bims-exocan Biomed News
on Exosomes roles in cancer
Issue of 2025–03–30
seven papers selected by
Muhammad Rizwan, COMSATS University



  1. J Transl Med. 2025 Mar 25. 23(1): 369
       BACKGROUND/AIM: Metastasis is the leading cause of mortality for colorectal cancer (CRC). Cancer-derived exosomes are widely recognized as the primary catalysts behind the development of pre-metastasis niche (PMN) in distal sites. However, the exact mechanism behind this process in CRC remains elusive. This study aimed to investigate the function and mechanisms underlying the role of exosomal miR-188-3p in activating hepatic stellate cells (HSCs) to develop the PMN and promote liver metastasis.
    METHODS: We extracted exosomes from CRC cells using ultracentrifugation. Exosomes were identified using transmission electron microscopy, nanoparticle tracking analysis, and Western blot. Exosome uptake was assessed using fluorescence tracing, exosome PKH67 staining, and real-time quantitative PCR. The effects of CRC cell-derived exosomes on HSCs migration were evaluated using Transwell migration and wound healing assays. Key differentially expressed miRNAs were screened from the GEO database, and bioinformatics prediction along with dual-luciferase reporter assays were used to identify downstream target genes of miR-188-3p. Downstream related proteins of the target genes were detected by Western blot. In vivo, the distribution of exosomes and activation of HSCs in the liver were explored by tail vein injection of exosomes into nude mice. Further, the impact of exosomal miR-188-3p on liver metastasis was investigated using a spleen injection liver metastasis model. Finally, the expression levels of miR-188-3p in exosomes from CRC patient plasma were determined by real-time quantitative PCR, and the relationship between the expression of miR-188-3p in plasma exosomes and CRC prognosis was analyzed.
    RESULTS: The expression level of miR-188-3p within plasma exosomes demonstrated a statistically significant increase in CRC with liver metastasis compared to those without liver metastases. We also demonstrated the transferability of miR-188-3p from CRC cells to HSCs cells via the exosomes. Exosomal miR-188-3p plays a pivotal role in orchestrating the establishment of PMN through targeting PHLPP2 to activate HSCs before tumor metastasis. Exosomal miR-188-3p was found to actively foster in vivo metastasis of CRC. Additionally, plasma exosomal miR-188-3p potentially serves as a viable blood-based biomarker for CRLM.
    CONCLUSION: Exosomal miR-188-3p derived from CRC cells can promote liver metastasis by activating HSCs to form a PMN through targeting PHLPP2 to activate the AKT/mTOR pathway. These results offer a new perspective on the mechanisms driving CRLM.
    Keywords:  Colorectal cancer; Exosome; Hepatic stellate cells; Liver metastasis; Pre-metastatic niche
    DOI:  https://doi.org/10.1186/s12967-025-06334-4
  2. Cancers (Basel). 2025 Mar 10. pii: 940. [Epub ahead of print]17(6):
      Exosomes have emerged as pivotal players in precision oncology, offering innovative solutions to longstanding challenges such as metastasis, therapeutic resistance, and immune evasion. These nanoscale extracellular vesicles facilitate intercellular communication by transferring bioactive molecules that mirror the biological state of their parent cells, positioning them as transformative tools for cancer diagnostics and therapeutics. Recent advancements in exosome engineering, artificial intelligence (AI)-driven analytics, and isolation technologies are breaking barriers in scalability, reproducibility, and clinical application. Bioengineered exosomes are being leveraged for CRISPR-Cas9 delivery, while AI models are enhancing biomarker discovery and liquid biopsy accuracy. Despite these advancements, key obstacles such as heterogeneity in exosome populations and the lack of standardized isolation protocols persist. This review synthesizes pioneering research on exosome biology, molecular engineering, and clinical translation, emphasizing their dual roles as both mediators of tumor progression and tools for intervention. It also explores emerging areas, including microbiome-exosome interactions and the integration of machine learning in exosome-based precision medicine. By bridging innovation with translational strategies, this work charts a forward-looking path for integrating exosomes into next-generation cancer care, setting it apart as a comprehensive guide to overcoming clinical and technological hurdles in this rapidly evolving field.
    Keywords:  biomarkers; cancer diagnostics; drug delivery; exosomes; extracellular vesicles (EVs); immune modulation; liquid biopsies; precision medicine; therapeutic resistance; tumor microenvironment (TME); tumor-derived exosomes (TDEs)
    DOI:  https://doi.org/10.3390/cancers17060940
  3. Cells. 2025 Mar 11. pii: 411. [Epub ahead of print]14(6):
      Cancer is one of the leading causes of death worldwide. Evidence indicates that extracellular vesicles are involved in cancer development and may be used as promising biomarkers in cancer detection. Concomitantly, saliva constitutes a non-invasive and inexpensive source of biomarkers. This systematic review investigates the use of salivary extracellular vesicles in detecting cancers located outside of the head and neck. PubMed, Web of Science, Scopus, and Embase were thoroughly searched from database inception to 16 July 2024. Data from sixteen eligible studies were analyzed, including glioblastoma, lung, esophageal, gastric, prostate, hepatocellular, breast, and pancreatobiliary tract cancers. The findings highlight strong diagnostic potential for lung and esophageal cancers, where specific exosomal RNAs and proteins demonstrated high accuracy in distinguishing cancer patients from healthy individuals. Additionally, biomarkers in glioblastoma showed prognostic value, while those in hepatocellular and pancreatobiliary cancers exhibited potential for early detection. However, gastric and prostate cancer biomarkers showed limited reliability, and breast cancer biomarkers require further validation. In conclusion, salivary extracellular vesicles present potential in non-invasive detection across multiple cancer types; however, their diagnostic power needs further research, including standardization and large-scale validation.
