Frontline Pembrolizumab Plus Trastuzumab/Chemotherapy Improves PFS in HER2+ Gastric/GEJ Cancer

News
Article

The addition of pembrolizumab to combination of trastuzumab and chemotherapy led to an improvement in progression-free survival vs placebo plus trastuzumab and chemotherapy in the first-line treatment of patients with metastatic HER2-positive gastric or gastroesophageal junction cancer, particularly in those whose tumors had a PD-L1 combined positive score.

Yelena Y. Janjigian, MD

Yelena Y. Janjigian, MD

The addition of pembrolizumab (Keytruda) to combination of trastuzumab (Herceptin) and chemotherapy led to an improvement in progression-free survival (PFS) vs placebo plus trastuzumab and chemotherapy in the first-line treatment of patients with metastatic HER2-positive gastric or gastroesophageal junction (GEJ) cancer, particularly in those whose tumors had a PD-L1 combined positive score (CPS) of 1 or more, according to updated data from the phase 3 KEYNOTE-811 trial (NCT03615326).1,2

Findings presented at the 2023 ESMO Congress and simultaneously published in The Lancet showed at the second interim analysis conducted at a median follow-up of 28.3 months (interquartile range [IQR], 19.4-34.3) for the pembrolizumab arm and 28.5 months (IQR, 20.1-34.3) for the placebo arm, patients treated with the pembrolizumab regimen (n = 350) experienced a median PFS of 10.0 months (95% CI, 8.6-11.7) vs 8.1 months (95% CI, 7.0-8.5) for those given the placebo regimen (n = 348; HR, 0.72; 95% CI, 0.60-0.87; P = .0002). In patients with a PD-L1 CPS of 1 or more, the median PFS was 10.8 months (95% CI, 8.5-12.5) for the pembrolizumab arm (n = 298) and 7.2 months (95% CI, 6.8-8.4) for the placebo arm (n = 296; HR, 0.70; 95% CI, 0.58-0.85).

Furthermore, findings from the third interim analysis at a median follow-up 38.4 months (IQR, 29.5-44.4) in the pembrolizumab group and 38.6 months (IQR, 30.2-44.4) in the placebo group, the median PFS was 10.0 months (95% CI, 8.6-12.2) vs 8.1 months (95% CI, 7.1-8.6) for the overall populations in the pembrolizumab and placebo arms, respectively (HR, 0.73; 95% CI, 0.61-0.87). In patients with a PD-L1 CPS of at least 1, the median PFS was 10.9 months (95% CI, 8.5-12.5) for the pembrolizumab arm vs 7.3 months (95% CI, 6.8-8.5) for the placebo arm (HR, 0.71; 95% CI, 0.59-0.86).

Additional data from the third interim analysis showed that the pembrolizumab regimen elicited a median overall survival (OS) of 20.0 months (95% CI, 17.8-22.1) vs 16.8 months (95% CI, 15.0-18.7) for the placebo regimen in the overall population (HR, 0.84; 95% CI, 0.70-1.01). The median OS in patients with a PD-L1 CPS of 1 or more was 20.0 months (95% CI, 17.9-22.7) vs 15.7 months (95% CI, 13.5-18.5) for the pembrolizumab and placebo arms, respectively (HR, 0.81; 95% CI, 0.67-0.98).

“The addition of pembrolizumab to first-line trastuzumab and chemotherapy led to meaningful improvement in PFS and overall response rate [ORR], particularly in patients with dual overexpression of HER2 and PD-L1 in their tumor,” lead study author Yelena Y. Janjigian, MD, chief of Gastrointestinal Oncology Service at Memorial Sloan Kettering Cancer Center in New York, New York, said in a presentation of the data.

In May 2021, the FDA granted accelerated approval to pembrolizumab in combination with trastuzumab and fluoropyrimidine- and platinum-containing chemotherapy for the first-line treatment of patients with locally advanced, unresectable, or metastatic HER2-positive gastric or GEJ adenocarcinoma, based on ORR data from the first interim analysis of KEYNOTE-811.3

At the 2023 ESMO Congress, investigators presented findings from additional interim analyses.1,2

The randomized, placebo-controlled study enrolled patients 18 years of age or older with a confirmed diagnosis of previously untreated, unresectable or metastatic gastric or GEJ adenocarcinoma with HER2 status of immunohistochemistry (IHC) 3+ or 2+/in situ hybridization–positive. Patients also needed to have measurable disease, an ECOG performance status of 0 or 1, a life expectancy of more than 6 months, and satisfactory organ function.

Patients were randomly assigned 1:1 to receive either 200 mg of intravenous (IV) pembrolizumab or placebo once every 3 weeks in combination with IV trastuzumab at 6 mg/kg once every 3 weeks (following a loading dose of 8 mg/kg) and chemotherapy consisting of either 800 mg/m2 of IV fluorouracil on days 1 to 5 of each 3-week cycle and 80 mg/m2 of IV cisplatin once every 3 weeks, or 1000 mg/m2 of oral capecitabine twice daily on days 1 to 14 of each 3-week cycle and 130 mg/m2 of IV oxaliplatin once every 3 weeks.

The treatment regimen continued for a maximum of 35 cycles (2 years) or until disease progression, intolerable toxicity, a decision by the investigator to halt treatment, or withdrawal of consent by the patient. Imaging assessments occurred at week 6 and then every 6 weeks thereafter.

Patients were stratified by geographic region, PD-L1 CPS, and chemotherapy choice.

The primary end points of the trial consisted of PFS per RECIST v1.1 criteria and OS in the intent-to-treat population. ORR and duration of response (DOR) were secondary end points. Notably, exploratory end points included health-related quality of life, identification of molecular, genetic, and genomic correlates of treatment, and PFS and ORR per iRECIST criteria.

