Read more

September 27, 2019
4 min read
Save

Atezolizumab extends OS for PD-L1-selected patients with non-small cell lung cancer

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.

David R. Spigel, MD
David R. Spigel

BARCELONA, Spain — Atezolizumab significantly prolonged OS compared with platinum-based chemotherapy for patients with newly diagnosed non-small cell lung cancer whose tumors expressed high levels of PD-L1, according to an interim survival analysis from the phase 3 IMpower110 study presented at European Society for Medical Oncology Congress.

Perspective from Naiyer A. Rizvi, MD, PhD

Safety outcomes also favored atezolizumab (Tecentriq, Genentech).

“Atezolizumab represents a promising first-line treatment option [for this patient population],” David R. Spigel, MD, chief scientific officer and lung cancer program director at Sarah Cannon Research Institute, said during his presentation.

Checkpoint inhibitors that target PD-1/PD-L1 — either alone or in combination with platinum-based doublet chemotherapy — can be used as first-line therapy for patients with metastatic NSCLC. PD-L1 expression typically informs the selection of the most appropriate agents.

Monotherapy with checkpoint inhibitors remains a viable treatment approach for patients who are not eligible for combination therapy, according to study background.

Spigel and colleagues conducted IMpower110 to evaluate atezolizumab — a monoclonal antibody designed to bind with PD-L1 — as first-line monotherapy for PD-L1-selected patients independent of tumor histology.

The trial included 572 chemotherapy-naive patients with stage IV nonsquamous or squamous NSCLC. All patients had PD-L1 expression of at least 1% on tumor cells (TC) or tumor-infiltrating immune cells (IC); measurable disease; and ECOG performance status of 0 or 1. No patients included in this analysis had ALK or EGFR mutations.

Researchers randomly assigned half of the patients to 1,200 mg IV atezolizumab every 3 weeks. Treatment continued until progressive disease or loss of clinical benefit.

The other half received platinum-based chemotherapy administered in four or six 21-day cycles. Patients with nonsquamous histology received cisplatin 75 mg/m2, or carboplatin area under the curve 6 plus pemetrexed 500 mg/m2 via IV every 3 weeks. Patients with squamous histology received cisplatin 75 mg/m2 plus gemcitabine 1,250 mg/m2, or carboplatin area under the curve 5 plus gemcitabine 1,000 mg/m2 via IV every 3 weeks.

Chemotherapy continued until progressive disease.

Investigators stratified patients by sex, ECOG performance status, histology and tumor PD-L1 status.

OS tested hierarchically served as the primary endpoint.

The primary efficacy populations included 554 patients classified as TC1/2/3 or IC1/2/3 (PD-L1 expression of 1% or greater by TC or IC); 328 patients classified as TC2/3 or IC2/3 (PD-L1 expression of 5% or greater by TC or IC); and 205 patients classified as TC3 or IC3 (PD-L1 expression of 50% or greater by TC, or 10% or greater by IC).

PAGE BREAK

Median follow-up was 15.7 months.

Results showed atezolizumab significantly improved median OS in the TC3 or IC3 group (20.2 months vs. 13.1 months; HR = 0.59; 95% CI, 0.4-0.89). This benefit persisted in all key subgroups.

Atezolizumab conferred an OS benefit in the TC2/3 or IC2/3 group (18.2 months vs. 14.9 months; HR = 0.72; 95% CI, 0.52-0.99), but it did not cross the prespecified testing boundary. Consequently, the TC1/2/3 or IC1/2/3 group was not formally tested.

A higher percentage of patients assigned chemotherapy received subsequent cancer therapies (49.5% vs. 29.6%).

Atezolizumab’s safety profile appeared consistent with prior observations, and no new safety signals were identified.

The safety analysis included 286 patients assigned atezolizumab and 263 patients assigned chemotherapy. Patients assigned chemotherapy were more likely to experience any treatment-related adverse events (85.2% vs. 60.5%), grade 3/grade 4 treatment-related adverse events (44.1% vs. 12.9%), grade 5 treatment-related serious adverse events (0.4% vs. 0%), and adverse events that led to treatment withdrawal (16.3% vs. 6.3%).

“IMpower110 will continue to the OS final analysis,” Spigel said. “Importantly, additional biomarker analyses will be presented at a future congress, including ongoing work with other assays to measure PD-1 expression ... as well as tumor mutation burden data.”– by Mark Leiser

Reference: Spigel D, et al. Abstract LBA78. Presented at: European Society for Medical Oncology Congress; Sept. 27-Oct. 1, 2019; Barcelona, Spain.

Disclosures: F. Hoffmann-La Roche funded this study. Spigel reports research funding to his institution from, consultant/advisory roles with payments to his institution from, or travel, accommodations or expenses from AbbVie, Boehringer Ingelheim, Bristol-Myers Squibb, Celgene, Eli Lilly, Genentech/Roche, Novartis, Pfizer, Takeda and several other pharmaceutical companies. Please see the abstract for all other authors’ relevant financial disclosures.