Exosomes — nanoscale vesicles naturally secreted by cells — have emerged as powerful candidates for targeted drug delivery. Offering advantages in biocompatibility, stability, and tissue specificity, they hold promise for revolutionizing the treatment of cancer, neurological disease, and beyond. Yet translating their potential into clinical success remains complex, with critical hurdles in isolation, loading efficiency, scalability, and regulatory classification. This review explores the evolving landscape of exosome-based therapeutics, surveying key challenges and the latest technological advances that are helping realize their promise.
The Case for Exosomes: Meeting the Demands of Modern Drug Delivery
The therapeutic landscape is rapidly evolving, with increasing demand for drug delivery systems that are not only effective and precise but also minimally invasive, biocompatible, and capable of overcoming traditional biological barriers. This demand has intensified in the context of complex diseases, such as cancer and neurodegeneration, which often require delivery of sensitive or large biomolecules — such as RNA, proteins, or gene editing tools — to difficult-to-reach sites. Conventional delivery platforms like synthetic nanoparticles, liposomes, and viral vectors, while valuable, carry limitations related to immunogenicity, scalability, stability, and off-target effects. In this context, exosomes have emerged as a promising alternative, offering a biologically derived, inherently functional vehicle for targeted drug delivery.
Exosomes are nanoscale extracellular vesicles, typically 30–150 nanometers in diameter, naturally secreted by most cell types. They originate from the endosomal system, forming as intraluminal vesicles within multivesicular bodies that are eventually released into the extracellular environment. Rich in lipids, proteins, and nucleic acids, exosomes play a central role in intercellular communication and can influence physiological and pathological processes systemically. Their endogenous origin equips them with several features that are highly desirable for drug delivery applications: they exhibit low immunogenicity, possess innate stability in circulation, and demonstrate the ability to cross biological barriers, such as the blood–brain barrier (BBB). These properties distinguish them from many synthetic carriers and make them uniquely suited to address delivery challenges across multiple therapeutic areas.
Interest in exosome-based delivery has grown particularly strong in fields like oncology, neurology, regenerative medicine, and immunotherapy. In cancer, exosomes are being explored as carriers of small molecule chemotherapeutics and RNA-based therapies, with the goal of reducing off-target toxicity and improving tumor penetration. In neurology, their natural capacity to traverse the BBB opens new avenues for delivering RNA- and protein-based treatments for conditions like Parkinson’s and Alzheimer’s diseases. In regenerative medicine, mesenchymal stem cell (MSC)-derived exosomes are being investigated for their ability to modulate inflammation and promote tissue repair, while in immunotherapy, engineered exosomes are under study for antigen delivery and immune cell reprogramming.
Although most exosome-based therapeutics remain in preclinical development, several candidates have progressed to early-phase clinical trials, and a number of biotech companies are working to develop platform technologies that enable scalable, GMP-compliant exosome manufacturing. As the field matures, it is becoming increasingly clear that exosomes are more than just an interesting biological phenomenon — they are a versatile and potentially transformative modality for drug delivery and therapeutic intervention, deserving of focused attention from both industry and regulatory bodies alike.
Harnessing Nature’s Delivery System
What Are Exosomes?
Exosomes are a distinct subclass of extracellular vesicles generated through a highly regulated endosomal pathway. They originate within cells as intraluminal vesicles formed by the inward budding of the limiting membrane of multivesicular bodies. Upon fusion of these multivesicular bodies with the plasma membrane, exosomes are released into the extracellular space, where they can interact with nearby or distant cells via circulation. Typically ranging in size from 30 to 150 nanometers, exosomes are distinguished by their enriched content of specific lipids, proteins, and nucleic acids reflective of their parent cells.
