51
|
Li L, Gao J, Sun Z, Li X, Wang N, Zhang R. Effects of CAR-T Cell Therapy on Immune Cells and Related Toxic Side Effect Analysis in Patients with Refractory Acute Lymphoblastic Leukemia. Mediators Inflamm 2023; 2023:2702882. [PMID: 37304661 PMCID: PMC10257545 DOI: 10.1155/2023/2702882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 05/08/2023] [Accepted: 05/17/2023] [Indexed: 06/13/2023] Open
Abstract
Objective To observe the effects of chimeric antigen receptor T (CAR-T) cell immunotherapy on immune cells and related toxic side effects in patients with refractory acute lymphoblastic leukemia (ALL). Methods A retrospective study was conducted in 35 patients with refractory ALL. The patients were treated with CAR-T cell therapy in our hospital from January 2020 to January 2021. The efficacy was evaluated at one and three months post treatments. The venous blood of the patients was collected before treatment, 1 month after treatment, and 3 months after treatment. The percentage of regulatory T cells (Treg cells), natural killer (NK) cells, and T lymphocyte subsets (CD3+, CD4+, and CD8+ T cells) was detected by flow cytometry. The ratio of CD4+/CD8+ was calculated. Patient's toxic side effects such as fever, chills, gastrointestinal bleeding, nervous system symptoms, digestive system symptoms, abnormal liver function, and blood coagulation dysfunction were monitored and recorded. The incidence of toxic and side effects was calculated, and the incidence of infection was recorded. Results After one month of CAR-T cell therapy in 35 patients with ALL, the efficacy evaluation showed that complete response (CR) patients accounted for 68.57%, CR with incomplete hematological recovery (CRi) patients accounted for 22.86%, and partial disease (PD) patients accounted for 8.57%, and the total effective rate was 91.43%. In addition, compared with that before treatment, the Treg cell level in CR+CRi patients treated for 1 month and 3 months decreased prominently, and the NK cell level increased dramatically (P < 0.05). Compared with that before treatment, the levels of CD3+, CD4+, and CD4+/CD8+ in patients with CR+CRi in the 1-month and 3-month groups were markedly higher, and the levels of CD4+/CD8+ in the 3-month group were memorably higher than those in the 1-month group (P < 0.05). During CAR-T cell therapy in 35 patients with ALL, fever accounted for 62.86%, chills for 20.00%, gastrointestinal bleeding for 8.57%, nervous system symptoms for 14.29%, digestive system symptoms for 28.57%, abnormal liver function for 11.43%, and coagulation dysfunction for 8.57%. These side effects were all relieved after symptomatic treatment. During the course of CAR-T therapy in 35 patients with ALL, 2 patients had biliary tract infection and 13 patients had lung infection. No correlations were found between the infection and age, gender, CRS grade, usage of glucocorticoids or tocilizumab, and laboratory indicators such as WBC, ANC, PLT, and Hb (P > 0.05). Conclusion CAR-T cell therapy had a good effect on patients with refractory ALL by regulating the immune function of the body via mediating the content of immune cells. CAR-T cell therapy may have therapeutic effect on refractory ALL patients with mild side effects and high safety.
Collapse
Affiliation(s)
- Lianlian Li
- Department of Hematology, Cangzhou People's Hospital, Cangzhou City, Hebei Province, China
| | - Jie Gao
- Department of Hematology, Cangzhou People's Hospital, Cangzhou City, Hebei Province, China
| | - Zhaojun Sun
- Department of Hematology, Cangzhou People's Hospital, Cangzhou City, Hebei Province, China
| | - Xiaolei Li
- Department of Hematology, Cangzhou People's Hospital, Cangzhou City, Hebei Province, China
| | - Ning Wang
- Department of Hematology, Cangzhou People's Hospital, Cangzhou City, Hebei Province, China
| | - Rui Zhang
- Department of Hematology, Cangzhou People's Hospital, Cangzhou City, Hebei Province, China
| |
Collapse
|
52
|
Mercadal S, Gomez CA, Lee CJ, Couriel DR. Infectious complications following CAR-t cell therapy for B cell non-Hodgkin lymphoma: a single-center experience and review of the literature. Ann Hematol 2023:10.1007/s00277-023-05131-7. [PMID: 37246975 DOI: 10.1007/s00277-023-05131-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/06/2023] [Indexed: 05/30/2023]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy targeting CD19 has significantly improved outcomes in the treatment of refractory or relapsed (R/R) B-cell non-Hodgkin lymphoma (NHL). Several risk factors including CAR-T cell-related toxicities and their treatments often lead to infectious complications (ICs); however, the pattern and timeline is not well established. We evaluated ICs in 48 patients with R/R B-cell NHL following CAR-T cell therapy at our institution. Overall, 15 patients experienced 22 infection events. Eight infections (4 bacterial, 3 viral and 1 fungal) occurred within the first 30 days and 14 infections (7 bacterial, 6 viral, 1 fungal) between days 31 to 180 following CAR-T infusion. Most infections were mild-to-moderate and fifteen infections involved the respiratory tract. Two patients developed mild-to-moderate COVID-19 infection and one patient a cytomegalovirus reactivation after CAR-T infusion. Two patients developed IFIs: one case each of fatal disseminated candidiasis and invasive pulmonary aspergillosis at day 16 and 77, respectively. Patients with more than 4 prior antitumor regimens and patient's ≥ 65 years had a higher infection rate. Infections in patients with relapsed/refractory B-cell NHL are common after CAR-T despite the use of infection prophylaxis. Age ≥ 65 years and having > 4 prior antitumor treatments were identified as risk factors for infection. Fungal infections carried significant impact in morbidity and mortality, suggesting a role for increase fungal surveillance and/or anti-mold prophylaxis following high-dose steroids and tocilizumab. Four of ten patients developed an antibody response following two doses of SARS-CoV-2 mRNA vaccine.
Collapse
Affiliation(s)
- Santiago Mercadal
- Transplant and Cellular Therapy Program, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA.
- Bone Marrow Transplant/Cellular Therapy and Regenerative Medicine, University of Utah, 2000 Cir of Hope Dr Ste 1950, Salt Lake City, Utah, 84112, USA.
| | - Carlos A Gomez
- Department of Internal Medicine, Division of Infectious Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Catherine J Lee
- Transplant and Cellular Therapy Program, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Daniel R Couriel
- Transplant and Cellular Therapy Program, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Bone Marrow Transplant/Cellular Therapy and Regenerative Medicine, University of Utah, 2000 Cir of Hope Dr Ste 1950, Salt Lake City, Utah, 84112, USA
| |
Collapse
|
53
|
Li Z, Que Y, Wang D, Lu J, Li C, Xu M, Wang Z, Yu Q, Long X, An N, Xiao Y, Li C. Recovery-model: A model for CAR T-cell-related thrombocytopenia in relapsed/refractory multiple myeloma. Thromb Res 2023; 227:62-70. [PMID: 37235950 DOI: 10.1016/j.thromres.2023.05.016] [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: 12/18/2022] [Revised: 05/02/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023]
Abstract
BACKGROUND Patients with multiple myeloma (MM) treated with anti-B cell maturation antigen (BCMA) and chimeric antigen receptor (CAR) T-cell therapy tend to show delayed platelet recovery. PATIENTS AND METHODS This single-center retrospective observational study included a cohort of patients with MM treated with anti-BCMA CAR-T cells in ChiCTR-OPC-16009113, ChiCTR1800018137, and ChiCTR1900021153. RESULTS Fifty-eight patients with MM treated with anti-BCMA CAR-T cells were included. Delayed platelet recovery (platelet count not recovering to 50 × 109/L within 28 days) was observed in 36 % of patients. Regression analysis identified several factors that influenced platelet recovery, and accordingly, a Recovery-Model was developed. A high Recovery-Model score indicates a greater risk of delayed platelet recovery after CAR-T cell infusion and reflects the risk of hematologic toxicity. The model's predictive biomarkers included baseline platelet count, baseline hemoglobin level, logarithm of baseline Ferritin level, and cytokine release syndrome grade. Finally, survival analysis showed a significant relationship between overall survival, delayed platelet recovery (p = 0.0457), and a high Recovery-Model score (p = 0.0011). CONCLUSIONS Inflammation-related factors and bone marrow reserves are associated with delayed platelet recovery. Therefore, we developed a model to predict the risk of delayed platelet recovery and hematological toxicity in relapsed/refractory patients with MM after anti-BCMA CAR-T cell treatment.
Collapse
Affiliation(s)
- Zhe Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yimei Que
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Di Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei 430030, China
| | - Jie Lu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Chunhui Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Menglei Xu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zhiqiong Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei 430030, China
| | - Qiuxia Yu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiaolu Long
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ning An
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yi Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei 430030, China
| | - Chunrui Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei 430030, China.
| |
Collapse
|
54
|
Perrone S, Lopedote P, De Sanctis V, Iamundo De Cumis I, Pulsoni A, Strati P. Novel Drugs and Radiotherapy in Relapsed Lymphomas: Abscopal Response and Beyond. Cancers (Basel) 2023; 15:2751. [PMID: 37345088 DOI: 10.3390/cancers15102751] [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: 03/20/2023] [Revised: 05/04/2023] [Accepted: 05/11/2023] [Indexed: 06/23/2023] Open
Abstract
Combined modality has represented a mainstay of treatment across many lymphoma histologies, given their sensitivity to both multi-agent chemotherapy and intermediate-dose radiotherapy. More recently, several new agents, including immunotherapies, have reshaped the therapeutic panorama of some lymphomas. In parallel, radiotherapy techniques have witnessed substantial improvement, accompanied by a growing understanding that radiation itself comes with an immune-mediated effect. Six decades after a metastatic lesion regression outside the irradiated field was first described, there is increasing evidence that a combination of radiotherapy and immunotherapy could boost an abscopal effect. This review focuses on the mechanisms underlying this interaction in the setting of lymphomas, and on the results of pivotal prospective studies. Furthermore, the available evidence on the concomitant use of radiotherapy and small molecules (i.e., lenalidomide, venetoclax, and ibrutinib), as well as brentuximab vedotin, and chimeric antigen receptor (CAR) T-cell therapy, is summarized. Currently, combining radiotherapy with new agents in patients who are affected by lymphomas appears feasible, particularly as a bridge to anti-CD19 autologous CAR T-cell infusion. However, more studies are required to assess these combinations, and preliminary data suggest only a synergistic rather than a curative effect.
Collapse
Affiliation(s)
- Salvatore Perrone
- Department of Hematology, S.M. Goretti Hospital, Polo Universitario Pontino, 04100 Latina, Italy
| | - Paolo Lopedote
- Department of Medicine, St Elizabeth's Medical Center, Boston University, Boston, MA 02135, USA
| | - Vitaliana De Sanctis
- Department of Radiation Oncology, Faculty of Medicina e Psicologia, Sant'Andrea Hospital, University of Rome "La Sapienza", 00185 Rome, Italy
| | - Ilenia Iamundo De Cumis
- Department of Radiation Oncology, A. Businco Hospital, ARNAS G. Brotzu, 09121 Cagliari, Italy
| | - Alessandro Pulsoni
- Department of Hematology, S.M. Goretti Hospital, Polo Universitario Pontino, 04100 Latina, Italy
| | - Paolo Strati
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
55
|
Zhang P, Huang L, Zheng M, Zhang C, Wan D, Wei J, Cao Y. Case Report: Active tuberculosis infection in CAR T-cell recipients post CAR T-cell therapy: a retrospective case series. Front Cell Infect Microbiol 2023; 13:1147454. [PMID: 37249982 PMCID: PMC10210154 DOI: 10.3389/fcimb.2023.1147454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/26/2023] [Indexed: 05/31/2023] Open
Abstract
High response rates in B-cell malignancies have been achieved with chimeric antigen receptor (CAR) T-cell therapy. Emerging reports indicate a risk of active tuberculosis (TB) with novel immunotherapy for tumors. However, studies of TB in patients post CAR T-cell therapy are limited. In this case series study, we describe five patients with active TB post CD19/CD22 target CAR T-cell therapy alone or following autologous stem cell transplantation (ASCT). One of the patients developed active TB within the first 30 days post CAR T-cell therapy, and fever was the dominant presenting symptom; extrapulmonary manifestations of active TB were common in the other four patients and manifested after the first 30 days of CAR T-cell therapy. Four of the five patients improved with anti-TB treatment, but one patient with isoniazid resistance died of central nervous system TB infection. Our study provides the first series report of active TB following CD19/CD22 target CAR T-cell therapy.
Collapse
Affiliation(s)
- Peiling Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, China
| | - Liang Huang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, China
| | - Miao Zheng
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, China
| | - Chao Zhang
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dongyi Wan
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jia Wei
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, China
| | - Yang Cao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, China
| |
Collapse
|
56
|
Sun T, Li D, Huang L, Zhu X. Inflammatory abrasion of hematopoietic stem cells: a candidate clue for the post-CAR-T hematotoxicity? Front Immunol 2023; 14:1141779. [PMID: 37223096 PMCID: PMC10200893 DOI: 10.3389/fimmu.2023.1141779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/21/2023] [Indexed: 05/25/2023] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has shown remarkable effects in treating various hematological malignancies. However, hematotoxicity, specifically neutropenia, thrombocytopenia, and anemia, poses a serious threat to patient prognosis and remains a less focused adverse effect of CAR-T therapy. The mechanism underlying lasting or recurring late-phase hematotoxicity, long after the influence of lymphodepletion therapy and cytokine release syndrome (CRS), remains elusive. In this review, we summarize the current clinical studies on CAR-T late hematotoxicity to clarify its definition, incidence, characteristics, risk factors, and interventions. Owing to the effectiveness of transfusing hematopoietic stem cells (HSCs) in rescuing severe CAR-T late hematotoxicity and the unignorable role of inflammation in CAR-T therapy, this review also discusses possible mechanisms of the harmful influence of inflammation on HSCs, including inflammatory abrasion of the number and the function of HSCs. We also discuss chronic and acute inflammation. Cytokines, cellular immunity, and niche factors likely to be disturbed in CAR-T therapy are highlighted factors with possible contributions to post-CAR-T hematotoxicity.
