86 results on '"Shouval, R."'
Search Results
2. Machine learning based predictive modeling for mortality 100 days post allogeneic hematopoietic stem cell transplantation (ALLO-HSCT) in acute leukemia: an EBMT-Acute Leukemia Working Party (ALWP) registry study [Abstract]
- Author
-
Shouval, R., Labopin, M., Bondi, O., Mishan-Shamay, H., Shimoni, A., Ciceri, F., Esteve, J., Giebel, S., Gorin, N.-C., Schmid, Christoph, Zakaria, I., Moukhtari, L., Polge, E., Aljurf, M., Kroger, N., Craddock, C., Bacigalupo, A., Cornelissen, J., Baron, F., Unger, R., Nagler, A., and Mohty, M.
- Published
- 2022
3. Radiotherapy for Relapse after Chimeric Antigen Receptor T Cell Therapy in Hematologic Malignancies
- Author
-
Hubbeling, H.G., primary, Silverman, E.A., additional, Tomas, A. Alarcon, additional, Tringale, K.R., additional, Wijetunga, N.A., additional, Hajj, C., additional, Mailankody, S., additional, Batlevi, C.L., additional, Dahi, P.B., additional, Giralt, S.A., additional, Lin, R.J., additional, Park, J.H., additional, Scordo, M., additional, Sauter, C.S., additional, Shah, G.L., additional, Perales, M.A., additional, Palomba, M.L., additional, Shouval, R., additional, Yahalom, J., additional, and Imber, B.S., additional
- Published
- 2022
- Full Text
- View/download PDF
4. Patterns of Relapse and Risk Reduction Following Tumor Debulking by Bridging Radiotherapy prior to Chimeric Antigen Receptor T Cell Therapy in Non-Hodgkin Lymphoma
- Author
-
Hubbeling, H.G., primary, Silverman, E.A., additional, Michaud, L., additional, Flynn, J., additional, Devlin, S., additional, Wijetunga, N.A., additional, Tomas, A. Alarcon, additional, Shouval, R., additional, Palomba, M.L., additional, Batlevi, C.L., additional, Dahi, P.B., additional, Park, J.H., additional, Scordo, M., additional, Sauter, C.S., additional, Shah, G.L., additional, Schoder, H., additional, Perales, M.A., additional, Hajj, C., additional, Yahalom, J., additional, and Imber, B.S., additional
- Published
- 2022
- Full Text
- View/download PDF
5. Improvement in Radiomic Features Following Bridging Therapy is Prognostic for CD19 Chimeric Antigen Receptor (CAR) T-Cell Therapy Outcome
- Author
-
Hubbeling, H.G., Leithner, D., Silverman, E.A., Flynn, J., Devlin, S., Shah, G.L., Fregonese, B., Bedmutha, A., Boardman, A., Dahi, P.B., Lin, R.J., Park, J.H., Scordo, M., Salles, G., Yahalom, J., Palomba, M.L., Schoder, H., Perales, M.A., Shouval, R., and Imber, B.S.
- Published
- 2024
- Full Text
- View/download PDF
6. P1108: HIGH COMPLETE RESPONSE RATE FOLLOWING POINT-OF-CARE ANTI CD19 CAR T-CELL THERAPY IN PATIENTS WITH RELAPSED/REFRACTORY FOLLICULAR LYMPHOMA
- Author
-
Fried, S., primary, Besser, M. J., additional, Shkury, E., additional, Yerushalmi, R., additional, Shem-Tov, N., additional, Danylesko, I., additional, Jacoby, E., additional, Itzhaki, O., additional, Shouval, R., additional, Kedmi, M., additional, Shimoni, A., additional, Nagler, A., additional, and Avigdor, A., additional
- Published
- 2022
- Full Text
- View/download PDF
7. CNS radiotherapy as bridging prior to CAR T‐cell therapy for hematologic malignancies.
- Author
-
Cederquist, G., Hubbeling, H., Tringale, K., Shah, G., Tomas, A. Alarcon, Shouval, R., Hajj, C., Fregonese, B., Lee, J., Dahi, P. B., Lin, R. J., Palomba, M. L., Perales, M., Salles, G., Falchi, L., Grommes, C., Scordo, M., Yahalom, J., and Imber, B. S.
- Subjects
HEMATOLOGIC malignancies ,T cells ,RADIOTHERAPY ,CYTOKINE release syndrome ,SCIENCE education - Abstract
CNS radiotherapy as bridging prior to CAR T-cell therapy for hematologic malignancies B Introduction: b Up to 80% of patients who receive CAR T-cell therapy for central nervous system lymphoma (CNSL) require bridging therapy, yet optimal regimens remain undefined. B Conclusion: b Preliminary data suggest CNS-BRT can achieve cytoreduction as bridging therapy prior to CAR T therapy, and may not increase the risk of high-grade CRS and ICANS, though the sample size is limited. [Extracted from the article]
- Published
- 2023
- Full Text
- View/download PDF
8. IMPACT OF RESPONSE TO SYSTEMIC BRIDGING THERAPY ON CLINICAL OUTCOMES AND CYTOKINE PROFILE IN PATIENTS RECEIVING CAR T‐CELL THERAPY FOR AGGRESSIVE B‐CELL LYMPHOMA.
- Author
-
Wills, B., Shouval, R., Flynn, J., Devlin, S. M., Shah, G. L., Scordo, M., P. B. Dahi, Lin, R. J., Imber, B., Parascondola, A., Saldia, A., Landego, I., Ip, A., Leslie, L., Suh, H., Salles, G., Perales, M., and Palomba, M. L.
- Subjects
TREATMENT effectiveness ,T cells ,CYTOKINES - Abstract
IMPACT OF RESPONSE TO SYSTEMIC BRIDGING THERAPY ON CLINICAL OUTCOMES AND CYTOKINE PROFILE IN PATIENTS RECEIVING CAR T-CELL THERAPY FOR AGGRESSIVE B-CELL LYMPHOMA B Background b : It is unclear whether a deeper response to bridging therapy (BT) before chimeric antigen receptor (CAR) T cell therapy improves CAR-T treatment outcome in large B cell lymphoma (LBCL). Further studies are required to evaluate which BT strategies may reduce tumor burden and optimize the inflammatory cytokine environment for improved outcomes after CAR -T cell therapy. [Extracted from the article]
- Published
- 2023
- Full Text
- View/download PDF
9. POINT‐OF‐CARE ANTI‐BCMA CAR T‐CELL THERAPY INDUCES ENCOURAGING RESPONSE RATES IN HIGH‐RISK RELAPSED/REFRACTORY MULTIPLE MYELOMA.
- Author
-
Magen, H., Fried, S., Itzhaki, O., Shem‐Tov, N., Danylesko, I., Yerushalmi, R., Marcus, R., Shouval, R., Nevo, L., Shapira‐Frommer, R., Nagler, A., Shimoni, A., Shkury, E., and Avigdor, A.
- Subjects
MULTIPLE myeloma ,T cells ,CHIMERIC antigen receptors ,CYTOKINE release syndrome ,POINT-of-care testing - Abstract
B Introduction: b Anti-BCMA chimeric antigen receptor (CAR) T-cell therapy showed excellent efficacy in patients with relapsed/refractory multiple myeloma (R/R MM). We report outcomes of phase 1b/2 single-center clinical trial of autologous POC anti-BCMA CAR T-cell therapy in patients (pts) with R/R MM treated with >=3 prior therapies (NCT05243212). POINT-OF-CARE ANTI-BCMA CAR T-CELL THERAPY INDUCES ENCOURAGING RESPONSE RATES IN HIGH-RISK RELAPSED/REFRACTORY MULTIPLE MYELOMA. [Extracted from the article]
- Published
- 2023
- Full Text
- View/download PDF
10. Predictors and implications of renal injury after CD19 chimeric antigen receptor T-cell therapy.
- Author
-
Boardman AP, Gutgarts V, Flynn J, Devlin SM, Goldman A, Tomas AA, Fein JA, Slingerland JB, Parascondola A, Lin RJ, Scordo M, Dahi PB, Giralt S, Palomba ML, Salles G, Nath K, Walji M, Corona M, Park JH, Shah GL, Perales MA, Jaffer-Sathick I, and Shouval R
- Abstract
Chimeric Antigen Receptor (CAR) T cells targeting CD19 induce durable remissions in patients with relapsed or refractory non-Hodgkin lymphoma (NHL), but many patients experience treatmentrelated toxicity. Cytokine release syndrome and immune effector cell-associated neurologic syndrome are extensively characterized. However, limited data exist on the burden, predictors, and implications of acute kidney injury (AKI) after CAR T cell therapy. On initial screening of the FDA adverse event reporting system, we identified a disproportionately high rate of renal adverse events among nearly 6,000 CAR T adverse event reports, suggesting it is clinically important in this patient population. In a subsequent single-center analysis of 399 NHL patients treated with CD19 CAR T cells, we found a substantial burden of AKI after CAR T infusion (10% and 5% of any grade and grade ≥2 AKI) with pre-renal causes being predominant (72%). Evolution to chronic kidney disease was rare, however, 3 patients required hemodialysis. Importantly, patients experiencing cytokine release syndrome and/or neurotoxicity as well as those with low serum albumin and high inflammatory cytokines, including IL-6 and TNF-alpha, were more likely to develop AKI. While pre-CAR T renal dysfunction was not associated with adverse outcomes, patients developing post-CAR T AKI had lower overall survival compared to their counterparts. Our findings indicate that renal dysfunction is a common toxicity of CAR T cell therapy with meaningful prognostic impact. Notably, the link between systemic inflammation and renal dysfunction, suggests that readily available biomarkers may inform on renal injury risk after CAR T cell therapy.
- Published
- 2024
- Full Text
- View/download PDF
11. Development of ALL-Hematotox (ALL-HT): Predicting post CAR T-cell hematotoxicity in B-cell acute lymphoblastic leukemia.
- Author
-
Nair MS, Silbert SK, Rejeski K, Wilson KA, Lamble AJ, Valtis YK, Yates B, Morales Arana A, Shouval R, Curran KJ, Gardner RA, Shalabi H, Annesley C, Park JH, Subklewe M, and Shah NN
- Abstract
Immune effector cell-associated hematotoxicity (ICAHT) is a major B-cell targeted chimeric antigen receptor (CAR) T-cell related toxicity. While ICAHT incidence and severity is documented in large B-cell lymphoma (LBCL), mantle cell lymphoma (MCL), and multiple myeloma (MM), ICAHT has not been described in B-cell acute lymphoblastic leukemia (B-ALL). Similarly, the CAR-HEMATOTOX (CAR-HT) model, designed to predict severe prolonged neutropenia (>14 days of ANC <500/µl), has been validated in LBCL, MCL, and MM, but not in B-ALL. As B-ALL bone marrow (BM) infiltration can impact cytopenias, we sought to describe ICAHT and assess CAR-HT for predicting hematotoxicity in B-ALL. In a cohort of 156 children and young adults with relapsed/refractory B-ALL, the median duration of severe neutropenia (ANC <500/µl) was 13 days (95% CI 10-16), with 83 (53%) experiencing grade >3 ICAHT. Applying CAR-HT, nearly 90% were classified as high-risk, demonstrating limited discriminative power and prompting further development. Using the association identified between BM disease burden and post-infusion neutropenia (r=0.64, p<0.0001), we developed the ALL-Hematotox (ALL-HT) score which substitutes BM disease burden for ferritin in CAR-HT. The ALL-HT score associated with severe prolonged neutropenia (area under the curve=0.84, p<0.0001), and appropriately discriminated high-risk patients (47%) who had more cumulative days of neutropenia (26 vs 4 days, p<0.0001), fewer rates of complete response (88% vs 98%, p=0.03), and shorter median overall survival (9.8 vs 24 months, logrank p=0.0002). ALL-HT was also validated in two independent cohorts. The ALL-HT score refines a widely accepted predictive model of post-infusion hematotoxicity, applicable in B-ALL., (Copyright © 2024 American Society of Hematology.)
