511 results on '"Immunotherapy, Adoptive"'
Search Results
2. Real-world and clinical trial outcomes in large B-cell lymphoma with axicabtagene ciloleucel across race and ethnicity.
- Author
-
Locke FL, Siddiqi T, Jacobson CA, Ghobadi A, Ahmed S, Miklos DB, Perales MA, Munoz J, Fingrut WB, Pennisi M, Gauthier J, Shadman M, Gowda L, Mirza AS, Abid MB, Hong S, Majhail NS, Kharfan-Dabaja MA, Khurana A, Badar T, Lin Y, Bennani NN, Herr MM, Hu ZH, Wang HL, Baer A, Baro E, Miao H, Spooner C, Xu H, and Pasquini MC
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Antigens, CD19 immunology, Antigens, CD19 therapeutic use, Ethnicity, Treatment Outcome, Black or African American, White, Asian, Clinical Trials as Topic, Biological Products therapeutic use, Immunotherapy, Adoptive, Lymphoma, Large B-Cell, Diffuse therapy
- Abstract
Abstract: Axicabtagene ciloleucel (axi-cel) is an autologous anti-CD19 chimeric antigen receptor (CAR) T-cell therapy approved for relapsed/refractory (R/R) large B-cell lymphoma (LBCL). Despite extensive data supporting its use, outcomes stratified by race and ethnicity groups are limited. Here, we report clinical outcomes with axi-cel in patients with R/R LBCL by race and ethnicity in both real-world and clinical trial settings. In the real-world setting, 1290 patients who received axi-cel between 2017 and 2020 were identified from the Center for International Blood and Marrow Transplant Research database; 106 and 169 patients were included from the ZUMA-1 and ZUMA-7 trials, respectively. Overall survival was consistent across race/ethnicity groups. However, non-Hispanic (NH) Black patients had lower overall response rate (OR, 0.37; 95% CI, 0.22-0.63) and lower complete response rate (OR, 0.57; 95% CI, 0.33-0.97) than NH White patients. NH Black patients also had a shorter progression-free survival vs NH White (HR, 1.41; 95% CI, 1.04-1.90) and NH Asian patients (HR, 1.67; 95% CI, 1.08-2.59). NH Asian patients had a longer duration of response than NH White (HR, 0.56; 95% CI, 0.33-0.94) and Hispanic patients (HR, 0.54; 95% CI, 0.30-0.97). There was no difference in cytokine release syndrome by race/ethnicity; however, higher rates of any-grade immune effector cell-associated neurotoxicity syndrome were observed in NH White patients than in other patients. These results provide important context when treating patients with R/R LBCL with CAR T-cell therapy across different racial and ethnic groups. ZUMA-1 and ZUMA-7 (ClinicalTrials.gov identifiers: #NCT02348216 and #NCT03391466, respectively) are registered on ClinicalTrials.gov., (© 2024 American Society of Hematology. Published by Elsevier Inc. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. Putting together the pieces: CAR into CD3ζ locus.
- Author
-
Casucci M
- Subjects
- Humans, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen genetics, Immunotherapy, Adoptive, CD3 Complex genetics, CD3 Complex immunology
- Published
- 2024
- Full Text
- View/download PDF
4. Cost-effectiveness of second-line axicabtagene ciloleucel in relapsed refractory diffuse large B-cell lymphoma
- Author
-
Swetha Kambhampati, Monica Saumoy, Yecheskel Schneider, Steve Serrao, Pejman Solaimani, Lihua Elizabeth Budde, Matthew G. Mei, Leslie L. Popplewell, Tanya Siddiqi, Jasmine Zain, Stephen J. Forman, Larry W. Kwak, Steven T. Rosen, Alexey V. Danilov, Alex F. Herrera, and Nikhil R. Thiruvengadam
- Subjects
Adult ,Receptors, Chimeric Antigen ,Cost-Benefit Analysis ,Antigens, CD19 ,Immunology ,Humans ,Lymphoma, Large B-Cell, Diffuse ,Cell Biology ,Hematology ,Immunotherapy, Adoptive ,Biochemistry ,United States ,Aged - Abstract
The ZUMA-7 (Efficacy of Axicabtagene Ciloleucel Compared to Standard of Care Therapy in Subjects With Relapsed/Refractory Diffuse Large B Cell Lymphoma) study showed that axicabtagene ciloleucel (axi-cel) improved event-free survival (EFS) compared with standard of care (SOC) salvage chemoimmunotherapy followed by autologous stem cell transplant in primary refractory/early relapsed diffuse large B-cell lymphoma (DLBCL); this led to its recent US Food and Drug Administration approval in this setting. We modeled a hypothetical cohort of US adults (mean age, 65 years) with primary refractory/early relapsed DLBCL by developing a Markov model (lifetime horizon) to model the cost-effectiveness of second-line axi-cel compared with SOC using a range of plausible long-term outcomes. EFS and OS were estimated from ZUMA-7. Outcome measures were reported in incremental cost-effectiveness ratios, with a willingness-to-pay (WTP) threshold of $150 000 per quality-adjusted life-year (QALY). Assuming a 5-year EFS of 35% with second-line axi-cel and 10% with SOC, axi-cel was cost-effective at a WTP of $150 000 per QALY ($93 547 per QALY). axi-cel was no longer cost-effective if its 5-year EFS was ≤26.4% or if it cost more than $972 061 at a WTP of $150 000. Second-line axi-cel was the cost-effective strategy in 73% of the 10 000 Monte Carlo iterations at a WTP of $150 000. If the absolute benefit in EFS is maintained over time, second-line axi-cel for aggressive relapsed/refractory DLBCL is cost-effective compared with SOC at a WTP of $150 000 per QALY. However, its cost-effectiveness is highly dependent on long-term outcomes. Routine use of second-line chimeric antigen receptor T-cell therapy would add significantly to health care expenditures in the United States (more than $1 billion each year), even when used in a high-risk subpopulation. Further reductions in the cost of chimeric antigen receptor T-cell therapy are needed to be affordable in many regions of the world.
- Published
- 2022
- Full Text
- View/download PDF
5. CAR T-cell therapy in highly aggressive B-cell lymphoma: emerging biological and clinical insights
- Author
-
Alaa Ali, Andre Goy, and Kieron Dunleavy
- Subjects
Receptors, Chimeric Antigen ,Antigens, CD19 ,Immunology ,Receptors, Antigen, T-Cell ,Humans ,Lymphoma, Large B-Cell, Diffuse ,Cell Biology ,Hematology ,Immunotherapy, Adoptive ,Biochemistry - Abstract
Recently, significant progress has been made in identifying novel therapies, beyond conventional immunochemotherapy strategies, with efficacy in B-cell lymphomas. One such approach involves targeting the CD19 antigen on B cells with autologous-derived chimeric antigen receptor (CAR) cells. This strategy is highly effective in patients with relapsed/refractory diffuse large B-cell lymphoma (DLBCL), as evidenced by recent regulatory approvals. Recent reports suggest that this is an effective strategy for high-grade B-cell lymphoma. The biological underpinnings of these entities and how they overlap with each other and DLBCL continue to be areas of intense investigation. Therefore, as more experience with CAR T-cell approaches is examined, it is interesting to consider how both tumor cell–specific and microenvironmental factors that define these highly aggressive subsets influence susceptibility to this approach.
- Published
- 2022
- Full Text
- View/download PDF
6. A TCR mimic CAR T cell specific for NDC80 is broadly reactive with solid tumors and hematologic malignancies
- Author
-
Martin G. Klatt, Tao Dao, Zhiyuan Yang, Jianying Liu, Sung Soo Mun, Megan M. Dacek, Hanzhi Luo, Thomas J. Gardner, Christopher Bourne, Leila Peraro, Zita E. H. Aretz, Tanya Korontsvit, Michael Lau, Michael G. Kharas, Cheng Liu, and David A. Scheinberg
- Subjects
HLA-A Antigens ,T-Lymphocytes ,Immunology ,Receptors, Antigen, T-Cell ,Cell Biology ,Hematology ,Immunotherapy, Adoptive ,Biochemistry ,Antibodies ,Cytoskeletal Proteins ,Mice ,Hematologic Neoplasms ,Neoplasms ,Animals ,Humans - Abstract
Target identification for chimeric antigen receptor (CAR) T-cell therapies remains challenging due to the limited repertoire of tumor-specific surface proteins. Intracellular proteins presented in the context of cell surface HLA provide a wide pool of potential antigens targetable through T-cell receptor mimic antibodies. Mass spectrometry (MS) of HLA ligands from 8 hematologic and nonhematologic cancer cell lines identified a shared, non-immunogenic, HLA-A*02–restricted ligand (ALNEQIARL) derived from the kinetochore-associated NDC80 gene. CAR T cells directed against the ALNEQIARL:HLA-A*02 complex exhibited high sensitivity and specificity for recognition and killing of multiple cancer types, especially those of hematologic origin, and were efficacious in mouse models against a human leukemia and a solid tumor. In contrast, no toxicities toward resting or activated healthy leukocytes as well as hematopoietic stem cells were observed. This shows how MS can inform the design of broadly reactive therapeutic T-cell receptor mimic CAR T-cell therapies that can target multiple cancer types currently not druggable by small molecules, conventional CAR T cells, T cells, or antibodies.
- Published
- 2022
- Full Text
- View/download PDF
7. CD19/22 CAR T cells in children and young adults with B-ALL: phase 1 results and development of a novel bicistronic CAR
- Author
-
Haneen Shalabi, Haiying Qin, Angela Su, Bonnie Yates, Pamela L. Wolters, Seth M. Steinberg, John A. Ligon, Sara Silbert, Kniya DéDé, Mehdi Benzaoui, Sophia Goldberg, Sooraj Achar, Dina Schneider, Shilpa A. Shahani, Lauren Little, Toni Foley, John C. Molina, Sandhya Panch, Crystal L. Mackall, Daniel W. Lee, Christopher D. Chien, Marie Pouzolles, Mark Ahlman, Constance M. Yuan, Hao-Wei Wang, Yanyu Wang, Jon Inglefield, Mary Anne Toledo-Tamula, Staci Martin, Steven L. Highfill, Gregoire Altan-Bonnet, David Stroncek, Terry J. Fry, Naomi Taylor, and Nirali N. Shah
- Subjects
Lymphoma, B-Cell ,Receptors, Chimeric Antigen ,T-Lymphocytes ,Antigens, CD19 ,Immunology ,Receptors, Antigen, T-Cell ,Cell Biology ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Burkitt Lymphoma ,Immunotherapy, Adoptive ,Biochemistry ,Mice ,Recurrence ,Animals ,Cytokines ,Humans ,Cytokine Release Syndrome - Abstract
Remission durability following single-antigen targeted chimeric antigen receptor (CAR) T-cells is limited by antigen modulation, which may be overcome with combinatorial targeting. Building upon our experiences targeting CD19 and CD22 in B-cell acute lymphoblastic leukemia (B-ALL), we report on our phase 1 dose-escalation study of a novel murine stem cell virus (MSCV)-CD19/CD22-4-1BB bivalent CAR T-cell (CD19.22.BBζ) for children and young adults (CAYA) with B-cell malignancies. Primary objectives included toxicity and dose finding. Secondary objectives included response rates and relapse-free survival (RFS). Biologic correlatives included laboratory investigations, CAR T-cell expansion and cytokine profiling. Twenty patients, ages 5.4 to 34.6 years, with B-ALL received CD19.22.BBζ. The complete response (CR) rate was 60% (12 of 20) in the full cohort and 71.4% (10 of 14) in CAR-naïve patients. Ten (50%) developed cytokine release syndrome (CRS), with 3 (15%) having ≥ grade 3 CRS and only 1 experiencing neurotoxicity (grade 3). The 6- and 12-month RFS in those achieving CR was 80.8% (95% confidence interval [CI]: 42.4%-94.9%) and 57.7% (95% CI: 22.1%-81.9%), respectively. Limited CAR T-cell expansion and persistence of MSCV-CD19.22.BBζ compared with EF1α-CD22.BBζ prompted laboratory investigations comparing EF1α vs MSCV promoters, which did not reveal major differences. Limited CD22 targeting with CD19.22.BBζ, as evaluated by ex vivo cytokine secretion and leukemia eradication in humanized mice, led to development of a novel bicistronic CD19.28ζ/CD22.BBζ construct with enhanced cytokine production against CD22. With demonstrated safety and efficacy of CD19.22.BBζ in a heavily pretreated CAYA B-ALL cohort, further optimization of combinatorial antigen targeting serves to overcome identified limitations (www.clinicaltrials.gov #NCT03448393).