Collapse
|
57
|
Ying Z, Yang H, Guo Y, Li W, Zou D, Zhou D, Wang Z, Zhang M, Wu J, Liu H, Wang C, Ma L, Yang S, Zhou Z, Qin Y, Song Y, Zhu J. Long-term outcomes of relmacabtagene autoleucel in Chinese patients with relapsed/refractory large B-cell lymphoma: Updated results of the RELIANCE study. Cytotherapy 2023; 25:521-529. [PMID: 36842849 DOI: 10.1016/j.jcyt.2022.10.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/09/2022] [Accepted: 10/27/2022] [Indexed: 02/28/2023]
Abstract
BACKGROUND AIMS The RELIANCE study has demonstrated the activity and safety of relmacabtagene autoleucel (relma-cel) (JW Therapeutics [Shanghai] Co, Ltd, Shanghai, China), a CD19-targeted chimeric antigen receptor T-cell product, in patients with heavily pre-treated relapsed/refractory large B-cell lymphoma (r/r LBCL). This study aimed to report the updated 2-year data of the RELIANCE study. METHODS The RELIANCE study (NCT04089215) was an open-label, multi-center, randomized, phase 1/2 registrational clinical trial conducted at 10 clinical sites in China. Adult patients with heavily pre-treated r/r LBCL were enrolled and received lymphodepletion chemotherapy followed by infusion of 100 × 106 or 150 × 106 relma-cel. The primary endpoint was objective response rate (ORR) at 3 months, as assessed by investigators. Secondary endpoints were duration of response (DoR), progression-free survival (PFS), overall survival (OS) and safety profiles. RESULTS From November 2017 to January 2022, a total of 68 patients were enrolled, and 59 patients received relma-cel infusion. As of March 29, 2022, a total of 59 patients had a median follow-up of 17.9 months (range, 0.3-25.6). ORR was 77.59% (95% confidence interval [CI], 64.73-87.49) and complete response rate was 53.45% (95% CI, 39.87-66.66). Median DoR was 20.3 months (95% CI, 4.86-not reached [NR]) and median PFS was 7.0 months (95% CI, 4.76-24.15). Median OS was NR and 1-year and 2-year OS rates were 75.0% and 69.3%, respectively. Three (5.1%) patients experienced grade ≥3 cytokine release syndrome and two (3.4%) patients had grade ≥3 neurotoxicity. CONCLUSIONS The updated data of the RELIANCE study demonstrate durable response with and manageable safety profile of relma-cel in patients with heavily pre-treated r/r LBCL.
Collapse
Affiliation(s)
- Zhitao Ying
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Haiyan Yang
- Department of Lymphoma, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, China
| | - Ye Guo
- Department of Oncology, Shanghai Dongfang Hospital, Tongji University, Shanghai, China
| | - Wenyu Li
- Department of Lymphoma, Guangdong Provincial People's Hospital, Guangdong Academy of Sciences, Guangzhou, China
| | - Dehui Zou
- Department of Lymphoma and Myeloma, State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union of Medical College, Tianjin, China
| | - Daobin Zhou
- Department of Hematopathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhao Wang
- Department of Hematopathology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Mingzhi Zhang
- Department of Oncology, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Jianqiu Wu
- Department of Medical Oncology, Jiangsu Institute of Cancer Research, Jiangsu Red Cross Cancer Center, Jiangsu Cancer Hospital, Nanjing Medical University, Nanjing, China
| | - Hui Liu
- Department of Hematology, Beijing Hospital, Beijing, China
| | - Chris Wang
- JW Therapeutics (Shanghai) Co, Ltd, Shanghai, China
| | - Laura Ma
- JW Therapeutics (Shanghai) Co, Ltd, Shanghai, China
| | - Su Yang
- JW Therapeutics (Shanghai) Co, Ltd, Shanghai, China
| | - Zisong Zhou
- JW Therapeutics (Shanghai) Co, Ltd, Shanghai, China
| | - Yun Qin
- JW Therapeutics (Shanghai) Co, Ltd, Shanghai, China
| | - Yuqin Song
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China.
| | - Jun Zhu
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
58
|
Lee D, Jordan AI, Menges MA, Lazaryan A, Nishihori T, Gaballa SR, Shah BD, Pinilla-Ibarz J, Baluch A, Klinkova OV, Chavez JC, Jain MD, Locke FL. Pneumococcal Conjugate Vaccine Does Not Induce Humoral Response When Administrated Within the Six Months After CD19 CAR T-Cell Therapy. Transplant Cell Ther 2023; 29:277.e1-277.e9. [PMID: 35970303 DOI: 10.1016/j.jtct.2022.08.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/25/2022] [Accepted: 08/06/2022] [Indexed: 10/15/2022]
Abstract
CD19 targeted chimeric antigen receptor-modified T cell therapy (CAR-T) leads to B cell aplasia and low serum immunoglobulin levels. Long-lived CD19-negative plasma cells may persist through the therapy and generate antibodies. There is a paucity of data describing how CAR-T impacts the persistence of antibodies against vaccine-related antigens and the degree to which CAR-T recipients may respond to vaccines. We characterized the effect of CAR-T on pneumococcal immunoglobulin G (IgG) titers and determine whether pneumococcal conjugate vaccine (PCV13) administered after CAR-T develops long-term humoral protection against pneumococcus. A retrospective chart review was performed to identify CAR-T recipients who had serum pneumococcal IgG titers drawn before (baseline) or at days +90, +180, +270, +360, or +540 after CAR-T. We then determined whether they received PCV13 vaccination at these timepoints. IgG concentration ≥1.3 μg/mL was considered protective for that serotype, and patients with ≥6/11 tested vaccine-specific serotypes meeting this threshold were deemed to have humoral protection against pneumococcus. Absolute pneumococcal IgG titers and the proportion of patients with humoral protection, stratified by serotype, and vaccination status were compared by paired nonparametric t-tests. Absolute counts for lymphocyte, CD4 T-cell, and CD19 cell and total IgG level, along with the rate of invasive pneumococcal infections, were measured at these timepoints. A total of 148 CAR-T recipients with pneumococcal IgG titers measured for at least one of the defined time points were identified. At baseline, 25% (19/76) patients with evaluable pneumococcal IgG titers met the definition of humoral protection. Among 44 patients with paired pneumococcal IgG titers at baseline and day+90, absolute IgG titers of all serotypes decreased (geometric mean = 0.41 and 0.32 µg/mL, respectively; P < .001). Thirteen patients were vaccinated following the titer blood draw at day+90 and had paired pneumococcal IgG titers at day+90 and day180. Absolute IgG titers of all vaccine specific serotypes in these vaccinated patients decreased from day+90 to day+180 (geometric mean = 0.36 and 0.29 µg/mL, respectively; P = .03). The proportion of patients meeting the criteria of humoral protection remained the same at day+180 despite vaccination at day+90. The results were similar among 8 patients vaccinated at day+180, as well as 7 patients consecutively vaccinated at day+90 and day+180 with corresponding pneumococcal IgG titers. When all vaccine-specific pneumococcal IgG titers were pooled together by timepoint regardless of vaccination status, the proportion of patients with humoral protection decreased until day+540. Some patients developed humoral protection after vaccination at day+360, maintained seroprotective IgG titers from baseline, or developed protection after receiving intravenous immunoglobulin treatment secondary to recurrent infections. Our study demonstrated that few large B cell lymphoma patients had humoral protection against pneumococcus at baseline, and existing IgG titers decreased after CAR-T. PCV13 vaccination at day+90 or day+180 after CAR-T did not increase humoral protection against pneumococcus. Only at day+540 was there evidence of humoral protection against pneumococcus in a modest proportion of patients. Clinical trials are needed to determine the optimal timing of vaccination, before or after CAR-T, to develop protective immunity against Streptococcus pneumoniae infections.
Collapse
Affiliation(s)
- Dasom Lee
- Department of Internal Medicine, University of South Florida, Tampa, Florida
| | - Aryanna I Jordan
- Department of Internal Medicine, University of South Florida, Tampa, Florida
| | - Meghan A Menges
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | - Alexandr Lazaryan
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | - Taiga Nishihori
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | - Sameh R Gaballa
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, Florida
| | - Bijal D Shah
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, Florida
| | | | - Aliyah Baluch
- Infectious Disease Division, Moffitt Cancer Center, Tampa, Florida
| | - Olga V Klinkova
- Infectious Disease Division, Moffitt Cancer Center, Tampa, Florida
| | - Julio C Chavez
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, Florida
| | - Michael D Jain
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida.
| |
Collapse
|
59
|
Wilson Dib R, Ariza-Heredia E, Spallone A, Chemaly RF. Respiratory Viral Infections in Recipients of Cellular Therapies: A Review of Incidence, Outcomes, Treatment, and Prevention. Open Forum Infect Dis 2023; 10:ofad166. [PMID: 37065990 PMCID: PMC10096899 DOI: 10.1093/ofid/ofad166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/22/2023] [Indexed: 04/18/2023] Open
Abstract
Respiratory viral infections (RVIs) are of major clinical importance in immunocompromised patients and represent a substantial cause of morbidity and mortality in patients with hematologic malignancies and those who have undergone hematopoietic cell transplantation. Similarly, patients receiving immunotherapy with CD19-targeted chimeric antigen receptor-modified T cells, natural killer cells, and genetically modified T-cell receptors are susceptible to RVIs and progression to lower respiratory tract infections. In adoptive cellular therapy recipients, this enhanced susceptibility to RVIs results from previous chemotherapy regimens such as lymphocyte-depleting chemotherapy conditioning regimens, underlying B-cell malignancies, immune-related toxicities, and secondary prolonged, profound hypogammaglobulinemia. The aggregated risk factors for RVIs have both immediate and long-term consequences. This review summarizes the current literature on the pathogenesis, epidemiology, and clinical aspects of RVIs that are unique to recipients of adoptive cellular therapy, the preventive and therapeutic options for common RVIs, and appropriate infection control and preventive strategies.
Collapse
Affiliation(s)
- Rita Wilson Dib
- Department of Internal Medicine, Division of Infectious Diseases, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ella Ariza-Heredia
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Amy Spallone
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Roy F Chemaly
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
60
|
Penack O, Peczynski C, Koenecke C, Polge E, Kuhnl A, Fegueux N, Daskalakis M, Kröger N, Dreger P, Besley C, Schanz U, Bloor A, Ganser A, Forcade E, Corral LL, Passweg JR, Novak U, Moiseev I, Schoemans H, Basak GW, Chabannon C, Sureda A, Averbuch D, Glass B, de la Camara R, Peric Z. Severe cytopenia after CD19 CAR T-cell therapy: a retrospective study from the EBMT Transplant Complications Working Party. J Immunother Cancer 2023; 11:e006406. [PMID: 37072350 PMCID: PMC10124318 DOI: 10.1136/jitc-2022-006406] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2023] [Indexed: 04/20/2023] Open
Abstract
We investigated the incidence and outcome of anti-CD19 chimeric antigen receptor (CAR) T-cells-associated Common Terminology Criteria for Adverse Events (CTCAE) ≥grade 3 cytopenia. In the EBMT CAR-T registry, we identified 398 adult patients with large B-cell lymphoma who had been treated with CAR-T-cells with axicel (62%) or tisacel (38%) before August 2021 and had cytopenia status documented for the first 100 days. Most patients had received two or three previous lines of therapy, however, 22.3% had received four or more. Disease status was progressive in 80.4%, stable in 5.0% and partial/complete remission in 14.6%. 25.9% of the patients had received a transplantation before. Median age was 61.4 years (min-max; IQR=18.7-81; (52.9-69.5)).The cumulative incidence of ≥grade 3 cytopenia was 9.0% at 30 days (95% CI (6.5 to 12.1)) and 12.1% at 100 days after CAR T-cell infusion (95% CI (9.1 to 15.5)). The median time from CAR-T infusion to cytopenia onset was 16.5 days (min-max; IQR=1-90; (4-29.8)). Grade 3 and grade 4 CTCAE cytopenia occurred in 15.2% and 84.8%, respectively. In 47.6% there was no resolution.Severe cytopenia had no significant impact on overall survival (OS) (HR 1.13 (95% CI 0.74 to 1.73), p=0.57). However, patients with severe cytopenia had a poorer progression-free survival (PFS) (HR 1.54 (95% CI 1.07 to 2.22), p=0.02) and a higher relapse incidence (HR 1.52 (95% CI 1.04 to 2.23), p=0.03). In those patients who developed severe cytopenia during the first 100 days (n=47), OS, PFS, relapse incidence and non-relapse mortality at 12 months after diagnosis of severe cytopenia were 53.6% (95% CI (40.3 to 71.2)), 20% (95% CI (10.4 to 38.6)), 73.5% (95% CI (55.2 to 85.2)) and 6.5% (95% CI (1.7 to 16.2)), respectively.In multivariate analysis of severe cytopenia risk factors, only year of CAR-T infusion (HR=0.61, 95% CI (0.39 to 0.95), p=0.028) and total number of treatment lines before CAR-T infusion (one or two lines vs three or more, HR=0.41, 95% CI (0.21 to 0.83), p=0.013) had a significant positive association with the incidence of cytopenia. Other factors, such as previous transplantation, disease status at time of CAR-T, patient age and patient sex, had no significant association.Our data provide insight on frequency and clinical relevance of severe cytopenia after CAR T-cell therapy 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; 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; INSERM UMR-S 938, Sorbonne University, Paris, France
| | - Andrea Kuhnl
- Departement of Haematological Medicine, Kings College Hospital, London, UK
| | - Nathalie Fegueux
- Département d'Hématologie Clinique, CHU Lapeyronie, Montpellier, Languedoc-Roussillon, France
| | - Michael Daskalakis
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nicolaus Kröger
- University Hospital Eppendorf, Bone Marrow Transplantation Centre, Hamburg, Germany
| | - Peter Dreger
- University of Heidelberg, Medizinische Klinik u. Poliklinik V, Heidelberg, Germany
- Department of Hematology, Oncology and Internal Medicine, the Medical University of Warsaw, Marseille, Poland
| | - Caroline Besley
- Departement of Paediatric Oncology/BMT, Bristol Royal Hospital for Children, Bristol, UK
| | - Urs Schanz
- University Hospital, Clinic of Hematology, Zurich, Switzerland
| | - Adrian Bloor
- Christie NHS Trust Hospital, Adult Leukaemia and Bone Marrow Transplant Unit, Manchester, UK
| | - Arnold Ganser
- Department of Haematology, Hemostasis, Oncology, Hannover Medical School, Hannover, Germany
| | | | | | | | - Urban Novak
- Department of Medical Oncology, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ivan Moiseev
- EBMT Transplant Complications Working Party, Paris, France
- 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, Marseille, Poland
| | - Christian Chabannon
- EBMT Cellular Therapy and Immunobiology Working Party, Leiden, The 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, Institut de Ciències Biomèdiques de Bellvitge (IDIBELL), Universitat de Barcelona, Barcelona 08908, Spain
| | - Dina Averbuch
- Faculty of Medicine, Department of Pediatric Infectious Diseases, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
- EBMT Infectious Diseases Working Party
| | - Bertram Glass
- Department of Hematology, Oncology, and Tumor Immunology, Helios Klinikum Berlin-Buch, Berlin, Germany
- EBMT Lymphoma Working Party
| | - Rafael de la Camara
- Faculty of Medicine, Department of Pediatric Infectious Diseases, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Haematology, Hospital Universitario de la Princesa, Madrid, Spain
| | - Zinaida Peric
- EBMT Transplant Complications Working Party, Paris, France
- University Hospital Centre Zagreb and School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
61
|
Wang J, Zhang M, Lyu H, Guo R, Xiao X, Bai X, Pu Y, Meng J, Li Q, Yuan T, Lu W, Zhao M. Low-dose administration of prednisone has a good effect on the treatment of prolonged hematologic toxicity post-CD19 CAR-T cell therapy. Front Immunol 2023; 14:1139559. [PMID: 36999027 PMCID: PMC10043253 DOI: 10.3389/fimmu.2023.1139559] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 02/27/2023] [Indexed: 03/15/2023] Open
Abstract
IntroductionHematologic toxicity (HT) is a joint adverse event after CAR-T cells infusion. Some patients experience prolonged hematologic toxicity (PHT), which is challenging to treat.MethodsWe collected clinical data from patients with relapsed refractory B-ALL treated with CD19 CAR-T cells. Patients with PHT who did not respond to erythropoietin, platelet receptor agonists, transfusion, or G-CSF and eventually received low-dose prednisone therapy were included in the analysis. We retrospectively analyzed the efficacy and safety of low-dose prednisone on PHT.ResultsAmong 109 patients treated with CD19 CAR-T cells, 78.9% (86/109) of patients were evaluated as PHT. Of these, 15 patients had persistent hematological toxicity after infusion (12 were grade 3/4 cytopenia, 12 were trilineage cytopenia and 3 were bilineage cytopenia), 2 developed cytopenia without apparent cause after D28. The initial prednisone dose was 0.5 mg/kg/day, and the median response time was 21 days (7-40 days). The recovery rate of blood count was 100%, and the complete recovery rate ranged from 60% to 66.67%. Especially exciting was that HT recurred in 6 patients after stopping prednisone. They were relieved again after the administration of prednisone. The median follow-up time was 14.97 months (4.1-31.2 months). Twelve-month duration of PFS and OS rates were 58.8% (±11.9%) and 64.7% (±11.6%). We did not observe any other side effects of prednisone apart from drug-controllable hyperglycemia and hypertension.DiscussionWe suggest that low-dose prednisone is a beneficial and tolerable therapy for PHT after CAR-T cells. The trials have been registered at www.chictr.org.cn as ChiCTR-ONN-16009862 (November 14, 2016) and ChiCTR1800015164 (March 11, 2018).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Wenyi Lu
- *Correspondence: Mingfeng Zhao, ; Wenyi Lu,
| | | |
Collapse
|
62
|
Nagler A, Perriello VM, Falini L, Falini B. How I treat refractory/relapsed diffuse large B-cell lymphomas with CD19-directed chimeric antigen receptor T cells. Br J Haematol 2023; 201:396-410. [PMID: 36916189 DOI: 10.1111/bjh.18724] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/10/2023] [Accepted: 02/17/2023] [Indexed: 03/15/2023]
Abstract
Chimeric antigen receptor (CAR) T cells targeting CD19 represent a promising salvage immunotherapy for relapsed/refractory diffuse large B-cell lymphoma (R/R DLBCL), offering ~40% of long-term responses. In everyday clinical practice, haematologists involved in CAR T cell treatment of patients with R/R DLBCL have to deal with diagnostically complex cases and difficult therapeutic choices. The availability of novel immunotherapeutic agents for R/R DLBCL and recent advances in understanding CAR T-cell failure mechanisms demand a rational approach to identify the best choice for bridging therapy and managing post-CAR T-cell therapy relapses. Moreover, positron emission tomography/computerised tomography may result in false-positive interpretation, highlighting the importance of post-treatment biopsy. In this review, we discuss all above issues, presenting four instructive cases, with the aim to provide criteria and new perspectives for CAR T-cell treatment of DLBCL.
