1
|
Vetsika EK, Katsianou MA, Sarantis P, Palamaris K, Papavassiliou AG, Piperi C. Pediatric gliomas immunity challenges and immunotherapy advances. Cancer Lett 2025; 618:217640. [PMID: 40090572 DOI: 10.1016/j.canlet.2025.217640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/18/2025]
Abstract
Pediatric gliomas, the most frequent brain tumors in children, are characterized by heterogeneity and a unique tumor immune microenvironment. They are categorized into different subtypes, including low-grade gliomas like pilocytic astrocytomas and high-grade gliomas such as diffuse midline gliomas and diffuse intrinsic pontine gliomas, each exhibiting distinct immunological profiles. The tumor immune microenvironment in pediatric gliomas is shaped by cellular and non-cellular components, including immune cells, cytokines, and the extracellular matrix, involved in tumor progression, immune evasion, and response to therapy. While pediatric low-grade gliomas often display an immunosuppressed microenvironment, high-grade gliomas are characterized by complex immune infiltrates and intricate immunosuppressive mechanisms. The blood-brain barrier further obscures immune cell recruitment and therapeutic delivery. Despite advances in understanding adult gliomas, the immunobiology of pediatric tumors is poorly investigated, with limited data on the interactions between glioma cells and immune populations such as T and natural killer cells, as well as tumor-associated macrophages. Herein, we provide an update of the current knowledge on tumor immune microenvironment interactions in pediatric gliomas, highlighting the immunosuppressive mechanisms and emerging immunotherapeutic strategies aiming at overcoming these barriers to improve clinical outcomes for affected children.
Collapse
Affiliation(s)
- Eleni-Kyriaki Vetsika
- Centre of New Biotechnologies and Precision Medicine (CNBPM), School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria A Katsianou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Sarantis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Kostas Palamaris
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 10679, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
2
|
Frigault MJ, Yao N, Berger TR, Wehrli M, Gallagher KME, Horick N, Graham CE, Jacobson CA, Chen YB, Leick MB, DeFilipp Z, El-Jawahri AR, Johnson PC, Dolaher M, Katsis K, Kim AI, Crombie J, Merryman RW, Cook D, Trailor M, Cho H, Jeffrey R, Shen R, Filosto S, Nater J, Getz G, Haradhvala NJ, Maus MV. Single-cell dynamics of breakthrough toxicities after anakinra prophylaxis for axicabtagene ciloleucel in lymphoma. Blood Adv 2025; 9:2122-2135. [PMID: 39928957 DOI: 10.1182/bloodadvances.2024015161] [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: 10/23/2024] [Revised: 01/09/2025] [Accepted: 01/22/2025] [Indexed: 02/12/2025] Open
Abstract
ABSTRACT Chimeric antigen receptor (CAR) T-cell (CAR-T) therapy is limited by cytokine release syndrome (CRS) and neurotoxicity (NT). We sought to use once-daily prophylactic anakinra, an interleukin-1 (IL-1) receptor antagonist, to prevent CRS/NT that would require hospitalization (grade ≥2) in patients receiving axicabtagene ciloleucel for large-cell lymphoma, with the goal of facilitating outpatient therapy and management. Our study, in line with others, demonstrates that once-daily prophylactic anakinra is insufficient to prevent the development of toxicities that would require hospitalization in most patients. As part of the initial study design, we prospectively incorporated single-cell RNA sequencing to gain insight into the molecular immune signaling associated with breakthrough CRS and NT despite anakinra prophylaxis. In patients who developed breakthrough CRS or NT, we found that interferon gamma (IFN-γ) pathways and ligand-receptor activities were significantly enriched, as were cytokine levels of IFN-γ and CXCL10 in CD14+ monocytes. This correlated with increased IFN-γ and other cytokines in the peripheral blood. In infused CAR-T products, IL-4 and IL-10 anti-inflammatory pathways were negatively associated with grade ≥2 toxicities, regardless of anakinra treatment. These data identify IFN-γ as a potential key mechanism in CAR-T-associated toxicities, which is not inhibited by anakinra but may be otherwise targetable. This trial was registered at www.ClinicalTrials.gov as #NCT04150913.
Collapse
Affiliation(s)
- Matthew J Frigault
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| | - Ning Yao
- Harvard Medical School, Boston, MA
| | - Trisha R Berger
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Marc Wehrli
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Kathleen M E Gallagher
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Nora Horick
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Charlotte E Graham
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Caron A Jacobson
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
- Dana-Farber Cancer Institute, Department of Hematology/Oncology, Boston, MA
| | - Yi-Bin Chen
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| | - Mark B Leick
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| | - Zachariah DeFilipp
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| | - Areej R El-Jawahri
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| | - P Connor Johnson
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| | - Maria Dolaher
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Katelin Katsis
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Austin I Kim
- Harvard Medical School, Boston, MA
- Dana-Farber Cancer Institute, Department of Hematology/Oncology, Boston, MA
| | - Jennifer Crombie
- Harvard Medical School, Boston, MA
- Dana-Farber Cancer Institute, Department of Hematology/Oncology, Boston, MA
| | - Reid W Merryman
- Harvard Medical School, Boston, MA
- Dana-Farber Cancer Institute, Department of Hematology/Oncology, Boston, MA
| | - Daniella Cook
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Michael Trailor
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Hana Cho
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Richard Jeffrey
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Rhine Shen
- Kite, a Gilead Company, Santa Monica, CA
| | | | | | - Gad Getz
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| | | | - Marcela V Maus
- Cellular Immunotherapy Program, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA
- Cancer Program, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
3
|
Freund BE, Feyissa AM, Betiku OE, Shar A, Drees C, Sherman W, Qin H, Britton JW, Barrios MS, Quinones-Hinojosa A, Tatum WO. Acute Symptomatic Seizures During CAR T-Cell Therapy for Hematologic Malignancies: Tri-Site Mayo Clinic Experience. Neurology 2025; 104:e213535. [PMID: 40215424 DOI: 10.1212/wnl.0000000000213535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 02/20/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Chimeric antigen receptor T-cell (CAR T-cell) therapy is associated with neurotoxicity, which may include acute symptomatic seizures (ASySs). Specific risk factors and short-term and long-term outcomes of ASyS associated with CAR T-cell therapy have not been well investigated. METHODS This retrospective cohort study evaluated incidence and risk factors for ASyS during CAR T-cell therapy. We included patients treated at Mayo Clinic in Minnesota, Florida, and Arizona who underwent CAR T-cell therapy for hematologic malignancies from October 2019 to November 2023. Pretreatment demographics, clinical information, type of CAR T-cell therapy, neuroimaging, laboratories during treatment, and clinical features during admission were analyzed. Data on treatment and prevalence of seizures, EEG, and survival at the last follow-up were assessed. T-tests and nonparametric testing were performed on categorical and continuous data, respectively. Multivariable analysis was also performed. RESULTS We included 180 patients (mean age 62.3 years, 57.2% women) with 8 (4.4%) developing ASyS at a mean of 8.0 ± 5.3 days after therapy. Earlier onset of cytotoxic release syndrome (odds ratio [OR] 1.81, 95% CI 0.62-2.99, p = 0.007), higher grade immune effector cell-associated neurotoxicity syndrome (ICANS) (OR -1.43, 95% CI -1.86 to -1.00, p < 0.001), focal neurologic deficits (OR 7.15, 95% CI 1.60-32.14, p = 0.007), and cefepime (OR 0.58, 95% CI 0.51-0.65, p = 0.022) exposure were significantly associated with a higher risk of ASyS. A multivariable model accounting for age and sex fit best using the lowest minimum immune effector cell encephalopathy score and highest ICANS grade (R2 = 0.555, χ2 = 28.507, p < 0.001). ASyS was associated with death at the last follow-up (OR 0.48, 95% CI 0.41-0.56, p = 0.007), although short-term outcomes were not affected by ASyS. Nonprotocolized antiseizure medication (ASM) prophylaxis did not affect ASyS incidence. DISCUSSION This study suggests a low risk of ASyS because of CAR T-cell therapy, with certain risk factors that may be predictive of ASyS and lack of a definitive and direct association of ASyS with outcomes. The current approach to ASM prophylaxis should be reconsidered when ICANS is encountered. This study is limited by its retrospective nature and the use of ASM prophylaxis in all patients with ICANS, which requires further study to assess its necessity.
Collapse
Affiliation(s)
- Brin E Freund
- Department of Neurology, Mayo Clinic, Jacksonville, FL
| | | | | | - Andy Shar
- Virginia Commonwealth University, Richmond
| | | | - Wendy Sherman
- Department of Neurology, Mayo Clinic, Jacksonville, FL
| | - Hong Qin
- Department of Hematology and Oncology, Mayo Clinic, Jacksonville, FL
| | | | | | | | | |
Collapse
|
4
|
Grunblatt E, Meng Z, Baldridge AS, Patel NP, Stanisic A, Feinstein MJ, Rao A, Gordon LI, Winter JN, Ma S, Mehta J, Singhal S, Karmali R, Akhter N. Variance in development of early and late cardiotoxicities in patients with lymphoma and myeloma receiving CAR T-cell therapies. Leuk Lymphoma 2025; 66:858-868. [PMID: 39772871 DOI: 10.1080/10428194.2024.2448713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/17/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025]
Abstract
Cardiovascular adverse events (CVAEs) are recognized complications of chimeric antigen receptor (CAR) T-cell therapies. However, data are lacking regarding subtypes of adverse events that develop in patients with different malignancies, and little is known about the timeframe in which different cardiotoxicities are most likely to occur post-CAR T-cell therapies. In this study, 211 patients, including 138 lymphoma patients and 66 myeloma patients who received CAR T-cell therapies were retrospectively identified. Of these, 42 patients (19.9%) developed CVAEs post-treatment. Myeloma patients predominantly experienced heart failure while lymphoma patients predominantly experienced arrhythmia. Severe CVAEs were observed even at >12 months post-treatment. Lower baseline global longitudinal strain was significantly associated with development of post-CAR T-cell therapy CVAEs in both lymphoma and myeloma patients. These findings highlight the spectra of post-CAR T-cell cardiotoxicities in lymphoma and myeloma patients and the importance of echocardiography for pretreatment risk stratification and long-term surveillance.
Collapse
Affiliation(s)
- Eli Grunblatt
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Zhiying Meng
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Abigail S Baldridge
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Nikita P Patel
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Alexander Stanisic
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Matthew J Feinstein
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Anjali Rao
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Leo I Gordon
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Jane N Winter
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Shuo Ma
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Jayesh Mehta
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Seema Singhal
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Reem Karmali
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Nausheen Akhter
- Division of Cardiovascular Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
5
|
Chen M, Zhou Y, Bao K, Chen S, Song G, Wang S. Multispecific Antibodies Targeting PD-1/PD-L1 in Cancer. BioDrugs 2025; 39:427-444. [PMID: 40106158 DOI: 10.1007/s40259-025-00712-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2025] [Indexed: 03/22/2025]
Abstract
The development of immune checkpoint inhibitors has revolutionized the treatment of patients with cancer. Targeting the programmed cell death protein 1 (PD-1)/programmed cell death 1 ligand 1(PD-L1) interaction using monoclonal antibodies has emerged as a prominent focus in tumor therapy with rapid advancements. However, the efficacy of anti-PD-1/PD-L1 treatment is hindered by primary or acquired resistance, limiting the effectiveness of single-drug approaches. Moreover, combining PD-1/PD-L1 with other immune drugs, targeted therapies, or chemotherapy significantly enhances response rates while exacerbating adverse reactions. Multispecific antibodies, capable of binding to different epitopes, offer improved antitumor efficacy while reducing drug-related side effects, serving as a promising therapeutic approach in cancer treatment. Several bispecific antibodies (bsAbs) targeting PD-1/PD-L1 have received regulatory approval, and many more are currently in clinical development. Additionally, tri-specific antibodies (TsAbs) and tetra-specific antibodies (TetraMabs) are under development. This review comprehensively explores the fundamental structure, preclinical principles, clinical trial progress, and challenges associated with bsAbs targeting PD-1/PD-L1.
Collapse
Affiliation(s)
- Miaomiao Chen
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China
| | - Yuli Zhou
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Kaicheng Bao
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China
| | - Siyu Chen
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China
| | - Guoqing Song
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China.
| | - Siliang Wang
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China.
| |
Collapse
|
6
|
Bhutani B, Sharma V, Ganguly NK, Rana R. Unravelling the modified T cell receptor through Gen-Next CAR T cell therapy in Glioblastoma: Current status and future challenges. Biomed Pharmacother 2025; 186:117987. [PMID: 40117901 DOI: 10.1016/j.biopha.2025.117987] [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: 12/23/2024] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025] Open
Abstract
PURPOSE Despite current technological advancements in the treatment of glioma, immediate alleviation of symptoms can be catered by therapeutic modalities, including surgery, chemotherapy, and combinatorial radiotherapy that exploit aberrations of glioma. Additionally, a small number of target antigens, their heterogeneity, and immune evasion are the potential reasons for developing targeted therapies. This oncologic milestone has catalyzed interest in developing immunotherapies against Glioblastoma to improve overall survival and cure patients with high-grade glioma. The next-gen CAR-T Cell therapy is one of the effective immunotherapeutic strategies in which autologous T cells have been modified to express receptors against GBM and it modulates cytotoxicity. METHODS In this review article, we examine preclinical and clinical outcomes, and limitations as well as present cutting-edge techniques to improve the function of CAR-T cell therapy and explore the possibility of combination therapy. FINDINGS To date, several CAR T-cell therapies are being evaluated in clinical trials for GBM and other brain malignancies and multiple preclinical studies have demonstrated encouraging outcomes. IMPLICATIONS CAR-T cell therapy represents a promising therapeutic paradigm in the treatment of solid tumors but a few limitations include, the blood-brain barrier (BBB), antigen escape, tumor microenvironment (TME), tumor heterogeneity, and its plasticity that suppresses immune responses weakens the ability of this therapy. Additional investigation is required that can accurately identify the targets and reflect the similar architecture of glioblastoma, thus optimizing the efficiency of CAR-T cell therapy; allowing for the selection of patients most likely to benefit from immuno-based treatments.
Collapse
Affiliation(s)
- Bhavya Bhutani
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Vyoma Sharma
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Nirmal Kumar Ganguly
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Rashmi Rana
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India.
| |
Collapse
|
7
|
Pagliuca S, Malard F, Mooyaart JE, Daskalakis M, Gabellier L, Yakoub-Agha I, Ram R, Besley C, Forcade E, Vucinic V, Corral LL, Vydra J, von Tresckow B, Amat P, Amrolia P, Vandenberghe P, Stölzel F, Sica S, Rubio MT, Hoogenboom JD, Ortiz-Maldonado V, Nagler A, Kuball J, Chabannon C, Ruggeri A. The landscape of immune monitoring in CAR-T cell therapy: A comprehensive review and survey study by the Cellular Therapy and Immunobiology Working Party of the EBMT. Blood Rev 2025; 71:101272. [PMID: 39986989 DOI: 10.1016/j.blre.2025.101272] [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: 11/10/2024] [Revised: 01/20/2025] [Accepted: 02/13/2025] [Indexed: 02/24/2025]
Abstract
Immune monitoring of cell therapies is a complex and evolving topic, particularly in the rapid expanding field of chimeric antigen receptor T (CAR-T) cell applications. Defining essential, recommended, and optional immune monitoring data post-CAR-T cell infusion is crucial to improve patient outcomes and inform post-treatment decisions. To address this gap, we conducted a survey-based study across centers affiliated with the European Society for Blood and Marrow Transplantation (EBMT), focusing on patients treated with European Medicines Agency (EMA)-approved CAR-T products. Building on a thorough review of the literature, we mapped the current landscape of immune monitoring practices and assessed their impact on clinical management. By defining the state of the art in the field, this work marks an initial step towards a structured harmonization process potentially able to enhance the management and outcomes of patients undergoing these immune cell therapies.