- Published
- 2022
- Full Text
- View/download PDF
8. Does BTKi improve CAR T-cell therapy in MCL?
- Author
-
Boardman AP and Palomba ML
- Subjects
- Adult, Humans, Immunotherapy, Adoptive, Piperidines, T-Lymphocytes, Lymphoma, Mantle-Cell, Adenine analogs & derivatives
- Published
- 2024
- Full Text
- View/download PDF
9. A dual-receptor T-cell platform with Ab-TCR and costimulatory receptor achieves specificity and potency against AML.
- Author
-
Dao T, Xiong G, Mun SS, Meyerberg J, Korontsvit T, Xiang J, Cui Z, Chang AY, Jarvis C, Cai W, Luo H, Pierson A, Daniyan A, Yoo S, Takao S, Kharas M, Kentsis A, Liu C, and Scheinberg DA
- Subjects
- Humans, T-Lymphocytes, Receptors, Antigen, T-Cell, Immunotherapy, Adoptive, Leukemia, Myeloid, Acute pathology, Single-Chain Antibodies
- Abstract
Abstract: Chimeric antigen receptor T-cell (CAR T) therapy has produced remarkable clinical responses in B-cell neoplasms. However, many challenges limit this class of agents for the treatment of other cancer types, in particular the lack of tumor-selective antigens for solid tumors and other hematological malignancies, such as acute myeloid leukemia (AML), which may be addressed without significant risk of severe toxicities while providing sufficient abundance for efficient tumor suppression. One approach to overcome this hurdle is dual targeting by an antibody-T-cell receptor (AbTCR) and a chimeric costimulatory signaling receptor (CSR) to 2 different antigens, in which both antigens are found together on the cancer cells but not together on normal cells. To explore this proof of concept in AML, we engineered a new T-cell format targeting Wilms tumor 1 protein (WT1) and CD33; both are highly expressed on most AML cells. Using an AbTCR comprising a newly developed TCR-mimic monoclonal antibody against the WT1 RMFPNAPYL (RMF) epitope/HLA-A2 complex, ESK2, and a secondary CSR comprising a single-chain variable fragment directed to CD33 linked to a truncated CD28 costimulatory fragment, this unique platform confers specific T-cell cytotoxicity to the AML cells while sparing healthy hematopoietic cells, including CD33+ myelomonocytic normal cells. These data suggest that this new platform, named AbTCR-CSR, through the combination of a AbTCR CAR and CSR could be an effective strategy to reduce toxicity and improve specificity and clinical outcomes in adoptive T-cell therapy in AML., (© 2024 American Society of Hematology. Published by Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
10. CAR T-cell therapy in multiple myeloma: mission accomplished?
- Author
-
Rasche L, Hudecek M, and Einsele H
- Subjects
- Humans, Immunotherapy, Adoptive, B-Cell Maturation Antigen metabolism, Antilymphocyte Serum, Multiple Myeloma therapy, Receptors, Chimeric Antigen
- Abstract
Abstract: B-cell maturation antigen (BCMA) chimeric antigen receptor (CAR) T cells are the most potent treatment against multiple myeloma (MM). Here, we review the increasing body of clinical and correlative preclinical data that support their inclusion into firstline therapy and sequencing before T-cell-engaging antibodies. The ambition to cure MM with (BCMA-)CAR T cells is informed by genomic and phenotypic analysis that assess BCMA expression for patient stratification and monitoring, steadily improving early diagnosis and management of side effects, and advances in rapid, scalable CAR T-cell manufacturing to improve access., (© 2024 American Society of Hematology. Published by Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
11. Exploiting the CD200-CD200R immune checkpoint axis in multiple myeloma to enhance CAR T-cell therapy.
- Author
-
Tang Y, Liu W, Kadu S, Johnson O, Hasanali ZS, Kelly A, Shestov A, Pajarillo R, Greenblatt E, Holmes M, Wang LP, Shih N, O'Connor RS, Ruella M, Garfall AL, Allman D, Vogl DT, Cohen A, June CH, and Sheppard NC
- Subjects
- Humans, Mice, Animals, CD28 Antigens metabolism, T-Lymphocytes, B-Cell Maturation Antigen metabolism, Neoplasm Recurrence, Local metabolism, Immunotherapy, Adoptive, Multiple Myeloma metabolism
- Abstract
Abstract: Patients with multiple myeloma (MM) treated with B-cell maturation antigen (BCMA)-specific chimeric antigen receptor (CAR) T cells usually relapse with BCMA+ disease, indicative of CAR T-cell suppression. CD200 is an immune checkpoint that is overexpressed on aberrant plasma cells (aPCs) in MM and is an independent negative prognostic factor for survival. However, CD200 is not present on MM cell lines, a potential limitation of current preclinical models. We engineered MM cell lines to express CD200 at levels equivalent to those found on aPCs in MM and show that these are sufficient to suppress clinical-stage CAR T-cells targeting BCMA or the Tn glycoform of mucin 1 (TnMUC1), costimulated by 4-1BB and CD2, respectively. To prevent CD200-mediated suppression of CAR T cells, we compared CRISPR-Cas9-mediated knockout of the CD200 receptor (CD200RKO), to coexpression of versions of the CD200 receptor that were nonsignaling, that is, dominant negative (CD200RDN), or that leveraged the CD200 signal to provide CD28 costimulation (CD200R-CD28 switch). We found that the CD200R-CD28 switch potently enhanced the polyfunctionality of CAR T cells, and improved cytotoxicity, proliferative capacity, CAR T-cell metabolism, and performance in a chronic antigen exposure assay. CD200RDN provided modest benefits, but surprisingly, the CD200RKO was detrimental to CAR T-cell activity, adversely affecting CAR T-cell metabolism. These patterns held up in murine xenograft models of plasmacytoma, and disseminated bone marrow predominant disease. Our findings underscore the importance of CD200-mediated immune suppression in CAR T-cell therapy of MM, and highlight a promising approach to enhance such therapies by leveraging CD200 expression on aPCs to provide costimulation via a CD200R-CD28 switch., (© 2024 American Society of Hematology. Published by Elsevier Inc. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
12. Is immune escape in the rearview mirror?
- Author
-
Naik S and Gottschalk S
- Subjects
- Immunotherapy, Adoptive, T-Lymphocytes
- Published
- 2024
- Full Text
- View/download PDF
13. CD19/CD22 targeting with cotransduced CAR T cells to prevent antigen-negative relapse after CAR T-cell therapy for B-cell ALL.
- Author
-
Ghorashian S, Lucchini G, Richardson R, Nguyen K, Terris C, Guvenel A, Oporto-Espuelas M, Yeung J, Pinner D, Chu J, Williams L, Ko KY, Walding C, Watts K, Inglott S, Thomas R, Connor C, Adams S, Gravett E, Gilmour K, Lal A, Kunaseelan S, Popova B, Lopes A, Ngai Y, Hackshaw A, Kokalaki E, Carulla MB, Mullanfiroze K, Lazareva A, Pavasovic V, Rao A, Bartram J, Vora A, Chiesa R, Silva J, Rao K, Bonney D, Wynn R, Pule M, Hough R, and Amrolia PJ
- Subjects
- Humans, Child, Immunotherapy, Adoptive, Recurrence, Antigens, CD19, T-Lymphocytes, Sialic Acid Binding Ig-like Lectin 2, Receptors, Chimeric Antigen genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma
- Abstract
Abstract: CD19-negative relapse is a leading cause of treatment failure after chimeric antigen receptor (CAR) T-cell therapy for acute lymphoblastic leukemia. We investigated a CAR T-cell product targeting CD19 and CD22 generated by lentiviral cotransduction with vectors encoding our previously described fast-off rate CD19 CAR (AUTO1) combined with a novel CD22 CAR capable of effective signaling at low antigen density. Twelve patients with advanced B-cell acute lymphoblastic leukemia were treated (CARPALL [Immunotherapy with CD19/22 CAR Redirected T Cells for High Risk/Relapsed Paediatric CD19+ and/or CD22+ Acute Lymphoblastic Leukaemia] study, NCT02443831), a third of whom had failed prior licensed CAR therapy. Toxicity was similar to that of AUTO1 alone, with no cases of severe cytokine release syndrome. Of 12 patients, 10 (83%) achieved a measurable residual disease (MRD)-negative complete remission at 2 months after infusion. Of 10 responding patients, 5 had emergence of MRD (n = 2) or relapse (n = 3) with CD19- and CD22-expressing disease associated with loss of CAR T-cell persistence. With a median follow-up of 8.7 months, there were no cases of relapse due to antigen-negative escape. Overall survival was 75% (95% confidence interval [CI], 41%-91%) at 6 and 12 months. The 6- and 12-month event-free survival rates were 75% (95% CI, 41%-91%) and 60% (95% CI, 23%-84%), respectively. These data suggest dual targeting with cotransduction may prevent antigen-negative relapse after CAR T-cell therapy., (© 2024 American Society of Hematology. Published by Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
14. Impact of CD19 CAR T-cell product type on outcomes in relapsed or refractory aggressive B-NHL
- Author
-
Jordan Gauthier, Nicolas Gazeau, Alexandre V. Hirayama, Joshua A. Hill, Vicky Wu, Aisling Cearley, Paula Perkins, Angela Kirk, Mazyar Shadman, Victor A. Chow, Ajay K. Gopal, Alexandria Hodges Dwinal, Staci Williamson, Jessie Myers, Andy Chen, Sarah Nagle, Brandon Hayes-Lattin, Levanto Schachter, David G. Maloney, Cameron J. Turtle, Mohamed L. Sorror, and Richard T. Maziarz
- Subjects
Lymphoma, B-Cell ,Receptors, Chimeric Antigen ,Clinical Trials, Phase I as Topic ,T-Lymphocytes ,Antigens, CD19 ,Immunology ,Cell Biology ,Hematology ,Immunotherapy, Adoptive ,Biochemistry ,Clinical Trials, Phase II as Topic ,Humans ,Blood Commentary ,Cytokine Release Syndrome ,Retrospective Studies - Abstract
CD19-targeted chimeric antigen receptor-engineered (CD19 CAR) T cells are novel therapies showing great promise for patients with relapsed or refractory (R/R) aggressive B-cell non-Hodgkin lymphoma (B-NHL). Single-arm studies showed significant variations in outcomes across distinct CD19 CAR T-cell products. To estimate the independent impact of the CAR T-cell product type on outcomes, we retrospectively analyzed data from 129 patients with R/R aggressive B-NHL treated with cyclophosphamide and fludarabine lymphodepletion followed by either a commercially available CD19 CAR T-cell therapy (axicabtagene ciloleucel [axicel] or tisagenlecleucel [tisacel]), or the investigational product JCAR014 on a phase 1/2 clinical trial (NCT01865617). After adjustment for age, hematopoietic cell transplantation-specific comorbidity index, lactate dehydrogenase (LDH), largest lesion diameter, and absolute lymphocyte count (ALC), CAR T-cell product type remained associated with outcomes in multivariable models. JCAR014 was independently associated with lower cytokine release syndrome (CRS) severity compared with axicel (adjusted odds ratio [aOR], 0.19; 95% confidence interval [CI]; 0.08-0.46), with a trend toward lower CRS severity with tisacel compared with axicel (aOR, 0.47; 95% CI, 0.21-1.06; P = .07). Tisacel (aOR, 0.17; 95% CI, 0.06-0.48) and JCAR014 (aOR, 0.17; 95% CI, 0.06-0.47) were both associated with lower immune effector cell-associated neurotoxicity syndrome severity compared with axicel. Lower odds of complete response (CR) were predicted with tisacel and JCAR014 compared with axicel. Although sensitivity analyses using either positron emission tomography- or computed tomography-based response criteria also suggested higher efficacy of axicel over JCAR014, the impact of tisacel vs axicel became undetermined. Higher preleukapheresis LDH, largest lesion diameter, and lower ALC were independently associated with lower odds of CR. We conclude that CD19 CAR T-cell product type independently impacts toxicity and efficacy in R/R aggressive B-NHL patients.