Collapse
Affiliation(s)
- Arnon Nagler
- Division of Hematology, Sheba Medical Center, Tel Hashomer, Israel
| | - Vincenzo Maria Perriello
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Lorenza Falini
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Brunangelo Falini
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, Perugia, Italy
| |
Collapse
|
63
|
Goldman A, Raschi E, Chapman J, Santomasso BD, Pasquini MC, Perales MA, Shouval R. Progressive multifocal leukoencephalopathy in patients treated with chimeric antigen receptor T cells. Blood 2023; 141:673-677. [PMID: 36332168 PMCID: PMC9979708 DOI: 10.1182/blood.2022017386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
Using 2 global postmarketing surveillance databases, Goldman and colleagues report that progressive multifocal leukoencephalopathy (PML), a viral disease associated with profound immunosuppression, occurs in approximately 0.9 cases per 1000 recipients of CD19-directed CAR T-cell therapy. The risk of PML appears higher with CAR T-cell therapy than other cancer therapies, but its precise role cannot be distinguished from antecedent therapies that these patients receive.
Collapse
Affiliation(s)
- Adam Goldman
- Department of Internal Medicine, The Talpiot Sheba Medical Leadership Program, Chaim Sheba Medical Center, Ramat-Gan, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Emanuel Raschi
- Pharmacology Unit, Department of Medical and Surgical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Joab Chapman
- Department of Neurology, The Chaim Sheba Medical Center, Ramat-Gan, Israel
| | - Bianca D. Santomasso
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY
- Adult Bone Marrow Transplantation Service and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marcelo C. Pasquini
- Department of Medicine, Center for International Blood and Bone Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Roni Shouval
- Adult Bone Marrow Transplantation Service and Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| |
Collapse
|
64
|
Cytokine profiles are associated with prolonged hematologic toxicities after B-cell maturation antigen targeted chimeric antigen receptor-T-cell therapy. Cytotherapy 2023; 25:192-201. [PMID: 36496302 DOI: 10.1016/j.jcyt.2022.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 10/20/2022] [Accepted: 11/03/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND AIMS The considerable efficacy of B-cell maturation antigen-targeted chimeric antigen receptor (CAR)-T-cell therapy has been extensively demonstrated in the treatment of relapsed or refractory multiple myeloma. Nevertheless, in clinical practice, prolonged hematologic toxicity (PHT) extends hospital stay and impairs long-term survival. METHODS This retrospective study reviewed 99 patients with relapsed or refractory multiple myeloma who underwent B-cell maturation antigen CAR-T-cell therapy at our institution between April 2018 and September 2021 (ChiCTR1800017404). RESULTS Among 93 evaluable patients, the incidence of prolonged hematologic toxicities was high after CAR-T-cell infusion, including 38.71% (36/93) of patients with prolonged neutropenia, 22.58% (21/93) with prolonged anemia and 59.14% (55/93) with prolonged thrombocytopenia. In addition, 9.68% (9/93) of patients experienced prolonged pancytopenia. Our multivariate analyses identified that cytokine profiles were independent risk factors for PHTs, whereas a sufficient baseline hematopoietic function and high CD4/CD8 ratio of CAR-T cells were protective factors for PHTs after CAR-T-cell infusion. Subgroup analyses found that the kinetics of post-CAR-T hematologic parameters were primarily determined by the collective effects of cytokine release syndrome and baseline hematopoietic functions, and showed influential weights for the three lineages. CONCLUSIONS Our findings improve the understanding of the impact of cytokines on hematopoietic functions, which could contribute to the mechanism investigation and exploration of potential intervention strategies.
Collapse
|
65
|
McNerney KO, Richards RM, Aguayo-Hiraldo P, Calkoen FG, Talano JA, Moskop A, Balduzzi A, Krajewski J, Dave H, Vatsayan A, Callahan C, Liu H, Li Y, Davis KL, Maude SL. SARS-CoV-2 infections in pediatric and young adult recipients of chimeric antigen receptor T-cell therapy: an international registry report. J Immunother Cancer 2023; 11:jitc-2022-005957. [PMID: 36707090 PMCID: PMC9884906 DOI: 10.1136/jitc-2022-005957] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2023] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Immunocompromised patients are at increased risk of SARS-CoV-2 infections. Patients undergoing chimeric antigen receptor (CAR) T-cell therapy for relapsed/refractory B-cell malignancies are uniquely immunosuppressed due to CAR T-mediated B-cell aplasia (BCA). While SARS-CoV-2 mortality rates of 33%-40% are reported in adult CAR T-cell recipients, outcomes in pediatric and young adult CAR T-cell recipients are limited. METHODS We created an international retrospective registry of CAR T recipients aged 0-30 years infected with SARS-CoV-2 within 2 months prior to or any time after CAR T infusion. SARS-CoV-2-associated illness was graded as asymptomatic, mild, moderate, or severe COVID-19, or multisystem inflammatory syndrome in children (MIS-C). To assess for risk factors associated with significant SARS-CoV-2 infections (infections requiring hospital admission for respiratory distress or supplemental oxygen), univariate and multivariable regression analyses were performed. RESULTS Nine centers contributed 78 infections in 75 patients. Of 70 SARS-CoV-2 infections occurring after CAR T infusion, 13 (18.6%) were classified as asymptomatic, 37 (52.9%) mild, 11 (15.7%) moderate, and 6 (8.6%) severe COVID-19. Three (4.3%) were classified as MIS-C. BCA was not significantly associated with infection severity. Prior to the emergence of the Omicron variant, of 47 infections, 19 (40.4%) resulted in hospital admission and 7 (14.9%) required intensive care, while after the emergence of the Omicron variant, of 23 infections, only 1 (4.3%) required admission and the remaining 22 (95.7%) had asymptomatic or mild COVID-19. Death occurred in 3 of 70 (4.3%); each death involved coinfection or life-threatening condition. In a multivariable model, factors associated with significant SARS-CoV-2 infection included having two or more comorbidities (OR 7.73, CI 1.05 to 74.8, p=0.048) and age ≥18 years (OR 9.51, CI 1.90 to 82.2, p=0.014). In the eight patients infected with SARS-CoV-2 before CAR T, half of these patients had their CAR T infusion delayed by 15-30 days. CONCLUSIONS In a large international cohort of pediatric and young adult CAR-T recipients, SARS-CoV-2 infections resulted in frequent hospital and intensive care unit admissions and were associated with mortality in 4.3%. Patients with two or more comorbidities or aged ≥18 years were more likely to experience significant illness. Suspected Omicron infections were associated with milder disease.
Collapse
Affiliation(s)
- Kevin Owen McNerney
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St Petersburg, Florida, USA,Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rebecca M Richards
- Hematology, Oncology, and Bone Marrow Transplant, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Paibel Aguayo-Hiraldo
- Cancer and Blood Disease Institute, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Friso G Calkoen
- Division of Pediatric Oncology, Princess Maxima Center, Utrecht, The Netherlands
| | - Julie-An Talano
- Division of Hematology/Oncology/Blood and Marrow TransplantationDepartment of Pediatrics, Medical College of Wisconsin and Children’s Wisconsin, Milwaukee, Wisconsin, USA
| | - Amy Moskop
- Division of Hematology/Oncology/Blood and Marrow TransplantationDepartment of Pediatrics, Medical College of Wisconsin and Children’s Wisconsin, Milwaukee, Wisconsin, USA
| | - Adriana Balduzzi
- Clinica Pediatrica Università degli Studi di Milano Bicocca, Fondazione IRCCS San Gerardo dai Tintori, Milan, Italy
| | - Jennifer Krajewski
- Pediatric Blood and Marrow Transplantation, Hackensack Meridian School of Medicine, Hackensack, New Jersey, USA
| | - Hema Dave
- Cancer Immunology and Microbial Oncology Research Program, Children's National Hospital, Washington, District of Columbia, USA
| | - Anant Vatsayan
- Center for Cancer and Blood Disorders, Children's National Hospital, Washington, District of Columbia, USA
| | - Colleen Callahan
- Division of Oncology and Cancer Immunotherapy Program, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Hongyan Liu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Yimei Li
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kara Lynn Davis
- Pediatrics, Stanford University School of Medicine, Stanford, California, USA,Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford, California, USA
| | - Shannon L Maude
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA,Division of Oncology, Center for Childhood Cancer Research and Cancer Immunotherapy Program, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
66
|
Zhao A, Zhao M, Qian W, Liang A, Li P, Liu H. Secondary myeloid neoplasms after CD19 CAR T therapy in patients with refractory/relapsed B-cell lymphoma: Case series and review of literature. Front Immunol 2023; 13:1063986. [PMID: 36713414 PMCID: PMC9880439 DOI: 10.3389/fimmu.2022.1063986] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/30/2022] [Indexed: 01/14/2023] Open
Abstract
Background Several chimeric antigen receptor T cells (CAR T) targeting CD19 have induced profound and prolonged remission for refractory/relapsed (R/R) B-cell lymphoma. The risk of secondary malignancies, especially myeloid neoplasms, is of particular concern in the CAR T community, which still remains unclear. Methods Four patients with R/R B-cell lymphoma after CD19 CAR T therapy diagnosed with secondary myeloid neoplasms (SMN) from 2 hospitals in eastern China were presented, including 3 with myelodysplastic syndrome (MDS) and 1 with acute myeloid leukemia (AML). Using single-cell RNA sequencing (scRNA-seq), we compared the cellular components of bone marrow (BM) samples obtained from one of these MDS patients and a health donor. We also provided a review of recently published literature concerning SMN risk of CAR T therapy. Results Relevant demographic, clinical, laboratory, therapeutic and outcome data were collected and presented by chart review. In our case series, the male-female ratio was 3.0 and the median age at MDS onset was 61.25 years old (range, 50-78). Median number of previous systemic therapies was 4.5 (range, 4-5), including autologous hematopoietic stem cell transplantation (auto-HSCT) in one patient. BM assessments prior to CAR T therapy confirmed normal hematopoiesis without myeloid neoplasms. Moreover, for 3 patients with SMN in our series, cytogenetic analysis predicted a relatively adverse outcome. In our experience and in the literature, treatment choices for the patients with SMN included allogeneic hematopoietic stem cell transplantation (allo-HSCT), hypomethylating agent (HMA), period filgrastim, transfusions and other supportive care. Finally, treatment responses of lymphoma, together with SMN, directly correlated with the overall survival of this community. Of note, it appeared that pathogenesis of MDS wasn't associated with the CAR T toxicities, since all 4 patients experienced a pretty mild CRS of grade 1-2. Additionally, scRNA-seq analysis described the transcriptional alteration of CD34+ cells, identified 13 T/NK clusters, and also indicated increased cytotoxic T cells in MDS BM. Conclusion Our study illustrated the onset and progression of SMN after CD19 CAR T therapy in patients with R/R B-cell lymphoma, which provides useful information of this uncommon later event.
