Hematol Oncol 2024; 42:e3239. [PMID: 38037872 DOI: 10.1002/hon.3239] [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: 04/09/2023] [Revised: 10/24/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023]
Abstract
Classic Hodgkin lymphoma (HL) is rare disease, with an incidence of approximately 85,000 patients globally per year and a predilection for adolescents and young adults (ages 15-39). Since the introduction of combination chemotherapy in the 1960's and radiation dating back to the early 1900's, therapeutic options and by extension, clinical outcomes have improved dramatically with 5-year overall survival (OS) approaching 90% today. [1](#ref-0001) Advances in understanding HL biology have additionally facilitated development of targeted agents and immunotherapy which have further improved short and long-term outcomes. Despite continued improvements in up-front and salvage therapy, long-term survivors of HL experience several treatment-associated late toxicities, thus, along with efforts to improve therapeutic efficacy, efforts to reduce late effects remain a high-priority in the field.
Collapse
Affiliation(s)
- Justine Kahn
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, New York, USA
| | - Bouthaina Dabaja
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Susan Wu
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kara Kelly
- Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Leanne Berkahn
- Leukaemia and Blood Cancer New Zealand, Auckland, New Zealand
| | | | - Anna Sureda
- Institut Catala D'Oncologia Badalona, Badalona, Spain
| | - Ann LaCasce
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
16
|
Grégoire C, Coutinho de Oliveira B, Caimi PF, Caers J, Melenhorst JJ. Chimeric antigen receptor T-cell therapy for haematological malignancies: Insights from fundamental and translational research to bedside practice. Br J Haematol 2024; 205:1699-1713. [PMID: 39262037 DOI: 10.1111/bjh.19751] [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: 06/09/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024]
Abstract
Autologous chimeric antigen receptor (CAR) T-cell therapy has revolutionized the treatment of lymphoid malignancies, leading to the approval of CD19-CAR T cells for B-cell lymphomas and acute leukaemia, and more recently, B-cell maturation antigen-CAR T cells for multiple myeloma. The long-term follow-up of patients treated in the early clinical trials demonstrates the possibility for long-term remission, suggesting a cure. This is associated with a low incidence of significant long-term side effects and a rapid improvement in the quality of life for responders. In contrast, other types of immunotherapies require prolonged treatments or carry the risk of long-term side effects impairing the quality of life. Despite impressive results, some patients still experience treatment failure or ultimately relapse, underscoring the imperative to improve CAR T-cell therapies and gain a better understanding of their determinants of efficacy to maximize positive outcomes. While the next-generation of CAR T cells will undoubtingly be more potent, there are already opportunities for optimization when utilizing the currently available CAR T cells. This review article aims to summarize the current evidence from clinical, translational and fundamental research, providing clinicians with insights to enhance their understanding and use of CAR T cells.
Collapse
Affiliation(s)
- Céline Grégoire
- Center for ImmunoTherapy and Precision Immuno-Oncology (CITI), Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Clinical Hematology and Laboratory of Hematology (GIGA I3), University Hospital Center of Liège and University of Liège, Liège, Belgium
| | - Beatriz Coutinho de Oliveira
- Center for ImmunoTherapy and Precision Immuno-Oncology (CITI), Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Paolo F Caimi
- Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio, USA
| | - Jo Caers
- Department of Clinical Hematology and Laboratory of Hematology (GIGA I3), University Hospital Center of Liège and University of Liège, Liège, Belgium
| | - Jan Joseph Melenhorst
- Center for ImmunoTherapy and Precision Immuno-Oncology (CITI), Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
17
|
Fu B, Liu R, Gao G, Lin Z, He A. Mechanisms and salvage treatments in patients with multiple myeloma relapsed post-BCMA CAR-T cell therapy. Front Immunol 2024; 15:1433774. [PMID: 39502704 PMCID: PMC11534873 DOI: 10.3389/fimmu.2024.1433774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has ushered in a new era for the treatment of multiple myeloma (MM). Numerous clinical studies, especially those involving B-cell maturation antigen (BCMA)-directed CAR-T, have shown remarkable efficacy in patients with relapsed or refractory multiple myeloma (R/R MM). However, a considerable number of patients still experience disease recurrence or progression after BCMA CAR-T treatment, which is attributed to various factors, including antigen escape, CAR-T manufacturing factors, T cell exhaustion, inhibitory effects of tumor microenvironment and impact of prior treatments. The scarcity of effective treatment options following post-CAR-T disease recurrence, coupled with the lack of well-established salvage regimens, leaves patients who do relapse facing a bleak prognosis. In recent years, some academic institutions have achieved certain results in salvage treatments of patients with relapse after BCMA CAR-T treatment through secondary infusion of BCMA CAR-T, changing to non-BCMA-directed CAR-T, double-target CAR-T, bispecific antibodies or other novel therapies. This review summarizes the mechanisms of resistance or relapse after BCMA CAR-T administration and the available data on current salvage treatments, hoping to provide ideas for optimizing clinical salvage therapies.
Collapse
Affiliation(s)
- Bingjie Fu
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Rui Liu
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Gongzhizi Gao
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Zujie Lin
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Aili He
- Department of Hematology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- National-Local Joint Engineering Research Center of Biodiagnostics & Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Xi’an Key Laboratory of Hematological Diseases, Xi’an, China
| |
Collapse
|
18
|
Shafagati N, Paul S, Rozati S, Sterling CH. Antibody-Based Therapies for Peripheral T-Cell Lymphoma. Cancers (Basel) 2024; 16:3489. [PMID: 39456582 PMCID: PMC11506347 DOI: 10.3390/cancers16203489] [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: 09/18/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
While antibody-based immunotherapeutic strategies have revolutionized the treatment of B-cell lymphomas, progress in T-cell lymphomas has suffered from suboptimal targets, disease heterogeneity, and limited effective treatment options. Nonetheless, recent advances in our understanding of T-cell biology, the identification of novel targets, and the emergence of new therapies provide hope for the future. In this review, we explore four areas of current and evolving antibody-based strategies for the treatment of peripheral T-cell lymphoma (PTCL): monoclonal antibodies (mAbs), bispecific antibodies (BsAs), chimeric antigen receptor T-cell therapy (CAR-T), and antibody-drug conjugates (ADCs). As part of this discussion, we will also include limitations, lessons learned, and potential future directions.
Collapse
Affiliation(s)
- Nazila Shafagati
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (S.P.); (S.R.)
| | | | | | - Cole H. Sterling
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (S.P.); (S.R.)
| |
Collapse
|
19
|
Andrea AE, Chiron A, Sarrabayrouse G, Bessoles S, Hacein-Bey-Abina S. A structural, genetic and clinical comparison of CAR-T cells and CAR-NK cells: companions or competitors? Front Immunol 2024; 15:1459818. [PMID: 39430751 PMCID: PMC11486669 DOI: 10.3389/fimmu.2024.1459818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/16/2024] [Indexed: 10/22/2024] Open
Abstract
In recent years, following the groundbreaking achievements of chimeric antigen receptor (CAR) T cell therapy in hematological cancers, and advancements in cell engineering technologies, the exploration of other immune cells has garnered significant attention. CAR-Therapy extended beyond T cells to include CAR natural killer (NK) cells and CAR-macrophages, which are firmly established in the clinical trial landscape. Less conventional immune cells are also making their way into the scene, such as CAR mucosal-associated invariant T (MAIT) cells. This progress is advancing precision medicine and facilitating the development of ready-to-use biological treatments. However, in view of the unique features of natural killer cells, adoptive NK cell immunotherapy has emerged as a universal, allogenic, "off-the shelf" therapeutic strategy. CAR-NK cytotoxic cells present targeted tumor specificity but seem to be devoid of the side effects associated with CAR-T cells. CAR-NK cells appear to be potentially promising candidates for cancer immunotherapy. However, their application is hindered by significant challenges, particularly the limited persistence of CAR-NK cells in the body, which poses a hurdle to their sustained effectiveness in treating cancer. Based upon the foregoing, this review discusses the current status and applications of both CAR-T cells and CAR-NK cells in hematological cancers, and provides a comparative analysis of the structure, genetics, and clinical outcomes between these two types of genetically modified immune cells.
