scholarly journals CD19 and CD30 CAR T-Cell Immunotherapy for High-Risk Classical Hodgkin’s Lymphoma

2021 ◽  
Vol 10 ◽  
Author(s):  
YuanBo Xue ◽  
Xun Lai ◽  
RuiLei Li ◽  
ChunLei Ge ◽  
BaoZhen Zeng ◽  
...  

BackgroundIn clinical applications of CAR T-cell therapy, life-threatening adverse events including cytokine release syndrome and neurotoxicity can lead to treatment failure. Outcomes of patients treated with anti-CD30 CAR T- cell have been disappointing in relapsing/refractory (r/r) classical Hodgkin’s Lymphoma (cHL).MethodsIn order to understand the applicable population of multiple CAR T-cell therapy, we examined the expression of CD19, CD20, and CD30 by immunohistochemistry (IHC) in 38 paraffin-embedded specimens of cHL. In the past two years, we found only one patient with cHL who is eligible for combined anti-CD19 and CD30 CAR T-cell treatment. This patient’s baseline characteristics were prone to severe adverse events. We treated this patient with low doses and multiple infusions of anti-CD19 and CD30 CAR T-cell.ResultsThe positive expression of CD19+ + CD30+ in Reed-Sternberg (RS) cells is approximately 5.2% (2/38). The patient we treated with combined anti-CD19 and CD30 CAR T-cell did not experience severe adverse events related to CAR T-cell therapy and received long term progression-free survival (PFS).ConclusionFor high risk r/r cHL patients, low doses of CAR T-cell used over different days at different times might be safe and effective. More clinical trials are warranted for CD19 and CD30 CAR T-cell combination therapy.

2021 ◽  
Vol 39 (15_suppl) ◽  
pp. TPS8053-TPS8053
Author(s):  
Saad Z. Usmani ◽  
Jesus G. Berdeja ◽  
Anna Truppel-Hartmann ◽  
Yizhou Fei ◽  
Honeylet Wortman-Vayn ◽  
...  

TPS8053 Background: High-risk (R-ISS stage III) newly diagnosed multiple myeloma (NDMM) has a poor prognosis (median PFS, 29 mo), highlighting the need for novel disease-targeting approaches (Palumbo A, et al. J Clin Oncol 2015;33:2863-2869). Ide-cel, a BCMA-directed CAR T cell therapy, demonstrated deep, durable responses in heavily pretreated patients (pts) with relapsed and refractory MM (RRMM; Raje N, et al. N Engl J Med 2019;380:1726-1737; Munshi NC, et al. J Clin Oncol 2020;38[suppl 15]. Abstract 8503), including those with high-risk (R-ISS stage III) RRMM. In this population, earlier use of ide cel—when there may be more bone marrow reserve, more healthy peripheral blood mononuclear cells, a less compromised immune status, and less extensive disease to debulk before cell therapy—may result in improved outcomes vs standard therapies and offer an opportunity to replace transplant with CAR T cell therapy. Methods: KarMMa-4 (NCT04196491), a multicenter, open-label, phase 1, single-arm study, is currently evaluating ide-cel in pts with high-risk NDMM, defined as R-ISS stage III (ISS stage III [serum ß2 microglobulin ≥ 5.5 mg/L] and cytogenetic abnormalities t(4;14), del(17p), and/or t(14;16) by interphase FISH; or ISS stage III and serum LDH > ULN). Pts must have received ≤ 3 cycles of the induction regimens listed below, be aged ≥ 18 years, and have ECOG PS 0-1. Nonsecretory MM and CNS involvement are exclusion criteria. Pts can enroll between induction cycles 1 and 3. Permitted cycle 1 regimens are carfilzomib + lenalidomide (LEN) + dexamethasone (DEX) ± daratumumab (DARA; KRd ± DARA), LEN + bortezomib (BOR) + DEX ± DARA (RVd ± DARA), or cyclophosphamide + BOR + DEX (CyBorD). Induction cycles 2-4 are limited to KRd or RVd, with DEX omitted during cycle 3. Pts will undergo T cell collection via leukapheresis after cycle 3, and ide-cel will be manufactured during cycle 4. Stem cell collection for future use may be conducted after cycle 3 (following leukapheresis) or 4 (before lymphodepletion). Ide-cel is infused after 2 days of rest following lymphodepletion with 3 days of fludarabine 30 mg/m2 + cyclophosphamide 300 mg/m2. LEN-based maintenance may be provided upon bone marrow recovery or 90 days after ide-cel infusion, whichever is later. Dose-limiting toxicity and safety are the primary endpoints. Secondary endpoints include complete response (CR) rate and overall response rate, duration of response, time to CR, time to start of maintenance, feasibility of initiating maintenance, PFS, overall survival, and pharmacokinetics. Exploratory endpoints include LEN maintenance safety, minimal residual disease, immunogenicity and biomarkers. The starting ide-cel target dose is 450 × 106 CAR+ T cells, with dose escalation/de-escalation (150, 300, and 800 × 106 CAR+ T cells). Upon determination of optimal target dose, 12 pts will be enrolled in the dose-expansion phase. Clinical trial information: NCT04196491.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 18-19
Author(s):  
Saad Z. Usmani ◽  
Jesus G. Berdeja ◽  
Anna Truppel-Hartmann ◽  
Marie-Laure Casadebaig ◽  
Honeylet Wortman-Vayn ◽  
...  