Collapse
Affiliation(s)
- Aiqi Zhao
- Department of Hematology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Mingzhe Zhao
- Department of Hematology, Jinhua Municipal Central Hospital, Jinhua, Zhejiang, China
| | - Wenbin Qian
- Department of Hematology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Aibin Liang
- Department of Hematology, Tongji Hospital of Tongji University, Shanghai, China
| | - Ping Li
- Department of Hematology, Tongji Hospital of Tongji University, Shanghai, China,*Correspondence: Ping Li, ; Hui Liu,
| | - Hui Liu
- Department of Hematology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China,*Correspondence: Ping Li, ; Hui Liu,
| |
Collapse
|
67
|
Eder LN, Martinovic D, Mazzeo P, Ganster C, Hasenkamp J, Thomson J, Trummer A, Haase D, Wulf G. Fatal Progression of Mutated TP53-Associated Clonal Hematopoiesis following Anti-CD19 CAR-T Cell Therapy. Curr Oncol 2023; 30:1146-1150. [PMID: 36661736 PMCID: PMC9858310 DOI: 10.3390/curroncol30010087] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/06/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
We present the case of a 64-year-old man diagnosed with large B-cell lymphoma who relapsed twice after standard-of-care therapy. Due to persisting cytopenia, Next generation sequencing analysis was performed, revealing a small TP53-mutated clone. As a third-line therapy, the patient was treated with CAR-T cells, which resulted in complete remission. However, this treatment also led to the expansion of the TP53-mutated clone and therapy-related myelodysplasia with a complex aberrant karyotype. This case may serve as a paradigmatic example of clonal hematopoietic progression in a patient undergoing CAR-T cell therapy, especially in the context of a TP53-mutated clone.
Collapse
Affiliation(s)
- Lea Naomi Eder
- Department of Hematology and Medical Oncology, University Medicine Goettingen, 37075 Göttingen, Germany
| | - Danilo Martinovic
- Department of Hematology and Medical Oncology, University Medicine Goettingen, 37075 Göttingen, Germany
| | - Paolo Mazzeo
- Department of Hematology and Medical Oncology, University Medicine Goettingen, 37075 Göttingen, Germany
| | - Christina Ganster
- Department of Hematology and Medical Oncology, University Medicine Goettingen, 37075 Göttingen, Germany
| | - Justin Hasenkamp
- Department of Hematology and Medical Oncology, University Medicine Goettingen, 37075 Göttingen, Germany
| | - Julia Thomson
- Department of Hematology and Medical Oncology, University Medicine Goettingen, 37075 Göttingen, Germany
| | - Arne Trummer
- Department of Hematology and Oncology, Städtisches Klinikum Braunschweig, 38114 Braunschweig, Germany
| | - Detlef Haase
- Department of Hematology and Medical Oncology, University Medicine Goettingen, 37075 Göttingen, Germany
| | - Gerald Wulf
- Department of Hematology and Medical Oncology, University Medicine Goettingen, 37075 Göttingen, Germany
| |
Collapse
|
68
|
Nath K, Wudhikarn K, Alarcon Tomas A, Perales MA. Safety evaluation of axicabtagene ciloleucel for relapsed or refractory large B-cell lymphoma. Expert Opin Drug Saf 2023; 22:5-15. [PMID: 36737060 PMCID: PMC9975047 DOI: 10.1080/14740338.2023.2177268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 02/02/2023] [Indexed: 02/05/2023]
Abstract
INTRODUCTION CD19-directed chimeric antigen receptor (CAR) T-cell therapy is a highly effective therapy for patients with relapsed/refractory large B-cell lymphoma (LBCL) and three CD19 CAR T-cell products (axicabtagene ciloleucel, tisagenlecleucel and lisocabtagene maraleucel) are currently approved for this indication. Despite the clinical benefit of CD19 directed CAR T-cell therapy, this treatment is associated with significant morbidity from treatment-emergent toxicities. AREAS COVERED This Review discusses the safety considerations of axicabtagene ciloleucel in patients with LBCL. This includes discussion of the frequently observed immune-mediated toxicities of cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. Additionally, we review CAR T-cell therapy related cytopenias, infection, organ dysfunction and the more recently described hemophagocytic lymphohistiocytosis. EXPERT OPINION A thorough understanding of the toxicities associated with CD19-directed CAR T-cell therapy will facilitate the optimal selection of patients for this therapy. Furthermore, knowledge of preventative measures of CAR T-cell related complications, and early recognition and appropriate intervention will lead to the safe administration of these therapies, and ultimately improved outcomes for our patients.
Collapse
Affiliation(s)
- Karthik Nath
- Cellular Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kitsada Wudhikarn
- Division of Hematology and Center of Excellence in Translational Hematology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Ana Alarcon Tomas
- Division of Hematology and Hemotherapy, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
69
|
Sim B, Ng JY, Teh BW, Talaulikar D. Immunoglobulin replacement in hematological malignancies: a focus on evidence, alternatives, dosing strategy, and cessation rule. Leuk Lymphoma 2023; 64:18-29. [PMID: 36218218 DOI: 10.1080/10428194.2022.2131424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Acquired hypogammaglobulinemia or secondary immunodeficiency (SID) occurs commonly in hematological malignancies with increasing incidence and complexity in the era of modern therapies. Despite current practice of immunoglobulin replacement (IgRT) in SID, the evidence is lacking, especially for newer treatments. We discuss the current evidence for IgRT in various disease groups including issues, such as actual or ideal body weight (IBW)-based dosing, length of treatment, antibiotic prophylaxis, and vaccination. Incidence of SID with newer treatment is lacking. While there is a trend toward decreased respiratory infections and hospitalizations with IgRT, this is not consistent across all disease course or treatment groups. Dosing and indications for cessation of IgRT are also inadequately characterized. Further randomized controlled trials (RCTs) and observational studies are required to assess the optimal indications, timing, and duration of IgRT to improve the efficacy, safety, and cost-effectiveness. Assessment of alternative and adjunctive therapies, such as vaccination and antibiotic prophylaxis could also improve the outcomes and costs.
Collapse
Affiliation(s)
- Beatrice Sim
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Jun Yen Ng
- Department of Hematology, ACT Pathology, Canberra Hospital, Canberra, Australia
| | - Benjamin W Teh
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Dipti Talaulikar
- Department of Hematology, ACT Pathology, Canberra Hospital, Canberra, Australia.,College of Health and Medicine, Australian National University, Canberra, Australia
| |
Collapse
|
70
|
Wang Z, Chen C, Wang L, Jia Y, Qin Y. Chimeric antigen receptor T-cell therapy for multiple myeloma. Front Immunol 2022; 13:1050522. [PMID: 36618390 PMCID: PMC9814974 DOI: 10.3389/fimmu.2022.1050522] [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: 09/21/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Multiple myeloma (MM) is a malignant plasma cell disorder that remains incurable for most patients, as persistent clonal evolution drives new mutations which confer MM high-risk signatures and resistance to standard care. The past two decades have significantly refashioned the therapeutic options for MM, especially adoptive T cell therapy contributing to impressive response rate and clinical efficacy. Despite great promises achieved from chimeric antigen receptor T-cell (CAR-T) therapy, the poor durability and severe toxicity (cytokine release syndrome and neurotoxicity) are still huge challenges. Therefore, relapsed/refractory multiple myeloma (RRMM), characterized by the nature of clinicopathologic and molecular heterogeneity, is frequently associated with poor prognosis. B Cell Maturation Antigen (BCMA) is the most successful target for CAR-T therapy, and other potential targets either for single-target or dual-target CAR-T are actively being studied in numerous clinical trials. Moreover, mechanisms driving resistance or relapse after CAR-T therapy remain uncharacterized, which might refer to T-cell clearance, antigen escape, and immunosuppressive tumor microenvironment. Engineering CAR T-cell to improve both efficacy and safety continues to be a promising area for investigation. In this review, we aim to describe novel tumor-associated neoantigens for MM, summarize the data from current MM CAR-T clinical trials, introduce the mechanism of disease resistance/relapse after CAR-T infusion, highlight innovations capable of enhanced efficacy and reduced toxicity, and provide potential directions to optimize manufacturing processes.
Collapse
Affiliation(s)
| | | | | | - Yongxu Jia
- *Correspondence: Yongxu Jia, ; Yanru Qin,
| | - Yanru Qin
- *Correspondence: Yongxu Jia, ; Yanru Qin,
| |
Collapse
|
71
|
Iqbal M, Bansal R, Yassine F, Gandhi S, Rosenthal A, Moustafa MA, Li Z, Craver EC, Mohty R, Murthy H, Ayala E, Tun H, Munoz J, Castro J, Lin Y, Kharfan-Dabaja MA. Impact of Rituximab and Corticosteroids on Late Cytopenias Post-Chimeric Antigen Receptor T Cell Therapy. Transplant Cell Ther 2022; 28:668.e1-668.e6. [PMID: 35842124 DOI: 10.1016/j.jtct.2022.07.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/26/2022] [Accepted: 07/07/2022] [Indexed: 11/18/2022]
Abstract
Chimeric antigen receptor (CAR) T cell therapy represents a significant advancement in the treatment of patients with relapsed/refractory B cell lymphoid malignancies. Cytokine release syndrome and immune effector cell-associated neurotoxicity represent the most acute serious adverse events post CAR T cell therapy but the occurrence and persistence of cytopenias post CAR T cell therapy represent a significant adverse event and a management challenge. While most patients typically recover blood counts by 30 days, a significant subset of patients have persistent or late cytopenias beyond 30 days. Patients receiving CAR T cell are heavily pre-treated and the impact of prior therapies on late cytopenias is not well understood. In this study, we found an association between increased number of rituximab infusions and/or cumulative rituximab dose received prior to CAR T cell infusion and persistent anemia and thrombocytopenia at 90 and 180 days afterwards. An overall increased number of prior lines of therapy was also associated with persistent lymphopenia and anemia at 90 days while receiving a prior autologous hematopoietic cell transplant was associated with a greater risk of neutropenia and lymphopenia.
Collapse
Affiliation(s)
- Madiha Iqbal
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida.
| | - Radhika Bansal
- Division Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Rochester, Minnesota
| | - Farah Yassine
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| | - Sangeetha Gandhi
- Division Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Rochester, Minnesota
| | - Allison Rosenthal
- Division Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Phoenix, Arizona
| | - Muhamad Alhaj Moustafa
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| | - Zhuo Li
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Jacksonville, Florida
| | - Emily C Craver
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Jacksonville, Florida
| | - Razan Mohty
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| | - Hemant Murthy
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| | - Ernesto Ayala
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| | - Han Tun
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| | - Javier Munoz
- Division Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Phoenix, Arizona
| | - Januario Castro
- Division Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Phoenix, Arizona
| | - Yi Lin
- Division Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Rochester, Minnesota
| | - Mohamed A Kharfan-Dabaja
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
72
|
Wang Y, Song Z, Geng Y, Gao L, Xu L, Tang G, Ni X, Chen L, Chen J, Wang T, Fu W, Feng D, Yu X, Wang L, Yang J. The risk factors and early predictive model of hematotoxicity after CD19 chimeric antigen receptor T cell therapy. Front Oncol 2022; 12:987965. [PMID: 36249041 PMCID: PMC9561932 DOI: 10.3389/fonc.2022.987965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Hematotoxicity is the most common long-term adverse event after chimeric antigen receptor T cell (CAR-T) therapy. Here, a total of 71 patients with relapsed or refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL) or large B-cell lymphoma (LBCL) were used to develop an early hematotoxicity predictive model and verify the accuracy of this model. The incidences of early hematotoxicity at 3 month following CAR-T infusion in B-ALL and LBCL were 45.5% and 38.5%, respectively. Multivariate analyses revealed that the severity of cytokine release syndrome (CRS) was an independent risk factor affecting early hematotoxicity. The analysis between the peak cytokine levels and early hematotoxicity suggested that tumor necrosis factor-α (TNF-α) and C-reactive protein (CRP) were closely associated with early hematotoxicity. Then, an early predictive model of hematotoxicity was constructed based on the peak contents of TNF-α and CRP. This model could diagnose early hematotoxicity with positive predictive values of 87.7% and 85.0% in training and validation cohorts, respectively. Lastly, we constructed the nomogram for clinical practice to predict the risk of early hematotoxicity, which performed well compared with the observed probability. This early predictive model is instrumental in the risk stratification of CAR-T recipients with hematotoxicity and early intervention for high-risk patients.
Collapse
Affiliation(s)
- Yang Wang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhiqiang Song
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yuke Geng
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Lei Gao
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Lili Xu
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Gusheng Tang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiong Ni
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Li Chen
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jie Chen
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Tao Wang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Weijia Fu
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Dongge Feng
- HuaDao Biopharma (Shanghai) Limited Corporation, Shanghai, China
| | - Xuejun Yu
- HuaDao Biopharma (Shanghai) Limited Corporation, Shanghai, China
| | - Libing Wang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
- *Correspondence: Libing Wang, ; Jianmin Yang,
| | - Jianmin Yang
- Department of Hematology, Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai, China
- *Correspondence: Libing Wang, ; Jianmin Yang,
| |
Collapse
|
73
|
Nogami A, Sasaki K. Therapeutic Advances in Immunotherapies for Hematological Malignancies. Int J Mol Sci 2022; 23:11526. [PMID: 36232824 PMCID: PMC9569660 DOI: 10.3390/ijms231911526] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/21/2022] [Accepted: 09/26/2022] [Indexed: 11/23/2022] Open
Abstract
Following the success of immunotherapies such as chimeric antigen receptor transgenic T-cell (CAR-T) therapy, bispecific T-cell engager therapy, and immune checkpoint inhibitors in the treatment of hematologic malignancies, further studies are underway to improve the efficacy of these immunotherapies and to reduce the complications associated with their use in combination with other immune checkpoint inhibitors and conventional chemotherapy. Studies of novel therapeutic strategies such as bispecific (tandem or dual) CAR-T, bispecific killer cell engager, trispecific killer cell engager, and dual affinity retargeting therapies are also underway. Because of these studies and the discovery of novel immunotherapeutic target molecules, the use of immunotherapy for diseases initially thought to be less promising to treat with this treatment method, such as acute myeloid leukemia and T-cell hematologic tumors, has become a reality. Thus, in this coming era of new transplantation- and chemotherapy-free treatment strategies, it is imperative for both scientists and clinicians to understand the molecular immunity of hematologic malignancies. In this review, we focus on the remarkable development of immunotherapies that could change the prognosis of hematologic diseases. We also review the molecular mechanisms, development processes, clinical efficacies, and problems of new agents.