Collapse
Affiliation(s)
- Alain E. Andrea
- Department of Biology, Faculty of Arts and Sciences, Saint George University of Beirut, Beirut, Lebanon
| | - Andrada Chiron
- Université Paris Cité, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Unité des Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris Saclay, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France
| | - Guillaume Sarrabayrouse
- Université Paris Cité, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Unité des Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France
| | - Stéphanie Bessoles
- Université Paris Cité, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Unité des Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France
| | - Salima Hacein-Bey-Abina
- Université Paris Cité, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Unité des Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris Saclay, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France
| |
Collapse
|
20
|
Brown MP. CAR spacers: Not just filling space. Mol Ther 2024; 32:3207-3208. [PMID: 39293429 PMCID: PMC11489524 DOI: 10.1016/j.ymthe.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 09/04/2024] [Accepted: 09/04/2024] [Indexed: 09/20/2024] Open
Affiliation(s)
- Michael P Brown
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA 5000, Australia; RAH Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia.
| |
Collapse
|
21
|
Kua L, Ng CH, Tan JW, Tan HC, Seh CC, Wong F, Ong R, Rooney CM, Tan J, Chen Q, Horak ID, Tan KW, Low L. Novel OX40 and 4-1BB derived spacers enhance CD30 CAR activity and safety in CD30 positive lymphoma models. Mol Ther 2024; 32:3504-3521. [PMID: 38946142 PMCID: PMC11489532 DOI: 10.1016/j.ymthe.2024.06.037] [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: 06/22/2023] [Revised: 04/26/2024] [Accepted: 06/28/2024] [Indexed: 07/02/2024] Open
Abstract
The chimeric antigen receptor (CAR) derived from the CD30 specific murine antibody, HRS-3, has produced promising clinical efficacy with a favorable safety profile in the treatment of relapsed or refractory CD30-positive lymphomas. However, persistence of the autologous CAR-T cells was brief, and many patients relapsed a year after treatment. The lack of persistence may be attributed to the use of a wild-type immunoglobulin (Ig)G1 spacer that can associate with Fc receptors. We first identified the cysteine-rich domain (CRD) 5 of CD30 as the primary binding epitope of HRS-3 and armed with this insight, attempted to improve the HRS-3 CAR functionality with a panel of novel spacer designs. We demonstrate that HRS-3 CARs with OX40 and 4-1BB derived spacers exhibited similar anti-tumor efficacy, circumvented interactions with Fc receptors, and secreted lower levels of cytokines in vitro than a CAR employing the IgG1 spacer. Humanization of the HRS-3 scFv coupled with the 4-1BB spacer preserved potent on-target, on-tumor efficacy, and on-target, off-tumor safety. In a lymphoma mouse model of high tumor burden, T cells expressing humanized HRS-3 CD30.CARs with the 4-1BB spacer potently killed tumors with low levels of circulating inflammatory cytokines, providing a promising candidate for future clinical development in the treatment of CD30-positive malignancies.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Cell Line, Tumor
- Disease Models, Animal
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Ki-1 Antigen/immunology
- Ki-1 Antigen/metabolism
- Lymphoma/therapy
- Lymphoma/immunology
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, OX40/metabolism
- Receptors, OX40/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
- Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Lindsay Kua
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | - Chee Hoe Ng
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | - Jin Wei Tan
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | | | | | - Fiona Wong
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | - Richard Ong
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, and Baylor College of Medicine, Houston, TX 77030, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Joel Tan
- Institute for Molecular and Cellular Biology, A∗STAR Singapore 138673, Singapore
| | - Qingfeng Chen
- Institute for Molecular and Cellular Biology, A∗STAR Singapore 138673, Singapore
| | - Ivan D Horak
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | - Kar Wai Tan
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | - Lionel Low
- Tessa Therapeutics Ltd, Singapore 138673, Singapore.
| |
Collapse
|
22
|
Zhou S, Yang Y, Jing Y, Zhu X. Generating advanced CAR-based therapy for hematological malignancies in clinical practice: targets to cell sources to combinational strategies. Front Immunol 2024; 15:1435635. [PMID: 39372412 PMCID: PMC11449748 DOI: 10.3389/fimmu.2024.1435635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has been a milestone breakthrough in the treatment of hematological malignancies, offering an effective therapeutic option for multi-line therapy-refractory patients. So far, abundant CAR-T products have been approved by the United States Food and Drug Administration or China National Medical Products Administration to treat relapsed or refractory hematological malignancies and exhibited unprecedented clinical efficiency. However, there were still several significant unmet needs to be progressed, such as the life-threatening toxicities, the high cost, the labor-intensive manufacturing process and the poor long-term therapeutic efficacy. According to the demands, many researches, relating to notable technical progress and the replenishment of alternative targets or cells, have been performed with promising results. In this review, we will summarize the current research progress in CAR-T eras from the "targets" to "alternative cells", to "combinational drugs" in preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Shu Zhou
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yuhang Yang
- The First Clinical Medical College, Wuhan University, Wuhan, China
| | - Yulu Jing
- The Second Clinical Medical College, Wuhan University, Wuhan, China
| | - Xiaoying Zhu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| |
Collapse
|
23
|
Schroers-Martin JG, Advani R. Integrating Novel Agents Into the Clinical Management of Classic Hodgkin Lymphoma. JCO Oncol Pract 2024:OP2400277. [PMID: 39265129 DOI: 10.1200/op.24.00277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/11/2024] [Accepted: 08/08/2024] [Indexed: 09/14/2024] Open
Abstract
Classic Hodgkin lymphoma (cHL) is highly curable at all stages. Research efforts over the past few decades have largely focused on interim PET-adapted strategies for therapy de-escalation or intensification. The overarching goals have been to increase cure rates, minimize potential therapy-related effects, and optimize survivorship. Better understanding of the biology of cHL has led to the development and approval of effective novel agents including the antibody-drug conjugate brentuximab vedotin and the checkpoint inhibitor immunotherapies. In this review, we discuss recent trial results and how these agents are integrated into clinical practice with the goal of further optimizing outcomes.
Collapse
Affiliation(s)
| | - Ranjana Advani
- Department of Medicine, Division of Oncology, Stanford University Medical Center, Stanford, CA
| |
Collapse
|
24
|
Anderson GSF, Chapman MA. T cell-redirecting therapies in hematological malignancies: Current developments and novel strategies for improved targeting. Mol Ther 2024; 32:2856-2891. [PMID: 39095991 PMCID: PMC11403239 DOI: 10.1016/j.ymthe.2024.07.028] [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: 03/25/2024] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024] Open
Abstract
T cell-redirecting therapies (TCRTs), such as chimeric antigen receptor (CAR) or T cell receptor (TCR) T cells and T cell engagers, have emerged as a highly effective treatment modality, particularly in the B and plasma cell-malignancy setting. However, many patients fail to achieve deep and durable responses; while the lack of truly unique tumor antigens, and concurrent on-target/off-tumor toxicities, have hindered the development of TCRTs for many other cancers. In this review, we discuss the recent developments in TCRT targets for hematological malignancies, as well as novel targeting strategies that aim to address these, and other, challenges.
Collapse
Affiliation(s)
| | - Michael A Chapman
- MRC Toxicology Unit, University of Cambridge, Cambridge CB2 1QR, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0XY, UK; Addenbrooke's Hospital, Cambridge Universities Foundation Trust, Cambridge CB2 0QQ, UK.
| |
Collapse
|
25
|
Spinner MA, Advani RH. Emerging immunotherapies in the Hodgkin lymphoma armamentarium. Expert Opin Emerg Drugs 2024; 29:263-275. [PMID: 38676917 DOI: 10.1080/14728214.2024.2349083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/25/2024] [Indexed: 04/29/2024]
Abstract
INTRODUCTION Brentuximab vedotin and PD-1 inhibitors have improved outcomes for classic Hodgkin lymphoma (cHL), but better therapies are needed for patients who relapse after these agents. Based on an improved understanding of cHL biology, there is a robust pipeline of novel therapies in development. In this review, we highlight emerging immunotherapeutic agents and combinations for cHL. AREAS COVERED We review clinical trials of novel PD-1/PD-L1 inhibitors beyond FDA-approved agents, checkpoint inhibitors targeting CTLA-4, LAG-3, TIM-3, TIGIT, and CD47/SIRPα, PD-1 inhibitor combinations with immunomodulatory agents and epigenetic modifying therapies, antibody-drug conjugates, bispecific antibodies, and cellular therapies including anti-CD30 CAR-T and allogeneic NK cell therapy. We review the key safety and efficacy data from published phase 1-2 studies and highlight trials in progress, including the first phase 3 trial for PD-1 inhibitor-refractory cHL. EXPERT OPINION Many novel immunotherapies hold great promise in cHL. Rational combinations with existing agents and next-generation antibody and CAR-T constructs may improve response rates and durability. Identifying biomarkers of response to these immunotherapies and using more sensitive tools to assess response, such as circulating tumor DNA, may further inform treatment decisions and enable a precision medicine approach in the future.
