Department Of Pharmacology, Dr. Moopan’s College of Pharmacy, Wayanad.
Exosomes are nanosized extracellular vesicles secreted by diverse cell types, playing pivotal roles in intercellular communication, disease progression, and therapeutic innovation. Their unique biogenesis, heterogeneous composition, and ability to transport proteins, lipids, and nucleic acids make them highly versatile mediators of physiological and pathological processes. This review provides a comprehensive overview of exosome biology, including their formation, maturation, isolation techniques, and protein composition. The functional significance of exosomes in health and disease pathogenesis—ranging from cancer and neurodegenerative disorders to fibrosis and immune modulation—is highlighted. Furthermore, their therapeutic potential is explored in the context of regenerative medicine, immunomodulation, and targeted drug delivery, underscoring their advantages as natural nanocarriers with biocompatibility and low immunogenicity. Advances in exosome-based therapies, challenges in clinical translation, and the regulatory landscape, including FDA status, are critically discussed. Finally, future directions emphasize personalized medicine, large-scale production, and emerging applications in diagnostics, aesthetics, and wellness. Collectively, exosomes represent a transformative frontier in modern medicine, bridging fundamental biology with translational therapeutics.
Exosomes are small extracellular vesicles (30–150 nm) secreted by a wide variety of cell types into the extracellular matrix. Once considered merely cellular waste products, they are now understood to play a crucial role in intercellular communication by transporting proteins, lipids, and nucleic acids to recipient cells, thereby influencing cellular behavior and function. Their natural ability to deliver bioactive molecules with high biocompatibility, low immunogenicity, and stability makes them highly attractive candidates for therapeutic applications. Compared to synthetic delivery systems such as liposomes or polymers, exosomes offer superior advantages in targeting specificity, reduced toxicity, and prolonged circulation. Despite their promise, isolating exosomes remains a technical challenge due to their heterogeneity of origin and variability in shape. Advances in proteomics and isolation technologies are steadily addressing these issues, enhancing yield, purity, and characterization. Functionally, exosomes are implicated in both physiological regulation and pathological processes, including cancer progression, cardiovascular disease, neurological disorders, and liver disease, through mechanisms such as modulation of protein function, gene expression, and immune signaling. In therapeutic contexts, exosomes represent a safer and less invasive alternative to conventional treatments, with potential applications in diagnostics, regenerative medicine, drug delivery, and personalized therapy. They hold the capacity to transform disease understanding, improve drug discovery, and revolutionize modern medicine. However, significant gaps remain in understanding their biology, mechanisms, and clinical translation, highlighting the need for further research to overcome technical, biological, and regulatory barriers.
Biogenesis Of Exosomes
Fig No:1 Biogenesis of exosomes
Exosomes are small extracellular vesicles generated within multivesicular bodies (MVBs) through inward budding of late endosomal membranes, which produce intraluminal vesicles (ILVs). These ILVs encapsulate proteins and cytosolic components and are either released as exosomes upon fusion of MVBs with the plasma membrane or sent to lysosomes for degradation. Exosomes typically appear cup-shaped under preparation artifacts but spheroid under TEM and exhibit densities ranging from 1.13 g/mL (B cell-derived) to 1.19 g/mL (epithelial cell-derived). Their formation is mainly attributed to the endosomal sorting complex required for transport (ESCRT) pathway, a coordinated protein machinery. ESCRT-0 recognizes ubiquitinated cargo, ESCRT-I and -II promote membrane deformation, and ESCRT-III drives vesicle budding and scission with the help of the ATPase Vps4. Proteins such as Alix, TSG101, and CHMP4 play crucial roles in both membrane remodeling and selective cargo sorting. However, alternative ESCRT-independent mechanisms also exist. These involve ceramide-enriched lipid microdomains, which induce negative membrane curvature and promote budding, as well as tetraspanins (CD81, CD9, CD63) that regulate receptor organization and cargo loading. Thus, both protein- and lipid-driven processes contribute to exosome biogenesis, varying with cell type and context. In contrast, other extracellular vesicles differ in origin and properties. Microvesicles (100–1000 nm, density 1.25–1.30 g/mL) bud directly from the plasma membrane, often produced by platelets, endothelial cells, or red blood cells. Apoptotic bodies (1–5 μm, density 1.18–1.28 g/mL) are released during apoptosis and contain organelles, membranes, and cytosolic fragments. Unlike these larger and heterogeneous vesicles, exosomes’ small, uniform structure allows them to avoid rapid clearance by the mononuclear phagocyte system, remain longer in circulation, and serve as highly effective mediators of cell–cell communication.
