Research on the tumor microenvironment, including immune, stromal, vascular, and extracellular matrix interactions

Tumor Microenvironment Dynamics

The tumor microenvironment (TME) plays a crucial role in cancer progression and therapy resistance. Recent studies have highlighted the impact of bile acids on cancer-associated fibroblasts (CAFs) in cholangiocarcinoma, where excessive bile acids activate GPBAR1 on CAFs, leading to the secretion of CXCL10. This process enhances epithelial-mesenchymal transition (EMT) and metastasis of cholangiocarcinoma cells while creating an immunosuppressive TME by recruiting neutrophils (ref: Huang doi.org/10.1016/j.ccell.2025.05.017/). In ovarian cancer, spatial proteo-transcriptomic profiling has revealed the molecular landscape of borderline ovarian tumors, elucidating the transition from non-invasive serous borderline tumors to low-grade serous cancer and its metastases, thus providing insights into the TME's role in tumor progression (ref: Schweizer doi.org/10.1016/j.ccell.2025.06.004/). Furthermore, multi-omic analyses of gallbladder cancer have identified distinct tumor microenvironments associated with disease progression, revealing spatial-temporal characterizations of cellular compositions that stratify clinical outcomes (ref: Zhou doi.org/10.1038/s41588-025-02236-9/). These findings underscore the complexity of the TME and its influence on tumor behavior and therapeutic responses.

Immunotherapy and Immune Response

Immunotherapy has emerged as a pivotal strategy in cancer treatment, yet challenges remain, particularly in enhancing immune responses against tumors. A combinatorial treatment of cisplatin and temozolomide has been shown to induce hypermutability and enhance immune surveillance in colorectal cancer models, suggesting a potential strategy to overcome immune resistance (ref: Vitiello doi.org/10.1016/j.ccell.2025.05.014/). In metastatic melanoma, patients treated with tumor-infiltrating lymphocytes reported improved health-related quality of life compared to those receiving ipilimumab, indicating the effectiveness of TIL therapy in enhancing patient outcomes (ref: Ten Ham doi.org/10.1016/j.annonc.2025.06.005/). Additionally, a phase 2 trial focusing on mobilizing antigen-presenting mast cells in anti-PD-1-refractory triple-negative breast cancer revealed that mast cell heterogeneity correlates with clinical benefits, emphasizing the need for personalized immunotherapeutic approaches (ref: Wu doi.org/10.1038/s41591-025-03776-7/). These studies collectively highlight the importance of understanding immune dynamics within the TME to optimize immunotherapeutic strategies.

Cancer-Associated Fibroblasts and Stromal Interactions

Cancer-associated fibroblasts (CAFs) are integral to the tumor microenvironment, influencing tumor progression and therapy resistance. In cholangiocarcinoma, bile acids activate CAFs, leading to an immunosuppressive environment that promotes tumor metastasis (ref: Huang doi.org/10.1016/j.ccell.2025.05.017/). The role of CAFs is further underscored in studies of soft-tissue sarcomas, where anti-angiogenic therapies combined with immune checkpoint inhibitors were shown to reshape the TME, enhancing T cell infiltration and tumor immunogenicity (ref: Toulmonde doi.org/10.1038/s41392-025-02278-9/). Additionally, the functional heterogeneity of CD4+ T cell subtypes in high-grade serous ovarian carcinoma has been characterized, revealing distinct distributions that could inform therapeutic strategies targeting the TME (ref: Zhang doi.org/10.5306/wjco.v16.i5.104138/). These findings illustrate the complex interactions between CAFs and immune cells within the TME, highlighting potential therapeutic targets.

Spatial and Multi-Omics Profiling

Spatial and multi-omics profiling techniques are revolutionizing our understanding of tumor heterogeneity and the tumor microenvironment. High-definition spatial transcriptomics has been employed to analyze immune cell populations in colorectal cancer, providing insights into cellular behavior and responses within the TME (ref: Oliveira doi.org/10.1038/s41588-025-02193-3/). Additionally, the integration of spatial proteomics and transcriptomics in borderline ovarian tumors has elucidated the molecular landscape and progression from non-invasive to invasive cancer forms (ref: Schweizer doi.org/10.1016/j.ccell.2025.06.004/). Multi-omic analyses in gallbladder cancer have identified distinct tumor microenvironments associated with disease progression, revealing spatial-temporal characterizations that stratify clinical outcomes (ref: Zhou doi.org/10.1038/s41588-025-02236-9/). These advanced profiling techniques are essential for uncovering the complexities of tumor biology and guiding personalized therapeutic approaches.