The exosomal membrane is composed of a lipid bilayer that incorporates various surface proteins, such as tetraspanins (CD63, CD81, CD9), heat shock proteins, integrins, and major histocompatibility complex (MHC) molecules. Internally, exosomes carry a diverse payload of bioactive molecules, including mRNA, microRNA (miRNA), long noncoding RNA, DNA fragments, and enzymes. These components contribute to exosomes’ critical function in mediating intercellular communication and regulating various physiological and pathological processes. Through uptake mechanisms such as endocytosis, phagocytosis, or direct membrane fusion, exosomes can deliver functional cargos to recipient cells, influencing gene expression, immune responses, and cellular behavior.1,2
Advantages for Drug Delivery
Several intrinsic properties make exosomes particularly attractive as drug delivery vehicles. Their biocompatibility is perhaps their most compelling feature: because they are derived from endogenous sources, exosomes are generally well tolerated and exhibit low immunogenicity compared with synthetic nanocarriers or viral vectors. This makes them ideal for repeated or long-term administration, especially in sensitive contexts like immunotherapy or neurodegenerative disease.
Exosomes also demonstrate remarkable stability in circulation. The lipid bilayer protects internal cargo from enzymatic degradation, enhancing the half-life of sensitive molecules, such as RNA or proteins. Moreover, exosomes possess natural targeting capabilities. Depending on their cell of origin, they may exhibit preferential tropism for specific tissues or pathological environments, enabling a degree of passive targeting without the need for external modifications.
One of the most significant advantages is their ability to cross challenging biological barriers, most notably the BBB. This characteristic opens up therapeutic possibilities in neurological and neuro-oncological applications that are otherwise constrained by delivery limitations.3–5 Together, these features position exosomes as a highly adaptable platform for a wide range of clinical needs.
Therapeutic Payloads
Exosomes can be loaded with a diverse array of therapeutic payloads, either through endogenous cell engineering or exogenous modification techniques. These payloads include small molecule drugs, therapeutic proteins, and a broad spectrum of nucleic acids, such as small interfering RNA (siRNA), messenger RNA (mRNA), miRNA, and gene-editing tools like CRISPR-Cas9 components. Their ability to deliver functional RNA species with high stability and efficiency has made them a particularly attractive vehicle for gene modulation therapies.
Depending on the intended application, exosomes may be derived from autologous sources to reduce immunogenic risk or engineered from donor or cell line sources to optimize scalability, targeting, and cargo compatibility. Engineered exosomes may also be modified to express targeting ligands or surface peptides that further enhance delivery precision. This dual potential — both as a natural carrier and a customizable platform — makes exosomes a compelling candidate for next-generation therapeutic delivery across a broad spectrum of diseases.6,7
Barriers to Translation: Overcoming Technical and Regulatory Hurdles
Despite the compelling advantages exosomes offer as drug delivery vehicles, the field faces several persistent challenges that must be addressed to support clinical translation and commercial viability. These challenges span technical, biological, and regulatory domains, reflecting the complexity of harnessing a naturally derived and highly heterogeneous modality for therapeutic use.
Isolation and Purification
One of the most fundamental challenges lies in the efficient and reproducible isolation of exosomes at clinically relevant scale and purity. Traditional methods, such as differential ultracentrifugation and ultrafiltration, while widely used in research settings, often suffer from poor yield and inconsistent purity. Size exclusion chromatography (SEC) and immunoaffinity capture techniques offer improvements in specificity but come with limitations in throughput, cost, and scalability. The lack of standardized, high-throughput protocols impedes cross-study comparability and hinders regulatory acceptance. Variability in exosome composition based on source cell type, culture conditions, and purification method further compounds these issues, making it difficult to define consistent product profiles across batches.8,9
Cargo Loading Efficiency
Efficiently loading therapeutic payloads into exosomes remains a critical bottleneck in drug development. Passive loading approaches, such as incubating exosomes with hydrophobic small molecules or nucleic acids, are limited by low encapsulation efficiency. Active loading techniques — including electroporation, sonication, and membrane permeabilization — offer improved uptake but introduce concerns about compromising exosome integrity. These methods can alter surface proteins, disrupt membrane stability, or cause aggregation, potentially affecting biodistribution, uptake, and safety profiles. Achieving high loading efficiency while maintaining exosome functionality and structural integrity remains an area of intense research and innovation.3,10
Source Cell Selection
The therapeutic utility of exosomes is intimately linked to their cellular origin. MSCs, dendritic cells, and tumor cells are among the most commonly used sources, each with distinct advantages and risks. MSC-derived exosomes are favored for their regenerative and anti-inflammatory properties and relatively low immunogenicity. Dendritic cell–derived exosomes have immunomodulatory potential useful in vaccine and cancer immunotherapy applications. However, tumor-derived exosomes — while often exhibiting high target affinity — pose concerns owing to their potential to promote tumor progression or metastasis if not adequately purified or engineered. Source cell selection must therefore balance efficacy, safety, and regulatory acceptance.
