201
|
Nagler A, Perriello VM, Falini L, Falini B. How I treat refractory/relapsed diffuse large B-cell lymphomas with CD19-directed chimeric antigen receptor T cells. Br J Haematol 2023; 201:396-410. [PMID: 36916189 DOI: 10.1111/bjh.18724] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/10/2023] [Accepted: 02/17/2023] [Indexed: 03/15/2023]
Abstract
Chimeric antigen receptor (CAR) T cells targeting CD19 represent a promising salvage immunotherapy for relapsed/refractory diffuse large B-cell lymphoma (R/R DLBCL), offering ~40% of long-term responses. In everyday clinical practice, haematologists involved in CAR T cell treatment of patients with R/R DLBCL have to deal with diagnostically complex cases and difficult therapeutic choices. The availability of novel immunotherapeutic agents for R/R DLBCL and recent advances in understanding CAR T-cell failure mechanisms demand a rational approach to identify the best choice for bridging therapy and managing post-CAR T-cell therapy relapses. Moreover, positron emission tomography/computerised tomography may result in false-positive interpretation, highlighting the importance of post-treatment biopsy. In this review, we discuss all above issues, presenting four instructive cases, with the aim to provide criteria and new perspectives for CAR T-cell treatment of DLBCL.
Collapse
Affiliation(s)
- Arnon Nagler
- Division of Hematology, Sheba Medical Center, Tel Hashomer, Israel
| | - Vincenzo Maria Perriello
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Lorenza Falini
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Brunangelo Falini
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, Perugia, Italy
| |
Collapse
|
202
|
Kitamura W, Asada N, Naoi Y, Abe M, Fujiwara H, Ennishi D, Nishimori H, Fujii K, Fujii N, Matsuoka KI, Yoshino T, Maeda Y. Bone marrow microenvironment disruption and sustained inflammation with prolonged haematologic toxicity after CAR T-cell therapy. Br J Haematol 2023. [PMID: 36890790 DOI: 10.1111/bjh.18747] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/10/2023]
Abstract
Mechanisms of prolonged cytopenia (PC) after chimeric antigen receptor (CAR) T-cell therapy, an emerging therapy for relapsed or refractory diffuse large B-cell lymphoma, remain elusive. Haematopoiesis is tightly regulated by the bone marrow (BM) microenvironment, called the 'niche'. To investigate whether alterations in the BM niche cells are associated with PC, we analysed CD271+ stromal cells in BM biopsy specimens and the cytokine profiles of the BM and serum obtained before and on day 28 after CAR T-cell infusion. Imaging analyses of the BM biopsy specimens revealed that CD271+ niche cells were severely impaired after CAR T-cell infusion in patients with PC. Cytokine analyses after CAR T-cell infusion showed that CXC chemokine ligand 12 and stem cell factor, niche factors essential for haematopoietic recovery, were significantly decreased in the BM of patients with PC, suggesting reduced niche cell function. The levels of inflammation-related cytokines on day 28 after CAR T-cell infusion were consistently high in the BM of patients with PC. Thus, we demonstrate for the first time that BM niche disruption and sustained elevation of inflammation-related cytokines in the BM following CAR T-cell infusion are associated with subsequent PC.
Collapse
Affiliation(s)
- Wataru Kitamura
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
- Department of Hematology, Oncology and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Noboru Asada
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - Yusuke Naoi
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Masaya Abe
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
- Department of Hematology, Oncology and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hideaki Fujiwara
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - Daisuke Ennishi
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan
| | - Hisakazu Nishimori
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - Keiko Fujii
- Division of Clinical Laboratory, Okayama University Hospital, Okayama, Japan
| | - Nobuharu Fujii
- Division of Blood Transfusion, Okayama University Hospital, Okayama, Japan
| | - Ken-Ichi Matsuoka
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - Tadashi Yoshino
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Yoshinobu Maeda
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
- Department of Hematology, Oncology and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
203
|
Benoit A, B Boies MH, Déry N, M Garcia L, Simard M, Poirier M, Delage R, Lortal Canguilhem B, Doyle C, Larouche JF, Couture F, Lemieux C. CAR T-Cells for the Treatment of Refractory or Relapsed Large B-Cell Lymphoma: A Single-Center Retrospective Canadian Study. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:203-210. [PMID: 36646606 DOI: 10.1016/j.clml.2022.12.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/18/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T-cells are an important new third-line treatment option for large B-cell lymphoma (LBCL). The objective response rates in pivotal early phase clinical trials with CAR T-cells were very promising. The objective of this study was to describe the efficacy results obtained with CAR T-cells infusions in our institution and to compare the toxicities of our cohort with those of pivotal trials and studies conducted in a real-life setting. PATIENTS AND METHODS Efficacy and safety data were retrospectively collected from 25 patients with LBCL treated with CAR T-cells therapy at CHU de Québec-Université Laval. A literature search was then performed to identify other efficacy or safety data from a real-life setting. RESULTS At 3 months post infusion, the objective response rate (ORR) in our population with tisagenlecleucel and axicabtagene-ciloleucel were 20% and 47%, respectively. Bulky disease was the only negative predictor of poor response at 3 months (0% vs. 53%, P = .03). Bulky disease was associated with a median PFS of 2 months compared to 5 months for non-bulky disease (P = .0009). Grade ≥ 3 hematological toxicities were greater in patients treated with axi-cel (60% vs. 20%, P = .048), without bone marrow involvement (55% vs. 0%, P =.046), without stage IV disease (72% vs. 21%, P =.02), with refractory disease (67% vs. 10%, P =.01) or having been affected by cytokine release syndrome (58% vs. 0%, P =.02). CONCLUSION The poor response rate at 3 months after infusion in our cohort was influenced mainly by bulky disease. Further studies are needed to better characterize the loss of efficacy of CAR T-cells because the majority of patients will relapse over time.
Collapse
Affiliation(s)
- Aurélie Benoit
- Department of pharmacy, CHU de Québec - Université Laval, Québec, Québec, Canada; Faculté de pharmacie, Université de Bordeaux, Bordeaux, Nouvelle Aquitaine, France; Unité pour l'usage optimal du médicament et la recherche (UGMR), CHU de Québec - Université Laval, Québec, Québec, Canada
| | - Marie-Hélène B Boies
- Department of pharmacy, CHU de Québec - Université Laval, Québec, Québec, Canada; Unité pour l'usage optimal du médicament et la recherche (UGMR), CHU de Québec - Université Laval, Québec, Québec, Canada
| | - Nicole Déry
- Department of pharmacy, CHU de Québec - Université Laval, Québec, Québec, Canada; Unité pour l'usage optimal du médicament et la recherche (UGMR), CHU de Québec - Université Laval, Québec, Québec, Canada
| | - Luciana M Garcia
- Department of medicine, CHU de Québec - Université Laval, Québec, Québec, Canada
| | - Mélanie Simard
- Department of pharmacy, CHU de Québec - Université Laval, Québec, Québec, Canada
| | - Mireille Poirier
- Department of pharmacy, CHU de Québec - Université Laval, Québec, Québec, Canada
| | - Robert Delage
- Department of medicine, CHU de Québec - Université Laval, Québec, Québec, Canada
| | | | - Catherine Doyle
- Department of medicine, CHU de Québec - Université Laval, Québec, Québec, Canada
| | | | - Félix Couture
- Department of medicine, CHU de Québec - Université Laval, Québec, Québec, Canada
| | - Christopher Lemieux
- Department of medicine, CHU de Québec - Université Laval, Québec, Québec, Canada.
| |
Collapse
|
204
|
Sasu BJ, Opiteck GJ, Gopalakrishnan S, Kaimal V, Furmanak T, Huang D, Goswami A, He Y, Chen J, Nguyen A, Balakumaran A, Shah NN, Hamadani M, Bone KM, Prashad S, Bowen MA, Pertel T, Embree HD, Gidwani SG, Chang D, Moore A, Leonard M, Amado RG. Detection of chromosomal alteration after infusion of gene-edited allogeneic CAR T cells. Mol Ther 2023; 31:676-685. [PMID: 36518079 PMCID: PMC10014221 DOI: 10.1016/j.ymthe.2022.12.004] [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: 09/13/2022] [Revised: 11/18/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
A chromosome 14 inversion was found in a patient who developed bone marrow aplasia following treatment with allogeneic chimeric antigen receptor (CAR) Tcells containing gene edits made with transcription activator-like effector nucleases (TALEN). TALEN editing sites were not involved at either breakpoint. Recombination signal sequences (RSSs) were found suggesting recombination-activating gene (RAG)-mediated activity. The inversion represented a dominant clone detected in the context of decreasing absolute CAR Tcell and overall lymphocyte counts. The inversion was not associated with clinical consequences and wasnot detected in the drug product administered to this patient or in any drug product used in this or other trials using the same manufacturing processes. Neither was the inversion detected in this patient at earlier time points or in any other patient enrolled in this or other trials treated with this or other product lots. This case illustrates that spontaneous, possibly RAG-mediated, recombination events unrelated to gene editing can occur in adoptive cell therapy studies, emphasizes the need for ruling out off-target gene editing sites, and illustrates that other processes, such as spontaneous V(D)J recombination, can lead to chromosomal alterations in infused cells independent of gene editing.
Collapse
Affiliation(s)
- Barbra J Sasu
- Allogene Therapeutics, South San Francisco, CA 94080, USA
| | | | | | - Vivek Kaimal
- Allogene Therapeutics, South San Francisco, CA 94080, USA
| | - Tom Furmanak
- Allogene Therapeutics, South San Francisco, CA 94080, USA
| | - David Huang
- Allogene Therapeutics, South San Francisco, CA 94080, USA
| | | | - Ying He
- Allogene Therapeutics, South San Francisco, CA 94080, USA
| | - Jiamin Chen
- Allogene Therapeutics, South San Francisco, CA 94080, USA
| | - Anh Nguyen
- Allogene Therapeutics, South San Francisco, CA 94080, USA
| | | | - Nirav N Shah
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mehdi Hamadani
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Kathleen M Bone
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Sacha Prashad
- Allogene Therapeutics, South San Francisco, CA 94080, USA
| | | | - Thomas Pertel
- Allogene Therapeutics, South San Francisco, CA 94080, USA
| | | | | | - David Chang
- Allogene Therapeutics, South San Francisco, CA 94080, USA
| | - Alison Moore
- Allogene Therapeutics, South San Francisco, CA 94080, USA
| | - Mark Leonard
- Allogene Therapeutics, South San Francisco, CA 94080, USA
| | - Rafael G Amado
- Allogene Therapeutics, South San Francisco, CA 94080, USA.
| |
Collapse
|
205
|
Lipsitt A, Beattie L, Harstead E, Li Y, Goorha S, Maron G, Gottschalk S, Triplett B, Naik S. Allogeneic CD34 + selected hematopoietic stem cell boost following CAR T-cell therapy in a patient with prolonged cytopenia and active infection. Pediatr Blood Cancer 2023; 70:e30166. [PMID: 36565276 DOI: 10.1002/pbc.30166] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/08/2022] [Accepted: 11/29/2022] [Indexed: 12/25/2022]
Abstract
Hematological toxicity (hematotoxicity) leading to peripheral cytopenias is a common long-term adverse effect following the use of CD19-chimeric antigen receptor (CD19-CAR) T-cell therapies. However, management remains unclear for patients whose cytopenias persist beyond 1 month after CAR T-cell infusion. We present the case of a 21-year old who received CD19-CAR T-cell therapy for relapse following a haploidentical transplant. He developed hematotoxicity and consequently multiple life-threatening infections. We administered a CD34+ hematopoietic stem cell boost (HSCB) from his transplant donor, which led to hematopoietic recovery and resolution of his infections without any effect on the activity of CD19-CAR T cells. CD34+ HSCB can be a safe and effective option to treat hematotoxicity following CD19-CAR T-cell therapy.
Collapse
Affiliation(s)
- Amanda Lipsitt
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Lisa Beattie
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Elaine Harstead
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Ying Li
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | - Gabriela Maron
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Brandon Triplett
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Swati Naik
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
206
|
Goldman A, Raschi E, Chapman J, Santomasso BD, Pasquini MC, Perales MA, Shouval R. Progressive multifocal leukoencephalopathy in patients treated with chimeric antigen receptor T cells. Blood 2023; 141:673-677. [PMID: 36332168 PMCID: PMC9979708 DOI: 10.1182/blood.2022017386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
Using 2 global postmarketing surveillance databases, Goldman and colleagues report that progressive multifocal leukoencephalopathy (PML), a viral disease associated with profound immunosuppression, occurs in approximately 0.9 cases per 1000 recipients of CD19-directed CAR T-cell therapy. The risk of PML appears higher with CAR T-cell therapy than other cancer therapies, but its precise role cannot be distinguished from antecedent therapies that these patients receive.
Collapse
Affiliation(s)
- Adam Goldman
- Department of Internal Medicine, The Talpiot Sheba Medical Leadership Program, Chaim Sheba Medical Center, Ramat-Gan, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Emanuel Raschi
- Pharmacology Unit, Department of Medical and Surgical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Joab Chapman
- Department of Neurology, The Chaim Sheba Medical Center, Ramat-Gan, Israel
| | - Bianca D. Santomasso
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY
- Adult Bone Marrow Transplantation Service and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marcelo C. Pasquini
- Department of Medicine, Center for International Blood and Bone Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Roni Shouval
- Adult Bone Marrow Transplantation Service and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| |
Collapse
|
207
|
Reinhardt B, Lee P, Sasine JP. Chimeric Antigen Receptor T-Cell Therapy and Hematopoiesis. Cells 2023; 12:531. [PMID: 36831198 PMCID: PMC9954220 DOI: 10.3390/cells12040531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Chimeric Antigen Receptor (CAR) T-cell therapy is a promising treatment option for patients suffering from B-cell- and plasma cell-derived hematologic malignancies and is being adapted for the treatment of solid cancers. However, CAR T is associated with frequently severe toxicities such as cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), macrophage activation syndrome (MAS), and prolonged cytopenias-a reduction in the number of mature blood cells of one or more lineage. Although we understand some drivers of these toxicities, their mechanisms remain under investigation. Since the CAR T regimen is a complex, multi-step process with frequent adverse events, ways to improve the benefit-to-risk ratio are needed. In this review, we discuss a variety of potential solutions being investigated to address the limitations of CAR T. First, we discuss the incidence and characteristics of CAR T-related cytopenias and their association with reduced CAR T-cell efficacy. We review approaches to managing or mitigating cytopenias during the CAR T regimen-including the use of growth factors, allogeneic rescue, autologous hematopoietic stem cell infusion, and alternative conditioning regimens. Finally, we introduce novel methods to improve CAR T-cell-infusion products and the implications of CAR T and clonal hematopoiesis.