- Published
- 2024
- Full Text
- View/download PDF
12. Scalable log-ratio lasso regression for enhanced microbial feature selection with FLORAL.
- Author
-
Fei T, Funnell T, Waters NR, Raj SS, Baichoo M, Sadeghi K, Dai A, Miltiadous O, Shouval R, Lv M, Peled JU, Ponce DM, Perales MA, Gönen M, and van den Brink MRM
- Subjects
- Humans, Hematopoietic Stem Cell Transplantation, Software, Computer Simulation, Microbiota genetics, Algorithms
- Abstract
Identifying predictive biomarkers of patient outcomes from high-throughput microbiome data is of high interest, while existing computational methods do not satisfactorily account for complex survival endpoints, longitudinal samples, and taxa-specific sequencing biases. We present FLORAL, an open-source tool to perform scalable log-ratio lasso regression and microbial feature selection for continuous, binary, time-to-event, and competing risk outcomes, with compatibility for longitudinal microbiome data as time-dependent covariates. The proposed method adapts the augmented Lagrangian algorithm for a zero-sum constraint optimization problem while enabling a two-stage screening process for enhanced false-positive control. In extensive simulation and real-data analyses, FLORAL achieved consistently better false-positive control compared to other lasso-based approaches and better sensitivity over popular differential abundance testing methods for datasets with smaller sample sizes. In a survival analysis of allogeneic hematopoietic cell transplant recipients, FLORAL demonstrated considerable improvement in microbial feature selection by utilizing longitudinal microbiome data over solely using baseline microbiome data., Competing Interests: Declaration of interests J.U.P. reports research funding, intellectual property fees, and travel reimbursement from Seres Therapeutics and consulting fees from DaVolterra, CSL Behring, Crestone, MaaT Pharma, Canaccord Genuity, and RA Capital. He serves on advisory boards of and holds equity in Postbiotics Plus Research and Prodigy Biosciences. He has filed intellectual property applications related to the microbiome (reference nos. 62/843,849, 62/977,908, and 15/756,845). D.M.P. has served as an advisory board member for Evive Biotechnology (Shanghai) Ltd. (formerly Generon [Shanghai] Corporation Ltd.); she served as advisory board member of or consultant to Sanofi, CareDx, Ceramedix, and Incyte; and she receives research funding from Takeda, Sanofi, and Incyte. M.-A.P. reports honoraria from Adicet, Allovir, Caribou Biosciences, Celgene, Bristol Myers Squibb, Equilium, Exevir, Incyte, Karyopharm, Kite/Gilead, Merck, Miltenyi Biotec, MorphoSys, Nektar Therapeutics, Novartis, Omeros, OrcaBio, Syncopation, VectivBio AG, and Vor Biopharma. He serves on data and safety monitoring boards for Cidara Therapeutics, Medigene, and Sellas Life Sciences and on the scientific advisory board of NexImmune. He has ownership interests in NexImmune and Omeros. He has received institutional research support for clinical trials from Incyte, Kite/Gilead, Miltenyi Biotec, Nektar Therapeutics, and Novartis. M.R.M.v.d.B. has received research support and stock options from Seres Therapeutics and stock options from Notch Therapeutics and Pluto Therapeutics; he has received royalties from Wolters Kluwer; he has consulted for, received honorarium from, or participated in advisory boards for Seres Therapeutics, Vor Biopharma, Rheos Medicines, Frazier Healthcare Partners, Nektar Therapeutics, Notch Therapeutics, Ceramedix, Lygenesis, Pluto Therapeutics, GlaxoSmithKline, Da Volterra, Thymofox, Garuda, Novartis (spouse), Synthekine (spouse), Beigene (spouse), and Kite (spouse); he has intellectual property licensing with Seres Therapeutics and Juno Therapeutics; and he holds a fiduciary role on the foundation board of DKMS (a nonprofit organization). MSKCC has institutional financial interests relative to Seres Therapeutics., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
13. Metabolic Tumor Volume Response after Bridging Therapy Determines Chimeric Antigen Receptor T-Cell Outcomes in Large B-Cell Lymphoma.
- Author
-
Hubbeling H, Leithner D, Silverman EA, Flynn J, Devlin S, Shah G, Fregonese B, Wills B, Bedmutha A, Alarcon Tomas A, Parascondola A, Saldia A, Landego I, Hajj C, Boardman AP, Dahi PB, Ghosh A, Giralt S, Lin RJ, Park J, Scordo M, Salles G, Yahalom J, Palomba ML, Schöder H, Perales MA, Shouval R, and Imber BS
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Adult, Prognosis, Treatment Outcome, Antigens, CD19 immunology, Young Adult, Retrospective Studies, Lymphoma, Large B-Cell, Diffuse therapy, Lymphoma, Large B-Cell, Diffuse immunology, Lymphoma, Large B-Cell, Diffuse pathology, Immunotherapy, Adoptive methods, Tumor Burden, Receptors, Chimeric Antigen immunology, Receptors, Chimeric Antigen metabolism
- Abstract
Purpose: Greater disease burden is a well-established predictor of poorer outcomes following chimeric antigen receptor T-cell (CAR T) therapy. Although bridging therapy (BT) is widely used between leukapheresis and CAR T infusion, limited data have evaluated the impact of BT on CAR T outcomes. In this study, we hypothesized that the quantitative dynamics of radiomic cytoreduction during bridging are prognostic., Experimental Design: Patients with large B-cell lymphoma treated with CD19-CAR T from 2016 to 2022 were included in the study. Metabolic tumor volume (MTV) was determined for all patients on pre-leukapheresis PET and on post-BT/pre-infusion PET in those who received BT. Patients were stratified into "High" and "Low" disease burden using an MTV cutpoint of 65.4cc established by maximally selected log-rank statistic for progression-free survival (PFS)., Results: Of 191 patients treated with CAR T, 144 (75%) received BT. In the BT cohort, 56% had a reduction in MTV post-BT. On multivariate analysis, the MTV trajectory across the bridging period remained significantly associated with PFS (P < 0.001); however, notably, patients with improved MTV (High->Low) had equivalent PFS compared with those with initially and persistently low MTV (Low->Low; HR for High->Low MTV: 2.74; 95% confidence interval, 0.82-9.18). There was a reduction in any grade immune effector cell-associated neurotoxicity syndrome in the High->Low MTV cohort as compared with the High->High MTV cohort (13% vs. 41%; P = 0.05)., Conclusions: This is the first study to use radiomics to quantify disease burden pre- and post-BT in a large real-world large B-cell lymphoma cohort. We demonstrate that effective BT can enable initially high-disease burden patients to achieve post-CAR T outcomes comparable with low-disease burden patients., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
14. Correction: Development and validation of an automated computational approach to grade immune effector cell-associated hematotoxicity.
- Author
-
Liang EC, Rejeski K, Fei T, Albittar A, Huang JJ, Portuguese AJ, Wu Q, Raj S, Subklewe M, Shouval R, and Gauthier J
- Published
- 2024
- Full Text
- View/download PDF
15. Anti-CD19 chimeric antigen receptor T-cell therapy has less efficacy in Richter transformation than in <I>de novo</I> large B-cell lymphoma and transformed low-grade B-cell lymphoma.
- Author
-
Benjamini O, Fried S, Shouval R, Flynn JR, Beyar-Katz O, Leslie LA, Zucherman T, Yerushalmi R, Shem-Tov N, Palomba ML, Danylesko I, Sdayoor I, Malka H, Itzhaki O, Suh H, Devlin SM, Marcus R, Dahi PB, Jacoby E, Shah GL, Sauter CS, Ip A, Perales MA, Nagler A, Shimoni A, Scordo M, and Avigdor A
- Subjects
- Humans, Male, Middle Aged, Female, Aged, Retrospective Studies, Adult, Treatment Outcome, Aged, 80 and over, Leukemia, Lymphocytic, Chronic, B-Cell therapy, Leukemia, Lymphocytic, Chronic, B-Cell mortality, Leukemia, Lymphocytic, Chronic, B-Cell immunology, Neoplasm Grading, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods, Lymphoma, Large B-Cell, Diffuse therapy, Lymphoma, Large B-Cell, Diffuse immunology, Lymphoma, Large B-Cell, Diffuse mortality, Antigens, CD19 immunology, Receptors, Chimeric Antigen immunology
- Abstract
The activity of anti-CD19 chimerci antigen receptor (CAR) T-cell therapy in chronic lymphocytic leukemia (CLL) with Richter's transformation (RT) to aggressive large B-cell lymphoma (LBCL) is largely unknown. In a multicenter retrospective study, we report the safety and efficacy of CAR T-cell therapy in patients with RT (N=30) compared to patients with aggressive B-cell lymphoma (N=283) and patients with transformed indolent non-Hodgkin lymphoma (iNHL) (N=141) between April 2016 and January 2023. Two-thirds of patients received prior therapy for CLL before RT and 89% of them received B-cell receptor and B-cell lymphoma 2 inhibitors. Toxicities of CAR T-cell therapy in RT were similar to other lymphomas, with no fatalities related to cytokine release syndrome or immune effector-cell associated neurotoxicity synderome. The 100-day overall response rate and complete response rates in patients with RT were 57% and 47%, respectively. With a median follow-up of 19 months, the median overall survival (OS) was 9.9 months in patients with RT compared to 18 months in de novo LBCL and not reached in patients with transformed iNHL. The OS at 12 months was 45% in patients with RT compared with 62% and 75% in patients with de novo LBCL and transformed iNHL, respectively. In a multivariate analysis, worse OS was associated with RT histology, elevated lactate dehydrogenase, and more prior lines of therapy. CAR T-cell therapy can salvage a proportion of patients with CLL and RT exposed to prior targeted agents; however, efficacy in RT is inferior compared to de novo LBCL and transformed iNHL.
- Published
- 2024
- Full Text
- View/download PDF
16. Cancer cachexia and weight loss prior to CAR T-cell therapy for lymphoma are independently associated with poor outcomes.
- Author
-
Valtis YK, Devlin SM, Shouval R, Rejeski K, Corona M, Luna De Abia A, Rivas-Delgado A, Luttwak E, Cassanello G, Landego I Dr, Schöder H, Bedmutha A, Boardman AP, Shah GL, Scordo M, Perales MA, Salles GA, Palomba ML, Shah UA, and Park JH
- Abstract
CAR T-cell therapy has transformed the care of lymphoma, yet many patients relapse. Several prognostic markers have been associated with CAR T cell outcomes, such as tumor burden, response to bridging chemotherapy, and laboratory parameters at the time of lymphodepletion or infusion. The effect of cancer cachexia and weight loss prior to CAR T cells on toxicity and outcomes is not well understood. Here, we present a retrospective single institution cohort study of 259 patients with lymphoma treated with CAR T-cells between 2017 and 2023. We observed that patients with a >5% decrease in their body mass index (BMI) in the 3 months preceding CAR T treatment (weight loss group; all meeting one of the commonly accepted definitions of cancer cachexia) had higher disease burden and inflammatory parameters (CRP, ferritin, IL6, TNFa) at time of lymphodepletion and CAR T-cell infusion. Patients with weight loss experienced higher rates of grade 3+ neurotoxicity and early hematotoxicity but those effects were not seen upon multivariable adjustment. However, in both univariate and multivariable analysis, patients with weight loss had worse response rates, overall survival, and event-free survival, indicating that weight loss is an independent poor prognostic factor. Our data suggest that weight loss in the 3 months preceding CAR T-cell therapy represents a worrisome "alarm signal" and potentially modifiable factor alongside tumor burden and inflammation and warrants further investigation in patients treated with CAR T therapy., (Copyright © 2024 American Society of Hematology.)