Tumor Metabolism and Microenvironment Interaction

The interplay between tumor metabolism and the microenvironment is critical for cancer progression and therapy response. Studies have shown that hypermutation induced by cisplatin and temozolomide can enhance immune surveillance in colorectal cancer, suggesting a metabolic shift that increases immunogenicity (ref: Vitiello doi.org/10.1016/j.ccell.2025.05.014/). Additionally, the role of lactate in modulating NK cell cytotoxicity within the TME has been highlighted, with strategies aimed at targeting lactylation showing promise in enhancing NK cell function (ref: Jin doi.org/10.1038/s41590-025-02178-8/). Furthermore, histone lactylation has been implicated in cholesterol-linked immunosuppression in pancreatic cancer, indicating that metabolic reprogramming can significantly impact immune responses (ref: Yang doi.org/10.1136/gutjnl-2024-334361/). These findings underscore the importance of metabolic pathways in shaping the TME and influencing therapeutic outcomes.

Therapeutic Strategies and Drug Delivery

Innovative therapeutic strategies and drug delivery systems are essential for improving cancer treatment efficacy. The combination of cisplatin and temozolomide has been shown to induce hypermutability and enhance immune responses in colorectal cancer, suggesting a promising approach to overcome resistance (ref: Vitiello doi.org/10.1016/j.ccell.2025.05.014/). In soft-tissue sarcomas, a phase 2 trial combining regorafenib and avelumab demonstrated the potential of targeting the TME to enhance T cell infiltration and immunogenicity (ref: Toulmonde doi.org/10.1038/s41392-025-02278-9/). Additionally, a novel long-acting IL-2 release platform has been developed to promote antitumor immune responses, highlighting advancements in drug delivery systems (ref: Han doi.org/10.1038/s43018-025-00993-4/). These studies illustrate the ongoing efforts to refine therapeutic strategies and enhance drug delivery mechanisms to improve patient outcomes.

Tumor Heterogeneity and Evolution

Tumor heterogeneity and evolution are critical factors influencing cancer progression and treatment response. Recent research has focused on the spatial proteo-transcriptomic profiling of borderline ovarian tumors, revealing insights into the transition from non-invasive to invasive cancer forms (ref: Schweizer doi.org/10.1016/j.ccell.2025.06.004/). In high-grade serous ovarian carcinoma, the functional heterogeneity of CD4+ T cell subtypes has been characterized, providing potential biomarkers for therapy (ref: Zhang doi.org/10.5306/wjco.v16.i5.104138/). Furthermore, the dynamics of the pancreatic ductal adenocarcinoma microenvironment have been investigated, highlighting the evolution of immunosuppressive mechanisms during metastasis (ref: Liu doi.org/10.1038/s41392-025-02265-0/). These findings emphasize the importance of understanding tumor evolution and heterogeneity to develop effective therapeutic strategies.

Cellular and Molecular Mechanisms in Cancer

Understanding the cellular and molecular mechanisms underlying cancer progression is essential for developing effective therapies. Bile acids have been shown to activate CAFs in cholangiocarcinoma, leading to an immunosuppressive TME that promotes metastasis (ref: Huang doi.org/10.1016/j.ccell.2025.05.017/). Additionally, the combinatorial treatment of cisplatin and temozolomide has been linked to hypermutation and enhanced immune surveillance in colorectal cancer, suggesting a mechanism for overcoming immune resistance (ref: Vitiello doi.org/10.1016/j.ccell.2025.05.014/). The role of HMGA1 in esophageal squamous cell carcinoma has also been elucidated, revealing its impact on inhibiting STING-mediated anti-tumor immunity (ref: He doi.org/10.1038/s41467-025-60221-6/). These studies highlight the complex interplay of molecular mechanisms in cancer and their implications for therapeutic strategies.

Key Highlights

Disclaimer: This is an AI-generated summarization. Please refer to the cited articles before making any clinical or scientific decisions.