Collapse
Affiliation(s)
- Michael A Spinner
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Ranjana H Advani
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
26
|
Miller A, Daum R, Wang T, Wu M, Tat C, Pfeiffer T, Navai S, Heczey A, Hegde M, Ahmed N, Whittle SB, Hill L, Martinez C, Krance R, Ramos CA, Rouce RH, Lulla P, Heslop HE, Omer B, Shekar M. Prolonged cytopenias after immune effector cell therapy and lymphodepletion in patients with leukemia, lymphoma and solid tumors. Cytotherapy 2024; 26:1026-1032. [PMID: 38819365 PMCID: PMC11344664 DOI: 10.1016/j.jcyt.2024.04.075] [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: 08/13/2023] [Revised: 04/10/2024] [Accepted: 04/29/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND AIMS The success of chimeric antigen receptor (CAR) T-cell therapy in treating B-cell malignancies has led to the evaluation of CAR T-cells targeting a variety of other malignancies. Although the efficacy of CAR T-cells is enhanced when administered post-lymphodepleting chemotherapy, this can trigger bone marrow suppression and sustained cytopenia after CD19.CAR T-cell therapy. Additionally, systemic inflammation associated with CAR T-cell activity may contribute to myelosuppression. Cytopenias, such as neutropenia and thrombocytopenia, elevate the risk of severe infections and bleeding, respectively. However, data on the incidence of prolonged cytopenias after immune effector therapy in the solid tumor context remain limited. OBJECTIVE We compared the incidence of prolonged cytopenias after immune effector therapy including genetically modified T-cells, virus-specific T-cells (VSTs) and NKT-cells, as well non-gene-modified VSTs for leukemia, lymphoma, and solid tumors (ST) to identify associated risk factors. METHODS A retrospective analysis was conducted of 112 pediatric and adult patients with relapsed and/or refractory cancers who received lymphodepleting chemotherapy followed by immune effector therapy. Patients treated with 13 distinct immune effector cell therapies through 11 single-center clinical trials and 2 commercial products over a 6-year period were categorized into 3 types of malignancies: leukemia, lymphoma and ST. We obtained baseline patient characteristics and adverse events data for each participant, and tracked neutrophil and platelet counts following lymphodepletion. RESULTS Of 112 patients, 104 (92.9%) experienced cytopenias and 88 (79%) experienced severe cytopenias. Patients with leukemia experienced significantly longer durations of severe neutropenia (median duration of 14 days) compared with patients with lymphoma (7 days) or ST (11 days) (P = 0.002). Patients with leukemia also had a higher incidence of severe thrombocytopenia (74.1%), compared with lymphoma (46%, P = 0.03) and ST (14.3%, P < 0.0001). Prolonged cytopenias were significantly associated with disease type (63% of patients with leukemia, 44% of patients with lymphoma, and 22.9% of patients with ST, P = 0.006), prior hematopoietic stem cell transplant (HSCT) (66.7% with prior HSCT versus 38.3% without prior HSCT, P = 0.039), and development of immune effector cell-associated neurotoxicity syndrome (ICANS) (75% with ICANS versus 38% without ICANS, P = 0.027). There was no significant association between prolonged cytopenias and cytokine release syndrome. CONCLUSIONS Immune effector recipients often experience significant cytopenias due to marrow suppression following lymphodepletion regardless of disease, but prolonged severe cytopenias are significantly less common after treatment of patients with lymphoma and solid tumors.
Collapse
Affiliation(s)
- Anne Miller
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Rachel Daum
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dell Medical School, University of Texas at Austin, Austin, TX, USA
| | - Tao Wang
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Mengfen Wu
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Candise Tat
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Thomas Pfeiffer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA; Department of Pediatrics, Division of Hematology/Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Shoba Navai
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Andras Heczey
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Meenakshi Hegde
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Nabil Ahmed
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Sarah B Whittle
- Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - LaQuisa Hill
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Caridad Martinez
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Robert Krance
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Carlos A Ramos
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Rayne H Rouce
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Premal Lulla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Bilal Omer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| | - Meghan Shekar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA; Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA.
| |
Collapse
|
27
|
Dou Z, Bonacci TR, Shou P, Landoni E, Woodcock MG, Sun C, Savoldo B, Herring LE, Emanuele MJ, Song F, Baldwin AS, Wan Y, Dotti G, Zhou X. 4-1BB-encoding CAR causes cell death via sequestration of the ubiquitin-modifying enzyme A20. Cell Mol Immunol 2024; 21:905-917. [PMID: 38937625 PMCID: PMC11291893 DOI: 10.1038/s41423-024-01198-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: 11/03/2023] [Accepted: 06/14/2024] [Indexed: 06/29/2024] Open
Abstract
CD28 and 4-1BB costimulatory endodomains included in chimeric antigen receptor (CAR) molecules play a critical role in promoting sustained antitumor activity of CAR-T cells. However, the molecular events associated with the ectopic and constitutive display of either CD28 or 4-1BB in CAR-T cells have been only partially explored. In the current study, we demonstrated that 4-1BB incorporated within the CAR leads to cell cluster formation and cell death in the forms of both apoptosis and necroptosis in the absence of CAR tonic signaling. Mechanistic studies illustrate that 4-1BB sequesters A20 to the cell membrane in a TRAF-dependent manner causing A20 functional deficiency that in turn leads to NF-κB hyperactivity, cell aggregation via ICAM-1 overexpression, and cell death including necroptosis via RIPK1/RIPK3/MLKL pathway. Genetic modulations obtained by either overexpressing A20 or releasing A20 from 4-1BB by deleting the TRAF-binding motifs of 4-1BB rescue cell cluster formation and cell death and enhance the antitumor ability of 4-1BB-costimulated CAR-T cells.
Collapse
Affiliation(s)
- Zhangqi Dou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | | | - Peishun Shou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Elisa Landoni
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Mark G Woodcock
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Division of Oncology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chuang Sun
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC, USA
| | - Laura E Herring
- Michael Hooker Proteomics Center, Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Michael J Emanuele
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Feifei Song
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Albert S Baldwin
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Yisong Wan
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.
| | - Xin Zhou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
28
|
Gu X, Zhang Y, Zhou W, Wang F, Yan F, Gao H, Wang W. Infusion and delivery strategies to maximize the efficacy of CAR-T cell immunotherapy for cancers. Exp Hematol Oncol 2024; 13:70. [PMID: 39061100 PMCID: PMC11282638 DOI: 10.1186/s40164-024-00542-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has achieved substantial clinical outcomes for tumors, especially for hematological malignancies. However, extending the duration of remission, reduction of relapse for hematological malignancies and improvement of the anti-tumor efficacy for solid tumors are challenges for CAR-T cells immunotherapy. Besides the endeavors to enhance the functionality of CAR-T cell per se, optimization of the infusion and delivery strategies facilitates the breakthrough of the hurdles that limited the efficacy of this cancer immunotherapy. Here, we summarized the infusion and delivery strategies of CAR-T cell therapies under pre-clinical study, clinical trials and on-market status, through which the improvements of safety and efficacy for hematological and solid tumors were analyzed. Of note, novel infusion and delivery strategies, including local-regional infusion, biomaterials bearing the CAR-T cells and multiple infusion technique, overcome many limitations of CAR-T cell therapy. This review provides hints to determine infusion and delivery strategies of CAR-T cell cancer immunotherapy to maximize clinical benefits.
Collapse
Affiliation(s)
- Xinyu Gu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, People's Republic of China
| | - Yalan Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, People's Republic of China
| | - Weilin Zhou
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, People's Republic of China
| | - Fengling Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, People's Republic of China
| | - Feiyang Yan
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, People's Republic of China
| | - Haozhan Gao
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, People's Republic of China
| | - Wei Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|
29
|
Olejarz W, Sadowski K, Szulczyk D, Basak G. Advancements in Personalized CAR-T Therapy: Comprehensive Overview of Biomarkers and Therapeutic Targets in Hematological Malignancies. Int J Mol Sci 2024; 25:7743. [PMID: 39062986 PMCID: PMC11276786 DOI: 10.3390/ijms25147743] [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: 06/30/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy is a novel anticancer therapy using autologous or allogeneic T-cells. To date, six CAR-T therapies for specific B-cell acute lymphoblastic leukemia (B-ALL), non-Hodgkin lymphomas (NHL), and multiple myeloma (MM) have been approved by the Food and Drug Administration (FDA). Significant barriers to the effectiveness of CAR-T therapy include cytokine release syndrome (CRS), neurotoxicity in the case of Allogeneic Stem Cell Transplantation (Allo-SCT) graft-versus-host-disease (GVHD), antigen escape, modest antitumor activity, restricted trafficking, limited persistence, the immunosuppressive microenvironment, and senescence and exhaustion of CAR-Ts. Furthermore, cancer drug resistance remains a major problem in clinical practice. CAR-T therapy, in combination with checkpoint blockades and bispecific T-cell engagers (BiTEs) or other drugs, appears to be an appealing anticancer strategy. Many of these agents have shown impressive results, combining efficacy with tolerability. Biomarkers like extracellular vesicles (EVs), cell-free DNA (cfDNA), circulating tumor (ctDNA) and miRNAs may play an important role in toxicity, relapse assessment, and efficacy prediction, and can be implicated in clinical applications of CAR-T therapy and in establishing safe and efficacious personalized medicine. However, further research is required to fully comprehend the particular side effects of immunomodulation, to ascertain the best order and combination of this medication with conventional chemotherapy and targeted therapies, and to find reliable predictive biomarkers.