- Published
- 2024
- Full Text
- View/download PDF
17. Shift from Widespread to Tailored Antifungal Prophylaxis in Lymphoma Patients Treated with CD19 CAR T Cell Therapy: Results from a Large Retrospective Cohort.
- Author
-
Melica G, Luna de Abia A, Shah GL, Devlin S, Corona M, Fein J, Dahi PB, Giralt SA, Lin RJ, Palomba ML, Parascondola A, Park J, Salles G, Saldia A, Scordo M, Shouval R, Perales MA, and Seo SK
- Abstract
Patients undergoing CD19 chimeric antigen receptor (CAR)-T cell therapy exhibit multiple immune deficits that may increase their susceptibility to infections. Invasive fungal infections (IFIs) are life-threatening events in the setting of hematologic diseases. However, there is ongoing debate regarding the optimal role and duration of antifungal prophylaxis in this specific patient population. The objective of this study was to provide a comprehensive overview of the evolution of IFI prophylactic strategies over time and to assess IFI incidence rates in a cohort of patients with relapsed or refractory (R/R) lymphoma treated with CAR-T cell therapy. A single-center retrospective study was conducted on a cohort of patients with R/R B cell lymphoma treated with CD19 CAR-T cell therapy between April 2016 and March 2023. Group A (April 2016-August 2020) consisted of patients primarily treated with fluconazole, irrespective of their individual IFI risk profile. In Group B (September 2020-March 2023) antifungal prophylaxis was recommended only for high-risk patients. Overall, 330 patients were included. Antifungal prophylaxis was prescribed to 119/142 (84%) patients in Group A and 58/188 (31%) in Group B (P < .001). Anti-mold azoles were prescribed to 8 (5.6%) patients in Group A and 21 (11.2%) patients in Group B. In Group A, 42 (29%) patients were switched to another antifungal, 9 (21%) because of toxicity, with 6 cases of transaminitis and 3 cases of prolonged QTc. In Group B, 21 (11.2%) patients were switched to the antifungal drug, mainly from fluconazole or micafungin to a mold-active agent following revised guidelines. No difference was found in liver toxicity between the two groups at infusion, day 10, and day 30. No significant differences were observed between the groups. IFIs following CAR-T cell therapy were rare, with 1 case of cryptococcal meningoencephalitis in group A (.7%) and 1 case of invasive aspergillosis in Group B (.5%), both occurring in patients on micafungin prophylaxis. In this large single-center cohort of patients with R/R lymphoma treated with CAR-T cells, we show that individualized prophylaxis, alongside careful management of CAR-T cell-related toxicities such as CRS, was associated with a very low IFI rate, avoiding the risk of unnecessary toxicities, drug-drug interactions, and high costs., (Copyright © 2024 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
18. CD19-directed CART therapy for T-cell/histiocyte-rich large B-cell lymphoma.
- Author
-
Pophali PA, Fein JA, Ahn KW, Allbee-Johnson M, Ahmed N, Awan FT, Farhan S, Grover NS, Hilal T, Iqbal M, Maakaron J, Modi D, Nasrollahi E, Schachter LG, Sauter C, Hamadani M, Herrera A, Shouval R, and Shadman M
- Subjects
- Humans, Male, Middle Aged, Female, Adult, Aged, T-Lymphocytes immunology, T-Lymphocytes metabolism, Histiocytes pathology, Treatment Outcome, Receptors, Chimeric Antigen therapeutic use, Young Adult, Antigens, CD19 immunology, Antigens, CD19 therapeutic use, Lymphoma, Large B-Cell, Diffuse therapy, Lymphoma, Large B-Cell, Diffuse drug therapy, Lymphoma, Large B-Cell, Diffuse mortality, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods
- Abstract
Abstract: T-cell/histiocyte-rich large B-cell lymphoma (THRLBCL) is a rare histologic variant of LBCL. Limited data regarding CD19-directed chimeric antigen receptor T-cell (CART) therapy in relapsed/refractory (R/R) THRLBCL suggest poor efficacy. We investigated CART outcomes for R/R THRLBCL through the Center for International Blood and Marrow Transplant Research registry. A total of 58 adult patients with R/R THRLBCL who received commercial CD19-CART therapy between 2018 and 2022 were identified. Most patients (67%) had early relapse of disease (45% primary refractory) with a median of 3 (range, 1-7) prior therapies and were treated with axicabtagene ciloleucel (69%). At median follow-up of 23 months after CART therapy, 2-year overall and progression-free survival were 42% (95% confidence interval [CI], 27-57) and 29% (95% CI, 17-43), respectively. In univariable analysis, poor performance status before CART therapy was associated with higher mortality (hazard ratio, 2.35; 95%CI, 1.02-5.5). The 2-year cumulative incidences of relapse/progression and nonrelapse mortality were 69% and 2%, respectively. Grade ≥3 cytokine release syndrome and immune effector cell-associated neurologic syndrome occurred in 7% and 15% of patients, respectively. In this largest analysis of CD19-CART therapy for R/R THRLBCL, ∼30% of patients were alive and progression free 2 years after CART therapy. Despite a high incidence of progression (69% at 2 years), these results suggest a subset of patients with R/R THRLBCL may have durable responses with CARTs., (© 2024 by The American Society of Hematology. 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
19. Second Primary Malignancies after CAR T-Cell Therapy: A Systematic Review and Meta-analysis of 5,517 Lymphoma and Myeloma Patients.
- Author
-
Tix T, Alhomoud M, Shouval R, Cliff ERS, Perales MA, Cordas Dos Santos DM, and Rejeski K
- Subjects
- Humans, Receptors, Chimeric Antigen immunology, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods, Lymphoma immunology, Lymphoma therapy, Multiple Myeloma therapy, Multiple Myeloma immunology, Neoplasms, Second Primary epidemiology, Neoplasms, Second Primary etiology
- Abstract
Purpose: Chimeric antigen receptor (CAR) T-cell therapy is a potent immunotherapy for hematologic malignancies, but patients can develop long-term adverse events, including second primary malignancies (SPM) that impact morbidity and mortality. To delineate the frequency and subtypes of SPMs following CAR-T in lymphoma and myeloma, we performed a systematic review and meta-analysis., Experimental Design: A literature search was conducted in the MEDLINE, Embase, and Cochrane CENTRAL databases. Following the extraction of SPM cases and assignment of malignant origin, we analyzed SPM point estimates using random effects models., Results: We identified 326 SPMs across 5,517 patients from 18 clinical trials and 7 real-world studies. With a median follow-up of 21.7 months, the overall SPM point estimate was 6.0% (95% confidence interval, 4.8%-7.4%). SPM estimates were associated with treatment setting (clinical trials > real-world studies), duration of follow-up, and number of prior treatment lines, which were each confirmed as independent study-level risk factors of SPM in a meta-regression model. A subgroup meta-analysis of the four trials that randomized CAR-T versus standard-of-care revealed a similar risk of SPM with either treatment strategy (P = 0.92). In a distribution analysis of SPM subtypes, hematologic malignancies were the most common entity (37%), followed by solid tumors (27%) and non-melanoma skin cancers (16%). T-cell malignancies represented a small minority of events (1.5%). We noted disease- and product-specific variations in SPM distribution., Conclusions: These data raise awareness of SPM as a clinically relevant long-term adverse event in patients receiving CAR T-cell therapy. However, our findings do not indicate that SPM frequency is higher with CAR-T versus previous standard-of-care strategies., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
20. CNS bridging radiotherapy achieves rapid cytoreduction before CAR T-cell therapy for aggressive B-cell lymphomas.
- Author
-
Cederquist GY, Schefflein J, Devlin SM, Shah GL, Shouval R, Hubbeling H, Tringale K, Alarcon Tomas A, Fregonese B, Hajj C, Boardman A, Luna De Abia A, Corona M, Cassanello G, Dahi PB, Lin RJ, Ghione P, Salles G, Perales MA, Palomba ML, Falchi L, Scordo M, Grommes C, Yahalom J, and Imber BS
- Subjects
- Humans, Middle Aged, Male, Female, Aged, Adult, Treatment Outcome, Receptors, Chimeric Antigen therapeutic use, Combined Modality Therapy, Immunotherapy, Adoptive methods, Immunotherapy, Adoptive adverse effects, Lymphoma, B-Cell therapy, Lymphoma, B-Cell radiotherapy, Central Nervous System Neoplasms therapy, Central Nervous System Neoplasms radiotherapy
- Abstract
Abstract: Chimeric antigen receptor (CAR) T-cell therapy (CART) for central nervous system lymphoma (CNSL) is a promising strategy, yet responses are frequently not durable. Bridging radiotherapy (BRT) is used for extracranial lymphoma in which it can improve CART outcomes through cytoreduction of high-risk lesions. We hypothesized that BRT would achieve similar, significant cytoreduction before CART for CNSL (CNS-BRT). We identified patients with CNSL with non-Hodgkin B-cell lymphoma who received CNS-BRT before commercial CART. Cytoreduction from CNS-BRT was calculated as change in lesion size before CART. Twelve patients received CNS-BRT, and the median follow-up among survivors is 11.8 months (interquartile range, 8.5-21.9). Ten patients had CNSL (9 secondary, 1 primary) and 2 patients had epidural disease (evaluable for toxicity). All 10 patients with CNSL had progressive disease at the time of CNS-BRT. Of 12 patients, 1 experienced grade ≥3 cytokine release syndrome, and 3 of 12 patients experienced grade ≥3 immune effector cell-associated neurotoxicity syndrome. CNS-BRT achieved a 74.0% (95% confidence interval, 62.0-86.0) mean reduction in lesion size from baseline (P = .014) at a median of 12 days from BRT completion and before CART infusion. Best CNS response included 8 complete responses, 1 partial response, and 1 progressive disease. Three patients experienced CNS relapse outside the BRT field. Preliminary data suggest CNS-BRT achieves rapid cytoreduction and is associated with a favorable CNS response and safety profile. These data support further study of BRT as a bridging modality for CNSL CART., (© 2024 by The American Society of Hematology. 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
21. CAF-induced physical constraints controlling T cell state and localization in solid tumours.
- Author
-
Arpinati L, Carradori G, and Scherz-Shouval R
- Subjects
- Humans, Animals, Signal Transduction, Tumor Microenvironment immunology, Neoplasms immunology, Neoplasms pathology, Cancer-Associated Fibroblasts immunology, Cancer-Associated Fibroblasts metabolism, Cancer-Associated Fibroblasts pathology, Extracellular Matrix metabolism, Extracellular Matrix immunology, T-Lymphocytes immunology
- Abstract
Solid tumours comprise cancer cells that engage in continuous interactions with non-malignant cells and with acellular components, forming the tumour microenvironment (TME). The TME has crucial and diverse roles in tumour progression and metastasis, and substantial efforts have been dedicated into understanding the functions of different cell types within the TME. These efforts highlighted the importance of non-cell-autonomous signalling in cancer, mediating interactions between the cancer cells, the immune microenvironment and the non-immune stroma. Much of this non-cell-autonomous signalling is mediated through acellular components of the TME, known as the extracellular matrix (ECM), and controlled by the cells that secrete and remodel the ECM - the cancer-associated fibroblasts (CAFs). In this Review, we delve into the complex crosstalk among cancer cells, CAFs and immune cells, highlighting the effects of CAF-induced ECM remodelling on T cell functions and offering insights into the potential of targeting ECM components to improve cancer therapies., (© 2024. Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