Collapse
Affiliation(s)
- Bryanna Reinhardt
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Patrick Lee
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Joshua P. Sasine
- Department of Medicine, Division of Hematology and Cellular Therapy, Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
208
|
Cytokine profiles are associated with prolonged hematologic toxicities after B-cell maturation antigen targeted chimeric antigen receptor-T-cell therapy. Cytotherapy 2023; 25:192-201. [PMID: 36496302 DOI: 10.1016/j.jcyt.2022.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 10/20/2022] [Accepted: 11/03/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND AIMS The considerable efficacy of B-cell maturation antigen-targeted chimeric antigen receptor (CAR)-T-cell therapy has been extensively demonstrated in the treatment of relapsed or refractory multiple myeloma. Nevertheless, in clinical practice, prolonged hematologic toxicity (PHT) extends hospital stay and impairs long-term survival. METHODS This retrospective study reviewed 99 patients with relapsed or refractory multiple myeloma who underwent B-cell maturation antigen CAR-T-cell therapy at our institution between April 2018 and September 2021 (ChiCTR1800017404). RESULTS Among 93 evaluable patients, the incidence of prolonged hematologic toxicities was high after CAR-T-cell infusion, including 38.71% (36/93) of patients with prolonged neutropenia, 22.58% (21/93) with prolonged anemia and 59.14% (55/93) with prolonged thrombocytopenia. In addition, 9.68% (9/93) of patients experienced prolonged pancytopenia. Our multivariate analyses identified that cytokine profiles were independent risk factors for PHTs, whereas a sufficient baseline hematopoietic function and high CD4/CD8 ratio of CAR-T cells were protective factors for PHTs after CAR-T-cell infusion. Subgroup analyses found that the kinetics of post-CAR-T hematologic parameters were primarily determined by the collective effects of cytokine release syndrome and baseline hematopoietic functions, and showed influential weights for the three lineages. CONCLUSIONS Our findings improve the understanding of the impact of cytokines on hematopoietic functions, which could contribute to the mechanism investigation and exploration of potential intervention strategies.
Collapse
|
209
|
Accorsi Buttini E, Farina M, Lorenzi L, Polverelli N, Radici V, Morello E, Colnaghi F, Almici C, Ferrari E, Bianchetti A, Leoni A, Re F, Bosio K, Bernardi S, Malagola M, Re A, Russo D. High risk-myelodysplastic syndrome following CAR T-cell therapy in a patient with relapsed diffuse large B cell lymphoma: A case report and literature review. Front Oncol 2023; 13:1036455. [PMID: 36741006 PMCID: PMC9897055 DOI: 10.3389/fonc.2023.1036455] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 01/02/2023] [Indexed: 01/22/2023] Open
Abstract
Background Chimeric antigen receptor (CAR) T-cell therapy represents the most advanced immunotherapy against relapsed/refractory B cell malignancies. While cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome are distinctive, known CAR T-cell acute adverse events, hematological toxicity has been increasingly reported. Cytopenia following CAR T-cell treatment is attributed in most cases to lymphodepletion regimens, bridging chemotherapy, or radiotherapy. However, when cytopenia becomes prolonged, the development of myelodysplastic syndrome (MDS) should be considered. Case presentation We report a case of high risk (HR)-MDS following CAR T-cell therapy in a patient with relapsed diffuse large B cell lymphoma. Eight months after CAR T-cell infusion, the blood count showed progressive, worsening cytopenia and the bone marrow biopsy revealed multilineage dysplasia without excess of blasts associated with chromosome 7 deletion and RUNX1 mutation. Next generation sequencing analysis, retrospectively performed on stored samples, showed a germ line CSF3R mutation, CEBPA clonal hematopoiesis, but no RUNX1 lesion. Conclusion We describe a case of HR-MDS, with deletion of chromosome 7 and acquisition of RUNX1 mutation, developing after CAR T-cell therapy in a patient with clonal hematopoiesis (CH). Previous chemotherapy favored MDS onset; however, we could not exclude the fact that the impairment of immunosurveillance related to either lymphodepletion or CAR T-cell infusion may play a role in MDS development. Thus, we designed a multicenter prospective study (ClonHema-CAR-T-Study) to investigate if cytopenia after CAR T-cell treatment may be due to underling CH as well as the presence of secondary myeloid malignancies.
Collapse
Affiliation(s)
- Eugenia Accorsi Buttini
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy,*Correspondence: Eugenia Accorsi Buttini,
| | - Mirko Farina
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Luisa Lorenzi
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Nicola Polverelli
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Vera Radici
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Enrico Morello
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Federica Colnaghi
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Camillo Almici
- Stem Cell Laboratory, Section of Hematology and Blood Coagulation, Clinical Chemistry Laboratory, Diagnostics Department, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Emilio Ferrari
- Stem Cell Laboratory, Section of Hematology and Blood Coagulation, Clinical Chemistry Laboratory, Diagnostics Department, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Andrea Bianchetti
- Stem Cell Laboratory, Section of Hematology and Blood Coagulation, Clinical Chemistry Laboratory, Diagnostics Department, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Alessandro Leoni
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy,Research Center Ail (CREA), Chair of Hematology Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Federica Re
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy,Research Center Ail (CREA), Chair of Hematology Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Katia Bosio
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy,Research Center Ail (CREA), Chair of Hematology Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Simona Bernardi
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy,Research Center Ail (CREA), Chair of Hematology Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Michele Malagola
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Alessandro Re
- Hematology Unit, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Domenico Russo
- Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, Department of Clinical and Experimental Science, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| |
Collapse
|
210
|
De Matteis S, Dicataldo M, Casadei B, Storci G, Laprovitera N, Arpinati M, Maffini E, Cortelli P, Guarino M, Vaglio F, Naddeo M, Sinigaglia B, Zazzeroni L, Guadagnuolo S, Tomassini E, Bertuccio SN, Messelodi D, Ferracin M, Bonafè M, Zinzani PL, Bonifazi F. Peripheral blood cellular profile at pre-lymphodepletion is associated with CD19-targeted CAR-T cell-associated neurotoxicity. Front Immunol 2023; 13:1058126. [PMID: 36726971 PMCID: PMC9886226 DOI: 10.3389/fimmu.2022.1058126] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/21/2022] [Indexed: 01/18/2023] Open
Abstract
Background Infusion of second generation autologous CD19-targeted chimeric antigen receptor (CAR) T cells in patients with R/R relapsed/refractory B-cell lymphoma (BCL) is affected by inflammatory complications, such as Immune Effector Cell-Associated Neurotoxicity Syndrome (ICANS). Current literature suggests that the immune profile prior to CAR-T infusion modifies the chance to develop ICANS. Methods This is a monocenter prospective study on 53 patients receiving approved CAR T-cell products (29 axi-cel, 24 tisa-cel) for R/R-BCL. Clinical, biochemical, and hematological variables were analyzed at the time of pre-lymphodepletion (pre-LD). In a subset of 21 patients whose fresh peripheral blood sample was available, we performed cytofluorimetric analysis of leukocytes and extracellular vesicles (EVs). Moreover, we assessed a panel of soluble plasma biomarkers (IL-6/IL-10/GDF-15/IL-15/CXCL9/NfL) and microRNAs (miR-146a-5p, miR-21-5p, miR-126-3p, miR-150-5p) which are associated with senescence and inflammation. Results Multivariate analysis at the pre-LD time-point in the entire cohort (n=53) showed that a lower percentage of CD3+CD8+ lymphocytes (38.6% vs 46.8%, OR=0.937 [95% CI: 0.882-0.996], p=0.035) and higher levels of serum C-reactive protein (CRP, 4.52 mg/dl vs 1.00 mg/dl, OR=7.133 [95% CI: 1.796-28], p=0.005) are associated with ICANS. In the pre-LD samples of 21 patients, a significant increase in the percentage of CD8+CD45RA+CD57+ senescent cells (median % value: 16.50% vs 9.10%, p=0.009) and monocytic-myeloid derived suppressor cells (M-MDSC, median % value: 4.4 vs 1.8, p=0.020) was found in ICANS patients. These latter also showed increased levels of EVs carrying CD14+ and CD45+ myeloid markers, of the myeloid chemokine CXCL-9, as well of the MDSC-secreted cytokine IL-10. Notably, the serum levels of circulating neurofilament light chain, a marker of neuroaxonal injury, were positively correlated with the levels of senescent CD8+ T cells, M-MDSC, IL-10 and CXCL-9. No variation in the levels of the selected miRNAs was observed between ICANS and no-ICANS patients. Discussion Our data support the notion that pre-CAR-T systemic inflammation is associated with ICANS. Higher proportion of senescence CD8+ T cells and M-MDSC correlate with early signs of neuroaxonal injury at pre-LD time-point, suggesting that ICANS may be the final event of a process that begins before CAR-T infusion, consequence to patient clinical history.
Collapse
Affiliation(s)
| | - Michele Dicataldo
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy,Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Beatrice Casadei
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy,Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Gianluca Storci
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | - Mario Arpinati
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Enrico Maffini
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Pietro Cortelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy,IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Maria Guarino
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Francesca Vaglio
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Maria Naddeo
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy,Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Barbara Sinigaglia
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Luca Zazzeroni
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | | | - Enrica Tomassini
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | | | - Daria Messelodi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Manuela Ferracin
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Massimiliano Bonafè
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy,*Correspondence: Massimiliano Bonafè,
| | - Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy,Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | | |
Collapse
|
211
|
Steinhardt MJ, Reinhardt L, Luu M, Danhof S, Hudecek M. CAR-T-Zell-basierte Immuntherapien in der Hämatoonkologie. DIE ONKOLOGIE 2023. [PMCID: PMC9842198 DOI: 10.1007/s00761-022-01299-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- M. J. Steinhardt
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Deutschland
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Oberdürrbacher Str. 6, 97080 Würzburg, Deutschland
| | - L. Reinhardt
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Deutschland
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Oberdürrbacher Str. 6, 97080 Würzburg, Deutschland
| | - M. Luu
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Deutschland
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Oberdürrbacher Str. 6, 97080 Würzburg, Deutschland
| | - S. Danhof
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Deutschland
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Oberdürrbacher Str. 6, 97080 Würzburg, Deutschland
| | - M. Hudecek
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Deutschland
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Oberdürrbacher Str. 6, 97080 Würzburg, Deutschland
| |
Collapse
|
212
|
Xu T, Karschnia P, Cadilha BL, Dede S, Lorenz M, Seewaldt N, Nikolaishvili E, Müller K, Blobner J, Teske N, Herold JJ, Rejeski K, Langer S, Obeck H, Lorenzini T, Mulazzani M, Zhang W, Ishikawa-Ankerhold H, Buchholz VR, Subklewe M, Thon N, Straube A, Tonn JC, Kobold S, von Baumgarten L. In vivo dynamics and anti-tumor effects of EpCAM-directed CAR T-cells against brain metastases from lung cancer. Oncoimmunology 2023; 12:2163781. [PMID: 36687005 PMCID: PMC9851202 DOI: 10.1080/2162402x.2022.2163781] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Lung cancer patients are at risk for brain metastases and often succumb to their intracranial disease. Chimeric Antigen Receptor (CAR) T-cells emerged as a powerful cell-based immunotherapy for hematological malignancies; however, it remains unclear whether CAR T-cells represent a viable therapy for brain metastases. Here, we established a syngeneic orthotopic cerebral metastasis model in mice by combining a chronic cranial window with repetitive intracerebral two-photon laser scanning-microscopy. This approach enabled in vivo-characterization of fluorescent CAR T-cells and tumor cells on a single-cell level over weeks. Intraparenchymal injection of Lewis lung carcinoma cells (expressing the tumor cell-antigen EpCAM) was performed, and EpCAM-directed CAR T-cells were injected either intravenously or into the adjacent brain parenchyma. In mice receiving EpCAM-directed CAR T-cells intravenously, we neither observed substantial CAR T-cell accumulation within the tumor nor relevant anti-tumor effects. Local CAR T-cell injection, however, resulted in intratumoral CAR T-cell accumulation compared to controls treated with T-cells lacking a CAR. This finding was accompanied by reduced tumorous growth as determined per in vivo-microscopy and immunofluorescence of excised brains and also translated into prolonged survival. However, the intratumoral number of EpCAM-directed CAR T-cells decreased during the observation period, pointing toward insufficient persistence. No CNS-specific or systemic toxicities of EpCAM-directed CAR T-cells were observed in our fully immunocompetent model. Collectively, our findings indicate that locally (but not intravenously) injected CAR T-cells may safely induce relevant anti-tumor effects in brain metastases from lung cancer. Strategies improving the intratumoral CAR T-cell persistence may further boost the therapeutic success.
