Checkpoint Inhibitors Enter ‘Real-World’ Settings for Relapsed/Refractory HL


Programmed death-1 (PD-1) inhibitors can play an important role in the treatment of relapsed/refractory classical Hodgkin lymphoma (HL) patients in “real-world” settings.

“In HL patients who progress after prior cytotoxic therapy, immunotherapy in a real-world setting induces outcomes that are similar to in prior clinical trials,” Steven M. Bair, MD, of the Abramson Cancer Center of the University of Pennsylvania in Philadelphia, told MedPage Today. “For those who do progress, our study shows they can respond to conventional strategies, which could provide a bridge to transplant.”

Approximately 8,260 patients are diagnosed with HL in the United States each year, according to statistics from the Leukemia & Lymphoma Society. While HL is a highly curable malignancy, 20% of patients are not able to be cured with conventional treatments. PD-1 inhibition has resulted in impressive activity in the relapsed/refractory setting, leading to FDA approval of nivolumab in 2016 and pembrolizumab in 2017, but there has been limited information on experience with these agents in HL patients outside of clinical trials.

At the American Society of Hematology (ASH) annual meeting, Bair presented a multicenter, retrospective analysis of relapsed/refractory HL patients treated with PD-1 inhibitors in a real-world setting. The study included 42 patients, median age of 34, from seven centers in the United States. At diagnosis, 60% of the patients had stage III/IV disease, half had previously undergone an autologous stem cell transplant (SCT), and 7% had prior allogeneic SCT.

Patients received a median of four lines of therapy prior to the PD-1 inhibitor; 85% had ABVD (doxorubicin, bleomycin, vinblastine, dacarbazine)/AVD as frontline therapy, and 95% had brentuximab vendotin. Nivolumab was the initial PD-1 inhibitor; three-quarters of patients received a dose of 3 mg/kg every 2 weeks. Concurrent therapies with PD-1 inhibitors were used at some point in 19% of patients.

Almost two-thirds of patients (62%) achieved an overall response to nivolumab as assessed by the treating physician, with 45% achieving a complete response (CR). The median time to best response was 3 months.

At a median follow-up of 13 months at the time of the analysis, the median progression-free survival (PFS) and overall survival (OS) had not been reached. The 12-month OS rate was 96%, and the 12-month PFS rate was 65%. The median duration of therapy was 9 months. The median duration of response in patients with CR was 13 months, and for those in partial response, 9 months.

Dose delays occurred in one-third of patients — most commonly due to toxicity, non-adherence, and patient preference, Bair reported. Dose delays did not affect PFS: Half the patients discontinued the original PD-1 inhibitor, primarily due to disease progression. Four patients died — all due to disease progression.

Among the 40 patients for whom toxicity data were available, nearly half had toxicity attributed to the PD-1 inhibitor. The most common toxicities were hypothyroidism, rash, pneumonitis/dyspnea, and colitis. Toxicities resulting in drug discontinuation included encephalitis, colitis, and skin graft-versus-host disease. Toxicities were managed with systemic steroids, thyroid replacement, drug discontinuation, dose delay, or local steroid therapy. There were no treatment-related deaths.

In the 21 patients who discontinued PD-1 inhibitor therapy, two-thirds had subsequent systemic therapies and 12 were evaluable for response. A clinical benefit of systemic therapy was seen immediately after PD-1 inhibitor therapy in 75% of patients, with five responding to cytotoxic chemotherapies.

“Patients can respond after failing initial therapy,” Bair said. “The thought is that checkpoint inhibitors sensitize tumors to cytotoxic therapy.”

In another presentation at ASH17, French researchers hypothesized that anti-PD-1 therapy may increase sensitivity to chemotherapy given after anti-PD-1 therapy (sequential strategy) or in combination with anti-PD-1 therapy after an unsatisfactory response to immunotherapy (concomitant strategy). The retrospective analysis of 30 relapsed/refractory HL patients showed that patients with an unsatisfactory response or progressive disease with anti-PD-1 therapy had a new objective response with chemotherapy alone (61%) or chemotherapy in combination with anti-PD-1 therapy (90%). This response could make some patients eligible for allograft, the researchers suggested.

 In the Bair study, four patients underwent SCT after PD-1 inhibitor therapy (three autologous, one allogeneic). Four patients were treated with pembrolizumab-containing regimens after nivolumab, and one of these patients had a clinical response.

“This is the first multi-center retrospective analysis to describe the real-world experience with PD-1 inhibitors in HL patients in the United States. There was good concordance in a real-world setting with clinical trials in terms of overall response rate, survival, and the toxicity profile in general,” Bair said, noting that the patterns of discontinuation and dose delays did not affect outcomes.