Targeting Specificity and Biodistribution
Although exosomes exhibit natural tissue tropism based on their origin, this intrinsic targeting is not always aligned with therapeutic goals. Off-target uptake or clearance by organs such as the liver, spleen, and lungs can reduce efficacy and introduce unintended effects. To overcome this, strategies such as surface engineering and ligand conjugation have been explored to enhance targeting specificity. Modifying exosome membranes with peptides, antibodies, or other ligands can improve homing to diseased tissues or specific cell types but introduces additional complexity in manufacturing and quality control. The success of these targeting strategies depends on careful design, stability of the modifications, and preservation of functional payload delivery.2,11
Scalability and Manufacturing
Translating exosome-based therapies from the laboratory to the clinic requires overcoming significant barriers in large-scale manufacturing. Scalable, GMP-compliant production systems are still in early stages of development, with limited standardization across upstream culture conditions and downstream purification processes. Batch-to-batch variability, low yields, and contamination with other extracellular vesicles or proteins remain concerns. Bioreactor-based culture systems, continuous purification methods, and improved analytics are being developed to address these issues, but achieving cost-effective, high-yield manufacturing that meets regulatory expectations is an ongoing challenge.12,13
Regulatory Uncertainty
A further obstacle is the evolving and often ambiguous regulatory landscape surrounding exosome-based therapeutics. It remains unclear in many jurisdictions whether exosome products should be classified as biologics, drugs, or medical devices, with significant implications for preclinical requirements, manufacturing standards, and clinical evaluation. Regulatory agencies have yet to establish harmonized guidelines for characterization, potency assays, release criteria, and long-term safety assessment. In the absence of clear pathways, developers face uncertainty and potential delays in advancing exosome therapies through clinical trials and into the market. Collaborative efforts between regulators, researchers, and industry stakeholders will be essential to build a framework that balances innovation with patient safety.1,8
Innovations Fueling Exosome Platform Development
To address the complex challenges associated with exosome-based drug delivery, researchers and developers are turning to a wide array of technological innovations. These approaches aim to improve targeting precision, enhance loading efficiency, streamline manufacturing, and ensure product consistency. Taken together, these advancements are helping to bridge the gap between exosomes’ biological potential and their therapeutic viability.
Bioengineering and Surface Modification
One of the most active areas of innovation involves modifying the exosomal membrane to improve targeting and therapeutic functionality. Surface engineering strategies include the conjugation of targeting ligands, peptides, or antibodies to the exosome surface, enabling active targeting of disease-specific receptors. These modifications can significantly enhance the precision of exosome delivery, particularly in oncology and neurology applications where tissue selectivity is critical.