Isolation And Characterization of Exosomes
The isolation and characterization of exosomes are crucial steps in both research and clinical applications, as they enable the accurate study of vesicle composition and function. A variety of isolation techniques are used, each offering a balance between yield, purity, and scalability. Ultracentrifugation-based methods remain the most widely adopted; differential ultracentrifugation separates vesicles based on size and density through sequential spins, though contamination with lipoproteins is a persistent issue. Density-gradient centrifugation, often using sucrose or iodixanol layers, improves purity but is labor-intensive and time-consuming. Size-based methods such as ultrafiltration employ membranes with defined pore sizes for rapid separation, while tangential flow filtration minimizes clogging and preserves vesicle integrity. Size-exclusion chromatography (SEC) uses porous gel matrices to separate exosomes from smaller contaminants like proteins and cytokines, and when combined with ultracentrifugation, further enhances purity. Polymer precipitation with polyethylene glycol (PEG) provides a simple and cost-effective approach but carries the risk of co-precipitating non-exosomal components such as lipoproteins and viruses. Immunoaffinity capture techniques use antibodies against surface markers like CD9 or CD63, immobilized on magnetic beads, to selectively isolate high-purity exosome subpopulations, although they may exclude vesicles lacking those specific markers. Recently, emerging technologies such as microfluidic platforms, nanolithography, and electro-deposition have enabled integrated isolation and analysis, offering high sensitivity and minimal sample requirements, and are increasingly applied in areas like cancer diagnostics. Once isolated, exosomes are characterized using complementary analytical techniques. Biophysical methods such as nanoparticle tracking analysis (NTA) measure size and concentration based on Brownian motion, while dynamic light scattering (DLS) provides size distribution data but is less reliable for heterogeneous samples. Imaging approaches including transmission electron microscopy (TEM), scanning electron microscopy (SEM), and atomic force microscopy (AFM) validate vesicle morphology, size, and surface topology at nanometer resolution. Molecular profiling techniques, such as flow cytometry, identify surface antigens but are limited by the detection threshold of conventional cytometers for particles below 300 nm, whereas western blotting confirms the presence of characteristic exosomal proteins like Alix and TSG101 to verify origin. Advanced analytical tools such as Raman spectroscopy allow non-destructive chemical fingerprinting of vesicles, and microfluidic biosensors enable real-time, high-throughput profiling of exosomal cargo. Together, these diverse strategies provide a comprehensive toolkit for the efficient isolation and detailed characterization of exosomes, supporting their growing relevance in diagnostics, prognostics, and therapeutic delivery systems.
Protein Composition O Exosomes
Fig No:2 Protein composition of exosomes
The protein composition of exosomes is highly specialized and reflects their biogenetic origin. Central protein families consistently identified in exosomes include tetraspanins (CD9, CD63, CD81), ESCRT-related proteins (e.g., Alix, TSG101), syntenin-1, and cytoskeletal proteins, which together contribute to exosomal structure and trafficking. In addition, other functionally relevant groups such as Rab GTPases, SNARE proteins, and cell type–specific proteins are frequently enriched. Exosomes are known to contain selected membrane and cytosolic proteins but are generally devoid of nuclear proteins when compared with their parental cells. Quantitative estimates suggest that a single exosome may contain approximately 20,000 protein molecules, although this number is influenced by vesicle size and the molecular packing parameters. Functionally, exosomal proteins participate in multiple steps of the tightly regulated process of exosome biogenesis. Endosomes, formed by the invagination of the plasma membrane, represent the precursors of exosomes. In the ESCRT-dependent pathway, the endosomal sorting complex required for transport (ESCRT) machinery is essential for the progression of early endosomes into multivesicular bodies (MVBs), which subsequently release intraluminal vesicles as exosomes.