Collapse
Affiliation(s)
- Ayako Nogami
- Department of Laboratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyoku, Tokyo 1138510, Japan
- Department of Hematology, Tokyo Medical and Dental University Hospital, 1-5-45 Yushima, Bunkyoku, Tokyo 1138510, Japan
| | - Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 428, Houston, TX 77030, USA
| |
Collapse
|
74
|
Si X, Gu T, Liu L, Huang Y, Han Y, Qian P, Huang H. Hematologic cytopenia post CAR T cell therapy: Etiology, potential mechanisms and perspective. Cancer Lett 2022; 550:215920. [PMID: 36122628 DOI: 10.1016/j.canlet.2022.215920] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/02/2022] [Accepted: 09/12/2022] [Indexed: 11/25/2022]
Abstract
Chimeric Antigen-Receptor (CAR) T-cell therapies have shown dramatic efficacy in treating relapsed and refractory cancers, especially B cell malignancies. However, these innovative therapies cause adverse toxicities that limit the broad application in clinical settings. Hematologic cytopenias, one frequently reported adverse event following CAR T cell treatment, are manifested as a disorder of hematopoiesis with decreased number of mature blood cells and subdivided into anemia, thrombocytopenia, leukopenia, and neutropenia, which increase the risk of infections, fatigue, bleeding, fever, and even fatality. Herein, we initially summarized the symptoms, etiology, risk factors and management of cytopenias. Further, we elaborated the cellular and molecular mechanisms underlying the initiation and progression of cytopenias following CAR T cell therapy based on previous studies about acquired cytopenias. Overall, this review will facilitate our understanding of the etiology of cytopenias and shed lights into developing new therapies against CAR T cell-induced cytopenias.
Collapse
Affiliation(s)
- Xiaohui Si
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Tianning Gu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Lianxuan Liu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Yue Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Yingli Han
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Pengxu Qian
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China.
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Hangzhou, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China.
| |
Collapse
|
75
|
Zhou J, Zhang Y, Shan M, Zong X, Geng H, Li J, Chen G, Yu L, Xu Y, Li C, Wu D. Cytopenia after chimeric antigen receptor T cell immunotherapy in relapsed or refractory lymphoma. Front Immunol 2022; 13:997589. [PMID: 36131934 PMCID: PMC9484486 DOI: 10.3389/fimmu.2022.997589] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/16/2022] [Indexed: 11/26/2022] Open
Abstract
Background Patients with relapsed or refractory (R/R) lymphomas have benefited from chimeric antigen receptor (CAR)-T-cell therapy. However, this treatment is linked to a high frequency of adverse events (AEs), such as cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and hematologic toxicity. There has been increasing interest in hematological toxicity in recent years, as it can result in additional complications, such as infection or hemorrhage, which remain intractable. Methods We conducted a retrospective, single-institution study to evaluate the patterns and outcomes of cytopenia following CAR-T-cell infusion and potential associated factors. Results Overall, 133 patients with R/R lymphoma who received CAR-T-cell therapy from June, 2017 to April, 2022 were included in this analysis. Severe neutropenia, anemia and thrombocytopenia occurred frequently (71, 30 and 41%, respectively) after CAR-T-cell infusion. A total of 98% of severe neutropenia and all severe thrombocytopenia cases occurred in the early phase. Early severe cytopenia was associated with CRS incidence and severity, as well as peak inflammatory factor (IL-6, C-reactive protein (CRP), and ferritin) levels. In multivariate analysis, prior hematopoietic stem cell transplantation (HSCT), baseline hemoglobin (HB), and lymphodepleting chemotherapy were independent adverse factors associated with early severe cytopenia. In addition, 18% and 35% of patients had late neutrophil- and platelet (PLT)-related toxicity, respectively. In multivariate analysis, lower baseline PLT count was an independent factor associated with late thrombocytopenia. More severe cytopenia was associated with higher infection rates and poorer survival. Conclusions This research indicates that improved selection of patients and management of CRS may help to decrease the severity of cytopenias and associated AEs and improve survival following CAR-T-cell therapy. Clinical Trial Registration https://www.clinicaltrials.gov/ct2/show/NCT03196830, identifier NCT03196830.
Collapse
Affiliation(s)
- Jin Zhou
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Ying Zhang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Meng Shan
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Xiangping Zong
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Hongzhi Geng
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Jiaqi Li
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Guanghua Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
| | - Lei Yu
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Yang Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
- *Correspondence: Depei Wu, ; Caixia Li, ; Yang Xu,
| | - Caixia Li
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
- *Correspondence: Depei Wu, ; Caixia Li, ; Yang Xu,
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Stem Cells and Biomedical Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China
- *Correspondence: Depei Wu, ; Caixia Li, ; Yang Xu,
| |
Collapse
|
76
|
Wudhikarn K, Perales MA. Infectious complications, immune reconstitution, and infection prophylaxis after CD19 chimeric antigen receptor T-cell therapy. Bone Marrow Transplant 2022; 57:1477-1488. [PMID: 35840746 PMCID: PMC9285870 DOI: 10.1038/s41409-022-01756-w] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/24/2022] [Accepted: 06/30/2022] [Indexed: 12/15/2022]
Abstract
CD19-targeted chimeric antigen receptor (CAR) T-cell becomes a breakthrough therapy providing excellent remission rates and durable disease control for patients with relapsed/refractory (R/R) hematologic malignancies. However, CAR T-cells have several potential side effects including cytokine release syndrome, neurotoxicities, cytopenia, and hypogammaglobulinemia. Infection has been increasingly recognized as a complication of CAR T-cell therapy. Several factors predispose CAR T-cell recipients to infection. Fortunately, although studies show a high incidence of infection post-CAR T-cells, most infections are manageable. In contrast to patients who undergo hematopoietic stem cell transplant, less is known about post-CAR T-cell immune reconstitution. Therefore, evidence regarding antimicrobial prophylaxis and vaccination strategies in these patients is more limited. As CAR T-cell therapy becomes the standard treatment for R/R B lymphoid malignancies, we should expect a larger impact of infections in these patients and the need for increased clinical attention. Studies exploring infection and immune reconstitution after CAR T-cell therapy are clinically relevant and will provide us with a better understanding of the dynamics of immune function after CAR T-cell therapy including insights into appropriate strategies for prophylaxis and treatment of infections in these patients. In this review, we describe infections in recipients of CAR T-cells, and discuss risk factors and potential mitigation strategies.
Collapse
Affiliation(s)
- Kitsada Wudhikarn
- Division of Hematology and Research Unit in Translational Hematology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| | - Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
77
|
Endowing universal CAR T-cell with immune-evasive properties using TALEN-gene editing. Nat Commun 2022; 13:3453. [PMID: 35773273 PMCID: PMC9247096 DOI: 10.1038/s41467-022-30896-2] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 05/24/2022] [Indexed: 12/29/2022] Open
Abstract
Universal CAR T-cell therapies are poised to revolutionize cancer treatment and to improve patient outcomes. However, realizing these advantages in an allogeneic setting requires universal CAR T-cells that can kill target tumor cells, avoid depletion by the host immune system, and proliferate without attacking host tissues. Here, we describe the development of a novel immune-evasive universal CAR T-cells scaffold using precise TALEN-mediated gene editing and DNA matrices vectorized by recombinant adeno-associated virus 6. We simultaneously disrupt and repurpose the endogenous TRAC and B2M loci to generate TCRαβ- and HLA-ABC-deficient T-cells expressing the CAR construct and the NK-inhibitor named HLA-E. This highly efficient gene editing process enables the engineered T-cells to evade NK cell and alloresponsive T-cell attacks and extend their persistence and antitumor activity in the presence of cytotoxic levels of NK cell in vivo and in vitro, respectively. This scaffold could enable the broad use of universal CAR T-cells in allogeneic settings and holds great promise for clinical applications.
Collapse
|
78
|
Zhang X, Zhu L, Zhang H, Chen S, Xiao Y. CAR-T Cell Therapy in Hematological Malignancies: Current Opportunities and Challenges. Front Immunol 2022; 13:927153. [PMID: 35757715 PMCID: PMC9226391 DOI: 10.3389/fimmu.2022.927153] [Citation(s) in RCA: 148] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 05/16/2022] [Indexed: 12/13/2022] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy represents a major breakthrough in cancer treatment, and it has achieved unprecedented success in hematological malignancies, especially in relapsed/refractory (R/R) B cell malignancies. At present, CD19 and BCMA are the most common targets in CAR-T cell therapy, and numerous novel therapeutic targets are being explored. However, the adverse events related to CAR-T cell therapy might be serious or even life-threatening, such as cytokine release syndrome (CRS), CAR-T-cell-related encephalopathy syndrome (CRES), infections, cytopenia, and CRS-related coagulopathy. In addition, due to antigen escape, the limited CAR-T cell persistence, and immunosuppressive tumor microenvironment, a considerable proportion of patients relapse after CAR-T cell therapy. Thus, in this review, we focus on the progress and challenges of CAR-T cell therapy in hematological malignancies, such as attractive therapeutic targets, CAR-T related toxicities, and resistance to CAR-T cell therapy, and provide some practical recommendations.
Collapse
Affiliation(s)
- Xiaomin Zhang
- Department of Hematology, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingling Zhu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Hui Zhang
- School of Medicine, Jishou University, Jishou, China
| | - Shanshan Chen
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yang Xiao
- Institute of Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Hematology, Shenzhen Qianhai Shekou Pilot Free Trade Zone Hospital, Shenzhen, China
| |
Collapse
|
79
|
Long-term safety for patients with tisagenlecleucel-treated relapsed/refractory diffuse large B-cell lymphoma. Blood Adv 2022; 6:4816-4820. [PMID: 35687492 PMCID: PMC9631665 DOI: 10.1182/bloodadvances.2021006193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 04/13/2022] [Indexed: 11/27/2022] Open
|
80
|
Gouni S, Rosenthal AC, Crombie JL, Ip A, Kamdar MK, Hess B, Feng L, Watson G, Ayers A, Neelapu SS, Khurana A, Lin Y, Iqbal M, Merryman RW, Strati P. A multicenter retrospective study of polatuzumab vedotin in patients with large B-cell lymphoma after CAR T-cell therapy. Blood Adv 2022; 6:2757-2762. [PMID: 35240681 PMCID: PMC9092406 DOI: 10.1182/bloodadvances.2021006801] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/25/2022] [Indexed: 11/24/2022] Open
Abstract
Polatuzumab vedotin (PV) is an antibody-drug conjugate targeting CD79b that is approved for patients with relapsed/refractory large B-cell lymphoma (LBCL). Patients who relapse after chimeric antigen receptor (CAR) T-cell therapy were not included in the registration study, and reports of PV use after CAR T cells are limited. This multicenter retrospective analysis included patients with LBCL who relapsed or progressed after CAR T-cell therapy and subsequently received PV with or without rituximab and bendamustine between July 2019 and May 2021. Response to treatment and progression were assessed based on the 2014 Lugano criteria. Fifty-seven patients were included in the study: 18 (32%) patients were primary refractory to CAR T-cell therapy, and 34 (60%) patients received PV-based therapy immediately after CAR T-cell therapy. PV was combined with rituximab in 54 (95%) patients and administered with bendamustine in 35 (61%) patients. A response was achieved in 25 (44%) patients, including complete remission in 8 (14%). No significant association between baseline characteristics and response was observed. After a median follow-up of 47 weeks (95% confidence interval [CI], 40-54), 46 (81%) patients had disease progression or died, and the median progression-free survival was 10 weeks (95% CI, 5-15). On a multivariate analysis, bone marrow involvement (hazard ratio, 5.2; 95% CI, 1.8-15; P = .003) and elevated lactate dehydrogenase levels (hazard ratio, 5.0; 95% CI, 1.4-16; P = .01) were associated with shorter progression-free survival. Studies aimed at better characterizing the intrinsic mechanism of resistance and identifying optimal consolidation strategies for these patients are warranted.
Collapse
Affiliation(s)
- Sushanth Gouni
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | - Andrew Ip
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ
| | | | - Brian Hess
- Division of Haematology, Medical University of South Carolina, Charleston, SC
| | - Lei Feng
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Grace Watson
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Amy Ayers
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sattva S. Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Yi Lin
- Division of Hematology, Mayo Clinic, Rochester, MN; and
| | - Madiha Iqbal
- Division of Hematology, Mayo Clinic, Jacksonville, FL
| | - Reid W. Merryman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Paolo Strati
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
81
|
Rejeski K, Perez A, Iacoboni G, Penack O, Bücklein V, Jentzsch L, Mougiakakos D, Johnson G, Arciola B, Carpio C, Blumenberg V, Hoster E, Bullinger L, Locke FL, von Bergwelt-Baildon M, Mackensen A, Bethge W, Barba P, Jain MD, Subklewe M. The CAR-HEMATOTOX risk-stratifies patients for severe infections and disease progression after CD19 CAR-T in R/R LBCL. J Immunother Cancer 2022; 10:jitc-2021-004475. [PMID: 35580927 PMCID: PMC9114843 DOI: 10.1136/jitc-2021-004475] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2022] [Indexed: 01/18/2023] Open
Abstract
Background CD19-directed chimeric antigen receptor T-cell therapy (CAR-T) represents a promising treatment modality for an increasing number of B-cell malignancies. However, prolonged cytopenias and infections substantially contribute to the toxicity burden of CAR-T. The recently developed CAR-HEMATOTOX (HT) score—composed of five pre-lymphodepletion variables (eg, absolute neutrophil count, platelet count, hemoglobin, C-reactive protein, ferritin)—enables risk stratification of hematological toxicity. Methods In this multicenter retrospective analysis, we characterized early infection events (days 0–90) and clinical outcomes in 248 patients receiving standard-of-care CD19 CAR-T for relapsed/refractory large B-cell lymphoma. This included a derivation cohort (cohort A, 179 patients) and a second independent validation cohort (cohort B, 69 patients). Cumulative incidence curves were calculated for all-grade, grade ≥3, and specific infection subtypes. Clinical outcomes were studied via Kaplan-Meier estimates. Results In a multivariate analysis adjusted for other baseline features, the HT score identified patients at high risk for severe infections (adjusted HR 6.4, 95% CI 3.1 to 13.1). HThigh patients more frequently developed severe infections (40% vs 8%, p<0.0001)—particularly severe bacterial infections (27% vs 0.9%, p<0.0001). Additionally, multivariate analysis of post-CAR-T factors revealed that infection risk was increased by prolonged neutropenia (≥14 days) and corticosteroid use (≥9 days), and decreased with fluoroquinolone prophylaxis. Antibacterial prophylaxis significantly reduced the likelihood of severe bacterial infections in HThigh (16% vs 46%, p<0.001), but not HTlow patients (0% vs 2%, p=n.s.). Collectively, HThigh patients experienced worse median progression-free (3.4 vs 12.6 months) and overall survival (9.1 months vs not-reached), and were hospitalized longer (median 20 vs 16 days). Severe infections represented the most common cause of non-relapse mortality after CAR-T and were associated with poor survival outcomes. A trend toward increased non-relapse mortality in HThigh patients was observed (8.0% vs 3.7%, p=0.09). Conclusions These data demonstrate the utility of the HT score to risk-stratify patients for infectious complications and poor survival outcomes prior to CD19 CAR-T. High-risk patients likely benefit from anti-infective prophylaxis and should be closely monitored for potential infections and relapse.