- Published
- 2022
- Full Text
- View/download PDF
15. Targeting cancer-associated fibroblasts in the bone marrow prevents resistance to CART-cell therapy in multiple myeloma
- Author
-
Reona Sakemura, Mehrdad Hefazi, Elizabeth L. Siegler, Michelle J. Cox, Daniel P. Larson, Michael J. Hansen, Claudia Manriquez Roman, Kendall J. Schick, Ismail Can, Erin E. Tapper, Paulina Horvei, Mohamad M. Adada, Evandro D. Bezerra, Lionel Aurelien Kankeu Fonkoua, Michael W. Ruff, Wendy K. Nevala, Denise K. Walters, Sameer A. Parikh, Yi Lin, Diane F. Jelinek, Neil E. Kay, P. Leif Bergsagel, and Saad S. Kenderian
- Subjects
Immunology ,Cell- and Tissue-Based Therapy ,virus diseases ,Cell Biology ,Hematology ,Fibroblasts ,Immunotherapy, Adoptive ,Biochemistry ,Cancer-Associated Fibroblasts ,Bone Marrow ,immune system diseases ,mental disorders ,Tumor Microenvironment ,Humans ,Multiple Myeloma - Abstract
Pivotal clinical trials of B-cell maturation antigen-targeted chimeric antigen receptor T (CART)-cell therapy in patients with relapsed/refractory multiple myeloma (MM) resulted in remarkable initial responses, which led to a recent US Food and Drug Administration approval. Despite the success of this therapy, durable remissions continue to be low, and the predominant mechanism of resistance is loss of CART cells and inhibition by the tumor microenvironment (TME). MM is characterized by an immunosuppressive TME with an abundance of cancer-associated fibroblasts (CAFs). Using MM models, we studied the impact of CAFs on CART-cell efficacy and developed strategies to overcome CART-cell inhibition. We showed that CAFs inhibit CART-cell antitumor activity and promote MM progression. CAFs express molecules such as fibroblast activation protein and signaling lymphocyte activation molecule family-7, which are attractive immunotherapy targets. To overcome CAF-induced CART-cell inhibition, CART cells were generated targeting both MM cells and CAFs. This dual-targeting CART-cell strategy significantly improved the effector functions of CART cells. We show for the first time that dual targeting of both malignant plasma cells and the CAFs within the TME is a novel strategy to overcome resistance to CART-cell therapy in MM.
- Published
- 2022
- Full Text
- View/download PDF
16. Efficacy and safety of CD19-specific CAR T cell–based therapy in B-cell acute lymphoblastic leukemia patients with CNSL
- Author
-
Yuekun Qi, Mingfeng Zhao, Yongxian Hu, Ying Wang, Ping Li, Jiang Cao, Ming Shi, Jiaqi Tan, Meng Zhang, Xia Xiao, Jieyun Xia, Sha Ma, Jianlin Qiao, Zhiling Yan, Hujun Li, Bin Pan, Wei Sang, Depeng Li, Zhenyu Li, Jianfeng Zhou, He Huang, Aibin Liang, Junnian Zheng, and Kailin Xu
- Subjects
Receptors, Chimeric Antigen ,T-Lymphocytes ,Antigens, CD19 ,Immunology ,Cell Biology ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Burkitt Lymphoma ,Immunotherapy, Adoptive ,Biochemistry ,Central Nervous System Neoplasms ,Acute Disease ,Humans ,Cytokine Release Syndrome - Abstract
Few studies have described chimeric antigen receptor (CAR) T-cell therapy for patients with B-cell acute lymphoblastic leukemia (B-ALL) with central nervous system leukemia (CNSL) because of concerns regarding poor response and treatment-related neurotoxicity. Our study included 48 patients with relapsed/refractory B-ALL with CNSL to evaluate the efficacy and safety of CD19-specific CAR T cell–based therapy. The infusion resulted in an overall response rate of 87.5% (95% confidence interval [CI], 75.3-94.1) in bone marrow (BM) disease and remission rate of 85.4% (95% CI, 72.8-92.8) in CNSL. With a median follow-up of 11.5 months (range, 1.3-33.3), the median event-free survival was 8.7 months (95% CI, 3.7-18.8), and the median overall survival was 16.0 months (95% CI, 13.5-20.1). The cumulative incidences of relapse in BM and CNS diseases were 31.1% and 11.3%, respectively, at 12 months (P = .040). The treatment was generally well tolerated, with 9 patients (18.8%) experiencing grade ≥3 cytokine release syndrome. Grade 3 to 4 neurotoxic events, which developed in 11 patients (22.9%), were associated with a higher preinfusion disease burden in CNS and were effectively controlled under intensive management. Our results suggest that CD19-specific CAR T cell–based therapy can induce similar high response rates in both BM and CNS diseases. The duration of remission in CNSL was longer than that in BM disease. CD19 CAR T-cell therapy may provide a potential treatment option for previously excluded patients with CNSL, with manageable neurotoxicity. The clinical trials were registered at www.clinicaltrials.gov as #NCT02782351 and www.chictr.org.cn as #ChiCTR-OPN-16008526.
- Published
- 2022
- Full Text
- View/download PDF
17. Impact of high-risk cytogenetics on outcomes for children and young adults receiving CD19-directed CAR T-cell therapy
- Author
-
Allison Barz Leahy, Kaitlin J. Devine, Yimei Li, Hongyan Liu, Regina Myers, Amanda DiNofia, Lisa Wray, Susan R. Rheingold, Colleen Callahan, Diane Baniewicz, Maria Patino, Haley Newman, Stephen P. Hunger, Stephan A. Grupp, David M. Barrett, and Shannon L. Maude
- Subjects
Antigens, CD19 ,Immunology ,Receptors, Antigen, T-Cell ,Infant ,Cell Biology ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Immunotherapy, Adoptive ,Biochemistry ,Young Adult ,Recurrence ,Cytogenetic Analysis ,Humans ,Philadelphia Chromosome ,Child - Abstract
Chimeric antigen receptor (CAR) T-cell therapy can induce durable remissions of relapsed/refractory B-acute lymphoblastic leukemia (ALL). However, case reports suggested differential outcomes mediated by leukemia cytogenetics. We identified children and young adults with relapsed/refractory CD19+ ALL/lymphoblastic lymphoma treated on 5 CD19-directed CAR T-cell (CTL019 or humanized CART19) clinical trials or with commercial tisagenlecleucel from April 2012 to April 2019. Patients were hierarchically categorized according to leukemia cytogenetics: High-risk lesions were defined as KMT2A (MLL) rearrangements, Philadelphia chromosome (Ph+), Ph-like, hypodiploidy, or TCF3/HLF; favorable as hyperdiploidy or ETV6/RUNX1; and intermediate as iAMP21, IKZF1 deletion, or TCF3/PBX1. Of 231 patients aged 1 to 29, 74 (32%) were categorized as high risk, 28 (12%) as intermediate, 43 (19%) as favorable, and 86 (37%) as uninformative. Overall complete remission rate was 94%, with no difference between strata. There was no difference in relapse-free survival (RFS; P = .8112), with 2-year RFS for the high-risk group of 63% (95% confidence interval [CI], 52-77). There was similarly no difference seen in overall survival (OS) (P = .5488), with 2-year OS for the high-risk group of 70% (95% CI, 60-82). For patients with KMT2A-rearranged infant ALL (n = 13), 2-year RFS was 67% (95% CI, 45-99), and OS was 62% (95% CI, 40-95), with multivariable analysis demonstrating no increased risk of relapse (hazard ratio, 0.70; 95% CI, 0.21-2.90; P = .7040) but a higher proportion of relapses associated with myeloid lineage switch and a 3.6-fold increased risk of all-cause death (95% CI, 1.04-12.75; P = .0434). CTL019/huCART19/tisagenlecleucel are effective at achieving durable remissions across cytogenetic categories. Relapsed/refractory patients with high-risk cytogenetics, including KMT2A-rearranged infant ALL, demonstrated high RFS and OS probabilities at 2 years.
