Beyond One-Size-Fits-All

The Quest for Universal Biomarkers in Cancer Immunotherapy

Biomarkers Immunotherapy Cancer Research Personalized Medicine

A Revolution With a Blind Spot

Imagine a revolutionary cancer treatment that doesn't attack cancer cells directly but instead empowers the body's own immune system to recognize and destroy tumors.

This is the promise of immunotherapy, particularly drugs known as immune checkpoint inhibitors that target molecules like CTLA-4 and PD-1. These treatments have transformed cancer care, producing remarkable, long-lasting responses in some patients with advanced cancers that were once considered untreatable 6 .

Yet, this breakthrough comes with a significant challenge: these powerful drugs don't work for everyone. In fact, they only benefit a subset of patients, while others endure potentially serious side effects with no therapeutic benefit 9 . This hit-or-miss approach has sparked an urgent scientific quest to find better ways to predict who will respond—a search for biological clues known as biomarkers. The ultimate goal? To find reliable, universal biomarkers that can guide treatment decisions across different cancer types, moving beyond the current "one-size-fits-all" approach to truly personalized cancer care.

The Current Biomarker Landscape: Pieces of the Puzzle

Biomarkers in immunotherapy fall into two main categories: prognostic biomarkers that provide information about a patient's overall cancer outcome regardless of treatment, and predictive biomarkers that specifically identify patients likely to respond to a particular therapy 4 . Several biomarkers have already entered clinical practice, though each has limitations.

PD-L1 Expression

Testing for PD-L1 levels on tumor cells through immunohistochemistry is the most commonly used biomarker today 2 5 .

Tumor Mutational Burden

Measures the total number of mutations in a tumor's DNA. More mutations create more targets for the immune system 2 .

Mismatch Repair Deficiency

Tumors with DNA repair defects become hyper-mutated and highly visible to the immune system 2 6 .

Note: "Although high tumor mutational burden (TMB), presence of tumor microsatellite instability (MSI) and mismatch-repair-deficient (dMMR) status, as well as high PD-L1 expression, in tumor cells are well established biomarkers, they are not perfect" 9 .

The Challenge of Generalizability: Why One Marker Doesn't Fit All

The core problem limiting current biomarkers is their lack of generalizability—what works well as a predictor in one cancer type often fails in another. This variability stems from several fundamental biological and technical challenges:

Tumor Heterogeneity

Tumors contain diverse cell populations with different characteristics. A small biopsy from one part may not represent the entire tumor's biology 5 .

Technical Variability

Differences in assay platforms, scoring systems, and cutoff thresholds create consistency problems 7 .

Biological Complexity

The immune response involves multiple overlapping pathways. A 2025 study found high PD-1 RNA expression correlated with other immune checkpoints 7 .

Case Study: Pancreatic Cancer

A 2025 study focusing on an Egyptian population found that specific genetic variants in the CTLA-4 gene were associated with increased risk of pancreatic ductal adenocarcinoma (PDAC) and higher levels of soluble CTLA-4 1 .

While promising for this specific population, it remains unclear whether these findings apply to other ethnic groups or cancer types.
Limitations of Current Biomarkers
Biomarker Key Strength Generalizability Challenge
PD-L1 Expression Clinically validated in multiple cancers Heterogeneous expression; different scoring systems across cancer types
Tumor Mutational Burden (TMB) Tissue-agnostic approval Cut-off values not uniform across cancers; affected by patient ancestry
Mismatch Repair Deficiency Strong predictor when present Only present in small subset of most cancer types
CTLA-4 Polymorphisms Identified in specific populations Variants may not predict response in other ethnic groups or cancer types 1

A Key Experiment: Mapping PD-1 Across Cancers

A landmark study published in 2025 in npj Genomic Medicine provides valuable insights into the generalizability challenge by systematically mapping PD-1 expression across different cancer types 7 . This research represents precisely the kind of comprehensive analysis needed to identify more universal biomarkers.