Collapse
Affiliation(s)
- Simona Pagliuca
- Hematology department, Nancy University Hospital, UMR 7365, CNRS, University of Lorraine, Vandoeuvre-lès-Nancy, France.
| | - Florent Malard
- Sorbonne Université, Centre de Recherche Saint-Antoine INSERM UMRs938, Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, AP-HP, Paris, France
| | | | | | | | | | - Ron Ram
- Tel Aviv Sourasky Medical Center and Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Caroline Besley
- University Hospitals Bristol and Weston NHSFT, Bristol, United Kingdom
| | | | | | - Lucía López Corral
- Hospital Clínico, CIBERONC. Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Jan Vydra
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Bastian von Tresckow
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center and German Cancer consortium (DKTK partner site Essen), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Paula Amat
- Hospital Clínico Universitario-INCLIVA de, Valencia, Spain
| | - Persis Amrolia
- Great Ormond Street Children's Hospital Paediatrics, London, United Kingdom
| | | | - Friedrich Stölzel
- University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | | | - Marie Thérèse Rubio
- Hematology department, Nancy University Hospital, UMR 7365, CNRS, University of Lorraine, Vandoeuvre-lès-Nancy, France
| | | | | | - Arnon Nagler
- Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Jürgen Kuball
- University Medical Center Utrecht, Utrecht, the Netherlands
| | - Christian Chabannon
- Institut Paoli-Calmettes Comprehensive Cancer Centre and Module Biothérapies du Centre d'Investigations Cliniques de Marseille, INSERM-Aix-Marseille Université-AP-HM-IPC, CBT-1409 Marseille, France
| | - Annalisa Ruggeri
- San Raffaele Scientific Institute, Hematology and Bone Marrow Transplantation Unit, Milan, Italy
| |
Collapse
|
8
|
Pondrelli F, Muccioli L, Mason F, Zenesini C, Ferri L, Asioli GM, Rossi S, Rinaldi R, Rondelli F, Nicodemo M, D'Angelo R, Barone V, Sambati L, Pensato U, Zinzani PL, Casadei B, Bonifazi F, Maffini E, Pierucci E, Cortelli P, Tinuper P, Bisulli F, Guarino M. EEG as a predictive biomarker of neurotoxicity in anti-CD19 CAR T-cell therapy. J Neurol 2025; 272:360. [PMID: 40278905 PMCID: PMC12031834 DOI: 10.1007/s00415-025-13102-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/29/2025] [Accepted: 04/13/2025] [Indexed: 04/26/2025]
Abstract
OBJECTIVE Immune effector cell-associated neurotoxicity syndrome (ICANS) is a potentially fatal complication of CD19-directed CAR T-cell therapy. The aim of this study was to investigate the role of EEG as a predictive biomarker of ICANS. METHODS In this prospective, monocentric, cohort study, consecutive refractory B-cell non-Hodgkin lymphoma patients undergoing CAR T-cell therapy had EEG assessments at fixed time points pre- and post-infusion. The risk of ICANS was evaluated according to EEG findings detected qualitatively, using a grading scale ranging from 0 (normal) to 3 (severely abnormal), and quantitatively, using power spectral and connectivity measures. RESULTS 307 EEGs from 68 patients have been qualitatively evaluated, of whom 238 were eligible for quantitative analysis. Neurotoxicity manifested in 22/68 (32.4%) patients. Pre-infusion EEG abnormalities (grade 1 and 2) were qualitatively detected in 8/68 (11.7%) patients, emerging as a risk factor for ICANS [HR 5.8 (95%CI 2.6-12.9)]. Quantitative analysis of pre-infusion EEGs did not yield significative results. Post-infusion qualitative EEG abnormalities were associated to a higher risk of ICANS development [HR 11.6 (4.4-30.5) for grade 2; HR 9.7 (2.6-36.6) for grade 3]. Concerning the quantitative analysis, in post-infusion EEGs higher theta energy [HR 1.10 (1.03-1.16)] and delta + theta/alfa ratio [HR 1.37 (1.11-1.67)] were associated to higher risk of ICANS, while higher beta energy resulted protective [HR 0.91 (0.85-0.97)]. CONCLUSIONS Our study establishes EEG as a predictive tool for identifying patients at risk for ICANS before CAR T-cell infusion, who may benefit from prophylactic treatments, and anticipating ICANS onset following infusion, enabling early intervention.
Collapse
Affiliation(s)
| | - Lorenzo Muccioli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| | - Federico Mason
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Corrado Zenesini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Lorenzo Ferri
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Gian Maria Asioli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Simone Rossi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Rita Rinaldi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | | | - Marianna Nicodemo
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Roberto D'Angelo
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Valentina Barone
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Luisa Sambati
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Umberto Pensato
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Department of Neurology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Beatrice Casadei
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | - Enrico Maffini
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | - Pietro Cortelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Paolo Tinuper
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Francesca Bisulli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Maria Guarino
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| |
Collapse
|
9
|
Esmaeilzadeh A, Hadiloo K, Yaghoubi S, Makoui MH, Mostanadi P. State of the art in CAR-based therapy: In vivo CAR production as a revolution in cell-based cancer treatment. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01056-7. [PMID: 40261561 DOI: 10.1007/s13402-025-01056-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/19/2025] [Indexed: 04/24/2025] Open
Abstract
Chimeric antigen receptor (CAR) therapy has successfully treated relapsed/refractory hematological cancers. This strategy can effectively target tumor cells. However, despite positive outcomes in clinical applications, challenges remain to overcome. These hurdles pertain to the production of the drugs, solid tumor resistance, and side effects related to the treatment. Some cases have been missed during the drug preparation due to manufacturing issues, prolonged production times, and high costs. These challenges mainly arise from the in vitro manufacturing process, so reevaluating this process could minimize the number of missed patients. The immune cells are traditionally collected and sent to the laboratory; after several steps, the cells are modified to express the CAR gene before being injected back into the patient's body. During the in vivo method, the CAR gene is introduced to the immune cells inside the body. This allows for treatment to begin sooner, avoiding potential failures in drug preparation and the associated high costs. In this review, we will elaborate on the production and treatment process using in vivo CAR, examine the benefits and challenges of this approach, and ultimately present the available solutions for incorporating this treatment into clinical practice.
Collapse
Affiliation(s)
- Abdolreza Esmaeilzadeh
- Pficell R&D Canadian Institution & Corporation, Profound Future Focused Innovative Cell and Gene Therapy, Pficell Canadian Institution and Corporation, Ontario, Canada.
- Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Kaveh Hadiloo
- Pficell R&D Canadian Institution & Corporation, Profound Future Focused Innovative Cell and Gene Therapy, Pficell Canadian Institution and Corporation, Ontario, Canada
- Department of Surgery, Velayat Clinical Research Development Unit, Qazvin University of Medical Sciences, Qazvin, Iran
- Department of Immunology, Student Research Committee, School of Medicine, Zanjan, Iran
| | - Sara Yaghoubi
- Department of Immunology, Student Research Committee, School of Medicine, Zanjan, Iran
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Parsa Mostanadi
- Department of Immunology, Student Research Committee, School of Medicine, Zanjan, Iran
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
10
|
Nie J, Zhou L, Tian W, Liu X, Yang L, Yang X, Zhang Y, Wei S, Wang DW, Wei J. Deep insight into cytokine storm: from pathogenesis to treatment. Signal Transduct Target Ther 2025; 10:112. [PMID: 40234407 PMCID: PMC12000524 DOI: 10.1038/s41392-025-02178-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: 08/09/2024] [Revised: 12/22/2024] [Accepted: 02/12/2025] [Indexed: 04/17/2025] Open
Abstract
Cytokine storm (CS) is a severe systemic inflammatory syndrome characterized by the excessive activation of immune cells and a significant increase in circulating levels of cytokines. This pathological process is implicated in the development of life-threatening conditions such as fulminant myocarditis (FM), acute respiratory distress syndrome (ARDS), primary or secondary hemophagocytic lymphohistiocytosis (HLH), cytokine release syndrome (CRS) associated with chimeric antigen receptor-modified T (CAR-T) therapy, and grade III to IV acute graft-versus-host disease following allogeneic hematopoietic stem cell transplantation. The significant involvement of the JAK-STAT pathway, Toll-like receptors, neutrophil extracellular traps, NLRP3 inflammasome, and other signaling pathways has been recognized in the pathogenesis of CS. Therapies targeting these pathways have been developed or are currently being investigated. While novel drugs have demonstrated promising therapeutic efficacy in mitigating CS, the overall mortality rate of CS resulting from underlying diseases remains high. In the clinical setting, the management of CS typically necessitates a multidisciplinary team strategy encompassing the removal of abnormal inflammatory or immune system activation, the preservation of vital organ function, the treatment of the underlying disease, and the provision of life supportive therapy. This review provides a comprehensive overview of the key signaling pathways and associated cytokines implicated in CS, elucidates the impact of dysregulated immune cell activation, and delineates the resultant organ injury associated with CS. In addition, we offer insights and current literature on the management of CS in cases of FM, ARDS, systemic inflammatory response syndrome, treatment-induced CRS, HLH, and other related conditions.
Collapse
Grants
- 82070217, 81873427 National Natural Science Foundation of China (National Science Foundation of China)
- 82100401 National Natural Science Foundation of China (National Science Foundation of China)
- 81772477, 81201848, 82473220 National Natural Science Foundation of China (National Science Foundation of China)
- 82330010,81630010,81790624 National Natural Science Foundation of China (National Science Foundation of China)
- National High Technology Research and Development Program of China, Grant number: 2021YFA1101500.
- The Hubei Provincial Natural Science Foundation (No.2024AFB050)
- Project of Shanxi Bethune Hospital, Grant Numbber: 2023xg02); Fundamental Research Program of Shanxi Province, Grant Numbber: 202303021211224
- The Key Scientific Research Project of COVID-19 Infection Emergency Treatment of Shanxi Bethune Hospital (2023xg01), 2023 COVID-19 Research Project of Shanxi Provincial Health Commission (No.2023XG001, No. 2023XG005), Four “Batches” Innovation Project of Invigorating Medical through Science and Technology of Shanxi Province (2023XM003), Cancer special Fund research project of Shanxi Bethune Hospital (No. 2020-ZL04), and External Expert Workshop Fund Program of Shanxi Provincial Health Commission(Proteomics Shanxi studio for Huanghe professor)
- Fundamental Research Program of Shanxi Province(No.202303021221192); 2023 COVID-19 Emergency Project of Shanxi Health Commission (Nos.2023XG001,2023XG005)
Collapse
Affiliation(s)
- Jiali Nie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Ling Zhou
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China
| | - Weiwei Tian
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Xiansheng Liu
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Liping Yang
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Xingcheng Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Branch of National Clinical Research Center for Infectious Diseases, Wuhan Pulmonary Hospital (Wuhan Tuberculosis Prevention and Control Institute), Wuhan, China.
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China.
| | - Jia Wei
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
11
|
Nguyen KA, Liu Z, Davies JS, McIntosh CP, Draper LM, Norberg SM, Rae Z, Achar SR, Altan-Bonnet G, Zhang L, Wu X, Meyer TJ, Kelly MC, Taylor N, Hinrichs CS, Ishii K. CD22 TCR-engineered T cells exert antileukemia cytotoxicity without causing inflammatory responses. SCIENCE ADVANCES 2025; 11:eadq4297. [PMID: 40203088 PMCID: PMC11980841 DOI: 10.1126/sciadv.adq4297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 03/04/2025] [Indexed: 04/11/2025]
Abstract
Chimeric antigen receptor (CAR) T cells effectively treat B cell malignancies. However, CAR-T cells cause inflammatory toxicities such as cytokine release syndrome (CRS), which is in contrast to T cell receptor (TCR)-engineered T cells against various antigens that historically have rarely been associated with CRS. To study whether and how differences in receptor types affect the propensity for eliciting inflammatory responses in a model system wherein TCR and CAR target equalized sources of clinically relevant antigen, we discovered a CD22-specific TCR and compared it to CD22 CAR. Both CD22 TCR-T and CD22 CAR-T cells eradicated leukemia in xenografts, but only CD22 CAR-T cells induced dose-dependent systemic inflammation. Compared to TCR-T cells, CAR-T cells disproportionately upregulated inflammatory pathways without concordant augmentation in pathways involved in direct cytotoxicity upon antigen engagement. These differences in antileukemia responses comparing TCR-T and CAR-T cells highlight the potential opportunity to improve therapeutic safety by using TCRs.
Collapse
MESH Headings
- Humans
- Animals
- Sialic Acid Binding Ig-like Lectin 2/immunology
- Sialic Acid Binding Ig-like Lectin 2/genetics
- Sialic Acid Binding Ig-like Lectin 2/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Mice
- Inflammation/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Leukemia/therapy
- Leukemia/immunology
- Leukemia/pathology
- Cytotoxicity, Immunologic
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Kilyna A. Nguyen
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Zhihui Liu
- Pediatric Oncology Branch, CCR, NCI, NIH, Bethesda, MD, USA
| | - John S. Davies
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
- Department of Safety Assessment, Genentech Inc., South San Francisco, CA, USA
| | - Crystal P. McIntosh
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Lindsey M. Draper
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Scott M. Norberg
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Zachary Rae
- Single Cell Analysis Facility, CCR, NCI, NIH, Bethesda, MD, USA
| | - Sooraj R. Achar
- Laboratory of Integrative Cancer Immunology, CCR, NCI, NIH, Bethesda, MD, USA
| | | | - Ling Zhang
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Xiaolin Wu
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, NCI, NIH, Frederick, MD, USA
| | - Thomas J. Meyer
- CCR Collaborative Bioinformatics Resource, CCR, NCI, NIH, Bethesda, MD, USA
| | | | - Naomi Taylor
- Pediatric Oncology Branch, CCR, NCI, NIH, Bethesda, MD, USA
| | - Christian S. Hinrichs
- Duncan and Nancy MacMillan Center of Excellence in Cancer Immunotherapy and Metabolism, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Kazusa Ishii
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
12
|
Gao P, Zhang Y, Ma J, Zhang Y. Immunotherapy in chronic lymphocytic leukemia: advances and challenges. Exp Hematol Oncol 2025; 14:53. [PMID: 40211406 PMCID: PMC11984025 DOI: 10.1186/s40164-025-00644-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Chronic lymphocytic leukemia (CLL) is characterized as a clonal proliferation of mature B lymphocytes with distinct immunophenotypic traits, predominantly affecting the middle-aged and elderly population. This condition is marked by an accumulation of lymphocytes within the peripheral blood, bone marrow, spleen, and lymph nodes. The associated immune dysregulation predisposes CLL patients to a higher risk of secondary malignancies and infections, which significantly contribute to morbidity and mortality rates. The advent of immunotherapy has revolutionized the prognosis of CLL, advancing treatment modalities and offering substantial benefits to patient outcomes. This review endeavors to synthesize and scrutinize the efficacy, merits, and limitations of the current immunotherapeutic strategies for CLL. The aim is to inform the selection of optimal treatment regimens tailored to individual patient needs. Furthermore, the review juxtaposes various therapeutic combinations to elucidate the comparative advantages of each approach, with the ultimate objective of enhancing patient prognosis and quality of life.