    Keywords:  biomarkers; cancer; diagnosis; extracellular vesicles; saliva
    DOI:  https://doi.org/10.3390/cells14060411
  4. Sci Rep. 2025 Mar 22. 15(1): 9928
      This study aimed to elucidate the role of microRNA-92a-3p (miR-92a-3p) in the pathogenesis of adenomyosis. We examined how miR-92a-3p, found in exosomes derived from ectopic lesions, influences the behaviour of endometrial cells, dorsal root ganglion (DRG), and human umbilical vein endothelial cells (HUVECs) and explored its potential as a non-invasive biomarker. Our findings revealed that miR-92a-3p was significantly upregulated in exosomes derived from ectopic adenomyotic lesions. This upregulation correlated with enhanced migration and invasion of eutopic endometrial cells, DRG, and HUVECs. Furthermore, this study demonstrated a significant correlation between miR-92a-3p levels in urinary exosomes and the clinical symptoms of adenomyosis, suggesting its potential as a non-invasive biomarker for the disease. This study elucidated an exosomal signalling process involving miR-92a-3p that drives pathological infiltration and angiogenesis to promote adenomyosis progression. Our findings highlight upregulated miR-92a-3p in biofluid exosomes as a promising non-invasive biomarker for diagnosing and monitoring adenomyosis and unveil novel targets and strategies for improved clinical management.
    Keywords:  Adenomyosis; Biomarker; Exosomes; miR-92a-3p
    DOI:  https://doi.org/10.1038/s41598-024-84608-5
  5. Acta Biochim Biophys Sin (Shanghai). 2025 Mar 28.
      Studies of cell-to-cell activities in the tumor microenvironment (TME) have identified multiple potential targets for oncotherapy. The interplay between tumor cells and neighboring cancer-associated fibroblasts (CAFs) persists in all stages of tumor progression. In this study, we reveal that exosomes from breast cancer cells can be endocytosed into fibroblasts and transform normal fibroblasts (NFs) into CAFs and that the ability of exosomes from highly metastatic breast cancer cells is greater than that of those from poorly metastatic breast cancer cells. Further investigation reveals that exosomes from highly metastatic breast cancer cells contain much more miR-105-5p than those from poorly metastatic breast cells do and that exosomal miR-105-5p facilitates the transformation of NFs to CAFs. A detailed study reveals that RBMY1A1-dependent sorting of miR-105-5p into fibroblasts and subsequent internalization of miR-105-5p promote the transformation of NFs to CAFs by downregulating LATS2 expression and activating NF-κB signaling, which concurrently facilitates the EMT of breast cancer cells. Thus, our results indicate that exosomal miR-105-5p may be a potential target for novel therapeutic strategies to prevent the coevolution of breast cancer cells and CAFs.
    Keywords:  breast cancer; cancer-associated fibroblasts; exosomes; miRNAs; tumor microenvironment
    DOI:  https://doi.org/10.3724/abbs.2025017
  6. Pharmaceuticals (Basel). 2025 Mar 05. pii: 371. [Epub ahead of print]18(3):
      Background: Identification of targetable biomarkers to improve early disease detection and overall patient outcomes is becoming an urgent need in clinical oncology. Ovarian cancer (OC) has one of the highest mortality rates among gynecological cancers. It is asymptomatic and almost always diagnosed at an advanced stage (III or IV), leading to a 5-year survival rate of approximately 35%. Methods: Current therapeutic approaches for OC are very limited and mainly consist of cytoreductive surgery and cisplatin plus taxane-based chemotherapy. No gender and tumor specific biomarkers are known. Exosomes, lipid bilayer vesicles of endocytic origin secreted by most cell types, represent sources of information for their involvement in the onset and progression of many diseases. Hence, research on exosome contents as tools and targets in precise oncology therapy provides knowledge essential to improving diagnosis and prognosis of the disease. Results: This review attempts to give an overview of how exosomes are implicated in ovarian carcinoma pathogenesis to trigger further cancer exosome-based investigations aimed at developing ovarian cancer fine-tuning diagnostic methodologies. Conclusions: It is essential to investigate exosome-based cancer drugs to advance understanding, improve treatment plans, create personalized strategies, ensure safety, and speed up clinical translation to increase patients' overall survival and quality of life. Papers published in PubMed and Web of Science databases in the last five years (2020-2024) were used as a bibliographic source.
    Keywords:  early diagnosis; exosomes; ovarian cancer; theragnostic; treatment
    DOI:  https://doi.org/10.3390/ph18030371
  7. J Nanobiotechnology. 2025 Mar 21. 23(1): 232
      Lung cancer, known for its high morbidity and mortality rates, remains one of the most critical health challenges globally. Conventional treatment options, such as chemotherapy and surgery, are often limited by high costs, significant side effects, and often yield a poor prognosis. Notably, recent research has shed light on the potential therapeutic roles of exosomes, which essentially influence lung cancer's development, diagnosis, treatment, and prognosis. Exosomes have been revealed for their exceptional properties, including natural intercellular communication, excellent biocompatibility, minimal toxicity, prolonged blood circulation ability, and biodegradability. These unique characteristics position exosomes as highly effective drug delivery systems, nanotherapeutics, and potential diagnostic and prognostic biomarkers in lung cancer. This review provides a comprehensive review of the physiological and pathological roles of exosomes in lung cancer, emphasizing their potential as innovative diagnostic biomarkers, therapeutics, and delivery platforms. By harnessing their unique properties, exosomes are poised to revolutionize the diagnosis and treatment of lung cancer, offering a promising avenue for more personalized and effective therapies.
    Keywords:  Diagonosis; Drug delivery; Exosomes; Lung cancer; Nanotherapeutics
    DOI:  https://doi.org/10.1186/s12951-025-03306-0