bims-exocan Biomed News
on Exosomes roles in cancer
Issue of 2025–12–14
seven papers selected by
Muhammad Rizwan, COMSATS University



  1. 3 Biotech. 2026 Jan;16(1): 15
      Pancreatic ductal adenocarcinoma (PDAC) accounts for 90% of pancreatic cancer (PC). The inefficient early detection and screening methods make PDAC the fourth deadliest cancer worldwide. The adjuvant and neoadjuvant therapies can manage the disease, but often with very low efficacy, resulting in a low 5-year survival rate of just 12%. Site-specific drug targeting and more precise early detection could be the way forward. Biological vehicles, like exosomes, a type of extracellular vesicle, play a crucial role in the development and metastasis of various types of cancer, including PC. By nature, exosomes are nano-sized vesicles secreted by most cells, including cancer cells. They carry biologically active molecules that facilitate cell-cell communication and signaling and are specific for each type of cancer, including PDACs. These PC-secreted exosomes have a unique molecular signature that is being investigated for PC diagnosis. Additionally, these vesicles could be engineered biologically, chemically, and immunologically to identify and target PC-affected sites for site-specific drug delivery. The strategic payload delivery capability of exosomes enhances the bioavailability and specificity of chemotherapeutic drugs. However, significant challenges remain in the clinical application of exosomes as drug carriers and biomarkers. This review summarizes the current understanding of the role of exosomes in PC development, contribution to metastasis, immunomodulation, and chemoresistance in PC. It emphasizes the therapeutic potential in tune with site-specific drug delivery and diagnostic applications of exosome-associated molecular signatures in PC detection.
    Keywords:  Biomarker; Chemoresistance; Drug delivery; Exosome; Immunotherapy; PDAC; Pancreatic cancer
    DOI:  https://doi.org/10.1007/s13205-025-04642-6
  2. Front Immunol. 2025 ;16 1644861
      Prostate cancer (PCa) remains a leading cause of cancer-related mortality in men worldwide, primarily due to its propensity for therapy resistance and metastasis. Emerging evidence underscores that exosomes, nanoscale extracellular vesicles, act as critical mediators of intercellular communication within the tumor microenvironment (TME), particularly via the non-coding RNAs (ncRNAs) they transport. These molecules include microRNAs (miRNAs), circular RNAs (circRNAs), and long non-coding RNAs (lncRNAs). Exosomal ncRNAs drive tumor progression, immune evasion, and therapy resistance by reprogramming neighboring stromal cells, immune cells, and malignant cells. This review systematically examines the multifaceted roles of exosomal ncRNAs in remodeling the prostate cancer tumor microenvironment, focusing on their communication between tumor cells, tumor-stromal cells (including immune cells), and within the pre-metastatic niche preceding bone metastasis. We emphasize their mechanisms of action and clinical relevance. These findings position exosomal ncRNAs as central drivers of prostate cancer progression, revealing novel diagnostic and therapeutic opportunities. Future research must address challenges in standardizing exosome isolation techniques, resolving spatiotemporal heterogeneity, and advancing clinical translation to fully realize the potential of exosome-based strategies in precision oncology.
    Keywords:  exosomes; immune microenvironment; non-coding RNA; prostate cancer; tumor microenvironment
    DOI:  https://doi.org/10.3389/fimmu.2025.1644861
  3. Transl Lung Cancer Res. 2025 Nov 30. 14(11): 5099-5117
       Background and Objective: Lung cancer remains the leading cause of cancer-related mortality worldwide, largely due to late-stage diagnosis and therapy resistance. Despite advances in targeted therapies and immunotherapies, the prognosis remains poor. There is a critical need for minimally invasive biomarkers that enable early detection, prognostic stratification and therapeutic monitoring. Exosomes, a subtype of extracellular vesicles, have emerged as promising candidates in this context given their role in intercellular communication and their selective cargo, which reflects the molecular state of tumor cells. This review aims to summarize the current evidence on the potential of exosomes and their cargo, particularly microRNAs (miRNAs), as diagnostic and prognostic biomarkers as well as therapeutic tools in lung cancer.
    Methods: A comprehensive literature search was conducted using PubMed/MEDLINE and Scopus databases between January 3 and April 30, 2025. Studies published from January 2000 to April 2024 were included if they addressed the role of exosomes in lung cancer diagnosis, prognosis, or therapy. Two reviewers independently screened titles and abstracts and full texts were assessed based on predefined inclusion criteria. Relevant articles were also identified through reference lists.
    Key Content and Findings: Exosomal miRNAs (microRNAs) have shown potential as biomarkers for early detection, disease subtype classification, prognosis and therapy resistance in lung cancer. Multiple studies have identified specific miRNA signatures associated with tumor burden, histological subtypes and clinical outcomes. Exosomes also contribute actively to oncogenesis through promoting angiogenesis, epithelial-mesenchymal transition, immune evasion and drug resistance. Furthermore, exosomes are being investigated both as therapeutic targets and delivery systems due to their ability to transfer functional biomolecules selectively and safely. Despite these advances, challenges remain regarding standardization of isolation methods, heterogeneity in miRNA signatures and clinical validation.
    Conclusions: Exosomes represent a dynamic and promising platform for improving the diagnosis, prognosis and treatment of lung cancer. Although technical and translational hurdles remain, their integration into clinical practice may enhance personalized and precision oncology strategies. Continued research and technological advancements are necessary to fully unlock their potential in routine cancer care.