Collapse
Affiliation(s)
- Pan Gao
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Yang Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Jun Ma
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Ya Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
| |
Collapse
|
13
|
Wermke M, Araurjo DM, Chatterjee M, Tsimberidou AM, Holderried TAW, Jazaeri AA, Reshef R, Bokemeyer C, Alsdorf W, Wetzko K, Brossart P, Aslan K, Backert L, Bunk S, Fritsche J, Gulde S, Hengler S, Hilf N, Hossain MB, Hukelmann J, Kalra M, Krishna D, Kursunel MA, Maurer D, Mayer-Mokler A, Mendrzyk R, Mohamed A, Pozo K, Satelli A, Letizia M, Schuster H, Schoor O, Wagner C, Rammensee HG, Reinhardt C, Singh-Jasuja H, Walter S, Weinschenk T, Luke JJ, Britten CM. Autologous T cell therapy for PRAME + advanced solid tumors in HLA-A*02 + patients: a phase 1 trial. Nat Med 2025:10.1038/s41591-025-03650-6. [PMID: 40205198 DOI: 10.1038/s41591-025-03650-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 03/10/2025] [Indexed: 04/11/2025]
Abstract
In contrast to chimeric antigen receptor T cells, T cell receptor (TCR)-engineered T cells can target intracellular tumor-associated antigens crucial for treating solid tumors. However, most trials published so far show limited clinical activity. Here we report interim data from a first-in-human, multicenter, open-label, 3 + 3 dose-escalation/de-escalation phase 1 trial studying IMA203, an autologous preferentially expressed antigen in melanoma (PRAME)-directed TCR T cell therapy in HLA-A*02+ patients with PRAME+ recurrent and/or refractory solid tumors, including melanoma and sarcoma. Primary objectives include the evaluation of safety and tolerability and the determination of the maximum tolerated dose (MTD) and/or recommended dose for extension. Secondary objectives include the evaluation of IMA203 TCR-engineered T cell persistence in peripheral blood, tumor response as well as duration of response. A total of 27 patients were enrolled in the phase 1a dose escalation and 13 patients in the phase 1b dose extension. IMA203 T cells were safe, and the MTD was not reached. Of the 41 patients receiving treatment (that is, who started lymphodepletion), severe cytokine release syndrome was observed in 4.9% (2/41), and severe neurotoxicity did not occur. In the 40 patients treated with IMA203, an overall response rate consisting of patients with unconfirmed or confirmed response (u/cORR) of 52.5% (21/40) and a cORR of 28.9% (11/38) was observed with a median duration of response of 4.4 months (range, 2.4-23.0, 95% confidence interval: 2.6-not reached) across multiple indications. Rapid T cell engraftment and long-term persistence of IMA203 T cells were observed. IMA203 T cells trafficked to all organs, and confirmed responses were more frequent in patients with higher dose. T cell exhaustion was not observed in the periphery; deep responses were enriched at higher PRAME expression; and higher T cell infiltration resulted in longer progression-free survival. Overall, IMA203 showed promising anti-tumor activity in multiple solid tumors, including refractory melanoma. ClinicalTrials.gov identifier: NCT03686124 .
Collapse
Affiliation(s)
- Martin Wermke
- Department of Medicine I, University Hospital Carl Gustav Carus TU Dresden, Dresden, Germany
- National Center for Tumor Diseases, Dresden, Germany
| | - Dejka M Araurjo
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Manik Chatterjee
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany
| | - Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tobias A W Holderried
- Department of Hematology, Oncology, Immunooncology, Stem Cell Transplantation, and Rheumatology, University Hospital Bonn, Bonn, Germany
| | - Amir A Jazaeri
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ran Reshef
- Columbia University Medical Center, New York, NY, USA
| | - Carsten Bokemeyer
- Department of Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Winfried Alsdorf
- Department of Oncology, Hematology, and Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katrin Wetzko
- Department of Medicine I, University Hospital Carl Gustav Carus TU Dresden, Dresden, Germany
| | - Peter Brossart
- Department of Hematology, Oncology, Immunooncology, Stem Cell Transplantation, and Rheumatology, University Hospital Bonn, Bonn, Germany
| | - Katrin Aslan
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | | | | | | | - Swapna Gulde
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | | | - Norbert Hilf
- Immatics Biotechnologies GmbH, Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jason J Luke
- Cancer Immunotherapeutics Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | | |
Collapse
|
14
|
Vonberg FW, Malik I, O'Reilly M, Hyare H, Carr AS, Roddie C. Neurotoxic complications of chimeric antigen receptor (CAR) T-cell therapy. J Neurol Neurosurg Psychiatry 2025:jnnp-2024-333924. [PMID: 40185628 DOI: 10.1136/jnnp-2024-333924] [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: 12/23/2024] [Accepted: 03/25/2025] [Indexed: 04/07/2025]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has revolutionised the treatment of haematological malignancies and has demonstrated efficacy in early trials for solid tumours, neurological and rheumatological autoimmune diseases. However, CAR-T is complicated in some patients by neurotoxicity syndromes including immune-effector cell-associated neurotoxicity syndrome, and the more recently described movement and neurocognitive treatment-emergent adverse events, and tumour inflammation-associated neurotoxicity. These neurotoxic syndromes remain poorly understood and are associated with significant morbidity and mortality. A multidisciplinary approach, including neurologists, haematologists and oncologists, is critical for the diagnosis and management of CAR-T neurotoxicity. This approach will be of increasing importance as the use of CAR-T expands, its applications increase and as novel neurotoxic syndromes emerge.
Collapse
Affiliation(s)
- Frederick W Vonberg
- National Hospital for Neurology and Neurosurgery, London, UK
- UCL Queen Square Institute of Neurology, London, UK
| | - Imran Malik
- University College London Hospitals NHS Foundation Trust, London, UK
| | - Maeve O'Reilly
- Haematology, University College London Hospitals NHS Foundation Trust, London, UK
- UCL Cancer Institute, London, UK
| | - Harpreet Hyare
- UCL Queen Square Institute of Neurology, London, UK
- Neuroradiology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Aisling S Carr
- UCL Queen Square Institute of Neurology, London, UK
- Neurology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Claire Roddie
- Haematology, University College London Hospitals NHS Foundation Trust, London, UK
- UCL Cancer Institute, London, UK
| |
Collapse
|
15
|
Chen B, Guo R, Niu Y, Guo W, Wang T. Haploidentical Hematopoietic Stem Cell Transplantation for Relapsed/Refractory Extramedullary Multiple Myeloma: A Conditioning Regimen Combining Selinexor and Helical Tomotherapy-A Case Report and Literature Review. J Blood Med 2025; 16:177-185. [PMID: 40196041 PMCID: PMC11972968 DOI: 10.2147/jbm.s495256] [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: 09/16/2024] [Accepted: 03/18/2025] [Indexed: 04/09/2025] Open
Abstract
Relapsed/refractory multiple myeloma (RRMM) and extramedullary multiple myeloma (EMM) present significant challenges for patients with multiple myeloma (MM) after their disease progresses.Despite notable advancements in treatments like autologous hematopoietic stem cell transplantation (ASCT) and chimeric antigen receptor (CAR)-T-cell therapy, most patients with RRMM and EMM face a short survival period. Currently, there are no effective treatments available. Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is one of the treatment options for MM. Reduced-intensity conditioning (RIC) regimens have largely replaced myeloablative conditioning (MAC) regimens. RIC is now preferred because it significantly lowers transplant-related mortality, which has dropped to 10-20%. However, RIC regimens are linked to higher relapse rates compared to MAC. To enhance the efficacy of allo-HSCT, it is essential to identify a safer and more effective conditioning regimen. We report a case of EMM involving the breast, supraclavicular region, mediastinum, and pleural effusion, among other sites. The patient did not respond to several treatments, including a proteasome inhibitor (PI) like bortezomib, immunomodulatory drugs (IMiDs) such as lenalidomide, and a monoclonal antibody targeting CD38, like daratumumab. Consequently, we recommended haploidentical hematopoietic stem cell transplantation as a salvage treatment. After undergoing allo-HSCT with a conditioning regimen that mainly included selinexor and helical tomotherapy, the patient achieved a complete remission(CR) and enjoyed long-term disease-free survival for 11 months. Along with existing literature, this case provides encouraging insights for future research on RRMM and EMM, and we anticipate more reports on allo-HSCT cases in the future.
Collapse
Affiliation(s)
- Bin Chen
- Department of Hematology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, People’s Republic of China
| | - Rongrong Guo
- Department of Hematology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, People’s Republic of China
| | - Yanyan Niu
- Department of Hematology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, People’s Republic of China
| | - Wei Guo
- Department of Hematology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, People’s Republic of China
| | - Tao Wang
- Department of Hematology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, People’s Republic of China
| |
Collapse
|
16
|
Daoudlarian D, Segot A, Latifyan S, Bartolini R, Joo V, Mederos N, Bouchaab H, Demicheli R, Abdelhamid K, Ferahta N, Doms J, Stalder G, Noto A, Mencarelli L, Mosimann V, Berthold D, Stravodimou A, Sartori C, Shabafrouz K, Thompson JA, Wang Y, Peters S, Pantaleo G, Obeid M. Tocilizumab and immune signatures for targeted management of cytokine release syndrome in immune checkpoint therapy. Ann Oncol 2025; 36:444-459. [PMID: 39701282 DOI: 10.1016/j.annonc.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/26/2024] [Accepted: 12/04/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND This study aimed to identify specific biomarkers in oncology patients experiencing immune-related cytokine release syndrome (irCRS)-like symptoms during immune checkpoint inhibitor (ICI) therapy, including severe cases like hemophagocytic lymphohistiocytosis (irHLH), and to distinguish these from sepsis. A secondary objective was to retrospectively analyze the efficacy of tocilizumab (TCZ) in treating corticosteroid (CS)-refractory high-grade irCRS. PATIENTS AND METHODS A cohort of 35 patients presenting with irCRS-like symptoms was studied, including 9 with irHLH-like manifestations and 8 with sepsis. Immune profiling was carried out using 48 mass cytometry markers, along with an analysis of 45 serum biomarkers, including 27 cytokines and 18 additional markers from the HScore. Twelve patients with high-grade irCRS refractory to CS were treated with TCZ. RESULTS Twenty-four biomarkers significantly distinguished between irHLH and grade 3 irCRS (P = 0.0027-0.0455). Hepatocyte growth factor (HGF) and ferritin had superior predictive values compared with the traditional HScore, both with a positive predictive value (PPV) and negative predictive value (NPV) of 100%. CXCL9 differentiated irHLH from grade 3 irCRS and predicted the need for TCZ treatment intensification (PPV = 90%, NPV = 100%). Additional biomarkers, including leukocyte count, neutrophils, ferritin, interleukin (IL)-6, IL-7, epidermal growth factor, fibrinogen, and granulocyte-macrophage colony-stimulating factor (GM-CSF), discriminated sepsis from high-grade irCRS (PPV = 75%-80%, NPV = 100%). Elevated frequencies of CXCR5+ or CCR4+ CD8 memory cells, CD38+ intermediate monocytes, and CD62L+ neutrophils were observed in high-grade irCRS compared with sepsis. All 12 patients with high-grade irCRS refractory to CS treated with TCZ experienced complete resolution. CONCLUSIONS This study highlights the importance of specific immunologic biomarkers in determining irCRS severity, predicting outcomes, and distinguishing between irHLH, irCRS, and sepsis. It also demonstrates the efficacy of TCZ in managing high-grade irCRS, underscoring the need for personalized therapeutic strategies based on these biomarkers.
Collapse
Affiliation(s)
- D Daoudlarian
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Medicine, Immunology and Allergy Service, University of Lausanne, Lausanne, Switzerland
| | - A Segot
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Service and Central Laboratory of Hematology, University of Lausanne, Lausanne, Switzerland
| | - S Latifyan
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - R Bartolini
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Medicine, Immunology and Allergy Service, University of Lausanne, Lausanne, Switzerland
| | - V Joo
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Medicine, Immunology and Allergy Service, University of Lausanne, Lausanne, Switzerland
| | - N Mederos
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - H Bouchaab
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - R Demicheli
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - K Abdelhamid
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - N Ferahta
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - J Doms
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Medicine, Immunology and Allergy Service, University of Lausanne, Lausanne, Switzerland
| | - G Stalder
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Service and Central Laboratory of Hematology, University of Lausanne, Lausanne, Switzerland; Service of Hematology, Institut Central des Hôpitaux, Hôpital du Valais, Sion, Switzerland
| | - A Noto
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Medicine, Immunology and Allergy Service, University of Lausanne, Lausanne, Switzerland
| | - L Mencarelli
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Medicine, Immunology and Allergy Service, University of Lausanne, Lausanne, Switzerland
| | - V Mosimann
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - D Berthold
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - A Stravodimou
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - C Sartori
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Medicine, Internal Medicine Service, University of Lausanne, Lausanne, Switzerland
| | - K Shabafrouz
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - J A Thompson
- Department of Medicine, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, USA
| | - Y Wang
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - S Peters
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Oncology, Medical Oncology Service, University of Lausanne, Lausanne, Switzerland
| | - G Pantaleo
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Medicine, Immunology and Allergy Service, University of Lausanne, Lausanne, Switzerland
| | - M Obeid
- Centre Hospitalier Universitaire Vaudois (CHUV), Department of Medicine, Immunology and Allergy Service, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
17
|
Santurio DS, Barros LRC, Glauche I, Fassoni AC. Mathematical modeling unveils the timeline of CAR-T cell therapy and macrophage-mediated cytokine release syndrome. PLoS Comput Biol 2025; 21:e1012908. [PMID: 40203243 PMCID: PMC11981663 DOI: 10.1371/journal.pcbi.1012908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/24/2025] [Indexed: 04/11/2025] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy holds significant potential for cancer treatment, although disease relapse and cytokine release syndrome (CRS) remain as frequent clinical challenges. To better understand the mechanisms underlying the temporal dynamics of CAR-T cell therapy response and CRS, we developed a novel multi-layer mathematical model incorporating antigen-mediated CAR-T cell expansion, antigen-negative resistance, and macrophage-associated cytokine release. Three key mechanisms of macrophage activation are considered: release of damage-associated molecular patterns, antigen-binding mediated activation, and CD40-CD40L contact. The model accurately describes 25 patient time courses with different responses and IL-6 cytokine kinetics. We successfully link the dynamic shape of the response to interpretable model parameters and investigate the influence of CAR-T cell dose and initial tumor burden on the occurrence of cytokine release and treatment outcome. By disentangling the timeline of macrophage activation, the model identified distinct contributions of each activation mechanism, suggesting the CD40-CD40L axis as a major driver of cytokine release and a clinically feasible target to control the activation process and modulate cytokine peak height. Our multi-layer model provides a comprehensive framework for understanding the complex interactions between CAR-T cells, tumor cells, and macrophages during therapy.
Collapse
Affiliation(s)
| | | | - Ingmar Glauche
- Institute for Medical Informatics and Biometry, Technische Universität Dresden, Dresden, Germany
| | - Artur c Fassoni
- Institute for Medical Informatics and Biometry, Technische Universität Dresden, Dresden, Germany
- Instituto de Matemática e Computação, Universidade Federal de Itajubá, Itajubá, Brazil
| |
Collapse
|
18
|
Long JP, Prakash R, Edelkamp P, Knafl M, Lionel AC, Nair R, Ahmed S, Strati P, Castillo LEM, Al-Zaki A, Chien K, Chihara D, Westin J, Khawaja F, Nastoupil LJ, Mulanovich V, Futreal A, Woodman SE, Daver NG, Flowers CR, Neelapu S, Manzano JG, Iyer SP. Cytokine Storms in COVID-19, Hemophagocytic Lymphohistiocytosis, and CAR-T Therapy. JAMA Netw Open 2025; 8:e253455. [PMID: 40193078 PMCID: PMC11976493 DOI: 10.1001/jamanetworkopen.2025.3455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 12/27/2024] [Indexed: 04/10/2025] Open
Abstract
Importance Cytokine storm (CS) is a hyperinflammatory syndrome causing multiorgan dysfunction and high mortality, especially in patients with malignant hematologic neoplasms. Triggers include malignant neoplasm-associated hemophagocytic lymphohistiocytosis (MN-HLH), cytokine release syndrome from chimeric antigen receptor T-cell therapy (CAR-T CRS), and COVID-19, but the underlying mechanisms of inflammation and their impact on outcomes are poorly understood. Objective To delineate the inflammatory patterns characterizing different CS etiologies and their association with clinical outcomes. Design, Setting, and Participants This retrospective cohort study was conducted at the MD Anderson Cancer Center in Houston, Texas, between March 1, 2020, and November 20, 2022, using the software-as-a-service Syntropy Foundry Platform. Participants were patients with malignant hematologic neoplasms who developed CS from COVID-19 (COVID-CS), MN-HLH, or CAR-T CRS. Exposure Diagnostic criteria for COVID-CS were developed based on surging inflammatory markers (interleukin-6, C-reactive protein, and ferritin), while diagnosis of MN-HLH and CAR-T CRS followed established guidelines. Main Outcomes and Measures The study compared cytokine levels, clinical characteristics, and survival outcomes across the 3 cohorts and focused on inflammatory markers, survival times, and key factors associated with survival identified through univariate and multivariable analyses. Results A total of 671 patients met the inclusion criteria. Of those, 220 (33%) had CAR-T CRS, 227 (34%) had COVID-CS, and 224 (33%) had MN-HLH. Patients were predominantly male (435 [65%]), and 461 (69%) were White, with significant differences in median age (CAR-T CRS, 63 [IQR, 54-71] years; COVID-CS, 63 [IQR, 52-72] years; MN-HLH, 55 [IQR, 41-65] years; P < .001) as well as number of admission days and underlying cancer type across cohorts. Marked variations in cytokine levels and survival outcomes were observed, with the MN-HLH cohort exhibiting the highest levels of inflammatory markers (eg, median TNF-α, 105 pg/mL [IQR, 38-201 pg/mL] for MN-HLH vs 23 pg/mL [IQR, 17-42 pg/mL] for COVID-CS) and lowest fibrinogen and albumin levels. The cohort with CAR-T CRS showed substantially longer survival times compared with the cohort with COVID-CS (hazard ratio [HR], 2.93; 95% CI, 1.95-4.41) and the cohort with MN-HLH (HR, 8.12; 95% CI, 5.51-12.00). Clustering analysis showed overlapping patterns between COVID-CS and CAR-T CRS, while MN-HLH formed a distinct cluster. Conclusions and Relevance This study of CS syndromes found distinct immune responses within each cohort. The distinct clinical patterns and outcomes associated with different CS etiologies emphasize the importance of early diagnosis and timely intervention.
