Category Ranking

98%

Total Visits

921

Avg Visit Duration

2 minutes

Citations

20

Article Abstract

Introduction: We previously conducted a phase I/Ib study (NCT03712943) with regorafenib and nivolumab in patients with refractory metastatic mismatch repair proficient (pMMR) colorectal cancer (CRC). This study aimed to investigate the role of Xerna™ TME Panel in predicting the treatment response.

Methods: Twenty-two archival pretreatment tumor samples were subjected to the Xerna™ TME Panel, a machine learning-based RNA-sequencing biomarker assay. The Xerna tumor microenvironment (TME) subtypes were evaluated for correlation with overall survival (OS), progression-free survival (PFS), disease control rate (DCR), and other biomarkers including KRAS, PD-L1, CD8 expression, and Treg cells in TME.

Results: Based on Xerna™ TME Panel, 4 patients with immune-active (IA) subtype and 6 patients with immune-suppressed subtype were classified as biomarker-positive, and five with angiogenic (A) subtype and seven with immune desert subtype were biomarker-negative. While not reaching statistical significance, Xerna TME biomarker-positive patients seemed to have longer median PFS (7.9 vs. 4.1 months, p = 0.254), median OS (15.75 vs. 11.9 months, p = 0.378), and higher DCR (70% vs. 58%, p = 0.675). The IA subtype in our cohort had higher levels of CD4+ FOXP3+ Treg cells, whereas the A subtype showed lower levels of Treg cells.

Conclusion: Xerna™ TME Panel analysis in patients with refractory metastatic pMMR CRC who were treated with regorafenib plus nivolumab might be of value for predictive clinical benefit. Further studies are needed to evaluate the predictive role of Xerna™ TME Panel analysis in patients with refractory metastatic pMMR CRC.

Download full-text PDF

Source
http://www.ncbi.nlm.nih.gov/pmc/articles/PMC11216360PMC
http://dx.doi.org/10.1159/000535599DOI Listing

Publication Analysis

Top Keywords

xerna™ tme
20
tme panel
20
regorafenib nivolumab
12
patients refractory
12
refractory metastatic
12
metastatic mismatch
8
mismatch repair
8
repair proficient
8
colorectal cancer
8
treated regorafenib
8

Similar Publications

An Activatable and Covalent Tumor-Associated Antigen Capturer Enabling Systemic Injection for Promoted Antitumor Immunity.

J Am Chem Soc

September 2025

Frontiers Science Center for New Organic Matter, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences and Academy for Advanced Interdisciplinary Studies, Nankai University, Tianjin 300071, PR China.

Antigen-capturing nanomaterials hold great promise for cancer immunotherapy; however, the need for tumor localized administration and limited antigen-binding affinity remains the "Achilles heel" of this strategy. Herein, we present a tumor microenvironment (TME)-activatable nanoplatform, TDR848@FPB, designed for systemic administration and enhanced covalent capture of tumor-associated antigens (TAAs), enabling effective immunotherapy with minimal off-target effects and independent of localized tumor administration. This platform encapsulates a photosensitizer-conjugated, light-activated toll-like receptor (TLR) agonist, which induces immunogenic cell death and triggers a pro-inflammatory TME conducive to antigen capture upon light irradiation.

View Article and Find Full Text PDF

The tumor microenvironment is a dynamic eco system where cellular interactions drive cancer progression. However, inferring cell-cell communication from non-spatial scRNA-seq data remains challenging due to incomplete li gand-receptor databases and noisy cell type annotations. H ere, we propose scGraphDap, a graph neural network frame work that integrates functional state pseudo-labels and graph structure learning to improve both cell type annotation an d CCC inference.

View Article and Find Full Text PDF

Systemic Delivery of an mRNA-Encoding, Tumor-Activated Interleukin-12 Lock to Eliminate Tumors and Avoid Immune-Related Adverse Events.

Nano Lett

September 2025

Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China.

Interleukin-12 (IL-12) is a robust proinflammatory cytokine that activates immune cells, such as T cells and natural killer cells, to induce antitumor immunity. However, the clinical application of recombinant IL-12 has been limited by systemic immune-related adverse events (irAEs) and rapid degradation. To address these challenges, we employed mRNA technology to encode a tumor-activated IL-12 "lock" fusion protein that offers both therapeutic efficacy and systemic safety.

View Article and Find Full Text PDF

The effect of CD40 agonist antibody therapy on the pancreatic cancer microenvironment.

Naunyn Schmiedebergs Arch Pharmacol

September 2025

Department of Gastroenterology, Jinhua Central Hospital, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, Zhejiang, China.

The fourth leading cause of cancer-related fatalities in the USA is pancreatic ductal adenocarcinoma (PDAC), a particularly deadly illness that is resistant to immunotherapy. One of the Main Obstacles in cancer research is developing better treatments for PDAC, which has the lowest 5-year survival rate of any malignancy. Anti-CTLA-4, anti-PD-L1, and anti-PD-1 immune checkpoint blockade medications also have poor results in these patients, which may indicate the presence of other immunosuppressive mechanisms in the pancreatic tumor microenvironment (TME).

View Article and Find Full Text PDF

Viral warfare: unleashing engineered oncolytic viruses to outsmart cancer's defenses.

Front Immunol

September 2025

Department of Pathological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States.

Oncolytic virotherapy (OVT) has emerged as a promising and innovative cancer treatment strategy that harnesses engineered viruses to selectively infect, replicate within, and destroys malignant cells while sparing healthy tissues. Beyond direct oncolysis, oncolytic viruses (OVs) exploit tumor-specific metabolic, antiviral, and immunological vulnerabilities to reshape the tumor microenvironment (TME) and initiate systemic antitumor immunity. Despite promising results from preclinical and clinical studies, several barriers, including inefficient intratumoral virus delivery, immune clearance, and tumor heterogeneity, continue to limit the therapeutic advantages of OVT as a standalone modality and hindered its clinical success.

View Article and Find Full Text PDF