1
|
Mikulska M, van Bömmel F, Mouliade C, Indolfi G, Kefalakes H, von Lilienfeld-Toal M, Pischke S, Hermine O, Moradpour D, Wedemeyer H, Berg T, Ljungman P, Mallet V. Updated recommendations for the management of hepatitis B, C, and E virus infections in patients with haematological malignancies and those undergoing haematopoietic cell transplantation: recommendations from the 9th European Conference on Infections in Leukaemia (ECIL-9). Lancet Haematol 2025; 12:e389-e399. [PMID: 40306834 DOI: 10.1016/s2352-3026(25)00049-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/09/2025] [Accepted: 02/14/2025] [Indexed: 05/02/2025]
Abstract
Viral hepatitis remains a global health challenge and immune status affects outcomes. In patients with haematological malignancies, including haematopoietic stem-cell transplantation recipients, viral hepatitis can be life-threatening due to the direct effects of the virus or the need to modify or delay chemotherapy. Additionally, haematopoietic stem-cell donors with past or current viral hepatitis infections might transmit the virus to recipients. The growing recognition of hepatitis E virus (HEV), advances in haematological therapies, and the availability of direct-acting antivirals for hepatitis C virus (HCV), led the 2022 9th European Conference on Infections in Leukaemia (ECIL-9) to update the 2013 ECIL-5 guidelines on viral hepatitis. The ECIL organising committee convened a panel of 13 impartial international experts (all authors of this Review) in viral hepatitis, both within and outside the fields of haematological malignancies and immunosuppression. The ECIL-9 panel conducted a review of the literature on hepatitis B virus (HBV), HCV, and HEV, grading the evidence based on the European Society for Clinical Microbiology and Infectious Diseases system. The panel identified key clinical questions and outcomes and built on the recommendations established during ECIL-5. A consensus conference was held in Sofia Antipolis, France, from Sept 15-17, 2022, bringing together 49 experts from 19 countries. The ECIL-9 panel presented the proposed recommendations, which were revised following expert discussions. A final consensus on updated guidelines was reached in a second plenary session. The updated ECIL-9 guidelines provide evidence-based recommendations on the prevention, screening, treatment, and long-term surveillance of viral hepatitis in patients with haematological malignancies and haematopoietic cell transplantation recipients.
Collapse
Affiliation(s)
- Malgorzata Mikulska
- Department of Health Sciences, Division of Infectious Diseases, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Florian van Bömmel
- Laboratory for Clinical and Experimental Hepatology, Department of Medicine II, Leipzig University Medical Center, Leipzig, Germany; Division of Hepatology, Department of Medicine II, Leipzig University Medical Center, Leipzig, Germany; University Liver Tumor Center, Leipzig University Medical Center, Leipzig, Germany
| | - Charlotte Mouliade
- Université Paris Cité, Paris, France; AP-HP Centre, Groupe Hospitalier Cochin Port Royal, DMU Cancérologie et spécialités médico-chirurgicales, Service d'Hépatologie, Paris, France
| | | | - Helenie Kefalakes
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Marie von Lilienfeld-Toal
- Institut für Diversitätsmedizin, Ruhr-Universität Bochum, Bochum, Germany; Hämatologie, Onkologie, Stammzelltransplantation und Zelltherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, Bochum, Germany; Department of Haematology, Oncology and Palliative Care, St Josef Hospital, Ruhr University, Bochum, Germany
| | - Sven Pischke
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Olivier Hermine
- Université Paris Cité, Paris, France; Department of Haematology, Necker Hospital, Assistance Publique Hôpitaux de Paris, Paris, France; Laboratory of Physiopathology of Haematological Disorders and their Treatment, Imagine Institute INSERM U 1163, Paris, France
| | - Darius Moradpour
- Division of Gastroenterology and Hepatology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Thomas Berg
- Division of Hepatology, Department of Medicine II, Leipzig University Medical Center, Leipzig, Germany
| | - Per Ljungman
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University Hospital Huddinge, Karolinska Comprehensive Cancer Center, Stockholm, Sweden; Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Vincent Mallet
- Université Paris Cité, Paris, France; AP-HP Centre, Groupe Hospitalier Cochin Port Royal, DMU Cancérologie et spécialités médico-chirurgicales, Service d'Hépatologie, Paris, France.
| |
Collapse
|
2
|
Deuse T, Schrepfer S. Progress and challenges in developing allogeneic cell therapies. Cell Stem Cell 2025; 32:513-528. [PMID: 40185072 DOI: 10.1016/j.stem.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 04/07/2025]
Abstract
The new era of cell therapeutics has started with autologous products to avoid immune rejection. However, therapeutics derived from allogeneic cells could be scaled and made available for a much larger patient population if immune rejection could reliably be overcome. In this review, we outline gene engineering concepts aimed at generating immune-evasive cells. First, we summarize the current state of allogeneic immune cell therapies, and second, we compile the still limited data for allogeneic cell replacement therapies. We emphasize the advances in this fast-developing field and provide an optimistic outlook for future allogeneic cell therapies.
Collapse
Affiliation(s)
- Tobias Deuse
- Department of Surgery, Division of Cardiothoracic Surgery, Transplant and Stem Cell Immunobiology (TSI)-Lab, University of California, San Francisco, San Francisco, CA, USA
| | - Sonja Schrepfer
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
3
|
Luan D, DeWolf S, Fei T, Raj S, Shah GL, Lareau CA, Alhomoud M, Salles G, Rivas-Delgado A, Rejeski K, Park JH, Luttwak E, Luna de Abia A, Corona M, Ntrivalas E, Cassanello G, Gomez-Llobell M, Parascondola A, Scordo M, Hsu KC, Palomba ML, Perales MA, Shouval R. Dynamics of Immune Reconstitution and Impact on Outcomes across CAR-T Cell Products in Large B-cell Lymphoma. Blood Cancer Discov 2025; 6:119-130. [PMID: 39666878 PMCID: PMC11876948 DOI: 10.1158/2643-3230.bcd-24-0163] [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: 07/02/2024] [Revised: 10/09/2024] [Accepted: 12/11/2024] [Indexed: 12/14/2024] Open
Abstract
SIGNIFICANCE This study reveals differences in IR patterns after CAR-T therapy in patients with large B-cell lymphoma, with early NK cell recovery emerging as a key predictor of survival. These findings provide potential future avenues of research for improving patient outcomes and tailoring post-therapy management strategies to mitigate relapse risk.
Collapse
Affiliation(s)
- Danny Luan
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Susan DeWolf
- Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Teng Fei
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sandeep Raj
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gunjan L. Shah
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Caleb A. Lareau
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mohammad Alhomoud
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gilles Salles
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Kai Rejeski
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine III – Hematology/Oncology, LMU University Hospital, Munich, Germany
| | - Jae H. Park
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Efrat Luttwak
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alejandro Luna de Abia
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Adult Bone Marrow Transplantation Service, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Magdalena Corona
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Evangelos Ntrivalas
- Hematopathology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Giulio Cassanello
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Marina Gomez-Llobell
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Allison Parascondola
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael Scordo
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Katharine C. Hsu
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - M. Lia Palomba
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Miguel-Angel Perales
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Roni Shouval
- Department of Medicine, Weill Cornell Medicine, New York, New York
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
4
|
Mir M, Faiz S, Bommakanti AG, Sheshadri A. Pulmonary Immunocompromise in Stem Cell Transplantation and Cellular Therapy. Clin Chest Med 2025; 46:129-147. [PMID: 39890284 DOI: 10.1016/j.ccm.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2025]
Abstract
Hematopoietic cell transplantation (HCT) and cellular therapies, such as chimeric-antigen receptor T-cell (CAR-T) treatments, are potentially curative treatments for certain hematologic malignancies and some nonmalignant disorders. However, pulmonary complications, both infectious and noninfectious, remain a significant cause of morbidity and mortality in patients who receive cellular therapies. This review article provides an overview of pulmonary complications encountered in the context of HCT and CAR-T. The authors discuss mechanisms of underlying immunocompromise that lead to a rise in infections. Additionally, they highlight key noninfectious complications of HCT that can mimic acute infections and suggest diagnostic approaches and preventive strategies to distinguish these entities promptly.
Collapse
Affiliation(s)
- Mahnoor Mir
- Divisions of Critical Care, Pulmonary and Sleep Medicine, McGovern Medical School at UTHealth, Houston, TX 77030, USA; Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Saadia Faiz
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anuradha G Bommakanti
- Divisions of Critical Care, Pulmonary and Sleep Medicine, McGovern Medical School at UTHealth, Houston, TX 77030, USA; Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ajay Sheshadri
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
5
|
Mallet V, Torres HA. Hepatitis E virus infection after CAR T-cell treatment: An important complication in patients already facing significant health challenges. Br J Haematol 2025; 206:1020-1021. [PMID: 39622628 DOI: 10.1111/bjh.19931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 03/08/2025]
Abstract
Cancer patients with haematological malignancies are at risk for chronic hepatitis E virus infection following chimeric antigen receptor (CAR) T-cell therapy. Strong clinical suspicion is essential for the early diagnosis and prompt treatment of this difficult-to-treat type of viral hepatitis. Commentary on: Schwarz et al. Chronic hepatitis E in a patient after CAR-T cell treatment for diffuse large B-cell lymphoma and rapid progression towards decompensated liver cirrhosis. Br J Haematol 2025; 206:977-980.
Collapse
Affiliation(s)
- Vincent Mallet
- Service Hépatologie, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris Université Paris Cité, Paris, France
| | - Harrys A Torres
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
6
|
Sassine J, Agudelo Higuita NI, Siegrist EA, Saeedi A, Corbisiero MF, Connelly P, Bastias AG, Dib RW, Henao-Cordero J, Chastain DB, Chiu CY, Henao-Martínez AF. Timeline and outcomes of viral and fungal infections after chimeric antigen receptor T-cell therapy: a large database analysis. Clin Microbiol Infect 2025; 31:466-472. [PMID: 39528086 DOI: 10.1016/j.cmi.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES This large database analysis aims to describe the incidence, timeline, and risk factors for viral and fungal infections after chimeric antigen receptor (CAR) T-cell therapy. METHODS We queried a global research network database, TriNetX, for patients who received CAR T-cell therapy, who were identified and followed for the development of viral and fungal infections. Baseline demographic, oncologic history, laboratory data and medication histories were collected. We evaluated risk factors for respiratory viral infections (RVIs), herpesvirus, fungal infections and mortality using Cox regression. RESULTS A total of 2256 patients who received CAR T-cell therapy were included, 1867 (82.7%) were CD19-targeted and 400 (17.7%) were B-cell maturation antigen-targeted. After CAR T-cell infusion, RVIs were the most prevalent (23.3%) with a median onset of 160 days (interquartile range [IQR]: 52-348 days), whereas herpesvirus and fungal infections were less frequent, occurring in 13.6% and 11.4% of cases with median onsets of 71 (IQR, 18-252) and 73 days (IQR, 14-236 days), respectively. On multivariable Cox regression, independent predictors of RVI included acute lymphoblastic leukaemia (hazard ratio [HR], 1.61), prior haematopoietic cell transplant (HCT; HR, 1.29), cytokine release syndrome (HR, 1.41), hemophagocytic lymphohistiocytosis (HR, 1.96) and glucocorticoids (HR, 3.37). Prior HCT (HR, 2.00), hypogammaglobulinemia (HR, 1.51), immune effector cell-associated neurotoxicity syndrome (HR, 1.52) and hemophagocytic lymphohistiocytosis (HR, 1.99) were associated with a higher risk of herpesviruses. Independent predictors of fungal infections included prior HCT (HR, 1.59), cytokine release syndrome (HR, 1.58) and hypogammaglobulinemia (HR, 1.40). Idecabtagene vicleucel was associated with a lower risk of herpesvirus and fungal infections (HR, 0.39 and 0.44, respectively). DISCUSSION In a large cohort of CAR T-cell therapy recipients, RVIs were the most common but occurred later, whereas herpesvirus and fungal infections were less frequent but occurred earlier. Prospective studies investigating prophylaxis and pre-emptive monitoring strategies are needed in this population.
Collapse
Affiliation(s)
- Joseph Sassine
- Infectious Diseases Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Nelson Iván Agudelo Higuita
- Infectious Diseases Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Instituto de Enfermedades Infecciosas y Parasitología Antonio Vidal, Tegucigalpa, Honduras
| | | | - Arman Saeedi
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Patrick Connelly
- Department of Computer Science, University of Colorado at Boulder, Boulder, CO, USA
| | - Alfonso G Bastias
- Department of Computer Science, University of Colorado at Boulder, Boulder, CO, USA
| | - Rita Wilson Dib
- Infectious Diseases Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - José Henao-Cordero
- Infectious Diseases Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Chia-Yu Chiu
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | |
Collapse
|
7
|
Umair MM, Lai X, Xue Y, Yao H. Influence of CAR T-cell therapy associated complications. Front Oncol 2025; 15:1494986. [PMID: 40052127 PMCID: PMC11882432 DOI: 10.3389/fonc.2025.1494986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/21/2025] [Indexed: 03/09/2025] Open
Abstract
Since the introduction of chimeric antigen receptor (CAR) T-cell therapy, it has elicited an immense response in both targeted and residual cancers. Its clinical efficacy is often accompanied by a group of side effects that may become serious because of factors such as tumor burden, the extent of lymphodepletion, and the type of co-stimulus. It is also crucial to know the common toxicities associated with CAR T-cell therapy, including cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), cardiotoxicity, metabolic disorders, pulmonary toxicity, macrophage activation syndrome (MAS), prolonged cytopenia, coagulation disorders, and potential off-target effects on various organs. If not well managed, these can be fatal. However, knowledge about molecular pathways, calcineurin inhibitors, IL-6 receptor antagonists, steroids, suppression of nitric oxide synthase, various therapeutic approaches, and other recent advances have been developed to mitigate the fatal results of various short-term and chronic adverse events related to CAR T-cell therapy. This study provides a comprehensive perspective on contemporary management strategies and presumed causative processes of CAR T-cell-related adverse effects, albeit with several limitations. When CAR T-cell complications, costs, and challenges of toxicity management are properly considered, the CAR T-cell therapy of the future will include a number of toxicity-escaping options.
Collapse
Affiliation(s)
- Mohammad Mussab Umair
- Cancer Biotherapy Center & Cancer Research Institute, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xun Lai
- Department of Hematology, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - YuanBo Xue
- Cancer Biotherapy Center, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hong Yao
- Cancer Biotherapy Center & Cancer Research Institute, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
8
|
Ow KV. CAR T-Cell Therapy Unveiled: Navigating Beyond CRS and ICANS to Address Delayed Complications and Optimize Management Strategies. J Adv Pract Oncol 2025; 16:1-15. [PMID: 39990042 PMCID: PMC11840332 DOI: 10.6004/jadpro.2025.16.7.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has ushered in a transformative era in the management of relapsed/refractory hematologic malignancies. The extensive phase II trials targeting relapsed/refractory non-Hodgkin lymphoma, including diverse subtypes such as diffuse large B-cell lymphoma, follicular lymphoma, and mantle cell lymphoma, along with multiple myeloma and B-cell acute lymphoblastic leukemia, have culminated in the endorsement of various CAR T-cell products for these specific indications by the US Food and Drug Administration. Although CAR T-cell therapy has achieved remarkable success, it is important to recognize that this innovative approach often gives rise to notable toxicities and is frequently associated with a distinctive pattern of adverse effects. Advanced practice providers, including advanced practice nurses and physician associates, involved in the care of these patients should be able to recognize these toxicities and be versed in treatment strategies to mitigate their impact.
Collapse
Affiliation(s)
- Karla V Ow
- From The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
9
|
Sassine J, Siegrist EA, Chemaly RF. Herpesvirus Infections After Chimeric Antigen Receptor T-Cell Therapy and Bispecific Antibodies: A Review. Viruses 2025; 17:133. [PMID: 39861922 PMCID: PMC11768728 DOI: 10.3390/v17010133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
In this narrative review, we explore the burden and risk factors of various herpesvirus infections in patients receiving chimeric antigen receptor T-cell (CAR-T) therapy or bispecific antibodies (BsAb) for the treatment of hematologic malignancies. Antiviral prophylaxis for herpes simplex/varicella zoster viruses became part of the standard of care in this patient population. Breakthrough infections may rarely occur, and the optimal duration of prophylaxis as well as the timing of recombinant zoster immunization remain to be explored. Clinically significant cytomegalovirus (CMV) infections can affect up to 10% of patients after CAR-T, depending on the CAR-T product target, post-CAR-T complications such as cytokine release syndrome and the need for glucocorticoid therapy. Surveillance and prophylactic strategies for CMV need to be developed, whereas the risk factors for and the burden of CMV infections after BsAb are not yet well-defined. Human herpes virus 6 reactivation and end organ disease such as encephalitis are rarely reported after CAR-T and have not yet been reported after BsAb; additional research is needed.