Collapse
Affiliation(s)
- James P. Long
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston
| | - Rishab Prakash
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Paul Edelkamp
- Department of Enterprise Data Engineering & Analytics, The University of Texas MD Anderson Cancer Center, Houston
| | - Mark Knafl
- Department of Enterprise Data Engineering & Analytics, The University of Texas MD Anderson Cancer Center, Houston
| | - Anath C. Lionel
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Ranjit Nair
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Sairah Ahmed
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Paolo Strati
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Luis E. Malpica Castillo
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Ajlan Al-Zaki
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Kelly Chien
- Department of Leukemia, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Dai Chihara
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Jason Westin
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Fareed Khawaja
- Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston
| | - Loretta J. Nastoupil
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Victor Mulanovich
- Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston
| | - Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Scott E. Woodman
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Naval G. Daver
- Department of Leukemia, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Christopher R. Flowers
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Sattva Neelapu
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Joanna-Grace Manzano
- Department of Hospital Medicine, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston
| | - Swaminathan P. Iyer
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
19
|
Zoref-Lorenz A, Witzig TE, Cerhan JR, Jordan MB. Malignancy-associated HLH: mechanisms, diagnosis, and treatment of a severe hyperinflammatory syndrome. Leuk Lymphoma 2025; 66:628-636. [PMID: 39656557 DOI: 10.1080/10428194.2024.2436037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 12/17/2024]
Abstract
Hemophagocytic lymphohistiocytosis (HLH) is a severe hyperinflammatory syndrome characterized by uncontrolled immune activation. While traditionally associated with genetic mutations affecting cytotoxic function, recent advances have highlighted the prevalence and significance of HLH in adults, particularly in hematologic malignancies. This review focuses on malignancy-associated HLH (M-HLH), a complex and challenging condition with a poor prognosis. The review explores four main subtypes of M-HLH: (1) HLH as the initial presentation of malignancy, (2) Chemotherapy Associated HLH, (3) Cytokine Release Syndrome (CRS) Associated HLH-like Syndrome, and (4) immune effector cell-associated HLH-like syndrome. Diagnosis is complicated by overlap with cancer symptoms and limitations of existing criteria. The Optimized HLH Inflammatory (OHI) index shows promise in early identification of hyperinflammation in new-onset hematologic malignancies. Treatment approaches must balance controlling hyperinflammation with addressing the underlying malignancy. Emerging therapies, including targeted agents like anakinra, ruxolitinib, and emapalumab, offer new management possibilities. This review examines the current understanding of M-HLH pathophysiology, diagnostic approaches, and treatment strategies for each subtype. It underscores the critical need for further research to unravel underlying mechanisms and establish evidence-based treatment protocols. Given the complexity of M-HLH, international collaborative efforts are essential to advance knowledge and improve patient outcomes.
Collapse
Affiliation(s)
- Adi Zoref-Lorenz
- Meir Medical Center, Hematology Institute, Tel Aviv University, Tel Aviv, Israel
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Thomas E Witzig
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - James R Cerhan
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Michael B Jordan
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Bone Marrow Transplantation, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
20
|
Santoro A, Angelico G, Travaglino A, Inzani F, Spadola S, Pettinato A, Mazzucchelli M, Bragantini E, Maccio L, Zannoni GF. The multiple facets of ovarian high grade serous carcinoma: A review on morphological, immunohistochemical and molecular features. Crit Rev Oncol Hematol 2025; 208:104603. [PMID: 39732305 DOI: 10.1016/j.critrevonc.2024.104603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/06/2024] [Accepted: 12/20/2024] [Indexed: 12/30/2024] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) is the most aggressive subtype of epithelial ovarian cancer and a leading cause of mortality among gynecologic malignancies. This review aims to comprehensively analyze the morphological, immunohistochemical, and molecular features of HGSOC, highlighting its pathogenesis and identifying biomarkers with diagnostic, prognostic, and therapeutic significance. Special emphasis is placed on the role of tumor microenvironment (TME) and genomic instability in shaping the tumor's behavior and therapeutic vulnerabilities. Key advancements, such as the identification of TP53 and BRCA mutations, the classification of homologous recombination repair (HRR) deficiencies, and the clinical implications of biomarkers like folate receptor alpha (FRα) and PD-L1 are discussed. These findings reveal actionable insights into targeted therapies, including immune checkpoint inhibitors and PARP inhibitors, which hold promise for improving outcomes in HGSOC. This synthesis of knowledge aims to bridge gaps in understanding HGSOC's multifaceted biology, enhance clinical decision-making, and foster the development of precision therapies.
Collapse
Affiliation(s)
- Angela Santoro
- Pathology Institute, Catholic University of Sacred Heart, Rome 00168, Italy; Pathology Unit, Department of Woman and Child's Health and Public Health Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome 00168, Italy
| | - Giuseppe Angelico
- Department of Medicine and Surgery, Kore University of Enna, Enna 94100, Italy
| | - Antonio Travaglino
- Pathology Unit, Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
| | - Frediano Inzani
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia and Fondazione IRCCS San Matteo Hospital, Pavia 27100, Italy
| | - Saveria Spadola
- Department of Medicine and Surgery, Kore University of Enna, Enna 94100, Italy
| | - Angela Pettinato
- Department of Pathological Anatomy, A.O.E. Cannizzaro, Via Messina, 829, Catania 95126, Italy
| | - Manuel Mazzucchelli
- Department of Medical and Surgical Sciences and Advanced Technologies "G.F. Ingrassia", Anatomic Pathology, University of Catania, Catania, Italy
| | - Emma Bragantini
- Unit of Surgical Pathology, Santa Chiara Hospital, APSS, Trento, Italy
| | - Livia Maccio
- Unit of Surgical Pathology, Santa Chiara Hospital, APSS, Trento, Italy
| | - Gian Franco Zannoni
- Pathology Institute, Catholic University of Sacred Heart, Rome 00168, Italy; Pathology Unit, Department of Woman and Child's Health and Public Health Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome 00168, Italy.
| |
Collapse
|
21
|
Maillie L, Nasta SD, Svoboda J, Barta SK, Chong EA, Garfall AL, Gill SI, Porter DL, Schuster SJ, Catania C, Frey NV, Landsburg DJ. Prolonged Neurologic Symptoms Following Immune Effector Cell-Associated Neurotoxicity Syndrome in Patients With Large B-cell Lymphoma Treated With Chimeric Antigen Receptor-Modified T Cell Therapy. Transplant Cell Ther 2025; 31:228-235. [PMID: 39848443 DOI: 10.1016/j.jtct.2025.01.884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/31/2024] [Accepted: 01/14/2025] [Indexed: 01/25/2025]
Abstract
While immune effector cell-associated neurotoxicity syndrome (ICANS) is a well-defined adverse effect associated with chimeric antigen receptor-modified T cell (CAR-T) therapy, some patients develop prolonged neurologic symptoms. Few studies have examined characteristics and outcomes of patients who develop such symptoms. The objective of this study was to provide an analysis of patients who developed ICANS in a single-center cohort of patients with large B-cell lymphoma (LBCL) who received commercial CAR-T and compare characteristics and outcomes between patients with vs. without subsequent prolonged neurologic symptoms. We examined a retrospective cohort of patients with LBCL treated with CAR-T at our institution who developed ICANS. Prolonged neurologic symptoms were defined as those lasting longer than four weeks. Thirty three of 278 (12%) LBCL patients treated with commercial CAR-T experienced ICANS. Nine patients (27%) experienced prolonged neurologic symptoms following ICANS, eight with ICANS grade ≥3 (high-grade) and one with ICANS grade <3 (low-grade). There were a range of symptoms experienced by these patients including difficulties with short-term memory, difficulties with long-term memory, aphasia, and tremors. The incidence of prolonged neurologic symptoms was greater in patients experiencing high-grade as compared to low-grade ICANS (42.1% vs. 7.1%, P = .049). However, no other pre-treatment characteristics or post-treatment outcomes were associated with development of prolonged neurologic symptoms following ICANS. In summary, nearly half of all patients with LBCL treated with CAR-T at our institution who developed high-grade ICANS experienced prolonged neurologic symptoms; however, pretreatment characteristics and post-treatment outcomes were not predictive of this clinical condition. Further work is needed to identify patients treated with CAR-T at risk for experiencing prolonged neurologic symptoms and developing strategies for evaluation and treatment of this toxicity.
Collapse
Affiliation(s)
- Luke Maillie
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Sunita D Nasta
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jakub Svoboda
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stefan K Barta
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elise A Chong
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alfred L Garfall
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Saar I Gill
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David L Porter
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stephen J Schuster
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christopher Catania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Noelle V Frey
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daniel J Landsburg
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
22
|
Shen R, Cao W, Wang L, Sheng L, Zhang Y, Wu W, Xu P, Cheng S, Liu M, Dong Y, Wang Y, Weng X, Jiang X, Song Q, Yi H, Li L, Chen S, Yan Z, Zhao W. Response-adapted zanubrutinib and tislelizumab as a potential strategy to enhance CD19 CAR T-cell therapy in relapsed/refractory large B-cell lymphoma: A retrospective observational study. Clin Transl Med 2025; 15:e70310. [PMID: 40268516 PMCID: PMC12017895 DOI: 10.1002/ctm2.70310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/30/2025] [Accepted: 04/08/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND CD19 chimeric antigen receptor (CAR) T-cell therapy is a potential treatment for relapsed/refractory (R/R) large B-cell lymphoma (LBCL). The combination of targeted therapeutic strategies, particularly bruton tyrosine kinase inhibitor zanubrutinib and programmed death-1 inhibitor tislelizumab, may improve clinical outcomes and modulate the tumour microenvironment (TME). METHODS We studied patients with R/R LBCL who received response-adapted zanubrutinib plus tislelizumab upon CD19 CAR T-cell therapy between June 2021 and March 2023. Patients were treated with zanubrutinib daily from leukapheresis to day 28 post-infusion; those achieving complete response continued zanubrutinib monotherapy for 3 months, while partial responders received combined zanubrutinib for 3 months and tislelizumab for up to 2 years. We evaluated the overall response rate (ORR), complete response rate (CRR), progression-free survival (PFS), overall survival (OS), and safety. DNA sequencing and RNA sequencing were performed on available tumour samples to analyse genetic aberrations and TME characteristics. RESULTS A total of 54 patients with LBCL were included, with a median follow-up of 23.6 months. The ORR at day 28, month 3, and month 6 were 94% (CRR 66%), 87% (CRR 80%), and 80% (CRR 76%), respectively. The 2-year PFS and 2-year OS rates were 68% and 76%, respectively. Median PFS and median OS were not reached. Grade ≥ 3 cytokine release syndrome occurred in 9% of patients, with no grade ≥ 3 neurotoxicity observed. Genomic and transcriptomic data indicated that this regimen was effective across genetic subtypes and abrogated T-cell exhaustion within the TME. However, tumour-infiltrating M2 macrophages with dysregulated lipid metabolism were associated with poor clinical outcome. CONCLUSIONS Response-adapted zanubrutinib and tislelizumab potentially enhances the efficacy of CAR T-cell therapy with a favourable safety profile in R/R LBCL, effectively counteracting T-cell exhaustion. Future studies should focus on targeting M2 macrophages by reprogramming lipid metabolism to further attenuate the immunosuppressive TME. HIGHLIGHTS Response-adapted zanubrutinib plus tislelizumab potentially enhances the efficacy of CAR T-cell therapy for R/R LBCL with acceptable safety profile. This regimen functions independently of genetic subtypes, rendering it more applicable for clinical practice with CAR T-cell therapy. This regimen effectively abrogates T-cell exhaustion, but fails to overcome the immunosuppressive effects of M2 macrophages, providing a rationale for remodelling TME to optimise CAR T-cell therapy.
Collapse
MESH Headings
- Humans
- Female
- Male
- Middle Aged
- Retrospective Studies
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/administration & dosage
- Aged
- Pyrimidines/therapeutic use
- Pyrimidines/pharmacology
- Pyrimidines/administration & dosage
- Piperidines/therapeutic use
- Piperidines/pharmacology
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/therapy
- Adult
- Immunotherapy, Adoptive/methods
- Antigens, CD19
- Pyrazoles
Collapse
Affiliation(s)
- Rong Shen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at ShanghaiRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wei‐Guo Cao
- Department of Radiation OncologyRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Li Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at ShanghaiRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Pôle de Recherches Sino‐Français en Science du Vivant et Génomique, Laboratory of Molecular PathologyShanghaiChina
| | - Ling‐Shuang Sheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at ShanghaiRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yi‐Lun Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at ShanghaiRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wen Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at ShanghaiRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Peng‐Peng Xu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at ShanghaiRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shu Cheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at ShanghaiRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Meng‐Ke Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at ShanghaiRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yan Dong
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at ShanghaiRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yue Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at ShanghaiRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiang‐Qin Weng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at ShanghaiRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xu‐Feng Jiang
- Department of Nuclear MedicineRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qi Song
- Department of RadiologyRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hong‐Mei Yi
- Department of PathologyRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lei Li
- Department of Critical Care MedicineRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Sheng Chen
- Department of NeurologyRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zi‐Xun Yan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at ShanghaiRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wei‐Li Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at ShanghaiRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Pôle de Recherches Sino‐Français en Science du Vivant et Génomique, Laboratory of Molecular PathologyShanghaiChina
| |
Collapse
|
23
|
Warrick KA, Vallez CN, Meibers HE, Pasare C. Bidirectional Communication Between the Innate and Adaptive Immune Systems. Annu Rev Immunol 2025; 43:489-514. [PMID: 40279312 DOI: 10.1146/annurev-immunol-083122-040624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
Effective bidirectional communication between the innate and adaptive immune systems is crucial for tissue homeostasis and protective immunity against infections. The innate immune system is responsible for the early sensing of and initial response to threats, including microbial ligands, toxins, and tissue damage. Pathogen-related information, detected primarily by the innate immune system via dendritic cells, is relayed to adaptive immune cells, leading to the priming and differentiation of naive T cells into effector and memory lineages. Memory T cells that persist long after pathogen clearance are integral for durable protective immunity. In addition to rapidly responding to reinfections, memory T cells also directly instruct the interacting myeloid cells to induce innate inflammation, which resembles microbial inflammation. As such, memory T cells act as newly emerging activators of the innate immune system and function independently of direct microbial recognition. While T cell-mediated activation of the innate immune system likely evolved as a protective mechanism to combat reinfections by virulent pathogens, the detrimental outcomes of this mechanism manifest in the forms of autoimmunity and other T cell-driven pathologies. Here, we review the complexities and layers of regulation at the interface between the innate and adaptive immune systems to highlight the implications of adaptive instruction of innate immunity in health and disease.