BACKGROUND: Despite the introduction of novel agents, patients with high-risk (Revised International Staging System [R-ISS] stage III) newly diagnosed multiple myeloma (NDMM) have a poor prognosis (median progression-free survival [PFS], 29 months), highlighting the need for novel disease-targeting approaches (Palumbo A, et al. J Clin Oncol. 2015;33:2863-2869). The BCMA-directed CAR T cell therapy ide-cel demonstrated deep, durable responses in heavily pretreated patients with relapsed and refractory MM (RRMM; Raje N, et al. N Engl J Med. 2019;380:1726-1737; Munshi NC, et al. J Clin Oncol. 2020;38[suppl] [abstract 8503]), including those with high-risk (R-ISS stage III) RRMM. Earlier use in this population-where there is potentially more bone marrow reserve, more healthy peripheral blood mononuclear cells, a less compromised immune status, and less extensive disease to debulk before cell therapy-may result in improved outcomes and offer an opportunity to replace transplant with CAR T cell therapy. KarMMa-4 is a multicenter, open-label, phase 1, single-arm study of ide-cel in patients with high-risk NDMM (R-ISS stage III disease per International Myeloma Working Group criteria). STUDY DESIGN: KarMMa-4 is enrolling patients with NDMM who have high-risk disease, defined as R-ISS stage III (ISS stage III [serum b2 microglobulin ≥ 5.5 mg/L] and cytogenetic abnormalities t(4;14), del(17p), and/or t(14;16) by interphase fluorescence in situ hybridization; or ISS stage III and serum lactate dehydrogenase > upper limit of normal). Patients must have received ≤ 3 cycles of the induction regimens listed below, be aged ≥ 18 years, and have Eastern Cooperative Oncology Group performance status 0-1. Patients with nonsecretory myeloma or central nervous system involvement are excluded. Patients can enroll between cycle 1 and cycle 3 of induction. Permitted induction regimens for cycle 1 are carfilzomib + lenalidomide + dexamethasone ± daratumumab (KRd ± DARA), lenalidomide + bortezomib + dexamethasone ± daratumumab (RVd ± DARA), or cyclophosphamide + bortezomib + dexamethasone (CyBorD). Induction regimens for cycles 2-4 are limited to KRd or RVd, with dexamethasone omitted during cycle 3. All patients will undergo leukapheresis for T cell collection after cycle 3, and ide-cel will be manufactured during cycle 4 of induction. Stem cell collection for future use may be conducted after cycle 3 (following leukapheresis) or cycle 4 (before lymphodepletion). Ide-cel is infused after 2 days of rest following lymphodepletion with 3 days of fludarabine 30 mg/m2 + cyclophosphamide 300 mg/m2. Lenalidomide-based maintenance may be provided upon bone marrow recovery or 90 days after ide-cel infusion, whichever is later. The primary endpoints are dose-limiting toxicity and safety. Secondary endpoints include complete response rate and overall response rate, duration of response, time to complete response, time to start of maintenance, feasibility of initiating maintenance, PFS, overall survival, and pharmacokinetics. Exploratory endpoints include safety of lenalidomide maintenance therapy, minimal residual disease, immunogenicity and biomarkers. The starting ide-cel target dose is 450 × 106 CAR+ T cells, with dose escalation/de-escalation (150, 300, and 800 × 106 CAR+ T cells). After identification of the optimal target dose, 12 patients will be enrolled in the dose-expansion phase. KarMMa-4 is registered at ClinicalTrials.gov: NCT04196491. Disclosures Usmani: Celgene: Other; Abbvie: Consultancy; Sanofi: Consultancy, Honoraria, Research Funding; Takeda: Consultancy, Honoraria, Other: Speaking Fees, Research Funding; Janssen: Consultancy, Honoraria, Other: Speaking Fees, Research Funding; SkylineDX: Consultancy, Research Funding; Seattle Genetics: Consultancy, Research Funding; Merck: Consultancy, Research Funding; Incyte: Research Funding; Pharmacyclics: Research Funding; Array Biopharma: Research Funding; GSK: Consultancy, Research Funding; BMS, Celgene: Consultancy, Honoraria, Other: Speaking Fees, Research Funding; Amgen: Consultancy, Honoraria, Other: Speaking Fees, Research Funding. Berdeja:Servier: Consultancy; Poseida: Research Funding; Lilly: Research Funding; Legend: Consultancy; Kite Pharma: Consultancy; Kesios: Research Funding; Karyopharm: Consultancy; Prothena: Consultancy; Acetylon: Research Funding; Amgen: Consultancy, Research Funding; Abbvie: Research Funding; Vivolux: Research Funding; Novartis: Research Funding; Bluebird: Research Funding; Bioclinica: Consultancy; BMS: Consultancy, Research Funding; Celgene: Consultancy, Research Funding; Cellularity: Research Funding; Constellation: Research Funding; CRISPR Therapeutics: Consultancy, Research Funding; CURIS: Research Funding; EMD Sorono: Research Funding; Genentech, Inc.: Research Funding; Janssen: Consultancy, Research Funding; Glenmark: Research Funding; Teva: Research Funding; Takeda: Consultancy, Research Funding. Truppel-Hartmann:bluebird bio, Inc: Current Employment, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company); F. Hoffmann La Roche: Current equity holder in publicly-traded company, Ended employment in the past 24 months. Casadebaig:Bristol-Myers Squibb Company: Current Employment. Wortman-Vayn:Bristol-Myers Squibb Company: Current Employment. Shelat:Bristol-Myers Squibb Company: Current Employment. Novick:Bristol-Myers Squibb Company: Current Employment. Shah:GSK, Amgen, Indapta Therapeutics, Sanofi, BMS, CareDx, Kite, Karyopharm: Consultancy; BMS, Janssen, Bluebird Bio, Sutro Biopharma, Teneobio, Poseida, Nektar: Research Funding.