Collapse
Affiliation(s)
- Joseph Sassine
- Infectious Diseases Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | - Roy F. Chemaly
- Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| |
Collapse
|
10
|
Stock S, Bücklein VL, Blumenberg V, Magno G, Emhardt A, Holzem AME, Cordas dos Santos DM, Schmidt C, Grießhammer S, Frölich L, Kobold S, von Bergwelt‐Baildon M, Rejeski K, Subklewe M. Prognostic significance of immune reconstitution following CD19 CAR T-cell therapy for relapsed/refractory B-cell lymphoma. Hemasphere 2025; 9:e70062. [PMID: 39807276 PMCID: PMC11726691 DOI: 10.1002/hem3.70062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/26/2024] [Accepted: 10/18/2024] [Indexed: 01/16/2025] Open
Abstract
Immune deficits after CD19 chimeric antigen receptor (CAR) T-cell therapy can be long-lasting, predisposing patients to infections and non-relapse mortality. In B-cell non-Hodgkin lymphoma (B-NHL), the prognostic impact of immune reconstitution (IR) remains ill-defined, and detailed cross-product comparisons have not been performed to date. In this retrospective observational study, we longitudinally characterized lymphocyte subsets and immunoglobulin levels in 105 B-NHL patients to assess patterns of immune recovery arising after CD19 CAR-T. Three key IR criteria were defined as CD4+ T helper (TH) cells > 200/µL, any detectable B cells, and serum immunoglobulin G (IgG) levels >4 g/L. After a median follow-up of 24.6 months, 38% of patients displayed TH cells, 11% showed any B cells, and 41% had IgG recovery. Notable product-specific differences emerged, including deeper TH cell aplasia with CD28z- versus longer B-cell aplasia with 41BBz-based products. Patients with any IR recovery experienced extended progression-free survival (PFS) (median 20.8 vs. 1.7 months, p < 0.0001) and overall survival (OS) (34.9 vs. 4.0 months, p < 0.0001). While landmark analysis at 90 days confirmed improved PFS in patients with any recovery (34.9 vs. 8.6 months, p = 0.005), no significant OS difference was noted. Notably, 72% of patients with refractory disease never displayed recovery of any IR criteria. Early progressors showed diminished IR at the time of progression/relapse compared to patients with late progression/recurrence (after Day 90). Our results highlight the profound immune deficits observed after CD19 CAR-T and shed light on the intersection of IR and efficacy in B-NHL. Importantly, IR was impaired considerably postprogression, carrying significant implications for subsequent T-cell-engaging therapies and treatment sequencing.
Collapse
Affiliation(s)
- Sophia Stock
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- Division of Clinical Pharmacology, Department of Medicine IVLMU University Hospital, LMU MunichMunichGermany
- German Cancer Consortium (DKTK)Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University HospitalMunichGermany
| | - Veit L. Bücklein
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- Laboratory for Translational Cancer Immunology, LMU Gene CenterMunichGermany
| | - Viktoria Blumenberg
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- Laboratory for Translational Cancer Immunology, LMU Gene CenterMunichGermany
- Cellular Immunotherapy ProgramMassachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown and Broad Institute of Harvard University and Massachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Giulia Magno
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
| | | | | | - David M. Cordas dos Santos
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- German Cancer Consortium (DKTK)Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University HospitalMunichGermany
- Dana‐Farber Cancer InstituteBostonMassachusettsUSA
| | - Christian Schmidt
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
| | | | - Lisa Frölich
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IVLMU University Hospital, LMU MunichMunichGermany
- German Cancer Consortium (DKTK)Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University HospitalMunichGermany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU)NeuherbergGermany
| | - Michael von Bergwelt‐Baildon
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- German Cancer Consortium (DKTK)Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University HospitalMunichGermany
| | - Kai Rejeski
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- German Cancer Consortium (DKTK)Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University HospitalMunichGermany
- Adult BMT and Cellular Therapy Service, Department of MedicineMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Marion Subklewe
- Department of Medicine IIILMU University HospitalLMU MunichMunichGermany
- German Cancer Consortium (DKTK)Partner Site Munich, a partnership between the DKFZ Heidelberg and LMU University HospitalMunichGermany
- Laboratory for Translational Cancer Immunology, LMU Gene CenterMunichGermany
| |
Collapse
|
11
|
Menegatti S, Lopez-Cobo S, Sutra Del Galy A, Fuentealba J, Silva L, Perrin L, Heurtebise-Chrétien S, Pottez-Jouatte V, Darbois A, Burgdorf N, Privat AL, Simon A, Laprie-Sentenac M, Saitakis M, Wick B, Webber BR, Moriarity BS, Lantz O, Amigorena S, Menger L. Ablation of FAS confers allogeneic CD3 - CAR T cells with resistance to rejection by T cells and natural killer cells. Nat Biomed Eng 2024; 8:1651-1664. [PMID: 39558141 DOI: 10.1038/s41551-024-01282-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 10/15/2024] [Indexed: 11/20/2024]
Abstract
Allogeneic chimaeric antigen receptor T cells (allo-CAR T cells) derived from healthy donors could provide rapid access to standardized and affordable batches of therapeutic cells if their rejection by the host's immune system is avoided. Here, by means of an in vivo genome-wide CRISPR knockout screen, we show that the deletion of Fas or B2m in allo- T cells increases their survival in immunocompetent mice. Human B2M- allo-CAR T cells become highly sensitive to rejection mediated by natural killer (NK) cells, whereas FAS- CAR T cells expressing normal levels of human leukocyte antigen I remain resistant to NK cells. CD3- FAS- CAR T cells outperformed CD3- B2M- CAR T cells in the control of leukaemia growth in mice under allogeneic pressure by T cells and NK cells. The partial protection of CD3- FAS- allo-CAR T cells from cellular rejection may improve the efficacy of allogeneic cellular therapies in patients with cancer.
Collapse
Affiliation(s)
- Silvia Menegatti
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France
- CellAction (Cell therapy Acceleration and Innovation), Institut Curie, Suresnes, France
| | - Sheila Lopez-Cobo
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France
| | | | - Jaime Fuentealba
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France
- CellAction (Cell therapy Acceleration and Innovation), Institut Curie, Suresnes, France
| | - Lisseth Silva
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France
- CellAction (Cell therapy Acceleration and Innovation), Institut Curie, Suresnes, France
| | - Laetitia Perrin
- Gustave Roussy, Paris-Saclay University, INSERM U1015, Villejuif, France
| | | | - Valentine Pottez-Jouatte
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France
- CellAction (Cell therapy Acceleration and Innovation), Institut Curie, Suresnes, France
| | - Aurelie Darbois
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France
| | - Nina Burgdorf
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France
| | - Anne-Laure Privat
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France
- CellAction (Cell therapy Acceleration and Innovation), Institut Curie, Suresnes, France
| | - Albane Simon
- Gustave Roussy, Paris-Saclay University, INSERM U1015, Villejuif, France
| | | | | | - Bryce Wick
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Beau R Webber
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Branden S Moriarity
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Olivier Lantz
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France
| | - Sebastian Amigorena
- Immunity and Cancer, Institut Curie, PSL University, INSERM U932, Paris, France.
- CellAction (Cell therapy Acceleration and Innovation), Institut Curie, Suresnes, France.
- Mnemo Therapeutics, Paris, France.
| | - Laurie Menger
- Gustave Roussy, Paris-Saclay University, INSERM U1015, Villejuif, France.
| |
Collapse
|
12
|
Jiang X, Han X, Jin F, An G, Hou J, He J, Wang Q, Wu W, Zhao Y, Jiang S, Li S, Xu Z, Zheng G, Yang Y, Chen Q, He D, Li Y, Cai Z. When Chinese patients with plasma cell disorders encountered the nationwide Omicron outbreak (December 2022): a real-world multicenter and multiregional study. Hematology 2024; 29:2411741. [PMID: 39373666 DOI: 10.1080/16078454.2024.2411741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 09/27/2024] [Indexed: 10/08/2024] Open
Abstract
OBJECTIVES This study aims to assess the impact of the nationwide Omicron outbreak in December 2022 on Chinese patients with plasma cell disorders (PCD), focusing on the clinical characteristics of PCD patients with COVID-19 and the risk factors contributing to adverse clinical courses (severity and hospitalization) and outcomes. METHODS A multicenter retrospective study was performed from December 1, 2022, to January 19, 2023. The study population includes 404 PCD patients, divided into a COVID-19 group (n = 342) and an uninfected group (n = 62). RESULTS The frequency of COVID-19 infection was 84.7% (342/404), and 16.4% (56/342) were severe COVID-19. Among the 277 patients with complete follow-up, 2 deaths (0.7%) were reported, while 231 (83.4%) recovered from COVID-19. Age > 65 (P = 0.02) and prior anti-CD38 monoclonal antibody (mAb) treatment within six months (P = 0.03) were independent risk factors for severe infection. Additionally, previous chimeric antigen receptor T-cell (CAR-T) therapy within six months was correlated with a higher risk of hospitalization (P = 0.04) and prolonged recovery time (P = 0.03). No significant protective effect of vaccination on infection or severe infection was observed (P > 0.05). CONCLUSIONS The latest Omicron outbreak results in higher rates of severe infection and mortality in PCD patients compared with the general population in China, highlighting the need to protect this vulnerable population during the pandemic. Recent use of anti-CD38 mAb and CAR-T therapy are associated with poorer clinical courses and outcomes of PCD patients with COVID-19.
Collapse
Affiliation(s)
- Xincheng Jiang
- Department of Hematology and Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiaoyan Han
- Department of Hematology and Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Fengyan Jin
- Department of Hematology, the First Hospital of Jilin University, Changchun, People's Republic of China
| | - Gang An
- National Clinical Research Center for Blood Diseases, State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, People's Republic of China
| | - Jian Hou
- Department of Hematology, Renji Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Jingsong He
- Department of Hematology and Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Qingming Wang
- Department of Hematology, the Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Wenjun Wu
- Department of Hematology and Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yi Zhao
- Department of Hematology and Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Songfu Jiang
- Department of Hematology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Shuchan Li
- Department of Hematology and Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Zhenshu Xu
- Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Gaofeng Zheng
- Department of Hematology and Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yang Yang
- Department of Hematology and Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Qingxiao Chen
- Department of Hematology and Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Donghua He
- Department of Hematology and Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yi Li
- Department of Hematology and Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Zhen Cai
- Department of Hematology and Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
13
|
Kampouri E, Reynolds G, Teh BW, Hill JA. Chimeric antigen receptor-T-cell therapies going viral: latent and incidental viral infections. Curr Opin Infect Dis 2024; 37:526-535. [PMID: 39361275 PMCID: PMC11932447 DOI: 10.1097/qco.0000000000001066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2024]
Abstract
PURPOSE OF REVIEW Infections are the leading cause of non-relapse mortality following chimeric antigen receptor (CAR)-T-cell therapy, with viral infections being frequent both in the early and late phases post-infusion. We review the epidemiology of viral infections and discuss critical approaches to prevention and management strategies in this setting. RECENT FINDINGS Herpesviruses dominate the early period. herpes simplex virus and varicella zoster virus infections are rare due to widespread antiviral prophylaxis, but cytomegalovirus (CMV) reactivation is increasingly observed, particularly in high-risk groups including B cell maturation antigen (BCMA)-CAR-T-cell therapy recipients and patients receiving corticosteroids. While CMV end-organ disease is rare, CMV is associated with increased mortality, emphasizing the need to evaluate the broader impact of CMV on long-term hematological, infection, and survival outcomes. Human herpesvirus-6 (HHV-6) has also emerged as a concern, with its diagnosis complicated by overlapping symptoms with neurotoxicity, underscoring the importance of considering viral encephalitis in differential diagnoses. Respiratory viruses are the most common late infections with a higher incidence after BCMA CAR-T-cell therapy. Vaccination remains a critical preventive measure against respiratory viruses but may be less immunogenic following CAR-T-cell therapy. The optimal timing, type of vaccine, and dosing schedule require further investigation. SUMMARY A better understanding of viral epidemiology and preventive trials are needed to improve infection prevention practices and outcomes following CAR-T-cell therapies.
Collapse
Affiliation(s)
- Eleftheria Kampouri
- Infectious Diseases Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Gemma Reynolds
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne
| | - Benjamin W. Teh
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Joshua A. Hill
- Vaccine and Infectious Disease Division
- Clinical Research Division, Fred Hutchinson Cancer Center
- Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
14
|
Palacios-Berraquero ML, Rodriguez-Marquez P, Calleja-Cervantes ME, Berastegui N, Zabaleta A, Burgos L, Alignani D, San Martin-Uriz P, Vilas-Zornoza A, Rodriguez-Diaz S, Inoges S, Lopez-Diaz de Cerio A, Huerga S, Tamariz E, Rifon J, Alfonso-Pierola A, Lasarte JJ, Paiva B, Hernaez M, Rodriguez-Otero P, San-Miguel J, Ezponda T, Rodriguez-Madoz JR, Prosper F. Molecular mechanisms promoting long-term cytopenia after BCMA CAR-T therapy in multiple myeloma. Blood Adv 2024; 8:5479-5492. [PMID: 39058976 PMCID: PMC11532743 DOI: 10.1182/bloodadvances.2023012522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 06/12/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
ABSTRACT Hematologic toxicity is a common side effect of chimeric antigen receptor T-cell (CAR-T) therapies, being particularly severe among patients with relapsed or refractory multiple myeloma (MM). In this study, we characterized 48 patients treated with B-cell maturation antigen (BCMA) CAR-T cells to understand kinetics of cytopenia, identify predictive factors, and determine potential mechanisms underlying these toxicities. We observed that overall incidence of cytopenia was 95.7%, and grade >3 thrombocytopenia and neutropenia, 1 month after infusion, was observed in 57% and 53% of the patients, respectively, being still present after 1 year in 4 and 3 patients, respectively. Baseline cytopenia and high peak inflammatory markers were highly correlated with cytopenia that persisted up to 3 months. To determine potential mechanisms underlying cytopenias, we evaluated the paracrine effect of BCMA CAR-T cells on hematopoietic stem and progenitor cell (HSPC) differentiation using an ex vivo myeloid differentiation model. Phenotypic analysis showed that supernatants from activated CAR-T cells (spCAR) halted HSPC differentiation, promoting more immature phenotypes, which could be prevented with a combination of interferon γ, tumor necrosis factor α/β, transforming growth factor β, interleukin-6 (IL-6) and IL-17 inhibitors. Single-cell RNA sequencing demonstrated upregulation of transcription factors associated with early stages of hematopoietic differentiation in the presence of spCAR (GATA2, RUNX1, CEBPA) and a decrease in the activity of key regulons involved in neutrophil and monocytic maturation (ID2 and MAFB). These results suggest that CAR-T activation induces HSPC maturation arrest through paracrine effects and provides potential treatments to mitigate the severity of this toxicity.
Collapse
Affiliation(s)
- Maria Luisa Palacios-Berraquero
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Paula Rodriguez-Marquez
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Maria Erendira Calleja-Cervantes
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Computational Biology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Nerea Berastegui
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Aintzane Zabaleta
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Leire Burgos
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Diego Alignani
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Patxi San Martin-Uriz
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Amaia Vilas-Zornoza
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Saray Rodriguez-Diaz
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Susana Inoges
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Immunology and Immunotherapy Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Ascensión Lopez-Diaz de Cerio
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Immunology and Immunotherapy Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Sofia Huerga
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Esteban Tamariz
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Jose Rifon
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Ana Alfonso-Pierola
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Juan Jose Lasarte
- Immunology and Immunotherapy Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Bruno Paiva
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Flow Cytometry Core, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
| | - Mikel Hernaez
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
- Computational Biology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Data Science and Artificial Intelligence Institute, Universidad de Navarra, Pamplona, Spain
| | - Paula Rodriguez-Otero
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Jesus San-Miguel
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Teresa Ezponda
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Juan Roberto Rodriguez-Madoz
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| | - Felipe Prosper
- Hematology and Cell Therapy Department, Cancer Center Clinica Universidad de Navarra, IdiSNA, Pamplona, Spain
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigacion Biomedica en Red de Cancer, Madrid, Spain
| |
Collapse
|
15
|
Yang Y, Peng H, Wang J, Li F. New insights into CAR T-cell hematological toxicities: manifestations, mechanisms, and effective management strategies. Exp Hematol Oncol 2024; 13:110. [PMID: 39521987 PMCID: PMC11549815 DOI: 10.1186/s40164-024-00573-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/12/2024] [Indexed: 11/16/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy represents a highly efficacious treatment modality demonstrated to enhance outcomes in patients afflicted with malignancies, particularly those enduring relapsed or refractory hematological malignancies. However, the escalating adoption of CAR T-cell therapy has unveiled several life-threatening toxicities, notably cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), infections, and hematological toxicities (HTs), thereby hindering the broad implementation of CAR T-cell therapy. HTs encompass a spectrum of adverse effects, including cytopenias, hemophagocytic lymphohistiocytosis (HLH), coagulopathies, and B-cell aplasia. While our comprehension of the underlying mechanisms governing CRS and ICANS is advancing, the intricate pathophysiology of HTs remains inadequately elucidated. Such knowledge gaps may precipitate suboptimal therapeutic decisions, potentially culminating in substantial medical resource depletion and detriment to patients' quality of life. In this comprehensive review, based on recent updated findings, we delineate various mechanisms contributing to HTs subsequent to CAR T-cell therapy, explicate manifestations of HTs, and proffer strategic interventions to mitigate this relevant clinical challenge.