    Keywords:  Lung cancer; biomarkers; extracellular vesicles (EVs); liquid biopsy; microRNA (miRNA)
    DOI:  https://doi.org/10.21037/tlcr-2025-557
  4. Cancer Cell Int. 2025 Dec 08.
      The reprogramming and polarization of cells within the tumor microenvironment (TME) are fundamental processes that critically influence the dynamics of tumor progression and response to therapy. These processes can either promote tumor evasion and metastasis or mount effective anti-tumor responses, thereby determining the overall tumor landscape. At the center of this dynamic interplay are exosomes that facilitate crucial intercellular communication by transferring bioactive molecules such as proteins, lipids, and nucleic acids. While the roles of individual TME components in cancer progression have been extensively characterized, the precise mechanisms through which the reprogramming of these elements favors tumor advancement remain inadequately elucidated. This review investigates the complex functions of exosomal signaling in the reprogramming and polarization of TME constituents, highlighting their dual capacity to orchestrate tumor-promoting and tumor-suppressing signals. This synthesis aims to elucidate how the modulation of TME cell behavior impacts cancer progression and contributes to the broader understanding of tumor biology.
    Keywords:  Cellular reprogramming; Exosomal dynamics; Intercellular communication; Polarization; TME
    DOI:  https://doi.org/10.1186/s12935-025-04105-1
  5. Genes Dis. 2026 Mar;13(2): 101682
      Exosome-associated tRNA-derived fragments (tRFs) play crucial roles in cancer progression, influencing cell proliferation, metastasis, immune evasion, and drug resistance. Due to their stability and specific expression in bodily fluids, exosomal tRFs hold great promise as non-invasive biomarkers for early cancer diagnosis, treatment monitoring, and prognosis evaluation. Additionally, tRFs present opportunities as therapeutic targets, with potential to modulate oncogenic processes, such as controlling gene expression and modulating cellular signaling pathways. This review explores the diverse functions of exosome-associated tRFs in tumor biology, highlighting their potential for clinical applications, including their use as diagnostic tools and their role in therapy. It also addresses the challenges that remain in standardizing detection methods and validating their efficacy in clinical settings, such as variability in isolation techniques and the need for large-scale studies. Advanced technologies and further research will be key to unlocking their potential in personalized cancer therapy, ultimately aiming to integrate tRFs into routine clinical practice for better patient outcomes.
    Keywords:  Cancer biomarkers; Clinical application; Exosome-associated tRFs; Personalized cancer therapy; Tumor progression
    DOI:  https://doi.org/10.1016/j.gendis.2025.101682
  6. Biomed Pharmacother. 2025 Dec 09. pii: S0753-3322(25)01083-2. [Epub ahead of print]193 118889
      Exosomes are a type of extracellular vesicle, shed by almost all reproductive tissues, and critically mediate intercellular communication in both physiological and pathological aspects of human reproduction. Exosomes can transport biological cargo such as proteins, lipids, and many RNA species, and thus they regulate reproduction-related events, including gametogenesis, fertilization, embryo implantation, and development. This review will emphasize the mechanistic roles of exosomes in the context of female and male reproductive physiology, as well as embryo-endometrial communication and immune modulation in early pregnancy. In assisted reproductive technologies (ART), exosomes are being considered for their diagnostic and therapeutic capacity. They are currently being developed as non-invasive biomarkers of oocyte quality, embryo viability, and endometrial receptivity, and have also been used to enhance embryo culture condition, improve cryopreservation outcomes, and deliver targeted therapies. In addition, in regenerative medicine applications for fertility treatment, particularly in endometrial repair, and restoration of ovarian function, exosomes are being used as flexible agents. However, despite recent advances, several challenges hinder clinical implementation, including inconsistencies in research methodologies, unclear regulatory frameworks, and limited clinical translation. Future avenues of exosome research need to concentrate on standardizing exosome isolation methods, the engineering of exosome vesicles for targeted delivery, the and incorporation of omics technologies for individualizing reproductive care. Collectively, exosomes are a new frontier in ART innovation, providing another modality for increasing reproductive success through precision medicine.
    Keywords:  Assisted Reproductive Technologies; Exosomes; Reproduction
    DOI:  https://doi.org/10.1016/j.biopha.2025.118889
  7. Cancer Med. 2025 Dec;14(23): e71421
       BACKGROUND: Cancer persists as a leading cause of global mortality, largely due to the immunosuppressive tumor microenvironment (TME) that facilitates tumor progression and therapy resistance. M2 macrophages dominate this immunosuppressive landscape, and emerging evidence highlights tumor-derived exosomes (TEXs) as critical mediators of macrophage M2 polarization via delivery of noncoding RNAs (ncRNAs), including miRNAs, lncRNAs, and circRNAs. These TEX-ncRNA networks activate key signaling pathways (e.g., JAK/STAT, PI3K/AKT, NF-κB) to sustain immunosuppression and pro-tumorigenic responses. Understanding the molecular intricacies of TEX-driven M2 polarization is essential for advancing immunotherapeutic strategies.
    METHODS: This review systematically analyzes literature (2019-2024, from PubMed and Web of Science) on the molecular mechanisms by which TEX-derived ncRNAs drive M2 polarization and their interplay with immunotherapies.
    RESULTS AND CONCLUSION: This review contains 142 citations, 60 of which are detailed examples of this mechanism. Our analysis of the literature shows that TEXs deliver specific ncRNAs to macrophages, reprogramming them toward an M2 phenotype via pathways such as PTEN/PI3Kγ, Wnt/β-catenin, and STAT3. This polarization amplifies immunosuppressive factor secretion and promotes tumor metastasis, chemoresistance, and immune evasion. These insights provide a theoretical foundation for novel TME-targeted therapies, potentially improving outcomes in refractory cancers.
    Keywords:  M2 macrophage polarization; exosomes; immune cells; immunosuppressive tumor microenvironment; immunotherapy resistance; non‐coding RNA
    DOI:  https://doi.org/10.1002/cam4.71421