American Association for Cancer Research
Browse
cir-23-0873_supplementary_table_s2_suppst2.pdf (19.18 kB)

Supplementary Table S2 from Machine Learning Links T-cell Function and Spatial Localization to Neoadjuvant Immunotherapy and Clinical Outcome in Pancreatic Cancer

Download (19.18 kB)
journal contribution
posted on 2024-05-02, 08:05 authored by Katie E. Blise, Shamilene Sivagnanam, Courtney B. Betts, Konjit Betre, Nell Kirchberger, Benjamin J. Tate, Emma E. Furth, Andressa Dias Costa, Jonathan A. Nowak, Brian M. Wolpin, Robert H. Vonderheide, Jeremy Goecks, Lisa M. Coussens, Katelyn T. Byrne

Supplementary Table S2. Tissue area sampled per patient out of total tissue area resected per patient (in mm2).

Funding

National Cancer Institute (NCI)

United States Department of Health and Human Services

Find out more...

Dana-Farber Cancer Institute Hale Family Center for Pancreatic Cancer Research

Lustgarten Foundation Dedicated Laboratory Program

Parker Institute for Cancer Immunotherapy (PICI)

Brenden-Colson Center for Pancreatic Care

Robert L. Fine Cancer Research Foundation

Prospect Creek Foundation

Knight Cancer Institute, Oregon Health and Science University (KCI)

History

ARTICLE ABSTRACT

Tumor molecular data sets are becoming increasingly complex, making it nearly impossible for humans alone to effectively analyze them. Here, we demonstrate the power of using machine learning (ML) to analyze a single-cell, spatial, and highly multiplexed proteomic data set from human pancreatic cancer and reveal underlying biological mechanisms that may contribute to clinical outcomes. We designed a multiplex immunohistochemistry antibody panel to compare T-cell functionality and spatial localization in resected tumors from treatment-naïve patients with localized pancreatic ductal adenocarcinoma (PDAC) with resected tumors from a second cohort of patients treated with neoadjuvant agonistic CD40 (anti-CD40) monoclonal antibody therapy. In total, nearly 2.5 million cells from 306 tissue regions collected from 29 patients across both cohorts were assayed, and over 1,000 tumor microenvironment (TME) features were quantified. We then trained ML models to accurately predict anti-CD40 treatment status and disease-free survival (DFS) following anti-CD40 therapy based on TME features. Through downstream interpretation of the ML models’ predictions, we found anti-CD40 therapy reduced canonical aspects of T-cell exhaustion within the TME, as compared with treatment-naïve TMEs. Using automated clustering approaches, we found improved DFS following anti-CD40 therapy correlated with an increased presence of CD44+CD4+ Th1 cells located specifically within cellular neighborhoods characterized by increased T-cell proliferation, antigen experience, and cytotoxicity in immune aggregates. Overall, our results demonstrate the utility of ML in molecular cancer immunology applications, highlight the impact of anti-CD40 therapy on T cells within the TME, and identify potential candidate biomarkers of DFS for anti-CD40–treated patients with PDAC.