ASH Clinical News May 2016 | Page 46

Literature Scan “Anti-CD19 CAR T cells can cause lasting remissions in a subset of patients with progressive B-cell malignancies after allogeneic stem cell transplantation,” James Kochenderfer, MD, investigator in the Experimental Transplantation and Immunology Branch of the National Institutes of Health and corresponding author on the study, told ASH Clinical News. “The fact that the CAR T cells caused remissions without chemotherapy clearly demonstrates the antimalignancy activity of the CAR T cells.” The phase I dose-escalation study included 20 patients treated between August 2010 and February 2015. Patients had measurable CD19+ B-cell malignancies including: • chronic lymphocytic leukemia (CLL; n=5) • diffuse large B-cell lymphoma (DLBCL; n=4) • mantle cell lymphoma (MCL; n=5) • Philadelphia chromosome (Ph)negative ALL (n=4) • Ph-positive ALL (n=1) • follicular lymphoma (FL) that had transformed to DLBCL (n=1) Patients had previously undergone human leukocyte antigen-matched sibling (n=13) or unrelated donor alloHCT (n=7). Chemotherapy and antibody therapies had to be stopped two weeks prior to CAR19 T-cell infusion. Disease stage was determined at least two weeks after the last therapy before CAR T-cell infusions, and patients were excluded if they had evidence of acute GVHD > grade 1 or moderate or severe chronic GVHD. Patients received a median of four post-transplant lines of therapy prior to enrollment (range = 1-24), and at least two months elapsed between the most recent DLI and CAR19 T-cell infusion. Subjects received a single infusion of CAR19 T cells obtained from the alloHCT donor’s T cells, and dose-escalation followed a standard, phase I, 3+3 design. Patients did not receive conditioning with chemotherapy prior to infusion. Following CAR19 T-cell therapy, the overall response rate (ORR) was 40 percent, with eight patients achieving remission: six CRs and two partial remissions. Patients with acute lymphocytic leukemia (ALL) experienced the highest response rate, with four of five patients obtaining minimal residual disease-negative CR. The longest ongoing CR was reached in a patient with chronic lymphocytic leukemia (30 months), and, of the two partial remissions, one Ibrutinib Plus Bendamustine and Rituximab Yields Positive Results for Patients With CLL/SLL Adding ibrutinib to a standard treatment regimen of bendamustine and rituximab for patients with relapsed/refractory chronic lymphocytic leukemia (CLL) or small lymphocytic lymphoma (SLL) led to significant improvements in progression-free survival (PFS) and overall response rate (ORR), according to a study led by Asher Chanan-Khan, MD, from the Mayo Clinic Cancer Center in Jacksonville, Florida, and published in Lancet Oncology. Safety profiles were similar between the three-drug regimen and the control arm, the authors noted, without added toxicities. “The results of this trial demonstrate that ibrutinib has added benefit beyond the current standard-of-care chemoimmunotherapy in previously treated patients with CLL or SLL,” Dr. Chanan-Khan and colleagues wrote. “Ibrutinib can be administered safely with bendamustine plus rituximab and represents an alternative option to traditional chemoimmunotherapy.” The international, double-blind, placebo-controlled, randomized, cross-over, phase III HELIOS trial included 578 adult patients enrolled at 133 sites in 21 countries in North America, Europe, Latin America, and Asia between September 2012 and January 2014. Patients were eligible for the study if they had: • Relapsed/refractory CLL or SLL following at least one previous line of therapy consisting of at least two cycles of a regimen containing chemotherapy • An Eastern Cooperative Oncology group performance status of 0-1 44 ASH Clinical News persisted for longer than 18 months. Eight patients had stable disease, with the longest duration of stable disease persisting for longer than 31 months following infusion. “Many of the patients who obtained remission after CAR19 T-cell infusions did not obtain remission after standard DLIs that contained higher T-cell doses, which demonstrates that in some cases, CAR19 T-cell infusion is superior to standard DLI at eradicating malignancy,” Dr. Brudno and colleagues observed. Peak blood CAR T-cell levels (measured by quantitative polymerase chain reaction) were higher among those who achieved remission compared with those who did not (p=0.001), “indicating that increasing the peak blood levels of CAR19 T cells in vivo is an important goal for future research,” the authors explained. Programmed cell death protein-1 (PD-1) expression, an inhibitory receptor expressed on T cells, was significantly elevated on CAR T cells after infusion, suggesting that administering PD-1 antagonists after CAR19 T-cell infusion might improve the rates of remission. The researchers reported no findings of new-onset acute GVHD following CAR T-cell infusion, despite 14 patients (70%) having had a history of GVHD sometime after alloHCT and prior to enrollment. GVHD occurred in only two patients on the trial: A case of mild chronic ocular GVHD developed approximately two years after CAR19 T-cell infusion and a case of slowly worsening chronic mild GVHD. The most commonly reported adverse events (AEs) included fever, tachycardia, and hypotension, which was similar to other trials assessing CAR T cells, the authors noted. Sixty percent of patients experienced grade 3/4 AEs, including an increase in serum creatine kinase in two patients that was associated with mu scle pain and weakness. Though the study is limited by its small sample size, Dr. Brudno and co-authors concluded that the findings “point toward a promising future when CAR T-cell therapy will be commonly used in transplant regimens to specifically target malignancy-associated antigens. CAR T cells could be administered as planned infusions along with or soon after stem cell infusions. Genetically targeted T cells will be an integral part of allogeneic transplant protocols to separate graft-versus-malignancy activity from GVHD.” Follow-up is also limited, and the study had no comparison group. REFERENCE Brudno JN, Somerville RP, Shi V, et al. Allogeneic T cells that express an anti-CD19 chimeric antigen receptor induce remissions of B-cell malignancies that progress after allogeneic hematopoietic stem-cell transplantation without causing graft-versus-host disease. J Clin Oncol. 2016;34:1112-21. • Lymph node disease >1.5 cm as measured by a computerized tomography (CT) scan • Absolute neutrophil count >1 x 109/L • Platelet count >50 x 109/L • Adequate liver and kidney function Patients were excluded from the study if they had del17p13 CLL or SLL; had received previous treatment with ibrutinib or other Bruton tyrosine kinase inhibitors; were refractory or relapsed within 24 months with a previous regimen containing bendamustine; received hematopoietic cell transplant; or had a history of a stroke, intracranial hemorrhage, or clinically significant cardiovascular disease within six months prior to the study. Eligible patients were randomized 1:1 to receive a four-week treatment regimen of: • bendamustine: 70 mg/m2 intravenously on days 2 and 3 in cycle 1 and days 1 and 2 in cycles 2-6 • rituximab: 375 mg/m2 on day 1 of cycle 1 and 500 mg/m2 on day 1 of cycles 2-6 • ibrutinib: 420 mg orally once daily (n=289) or placebo (n=289) “A large proportion of [these patients had] high-risk features such as unmutated IGHV status (81% of 519 patients), del11q (26%), and bulky disease (56%),” the authors added. “The median time between previous treatment and study treatment was 24 months or less in both treatment groups, indicating a population with poor prognosis.” Patients receiving placebo were allowed to crossover to the ibrutinib arm if disease progression was confirmed. In addition to PFS (the study’s primary endpoint), secondary endpoints of the study included overall survival (OS), ORR, proportion of patients with a negative response for minimal residual disease (MRD), and safety. The majority of patients in both the ibrutinib and placebo cohorts received the maximum six cycles of treatment (81% [n=235] and 77% [n=222], respectively). Patients in the ibrutinib cohort had a median of 14.7 months of treatment exposure compared with 12.8 May 2016