Collapse
Affiliation(s)
- Yuanyuan Yang
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hongwei Peng
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, National Clinical Research Center for Blood Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Fei Li
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
- Jiangxi Clinical Research Center for Hematologic Disease, Nanchang, Jiangxi, China.
- Institute of Lymphoma and Myeloma, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
16
|
Deng L, Yu X, Song X, Guan R, Li W, Hou Y, Shao Y, Zhao Y, Wang J, Liu Y, Xiao Q, Xin B, Zhou F. Efficacy and risk of donor-derived CAR-T treatment of relapsed B-cell acute lymphoblastic leukemia after hematopoietic stem cell transplantation. Cytotherapy 2024; 26:1301-1307. [PMID: 38888526 DOI: 10.1016/j.jcyt.2024.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024]
Abstract
The one-year survival rate for patients experiencing a relapse of B-cell acute lymphocytic leukemia (B-ALL) following hematopoietic stem cell transplantation (HSCT) is approximately 30%. Patients experiencing a relapse after allogeneic HSCT frequently encounter difficulties in obtaining autologous CAR-T products. We conducted a study involving 14 patients who received donor-derived CAR-T therapy for relapsed B-ALL following HSCT between August 2019 and May 2023 in our center. The results revealed a CR/CRi rate of 78.6% (11/14), a GVHD rate of 21.4% (3/14), and a 1-year overall survival (OS) rate of 56%. Decreased bone marrow donor cell chimerism in 9 patients recovered after CAR-T therapy. The main causes of death were disease progression and infection. Further analysis showed that GVHD (HR 7.224, 95% CI 1.42-36.82, P = 0.017) and platelet recovery at 30 days (HR 6.807, 95% CI 1.61-28.83, P = 0.009) are significantly associated with OS after CAR-T therapy. Based on the findings, we conclude that donor-derived CAR-T cells are effective in treating relapsed B-ALL patients following HSCT. Additionally, GVHD and poor platelet recovery impact OS, but further verification with a larger sample size is needed.
Collapse
Affiliation(s)
- Lei Deng
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Xiaolin Yu
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Xiaocheng Song
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Rui Guan
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Wenjun Li
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Yixi Hou
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Yan Shao
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Yuerong Zhao
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Jing Wang
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Yue Liu
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Qianqian Xiao
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Bo Xin
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China
| | - Fang Zhou
- Department of Hematology, The 960th Hospital of The Chinese People's Liberation Army Joint Logistics Support Force, Jinan, China.
| |
Collapse
|
17
|
Daunov M, van Besien K. High-Dose Chemotherapy and Autologous or Allogeneic Transplantation in Aggressive B-Cell Lymphoma-Is There Still a Role? Cells 2024; 13:1780. [PMID: 39513887 PMCID: PMC11545473 DOI: 10.3390/cells13211780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/15/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Novel therapies such as CAR-T, BTK inhibitors and PD-1 inhibitors have changed the management of aggressive B-cell lymphomas. Nonetheless, these novel therapies have their own risk of late toxicities including second malignancies. They also create a subgroup of patients with relapse, treatment failure, or indefinite maintenance. We discuss the current role of autologous and allogeneic stem cell transplantation in this context. In patients with recurrent diffuse large B-cell lymphoma, CAR-T cell treatment has largely replaced autologous transplant. Autologous transplant should be considered in patients with late relapses and in selected patients with T-cell-rich B-cell lymphoma, where CAR-T cell therapy may be less effective. It also remains the treatment of choice for consolidation of patients with primary CNS lymphoma. In mantle cell lymphoma, intensive chemotherapy combined with BTK inhibitors and rituximab results in excellent outcomes, and the role of autologous transplantation is declining. In Hodgkin's lymphoma, autologous transplant consolidation remains the standard of care for patients who failed initial chemotherapy. Allogeneic transplantation has lower relapse rates but more complications and higher non-relapse mortality than autologous transplantation. It is usually reserved for patients who fail autologous transplantation or in whom autologous stem cells cannot be collected. It may also have an important role in patients who fail CAR-T therapies. The increasing complexity of care and evolving sequencing of therapies for patients with aggressive B-cell lymphomas only emphasizes the importance of appropriate patient selection and optimal timing of stem cell transplantation.
Collapse
Affiliation(s)
| | - Koen van Besien
- University Hospitals, Seidman Cancer Center, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA;
| |
Collapse
|
18
|
Jain MD, Spiegel JY, Nastoupil LJ, Tamaresis J, Ghobadi A, Lin Y, Lekakis L, Reagan P, Oluwole O, McGuirk J, Deol A, Dorritie KA, Sehgal AR, Goy A, Hill BT, Andreadis C, Munoz J, Ulrickson M, Westin J, Chavez JC, Patel D, Jacobs MT, Bansal R, Bennani NN, Patel VG, Rapoport AP, Vose JM, Miklos DB, Neelapu SS, Locke FL, Lunning M, Dahiya S. Five-Year Follow-Up of Standard-of-Care Axicabtagene Ciloleucel for Large B-Cell Lymphoma: Results From the US Lymphoma CAR T Consortium. J Clin Oncol 2024; 42:3581-3592. [PMID: 39094076 PMCID: PMC11483191 DOI: 10.1200/jco.23.02786] [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: 12/26/2023] [Revised: 04/29/2024] [Accepted: 05/20/2024] [Indexed: 08/04/2024] Open
Abstract
PURPOSE Axicabtagene ciloleucel (axi-cel) is an autologous CD19 chimeric antigen receptor (CAR) T-cell therapy that is approved for the treatment of relapsed or refractory large B-cell lymphoma. Little is known about the long-term survivorship after CAR T-cell therapy. METHODS We previously reported the results of 298 patients who were leukapheresed with the intent to receive standard-of-care axi-cel (n = 275 infused) after two or more previous lines of therapy at a median follow-up of 12.9 months. Here, we report extended follow-up of this cohort to a median of 58 months, with a focus on late survivorship events. RESULTS Among axi-cel-infused patients, progression-free survival at 5 years was 29% and overall survival (OS) at 5 years was 40%. The 5-year lymphoma-specific survival was 53% with infrequent late relapses. However, the 5-year nonrelapse mortality (NRM) was 16.2%, with over half of NRM events occurring beyond 2 years. Patients who were 60 years and older had a lower risk of relapse (P = .02), but a higher risk of NRM compared with patients younger than 60 years (NRM odds ratio, 4.5 [95% CI, 2.1 to 10.8]; P < .001). Late NRM was mainly due to infections and subsequent malignant neoplasms (SMNs). In total, SMNs occurred in 24 patients (9%), including therapy-related myeloid neoplasms (n = 15), solid tumors (n = 7), and unrelated lymphoid malignancies (n = 2). CONCLUSION In the standard-of-care setting, axi-cel exhibits outcomes consistent with those reported in clinical trials, with sustained, durable responses observed at the 5-year time point. However, late infections and the development of SMN are key survivorship issues that reduce long-term survival after CAR T-cell therapy, particularly in the elderly.
Collapse
MESH Headings
- Humans
- Middle Aged
- Male
- Female
- Biological Products/therapeutic use
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/mortality
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Aged
- Antigens, CD19/immunology
- Antigens, CD19/therapeutic use
- Adult
- Follow-Up Studies
- United States
- Young Adult
- Aged, 80 and over
- Standard of Care
- Receptors, Chimeric Antigen/therapeutic use
- Receptors, Chimeric Antigen/immunology
Collapse
Affiliation(s)
| | - Jay Y Spiegel
- University of Miami Miller School of Medicine, Miami, FL
| | | | | | - Armin Ghobadi
- Washington University School of Medicine and Siteman Cancer Center, St Louis, MO
| | - Yi Lin
- Mayo Clinic, Rochester, MN
| | | | | | | | | | - Abhinav Deol
- Karmanos Center Institute/Wayne State University, Detroit, MI
| | | | | | - Andre Goy
- John Theurer Cancer Center, Hackensack Meridian Health, Hackensack, NJ
| | | | | | | | | | - Jason Westin
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Dilan Patel
- Washington University School of Medicine and Siteman Cancer Center, St Louis, MO
| | - Miriam T Jacobs
- Washington University School of Medicine and Siteman Cancer Center, St Louis, MO
| | | | | | | | - Aaron P Rapoport
- University of Maryland School of Medicine and Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | - Julie M Vose
- University of Nebraska Medical Center, Omaha, NE
| | | | | | | | | | - Saurabh Dahiya
- Stanford University Medical Center, Stanford, CA
- University of Maryland School of Medicine and Greenebaum Comprehensive Cancer Center, Baltimore, MD
| |
Collapse
|
19
|
Hou Z, Meng D, Ruan M, Liang X, Ni J, Jifei D, Wu Z, Xia R, Ge J, Long Z. A lesson from fatal invasive fungal infections after CAR-T cell therapy: a case report and literature review. Immunotherapy 2024; 16:1021-1027. [PMID: 39373599 PMCID: PMC11492695 DOI: 10.1080/1750743x.2024.2404381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 09/11/2024] [Indexed: 10/08/2024] Open
Abstract
CD19 chimeric antigen receptor T (CAR-T) cell therapy represents an effective approach to treating patients with relapsed or refractory B-cell hematologic malignancies. Nevertheless, owing to the immunosuppressive effects of this regimen, patients undergoing CD19 CAR-T cell therapy may face an elevated risk of invasive fungal infections, which involve fungi penetrating the host's tissues or bloodstream, leading to life-threating infectious diseases. Herein, we present the case of a 17-year-old male diagnosed with acute lymphoblastic leukemia, who subsequently experienced a fatal invasive fungal infection following administration of CAR-T cell therapy. Furthermore, we delve into the identification of risk factors, implementation of preventive measures and exploration of therapeutic interventions for invasive fungal infections after CAR-T cell therapy.
Collapse
Affiliation(s)
- Zhengfeng Hou
- Department of hematology, the first affiliated hospital of Anhui medical university, Hefei, 230022, China
| | - Danchen Meng
- Department of hematology, the first affiliated hospital of Anhui medical university, Hefei, 230022, China
| | - Min Ruan
- Department of hematology, the first affiliated hospital of Anhui medical university, Hefei, 230022, China
| | - Xinglin Liang
- Department of hematology, the first affiliated hospital of Anhui medical university, Hefei, 230022, China
| | - Jing Ni
- Department of hematology, the first affiliated hospital of Anhui medical university, Hefei, 230022, China
| | - Dai Jifei
- Department of hematology, the first affiliated hospital of Anhui medical university, Hefei, 230022, China
| | - Zhonghui Wu
- Department of hematology, the first affiliated hospital of Anhui medical university, Hefei, 230022, China
| | - Ruixiang Xia
- Department of hematology, the first affiliated hospital of Anhui medical university, Hefei, 230022, China
| | - Jian Ge
- Department of hematology, the first affiliated hospital of Anhui medical university, Hefei, 230022, China
| | - Zhangbiao Long
- Department of hematology, the first affiliated hospital of Anhui medical university, Hefei, 230022, China
| |
Collapse
|
20
|
Lorenc R, Shouval R, Flynn JR, Devlin SM, Saldia A, De Abia AL, De Lapuerta MC, Tomas AA, Cassanello G, Leslie LA, Rejeski K, Lin RJ, Scordo M, Shah GL, Palomba ML, Salles G, Park J, Giralt SA, Perales MA, Ip A, Dahi PB. Subsequent Malignancies After CD19-Targeted Chimeric Antigen Receptor T Cells in Patients With Lymphoma. Transplant Cell Ther 2024; 30:990-1000. [PMID: 38972512 PMCID: PMC11427145 DOI: 10.1016/j.jtct.2024.06.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/23/2024] [Accepted: 06/30/2024] [Indexed: 07/09/2024]
Abstract
Chimeric antigen receptor (CAR) T cells are an established treatment for B cell non-Hodgkin lymphomas (B-NHL). With the remarkable success in improving survival, understanding the late effects of CAR T cell therapy is becoming more relevant. The aim of this study is to determine the incidence of subsequent malignancies in adult patients with B-NHL. We retrospectively studied 355 patients from 2 different medical centers treated with four different CAR T cell products from 2016 to 2022. The overall cumulative incidence for subsequent malignancies at 36 months was 14% (95% CI: 9.2%, 19%). Subsequent malignancies were grouped into 3 primary categories: solid tumor, hematologic malignancy, and dermatologic malignancy with cumulative incidences at 36 months of 6.1% (95% CI: 3.1%-10%), 4.5% (95% CI: 2.1%-8.1%) and 4.2% (95% CI: 2.1%-7.5%) respectively. Notably, no cases of T cell malignancies were observed. In univariable analysis, increasing age was associated with higher risk for subsequent malignancy. While the overall benefits of CAR T products continue to outweigh their potential risks, more studies and longer follow ups are needed to further demonstrate the risks, patterns, and molecular pathways that lead to the development of subsequent malignancies.
Collapse
Affiliation(s)
- Rachel Lorenc
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Roni Shouval
- Department of Medicine, Weill Cornell Medical College, New York, New York; Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jessica R Flynn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sean M Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Amethyst Saldia
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alejandro Luna De Abia
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Adult Bone Marrow Transplantation Unit. Hospital Universitario Ramón y Cajal, Madrid, Spain
| | | | - Ana Alarcon Tomas
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Hospital Universitario Gregorio Marañón, Madrid, Spain
| | - Giulio Cassanello
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Oncology and Hemato-Oncology, University of Milan, Italy; Lymphoma Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lori A Leslie
- Lymphoma Service, Hackensack Meridian Health, New Jersey, New Jersey
| | - Kai Rejeski
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard J Lin
- Department of Medicine, Weill Cornell Medical College, New York, New York; Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael Scordo
- Department of Medicine, Weill Cornell Medical College, New York, New York; Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gunjan L Shah
- Department of Medicine, Weill Cornell Medical College, New York, New York; Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - M Lia Palomba
- Department of Medicine, Weill Cornell Medical College, New York, New York; Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gilles Salles
- Department of Medicine, Weill Cornell Medical College, New York, New York; Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jae Park
- Department of Medicine, Weill Cornell Medical College, New York, New York; Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sergio A Giralt
- Department of Medicine, Weill Cornell Medical College, New York, New York; Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Miguel-Angel Perales
- Department of Medicine, Weill Cornell Medical College, New York, New York; Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andrew Ip
- Lymphoma Service, Hackensack Meridian Health, New Jersey, New Jersey
| | - Parastoo B Dahi
- Department of Medicine, Weill Cornell Medical College, New York, New York; Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
21
|
Shahid Z, Jain T, Dioverti V, Pennisi M, Mikkilineni L, Thiruvengadam SK, Shah NN, Dadwal S, Papanicolaou G, Hamadani M, Carpenter PA, Alfaro GM, Seo SK, Hill JA. Best Practice Considerations by The American Society of Transplant and Cellular Therapy: Infection Prevention and Management After Chimeric Antigen Receptor T Cell Therapy for Hematological Malignancies. Transplant Cell Ther 2024; 30:955-969. [PMID: 39084261 DOI: 10.1016/j.jtct.2024.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is rapidly advancing, offering promising treatments for patients with hematological malignancy. However, associated infectious complications remain a significant concern because of their contribution to patient morbidity and non-relapse mortality. Recent epidemiological insights shed light on risk factors for infections after CAR T-cell therapy. However, the available evidence is predominantly retrospective, highlighting a need for further prospective studies. Institutions are challenged with managing infections after CAR T-cell therapy but variations in the approaches taken underscore the importance of standardizing infection prevention and management protocols across different healthcare settings. Therefore, the Infectious Diseases Special Interest Group of the American Society of Transplantation and Cellular Therapy assembled an expert panel to develop best practice considerations. The aim was to guide healthcare professionals in optimizing infection prevention and management for CAR T-cell therapy recipients and advocates for early consultation of Infectious Diseases during treatment planning phases given the complexities involved. By synthesizing current evidence and expert opinion these best practice considerations provide the basis for understanding infection risk after CAR T-cell therapies and propose risk-mitigating strategies in children, adolescents, and adults. Continued research and collaboration will be essential to refining and effectively implementing these recommendations.
