MedPath

Blood-Based Protein Analysis Achieves 85.8% Accuracy in Predicting Triple-Negative Breast Cancer Immunotherapy Response

15 days ago3 min read

Key Insights

  • Researchers at Fudan University developed the Plasma Immuno Prediction Score (PIPscore) using six immune-related plasma proteins to predict immunotherapy response in triple-negative breast cancer with 85.8% accuracy.

  • The study identified key biomarkers ARG1, NOS3, and CD28 from analysis of 92 proteins in 195 TNBC patients, offering a non-invasive alternative to tumor biopsies for treatment prediction.

  • The PIPscore demonstrated 96% precision in predicting 12-month progression-free survival, potentially enabling personalized treatment strategies and early identification of non-responders.

Researchers at Fudan University Shanghai Cancer Center have developed a groundbreaking blood-based test that predicts immunotherapy response in triple-negative breast cancer (TNBC) patients with 85.8% accuracy. The study, involving 195 patients, introduces the Plasma Immuno Prediction Score (PIPscore), which analyzes immune-related proteins in blood samples to forecast treatment outcomes before therapy begins.

Novel Biomarker Discovery Through Comprehensive Protein Analysis

The research team conducted systematic analysis of 92 immune-related proteins circulating in plasma samples collected before, during, and after immunotherapy treatment. Through this comprehensive profiling, investigators identified several key biomarkers that correlate strongly with treatment outcomes, most notably ARG1, NOS3, and CD28 proteins.
"Our findings transcend the tumor microenvironment, highlighting systemic immunity as the pivotal driver of immunotherapy outcomes in TNBC," stated Dr. Yizhou Jiang, co-corresponding author of the study. "By distilling complex plasma proteomics into the clinically actionable PIPscore, we have forged a bridge connecting cutting-edge research with tangible therapeutic decision-making."
The PIPscore integrates six immune-related plasma proteins into a sophisticated predictive model that stratifies patients into high- and low-response categories prior to treatment initiation. This approach enables oncologists to tailor therapies more effectively while sparing non-responders from immunotherapy-associated toxicities and financial burdens.

Superior Performance in Prognostic Assessment

The model demonstrated remarkable prognostic capabilities, achieving 96% precision in predicting 12-month progression-free survival. This performance represents a significant advancement over current biomarkers such as PD-L1 expression and tumor mutational burden, which are fraught with inconsistency and require invasive sampling procedures.
Post-treatment samples from patients who achieved pathologic complete response exhibited elevated levels of immune-stimulatory molecules like CXCL9 and interferon-gamma (IFN-γ), emphasizing active immune engagement. The observed expression patterns revealed nuanced immune dynamics: ARG1 and CD28 were upregulated in responders, while NOS3 was downregulated in responders.

Addressing Critical Clinical Limitations

Triple-negative breast cancer, characterized by the absence of estrogen receptor, progesterone receptor, and HER2 expression, has long defied targeted therapies. This aggressive breast cancer subtype leaves immunotherapy as a primary treatment option, though outcomes remain unpredictable.
The plasma proteomics approach circumvents intrinsic heterogeneity and sampling bias of tumor biopsies by offering a dynamic view of systemic immunity. This perspective proves critical for understanding the complex interplay between the tumor microenvironment and host immune mechanisms.

Mechanistic Insights Through Multi-Omic Integration

Integration of single-cell RNA sequencing data provided granular perspective linking circulating protein levels with cellular heterogeneity within the tumor microenvironment. The inverse relationship between NOS3 plasma concentrations and intratumoral CD8+ T-cell abundance underscores the relevance of systemic immunosuppression markers.
The research revealed that proteins like ARG1 play crucial roles in arginine metabolism pathways that potentiate T-cell functionality, while elevated NOS3 may contribute to an immunosuppressive milieu by limiting CD8+ T-cell infiltration into tumors. This holistic, multi-omic approach bridges peripheral blood immune signatures with intratumoral cellular landscapes.

Clinical Implementation and Future Applications

The non-invasive nature of plasma-based monitoring enables frequent, real-time assessment of immune status without risks and discomfort associated with repeated biopsies. Dynamic monitoring of PIPscore during treatment courses may facilitate timely therapeutic modifications, maximizing benefit while minimizing unnecessary exposure to ineffective treatments.
The study employed state-of-the-art high-sensitivity immunoassays for protein quantification, ensuring detection of low-abundance proteins critical to immune function. Validation of proteomic data through enzyme-linked immunosorbent assays (ELISA) bolstered platform reliability.
The research implications extend beyond TNBC, suggesting broader applicability of plasma proteomic profiling in predicting immunotherapy responses across diverse malignancies. Given variability in patient responses to immune checkpoint inhibitors in cancers such as melanoma, lung, and bladder carcinoma, non-invasive predictive tools like the PIPscore could substantially enhance personalized treatment paradigms and resource allocation.
Subscribe Icon

Stay Updated with Our Daily Newsletter

Get the latest pharmaceutical insights, research highlights, and industry updates delivered to your inbox every day.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.