Collapse
Affiliation(s)
- Kathrynne A Warrick
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| | - Charles N Vallez
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| | - Hannah E Meibers
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| | - Chandrashekhar Pasare
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA ;
| |
Collapse
|
24
|
Cao L, Liu Y, Lin G. Strategies for Altering Delivery Technologies to Optimize CAR Therapy. Int J Mol Sci 2025; 26:3206. [PMID: 40244018 PMCID: PMC11989270 DOI: 10.3390/ijms26073206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has been proven to be an effective strategy for the treatment of hematological malignancies. At present, how to prepare CAR-T cells efficiently, quickly, and safely is one of the urgent problems to be solved. The durability and activity of engineered T cells in solid tumors need to be further improved, and the strategy of T cells penetrating the tumor microenvironment also needs to be improved. In addition, although the problems mainly caused by T-cell biology are being solved, the manufacturing mode and process still need to be improved to ensure that CAR-T cell therapy can be widely used. This paper summarizes some strategies that can improve the efficacy of CAR-T cells.
Collapse
Affiliation(s)
- Lili Cao
- Student Counseling Center, Shandong University, Jinan 250012, China;
| | - Yingying Liu
- School of Pharmaceutical Science, Shandong University, Jinan 250012, China;
| | - Guimei Lin
- School of Pharmaceutical Science, Shandong University, Jinan 250012, China;
| |
Collapse
|
25
|
Lee JC, Johnson WT, Hines M, Shah NN. Immune Effector Cell-associated Hemophagocytic Lymphohistiocytosis-like Syndrome (IEC-HS). Hematol Oncol Clin North Am 2025:S0889-8588(25)00020-6. [PMID: 40158936 DOI: 10.1016/j.hoc.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Immune effector cell-associated hemophagocytic lymphohistiocytosis-like syndrome (IEC-HS) is a complication of adoptive cell therapy. Presenting with clinical manifestations of hyperinflammation and surrogate indicators of hyperinflammation such as elevations in serum ferritin and hepatic transaminases, decreasing cell counts, and hypofibrinogenemia, IEC-HS resembles primary and other forms of secondary hemophagocytic lymphohistiocytosis. Nonetheless, this is an iatrogenic complication resulting from the induction of hyperinflammatory pathways during T-cell-mediated anticancer targeting. Distinct from cytokine release syndrome, IEC-associated neurotoxicity syndrome, and IEC-associated hematotoxicity, IEC-HS can be life-threatening. Identification of IEC-HS, optimization of treatment strategies, and use of supportive care are critical to improving outcomes.
Collapse
Affiliation(s)
- Jerry C Lee
- Hematology, Blood and Marrow Transplantation, and Cellular Therapy Program, Division of Hematology/Oncology, Department of Medicine, University of California, UCSF Box 0345, 400 Parnassus Avenue, San Francisco, CA 94143, USA
| | - William T Johnson
- Lymphoma Service and Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Melissa Hines
- Division of Critical Care Medicine, Department of Pediatric Medicine, St. Jude Children's Research Hospital, MS #734262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, MD, USA.
| |
Collapse
|
26
|
Arvanitis P, Tziotis A, Papadimatos S, Farmakiotis D. Pathogenesis, Diagnosis, and Management of Cytokine Release Syndrome in Patients with Cancer: Focus on Infectious Disease Considerations. Curr Oncol 2025; 32:198. [PMID: 40277755 PMCID: PMC12026323 DOI: 10.3390/curroncol32040198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025] Open
Abstract
Background: Cytokine Release Syndrome (CRS) is a hyperinflammatory state triggered by immune therapies like CAR T-cell therapy and bispecific T-cell engagers (BiTEs). Characterized by excessive cytokine release, CRS often mimics infectious and inflammatory conditions, complicating diagnosis and treatment. Immunosuppressive therapies used for CRS further elevate the risk of secondary infections. Methods: A systematic search of PubMed and EMBASE was conducted using terms related to "cytokine release syndrome", "cytokine storm", "infections", and "management". Studies were included if they described infectious complications, diagnostic mimics, or therapeutic approaches related to CRS. Results: Of 19,634 studies, 2572 abstracts were reviewed. Infections occurred in up to 23% of patients post-CAR T therapy and 24% post-BiTE therapy. Pathogens included gram-positive and gram-negative bacteria, herpesviruses (e.g., CMV, HSV), fungi (e.g., Candida, Aspergillus), and parasites (e.g., Toxoplasma gondii). CRS mimics also included non-infectious inflammatory syndromes. Differentiation remains challenging, but cytokine profiling and biomarkers (e.g., ferritin, CRP, sIL-2Rα) may aid in diagnosis. Treatments included tocilizumab, corticosteroids, and empiric antimicrobials. Prophylactic strategies were inconsistently reported. Conclusions: Effective CRS management requires early recognition, differentiation from infectious mimics, and collaboration between oncology and infectious disease (ID) specialists. A multidisciplinary, collaborative, and structured approach, including dedicated ID input and pre-treatment evaluation, is essential for optimizing CRS management and patient outcomes.
Collapse
Affiliation(s)
- Panos Arvanitis
- Division of Infectious Diseases, The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Andreas Tziotis
- Beth Israel Deaconess Medical Center Division of Gastroenterology, Boston, MA 02115, USA; (A.T.); (S.P.)
| | - Spyridon Papadimatos
- Beth Israel Deaconess Medical Center Division of Gastroenterology, Boston, MA 02115, USA; (A.T.); (S.P.)
| | - Dimitrios Farmakiotis
- Beth Israel Deaconess Medical Center Division of Infectious Diseases, Boston, MA 02115, USA
| |
Collapse
|
27
|
Li YR, Zhu Y, Chen Y, Yang L. The clinical landscape of CAR-engineered unconventional T cells. Trends Cancer 2025:S2405-8033(25)00069-X. [PMID: 40155286 DOI: 10.1016/j.trecan.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
Unconventional T cells, such as invariant natural killer T (iNKT), γδ T, and mucosal-associated invariant T (MAIT) cells, play a pivotal role in bridging innate and adaptive immunity. Their capacity for rapid tumor targeting and effective modulation of the tumor microenvironment (TME) makes them promising candidates for cancer immunotherapy. Advances in chimeric antigen receptor (CAR) engineering have further highlighted their therapeutic potential, particularly for treating challenging cancers. Notably, these cells exhibit favorable safety profiles, enhancing their viability as off-the-shelf therapeutic options. We provide a comprehensive analysis of the clinical applications of CAR-engineered unconventional T cells, focusing on genetic modifications, manufacturing processes, preconditioning regimens, and dosing strategies. We discuss successful examples from recent clinical trials and explore future directions for utilizing these cells in cancer therapy and beyond.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology, and Molecular Genetics (MIMG), University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA.
| | - Yichen Zhu
- Department of Microbiology, Immunology, and Molecular Genetics (MIMG), University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Yuning Chen
- Department of Microbiology, Immunology, and Molecular Genetics (MIMG), University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Lili Yang
- Department of Microbiology, Immunology, and Molecular Genetics (MIMG), University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; Parker Institute for Cancer Immunotherapy, University of California Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
28
|
Schwarzlmueller P, Triebig A, Assié G, Jouinot A, Theurich S, Maier T, Beuschlein F, Kobold S, Kroiss M. Steroid hormones as modulators of anti-tumoural immunity. Nat Rev Endocrinol 2025:10.1038/s41574-025-01102-2. [PMID: 40128599 DOI: 10.1038/s41574-025-01102-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/27/2025] [Indexed: 03/26/2025]
Abstract
Immune evasion is a hallmark of cancer progression but the role of steroid hormones in this evasion has long been underrated. This oversight is particularly notable for glucocorticoids given that exogenous glucocorticoids remain a cornerstone therapy in various oncological treatment regimens, supportive care and treatment of immune-related adverse events caused by immune-checkpoint inhibitors. Cortisol, the main endogenous glucocorticoid in humans, is secreted by the adrenal cortex in response to stress. Additionally, cortisol and its inactive metabolite cortisone can be interconverted to further modulate tissue-dependent glucocorticoid action. In the past 5 years, intratumoural production of glucocorticoids, by both immune and tumour cells, has been shown to support tumour immune evasion. Here, we summarize current progress at the crossroads of endocrinology and immuno-oncology. We outline the known effects of steroid hormones on different immune cell types with a focus on glucocorticoids and androgens. We conclude with options for pharmaceutical intervention, including the engineering of cell-based therapies that resist the immunosuppressive action of steroid hormones. Overall, local steroid production and metabolism are emerging elements of tumour immune suppression that are potentially amenable to therapeutic intervention. Targeting steroid hormones to enhance anticancer therapies could increase their efficacy but will require expertise in endocrine care.
Collapse
Affiliation(s)
| | - Alexandra Triebig
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Guillaume Assié
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
- Department of Endocrinology and National Reference Center for Rare Adrenal Disorders, Hôpital Cochin, Paris, France
| | - Anne Jouinot
- Department of Endocrinology and National Reference Center for Rare Adrenal Disorders, Hôpital Cochin, Paris, France
- Université Paris Cité, Institut Cochin, Paris, France
| | - Sebastian Theurich
- Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital, Munich, Germany
- Cancer- and Immunometabolism Research Group, Gene Center, Ludwig Maximilian University (LMU), Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- German Cancer Consortium (DKTK), Munich Site, Heidelberg, Germany
| | - Tanja Maier
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Felix Beuschlein
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ), Zurich, Switzerland
- The LOOP Zurich - Medical Research Center, Zurich, Switzerland
| | - Sebastian Kobold
- German Cancer Consortium (DKTK), Munich Site, Heidelberg, Germany
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Matthias Kroiss
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany.
- Bavarian Cancer Research Center (BZKF), Munich, Germany.
- Kroiss Endokrinologie & Diabetologie, Schweinfurt, Germany.
| |
Collapse
|
29
|
Sabha Y, Kim SH, Tseng HC, Byrne ME, Tsao WC, Lee SH, Zhou Z, Jang MH, Liu D. CD147-CAR-NK cell therapy shows minimal toxicities in human CD147 transgenic mouse model with solid tumors. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200957. [PMID: 40160933 PMCID: PMC11952776 DOI: 10.1016/j.omton.2025.200957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 11/28/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025]
Abstract
The toxicity of chimeric antigen receptor-natural killer (CAR-NK) therapy has not been tested in solid tumors, compared with CAR-T therapy side by side. To address this, we investigated the CD147-CAR-NK "on-target/off-tumor" toxicity and neurotoxicity in human CD147-transgenic (hCD147TG) mice with hepatocellular carcinoma (HCC). We first tested the in vitro cytotoxicity of CD147-CAR-NK against CD147+ tumor and CD147+ healthy cells. Both CD147-CAR-NK cells and CD147-IL15-CAR-NK (autocrine expressing interleukin [IL]-15) can kill tumor cells specifically but not CD147+ healthy lung and spleen tissue from hCD147TG mice. In vivo assays show minimal systemic toxicities against CD147+ healthy tissues but 1-week-longer persistence times in tumor than non-tumor tissues. To evaluate neurotoxicity, we compared the expression of ionized calcium-binding adaptor protein 1 (IBA1), glial fibrillary acidic protein (GFAP), and inducible nitric oxide synthase (iNOS) between CD147-CAR-T- and CD147-CAR-NK-treated hCD147TG mice with HCC. Both CD147-CAR-T- and CD147-CAR-NK-treated mice exhibited higher GFAP and IBA1 expression than control groups. CD147-CAR-T-treated mice showed an increase in iNOS compared to the control groups. The behavioral studies testing spatial memory showed that mice treated with CD147-CAR-NK exhibit better memory function than CD147-CAR-T-treated mice. This study provides a deeper understanding of the CD147-CAR-NK systemic toxicities and neurotoxicity of CD147-CAR-NK relative to CD147-CAR-T therapy.
Collapse
Affiliation(s)
- Youssef Sabha
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University New Jersey Medical School, 180 South Orange Avenue, Newark, NJ 07103, USA
| | - Sang Hoon Kim
- Department of Neurosurgery, Robert Wood Johnson Medical School, The State University of New Jersey, 661 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Hsiang-chi Tseng
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University New Jersey Medical School, 180 South Orange Avenue, Newark, NJ 07103, USA
| | - Maeve Elizabeth Byrne
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University New Jersey Medical School, 180 South Orange Avenue, Newark, NJ 07103, USA
| | - Wei-Chung Tsao
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University New Jersey Medical School, 180 South Orange Avenue, Newark, NJ 07103, USA
| | - Sang Hoon Lee
- Department of Neurosurgery, Robert Wood Johnson Medical School, The State University of New Jersey, 661 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Zhongren Zhou
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, The State University of New Jersey, 1 Robert Wood Johnson Place, New Brunswick, NJ 08901, USA
| | - Mi-Hyeon Jang
- Department of Neurosurgery, Robert Wood Johnson Medical School, The State University of New Jersey, 661 Hoes Lane West, Piscataway, NJ 08854, USA
| | - Dongfang Liu
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University New Jersey Medical School, 180 South Orange Avenue, Newark, NJ 07103, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, 205 South Orange Avenue, Newark, NJ 07101, USA
| |
Collapse
|
30
|
Renninger J, Kurz L, Stein H. Mitigation and Management of Common Toxicities Associated with the Administration of CAR-T Therapies in Oncology Patients. Drug Saf 2025:10.1007/s40264-025-01538-5. [PMID: 40108072 DOI: 10.1007/s40264-025-01538-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2025] [Indexed: 03/22/2025]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapies are one of the main approaches among targeted cellular therapies. Despite the potential benefit and durable responses observed in some patients receiving CAR-T therapies, serious and potentially fatal toxicities remain a major challenge. The most common CAR-T-associated toxicities include cytokine release syndrome (CRS), neurotoxicity, cytopenias, and infections. While CRS and neurotoxicity are generally managed with tocilizumab and corticosteroids, respectively, high-grade toxicities can be life-threatening. Close postinfusion monitoring and assessment of clinical laboratory parameters, patient-related and clinical risk factors (e.g., age, tumor burden, comorbidities, baseline laboratory parameters, and underlying abnormalities), and therapy-related risk factors (e.g., CAR-T type, dose, and CAR-T-induced toxicity) are effective strategies to mitigate the toxicities. Clinical laboratory parameters, including various cytokines, have been identified for CRS (interleukin [IL]-1, IL-2, IL-5, IL-6, IL-8, IL-10, C-reactive protein [CRP], interferon [IFN]-γ, ferritin, granulocyte-macrophage colony-stimulating factor [GM-CSF], and monocyte chemoattractant protein-1), neurotoxicity (IL-1, IL-2, IL-6, IL-15, tumor necrosis factor [TNF]-α, GM-CSF, and IFN-γ), cytopenias (IL-2, IL-4, IL-6, IL-10, IFN-γ, ferritin, and CRP), and infections (IL-8, IL-1β, CRP, IFN-γ, and procalcitonin). CAR-T-associated toxicities can be monitored and treated to mitigate the risk to patients. Assessment of alterations in clinical laboratory parameter values that are correlated with CAR-T-associated toxicities may predict development and/or severity of a given toxicity, which can improve patient management strategies and ultimately enable the patients to better tolerate these therapies.