22. HSF1 renders NK cells too stressed to respond.
- Author
-
Gruper Y, Ben-Shmuel A, and Scherz-Shouval R
- Published
- 2024
- Full Text
- View/download PDF
23. Subsequent Malignancies After CD19-Targeted Chimeric Antigen Receptor T Cells in Patients With Lymphoma.
- Author
-
Lorenc R, Shouval R, Flynn JR, Devlin SM, Saldia A, De Abia AL, De Lapuerta MC, Tomas AA, Cassanello G, Leslie LA, Rejeski K, Lin RJ, Scordo M, Shah GL, Palomba ML, Salles G, Park J, Giralt SA, Perales MA, Ip A, and Dahi PB
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Adult, Retrospective Studies, Neoplasms, Second Primary immunology, Neoplasms, Second Primary epidemiology, Young Adult, Aged, 80 and over, Immunotherapy, Adoptive adverse effects, Receptors, Chimeric Antigen immunology, Antigens, CD19 immunology
- Abstract
Chimeric antigen receptor (CAR) T cells are an established treatment for B cell non-Hodgkin lymphomas (B-NHL). With the remarkable success in improving survival, understanding the late effects of CAR T cell therapy is becoming more relevant. The aim of this study is to determine the incidence of subsequent malignancies in adult patients with B-NHL. We retrospectively studied 355 patients from 2 different medical centers treated with four different CAR T cell products from 2016 to 2022. The overall cumulative incidence for subsequent malignancies at 36 months was 14% (95% CI: 9.2%, 19%). Subsequent malignancies were grouped into 3 primary categories: solid tumor, hematologic malignancy, and dermatologic malignancy with cumulative incidences at 36 months of 6.1% (95% CI: 3.1%-10%), 4.5% (95% CI: 2.1%-8.1%) and 4.2% (95% CI: 2.1%-7.5%) respectively. Notably, no cases of T cell malignancies were observed. In univariable analysis, increasing age was associated with higher risk for subsequent malignancy. While the overall benefits of CAR T products continue to outweigh their potential risks, more studies and longer follow ups are needed to further demonstrate the risks, patterns, and molecular pathways that lead to the development of subsequent malignancies., (Copyright © 2024 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
24. Dermatologic Adverse Events Associated With Chimeric Antigen Receptor T-Cell Therapy: A Pharmacovigilance Analysis of the FDA Reporting System.
- Author
-
Storgard R, Dusza S, Shouval R, Scordo M, and Markova A
- Subjects
- Humans, United States epidemiology, Male, Female, Middle Aged, Adult, Adolescent, Aged, Receptors, Chimeric Antigen, Receptors, Antigen, T-Cell therapeutic use, Young Adult, Skin Diseases chemically induced, Skin Diseases therapy, Lymphoma, Large B-Cell, Diffuse drug therapy, Biological Products adverse effects, Pharmacovigilance, Immunotherapy, Adoptive adverse effects, United States Food and Drug Administration, Adverse Drug Reaction Reporting Systems statistics & numerical data
- Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy, including axicabtagene ciloleucel (axi-cel) and tisagenlecleucel (tisa-cel), has demonstrated significant efficacy in treating refractory or relapsed diffuse large B-cell lymphoma and B-cell acute lymphoblastic leukemia. Though adverse events such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) are well characterized, the dermatologic adverse event (DAE) profile is less thoroughly described. This study aims to provide the first comprehensive analysis of DAEs associated with axi-cel and tisa-cel using real-world data from the FDA Adverse Event Reporting System (FAERS) database. FAERS database reports citing axi-cel or tisa-cel in patients aged 16 years or older were included, excluding duplicate reports and off-label indications. Disproportionality analysis by reporting odds ratio (ROR) was utilized to detect increased reporting of drug-adverse event combinations. Of the 11,256,845 reports in the FAERS database, 5559 identified CAR-T therapy as the primary suspected drug. After exclusions, 3,666 reports were analyzed (2,168 for axi-cel and 1,498 for tisa-cel). Among these, 2.7% of axi-cel and 5.1% of tisa-cel cases reported DAEs. There was a statistically significant increased reporting of 2 DAE groups associated with CAR-T therapy: severe cutaneous eruptions (ROR 5.18, 95% CI 1.29, 20.76) and vascular cutaneous (ROR 2.91, 95% CI 1.51, 5.60). The median time to DAE onset was 3 days after CAR T-cell infusion. Death was a reported outcome in 11.9% and 13.0% of axi-cel and tisa-cel DAE cases, respectively, and in 50% and 25% of severe cutaneous eruptions and vascular cutaneous cases, respectively. This study reveals a significantly increased reporting rate of severe cutaneous eruptions and vascular cutaneous DAEs associated with CAR-T therapy, with both event groups associated with high mortality. These results emphasize the importance of monitoring dermatologic toxicities in clinical practice to ensure timely identification and management of potentially severe adverse events., (Copyright © 2024 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
25. Point of care CD19 chimeric antigen receptor (CAR) T-cells for relapsed/refractory acute myeloid leukemia (AML) with aberrant CD19 antigen expression.
- Author
-
Danylesko I, Shem-Tov N, Yerushalmi R, Jacoby E, Toren A, Shouval R, Itzhaki O, Avigdor A, Shimoni A, and Nagler A
- Abstract
Relapsed/refractory (r/r) acute myeloid leukemia (AML) is associated with poor prognosis. CD19 is a B-cell marker, is aberrantly expressed in AML, mostly with t(8; 21)(q22; q22.1). Here we report the results of a phase 2 study giving point of care produced CD19 CAR T- cells for r/r AML with aberrant expression of CD19 (NCT04257175). Lymphodepletion included fludarabine and cyclophosphamide The response was evaluated by bone marrow (BM) aspiration on day 28. Six patients (5 adults and 1 child) were included. Median number of previous chemotherapy lines was 4 (range, 3-8) and four patients received CAR T-cells 8-18 months post allogeneic hematopoietic stem cell transplantation (allo-HSCT). Cytokine release syndrome (CRS) of any grade occurred in all patients, and 1 patient had grade 3 CRS. Immune effector cell-associated neurotoxicity syndrome (ICANS) occurred in 2 patients at low grades. Tocilizumab was administered to 2 patients and corticosteroids to 3 patients. Four patients achieved a complete remission (CR), while 2/6 progressed (PD). Three patients (2 with CR and 1 with PD) underwent allo-HSCT (it was the second transplant in 2) 2-5 months post CAR T-cells infusion. The median duration of response in patients achieving CR was 8.5 (range; 3-14) months. However, all patients eventually died within 5 (1-18) months. In conclusion, CD19 CAR T- cell treatment for AML is feasible and safe. However, the response is short and should be followed by allo-HSCT. Hopefully, future long term results will be improved by combining the CAR T- cell therapy with the emerging novel effective anti-leukemic compounds., Competing Interests: Declaration of competing interest The authors declare that they have no relevant conflict of interest and no competing financial interests., (Copyright © 2024. Published by Elsevier Masson SAS.)
- Published
- 2024
- Full Text
- View/download PDF
26. Microbial metabolite-guided CAR T cell engineering enhances anti-tumor immunity via epigenetic-metabolic crosstalk.
- Author
-
Staudt S, Nikolka F, Perl M, Franz J, Leblay N, Yuan XK, Warmuth L, Fantes MA, Skorupskaitė A, Fei T, Bromberg M, Martin-Uriz PS, Rodriguez-Madoz JR, Ziegler-Martin K, Gholam NA, Benz P, Tran PH, Freitag F, Riester Z, Stein-Thoeringer C, Schmitt M, Kleigrewe K, Weber J, Mangold K, Einsele H, Prosper F, Ellmeier W, Busch D, Visekruna A, Slingerland J, Shouval R, Hiller K, van den Brink M, Pausch P, Neri P, Hudecek M, Poeck H, and Luu M
- Abstract
Emerging data have highlighted a correlation between microbiome composition and cancer immunotherapy outcome. While commensal bacteria and their metabolites are known to modulate the host environment, contradictory effects and a lack of mechanistic understanding impede the translation of microbiome-based therapies into the clinic. In this study, we demonstrate that abundance of the commensal metabolite pentanoate is predictive for survival of chimeric antigen receptor (CAR) T cell patients in two independent cohorts. Its implementation in the CAR T cell manufacturing workflow overcomes solid tumor microenvironments in immunocompetent cancer models by hijacking the epigenetic-metabolic crosstalk, reducing exhaustion and promoting naive-like differentiation. While synergy of clinically relevant drugs mimicked the phenotype of pentanoate-engineered CAR T cells in vitro , in vivo challenge showed inferior tumor control. Metabolic tracing of
13 C-pentanoate revealed citrate generation in the TCA cycle via the acetyl- and succinyl-CoA entry points as a unique feature of the C5 aliphatic chain. Inhibition of the ATP-citrate lyase, which links metabolic output and histone acetylation, led to accumulation of pentanoate-derived citrate from the succinyl-CoA route and decreased functionality of SCFA-engineered CAR T cells. Our data demonstrate that microbial metabolites are incorporated as epigenetic imprints and implementation into CAR T cell production might serve as embodiment of the microbiome-host axis benefits for clinical applications., Competing Interests: ML, MH and AV are listed as inventors on patent application WO2021/058811A1. MH is listed as an inventor on patent applications and granted patents related to CAR-T technologies that have been filed by the Fred Hutchinson Cancer Research Center, Seattle, WA and by the University of Würzburg, Würzburg, Germany. MH is a co-founder and equity owner of T-CURX GmbH, Würzburg, Germany. MH received honoraria from Celgene/BMS, Janssen, Kite/Gilead. MvdB has received research support and stock options from Seres Therapeutics and stock options from Notch Therapeutics and Pluto Therapeutics; he has received royalties from Wolters Kluwer; has consulted, received honorarium from or participated in advisory boards for Seres Therapeutics, Rheos Medicines, Ceramedix, Pluto Therapeutics, Thymofox, Garuda, Novartis (Spouse), Synthekine (Spouse), Beigene (Spouse), Kite (Spouse); he has IP Licensing with Seres Therapeutics and Juno Therapeutics; and holds a fiduciary role on the Foundation Board of DKMS (a nonprofit organization). PN received honoraria from BMS, Janssen, Sanofi and Pfizer as a consultant/advisory board member. MAF received honoraria from Novartis and Sanofi and travel grants from Sanofi. HP is a consultant for Gilead, Abbvie, Pfizer, Novartis, Servier, and Bristol Myers-Squibb. The remaining authors declare no financial conflict of interest.- Published