Collapse
Affiliation(s)
- Tao Xu
- Department of Neurology, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Philipp Karschnia
- Department of Neurosurgery, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany,CONTACT Philipp Karschnia
| | - Bruno Loureiro Cadilha
- Department of Medicine IV, Division of Clinical Pharmacology and Center of Integrated Protein Science Munich, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sertac Dede
- Department of Neurology, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Michael Lorenz
- Department of Medicine I, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Niklas Seewaldt
- Department of Neurosurgery, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Elene Nikolaishvili
- Department of Neurosurgery, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Katharina Müller
- Department of Neurology, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jens Blobner
- Department of Neurosurgery, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Nico Teske
- Department of Neurosurgery, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Julika J. Herold
- Department of Neurosurgery, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Kai Rejeski
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany,Department of Medicine III, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sigrid Langer
- Department of Neurology, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Hannah Obeck
- Department of Medicine IV, Division of Clinical Pharmacology and Center of Integrated Protein Science Munich, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Theo Lorenzini
- Department of Medicine IV, Division of Clinical Pharmacology and Center of Integrated Protein Science Munich, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Matthias Mulazzani
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Wenlong Zhang
- Department of Neurology, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Hellen Ishikawa-Ankerhold
- Department of Medicine I, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Veit R. Buchholz
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universitaet Muenchen (TUM), Munich, Germany
| | - Marion Subklewe
- Department of Medicine III, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Niklas Thon
- Department of Neurosurgery, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Andreas Straube
- Department of Neurology, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Joerg-Christian Tonn
- Department of Neurosurgery, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Sebastian Kobold
- Department of Medicine IV, Division of Clinical Pharmacology and Center of Integrated Protein Science Munich, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Louisa von Baumgarten
- Department of Neurology, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany,Department of Neurosurgery, University Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany,Louisa von Baumgarten Department of Neurosurgery, Division of Neuro-Oncology, University Hospital of the Ludwig-Maximilians-University Munich, Marchioninistrasse 15/81377, Munich, Germany
| |
Collapse
|
213
|
Fowler NH, Dickinson M, Ghosh M, Chen AI, Andreadis C, Tiwari R, Masood A, Ramos R, Jousseaume E, Thieblemont C, Dreyling M, Schuster SJ. Assessment of Healthcare Resource Utilization and Hospitalization Costs in Patients With Relapsed or Refractory Follicular Lymphoma Undergoing CAR-T Cell Therapy With Tisagenlecleucel: Results From the ELARA Study. Transplant Cell Ther 2023; 29:60.e1-60.e4. [PMID: 36182104 DOI: 10.1016/j.jtct.2022.09.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/12/2022] [Accepted: 09/22/2022] [Indexed: 02/09/2023]
Abstract
Follicular lymphoma (FL) is generally considered an indolent disease, although patients with relapsing FL experience progressively shorter durations of response to second or later lines of therapy. The ongoing ELARA trial in adult patients with relapsed/refractory (r/r) FL treated with tisagenlecleucel demonstrated an overall response rate of 86.2% and a complete response rate of 69.1%, with no treatment-related deaths. Tisagenlecleucel was administered in the outpatient setting in 18% of patients in ELARA; however, there is limited knowledge concerning the impact of inpatient versus outpatient tisagenlecleucel administration on healthcare resource utilization (HCRU) among patients with r/r FL. Here, we present the first HCRU analysis among patients with r/r FL who received tisagenlecleucel in the Phase II, single-arm, multicenter ELARA trial. HCRU was characterized using hospitalization data from day 1 to month 2 after tisagenlecleucel infusion. Information on length of stay, facility use, and discharge was assessed in patients who received tisagenlecleucel in the outpatient or inpatient setting. All costs were inflated to 2020 US dollars. As of August 3, 2021 (20-month median follow-up), 17/97 (18%) r/r FL patients were infused in an outpatient setting. Patients infused in the outpatient setting generally had favorable Eastern Cooperative Oncology Group performance status and Follicular Lymphoma International Prognostic Index scores, and less bulky disease at baseline. However, the outpatients had higher proportions of patients with grade 3A FL, primary refractory disease, and >5 lines of prior therapy compared with inpatients. Forty-one percent of patients treated in the outpatient setting did not require hospitalization within 30 days after infusion, and outpatients who did require hospitalization had a shorter average length of stay compared with inpatients (5 versus 13 days). No outpatients required intensive care unit (ICU) admission, whereas 9% of inpatients were admitted to the ICU. The mean postinfusion hospitalization costs were $7477 and $40,054 in the outpatient and inpatient settings, respectively. Efficacy between both groups was similar. Tisagenlecleucel can be safely administered to some patients in the outpatient setting, which may reduce HCRU for patients with r/r FL.
Collapse
Affiliation(s)
- Nathan Hale Fowler
- The University of Texas MD Anderson Cancer Center, Houston, Texas; BostonGene, Waltham, Massachusetts.
| | - Michael Dickinson
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, and the Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Monalisa Ghosh
- Michigan Medicine University of Michigan, Ann Arbor, Michigan
| | - Andy I Chen
- Oregon Health and Science University, Portland, Oregon
| | - Charalambos Andreadis
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Ranjan Tiwari
- Novartis Healthcare Pvt. Ltd, Hyderabad, Telangana, India
| | - Aisha Masood
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey
| | - Roberto Ramos
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey
| | | | | | - Martin Dreyling
- Klinikum Der Universität München-Grosshadern, Medizinische Klinik und Poliklinik III, München, Germany
| | - Stephen J Schuster
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
214
|
Nath K, Wudhikarn K, Alarcon Tomas A, Perales MA. Safety evaluation of axicabtagene ciloleucel for relapsed or refractory large B-cell lymphoma. Expert Opin Drug Saf 2023; 22:5-15. [PMID: 36737060 PMCID: PMC9975047 DOI: 10.1080/14740338.2023.2177268] [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: 08/04/2022] [Accepted: 02/02/2023] [Indexed: 02/05/2023]
Abstract
INTRODUCTION CD19-directed chimeric antigen receptor (CAR) T-cell therapy is a highly effective therapy for patients with relapsed/refractory large B-cell lymphoma (LBCL) and three CD19 CAR T-cell products (axicabtagene ciloleucel, tisagenlecleucel and lisocabtagene maraleucel) are currently approved for this indication. Despite the clinical benefit of CD19 directed CAR T-cell therapy, this treatment is associated with significant morbidity from treatment-emergent toxicities. AREAS COVERED This Review discusses the safety considerations of axicabtagene ciloleucel in patients with LBCL. This includes discussion of the frequently observed immune-mediated toxicities of cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. Additionally, we review CAR T-cell therapy related cytopenias, infection, organ dysfunction and the more recently described hemophagocytic lymphohistiocytosis. EXPERT OPINION A thorough understanding of the toxicities associated with CD19-directed CAR T-cell therapy will facilitate the optimal selection of patients for this therapy. Furthermore, knowledge of preventative measures of CAR T-cell related complications, and early recognition and appropriate intervention will lead to the safe administration of these therapies, and ultimately improved outcomes for our patients.
Collapse
Affiliation(s)
- Karthik Nath
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kitsada Wudhikarn
- Division of Hematology and Center of Excellence in Translational Hematology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Ana Alarcon Tomas
- Division of Hematology and Hemotherapy, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
215
|
Schuessler-Lenz M, Herberts C, Reischl I, Ruiz S, Celis P, Beuneu C, Kjeken R, Timón M. Marketing Regulatory Oversight of Advanced Therapy Medicinal Products in Europe. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1430:1-21. [PMID: 37526839 DOI: 10.1007/978-3-031-34567-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Advanced therapy medicinal products (ATMP) in the European Union (EU) are regulated by Regulation 1394/2007 and comprise gene and cell therapy and tissue-engineered products. Under this framework, ATMP are authorised by the centralised procedure, coordinated by the European Medicines Agency (EMA), whereas clinical trial authorisations remain at the remit of each National Competent Authority. The Committee for Advanced Therapies is responsible for the scientific evaluation of the marketing authorisation applications and for generating a draft opinion that goes to the Committee for Human Medicinal Products for a final opinion. For every application, data and information relating to manufacturing processes and quality control of the active substance and final product have to be submitted for assessment together with data from non-clinical and clinical safety and efficacy studies. Technical requirements for ATMP are defined in the legislation, and guidance for different products is available through several EMA/CAT guidelines.Due to the diverse and complex nature of ATMP, a need for some regulatory flexibility was recognised. Thus, a risk-based approach was introduced in Regulation 1394/2007 allowing adapted regulatory requirements. This has led, for instance, to the development of good manufacturing practice (GMP) guidelines specific for ATMP. This, together with enhanced regulatory support, has allowed an increasing number of successful marketing authorisation applications resulting in 25 licensed ATMP in the EU, mainly gene therapy medicinal products. The promise of messenger RNA and genome editing technologies as therapeutic tools make the future for these innovative medicinal products look even brighter.This chapter reviews the regulatory landscape together with some of the support initiatives developed for ATMP in the EU.
Collapse
Affiliation(s)
- Martina Schuessler-Lenz
- Division Haematology/Cell and Gene Therapies, Paul-Ehrlich-Institute, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Carla Herberts
- Division Europe, Medicines Evaluation Board, RG, Utrecht, Netherlands
| | - Ilona Reischl
- Division Clinical Trials, Institute Surveillance, Austrian Medicines and Medical Devices Agency (AGES MEA), Vienna, Austria
| | - Sol Ruiz
- Division Biological Products, Advanced Therapies and Biotechnology, Spanish Medicines Agency, Madrid, Spain
| | - Patrick Celis
- Advanced Therapies Office, European Medicines Agency, Amsterdam, Netherlands
| | - Claire Beuneu
- Federal Agency for Medicines and Health Products, Brussels, Belgium
| | | | - Marcos Timón
- Division Biological Products, Advanced Therapies and Biotechnology, Spanish Medicines Agency, Madrid, Spain.
| |
Collapse
|
216
|
Logue JM, Peres LC, Hashmi H, Colin-Leitzinger CM, Shrewsbury AM, Hosoya H, Gonzalez RM, Copponex C, Kottra KH, Hovanky V, Sahaf B, Patil S, Lazaryan A, Jain MD, Baluch A, Klinkova OV, Bejanyan N, Faramand RG, Elmariah H, Khimani F, Davila ML, Mishra A, Blue BJ, Grajales-Cruz AF, Castaneda Puglianini OA, Liu HD, Nishihori T, Freeman CL, Brayer JB, Shain KH, Baz RC, Locke FL, Alsina M, Sidana S, Hansen DK. Early cytopenias and infections after standard of care idecabtagene vicleucel in relapsed or refractory multiple myeloma. Blood Adv 2022; 6:6109-6119. [PMID: 35939783 PMCID: PMC9768247 DOI: 10.1182/bloodadvances.2022008320] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 12/31/2022] Open
Abstract
Idecabtagene vicleucel (ide-cel) was FDA-approved in March 2021 for the treatment of relapsed/refractory multiple myeloma after 4 lines of therapy. On the KarMMa trial, grade ≥ 3 cytopenias and infections were common. We sought to characterize cytopenias and infections within 100 days after ide-cel in the standard-of-care (SOC) setting. This multi-center retrospective study included 52 patients who received SOC ide-cel; 47 reached day-90 follow-up. Data were censored at day 100. Grade ≥ 3 cytopenia was present among 65% of patients at day 30 and 40% of patients at day 90. Granulocyte colony stimulating factor (G-CSF) was administered to 88%, packed red blood cell transfusions to 63%, platelet transfusions to 42%, thrombopoietin (TPO) agonists to 21%, intravenous immunoglobulin to 13%, and CD34+ stem cell boosts to 8%. At day 100, 19% and 13% of patients had ongoing use of TPO agonists and G-CSF, respectively. Infections occurred in 54% of patients and were grade ≥ 3 in 23%. Earlier infections in the first 30 days were typically bacterial (68%) and severe (50%). Later infections between days 31 and 100 were 50% bacterial and 42% viral; only 13% were grade ≥ 3. On univariate analysis, high pre-CAR-T marrow myeloma burden (≥ 50%), circulating plasma cells at pre-lymphodepletion (LD), and grade ≥ 3 anemia at pre-LD were associated with grade ≥ 3 cytopenia at both days 30 and 90. Longer time from last bridging treatment to LD was the only significant risk factor for infection.