Collapse
Affiliation(s)
- Jonathan Renninger
- GSK Safety Evaluation and Risk Management, Global Safety, Philadelphia, PA, USA.
| | - Lisa Kurz
- GSK Safety Evaluation and Risk Management, Global Safety, Upper Providence, PA, USA
| | - Heather Stein
- GSK Safety Evaluation and Risk Management, Global Safety, Cambridge, MA, USA
| |
Collapse
|
31
|
Erasha AM, EL-Gendy H, Aly AS, Fernández-Ortiz M, Sayed RKA. The Role of the Tumor Microenvironment (TME) in Advancing Cancer Therapies: Immune System Interactions, Tumor-Infiltrating Lymphocytes (TILs), and the Role of Exosomes and Inflammasomes. Int J Mol Sci 2025; 26:2716. [PMID: 40141358 PMCID: PMC11942452 DOI: 10.3390/ijms26062716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Understanding how different contributors within the tumor microenvironment (TME) function and communicate is essential for effective cancer detection and treatment. The TME encompasses all the surroundings of a tumor such as blood vessels, fibroblasts, immune cells, signaling molecules, exosomes, and the extracellular matrix (ECM). Subsequently, effective cancer therapy relies on addressing TME alterations, known drivers of tumor progression, immune evasion, and metastasis. Immune cells and other cell types act differently under cancerous conditions, either driving or hindering cancer progression. For instance, tumor-infiltrating lymphocytes (TILs) include lymphocytes of B and T cell types that can invade malignancies, bringing in and enhancing the ability of immune system to recognize and destroy cancer cells. Therefore, TILs display a promising approach to tackling the TME alterations and have the capability to significantly hinder cancer progression. Similarly, exosomes and inflammasomes exhibit a dual effect, resulting in either tumor progression or inhibition depending on the origin of exosomes, type of inflammasome and tumor. This review will explore how cells function in the presence of a tumor, the communication between cancer cells and immune cells, and the role of TILs, exosomes and inflammasomes within the TME. The efforts in this review are aimed at garnering interest in safer and durable therapies for cancer, in addition to providing a promising avenue for advancing cancer therapy and consequently improving survival rates.
Collapse
Affiliation(s)
- Atef M. Erasha
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sadat City University, Sadat City 32897, Egypt;
| | - Hanem EL-Gendy
- Department of Pharmacology, Faculty of Veterinary Medicine, Sadat City University, Sadat City 32897, Egypt;
| | - Ahmed S. Aly
- Department of Animal Production, Faculty of Agriculture, Ain Shams University, Cairo 11241, Egypt;
| | - Marisol Fernández-Ortiz
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA
| | - Ramy K. A. Sayed
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, Sohag 82524, Egypt;
| |
Collapse
|
32
|
Cai J, Liu Z, Chen S, Zhang J, Li H, Wang X, Yang F, Wang S, Li X, Li Y, Chen K, Wang J, Sun M, Qiu M. Engineered circular RNA-based DLL3-targeted CAR-T therapy for small cell lung cancer. Exp Hematol Oncol 2025; 14:35. [PMID: 40075480 PMCID: PMC11905684 DOI: 10.1186/s40164-025-00625-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
PURPOSE Circular RNA (circRNA) has emerged as a promising RNA therapeutic molecule due to its enhanced stability and prolonged protein expression compared to messenger RNA (mRNA). Using circRNA to construct transient Chimeric Antigen Receptor (CAR)-T cells can mitigate the limitations of conventional viral vector-based CAR-T approaches, such as complex process and long-term side effects. METHODS The study first reconfirmed the advantageous properties of circRNA, focusing on its stability and protein expression efficiency. Electroporation conditions were then optimized for the efficient delivery of circRNA into human primary T cells. Subsequently, a circRNA encoding the anti-Delta-like Ligand 3 (DLL3) CAR was constructed, and CAR-T cells were generated via electroporation. The efficacy of circRNA-based CAR-T cells was compared to mRNA-based CAR-T cells in both in vitro and in vivo models, including subcutaneous and orthotopic small cell lung cancer (SCLC) mouse models. RESULTS CircRNA-based CAR-T cells demonstrated superior efficacy against SCLC compared to mRNA-based CAR-T cells. In vitro experiments showed enhanced tumor-killing effects, while in vivo studies revealed complete elimination of human SCLC tumors in both subcutaneous and orthotopic mouse models. These results underscored the therapeutic advantages of circRNA in CAR-T cell therapy. CONCLUSIONS This study validated the feasibility of the circRNA-electroporation strategy in CAR-T cell therapy and offered a potentially effective approach for treating SCLC, highlighting the potential of circRNA-based technologies in advancing cell therapies.
Collapse
Affiliation(s)
- Jingsheng Cai
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
- Department of Thoracic Surgery, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China
| | - Zheng Liu
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
- Department of Thoracic Surgery, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China
| | - Shaoyi Chen
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
- Department of Thoracic Surgery, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China
| | - Jingwei Zhang
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
- Department of Thoracic Surgery, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China
| | - Haoran Li
- Department of Thoracic Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital& Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China (UESTC), Chengdu, 610041, China
| | - Xun Wang
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
- Department of Thoracic Surgery, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China
| | - Feng Yang
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
- Department of Thoracic Surgery, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China
| | - Shaodong Wang
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
- Department of Thoracic Surgery, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China
| | - Xiao Li
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
- Department of Thoracic Surgery, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China
| | - Yun Li
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
- Department of Thoracic Surgery, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China
| | - Kezhong Chen
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China
- Department of Thoracic Surgery, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China
| | - Jun Wang
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China.
- Department of Thoracic Surgery, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China.
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China.
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China.
| | - Ming Sun
- Department of Oncology Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, No. 16 Baita West Road, Suzhou, 215001, China.
| | - Mantang Qiu
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, 100044, China.
- Department of Thoracic Surgery, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China.
- Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Chinese Academy of Medical Sciences, 2021RU002, Peking University People's Hospital, Beijing, 100044, China.
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China.
| |
Collapse
|
33
|
He J, Connors J, Meador A, Xu S, Meador H, Jiang H, Fueyo J, Gomez-Manzano C, Friedman GK, Zaky W, Sadighi Z, Slopis JM, Ahmad AH. Immunotherapy-related neurotoxicity in the central nervous system of children with cancer. Neuro Oncol 2025; 27:625-643. [PMID: 39535217 PMCID: PMC11889721 DOI: 10.1093/neuonc/noae243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Indexed: 11/16/2024] Open
Abstract
Significant gaps remain in our understanding of immunotherapy-related neurotoxicity in pediatric patients, largely because much of our knowledge comes from studies in adults. Accurately identifying the adverse effects of immunotherapy in children is also challenging, owing to variations in terminology and grading systems. Moreover, the manifestation of immunotherapy-related neurotoxicity differs greatly across different diseases, various modalities, dosages, and delivery methods. Combining immunotherapy with other treatments might improve outcomes but introduces new complexities and potential for increased toxicities. Additionally, pediatric patients with intracranial malignancy have unique responses to immunotherapies and distinct neurotoxicity compared to those with extracranial malignancy. Consequently, we must enhance our understanding of the pathophysiology, prevalence, severity, and management of immunotherapy's neurotoxic effects in this vulnerable group. This review consolidates the current knowledge of immunotherapy-related neurotoxicity in pediatric oncology, highlighting various types of neurotoxicity including cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and tumor inflammation-associated neurotoxicity (TIAN), among others. Furthermore, we examine the unique features of neurotoxicity associated with adoptive cellular therapy (ACT), antibody-based therapies, immune checkpoint inhibitors (ICIs), oncolytic viruses (OV), and cancer vaccines.
Collapse
Affiliation(s)
- Jiasen He
- Section of Pediatric Neuro-Oncology, Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeremy Connors
- Section of Stem Cell Transplant, Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Andrew Meador
- Texas A&M University School of Medicine, Bryan, Texas, USA
| | - Shuo Xu
- Section of Pediatric Hematology Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Heather Meador
- Section of Pediatric Neuro-Oncology, Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hong Jiang
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Juan Fueyo
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Candelaria Gomez-Manzano
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gregory K Friedman
- Section of Pediatric Neuro-Oncology, Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wafik Zaky
- Section of Pediatric Neuro-Oncology, Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zsila Sadighi
- Section of Pediatric Neuro-Oncology, Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - John M Slopis
- Section of Pediatric Neuro-Oncology, Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ali H Ahmad
- Section of Pediatric Critical Care, Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
34
|
Adams RC, MacDonald KPA, Hill GR. The contribution of the monocyte-macrophage lineage to immunotherapy outcomes. Blood 2025; 145:1010-1021. [PMID: 39576958 DOI: 10.1182/blood.2024025680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/21/2024] [Accepted: 11/04/2024] [Indexed: 11/24/2024] Open
Abstract
ABSTRACT Macrophages execute core functions in maintaining tissue homeostasis, in which their extensive plasticity permits a spectrum of functions from tissue remodeling to immune defense. However, perturbations to tissue-resident macrophages during disease, and the subsequent emergence of monocyte-derived macrophages, can hinder tissue recovery and promote further damage through inflammatory and fibrotic programs. Gaining a fundamental understanding of the critical pathways defining pathogenic macrophage populations enables the development of targeted therapeutic approaches to improve disease outcomes. In the setting of chronic graft-versus-host disease (cGVHD), which remains the major complication of allogeneic hematopoietic stem cell transplantation, colony-stimulating factor 1 (CSF1)-dependent donor-derived macrophages have been identified as key pathogenic mediators of fibrotic skin and lung disease. Antibody blockade of the CSF1 receptor (CSF1R) to induce macrophage depletion showed remarkable capacity to prevent fibrosis in preclinical models and has subsequently demonstrated impressive efficacy for improving cGVHD in ongoing clinical trials. Similarly, macrophage depletion approaches are currently under investigation for their potential to augment responses to immune checkpoint inhibition. Moreover, both monocyte and tissue-resident macrophage populations have recently been implicated as mediators of the numerous toxicities associated with chimeric antigen receptor T-cell therapy, further highlighting potential avenues of macrophage-based interventions to improve clinical outcomes. Herein, we examine the current literature on basic macrophage biology and contextualize this in the setting of cellular and immunotherapy. Additionally, we highlight mechanisms by which macrophages can be targeted, largely by interfering with the CSF1/CSF1R signaling axis, for therapeutic benefit in the context of both cellular and immunotherapy.
Collapse
Affiliation(s)
- Rachael C Adams
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Kelli P A MacDonald
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Geoffrey R Hill
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| |
Collapse
|
35
|
Li R, Grosskopf AK, Joslyn LR, Stefanich EG, Shivva V. Cellular Kinetics and Biodistribution of Adoptive T Cell Therapies: from Biological Principles to Effects on Patient Outcomes. AAPS J 2025; 27:55. [PMID: 40032717 DOI: 10.1208/s12248-025-01017-w] [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: 11/12/2024] [Accepted: 01/06/2025] [Indexed: 03/05/2025] Open
Abstract
Cell-based immunotherapy has revolutionized cancer treatment in recent years and is rapidly expanding as one of the major therapeutic options in immuno-oncology. So far ten adoptive T cell therapies (TCTs) have been approved by the health authorities for cancer treatment, and they have shown remarkable anti-tumor efficacy with potent and durable responses. While adoptive T cell therapies have shown success in treating hematological malignancies, they are lagging behind in establishing promising efficacy in treating solid tumors, partially due to our incomplete understanding of the cellular kinetics (CK) and biodistribution (including tumoral penetration) of cell therapy products. Indeed, recent clinical studies have provided ample evidence that CK of TCTs can influence clinical outcomes in both hematological malignancies and solid tumors. In this review, we will discuss the current knowledge on the CK and biodistribution of anti-tumor TCTs. We will first describe the typical CK and biodistribution characteristics of these "living" drugs, and the biological factors that influence these characteristics. We will then review the relationships between CK and pharmacological responses of TCT, and potential strategies in enhancing the persistence and tumoral penetration of TCTs in the clinic. Finally, we will also summarize bioanalytical methods, preclinical in vitro and in vivo tools, and in silico modeling approaches used to assess the CK and biodistribution of TCTs.
Collapse
Affiliation(s)
- Ran Li
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA.
| | - Abigail K Grosskopf
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Louis R Joslyn
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Eric Gary Stefanich
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Vittal Shivva
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA.
| |
Collapse
|
36
|
Hoogland AI, Li X, Modi K, Welniak T, Rodriguez Y, Irizarry-Arroyo N, Oswald LB, Snider JT, Wade SW, Chavez J, Corallo S, Booth-Jones M, Jain MD, Locke FL, Jim HSL. Real-World Patient-Reported and Neurocognitive Outcomes in the Year After Axicabtagene Ciloleucel. Transplant Cell Ther 2025; 31:157.e1-157.e13. [PMID: 39733840 DOI: 10.1016/j.jtct.2024.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/13/2024] [Accepted: 12/24/2024] [Indexed: 12/31/2024]
Abstract
Axicabtagene ciloleucel (axi-cel), a chimeric antigen receptor T-cell therapy, has significantly improved clinical outcomes in adult patients with relapsed/refractory large B-cell lymphoma. However, few studies have examined patient-reported outcomes (PROs) or neurocognitive performance in patients treated with axi-cel. Moreover, no longitudinal PRO study has reported on patients treated with axi-cel as standard of care in the United States, to our knowledge. This paper reports on real-world changes in PROs (i.e., quality of life [QOL] and perceived cognition) and objective neurocognitive performance before treatment with axi-cel and in the first year after. Patients scheduled to receive axi-cel as standard of care were recruited from a single cancer center between March 2020 and June 2022. QOL was assessed using the EORTC QLQ-C30 and EQ-5D-5L at baseline recruitment (ie, prior to conditioning chemotherapy before axi-cel), and at 7, 14, 30, 60, 90, 180, and 360 days after receiving axi-cel. Perceived cognition was assessed using the Patient-Reported Outcomes Measurement Information System Cognitive Function 4a scale. Objective neurocognitive performance was assessed using a battery of tests at baseline, and 30, 90, and 360 days after receiving axi-cel. Random-effects mixed models evaluated changes in QOL, perceived cognition, and neurocognitive performance using all available data. Clinically meaningful change in QOL was defined as a difference of 10 points on the EORTC QLQ-C30. Clinically meaningful change in perceived cognition or neurocognitive performance was defined as a difference of 5 points. On average, participants (N = 53) were 63 years of age (SD = 13), and predominantly male (62%), White (92%), and college graduates (60%). Participants reported statistically significant improvements from baseline to day 360 in overall QOL, physical functioning, role functioning, and social functioning (Ps < .05) after axi-cel, despite clinically significant worsening in the first 14 days. For role functioning and social functioning, improvements also met criteria for clinical significance. There were no statistically (Ps > .05) or clinically significant changes in perceived cognition over time. Despite some transient declines, neurocognitive performance generally returned to or exceeded baseline levels by day 360 (Ps < .01). However, visuospatial ability worsened by day 90 and did not recover to baseline levels by day 360 (P < .0001). These real-world data suggest that axi-cel is associated with significant improvements in overall QOL in the first year after infusion. These data are generally consistent with, or exceed, improvements in QOL reported from clinical trials of axi-cel therapy. Despite transient worsening in the acute period after treatment, neurocognitive performance in most domains also recovered to pretreatment levels by 1 year after infusion. These findings extend previous research by reporting on patients' perspectives on axi-cel therapy received as standard of care in the real-world setting and neurocognitive changes after treatment with axi-cel.
Collapse
Affiliation(s)
- Aasha I Hoogland
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida
| | - Xiaoyin Li
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida
| | - Karnav Modi
- Department of Blood and Marrow Transplantation and Cellular Therapy, Moffitt Cancer Center, Tampa, Florida
| | - Taylor Welniak
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida
| | - Yvelise Rodriguez
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida
| | | | - Laura B Oswald
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida
| | | | - Sally W Wade
- Wade Outcomes Research and Consulting, Salt Lake City, Utah
| | - Julio Chavez
- Malignant Hematology Program, Moffitt Cancer Center, Tampa, Florida
| | | | | | - Michael D Jain
- Department of Blood and Marrow Transplantation and Cellular Therapy, Moffitt Cancer Center, Tampa, Florida
| | - Frederick L Locke
- Department of Blood and Marrow Transplantation and Cellular Therapy, Moffitt Cancer Center, Tampa, Florida
| | - Heather S L Jim
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, Florida.
| |
Collapse
|
37
|
Maganzini N, Reschke A, Cartwright AP, Gidi Y, Thompson IAP, Yee S, Hariri A, Dory C, Rosenberg-Hasson Y, Pan J, Eisenstein M, Vučković J, Cornell TT, Soh HT. Rapid, Sensitive Detection of Protein Biomarkers in Minimally-Processed Blood Products with a Monolithic Sandwich Immunoassay Reagent. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2412613. [PMID: 39910829 DOI: 10.1002/adma.202412613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/20/2024] [Indexed: 02/07/2025]
Abstract
For more than fifty years, the enzyme-linked immunosorbent assay (ELISA) serves as the gold standard for protein biomarker detection. However, conventional ELISA requires considerable sample preparation including reagent addition, incubation, and washing steps, limiting its usefulness at the point-of-care. In this work, the "instant ELISA" (fluorophore-linked immunosorbent assay) biosensor that can measure protein biomarkers in the picomolar range within 15 min in undiluted plasma or serum with no sample preparation is described. The sensor leverages a synthetic reagent termed the "monolithic dual-antibody clamp" (MDAC) which preserves the specificity, sensitivity, and generalizability of an ELISA, but produces a fluorescence signal as two surface-tethered antibodies form a "sandwich" by binding to two distinct epitopes on the target. As exemplars, picomolar quantification of tumor necrosis factor alpha (TNFα) and monocyte chemotactic protein (MCP)-1, the latter of which is a useful prognostic indicator of cytokine release syndrome in patient plasma samples during chimeric antigen receptor T cell therapy are demonstrated.