Collapse
Affiliation(s)
- Kai Rejeski
- Department of Medicine III, Hematology and Oncology, University Hospital, LMU Munich, Munich, Germany.,Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany.,German Cancer Consortium (DKTK) Munich Site, and German Cancer Research Center, Heidelberg, Germany
| | - Ariel Perez
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida, USA.,Blood & Marrow Transplant Program, Miami Cancer Institute, Miami, Florida, USA
| | - Gloria Iacoboni
- Department of Hematology, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, Universitat Autònoma of Barcelona (UAB), Department of Medicin, Barcelona, Spain
| | - Olaf Penack
- Department of Hematology, Oncology and Tumorimmunology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Cancer Consortium (DKTK) Berlin Site, and German Cancer Research Center, Heidelberg, Germany
| | - Veit Bücklein
- Department of Medicine III, Hematology and Oncology, University Hospital, LMU Munich, Munich, Germany.,Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Liv Jentzsch
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Dimitrios Mougiakakos
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Grace Johnson
- USF Morsani College of Medicine, Tampa, Florida, USA
| | - Brian Arciola
- USF Morsani College of Medicine, Tampa, Florida, USA
| | - Cecilia Carpio
- Department of Hematology, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, Universitat Autònoma of Barcelona (UAB), Department of Medicin, Barcelona, Spain
| | - Viktoria Blumenberg
- Department of Medicine III, Hematology and Oncology, University Hospital, LMU Munich, Munich, Germany.,Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Eva Hoster
- Institute for Medical Information Processing, Biometry and Epidemiology (IBE), LMU Munich, Munich, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology and Tumorimmunology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Cancer Consortium (DKTK) Berlin Site, and German Cancer Research Center, Heidelberg, Germany
| | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida, USA
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, Hematology and Oncology, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium (DKTK) Munich Site, and German Cancer Research Center, Heidelberg, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology and Oncology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Wolfgang Bethge
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Pere Barba
- Department of Hematology, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, Universitat Autònoma of Barcelona (UAB), Department of Medicin, Barcelona, Spain
| | - Michael D Jain
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida, USA
| | - Marion Subklewe
- Department of Medicine III, Hematology and Oncology, University Hospital, LMU Munich, Munich, Germany .,Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany.,German Cancer Consortium (DKTK) Munich Site, and German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
82
|
Busca A, Salmanton-García J, Corradini P, Marchesi F, Cabirta A, Di Blasi R, Dulery R, Lamure S, Farina F, Weinbergerová B, Batinić J, Nordlander A, López-García A, Drgoňa Ľ, Espigado-Tocino I, Falces-Romero I, García-Sanz R, García-Vidal C, Guidetti A, Khanna N, Kulasekararaj A, Maertens J, Hoenigl M, Klimko N, Koehler P, Pagliuca A, Passamonti F, Cornely OA, Pagano L. COVID-19 and CAR T cells: a report on current challenges and future directions from the EPICOVIDEHA survey by EHA-IDWP. Blood Adv 2022; 6:2427-2433. [PMID: 34749396 PMCID: PMC8575532 DOI: 10.1182/bloodadvances.2021005616] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/24/2021] [Indexed: 12/15/2022] Open
Abstract
The EHA-IDWP developed an observational registry collecting data on COVID-19 infection in patients who received CAR T-cell therapy. Prevalence of COVID-19 was 4.8%, and overall mortality was 50%, highlighting the need for prevention of infection in these patients.
Collapse
Affiliation(s)
- Alessandro Busca
- Stem Cell Transplant Center, Azienda Ospedaliera Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Jon Salmanton-García
- Department I of Internal Medicine, Excellence Center for Medical Mycology (ECMM), Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University Hospital Cologne, Cologne, Germany
| | - Paolo Corradini
- University of Milan and Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesco Marchesi
- Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Alba Cabirta
- Department of Hematology, Vall d’Hebron Hospital Universitari, Vall d’Hebron Istitute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Roberta Di Blasi
- Hôpital Saint Louis, Assistance Publique–Hôpitaux de Paris (AP-HP), Paris, France
| | - Remy Dulery
- Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, AP-HP, Unité Mixte de Recherche (UMR) 938, Sorbonne Universitè, Paris, France
| | - Sylvain Lamure
- Department of Clinical Hematology, Montpellier University Hospital, IGMM UMR1535 CNRS, University of Montpellier, Montpellier, France
| | | | - Barbora Weinbergerová
- Department of Internal Medicine-Hematology and Oncology, Masaryk University Hospital Brno, Brno, Czech Republic
| | - Josip Batinić
- University Hospital Centre Zagreb, Zagreb, Croatia
- Croatian Cooperative Group for Hematological Diseases (CROHEM), Zagreb, Croatia
- Faculty of Medicine, University of Zagreb, Zagreb, Croatia
| | - Anna Nordlander
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | | | - Ľuboš Drgoňa
- Comenius University and National Cancer Institute, Bratislava, Slovakia
| | | | | | | | | | - Anna Guidetti
- Division of Hematology and Bone Marrow Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, University of Milano, Milan, Italy
| | - Nina Khanna
- Division of Infectious Diseases and Hospital Epidemiology, Department of Clinical Research, University and University Hospital of Basel, Basel, Switzerland
| | | | | | - Martin Hoenigl
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, San Diego, CA
- Clinical and Translational Fungal Working Group, University of California San Diego, La Jolla, CA
- Division of Infectious Diseases, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Nikolai Klimko
- Department of Clinical Mycology, Allergy and Immunology, North Western State Medical University, St Petersburg, Russia
| | - Philipp Koehler
- Department I of Internal Medicine, Excellence Center for Medical Mycology (ECMM), Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University Hospital Cologne, Cologne, Germany
| | - Antonio Pagliuca
- Department of Hematological Medicine, Kings College Hospital, National Health Service Foundation Trust, London, United Kingdom
| | - Francesco Passamonti
- Department of Medicine and Surgery, University of Insubria and Azienda Socio Sanitaria Territoriale Sette Laghi, Ospedale di Circolo of Varese, Varese, Italy
| | - Oliver A. Cornely
- Department I of Internal Medicine, Excellence Center for Medical Mycology (ECMM), Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University Hospital Cologne, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinical Trials Centre Cologne, Cologne, Germany
- German Centre for Infection Research, Partner Site Bonn-Cologne, Cologne, Germany
| | - Livio Pagano
- Hematology, IRCCS Fondazione Policlinico Universitario Agostino Gemelli, Rome, Italy; and
- Hematology, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
83
|
Kampouri E, Walti CS, Gauthier J, Hill JA. Managing Hypogammaglobulinemia in Patients Treated with CAR-T-cell Therapy: Key Points for Clinicians. Expert Rev Hematol 2022; 15:305-320. [PMID: 35385358 DOI: 10.1080/17474086.2022.2063833] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The unprecedented success of chimeric antigen receptor (CAR)-T-cell therapy in the management of B-cell malignancies comes with a price of specific side effects. Healthy B-cell depletion is an anticipated 'on-target' 'off-tumor' side effect and can contribute to severe and prolonged hypogammaglobulinemia. Evidence-based guidelines for the use of immunoglobulin replacement therapy (IGRT) for infection prevention are lacking in this population. AREAS COVERED This article reviews the mechanisms and epidemiology of hypogammaglobulinemia and antibody deficiency, association with infections, and strategies to address these issues in CD19- and BCMA-CAR-T-cell recipients. EXPERT OPINION CD19 and BCMA CAR-T-cell therapy result in unique immune deficits due to depletion of specific B-lineage cells and may require different infection prevention strategies. Hypogammaglobulinemia before and after CAR-T-cell therapy is frequent, but data on the efficacy and cost-effectiveness of IGRT are lacking. Monthly IGRT should be prioritized for patients with severe or recurrent bacterial infections. IGRT may be more broadly necessary to prevent infections in BCMA-CAR-T-cell recipients and children with severe hypogammaglobulinemia irrespective of infection history. Vaccinations are indicated to augment humoral immunity and can be immunogenic despite cytopenias; re-vaccination(s) may be required. Controlled trials are needed to better understand the role of IGRT and vaccines in this population.
Collapse
Affiliation(s)
- Eleftheria Kampouri
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Infectious Diseases Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Carla S Walti
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Infectious Disease and Hospital Epidemiology Division, University Hospital Basel, Basel, Switzerland
| | - Jordan Gauthier
- Department of Medicine, University of Washington, Seattle, Washington, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Joshua A Hill
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
84
|
Sharma N, Reagan PM, Liesveld JL. Cytopenia after CAR-T Cell Therapy-A Brief Review of a Complex Problem. Cancers (Basel) 2022; 14:1501. [PMID: 35326654 PMCID: PMC8946106 DOI: 10.3390/cancers14061501] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/02/2022] [Accepted: 03/07/2022] [Indexed: 02/06/2023] Open
Abstract
Chimeric Antigen Receptor T-cell (CAR-T) immunotherapy has emerged as an efficacious and life extending treatment modality with high response rates and durable remissions in patients with relapsed and refractory non-Hodgkin lymphoma (NHL), follicular lymphoma, and B-cell acute lymphoblastic leukemia (B-ALL) as well as in other diseases. Prolonged or recurrent cytopenias after CAR-T therapy have increasingly been reported at varying rates, and the pathogenesis of this complication is not yet well-understood but is likely contributed to by multiple factors. Current studies reported are primarily retrospective, heterogeneous in terms of CAR-Ts used and diseases treated, non-uniform in definitions of cytopenias and durations for end points, and vary in terms of recommended management. Prospective studies and correlative laboratory studies investigating the pathophysiology of prolonged cytopenias will enhance our understanding of this phenomenon. This review summarizes knowledge of these cytopenias to date.
Collapse
Affiliation(s)
- Naman Sharma
- Department of Hematology-Oncology, Baystate Medical Center, University of Massachusetts Medical School, Springfield, MA 100107, USA;
| | - Patrick M. Reagan
- Department of Medicine, Hematology-Oncology, James P. Wilmot Cancer Institute, University of Rochester, Rochester, NY 14642, USA;
| | - Jane L. Liesveld
- Department of Medicine, Hematology-Oncology, James P. Wilmot Cancer Institute, University of Rochester, Rochester, NY 14642, USA;
| |
Collapse
|
85
|
Hayden PJ, Roddie C, Bader P, Basak GW, Bonig H, Bonini C, Chabannon C, Ciceri F, Corbacioglu S, Ellard R, Sanchez-Guijo F, Jäger U, Hildebrandt M, Hudecek M, Kersten MJ, Köhl U, Kuball J, Mielke S, Mohty M, Murray J, Nagler A, Rees J, Rioufol C, Saccardi R, Snowden JA, Styczynski J, Subklewe M, Thieblemont C, Topp M, Ispizua ÁU, Chen D, Vrhovac R, Gribben JG, Kröger N, Einsele H, Yakoub-Agha I. Management of adults and children receiving CAR T-cell therapy: 2021 best practice recommendations of the European Society for Blood and Marrow Transplantation (EBMT) and the Joint Accreditation Committee of ISCT and EBMT (JACIE) and the European Haematology Association (EHA). Ann Oncol 2022; 33:259-275. [PMID: 34923107 DOI: 10.1016/j.annonc.2021.12.003] [Citation(s) in RCA: 220] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Several commercial and academic autologous chimeric antigen receptor T-cell (CAR-T) products targeting CD19 have been approved in Europe for relapsed/refractory B-cell acute lymphoblastic leukemia, high-grade B-cell lymphoma and mantle cell lymphoma. Products for other diseases such as multiple myeloma and follicular lymphoma are likely to be approved by the European Medicines Agency in the near future. DESIGN The European Society for Blood and Marrow Transplantation (EBMT)-Joint Accreditation Committee of ISCT and EBMT (JACIE) and the European Haematology Association collaborated to draft best practice recommendations based on the current literature to support health care professionals in delivering consistent, high-quality care in this rapidly moving field. RESULTS Thirty-six CAR-T experts (medical, nursing, pharmacy/laboratory) assembled to draft recommendations to cover all aspects of CAR-T patient care and supply chain management, from patient selection to long-term follow-up, post-authorisation safety surveillance and regulatory issues. CONCLUSIONS We provide practical, clinically relevant recommendations on the use of these high-cost, logistically complex therapies for haematologists/oncologists, nurses and other stakeholders including pharmacists and health sector administrators involved in the delivery of CAR-T in the clinic.
Collapse
Affiliation(s)
- P J Hayden
- Department of Haematology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - C Roddie
- UCL Cancer Institute, London, UK; University College London Hospital NHS Foundation Trust, London, UK.
| | - P Bader
- Clinic for Children and Adolescents, University Children's Hospital, Frankfurt, Germany
| | - G W Basak
- Medical University of Warsaw, Department of Hematology, Transplantation and Internal Medicine, Warsaw, Poland
| | - H Bonig
- Institute for Transfusion Medicine and Immunohematology of Goethe University and German Red Cross Blood Service, Frankfurt, Germany
| | - C Bonini
- Ospedale San Raffaele and Università Vita-Salute San Raffaele, Milan, Italy
| | - C Chabannon
- Aix-Marseille université, Inserm CBT-1409, Institut Paoli-Calmettes, centre de thérapie cellulaire, unité de transplantation et de thérapie cellulaire, département de biologie du cancer, Marseille, France
| | - F Ciceri
- Università Vita-Salute San Raffaele, IRCCS Ospedale San Raffaele, Milan, Italy
| | - S Corbacioglu
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Franz-Josef-Strauss-Allee 11, University Hospital of Regensburg, Regensburg, Germany
| | - R Ellard
- Royal Marsden Hospital, Fulham Rd, London, UK
| | - F Sanchez-Guijo
- IBSAL-Hospital Universitario de Salamanca, CIC, Universidad de Salamanca, Salamanca, Spain
| | - U Jäger
- Clinical Department for Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - M Hildebrandt
- Department of Transfusion Medicine, Cell Therapeutics and Haemostaseology, LMU University Hospital Grosshadern, Munich
| | - M Hudecek
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - M J Kersten
- Department of Hematology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam and LYMMCARE, Amsterdam, the Netherlands
| | - U Köhl
- Fraunhofer Institute for Cell Therapy and Immunology (IZI) and Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany; Institute of Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| | - J Kuball
- Department of Hematology and Centre for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - S Mielke
- Karolinska Institutet and University Hospital, Department of Laboratory Medicine/Department of Cell Therapy and Allogeneic Stem Cell Transplantation (CAST), Stockholm, Sweden
| | - M Mohty
- Hôpital Saint-Antoine, APHP, Sorbonne Université, INSERM UMRs 938, Paris, France
| | - J Murray
- Christie Hospital NHS Trust, Manchester, UK
| | - A Nagler
- The Chaim Sheba Medical Center, Tel-Hashomer, affiliated with the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - J Rees
- University College London Hospital NHS Foundation Trust, London, UK; UCL Institute of Neurology, University College of London Hospitals NHS Foundation Trust, London, UK
| | - C Rioufol
- Hospices Civils de Lyon, UCBL1, EMR 3738 CICLY, Lyon, France
| | - R Saccardi
- Cell Therapy and Transfusion Medicine Department, Careggi University Hospital, Florence, Italy
| | - J A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - J Styczynski
- Department of Pediatric Hematology and Oncology, Collegium Medicum, Nicolaus Copernicus University Torun, Bydgoszcz, Poland
| | - M Subklewe
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - C Thieblemont
- AP-HP, Saint-Louis Hospital, Hemato-oncology, University of Paris, Paris, France
| | - M Topp
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Á U Ispizua
- Department of Hematology, ICMHO, Hospital Clínic de Barcelona, Barcelona, Spain
| | - D Chen
- University College London Hospital NHS Foundation Trust, London, UK; Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - R Vrhovac
- Department of Haematology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - J G Gribben
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - N Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg, Germany
| | - H Einsele
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - I Yakoub-Agha
- CHU de Lille, Univ Lille, INSERM U1286, Infinite, Lille, France
| |
Collapse
|
86
|
Luo W, Li C, Zhang Y, Du M, Kou H, Lu C, Mei H, Hu Y. Adverse effects in hematologic malignancies treated with chimeric antigen receptor (CAR) T cell therapy: a systematic review and Meta-analysis. BMC Cancer 2022; 22:98. [PMID: 35073859 PMCID: PMC8785493 DOI: 10.1186/s12885-021-09102-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/12/2021] [Indexed: 12/13/2022] Open
Abstract
Abstract
Background
Recently, chimeric antigen receptor-modified (CAR) T cell therapy for hematological malignancies has shown clinical efficacy. Hundreds of clinical trials have been registered and lots of studies have shown hematologic toxic effects were very common. The main purpose of this review is to systematically analyze hematologic toxicity in hematologic malignancies treated with CAR-T cell therapy.