Collapse
Affiliation(s)
- Jennifer M. Logue
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Lauren C. Peres
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Hamza Hashmi
- Division of Hematology and Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC
| | | | - Alexandria M. Shrewsbury
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Hitomi Hosoya
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Rebecca M. Gonzalez
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Christina Copponex
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Krista H. Kottra
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Vanna Hovanky
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Bita Sahaf
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Sunita Patil
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Aleksandr Lazaryan
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Michael D. Jain
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Aliyah Baluch
- Division of Infectious Diseases, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Olga V. Klinkova
- Division of Infectious Diseases, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Nelli Bejanyan
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Rawan G. Faramand
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Hany Elmariah
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Farhad Khimani
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Marco L. Davila
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Asmita Mishra
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Brandon J. Blue
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Ariel F. Grajales-Cruz
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Omar A. Castaneda Puglianini
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Hien D. Liu
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Taiga Nishihori
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Ciara L. Freeman
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Jason B. Brayer
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Kenneth H. Shain
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Rachid C. Baz
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Frederick L. Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Melissa Alsina
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Surbhi Sidana
- Division of Blood and Marrow Transplantation and Cellular Therapy, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Doris K. Hansen
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| |
Collapse
|
217
|
Hernani R, Benzaquén A, Solano C. Toxicities following CAR-T therapy for hematological malignancies. Cancer Treat Rev 2022; 111:102479. [DOI: 10.1016/j.ctrv.2022.102479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/07/2022] [Accepted: 10/18/2022] [Indexed: 11/25/2022]
|
218
|
Lolli G, Ursi M, Dicataldo M, Casadei B, Storci G, Argnani L, Bonifazi F, Zinzani PL. Allogeneic stem cell transplantation is capable of healing a case of bone marrow aplasia following CAR-T cell therapy in a lymphoma patient. Leuk Lymphoma 2022; 63:3012-3014. [PMID: 35866874 DOI: 10.1080/10428194.2022.2102622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Ginevra Lolli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy.,Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Margherita Ursi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy.,Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Michele Dicataldo
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy.,IRCCS Azienda-Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Beatrice Casadei
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy.,Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Gianluca Storci
- IRCCS Azienda-Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Lisa Argnani
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | | | - Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy.,Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| |
Collapse
|
219
|
Oligoclonal T-cell expansion in a patient with bone marrow failure after CD19 CAR-T therapy for Richter-transformed DLBCL. Blood 2022; 140:2175-2179. [PMID: 35776908 PMCID: PMC9837444 DOI: 10.1182/blood.2022017015] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/18/2022] [Indexed: 01/21/2023] Open
|
220
|
Rial Saborido J, Völkl S, Aigner M, Mackensen A, Mougiakakos D. Role of CAR T Cell Metabolism for Therapeutic Efficacy. Cancers (Basel) 2022; 14:5442. [PMID: 36358860 PMCID: PMC9658570 DOI: 10.3390/cancers14215442] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/30/2022] [Accepted: 11/02/2022] [Indexed: 08/08/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cells hold enormous potential. However, a substantial proportion of patients receiving CAR T cells will not reach long-term full remission. One of the causes lies in their premature exhaustion, which also includes a metabolic anergy of adoptively transferred CAR T cells. T cell phenotypes that have been shown to be particularly well suited for CAR T cell therapy display certain metabolic characteristics; whereas T-stem cell memory (TSCM) cells, characterized by self-renewal and persistence, preferentially meet their energetic demands through oxidative phosphorylation (OXPHOS), effector T cells (TEFF) rely on glycolysis to support their cytotoxic function. Various parameters of CAR T cell design and manufacture co-determine the metabolic profile of the final cell product. A co-stimulatory 4-1BB domain promotes OXPHOS and formation of central memory T cells (TCM), while T cells expressing CARs with CD28 domains predominantly utilize aerobic glycolysis and differentiate into effector memory T cells (TEM). Therefore, modification of CAR co-stimulation represents one of the many strategies currently being investigated for improving CAR T cells' metabolic fitness and survivability within a hostile tumor microenvironment (TME). In this review, we will focus on the role of CAR T cell metabolism in therapeutic efficacy together with potential targets of intervention.
Collapse
Affiliation(s)
- Judit Rial Saborido
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Simon Völkl
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Michael Aigner
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität and University Hospital Erlangen, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität and University Hospital Erlangen, 91054 Erlangen, Germany
| | - Dimitrios Mougiakakos
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander-Universität and University Hospital Erlangen, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität and University Hospital Erlangen, 91054 Erlangen, Germany
- Medical Center, Department of Hematology and Oncology, Otto-von-Guericke University, 39120 Magdeburg, Germany
| |
Collapse
|
221
|
Effect of granulocyte colony-stimulating factor on toxicities after CAR T cell therapy for lymphoma and myeloma. Blood Cancer J 2022; 12:146. [PMID: 36316312 PMCID: PMC9622902 DOI: 10.1038/s41408-022-00741-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/30/2022] [Accepted: 10/06/2022] [Indexed: 11/17/2022] Open
Abstract
Chimeric antigen receptor T cells (CAR T) are groundbreaking therapies but may cause significant toxicities including cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and cytopenias. Granulocyte colony-stimulating factor (G-CSF) is often used to mitigate neutropenia after CAR T, but there is no consensus recommended strategy due to hypothesized, but largely unknown risks of exacerbating toxicities. To investigate the impact of G-CSF, we retrospectively analyzed 197 patients treated with anti-CD19 CAR T for lymphoma and 47 patients treated with anti-BCMA CAR T for multiple myeloma. In lymphoma, 140 patients (71%) received prophylactic G-CSF before CAR T (mostly pegylated G-CSF) and were compared with 57 patients (29%) treated with G-CSF after CAR T or not exposed. Prophylactic G-CSF was associated with faster neutrophil recovery (3 vs. 4 days, P < 0.01) but did not reduce recurrent neutropenia later. Prophylactic G-CSF was associated with increased grade ≥2 CRS (HR 2.15, 95% CI 1.11-4.18, P = 0.02), but not ICANS. In multiple myeloma, prophylactic G-CSF was not used; patients were stratified by early G-CSF exposure (≤2 days vs. ≥3 days after CAR T or no exposure), with no significant difference in toxicities. Future trials should clarify the optimal G-CSF strategy to improve outcomes after CAR T.
Collapse
|
222
|
Mackensen A, Müller F, Mougiakakos D, Böltz S, Wilhelm A, Aigner M, Völkl S, Simon D, Kleyer A, Munoz L, Kretschmann S, Kharboutli S, Gary R, Reimann H, Rösler W, Uderhardt S, Bang H, Herrmann M, Ekici AB, Buettner C, Habenicht KM, Winkler TH, Krönke G, Schett G. Anti-CD19 CAR T cell therapy for refractory systemic lupus erythematosus. Nat Med 2022; 28:2124-2132. [PMID: 36109639 DOI: 10.1038/s41591-022-02017-5] [Citation(s) in RCA: 444] [Impact Index Per Article: 148.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/18/2022] [Indexed: 02/06/2023]
Abstract
Systemic lupus erythematosus (SLE) is a life-threatening autoimmune disease characterized by adaptive immune system activation, formation of double-stranded DNA autoantibodies and organ inflammation. Five patients with SLE (four women and one man) with a median (range) age of 22 (6) years, median (range) disease duration of 4 (8) years and active disease (median (range) SLE disease activity index Systemic Lupus Erythematosus Disease Activity Index: 16 (8)) refractory to several immunosuppressive drug treatments were enrolled in a compassionate-use chimeric antigen receptor (CAR) T cell program. Autologous T cells from patients with SLE were transduced with a lentiviral anti-CD19 CAR vector, expanded and reinfused at a dose of 1 × 106 CAR T cells per kg body weight into the patients after lymphodepletion with fludarabine and cyclophosphamide. CAR T cells expanded in vivo, led to deep depletion of B cells, improvement of clinical symptoms and normalization of laboratory parameters including seroconversion of anti-double-stranded DNA antibodies. Remission of SLE according to DORIS criteria was achieved in all five patients after 3 months and the median (range) Systemic Lupus Erythematosus Disease Activity Index score after 3 months was 0 (2). Drug-free remission was maintained during longer follow-up (median (range) of 8 (12) months after CAR T cell administration) and even after the reappearance of B cells, which was observed after a mean (±s.d.) of 110 ± 32 d after CAR T cell treatment. Reappearing B cells were naïve and showed non-class-switched B cell receptors. CAR T cell treatment was well tolerated with only mild cytokine-release syndrome. These data suggest that CD19 CAR T cell transfer is feasible, tolerable and highly effective in SLE.
Collapse
Affiliation(s)
- Andreas Mackensen
- Department of Internal Medicine 5-Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Fabian Müller
- Department of Internal Medicine 5-Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Dimitrios Mougiakakos
- Department of Internal Medicine 5-Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany.,Department of Hematology and Oncology, Otto-von-Guericke University Magdeburg (OVGU), Magdeburg, Germany
| | - Sebastian Böltz
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany.,Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Artur Wilhelm
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany.,Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Michael Aigner
- Department of Internal Medicine 5-Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Simon Völkl
- Department of Internal Medicine 5-Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - David Simon
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany.,Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Arnd Kleyer
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany.,Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Luis Munoz
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany.,Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sascha Kretschmann
- Department of Internal Medicine 5-Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Soraya Kharboutli
- Department of Internal Medicine 5-Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Regina Gary
- Department of Internal Medicine 5-Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Hannah Reimann
- Department of Internal Medicine 5-Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Wolf Rösler
- Department of Internal Medicine 5-Hematology and Oncology, Friedrich Alexander University Erlangen-Nuremberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Stefan Uderhardt
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany.,Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Martin Herrmann
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany.,Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Arif Bülent Ekici
- Institute of Human Genetics, Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christian Buettner
- Institute of Human Genetics, Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Thomas H Winkler
- Division of Genetics, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Gerhard Krönke
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany.,Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany. .,Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany.
| |
Collapse
|
223
|
Joseph A, Lafarge A, Mabrouki A, Abdel-Nabey M, Binois Y, Younan R, Azoulay E. Severe infections in recipients of cancer immunotherapy: what intensivists need to know. Curr Opin Crit Care 2022; 28:540-550. [PMID: 35950720 DOI: 10.1097/mcc.0000000000000978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Given the increased number of cancer patients admitted in the ICU and the growing importance of immunotherapy in their therapeutic arsenal, intensivists will be increasingly confronted to patients treated with immunotherapies who will present with complications, infectious and immunologic. RECENT FINDINGS Apart from their specific immunologic toxicities, cancer immunotherapy recipients also have specific immune dysfunction and face increased infectious risks that may lead to intensive care unit admission. SUMMARY Chimeric antigen receptor T-cell therapy is associated with profound immunosuppression and the risks of bacterial, fungal and viral infections vary according to the time since infusion.Immune checkpoint blockers are associated with an overall favorable safety profile but associations of checkpoint blockers and corticosteroids and immunosuppressive drugs prescribed to treat immune-related adverse events are associated with increased risks of bacterial and fungal infections.The T-cell engaging bispecific therapy blinatumomab causes profound B-cell aplasia, hypogammaglobulinemia and neutropenia, but seems to be associated with fewer infectious adverse events compared with standard intensive chemotherapy.Lastly, intravesical administration of Bacillus Calmette-Guérin (BCG) can lead to disseminated BCGitis and severe sepsis requiring a specific antibiotherapy, often associated with corticosteroid treatment.
Collapse
Affiliation(s)
- Adrien Joseph
- Medical Intensive Care Unit, Saint-Louis Teaching Hospital, Public Assistance Hospitals of Paris, Paris, France
| | | | | | | | | | | | | |
Collapse
|
224
|
Iqbal M, Bansal R, Yassine F, Gandhi S, Rosenthal A, Moustafa MA, Li Z, Craver EC, Mohty R, Murthy H, Ayala E, Tun H, Munoz J, Castro J, Lin Y, Kharfan-Dabaja MA. Impact of Rituximab and Corticosteroids on Late Cytopenias Post-Chimeric Antigen Receptor T Cell Therapy. Transplant Cell Ther 2022; 28:668.e1-668.e6. [PMID: 35842124 DOI: 10.1016/j.jtct.2022.07.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/26/2022] [Accepted: 07/07/2022] [Indexed: 11/18/2022]
Abstract
Chimeric antigen receptor (CAR) T cell therapy represents a significant advancement in the treatment of patients with relapsed/refractory B cell lymphoid malignancies. Cytokine release syndrome and immune effector cell-associated neurotoxicity represent the most acute serious adverse events post CAR T cell therapy but the occurrence and persistence of cytopenias post CAR T cell therapy represent a significant adverse event and a management challenge. While most patients typically recover blood counts by 30 days, a significant subset of patients have persistent or late cytopenias beyond 30 days. Patients receiving CAR T cell are heavily pre-treated and the impact of prior therapies on late cytopenias is not well understood. In this study, we found an association between increased number of rituximab infusions and/or cumulative rituximab dose received prior to CAR T cell infusion and persistent anemia and thrombocytopenia at 90 and 180 days afterwards. An overall increased number of prior lines of therapy was also associated with persistent lymphopenia and anemia at 90 days while receiving a prior autologous hematopoietic cell transplant was associated with a greater risk of neutropenia and lymphopenia.
Collapse
Affiliation(s)
- Madiha Iqbal
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida.
| | - Radhika Bansal
- Division Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Rochester, Minnesota
| | - Farah Yassine
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| | - Sangeetha Gandhi
- Division Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Rochester, Minnesota
| | - Allison Rosenthal
- Division Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Phoenix, Arizona
| | - Muhamad Alhaj Moustafa
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| | - Zhuo Li
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Jacksonville, Florida
| | - Emily C Craver
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Jacksonville, Florida
| | - Razan Mohty
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| | - Hemant Murthy
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| | - Ernesto Ayala
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| | - Han Tun
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| | - Javier Munoz
- Division Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Phoenix, Arizona
| | - Januario Castro
- Division Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Phoenix, Arizona
| | - Yi Lin
- Division Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Rochester, Minnesota
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
225
|
Wang Y, Song Z, Geng Y, Gao L, Xu L, Tang G, Ni X, Chen L, Chen J, Wang T, Fu W, Feng D, Yu X, Wang L, Yang J. The risk factors and early predictive model of hematotoxicity after CD19 chimeric antigen receptor T cell therapy. Front Oncol 2022; 12:987965. [PMID: 36249041 PMCID: PMC9561932 DOI: 10.3389/fonc.2022.987965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Hematotoxicity is the most common long-term adverse event after chimeric antigen receptor T cell (CAR-T) therapy. Here, a total of 71 patients with relapsed or refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL) or large B-cell lymphoma (LBCL) were used to develop an early hematotoxicity predictive model and verify the accuracy of this model. The incidences of early hematotoxicity at 3 month following CAR-T infusion in B-ALL and LBCL were 45.5% and 38.5%, respectively. Multivariate analyses revealed that the severity of cytokine release syndrome (CRS) was an independent risk factor affecting early hematotoxicity. The analysis between the peak cytokine levels and early hematotoxicity suggested that tumor necrosis factor-α (TNF-α) and C-reactive protein (CRP) were closely associated with early hematotoxicity. Then, an early predictive model of hematotoxicity was constructed based on the peak contents of TNF-α and CRP. This model could diagnose early hematotoxicity with positive predictive values of 87.7% and 85.0% in training and validation cohorts, respectively. Lastly, we constructed the nomogram for clinical practice to predict the risk of early hematotoxicity, which performed well compared with the observed probability. This early predictive model is instrumental in the risk stratification of CAR-T recipients with hematotoxicity and early intervention for high-risk patients.