Collapse
Affiliation(s)
- Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Karol Sadowski
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Daniel Szulczyk
- Chair and Department of Biochemistry, The Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Grzegorz Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| |
Collapse
|
30
|
Cao F, Xiu Y, Mohnasky M, Serody JS, Armistead P, Dotti G, Smith M, Huggins J, Messina J, Ramachandran B, Saullo J, Stromberg J, Saha MK, Walsh M, Savoldo B, Grover N, Henderson HI, Andermann TM. Infectious Complications Following CD30 Chimeric Antigen Receptor T-Cell Therapy in Adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.10.24310235. [PMID: 39040188 PMCID: PMC11261934 DOI: 10.1101/2024.07.10.24310235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Infections are increasingly recognized as a common complication of chimeric antigen receptor (CAR) T-cell therapy. The incidence of clinically-defined infection after CD19.CAR T-cell therapy for relapsed/refractory lymphoma ranges from 60-90% in the first year after CAR T-cell therapy and is the most common cause for non-relapse mortality. However, infectious risk after CAR T-cell therapy targeting other malignancies is not well understood. Herein, we report for the first time, infectious complications after CD30.CAR T-cell treatment for patients with Hodgkin's lymphoma and peripheral T-cell lymphoma. Since CD30 is only expressed on a subset of activated T and B-cells, we hypothesized that CD30.CAR T-cell patients would have reduced incidence and severity of infections after infusion compared to CD19.CAR T-cell patients. We retrospectively evaluated all 64 patients who received CD30.CAR T-cells at a single institution between 2016-2021, and assessed infections within one year after cell infusion, comparing these data to a contemporary cohort of 50 patients who received CD19.CAR T-cells at the same institution between 2018-2021. 23 CD30.CAR T-cell patients (36%) and 18 CD19.CAR T-cell patients (36%) developed a microbiologically confirmed infection. Infection severity and bacterial infections were higher in the CD19.CAR T-cell group compared to CD30.CAR T-cell recipients who more commonly had grade 1 respiratory viral infections. Our data reflect expected outcomes for severity and infection type in CD19.CAR T-cell patients and provide a benchmark for comparison with the novel CD30.CAR T-cell product. Although our findings require replication in a larger cohort, they have implications for antimicrobial prophylaxis guidelines after CD30.CAR T-cell therapy. KEY POINTS 1) The incidence of infections within the first year after CD30.CAR T-cell therapy was equivalent to that following CD19.CAR T-cell therapy2) Viral infections were more common after CD30.CAR T-cell therapy but bacterial infections predominated after CD19.CAR T-cell therapy.
Collapse
|
31
|
Chang SR, Chou CH, Tu HF, Liu CJ, Chang KW, Lin SC. The expression of immune co-stimulators as a prognostic predictor of head and neck squamous cell carcinomas and oral squamous cell carcinomas. J Dent Sci 2024; 19:1380-1388. [PMID: 39035328 PMCID: PMC11259670 DOI: 10.1016/j.jds.2024.05.004] [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: 04/28/2024] [Revised: 05/06/2024] [Indexed: 07/23/2024] Open
Abstract
Background/purpose T cells require second immune checkpoint molecules for activation and immune memory after antigen presentation. We found that inducible co-stimulator (ICOS) has been a favorable prognostic factor amongst B7 immune checkpoint co-stimulators (ICSs) families in head and neck squamous cell carcinoma (HNSCC) and oral SCC (OSCC). Materials and methods This study analyzed the expression of non-B7 tumor necrosis factor (TNF) superfamily ICSs in the Cancer Genome Atlas (TCGA) HNSCC cohort, our OSCC cohort, and TCGA pan-cancer datasets. The correlation in expression, prognosis, and immune status was assessed. Results The higher expression of CD27, CD30, CD40L, death domain 3 (DR3), and OX40, presumably on the T cell surface, defined better overall survival of HNSCC patients. Besides, CD27, CD30, CD40L, and OX40 were highly correlated with ICOS expression in tumors. CD27, CD40L, and DR3 expression are higher in HPV+ HNSCC tumors than in HPV- tumors. The combined expression level of CD27/OX40 or CD27/CD40L/OX40 enables the potent survival prediction of small, less nodal involvement, early stage, and HPV + tumor subsets. Tumors expressing high CD27, CD30, CD40L, ICOS, and OX40 exhibited enhanced immune cell infiltration. The high correlation in the expression of these ICSs was also noted in the vast majority of tumor types in TCGA datasets. Conclusion The findings of this study not only confirm the potential of the concordant stimulation of CD27, CD30, CD40L, ICOS, and OX40 as a crucial strategy in cancer immunotherapy but also inspire further exploration into the field, highlighting the promising future of cancer treatment.
Collapse
Affiliation(s)
- Shi-Rou Chang
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chung-Hsien Chou
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsi-Feng Tu
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chung-Ji Liu
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Dentistry, Taipei Mackay Memorial Hospital, Taipei, Taiwan
| | - Kuo-Wei Chang
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shu-Chun Lin
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Dentistry, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
32
|
Brudno JN, Kochenderfer JN. Current understanding and management of CAR T cell-associated toxicities. Nat Rev Clin Oncol 2024; 21:501-521. [PMID: 38769449 PMCID: PMC11529341 DOI: 10.1038/s41571-024-00903-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2024] [Indexed: 05/22/2024]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has revolutionized the treatment of several haematological malignancies and is being investigated in patients with various solid tumours. Characteristic CAR T cell-associated toxicities such as cytokine-release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) are now well-recognized, and improved supportive care and management with immunosuppressive agents has made CAR T cell therapy safer and more feasible than it was when the first regulatory approvals of such treatments were granted in 2017. The increasing clinical experience with these therapies has also improved recognition of previously less well-defined toxicities, including movement disorders, immune effector cell-associated haematotoxicity (ICAHT) and immune effector cell-associated haemophagocytic lymphohistiocytosis-like syndrome (IEC-HS), as well as the substantial risk of infection in patients with persistent CAR T cell-induced B cell aplasia and hypogammaglobulinaemia. A more diverse selection of immunosuppressive and supportive-care pharmacotherapies is now being utilized for toxicity management, yet no universal algorithm for their application exists. As CAR T cell products targeting new antigens are developed, additional toxicities involving damage to non-malignant tissues expressing the target antigen are a potential hurdle. Continued prospective evaluation of toxicity management strategies and the design of less-toxic CAR T cell products are both crucial for ongoing success in this field. In this Review, we discuss the evolving understanding and clinical management of CAR T cell-associated toxicities.
Collapse
Affiliation(s)
- Jennifer N Brudno
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - James N Kochenderfer
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
33
|
Bharadwaj S, Lau E, Hamilton MP, Goyal A, Srinagesh H, Jensen A, Lee D, Mallampet J, Elkordy S, Syal S, Patil S, Latchford T, Sahaf B, Arai S, Johnston LJ, Lowsky R, Negrin R, Rezvani AR, Shizuru J, Meyer EH, Shiraz P, Mikkilineni L, Weng WK, Smith M, Sidana S, Muffly L, Maecker HT, Frank MJ, Mackall C, Miklos D, Dahiya S. Bendamustine is a safe and effective lymphodepletion agent for axicabtagene ciloleucel in patients with refractory or relapsed large B-cell lymphoma. J Immunother Cancer 2024; 12:e008975. [PMID: 38955420 PMCID: PMC11218002 DOI: 10.1136/jitc-2024-008975] [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] [Accepted: 06/07/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Fludarabine in combination with cyclophosphamide (FC) is the standard lymphodepletion regimen for CAR T-cell therapy (CAR T). A national fludarabine shortage in 2022 necessitated the exploration of alternative regimens with many centers employing single-agent bendamustine as lymphodepletion despite a lack of clinical safety and efficacy data. To fill this gap in the literature, we evaluated the safety, efficacy, and expansion kinetics of bendamustine as lymphodepletion prior to axicabtagene ciloleucel (axi-cel) therapy. METHODS 84 consecutive patients with relapsed or refractory large B-cell lymphoma treated with axi-cel and managed with a uniform toxicity management plan at Stanford University were studied. 27 patients received alternative lymphodepletion with bendamustine while 57 received FC. RESULTS Best complete response rates were similar (73.7% for FC and 74% for bendamustine, p=0.28) and there was no significant difference in 12-month progression-free survival or overall survival estimates (p=0.17 and p=0.62, respectively). The frequency of high-grade cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome was similar in both the cohorts. Bendamustine cohort experienced lower proportions of hematological toxicities and antibiotic use for neutropenic fever. Immune reconstitution, as measured by quantitative assessment of cellular immunity, was better in bendamustine cohort as compared with FC cohort. CAR T expansion as measured by peak expansion and area under the curve for expansion was comparable between cohorts. CONCLUSIONS Bendamustine is a safe and effective alternative lymphodepletion conditioning for axi-cel with lower early hematological toxicity and favorable immune reconstitution.