Exosomes As Mediators of Cellular Communication and Disease Pathogenesis
Exosomes serve as critical mediators of intercellular communication by transporting a wide range of bioactive molecules, including proteins, lipids, mRNAs, miRNAs, and DNA, thereby regulating the fate and behaviour of recipient cells at both local and systemic levels. Communication occurs through several mechanisms: exosomes may interact with recipient cells via adhesion molecules that activate signalling cascades, fuse directly with the plasma membrane to release their cargo, or undergo internalization through receptor-mediated endocytosis, phagocytosis, or macropinocytosis. Through these pathways, exosomes deliver genetic material, proteins, and lipids that modulate gene expression and signalling networks, thereby influencing a broad spectrum of physiological and pathological processes. Their functions extend to immune modulation, cell proliferation and survival, tissue remodelling and angiogenesis, and the progression of diseases such as tumour growth and metastasis. Importantly, exosomal communication demonstrates greater specificity and targeting than classical paracrine or endocrine signalling, as they are capable of transmitting complex molecular “messages” to distant recipient cells. Beyond their roles in physiological regulation, exosomes are deeply implicated in disease pathogenesis, where they act as conveyors of pathogenic molecules and modulators of cellular responses. In cancer, tumour-derived exosomes transfer oncogenic proteins, RNAs, and regulatory factors that remodel the tumour microenvironment, enhance angiogenesis, drive epithelial–mesenchymal transition (EMT), and promote metastasis. For example, hepatocellular carcinoma (HCC)-derived exosomes activate the FAK/Src–p38 MAPK and Wnt/β-catenin pathways to stimulate EMT while simultaneously suppressing immune surveillance by impairing NK cell activity, inducing T-cell apoptosis, and promoting M2 macrophage polarization. In neurodegenerative diseases such as Alzheimer’s and Parkinson’s, exosomes facilitate the spread of β-amyloid and α-synuclein aggregates between neurons, disrupting proteostasis and accelerating disease progression. Similarly, in fibrotic diseases, exosomes regulate fibroblast activation, immune cell migration, and extracellular matrix deposition. Macrophage-derived exosomes, for instance, deliver angiotensin II receptors to fibroblasts, driving myofibroblast differentiation in pulmonary fibrosis, while hepatocyte-stromal exosomal crosstalk promotes fibrotic remodelling and tumour progression in liver disease. Moreover, exosomes exert dual roles in immune regulation, either stimulating immune responses through MHC–antigen presentation or dampening immunity by transferring immunosuppressive cargo such as PD-L1 and TGF-β. Given their stability, targeting specificity, and ability to carry diverse molecular cargo, exosomes are increasingly being explored as therapeutic agents and diagnostic biomarkers. Strategies such as blocking oncogenic exosomal miRNAs (e.g., miRNA-21) or enhancing tumour-suppressive exosomal miRNAs (e.g., let-7) show promise in treating fibrosis and cancer. Collectively, these findings highlight exosomes as multifaceted regulators of cell communication that function not only as drivers of pathogenesis but also as potential targets and vehicles for therapy, underscoring their profound importance in both health and disease.
Therapeutic Potential of Exosomes in Regenerative Medicine and Targeted Drug Delivery
Fig No.3 Therapeutic Potential of Exosomes
Exosomes are increasingly recognized as powerful therapeutic tools due to their capacity to deliver bioactive molecules, regulate immune responses, and cross biological barriers. Their applications span regenerative medicine, immunomodulation, targeted drug delivery, and cancer therapy. In regenerative medicine, stem cell-derived exosomes (mesenchymal and neural) facilitate tissue repair, neuroprotection, and angiogenesis. For instance, MSC-derived exosomes aid spinal cord injury recovery by shifting microglial polarization (M1 to M2) through TLR4/NF-κB/PI3K/Akt signaling and promoting angiogenesis via VEGF-A delivery, while neural stem cell-derived exosomes reduce neuroinflammation in Alzheimer’s and Parkinson’s disease through anti-inflammatory miRNAs such as miR-216a-5p. In cardiovascular repair, they mitigate atherosclerosis and heart failure by modulating macrophage activity and suppressing inflammation. Their immunomodulatory potential is equally significant. In multiple sclerosis, MSC-derived exosomes inhibit TLR2/IRAK1/NF-κB signaling to reduce demyelination while promoting oligodendrocyte regeneration. In pulmonary disorders like ARDS, they suppress alveolar inflammation and enhance epithelial regeneration, supporting their role in autoimmune and inflammatory diseases. A major therapeutic frontier is drug delivery, where exosomes outperform synthetic carriers owing to their biocompatibility, stability, natural targeting capacity, and ability to traverse the blood–brain barrier. They can be engineered via passive loading, active loading (electroporation, sonication), or cell engineering to deliver chemotherapeutics (e.g., doxorubicin), nucleic acids (miRNAs, mRNAs), proteins, or gene-editing agents such as CRISPR-Cas systems. This flexibility has fueled interest in their use for neurological therapy, cancer treatment, and gene therapy. In oncology, engineered exosomes can deliver PD-1/PD-L1 inhibitors, tumor antigens, and immunostimulatory molecules, enhancing T-cell activation and reducing tumor immunosuppression. Furthermore, hybrid exosome–nanoparticle systems improve stability, loading efficiency, and pharmacokinetics, broadening therapeutic scope. In summary, exosomes offer unique advantages—low immunogenicity, cargo protection, precise targeting, and barrier-crossing ability—making them an exceptional platform for next-generation therapies in regenerative medicine, oncology, neurology, cardiovascular repair, and immunomodulation.
