Brexucabtagene Autoleucel Approved for Refractory B-Cell Acute Lymphoblastic Leukemia

Article

ZUMA-3 showed that a single infusion of the CAR T-cell therapy elicited a high and durable response rate in heavily pretreated patients with relapsed/refractory B-ALL.

This content originally appeared on our sister site, OncLive.

Brexucabtagene autoleucel (Tecartus; Gilead) has been approved by the FDA as a treatment for adults with relapsed/refractory B-cell precursor acute lymphoblastic leukemia (B-ALL).1 This is the first approval of a CAR T-cell therapy for this population.

The approval is based on results from the phase 1/2 ZUMA-3 study (NCT02614066) which showed that a single infusion of the CAR T-cell therapy elicited a high and durable response rate in heavily pretreated patients with relapsed/refractory B-ALL, the majority of whom had a high disease burden, at a median follow-up of 16.4 months (range, 10.3-22.1).2

Specifically, data from the phase 2 portion of the trial presented during the 2021 ASCO Annual Meeting showed that brexucabtagene autoleucel elicited a complete response/complete response with incomplete blood count recovery (CR/CRi) rate of 70.9%, which included a CR rate of 56.4%.

The international, multicenter, phase 1/2 trial enrolled patients with relapsed/refractory B-ALL and bone marrow blasts that were greater than 5% who were at least 18 years of age. Notably, patients were not able to have previously received blinatumomab (Blincyto).

READ MORE: CAR T-Cell Therapy Axi-Cel Seeks Expanded Indication for Second-Line LBC Lymphoma

Earlier results from the phase 1 portion of the research showed that brexucabtagene autoleucel had a manageable toxicity profile with an overall CR/CRi rate of 83%. The recommended phase 2 dose of the CAR T-cell product was determined to be 1 x 106 CAR T cells/kg.3

Seventy-one adult patients were enrolled to the phase 2 portion of the trial and they received conditioning chemotherapy comprised of fludarabine 25 mg/m2 on days -4, -3, and -2, as well as cyclophosphamide at 900 mg/m2 on day -2. A single infusion of the CAR T-cell therapy was administered on day 0 at a dose of 1 x 106 anti-CD19 CAR T cells/kg.

The primary end point of the trial was CR/CRi rate by central assessment, and key secondary end points included minimal residual disease (MRD) negativity rate, duration of response (DOR), relapse-free survival (RFS), overall survival, safety, and CAR T-cell levels in the blood and cytokine levels in serum.

Of the 71 patients enrolled to the trial, 14 patients did not receive treatment because of toxicities (n = 7), eligibility criteria were not met (n = 3), product was not available (n = 1), patient consent was withdrawn (n = 1), or for other reasons not specified (n = 2). A total of 57 patients were given conditioning chemotherapy; 2 of these patients did not receive treatment after this, which left 55 patients who received the CAR T-cell therapy.

Among the study participants, the median age was 40 years (range, 19-84), and the majority were male (60%) with an ECOG performance status of 1 (71%). Additionally, 27% of patients had Philadelphia chromosome positivity and all had central nervous system disease at baseline. Patients received a median of 2 prior therapies (range, 1-8) and 47% of patients had undergone 3 or more prior lines of treatment.

The majority of patients has relapsed on, or were refractory to, 2 or more prior systemic lines of treatment (78%); 45% of patients had received prior treatment with blinatumomab, 22% previously received inotuzumab ozogamicin (Besponsa), and 42% had undergone prior autologous stem cell transplant (ASCT). Moreover, the median rate of bone marrow blasts at screening was 65% (range, 5%-100%), and the median rate at preconditioning after bridging chemotherapy was 59% (range, 0%-98%).

Moreover, the median time to initial CR/CRi was 1.1 months (range, 0.85-2.99). In responders, the MRD negativity rate with the CAR T-cell therapy was 97%, with a sample unavailable for 1 patient. Eighteen percent of patients received ASCT at a median of 98 days (range, 60-207) after having received the CAR T product.

The median DOR was 12.8 months, with or without censoring patients at subsequent ASCT. As of data cutoff, 31% of the patients who achieved a CR or CRi experienced ongoing remissions without receiving subsequent ASCT. Additionally, the median RFS was 11.6 months and 14.2 months for those who achieved a CR/CRi.

Regarding safety, the most common grade 3 or higher adverse effects (AEs) included anemia (49%) and pyrexia (36%). Ten cases of grade 5 AEs were reported, 4 of which were ALL; 2 were treatment-emergent from the CAR T-cell product and included brain herniation and septic shock; 3 occurred after initiation of another cancer treatment and included fungal pneumonia, sepsis, and respiratory failure; and 1 was pneumonia unrelated to treatment with brexucabtagene autoleucel.

No grade 5 cytokine release syndrome (CRS) or other neurological AEs were reported. However, 24% of patients experienced grade 3 or higher CRS and 25% reported other grade 3 or higher neurological events. To treat these AEs, patients were given tocilizumab (Actemra; 80%), steroids (75%), and vasopressors (40%).

References
1. U.S. FDA Approves Kite’s Tecartus® as the First and Only Car T for Adults With Relapsed or Refractory B-cell Acute Lymphoblastic Leukemia. Kite. News release. Published October 1, 2021. Accessed October 1, 2021. https://bwnews.pr/3iq0Chg.
2. Shah B, Ghobadi A, Oluwole O, et al. Phase 2 results of the ZUMA-3 study evaluating KTE-X19, an anti-CD19 chimeric antigen receptor T-cell therapy, in adult patients with relapsed/refractory B-cell acute lymphoblastic leukemia. J Clin Oncol. 2021;39(15):7002-7002. doi:10.1200/JCO.2021.39.15_suppl.7002
3. Shah BD, Bishop MR, Oluwole OO, et al. End of phase I results of ZUMA-3, a phase 1/2 study of KTE-X19, anti-CD19 chimeric antigen receptor (CAR) T cell therapy, in adult patients (pts) with relapsed/refractory (R/R) acute lymphoblastic leukemia (ALL). J Clin Oncol. 2019;37(suppl; abstr 70060). doi:10.1200/JCO.2019.37.15_suppl.7006
Related Videos
Genovefa (Zenia) Papanicolaou, MD, an infectious diseases specialist at Memorial Sloan Kettering Cancer Center
Akshay Sharma, MBBS, a bone marrow transplant physician at St. Jude Children’s Research Hospital
John DiPersio, MD, PhD, the director of the Center for Gene and Cellular Immunotherapy at Washington University School of Medicine
Aude Chapuis, MD, an associate professor in the Translational Science and Therapeutics Division at Fred Hutch Cancer Center
Amar Kelkar, MD, a stem cell transplantation physician at the Dana-Farber Cancer Institute
David Porter, MD
© 2024 MJH Life Sciences

All rights reserved.