- 2024
- Full Text
- View/download PDF
27. A systematic review and meta-analysis of nonrelapse mortality after CAR T cell therapy.
- Author
-
Cordas Dos Santos DM, Tix T, Shouval R, Gafter-Gvili A, Alberge JB, Cliff ERS, Theurich S, von Bergwelt-Baildon M, Ghobrial IM, Subklewe M, Perales MA, and Rejeski K
- Subjects
- Humans, Biological Products administration & dosage, Biological Products adverse effects, Receptors, Chimeric Antigen immunology, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods, Lymphoma therapy, Lymphoma immunology, Lymphoma mortality, Multiple Myeloma therapy, Multiple Myeloma immunology, Multiple Myeloma mortality
- Abstract
Although chimeric antigen receptor (CAR) T cell therapy represents a transformative immunotherapy, it is also associated with distinct toxicities that contribute to morbidity and mortality. In this systematic review and meta-analysis, we searched MEDLINE, Embase and CINAHL (Cochrane) for reports of nonrelapse mortality (NRM) after CAR T cell therapy in lymphoma and multiple myeloma up to March 2024. After extraction of causes and numbers of death, we analyzed NRM point estimates using random-effect models. We identified 7,604 patients across 18 clinical trials and 28 real-world studies. NRM point estimates varied across disease entities and were highest in patients with mantle-cell lymphoma (10.6%), followed by multiple myeloma (8.0%), large B cell lymphoma (6.1%) and indolent lymphoma (5.7%). Entity-specific meta-regression models for large B cell lymphoma and multiple myeloma revealed that axicabtagene ciloleucel and ciltacabtagene autoleucel were independently associated with increased NRM point estimates, respectively. Of 574 reported nonrelapse deaths, over half were attributed to infections (50.9%), followed by other malignancies (7.8%) and cardiovascular/respiratory events (7.3%). Conversely, the CAR T cell-specific side effects, immune effector cell-associated neurotoxicity syndrome/neurotoxicity, cytokine release syndrome and hemophagocytic lymphohistiocytosis, represented only a minority of nonrelapse deaths (cumulatively 11.5%). Our findings underline the critical importance of infectious complications after CAR T cell therapy and support the comprehensive reporting of NRM, including specific causes and long-term outcomes., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2024
- Full Text
- View/download PDF
28. Atrial arrhythmias following CAR-chimeric antigen receptor T-cell therapy: Incidence, risk factors and biomarker profile.
- Author
-
Shouval R, Goldman A, Flynn JR, El-Moghraby A, Rehman M, Devlin SM, Corona M, Landego I, Lin RJ, Scordo M, Raj SS, Giralt SA, Palomba ML, Dahi PB, Walji M, Salles G, Nath K, Geyer MB, Park JH, Fein JA, Kosmidou I, Shah GL, Liu JE, Perales MA, and Mahmood SS
- Subjects
- Humans, Male, Female, Middle Aged, Risk Factors, Incidence, Aged, Retrospective Studies, Adult, Biomarkers blood, Lymphoma, Non-Hodgkin therapy, Lymphoma, Non-Hodgkin epidemiology, Lymphoma, Non-Hodgkin immunology, Receptors, Chimeric Antigen, Antigens, CD19 immunology, Immunotherapy, Adoptive adverse effects, Arrhythmias, Cardiac etiology, Arrhythmias, Cardiac therapy
- Abstract
Recent reports have raised concerns about the association of chimeric antigen receptor T cell (CAR-T) with non-negligible cardiotoxicity, particularly atrial arrhythmias. First, we conducted a pharmacovigilance study to assess the reporting of atrial arrhythmias following CD19-directed CAR-T. Subsequently, to determine the incidence, risk factors and outcomes of atrial arrhythmias post-CAR-T, we compiled a retrospective single-centre cohort of non-Hodgkin lymphoma patients. Only commercial CAR-T products were considered. Atrial arrhythmias were nearly fourfold more likely to be reported after CAR-T therapy compared to all other cancer patients in the FAERS (adjusted ROR = 3.76 [95% CI 2.67-5.29]). Of the 236 patients in our institutional cohort, 23 (10%) developed atrial arrhythmias post-CAR-T, including 12 de novo arrhythmias, with most (83%) requiring medical intervention. Atrial arrhythmias frequently co-occurred with cytokine release syndrome and were associated with higher post-CAR-T infusion peak levels of IL-10, TNF-alpha and LDH, and lower trough levels of fibrinogen. In a multivariable analysis, risk factors for atrial arrhythmia were history of atrial arrhythmia (OR = 6.80 [2.39-19.6]) and using CAR-T product with a CD28-costimulatory domain (OR = 5.17 [1.72-18.6]). Atrial arrhythmias following CD19-CAR-T therapy are prevalent and associated with elevated inflammatory biomarkers, a history of atrial arrhythmia and the use of a CAR-T product with a CD28 costimulatory domain., (© 2024 British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
29. Multi-parametric atlas of the pre-metastatic liver for prediction of metastatic outcome in early-stage pancreatic cancer.
- Author
-
Bojmar L, Zambirinis CP, Hernandez JM, Chakraborty J, Shaashua L, Kim J, Johnson KE, Hanna S, Askan G, Burman J, Ravichandran H, Zheng J, Jolissaint JS, Srouji R, Song Y, Choubey A, Kim HS, Cioffi M, van Beek E, Sigel C, Jessurun J, Velasco Riestra P, Blomstrand H, Jönsson C, Jönsson A, Lauritzen P, Buehring W, Ararso Y, Hernandez D, Vinagolu-Baur JP, Friedman M, Glidden C, Firmenich L, Lieberman G, Mejia DL, Nasar N, Mutvei AP, Paul DM, Bram Y, Costa-Silva B, Basturk O, Boudreau N, Zhang H, Matei IR, Hoshino A, Kelsen D, Sagi I, Scherz A, Scherz-Shouval R, Yarden Y, Oren M, Egeblad M, Lewis JS, Keshari K, Grandgenett PM, Hollingsworth MA, Rajasekhar VK, Healey JH, Björnsson B, Simeone DM, Tuveson DA, Iacobuzio-Donahue CA, Bromberg J, Vincent CT, O'Reilly EM, DeMatteo RP, Balachandran VP, D'Angelica MI, Kingham TP, Allen PJ, Simpson AL, Elemento O, Sandström P, Schwartz RE, Jarnagin WR, and Lyden D
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Biopsy, Neoplasm Staging, Pancreatectomy, Extracellular Traps metabolism, Prognosis, Pancreatic Neoplasms pathology, Pancreatic Neoplasms genetics, Pancreatic Neoplasms surgery, Liver Neoplasms secondary, Liver Neoplasms pathology, Liver Neoplasms genetics, Liver pathology, Liver metabolism
- Abstract
Metastasis occurs frequently after resection of pancreatic cancer (PaC). In this study, we hypothesized that multi-parametric analysis of pre-metastatic liver biopsies would classify patients according to their metastatic risk, timing and organ site. Liver biopsies obtained during pancreatectomy from 49 patients with localized PaC and 19 control patients with non-cancerous pancreatic lesions were analyzed, combining metabolomic, tissue and single-cell transcriptomics and multiplex imaging approaches. Patients were followed prospectively (median 3 years) and classified into four recurrence groups; early (<6 months after resection) or late (>6 months after resection) liver metastasis (LiM); extrahepatic metastasis (EHM); and disease-free survivors (no evidence of disease (NED)). Overall, PaC livers exhibited signs of augmented inflammation compared to controls. Enrichment of neutrophil extracellular traps (NETs), Ki-67 upregulation and decreased liver creatine significantly distinguished those with future metastasis from NED. Patients with future LiM were characterized by scant T cell lobular infiltration, less steatosis and higher levels of citrullinated H3 compared to patients who developed EHM, who had overexpression of interferon target genes (MX1 and NR1D1) and an increase of CD11B
+ natural killer (NK) cells. Upregulation of sortilin-1 and prominent NETs, together with the lack of T cells and a reduction in CD11B+ NK cells, differentiated patients with early-onset LiM from those with late-onset LiM. Liver profiles of NED closely resembled those of controls. Using the above parameters, a machine-learning-based model was developed that successfully predicted the metastatic outcome at the time of surgery with 78% accuracy. Therefore, multi-parametric profiling of liver biopsies at the time of PaC diagnosis may determine metastatic risk and organotropism and guide clinical stratification for optimal treatment selection., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2024
- Full Text
- View/download PDF
30. Development and validation of an automated computational approach to grade immune effector cell-associated hematotoxicity.
- Author
-
Liang EC, Rejeski K, Fei T, Albittar A, Huang JJ, Portuguese AJ, Wu Q, Raj S, Subklewe M, Shouval R, and Gauthier J
- Subjects
- Humans, Immunotherapy, Adoptive methods, Immunotherapy, Adoptive adverse effects
- Abstract
Hematologic toxicity frequently complicates chimeric antigen receptor (CAR) T-cell therapy, resulting in significant morbidity and mortality. In an effort to standardize reporting, the European Hematology Association (EHA) and European Society of Blood and Marrow Transplantation (EBMT) devised the immune effector cell-associated hematotoxicity (ICAHT) grading system, distinguishing between early (day 0-30) and late (after day +30) events based on neutropenia depth and duration. However, manual implementation of ICAHT grading criteria is time-consuming and susceptible to subjectivity and error. To address these challenges, we introduce a novel computational approach, utilizing the R programming language, to automate early and late ICAHT grading. Given the complexities of early ICAHT grading, we benchmarked our approach both manually and computationally in two independent cohorts totaling 1251 patients. Our computational approach offers significant implications by streamlining grading processes, reducing manual time and effort, and promoting standardization across varied clinical settings. We provide this tool to the scientific community alongside a comprehensive implementation guide, fostering its widespread adoption and enhancing reporting consistency for ICAHT., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
31. T-Cell Malignant Neoplasms After Chimeric Antigen Receptor T-Cell Therapy.
- Author
-
Storgard R, Rejeski K, Perales MA, Goldman A, and Shouval R
- Subjects
- Humans, T-Lymphocytes immunology, Male, Female, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods, Receptors, Chimeric Antigen immunology
- Published
- 2024
- Full Text
- View/download PDF
32. Conventional and novel [ 18 F]FDG PET/CT features as predictors of CAR-T cell therapy outcome in large B-cell lymphoma.