Collapse
Affiliation(s)
- Sushma Bharadwaj
- Stanford University School of Medicine, Stanford, California, USA
| | - Eric Lau
- Stanford University School of Medicine, Stanford, California, USA
| | - Mark P Hamilton
- Stanford University School of Medicine, Stanford, California, USA
| | - Anmol Goyal
- Stanford University School of Medicine, Stanford, California, USA
| | - Hrishi Srinagesh
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Dasom Lee
- Stanford University School of Medicine, Stanford, California, USA
| | - Jayasindhu Mallampet
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, California, USA
| | - Sarah Elkordy
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, California, USA
| | - Shriya Syal
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, California, USA
| | - Sunita Patil
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, California, USA
| | | | - Bita Sahaf
- Cancer Institute, Stanford University School of Medicine, Palo Alto, California, USA
| | - Sally Arai
- Stanford University School of Medicine, Stanford, California, USA
| | - Laura J Johnston
- Stanford University School of Medicine, Stanford, California, USA
| | - Robert Lowsky
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Andrew R Rezvani
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Everett H Meyer
- Stanford University School of Medicine, Stanford, California, USA
| | - Parveen Shiraz
- Stanford University School of Medicine, Stanford, California, USA
| | | | - Wen-Kai Weng
- Stanford University School of Medicine, Stanford, California, USA
| | - Melody Smith
- Stanford University School of Medicine, Stanford, California, USA
| | - Surbhi Sidana
- Stanford University School of Medicine, Stanford, California, USA
| | - Lori Muffly
- Stanford University, Palo Alto, California, USA
| | | | - Matthew J Frank
- Stanford University School of Medicine, Stanford, California, USA
| | | | | | - Saurabh Dahiya
- Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
34
|
Pandit S, Agarwalla P, Song F, Jansson A, Dotti G, Brudno Y. Implantable CAR T cell factories enhance solid tumor treatment. Biomaterials 2024; 308:122580. [PMID: 38640784 PMCID: PMC11125516 DOI: 10.1016/j.biomaterials.2024.122580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/11/2024] [Accepted: 04/13/2024] [Indexed: 04/21/2024]
Abstract
Chimeric Antigen Receptor (CAR) T cell therapy has produced revolutionary success in hematological cancers such as leukemia and lymphoma. Nonetheless, its translation to solid tumors faces challenges due to manufacturing complexities, short-lived in vivo persistence, and transient therapeutic impact. We introduce 'Drydux' - an innovative macroporous biomaterial scaffold designed for rapid, efficient in-situ generation of tumor-specific CAR T cells. Drydux expedites CAR T cell preparation with a mere three-day turnaround from patient blood collection, presenting a cost-effective, streamlined alternative to conventional methodologies. Notably, Drydux-enabled CAR T cells provide prolonged in vivo release, functionality, and enhanced persistence exceeding 150 days, with cells transitioning to memory phenotypes. Unlike conventional CAR T cell therapy, which offered only temporary tumor control, equivalent Drydux cell doses induced lasting tumor remission in various animal tumor models, including systemic lymphoma, peritoneal ovarian cancer, metastatic lung cancer, and orthotopic pancreatic cancer. Drydux's approach holds promise in revolutionizing solid tumor CAR T cell therapy by delivering durable, rapid, and cost-effective treatments and broadening patient accessibility to this groundbreaking therapy.
Collapse
Affiliation(s)
- Sharda Pandit
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA; Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Pritha Agarwalla
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA; Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Feifei Song
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anton Jansson
- Department of Product Development, Production and Design, School of Engineering, Jönköping University, Sweden
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yevgeny Brudno
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA; Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
35
|
Mohammadian Gol T, Zahedipour F, Trosien P, Ureña-Bailén G, Kim M, Antony JS, Mezger M. Gene therapy in pediatrics - Clinical studies and approved drugs (as of 2023). Life Sci 2024; 348:122685. [PMID: 38710276 DOI: 10.1016/j.lfs.2024.122685] [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: 01/19/2024] [Revised: 04/17/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
Gene therapy in pediatrics represents a cutting-edge therapeutic strategy for treating a range of genetic disorders that manifest in childhood. Gene therapy involves the modification or correction of a mutated gene or the introduction of a functional gene into a patient's cells. In general, it is implemented through two main modalities namely ex vivo gene therapy and in vivo gene therapy. Currently, a noteworthy array of gene therapy products has received valid market authorization, with several others in various stages of the approval process. Additionally, a multitude of clinical trials are actively underway, underscoring the dynamic progress within this field. Pediatric genetic disorders in the fields of hematology, oncology, vision and hearing loss, immunodeficiencies, neurological, and metabolic disorders are areas for gene therapy interventions. This review provides a comprehensive overview of the evolution and current progress of gene therapy-based treatments in the clinic for pediatric patients. It navigates the historical milestones of gene therapies, currently approved gene therapy products by the U.S. Food and Drug Administration (FDA) and/or European Medicines Agency (EMA) for children, and the promising future for genetic disorders. By providing a thorough compilation of approved gene therapy drugs and published results of completed or ongoing clinical trials, this review serves as a guide for pediatric clinicians to get a quick overview of the situation of clinical studies and approved gene therapy products as of 2023.
Collapse
Affiliation(s)
- Tahereh Mohammadian Gol
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Fatemeh Zahedipour
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Paul Trosien
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Guillermo Ureña-Bailén
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Miso Kim
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Justin S Antony
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Markus Mezger
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
36
|
Quach DH, Ganesh HR, Briones YD, Nouraee N, Ma A, Hadidi YF, Sharma S, Rooney CM. Rejection resistant CD30.CAR-modified Epstein-Barr virus-specific T cells as an off-the-shelf platform for CD30 + lymphoma. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200814. [PMID: 38966037 PMCID: PMC11223124 DOI: 10.1016/j.omton.2024.200814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/14/2024] [Accepted: 05/10/2024] [Indexed: 07/06/2024]
Abstract
Off-the-shelf (OTS) adoptive T cell therapies have many benefits such as immediate availability, improved access and reduced cost, but face the major challenges of graft-vs-host disease (GVHD) and graft rejection, mediated by alloreactive T cells present in the graft and host, respectively. We have developed a platform for OTS T cell therapies by using Epstein-Bar virus (EBV)-specific T cells (EBVSTs) expressing a chimeric antigen receptor (CAR) targeting CD30. Allogeneic EBVSTs have not caused GVHD in several clinical trials, while the CD30.CAR, that is effective for the treatment of lymphoma, can also target alloreactive T cells that upregulate CD30 on activation. Although EBVSTs express high levels of CD30, they were protected from fratricide in cis, by the CD30.CAR. Hence, they could proliferate extensively and maintained function both through their native EBV-specific T cell receptor and the CD30.CAR. The CD30.CAR enabled EBVSTs to persist in co-cultures with naive and primed alloreactive T cells and eliminate activated natural killer cells that can also be alloreactive. In conclusion, we show that CD30.CAR EBVSTs have the potential to be an effective OTS therapy against CD30+ tumors and, if successful, could then be used as a platform to target other tumor antigens.