More advanced techniques, such as “click chemistry,” enable covalent attachment of targeting moieties or imaging agents under mild conditions, preserving the structural integrity of the exosomes. In parallel, membrane fusion approaches are being explored to combine natural exosome membranes with synthetic liposomes or other nanocarriers, thereby integrating the advantages of both systems while enabling greater customization of surface properties and payload capacity.10,11
Synthetic or Hybrid Exosomes
To overcome limitations in scalability, reproducibility, and cargo loading, researchers are also exploring the development of synthetic or hybrid exosomes. These structures mimic the size, shape, and surface composition of natural exosomes but are produced using bottom-up techniques that facilitate greater control over composition and function. One approach involves fusing natural exosomes with liposomes, creating hybrid vesicles that retain exosomal targeting properties while gaining the stability and modularity of synthetic nanocarriers.
In addition, microfluidic platforms have been employed to generate exosome mimetics in a highly reproducible and scalable manner. These systems can be fine-tuned to yield vesicles with specific size distributions, membrane markers, and encapsulation characteristics, offering a promising pathway toward standardized, GMP-compliant production of functionalized exosome-like particles.6
Advanced Loading Technologies
Improving the efficiency and reliability of therapeutic cargo loading remains a central focus of innovation. Rather than relying on post hoc loading techniques, some strategies involve genetically engineering the donor cells to express therapeutic proteins or nucleic acids that are naturally sorted into exosomes during their biogenesis. This endogenous loading approach avoids the risk of damaging the vesicle structure and may yield higher and more consistent payload levels.
Microfluidic electroporation and nanoporation platforms offer another promising avenue. These systems apply controlled electric fields or mechanical forces to temporarily permeabilize exosome membranes, allowing precise introduction of therapeutic cargo without causing aggregation or irreversible membrane disruption. Such methods enable real-time control of loading parameters and can be integrated into scalable production workflows.9,10
AI and Multi-Omic Tools for Quality Control
As exosome therapies progress toward clinical application, quality control and characterization have become top priorities. Advanced analytical technologies — particularly those integrating proteomics, lipidomics, and transcriptomics — are being deployed to develop comprehensive exosome profiles. These multi-omic signatures can serve as fingerprints for product identity, potency, and safety.
Artificial intelligence (AI) and machine learning models are increasingly being used to interpret these complex datasets and identify subtle patterns that correlate with functional outcomes. This allows for more consistent batch-to-batch performance, early detection of manufacturing deviations, and predictive modeling of therapeutic efficacy. By combining data-driven analytics with high-resolution molecular profiling, developers are building a more robust framework for regulatory compliance and clinical translation.4,13
From Bench to Bedside: Emerging Applications Across Therapeutic Areas
The versatility of exosomes as drug delivery vehicles is reflected in the wide range of therapeutic applications under investigation. Their ability to carry diverse payloads, modulate the immune system, and navigate biological barriers has spurred interest across multiple disease areas. In both preclinical and early clinical studies, exosome-based approaches are being explored to address unmet needs in oncology, neurology, immunotherapy, and regenerative medicine.
Oncology
Cancer remains one of the most promising and intensively studied areas for exosome-based drug delivery. Exosomes can be harnessed to deliver chemotherapeutic agents, RNA-based therapies, or immune-modulating factors directly to tumor cells. Their endogenous membrane composition allows them to evade immune detection and exploit natural uptake pathways, enhancing accumulation in the tumor microenvironment.
By incorporating targeting ligands or leveraging the natural tropism of exosomes derived from tumor or immune cells, researchers have achieved selective delivery of siRNA, miRNA, and anticancer drugs to malignant tissues. This has shown potential not only for direct tumor cell killing but also for modifying the tumor microenvironment, reprogramming immune responses, and overcoming resistance mechanisms. Compared with traditional nanoparticle formulations, exosomes have demonstrated a superior ability to reduce off-target toxicity while improving therapeutic index.2,7
Neurological Disorders
Exosomes’ ability to cross the BBB positions them uniquely for the treatment of neurological diseases — an area that remains notoriously difficult for conventional delivery technologies. Studies have demonstrated that exosomes can be engineered or selected to deliver therapeutic RNA molecules, neuroprotective peptides, or anti-inflammatory agents to brain tissue.