- Published
- 2022
- Full Text
- View/download PDF
18. Genetic ablation of PRDM1 in antitumor T cells enhances therapeutic efficacy of adoptive immunotherapy
- Author
-
Zhiwen Wu, Toshiaki Yoshikawa, Yusuke Takahashi, Satoshi Inoue, Hirokazu Matsushita, Hitomi Kasuya, Hiroaki Kuroda, Yuki Kagoya, Shiro Suzuki, and Waki Hosoda
- Subjects
medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,Biology ,Lymphocyte Activation ,Immunotherapy, Adoptive ,Biochemistry ,Chimeric antigen receptor ,Gene Knockout Techniques ,Cytokine ,Antigen ,Cancer immunotherapy ,Neoplasms ,PRDM1 ,Cancer research ,medicine ,Cytokines ,Humans ,CRISPR ,Cytokine secretion ,Positive Regulatory Domain I-Binding Factor 1 ,Epigenetics - Abstract
Adoptive cancer immunotherapy can induce objective clinical efficacy in patients with advanced cancer; however, a sustained response is achieved in a minority of cases. The persistence of infused T cells is an essential determinant of a durable therapeutic response. Antitumor T cells undergo a genome-wide remodeling of the epigenetic architecture upon repeated antigen encounters, which inevitably induces progressive T-cell differentiation and the loss of longevity. In this study, we identified PR domain zinc finger protein 1 (PRDM1) ie, Blimp-1, as a key epigenetic gene associated with terminal T-cell differentiation. The genetic knockout of PRDM1 by clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) supported the maintenance of an early memory phenotype and polyfunctional cytokine secretion in repeatedly stimulated chimeric antigen receptor (CAR)-engineered T cells. PRDM1 disruption promoted the expansion of less differentiated memory CAR-T cells in vivo, which enhanced T-cell persistence and improved therapeutic efficacy in multiple tumor models. Mechanistically, PRDM1-ablated T cells displayed enhanced chromatin accessibility of the genes that regulate memory formation, thereby leading to the acquisition of gene expression profiles representative of early memory T cells. PRDM1 knockout also facilitated maintaining an early memory phenotype and cytokine polyfunctionality in T-cell receptor-engineered T cells as well as tumor-infiltrating lymphocytes. In other words, targeting PRDM1 enabled the generation of superior antitumor T cells, which is potentially applicable to a wide range of adoptive cancer immunotherapies.
- Published
- 2022
- Full Text
- View/download PDF
19. Systemic IL-15 promotes allogeneic cell rejection in patients treated with natural killer cell adoptive therapy
- Author
-
Ethan McClain, Sweta Desai, Amanda F. Cashen, Mark P. Foster, Peter Westervelt, Patrick Soon-Shiong, Michelle Becker-Hapak, Keith Stockerl-Goldstein, Jeffrey S. Miller, Miriam T. Jacobs, Mark A. Schroeder, Pamela Wong, Camille N. Abboud, Patrick Pence, Geoffrey L. Uy, Feng Gao, Claudio G. Brunstein, Melissa M. Berrien-Elliott, Meagan A. Jacoby, Iskra Pusic, Sarah Cooley, John F. DiPersio, and Todd A. Fehniger
- Subjects
Male ,Adoptive cell transfer ,Recombinant Fusion Proteins ,T cell ,Lymphocyte ,Immunology ,Antineoplastic Agents ,CD8-Positive T-Lymphocytes ,Immunotherapy, Adoptive ,Biochemistry ,Natural killer cell ,Cell therapy ,Humans ,Medicine ,Cytotoxic T cell ,Interleukin-15 ,business.industry ,Allogeneic Cells ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Hematology ,Killer Cells, Natural ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Interleukin 15 ,Cancer research ,Female ,business ,CD8 - Abstract
Natural killer (NK) cells are a promising alternative to T cells for cancer immunotherapy. Adoptive therapies with allogeneic, cytokine-activated NK cells are being investigated in clinical trials. However, the optimal cytokine support after adoptive transfer to promote NK cell expansion, and persistence remains unclear. Correlative studies from 2 independent clinical trial cohorts treated with major histocompatibility complex-haploidentical NK cell therapy for relapsed/refractory acute myeloid leukemia revealed that cytokine support by systemic interleukin-15 (IL-15; N-803) resulted in reduced clinical activity, compared with IL-2. We hypothesized that the mechanism responsible was IL-15/N-803 promoting recipient CD8 T-cell activation that in turn accelerated donor NK cell rejection. This idea was supported by increased proliferating CD8+ T-cell numbers in patients treated with IL-15/N-803, compared with IL-2. Moreover, mixed lymphocyte reactions showed that IL-15/N-803 enhanced responder CD8 T-cell activation and proliferation, compared with IL-2 alone. Additionally, IL-15/N-803 accelerated the ability of responding T cells to kill stimulator-derived memory-like NK cells, demonstrating that additional IL-15 can hasten donor NK cell elimination. Thus, systemic IL-15 used to support allogeneic cell therapy may paradoxically limit their therapeutic window of opportunity and clinical activity. This study indicates that stimulating patient CD8 T-cell allo-rejection responses may critically limit allogeneic cellular therapy supported with IL-15. This trial was registered at www.clinicaltrials.gov as #NCT03050216 and #NCT01898793.
- Published
- 2022
- Full Text
- View/download PDF
20. Pembrolizumab for B-cell lymphomas relapsing after or refractory to CD19-directed CAR T-cell therapy
- Author
-
Simon F. Lacey, Stephen J. Schuster, Elise A. Chong, Sunita D. Nasta, Jakub Svoboda, Marco Ruella, Siddharth Bhattacharyya, E. John Wherry, Cécile Alanio, and Daniel J. Landsburg
- Subjects
Adult ,Male ,Oncology ,medicine.medical_specialty ,Lymphoma, B-Cell ,T cell ,Antigens, CD19 ,Immunology ,Pembrolizumab ,Neutropenia ,Antibodies, Monoclonal, Humanized ,Immunotherapy, Adoptive ,Biochemistry ,CD19 ,Antineoplastic Agents, Immunological ,Refractory ,Antigen ,Internal medicine ,medicine ,Humans ,Prospective Studies ,Adverse effect ,B cell ,Aged ,Salvage Therapy ,Receptors, Chimeric Antigen ,biology ,business.industry ,Cell Biology ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,medicine.anatomical_structure ,biology.protein ,Female ,Blood Commentary ,Neoplasm Recurrence, Local ,business ,Follow-Up Studies - Abstract
CD19-directed chimeric antigen receptor–modified (CAR T) T cells achieve durable remissions in about 30% to 40% of relapsed/refractory large B-cell lymphomas. T-cell exhaustion and/or an immunosuppressive tumor microenvironment may contribute to CAR T-cell failure. Pembrolizumab, an anti-PD1 immune checkpoint inhibitor, may reverse T-cell exhaustion after CAR T-cell therapy. We treated 12 patients with B-cell lymphomas who were either refractory to (n = 9) or relapsed after (n = 3) CD19-directed CAR T-cell (4-1BB–costimulated) therapy with pembrolizumab 200 mg IV every 3 weeks. Median time from CAR T-cell infusion to first pembrolizumab dose was 3.3 months (range, 0.4-42.8 months). Pembrolizumab was well tolerated, and the only grade ≥3 adverse events related to pembrolizumab were neutropenia (n = 3; 25%). Best overall response rate after pembrolizumab was 25% (3 of 12 patients; 1 complete response; 2 partial responses). One (8%) patient had stable disease; thus, 4 of 12 (33%) patients had clinical benefit. After pembrolizumab, 4 patients with clinical benefit had an increase in percentage of CAR T cells by mass cytometry by time of flight (CyTOF); 3 of 4 of these patients also had increases in CAR19 transgene levels by quantitative polymerase chain reaction. Deep immune profiling using CyTOF revealed increased CAR T-cell activation and proliferation and less T-cell exhaustion in clinical responders. Together, PD1 blockade with pembrolizumab after CD19-directed CAR T-cell therapy appears safe and may achieve clinical responses in some patients with B-cell lymphomas refractory to or relapsed after CAR T-cell therapy. This trial was registered at www.clinicaltrials.gove as #NCT02650999.
- Published
- 2022
- Full Text
- View/download PDF
21. EBV+ lymphoproliferative diseases: opportunities for leveraging EBV as a therapeutic target
- Author
-
Keri Toner and Catherine M. Bollard
- Subjects
Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,business.industry ,T cell ,Immunology ,Cell Biology ,Hematology ,Immunotherapy, Adoptive ,Biochemistry ,Lymphoproliferative Disorders ,Virus ,Proliferative response ,Immune therapy ,Human tumor ,Virus-Mediated Hematologic Disease ,medicine.anatomical_structure ,Immunity ,hemic and lymphatic diseases ,Humans ,Medicine ,In patient ,Lymphoproliferative disease ,business - Abstract
Epstein-Barr virus (EBV) is a ubiquitous human tumor virus, which contributes to the development of lymphoproliferative disease, most notably in patients with impaired immunity. EBV-associated lymphoproliferation is characterized by expression of latent EBV proteins and ranges in severity from a relatively benign proliferative response to aggressive malignant lymphomas. The presence of EBV can also serve as a unique target for directed therapies for the treatment of EBV lymphoproliferative diseases, including T cell–based immune therapies. In this review, we describe the EBV-associated lymphoproliferative diseases and particularly focus on the therapies that target EBV.