Collapse
Affiliation(s)
- David H. Quach
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Haran R. Ganesh
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - Yolanda D. Briones
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - Nazila Nouraee
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - Audrey Ma
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - Yezan F. Hadidi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
| | - Sandhya Sharma
- Graduate program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cliona M. Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital, and Houston Methodist Hospital, Houston, TX 77030, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston TX 77030, USA
- Department of Molecular Virology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
37
|
Guruprasad P, Carturan A, Zhang Y, Cho JH, Kumashie KG, Patel RP, Kim KH, Lee JS, Lee Y, Kim JH, Chung J, Joshi A, Cohen I, Shestov M, Ghilardi G, Harris J, Pajarillo R, Angelos M, Lee YG, Liu S, Rodriguez J, Wang M, Ballard HJ, Gupta A, Ugwuanyi OH, Hong SJA, Bochi-Layec AC, Sauter CT, Chen L, Paruzzo L, Kammerman S, Shestova O, Liu D, Vella LA, Schuster SJ, Svoboda J, Porazzi P, Ruella M. The BTLA-HVEM axis restricts CAR T cell efficacy in cancer. Nat Immunol 2024; 25:1020-1032. [PMID: 38831106 DOI: 10.1038/s41590-024-01847-4] [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: 05/08/2023] [Accepted: 04/17/2024] [Indexed: 06/05/2024]
Abstract
The efficacy of T cell-based immunotherapies is limited by immunosuppressive pressures in the tumor microenvironment. Here we show a predominant role for the interaction between BTLA on effector T cells and HVEM (TNFRSF14) on immunosuppressive tumor microenvironment cells, namely regulatory T cells. High BTLA expression in chimeric antigen receptor (CAR) T cells correlated with poor clinical response to treatment. Therefore, we deleted BTLA in CAR T cells and show improved tumor control and persistence in models of lymphoma and solid malignancies. Mechanistically, BTLA inhibits CAR T cells via recruitment of tyrosine phosphatases SHP-1 and SHP-2, upon trans engagement with HVEM. BTLA knockout thus promotes CAR signaling and subsequently enhances effector function. Overall, these data indicate that the BTLA-HVEM axis is a crucial immune checkpoint in CAR T cell immunotherapy and warrants the use of strategies to overcome this barrier.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Cell Line, Tumor
- Immunotherapy, Adoptive/methods
- Mice, Knockout
- Neoplasms/immunology
- Neoplasms/therapy
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/genetics
- Receptors, Tumor Necrosis Factor, Member 14/metabolism
- Receptors, Tumor Necrosis Factor, Member 14/immunology
- Receptors, Tumor Necrosis Factor, Member 14/genetics
- Signal Transduction
- T-Lymphocytes, Regulatory/immunology
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Puneeth Guruprasad
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Alberto Carturan
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Yunlin Zhang
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Jong Hyun Cho
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | | | - Ruchi P Patel
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Ki-Hyun Kim
- R&D Center, AbClon Inc., Seoul, Republic of Korea
| | - Jong-Seo Lee
- R&D Center, AbClon Inc., Seoul, Republic of Korea
| | - Yoon Lee
- R&D Center, AbClon Inc., Seoul, Republic of Korea
| | | | - Junho Chung
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Akshita Joshi
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Ivan Cohen
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Maksim Shestov
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Guido Ghilardi
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Jaryse Harris
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Raymone Pajarillo
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Mathew Angelos
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Yong Gu Lee
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- College of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Republic of Korea
| | - Shan Liu
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jesse Rodriguez
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael Wang
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Hatcher J Ballard
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Aasha Gupta
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Ositadimma H Ugwuanyi
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Seok Jae Albert Hong
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Audrey C Bochi-Layec
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher T Sauter
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Linhui Chen
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Luca Paruzzo
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Shane Kammerman
- Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Olga Shestova
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Dongfang Liu
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Laura A Vella
- Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Stephen J Schuster
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Jakub Svoboda
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrizia Porazzi
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Marco Ruella
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
38
|
Shmidt D, Mamonkin M. CAR T Cells in T Cell Acute Lymphoblastic Leukemia and Lymphoblastic Lymphoma. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024:S2152-2650(24)00211-8. [PMID: 38955579 DOI: 10.1016/j.clml.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 05/28/2024] [Indexed: 07/04/2024]
Abstract
Chimeric antigen receptor (CAR T) therapy produced excellent activity in patients with relapsed/refractory B-lineage malignancies. However, extending these therapies to T cell cancers requires overcoming unique challenges. In the recent years, multiple approaches have been developed in preclinical models and some were tested in clinical trials in patients with treatment-refractory T-cell malignanices with promising early results. Here, we review main hurdles impeding the success of CAR T therapy in T-cell acute lymphoblastic leukemia/lymphoma (T-ALL/LBL), discuss potential solutions, and summarize recent progress in both preclinical and clinical development of CAR T therapy for these diseases.
Collapse
Affiliation(s)
- Daniil Shmidt
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Maksim Mamonkin
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX.
| |
Collapse
|
39
|
Heneghan MB, Belsky JA, Milgrom SA, Forlenza CJ. The pediatric approach to Hodgkin lymphoma. Semin Hematol 2024:S0037-1963(24)00061-1. [PMID: 38851951 DOI: 10.1053/j.seminhematol.2024.05.003] [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: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 06/10/2024]
Abstract
Hodgkin lymphoma (HL) occurs throughout the lifespan but is one of the most common cancers in adolescents and young adults (AYA; 15-39 years). HL has become a highly curable disease with survival rates surpassing 90%, including patients with high-risk and advanced stage disease. Unfortunately, intensive treatment carries a risk of short- and long-term toxicity. Given the decades pediatric HL survivors are expected to live after treatment, the pediatric approach to treatment has focused on improving the therapeutic index through response adapted treatment and more recently the incorporation of novel agents. The efforts of pediatric and medical oncologists in research and clinical trial development have long occurred in parallel, but recent efforts have laid the foundation for collaboration with the goal of standardizing AYA care and allowing earlier incorporation of novel therapy for younger patients. This review focuses on the evolution of the management of pediatric HL including epidemiology, biology, and approaches to upfront and salvage treatment regimens.
Collapse
Affiliation(s)
- Mallorie B Heneghan
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Utah/Primary Children's Hospital, Salt Lake City, UT.
| | - Jennifer A Belsky
- Department of Pediatrics, Riley Hospital for Children/Indiana University School of Medicine, Indianapolis, IN
| | - Sarah A Milgrom
- Department or Radiation Oncology, University of Colorado School of Medicine, Aurora, CO
| | | |
Collapse
|
40
|
Benevolo Savelli C, Bisio M, Legato L, Fasano F, Santambrogio E, Nicolosi M, Morra D, Boccomini C, Freilone R, Botto B, Novo M. Advances in Hodgkin Lymphoma Treatment: From Molecular Biology to Clinical Practice. Cancers (Basel) 2024; 16:1830. [PMID: 38791909 PMCID: PMC11120540 DOI: 10.3390/cancers16101830] [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: 04/06/2024] [Revised: 05/05/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Classical Hodgkin Lymphoma (cHL) is a highly curable disease, but around 20% of patients experience progression or relapse after standard frontline chemotherapy regimens. Salvage regimens followed by autologous stem cell transplants represent the historical treatment approach for these cases. In the last decade, with the increasing understanding of cHL biology and tumor microenvironment role in disease course, novel molecules have been introduced in clinical practice, improving outcomes in the relapsed/refractory setting. The anti-CD30 antibody-drug conjugated brentuximab vedotin and PD-1/PD-L1 checkpoint inhibitors represent nowadays curative options for chemorefractory patients, and randomized trials recently demonstrated their efficacy in frontline immune-chemo-combined modalities. Several drugs able to modulate the patients' T-lymphocytes and NK cell activity are under development, as well as many anti-CD30 chimeric antigen receptor T-cell products. Multiple tumor aberrant epigenetic mechanisms are being investigated as targets for antineoplastic compounds such as histone deacetylase inhibitors and hypomethylating agents. Moreover, JAK2 inhibition combined with anti-PD1 blockade revealed a potential complementary therapeutic pathway in cHL. In this review, we will summarize recent findings on cHL biology and novel treatment options clinically available, as well as promising future perspectives in the field.
Collapse
Affiliation(s)
- Corrado Benevolo Savelli
- Hematology Division, A.O.U. Città della Salute e della Scienza di Torino, C.so Bramante 88, 10126 Turin, Italy; (M.B.); (L.L.); (F.F.); (E.S.); (M.N.); (D.M.); (C.B.); (R.F.); (B.B.)
| | | | | | | | | | | | | | | | | | | | - Mattia Novo
- Hematology Division, A.O.U. Città della Salute e della Scienza di Torino, C.so Bramante 88, 10126 Turin, Italy; (M.B.); (L.L.); (F.F.); (E.S.); (M.N.); (D.M.); (C.B.); (R.F.); (B.B.)
| |
Collapse
|
41
|
Veyri M. The value of anti-CD30 CAR T cells in Hodgkin lymphoma. Lancet Haematol 2024; 11:e314-e316. [PMID: 38555922 DOI: 10.1016/s2352-3026(24)00094-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 04/02/2024]
Affiliation(s)
- Marianne Veyri
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Sante Publique (IPLESP), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Service d'Oncologie Médicale, Paris 75013, France.
