Sequential CD19- and CD22-Directed CAR T-Cell Therapy Improves Long-Term Remission in Relapsed/Refractory B-ALL

Article

Sequential administration of CAR T-cell therapy targeting both the CD19 and CD22 antigens demonstrated high complete remission rates and improved long-term survival in patients with relapsed/refractory B-cell acute lymphoblastic leukemia following allogeneic hematopoietic stem cell transplant.

blood cells

Sequential administration of CAR T-cell therapy targeting both the CD19 and CD22 antigens demonstrated high complete remission (CR) rates and improved long-term survival in patients with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL) following allogeneic hematopoietic stem cell transplant (HSCT), according to findings presented at the 2019 European Society for Blood and Marrow Transplantation Annual Meeting.

Thirty days after the first CAR T-cell infusion, 27 (84.4%) patients achieved complete remission (CR), and 100% of these patients were minimum residual disease (MRD) negative. Partial responses following treatment were observed in 4 (12.5%) patients with extramedullary disease (EMD).

“Sequentially combined CD19- and CD22-[directed] CAR T-cell therapies significantly improved outcomes with the rate of overall survival and event-free survival being 87.5% and 58.3%, respectively, at a median follow-up of 10 months,” said lead study author Shuangyou Liu, MD, in a presentation during the meeting.

CAR T-cell therapy targeting CD19 has shown significant efficacy in patients with relapsed/refractory B-ALL; however, long-term remission has not been achieved with single target CAR T-cell therapy, Liu explained.

In the study, Liu and investigators from Beijing Boren Hospital in Beijing, China evaluated whether sequentially applied CD19- and CD22-targeted CAR T cells could improve remission rates in patients with relapsed B-ALL after receiving HSCT. Thirty-two patients with B-ALL and both CD19 and CD22 expression on blast cells who relapsed following allogeneic HSCT were enrolled. Patients had no or minimal graft-versus-host disease (GVHD).

Recipient-derived T cells were collected and transfected by a lentiviral vector encoding the CAR composed of CD3ζ and 4-1BB. The CD19-targeted CAR T-cell product was humanized or murinized for patients previously treated with murinized or humanized CD19-targeted CAR T-cell therapy, respectively, and humanized CD22-directed CAR T-cell therapy was used for all transplanted patients.

Patients underwent fludarabine lymphodepletion with or without cyclophosphamide prior to each infusion. Treatment was evaluated on day 30 and every 1 to 2 months thereafter. MRD was evaluated by flow cytometry and quantitative polymerase chain reaction was used to detect fusion genes; EMD was assessed using PET-CT, CT, or MRI.

The median age was 22 (range, 2.3-55) years. Relapse was detected in the bone marrow in 21 (65.6%) patients, 4 (12.5%) patients had EMD, and 7 (21.9%) patients had both EMD and bone marrow involvement. Complex chromosomal aberrations were detected in 5 (15.6%) patients. Two patients had MLL-AF4+ fusion genes, 3 patients each had E2A-PBX1 and TEL-AML1 fusions genes, and 5 patients had BCR-ABL-1; of these, 3 patients had T3151 mutation. Eighteen (56.2%) patients accepted immediate salvage therapy when necessary.

Treatment failure to chemotherapy, donor lymphocyte infusion, interferon, and murinized CD19-directed CAR T-cell therapy had occurred in 18 patients.

All 32 patients were expected to receive sequentially applied CD19- and CD22-directed CAR T-cell therapies with an interval of 1.5 to 6 months between CAR-T treatments. Of the 32 patients, 26 (81.2%) patients first received CD19-directed CAR T-cell infusion at a median dose of 1.3 x 105 cells/kg (range, 04-9.2 x 105 cells/kg) and 6 (18.8%) patients first received CD22-directed CAR T-cell therapy at a median dose of 1.9 x 105 cells/kg (range, 0.55-6.6 x 105 cells/kg). Twenty-four (75%) patients received both CAR T-cell constructs.

“For B-ALL patients relapsing after allogeneic HSCT, single CD19- or CD22-[directed] infusion resulted in a high CR rate,” Liu remarked.

As previously mentioned, the CR rate was 84.4% and the PR rate was 12.5% 30 days after the first CAR T-cell treatment. One (3.1%) patient died of severe cytokine release syndrome (CRS) and acute hepatic GVHD. CRS was reported in 28 (87.5%) patients; of these, 24 CRS cases were grade 1/2 and 4 cases were grade 3/4. Eight (25%) patients had GVHD; of these, 5 patients had skin and oral involvement, 2 patients had severe acute hepatic GVHD, and one patient had extensive chronic GVHD.

Prior to the second CAR T-cell treatment, 21 patients had achieved CR with the first CAR T-cell infusion; of these, 18 maintained absence of MRD and 3 patients became positive for MRD before the second infusion. Of the 3 patients demonstrating PR after the first CAR T-cell treatment who underwent a second infusion, 2 patients achieved CR and 1 had PR at the day 30 evaluation. All patients receiving a second infusion had either no or grade 1 CRS and no GVHD was reported.

Survival outcomes were improved among the 24 patients who received both CD19- and CD22-directed CAR T-cell infusions. At a median follow-up of 10 months (range, 4.5-15), 21 (87.5%) patients were still alive. No median OS had been calculated; however, 75% of patients demonstrated a 13-month OS. The EFS rate was 58.3% and the median EFS was 12.5 months.

At the time of follow-up, 7 patients had relapsed, 3 of which died of disease progression. Fourteen patients maintained MRD-negative EFS. One patient died of extensive chronic GVHD, 1 patient withdrew from the study, and 1 suffered severe neurologic damage. The remaining 4 patients await the second CAR T-cell infusion.

Liu cautioned that CAR T-cell induced GVHD should be cautiously monitored and that the effect of sequential CAR T-cell therapy was “not good” in patients with multiple EMD.

Liu S, Deng B, Lin Y, et al. Sequential CD19- and CD22-CART cell therapies for relapsed B-cell acute lymphoblastic leukemia after allogeneic hematopoietic stem cell transplantation: 10-month median follow-up. Presented at: 2019 European Society for Blood and Marrow Transplantation Annual Meeting; March 24 to 27, 2019; Frankfurt, Germany. Abstract GS2-4.

Related Videos
Omid Hamid, MD
Paula Cannon, PhD, the president elect of ASGCT and a distinguished professor of microbiology at Keck School of Medicine of USC
George Tachas, PhD
Alexandra Gomez-Arteaga, MD
Pietro Genovese, PhD, the principal investigator at the Gene Therapy Program of Dana-Farber/Boston Children’s Cancer and Blood Disorder Center
Akshay Sharma, MBBS, a bone marrow transplant physician at St. Jude Children’s Research Hospital
M. Peter Marinkovich, MD, on Bringing RDEB Treatment to the Local Level
Caspian Oliai, MD, MS, the medical director of the UCLA Bone Marrow Transplantation Stem Cell Processing Center
© 2024 MJH Life Sciences

All rights reserved.