Collapse
Affiliation(s)
- Nicolò Maganzini
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Agnes Reschke
- Department of Pediatric Critical Care, Stanford University, Stanford, CA, 94305, USA
| | - Alyssa P Cartwright
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Yasser Gidi
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | | | - Steven Yee
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Amani Hariri
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Constantin Dory
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | | | - Jing Pan
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Michael Eisenstein
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Jelena Vučković
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | | | - Hyongsok Tom Soh
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
38
|
Elmeliegy M, Viqueira A, Vandendries E, Hickman A, Conte U, Irby D, Hibma J, Lon HK, Piscitelli J, Soltantabar P, Skoura A, Jiang S, Wang D. Dose Optimization of Elranatamab to Mitigate the Risk of Cytokine Release Syndrome in Patients with Multiple Myeloma. Target Oncol 2025; 20:349-359. [PMID: 40000533 PMCID: PMC11933221 DOI: 10.1007/s11523-025-01134-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND Elranatamab is a BCMA-CD3 bispecific antibody approved for the treatment of relapsed or refractory multiple myeloma. Cytokine release syndrome is one of the most common adverse events associated with bispecific antibodies. OBJECTIVE We aimed to determine the optimal elranatamab dosing regimen for mitigating cytokine release syndrome. PATIENTS AND METHODS Safety, pharmacokinetics, and exposure-response were analyzed across four clinical studies (MagnetisMM-1, MagnetisMM-2, MagnetisMM-3, and MagnetisMM-9). Different priming regimens evaluated across these studies included a one-step-up dose priming regimen of 44 mg with or without premedication, a two-step-up dose priming regimen of 12 mg on day 1 and 32 mg on day 4 with premedication, and a two-step-up dose priming regimen of 4 mg on day 1 and 20 mg on day 4 with premedication. RESULTS The maximum elranatamab serum concentration on day 1 was positively associated with any-grade and grade ≥ 2 cytokine release syndrome. A slower time to maximum serum concentration and a lower dose-normalized maximum serum concentration were observed with subcutaneous versus intravenous administration, supporting subcutaneous dosing to help mitigate cytokine release syndrome. CONCLUSIONS Based on the incidence, severity, and predictable profile of cytokine release syndrome, the 12/32-mg priming-dose regimen with premedication was determined to be the optimal regimen before the first full dose of 76 mg on day 8. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov identifiers: NCT03269136, NCT04798586, NCT04649359, and NCT05014412.
Collapse
Affiliation(s)
- Mohamed Elmeliegy
- Oncology Research and Development, Pfizer Inc., San Diego, CA, 92121, USA.
| | - Andrea Viqueira
- Oncology Research and Development, Pfizer SLU, Madrid, Spain
| | - Erik Vandendries
- Oncology Research and Development, Pfizer Inc., Cambridge, MA, USA
| | - Anne Hickman
- Oncology Research and Development, Pfizer Inc., Groton, CT, USA
| | - Umberto Conte
- Oncology Research and Development, Pfizer Inc., New York, NY, USA
| | - Donald Irby
- Pfizer Research and Development, Pfizer Inc., La Jolla, CA, USA
| | - Jennifer Hibma
- Pfizer Research and Development, Pfizer Inc., La Jolla, CA, USA
| | - Hoi-Kei Lon
- Oncology Research and Development, Pfizer Inc., San Diego, CA, 92121, USA
| | - Joseph Piscitelli
- Oncology Research and Development, Pfizer Inc., San Diego, CA, 92121, USA
| | - Pooneh Soltantabar
- Oncology Research and Development, Pfizer Inc., San Diego, CA, 92121, USA
| | - Athanasia Skoura
- Oncology Research and Development, Pfizer Inc., New York, NY, USA
| | - Sibo Jiang
- Oncology Research and Development, Pfizer Inc., San Diego, CA, 92121, USA
| | - Diane Wang
- Oncology Research and Development, Pfizer Inc., San Diego, CA, 92121, USA
| |
Collapse
|
39
|
Hensley MK, Dela Cruz CS. Host-Directed Adjunctive Therapies in Immunocompromised Patients with Pneumonia. Clin Chest Med 2025; 46:37-48. [PMID: 39890291 DOI: 10.1016/j.ccm.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2025]
Abstract
Immunocompromised (IC) hosts represent a unique patient population at risk for not only typical pathogens, but also opportunistic microorganisms. While antimicrobials remain the main treatment, new investigations have demonstrated the importance of host-response to pathogens. In this article, we highlight previously discovered and new areas of investigation for adjunctive host-response treatments for IC host pneumonia.
Collapse
Affiliation(s)
- Matthew K Hensley
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Charles S Dela Cruz
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
40
|
Wang JM, Jiang HW, Zhang YQ, Hu Y, Mei H. Hemophagocytic lymphohistiocytosis post chimeric antigen receptor T cell therapies. Expert Rev Clin Immunol 2025; 21:277-289. [PMID: 39727348 DOI: 10.1080/1744666x.2024.2444673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
INTRODUCTION Besides cytokine release syndromes (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), immune effector cell-associated HLH-like syndrome (IEC-HS) is increasingly recognized across CAR-T recipients. This emergent and fatal syndrome is difficult to separate from other disorders during the early phase, and urgently requires more integrated diagnostic and therapeutic frameworks. AREAS COVERED Existing literature has pointed out the potential role of unbridled proliferation of cytotoxic T lymphocytes, lymphopenia of natural killing cells, and hypercytokinemia in triggering the IEC-HS. The onset time of IEC-HS usually overlaps with CRS or be delayed from CRS. Clinical features include hyperferritinemia, hepatic and renal dysfunctions, cytopenias, coagulopathy, and hemophagocytosis. Multiple diagnostic criteria are based predominantly on ferritin elevation and prerequisite CRS. Corticosteroids are the cornerstone for IEC-HS treatment, while cytokine-targeted agents and pathway inhibitors offer great promise in alleviating IEC-HS syndromes. EXPERT OPINIONS Several controversial predisposing factors of IEC-HS such as disease burden should be further investigated. Future research is anticipated to identify the real-time biomarkers, as well as develop a more sophisticated grading and management network.
Collapse
Affiliation(s)
- Jing-Ming Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Hui-Wen Jiang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Yin-Qiang Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| |
Collapse
|
41
|
Wu AS, Hophing L, Gosse P, Motamed M, Bhella SD, Stewart K, Fasano A. Parkinsonism and Bilateral Facial Palsy after Chimeric Antigen Receptor T-Cell Therapy. Mov Disord Clin Pract 2025; 12:371-374. [PMID: 39780686 PMCID: PMC11952937 DOI: 10.1002/mdc3.14335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 12/01/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Affiliation(s)
- Annie Siyu Wu
- Division of Internal Medicine, Department of MedicineUniversity of TorontoTorontoOntarioCanada
| | - Lauren Hophing
- Division of Neurology, Department of MedicineUniversity of TorontoTorontoOntarioCanada
| | - Paula Gosse
- Division of Neurology, Department of MedicineUniversity of TorontoTorontoOntarioCanada
| | - Mehras Motamed
- Division of Internal Medicine, Department of MedicineUniversity of TorontoTorontoOntarioCanada
| | - Sita Devi Bhella
- Division of Medical Oncology and Hematology, Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioCanada
| | - Keith Stewart
- Division of Medical Oncology and Hematology, Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioCanada
| | - Alfonso Fasano
- Edmond J. Safra Program in Parkinson's Disease, Morton and Gloria Shulman Movement Disorders Clinic, Dvision of NeurologyToronto Western Hospital, UHN, Krembil Brain Institute, University of TorontoTorontoOntarioCanada
- IRCCS Humanitas Research HospitalRozzanoMilanItaly
| |
Collapse
|
42
|
Consoli V, Sorrenti V, Gulisano M, Spampinato M, Vanella L. Navigating heme pathways: the breach of heme oxygenase and hemin in breast cancer. Mol Cell Biochem 2025; 480:1495-1518. [PMID: 39287890 PMCID: PMC11842487 DOI: 10.1007/s11010-024-05119-5] [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: 08/06/2024] [Accepted: 09/07/2024] [Indexed: 09/19/2024]
Abstract
Breast cancer remains a significant global health challenge, with diverse subtypes and complex molecular mechanisms underlying its development and progression. This review comprehensively examines recent advances in breast cancer research, with a focus on classification, molecular pathways, and the role of heme oxygenases (HO), heme metabolism implications, and therapeutic innovations. The classification of breast cancer subtypes based on molecular profiling has significantly improved diagnosis and treatment strategies, allowing for tailored approaches to patient care. Molecular studies have elucidated key signaling pathways and biomarkers implicated in breast cancer pathogenesis, shedding light on potential targets for therapeutic intervention. Notably, emerging evidence suggests a critical role for heme oxygenases, particularly HO-1, in breast cancer progression and therapeutic resistance, highlighting the importance of understanding heme metabolism in cancer biology. Furthermore, this review highlights recent advances in breast cancer therapy, including targeted therapies, immunotherapy, and novel drug delivery systems. Understanding the complex interplay between breast cancer subtypes, molecular pathways, and innovative therapeutic approaches is essential for improving patient outcomes and developing more effective treatment strategies in the fight against breast cancer.
Collapse
Affiliation(s)
- Valeria Consoli
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
- CERNUT - Research Centre on Nutraceuticals and Health Products, University of Catania, 95125, Catania, Italy
| | - Valeria Sorrenti
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
- CERNUT - Research Centre on Nutraceuticals and Health Products, University of Catania, 95125, Catania, Italy
| | - Maria Gulisano
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
| | - Mariarita Spampinato
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
| | - Luca Vanella
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy.
- CERNUT - Research Centre on Nutraceuticals and Health Products, University of Catania, 95125, Catania, Italy.
| |
Collapse
|
43
|
Du M, Mayombo RTM, Liu J, Zhang Y, Liao D, Hu Y, Mei H. The impact of obesity and its related underlying diseases on cytokine release syndrome and the efficacy of CAR-T therapy in treating B-cell malignancies. Ann Hematol 2025; 104:1887-1895. [PMID: 40195173 DOI: 10.1007/s00277-025-06338-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 03/24/2025] [Indexed: 04/09/2025]
Abstract
Chimeric Antigen Receptor T-cell (CAR-T) therapy has revolutionized treatment for relapsed/refractory B-cell malignancies, including B-cell Acute Lymphoblastic Leukemia (B-ALL) and Diffuse Large B-Cell Lymphoma (DLBCL). However, the influence of obesity and related comorbidities on treatment outcomes and toxicity profiles remains unclear. This retrospective study included 115 patients treated with CAR-T therapy at Union Hospital, Tongji Medical College, Huazhong University of Science and Technology from 2017 to October 2023. Patients were stratified into high-risk and low-risk groups based on Body Mass Index (BMI) and the presence of obesity-related comorbidities. Clinical outcomes, including Cytokine Release Syndrome (CRS) and Immune effector Cell-Associated Neurotoxicity Syndrome (ICANS) severity, treatment efficacy, Overall Survival (OS), and Progression-Free Survival (PFS), were analyzed. Logistic regression models assessed the relationships between covariates and clinical outcomes. The median BMI was 21.91 (IQR 19.265-24.365). Among the patients, 32 were overweight, and only one had a BMI over 30. Severe CRS occurred in 16 patients, with a higher proportion in those with obesity or related conditions (10.4% vs. 3.5%, p = 0.01). Hyperlipidemia significantly increased the risk of severe CRS (OR = 3.730, CI [1.204-11.556], p = 0.022). However, being overweight, having diabetes, hypertension, coronary heart disease, or fatty liver were not significantly associated with severe CRS. Elevated total cholesterol was moderately correlated with increased Interleukin 6 (IL-6) levels (R = 0.637, p < 0.001) and weakly with Interferon gamma (IFN-γ) (R = 0.337, p < 0.001). Besides, overweight patients had a lower proportion of CAR-T cells post-infusion (OR = 0.98, CI [0.961-1.0], p = 0.048). Obesity and related comorbidities did not significantly impact treatment efficacy. However, hyperlipidemia was associated with an increased risk of severe CRS, emphasizing the need for tailored risk management strategies in CAR-T therapy. Clinical trial: NCT02965092/ NCT03366350/ NCT04008251(ClinicalTrials.gov).
Collapse
Affiliation(s)
- Mengyi Du
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Rosanna Tryphene Massounga Mayombo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Jiachen Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Yinqiang Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Danying Liao
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, 430022, China.
| |
Collapse
|
44
|
M CB, S BP, A C, A AS, A V, S R, I J. Role of procalcitonin, C-reactive protein and ferritin in cytokine release syndrome after CAR T-cell therapy in children and young adults. Biomarkers 2025; 30:115-122. [PMID: 39817702 DOI: 10.1080/1354750x.2025.2454471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/12/2025] [Indexed: 01/18/2025]
Abstract
PURPOSE Chimeric antigen receptor (CAR) T-cell CD19 therapy has changed the treatment paradigm for patients with relapsed/refractory B-cell acute lymphoblastic leukemia. It is frequently associated with potentially severe toxicities: cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), and admission to PICU is often required. Some biomarkers seem to correlate with CRS severity. Our goal is to elucidate the role of procalcitonin (PCT), C-reactive protein (CRP) and ferritin in the context of CRS following CAR T-cell infusion to predict its severity and PICU admission. METHODS Prospective observational study (2016-2022) in children and young adult who received CAR T-cell therapy (Tisagenlecleucel/ARI-0001). We collected epidemiologic data, specific CAR T-cell toxicities, PICU admission, biomarker results (PCT, CRP and ferritin), length of stay and mortality. Biomarkers were analyzed considering two values: the highest value during ward admission, and the highest overall value including PICU admission. RESULTS Seventy-seven patients were included. Median age at infusion was 9.1 years (IQR 6-13), 49.4% were females. Before CAR T-cell infusion, the median bone marrow blast was 9% (IQR 0-59). The most frequent toxicity was CRS in 62 patients (80.5%), it was severe in 18 cases (23.4%). Fourteen patients (18.1%) had ICANS. Thirty-one patients (40.3%) required admission to the PICU. PCT and ferritin were higher in patients admitted to PICU (PCT 0.8 ng/mL vs 0.15 ng/mL, p < 0.001, ferritin 5490 vs. 2900 µg/L, p < 0.019). The proposed cut-off for PCT to predict admission to PICU is 0.55 ng/mL, presenting a sensitivity of 67.7% and a specificity of 86.7%. The maximum value of three biomarkers was higher in those who presented any primary outcome: development of severe CRS, the need for admission to PICU, and in-hospital mortality. Biomarkers were higher in those who needed inotropic or respiratory support. CONCLUSIONS PCT levels increase after CAR-T cell therapy in the setting of systemic inflammation and could be a predictor of PICU admission and evolution to death. Further research studying its role in the context of CRS and the differential diagnosis between infection and CRS is needed to better understand the biology of this biomarker and to define its value in clinical practice.