Methods
We searched databases including PubMed, Web of Science, Embase and Cochrane up to January 2021. For safety analysis of overall hematologic toxicity, the rate of neutrophil, thrombocytopenia and anemia were calculated. Subgroup analysis was performed for age, pathological type, target antigen, co-stimulatory molecule, history of hematopoietic stem cell transplantation (HSCT) and prior therapy lines. The incidence rate of aspartate transferase (AST) increased, alanine transaminase (ALT) increased, serum creatine increased, APTT prolonged and fibrinogen decreased were also calculated.
Results
Overall, 52 studies involving 2004 patients were included in this meta-analysis. The incidence of any grade neutropenia, thrombocytopenia and anemia was 80% (95% CI: 68–89%), 61% (95% CI: 49–73%), and 68% (95%CI: 54–80%) respectively. The incidences of grade ≥ 3 neutropenia, thrombocytopenia and anemia were 60% (95% CI: 49–70%), 33% (95% CI: 27–40%), and 32% (95%CI: 25–40%) respectively. According to subgroup analysis and the corresponding Z test, hematological toxicity was more frequent in younger patients, in patients with ≥4 median lines of prior therapy and in anti-CD19 cases. The subgroup analysis of CD19 CAR-T cell constructs showed that 41BB resulted in less hematological toxicity than CD28.
Conclusion
CAR-T cell therapy has dramatical efficacy in hematological malignancies, but the relevant adverse effects remain its obstacle. The most common ≥3 grade side effect is hematological toxicity, and some cases die from infections or severe hemorrhage in early period. In long-term follow-up, hematological toxicity is less life-threatening generally and most suffered patients recover to adequate levels after 3 months. To prevent life-threatening infections or bleeding events, clinicians should pay attention to intervention of hematological toxicity in the early process of CAR-T cell therapy.
Collapse
|
87
|
Liu H, Li P, Zhao A, Lei W, Liang A, Qian W. Incidence and prophylaxis of herpes zoster in relapsed or refractory B-cell lymphoma patients after CD19-specific CAR-T cell therapy. Leuk Lymphoma 2021; 63:1001-1004. [PMID: 34842037 DOI: 10.1080/10428194.2021.2010062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Hui Liu
- Department of Hematology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Ping Li
- Department of Hematology, Tongji Hospital of Tongji University, Shanghai, People's Republic of China
| | - Aiqi Zhao
- Department of Hematology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Wen Lei
- Department of Hematology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Aibin Liang
- Department of Hematology, Tongji Hospital of Tongji University, Shanghai, People's Republic of China
| | - Wenbin Qian
- Department of Hematology, the Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China.,National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University
| |
Collapse
|
88
|
Pochon C, Courbon C, Bay JO, Moreau AS, Paul F, Picard M, Sterin A, Tudesq JJ, Vicente C, Yakoub-Agha M, Yakoub-Agha I. [Complications other than infections, CRS and ICANS following CAR T-cells therapy: Recommendations of the Francophone Society of bone marrow transplantation and cell therapy (SFGM-TC)]. Bull Cancer 2021; 108:S98-S103. [PMID: 34802718 DOI: 10.1016/j.bulcan.2021.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/15/2021] [Indexed: 10/19/2022]
Abstract
CAR-T cells are an innovative treatment for an increasing number of patients, particularly since the extension of their indication to mantle lymphoma and multiple myeloma. Several complications of CAR T-cell therapy, that were first described as exceptional, have now been reported in series of patients, since its first clinical use in 2011. Among them, cardiac complications, delayed cytopenias, acute and chronic Graft versus Host Disease, and tumoral lysis syndrome are recognized as specific potent complications following CAR T-cells infusion. During the twelfth edition of practice harmonization workshops of the Francophone society of bone marrow transplantation and cellular therapy (SFGM-TC), a working group focused its work on the management of these complications with focuses the epidemiology, the physiopathology and the risk factors of these 4 side effects. Our recommendations apply to commercial CAR-T cells, in order to guide strategies for the management of complications associated with this new therapeutic approach.
Collapse
Affiliation(s)
- Cécile Pochon
- CHRU de Nancy, service d'onco-hématologie pédiatrique, 54500 Vandœuvre-Lès-Nancy, France; Université de Lorraine, UMR 7365 CNRS-UL IMoPA, campus biologie santé, 9, avenue de la Forêt-de-Haye, 54505 Vandœuvre-Lès-Nancy, France.
| | - Corinne Courbon
- Institut de cancérologie de la Loire, service d'hématologie, 42270 Saint-Priest-en-Jarez, France
| | - Jacques-Olivier Bay
- CHU de Clermont-Ferrand, service de thérapie cellulaire et d'hématologie clinique adulte, site Estaing, 1, place Lucie-Aubrac, 63000 Clermont-Ferrand, France
| | - Anne-Sophie Moreau
- Hôpital Salengro, CHU de Lille, service de médecine intensive réanimation, rue Émile-Laine, 59037 Lille cedex, France
| | - Franciane Paul
- Université Montpellier, CHU de Montpellier, département d'hématologie clinique, Montpellier, France
| | - Muriel Picard
- CHU de Toulouse, réanimation polyvalente IUCT-oncopole, Toulouse, France
| | - Arthur Sterin
- Hôpital La Timone Enfants, service hémato-immunologie pédiatrique, 13005 Marseille, France
| | - Jean-Jacques Tudesq
- Université Montpellier, CHU de Montpellier, département d'hématologie clinique, Montpellier, France
| | - Céline Vicente
- CHU de Toulouse, service d'hématologie, IUCT-oncopole, 1, avenue Joliot-Curie, 31059 Toulouse, France
| | | | | |
Collapse
|
89
|
Clé DV, Hirayama AV, Alencar AJ, Costa LJ, Feliciano JVP, Mattos ER, Cordeiro AC, Salvino MA, Barros GMN, de Lima M, Scheinberg P, Guerino-Cunha RL. Associação Brasileira de Hematologia, Hemoterapia e Terapia Celular Consensus on genetically modified cells. I: Structuring centers for the multidisciplinary clinical administration and management of CAR-T cell therapy patients. Hematol Transfus Cell Ther 2021; 43 Suppl 2:S3-S12. [PMID: 34794793 PMCID: PMC8606713 DOI: 10.1016/j.htct.2021.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 09/14/2021] [Indexed: 01/16/2023] Open
Abstract
Chimeric antigen receptor T-cells (CAR-T cells) are a new modality of oncological treatment which has demonstrated impressive response in refractory or relapsed diseases, such as acute lymphoblastic leukemia (ALL), lymphomas, and myeloma but is also associated with unique and potentially life-threatening toxicities. The most common adverse events (AEs) include cytokine release syndrome (CRS), neurological toxicities, such as the immune effector cell-associated neurotoxicity syndrome (ICANS), cytopenias, infections, and hypogammaglobulinemia. These may be severe and require admission of the patient to an intensive care unit. However, these AEs are manageable when recognized early and treated by a duly trained team. The objective of this article is to report a consensus compiled by specialists in the fields of oncohematology, bone marrow transplantation, and cellular therapy describing recommendations on the Clinical Centers preparation, training of teams that will use CAR-T cells, and leading clinical questions as to their use and the management of potential complications.
Collapse
Affiliation(s)
- Diego V Clé
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil.
| | | | - Alvaro J Alencar
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Luciano J Costa
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - João V P Feliciano
- Hospital de Base, Faculdade de Medicina de Rio Preto (HB FAMERP), São José do Rio Preto, SP, Brazil
| | | | | | | | | | | | | | - Renato L Guerino-Cunha
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (FMRP-USP), Ribeirão Preto, SP, Brazil
| |
Collapse
|
90
|
Johnsrud A, Craig J, Baird J, Spiegel J, Muffly L, Zehnder J, Tamaresis J, Negrin R, Johnston L, Arai S, Shizuru J, Lowsky R, Meyer E, Weng WK, Shiraz P, Rezvani A, Latchford T, Mackall C, Miklos D, Frank M, Sidana S. Incidence and risk factors associated with bleeding and thrombosis following chimeric antigen receptor T-cell therapy. Blood Adv 2021; 5:4465-4475. [PMID: 34521106 PMCID: PMC8579267 DOI: 10.1182/bloodadvances.2021004716] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/22/2021] [Indexed: 01/16/2023] Open
Abstract
Bleeding and thrombotic events are an emerging toxicity associated with chimeric antigen receptor (CAR) therapies. To determine their incidence, we retrospectively analyzed consecutive adult patients (N = 127) with large B-cell lymphoma (LBCL) or B-cell acute lymphoblastic leukemia (B-ALL) treated from 2017 through 2020 with axicabtagene ciloleucel (axi-cel; n = 89) or a bispecific CD19/CD22 CAR (n = 38). Twelve (9.4%) and 8 (6.3%) patients developed bleeding and thrombosis within the first 3 months, respectively. In the axi-cel subgroup, these occurred in 11.2% and 6.7%, respectively. Bleeding occurred between days 8 and 30 (median, 17.5) and thrombosis between days 2 and 91 (median, 29). Bleeding sites included genitourinary, soft tissue, intracranial, gastrointestinal, and pulmonary and were associated with features of consumptive coagulopathy. On univariate analysis, patients with bleeding were older, had lower baseline platelets (86 × 103/μL vs 178 × 103/μL; P < .01), lower platelet and fibrinogen nadirs , and elevated lactate dehydrogenase. Immune effector cell (IEC)-associated neurotoxicity syndrome (ICANS) grade ≥3 was associated with increased bleeding (50% vs 15%; P = .01), thrombosis (50% vs 16%; P = .04), prothrombin time prolongation, hypofibrinogenemia, and elevated D-dimer. Low pretreatment platelet counts were associated with bleeding in a multivariate logistic regression model. Patients with thrombocytopenia or severe ICANS are at increased risk of bleeding and should be closely monitored, particularly within the first month after CAR therapy. Future studies in larger cohorts should assess risk factors for systemic coagulopathies in CAR T therapy, including their association with neurotoxicity.
Collapse
Affiliation(s)
- Andrew Johnsrud
- Division of Blood and Marrow Transplantation and Cellular Therapy; and
| | - Juliana Craig
- Division of Blood and Marrow Transplantation and Cellular Therapy; and
| | - John Baird
- Division of Blood and Marrow Transplantation and Cellular Therapy; and
| | - Jay Spiegel
- Division of Blood and Marrow Transplantation and Cellular Therapy; and
| | - Lori Muffly
- Division of Blood and Marrow Transplantation and Cellular Therapy; and
| | | | - John Tamaresis
- Department of Biomedical Data Science, Stanford University, Stanford, CA
| | - Robert Negrin
- Division of Blood and Marrow Transplantation and Cellular Therapy; and
| | - Laura Johnston
- Division of Blood and Marrow Transplantation and Cellular Therapy; and
| | - Sally Arai
- Division of Blood and Marrow Transplantation and Cellular Therapy; and
| | - Judith Shizuru
- Division of Blood and Marrow Transplantation and Cellular Therapy; and
| | - Robert Lowsky
- Division of Blood and Marrow Transplantation and Cellular Therapy; and
| | - Everett Meyer
- Division of Blood and Marrow Transplantation and Cellular Therapy; and
| | - Wen-Kai Weng
- Division of Blood and Marrow Transplantation and Cellular Therapy; and
| | - Parveen Shiraz
- Division of Blood and Marrow Transplantation and Cellular Therapy; and
| | - Andrew Rezvani
- Division of Blood and Marrow Transplantation and Cellular Therapy; and
| | - Theresa Latchford
- Division of Blood and Marrow Transplantation and Cellular Therapy; and
| | - Crystal Mackall
- Division of Blood and Marrow Transplantation and Cellular Therapy; and
| | - David Miklos
- Division of Blood and Marrow Transplantation and Cellular Therapy; and
| | - Matthew Frank
- Division of Blood and Marrow Transplantation and Cellular Therapy; and
| | - Surbhi Sidana
- Division of Blood and Marrow Transplantation and Cellular Therapy; and
| |
Collapse
|
91
|
Al-Juhaishi T, Ahmed S. CAR-T in B-Cell Lymphomas: The Past, Present, and Future. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 22:e261-e268. [PMID: 34782260 DOI: 10.1016/j.clml.2021.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/27/2021] [Accepted: 10/09/2021] [Indexed: 01/01/2023]
Abstract
Aggressive B-cell lymphomas including diffuse large B-cell lymphoma make up the majority of non-Hodgkin's lymphoma globally. While more than half of these patients can be cured with modern chemoimmunotherapy regimens, the outcomes of relapsed or refractory disease continue to be very poor. Despite significant developments in targeted cancer therapies and immuno-oncology, the attainability of a cure remained an elusive goal outside of incorporating high doses of chemotherapy followed by hematopoietic stem cell transplantation, for patients who have chemosensitive disease. The development of chimeric antigen receptor T-cell therapy changed that paradigm and introduced a new field of therapeutic possibilities for these patients. In this review, we will discuss the current state of this therapeutic modality in B-cell lymphomas and provide opinions on where future efforts need to focus in order to further improve their clinical utility.