Collapse
Affiliation(s)
- Yang Wang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhiqiang Song
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yuke Geng
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Lei Gao
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Lili Xu
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Gusheng Tang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiong Ni
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Li Chen
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jie Chen
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Tao Wang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Weijia Fu
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Dongge Feng
- HuaDao Biopharma (Shanghai) Limited Corporation, Shanghai, China
| | - Xuejun Yu
- HuaDao Biopharma (Shanghai) Limited Corporation, Shanghai, China
| | - Libing Wang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
- *Correspondence: Libing Wang, ; Jianmin Yang,
| | - Jianmin Yang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
- *Correspondence: Libing Wang, ; Jianmin Yang,
| |
Collapse
|
226
|
Vic S, Lemoine J, Armand P, Lemonnier F, Houot R. Transplant-ineligible but chimeric antigen receptor T-cells eligible: a real and relevant population. Eur J Cancer 2022; 175:246-253. [PMID: 36166850 DOI: 10.1016/j.ejca.2022.08.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/11/2022] [Accepted: 08/16/2022] [Indexed: 11/03/2022]
Abstract
Autologous stem cell transplantation (ASCT) and chimeric antigen receptor (CAR) T-cells are two therapeutic options for relapsed/refractory diffuse large B-cell lymphoma. Both are intensive and potentially curative therapies but differ in their efficacy and toxicity. ASCT may be offered to 'fit' patients (i.e. usually young with limited comorbidities) with chemosensitive disease. On the other hand, real world studies have shown that CAR T-cells may be safely administered to less fit and older patients. Thus, there is a potentially significant population of patients who may be offered CAR T-cell therapy despite not being eligible for ASCT. As the relative role of ASCT and CAR T-cells evolves, recognising and defining this population may be increasingly relevant. Here, we review criteria which may help identify this 'ASCT-ineligible but CAR T-cells eligible' population of patients.
Collapse
Affiliation(s)
- Samuel Vic
- Department of Hematology, CHU de Rennes, Université de Rennes, Rennes, France
| | - Jean Lemoine
- Department of Hematology, AP-HP, Université de Paris, Paris, France
| | - Philippe Armand
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - François Lemonnier
- Department of Hematology, AP-HP, Hôpitaux Universitaires Henri Mondor, Créteil, France
| | - Roch Houot
- Department of Hematology, CHU de Rennes, Université de Rennes, Rennes, France.
| |
Collapse
|
227
|
Management of prolonged cytopenia following CAR T-cell therapy. Bone Marrow Transplant 2022; 57:1839-1841. [PMID: 36131002 DOI: 10.1038/s41409-022-01771-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 07/23/2022] [Accepted: 07/26/2022] [Indexed: 11/08/2022]
|
228
|
Si X, Gu T, Liu L, Huang Y, Han Y, Qian P, Huang H. Hematologic cytopenia post CAR T cell therapy: Etiology, potential mechanisms and perspective. Cancer Lett 2022; 550:215920. [PMID: 36122628 DOI: 10.1016/j.canlet.2022.215920] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/02/2022] [Accepted: 09/12/2022] [Indexed: 11/25/2022]
Abstract
Chimeric Antigen-Receptor (CAR) T-cell therapies have shown dramatic efficacy in treating relapsed and refractory cancers, especially B cell malignancies. However, these innovative therapies cause adverse toxicities that limit the broad application in clinical settings. Hematologic cytopenias, one frequently reported adverse event following CAR T cell treatment, are manifested as a disorder of hematopoiesis with decreased number of mature blood cells and subdivided into anemia, thrombocytopenia, leukopenia, and neutropenia, which increase the risk of infections, fatigue, bleeding, fever, and even fatality. Herein, we initially summarized the symptoms, etiology, risk factors and management of cytopenias. Further, we elaborated the cellular and molecular mechanisms underlying the initiation and progression of cytopenias following CAR T cell therapy based on previous studies about acquired cytopenias. Overall, this review will facilitate our understanding of the etiology of cytopenias and shed lights into developing new therapies against CAR T cell-induced cytopenias.
Collapse
Affiliation(s)
- Xiaohui Si
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Tianning Gu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Lianxuan Liu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Yue Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Yingli Han
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Pengxu Qian
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China.
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China.
| |
Collapse
|
229
|
Zhou J, Zhang Y, Shan M, Zong X, Geng H, Li J, Chen G, Yu L, Xu Y, Li C, Wu D. Cytopenia after chimeric antigen receptor T cell immunotherapy in relapsed or refractory lymphoma. Front Immunol 2022; 13:997589. [PMID: 36131934 PMCID: PMC9484486 DOI: 10.3389/fimmu.2022.997589] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/16/2022] [Indexed: 11/26/2022] Open
Abstract
Background Patients with relapsed or refractory (R/R) lymphomas have benefited from chimeric antigen receptor (CAR)-T-cell therapy. However, this treatment is linked to a high frequency of adverse events (AEs), such as cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and hematologic toxicity. There has been increasing interest in hematological toxicity in recent years, as it can result in additional complications, such as infection or hemorrhage, which remain intractable. Methods We conducted a retrospective, single-institution study to evaluate the patterns and outcomes of cytopenia following CAR-T-cell infusion and potential associated factors. Results Overall, 133 patients with R/R lymphoma who received CAR-T-cell therapy from June, 2017 to April, 2022 were included in this analysis. Severe neutropenia, anemia and thrombocytopenia occurred frequently (71, 30 and 41%, respectively) after CAR-T-cell infusion. A total of 98% of severe neutropenia and all severe thrombocytopenia cases occurred in the early phase. Early severe cytopenia was associated with CRS incidence and severity, as well as peak inflammatory factor (IL-6, C-reactive protein (CRP), and ferritin) levels. In multivariate analysis, prior hematopoietic stem cell transplantation (HSCT), baseline hemoglobin (HB), and lymphodepleting chemotherapy were independent adverse factors associated with early severe cytopenia. In addition, 18% and 35% of patients had late neutrophil- and platelet (PLT)-related toxicity, respectively. In multivariate analysis, lower baseline PLT count was an independent factor associated with late thrombocytopenia. More severe cytopenia was associated with higher infection rates and poorer survival. Conclusions This research indicates that improved selection of patients and management of CRS may help to decrease the severity of cytopenias and associated AEs and improve survival following CAR-T-cell therapy. Clinical Trial Registration https://www.clinicaltrials.gov/ct2/show/NCT03196830, identifier NCT03196830.
Collapse
Affiliation(s)
- Jin Zhou
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Ying Zhang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Meng Shan
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Xiangping Zong
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Hongzhi Geng
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Jiaqi Li
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Guanghua Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Lei Yu
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Yang Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
- *Correspondence: Depei Wu, ; Caixia Li, ; Yang Xu,
| | - Caixia Li
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
- *Correspondence: Depei Wu, ; Caixia Li, ; Yang Xu,
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
- *Correspondence: Depei Wu, ; Caixia Li, ; Yang Xu,
| |
Collapse
|
230
|
Perales MA, Anderson LD, Jain T, Kenderian SS, Oluwole OO, Shah GL, Svoboda J, Hamadani M. Role of CD19 Chimeric Antigen Receptor T Cells in Second-Line Large B Cell Lymphoma: Lessons from Phase 3 Trials. An Expert Panel Opinion from the American Society for Transplantation and Cellular Therapy. Transplant Cell Ther 2022; 28:546-559. [PMID: 35768052 PMCID: PMC9427727 DOI: 10.1016/j.jtct.2022.06.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 12/25/2022]
Abstract
Since 2017, 3 CD19-directed chimeric antigen receptor (CAR) T cell therapies-axicabtagene ciloleucel, tisagenlecleucel, and lisocabtagene maraleucel-have been approved for relapsed/refractory aggressive diffuse large B cell lymphoma after 2 lines of therapy. Recently, 3 prospective phase 3 randomized clinical trials were conducted to define the optimal second-line treatment by comparing each of the CAR T cell products to the current standard of care: ZUMA-7 for axicabtagene ciloleucel, BELINDA for tisagenlecleucel, and TRANSFORM for lisocabtagene maraleucel. These 3 studies, although largely addressing the same question, had different outcomes, with ZUMA-7 and TRANSFORM demonstrating significant improvement with CD19 CAR T cells in second-line therapy compared with standard of care but BELINDA not showing any benefit. The US Food and Drug Administration has now approved axicabtagene ciloleucel and lisocabtagene maraleucel for LBCL that is refractory to first-line chemoimmunotherapy or relapse occurring within 12 months of first-line chemoimmunotherapy. Following the reporting of these practice changing studies, here a group of experts convened by the American Society for Transplantation and Cellular Therapy provides a comprehensive review of the 3 studies, emphasizing potential differences, and shares perspectives on what these results mean to clinical practice in this new era of treatment of B cell lymphomas.
Collapse
Affiliation(s)
- Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York.
| | - Larry D Anderson
- Hematologic Malignancies, Transplantation, and Cellular Therapy Program, Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| | - Tania Jain
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Saad S Kenderian
- T Cell Engineering, Mayo Clinic, Mayo Clinic Graduate School of Biomedical Sciences, Division of Hematology, Department of Immunology and Department of Molecular Medicine, Rochester, Minnesota
| | - Olalekan O Oluwole
- Division of Hematology/Oncology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Gunjan L Shah
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Jakub Svoboda
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mehdi Hamadani
- BMT & Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
231
|
Ghilardi G, Chong EA, Svoboda J, Wohlfarth P, Nasta SD, Williamson S, Landsburg JD, Gerson JN, Barta SK, Pajarillo R, Myers J, Chen AI, Schachter L, Yelton R, Ballard HJ, Hodges Dwinal A, Gier S, Victoriano D, Weber E, Napier E, Garfall A, Porter DL, Jäger U, Maziarz RT, Ruella M, Schuster SJ. Bendamustine is safe and effective for lymphodepletion before tisagenlecleucel in patients with refractory or relapsed large B-cell lymphomas. Ann Oncol 2022; 33:916-928. [PMID: 35690221 PMCID: PMC11332249 DOI: 10.1016/j.annonc.2022.05.521] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Anti-CD19 chimeric antigen receptor T-cell immunotherapy (CAR-T) is now a standard treatment of relapsed or refractory B-cell non-Hodgkin lymphomas; however, a significant portion of patients do not respond to CAR-T and/or experience toxicities. Lymphodepleting chemotherapy is a critical component of CAR-T that enhances CAR-T-cell engraftment, expansion, cytotoxicity, and persistence. We hypothesized that the lymphodepletion regimen might affect the safety and efficacy of CAR-T. PATIENTS AND METHODS We compared the safety and efficacy of lymphodepletion using either fludarabine/cyclophosphamide (n = 42) or bendamustine (n = 90) before tisagenlecleucel in two cohorts of patients with relapsed or refractory large B-cell lymphomas treated consecutively at three academic institutions in the United States (University of Pennsylvania, n = 90; Oregon Health & Science University, n = 35) and Europe (University of Vienna, n = 7). Response was assessed using the Lugano 2014 criteria and toxicities were assessed by the Common Terminology Criteria for Adverse Events (CTCAE) version 5.0 and, when possible, the American Society for Transplantation and Cellular Therapy (ASTCT) consensus grading. RESULTS Fludarabine/cyclophosphamide led to more profound lymphocytopenia after tisagenlecleucel infusion compared with bendamustine, although the efficacy of tisagenlecleucel was similar between the two groups. We observed significant differences, however, in the frequency and severity of adverse events. In particular, patients treated with bendamustine had lower rates of cytokine release syndrome and neurotoxicity. In addition, higher rates of hematological toxicities were observed in patients receiving fludarabine/cyclophosphamide. Bendamustine-treated patients had higher nadir neutrophil counts, hemoglobin levels, and platelet counts, as well as a shorter time to blood count recovery, and received fewer platelet and red cell transfusions. Fewer episodes of infection, neutropenic fever, and post-infusion hospitalization were observed in the bendamustine cohort compared with patients receiving fludarabine/cyclophosphamide. CONCLUSIONS Bendamustine for lymphodepletion before tisagenlecleucel has efficacy similar to fludarabine/cyclophosphamide with reduced toxicities, including cytokine release syndrome, neurotoxicity, infectious and hematological toxicities, as well as reduced hospital utilization.
Collapse
Affiliation(s)
- G Ghilardi
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, USA; Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, USA
| | - E A Chong
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, USA; Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, USA
| | - J Svoboda
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, USA; Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, USA
| | - P Wohlfarth
- Medical University of Vienna, Division of Hematology and Hemostaseology, Department of Medicine I Wien, Comprehensive Cancer Center, Vienna, Austria
| | - S D Nasta
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, USA
| | - S Williamson
- Oregon Health & Science University Knight Cancer Institute, Adult Blood and Marrow Stem Cell Transplant & Cell Therapy Program, Portland, USA
| | - J D Landsburg
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, USA
| | - J N Gerson
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, USA
| | - S K Barta
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, USA; Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, USA
| | - R Pajarillo
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, USA; Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, USA
| | - J Myers
- Oregon Health & Science University Knight Cancer Institute, Adult Blood and Marrow Stem Cell Transplant & Cell Therapy Program, Portland, USA
| | - A I Chen
- Oregon Health & Science University Knight Cancer Institute, Adult Blood and Marrow Stem Cell Transplant & Cell Therapy Program, Portland, USA
| | - L Schachter
- Oregon Health & Science University Knight Cancer Institute, Adult Blood and Marrow Stem Cell Transplant & Cell Therapy Program, Portland, USA
| | - R Yelton
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, USA; Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia, USA
| | - H J Ballard
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, USA
| | - A Hodges Dwinal
- Oregon Health & Science University Knight Cancer Institute, Adult Blood and Marrow Stem Cell Transplant & Cell Therapy Program, Portland, USA
| | - S Gier
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, USA
| | - D Victoriano
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, USA
| | - E Weber
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, USA
| | - E Napier
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, USA
| | - A Garfall
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, USA
| | - D L Porter
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, USA
| | - U Jäger
- Medical University of Vienna, Division of Hematology and Hemostaseology, Department of Medicine I Wien, Comprehensive Cancer Center, Vienna, Austria
| | - R T Maziarz
- Oregon Health & Science University Knight Cancer Institute, Adult Blood and Marrow Stem Cell Transplant & Cell Therapy Program, Portland, USA
| | - M Ruella
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, USA; Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, USA
| | - S J Schuster
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, USA; Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, USA.
| |
Collapse
|
232
|
Maakaron JE, Hu M, El Jurdi N. Chimeric antigen receptor T cell therapy for cancer: clinical applications and practical considerations. BRITISH MEDICAL JOURNAL 2022. [DOI: 10.1136/bmj-2021-068956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Abstract
Chimeric antigen receptor T cells have revolutionized the treatment of hematological malignancies during the past five years, boasting impressive response rates and durable remissions for patients who previously had no viable options. In this review, we provide a brief historical overview of their development. We focus on the practical aspects of a patient’s journey through this treatment and the unique toxicities and current best practices to manage those. We then discuss the key registration trials that have led to approvals for the treatment of relapsed/refractory acute lymphoblastic leukemia (ALL), diffuse large B cell lymphoma (DLBCL), follicular lymphoma, mantle cell lymphoma (MCL), and multiple myeloma. Finally, we consider the future development and research directions of this cutting edge therapy.