2021 ◽  
Vol 19 (11.5) ◽  
pp. 1331-1333
Author(s):  
Mazyar Shadman

Mantle cell lymphoma remains incurable despite recent treatment advances, and most patients experience relapsed/refractory disease. BTK inhibitors are the preferred choice in the relapsed setting, especially in patients with early relapse. For patients with high-risk features such as TP53 mutation, early referral for CAR T-cell therapy should be considered, even in those with stable disease on a BTK inhibitor. Patients without high-risk features may be monitored and initiate CAR T-cell therapy after clinical disease progression. CAR T-cell therapy is an effective treatment with high rate of complete remissions. For patients who do not achieve a complete remission 3 months after CAR-T therapy, bridging therapy with chemotherapy or targeted therapy agents and referral for allogeneic transplant are recommended.


Author(s):  
Ekaterina Vorozheikina ◽  
Magdalena Ruiz ◽  
Maria Leticia Solari ◽  
Dmitry Ostasevich ◽  
Luis Mendoza

Anti-CD19 chimeric antigen receptor (CAR) T-cells represent a novel immunotherapy that has shown remarkable success in the treatment of adult relapsed or refractory (R/R) B-cell non-Hodgkin's lymphoma, adult R/R mantle cell lymphoma, and R/R acute paediatric lymphoblastic leukaemia. One barrier to the widespread use of CAR T-cell therapy is toxicity, primarily cytokine release syndrome (CRS) with a variable grade of severity. The main manifestations of CRS are fever, hypotension, cytopenia, organ dysfunction among others. Neurological toxicities vary widely and range from headaches to encephalopathy. In addition, anti-CD19 CAR T-cell therapy provokes an array of less frequent events, such as coagulopathies, delayed cytopenia, and cardiovascular toxicities. In general, toxicities are usually reversible and resolve on their own in most cases, though severe cases may require intensive care and immunosuppressive therapy. Deaths due to CRS, neurologic toxicity and infectious complications have been reported, which highlights the gravity of these syndromes and the critical nature of appropriate intervention. In this paper, we look at all available FDA- and EMA-approved information about the pathophysiology, clinical manifestations, risk factor reviews of existing toxicity grading systems, current management strategies, and guidelines for anti-CD19 CAR T-cell toxicities. We also present new approaches, which are under investigation, to mitigate these adverse events.