Collapse
Affiliation(s)
- Zainab Shahid
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Tania Jain
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Veronica Dioverti
- Division of Infectious Disease, Department of Medicine, John Hopkins School of Medicine, Baltimore, Maryland
| | - Martini Pennisi
- Division of Hematology and Stem Cell Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Lekha Mikkilineni
- Division of Bone and Marrow Transplant & Cellular Therapies, Stanford School of Medicine, Palo Alto, California
| | - Swetha Kambhampati Thiruvengadam
- Division of Lymphoma, Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sanjeet Dadwal
- Division of Infectious Disease, Department of Medicine, City of Hope National Medical Center, Duarte, California
| | - Genovefa Papanicolaou
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mehdi Hamadani
- Bone Marrow Transplant & Cellular Therapy Program, Medical College of Wisconsin, Milwaukee, Wisconsin; Center for International Blood and Marrow Transplant Research, Milwaukee, Wisconsin
| | - Paul A Carpenter
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Gabriela Maron Alfaro
- Department of Infectious Diseases, St. Jude Children's Research Hospital and Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Susan K Seo
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joshua A Hill
- Vaccine and Infectious Disease Division, Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| |
Collapse
|
22
|
Angelidakis G, Chemaly RF, Sahasrabhojane PV, Morado-Aramburo O, Jiang Y, Bhatti MM, Shpall E, Hosing C, Jain P, Mahadeo KM, Khawaja F, Elhajj P, Wargo JA, Jenq RR, Ajami NJ, Kebriaei P, Ariza-Heredia EJ. Humoral Immunity and Antibody Responses against Diphtheria, Tetanus, and Pneumococcus after Immune Effector Cell Therapies: A Prospective Study. Vaccines (Basel) 2024; 12:1070. [PMID: 39340100 PMCID: PMC11436035 DOI: 10.3390/vaccines12091070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/07/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Patients undergoing immune effector cell therapy (IECT) are at high risk for infections. We assessed seropositivity against pneumococcus, tetanus, and diphtheria in patients before and after IECT and the patients' response to vaccination. We enrolled patients who underwent IECT from January 2020 to March 2022. Antibody levels for diphtheria, tetanus, and pneumococcus were measured before IECT, at 1 month, and 3-6 months after. Eligible patients were vaccinated after IECT. In non-seroprotected patients, we discontinued testing. Before IECT, most patients had seroprotective antibody levels against tetanus (68/69, 99%) and diphtheria (65/69, 94%), but fewer did against pneumococcus (24/67, 36%). After IECT, all patients had seroprotective antibody levels for tetanus at 1 month (68/68) and 3-6 months (56/56). For diphtheria, 65/65 patients (100%) had seroprotective antibody levels at 1 month, and 48/53 (91%) did at 3-6 months. For pneumococcus, seroprotective antibody levels were identified in 91% (21/23) of patients at 1 month and 79% (15/19) at 3-6 months following IECT. Fifteen patients received a pneumococcal vaccine after IECT, but none achieved seroprotective response. One patient received the tetanus-diphtheria vaccine and had a seroprotective antibody response. Because some patients experience loss of immunity after IECT, studies evaluating vaccination strategies post-IECT are needed.
Collapse
Affiliation(s)
- Georgios Angelidakis
- Departments of Infectious Diseases, Infection Control and Employee Health, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Roy F Chemaly
- Departments of Infectious Diseases, Infection Control and Employee Health, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pranoti V Sahasrabhojane
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Platform for Innovative Microbiome and Translational Research, The University of Texas MD Anderson Cancer, Houston, TX 77030, USA
| | - Oscar Morado-Aramburo
- Departments of Infectious Diseases, Infection Control and Employee Health, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ying Jiang
- Departments of Infectious Diseases, Infection Control and Employee Health, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Micah M Bhatti
- Department of Clinical Microbiology, Division of Pathology/Lab Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elizabeth Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chitra Hosing
- Department of Stem Cell Transplantation and Cellular Therapy, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Preetesh Jain
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kris Michael Mahadeo
- Department of Pediatrics, Division of Pediatric Transplant and Cellular Therapy, Duke University School of Medicine, Durham, NC 27705, USA
| | - Fareed Khawaja
- Departments of Infectious Diseases, Infection Control and Employee Health, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Peter Elhajj
- Departments of Infectious Diseases, Infection Control and Employee Health, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer A Wargo
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Platform for Innovative Microbiome and Translational Research, The University of Texas MD Anderson Cancer, Houston, TX 77030, USA
- Department of Surgical Oncology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert R Jenq
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Platform for Innovative Microbiome and Translational Research, The University of Texas MD Anderson Cancer, Houston, TX 77030, USA
| | - Nadim J Ajami
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Platform for Innovative Microbiome and Translational Research, The University of Texas MD Anderson Cancer, Houston, TX 77030, USA
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ella J Ariza-Heredia
- Departments of Infectious Diseases, Infection Control and Employee Health, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
23
|
Bouziana S, Bouzianas D. The Current Landscape of Secondary Malignancies after CAR T-Cell Therapies: How Could Malignancies Be Prevented? Int J Mol Sci 2024; 25:9518. [PMID: 39273462 PMCID: PMC11395546 DOI: 10.3390/ijms25179518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapies have revolutionised the field of haematological malignancies by achieving impressive remission rates in patients with highly refractory haematological malignancies, improving overall survival. To date, six commercial anti-CD19 and anti-BCMA CAR T-cell products have been approved by the Food and Drug Administration (FDA) for the treatment of relapsed/refractory B-cell haematological malignancies and multiple myeloma. The indications for CAR T-cell therapies are gradually expanding, with these therapies being investigated in a variety of diseases, including non-malignant ones. Despite the great success, there are several challenges surrounding CAR T-cell therapies, such as non-durable responses and high-grade toxicities. In addition, a new safety concern was added by the FDA on 28 November 2023 following reports of T-cell malignancies in patients previously treated with either anti-CD19 or anti-BCMA autologous CAR T-cell therapies both in clinical trials and in the real-world setting. Since then, several reports have been published presenting the incidence and analysing the risks of other secondary malignancies after CAR T-cell therapies. In this opinion article, the current landscape of secondary malignancies after CAR T-cell therapies is presented, along with a proposed strategy for future research aiming at potentially diminishing or abrogating the risk of developing secondary malignancies after CAR T-cell therapies.
Collapse
Affiliation(s)
- Stella Bouziana
- Department of Hematology, King’s College Hospital, London SE59RS, UK
| | - Dimitrios Bouzianas
- BReMeL, Biopharmaceutical and Regenerative Medicine Laboratories, 55534 Thessaloniki, Greece;
| |
Collapse
|
24
|
Kitamura W, Asada N, Ikegawa S, Fujiwara H, Kamoi C, Ennishi D, Nishimori H, Fujii K, Fujii N, Matsuoka KI, Maeda Y. Activated CD4 + T Cell Proportion in the Peripheral Blood Correlates with the Duration of Cytokine Release Syndrome and Predicts Clinical Outcome after Chimeric Antigen Receptor T Cell Therapy. Intern Med 2024; 63:1863-1872. [PMID: 38945932 PMCID: PMC11272506 DOI: 10.2169/internalmedicine.2556-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 10/16/2023] [Indexed: 07/02/2024] Open
Abstract
Objective Chimeric antigen receptor (CAR) T cell therapy is an emerging and effective therapy for relapsed or refractory diffuse large B cell lymphoma (R/R DLBCL). The characteristic toxicities of CAR T cell therapy include cytokine release syndrome (CRS) and prolonged cytopenia. We investigated the factors associated with these complications after CAR T cell therapy by analyzing lymphocyte subsets following CAR T cell infusion. Methods We retrospectively analyzed peripheral blood samples on days 7, 14, and 28 after tisagenlecleucel (tisa-cel) infusion by flow cytometry at our institution between June 2020 and September 2022. Patients Thirty-five patients with R/R DLBCL who received tisa-cel therapy were included. Results A flow cytometry-based analysis of blood samples from these patients revealed that the proportion of CD4+CD25+CD127+ T cells (hereafter referred to as "activated CD4+ T cells" ) among the total CD4+ T cells on day 7 after tisa-cel infusion correlated with the duration of CRS (r=0.79, p<0.01). In addition, a prognostic analysis of the overall survival (OS) using time-dependent receiver operating characteristic curves indicated a significantly more favorable OS and progression-free survival of patients with a proportion of activated CD4+ T cells among the total CD4+ T cells <0.73 (p=0.01, and p<0.01, respectively). Conclusion These results suggest that the proportion of activated CD4+ T cells on day 7 after tisa-cel infusion correlates with the CRS duration and predicts clinical outcomes after CAR T cell therapy. Further studies with a larger number of patients are required to validate these observations.
Collapse
MESH Headings
- Humans
- Male
- Female
- Cytokine Release Syndrome/blood
- Cytokine Release Syndrome/etiology
- Cytokine Release Syndrome/therapy
- Cytokine Release Syndrome/immunology
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Middle Aged
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/blood
- Lymphoma, Large B-Cell, Diffuse/immunology
- Aged
- Retrospective Studies
- CD4-Positive T-Lymphocytes/immunology
- Adult
- Treatment Outcome
- Receptors, Chimeric Antigen/immunology
- Prognosis
- Receptors, Antigen, T-Cell
Collapse
Affiliation(s)
- Wataru Kitamura
- Department of Hematology and Oncology, Okayama University Hospital, Japan
| | - Noboru Asada
- Department of Hematology and Oncology, Okayama University Hospital, Japan
| | - Shuntaro Ikegawa
- Department of Hematology and Oncology, Okayama University Hospital, Japan
- Division of Blood Transfusion, Okayama University Hospital, Japan
| | - Hideaki Fujiwara
- Department of Hematology and Oncology, Okayama University Hospital, Japan
| | - Chihiro Kamoi
- Department of Hematology and Oncology, Okayama University Hospital, Japan
- Division of Blood Transfusion, Okayama University Hospital, Japan
| | - Daisuke Ennishi
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, Japan
| | - Hisakazu Nishimori
- Department of Hematology and Oncology, Okayama University Hospital, Japan
| | - Keiko Fujii
- Division of Clinical Laboratory, Okayama University Hospital, Japan
| | - Nobuharu Fujii
- Division of Blood Transfusion, Okayama University Hospital, Japan
| | - Ken-Ichi Matsuoka
- Department of Hematology and Oncology, Okayama University Hospital, Japan
| | - Yoshinobu Maeda
- Department of Hematology and Oncology, Okayama University Hospital, Japan
| |
Collapse
|
25
|
Cordeiro AC, Durisek G, Batista MV, Schmidt J, de Lima M, Bezerra E. Late events after anti-CD19 CAR T-cell therapy for relapsed/refractory B-cell non-Hodgkin lymphoma. Front Oncol 2024; 14:1404351. [PMID: 38919524 PMCID: PMC11196778 DOI: 10.3389/fonc.2024.1404351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 05/27/2024] [Indexed: 06/27/2024] Open
Abstract
Background The short-term complications from chimeric antigen receptor T-cell therapy (CART) are well characterized, but the long-term complications still need to be further investigated. Therefore, herein, we will review the currently available literature published on the late adverse events following CART. Methods We reviewed published data available from pivotal trials and real-world experiences with anti-CD19 CART (CART19) for adults with lymphoma. We defined late events as occurring or persisting beyond 1 month after CART infusion. We focused our literature review on the following late-event outcomes post-CART19: cytopenia, immune reconstitution, infections, and subsequent malignancies. Results Grade 3-4 cytopenia beyond 30 days occurs in 30%-40% of patients and beyond 90 days in 3%-22% of patients and is usually managed with growth-factor and transfusion support, along with neutropenic prophylaxis. B-cell aplasia and hypogammaglobulinemia are expected on-target off-tumor effects of CART19, 44%-53% of patients have IgG < 400 mg/dL, and approximately 27%-38% of patients receive intravenous immunoglobulin (IVIG) replacement. Infections beyond the initial month from CART19 are not frequent and rarely severe, but they are more prevalent and severe when patients receive subsequent therapies post-CART19 for their underlying disease. Late neurotoxicity and neurocognitive impairment are uncommon, and other causes should be considered. T-cell lymphoma (TCL) after CART is an extremely rare event and not necessarily related to CAR transgene. Myeloid neoplasm is not rare post-CART, but unclear causality given heavily pretreated patient population is already at risk for therapy-related myeloid neoplasm. Conclusion CART19 is associated with clinically significant long-term effects such as prolonged cytopenia, hypogammaglobulinemia, and infections that warrant clinical surveillance, but they are mostly manageable with a low risk of non-relapse mortality. The risk of subsequent malignancies post-CART19 seems low, and the relationship with CART19 and/or prior therapies is unclear; but regardless of the possible causality, this should not impact the current benefit-risk ratio of CART19 for relapsed/refractory B-cell non-Hodgkin lymphoma (NHL).
Collapse
Affiliation(s)
| | - George Durisek
- College of Medicine, The Ohio State University, Columbus, OH, United States
| | | | - Jayr Schmidt
- Hematology Division, AC Camargo Cancer Center, São Paulo, SP, Brazil
| | - Marcos de Lima
- Division of Hematology, The Ohio State University, Columbus, OH, United States
| | - Evandro Bezerra
- Division of Hematology, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
26
|
Melody M, Epperla N, Shouse G, Romancik J, Allen P, Moyo TK, Kenkre V, Ollila T, Fitzgerald L, Hess B, David K, Herr MM, Odetola O, Lin A, Moreira J, Ma S, Winter JN, Roy I, Stephens D, Danilov A, Shah NN, Barta SK, Cortese M, Cohen JB, Gordon LI, Karmali R. Subsequent malignant neoplasms in patients previously treated with anti-CD19 CAR T-cell therapy. Blood Adv 2024; 8:2327-2331. [PMID: 38498727 PMCID: PMC11126789 DOI: 10.1182/bloodadvances.2024012573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/20/2024] Open
Affiliation(s)
- Megan Melody
- Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Narendranath Epperla
- Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH
| | | | | | - Pamela Allen
- Winship Cancer Institute, Emory University, Atlanta, GA
| | | | - Vaishalee Kenkre
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, WI
| | - Thomas Ollila
- Lifespan Cancer Institute, Brown University, Providence, RI
| | | | - Brian Hess
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC
| | - Kevin David
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ
| | - Megan M. Herr
- Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | | | - Adam Lin
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - Jonathan Moreira
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - Shuo Ma
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - Jane N. Winter
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - Ishan Roy
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
- Shirley Ryan Ability Lab, Chicago, IL
| | - Deborah Stephens
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | - Nirav N. Shah
- MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI
| | - Stefan K. Barta
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | | | | | - Leo I. Gordon
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - Reem Karmali
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| |
Collapse
|
27
|
Ahmed N, Oluwole O, Mahmoudjafari Z, Suleman N, McGuirk JP. Managing Infection Complications in the Setting of Chimeric Antigen Receptor T cell (CAR-T) Therapy. Clin Hematol Int 2024; 6:31-45. [PMID: 38817309 PMCID: PMC11086990 DOI: 10.46989/001c.115932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/23/2024] [Indexed: 06/01/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR T-cell) therapy has changed the paradigm of management of non-Hodgkin's lymphoma (NHL) and Multiple Myeloma. Infection complications have emerged as a concern that can arise in the setting of therapy and lead to morbidity and mortality. In this review, we classified infection complications into three categories, pre-infusion phase from the time pre- lymphodepletion (LD) up to day zero, early phase from day of infusion to day 30 post-infusion, and late phase after day 30 onwards. Infections arising in the pre-infusion phase are closely related to previous chemotherapy and bridging therapy. Infections arising in the early phase are more likely related to LD chemo and the expected brief period of grade 3-4 neutropenia. Infections arising in the late phase are particularly worrisome because they are associated with adverse risk features including prolonged neutropenia, dysregulation of humoral and adaptive immunity with lymphopenia, hypogammaglobinemia, and B cell aplasia. Bacterial, respiratory and other viral infections, protozoal and fungal infections can occur during this time . We recommend enhanced supportive care including prompt recognition and treatment of neutropenia with growth factor support, surveillance testing for specific viruses in the appropriate instance, management of hypogammaglobulinemia with repletion as appropriate and extended antimicrobial prophylaxis in those at higher risk (e.g. high dose steroid use and prolonged cytopenia). Finally, we recommend re-immunizing patients post CAR-T based on CDC and transplant guidelines.
