A Novel Immunomodulatory 27-Gene Signature to Predict Response to Neoadjuvant Immunochemotherapy for Primary Triple-Negative Breast Cancer

Simple Summary Through analysis of specimens from patients with primary triple-negative breast cancer (TNBC) enrolled in a neoadjuvant clinical trial assessing durvalumab with chemotherapy, we confirmed a novel 27-gene immuno-oncology (IO) signature that generates an IO score to predict the pathologic complete response (pCR) of primary TNBC to neoadjuvant immunotherapy with the PD-L1 blocker durvalumab with chemotherapy. Combining the 27-gene IO signature with PD-L1 immunohistochemistry strengthened the model’s predictive power of the pCR. Furthermore, the comprehensive computational analysis revealed that the 27-gene IO signature corresponded with an immunogenic tumor microenvironment. Abstract A precise predictive biomarker for TNBC response to immunochemotherapy is urgently needed. We previously established a 27-gene IO signature for TNBC derived from a previously established 101-gene model for classifying TNBC. Here we report a pilot study to assess the performance of a 27-gene IO signature in predicting the pCR of TNBC to preoperative immunochemotherapy. We obtained RNA sequencing data from the primary tumors of 55 patients with TNBC, who received neoadjuvant immunochemotherapy with the PD-L1 blocker durvalumab. We determined the power and accuracy in predicting pCR for the immunomodulatory (IM) subtype identified by the 101-gene model, the 27-gene IO signature, and PD-L1 expression by immunohistochemistry (IHC). The pCR rate was 45% (25/55). The odds ratios for pCR were as follows: IM subtype by 101-gene model, 3.14 (p = 0.054); 27-gene IO signature, 4.13 (p = 0.012); PD-L1 expression by IHC, 2.63 (p = 0.106); 27-gene IO signature in combination with PD-L1 expression by IHC, 6.53 (p = 0.003). The 27-gene IO signature has the potential to predict the pCR of primary TNBC to neoadjuvant immunochemotherapy. Further analysis in a large cohort is needed.

[1]  P. Fasching,et al.  VP7-2021: KEYNOTE-522: Phase III study of neoadjuvant pembrolizumab + chemotherapy vs. placebo + chemotherapy, followed by adjuvant pembrolizumab vs. placebo for early-stage TNBC , 2021 .

[2]  A. Schneeweiss,et al.  First-line atezolizumab plus nab-paclitaxel for unresectable, locally advanced, or metastatic triple-negative breast cancer: IMpassion130 final overall survival analysis. , 2021, Annals of oncology : official journal of the European Society for Medical Oncology.

[3]  B. Ring,et al.  A novel immuno-oncology algorithm measuring tumor microenvironment to predict response to immunotherapies , 2021, Heliyon.

[4]  Ahn, Jin Hee,et al.  Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial , 2020, The Lancet.

[5]  H. Iwata,et al.  LBA11 IMpassion031: Results from a phase III study of neoadjuvant (neoadj) atezolizumab + chemotherapy in early triple-negative breast cancer (TNBC) , 2020 .

[6]  L. Pusztai,et al.  PD-L1 Protein Expression on Both Tumor Cells and Macrophages are Associated with Response to Neoadjuvant Durvalumab with Chemotherapy in Triple-negative Breast Cancer , 2020, Clinical Cancer Research.

[7]  E. Richardson,et al.  Revisiting the role of CD4+ T cells in cancer immunotherapy—new insights into old paradigms , 2020, Cancer Gene Therapy.

[8]  M. Lenardo,et al.  A guide to cancer immunotherapy: from T cell basic science to clinical practice , 2020, Nature Reviews Immunology.

[9]  A. Gown,et al.  The path to a better biomarker: application of a risk management framework for the implementation of PD‐L1 and TILs as immuno‐oncology biomarkers in breast cancer clinical trials and daily practice , 2020, The Journal of pathology.

[10]  P. Fasching,et al.  Pembrolizumab for Early Triple-Negative Breast Cancer. , 2020, The New England journal of medicine.

[11]  I. Melero,et al.  Dendritic cells in cancer immunology and immunotherapy , 2019, Nature Reviews Immunology.

[12]  Brian Ruffell,et al.  Macrophages as regulators of tumour immunity and immunotherapy , 2019, Nature Reviews Immunology.

[13]  A. Butte,et al.  xCell: digitally portraying the tissue cellular heterogeneity landscape , 2017, bioRxiv.

[14]  M. Atkins,et al.  Predictive biomarkers for checkpoint inhibitor-based immunotherapy. , 2016, The Lancet. Oncology.

[15]  Jun S. Liu,et al.  Comprehensive analyses of tumor immunity: implications for cancer immunotherapy , 2016, Genome Biology.

[16]  L. Pusztai,et al.  Quantitative assessment of the spatial heterogeneity of tumor-infiltrating lymphocytes in breast cancer , 2016, Breast Cancer Research.

[17]  Harold L. Moses,et al.  Refinement of Triple-Negative Breast Cancer Molecular Subtypes: Implications for Neoadjuvant Chemotherapy Selection , 2016, PloS one.

[18]  B. Ring,et al.  Generation of an algorithm based on minimal gene sets to clinically subtype triple negative breast cancer patients , 2016, BMC Cancer.

[19]  Ash A. Alizadeh,et al.  Robust enumeration of cell subsets from tissue expression profiles , 2015, Nature Methods.

[20]  R. Emerson,et al.  PD-1 blockade induces responses by inhibiting adaptive immune resistance , 2014, Nature.

[21]  Funda Meric-Bernstam,et al.  Differential Response to Neoadjuvant Chemotherapy Among 7 Triple-Negative Breast Cancer Molecular Subtypes , 2013, Clinical Cancer Research.

[22]  X. Chen,et al.  Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. , 2011, The Journal of clinical investigation.

[23]  S. Narod,et al.  Triple-Negative Breast Cancer: Clinical Features and Patterns of Recurrence , 2007, Clinical Cancer Research.