Novel PD-1/PD-L1 Inhibitor Combo Promotes Antitumor Activity in Urothelial Carcinoma

Publication
Article
Supplements and Featured PublicationsESMO 2020 Congress: Focus in Genitourinary Cancers
Volume 1
Issue 1

The investigational PD-L1 inhibitor BGB-A333 in combination with the PD-1 inhibitor tislelizumab demonstrated marked antitumor activity, durable clinical responses, and a tolerable safety profile in patients with urothelial carcinoma.

Juan Martin-Liberal, MD, PhD

The investigational PD-L1 inhibitor BGB-A333 in combination with the PD-1 inhibitor tislelizumab demonstrated marked antitumor activity, durable clinical responses, and a tolerable safety profile in patients with urothelial carcinoma, according to findings from the phase 1/2 BGB-900-101 study that were presented during the 2020 ESMO Congress.1

In the overall patient population (n = 12) of the phase 2B dose-expansion portion of the study, the combination induced an overall response rate (ORR) of 42% (95% CI, 15.2%-72.3%), consisting of 3 complete responses (CRs), and 2 partial responses (PRs). Additionally, 4 patients achieved stable disease (SD), 2 patients had progressive disease, and 1 patient was not evaluable.

Among patients with PD-L1–high urothelial carcinoma, the ORR was 67% (95% CI, 22.3%-95.7%), with CRs in 2 patients, PRs in 2 patients, and SD in 2 patients. The ORR in patients with PD-L1–low disease was 17%.

The median duration of response was 9.1 months (95% CI, 6.0-9.6).

“Simultaneous PD-L1 and PD-1 blockade has been hypothesized to provide synergistic antitumor effects, as inhibitors may have distinct mechanisms of action,” said lead study author Juan Martin-Liberal, MD, PhD, a consultant medical oncologist in the Melanoma, Sarcoma, and Genitourinary Tumors Unit at the Institut Catala d’Oncologia Hospitalet, in Barcelona, Spain. “[Although] these differences should be taken cautiously given the sample size, these data have provided insights into combining tislelizumab, a clinical stage anti–PD-1 antibody, with anti–PD-L1 antibodies.”

In previous findings from the part 1A dose-escalation portion of the study, the recommended dose was determined to be 1350 mg of intravenous (IV) BGB-A333 and 200 mg of IV tislelizumab every 3 weeks.2

Patients who were eligible for the phase 2B portion of the study had to have locally advanced or metastatic urothelial carcinoma that had progressed after at least 1 platinum-containing regimen. These patients received the recommended dose for a median duration of 6.2 months. The median follow-up was 10 months.

Patients had a median of 69.5, and 92% of patients were male. Ten patients had received 1 prior line of systemic therapy.

PD-L1–high status was defined as PD-L1 staining on 25% or more of tumor or immune cells with the VENTANA SP263 assay.

Additionally, radiographic assessments were performed every 9 weeks during the first year of treatment and every 12 weeks thereafter.

Additional findings showed that disease control rates (DCRs) also varied based on PD-L1 status. In the overall patient population, the DCR was 75% (95% CI, 42.8%-94.5%). In PD-L1–high patients, the DCR was 100% (95% CI, 54.1%-100%) compared with 50% (95% CI, 11.8%-88.2%) in PD-L1–low patients.

The median progression-free survival (PFS) was 6.1 months in the overall population (95% CI, 1.9-11.0); a total of 10 patients (83.3%) experienced a PFS event. In the PD-L1–high subgroup, the median PFS was 10 months (95% CI, 4.0-11.0) compared with 4.1 months (95% CI, 1.2-11.5) in the PD-L1–low subgroup. Moreover, PFS events occurred in 4 (66.7%) and 6 (100%) patients, respectively.

Regarding safety across the entire study population (n = 39), 20 patients experienced any-grade treatment-related adverse effects (TRAEs). The most common TRAEs were fatigue (n = 5), maculopapular rash (n = 4), myalgia (n = 4), nausea (n = 4), pruritis (n = 3), asthenia (n = 2), back pain (n = 2), and diarrhea (n = 2). A total of 7 patients experienced a grade 3 or higher TRAE, including 1 report of fatigue and 1 report of maculopapular rash.

Immune-related AEs occurred in 2 patients who were enrolled in the phase 2B portion of the study and consisted of grade 3 endocrine disorders, grade 3 hypophysitis, grade 2 musculoskeletal and connective tissue disorder, and grade 2 myositis.

No patients experienced a fatal TRAEs.

Ultimately, the safety profile of the combination was consistent with previously reported findings across multiple tumor types, concluded Martin-Liberal.

References:

  1. Martin-Liberal H, Fong P, Moreno V, et al. BGB-A333, an anti-PD-L1 monoclonal antibody, in combination with tislelizumab in patients with urothelial carcinoma. Ann Oncol. 2020;31(4). Abstract: 535MO.
  2. Desai J, Voskoboynik M, Markman B, et al. Preliminary safety and efficacy data of BGB-A333, an anti-PD-L1 monoclonal antibody, alone and in combination with tislelizumab in patients with advanced solid tumors. Cancer Research. 2020;80(16). doi:10.1158/1538-7445.AM2020-CT253
Related Videos
Samer A. Srour, MB ChB, MS
Nizar M. Tannir, MD, FACP, professor; Ransom Horne, Jr. Professor for Cancer Research, Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center
Samer A. Srour, MB ChB, MS
Samer A. Srour, MB ChB, MS
Petros Grivas, MD, PhD, professor, Clinical Research Division, Fred Hutchinson Cancer Center; professor, Division of Hematology and Oncology, University of Washington (UW) School of Medicine; clinical director, Genitourinary Cancers Program, UW Medicine
A panel of 5 experts on renal cell carcinoma
Chandler H. Park, MD, an expert on renal cell carcinoma
Pasi A. Jänne, MD, PhD, discusses an exploratory analysis from the FLAURA2 trial of osimertinib plus chemotherapy in treatment-naive, EGFR-mutant NSCLC.
Eric S. Christenson, MD
Benjamin Garmezy, MD