Collapse
Affiliation(s)
- Caballero-Bellón M
- Oncology Department, Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu-University of Barcelona, Barcelona, Spain
| | - Bobillo-Perez S
- Pediatric Intensive Care Unit, Hospital Sant Joan de Déu-University of Barcelona, Barcelona, Spain
- Immunological and Respiratory Disorders in the Paediatric Critical Patient Research Group, Institut de Recerca Sant Joan de Déu-University of Barcelona, Barcelona, Spain
| | - Català A
- Leukemia and Lymphoma Department. CAR T-cell Unit, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Leukemia and Pediatric Hematologic Disorders. Pediatric Cancer Research Group, Institut de Recerca Sant Joan de Déu, Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Alonso-Saladrigues A
- Leukemia and Lymphoma Department. CAR T-cell Unit, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Valls A
- Laboratory Department, Hospital Sant Joan de Déu-University of Barcelona, Barcelona, Spain
| | - Rives S
- Leukemia and Lymphoma Department. CAR T-cell Unit, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Leukemia and Pediatric Hematologic Disorders. Pediatric Cancer Research Group, Institut de Recerca Sant Joan de Déu, Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Jordan I
- Pediatric Intensive Care Unit, Hospital Sant Joan de Déu-University of Barcelona, Barcelona, Spain
- Paediatric Infectious Diseases Research Group, Institut de Recerca Sant Joan de Déu, Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| |
Collapse
|
45
|
Kim WS, Kim TM, Cho SG, Jarque I, Iskierka-Jażdżewska E, Poon LM, Prince HM, Zhang H, Cao J, Zhang M, Tessoulin B, Oh SY, Lim F, Carpio C, Tan TD, Ayyappan S, Gutierrez A, Cai J, Ufkin M, Shariff S, Brouwer-Visser J, Chaudhry A, Mohamed H, Ambati S, Walewski J. Odronextamab monotherapy in patients with relapsed/refractory diffuse large B cell lymphoma: primary efficacy and safety analysis in phase 2 ELM-2 trial. NATURE CANCER 2025; 6:528-539. [PMID: 40097657 PMCID: PMC12003196 DOI: 10.1038/s43018-025-00921-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 01/23/2025] [Indexed: 03/19/2025]
Abstract
The phase 2, multicohort, ongoing ELM-2 study evaluates odronextamab, a CD20×CD3 bispecific antibody, in patients with relapsed/refractory (R/R) B cell non-Hodgkin lymphoma after ≥2 lines of therapy. Here primary analysis of the diffuse large B cell lymphoma (DLBCL) cohort is reported. Patients received intravenous odronextamab in 21-day cycles until progression or unacceptable toxicity, with cycle 1 step-up dosing to mitigate cytokine release syndrome (CRS) risk. The primary endpoint was objective response rate (ORR). Secondary endpoints included complete response (CR) rate, duration of response, progression-free survival (PFS) and overall survival. A total of 127 patients were enrolled. At the 29.9-month efficacy follow-up, the ORR was 52.0% and CR rate was 31.5%. Median durations of response and CR were 10.2 and 17.9 months, respectively. Undetectable minimal residual disease at cycle 4 day 15 was associated with PFS benefit. With a step-up of 0.7 to 4 to 20 mg (n = 60), CRS was the most common treatment-emergent adverse event (53.3% (grade ≥3, 1.7%)). No immune effector cell-associated neurotoxicity syndrome was reported. Infections were reported in 82/127 (64.6%) patients (grade ≥3, 38.6%; coronavirus disease 2019, 18.1% (grade ≥3, 12.6%)). In conclusion, odronextamab showed encouraging efficacy in heavily pretreated R/R DLBCL and generally manageable safety with supportive care. Clinical trial registration: NCT03888105 .
Collapse
Affiliation(s)
- Won Seog Kim
- Sungkyunkwan University School of Medicine, Samsung Medical Center Division of Hematology-Oncology, Seoul, South Korea.
| | - Tae Min Kim
- Seoul National University Hospital, Seoul National University Cancer Research Institute, Seoul, South Korea
| | - Seok-Goo Cho
- Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Isidro Jarque
- Hematology Department, Hospital Universitari i Politècnic La Fe, Centro de Investigación Biomédica en Red de Cáncer-CIBERONC, Valencia, Spain
| | | | - Li Mei Poon
- Department of Haematology-Oncology, National University Cancer Institute, National University Hospital, Singapore, Singapore
| | - H Miles Prince
- Epworth HealthCare and University of Melbourne, Melbourne, Victoria, Australia
| | - Huilai Zhang
- Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Junning Cao
- Fudan University Shanghai Cancer Center, Shanghai, China
| | - Mingzhi Zhang
- Department of Oncology, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Benoît Tessoulin
- Hematology Department, Nantes University Hospital, Nantes, France
| | | | | | - Cecilia Carpio
- Department of Hematology, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, Autonomous University of Barcelona (UAB), Barcelona, Spain
| | - Tran-Der Tan
- Hematology and Medical Oncology, Koo Foundation Sun Yat Sen Cancer Center, Taipei City, Taiwan
| | | | - Antonio Gutierrez
- Department of Hematology, Hospital Universitario Son Espases, IdISBa, Palma, Spain
| | - Jingxian Cai
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | | | | | | | | | | | - Jan Walewski
- Narodowy Instytut Onkologii im. Marii Skłodowskiej-Curie Państwowy Instytut Badawczy, Warszawa, Poland
| |
Collapse
|
46
|
Paul S, Jabbour E, Nichols ED, Short NJ, Kantarjian H. Blinatumomab for the treatment of acute lymphoblastic leukemia in a real-world setting: clinical vignettes. Leuk Lymphoma 2025; 66:389-399. [PMID: 39611241 DOI: 10.1080/10428194.2024.2426052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/07/2024] [Accepted: 10/28/2024] [Indexed: 11/30/2024]
Abstract
Blinatumomab, a CD19/CD3 bispecific T-cell engager; inotuzumab ozogamicin (INO), a CD22 antibody drug conjugate; and chimeric-antigen receptor (CAR) T-cell constructs are novel immune-therapeutic options for treatment of acute lymphoblastic leukemia (ALL). The use of blinatumomab has recently expanded to multiple B-ALL treatment settings. Despite the efficacy of blinatumomab, its use can be challenging in the real-world because of limited experience with its administration and management of toxicities. Optimal use and sequencing of blinatumomab is critical to improve outcomes, reduce undesired toxicities, and decrease discontinuation rates related to such toxicities. Herein, we discuss strategies to address the unique adverse effects of blinatumomab and ways to optimize its administration and integration into the treatment backbone of B-ALL. We outline our approach to combining and sequencing blinatumomab with other immunotherapies, such as INO and CD19 CAR T-cells, and provide recommendations for the management of toxicities and dose-optimization of blinatumomab therapy in clinical practice.
Collapse
Affiliation(s)
- Shilpa Paul
- Division of Pharmacy, M.D. Anderson Cancer Center, The University of Texas, Houston, TX, USA
| | - Elias Jabbour
- Department of Leukemia, M.D. Anderson Cancer Center, The University of Texas, Houston, TX, USA
| | - E Dan Nichols
- Division of Pharmacy, M.D. Anderson Cancer Center, The University of Texas, Houston, TX, USA
| | - Nicholas J Short
- Department of Leukemia, M.D. Anderson Cancer Center, The University of Texas, Houston, TX, USA
| | - Hagop Kantarjian
- Department of Leukemia, M.D. Anderson Cancer Center, The University of Texas, Houston, TX, USA
| |
Collapse
|
47
|
Sassine J, Agudelo Higuita NI, Siegrist EA, Saeedi A, Corbisiero MF, Connelly P, Bastias AG, Dib RW, Henao-Cordero J, Chastain DB, Chiu CY, Henao-Martínez AF. Timeline and outcomes of viral and fungal infections after chimeric antigen receptor T-cell therapy: a large database analysis. Clin Microbiol Infect 2025; 31:466-472. [PMID: 39528086 DOI: 10.1016/j.cmi.2024.11.008] [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: 08/28/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES This large database analysis aims to describe the incidence, timeline, and risk factors for viral and fungal infections after chimeric antigen receptor (CAR) T-cell therapy. METHODS We queried a global research network database, TriNetX, for patients who received CAR T-cell therapy, who were identified and followed for the development of viral and fungal infections. Baseline demographic, oncologic history, laboratory data and medication histories were collected. We evaluated risk factors for respiratory viral infections (RVIs), herpesvirus, fungal infections and mortality using Cox regression. RESULTS A total of 2256 patients who received CAR T-cell therapy were included, 1867 (82.7%) were CD19-targeted and 400 (17.7%) were B-cell maturation antigen-targeted. After CAR T-cell infusion, RVIs were the most prevalent (23.3%) with a median onset of 160 days (interquartile range [IQR]: 52-348 days), whereas herpesvirus and fungal infections were less frequent, occurring in 13.6% and 11.4% of cases with median onsets of 71 (IQR, 18-252) and 73 days (IQR, 14-236 days), respectively. On multivariable Cox regression, independent predictors of RVI included acute lymphoblastic leukaemia (hazard ratio [HR], 1.61), prior haematopoietic cell transplant (HCT; HR, 1.29), cytokine release syndrome (HR, 1.41), hemophagocytic lymphohistiocytosis (HR, 1.96) and glucocorticoids (HR, 3.37). Prior HCT (HR, 2.00), hypogammaglobulinemia (HR, 1.51), immune effector cell-associated neurotoxicity syndrome (HR, 1.52) and hemophagocytic lymphohistiocytosis (HR, 1.99) were associated with a higher risk of herpesviruses. Independent predictors of fungal infections included prior HCT (HR, 1.59), cytokine release syndrome (HR, 1.58) and hypogammaglobulinemia (HR, 1.40). Idecabtagene vicleucel was associated with a lower risk of herpesvirus and fungal infections (HR, 0.39 and 0.44, respectively). DISCUSSION In a large cohort of CAR T-cell therapy recipients, RVIs were the most common but occurred later, whereas herpesvirus and fungal infections were less frequent but occurred earlier. Prospective studies investigating prophylaxis and pre-emptive monitoring strategies are needed in this population.
Collapse
Affiliation(s)
- Joseph Sassine
- Infectious Diseases Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Nelson Iván Agudelo Higuita
- Infectious Diseases Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Instituto de Enfermedades Infecciosas y Parasitología Antonio Vidal, Tegucigalpa, Honduras
| | | | - Arman Saeedi
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Patrick Connelly
- Department of Computer Science, University of Colorado at Boulder, Boulder, CO, USA
| | - Alfonso G Bastias
- Department of Computer Science, University of Colorado at Boulder, Boulder, CO, USA
| | - Rita Wilson Dib
- Infectious Diseases Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - José Henao-Cordero
- Infectious Diseases Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Chia-Yu Chiu
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | |
Collapse
|
48
|
Dai Z, Yu N, Cao Y, Zhou J, Zhang Y, Wang N, Zhou X. A successful haploidentical transplantation without conditioning regimen for a relapsed/refractory multiple myeloma patient after chimeric antigen receptor T-cell therapy. Cytotherapy 2025; 27:295-299. [PMID: 39729053 DOI: 10.1016/j.jcyt.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 10/10/2024] [Accepted: 10/16/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND AIMS With novel therapies improving prognosis, the complications of multiple myeloma after multi-line treatment, particularly myelosuppression, have become a crucial determinant of long-term outcomes. Non-myeloablative allogeneic hematopoietic stem cell transplantation is a feasible option, but the transplant-related mortality rate remains high. Our study presents a relapsed/refractory multiple myeloma patient with a 9-year disease history. METHODS The patient underwent multiple chemotherapy treatments and achieved partial remission. The patient then received two B-cell maturation antigen-targeting chimeric antigen receptor (CAR) T-cell treatments with lymphodepletion conditioning of fludarabine and cyclophosphamide. RESULTS At the fifth month after the second CAR T-cell treatment, the patient achieved complete remission but developed refractory myelosuppression (grade 4 according to Common Terminology Criteria for Adverse Events 5.0) after several severe infections. In order to facilitate hematopoietic recovery, her daughter's stem cells were infused into her, which fortunately implanted without conditioning. After thrombotic microangiopathy and acute graft-versus-host disease, the patient was discharged with consistent full donor chimerism. We assume this engraftment may be attributed to the patient's severe hematopoietic failure and further host lymphodepletion by fludarabine. CONCLUSIONS This study highlights the potential of allogeneic hematopoietic stem cell transplantation with reduced conditioning intensity or even the omission of conditioning, particularly for a relapsed/refractory multiple myeloma patient who struggles with severe myelosuppression after long-term treatment.
Collapse
Affiliation(s)
- Zigang Dai
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Nanzhou Yu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yang Cao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Na Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| | - Xiaoxi Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
49
|
Jamroziak K, Zielonka K, Khwaja J, Wechalekar AD. Update on B-cell maturation antigen-directed therapies in AL amyloidosis. Br J Haematol 2025; 206:817-831. [PMID: 39748220 DOI: 10.1111/bjh.19960] [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: 08/19/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025]
Abstract
Systemic light chain (AL) amyloidosis is a rare clonal plasma cell disorder characterized by the production of amyloidogenic immunoglobulin light chains, which causes the formation and deposition of amyloid fibrils, leading to multi-organ dysfunction. Current treatment is directed at the underlying plasma cell clone to achieve a profound reduction in the monoclonal free light chain production. The standard-of-care first-line therapy is a combination of daratumumab, cyclophosphamide, bortezomib and dexamethasone (D-VCd regimen), resulting in high rates of haematological and organ responses. However, AL amyloidosis remains incurable, and all patients inevitably relapse. Hence, novel treatment options are needed for patients with an inadequate response or relapsed/refractory disease. B-cell maturation antigen (BCMA) is a tumour necrosis factor (TNF receptor superfamily receptor overexpressed on plasma cells in multiple myeloma (MM) and AL amyloidosis. Recently, several novel anti-BCMA immunotherapies have been approved for the treatment of relapsed/refractory MM, including antibody-drug conjugate belantamab mafodotin, bispecific antibodies teclistamab and elranatamab and chimeric antigen receptor T-cell therapies idecabtagene vicleucel and ciltacabtagene autoleucel. Despite lower expression than in MM, BCMA is also a promising target in AL amyloidosis. This review aims to provide up-to-date information on the efficacy and toxicity of anti-BCMA therapy in AL amyloidosis.
Collapse
Affiliation(s)
- Krzysztof Jamroziak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Klaudia Zielonka
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Jahanzaib Khwaja
- Department of Haematology, University College London Hospital, London, UK
| | | |
Collapse
|
50
|
Fleury I, MacDonald D, Shafey M, Christofides A, Sehn LH. Optimal Use of Bispecific Antibodies for the Treatment of Diffuse Large B-Cell Lymphoma in Canada. Curr Oncol 2025; 32:142. [PMID: 40136346 PMCID: PMC11941342 DOI: 10.3390/curroncol32030142] [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: 01/24/2025] [Revised: 02/24/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
CAR-T cell therapy has significantly improved outcomes for patients with relapsed or refractory (R/R) diffuse large B-cell lymphoma (DLBCL), but challenges such as limited resources, manufacturing timelines, and notable toxicities persist. Bispecific antibodies (BsAbs), including glofitamab and epcoritamab, have demonstrated promising efficacy and represent a new treatment option in patients who are unsuitable for or have relapsed following CAR-T therapy. Bispecific antibodies have a manageable safety profile and are generally more widely accessible than CAR-T cell therapy. Case discussions in this paper illustrate the potential real-world application of BsAbs, highlighting their role in treating patients who have relapsed after or are unable to undergo CAR-T cell therapy. Overall, glofitamab and epcoritamab represent valuable treatment options in the evolving landscape of R/R DLBCL.
Collapse
Affiliation(s)
- Isabelle Fleury
- Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - David MacDonald
- Division of Hematology, University of Ottawa, Ottawa, ON K1N 6N5, Canada;
| | - Mona Shafey
- Department of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | | | - Laurie H. Sehn
- BC Cancer Centre for Lymphoid Cancer, The University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| |
Collapse
|