- Published
- 2022
- Full Text
- View/download PDF
22. CAR T-cell therapy in primary central nervous system lymphoma: the clinical experience of the French LOC network
- Author
-
Marion Alcantara, Caroline Houillier, Marie Blonski, Marie-Thérèse Rubio, Lise Willems, Agathe Waultier Rascalou, Magali Le Garff-Tavernier, Karim Maloum, Clotilde Bravetti, Laetitia Souchet, Damien Roos-Weil, Véronique Morel, Madalina Uzunov, Carole Metz, Meriem Dhib-Charfi, Stéphanie Nguyen, Natalia Shor, Dimitri Psimaras, Nicolas Weiss, Nathalie Jacque, Silvia Solorzano, Nicolas Gauthier, Marie Le Cann, Françoise Norol, Carole Soussain, and Sylvain Choquet
- Subjects
Central Nervous System ,Male ,Immunology ,Cell Biology ,Hematology ,Middle Aged ,Immunotherapy, Adoptive ,Survival Analysis ,Biochemistry ,Central Nervous System Neoplasms ,Cohort Studies ,Humans ,Female ,France ,Lymphoma, Large B-Cell, Diffuse ,Letter to Blood ,Aged - Published
- 2022
- Full Text
- View/download PDF
23. Buckling up against COVID-19 after CAR T-cell therapy
- Author
-
Jeffery Auletta
- Subjects
Immunity, Cellular ,T-Lymphocytes ,Antigens, CD19 ,Immunology ,COVID-19 ,Humans ,Cell Biology ,Hematology ,Immunotherapy, Adoptive ,Biochemistry ,BNT162 Vaccine - Published
- 2022
- Full Text
- View/download PDF
24. Investigation of product-derived lymphoma following infusion of piggyBac-modified CD19 chimeric antigen receptor T cells
- Author
-
Brian S. Gloss, Kavitha Gowrishankar, Piers Blombery, Raymond H. Y. Louie, Kenneth P. Micklethwaite, Emily Blyth, Fritz J. Sedlazeck, Janine Street, David Gottlieb, Matthew MacKay, Christopher E. Mason, David Bishop, Leili Moezzi, Gaurav Sutrave, Curtis Cai, Ziduo Li, Leighton Clancy, Fabio Luciani, Jonathan Foox, and Melanie A Mach
- Subjects
Male ,Lymphoma, B-Cell ,Lymphoma ,T-Lymphocytes ,Transgene ,Immunology ,Receptors, Antigen, T-Cell ,Immunotherapy, Adoptive ,Biochemistry ,CD19 ,Viral vector ,Leukemia, B-Cell ,medicine ,Humans ,Transgenes ,Copy-number variation ,Aged ,biology ,business.industry ,Point mutation ,Gene Transfer Techniques ,Cell Biology ,Hematology ,medicine.disease ,Chimeric antigen receptor ,Gene Expression Regulation, Neoplastic ,PiggyBac Transposon System ,DNA Transposable Elements ,Cancer research ,biology.protein ,Transcriptome ,business - Abstract
We performed a phase 1 clinical trial to evaluate outcomes in patients receiving donor-derived CD19-specific chimeric antigen receptor (CAR) T cells for B-cell malignancy that relapsed or persisted after matched related allogeneic hemopoietic stem cell transplant. To overcome the cost and transgene-capacity limitations of traditional viral vectors, CAR T cells were produced using the piggyBac transposon system of genetic modification. Following CAR T-cell infusion, 1 patient developed a gradually enlarging retroperitoneal tumor due to a CAR-expressing CD4+ T-cell lymphoma. Screening of other patients led to the detection, in an asymptomatic patient, of a second CAR T-cell tumor in thoracic para-aortic lymph nodes. Analysis of the first lymphoma showed a high transgene copy number, but no insertion into typical oncogenes. There were also structural changes such as altered genomic copy number and point mutations unrelated to the insertion sites. Transcriptome analysis showed transgene promoter–driven upregulation of transcription of surrounding regions despite insulator sequences surrounding the transgene. However, marked global changes in transcription predominantly correlated with gene copy number rather than insertion sites. In both patients, the CAR T-cell–derived lymphoma progressed and 1 patient died. We describe the first 2 cases of malignant lymphoma derived from CAR gene–modified T cells. Although CAR T cells have an enviable record of safety to date, our results emphasize the need for caution and regular follow-up of CAR T recipients, especially when novel methods of gene transfer are used to create genetically modified immune therapies. This trial was registered at www.anzctr.org.au as ACTRN12617001579381.
- Published
- 2021
- Full Text
- View/download PDF
25. Mitigating inequity: ethically prioritizing patients for CAR T-cell therapy.
- Author
-
Bell JAH, Jeffries GA, and Chen CI
- Subjects
- Humans, Immunotherapy, Adoptive, Patients, Health Priorities, Receptors, Chimeric Antigen
- Abstract
Manufacturing capacity and institutional infrastructure to deliver chimeric antigen receptor T-cell therapies (CAR-T) are pressured to keep pace with the growing number of approved products and expanding eligible patient population for this potentially life-saving therapy. Consequently, many cell therapy programs must make difficult decisions about which patient should get the next available treatment slot. This situation requires an ethical framework to ensure fair and equitable decision-making. In this perspective, we discuss the application of Accountability for Reasonableness (A4R), a priority-setting framework grounded in procedural justice, to the problem of limited CAR-T slots at our institution. We formed a multidisciplinary working group spanning several hematological malignancies. Through multiple rounds of partner engagement, we used A4R guiding principles to identify 4 main criteria to prioritize patients for CAR-T: medical benefit, safety/risk of complications, psychosocial factors, and medical urgency. Associated measures/tools and an implementation process were developed. We discuss further how ethical principles of fairness and equity demand a consistent approach within health systems that does not disadvantage medically underserved or underrepresented populations and supports overcoming barriers to care. In our commitment to transparency and collaboration, we make our tools available to others, ideally to be used to engage in their own A4R process, adapting the tools to their unique environments. Our hope is that our preliminary work will support the advancement of further study in this area globally, aiming for justice in resource allocation for all potential CAR-T candidates, wherever they may seek care., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
26. CAR Traffic jam: who can use the fast lane?
- Author
-
Kuipers MT and Kersten MJ
- Subjects
- Humans, Safety, Immunotherapy, Adoptive
- Published
- 2023
- Full Text
- View/download PDF
27. Chemotherapy-induced reversal of ciltacabtagene autoleucel-associated movement and neurocognitive toxicity.
- Author
-
Graham CE, Lee WH, Wiggin HR, Supper VM, Leick MB, Birocchi F, Yee AJ, Petrichenko A, Everett J, Bushman FD, Sadrzadeh H, Rapalino O, Chiu D, Arrillaga-Romany I, Maus MV, Frigault MJ, and Gallagher KME
- Subjects
- Humans, Cognition, Immunotherapy, Adoptive, Antineoplastic Agents adverse effects, Multiple Myeloma
- Published
- 2023
- Full Text
- View/download PDF
28. BCMA-CAR T-cell treatment-associated parkinsonism.
- Author
-
Gust J
- Subjects
- Humans, B-Cell Maturation Antigen, Immunotherapy, Adoptive, T-Lymphocytes, Receptors, Chimeric Antigen, Parkinsonian Disorders therapy, Antineoplastic Agents
- Published
- 2023
- Full Text
- View/download PDF
29. Lulla PD. CAR T cells and autologous transplantation can coexist for DLBCL. Blood. 2021;139(9):1266-1267
- Subjects
T-Lymphocytes ,Antigens, CD19 ,Humans ,Blood Commentary ,Lymphoma, Large B-Cell, Diffuse ,Transplantation, Autologous ,Immunotherapy, Adoptive - Published
- 2022
30. Failure after CAR T-cell treatment in BCL
- Subjects
T-Lymphocytes ,Antigens, CD19 ,Receptors, Antigen, T-Cell ,Immunotherapy, Adoptive - Published
- 2022
31. Axi-cel in LBCL: fulfill two needs with one deed
- Author
-
Nilanjan Ghosh
- Subjects
Immunology ,Antigens, CD19 ,Humans ,Cell Biology ,Hematology ,Lymphoma, Large B-Cell, Diffuse ,Patient Reported Outcome Measures ,Biochemistry ,Immunotherapy, Adoptive - Published
- 2022
32. Treating CAR-T relapses: check not checkmate
- Author
-
Jay Y. Spiegel and Krishna V. Komanduri
- Subjects
Receptors, Chimeric Antigen ,Recurrence ,Clinical Trials and Observations ,Immunology ,Humans ,Cell Biology ,Hematology ,Immunotherapy, Adoptive ,Biochemistry - Abstract
CD19-directed chimeric antigen receptor–modified (CAR T) T cells achieve durable remissions in about 30% to 40% of relapsed/refractory large B-cell lymphomas. T-cell exhaustion and/or an immunosuppressive tumor microenvironment may contribute to CAR T-cell failure. Pembrolizumab, an anti-PD1 immune checkpoint inhibitor, may reverse T-cell exhaustion after CAR T-cell therapy. We treated 12 patients with B-cell lymphomas who were either refractory to (n = 9) or relapsed after (n = 3) CD19-directed CAR T-cell (4-1BB–costimulated) therapy with pembrolizumab 200 mg IV every 3 weeks. Median time from CAR T-cell infusion to first pembrolizumab dose was 3.3 months (range, 0.4-42.8 months). Pembrolizumab was well tolerated, and the only grade ≥3 adverse events related to pembrolizumab were neutropenia (n = 3; 25%). Best overall response rate after pembrolizumab was 25% (3 of 12 patients; 1 complete response; 2 partial responses). One (8%) patient had stable disease; thus, 4 of 12 (33%) patients had clinical benefit. After pembrolizumab, 4 patients with clinical benefit had an increase in percentage of CAR T cells by mass cytometry by time of flight (CyTOF); 3 of 4 of these patients also had increases in CAR19 transgene levels by quantitative polymerase chain reaction. Deep immune profiling using CyTOF revealed increased CAR T-cell activation and proliferation and less T-cell exhaustion in clinical responders. Together, PD1 blockade with pembrolizumab after CD19-directed CAR T-cell therapy appears safe and may achieve clinical responses in some patients with B-cell lymphomas refractory to or relapsed after CAR T-cell therapy. This trial was registered at www.clinicaltrials.gove as #NCT02650999.
- Published
- 2022
- Full Text
- View/download PDF
33. Proof of concept for a rapidly switchable universal CAR-T platform with UniCAR-T-CD123 in relapsed/refractory AML
- Author
-
Michael Pehl, Sabrina Kraus, Gerhard Ehninger, Marc Cartellieri, Carla Kreissig, Malte von Bonin, Maria-Elisabeth Goebeler, Martin Wermke, Martin Bornhäuser, Ralf C. Bargou, Jan Moritz Middeke, Jan Koedam, Hermann Einsele, and Armin Ehninger
- Subjects
Aged, 80 and over ,Male ,Oncology ,medicine.medical_specialty ,Receptors, Chimeric Antigen ,business.industry ,Immunology ,Cell Biology ,Hematology ,Middle Aged ,Immunotherapy, Adoptive ,Proof of Concept Study ,Biochemistry ,Leukemia, Myeloid, Acute ,Text mining ,Proof of concept ,Internal medicine ,Relapsed refractory ,Humans ,Medicine ,Car t cells ,Letter to Blood ,business ,Aged - Published
- 2021
- Full Text
- View/download PDF
34. Safety of CAR T-cell therapy in kidney transplant recipients
- Author
-
Sreedhar Mandayam, Ranjit Nair, Raphael E Steiner, Kartik Devashish, Sherry Adkins, Swaminathan P. Iyer, Angelina Edwards, Sattva S. Neelapu, Paolo Strati, Misha Hawkins, Neeraj Saini, Omar Mamlouk, and Sairah Ahmed
- Subjects
Adult ,Graft Rejection ,Male ,Oncology ,medicine.medical_specialty ,business.industry ,Immunology ,MEDLINE ,Cell Biology ,Hematology ,Prognosis ,Immunotherapy, Adoptive ,Kidney Transplantation ,Biochemistry ,Kidney transplant ,Transplant Recipients ,Therapeutic immunosuppression ,Internal medicine ,medicine ,Humans ,CAR T-cell therapy ,Lymphoma, Large B-Cell, Diffuse ,Letter to Blood ,business - Published
- 2021
- Full Text
- View/download PDF
35. CD22-directed CAR T-cell therapy induces complete remissions in CD19-directed CAR–refractory large B-cell lymphoma
- Author
-
Warren D. Reynolds, Bita Sahaf, Zachary Ehlinger, Steven A. Feldman, Matthew J. Frank, Andrew R. Rezvani, Surbhi Sidana, Jay Y. Spiegel, Allison P. Jacob, Lori Muffly, Kara L. Davis, Shabnum Patel, Jean Oak, Eric H. Yang, Ilan R. Kirsch, Robert Lowsky, Laura Johnston, Juliana Craig, John S. Tamaresis, Michael G. Ozawa, John H. Baird, Sheren F. Younes, David B. Miklos, Parveen Shiraz, Chelsea D. Mullins, Crystal L. Mackall, Robert S. Negrin, Liora M. Schultz, Wen-Kai Weng, Everett Meyer, Sally Arai, Yasodha Natkunam, and Sneha Ramakrishna
- Subjects
Oncology ,medicine.medical_specialty ,Clinical Trials and Observations ,Sialic Acid Binding Ig-like Lectin 2 ,Antigens, CD19 ,Immunology ,Immunotherapy, Adoptive ,Biochemistry ,CD19 ,Refractory ,Internal medicine ,medicine ,Humans ,Adverse effect ,B-cell lymphoma ,Clinical Trials, Phase I as Topic ,biology ,business.industry ,Remission Induction ,CD22 ,Cell Biology ,Hematology ,Prognosis ,medicine.disease ,Chimeric antigen receptor ,Lymphoma ,biology.protein ,Chimeric Antigen Receptor T-Cell Therapy ,Lymphoma, Large B-Cell, Diffuse ,business - Abstract
The prognosis of patients with large B-cell lymphoma (LBCL) that progresses after treatment with chimeric antigen receptor (CAR) T-cell therapy targeting CD19 (CAR19) is poor. We report on the first 3 consecutive patients with autologous CAR19-refractory LBCL who were treated with a single infusion of autologous 1 × 106 CAR+ T cells per kilogram targeting CD22 (CAR22) as part of a phase 1 dose-escalation study. CAR22 therapy was relatively well tolerated, without any observed nonhematologic adverse events higher than grade 2. After infusion, all 3 patients achieved complete remission, with all responses continuing at the time of last follow-up (mean, 7.8 months; range, 6-9.3). Circulating CAR22 cells demonstrated robust expansion (peak range, 85.4-350 cells per microliter), and persisted beyond 3 months in all patients with continued radiographic responses and corresponding decreases in circulating tumor DNA beyond 6 months after infusion. Further accrual at a higher dose level in this phase 1 dose-escalation study is ongoing and will explore the role of this therapy in patients in whom prior CAR T-cell therapies have failed. This trial is registered on clinicaltrials.gov as #NCT04088890.