Collapse
|
233
|
de Tena PS, Bailén R, Oarbeascoa G, Gómez‐Centurión I, Pérez‐Corral A, Carbonell D, Martínez‐Laperche C, Sancho M, Bastos‐Oreiro M, Conde‐Royo D, Fernández‐Caldas P, Muñoz C, Sabell S, Buño I, Anguita J, Díez‐Martín JL, Kwon M. Allogeneic
CD34
‐selected stem cell boost as salvage treatment of life‐threatening infection and severe cytopenias after
CAR‐T
cell therapy. Transfusion 2022; 62:2143-2147. [DOI: 10.1111/trf.17071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/02/2022] [Accepted: 06/05/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Pablo Silva de Tena
- Hemathology and Hemotherapy Department Hospital General Universitario Gregorio Marañón Madrid Spain
| | - Rebeca Bailén
- Hemathology and Hemotherapy Department Hospital General Universitario Gregorio Marañón Madrid Spain
- Department of Translational Oncology Instituto de Investigación Sanitaria Gregorio Marañón Madrid Spain
| | - Gillen Oarbeascoa
- Hemathology and Hemotherapy Department Hospital General Universitario Gregorio Marañón Madrid Spain
- Department of Translational Oncology Instituto de Investigación Sanitaria Gregorio Marañón Madrid Spain
| | - Ignacio Gómez‐Centurión
- Hemathology and Hemotherapy Department Hospital General Universitario Gregorio Marañón Madrid Spain
- Department of Translational Oncology Instituto de Investigación Sanitaria Gregorio Marañón Madrid Spain
| | - Ana Pérez‐Corral
- Hemathology and Hemotherapy Department Hospital General Universitario Gregorio Marañón Madrid Spain
- Department of Translational Oncology Instituto de Investigación Sanitaria Gregorio Marañón Madrid Spain
| | - Diego Carbonell
- Hemathology and Hemotherapy Department Hospital General Universitario Gregorio Marañón Madrid Spain
- Department of Translational Oncology Instituto de Investigación Sanitaria Gregorio Marañón Madrid Spain
| | - Carolina Martínez‐Laperche
- Hemathology and Hemotherapy Department Hospital General Universitario Gregorio Marañón Madrid Spain
- Department of Translational Oncology Instituto de Investigación Sanitaria Gregorio Marañón Madrid Spain
| | - Milagros Sancho
- Intensive Care Unit Hospital General Universitario Gregorio Marañón Madrid Spain
| | - Mariana Bastos‐Oreiro
- Hemathology and Hemotherapy Department Hospital General Universitario Gregorio Marañón Madrid Spain
- Department of Translational Oncology Instituto de Investigación Sanitaria Gregorio Marañón Madrid Spain
| | - Diego Conde‐Royo
- Hemathology and Hemotherapy Department Hospital General Universitario Gregorio Marañón Madrid Spain
| | - Paula Fernández‐Caldas
- Hemathology and Hemotherapy Department Hospital General Universitario Gregorio Marañón Madrid Spain
| | - Cristina Muñoz
- Hemathology and Hemotherapy Department Hospital General Universitario Gregorio Marañón Madrid Spain
- Department of Translational Oncology Instituto de Investigación Sanitaria Gregorio Marañón Madrid Spain
| | - Santiago Sabell
- Hemathology and Hemotherapy Department Hospital General Universitario Gregorio Marañón Madrid Spain
- Department of Translational Oncology Instituto de Investigación Sanitaria Gregorio Marañón Madrid Spain
| | - Ismael Buño
- Hemathology and Hemotherapy Department Hospital General Universitario Gregorio Marañón Madrid Spain
- Department of Translational Oncology Instituto de Investigación Sanitaria Gregorio Marañón Madrid Spain
| | - Javier Anguita
- Hemathology and Hemotherapy Department Hospital General Universitario Gregorio Marañón Madrid Spain
- Department of Translational Oncology Instituto de Investigación Sanitaria Gregorio Marañón Madrid Spain
- Department of Medicine Universidad Complutense de Madrid Madrid Spain
| | - José Luis Díez‐Martín
- Hemathology and Hemotherapy Department Hospital General Universitario Gregorio Marañón Madrid Spain
- Department of Translational Oncology Instituto de Investigación Sanitaria Gregorio Marañón Madrid Spain
- Department of Medicine Universidad Complutense de Madrid Madrid Spain
| | - Mi Kwon
- Hemathology and Hemotherapy Department Hospital General Universitario Gregorio Marañón Madrid Spain
- Department of Translational Oncology Instituto de Investigación Sanitaria Gregorio Marañón Madrid Spain
| |
Collapse
|
234
|
Berger SC, Fehse B, Akyüz N, Geffken M, Wolschke C, Janson D, Gagelmann N, Luther M, Wichmann D, Frenzel C, Thayssen G, Alegiani A, Badbaran A, Zeschke S, Dierlamm J, Kröger N, Ayuk FA. Molecular monitoring of T-cell kinetics and migration in severe neurotoxicity after real-world CD19-specific chimeric antigen receptor T cell therapy. Haematologica 2022; 108:444-456. [PMID: 35950534 PMCID: PMC9890009 DOI: 10.3324/haematol.2022.281110] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Indexed: 02/03/2023] Open
Abstract
CD19-specific chimeric antigen receptor (CD19-CAR) T-cell therapies mediate durable responses in late-stage B-cell malignancies, but can be complicated by a potentially severe immune effector cell-associated neurotoxicity syndrome (ICANS). Despite broad efforts, the precise mechanisms of ICANS are not entirely known, and resistance to current ICANSdirected therapies (especially corticosteroids) has been observed. Recent data suggest that inflammatory cytokines and/or targeting of cerebral CD19-expressing pericytes can disrupt the blood-brain barrier and facilitate influx of immune cells, including CAR T cells. However, specific tools for CD19-CAR T-cell analysis within often minute samples of cerebrospinal fluid (CSF) are not broadly available. Here, we applied our recently developed digital polymerase chain reaction assays to monitor CD19-CAR T-cell kinetics in CSF and blood in real-world patients with neurotoxicity. Consistently, we observed a CAR T-cell enrichment within CSF in ICANS patients with further progressive accumulation despite intense corticosteroid- containing immuno-chemotherapies in a subset of patients with prolonged and therapy-resistant grade 3-4 neurotoxicity. We used next-generation T-cell receptor-b sequencing to assess the repertoire of treatment-refractory cells. Longitudinal analysis revealed a profound skewing of the T-cell receptor repertoire, which at least partly reflected selective expansion of infused T-cell clones. Interestingly, a major fraction of eventually dominating hyperexpanded T-cell clones were of non-CAR T-cell derivation. These findings hint to a role of therapy-refractory T-cell clones in severe ICANS development and prompt future systematic research to determine if CAR T cells may serve as 'door openers' and to further characterize both CAR-positive and non-CAR T cells to interrogate the transcriptional signature of these possibly pathologic T cells.
Collapse
Affiliation(s)
| | - Boris Fehse
- Department of Stem Cell Transplantation,Research Department Cell and Gene Therapy
| | | | | | | | | | | | | | | | | | - Guenther Thayssen
- Department of Neurology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Anna Alegiani
- Department of Neurology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany,°Current address: Department of Neurology with Stroke Unit, Asklepios Clinic Altona, Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
235
|
Prolonged thrombocytopenia after CAR T-cell therapy: the role of thrombopoietin receptor agonists. Blood Adv 2022; 7:537-540. [PMID: 35930692 PMCID: PMC9979763 DOI: 10.1182/bloodadvances.2022008066] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/13/2022] [Accepted: 07/17/2022] [Indexed: 11/20/2022] Open
|
236
|
Brudno JN, Natrakul D, Lam N, Dulau-Florea A, Yuan CM, Kochenderfer JN. Acute and delayed cytopenias following CAR T-cell therapy: an investigation of risk factors and mechanisms. Leuk Lymphoma 2022; 63:1849-1860. [PMID: 35389319 PMCID: PMC11627212 DOI: 10.1080/10428194.2022.2056172] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 03/02/2022] [Accepted: 03/09/2022] [Indexed: 10/18/2022]
Abstract
Prolonged myelosuppression after chimeric antigen receptor (CAR) T-cell therapy is common and poorly understood. A retrospective analysis of 43 patients was conducted to investigate factors contributing to CAR T-cell-related cytopenias. Thirty-five patients were evaluable for analysis of delayed cytopenias occurring after initial hematologic recovery. Time to hematologic recovery (TTHR) was defined as number of days after CAR T-cell infusion for recovery to hemoglobin ≥8.0 g/dL, platelets ≥50.0 k/µL, and neutrophil count ≥1.0 k/µL without transfusions or growth factors for 7 days. Baseline percent bone marrow (BM) malignancy involvement correlated with TTHR (p = .0047). Patients with grades 3-4 cytokine-release syndrome (CRS) had longer TTHR than those with grades 0-2 CRS (p = .0479). Patients who developed prolonged or delayed cytopenias after anti-BCMA CAR T cells had a higher percentage of BM aspirate CAR+ cells at 2 months (n = 10; p = .0159).
Collapse
Affiliation(s)
- Jennifer N. Brudno
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Danielle Natrakul
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Norris Lam
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alina Dulau-Florea
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Constance M. Yuan
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James N. Kochenderfer
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
237
|
Wudhikarn K, Perales MA. Infectious complications, immune reconstitution, and infection prophylaxis after CD19 chimeric antigen receptor T-cell therapy. Bone Marrow Transplant 2022; 57:1477-1488. [PMID: 35840746 PMCID: PMC9285870 DOI: 10.1038/s41409-022-01756-w] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/24/2022] [Accepted: 06/30/2022] [Indexed: 12/15/2022]
Abstract
CD19-targeted chimeric antigen receptor (CAR) T-cell becomes a breakthrough therapy providing excellent remission rates and durable disease control for patients with relapsed/refractory (R/R) hematologic malignancies. However, CAR T-cells have several potential side effects including cytokine release syndrome, neurotoxicities, cytopenia, and hypogammaglobulinemia. Infection has been increasingly recognized as a complication of CAR T-cell therapy. Several factors predispose CAR T-cell recipients to infection. Fortunately, although studies show a high incidence of infection post-CAR T-cells, most infections are manageable. In contrast to patients who undergo hematopoietic stem cell transplant, less is known about post-CAR T-cell immune reconstitution. Therefore, evidence regarding antimicrobial prophylaxis and vaccination strategies in these patients is more limited. As CAR T-cell therapy becomes the standard treatment for R/R B lymphoid malignancies, we should expect a larger impact of infections in these patients and the need for increased clinical attention. Studies exploring infection and immune reconstitution after CAR T-cell therapy are clinically relevant and will provide us with a better understanding of the dynamics of immune function after CAR T-cell therapy including insights into appropriate strategies for prophylaxis and treatment of infections in these patients. In this review, we describe infections in recipients of CAR T-cells, and discuss risk factors and potential mitigation strategies.
Collapse
Affiliation(s)
- Kitsada Wudhikarn
- Division of Hematology and Research Unit in Translational Hematology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| | - Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
238
|
CD34+-selected stem cell boost can safely improve cytopenias following CAR-T therapy. Blood Adv 2022; 6:4715-4718. [PMID: 35790110 PMCID: PMC9631677 DOI: 10.1182/bloodadvances.2022007572] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/28/2022] [Indexed: 11/29/2022] Open
|
239
|
Dowling MR, Dickinson M. Post CAR-T cytopenia: poorly understood and clinically challenging. Leuk Lymphoma 2022; 63:1774-1776. [DOI: 10.1080/10428194.2022.2095631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Mark R. Dowling
- Clinical Haematology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Clinical Haematology, Royal Melbourne Hospital, Parkville, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Michael Dickinson
- Clinical Haematology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Clinical Haematology, Royal Melbourne Hospital, Parkville, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
240
|
Schroeder BA, Jess J, Sankaran H, Shah NN. Clinical trials for chimeric antigen receptor T-cell therapy: lessons learned and future directions. Curr Opin Hematol 2022; 29:225-232. [PMID: 35787551 PMCID: PMC9354650 DOI: 10.1097/moh.0000000000000723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize the status and utilization of chimeric antigen receptor T-cell (CAR-T) therapy based on the most recent clinical trials in patients with leukemia and lymphoma. Additionally, this review will highlight limitations in current strategies, discuss efforts in toxicity mitigation, and outline future directions for investigation. RECENT FINDINGS CD19 targeted CAR-T-cell therapy (CD19-CAR) is highly effective in patients with relapsed/refractory (r/r) B-cell hematologic malignancies. However, multiple challenges have arisen, particularly life-threatening adverse events, such as cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. Despite these challenges, recent CD19-CAR trials, including two randomized studies, have demonstrated both impressive initial results along with durable responses. Combined with results emerging from 'real-world' experience, the efficacy of CAR-T-cells is high, propelling CAR-T-cells studies targeting alternate B-cell antigens [e.g. CD20, CD22 and CD269 (BCMA)] and other targets for hematologic malignancies, along with solid and CNS tumors. SUMMARY Given the benefit for CD19-CAR, determining the appropriate place in utilization for both an individual patient's treatment course and more broadly in the generalized treatment paradigm is critically needed. We discuss the most recent trials exploring this topic and future directions in the field.