Collapse
Affiliation(s)
| | - Sairah Ahmed
- University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
92
|
Marasco V, Carniti C, Guidetti A, Farina L, Magni M, Miceli R, Calabretta L, Verderio P, Ljevar S, Serpenti F, Morelli D, Apolone G, Ippolito G, Agrati C, Corradini P. T-cell immune response after mRNA SARS-CoV-2 vaccines is frequently detected also in the absence of seroconversion in patients with lymphoid malignancies. Br J Haematol 2021; 196:548-558. [PMID: 34649298 PMCID: PMC8653177 DOI: 10.1111/bjh.17877] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/08/2021] [Accepted: 09/22/2021] [Indexed: 12/27/2022]
Abstract
Patients affected by lymphoid malignancies (LM) are frequently immune‐compromised, suffering increased mortality from COVID‐19. This prospective study evaluated serological and T‐cell responses after complete mRNA vaccination in 263 patients affected by chronic lymphocytic leukaemia, B‐ and T‐cell lymphomas and multiple myeloma. Results were compared with those of 167 healthy subjects matched for age and sex. Overall, patient seroconversion rate was 64·6%: serological response was lower in those receiving anti‐cancer treatments in the 12 months before vaccination: 55% vs 81·9% (P < 0·001). Anti‐CD20 antibody plus chemotherapy treatment was associated with the lowest seroconversion rate: 17·6% vs. 71·2% (P < 0·001). In the multivariate analysis conducted in the subgroup of patients on active treatment, independent predictors for seroconversion were: anti‐CD20 treatment (P < 0·001), aggressive B‐cell lymphoma diagnosis (P = 0·002), and immunoglobulin M levels <40 mg/dl (P = 0·030). The T‐cell response was evaluated in 99 patients and detected in 85 of them (86%). Of note, 74% of seronegative patients had a T‐cell response, but both cellular and humoral responses were absent in 13·1% of cases. Our findings raise some concerns about the protection that patients with LM, particularly those receiving anti‐CD20 antibodies, may gain from vaccination. These patients should strictly maintain all the protective measures.
Collapse
Affiliation(s)
| | - Cristiana Carniti
- Division of Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Italy
| | - Anna Guidetti
- School of Medicine, University of Milano, Italy.,Division of Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Italy
| | - Lucia Farina
- Division of Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Italy
| | - Martina Magni
- Division of Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Italy
| | - Rosalba Miceli
- Department of Clinical Epidemiology and Trial Organization, Fondazione IRCCS Istituto Nazionale dei Tumori, Italy
| | | | - Paolo Verderio
- Unit of Bioinformatics and Biostatistics, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Italy
| | - Silva Ljevar
- Department of Clinical Epidemiology and Trial Organization, Fondazione IRCCS Istituto Nazionale dei Tumori, Italy
| | | | - Daniele Morelli
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Italy
| | - Giovanni Apolone
- Scientific Directorate, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Giuseppe Ippolito
- National Institute for Infectious Diseases "Lazzaro Spallanzani" I.R.C.C.S, Italy
| | - Chiara Agrati
- Cellular Immunology Laboratory, INMI L Spallanzani, Rome, Italy
| | - Paolo Corradini
- School of Medicine, University of Milano, Italy.,Division of Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Italy
| |
Collapse
|
93
|
Handley G, Khawaja F, Kondapi DS, Lee HJ, Kaufman GP, Neelapu SS, Fayad LE, Tummala S, Chi L, Strati P, Mulanovich VE. Human herpesvirus 6 myelitis after chimeric antigen receptor T-cell therapy. Int J Infect Dis 2021; 112:327-329. [PMID: 34600133 DOI: 10.1016/j.ijid.2021.09.061] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/30/2021] [Accepted: 09/26/2021] [Indexed: 11/16/2022] Open
Abstract
This article reports a fatal case of human herpesvirus 6 (HHV-6) myelitis following CD19-targeted chimeric antigen receptor T-cell therapy. Infection from HHV-6 reactivation after haematopoietic stem cell transplant is established, and outside of this population is limited to case reports. The patient developed cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome that responded to corticosteroids both clinically and on imaging. Subsequently, ascending flaccid paralysis developed, leading to neuromuscular respiratory failure and, ultimately, death. Disease progression was refractory to foscarnet and multiple immunomodulating agents. HHV-6 should be considered in patients with encephalitis and myelitis after adoptive T-cell therapy.
Collapse
Affiliation(s)
- Guy Handley
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Medicine, Division of Infectious Disease and International Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| | - Fareed Khawaja
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Divya S Kondapi
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hun J Lee
- Department of Lymphoma-Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gregory P Kaufman
- Department of Lymphoma-Myeloma, 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
| | - Luis E Fayad
- Department of Lymphoma-Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sudhakar Tummala
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linda Chi
- Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paolo Strati
- Department of Lymphoma-Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Victor E Mulanovich
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
94
|
Cousin E, Belicard F, Michel L, Pronier C, Lassalle N, Lamy T, Houot R, Lhomme F. Severe cytomegalovirus disease with encephalitis after CAR-T cell therapy: A rare but potentially fatal complication. J Med Virol 2021; 93:6398-6403. [PMID: 34370316 DOI: 10.1002/jmv.27257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/02/2021] [Indexed: 11/09/2022]
Abstract
Cytomegalovirus (CMV) is a ubiquitous herpes virus that develops lifelong latency following primary infection and can be reactivated following immune suppression. CMV encephalopathy has been described in few reports after hematopoietic stem cell transplantation and in patients with acquired immunodeficiency syndrome. To the best of our knowledge, CMV encephalopathy following CAR-T cells infusion had not been previously reported. Initial CMV viral load and monitoring are crucial in patients with CAR-T cells to allow early intervention with aggressive antiviral treatment without delay if needed.
Collapse
Affiliation(s)
- Elie Cousin
- Department of Hematology, Rennes University Hospital, Rennes, France
| | - Félicie Belicard
- Department of Hematology, Rennes University Hospital, Rennes, France
| | - Laure Michel
- Department of Neurology, Rennes University Hospital, Rennes, France.,Clinical Neuroscience Centre, Rennes University Hospital, Rennes University, Rennes, France
| | | | - Nicolas Lassalle
- Department of Radiology, Rennes University Hospital, Rennes, France
| | - Thierry Lamy
- Department of Hematology, Rennes University Hospital, Rennes, France
| | - Roch Houot
- Department of Hematology, Rennes University Hospital, Rennes, France
| | - Faustine Lhomme
- Department of Hematology, Rennes University Hospital, Rennes, France
| |
Collapse
|
95
|
Tang K, Nastoupil LJ. Real-World Experiences of CAR T-Cell Therapy for Large B-Cell Lymphoma: How Similar Are They to the Prospective Studies? JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2021; 4:150-159. [PMID: 35663108 PMCID: PMC9138439 DOI: 10.36401/jipo-21-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/02/2021] [Accepted: 06/16/2021] [Indexed: 05/06/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has emerged as a revolutionary treatment option for highly aggressive B cell malignancies. Clinical trials of CD19 CAR T cells for the management of relapsed and/or refractory non-Hodgkin lymphoma (NHL) have shown markedly improved survival and response rates. The goal of this review is to evaluate whether the results from these clinical trials are reflective of real-world practices through the analysis of published literature of the commercially available CAR T cell products. We have found that despite the significantly different patient characteristics, the adverse events and response rates of real-world patients were similar to those of the clinical trials. Of interest, several groups excluded from the clinical trials, such as patients with HIV infection, chronic viral hepatitis, and secondary CNS (central nervous system) lymphoma, had case reports of promising outcomes.
Collapse
Affiliation(s)
- Kevin Tang
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Loretta J. Nastoupil
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
96
|
Abstract
Chimeric antigen receptor-engineer (CAR) T-cell therapy is a promising novel immunotherapy that has the potential to revolutionize cancer treatment. With four CAR T-cell therapies receiving FDA approval within the last 5 years, the role of CAR T-cells is anticipated to continue to evolve and expand. However, various aspects of CAR T-cell therapies remain poorly understood, and the therapies are associated with severe side effects [including cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity (ICANS)] that require prompt diagnosis and intervention. In this review, we discuss the role of imaging in diagnosing and monitoring toxicities from CAR T-cell therapies and explore the application of various imaging techniques, including use of PET/CT with novel radiotracers, to predict and assess treatment response and adverse effects. It is important for radiologists to recognize the imaging findings associated with each syndrome, as well as the typical and atypical treatment response patterns associated with CAR T-cell therapy. Given the expected increase in use of CAR T-cells in the near future, radiologists should familiarize themselves with the imaging findings encountered in these novel therapies, to provide comprehensive and up-to-date guidance for clinical management.
Collapse
|
97
|
Shalabi H, Gust J, Taraseviciute A, Wolters PL, Leahy AB, Sandi C, Laetsch TW, Wiener L, Gardner RA, Nussenblatt V, Hill JA, Curran KJ, Olson TS, Annesley C, Wang HW, Khan J, Pasquini MC, Duncan CN, Grupp SA, Pulsipher MA, Shah NN. Beyond the storm - subacute toxicities and late effects in children receiving CAR T cells. Nat Rev Clin Oncol 2021; 18:363-378. [PMID: 33495553 PMCID: PMC8335746 DOI: 10.1038/s41571-020-00456-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2020] [Indexed: 12/15/2022]
Abstract
As clinical advances with chimeric antigen receptor (CAR) T cells are increasingly described and the potential for extending their therapeutic benefit grows, optimizing the implementation of this therapeutic modality is imperative. The recognition and management of cytokine release syndrome (CRS) marked a milestone in this field; however, beyond the understanding gained in treating CRS, a host of additional toxicities and/or potential late effects of CAR T cell therapy warrant further investigation. A multicentre initiative involving experts in paediatric cell therapy, supportive care and/or study of late effects from cancer and haematopoietic stem cell transplantation was convened to facilitate the comprehensive study of extended CAR T cell-mediated toxicities and establish a framework for new systematic investigations of CAR T cell-related adverse events. Together, this group identified six key focus areas: extended monitoring of neurotoxicity and neurocognitive function, psychosocial considerations, infection and immune reconstitution, other end organ toxicities, evaluation of subsequent neoplasms, and strategies to optimize remission durability. Herein, we present the current understanding, gaps in knowledge and future directions of research addressing these CAR T cell-related outcomes. This systematic framework to study extended toxicities and optimization strategies will facilitate the translation of acquired experience and knowledge for optimal application of CAR T cell therapies.
Collapse
Affiliation(s)
- Haneen Shalabi
- Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Juliane Gust
- Seattle Children's Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington Seattle, Seattle, WA, USA
| | - Agne Taraseviciute
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Pamela L Wolters
- Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Allison B Leahy
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carlos Sandi
- Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
- St. Baldrick's Foundation, Monrovia, CA, USA
| | - Theodore W Laetsch
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pediatrics and Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - Lori Wiener
- Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Rebecca A Gardner
- Seattle Children's Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington Seattle, Seattle, WA, USA
| | - Veronique Nussenblatt
- National Institute of Allergy and Infectious Disease, Clinical Center, NIH, Bethesda, MD, USA
| | - Joshua A Hill
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Kevin J Curran
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Timothy S Olson
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Colleen Annesley
- Seattle Children's Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington Seattle, Seattle, WA, USA
| | - Hao-Wei Wang
- Laboratory of Pathology, NCI, NIH, Bethesda, MD, USA
| | - Javed Khan
- Oncogenomics Section, Genetics Branch, NCI, NIH, Bethesda, MD, USA
| | - Marcelo C Pasquini
- Blood and Marrow Transplant and Cellular Therapy Program, Medical College of Wisconsin, Milwaukee, WI, USA
- Center for International Blood and Marrow Transplant Research, Milwaukee, WI, USA
| | - Christine N Duncan
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Stephan A Grupp
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael A Pulsipher
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA.
| |
Collapse
|
98
|
Immune reconstitution and infectious complications following axicabtagene ciloleucel therapy for large B-cell lymphoma. Blood Adv 2021; 5:143-155. [PMID: 33570626 DOI: 10.1182/bloodadvances.2020002732] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy targeting CD19 has significantly improved outcomes in the treatment of refractory or relapsed large B-cell lymphoma (LBCL). We evaluated the long-term course of hematologic recovery, immune reconstitution, and infectious complications in 41 patients with LBCL treated with axicabtagene ciloleucel (axi-cel) at a single center. Grade 3+ cytopenias occurred in 97.6% of patients within the first 28 days postinfusion, with most resolved by 6 months. Overall, 63.4% of patients received a red blood cell transfusion, 34.1% of patients received a platelet transfusion, 36.6% of patients received IV immunoglobulin, and 51.2% of patients received growth factor (granulocyte colony-stimulating factor) injections beyond the first 28 days postinfusion. Only 40% of patients had recovered detectable CD19+ B cells by 1 year, and 50% of patients had a CD4+ T-cell count <200 cells per μL by 18 months postinfusion. Patients with durable responses to axi-cel had significantly longer durations of B-cell aplasia, and this duration correlated strongly with the recovery of CD4+ T-cell counts. There were significantly more infections within the first 28 days compared with any other period of follow-up, with the majority being mild-moderate in severity. Receipt of corticosteroids was the only factor that predicted risk of infection in a multivariate analysis (hazard ratio, 3.69; 95% confidence interval, 1.18-16.5). Opportunistic infections due to Pneumocystis jirovecii and varicella-zoster virus occurred up to 18 months postinfusion in patients who prematurely discontinued prophylaxis. These results support the use of comprehensive supportive care, including long-term monitoring and antimicrobial prophylaxis, beyond 12 months after axi-cel treatment.
Collapse
|
99
|
Safety of CAR T-cell therapy in kidney transplant recipients. Blood 2021; 137:2558-2562. [PMID: 33512386 DOI: 10.1182/blood.2020008759] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/09/2020] [Indexed: 12/24/2022] Open
|
100
|
Miao L, Zhang Z, Ren Z, Li Y. Reactions Related to CAR-T Cell Therapy. Front Immunol 2021; 12:663201. [PMID: 33995389 PMCID: PMC8113953 DOI: 10.3389/fimmu.2021.663201] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
The application of chimeric antigen receptor (CAR) T-cell therapy as a tumor immunotherapy has received great interest in recent years. This therapeutic approach has been used to treat hematological malignancies solid tumors. However, it is associated with adverse reactions such as, cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), off-target effects, anaphylaxis, infections associated with CAR-T-cell infusion (CTI), tumor lysis syndrome (TLS), B-cell dysplasia, hemophagocytic lymphohistiocytosis (HLH)/macrophage activation syndrome (MAS) and coagulation disorders. These adverse reactions can be life-threatening, and thus they should be identified early and treated effectively. In this paper, we review the adverse reactions associated with CAR-T cells, the mechanisms driving such adverse reactions, and strategies to subvert them. This review will provide important reference data to guide clinical application of CAR-T cell therapy.
Collapse
Affiliation(s)
- Lele Miao
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Zhengchao Zhang
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Zhijian Ren
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Yumin Li
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|