Author(s):  
Marjorie E. Zettler ◽  
Bruce A. Feinberg ◽  
Eli G. Phillips Jr ◽  
Andrew J. Klink ◽  
Sonam Mehta ◽  
...  

2020 ◽  
Vol 4 (10) ◽  
pp. 2325-2338 ◽  
Author(s):  
Xian Zhang ◽  
Xin-an Lu ◽  
Junfang Yang ◽  
Gailing Zhang ◽  
Jingjing Li ◽  
...  

Abstract Anti-CD19 chimeric antigen receptor (CAR) T-cell therapy is effective in patients with advanced B-cell acute lymphoblastic leukemia (B-ALL). However, efficacy data is sparse in subgroups of patients with high-risk features such as BCR-ABL+, TP53 mutation, extramedullary disease (including central nervous system leukemia) or posttransplant relapse. It is also uncertain whether there is an added benefit of transplantation after anti-CD19 CAR T-cell therapy. We conducted a phase 1/2 study of 115 enrolled patients with CD19+ B-ALL. A total of 110 patients were successfully infused with anti-CD19 CAR T cells. In all, 93% of patients achieved a morphologic complete remission, and 87% became negative for minimal residual disease. Efficacy was seen across all subgroups. One-year leukemia-free survival (LFS) was 58%, and 1-year overall survival (OS) was 64% for the 110 patients. Seventy-five nonrandomly selected patients (73.5%) subsequently received an allogeneic hematopoietic stem cell transplant (allo-HSCT). LFS (76.9% vs 11.6%; P < .0001; 95% confidence interval [CI], 11.6-108.4) and OS (79.1% vs 32.0%; P < .0001; 95% CI, 0.02-0.22) were significantly better among patients who subsequently received allo-HSCT compared with those receiving CAR T-cell therapy alone. This was confirmed in multivariable analyses (hazard ratio, 16.546; 95% CI, 5.499-49.786). Another variate that correlated with worse outcomes was TP53 mutation (hazard ratio, 0.235; 95% CI, 0.089-0.619). There were no differences in complete remission rate, OS, or LFS between groups of patients age 2 to 14 years or age older than 14 years. Most patients had only mild cytokine release syndrome and neurotoxicity. Our data indicate that anti-CD19 CAR T-cell therapy is safe and effective in all B-ALL subgroups that have high-risk features. The benefit of a subsequent allo-HSCT requires confirmation because of nonrandom allocation. This trial was registered at www.clinicaltrials.gov as #NCT03173417.


2020 ◽  
Vol 20 (3) ◽  
pp. e118-e123 ◽  
Author(s):  
Swetha Kambhampati ◽  
Lissa Gray ◽  
Bita Fakhri ◽  
Mimi Lo ◽  
Khoan Vu ◽  
...  

Author(s):  
Ekaterina Vorozheikina ◽  
Magdalena Ruiz ◽  
Leticia Solari ◽  
Dmitry Ostasevich ◽  
Luis Mendoza

Anti-CD19 chimeric antigen receptor (CAR) T-cells represent a novel immunotherapy that has shown remarkable success in the treatment of adult relapsed or refractory (R/R) B-cell non-Hodgkin's lymphoma, adult R/R mantle cell lymphoma, and R/R acute paediatric lymphoblastic leukaemia. One barrier to the widespread use of CAR T-cell therapy is toxicity, primarily cytokine release syndrome (CRS) with a variable grade of severity. The main manifestations of CRS are fever, hypotension, cytopenia, organ dysfunction among others. Neurological toxicities vary widely and range from headaches to encephalopathy. In addition, anti-CD19 CAR T-cell therapy provokes an array of less frequent events, such as coagulopathies, delayed cytopenia, and cardiovascular toxicities. In general, toxicities are usually reversible and resolve on their own in most cases, though severe cases may require intensive care and immunosuppressive therapy. Deaths due to CRS, neurologic toxicity and infectious complications have been reported, which highlights the gravity of these syndromes and the critical nature of appropriate intervention. In this paper, we look at all available FDA- and EMA-approved information about the pathophysiology, clinical manifestations, risk factor reviews of existing toxicity grading systems, current management strategies, and guidelines for anti-CD19 CAR T-cell toxicities. We also present new approaches, which are under investigation, to mitigate these adverse events.


Sign in / Sign up

Export Citation Format

Share Document