- Published
- 2021
- Full Text
- View/download PDF
36. How I treat chronic lymphocytic leukemia after venetoclax
- Author
-
Constantine S. Tam, John F. Seymour, and Thomas E Lew
- Subjects
Oncology ,medicine.medical_specialty ,Chronic lymphocytic leukemia ,Immunology ,Immunotherapy, Adoptive ,Biochemistry ,chemistry.chemical_compound ,Obinutuzumab ,Internal medicine ,medicine ,Humans ,Sulfonamides ,Venetoclax ,business.industry ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Hematology ,Allografts ,Bridged Bicyclo Compounds, Heterocyclic ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Fludarabine ,Transplantation ,Proto-Oncogene Proteins c-bcl-2 ,chemistry ,Drug Resistance, Neoplasm ,Ibrutinib ,Rituximab ,Tumor Suppressor Protein p53 ,business ,Progressive disease ,medicine.drug - Abstract
Venetoclax-based regimens have expanded the therapeutic options for patients with chronic lymphocytic leukemia (CLL), frequently achieving remissions with undetectable measurable residual disease (uMRD) and facilitating time-limited treatment without utilizing chemotherapy. Although response rates are high and durable disease control is common, longer-term follow-up of patients with relapsed and refractory (RR) disease, especially in the presence of TP53 aberrations, demonstrates frequent disease resistance and progression. Although the understanding of venetoclax resistance remains incomplete, progressive disease (PD) is typified by oligoclonal leukemic populations with distinct resistance mechanisms, including BCL2 mutations, upregulation of alternative BCL2 family proteins and genomic instability. Although most commonly observed in heavily pre-treated patients with disease refractory to fludarabine and harboring complex karyotype (CK), Richter transformation (RT) presents a distinct and challenging manifestation of venetoclax resistance. For patients with progressive CLL after venetoclax, treatment options include B-cell receptor pathway inhibitors (BCRis), allogeneic stem cell transplantation (SCT), chimeric antigen receptor (CAR) T-cells, and venetoclax re-treatment for those with disease relapsing after time-limited therapy. However, data to inform clinical decisions for these patients are limited. We review the biology of venetoclax resistance and outline an approach to the common clinical scenarios encountered after venetoclax-based therapy that will increasingly confront practising clinicians.
- Published
- 2021
- Full Text
- View/download PDF
37. Interventions and outcomes of adult patients with B-ALL progressing after CD19 chimeric antigen receptor T-cell therapy
- Author
-
Brigitte Senechal, Mark B. Geyer, Marco L. Davila, Kevin J. Curran, Mithat Gonen, Mikhail Roshal, Isabelle Riviere, Michel Sadelain, Peter Maslak, Jessica Flynn, Kitsada Wudhikarn, Claudia Diamonte, Renier J. Brentjens, Jae H. Park, Xiuyan Wang, and Elizabeth Halton
- Subjects
Adult ,Male ,Oncology ,medicine.medical_specialty ,Clinical Trials and Observations ,Immunology ,Psychological intervention ,Immunotherapy, Adoptive ,Biochemistry ,Disease-Free Survival ,CD19 ,Refractory ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Internal medicine ,Antibodies, Bispecific ,medicine ,Humans ,Inotuzumab Ozogamicin ,Aged ,Salvage Therapy ,Response rate (survey) ,biology ,business.industry ,Cell Biology ,Hematology ,Middle Aged ,Chimeric antigen receptor ,Survival Rate ,Natural history ,biology.protein ,Female ,Chimeric Antigen Receptor T-Cell Therapy ,Blinatumomab ,business ,human activities ,medicine.drug - Abstract
CD19-targeted chimeric antigen receptor (CAR) T-cell therapy has become a breakthrough treatment of patients with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL). However, despite the high initial response rate, the majority of adult patients with B-ALL progress after CD19 CAR T-cell therapy. Data on the natural history, management, and outcome of adult B-ALL progressing after CD19 CAR T cells have not been described in detail. Herein, we report comprehensive data of 38 adult patients with B-ALL who progressed after CD19 CAR T therapy at our institution. The median time to progression after CAR T-cell therapy was 5.5 months. Median survival after post–CAR T progression was 7.5 months. A high disease burden at the time of CAR T-cell infusion was significantly associated with risk of post–CAR T progression. Thirty patients (79%) received salvage treatment of post–CAR T disease progression, and 13 patients (43%) achieved complete remission (CR), but remission duration was short. Notably, 7 (58.3%) of 12 patients achieved CR after blinatumomab and/or inotuzumab administered following post–CAR T failure. Multivariate analysis revealed that a longer remission duration from CAR T cells was associated with superior survival after progression following CAR T-cell therapy. In summary, overall prognosis of adult B-ALL patients progressing after CD19 CAR T cells was poor, although a subset of patients achieved sustained remissions to salvage treatments, including blinatumomab, inotuzumab, and reinfusion of CAR T cells. Novel therapeutic strategies are needed to reduce risk of progression after CAR T-cell therapy and improve outcomes of these patients.
- Published
- 2021
- Full Text
- View/download PDF
38. CD70-specific CAR T cells have potent activity against acute myeloid leukemia without HSC toxicity
- Author
-
Bilal Omer, Linus Angenendt, Christoph Schliemann, Carsten Müller-Tidow, David Nikolov Sedloev, Kathan Parikh, Sandhya Sharma, Michael Schmitt, Qian Chen, Cliona M. Rooney, Tim Sauer, and Stephen Gottschalk
- Subjects
Adoptive cell transfer ,Immunobiology and Immunotherapy ,THP-1 Cells ,T-Lymphocytes ,medicine.medical_treatment ,Immunology ,Biology ,Immunotherapy, Adoptive ,Biochemistry ,Antigen ,medicine ,Humans ,Cytotoxic T cell ,Receptors, Chimeric Antigen ,Myeloid leukemia ,Cell Biology ,Hematology ,Immunotherapy ,Chimeric antigen receptor ,Neoplasm Proteins ,Leukemia, Myeloid, Acute ,Haematopoiesis ,HEK293 Cells ,Cancer research ,Chimeric Antigen Receptor T-Cell Therapy ,CD27 Ligand ,Single-Chain Antibodies - Abstract
The prognosis of patients with acute myeloid leukemia (AML) remains dismal, highlighting the need for novel innovative treatment strategies. The application of chimeric antigen receptor (CAR) T-cell therapy to patients with AML has been limited, in particular by the lack of a tumor-specific target antigen. CD70 is a promising antigen to target AML, as it is expressed on most leukemic blasts, whereas little or no expression is detectable in normal bone marrow samples. To target CD70 on AML cells, we generated a panel of CD70-CAR T cells that contained a common single-chain variable fragment (scFv) for antigen detection, but differed in size and flexibility of the extracellular spacer and in the transmembrane and the costimulatory domains. These CD70scFv CAR T cells were compared with a CAR construct that contained human CD27, the ligand of CD70 fused to the CD3ζ chain (CD27z). The structural composition of the CAR strongly influenced expression levels, viability, expansion, and cytotoxic capacities of CD70scFv-based CAR T cells, but CD27z-CAR T cells demonstrated superior proliferation and antitumor activity in vitro and in vivo, compared with all CD70scFv-CAR T cells. Although CD70-CAR T cells recognized activated virus-specific T cells (VSTs) that expressed CD70, they did not prevent colony formation by normal hematopoietic stem cells. Thus, CD70-targeted immunotherapy is a promising new treatment strategy for patients with CD70-positive AML that does not affect normal hematopoiesis but will require monitoring of virus-specific T-cell responses.