Exosome-Based Therapy
Exosome based therapy is an emerging opportunity in targeted drug delivery, regenerative medicine and it is a natural extracellular vesicle to treat various diseases.
The exosome-based therapy has various advantages
Major advantages over Traditional Therapies
Ethical safety: Avoid controversies surrounding stem cell utilization.
Clinical Applications:
Regenerative Medicine
Cancer Therapy
Disease-Specific Treatments
Recent Advancements
Challenges And Limitations of Exosome Based Therapy
1. Manufacturing and Standardization Challenges
2. Biological and Technical Limitations
3. Regulatory and Clinical Challenges
4. Additional Considerations
Column 1: Sr No, Column 2: Challenge type, Column 3: Description.
Sr, No. |
Challenge Type |
Description |
1 |
Manufacturing |
Standardization, scalability problems, potential for contamination |
2 |
Biological/Technical |
Heterogeneity, constrained drug loading, short half-life, inadequate tissue targeting
|
3 |
Regulatory/Clinical |
Regulatory uncertainty, expense of clinical trials, limited large-scale data
|
4 |
Ethical/Quality |
Ethical sourcing, high-quality control demands
|
5 |
Potential for Off-Target Effects |
Nonspecific cargo loading |
FDA Approval Status
Major Types and Strategies of Exosome Formulations
Intravenous administration results in rapid hepatic clearance, making exosomes particularly useful for liver-targeted therapies. To expand systemic applications, novel strategies are under investigation to minimize liver capture and enhance targeting to other organs.
Exosomes can be bioengineered to carry therapeutic RNAs, proteins, or small molecules, and may be modified to display targeting ligands on their surface. These strategies improve selective tissue targeting, intracellular uptake, and therapeutic activity.
Synthetic or semi-synthetic vesicles designed to mimic the structure and function of natural exosomes. They address challenges such as low natural yield, immunogenicity, and toxicity while allowing scalable, reproducible production.
Therapeutics may be incorporated into exosomes either indirectly (loading parental cells, which then release drug-loaded exosomes) or directly (loading isolated exosomes through techniques such as co-incubation or electroporation). Challenges remain regarding loading efficiency, membrane integrity, and drug stability.
Advanced purification methods (e.g., anion-exchange chromatography) and removal of impurities improve exosome shelf-life and product stability, which is critical for commercial scalability and clinical reliability.
Specialized exosome formulations are under development for targeted indications—for example, regulatory T-cell-derived exosomes engineered to deliver VEGF antibodies for ocular disorders such as choroidal neovascularization, improving precision and efficacy.
Column 1: Sr No, Colum 2: Brand or Product Nate, Column 3: Company/Developer, Column 4: Source/Type, Column 5: Main Application/Indication, Column 6: Status/Notes.
Sr. No. |
Brand/ Product Name |
Company/ Developer |
Source/ Type |
Main Application/ Indication |
Status / Notes |
1 |
XoGlo® |
Kimera Labs |
MSC-derived exosomes |
Regenerative, anti-inflammatory, cosmeceuticals |
not FDA-approved |
2 |
ExoFlo |
Direct Biologics |
Bone marrow MSC-derived EVs |
Anti-inflammatory, regenerative |
Clinical trials; not FDA-approved |
3 |
DeliverEX platform |
Evox Therapeutics |
Engineered exosomes |
Rare genetic/metabolic diseases |
Pipeline; multiple candidates |
4 |
AGLE-102 |
Aegle Therapeutics |
Not Specified (likely MSC derived) |
Dermatology, Wound healing |
Clinical pipeline |
5 |
EXO-CD24 (Nano24) |
Nano 24 (OBCTCD 24) |
Not Specified |
Inflammatory/immune modulation |
Clinical pipeline |
6 |
CAP-1002 |
Capricor therapeutics |
Cardio sphere-derived exosomes |
Cardiovascular, Duchenne muscular dystrophy |
Clinical pipeline |
7 |
Exo-101 |
Exogenous Therapeutics |
Umbilical cord blood derived exosomes |
Tissue regeneration, inflammatory diseases |
Preclinical/clinical GMP manufacturing |
8 |
CALECIM Professional Serum |
Cell Research Corporation |
Umbilical cord lining- derived exosomes |
Skin rejuvenation (cosmetic) |
Cosmetceutical;not FDA-approved |
9 |
Lifeline Intense Moisture Serum |
Lifeline Skin Care |
Stem cell-derived exosomes |
Skin care (cosmetic) |
cosmetceutical |
Future Directions and Clinical Trial Landscape of Exosome-Based Therapies
Exosomes are rapidly emerging as versatile platforms for therapeutics, diagnostics, and regenerative applications, with ongoing research pointing to a transformative future in precision medicine.