Methodology: A Cross-Cancer Atlas
  • 514 patients with advanced or metastatic disease
  • 31 different cancer types
  • Used transcriptomic analysis to measure PD-1 RNA expression
  • Stratified expression into high (75th-100th), moderate (25th-74th), and low (0-24th percentile) categories
Key Findings
  • PD-1 expression varies widely across and within cancer types
  • High PD-1 expression independently predicted longer overall survival in immunotherapy patients
  • Hazard ratio of 0.40 indicates 60% reduction in risk of death
  • Not merely prognostic - specifically predicted immunotherapy response
PD-1 Expression Across Selected Cancers 7
Cancer Type High PD-1 Expression Moderate PD-1 Expression Low PD-1 Expression
Pancreatic Cancer 21.82% 49.09% 29.09%
Liver/Bile Duct 21.05% 47.37% 31.58%
Colorectal Cancer 17.14% 47.86% 35.00%
Breast Cancer 16.33% 53.06% 30.61%
Lung Cancer 15.00% 50.00% 35.00%
Occult Primary 7.59% 43.04% 49.37%
Significance: PD-1 expression maintained its predictive power across various cancer types, suggesting it might serve as a more universal biomarker than current options. The study also found that high PD-1 expression correlated with other immune checkpoints (CTLA-4, LAG-3, and TIGIT), indicating it may identify tumors with broadly active immune responses 7 .

The Scientist's Toolkit: Key Research Reagent Solutions

The search for generalizable biomarkers relies on sophisticated laboratory tools and reagents. Here are some essential components of the biomarker discovery toolkit:

Research Tool Function in Biomarker Research Application Example
TaqMan™ SNP Genotyping Assays Detects specific genetic variations Analyzing CTLA-4 polymorphisms in pancreatic cancer risk 1
ELISA Kits Measures protein concentrations in solutions Quantifying soluble CTLA-4 levels in patient serum 1
OmniSeq Immune Panel Comprehensive immune gene expression profiling Mapping PD-1 expression across 31 cancer types 7
TaqMan™ Genotyping Master Mix Essential component for genetic amplification Genotyping CTLA-4 variants in PCR experiments 1
Multiplex Immunofluorescence Simultaneously visualizes multiple protein markers Analyzing spatial relationships between immune cells in tumors 5
RNA Extraction Kits Isolates high-quality RNA from patient samples Preparing samples for transcriptomic analysis of PD-1 3

The Future of Biomarkers: Integration and Innovation

As the limitations of single biomarkers become increasingly apparent, the field is shifting toward integrated approaches that combine multiple data types. The future lies in composite biomarkers that simultaneously consider tumor genetics, immune environment, and patient factors 5 9 .

Peripheral Blood Biomarkers

Instead of relying solely on invasive tumor biopsies, researchers are developing blood-based biomarkers that provide a "liquid biopsy" of the cancer immune environment.

These include circulating immune cells, soluble checkpoint proteins, and other inflammatory markers 5 . For instance, the relative eosinophil count (REC) in peripheral blood has shown promise as a predictor of response to CTLA-4 blockade in melanoma patients 2 .

AI and Multi-Omics

The integration of multiple data layers—genomics, transcriptomics, proteomics—coupled with advanced computational analysis represents the cutting edge of biomarker discovery.

One study demonstrated approximately 15% improvement in predictive accuracy using multi-omics with machine learning models 2 . These approaches can analyze the complex interplay between tumors and the immune system in ways that were previously impossible.

Collaboration is Key: The road to generalizable biomarkers will require collaboration and data sharing on an unprecedented scale. Initiatives like the National Cancer Institute's Cancer Immune Monitoring and Analysis Centers–Cancer Immunologic Data Commons (CIMAC-CIDC) Network are working to harmonize methods and integrate large datasets from multiple immunotherapy trials 9 .

Conclusion: The Path to Truly Personalized Immunotherapy

The quest for generalizable biomarkers of response to CTLA-4 and PD-1 blockade represents one of the most important frontiers in cancer research. While significant challenges remain—tumor heterogeneity, technical variability, and biological complexity—the scientific community is making steady progress.

The transition from single, inflexible biomarkers to adaptive, multi-parameter models will ultimately allow clinicians to tailor immunotherapy with unprecedented precision. As research continues to unravel the complex dialogue between tumors and the immune system, the vision of predicting immunotherapy response for each individual patient, regardless of their cancer type, moves closer to reality.

Future success will depend on continuing to map the immune landscape across diverse cancers and populations, standardizing assessment methods, and embracing integrated analytical approaches. With these advances, the revolutionary potential of immunotherapy may finally be accessible to all patients who can benefit, ushering in a new era of truly personalized cancer care.

The future for ICIs is undeniably bright, with promising recent results in the neoadjuvant setting and for inhibitors of targets beyond PD-1–PD-L1 and CTLA-4. Intensifying efforts to enhance data standardization, sharing of existing trial datasets, and prospective validation of candidate biomarkers in diverse populations will be crucial for the development of more-effective biomarkers. 9

References