- Author
-
Leithner D, Flynn JR, Devlin SM, Mauguen A, Fei T, Zeng S, Zheng J, Imber BS, Hubbeling H, Mayerhoefer ME, Bedmutha A, Luttwak E, Corona M, Dahi PB, Luna de Abia A, Landego I, Lin RJ, Palomba ML, Scordo M, Park JH, Tomas AA, Salles G, Lafontaine D, Michaud L, Shah GL, Perales MA, Shouval R, and Schöder H
- Subjects
- Humans, Male, Female, Aged, Middle Aged, Adult, Treatment Outcome, Aged, 80 and over, Radiopharmaceuticals, Prognosis, Retrospective Studies, Fluorodeoxyglucose F18, Positron Emission Tomography Computed Tomography methods, Immunotherapy, Adoptive methods, Lymphoma, Large B-Cell, Diffuse therapy, Lymphoma, Large B-Cell, Diffuse diagnostic imaging
- Abstract
Relapse and toxicity limit the effectiveness of chimeric antigen receptor T-cell (CAR-T) therapy for large B-cell lymphoma (LBCL), yet biomarkers that predict outcomes and toxicity are lacking. We examined radiomic features extracted from pre-CAR-T
18 F-fluorodeoxyglucose positron emission tomography/computed tomography ([18 F]FDG PET/CT) scans (n = 341) of 180 patients (121 male; median age, 66 years). Three conventional (maximum standardized uptake value [SUVmax], metabolic tumor volume [MTV], total lesion glycolysis [TLG]) and 116 novel radiomic features were assessed, along with inflammatory markers, toxicities, and outcomes. At both pre-apheresis and pre-infusion time points, conventional PET features of disease correlated with elevated inflammatory markers. At pre-infusion, MTV was associated with grade ≥ 2 cytokine release syndrome (odds ratio [OR] for 100 mL increase: 1.08 [95% confidence interval (CI), 1.01-1.20], P = 0.031), and SUVmax was associated with failure to achieve complete response (CR) (OR 1.72 [95% CI, 1.24-2.43], P < 0.001). Higher pre-apheresis and pre-infusion MTV values were associated with shorter progression-free survival (PFS) (HR for 10-unit increase: 1.11 [95% CI, 1.05-1.17], P < 0.001; 1.04 [95% CI, 1.02-1.07], P < 0.001) and shorter overall survival (HR for 100-unit increase: 1.14 [95% CI, 1.07-1.21], P < 0.001; 1.04 [95% CI, 1.02-1.06], P < 0.001). A combined MTV and LDH measure stratified patients into high and low PFS risk groups. Multiple pre-infusion novel radiomic features were associated with CR. These quantitative conventional [18 F]FDG PET/CT features obtained before CAR-T cell infusion, which were correlated with inflammation markers, may provide prognostic biomarkers for CAR-T therapy efficacy and toxicity. The use of conventional and novel radiomic features may thus help identify high-risk patients for earlier interventions., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
33. Unlocking the Role of Age-Related Changes to Fibroblasts in Pancreatic Cancer.
- Author
-
Isaacson A, Barki D, and Scherz-Shouval R
- Subjects
- Humans, Aged, Pancreas, Fibroblasts, Signal Transduction, Pancreatic Neoplasms genetics, Carcinoma, Pancreatic Ductal genetics
- Abstract
Pancreatic cancer prevalence increases with age, and disease prognosis is poorer in older individuals. The increased prevalence is driven, undoubtedly, by the multistep accumulation of oncogenic mutations in cancer cells with age. However, fibroblasts are major constituents and key players in pancreatic cancer, and they too undergo age-related changes that may contribute to disease severity. In this issue of Cancer Research, Zabransky and colleagues set out to dissect the effect of age-related changes in pancreatic fibroblasts on pancreatic ductal adenocarcinoma growth and metastasis. They discovered that aged fibroblasts secrete GDF-15, which in turn activates AKT signaling and accelerates tumor progression. These findings provide a mechanistic role for aged fibroblasts in pancreatic cancer, underpinning the importance of normal physiologic processes in tumor progression. See related article by Zabransky et al., p. 1221., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
34. Loss of EIF4G2 mediates aggressiveness in distinct human endometrial cancer subpopulations with poor survival outcome in patients.
- Author
-
Meril S, Muhlbauer Avni M, Lior C, Bahlsen M, Olender T, Savidor A, Krausz J, Belhanes Peled H, Birisi H, David N, Bialik S, Scherz-Shouval R, Ben David Y, and Kimchi A
- Subjects
- Female, Humans, Proteomics, Cell Line, Eukaryotic Initiation Factor-4G genetics, Eukaryotic Initiation Factor-4G metabolism, Kinesins genetics, Endometrial Neoplasms genetics, Endometrial Neoplasms pathology
- Abstract
The non-canonical translation initiation factor EIF4G2 plays essential roles in cellular stress responses via translation of selective mRNA cohorts. Currently there is limited and conflicting information regarding its involvement in cancer development and progression. Here we assessed its role in endometrial cancer (EC), in a cohort of 280 EC patients across different types, grades, and stages, and found that low EIF4G2 expression highly correlated with poor overall- and recurrence-free survival in Grade 2 EC patients, monitored over a period of up to 12 years. To establish a causative connection between low EIF4G2 expression and cancer progression, we stably knocked-down EIF4G2 in two human EC cell lines in parallel. EIF4G2 depletion resulted in increased resistance to conventional therapies and increased the prevalence of molecular markers for aggressive cell subsets, altering their transcriptional and proteomic landscapes. Prominent among the proteins with decreased abundance were Kinesin-1 motor proteins, KIF5B and KLC1, 2, 3. Multiplexed imaging of the EC patient tumor cohort showed a correlation between decreased expression of the kinesin proteins, and poor survival in patients with tumors of certain grades and stages. These findings reveal potential novel biomarkers for Grade 2 EC with ramifications for patient stratification and therapeutic interventions., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
35. Fecal microbiota transplantation in capsules for the treatment of steroid refractory and steroid dependent acute graft vs. host disease: a pilot study.
- Author
-
Youngster I, Eshel A, Geva M, Danylesko I, Henig I, Zuckerman T, Fried S, Yerushalmi R, Shem-Tov N, Fein JA, Bomze D, Shimoni A, Koren O, Shouval R, and Nagler A
- Subjects
- Humans, Fecal Microbiota Transplantation adverse effects, Pilot Projects, Prospective Studies, Gastrointestinal Tract, Steroids, Hematopoietic Stem Cell Transplantation adverse effects, Graft vs Host Disease etiology
- Abstract
Acute graft-versus-host disease (aGvHD) is a serious complication of allogeneic hematopoietic stem-cell transplantation with limited treatment options. The gut microbiome plays a critical role in aGvHD pathogenesis. Fecal microbiota transplantation (FMT) has emerged as a potential therapeutic approach to restore gut microbial diversity. In this prospective pilot study, 21 patients with steroid-resistant or steroid-dependent lower gastrointestinal aGvHD received FMT in capsule form. At 28 days after the first FMT, the overall response rate was 52.4%, with 23.8% complete and 28.6% partial responses. However, sustained responses were infrequent, with only one patient remaining aGvHD-free long-term. FMT was generally well-tolerated. Microbiome analysis revealed dysbiosis in pre-FMT patient stool samples, with distinct microbial characteristics compared to donors. Following FMT, there was an increase in beneficial Clostridiales and a decrease in pathogenic Enterobacteriales. These findings highlight the potential of FMT as a treatment option for steroid-resistant aGvHD. Trial registration number NCT #03214289., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
36. Systematic evaluation of donor-KIR/recipient-HLA interactions in HLA-matched hematopoietic cell transplantation for AML.
- Author
-
Fein JA, Shouval R, Krieger E, Spellman SR, Wang T, Baldauf H, Fleischhauer K, Kröger N, Horowitz M, Maiers M, Miller JS, Mohty M, Nagler A, Weisdorf D, Malmberg KJ, Toor AA, Schetelig J, Romee R, and Koreth J
- Subjects
- Adult, Humans, Receptors, KIR genetics, Chronic Disease, Unrelated Donors, Recurrence, Hematopoietic Stem Cell Transplantation, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy
- Abstract
Abstract: In acute myeloid leukemia (AML), donor natural killer cell killer immunoglobulin-like receptors (KIR) and recipient HLA interactions may contribute to the graft-versus-leukemia effect of allogeneic hematopoietic cell transplantation (HCT). Analyses of individual KIR/HLA interactions, however, have yielded conflicting findings, and their importance in the HLA-matched unrelated donor (MUD) setting remains controversial. We systematically studied outcomes of individual donor-KIR/recipient-HLA interactions for HCT outcomes and empirically evaluated prevalent KIR genotypes for clinical benefit. Adult patients with AML (n = 2025) who received HCT with MUD grafts in complete remission reported to the Center for International Blood and Marrow Transplantation were evaluated. Only the donor-2DL2+/recipient-HLA-C1+ pair was associated with reduced relapse (hazard ratio [HR], 0.79; 95% confidence interval [CI], 0.67-0.93; P = .006) compared with donor-2DL2-/recipient-HLA-C1+ pair. However, no association was found when comparing HLA-C groups among KIR-2DL2+-graft recipients. We identified 9 prevalent donor KIR genotypes in our cohort and screened them for association with relapse risk. Genotype 5 (G5) in all recipients and G3 in Bw4+ recipients were associated with decreased relapse risk (HR, 0.52; 95% CI, 0.35-0.78; P = .002; and HR, 0.32; 95% CI, 0.14-0.72; P = .006; respectively) and G2 (HR 1.63, 95% CI, 1.15-2.29; P = .005) with increased relapse risk in C1-homozygous recipients, compared with other patients with the same ligand. However, we could not validate these findings in an external data set of 796 AML transplants from the German transplantation registry. Neither a systematic evaluation of known HLA-KIR interactions nor an empiric assessment of prevalent KIR genotypes demonstrated clinically actionable associations; therefore, these data do not support these KIR-driven strategies for MUD selection in AML., (© 2024 by The American Society of Hematology. 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
37. Longitudinal clinical data improve survival prediction after hematopoietic cell transplantation using machine learning.
- Author
-
Zhou Y, Smith J, Keerthi D, Li C, Sun Y, Mothi SS, Shyr DC, Spitzer B, Harris A, Chatterjee A, Chatterjee S, Shouval R, Naik S, Bertaina A, Boelens JJ, Triplett BM, Tang L, and Sharma A
- Subjects
- Young Adult, Humans, Child, Transplantation, Homologous adverse effects, Bayes Theorem, Retrospective Studies, Prognosis, Hematopoietic Stem Cell Transplantation adverse effects
- Abstract
Abstract: Serial prognostic evaluation after allogeneic hematopoietic cell transplantation (allo-HCT) might help identify patients at high risk of lethal organ dysfunction. Current prediction algorithms based on models that do not incorporate changes to patients' clinical condition after allo-HCT have limited predictive ability. We developed and validated a robust risk-prediction algorithm to predict short- and long-term survival after allo-HCT in pediatric patients that includes baseline biological variables and changes in the patients' clinical status after allo-HCT. The model was developed using clinical data from children and young adults treated at a single academic quaternary-care referral center. The model was created using a randomly split training data set (70% of the cohort), internally validated (remaining 30% of the cohort) and then externally validated on patient data from another tertiary-care referral center. Repeated clinical measurements performed from 30 days before allo-HCT to 30 days afterwards were extracted from the electronic medical record and incorporated into the model to predict survival at 100 days, 1 year, and 2 years after allo-HCT. Naïve-Bayes machine learning models incorporating longitudinal data were significantly better than models constructed from baseline variables alone at predicting whether patients would be alive or deceased at the given time points. This proof-of-concept study demonstrates that unlike traditional prognostic tools that use fixed variables for risk assessment, incorporating dynamic variability using clinical and laboratory data improves the prediction of mortality in patients undergoing allo-HCT., (© 2024 by The American Society of Hematology. 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
38. p53 deficient breast cancer cells reprogram preadipocytes toward tumor-protective immunomodulatory cells.
- Author
-
Hassin O, Sernik M, Seligman A, Vogel FCE, Wellenstein MD, Smollich J, Halperin C, Pirona AC, Toledano LN, Caballero CD, Schlicker L, Salame TM, Sarusi Portuguez A, Aylon Y, Scherz-Shouval R, Geiger T, de Visser KE, Schulze A, and Oren M
- Subjects
- Humans, Female, Genes, p53, Adipose Tissue metabolism, Adipocytes metabolism, Tumor Microenvironment genetics, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Breast Neoplasms pathology
- Abstract
The TP53 gene is mutated in approximately 30% of all breast cancer cases. Adipocytes and preadipocytes, which constitute a substantial fraction of the stroma of normal mammary tissue and breast tumors, undergo transcriptional, metabolic, and phenotypic reprogramming during breast cancer development and play an important role in tumor progression. We report here that p53 loss in breast cancer cells facilitates the reprogramming of preadipocytes, inducing them to acquire a unique transcriptional and metabolic program that combines impaired adipocytic differentiation with augmented cytokine expression. This, in turn, promotes the establishment of an inflammatory tumor microenvironment, including increased abundance of Ly6C+ and Ly6G+ myeloid cells and elevated expression of the immune checkpoint ligand PD-L1. We also describe a potential gain-of-function effect of common p53 missense mutations on the inflammatory reprogramming of preadipocytes. Altogether, our study implicates p53 deregulation in breast cancer cells as a driver of tumor-supportive adipose tissue reprogramming, expanding the network of non-cell autonomous mechanisms whereby p53 dysfunction may promote cancer. Further elucidation of the interplay between p53 and adipocytes within the tumor microenvironment may suggest effective therapeutic targets for the treatment of breast cancer patients., Competing Interests: Competing interests statement:K.E.d.V. is consultant for Macomics.
