Approximately 3% to 10% of patients with pancreatic ductal adenocarcinoma (PDAC) have pathogenic germline variants (PGVs) leading to homologous recombination deficiency (HRD), and approximately 15% to 17% have similar somatic alterations.1 These patients have increased sensitivity to platinum-containing chemotherapy and PARP inhibitors.1,2 There is increased genomic instability in this subgroup of patients; in a pan-cancer analysis, PDACs with biallelic loss of BRCA1 and BRCA2 (BRCA1/2) had a higher median tumor mutation burden (4.3 vs 1.7 mutations/Mb) and higher genomic loss of heterozygosity than wild-type tumors.3 Similar genomic instability leading to tumor-inflamed phenotype has been described in BRCA1/2-variant breast, ovarian, and prostate cancers.4 Immune checkpoint inhibitors (ICIs) have been ineffective in unselected patients with PDAC.5 Given the molecular rationale for increased susceptibility to ICIs in patients with PDAC associated with HRD PGVs, we investigated whether this subgroup may be sensitive to immunotherapy strategies.
This retrospective single-institution case series included patients with chemotherapy-refractory metastatic PDAC or biliary cancer who were treated between April 2018 and February 2021 with combination ipilimumab/nivolumab and had PGVs in HRD genes (detected by a Clinical Laboratory Improvement Amendments–approved assay). This study was approved by the University of Miami institutional review board, and a waiver of informed consent was given to collect deidentified data retrospectively. This study followed the reporting guideline for case series.
Patients received ipilimumab, 1 mg/kg, with nivolumab, 3 mg/kg, every 21 days for 4 doses, followed by nivolumab, 480 mg, every 28 days. Available pretreatment tumor specimens were analyzed by immunohistochemistry and RNA sequencing using the PanCancer IO 360 Panel. Data were analyzed using Prism, version 7, and 2-sided P < .05 was the level of significance.
A total of 12 patients were included (7 [58%] men; median age, 66 [range, 47-73] years) (Table). Four patients achieved a complete response (CR) to therapy, 1 had a partial response, and 2 had stable disease. The objective response rate was 42%, with a disease control rate of 58%. The 4 patients who achieved CR remained without evidence of disease 11 to 41 months after starting therapy. Treatment-related adverse events were consistent with the known toxic effects of combination ipilimumab/nivolumab.
Adequate archival biopsy results were available for 4 patients (2 with CRs and 2 with progressive disease). Responders had higher density of tumor-infiltrating lymphocytes than nonresponders. Expression of chemokines known to enable trafficking of adaptive effector immune populations was significantly higher in responders than in nonresponders: CCL4 (mean [SE], 6.17 [0.12] vs 4.20 [0.11]; P < .001), CXCL9 (median, 7.44 [IQR, 7.09-7.79] vs 4.70 [IQR, 1.86-7.54]; P = .03), and CXCL10 (mean [SE], 6.81 [0.13] vs 4.51 [0.39]; P = .003) (Figure). The expression of these chemokines was previously shown to correlate with a T-cell–inflamed phenotype in PDAC.6
This case series showed that ipilimumab/nivolumab therapy was associated with clinical benefit in a biomarker-selected group of patients with PDAC and PGVs in genes encoding for HRD, with pretreatment biopsy analysis supporting biological plausibility. Most patients had platinum and PARP inhibitor–refractory disease and would typically have an unfavorable prognosis but showed marked improvement with ipilimumab/nivolumab therapy. Of the 4 patients who achieved CR, 3 discontinued therapy after 2 years and all remained without evidence of disease. Limitations of the study were the small number of patients and few tumor specimens for analysis; however, differences were revealed in the pretreatment tumor characteristics between responders and nonresponders, including more tumor-infiltrating lymphocytes and a T-cell–inflamed signature on RNA expression analysis in responders.
Typically, PDAC is characterized by few or no infiltrating immune effector cells, low antigenicity, and multiple immunosuppressive factors in the tumor microenvironment. Treatment with ICIs has not shown meaningful benefit for PDAC except for microsatellite instability–high tumors. This study showed an association between germline HRD status and sensitivity to ICIs, advancing previous evidence of an association between BRCA1/2 variants in other tumors and immunotherapy response.
Accepted for Publication: January 31, 2022.
Published Online: April 21, 2022. doi:10.1001/jamaoncol.2022.0611
Corresponding Author: Peter J. Hosein, MD, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, 1121 NW 14th St, Ste 245A, Miami, FL 33136 (phosein@miami.edu).
Author Contributions: Drs Terrero and Hosein had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.
Concept and design: Terrero, Merchant, Hosein.
Acquisition, analysis, or interpretation of data: All authors.
Drafting of the manuscript: Terrero, Datta, Dennison, Hosein.
Critical revision of the manuscript for important intellectual content: Terrero, Datta, Sussman, Lohse, Merchant, Hosein.
Statistical analysis: Datta, Dennison, Hosein.
Obtained funding: Datta, Hosein.
Administrative, technical, or material support: Terrero, Merchant, Hosein.
Supervision: Datta, Sussman, Merchant, Hosein.
Conflict of Interest Disclosures: None reported.
Funding/Support: This work was supported by the KL2 Career Development Grant from the Miami Clinical and Translational Science Institute under grant UL1TR002736 from the National Institutes of Health (NIH), the Stanley J. Glaser Foundation, the Franklin H. Martin, MD, FACS, Faculty Research Fellowship of the American College of Surgeons, and the Association for Academic Surgery Joel J. Roslyn Faculty Research Award (Dr Datta); Hyundai Hope on Wheels, the Sebastian Strong Foundation, the Wallace H. Coulter Center Foundation for Translational Research, and the Live Like Bella Childhood Cancer Foundation (Dr Lohse); grants R01 CA161976 and T32 CA211034 from the NIH, Pancreatic Cancer Action Network–American Association for Cancer Research Clinical and Translational Cancer Research Fellowship 15-65-25-MERC, and the Sylvester Comprehensive Cancer Center (Dr Merchant); the Helene & Bernard Herskowitz Fund for Pancreatic Cancer Research, the Esther Finkelstein Family Fund for Pancreatic Cancer Research, and the Luis Rios Family Fund for Pancreatic Cancer Research (Dr Hosein); and Cancer Center Core Grant P30CA240139 from the National Cancer Institute, NIH.
Role of the Funder/Sponsor: The funders had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication.
3.Sokol
ES, Pavlick
D, Khiabanian
H,
et al. Pan-cancer analysis of
BRCA1 and
BRCA2 genomic alterations and their association with genomic instability as measured by genome-wide loss of heterozygosity.
JCO Precis Oncol. 2020;4:442-465. doi:
10.1200/PO.19.00345
PubMedGoogle ScholarCrossref 4.Strickland
KC, Howitt
BE, Shukla
SA,
et al. Association and prognostic significance of
BRCA1/2-mutation status with neoantigen load, number of tumor-infiltrating lymphocytes and expression of PD-1/PD-L1 in high grade serous ovarian cancer.
Oncotarget. 2016;7(12):13587-13598. doi:
10.18632/oncotarget.7277
PubMedGoogle ScholarCrossref