Preclinical investigations have explored the use of exosome-delivered siRNA and miRNA to silence pathogenic genes implicated in disorders such as Alzheimer’s disease, Parkinson’s disease, and glioblastoma. In addition to their cargo-carrying capacity, exosomes may also exert intrinsic neuroprotective effects, especially when derived from neural or stem cell sources. Their dual role as carriers and potential modulators of disease makes them an especially attractive platform for neurotherapeutics.5
Immunomodulation and Vaccines
Exosomes also hold promise in modulating immune responses, both as therapeutic agents in autoimmune and inflammatory diseases and as delivery vehicles for vaccines. Their natural role in antigen presentation and immune communication has inspired efforts to harness them as adjuvants or antigen delivery systems that can enhance the activation of specific immune cell populations.
In the context of infectious disease and cancer immunotherapy, exosomes engineered to carry tumor antigens or viral peptides have demonstrated the ability to prime T cells and stimulate cytotoxic immune responses. Conversely, in autoimmune and inflammatory conditions, exosomes derived from tolerogenic or stem cell sources have been investigated for their ability to suppress aberrant immune activation and restore homeostasis.1,3
Regenerative Medicine
Exosome-based therapies are also being developed to support tissue regeneration and repair, particularly in areas where conventional cell therapies face challenges in engraftment or long-term viability. Exosomes derived from MSCs have shown regenerative potential across a variety of tissues, including skin, cartilage, heart, and liver.
These MSC-derived exosomes can influence local inflammation, promote angiogenesis, and stimulate resident stem cell activity without the risks associated with direct stem cell transplantation. Applications in wound healing, osteoarthritis, and myocardial infarction are being actively explored, with early studies demonstrating improved functional recovery and reduced fibrosis. The ability to isolate and standardize exosome preparations from well-characterized MSC lines offers a scalable, cell-free alternative to conventional regenerative approaches.8,13
Charting the Future of Exosome Therapeutics
As the field of exosome-based drug delivery matures, momentum continues to build across academic, clinical, and commercial domains. A growing number of biotechnology companies are advancing exosome platform technologies designed to optimize production, targeting, and payload delivery. These platforms are being tailored for applications ranging from oncology and neurology to vaccine development and gene editing, often with a focus on scalable manufacturing and regulatory readiness. Several early-stage clinical trials are now underway, and the entry of large pharmaceutical players into the exosome space suggests increasing confidence in the modality’s long-term potential.
One of the most promising trends is the integration of exosome technology with other advanced therapeutic platforms. Exosomes are being explored as vehicles for delivering CRISPR-Cas9 components, enabling gene editing with reduced immunogenicity and improved cellular uptake compared to viral vectors. Similarly, efforts are underway to combine exosome delivery with engineered cell therapies, using exosomes to modulate the tumor microenvironment, enhance trafficking to disease sites, or deliver immunostimulatory agents. These synergies illustrate exosomes’ role not just as a standalone modality, but as a versatile enabler of complex, multimodal treatment strategies.
However, widespread clinical adoption will require continued progress on regulatory and standardization fronts. Regulatory bodies such as the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) are still in the process of determining how to classify and evaluate exosome-based products, with implications for clinical trial design, safety testing, and quality control. In the absence of clear guidelines, the International Society for Extracellular Vesicles (ISEV) and other stakeholder organizations have begun to develop consensus standards for exosome characterization, potency, and release criteria. Adoption of such standards will be essential to harmonizing expectations across jurisdictions and accelerating regulatory review.
To bring exosome-based delivery into the mainstream, several parallel developments must converge. Technological innovation must continue to address remaining barriers in manufacturing, targeting, and consistency. Clinical trials will need to produce clear evidence of safety, efficacy, and superiority over existing delivery systems. And regulatory frameworks must evolve to provide a transparent and predictable path to approval. Achieving this will require sustained collaboration among scientists, clinicians, regulators, and commercial developers. If these elements align, exosomes could become a foundational platform for the next generation of precision therapeutics.