- Published
- 2023
- Full Text
- View/download PDF
39. Enhanced Feature Selection for Microbiome Data using FLORAL: Scalable Log-ratio Lasso Regression.
- Author
-
Fei T, Funnell T, Waters NR, Raj SS, Sadeghi K, Dai A, Miltiadous O, Shouval R, Lv M, Peled JU, Ponce DM, Perales MA, Gönen M, and van den Brink MRM
- Abstract
Identifying predictive biomarkers of patient outcomes from high-throughput microbiome data is of high interest, while existing computational methods do not satisfactorily account for complex survival endpoints, longitudinal samples, and taxa-specific sequencing biases. We present FLORAL (https://vdblab.github.io/FLORAL/), an open-source computational tool to perform scalable log-ratio lasso regression and microbial feature selection for continuous, binary, time-to-event, and competing risk outcomes, with compatibility of longitudinal microbiome data as time-dependent covariates. The proposed method adapts the augmented Lagrangian algorithm for a zero-sum constraint optimization problem while enabling a two-stage screening process for extended false-positive control. In extensive simulation and real-data analyses, FLORAL achieved consistently better false-positive control compared to other lasso-based approaches, and better sensitivity over popular differential abundance testing methods for datasets with smaller sample size. In a survival analysis in allogeneic hematopoietic-cell transplant, we further demonstrated considerable improvement by FLORAL in microbial feature selection by utilizing longitudinal microbiome data over only using baseline microbiome data., Competing Interests: Authors’ Disclosures J.U. Peled reports research funding, intellectual property fees, and travel reimbursement from Seres Therapeutics, and consulting fees from DaVolterra, CSL Behring, Crestone Inc, and from MaaT Pharma. He serves on an Advisory board of and holds equity in Postbiotics Plus Research. He has filed intellectual property applications related to the microbiome (reference numbers #62/843,849, #62/977,908, and #15/756,845). D.M. Ponce has served as advisory board member for Evive Biotechnology (Shanghai) Ltd (formerly Generon [Shanghai] Corporation Ltd), she served as advisory board member or consultant of Sanofi Corporation, CareDx, Ceramedix, Incyte, and receives research funding from Takeda Corporation and Incyte. M.-A. Perales reports honoraria from Adicet, Allovir, Caribou Biosciences, Celgene, Bristol-Myers Squibb, Equilium, Exevir, Incyte, Karyopharm, Kite/Gilead, Merck, Miltenyi Biotec, MorphoSys, Nektar Therapeutics, Novartis, Omeros, OrcaBio, Syncopation, VectivBio AG, and Vor Biopharma. He serves on DSMBs for Cidara Therapeutics, Medigene, and Sellas Life Sciences, and the scientific advisory board of NexImmune. He has ownership interests in NexImmune and Omeros. He has received institutional research support for clinical trials from Incyte, Kite/Gilead, Miltenyi Biotec, Nektar Therapeutics, and Novartis. M.R.M. van den Brink has received research support and stock options from Seres Therapeutics and stock options from Notch Therapeutics and Pluto Therapeutics; he has received royalties from Wolters Kluwer; has consulted, received honorarium from or participated in advisory boards for Seres Therapeutics, Vor Biopharma, Rheos Medicines, Frazier Healthcare Partners, Nektar Therapeutics, Notch Therapeutics, Ceramedix, Lygenesis, Pluto Therapeutics, GlaskoSmithKline, Da Volterra, Thymofox, Garuda, Novartis (Spouse), Synthekine (Spouse), Beigene (Spouse), Kite (Spouse); he has IP Licensing with Seres Therapeutics and Juno Therapeutics; and holds a fiduciary role on the Foundation Board of DKMS (a nonprofit organization). Memorial Sloan Kettering Cancer Center (MSK) has institutional financial interests relative to Seres Therapeutics.
- Published
- 2023
- Full Text
- View/download PDF
40. The sum of the parts: what we can and cannot learn from comorbidity scores in allogeneic transplantation.
- Author
-
Shouval R and Fein JA
- Subjects
- Humans, Transplantation, Homologous methods, Comorbidity, Retrospective Studies, Hematopoietic Stem Cell Transplantation methods
- Abstract
Allogeneic hematopoietic cell transplantation (alloHCT) requires the comprehensive evaluation of patients across multiple dimensions. Among the factors considered, comorbidities hold great significance in the pretransplant assessment. As many as 40% of alloHCT recipients will have a high burden of comorbidities in contemporary cohorts. To ensure a standardized evaluation, several comorbidity scores have been developed; however, they exhibit variations in properties and performance. This review examines the strengths and weaknesses associated with these comorbidity scores, critically appraising these models and proposing a framework for their application in considering the alloHCT candidate. Furthermore, we introduce the concept that comorbidities may have specific effects depending on the chosen transplantation approach and outline the findings of key studies that consider the impact of individual comorbidities on alloHCT outcomes. We suggest that a personalized transplantation approach should not rely solely on the overall burden of comorbidities but should also take into account the individual comorbidities themselves, along with other patient, disease, and transplantation-related factors., (Copyright © 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
41. Point-of-care anti-CD19 chimeric antigen receptor T-cell therapy for relapsed/refractory follicular lymphoma.
- Author
-
Fried S, Shkury E, Itzhaki O, Sdayoor I, Yerushalmi R, Shem-Tov N, Danylesko I, Jacoby E, Shouval R, Kedmi M, Marcus R, Nagler A, Shimoni A, and Avigdor A
- Subjects
- Humans, Point-of-Care Systems, Immunotherapy, Adoptive adverse effects, Cell- and Tissue-Based Therapy, Antigens, CD19, Receptors, Chimeric Antigen, Lymphoma, Follicular therapy, Lymphoma, Follicular drug therapy
- Abstract
Patients with relapsed/refractory follicular lymphoma (R/R-FL) often require multiple treatment lines. We performed a phase 1b/2 single-center clinical trial of autologous point-of-care anti-CD19 chimeric antigen receptor (CAR) T-cells in R/R-FL patients treated patients with ≥ 2 treatment lines. All 26 patients enrolled received CAR T-cell infusion at a median of 11 days after leukapheresis. Seventy-seven percent of patients had POD24. At enrollment, disease stage was III-IV in 85% of the patients, 77% had high-risk FLIPI score, and 77% had progressive disease. Grade III-IV cytokine release and immune effector cell-associated neurotoxicity syndromes occurred in 12% and 16% of the patients, respectively. Overall response rate at 1-month was 88%. The median follow-up was 15.4 months. One-year overall and progression-free survival were 100% and 63%, respectively. In conclusion, point-of-care CAR T-cell, manufactured within 11 days, induced a high response rate with an acceptable safety profile in patients with high-risk R/R-FL.
- Published
- 2023
- Full Text
- View/download PDF
42. The Simplified Comorbidity Index predicts non-relapse mortality in reduced-intensity conditioning allogeneic haematopoietic cell transplantation.
- Author
-
Elias S, Brown S, Devlin SM, Barker JN, Cho C, Chung DJ, Dahi PB, Giralt S, Gyurkocza B, Jakubowski AA, Lahoud OB, Landau H, Lin RJ, Papadopoulos EB, Politikos I, Ponce DM, Scordo M, Shaffer BC, Shah GL, Tamari R, Young JW, Perales MA, and Shouval R
- Subjects
- Humans, Comorbidity, Recurrence, Retrospective Studies, Transplantation Conditioning methods, Transplantation, Homologous methods, Mortality, Hematopoietic Stem Cell Transplantation methods, Tissue Donors
- Abstract
Comorbidity assessment before allogeneic haematopoietic cell transplantation (allo-HCT) is essential for estimating non-relapse mortality (NRM) risk. We previously developed the Simplified Comorbidity Index (SCI), which captures a small number of 'high-yield' comorbidities and older age. The SCI was predictive of NRM in myeloablative CD34-selected allo-HCT. Here, we evaluated the SCI in a single-centre cohort of 327 patients receiving reduced-intensity conditioning followed by unmanipulated allografts from HLA-matched donors. Among the SCI factors, age above 60, mild renal impairment, moderate pulmonary disease and cardiac disease were most frequent. SCI scores ranged from 0 to 8, with 39%, 20%, 20% and 21% having scores of 0-1, 2, 3 and ≥4 respectively. Corresponding cumulative incidences of 3-year NRM were 11%, 16%, 22% and 27%; p = 0.03. In multivariable models, higher SCI scores were associated with incremental risks of all-cause mortality and NRM. The SCI had an area under the receiver operating characteristic curve of 65.9%, 64.1% and 62.9% for predicting 1-, 2- and 3-year NRM versus 58.4%, 60.4% and 59.3% with the haematopoietic cell transplantation comorbidity index. These results demonstrate for the first time that the SCI is predictive of NRM in patients receiving allo-HCT from HLA-matched donors after reduced-intensity conditioning., (© 2023 British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
43. Clinical efficacy and safety of chimeric antigen receptor T-cell therapy for mantle cell lymphoma with secondary central nervous system involvement.
- Author
-
Ryan CE, Zon RL, Redd R, Fisher DC, Shouval R, Kumar A, Crombie JL, Sadrzadeh H, Kim AI, Nayak L, Chukwueke UN, Jacobson CA, Frigault MJ, Palomba ML, Armand P, Epstein-Peterson Z, and Merryman RW
- Subjects
- Adult, Humans, Immunotherapy, Adoptive adverse effects, Immunotherapy, Adoptive methods, Receptors, Antigen, T-Cell therapeutic use, T-Lymphocytes, Treatment Outcome, Antigens, CD19, Central Nervous System, Cell- and Tissue-Based Therapy, Lymphoma, Mantle-Cell drug therapy, Receptors, Chimeric Antigen therapeutic use, Neurotoxicity Syndromes drug therapy
- Abstract
Data describing outcomes of chimeric antigen receptor (CAR) T-cell therapy in patients with secondary central nervous system (SCNS) involvement of mantle cell lymphoma (MCL) are limited. We identified 10 patients with MCL and SCNS involvement treated with anti-CD19 CAR T-cell therapy at three US academic centres. Frequent objective responses were observed in the CNS (86%) and systemically (90%), and the 1-year progression-free survival was 47%. Seven patients developed immune-effector-cell-associated-neurotoxicity-syndrome (n = 2 Grade 1, n = 5 Grade 3). Our results suggest that anti-CD19 CAR T-cell therapy in this setting is feasible and additional data regarding neurotoxicity in this population may be warranted., (© 2023 British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
44. Breast Cancer-Secreted Factors Promote Lung Metastasis by Signaling Systemically to Induce a Fibrotic Premetastatic Niche.