“Systemic therapies after PD-1 inhibitor discontinuation appear to be active. Our results support the important role of PD-1 inhibitor therapy in relapsed/refractory HL and provide insight into practice patterns,” he continued. “When HL patients fail cytotoxic therapy, they have an option of salvage immunotherapy.”

Researchers are now exploring further regimens, sequences, and combinations with immunotherapy and cytotoxic therapy that could be a bridge to transplant, Bair noted. “We have a possible paradigm to treat HL that is progressing rapidly, and are also starting to see immunotherapy move into the frontline setting in HL.”

Brentuximab vedotin combined with ABVD or AVD for patients with newly diagnosed Hodgkin’s lymphoma: a phase 1, open-label, dose-escalation study.


Summary

Background

Roughly 70–80% of patients with advanced stage Hodgkin’s lymphoma are cured with various first-line and second-line treatments, including ABVD, BEACOPP, and stem-cell transplantation. Brentuximab vedotin has shown significant clinical activity, with a manageable safety profile, in patients with relapsed or refractory Hodgkin’s lymphoma. We aimed to assess the safety and early clinical efficacy of this drug as first-line treatment in combination with standard or modified-standard treatment in patients with previously untreated Hodgkin’s lymphoma.

Methods

We did a phase 1, open-label, dose-escalation safety study comparing brentuximab vedotin in combination with standard (ABVD) or a modified-standard (AVD) treatment. Patients were enrolled into the groups sequentially. Main entry criteria were newly diagnosed, treatment-naive, CD30-positive patients with Hodgkin’s lymphoma who had histologically confirmed stage IIA bulky disease or stage IIB–IV disease and an Eastern Cooperative Oncology Group performance status of two or less. Patients received doses of 0·6, 0·9, or 1·2 mg/kg brentuximab vedotin by intravenous infusion every 2 weeks with either ABVD (25 mg/m2 doxorubicin, 10 units/m2 bleomycin, 6 mg/m2 vinblastine, and 375 mg/m2 dacarbazine) or AVD (ABVD modified regimen without the inclusion of bleomycin) for up to six cycles. Our primary objectives were to assess the safety profile and establish the maximum tolerated dose (MTD) of brentuximab vedotin in combination with ABVD and AVD. The safety profile and MTD was assessed for the safety population. The study has completed and the final analysis is presented. This study was registered with ClinicalTrials.gov, number NCT01060904.

Findings

Between Jan 29, 2010, and Sept 17, 2012, 51 patients were enrolled and received at least one dose of brentuximab vedotin. The maximum tolerated dose of brentuximab vedotin when combined with ABVD or AVD was not exceeded at 1·2 mg/kg. 21 (95%) of 22 patients given brentuximab vedotin and ABVD achieved complete remission, as did 24 (96%) of 25 patients given brentuximab vedotin and AVD. Adverse events were generally grade 1 or 2; however, an unacceptable number of patients in the brentuximab vedotin and ABVD groups had pulmonary toxic effects (11 [44%] of 25), which exceeded the historical incidence for ABVD alone. No patients experienced pulmonary toxic effects when treated with brentuximab vedotin plus AVD. The most common grade 3 or worse events were neutropenia (20 [80%] of 25 patients in the brentuximab vedotin and ABVD group vs 20 [77%] of 26 patients in the brentuximab vedotin and AVD group), anaemia (five [20%] vs three [12%]), febrile neutropenia (five [20%] vs two [8%]), pulmonary toxic effects (six [24%] vs 0), syncope (three [12%] vstwo [8%]), dyspnoea (three [12%] vs one [4%]), pulmonary embolism (three [12%] vs 0), fatigue (one [4%] each), and leucopenia (one [4%] each). Serious events occured in 41% of all patients (14 [56%] in the brentuximab vedotin and ABVD group and seven [27%] in the brentuximab vedotin and AVD group). Serious events occurring in 10% of patients or more overall were febrile neutropenia (four [16%] in the brentuximab vedotin and ABVD group vs two [8%] in the brentuximab vedotin and AVD group), and, in the brentuximab vedotin and ABVD group only, pulmonary toxic effects (six [24%]).

Interpretation

Brentuximab vedotin should not be given with bleomycin in general or specifically as first-line therapy for patients with treatment naive, advanced stage Hodgkin’s lymphoma. 1·2 mg/kg brentuximab vedotin combined with AVD given every 2 weeks was generally well tolerated by patients. At present, a phase 3 trial comparing brentuximab vedotin plus AVD to ABVD alone is ongoing (ClinicalTrials.gov, number NCT01712490) and will formally assess whether brentuximab vedotin plus AVD might redefine therapy in treatment-naive patients with Hodgkin’s lymphoma.

Source:http://www.thelancet.com