Collapse
Affiliation(s)
- Nausheen Ahmed
- Hematologic Malignancies and Cellular TherapeuticsUniversity of Kansas Cancer Center
| | - Olalekan Oluwole
- Medicine, Hematology and OncologyVanderbilt University Medical Center
| | - Zahra Mahmoudjafari
- Hematologic Malignancies and Cellular TherapeuticsUniversity of Kansas Cancer Center
| | - Nahid Suleman
- Hematologic Malignancies and Cellular TherapeuticsUniversity of Kansas Cancer Center
| | - Joseph P McGuirk
- Hematologic Malignancies and Cellular TherapeuticsUniversity of Kansas Cancer Center
| |
Collapse
|
28
|
Shi M, Wang J, Huang H, Liu D, Cheng H, Wang X, Chen W, Yan Z, Sang W, Qi K, Li D, Zhu F, Li Z, Qiao J, Wu Q, Zeng L, Fei X, Gu W, Miao Y, Xu K, Zheng J, Cao J. Bispecific CAR T cell therapy targeting BCMA and CD19 in relapsed/refractory multiple myeloma: a phase I/II trial. Nat Commun 2024; 15:3371. [PMID: 38643278 PMCID: PMC11032309 DOI: 10.1038/s41467-024-47801-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/12/2024] [Indexed: 04/22/2024] Open
Abstract
Despite the high therapeutic response achieved with B-cell maturation antigen (BCMA)-specific chimeric antigen receptor (CAR) T-cell therapy in relapsed and refractory multiple myeloma (R/R MM), primary resistance and relapse exist with single-target immunotherapy. Here, we design bispecific BC19 CAR T cells targeting BCMA/CD19 and evaluate antimyeloma activity in vitro and in vivo. Preclinical results indicate that BC19 CAR specifically recognize target antigens, and BC19 CAR T cells mediate selective killing of BCMA or CD19-positive cancer cells. BC19 CAR T cells also exhibit potent antigen-specific anti-tumor activity in xenograft mouse models. We conduct an open-label, single-arm, phase I/II study of BC19 CAR T cells in 50 patients with R/R MM (ChiCTR2000033567). The primary endpoint was safety. BC19 CAR T cells are well tolerated with grade 3 or higher cytokine release syndrome in 8% of patients and grade 1 neurotoxic events in 4% of patients, which meet the pre-specified primary endpoint. Secondary endpoints include overall response rate (92%), median progression-free survival (19.7 months), median overall survival (19.7 months) and median duration of response (not reached). Our study demonstrates that bispecific BC19 CAR T cells are feasible, safe and effective in treating patients with R/R MM.
Collapse
Affiliation(s)
- Ming Shi
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, China
| | - Jiaojiao Wang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Hongming Huang
- Department of Hematology, The Affiliated Hospital of Nantong University, Nantong, 226000, China
| | - Dan Liu
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, China
- Jiangsu Bone Marrow Stem Cell Institute, Xuzhou, 221002, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, 221002, China
| | - Hai Cheng
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Xu Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, 221002, China
| | - Wei Chen
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Zhiling Yan
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Wei Sang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Kunming Qi
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Depeng Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Feng Zhu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Zhenyu Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Jianlin Qiao
- Jiangsu Bone Marrow Stem Cell Institute, Xuzhou, 221002, China
| | - Qingyun Wu
- Jiangsu Bone Marrow Stem Cell Institute, Xuzhou, 221002, China
| | - Lingyu Zeng
- Jiangsu Bone Marrow Stem Cell Institute, Xuzhou, 221002, China
| | - Xiaoming Fei
- Department of Hematology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, China
| | - Weiying Gu
- Department of Hematology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, China
| | - Yuqing Miao
- Department of Hematology, Yancheng No. People's Hospital, Yancheng, 224006, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.
| | - Junnian Zheng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, 221002, China.
| | - Jiang Cao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.
| |
Collapse
|
29
|
Degagné É, Donohoue PD, Roy S, Scherer J, Fowler TW, Davis RT, Reyes GA, Kwong G, Stanaway M, Larroca Vicena V, Mutha D, Guo R, Edwards L, Schilling B, Shaw M, Smith SC, Kohrs B, Kufeldt HJ, Churchward G, Ruan F, Nyer DB, McSweeney K, Irby MJ, Fuller CK, Banh L, Toh MS, Thompson M, Owen AL, An Z, Gradia S, Skoble J, Bryan M, Garner E, Kanner SB. High-Specificity CRISPR-Mediated Genome Engineering in Anti-BCMA Allogeneic CAR T Cells Suppresses Allograft Rejection in Preclinical Models. Cancer Immunol Res 2024; 12:462-477. [PMID: 38345397 PMCID: PMC10985478 DOI: 10.1158/2326-6066.cir-23-0679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/16/2023] [Accepted: 01/31/2024] [Indexed: 04/04/2024]
Abstract
Allogeneic chimeric antigen receptor (CAR) T cell therapies hold the potential to overcome many of the challenges associated with patient-derived (autologous) CAR T cells. Key considerations in the development of allogeneic CAR T cell therapies include prevention of graft-vs-host disease (GvHD) and suppression of allograft rejection. Here, we describe preclinical data supporting the ongoing first-in-human clinical study, the CaMMouflage trial (NCT05722418), evaluating CB-011 in patients with relapsed/refractory multiple myeloma. CB-011 is a hypoimmunogenic, allogeneic anti-B-cell maturation antigen (BCMA) CAR T cell therapy candidate. CB-011 cells feature 4 genomic alterations and were engineered from healthy donor-derived T cells using a Cas12a CRISPR hybrid RNA-DNA (chRDNA) genome-editing technology platform. To address allograft rejection, CAR T cells were engineered to prevent endogenous HLA class I complex expression and overexpress a single-chain polyprotein complex composed of beta-2 microglobulin (B2M) tethered to HLA-E. In addition, T-cell receptor (TCR) expression was disrupted at the TCR alpha constant locus in combination with the site-specific insertion of a humanized BCMA-specific CAR. CB-011 cells exhibited robust plasmablast cytotoxicity in vitro in a mixed lymphocyte reaction in cell cocultures derived from patients with multiple myeloma. In addition, CB-011 cells demonstrated suppressed recognition by and cytotoxicity from HLA-mismatched T cells. CB-011 cells were protected from natural killer cell-mediated cytotoxicity in vitro and in vivo due to endogenous promoter-driven expression of B2M-HLA-E. Potent antitumor efficacy, when combined with an immune-cloaking armoring strategy to dampen allograft rejection, offers optimized therapeutic potential in multiple myeloma. See related Spotlight by Caimi and Melenhorst, p. 385.
Collapse
Affiliation(s)
| | | | - Suparna Roy
- Caribou Biosciences, Inc., Berkeley, California
| | | | | | | | | | | | | | | | - Devin Mutha
- Caribou Biosciences, Inc., Berkeley, California
| | - Raymond Guo
- Caribou Biosciences, Inc., Berkeley, California
| | | | | | - McKay Shaw
- Caribou Biosciences, Inc., Berkeley, California
| | | | - Bryan Kohrs
- Caribou Biosciences, Inc., Berkeley, California
| | | | | | - Finey Ruan
- Caribou Biosciences, Inc., Berkeley, California
| | | | | | | | | | - Lynda Banh
- Caribou Biosciences, Inc., Berkeley, California
| | | | | | | | - Zili An
- Caribou Biosciences, Inc., Berkeley, California
| | | | | | - Mara Bryan
- Caribou Biosciences, Inc., Berkeley, California
| | | | | |
Collapse
|
30
|
Gambella M, Carlomagno S, Mangerini R, Colombo N, Parodi A, Ghiggi C, Giannoni L, Coviello E, Setti C, Luchetti S, Serio A, Laudisi A, Passannante M, Bo A, Tedone E, Sivori S, Angelucci E, Raiola AM. Early CAR - CD4 + T-lymphocytes recovery following CAR-T cell infusion: A worse outcome in diffuse large B cell lymphoma. EJHAEM 2024; 5:360-368. [PMID: 38633118 PMCID: PMC11020131 DOI: 10.1002/jha2.871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/05/2024] [Indexed: 04/19/2024]
Abstract
CAR- CD4+ T cell lymphopenia is an emerging issue following CAR-T cell therapy. We analyzed the determinants of CD4+ T cell recovery and a possible association with survival in 31 consecutive patients treated with commercial CAR-T for diffuse large B-cell (DLBCL) or mantle cell lymphoma. Circulating immune subpopulations were characterized through multiparametric-flow cytometry. Six-month cumulative incidence of CAR- CD4+ T cell recovery (≥200 cells/μL) was 0.43 (95% confidence interval [CI]: 0.28-0.65). Among possible determinants of CD4+ T cell recovery, we recognized infusion of a 4-1BB product (tisagenlecleucel, TSA) in comparison with a CD28 (axicabtagene/brexucabtagene, AXI/BRX) (hazard ratio [HR] [95% CI]: 5.79 [1.16-24.12] p = 0.016). Higher CD4+ T cell counts resulted with TSA at month-1, -2 and -3. Moderate-to-severe infections were registered with prolonged CD4+ T cell lymphopenia. Early, month-1 CD4+ T cell recovery was associated with a worse outcome in the DLBCL cohort, upheld in a multivariate regression model for overall survival (HR: 4.46 [95% CI: 1.12-17.71], p = 0.03). We conclude that a faster CAR- CD4+ T cell recovery is associated with TSA as compared to AXI/BRX. Month-1 CAR- CD4+ T cell subset recovery could represent a "red flag" for CAR-T cell therapy failure in DLBCL patients.
Collapse
Affiliation(s)
- Massimiliano Gambella
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
- Department of Experimental Medicine (DIMES)University of GenoaGenovaItaly
| | | | - Rosa Mangerini
- Anatomia Patologica OspedalieraIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Nicoletta Colombo
- Anatomia Patologica OspedalieraIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Alessia Parodi
- Anatomia Patologica OspedalieraIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Chiara Ghiggi
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Livia Giannoni
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Elisa Coviello
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Chiara Setti
- Department of Experimental Medicine (DIMES)University of GenoaGenovaItaly
| | - Silvia Luchetti
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Alberto Serio
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Antonella Laudisi
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Monica Passannante
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Alessandra Bo
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Elisabetta Tedone
- Anatomia Patologica OspedalieraIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Simona Sivori
- Department of Experimental Medicine (DIMES)University of GenoaGenovaItaly
- IRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Emanuele Angelucci
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Anna Maria Raiola
- Ematologia e Terapie CellulariIRCCS Ospedale Policlinico San MartinoGenovaItaly
| |
Collapse
|
31
|
Oluwole OO, Forcade E, Muñoz J, de Guibert S, Vose JM, Bartlett NL, Lin Y, Deol A, McSweeney P, Goy AH, Kersten MJ, Jacobson CA, Farooq U, Minnema MC, Thieblemont C, Timmerman JM, Stiff P, Avivi I, Tzachanis D, Zheng Y, Vardhanabhuti S, Nater J, Shen RR, Miao H, Kim JJ, van Meerten T. Long-term outcomes of patients with large B-cell lymphoma treated with axicabtagene ciloleucel and prophylactic corticosteroids. Bone Marrow Transplant 2024; 59:366-372. [PMID: 38177222 PMCID: PMC10920180 DOI: 10.1038/s41409-023-02169-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 01/06/2024]
Abstract
ZUMA-1 safety management cohort 6 investigated the impact of prophylactic corticosteroids and earlier corticosteroids and/or tocilizumab on the incidence and severity of cytokine release syndrome (CRS) and neurologic events (NEs) following axicabtagene ciloleucel (axi-cel) in patients with relapsed/refractory large B-cell lymphoma (R/R LBCL). Prior analyses of cohort 6 with limited follow-up demonstrated no Grade ≥3 CRS, a low rate of NEs, and high response rates, without negatively impacting axi-cel pharmacokinetics. Herein, long-term outcomes of cohort 6 (N = 40) are reported (median follow-up, 26.9 months). Since the 1-year analysis (Oluwole, et al. Blood. 2022;138[suppl 1]:2832), no new CRS was reported. Two new NEs occurred in two patients (Grade 2 dementia unrelated to axi-cel; Grade 5 axi-cel-related leukoencephalopathy). Six new infections and eight deaths (five progressive disease; one leukoencephalopathy; two COVID-19) occurred. Objective and complete response rates remained at 95% and 80%, respectively. Median duration of response and progression-free survival were reached at 25.9 and 26.8 months, respectively. Median overall survival has not yet been reached. Eighteen patients (45%) remained in ongoing response at data cutoff. With ≥2 years of follow-up, prophylactic corticosteroids and earlier corticosteroids and/or tocilizumab continued to demonstrate CRS improvement without compromising efficacy outcomes, which remained high and durable.
Collapse
Affiliation(s)
| | - Edouard Forcade
- Service d'Hématologie Clinique et Thérapie Cellulaire, Centre Hospitalier Universitaire de Bordeaux, F-33000, Bordeaux, France
| | - Javier Muñoz
- Banner MD Anderson Cancer Center, Gilbert, AZ, USA
| | - Sophie de Guibert
- Hématologie Clinique, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Julie M Vose
- University of Nebraska Medical Center, Omaha, NE, USA
| | - Nancy L Bartlett
- Washington University School of Medicine and Siteman Cancer Center, St Louis, MO, USA
| | - Yi Lin
- Mayo Clinic, Rochester, MN, USA
| | - Abhinav Deol
- Karmanos Cancer Center, Wayne State University, Detroit, MI, USA
| | | | - Andre H Goy
- John Theurer Cancer Center, Hackensack, NJ, USA
| | - Marie José Kersten
- Amsterdam UMC, Location University of Amsterdam, Cancer Center Amsterdam, Amsterdam (on behalf of HOVON/LLPC), The Netherlands
| | | | | | - Monique C Minnema
- University Medical Center Utrecht (on behalf of HOVON/LLPC), Utrecht, The Netherlands
| | - Catherine Thieblemont
- Paris University, Assistance publique-Hôpitaux de Paris, Hemato-oncology, F-75010, Paris, France
| | - John M Timmerman
- University of California, Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Patrick Stiff
- Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Irit Avivi
- Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Yan Zheng
- Kite, a Gilead Company, Santa Monica, CA, USA
| | | | - Jenny Nater
- Kite, a Gilead Company, Santa Monica, CA, USA
| | | | - Harry Miao
- Kite, a Gilead Company, Santa Monica, CA, USA
| | - Jenny J Kim
- Kite, a Gilead Company, Santa Monica, CA, USA
| | - Tom van Meerten
- University Medical Center Groningen, Groningen (on behalf of HOVON/LLPC), The Netherlands
| |
Collapse
|
32
|
Garcia-Pouton N, Ortiz-Maldonado V, Peyrony O, Chumbita M, Aiello TF, Monzo-Gallo P, Lopera C, Puerta-Alcalde P, Magnano L, Martinez-Cibrian N, Pitart C, Juan M, Delgado J, Fernandez De Larrea C, Soriano Á, Urbano-Ispizua Á, Garcia-Vidal C. Infection epidemiology in relation to different therapy phases in patients with haematological malignancies receiving CAR T-cell therapy. Eur J Haematol 2024; 112:371-378. [PMID: 37879842 DOI: 10.1111/ejh.14122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 10/27/2023]
Abstract
BACKGROUND We described the real-life epidemiology and causes of infections on the different therapy phases in patients undergoing chimeric antigen receptor (CAR) T-cells directed towards CD19+ or BCMA+ cells. METHODS All consecutive patients receiving CAR T-cell therapy at our institution were prospectively followed-up. We performed various comparative analyses of all patients and subgroups with and without infections. RESULTS Ninety-one adults mainly received CAR T-cell therapy for acute leukaemia (53%) and lymphoma (33%). We documented a total of 77 infections in 47 (52%) patients, 37 (48%) during the initial neutropenic phase and 40 (52%) during the non-neutropenic phase. Infections during the neutropenic phase were mainly due to bacterial (29, 78%): catheter infections (11 [38%] cases), endogenous source (5 [17%]), and Clostridioides difficile (5 [17%]). Patients receiving corticosteroids after CAR T-cell therapy had a higher risk of endogenous infection (100% vs. 16%; p = .006). During the non-neutropenic phase, bacterial infections remained very frequent (24, 60%), mainly with catheter source (8, 33%). Respiratory tract infections were common (17, 43%). CONCLUSIONS Infections after CAR T-cell therapy were frequent. During the neutropenic phase, it is essential to prevent nosocomial infections and balance the use of antibiotics to lower endogenous bacteraemia and Clostridial infection rates.