Collapse
Affiliation(s)
- Brett A Schroeder
- National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Jennifer Jess
- Pediatric Oncology Branch, Center for Cancer Research (CCR)
| | - Hari Sankaran
- Biometric Research Program, Division of Cancer Treatment and Diagnosis
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research (CCR)
| |
Collapse
|
241
|
Kwon M, Iacoboni G, Reguera JL, Corral LL, Morales RH, Ortiz-Maldonado V, Guerreiro M, Caballero AC, Domínguez MLG, Pina JMS, Mussetti A, Sancho JM, Bastos-Oreiro M, Catala E, Delgado J, Henriquez HL, Sanz J, Calbacho M, Bailén R, Carpio C, Ribera JM, Sureda A, Briones J, Hernandez-Boluda JC, Cebrián NM, Martin JLD, Martín A, Barba P. Axicabtagene ciloleucel compared to tisagenlecleucel for the treatment of aggressive B-cell lymphoma. Haematologica 2022; 108:110-121. [PMID: 35770532 PMCID: PMC9827173 DOI: 10.3324/haematol.2022.280805] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Indexed: 02/05/2023] Open
Abstract
Axicabtagene ciloleucel (axi-cel) and tisagenlecleucel (tisa-cel) are CD19-targeted chimeric antigen receptor (CAR) T cells approved for relapsed/refractory (R/R) large B-cell lymphoma (LBCL). We performed a retrospective study to evaluate safety and efficacy of axi-cel and tisa-cel outside the setting of a clinical trial. Data from consecutive patients with R/R LBCL who underwent apheresis for axi-cel or tisa-cel were retrospectively collected from 12 Spanish centers. A total of 307 patients underwent apheresis for axi-cel (n=152) and tisa-cel (n=155) from November 2018 to August 2021, of which 261 (85%) received a CAR T infusion (88% and 82%, respectively). Median time from apheresis to infusion was 41 days for axi-cel and 52 days for tisa-cel (P=0.006). None of the baseline characteristics were significantly different between both cohorts. Both cytokine release syndrome and neurologic events (NE) were more frequent in the axi-cel group (88% vs. 73%, P=0.003, and 42% vs. 16%, P<0.001, respectively). Infections in the first 6 months post-infusion were also more common in patients treated with axi-cel (38% vs. 25%, P=0.033). Non-relapse mortality was not significantly different between the axi-cel and tisa-cel groups (7% and 4%, respectively, P=0.298). With a median follow-up of 9.2 months, median PFS and OS were 5.9 and 3 months, and 13.9 and 11.2 months for axi-cel and tisa-cel, respectively. The 12-month PFS and OS for axi-cel and tisa-cel were 41% and 33% (P=0.195), 51% and 47% (P=0.191), respectively. Factors associated with lower OS in the multivariate analysis were increased lactate dehydrogenase, ECOG ≥2 and progressive disease before lymphodepletion. Safety and efficacy results in our real-world experience were comparable with those reported in the pivotal trials. Patients treated with axi-cel experienced more toxicity but similar non-relapse mortality compared with those receiving tisa-cel. Efficacy was not significantly different between both products.
Collapse
Affiliation(s)
- Mi Kwon
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid,MK and GI contributed equally as co-first authors
| | - Gloria Iacoboni
- Department of Hematology, Vall d’Hebron University Hospital, Experimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona,Department of Medicine, Universitat Autonoma de Barcelona, Bellaterra,MK and GI contributed equally as co-first authors
| | - Juan Luis Reguera
- Department of Hematology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Sevilla
| | - Lucía López Corral
- Department of Hematology, Hospital Clínico Universitario de Salamanca, IBSAL, Salamanca
| | - Rafael Hernani Morales
- Department of Hematology, Hospital Clínico Universitario de Valencia, Instituto de Investigación Sanitaria INCLIVA, Valencia
| | | | - Manuel Guerreiro
- Department of Hematology, Hospital Universitario y Politécnico La Fe, Valencia
| | | | | | | | - Alberto Mussetti
- Department of Hematology, Hospital Duran i Reynals, Instituto Catalán de Oncología, Barcelona
| | - Juan Manuel Sancho
- Department of Hematology, Hospital Universitari Germans Trias i Pujol, Instituto Catalán de Oncología, Josep Carreras Research Institute, Badalona
| | - Mariana Bastos-Oreiro
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid
| | - Eva Catala
- Department of Hematology, Vall d’Hebron University Hospital, Experimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona,Department of Medicine, Universitat Autonoma de Barcelona, Bellaterra
| | - Javier Delgado
- Department of Hematology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Sevilla
| | - Hugo Luzardo Henriquez
- Department of Hematology, Hospital Universitario de Gran Canaria Doctor Negrín, Las Palmas de Gran Canaria
| | - Jaime Sanz
- Department of Hematology, Hospital Universitario y Politécnico La Fe, Valencia
| | - María Calbacho
- Department of Hematology, Hospital Universitario 12 de Octubre, Madrid
| | - Rebeca Bailén
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid
| | - Cecilia Carpio
- Department of Hematology, Vall d’Hebron University Hospital, Experimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona,Department of Medicine, Universitat Autonoma de Barcelona, Bellaterra
| | - Jose Maria Ribera
- Department of Hematology, Hospital Universitari Germans Trias i Pujol, Instituto Catalán de Oncología, Josep Carreras Research Institute, Badalona
| | - Anna Sureda
- Department of Hematology, Hospital Duran i Reynals, Instituto Catalán de Oncología, Barcelona
| | - Javier Briones
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona
| | - Juan Carlos Hernandez-Boluda
- Department of Hematology, Hospital Clínico Universitario de Valencia, Instituto de Investigación Sanitaria INCLIVA, Valencia
| | | | - Jose Luis Diez Martin
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid,Instituto de Investigación Sanitaria Gregorio Marañón, Madrid,Universidad Complutense de Madrid, Madrid, Spain
| | - Alejandro Martín
- Department of Hematology, Hospital Clínico Universitario de Salamanca, IBSAL, Salamanca
| | - Pere Barba
- Department of Hematology, Vall d’Hebron University Hospital, Experimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona,Department of Medicine, Universitat Autonoma de Barcelona, Bellaterra
| |
Collapse
|
242
|
Zhang X, Zhu L, Zhang H, Chen S, Xiao Y. CAR-T Cell Therapy in Hematological Malignancies: Current Opportunities and Challenges. Front Immunol 2022; 13:927153. [PMID: 35757715 PMCID: PMC9226391 DOI: 10.3389/fimmu.2022.927153] [Citation(s) in RCA: 146] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 05/16/2022] [Indexed: 12/13/2022] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy represents a major breakthrough in cancer treatment, and it has achieved unprecedented success in hematological malignancies, especially in relapsed/refractory (R/R) B cell malignancies. At present, CD19 and BCMA are the most common targets in CAR-T cell therapy, and numerous novel therapeutic targets are being explored. However, the adverse events related to CAR-T cell therapy might be serious or even life-threatening, such as cytokine release syndrome (CRS), CAR-T-cell-related encephalopathy syndrome (CRES), infections, cytopenia, and CRS-related coagulopathy. In addition, due to antigen escape, the limited CAR-T cell persistence, and immunosuppressive tumor microenvironment, a considerable proportion of patients relapse after CAR-T cell therapy. Thus, in this review, we focus on the progress and challenges of CAR-T cell therapy in hematological malignancies, such as attractive therapeutic targets, CAR-T related toxicities, and resistance to CAR-T cell therapy, and provide some practical recommendations.
Collapse
Affiliation(s)
- Xiaomin Zhang
- Department of Hematology, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingling Zhu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Hui Zhang
- School of Medicine, Jishou University, Jishou, China
| | - Shanshan Chen
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yang Xiao
- Institute of Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Hematology, Shenzhen Qianhai Shekou Pilot Free Trade Zone Hospital, Shenzhen, China
| |
Collapse
|
243
|
Safety and feasibility of stem cell boost as a salvage therapy for severe hematotoxicity after CD19 CAR T-cell therapy. Blood Adv 2022; 6:4719-4725. [PMID: 35793454 DOI: 10.1182/bloodadvances.2022007776] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/15/2022] [Indexed: 11/20/2022] Open
Abstract
CD19 CAR T-cells represent a practice-changing treatment modality for advanced B-cell malignancies. However, refractory cytopenias have emerged as a potentially life-threatening complication that can persist long after lymphodepleting chemotherapy. Whether stem cell rescue is feasible and efficacious after CAR-T has not been addressed. In this retrospective multi-center study, we describe clinical characteristics and outcomes of 13 patients with hyporegenerative bone marrow (BM) failure after CD19 CAR-T, which received a previously collected stem cell graft (10 autologous, 3 allogeneic) to rescue cytopenias. Interestingly, patients already presented with impaired hematopoietic function and high levels of systemic inflammation prior to lymphodepleting chemotherapy, as reflected by high CAR-HEMATOTOX scores (median 4). The median duration of severe neutropenia prior to stem cell boost was 41 days (interquartile range 16-50). The indication for boost was severe pancytopenia (aplasia) in 7 cases and persistent isolated neutropenia/thrombocytopenia in 6 cases. Median day of stem cell boost was day 55 after CAR-T (median 3.1x106/kg CD34+ cells). Engraftment rates were high (neutrophil: 92%, platelet: 70%), with a median time to neutrophil- and platelet engraftment of 15 and 21 days, respectively. Two patients died of invasive fungal infections (day 4 and 17 after stem cell boost). The 1-year progression-free (PFS) and overall survival (OS) rates were 42% and 51%, respectively. These data indicate that the transplantation of available stem cell products for post-CAR-T cytopenias is clinically feasible, safe and efficacious. Further studies are needed to assess, whether a pre-emptive collection of stem cells can be justified in selected high-risk patients.
Collapse
|
244
|
Du M, Huang L, Kou H, Li C, Hu Y, Mei H. Case Report: ITP Treatment After CAR-T Cell Therapy in Patients With Multiple Myeloma. Front Immunol 2022; 13:898341. [PMID: 35784357 PMCID: PMC9244693 DOI: 10.3389/fimmu.2022.898341] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/11/2022] [Indexed: 11/26/2022] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy is an attractive strategy for patients with relapsed or refractory hematological malignancies including multiple myeloma (MM). T cells are engineered to attack malignant cells that express tumor-associated antigens and better efficacy could be achieved. However, cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and hematologic toxicity are still challenges for CAR-T cell therapy. Among them, hematologic toxicity including thrombocytopenia has a longer duration and lasting effect during and after the treatment for some patients. Here, we present 3 cases of hematologic toxicity manifested as refractory thrombocytopenia with platelet autoantibodies positive and plasma thrombopoietin (TPO) concentration elevated after bispecific CAR-T cell therapy in relapsed/refractory (R/R) MM patients who were successfully treated with standard therapy of immune thrombocytopenia (ITP). Without clear pathogenesis or guidance on therapy published, our cases provide a reference for the treatment of thrombocytopenia after CAR-T cell therapy and inspire exploration of the underlying pathophysiological mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Yu Hu
- *Correspondence: Heng Mei, ; Yu Hu,
| | - Heng Mei
- *Correspondence: Heng Mei, ; Yu Hu,
| |
Collapse
|
245
|
Hematopoietic stem cell boost for persistent neutropenia after CAR T-cell therapy: a GLA/DRST study. Blood Adv 2022; 7:555-559. [PMID: 35696759 PMCID: PMC9984300 DOI: 10.1182/bloodadvances.2022008042] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 11/20/2022] Open
Abstract
Hematotoxicity after chimeric antigen receptor (CAR) T-cell therapy is associated with infection and death but management remains unclear. We report results of 31 patients receiving hematopoietic stem cell boost (HSCB; 30 autologous, 1 allogeneic) for either sustained severe neutropenia of grade 4 (<0.5 × 109/L), sustained moderate neutropenia (≤1.5 × 109/L) and high risk of infection, or neutrophil count ≤2.0 × 109/L and active infection. Median time from CAR T-cell therapy to HSCB was 43 days and median absolute neutrophil count at time of HSCB was 0.2. Median duration of neutropenia before HSCB was 38 days (range, 7-151). Overall neutrophil response rate (recovery or improvement) was observed in 26 patients (84%) within a median of 9 days (95% confidence interval, 7-14). Time to response was significantly associated with the duration of prior neutropenia (P = .007). All nonresponders died within the first year after HSCB. One-year overall survival for all patients was 59% and significantly different for neutropenia (≤38 days; 85%) vs neutropenia >38 days before HSCB (44%; P = .029). In conclusion, early or prophylactic HSCB showed quick response and improved outcomes for sustained moderate to severe neutropenia after CAR-T.