- Author
-
Cohen N, Mundhe D, Deasy SK, Adler O, Ershaid N, Shami T, Levi-Galibov O, Wassermann R, Scherz-Shouval R, and Erez N
- Subjects
- Animals, Female, Mice, Humans, Fibrosis metabolism, Activins metabolism, Cancer-Associated Fibroblasts metabolism, Cancer-Associated Fibroblasts pathology, Cell Line, Tumor, Lung Neoplasms secondary, Lung Neoplasms metabolism, Lung Neoplasms pathology, Lung Neoplasms genetics, Breast Neoplasms pathology, Breast Neoplasms metabolism, Breast Neoplasms genetics, Tumor Microenvironment, Signal Transduction
- Abstract
Metastatic cancer is largely incurable and is the main cause of cancer-related deaths. The metastatic microenvironment facilitates formation of metastases. Cancer-associated fibroblasts (CAF) are crucial players in generating a hospitable metastatic niche by mediating an inflammatory microenvironment. Fibroblasts also play a central role in modifying the architecture and stiffness of the extracellular matrix (ECM). Resolving the early changes in the metastatic niche could help identify approaches to inhibit metastatic progression. Here, we demonstrate in mouse models of spontaneous breast cancer pulmonary metastasis that fibrotic changes and rewiring of lung fibroblasts occurred at premetastatic stages, suggesting systemic influence by the primary tumor. Activin A (ActA), a TGFβ superfamily member, was secreted from breast tumors and its levels in the blood were highly elevated in tumor-bearing mice. ActA upregulated the expression of profibrotic factors in lung fibroblasts, leading to enhanced collagen deposition in the lung premetastatic niche. ActA signaling was functionally important for lung metastasis, as genetic targeting of ActA in breast cancer cells significantly attenuated lung metastasis and improved survival. Moreover, high levels of ActA in human patients with breast cancer were associated with lung metastatic relapse and poor survival. This study uncovers a novel mechanism by which breast cancer cells systemically rewire the stromal microenvironment in the metastatic niche to facilitate pulmonary metastasis., Significance: ActA mediates cross-talk between breast cancer cells and cancer-associated fibroblasts in the lung metastatic niche that enhances fibrosis and metastasis, implicating ActA as a potential therapeutic target to inhibit metastatic relapse., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
45. Identifying an optimal fludarabine exposure for improved outcomes after axi-cel therapy for aggressive B-cell non-Hodgkin lymphoma.
- Author
-
Scordo M, Flynn JR, Gonen M, Devlin SM, Parascondola A, Tomas AA, Shouval R, Brower J, Porter DL, Schuster SJ, Bachanova V, Maakaron J, Maziarz RT, Chen AI, Nastoupil LJ, McGuirk JP, Oluwole OO, Ip A, Leslie LA, Bishop MR, Riedell PA, and Perales MA
- Subjects
- Adult, Humans, Recurrence, Immunotherapy, Adoptive adverse effects, Lymphoma, Non-Hodgkin drug therapy
- Abstract
Fludarabine is one of the most common agents given for lymphodepletion before CD19 chimeric antigen receptor T cells, but its optimal therapeutic intensity is unknown. Using data from a multicenter consortium, we estimated fludarabine exposure (area under the curve [AUC]) using a population pharmacokinetic (PK) model in 199 adult patients with aggressive B-cell non-Hodgkin lymphomas who received commercial axicabtagene ciloleucel (Axi-cel). We evaluated the association of estimated fludarabine AUC with key outcomes, aiming to find an AUC that optimized efficacy and tolerability. We identified low (<18 mg × hour/L [mgh/L]), optimal (18-20 mgh/L), and high (>20 mgh/L) AUC groups for analyses; the 6-month cumulative incidences of relapse/progression of disease (relapse/POD) by AUC groups were 54% (45%-62%), 28% (15%-44%), and 30% (14%-47%), respectively; and the 1-year progression-free survival (PFS) rates were 39% (31%-48%), 66% (52%-84%), and 46% (30%-70%) and the overall survival (OS) rates were 58% (50%-67%), 77% (64%-92%), and 66% (50%-87%), respectively. In multivariable analyses compared with low AUC, an optimal AUC was associated with the highest PFS (hazard ratio [HR], 0.52; 0.3-0.91; P = .02) and lowest risk of relapse/POD (HR, 0.46; 0.25-0.84; P = .01) without an increased risk of any-grade cytokine release syndrome (HR, 1.1; 0.7-1.6; P = .8) or and immune effector cell-associated neurotoxicity syndrome (ICANS) (HR, 1.36; 0.83-2.3; P = .2). A high AUC was associated with the greatest risk of any-grade ICANS (HR, 1.9; 1.1-3.2; P = .02). Although the main cause of death in all groups was relapse/POD, nonrelapse-related deaths, including 3 deaths from ICANS, were more frequent in the high AUC group. These findings suggest that PK-directed fludarabine dosing to achieve an optimal AUC may result in improved outcomes for patients receiving axi-cel., (© 2023 by The American Society of Hematology. 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
- 2023
- Full Text
- View/download PDF
46. The tumor microenvironment shows a hierarchy of cell-cell interactions dominated by fibroblasts.
- Author
-
Mayer S, Milo T, Isaacson A, Halperin C, Miyara S, Stein Y, Lior C, Pevsner-Fischer M, Tzahor E, Mayo A, Alon U, and Scherz-Shouval R
- Subjects
- Female, Humans, Fibroblasts, Biological Transport, Cell Communication, Tumor Microenvironment, Cancer-Associated Fibroblasts
- Abstract
The tumor microenvironment (TME) is comprised of non-malignant cells that interact with each other and with cancer cells, critically impacting cancer biology. The TME is complex, and understanding it requires simplifying approaches. Here we provide an experimental-mathematical approach to decompose the TME into small circuits of interacting cell types. We find, using female breast cancer single-cell-RNA-sequencing data, a hierarchical network of interactions, with cancer-associated fibroblasts (CAFs) at the top secreting factors primarily to tumor-associated macrophages (TAMs). This network is composed of repeating circuit motifs. We isolate the strongest two-cell circuit motif by culturing fibroblasts and macrophages in-vitro, and analyze their dynamics and transcriptomes. This isolated circuit recapitulates the hierarchy of in-vivo interactions, and enables testing the effect of ligand-receptor interactions on cell dynamics and function, as we demonstrate by identifying a mediator of CAF-TAM interactions - RARRES2, and its receptor CMKLR1. Thus, the complexity of the TME may be simplified by identifying small circuits, facilitating the development of strategies to modulate the TME., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
47. Learning from the microbes: exploiting the microbiome to enforce T cell immunotherapy.
- Author
-
Staudt S, Ziegler-Martin K, Visekruna A, Slingerland J, Shouval R, Hudecek M, van den Brink M, and Luu M
- Subjects
- Humans, T-Lymphocytes, Neoplasm Recurrence, Local, Immunotherapy, Tumor Microenvironment, Receptors, Chimeric Antigen genetics, Microbiota
- Abstract
The opportunities genetic engineering has created in the field of adoptive cellular therapy for cancer are accelerating the development of novel treatment strategies using chimeric antigen receptor (CAR) and T cell receptor (TCR) T cells. The great success in the context of hematologic malignancies has made especially CAR T cell therapy a promising approach capable of achieving long-lasting remission. However, the causalities involved in mediating resistance to treatment or relapse are still barely investigated. Research on T cell exhaustion and dysfunction has drawn attention to host-derived factors that define both the immune and tumor microenvironment (TME) crucially influencing efficacy and toxicity of cellular immunotherapy. The microbiome, as one of the most complex host factors, has become a central topic of investigations due to its ability to impact on health and disease. Recent findings support the hypothesis that commensal bacteria and particularly microbiota-derived metabolites educate and modulate host immunity and TME, thereby contributing to the response to cancer immunotherapy. Hence, the composition of microbial strains as well as their soluble messengers are considered to have predictive value regarding CAR T cell efficacy and toxicity. The diversity of mechanisms underlying both beneficial and detrimental effects of microbiota comprise various epigenetic, metabolic and signaling-related pathways that have the potential to be exploited for the improvement of CAR T cell function. In this review, we will discuss the recent findings in the field of microbiome-cancer interaction, especially with respect to new trajectories that commensal factors can offer to advance cellular immunotherapy., Competing Interests: ML, MH, and AV are inventors on a patent application related to the use of pentanoate that has been filed by Philipps-University Marburg and Julius-Maximilians University Würzburg WO2021/058811A1. MH is inventor on patent applications related to CAR T cell manufacturing. MH is listed as an inventor on patent applications and granted patents related to CAR-T technologies that have been filed by the Fred Hutchinson Cancer Research Center, Seattle, WA and by the University of Würzburg, Würzburg, Germany. MH is a co-founder and equity owner of T-CURX GmbH, Würzburg, Germany. MH received honoraria from Celgene/BMS, Janssen, Kite/Gilead. MvdB has received research support and stock options from Seres Therapeutics and stock options from Notch Therapeutics and Pluto Therapeutics; he has received royalties from Wolters Kluwer; has consulted, received honorarium from or participated in advisory boards for Seres Therapeutics, Rheos Medicines, Ceramedix, Pluto Therapeutics, Thymofox, Garuda, Novartis (Spouse), Synthekine (Spouse), Beigene (Spouse), Kite (Spouse); he has IP Licensing with Seres Therapeutics and Juno Therapeutics; and holds a fiduciary role on the Foundation Board of DKMS (a nonprofit organization). The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Staudt, Ziegler-Martin, Visekruna, Slingerland, Shouval, Hudecek, van den Brink and Luu.)
- Published
- 2023
- Full Text
- View/download PDF
48. Harnessing the Gut Microbiota to Potentiate the Efficacy of CAR T Cell Therapy.
- Author
-
Gabrielli G, Shouval R, Ghilardi G, van den Brink M, and Ruella M
- Abstract
Competing Interests: The authors have no conflicts of interest to disclose.
- Published
- 2023
- Full Text
- View/download PDF
49. Keeping IPMNs in Check: A Novel Role for the Transcription Factor NKX6-2 in Preserving an Indolent Cell Identity in Pancreatic Cystic Lesions.
- Author
-
Ben-Shmuel A and Scherz-Shouval R
- Subjects
- Animals, Transcriptome, Pancreas pathology, Cell Differentiation, Transcription Factors genetics, Pancreatic Intraductal Neoplasms, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal pathology, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Pancreatic Cyst genetics
- Abstract
Summary: In this issue of Cancer Discovery, Sans and colleagues identify the transcription factor NKX6-2 as a principal element in maintaining the low-grade gastric cell phenotype of intraductal papillary mucinous neoplasms (IPMN) in the pancreas. Their discoveries in patient cohorts and dissection in animal models provide a novel molecular understanding underpinning IPMN differentiation, with implications for risk stratification and therapeutic intervention in pancreatic cancer. See related article by Sans et al., p. 1844 (7)., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
50. Prospective geriatric assessment and geriatric consultation in CAR T-cell therapy for older patients with lymphoma.
- Author
-
Lin RJ, Kim SJ, Brown S, Elko TA, Ruiz JD, Hanley DM, Lia Palomba M, Perales MA, Shah GL, Dahi PB, Scordo M, Sauter CS, Batlevi CL, Tomas AA, Shouval R, Lee N, Pavkovic EA, Engstler DE, Park JH, Salles GA, Devlin SM, Korc-Grodzicki B, Hamlin PA, and Giralt SA
- Subjects
- Humans, Aged, Geriatric Assessment, Prospective Studies, Referral and Consultation, Immunotherapy, Adoptive adverse effects, Lymphoma
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.