Collapse
Affiliation(s)
- Nicol Garcia-Pouton
- Infectious Diseases Department, Hospital Clinic of Barcelona-IDIBAPS, University of Barcelona, Barcelona, Spain
| | | | - Oliver Peyrony
- Infectious Diseases Department, Hospital Clinic of Barcelona-IDIBAPS, University of Barcelona, Barcelona, Spain
- Emergency Department, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Mariana Chumbita
- Infectious Diseases Department, Hospital Clinic of Barcelona-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Tommaso Francesco Aiello
- Infectious Diseases Department, Hospital Clinic of Barcelona-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Patricia Monzo-Gallo
- Infectious Diseases Department, Hospital Clinic of Barcelona-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Carlos Lopera
- Infectious Diseases Department, Hospital Clinic of Barcelona-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Pedro Puerta-Alcalde
- Infectious Diseases Department, Hospital Clinic of Barcelona-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Laura Magnano
- Haematology Department, Hospital Clínic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Nuria Martinez-Cibrian
- Haematology Department, Hospital Clínic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Cristina Pitart
- Microbiology Department, Hospital Clinic, University of Barcelona, ISGLOBAL, Barcelona, Spain
| | - Manel Juan
- Immunology Department, Hospital Clínic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Julio Delgado
- Haematology Department, Hospital Clínic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | | | - Álex Soriano
- Infectious Diseases Department, Hospital Clinic of Barcelona-IDIBAPS, University of Barcelona, Barcelona, Spain
- CIBERINF, CIBER in Infectious Diseases, Barcelona, Spain
| | - Álvaro Urbano-Ispizua
- Haematology Department, Hospital Clínic-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Carolina Garcia-Vidal
- Infectious Diseases Department, Hospital Clinic of Barcelona-IDIBAPS, University of Barcelona, Barcelona, Spain
- CIBERINF, CIBER in Infectious Diseases, Barcelona, Spain
| |
Collapse
|
33
|
Galli E, Frioni F, Malara T, Attardi E, Bellesi S, Hohaus S, Sica S, Sorà F, Chiusolo P. FLT3 Mutated Acute Myeloid Leukemia after CD19 CAR-t Cells. Mediterr J Hematol Infect Dis 2024; 16:e2024029. [PMID: 38468840 PMCID: PMC10927208 DOI: 10.4084/mjhid.2024.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/12/2024] [Indexed: 03/13/2024] Open
Abstract
Chimeric Antigen Receptor T-cells have improved the life expectancy of severely pretreated patients with aggressive hematological cancers; for this reason, therapy-related myeloid leukemias are becoming of great concern in this field, despite their clonal phylogenesis and mutational landscape have not been fully explored yet. This case discusses a 33-year-old man with refractory large B-cell lymphoma, treated with Chimeric Antigen Receptor T-cell (CAR-T) therapy as the 7th line of treatment. Despite a persistent partial response, the patient developed therapy-related acute myeloid leukemia (t-AML) six months post-CAR-T, revealing pre-existing clonal hematopoiesis. The myeloid malignancy exhibited an unusual hypocellular/dysplastic pattern, progressing to an established blast phase with cytopenia. Treatment with demethylating agents and BCL2 inhibitors proved ineffective, leading to t-AML with hyperleukocytosis and FLT3-ITD gain, resulting in the patient's death. This case underscores the impact of severe pretreatment and bone marrow impairment in CAR-T-associated t-AML, emphasizing their role over insertional mutagenesis. Furthermore, it highlights the retention of classic therapy-related leukemia characteristics, including the potential for acquiring FLT3 mutations and displaying dysplastic morphology in these secondary leukemias.
Collapse
Affiliation(s)
- Eugenio Galli
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma
| | - Filippo Frioni
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Roma
| | - Tanja Malara
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma
| | - Enrico Attardi
- Dipartimento di Oncoematologia, Fondazione PTV Policlinico Tor Vergata, Roma
| | - Silvia Bellesi
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma
| | - Stefan Hohaus
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Roma
| | - Simona Sica
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Roma
| | - Federica Sorà
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Roma
| | - Patrizia Chiusolo
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Roma
| |
Collapse
|
34
|
Galli E, Fresa A, Bellesi S, Metafuni E, Maiolo E, Pansini I, Frioni F, Autore F, Limongiello MA, Innocenti I, Giammarco S, Chiusolo P, Zini G, Sorà F. Hematopoiesis and immune reconstitution after CD19 directed chimeric antigen receptor T-cells (CAR-T): A comprehensive review on incidence, risk factors and current management. Eur J Haematol 2024; 112:184-196. [PMID: 37491951 DOI: 10.1111/ejh.14052] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 07/27/2023]
Abstract
Impaired function of hematopoiesis after treatment with chimeric antigen T-cells (CAR-T) is a frequent finding and can interest a wide range of patients, regardless of age and underlying disease. Trilinear cytopenias, as well as hypogammaglobulinemia, B-cell aplasia, and T-cell impairment, can severely affect the infectious risk of CAR-T recipients, as well as their quality of life. In this review, we provide an overview of defects in hematopoiesis after CAR-T, starting with a summary of different definitions and thresholds. We then move to summarize the main pathogenetic mechanisms of cytopenias, and we offer insight into cytomorphological aspects, the role of clonal hematopoiesis, and the risk of secondary myeloid malignancies. Subsequently, we expose the major findings and reports on T-cell and B-cell quantitative and functional impairment after CAR-T. Finally, we provide an overview of current recommendations and leading experiences regarding the management of cytopenias and defective B- and T-cell function.
Collapse
Affiliation(s)
- Eugenio Galli
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alberto Fresa
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Silvia Bellesi
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Elisabetta Metafuni
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Elena Maiolo
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Ilaria Pansini
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Filippo Frioni
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francesco Autore
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Maria Assunta Limongiello
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Idanna Innocenti
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Sabrina Giammarco
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Patrizia Chiusolo
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gina Zini
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Federica Sorà
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
35
|
Infante MS, Nemirovsky D, Devlin S, DeWolf S, Tamari R, Dahi PB, Lee YJ, Chung DJ, Politikos I, Barker J, Giralt SA, Babady NE, Ramanathan L, Papanicolaou GA, Seo S, Kamboj M, Perales MA, Shah GL. Outcomes and Management of the SARS-CoV2 Omicron Variant in Recipients of Hematopoietic Cell Transplantation and Chimeric Antigen Receptor T Cell Therapy. Transplant Cell Ther 2024; 30:116.e1-116.e12. [PMID: 37806446 PMCID: PMC11220618 DOI: 10.1016/j.jtct.2023.09.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 10/10/2023]
Abstract
Hematopoietic cell transplantation (HCT) and chimeric antigen receptor T cell therapy (CAR-T) recipients who develop Coronavirus disease 2019 (COVID-19) can have decreased overall survival (OS), likely due to disease-inherent and therapy-related immunodeficiency. The availability of COVID-19-directed therapies and vaccines have improved COVID-19-related outcomes, but immunocompromised individuals remain vulnerable. Specifically, the effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variant infections, including Omicron and its sublineages, particularly in HCT recipients, remain to be defined. The aim of this study was to compare the impact of SARS-CoV-2 Omicron infections in HCT/CAR-T recipients with outcomes previously reported for ancestral SARS-CoV-2 infections early in the pandemic (March to June 2020). This was a retrospective analysis of adult HCT/CAR-T recipients diagnosed with COVID-19 at Memorial Sloan Kettering Cancer Center between July 2021 and July 2022. We identified 353 patients (172 autologous HCT recipients [49%], 152 allogeneic HCT recipients [43%], and 29 CAR-T recipients [8%]), with a median time from HCT/CAR-T to SARS-CoV-2 infection of 1010 days (interquartile range, 300 to 2046 days). Forty-one patients (12%) were diagnosed with COVID-19 during the delta wave, and 312 patients (88%) were diagnosed during the Omicron wave. Risk factors associated with increased odds of COVID-19-related hospitalization were the presence of 2 or more comorbidities (odds ratio [OR], 4.9; 95% confidence interval [CI], 2.4 to 10.7; P < .001), CAR-T therapy compared to allogeneic HCT (OR, 7.7; 95% CI, 3.0 to 20.0; P < .001), hypogammaglobulinemia (OR, 2.71; 95% CI, 1.06 to 6.40; P = .027), and age at COVID-19 diagnosis (OR, 1.03; 95% CI, 1.0 to 1.05; P = .04). In contrast, infection during the Omicron variant BA5/BA4-dominant period compared to variant BA1 (OR, .21; 95% CI, .03 to .73; P = .037) and more than 3 years from HCT/CAR-T therapy to COVID-19 diagnosis compared to early infection at <100 days (OR, .31; 95% CI, .12 to .79; P = .011) were associated with a decreased odds for hospitalization. The OS at 12 months from COVID-19 diagnosis was 89% (95% CI, 84% to 94%), with 6 of 26 deaths attributable to COVID-19. Patients with the ancestral strain of SAR-CoV-2 had a lower OS at 12 months, with 73% (95% CI, 62% to 84%) versus 89% (95% CI, 84% to 94%; P < .001) in the Omicron cohort. Specific COVID-19 treatment was administered in 62% of patients, and 84% were vaccinated with mRNA COVID-19 vaccines. Vaccinated patients had significantly better OS than unvaccinated patients (90% [95% CI, 86% to 95%] versus 82% [95% CI, 72% to 94%] at 12 months; P = .003). No significant difference in OS was observed in patients infected with the Omicron and those infected with the Delta variant (P = .4) or treated with specific COVID-19 treatments compared with those not treated (P = .2). We observed higher OS in HCT and CAR-T recipients infected with the Omicron variants compared to those infected with the ancestral strain of SARS-CoV2. The use of COVID-19 antivirals, mAbs, and vaccines might have contributed to the improved outcomes.
Collapse
Affiliation(s)
- Maria-Stefania Infante
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; University Hospital Infanta Leonor, Madrid, Spain.
| | - David Nemirovsky
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sean Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Susan DeWolf
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Roni Tamari
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Parastoo B Dahi
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Yeon Joo Lee
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - David J Chung
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Ioannis Politikos
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Juliet Barker
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Sergio A Giralt
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - N Esther Babady
- Clinical Microbiology Service, Department of Laboratory Medicine, Memorial SLoan Kettering Cancer Center, New York, New York
| | - Lakshmi Ramanathan
- Clinical Chemistry Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Genovefa A Papanicolaou
- Department of Medicine, Weill Cornell Medical College, New York, New York; Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Susan Seo
- Department of Medicine, Weill Cornell Medical College, New York, New York; Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mini Kamboj
- Department of Medicine, Weill Cornell Medical College, New York, New York; Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Gunjan L Shah
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
36
|
Rejeski K, Subklewe M, Locke FL. Recognizing, defining, and managing CAR-T hematologic toxicities. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:198-208. [PMID: 38066881 PMCID: PMC10727074 DOI: 10.1182/hematology.2023000472] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Autologous CAR-T cell therapy (CAR-T) has improved outcomes for patients with B-cell malignancies. It is associated with the well-described canonical toxicities cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), which may be abrogated by corticosteroids and the anti-IL6 receptor antagonist tocilizumab. Practitioners and researchers should be aware of additional toxicities. Here we review current understanding and management of hematologic toxicities after CAR-T, including cytopenias, coagulopathies, bleeding and clotting events, hemophagocytic-lymphohistiocytosis, and tumor lysis syndrome. We pay particular attention to cytopenias, recently termed immune effector cell-associated hematological toxicity (ICAHT). While the "H" is silent, hematotoxicity is not: ICAHT has the highest cumulative incidence of all immune adverse events following CAR-T. Early cytopenia (day 0-30) is closely linked to lymphodepleting chemotherapy and CRS-related inflammatory stressors. Late ICAHT (after day 30) can present either with or without antecedent count recovery (e.g., "intermittent" vs "aplastic" phenotype), and requires careful evaluation and management strategies. Growth factor support is the mainstay of treatment, with recent evidence demonstrating safety and feasibility of early granulocyte colony-stimulating factor (G-CSF) (e.g., within week 1). In G-CSF refractory cases, autologous stem cell boosts represent a promising treatment avenue, if available. The CAR-HEMATOTOX scoring system, validated for use across lymphoid malignancies (B-NHL, multiple myeloma), enables pretherapeutic risk assessment and presents the potential for risk-adapted management. Recent expert panels have led to diagnostic scoring criteria, severity grading systems, and management strategies for both ICAHT and the recently termed immune effector cell-associated hemophagocytic lymphohistiocytosis-like syndrome (IEC-HS), now clarified and defined as a distinct entity from CRS.
Collapse
Affiliation(s)
- Kai Rejeski
- Department of Medicine III, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marion Subklewe
- Department of Medicine III, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL
| |
Collapse
|
37
|
Epperly R, Giordani VM, Mikkilineni L, Shah NN. Early and Late Toxicities of Chimeric Antigen Receptor T-Cells. Hematol Oncol Clin North Am 2023; 37:1169-1188. [PMID: 37349152 PMCID: PMC10592597 DOI: 10.1016/j.hoc.2023.05.010] [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] [Indexed: 06/24/2023]
Abstract
As chimeric antigen receptor (CAR) T-cell therapy is increasingly integrated into clinical practice across a range of malignancies, identifying and treating inflammatory toxicities will be vital to success. Early experiences with CD19-targeted CAR T-cell therapy identified cytokine release syndrome and neurotoxicity as key acute toxicities and led to unified initiatives to mitigate the influence of these complications. In this section, we provide an update on the current state of CAR T-cell-related toxicities, with an emphasis on emerging acute toxicities affecting additional organ systems and considerations for delayed toxicities and late effects.
Collapse
Affiliation(s)
- Rebecca Epperly
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 1130, Memphis, TN 38105, USA
| | - Victoria M Giordani
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Building 10, Room 1W-3750, 9000 Rockville Pike MSC 1104, Bethesda, MD 20892, USA; Pediatric Hematology/Oncology, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Lekha Mikkilineni
- Blood and Marrow Transplantation & Cellular Therapy, Stanford University, Palo Alto, CA, USA; Stanford School of Medicine, 300 Pasteur Drive, Room H0101, Stanford, CA 94305, USA
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Building 10, Room 1W-3750, 9000 Rockville Pike MSC 1104, Bethesda, MD 20892, USA.
| |
Collapse
|
38
|
Lee C, Lin T, Yao M, Hsiao L, Ko B, Liu C, Chen T. Allogeneic hematopoietic stem cell transplantation for B-cell lymphoma in Taiwan. Cancer Med 2023; 12:21761-21769. [PMID: 38018321 PMCID: PMC10757116 DOI: 10.1002/cam4.6741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/05/2023] [Accepted: 11/13/2023] [Indexed: 11/30/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is considered for patients with high-risk B-cell lymphoma and relapsed or refractory disease. This study aimed to analyze the long-term follow-up data of patients who underwent allo-HSCT in Taiwan. This was a retrospective observational study using data from the Taiwan Society of Blood and Marrow Transplantation database. A total of 105 patients who underwent allo-HSCT because of high-risk, relapsed, or refractory disease between 2010 and 2019 were included. Forty-five percent of the patients previously underwent autologous stem cell transplantation (ASCT). The median follow-up duration was 18.6 months. The probability of 3-year progression-free survival and overall survival (OS) was 34.5% and 37%, respectively. The probability of 1-year non-relapse mortality was 31.4%, and the major cause was infection (75.8%). The multivariable analysis showed that not in remission at the time of transplantation and the absence of graft-versus-host disease (GVHD) were factors associated with inferior OS. The probability of 3-year OS in patients with diffuse large B-cell lymphoma who underwent allo-HSCT and allo-HSCT after ASCT was 40.2% and 25.2%, respectively. Allo-HSCT could be a salvage therapeutic option for relapsed or refractory B-cell lymphoma. Complete remission at the time of allo-HSCT and the presence of GVHD are independent variables for overall survival.
