Association Between Biomarkers and Clinical Outcomes of Pembrolizumab Monotherapy in Patients With Metastatic Triple-Negative Breast Cancer: KEYNOTE-086 Exploratory Analysis.

PURPOSE In the two-cohort phase II KEYNOTE-086 study (ClinicalTrials.gov identifier: NCT02447003), first-line and second-line or later pembrolizumab monotherapy demonstrated antitumor activity in metastatic triple-negative breast cancer (mTNBC; N = 254). This exploratory analysis evaluates the association between prespecified molecular biomarkers and clinical outcomes. METHODS Cohort A enrolled patients with disease progression after one or more systemic therapies for metastatic disease irrespective of PD-L1 status; Cohort B enrolled patients with previously untreated PD-L1-positive (combined positive score [CPS] ≥ 1) metastatic disease. The association between the following biomarkers as continuous variables and clinical outcomes (objective response rate [ORR], progression-free survival [PFS], and overall survival [OS]) was evaluated: PD-L1 CPS (immunohistochemistry), cluster of differentiation 8 (CD8; immunohistochemistry), stromal tumor-infiltrating lymphocyte (sTIL; hematoxylin and eosin staining), tumor mutational burden (TMB; whole-exome sequencing [WES]), homologous recombination deficiency-loss of heterozygosity, mutational signature 3 (WES), mutational signature 2 (apolipoprotein B mRNA editing catalytic polypeptide-like; WES), T-cell-inflamed gene expression profile (TcellinfGEP; RNA sequencing), and 10 non-TcellinfGEP signatures (RNA sequencing); Wald test P values were calculated, and significance was prespecified at α = 0.05. RESULTS In the combined cohorts (A and B), PD-L1 (P = .040), CD8 (P < .001), sTILs (P = .012), TMB (P = .007), and TcellinfGEP (P = .011) were significantly associated with ORR; CD8 (P < .001), TMB (P = .034), Signature 3 (P = .009), and TcellinfGEP (P = .002) with PFS; and CD8 (P < .001), sTILs (P = .004), TMB (P = .025), and TcellinfGEP (P = .001) with OS. None of the non-TcellinfGEP signatures were associated with outcomes of pembrolizumab after adjusting for the TcellinfGEP. CONCLUSION In this exploratory biomarker analysis from KEYNOTE-086, baseline tumor PD-L1, CD8, sTILs, TMB, and TcellinfGEP were associated with improved clinical outcomes of pembrolizumab and may help identify patients with mTNBC who are most likely to respond to pembrolizumab monotherapy.

[1]  J. Lunceford,et al.  Tumor mutational burden predicts the efficacy of pembrolizumab monotherapy: a pan-tumor retrospective analysis of participants with advanced solid tumors , 2022, Journal for ImmunoTherapy of Cancer.

[2]  S. Loi,et al.  LBA16 KEYNOTE-355: Final results from a randomized, double-blind phase III study of first-line pembrolizumab + chemotherapy vs placebo + chemotherapy for metastatic TNBC , 2021, Annals of Oncology.

[3]  Trevor J Pugh,et al.  Pan-cancer analysis of longitudinal metastatic tumors reveals genomic alterations and immune landscape dynamics associated with pembrolizumab sensitivity , 2021, Nature Communications.

[4]  Anh Nguyen-Duc,et al.  Atezolizumab and nab-Paclitaxel in Advanced Triple-Negative Breast Cancer: Biomarker Evaluation of the IMpassion130 Study , 2021, Journal of the National Cancer Institute.

[5]  P. Hegde,et al.  Comparison of PD-L1 protein expression between primary tumors and metastatic lesions in triple negative breast cancers , 2020, Journal for ImmunoTherapy of Cancer.

[6]  V. Di Lauro,et al.  Biomarkers in Triple-Negative Breast Cancer: State-of-the-Art and Future Perspectives , 2020, International journal of molecular sciences.

[7]  Roger R. Wang,et al.  Multi-panel immunofluorescence analysis of tumor infiltrating lymphocytes in triple negative breast cancer: Evolution of tumor immune profiles and patient prognosis , 2020, PloS one.

[8]  O. Cohen,et al.  Prevalence and mutational determinants of high tumor mutation burden in breast cancer , 2019, bioRxiv.

[9]  G. Curigliano,et al.  Recent advances in triple negative breast cancer: the immunotherapy era , 2019, BMC Medicine.

[10]  E. Winer,et al.  Pembrolizumab monotherapy for previously untreated, PD-L1-positive, metastatic triple-negative breast cancer: cohort B of the phase II KEYNOTE-086 study , 2019, Annals of oncology : official journal of the European Society for Medical Oncology.

[11]  S. Loi,et al.  Pembrolizumab monotherapy for previously treated metastatic triple-negative breast cancer: cohort A of the phase II KEYNOTE-086 study , 2019, Annals of oncology : official journal of the European Society for Medical Oncology.

[12]  Y. Shentu,et al.  Use of archival versus newly collected tumor samples for assessing PD-L1 expression and overall survival: an updated analysis of KEYNOTE-010 trial , 2019, Annals of oncology : official journal of the European Society for Medical Oncology.

[13]  E. Elgabry,et al.  Long-term Clinical Outcomes and Biomarker Analyses of Atezolizumab Therapy for Patients With Metastatic Triple-Negative Breast Cancer: A Phase 1 Study , 2019, JAMA oncology.

[14]  J. Lunceford,et al.  Pan-tumor genomic biomarkers for PD-1 checkpoint blockade–based immunotherapy , 2018, Science.

[15]  P. Khosravi-Shahi,et al.  Metastatic triple negative breast cancer: Optimizing treatment options, new and emerging targeted therapies , 2018, Asia-Pacific journal of clinical oncology.

[16]  L. Dirix,et al.  Avelumab, an anti-PD-L1 antibody, in patients with locally advanced or metastatic breast cancer: a phase 1b JAVELIN Solid Tumor study , 2017, Breast Cancer Research and Treatment.

[17]  P. Marchetti,et al.  PD-L1 Expression in TNBC: A Predictive Biomarker of Response to Neoadjuvant Chemotherapy? , 2017, BioMed research international.

[18]  Toby C. Cornish,et al.  PD-L1 (B7-H1) expression and the immune tumor microenvironment in primary and metastatic breast carcinomas. , 2016, Human pathology.

[19]  T. Nielsen,et al.  The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. , 2015, Annals of oncology : official journal of the European Society for Medical Oncology.

[20]  David T. W. Jones,et al.  Signatures of mutational processes in human cancer , 2013, Nature.