Personalized Drug Delivery and Targeted Therapies: Advances in exosome engineering are enhancing cargo loading, targeting specificity, and therapeutic potency, allowing for precise delivery of drugs, genes, and immune modulators. Integration with gene editing tools such as CRISPR/Cas9 and parent cell engineering further expands their potential.
Diagnostics and Liquid Biopsy: Exosomes are increasingly studied as non-invasive biomarkers for cancer detection, prognosis, and disease monitoring, particularly in breast, lung, colorectal, and prostate cancers. Coupled with AI and big data analytics, exosome-assisted multiplexed detection promises greater diagnostic precision and early intervention capabilities.
Regenerative Medicine and Tissue Repair: Clinical trials are underway testing exosome therapies for osteoarthritis, myocardial infarction, wound healing, and organ regeneration. Their ability to regulate inflammation and promote tissue repair positions exosomes as promising candidates for next-generation regenerative medicine.
Neurological and Immune Disorders : Exosome therapies are in development for neurodegenerative diseases (e.g., Parkinson’s, Alzheimer’s, stroke) and immune-mediated disorders, leveraging their neuroprotective and immunomodulatory activities to achieve therapeutic benefits.
Aesthetics and Wellness : Beyond clinical medicine, exosomes are entering the aesthetics sector, showing promise in skin rejuvenation, anti-aging, and wound healing. As production scalability improves, bioengineered exosomes are expected to become more accessible in routine skincare and wellness products.
Manufacturing and Scalability : Technological advances such as hollow-fiber bioreactors, microfluidic isolation, and scalable purification methods are addressing the challenges of low yield and high manufacturing costs. These developments pave the way for large-scale, GMP-grade production of exosome formulations suitable for widespread clinical use.
Challenges Ahead: Despite advances, key hurdles remain: standardizing quality control, improving reproducibility, enhancing targeting specificity, addressing immunogenicity concerns, and generating robust clinical trial data. Collaborative efforts between academia, industry, and regulatory bodies will be essential to ensure safe and effective clinical translation.
Pipeline and Clinical Trials (as of July 2025) : The global pipeline of exosome therapies is expanding rapidly, with over 100 active clinical trials exploring applications in oncology, neurology, immune modulation, and regenerative medicine. Notable programs include:
Therapeutic Areas in Development
CONCLUSION
Exosomes represent a transformative frontier in modern medicine, uniquely positioned at the intersection of cellular communication, diagnostics, and therapeutics. Their nanoscale architecture, ability to encapsulate diverse bioactive cargo, and natural biocompatibility provide significant advantages over synthetic delivery systems. Current evidence underscores their therapeutic promise in regenerative medicine, oncology, neurology, and immune modulation, with early clinical trials demonstrating safety and feasibility. However, the translation of exosome-based therapies into clinical practice is constrained by challenges in large-scale manufacturing, standardization, targeted delivery, and regulatory approval. Advances in bioengineering, high-throughput isolation, and GMP-grade production are beginning to address these barriers, paving the way for next-generation, personalized, and disease-specific applications. As the clinical pipeline expands, collaborative efforts between academia, industry, and regulatory agencies will be critical to establish robust safety, efficacy, and quality frameworks. Collectively, exosomes hold immense potential to redefine therapeutic strategies, serving not only as powerful nanocarriers but also as biomarkers and active modulators of disease biology, thereby shaping the future of precision and regenerative medicine.
Conflict Of Interest
The authors declare that they have no conflict of interest.
REFERENCES
Santilna*, Asna Nourin P., Exosomes and Their Therapeutic Promise in Modern Medicine, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 8, 2730-2743 https://doi.org/10.5281/zenodo.16948015