Building the Smart Courier System of Precision Medicine
Exosomes represent one of the most exciting frontiers in drug delivery science — naturally evolved nanocarriers capable of transporting complex therapeutic payloads across biological barriers with remarkable precision and minimal immunogenicity. Their intrinsic biocompatibility, stability, and targeting potential position them as an elegant solution to many of the challenges faced by current delivery technologies, particularly in areas such as oncology, neurology, immunotherapy, and regenerative medicine.
However, realizing this promise requires overcoming significant technical and translational hurdles. Challenges in scalable manufacturing, standardized isolation and characterization, efficient loading, and regulatory ambiguity continue to temper expectations. At the same time, emerging innovations — from surface engineering and synthetic analogues to advanced analytics and AI-driven quality control — are rapidly expanding the toolkit available to exosome developers and helping to close the gap between research potential and clinical application.
The future of exosome-based therapeutics will depend on deep collaboration across disciplines. Biotech innovators, materials scientists, clinicians, and regulatory experts must work together to develop robust, reproducible platforms that meet the complex demands of modern drug development. Investment in infrastructure, interdisciplinary research, and regulatory engagement will be essential.
If these efforts succeed, exosomes could take their place as the “smart courier system” of precision medicine — capable of delivering the right therapeutic to the right place at the right time, with unprecedented specificity and safety. Their journey from biological curiosity to clinical mainstay is well underway, and with continued focus and innovation, their impact on the future of medicine may be profound.
References
1. Chen, Huizi, et al. “Exosomes, a New Star for Targeted Delivery.” Front. Cell Dev. Biol. 7 Oct. 2021.
2. Kim, Hyosuk, et al. “Recent Advances in Exosome-Based Drug Delivery for Cancer Therapy.” Cancers. 13: 4435 (2021).
3. Kar, Rishav, et al. “Exosome-Based Smart Drug Delivery Tool for Cancer Theranostics.” ACS Biomaterials Science & Engineering. 9: 577–594 (2023).
4. Lee, Jaewook, et al. “Exosome-based drug delivery systems and their therapeutic applications.” RSC Advances. 2022.
5. Luan, Xin, et al. “Engineering exosomes as refined biological nanoplatforms for drug delivery.” Acta Pharmacologica Sinica. 38: 754–763 (2017).
6. Paul, Swastika, et al. “ExoDS: a versatile exosome-based drug delivery platform to target cancer cells and cancer stem cells.” Front. Bioeng. Biotechnol. 4 Jun. 2024.
7. Huang, Ruotong, Jianming Zhou, and Shuying Chen. “Exosome-Based Drug Delivery Systems.” In Exosome Research - Biochemistry, Biomarkers and Perspectives in Therapy. 11 Feb. 2024.
8. Rezaie, Jafar, Maryan Feghhi, and Tahereh Etemadi. “A review on exosomes application in clinical trials: perspective, questions, and challenges.” Cell Communication and Signaling. 20: 145 (2022).
9. Kim, Hyo In, et al. “Recent advances in extracellular vesicles for therapeutic cargo delivery.” Experimental & Molecular Medicine. 56: 836–849 (2024).
10. Palakurthi, Sushesh Srivatsa, et al. “A comprehensive review of challenges and advances in exosome-based drug delivery systems.” Nanoscale Adv. 6: 6803–5826 (2024).
11. Ferreira, Débora, João Nuno Moreira, and Lígia R. Rodrigues. “New advances in exosome-based targeted drug delivery systems.” Critical Reviews in Oncology/Hematology. 172: 103628 (2022).
12. Koh, Hee Byung, et al. “Exosome-Based Drug Delivery: Translation from Bench to Clinic.” Pharmaceutics. 15: 2042 (2023).
13. Sharma, Vriti and Chitrangada Das Mukhopadhyay. “Exosome as drug delivery system: Current advancements.” Extracellular Vesicle. 3: 100032 (2024).