| |
Collapse
|
42
|
Grover NS, Hucks G, Riches ML, Ivanova A, Moore DT, Shea TC, Seegars MB, Armistead PM, Kasow KA, Beaven AW, Dittus C, Coghill JM, Jamieson KJ, Vincent BG, Wood WA, Cheng C, Morrison JK, West J, Cavallo T, Dotti G, Serody JS, Savoldo B. Anti-CD30 CAR T cells as consolidation after autologous haematopoietic stem-cell transplantation in patients with high-risk CD30 + lymphoma: a phase 1 study. Lancet Haematol 2024; 11:e358-e367. [PMID: 38555923 PMCID: PMC11238265 DOI: 10.1016/s2352-3026(24)00064-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T cells targeting CD30 are safe and have promising activity when preceded by lymphodepleting chemotherapy. We aimed to determine the safety of anti-CD30 CAR T cells as consolidation after autologous haematopoietic stem-cell transplantation (HSCT) in patients with CD30+ lymphoma at high risk of relapse. METHODS This phase 1 dose-escalation study was performed at two sites in the USA. Patients aged 3 years and older, with classical Hodgkin lymphoma or non-Hodgkin lymphoma with CD30+ disease documented by immunohistochemistry, and a Karnofsky performance score of more than 60% planned for autologous HSCT were eligible if they were considered high risk for relapse as defined by primary refractory disease or relapse within 12 months of initial therapy or extranodal involvement at the start of pre-transplantation salvage therapy. Patients received a single infusion of CAR T cells (2 × 107 CAR T cells per m2, 1 × 108 CAR T cells per m2, or 2 × 108 CAR T cells per m2) as consolidation after trilineage haematopoietic engraftment (defined as absolute neutrophil count ≥500 cells per μL for 3 days, platelet count ≥25 × 109 platelets per L without transfusion for 5 days, and haemoglobin ≥8 g/dL without transfusion for 5 days) following carmustine, etoposide, cytarabine, and melphalan (BEAM) and HSCT. The primary endpoint was the determination of the maximum tolerated dose, which was based on the rate of dose-limiting toxicity in patients who received CAR T-cell infusion. This study is registered with ClinicalTrials.gov (NCT02663297) and enrolment is complete. FINDINGS Between June 7, 2016, and Nov 30, 2020, 21 patients were enrolled and 18 patients (11 with Hodgkin lymphoma, six with T-cell lymphoma, one with grey zone lymphoma) were infused with anti-CD30 CAR T cells at a median of 22 days (range 16-44) after autologous HSCT. There were no dose-limiting toxicities observed, so the highest dose tested, 2 × 108 CAR T cells per m2, was determined to be the maximum tolerated dose. One patient had grade 1 cytokine release syndrome. The most common grade 3-4 adverse events were lymphopenia (two [11%] of 18) and leukopenia (two [11%] of 18). There were no treatment-related deaths. Two patients developed secondary malignancies approximately 2 years and 2·5 years following treatment (one stage 4 non-small cell lung cancer and one testicular cancer), but these were judged unrelated to treatment. At a median follow-up of 48·2 months (IQR 27·5-60·7) post-infusion, the median progression-free survival for all treated patients (n=18) was 32·3 months (95% CI 4·6 months to not estimable) and the median progression-free survival for treated patients with Hodgkin lymphoma (n=11) has not been reached. The median overall survival for all treated patients has not been reached. INTERPRETATION Anti-CD30 CAR T-cell infusion as consolidation after BEAM and autologous HSCT is safe, with low rates of toxicity and encouraging preliminary activity in patients with Hodgkin lymphoma at high risk of relapse, highlighting the need for larger studies to confirm these findings. FUNDING National Heart Lung and Blood Institute, University Cancer Research Fund at the Lineberger Comprehensive Cancer Center.
Collapse
Affiliation(s)
- Natalie S Grover
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - George Hucks
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Pediatrics, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - Marcie L Riches
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - Anastasia Ivanova
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Biostatistics, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - Dominic T Moore
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Biostatistics, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - Thomas C Shea
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - Mary Beth Seegars
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
| | - Paul M Armistead
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - Kimberly A Kasow
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Pediatrics, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - Anne W Beaven
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - Christopher Dittus
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - James M Coghill
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - Katarzyna J Jamieson
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - Benjamin G Vincent
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Program in Computational Medicine, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - William A Wood
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - Catherine Cheng
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - Julia Kaitlin Morrison
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - John West
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - Tammy Cavallo
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - Jonathan S Serody
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Program in Computational Medicine, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA; Department of Pediatrics, University of North Carolina at Chapel Hill, Chapell Hill, NC, USA.
| |
Collapse
|
43
|
Massaro F, Andreozzi F, Abrassart T, Castiaux J, Massa H, Rizzo O, Vercruyssen M. Beyond Chemotherapy: Present and Future Perspectives in the Treatment of Lymphoproliferative Disorders. Biomedicines 2024; 12:977. [PMID: 38790939 PMCID: PMC11117538 DOI: 10.3390/biomedicines12050977] [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: 03/27/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
Over the past three decades, the treatment of lymphoproliferative disorders has undergone profound changes, notably due to the increasing availability of innovative therapies with the potential to redefine clinical management paradigms. A major impact is related to the development of monoclonal antibodies, checkpoint inhibitors, bispecific antibodies, and chimeric antigen receptor T (CAR-T) cell therapies. This review discusses the current landscape of clinical trials targeting various hematological malignancies, highlighting promising early-phase results and strategies to overcome resistance. Lymphoproliferative disorders encompass a range of conditions: while in Hodgkin lymphoma (HL) the goal is to reduce chemotherapy-related toxicity by integrating immunotherapy into the frontline setting, peripheral T cell lymphoma (PTCL) lacks effective targeted therapies. The review emphasizes a shifting therapeutic landscape towards precision medicine and treatment modalities that are less toxic yet more effective.
Collapse
Affiliation(s)
- Fulvio Massaro
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium; (F.A.); (T.A.); (J.C.); (H.M.); (O.R.); (M.V.)
| | | | | | | | | | | | | |
Collapse
|
44
|
Pang Y, Ghosh N. Novel and multiple targets for chimeric antigen receptor-based therapies in lymphoma. Front Oncol 2024; 14:1396395. [PMID: 38711850 PMCID: PMC11070555 DOI: 10.3389/fonc.2024.1396395] [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: 03/05/2024] [Accepted: 04/08/2024] [Indexed: 05/08/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy targeting CD19 in B-cell non-Hodgkin lymphoma (NHL) validates the utility of CAR-based therapy for lymphomatous malignancies. Despite the success, treatment failure due to CD19 antigen loss, mutation, or down-regulation remains the main obstacle to cure. On-target, off-tumor effect of CD19-CAR T leads to side effects such as prolonged B-cell aplasia, limiting the application of therapy in indolent diseases such as chronic lymphocytic leukemia (CLL). Alternative CAR targets and multi-specific CAR are potential solutions to improving cellular therapy outcomes in B-NHL. For Hodgkin lymphoma and T-cell lymphoma, several cell surface antigens have been studied as CAR targets, some of which already showed promising results in clinical trials. Some antigens are expressed by different lymphomas and could be used for designing tumor-agnostic CAR. Here, we reviewed the antigens that have been studied for novel CAR-based therapies, as well as CARs designed to target two or more antigens in the treatment of lymphoma.
Collapse
Affiliation(s)
- Yifan Pang
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Wake Forest School of Medicine, Charlotte, NC, United States
| | | |
Collapse
|
45
|
Xiong Q, Wang H, Shen Q, Wang Y, Yuan X, Lin G, Jiang P. The development of chimeric antigen receptor T-cells against CD70 for renal cell carcinoma treatment. J Transl Med 2024; 22:368. [PMID: 38637886 PMCID: PMC11025280 DOI: 10.1186/s12967-024-05101-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/15/2024] [Indexed: 04/20/2024] Open
Abstract
In this study, we investigated CD70 as a promising target for renal cell carcinoma (RCC) therapy and developed a potent chimeric antigen receptor T (CAR-T) cells for potential clinical testing. CD70, found to be highly expressed in RCC tumors, was associated with decreased survival. We generated CAR-T cells expressing VHH sequence of various novel nanobodies from immunized alpaca and a single-chain variable fragment (scFv) derived from human antibody (41D12). In our in vitro experiments, anti-CD70 CAR-T cells effectively eliminated CD70-positive tumor cells while sparing CD70-negative cells. The nanobody-based CAR-T cells demonstrated significantly higher production of cytokines such as IL-2, IFN-γ and TNF-ɑ during co-culture, indicating their potential for enhanced functionality. In xenograft mouse model, these CAR-T cells exhibited remarkable anti-tumor activity, leading to the eradication of RCC tumor cells. Importantly, human T cell expansion after infusion was significantly higher in the VHH groups compared to the scFv CAR-T group. Upon re-challenging mice with RCC tumor cells, the VHH CAR-T treated group remained tumor-free, suggesting a robust and long-lasting anti-tumor response. These findings provide strong support for the potential of nanobody-based CD70 CAR-T cells as a promising therapeutic option for RCC. This warrants further development and consideration for future clinical trials and applications.