Collapse
|
246
|
Rejeski K, Perez A, Iacoboni G, Penack O, Bücklein V, Jentzsch L, Mougiakakos D, Johnson G, Arciola B, Carpio C, Blumenberg V, Hoster E, Bullinger L, Locke FL, von Bergwelt-Baildon M, Mackensen A, Bethge W, Barba P, Jain MD, Subklewe M. The CAR-HEMATOTOX risk-stratifies patients for severe infections and disease progression after CD19 CAR-T in R/R LBCL. J Immunother Cancer 2022; 10:jitc-2021-004475. [PMID: 35580927 PMCID: PMC9114843 DOI: 10.1136/jitc-2021-004475] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2022] [Indexed: 01/18/2023] Open
Abstract
Background CD19-directed chimeric antigen receptor T-cell therapy (CAR-T) represents a promising treatment modality for an increasing number of B-cell malignancies. However, prolonged cytopenias and infections substantially contribute to the toxicity burden of CAR-T. The recently developed CAR-HEMATOTOX (HT) score—composed of five pre-lymphodepletion variables (eg, absolute neutrophil count, platelet count, hemoglobin, C-reactive protein, ferritin)—enables risk stratification of hematological toxicity. Methods In this multicenter retrospective analysis, we characterized early infection events (days 0–90) and clinical outcomes in 248 patients receiving standard-of-care CD19 CAR-T for relapsed/refractory large B-cell lymphoma. This included a derivation cohort (cohort A, 179 patients) and a second independent validation cohort (cohort B, 69 patients). Cumulative incidence curves were calculated for all-grade, grade ≥3, and specific infection subtypes. Clinical outcomes were studied via Kaplan-Meier estimates. Results In a multivariate analysis adjusted for other baseline features, the HT score identified patients at high risk for severe infections (adjusted HR 6.4, 95% CI 3.1 to 13.1). HThigh patients more frequently developed severe infections (40% vs 8%, p<0.0001)—particularly severe bacterial infections (27% vs 0.9%, p<0.0001). Additionally, multivariate analysis of post-CAR-T factors revealed that infection risk was increased by prolonged neutropenia (≥14 days) and corticosteroid use (≥9 days), and decreased with fluoroquinolone prophylaxis. Antibacterial prophylaxis significantly reduced the likelihood of severe bacterial infections in HThigh (16% vs 46%, p<0.001), but not HTlow patients (0% vs 2%, p=n.s.). Collectively, HThigh patients experienced worse median progression-free (3.4 vs 12.6 months) and overall survival (9.1 months vs not-reached), and were hospitalized longer (median 20 vs 16 days). Severe infections represented the most common cause of non-relapse mortality after CAR-T and were associated with poor survival outcomes. A trend toward increased non-relapse mortality in HThigh patients was observed (8.0% vs 3.7%, p=0.09). Conclusions These data demonstrate the utility of the HT score to risk-stratify patients for infectious complications and poor survival outcomes prior to CD19 CAR-T. High-risk patients likely benefit from anti-infective prophylaxis and should be closely monitored for potential infections and relapse.
Collapse
Affiliation(s)
- Kai Rejeski
- Department of Medicine III, Hematology and Oncology, University Hospital, LMU Munich, Munich, Germany.,Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany.,German Cancer Consortium (DKTK) Munich Site, and German Cancer Research Center, Heidelberg, Germany
| | - Ariel Perez
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida, USA.,Blood & Marrow Transplant Program, Miami Cancer Institute, Miami, Florida, USA
| | - Gloria Iacoboni
- Department of Hematology, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, Universitat Autònoma of Barcelona (UAB), Department of Medicin, Barcelona, Spain
| | - Olaf Penack
- Department of Hematology, Oncology and Tumorimmunology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Cancer Consortium (DKTK) Berlin Site, and German Cancer Research Center, Heidelberg, Germany
| | - Veit Bücklein
- Department of Medicine III, Hematology and Oncology, University Hospital, LMU Munich, Munich, Germany.,Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Liv Jentzsch
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Dimitrios Mougiakakos
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Grace Johnson
- USF Morsani College of Medicine, Tampa, Florida, USA
| | - Brian Arciola
- USF Morsani College of Medicine, Tampa, Florida, USA
| | - Cecilia Carpio
- Department of Hematology, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, Universitat Autònoma of Barcelona (UAB), Department of Medicin, Barcelona, Spain
| | - Viktoria Blumenberg
- Department of Medicine III, Hematology and Oncology, University Hospital, LMU Munich, Munich, Germany.,Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Eva Hoster
- Institute for Medical Information Processing, Biometry and Epidemiology (IBE), LMU Munich, Munich, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology and Tumorimmunology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Cancer Consortium (DKTK) Berlin Site, and German Cancer Research Center, Heidelberg, Germany
| | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida, USA
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, Hematology and Oncology, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium (DKTK) Munich Site, and German Cancer Research Center, Heidelberg, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Wolfgang Bethge
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Pere Barba
- Department of Hematology, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, Universitat Autònoma of Barcelona (UAB), Department of Medicin, Barcelona, Spain
| | - Michael D Jain
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida, USA
| | - Marion Subklewe
- Department of Medicine III, Hematology and Oncology, University Hospital, LMU Munich, Munich, Germany .,Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany.,German Cancer Consortium (DKTK) Munich Site, and German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
247
|
Kuhnl A, Roddie C, Kirkwood AA, Tholouli E, Menne T, Patel A, Besley C, Chaganti S, Sanderson R, O'Reilly M, Norman J, Osborne W, Bloor A, Lugthart S, Malladi R, Patten PEM, Neill L, Martinez-Cibrian N, Kennedy H, Phillips EH, Jones C, Sharplin K, El-Sharkawi D, Latif AL, Mathew A, Uttenthal B, Stewart O, Marzolini MAV, Townsend W, Cwynarski K, Ardeshna K, Ardavan A, Robinson K, Pagliuca A, Collins GP, Johnson R, McMillan A. A national service for delivering CD19 CAR-Tin large B-cell lymphoma - The UK real-world experience. Br J Haematol 2022; 198:492-502. [PMID: 35485402 DOI: 10.1111/bjh.18209] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/22/2022] [Accepted: 04/08/2022] [Indexed: 01/01/2023]
Abstract
CD19 CAR-T have emerged as a new standard treatment for relapsed/refractory (r/r) large B-cell lymphoma (LBCL). CAR-T real-world (RW) outcomes published to date suggest significant variability across countries. We provide results of a large national cohort of patients intended to be treated with CAR-T in the UK. Consecutive patients with r/r LBCL approved for CAR-T by the National CAR-T Clinical Panel between December 2018 and November 2020 across all UK CAR-T centres were included. 404/432 patients were approved [292 axicabtagene ciloleucel (axi-cel), 112 tisagenlecleucel (tisa-cel)], 300 (74%) received the cells. 110/300 (38.3%) patients achieved complete remission (CR) at 6 months (m). The overall response rate was 77% (52% CR) for axi-cel, 57% (44% CR) for tisa-cel. The 12-month progression-free survival was 41.8% (axi-cel) and 27.4% (tisa-cel). Median overall survival for the intention-to-treat population was 10.5 m, 16.2 m for infused patients. The incidence of grade ≥3 cytokine release syndrome and neurotoxicity were 7.6%/19.6% for axi-cel and 7.9%/3.9% for tisa-cel. This prospective RW population of CAR-T eligible patients offers important insights into the clinical benefit of CD19 CAR-T in LBCL in daily practice. Our results confirm long-term efficacy in patients receiving treatment similar to the pivotal trials, but highlight the significance of early CAR-T failure.
Collapse
Affiliation(s)
- Andrea Kuhnl
- Department of Haematology, King's College Hospital, London, UK
| | - Claire Roddie
- Department of Haematology, University College London Hospitals, London, UK.,UCL Cancer Institute, University College London, London, UK
| | - Amy A Kirkwood
- Cancer Research UK & UCL Cancer Trials Centre, UCL Cancer Institute, University College London, London, UK
| | - Eleni Tholouli
- Department of Haematology, Manchester Royal Infirmary, Manchester, UK
| | - Tobias Menne
- Department of Haematology, Freeman Hospital, Newcastle, UK
| | - Amit Patel
- Department of Haematology, The Christie Hospital, Manchester, UK
| | - Caroline Besley
- Department of Haematology, University Hospitals Bristol and Weston, Bristol, UK
| | - Sridhar Chaganti
- Department of Haematology, Queen Elizabeth Hospital, Birmingham, UK
| | - Robin Sanderson
- Department of Haematology, King's College Hospital, London, UK
| | - Maeve O'Reilly
- Department of Haematology, University College London Hospitals, London, UK
| | - Jane Norman
- Department of Haematology, Manchester Royal Infirmary, Manchester, UK
| | - Wendy Osborne
- Department of Haematology, Freeman Hospital, Newcastle, UK
| | - Adrian Bloor
- Department of Haematology, The Christie Hospital, Manchester, UK
| | - Sanne Lugthart
- Department of Haematology, University Hospitals Bristol and Weston, Bristol, UK
| | - Ram Malladi
- Department of Haematology, Queen Elizabeth Hospital, Birmingham, UK.,Department of Haematology, Addenbrookes Hospital, Cambridge, UK
| | - Piers E M Patten
- Department of Haematology, King's College Hospital, London, UK.,Comprehensive Cancer Centre, King's College London, London, UK
| | - Lorna Neill
- Department of Haematology, University College London Hospitals, London, UK
| | | | - Hannah Kennedy
- Department of Haematology, Freeman Hospital, Newcastle, UK
| | - Elizabeth H Phillips
- Department of Medical Oncology, The Christie Hospital, Manchester, UK.,Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Ceri Jones
- Department of Haematology, University Hospital of Wales, Cardiff, UK
| | - Kirsty Sharplin
- Department of Haematology, University Hospitals Bristol and Weston, Bristol, UK
| | | | | | - Amrith Mathew
- Department of Haematology, Queen Elizabeth Hospital, Birmingham, UK
| | | | - Orla Stewart
- Department of Haematology, King's College Hospital, London, UK
| | | | - William Townsend
- Department of Haematology, University College London Hospitals, London, UK
| | - Kate Cwynarski
- Department of Haematology, University College London Hospitals, London, UK
| | - Kirit Ardeshna
- Department of Haematology, University College London Hospitals, London, UK
| | - Arzhang Ardavan
- NCRI Consumer Forum, London, UK.,Department of Physics, University of Oxford, UK
| | | | | | - Graham P Collins
- Department of Haematology, Oxford University Hospital, Oxford, UK
| | | | - Andrew McMillan
- Department of Haematology, Nottingham University Hospitals, Nottingham, UK
| |
Collapse
|
248
|
Kampouri E, Walti CS, Gauthier J, Hill JA. Managing Hypogammaglobulinemia in Patients Treated with CAR-T-cell Therapy: Key Points for Clinicians. Expert Rev Hematol 2022; 15:305-320. [PMID: 35385358 DOI: 10.1080/17474086.2022.2063833] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The unprecedented success of chimeric antigen receptor (CAR)-T-cell therapy in the management of B-cell malignancies comes with a price of specific side effects. Healthy B-cell depletion is an anticipated 'on-target' 'off-tumor' side effect and can contribute to severe and prolonged hypogammaglobulinemia. Evidence-based guidelines for the use of immunoglobulin replacement therapy (IGRT) for infection prevention are lacking in this population. AREAS COVERED This article reviews the mechanisms and epidemiology of hypogammaglobulinemia and antibody deficiency, association with infections, and strategies to address these issues in CD19- and BCMA-CAR-T-cell recipients. EXPERT OPINION CD19 and BCMA CAR-T-cell therapy result in unique immune deficits due to depletion of specific B-lineage cells and may require different infection prevention strategies. Hypogammaglobulinemia before and after CAR-T-cell therapy is frequent, but data on the efficacy and cost-effectiveness of IGRT are lacking. Monthly IGRT should be prioritized for patients with severe or recurrent bacterial infections. IGRT may be more broadly necessary to prevent infections in BCMA-CAR-T-cell recipients and children with severe hypogammaglobulinemia irrespective of infection history. Vaccinations are indicated to augment humoral immunity and can be immunogenic despite cytopenias; re-vaccination(s) may be required. Controlled trials are needed to better understand the role of IGRT and vaccines in this population.
Collapse
Affiliation(s)
- Eleftheria Kampouri
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Infectious Diseases Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Carla S Walti
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Infectious Disease and Hospital Epidemiology Division, University Hospital Basel, Basel, Switzerland
| | - Jordan Gauthier
- Department of Medicine, University of Washington, Seattle, Washington, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Joshua A Hill
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
249
|
ABCs of ADCs in Management of Relapsed/refractory diffuse large B-cell lymphoma. Blood Rev 2022; 56:100967. [DOI: 10.1016/j.blre.2022.100967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 11/19/2022]
|
250
|
Sharma N, Reagan PM, Liesveld JL. Cytopenia after CAR-T Cell Therapy-A Brief Review of a Complex Problem. Cancers (Basel) 2022; 14:1501. [PMID: 35326654 PMCID: PMC8946106 DOI: 10.3390/cancers14061501] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/02/2022] [Accepted: 03/07/2022] [Indexed: 02/06/2023] Open
Abstract
Chimeric Antigen Receptor T-cell (CAR-T) immunotherapy has emerged as an efficacious and life extending treatment modality with high response rates and durable remissions in patients with relapsed and refractory non-Hodgkin lymphoma (NHL), follicular lymphoma, and B-cell acute lymphoblastic leukemia (B-ALL) as well as in other diseases. Prolonged or recurrent cytopenias after CAR-T therapy have increasingly been reported at varying rates, and the pathogenesis of this complication is not yet well-understood but is likely contributed to by multiple factors. Current studies reported are primarily retrospective, heterogeneous in terms of CAR-Ts used and diseases treated, non-uniform in definitions of cytopenias and durations for end points, and vary in terms of recommended management. Prospective studies and correlative laboratory studies investigating the pathophysiology of prolonged cytopenias will enhance our understanding of this phenomenon. This review summarizes knowledge of these cytopenias to date.
Collapse
Affiliation(s)
- Naman Sharma
- Department of Hematology-Oncology, Baystate Medical Center, University of Massachusetts Medical School, Springfield, MA 100107, USA;
| | - Patrick M. Reagan
- Department of Medicine, Hematology-Oncology, James P. Wilmot Cancer Institute, University of Rochester, Rochester, NY 14642, USA;
| | - Jane L. Liesveld
- Department of Medicine, Hematology-Oncology, James P. Wilmot Cancer Institute, University of Rochester, Rochester, NY 14642, USA;
| |
Collapse
|