- Published
- 2021
- Full Text
- View/download PDF
39. Is autologous transplant in relapsed DLBCL patients achieving only a PET+ PR appropriate in the CAR T-cell era?
- Author
-
Timothy S. Fenske, Carlos Litovich, Kwang Woo Ahn, Mehdi Hamadani, Nirav N. Shah, Craig S. Sauter, and Yizeng He
- Subjects
Adult ,Male ,Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Immunology ,Hematopoietic stem cell transplantation ,Immunotherapy, Adoptive ,Transplantation, Autologous ,Biochemistry ,Young Adult ,Chemoimmunotherapy ,Positron Emission Tomography Computed Tomography ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Survival analysis ,Aged ,Transplantation ,Errata ,business.industry ,Hazard ratio ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Survival Analysis ,Confidence interval ,Lymphoma ,Treatment Outcome ,Cohort ,Female ,Lymphoma, Large B-Cell, Diffuse ,Neoplasm Recurrence, Local ,business - Abstract
For relapsed chemosensitive diffuse large B-cell lymphoma (DLBCL), consolidation with autologous hematopoietic cell transplantation (auto-HCT) is a standard option. With the approval of anti-CD19 chimeric antigen receptor T cells in 2017, the Center for International Blood and Marrow Transplant Research (CIBMTR) reported an ?∼10% decrease in the number of auto-HCTs for DLBCL in the United States. Using the CIBMTR database, we identified 249 relapsed DLBCL patients undergoing auto-HCT from 2003 to 2013 with a positive positron emission tomography/computed tomography (PET/CT)+ partial response prior to transplant were identified. The study cohort was divided into 2 groups: early chemoimmunotherapy failure (ECF), defined as patients with primary refractory disease (PRefD) or relapse within 12 months of diagnosis and late chemoimmunotherapy failure, defined as patients relapsing after ≥12 months. Primary outcome was overall survival (OS). Secondary outcomes included progression-free survival (PFS) and relapse. Atotal of 182 patients had ECF, whereas 67 did not. Among ECF cohort, 79% had PRefD. The adjusted 5-year probabilities for PFS and OS (ECF vs no ECF) were not different: 41% vs 41% (P = .93) and 51% vs 63% (P = .09), respectively. On multivariate analysis, ECF patients had an increased risk for death (hazard ratio, 1.61; 95% confidence interval, 1.05-2.46; P = .03) but not for PFS or relapse. In conclusion, for relapsed chemosensitive DLBCL patients with residual PET/CT+ disease prior to auto-HCT, the adjusted 5-year PFS (41%) was comparable, irrespective of time to relapse. These data support ongoing application of auto-HCT in chemosensitive DLBCL.
- Published
- 2021
- Full Text
- View/download PDF
40. Targeting a cytokine checkpoint enhances the fitness of armored cord blood CAR-NK cells
- Author
-
Nedyalko Petrov, Junjun Lu, Ana Karen Nunez Cortes, Mustafa H Bdiwi, Matthew S. McNeill, Lorenzo Brunetti, Li Li, Gonca Ozcan, Nobuhiko Imahashi, Ken Chen, Mollie S. Schubert, Duncan Mak, Lucila Nassif Kerbauy, Mark A. Behlke, Luis Muniz-Feliciano, Leiser Silva, Elizabeth J. Shpall, Enli Liu, Ali Rezvan, Natalie W. Fowlkes, Elif Gokdemir, Garrett R. Rettig, Richard Skinner, Xin Ru Jiang, Jing Wang, Vakul Mohanty, Mohsen Fathi, Yifei Shen, Yuanxin Xi, Mayela Mendt, Shahram Kordasti, Natalia Baran, Francesca Lorraine Wei Inng Lim, Nadima Uprety, Sonny Ang, Sunil Acharya, Pinaki P. Banerjee, Marina Konopleva, May Daher, Richard E. Champlin, Gavin Kurgan, Heng Li, Hila Shaim, Rolf Turk, Mecit Kaplan, Xinhai Wan, Mayra Shanley, Emily Ensley, David Marin, Rafet Basar, Navin Varadarajan, Shangrong Lyu, Katayoun Rezvani, Ye Li, and Vandana Nandivada
- Subjects
Cell cycle checkpoint ,medicine.medical_treatment ,Antigens, CD19 ,Immunology ,Suppressor of Cytokine Signaling Proteins ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,Immunotherapy, Adoptive ,Biochemistry ,Gene Knockout Techniques ,Mice ,Cancer immunotherapy ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Immune Checkpoint Inhibitors ,Interleukin-15 ,Receptors, Chimeric Antigen ,Cell Biology ,Hematology ,Immunotherapy ,Fetal Blood ,Burkitt Lymphoma ,Xenograft Model Antitumor Assays ,Aerobiosis ,Immune checkpoint ,Chimeric antigen receptor ,Neoplasm Proteins ,Killer Cells, Natural ,Cytokine ,Interleukin 15 ,Cancer research ,CRISPR-Cas Systems ,Glycolysis ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Immune checkpoint therapy has resulted in remarkable improvements in the outcome for certain cancers. To broaden the clinical impact of checkpoint targeting, we devised a strategy that couples targeting of the cytokine-inducible Src homology 2–containing (CIS) protein, a key negative regulator of interleukin 15 (IL-15) signaling, with fourth-generation “armored” chimeric antigen receptor (CAR) engineering of cord blood–derived natural killer (NK) cells. This combined strategy boosted NK cell effector function through enhancing the Akt/mTORC1 axis and c-MYC signaling, resulting in increased aerobic glycolysis. When tested in a lymphoma mouse model, this combined approach improved NK cell antitumor activity more than either alteration alone, eradicating lymphoma xenografts without signs of any measurable toxicity. We conclude that targeting a cytokine checkpoint further enhances the antitumor activity of IL-15–secreting armored CAR-NK cells by promoting their metabolic fitness and antitumor activity. This combined approach represents a promising milestone in the development of the next generation of NK cells for cancer immunotherapy. Key Points: • CRISPR-Cas9 CISH deletion enhances the metabolic fitness and antitumor activity of armored IL-15–secreting CB-derived CAR-NK cells.
- Published
- 2021
- Full Text
- View/download PDF
41. Oligoclonal T-cell expansion in a patient with bone marrow failure after CD19 CAR-T therapy for Richter-transformed DLBCL
- Author
-
Kai Rejeski, Zhijie Wu, Viktoria Blumenberg, Wolfgang G. Kunz, Susanna Müller, Sachiko Kajigaya, Shouguo Gao, Veit L. Bücklein, Lisa Frölich, Christian Schmidt, Michael von Bergwelt-Baildon, Xingmin Feng, Neal S. Young, and Marion Subklewe
- Subjects
Receptors, Chimeric Antigen ,T-Lymphocytes ,Immunology ,Antigens, CD19 ,Receptors, Antigen, T-Cell ,Humans ,Cell Biology ,Hematology ,Bone Marrow Failure Disorders ,Biochemistry ,Immunotherapy, Adoptive - Published
- 2022
42. CAR-T for follicular lymphoma: are we good to go?
- Author
-
Robin Gasiorowski and Judith Trotman
- Subjects
Receptors, Chimeric Antigen ,Immunology ,Antigens, CD19 ,Humans ,Cell Biology ,Hematology ,Biochemistry ,Immunotherapy, Adoptive ,Lymphoma, Follicular - Published
- 2022
43. NOTCH1 signaling during CD4+ T-cell activation alters transcription factor networks and enhances antigen responsiveness
- Author
-
Alec B. Wilkens, Elena C. Fulton, Margot J. Pont, Gabriel O. Cole, Isabel Leung, Sylvia M. Stull, Matthew R. Hart, Irwin D. Bernstein, Scott N. Furlan, and Stanley R. Riddell
- Subjects
CD4-Positive T-Lymphocytes ,Receptors, Chimeric Antigen ,Lymphoma ,Immunology ,Receptors, Antigen, T-Cell ,Cell Biology ,Hematology ,Biochemistry ,Immunotherapy, Adoptive ,Mice ,Humans ,Animals ,Receptor, Notch1 ,Transcription Factors - Abstract
Adoptive transfer of T cells expressing chimeric antigen receptors (CAR-T) effectively treats refractory hematologic malignancies in a subset of patients but can be limited by poor T-cell expansion and persistence in vivo. Less differentiated T-cell states correlate with the capacity of CAR-T to proliferate and mediate antitumor responses, and interventions that limit tumor-specific T-cell differentiation during ex vivo manufacturing enhance efficacy. NOTCH signaling is involved in fate decisions across diverse cell lineages and in memory CD8+ T cells was reported to upregulate the transcription factor FOXM1, attenuate differentiation, and enhance proliferation and antitumor efficacy in vivo. Here, we used a cell-free culture system to provide an agonistic NOTCH1 signal during naïve CD4+ T-cell activation and CAR-T production and studied the effects on differentiation, transcription factor expression, cytokine production, and responses to tumor. NOTCH1 agonism efficiently induced a stem cell memory phenotype in CAR-T derived from naïve but not memory CD4+ T cells and upregulated expression of AhR and c-MAF, driving heightened production of interleukin-22, interleukin-10, and granzyme B. NOTCH1-agonized CD4+ CAR-T demonstrated enhanced antigen responsiveness and proliferated to strikingly higher frequencies in mice bearing human lymphoma xenografts. NOTCH1-agonized CD4+ CAR-T also provided superior help to cotransferred CD8+ CAR-T, driving improved expansion and curative antitumor responses in vivo at low CAR-T doses. Our data expand the mechanisms by which NOTCH can shape CD4+ T-cell behavior and demonstrate that activating NOTCH1 signaling during genetic modification ex vivo is a potential strategy for enhancing the function of T cells engineered with tumor-targeting receptors.
- Published
- 2022
44. CARs put age in the rearview mirror.
- Author
-
Freeman CL and Locke FL
- Subjects
- Humans, Aged, Immunotherapy, Adoptive, Lymphoma, Large B-Cell, Diffuse, Lymphoma, Non-Hodgkin
