December 03, 2016
5 min read
Save

Anti–CD22 CAR T-cell therapy induces responses in relapsed, refractory ALL

You've successfully added to your alerts. You will receive an email when new content is published.

Click Here to Manage Email Alerts

We were unable to process your request. Please try again later. If you continue to have this issue please contact customerservice@slackinc.com.

SAN DIEGO — Anti–CD22 chimeric antigen receptor T-cell therapy appeared active and safe for children and young adults with relapsed or refractory acute lymphoblastic leukemia, according to interim results of a first-in-human study presented at the ASH Annual Meeting and Exposition.

Perspective from Frederick Locke, MD

Researchers reported an 80% complete remission rate among patients treated at the two highest dose levels. Those responses occurred in patients who were naive to chimeric antigen receptor (CAR) therapy, as well as those who underwent prior treatment with anti–CD19 CAR therapy and achieved CD19–negative status.

Terry J. Fry

“This is the first successful salvage CAR therapy for CD19–negative B-ALL, and our preliminary experience suggests a potency that is at least comparable to the phase 1 experience with CD19 at the same dose level,” Terry J. Fry, MD, head of the hematologic malignancies section of the pediatric oncology branch at NCI’s Center for Cancer Research, said during a press conference.

Anti–CD19 CAR therapy has been highly effective for patients with relapsed or refractory ALL, inducing complete remission rates between 80% and 90% in clinical trials. However, some patients do not respond. Also, loss of CD19 expression occurs in a substantial number of patients, potentially limiting the therapeutic benefit.

CD22 — widely expressed on B-lineage leukemia and lymphomas — is a well-defined alternative target in B-ALL, Fry said.

Fry and colleagues developed an anti–CD22 CAR, and then evaluated the safety of administering escalating doses of these cells to children and young adults with relapsed or refractory CD22–positive B-cell malignancies.

The regimen’s antileukemia effects, a measurement of persistence of anti–CD22 CAR T cells and an evaluation of cytokine profiles served as secondary objectives.

Study participants underwent lymphodepleting chemotherapy on days –4, –3 and –2. The regimen consisted of three doses of fludarabine 25 mg/m2 and one dose of cyclophosphamide 900 mg/m2.

Cell infusion began on day 0. Researchers established three dose levels: 3 x 105 transduced T cells/kg (n = 6), 1 x 106 transduced T cells/kg (n = 9), and 3 x 106 transduced T cells/kg (n = 2).

Fry presented results from 16 patients (median age, 19.5 years; range, 7-30; male, n = 9), all of whom had relapsed or refractory ALL and had undergone at least one prior allogeneic hematopoietic stem cell transplant.

Eleven of these patients had undergone prior treatment with anti–CD19 CAR T-cell therapy and five had undergone treatment with blinatumomab (Blincyto, Amgen), a bispecific T-cell engaging antibody construct that links CD3–positive T cells with CD19–positive B cells. At the conclusion of these prior treatments, nine patients either were CD19 negative or had reduced CD19 expression.

Results of the anti–CD22 CAR therapy analysis showed one of six patients treated at the lowest dose attained remission. Eight of 10 participants treated at one of the higher dose levels — comparable to those used by CD19 CAR T-cell programs — achieved complete remission with no evidence of residual disease at 1 month after infusion.

Six of nine patients who achieved remission later relapsed, primarily due to declines in CD22 expression on the cells. At the time of reporting, three patients remained in remission, including one who had been in remission for more than 1 year.

Thirteen patients experienced cytokine release syndrome; however, all cases were mild and none exceeded grade 2. Fever and low blood pressure also were common. One patient died of sepsis that occurred after resolution of cytokine release syndrome.

Trial accrual is ongoing.

The results raise the question about whether anti–CD22 therapy is best used after relapse of initial CAR therapy, or if combined use of anti–CD19 and anti–CD22 CAR targeting approaches could be more effective.

“This may not be best used as a salvage therapy,” Fry said. “We are beginning to think about how this could potentially be included with CD19 in upfront CAR treatment.” – by Mark Leiser

 

Reference: Shah NN, et al. Abstract 650. Presented at: ASH Annual Meeting and Exposition; Dec. 3-6, 2016; San Diego.

 

Disclosure: The NCI funded this study. Fry reports no relevant financial disclosures. Other researchers report honoraria from and patents/royalties with Juno; equity ownership in Unum Therapeutics; and board/advisory committee roles with Adaptimmune, GlaxoSmithKline, Pfizer-Servier, Roche, Unum Therapeutics and Vor Pharmaceuticals.