Collapse
Affiliation(s)
- Chun‐Hui Lee
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung UniversityTainanTaiwan
- Department of OncologyNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainanTaiwan
| | - Tzu‐Chien Lin
- Department of OncologyNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainanTaiwan
- Division of Hematology, Department of Internal MedicineNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainanTaiwan
| | - Ming Yao
- Division of Hematology, Department of Internal MedicineNational Taiwan University Hospital, College of MedicineTaipeiTaiwan
| | - Liang‐Tsai Hsiao
- Division of Hematology, Department of Internal MedicineTaipei Veterans General HospitalTaipeiTaiwan
| | - Bor‐Sheng Ko
- Division of Hematology, Department of Internal MedicineNational Taiwan University Hospital, College of MedicineTaipeiTaiwan
- Department of Hematological OncologyNational Taiwan University Cancer CenterTaipeiTaiwan
| | - Chia‐Jen Liu
- Division of Hematology, Department of Internal MedicineTaipei Veterans General HospitalTaipeiTaiwan
- Institute of Emergency and Critical Care Medicine, School of MedicineNational Yang‐Ming Chiao Tung UniversityTaipeiTaiwan
| | - Tsai‐Yun Chen
- Division of Hematology, Department of Internal MedicineNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainanTaiwan
- Center for Cell TherapyNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainanTaiwan
| |
Collapse
|
39
|
Liu Q, Hu T, Li H, Shen Y, Wu D, Ye B. Prolonged haematologic toxicity in CAR-T-cell therapy: A review. J Cell Mol Med 2023; 27:3662-3671. [PMID: 37702530 PMCID: PMC10718150 DOI: 10.1111/jcmm.17930] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 09/14/2023] Open
Abstract
Chimeric antigen receptor-T-cell (CAR-T-cell) therapy is a novel immunotherapy with encouraging results for treatment of relapsed/refractory haematologic malignancies. With increasing use, our understanding of immune-mediated side effects such as cytokine release syndrome and neurotoxicity has improved; nevertheless, prolonged haematologic toxicity (PHT), with a high incidence rate, remains underrecognized. Owing to heterogeneity in populations, the CAR-T cells used and diseases treated as well as differences in the definition of PHT, its rate, risk factors and management vary across studies. In this review, we provide a narrative of PHT occurring in patients following CAR-T-cell therapy; evidence of PHT treatment strategies is also presented, with the aim of contributing to systematic understanding of PHT.
Collapse
Affiliation(s)
- Qi Liu
- Department of HematologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
- The First School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouChina
| | - Tonglin Hu
- Department of HematologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
- The First School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouChina
| | - Hangchao Li
- Department of HematologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
- The First School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouChina
| | - Yingying Shen
- Department of HematologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
- The First School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouChina
| | - Dijiong Wu
- Department of HematologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
- The First School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouChina
| | - Baodong Ye
- Department of HematologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
- The First School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouChina
| |
Collapse
|
40
|
Strati P, Spiotto MT. Incorporating Immunotherapy with Radiotherapy for Lymphomas. LYMPHATICS 2023; 1:273-286. [PMID: 39917366 PMCID: PMC11800356 DOI: 10.3390/lymphatics1030018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
Abstract
Radiotherapy and/or chemotherapy have been used for nearly 100 years to treat lymphoma. Recently, immunotherapy has been incorporated into the treatment of lymphomas. Here, we will review both the role of immunotherapy in lymphoma as well as the feasibility of incorporating immunotherapies with conventional lymphoma treatments, especially radiotherapy. Immunotherapy agents include checkpoint inhibitors that target the PD-1/PD-L1 axis, CTLA-4, or CD47. In addition, other immunotherapy agents such as bi-specific antibodies and CD19 CAR-T cell therapy are being implemented in various non-Hodgkin's lymphomas. Extrapolating from observations in other disease sites and incorporating immunotherapy with conventional treatments of lymphoma, including radiotherapy, may have opposing effects. Radiotherapy may stimulate anti-tumor immune responses that synergize with immunotherapies. In contrast, radiotherapy, as well as chemotherapy, may also induce local and systemic immune dysfunction which reduces the efficacy of immunotherapies. With newer radiation treatment techniques and limited radiation fields, it is likely that the efficacy of immunotherapy can be maintained when included with conventional treatments. Therefore, there remains an unmet need to better understand the role of immunotherapy alone and in combination with current treatments in lymphoma patients.
Collapse
Affiliation(s)
- Paolo Strati
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael T. Spiotto
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
41
|
Walker B, Zimmer AJ, Stohs EJ, Lunning M, Lyden E, Abbas A. Infectious complications among CD19 CAR-T cell therapy recipients: A single-center experience. Transpl Infect Dis 2023; 25 Suppl 1:e14191. [PMID: 37987114 DOI: 10.1111/tid.14191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/04/2023] [Accepted: 10/30/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND CD19 chimeric antigen receptor (CAR)-T cell therapy has emerged as an effective treatment in those with refractory or relapsed lymphoma. CD19 CAR-T cell therapy can cause direct and indirect toxic adverse effects and increased risk for infection. Infectious complications and optimal antimicrobial prophylaxis strategies are an ongoing area of investigation. METHODS A single-center retrospective cohort study was conducted to review recipients of CD19 CAR-T cell therapy between April 2018 and December 2020. Patient characteristics and clinical outcomes were extracted from the electronic health records. RESULTS Infectious complications were identified in 18/50 (36%) recipients with 31 episodes of infection. The median time to infection was 225 days (range 0-614). Bacterial infections were most common with bloodstream infection followed by sinusitis and skin and soft tissue infection. Eight viral infections were identified, most being respiratory viral illnesses. Two fungal infections were identified: Pneumocystis jirovecii pneumonia (PJP) and disseminated fusariosis. Seventeen infections (54.8%) were classified as severe: leading to death, requiring hospitalization, need for empiric intravenous antibiotics, or significant alteration in hospital course. No characteristics were found to be statistically significant risks for infection, although a trend toward significance was seen in prior autologous stem cell transplant recipients (p = .12) and those with recurrent neutropenia (p = .14). Three patients (6%) died from infection. CONCLUSION Infections were common after CD19 CAR-T cell therapy and occurred beyond the first year. Further multicenter studies are needed to define infectious risks and optimize antimicrobial prophylaxis recommendations in recipients of CD19 CAR-T cell therapy.
Collapse
Affiliation(s)
- Bryan Walker
- Division of Infectious Disease, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Andrea J Zimmer
- Division of Infectious Disease, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Erica J Stohs
- Division of Infectious Disease, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Matthew Lunning
- Division of Hematology and Oncology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Elizabeth Lyden
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Anum Abbas
- Division of Infectious Disease, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
42
|
Kampouri E, Little JS, Rejeski K, Manuel O, Hammond SP, Hill JA. Infections after chimeric antigen receptor (CAR)-T-cell therapy for hematologic malignancies. Transpl Infect Dis 2023; 25 Suppl 1:e14157. [PMID: 37787373 DOI: 10.1111/tid.14157] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/30/2023] [Accepted: 09/10/2023] [Indexed: 10/04/2023]
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-T-cell therapies have revolutionized the management of acute lymphoblastic leukemia, non-Hodgkin lymphoma, and multiple myeloma but come at the price of unique toxicities, including cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, and long-term "on-target off-tumor" effects. METHODS All of these factors increase infection risk in an already highly immunocompromised patient population. Indeed, infectious complications represent the key determinant of non-relapse mortality after CAR-T cells. The temporal distribution of these risk factors shapes different infection patterns early versus late post-CAR-T-cell infusion. Furthermore, due to the expression of their targets on B lineage cells at different stages of differentiation, CD19, and B-cell maturation antigen (BCMA) CAR-T cells induce distinct immune deficits that could require different prevention strategies. Infection incidence is the highest during the first month post-infusion and subsequently decreases thereafter. However, infections remain relatively common even a year after infusion. RESULTS Bacterial infections predominate early after CD19, while a more equal distribution between bacterial and viral causes is seen after BCMA CAR-T-cell therapy, and fungal infections are universally rare. Cytomegalovirus (CMV) and other herpesviruses are increasingly breported, but whether routine monitoring is warranted for all, or a subgroup of patients, remains to be determined. Clinical practices vary substantially between centers, and many areas of uncertainty remain, including CMV monitoring, antibacterial and antifungal prophylaxis and duration, use of immunoglobulin replacement therapy, and timing of vaccination. CONCLUSION Risk stratification tools are available and may help distinguish between infectious and non-infectious causes of fever post-infusion and predict severe infections. These tools need prospective validation, and their integration in clinical practice needs to be systematically studied.
Collapse
Affiliation(s)
- Eleftheria Kampouri
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Infectious Diseases Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jessica S Little
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Division of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Kai Rejeski
- Department of Medicine III-Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Munich site, and German Cancer Research Center, Heidelberg, Germany
| | - Oriol Manuel
- Infectious Diseases Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Sarah P Hammond
- Division of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Divisions of Hematology/Oncology and Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Joshua A Hill
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
43
|
Li A, Feng R. [CAR-T cell therapy-related long-term cytopenias]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2023; 44:870-875. [PMID: 38049346 PMCID: PMC10694071 DOI: 10.3760/cma.j.issn.0253-2727.2023.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Indexed: 12/06/2023]
Affiliation(s)
- A Li
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - R Feng
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
44
|
Kawata K, Shima H, Shinjoh M, Yamazaki F, Kurosawa T, Yaginuma M, Takada H, Shimada H. Pneumocystis jirovecii pneumonia after CD4+ T-cell recovery subsequent to CD19-targeted chimeric antigen receptor T-cell therapy: A case report and brief review of literature. Cancer Rep (Hoboken) 2023; 6:e1885. [PMID: 37563749 PMCID: PMC10598253 DOI: 10.1002/cnr2.1885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/20/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND CD19-targeted chimeric antigen receptor (CAR)-T cell therapy involves administration of patient-derived T cells that target B cells, resulting in B-cell depletion and aplasia. In immunity against Pneumocystis jirovecii (Pj), CD4+ T cells and, more recently, B cells, are generally considered important. Antigen presentation by B cells to CD4+ T cells is particularly important. Trimethoprim-sulfamethoxazole (TMP/SMX) for Pj pneumonia (PJP) prophylaxis is generally discontinued when the CD4+ T-cell count is >200/μL. Here we report the first case, to our knowledge, of PJP in a patient with a CD4+ T cell count of >200/μL after CAR-T cell therapy. CASE A 14-year-old girl developed hemophagocytic lymphohistiocytosis (HLH) after cord blood transplantation (CBT) for relapsed precursor B-cell acute lymphoblastic leukemia (B-ALL). Twenty-one months after CBT, she was diagnosed with combined second relapse in the bone marrow and central nervous system. The patient was treated with CD19-targeted CAR-T cell therapy for the relapse. After CAR-T cell therapy, the patient remained in remission and continued to receive TMP/SMX for PJP prophylaxis. Seven months after CAR-T cell therapy, CD4+ T cells recovered and TMP/SMX was discontinued. The B-cell aplasia persisted. Ten months after CAR-T cell therapy, the patient developed PJP. The patient was also considered to have macrophage hyperactivation at the onset of PJP. Treatment with immunoglobulin, TMP/SMX, and prednisolone was initiated, and the patient's symptoms rapidly ameliorated. CONCLUSION The patient in the present case developed PJP despite a CD4+ T-cell count of >200/μL after CAR-T cell therapy, probably because of inadequate CD4+ T-cell activation caused by B-cell depletion after CAR-T cell therapy and repeated abnormal macrophage immune responses after CBT. It is important to determine the duration of TMP/SMX for prophylaxis after CAR-T cell therapy according to each case, as well as the CD4+ T-cell count.
Collapse
Affiliation(s)
- Kento Kawata
- Department of PediatricsKeio University School of MedicineTokyoJapan
| | - Haruko Shima
- Department of PediatricsKeio University School of MedicineTokyoJapan
| | - Masayoshi Shinjoh
- Department of PediatricsKeio University School of MedicineTokyoJapan
| | - Fumito Yamazaki
- Department of PediatricsKeio University School of MedicineTokyoJapan
| | - Takumi Kurosawa
- Department of PediatricsKeio University School of MedicineTokyoJapan
| | - Mizuki Yaginuma
- Department of PediatricsKeio University School of MedicineTokyoJapan
| | - Hiroshi Takada
- Department of PediatricsKeio University School of MedicineTokyoJapan
| | - Hiroyuki Shimada
- Department of PediatricsKeio University School of MedicineTokyoJapan
| |
Collapse
|
45
|
Penack O, Peczynski C, Koenecke C, Polge E, Sanderson R, Yakoub-Agha I, Fegueux N, Daskalakis M, Collin M, Dreger P, Kröger N, Schanz U, Bloor A, Ganser A, Besley C, Wulf GG, Novak U, Moiseev I, Schoemans H, Basak GW, Chabannon C, Sureda A, Glass B, Peric Z. Organ complications after CD19 CAR T-cell therapy for large B cell lymphoma: a retrospective study from the EBMT transplant complications and lymphoma working party. Front Immunol 2023; 14:1252811. [PMID: 37828980 PMCID: PMC10565347 DOI: 10.3389/fimmu.2023.1252811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023] Open
Abstract
We investigated ≥ grade 3 (CTC-AE) organ toxicities for commercial CD19 chimeric antigen receptor T cell (CAR-T cell) products in 492 patients (Axi-Cel; n = 315; Tisa-Cel; n = 177) with Large B-cell Lymphoma in the European Society for Blood and Marrow Transplantation (EBMT) CAR-T registry. The incidence of ≥ grade 3 organ toxicities during the first 100 days after CAR-T was low and the most frequent were: renal (3.0%), cardiac (2.3%), gastro-intestinal (2.3%) and hepatic (1.8%). The majority occurred within three weeks after CAR-T cell therapy. Overall survival was 83.1% [79.8-86.5; 95% CI] at 3 months and 53.5% [49-58.4; 95% CI] at one year after CAR-T. The most frequent cause of death was tumour progression (85.1%). Non-relapse mortality was 3.1% [2.3-4.1; 95% CI] at 3 months and 5.2% [4.1-6.5; 95% CI] at one year after CAR-T. The most frequent causes of non-relapse mortality were cell-therapy-related toxicities including organ toxicities (6.4% of total deaths) and infections (4.4% of total deaths). Our data demonstrates good safety in the European real-world setting.
Collapse
Affiliation(s)
- Olaf Penack
- Medical Clinic, Department for Haematology, Oncology and Tumorimmunology, Charité Universitätsmedizin Berlin, Berlin, Germany
- EBMT Transplant Complications Working Party, Paris, France
| | - Christophe Peczynski
- EBMT Transplant Complications Working Party, Paris, France
- EBMT Paris Study Office, Department of Haematology, Saint Antoine Hospital, Paris, France
- INSERM UMR-S 938, Sorbonne University, Paris, France
| | - Christian Koenecke
- EBMT Transplant Complications Working Party, Paris, France
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Emmanuelle Polge
- EBMT Transplant Complications Working Party, Paris, France
- EBMT Paris Study Office, Department of Haematology, Saint Antoine Hospital, Paris, France
- INSERM UMR-S 938, Sorbonne University, Paris, France
| | - Robin Sanderson
- Kings College Hospital, Departement of Haematological Medicine, London, United Kingdom
| | | | - Nathalie Fegueux
- CHU Lapeyronie, Département d`Hématologie Clinique, Montpellier, France
| | - Michael Daskalakis
- Department of Hematology, University Hospital Bern, Bern, Switzerland
- Department of Oncology, University Hospital Bern, Bern, Switzerland
| | - Matthew Collin
- Adult HSCT Unit, Northern Centre for Bone Marrow Transplantation, Newcastle upon Tyne, United Kingdom
| | - Peter Dreger
- Department of Hematology, University of Heidelberg, Heidelberg, Germany
| | - Nicolaus Kröger
- Bone Marrow Transplantation Centre, University Hospital Eppendorf, Hamburg, Germany
| | - Urs Schanz
- Clinic of Hematology, University Hospital, Zurich, Switzerland
| | - Adrian Bloor
- Christie NHS Trust Hospital, Adult Leukaemia and Bone Marrow Transplant Unit, Manchester, United Kingdom
| | - Arnold Ganser
- Department of Haematology, Hemostasis, Oncology, Hannover Medical School, Hannover, Germany
| | - Caroline Besley
- Department of Paediatric Oncology, Bristol Royal Hospital for Children, Bristol, United Kingdom
- Department of BMT, Bristol Royal Hospital for Children, Bristol, United Kingdom
| | - Gerald G. Wulf
- Universitaetsmedizin Goettingen, Klinik für Hämatologie und Medizinische Onkologie, Göttingen, Germany
| | - Urban Novak
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ivan Moiseev
- EBMT Transplant Complications Working Party, Paris, France
- Department of Hematology, First Pavlov State Medical University of St. Petersburg, St. Petersburg, Russia
| | - Hélène Schoemans
- EBMT Transplant Complications Working Party, Paris, France
- Department of Hematology, University Hospitals Leuven and KU Leuven, Leuven, Belgium
| | - Grzegorz W. Basak
- EBMT Transplant Complications Working Party, Paris, France
- Department of Hematology, Oncology and Internal Medicine, The Medical University of Warsaw, Warsaw, Poland
| | - Christian Chabannon
- EBMT Cellular Therapy and Immunobiology Working Party, Leiden, Netherlands
- Institut Paoli-Calmettes Comprehensive Cancer Centre, Inserm CBT-1409, Aix-Marseille Université, Marseille, France
| | - Anna Sureda
- Clinical Hematology Department, Institut Català d’Oncologia-Hospitalet, Barcelona, Spain
- Institut de Ciències Biomèdiques de Bellvitge (IDIBELL), Universitat de Barcelona, Barcelona, Spain
| | - Bertram Glass
- EBMT Lymphoma Working Party, Leiden, Netherlands
- Department of Hematology, Oncology, and Tumor ImmunologyKlinikum Berlin-Buch, Helios, Berlin, Germany
| | - Zinaida Peric
- EBMT Transplant Complications Working Party, Paris, France
- University Hospital Centre Zagreb and School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
46
|
Strati P, Gregory T, Majhail NS, Jain N. Chimeric Antigen Receptor T-Cell Therapy for Hematologic Malignancies: A Practical Review. JCO Oncol Pract 2023; 19:706-713. [PMID: 37406255 DOI: 10.1200/op.22.00819] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/09/2023] [Accepted: 05/24/2023] [Indexed: 07/07/2023] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has become an established therapeutic approach for the treatment of hematologic malignancies. The field continues to evolve rapidly and newer-generation constructs are being designed to enhance proliferative capacity, and achieve long-term persistence and greater efficacy with an overall lower incidence of toxicity. Initial clinical application of CAR-T therapies has focused on relapsed and/or refractory hematologic malignancies, and Food and Drug Administration-approved CAR-T products targeting CD19 are available for B-cell acute lymphoblastic leukemia and low- and high-grade B-cell non-Hodgkin lymphoma, and targeting B-cell maturation antigen are available for multiple myeloma. Cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome have been recognized as class specific toxicities associated with these novel therapies. In this review, we focus on the clinical application of CAR-T therapies in adult patients with hematologic malignancies, including access issues, outpatient administration, and appropriate timing for referring a patient to a CAR-T treatment center.