Collapse
Affiliation(s)
- Qinghui Xiong
- Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Shanghai, 201203, China.
| | - Haiying Wang
- Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Shanghai, 201203, China.
| | - Qiushuang Shen
- Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Shanghai, 201203, China
| | - Yan Wang
- Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Shanghai, 201203, China
| | - Xiujie Yuan
- Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Shanghai, 201203, China
| | - Guangyao Lin
- Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Shanghai, 201203, China
| | - Pengfei Jiang
- Shanghai HRAIN Biotechnology Co., Ltd., 1238 Zhangjiang Road, Shanghai, 201203, China
| |
Collapse
|
46
|
Khaliulin MR, Safin RN, Kunst MA, Bulatov ER. The use of T-cells with chimeric antigen receptor (CAR-T) in combination with chemotherapy and radiotherapy for the treatment of solid tumors. ADVANCES IN MOLECULAR ONCOLOGY 2024; 11:31-45. [DOI: 10.17650/2313-805x-2024-11-1-31-45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The introduction of chimeric antigen receptor (CAR) T-cell therapy has revolutionized the treatment of hematological diseases, particularly in combating blood cancer. The success of this cell therapy approach has led to the development of approximately seven commercial CAR-T based drugs. However, the application of CAR-T therapy for solid tumors has proven to be less effective due to challenges such as the varied antigens in solid tumors, an immunosuppressive tumor environment, limited immune cell infiltration, reduced CAR-T cell activity and toxicity issues. To solve these problems, scientists are making efforts to improve and improve the methods of treatment of solid tumors. Chemotherapy is the standard treatment for a large number of malignant neoplasms. It is also used before starting cell therapy for lymphodepletion and better engraftment of injected CAR-T cells. It has been shown that chemotherapy can reduce the immunosuppressive effect of the tumor microenvironment, destroy the stroma, and promote better infiltration of the tumor by CAR-T cells, improving their survival, persistence, cytotoxicity, and influencing the metabolism of immune cells inside the tumor. The effectiveness of combining chemotherapy and CAR-T cell therapy relies on various factors such as tumor type, dosage, treatment schedule, CAR-T cell composition, and individual biological traits. Similarly, radiation therapy can enhance tumor cell vulnerability to specific treatments while also supporting tumor cell survival.In this review, we discuss the use of CAR-T therapy to combat solid tumors, regarding the challenges of treating solid tumors, ways to overcome them, and also touch upon the possibility of using combination treatments to improve the effectiveness of cell therapy.
Collapse
Affiliation(s)
| | - R. N. Safin
- Republican Clinical Oncology Dispensary named after Prof. M.Z. Sigal Russia
| | - M. A. Kunst
- Republican Clinical Hospital of the Ministry of Health of the Republic of Tatarstan
| | - E. R. Bulatov
- Kazan (Volga Region) Federal University; Shemyakin–Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences
| |
Collapse
|
47
|
Zhang W, Liu M, Li W, Song Y. Immune cells in the B-cell lymphoma microenvironment: From basic research to clinical applications. Chin Med J (Engl) 2024; 137:776-790. [PMID: 38269619 PMCID: PMC10997228 DOI: 10.1097/cm9.0000000000002919] [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: 08/29/2023] [Indexed: 01/26/2024] Open
Abstract
ABSTRACT B-cell lymphoma is a group of hematological malignancies characterized by variable genetic and biological features and clinical behaviors. The tumor microenvironment (TME) is a complex network in tumors, which consists of surrounding blood vessels, extracellular matrix, immune and non-immune cells, and signaling molecules. Increasing evidence has shown that the TME, especially immune cells within, is a double-edged sword, acting either as a tumor killer or as a promoter of tumor progression. These pro-tumor activities are driven by subpopulations of immune cells that express typical markers but have unique transcriptional characteristics, making tumor-associated immune cells good targets for human anti-cancer therapy by ablating immunosuppressive cells or enhancing immune-activated cells. Thus, exploring the role of immune cells in the TME provides distinct insights for immunotherapy in B-cell lymphoma. In this review, we elucidated the interaction between immune cells and tumor cells and their function in the initiation, progression, and prognosis of B-cell lymphoma, from preclinical experiments to clinical trials. Furthermore, we outlined potential therapeutic approaches and discussed the potential clinical value and future perspectives of targeting immune cells in patients with B-cell lymphoma.
Collapse
Affiliation(s)
- Wenli Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, Henan 450000, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Mengmeng Liu
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Research and Foreign Affairs, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Wei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, Henan 450000, China
| | - Yongping Song
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, Henan 450000, China
| |
Collapse
|
48
|
Saha P, Ettel P, Weichhart T. Leveraging macrophage metabolism for anticancer therapy: opportunities and pitfalls. Trends Pharmacol Sci 2024; 45:335-349. [PMID: 38494408 DOI: 10.1016/j.tips.2024.02.005] [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: 01/26/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/19/2024]
Abstract
Tumor-associated macrophages (TAMs) constitute an important part of the tumor microenvironment (TME) that regulates tumor progression. Tumor-derived signals, hypoxia, and competition for nutrients influence TAMs to reprogram their cellular metabolism. This altered metabolic profile creates a symbiotic communication between tumor and other immune cells to support tumor growth. In addition, the metabolic profile of TAMs regulates the expression of immune checkpoint molecules. The dynamic plasticity also allows TAMs to reshape their metabolism in response to modern therapeutic strategies. Therefore, over the years, a significant number of approaches have been implicated to reprogram cancer-promoting metabolism in TAMs. In this review, we discuss the current strategies and pitfalls, along with upcoming promising opportunities in leveraging TAM metabolism for developing better therapeutic approaches against cancer.
Collapse
Affiliation(s)
- Piyal Saha
- Institute for Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Straße 10, 1090 Vienna, Austria
| | - Paul Ettel
- Institute for Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Straße 10, 1090 Vienna, Austria
| | - Thomas Weichhart
- Institute for Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Straße 10, 1090 Vienna, Austria.
| |
Collapse
|
49
|
Ladbury C, Hao C, Mei M, Herrera A, Green G, Dandapani S. Role of Radiation in Combination With CD30-Directed Chimeric Antigen Receptor T-Cell Therapy for Relapsed/Refractory Hodgkin Lymphoma. Adv Radiat Oncol 2024; 9:101428. [PMID: 38406393 PMCID: PMC10882125 DOI: 10.1016/j.adro.2023.101428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 12/14/2023] [Indexed: 02/27/2024] Open
Affiliation(s)
- Colton Ladbury
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, California
| | - Claire Hao
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, California
| | - Matthew Mei
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California
| | - Alex Herrera
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California
| | - Garth Green
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, California
| | - Savita Dandapani
- Department of Radiation Oncology, City of Hope National Medical Center, Duarte, California
| |
Collapse
|
50
|
Hill LC, Rouce RH, Wu MJ, Wang T, Ma R, Zhang H, Mehta B, Lapteva N, Mei Z, Smith TS, Yang L, Srinivasan M, Burkhardt PM, Ramos CA, Lulla P, Arredondo M, Grilley B, Heslop HE, Brenner MK, Mamonkin M. Antitumor efficacy and safety of unedited autologous CD5.CAR T cells in relapsed/refractory mature T-cell lymphomas. Blood 2024; 143:1231-1241. [PMID: 38145560 PMCID: PMC10997912 DOI: 10.1182/blood.2023022204] [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: 09/13/2023] [Revised: 11/15/2023] [Accepted: 11/27/2023] [Indexed: 12/27/2023] Open
Abstract
ABSTRACT Despite newer targeted therapies, patients with primary refractory or relapsed (r/r) T-cell lymphoma have a poor prognosis. The development of chimeric antigen receptor (CAR) T-cell platforms to treat T-cell malignancies often requires additional gene modifications to overcome fratricide because of shared T-cell antigens on normal and malignant T cells. We developed a CD5-directed CAR that produces minimal fratricide by downmodulating CD5 protein levels in transduced T cells while retaining strong cytotoxicity against CD5+ malignant cells. In our first-in-human phase 1 study (NCT0308190), second-generation autologous CD5.CAR T cells were manufactured from patients with r/r T-cell malignancies. Here, we report safety and efficacy data from a cohort of patients with mature T-cell lymphoma (TCL). Among the 17 patients with TCL enrolled, CD5 CAR T cells were successfully manufactured for 13 out of 14 attempted lines (93%) and administered to 9 (69%) patients. The overall response rate (complete remission or partial response) was 44%, with complete responses observed in 2 patients. The most common grade 3 or higher adverse events were cytopenias. No grade 3 or higher cytokine release syndrome or neurologic events occurred. Two patients died during the immediate toxicity evaluation period due to rapidly progressive disease. These results demonstrated that CD5.CAR T cells are safe and can induce clinical responses in patients with r/r CD5-expressing TCLs without eliminating endogenous T cells or increasing infectious complications. More patients and longer follow-up are needed for validation. This trial was registered at www.clinicaltrials.gov as #NCT0308190.
Collapse
Affiliation(s)
- LaQuisa C. Hill
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - Rayne H. Rouce
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Mengfen J. Wu
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX
- Biostatistics Shared Resource, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX
| | - Tao Wang
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX
- Biostatistics Shared Resource, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX
| | - Royce Ma
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX
| | - Huimin Zhang
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - Birju Mehta
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - Natalia Lapteva
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - Zhuyong Mei
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - Tyler S. Smith
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX
| | - Lina Yang
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX
| | - Madhuwanti Srinivasan
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX
| | - Phillip M. Burkhardt
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX
| | - Carlos A. Ramos
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - Premal Lulla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| | - Martha Arredondo
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX
| | - Bambi Grilley
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX
| | - Helen E. Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Malcolm K. Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Maksim Mamonkin
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| |
Collapse
|