- Published
- 2023
- Full Text
- View/download PDF
45. Immune effector cell-associated hematotoxicity: EHA/EBMT consensus grading and best practice recommendations.
- Author
-
Rejeski K, Subklewe M, Aljurf M, Bachy E, Balduzzi A, Barba P, Bruno B, Benjamin R, Carrabba MG, Chabannon C, Ciceri F, Corradini P, Delgado J, Di Blasi R, Greco R, Houot R, Iacoboni G, Jäger U, Kersten MJ, Mielke S, Nagler A, Onida F, Peric Z, Roddie C, Ruggeri A, Sánchez-Guijo F, Sánchez-Ortega I, Schneidawind D, Schubert ML, Snowden JA, Thieblemont C, Topp M, Zinzani PL, Gribben JG, Bonini C, Sureda A, and Yakoub-Agha I
- Subjects
- Consensus, Immunotherapy, Adoptive, Immunologic Factors, Hematopoietic Stem Cell Transplantation, Hematology
- Abstract
Hematological toxicity is the most common adverse event after chimeric antigen receptor (CAR) T-cell therapy. Cytopenias can be profound and long-lasting and can predispose for severe infectious complications. In a recent worldwide survey, we demonstrated that there remains considerable heterogeneity in regard to current practice patterns. Here, we sought to build consensus on the grading and management of immune effector cell-associated hematotoxicity (ICAHT) after CAR T-cell therapy. For this purpose, a joint effort between the European Society for Blood and Marrow Transplantation (EBMT) and the European Hematology Association (EHA) involved an international panel of 36 CAR T-cell experts who met in a series of virtual conferences, culminating in a 2-day meeting in Lille, France. On the basis of these deliberations, best practice recommendations were developed. For the grading of ICAHT, a classification system based on depth and duration of neutropenia was developed for early (day 0-30) and late (after day +30) cytopenia. Detailed recommendations on risk factors, available preinfusion scoring systems (eg, CAR-HEMATOTOX score), and diagnostic workup are provided. A further section focuses on identifying hemophagocytosis in the context of severe hematotoxicity. Finally, we review current evidence and provide consensus recommendations for the management of ICAHT, including growth factor support, anti-infectious prophylaxis, transfusions, autologous hematopoietic stem cell boost, and allogeneic hematopoietic cell transplantation. In conclusion, we propose ICAHT as a novel toxicity category after immune effector cell therapy, provide a framework for its grading, review literature on risk factors, and outline expert recommendations for the diagnostic workup and short- and long-term management., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
46. SARS-CoV-2–specific T cells are rapidly expanded for therapeutic use and target conserved regions of the membrane protein
- Author
-
Ashley Geiger, Hua Liang, Jessica Durkee-Shock, Catherine M. Bollard, Allistair Abraham, C. Russell Cruz, Eva M. Stevenson, Patrick J. Hanley, Fahmida Hoq, R. Brad Jones, Maja Stanojevic, Christopher A. Lazarski, Anushree Datar, Kajal Chaudhry, Zoe Shancer, Haili Lang, Jeffrey I. Cohen, Emily K. Reynolds, Mariah Jensen-Wachspress, Madeline Terpilowski, Ping-Hsien Lee, Kathleen Webber, Robert Ulrey, Kathleen Ferrer, Vaishnavi V. Kankate, Krista Gangler, Katherine M. Harris, Nan Zhang, Stéphanie Val, Peter D. Burbelo, Uduak Ekanem, Michael D. Keller, and Lesia K. Dropulic
- Subjects
Adult ,CD4-Positive T-Lymphocytes ,Male ,Immunobiology and Immunotherapy ,medicine.medical_treatment ,T-Lymphocytes ,Immunology ,Cell Culture Techniques ,Epitopes, T-Lymphocyte ,Inflammation ,Biochemistry ,Immunotherapy, Adoptive ,Epitope ,Viral Proteins ,Young Adult ,Interferon ,Immunity ,medicine ,Humans ,Pandemics ,Aged ,business.industry ,Immunodominant Epitopes ,SARS-CoV-2 ,COVID-19 ,Membrane Proteins ,Cell Biology ,Hematology ,Immunotherapy ,Middle Aged ,Virology ,In vitro ,COVID-19 Drug Treatment ,Vaccination ,Membrane protein ,Female ,medicine.symptom ,business ,medicine.drug - Abstract
T-cell responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been described in recovered patients, and may be important for immunity following infection and vaccination as well as for the development of an adoptive immunotherapy for the treatment of immunocompromised individuals. In this report, we demonstrate that SARS-CoV-2–specific T cells can be expanded from convalescent donors and recognize immunodominant viral epitopes in conserved regions of membrane, spike, and nucleocapsid. Following in vitro expansion using a good manufacturing practice-compliant methodology (designed to allow the rapid translation of this novel SARS-CoV-2 T-cell therapy to the clinic), membrane, spike, and nucleocapsid peptides elicited interferon-γ production, in 27 (59%), 12 (26%), and 10 (22%) convalescent donors (respectively), as well as in 2 of 15 unexposed controls. We identified multiple polyfunctional CD4-restricted T-cell epitopes within a highly conserved region of membrane protein, which induced polyfunctional T-cell responses, which may be critical for the development of effective vaccine and T-cell therapies. Hence, our study shows that SARS-CoV-2 directed T-cell immunotherapy targeting structural proteins, most importantly membrane protein, should be feasible for the prevention or early treatment of SARS-CoV-2 infection in immunocompromised patients with blood disorders or after bone marrow transplantation to achieve antiviral control while mitigating uncontrolled inflammation., Key Points • Coronavirus-specific polyfunctional T cells can be expanded from convalescent individuals for use for patients after bone marrow transplant. • SARS-CoV-2 T-cell products target structural viral proteins, including commonly recognized regions in the C terminus of membrane protein.
- Published
- 2020
47. CAR-modified memory-like NK cells exhibit potent responses to NK-resistant lymphomas
- Author
-
Timothy Schappe, Pamela Wong, Marco L. Davila, Neha Mehta-Shah, Amanda F. Cashen, Wei Meng, John F. DiPersio, Matthew L. Cooper, Brad S. Kahl, Maximilian Schaettler, Jennifer Tran, Melissa M. Berrien-Elliott, Feng Gao, Lynne Marsala, Carly Neal, Nancy D. Marin, Miriam Y. Kim, Margery Gang, Nancy L. Bartlett, Todd A. Fehniger, and Mark P. Foster
- Subjects
Cytotoxicity, Immunologic ,Lymphoma ,Immunobiology and Immunotherapy ,medicine.medical_treatment ,Immunology ,Cell ,Biology ,Immunotherapy, Adoptive ,Biochemistry ,Mice ,medicine ,Animals ,Humans ,Receptor ,Cytotoxicity ,Receptors, Chimeric Antigen ,Degranulation ,Cell Biology ,Hematology ,Immunotherapy ,medicine.disease ,Xenograft Model Antitumor Assays ,Chimeric antigen receptor ,Killer Cells, Natural ,Leukemia ,medicine.anatomical_structure ,Cancer research - Abstract
Natural killer (NK) cells are a promising cellular immunotherapy for cancer. Cytokine-induced memory-like (ML) NK cells differentiate after activation with interleukin-12 (IL-12), IL-15, and IL-18, exhibit potent antitumor responses, and safely induce complete remissions in patients with leukemia. However, many cancers are not fully recognized via NK cell receptors. Chimeric antigen receptors (CARs) have been used to enhance tumor-specific recognition by effector lymphocytes. We hypothesized that ML differentiation and CAR engineering would result in complementary improvements in NK cell responses against NK-resistant cancers. To test this idea, peripheral blood ML NK cells were modified to express an anti-CD19 CAR (19-CAR-ML), which displayed significantly increased interferon γ production, degranulation, and specific killing against NK-resistant lymphoma lines and primary targets compared with nonspecific control CAR-ML NK cells or conventional CAR NK cells. The 19-CAR and ML responses were synergistic and CAR specific and required immunoreceptor tyrosine-based activation motif signaling. Furthermore, 19-CAR-ML NK cells generated from lymphoma patients exhibited improved responses against their autologous lymphomas. 19-CAR-ML NK cells controlled lymphoma burden in vivo and improved survival in human xenograft models. Thus, CAR engineering of ML NK cells enhanced responses against resistant cancers and warrants further investigation, with the potential to broaden ML NK cell recognition against a variety of NK cell–resistant tumors.
- Published
- 2020
- Full Text
- View/download PDF
48. How I prevent infections in patients receiving CD19-targeted chimeric antigen receptor T cells for B-cell malignancies
- Author
-
Joshua A. Hill and Susan K. Seo
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Transplantation Conditioning ,T-Lymphocytes ,medicine.medical_treatment ,Antigens, CD19 ,Immunology ,Infections ,Immunotherapy, Adoptive ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Transplantation Immunology ,Internal medicine ,Humans ,Medicine ,Molecular Targeted Therapy ,Practice Patterns, Physicians' ,Multiple myeloma ,B cell ,B-Lymphocytes ,Infection Control ,Receptors, Chimeric Antigen ,business.industry ,How I Treat ,Immunosuppression ,Cell Biology ,Hematology ,Immunotherapy ,medicine.disease ,Chimeric antigen receptor ,030104 developmental biology ,medicine.anatomical_structure ,Hematologic Neoplasms ,030220 oncology & carcinogenesis ,Chimeric Antigen Receptor T-Cell Therapy ,business - Abstract
Adoptive immunotherapy using B-cell–targeted chimeric antigen receptor (CAR)-modified T cells to treat hematologic malignancies is transforming cancer care for patients with refractory or relapsed diseases. Recent and anticipated regulatory approval for products targeting acute lymphoblastic leukemia, lymphomas, and multiple myeloma have led to global implementation of these novel treatments. The rapidity of commercial utilization of CAR–T-cell therapy has created a largely unexplored gap in patient supportive-care approaches. Such approaches are critical in these complex patients given their high net state of immunosuppression prior to CAR–T-cell infusion coupled with unique acute and persistent insults to their immune function after CAR–T-cell infusion. In this “How I Treat” article, we focus on key questions that arise during 3 phases of management for patients receiving CD19-targeted CAR-T cells: pre CAR–T-cell infusion, immediate post CAR–T-cell infusion, and long-term follow-up. A longitudinal patient case is presented for each phase to highlight fundamental issues including infectious diseases screening, antimicrobial prophylaxis, immunoglobulin supplementation, risk factors for infection, and vaccination. We hope this discussion will provide a framework for institutions and health care providers to formulate their own approach to preventing infections in light of the paucity of data specific to this treatment modality.
- Published
- 2020
- Full Text
- View/download PDF
49. Entecavir prophylaxis for hepatitis B virus reactivation in patients with CAR T-cell therapy
- Author
-
Chaojiang Gu, Shangkun Zhang, Tongcun Zhang, Yang Cao, Na Wang, Lifang Huang, Wenyue Cao, Hao Xu, Yi Xiao, Liang Huang, Chunrui Li, Dengju Li, Jia Wei, Yicheng Zhang, Min Xiao, and Jianfeng Zhou
- Subjects
Adult ,Male ,Hepatitis B virus ,Guanine ,Adolescent ,medicine.medical_treatment ,Immunology ,medicine.disease_cause ,Immunotherapy, Adoptive ,Biochemistry ,medicine ,Humans ,In patient ,business.industry ,Cell Biology ,Hematology ,Immunotherapy ,Entecavir ,Middle Aged ,Hepatitis B ,Virology ,CAR T-cell therapy ,Female ,Virus Activation ,business ,medicine.drug - Published
- 2020
- Full Text
- View/download PDF
50. Long-term outcomes of Sleeping Beauty–generated CD19-specific CAR T-cell therapy for relapsed-refractory B-cell lymphomas
- Author
-
Helen Huls, G. Bardelli, J. Buck, E. de Groot, Partow Kebriaei, Amin M. Alousi, Samer A. Srour, Chitra Hosing, David Marin, William G. Wierda, Harjeet Singh, Y. Nieto, Hagop M. Kantarjian, Jessica McCarty, Laurence J.N. Cooper, U. Popat, Elizabeth J. Shpall, Richard E. Champlin, Stefan O. Ciurea, Gabriella Rondon, Katy Rezvani, and M.H. Qazilbash
- Subjects
Adult ,Male ,Oncology ,medicine.medical_specialty ,Lymphoma, B-Cell ,T-Lymphocytes ,medicine.medical_treatment ,Antigens, CD19 ,Immunology ,Receptors, Antigen, T-Cell ,Immunotherapy, Adoptive ,Biochemistry ,CD19 ,Antigen ,Antigens, Neoplasm ,Internal medicine ,Long term outcomes ,medicine ,Humans ,Letter to Blood ,Receptor ,B cell ,Receptors, Chimeric Antigen ,biology ,business.industry ,Cell Biology ,Hematology ,Immunotherapy ,Middle Aged ,medicine.disease ,Lymphoma ,Treatment Outcome ,medicine.anatomical_structure ,Relapsed refractory ,biology.protein ,business - Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.