Collapse
Affiliation(s)
- Paolo Strati
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Tara Gregory
- Colorado Blood Cancer Institute, Denver, CO
- Sarah Cannon Transplant and Cellular Therapy Program at Presbyterian/St Luke's Medical Center, Denver, CO
| | - Navneet S Majhail
- Sarah Cannon, Nashville, TN
- Sarah Cannon Transplant and Cellular Therapy Program at TriStar Centennial, Nashville, TN
| | - Nitin Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
47
|
Dhaliwal A, Ravi S. Myelodysplastic Syndrome After Anti-CD19 Chimeric Antigen Receptor T-cell Therapy: A Case Series. Cureus 2023; 15:e44677. [PMID: 37809221 PMCID: PMC10550779 DOI: 10.7759/cureus.44677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2023] [Indexed: 10/10/2023] Open
Abstract
The utility of CD19-targeted chimeric antigen receptor T-cell (CAR-T cell) therapy in the management of refractory/relapsed B-cell malignancies has increased tremendously in recent times. In addition to cytokine release syndrome (CRS), neurotoxicity, and infections, CAR-T cell patients develop cytopenias, with about 15% of the patients continuing to have severe cytopenias up to three months after treatment. Retrospective reviews have reported the development of myelodysplastic syndrome (MDS) in patients undergoing CAR-T cell therapy. Here, we describe four cases of MDS and/or clonal cytopenias of undetermined significance (CCUS), developing after CAR-T cell therapy. A retrospective review of four patients with relapsed/refractory B-cell lymphomas treated with CD19-directed autologous CAR-T cell was conducted at our institution. The median age was 72.5 years (range 63-76). Three of the four patients had double-hit diffuse large B-cell lymphoma (DLBCL). The median number of lines of therapy before CAR-T cell was three. Only one patient had a prior autologous stem cell transplant (ASCT). The median time to diagnosis of MDS/CCUS from CAR-T cell therapy was three months. Two cases of CCUS diagnosed were at one- and two-month post-CAR-T cell, and two cases of MDS were diagnosed at 10 and 26 months. None of the patients had dysplastic clones before the initiation of CAR-T cell therapy. Only one patient was found to have CCUS-developed CRS post-CAR-T cell requiring treatment with tocilizumab and steroids. Three patients showed complete response, with one showing a very good partial response. All the patients were in remission with no additional therapies post-CAR-T cell. One patient died secondary to COVID-19-related complications. Four patients with prolonged cytopenias were found to have either MDS or CCUS after CAR-T cell therapy. Two CCUS cases underwent bone marrow evaluation early in the course of cytopenias and may develop into MDS, acute myeloid leukemia (AML), or myeloproliferative neoplasm over time. Our retrospective case series review, compared to previous studies, constitutes of patients with no prior clonal hematopoiesis-related cytogenetic abnormalities, fewer lines of therapy, and only one patient with previous hematopoietic stem cell transplantation (HSCT). Based on the upcoming data and our review, a bone marrow biopsy with next-generation sequencing (NGS) is imperative in patients with prolonged cytopenias after CAR-T cell therapy. A diagnosis of CCUS/MDS in these cases can help guide treatment.
Collapse
Affiliation(s)
- Armaan Dhaliwal
- Department of Internal Medicine, University of Arizona College of Medicine, Tucson, USA
| | - Soumiya Ravi
- Department of Internal Medicine, University of Arizona College of Medicine, Tucson, USA
| |
Collapse
|
48
|
Strati P, Li X, Deng Q, Marques-Piubelli ML, Henderson J, Watson G, Deaton L, Cain T, Yang H, Ravanmehr V, Fayad LE, Iyer SP, Nastoupil LJ, Hagemeister FB, Parra ER, Saini N, Takahashi K, Fowler NH, Westin JR, Steiner RE, Nair R, Flowers CR, Wang L, Ahmed S, Al-Atrash G, Vega F, Neelapu SS, Green MR. Prolonged cytopenia following CD19 CAR T cell therapy is linked with bone marrow infiltration of clonally expanded IFNγ-expressing CD8 T cells. Cell Rep Med 2023; 4:101158. [PMID: 37586321 PMCID: PMC10439270 DOI: 10.1016/j.xcrm.2023.101158] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 06/13/2023] [Accepted: 07/18/2023] [Indexed: 08/18/2023]
Abstract
Autologous anti-CD19 chimeric antigen receptor T cell (CAR T) therapy is highly effective in relapsed/refractory large B cell lymphoma (rrLBCL) but is associated with toxicities that delay recovery. While the biological mechanisms of cytokine release syndrome and neurotoxicity have been investigated, the pathophysiology is poorly understood for prolonged cytopenia, defined as grade ≥3 cytopenia lasting beyond 30 days after CAR T infusion. We performed single-cell RNA sequencing of bone marrow samples from healthy donors and rrLBCL patients with or without prolonged cytopenia and identified significantly increased frequencies of clonally expanded CX3CR1hi cytotoxic T cells, expressing high interferon (IFN)-γ and cytokine signaling gene sets, associated with prolonged cytopenia. In line with this, we found that hematopoietic stem cells from these patients expressed IFN-γ response signatures. IFN-γ deregulates hematopoietic stem cell self-renewal and differentiation and can be targeted with thrombopoietin agonists or IFN-γ-neutralizing antibodies, highlighting a potential mechanism-based approach for the treatment of CAR T-associated prolonged cytopenia.
Collapse
Affiliation(s)
- Paolo Strati
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xubin Li
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qing Deng
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mario L Marques-Piubelli
- Department Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jared Henderson
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Grace Watson
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Laurel Deaton
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Taylor Cain
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Haopeng Yang
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vida Ravanmehr
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luis E Fayad
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Swaminathan P Iyer
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Loretta J Nastoupil
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Frederick B Hagemeister
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Edwin R Parra
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neeraj Saini
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Koichi Takahashi
- Department Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nathan H Fowler
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason R Westin
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Raphael E Steiner
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ranjit Nair
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher R Flowers
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sairah Ahmed
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gheath Al-Atrash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Francisco Vega
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sattva S Neelapu
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Michael R Green
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
49
|
Zeng K, Huang M, Lyu MA, Khoury JD, Ahmed S, Patel KK, Dropulić B, Reese-Koc J, Caimi PF, Sadeghi T, Lima MD, Flowers CR, Parmar S. Adjunct Therapy with T Regulatory Cells Decreases Inflammation and Preserves the Anti-Tumor Activity of CAR T Cells. Cells 2023; 12:1880. [PMID: 37508543 PMCID: PMC10377823 DOI: 10.3390/cells12141880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
With greater accessibility and an increased number of patients being treated with CAR T cell therapy, real-world toxicity continues to remain a significant challenge to its widespread adoption. We have previously shown that allogeneic umbilical cord blood-derived (UCB) regulatory T cells (Tregs) can resolve inflammation and treat acute and immune-mediated lung injuries. Allogeneic, cryopreserved UCB Tregs have shown a clinical benefit in patients suffering from COVID-19 acute respiratory distress syndrome. The unique properties of UCB Treg cells include a lack of plasticity under inflammatory micro-environments, no requirement for HLA matching, a long shelf life of cryopreserved cells, and immediate product availability, which makes them attractive for treating acute inflammatory syndromes. Therefore, we hypothesized that adjunct therapy with UCB Tregs may resolve the undesirable inflammation responsible for CAR T cell therapy-associated toxicity. In in vitro analysis, no interference from the addition of UCB Tregs was observed on CD19 CAR T cells' ability to kill CD19 Raji cells at different CAR T: Raji cell ratios of 8:1 (80.4% vs. 81.5%); 4:1 (62.0% vs. 66.2%); 2:1 (50.1% vs. 54.7%); and 1:1 (35.4% vs. 44.1%). In the xenogeneic B-cell lymphoma model, multiple injections of UCB Tregs were administered 3 days after CD19 CAR T cell injection, and no detrimental effect of add-on Tregs was noted on the circulating CD8+ T effector cells. The distribution of CAR T cells in multiple organs remained unaffected by the addition of the UCB Tregs. Specifically, no difference in the overall tumor burden was detected between the UCB Treg + CAR T vs. CAR T alone recipients. No tumor was detected in the liver or bone marrow in CAR T cells + UCB Tregs recipients, with a notable corresponding decrease in multiple circulating inflammatory cytokines when compared to CART alone recipients. Here we show the proof of concept for adjunct therapy with UCB Tregs to mitigate the hyper-inflammatory state induced by CAR T cells without any interference in their on-target anti-tumor activity. Administration of UCB Tregs after CAR T cells allows sufficient time for their synapse formation with tumor cells and exerts cytotoxicity, such that the UCB Tregs are diverted to interact with the antigen-presenting cells at the site of inflammation. Such a differential distribution of cells would allow for a two-pronged strategy of a UCB Treg "cooling blanket" effect and lay the groundwork for clinical study.
Collapse
Affiliation(s)
- Ke Zeng
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Meixian Huang
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mi-Ae Lyu
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Joseph D Khoury
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sairah Ahmed
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Krina K Patel
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Jane Reese-Koc
- Department of Cellular Therapy, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Paolo F Caimi
- Department of Hematology and Medical Oncology, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | - Marcos de Lima
- Division of Hematology, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Christopher R Flowers
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Simrit Parmar
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
50
|
Westin JR, Oluwole OO, Kersten MJ, Miklos DB, Perales MA, Ghobadi A, Rapoport AP, Sureda A, Jacobson CA, Farooq U, van Meerten T, Ulrickson M, Elsawy M, Leslie LA, Chaganti S, Dickinson M, Dorritie K, Reagan PM, McGuirk J, Song KW, Riedell PA, Minnema MC, Yang Y, Vardhanabhuti S, Filosto S, Cheng P, Shahani SA, Schupp M, To C, Locke FL. Survival with Axicabtagene Ciloleucel in Large B-Cell Lymphoma. N Engl J Med 2023; 389:148-157. [PMID: 37272527 DOI: 10.1056/nejmoa2301665] [Citation(s) in RCA: 192] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
BACKGROUND In an analysis of the primary outcome of this phase 3 trial, patients with early relapsed or refractory large B-cell lymphoma who received axicabtagene ciloleucel (axi-cel), an autologous anti-CD19 chimeric antigen receptor T-cell therapy, as second-line treatment had significantly longer event-free survival than those who received standard care. Data were needed on longer-term outcomes. METHODS In this trial, we randomly assigned patients with early relapsed or refractory large B-cell lymphoma in a 1:1 ratio to receive either axi-cel or standard care (two to three cycles of chemoimmunotherapy followed by high-dose chemotherapy with autologous stem-cell transplantation in patients who had a response). The primary outcome was event-free survival, and key secondary outcomes were response and overall survival. Here, we report the results of the prespecified overall survival analysis at 5 years after the first patient underwent randomization. RESULTS A total of 359 patients underwent randomization to receive axi-cel (180 patients) or standard care (179 patients). At a median follow-up of 47.2 months, death had been reported in 82 patients in the axi-cel group and in 95 patients in the standard-care group. The median overall survival was not reached in the axi-cel group and was 31.1 months in the standard-care group; the estimated 4-year overall survival was 54.6% and 46.0%, respectively (hazard ratio for death, 0.73; 95% confidence interval [CI], 0.54 to 0.98; P = 0.03 by stratified two-sided log-rank test). This increased survival with axi-cel was observed in the intention-to-treat population, which included 74% of patients with primary refractory disease and other high-risk features. The median investigator-assessed progression-free survival was 14.7 months in the axi-cel group and 3.7 months in the standard-care group, with estimated 4-year percentages of 41.8% and 24.4%, respectively (hazard ratio, 0.51; 95% CI, 0.38 to 0.67). No new treatment-related deaths had occurred since the primary analysis of event-free survival. CONCLUSIONS At a median follow-up of 47.2 months, axi-cel as second-line treatment for patients with early relapsed or refractory large B-cell lymphoma resulted in significantly longer overall survival than standard care. (Funded by Kite; ZUMA-7 ClinicalTrials.gov number, NCT03391466.).
Collapse
Affiliation(s)
- Jason R Westin
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Olalekan O Oluwole
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Marie José Kersten
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - David B Miklos
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Miguel-Angel Perales
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Armin Ghobadi
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Aaron P Rapoport
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Anna Sureda
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Caron A Jacobson
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Umar Farooq
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Tom van Meerten
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Matthew Ulrickson
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Mahmoud Elsawy
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Lori A Leslie
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Sridhar Chaganti
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Michael Dickinson
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Kathleen Dorritie
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Patrick M Reagan
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Joseph McGuirk
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Kevin W Song
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Peter A Riedell
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Monique C Minnema
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Yin Yang
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Saran Vardhanabhuti
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Simone Filosto
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Paul Cheng
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Shilpa A Shahani
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Marco Schupp
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Christina To
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| | - Frederick L Locke
- From University of Texas M.D. Anderson Cancer Center, Houston (J.R.W.); Vanderbilt-Ingram Cancer Center, Nashville (O.O.O.); Amsterdam University Medical Center (UMC), University of Amsterdam, Cancer Center Amsterdam, Amsterdam (M.J.K.), UMC Groningen, Groningen (T.M.), and UMC Utrecht, Utrecht (M.C.M.) - all in the Netherlands; Stanford University School of Medicine, Stanford (D.B.M.), and Kite, Santa Monica (Y.Y., S.V., S.F., P.C., S.A.S., M.S., C.T.) - both in California; Memorial Sloan Kettering Cancer Center, New York (M.-A.P.), and University of Rochester School of Medicine, Rochester (P.M.R.) - both in New York; Washington University School of Medicine, St. Louis (A.G.); Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore (A.P.R.); Servei d'Hematologia Clínica, Institut Català d'Oncologia-Hospitalet, Institut de Recerca Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona (A.S.B.); Dana-Farber Cancer Institute, Boston (C.A.J.); University of Iowa, Iowa City (U.F.); Banner M.D. Anderson Cancer Center, Gilbert, AZ (M.U.); the Division of Hematology and Hematologic Oncology, Department of Medicine, Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS (M.E.), and Vancouver General Hospital, BC Cancer, University of British Columbia, Vancouver (K.W.S.) - both in Canada; John Theurer Cancer Center, Hackensack, NJ (L.A.L.); the Centre for Clinical Haematology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom (S.C.); Peter MacCallum Cancer Centre, Royal Melbourne Hospital, and the University of Melbourne, Melbourne (M.D.); UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh (K.D.); University of Kansas Cancer Center, Kansas City (J.M.); David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago (P.A.R.); and Moffitt Cancer Center, Tampa, FL (F.L.L.)
| |
Collapse
|