The median age of patients were 62 years (range, 19-85) and 63 years (range, 32-85) in the pembrolizumab and placebo groups, respectively; a majority of patients across both treatment groups were male (81% and 80% for pembrolizumab and placebo, respectively) and had an ECOG performance state of 1 (58% and 58%). Across both groups, 32% of patients were from Australia, Europe, Israel, and North America, 34% were from Asia, and 34% were from the rest of the world.

Regarding histology , patients in the pembrolizumab cohort had diffuse (19%), intestinal (57%), or unknown (24%) subtypes. Patients in the placebo group had diffuse (15%), intestinal (54%), or unknown (31%) subtypes. Furthermore, 69% and 65% of patients in the pembrolizumab and placebo groups had a primary tumor location of the stomach, respectively. Notably, 85% of patients in both groups had a PD-L1 CPS of 1 or more. Two percent of patients in the pembrolizumab group vs 1% in the placebo group had microsatellite instability–high disease.

The chemotherapy of choice was capecitabine/oxaliplatin in 85% and 85% of patients in the pembrolizumab and placebo groups, respectively, and fluoropyrimidine/cisplatin was used in in 15% and 14% of patients, respectively.

Additional data showed that the ORR was 73% (95% CI, 68%-77%) in patients treated with pembrolizumab, including a complete response (CR) rate of 17%, a partial response (PR) rate of 56%, and stable disease (SD) rate of 19%. The disease control rate (DCR) was 92% (95% CI, 88%-94%) and the median DOR was 11.3 months (range, 1.1+ to 49.7+).

The ORR in patients treated with placebo was 60% (95% CI, 55%-65%), including a CR rate of 11%, a PR rate of 49%, a SD rate of 27%, a DCR of 87% (95% CI, 83%-91%), and a median DOR of 9.5 months (1.4+ to 48.7+).

Patients who were randomly assigned to the study who received at least 1 dose of the treatment were evaluated for safety. In the pembrolizumab group (n = 350) and placebo group (n = 346), 99% and 100% of patients experienced any-grade treatment-related adverse effects (TRAEs), respectively. The rates of grade 3/4 TRAEs were 58% and 50%, respectively. The rates of serious TRAEs were 26% and 23%, respectively. Four patients (1%) in the pembrolizumab arm and 3 patients (1%) in the placebo arm experienced grade 5 TRAEs. Discontinuation of any drug due to TRAEs occurred in 36% and 33% of patients treated with pembrolizumab vs placebo, respectively.

The most frequent TRAEs of any grade included diarrhea at 47% in the pembrolizumab group and 42% in the placebo group, nausea at 44% in the pembrolizumab group and 44% in the placebo group, and anemia at 31% in the pembrolizumab group and 33% in the placebo group.

Janjigian concluded by saying that although the data PFS suggest consideration of this regimen as a first-line treatment option for patients with metastatic HER2-positive gastric/GEJ cancer, the OS benefits of this regimen are yet to be determined.

“Based on these results, now this combination of pembrolizumab, trastuzumab, and chemotherapy in the first-line setting for HER2-positive and PD-L1–positive tumors is approved in Europe. The [OS] analysis is ongoing, though the study is complete,” Janjigian said.

Clinicians referring a patient to MSK can do so by visiting msk.org/refer, emailing referapatient@mskcc.org, or by calling 833-315-2722.

References

  1. Janjigian YY, Kawazoe A, Bai Y, et al. Pembrolizumab plus trastuzumab and chemotherapy for HER2+ metastatic gastric or gastroesophageal junction (mG/GEJ) adenocarcinoma: Survival results from the phase III, randomized, double-blind, placebo-controlled KEYNOTE-811 study. Ann Oncol. 2023;34(suppl 2):S851-S852. doi:10.1016/j.annonc.2023.09.1424
  2. Janjigian YY, Kawazoe A, Bai Y, et al. Pembrolizumab plus trastuzumab and chemotherapy for HER2+ metastatic gastric or gastroesophageal junction (mG/GEJ) adenocarcinoma: survival results from the phase 3, randomized, double-blind, placebo-controlled KEYNOTE-811 study. Lancet Oncol. Published online October 20, 2023. doi: 10.1016/S0140-6736(23)02033-0
  3. FDA grants accelerated approval to pembrolizumab for HER2-positive gastric cancer. News release. FDA. May 5, 2021. Accessed October 20, 2023. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-pembrolizumab-her2-positive-gastric-cancer
Related Videos
Saad Z. Usmani, MD, MBA, FACP, FASCO
Saad Z. Usmani, MD, MBA, FACP, FASCO, chief, Myeloma Service, Memorial Sloan Kettering Cancer Center
A panel of 5 experts on renal cell carcinoma
Chandler H. Park, MD, an expert on renal cell carcinoma
Video 6 - "Patient Case 2: A 62-Year-Old Woman with Metastatic Rectal Cancer"
Video 5 - "Adverse Events Associated With TAS-102 Plus Bevacizumab in CRC"
Michael J. Overman, MD
Saad Z. Usmani, MD, MBA, FACP, FASCO, chief, Myeloma Service, Memorial Sloan Kettering Cancer Center
Ilyas Sahin, assistant professor, Medicine, Department of Medicine, Division of Hematology & Oncology, University of Florida College of Medicine
Pasi A. Jänne, MD, PhD, discusses an exploratory analysis from the FLAURA2 trial of osimertinib plus chemotherapy in treatment-naive, EGFR-mutant NSCLC.