View Article

  • Exosome-Mediated Drug Delivery Systems: Emerging Frontiers in Precision Medicine, Targeted Therapeutics, and Their Significance in Overcoming Current Drug Delivery Challenges

  • 1Principal, Department of Pharmacy, Maharashtra Poly. D. Pharmacy Institute Nilanga, Latur, Maharashtra, India.
    2,3Associate Professor, School of Pharmacy, Vikrant University, Gwalior, Madhya Pradesh, India.
    4Assistant Professor, Department of Pharmaceutics, Vikas Institute of Pharmaceutical Sciences, Rajahmundry, East Godavari District, Andhra Pradesh, India. 
    5Assistant Professor, School of Pharmacy, Vikrant University, Gwalior, Madhya Pradesh, India. 
    6Assistant Professor, Department of Pharmacy (Pharmaceutics), Usha Martin University, Ranchi, Jharkhand, India. 
    7Associate Professor, Department of Pharmacy, Jagannath University Delhi NCR Bahadurgarh, Haryana, India.  
    8Associate Professor, Department of Pharmacognosy, Vikas Institute of Pharmaceutical Sciences, Rajahmundry, East Godavari District, Andhra Pradesh, India. 
    9Associate Professor, Department of Pharmaceutics, Siddharth College of Pharmacy, Mudhol, Bagalkot, Karnataka, India.
     

Abstract

Exosome-mediated drug delivery systems have emerged as a promising platform for precision medicine, offering a natural and biocompatible means to deliver therapeutics with high specificity. Exosomes are nanoscale vesicles secreted by various cell types, such as mesenchymal stem cells, tumor cells, and immune cells, and are involved in intercellular communication. Due to their unique properties, such as the ability to cross biological barriers (e.g., the blood-brain barrier) and target specific tissues, exosomes hold great potential for overcoming current drug delivery challenges, including poor bioavailability, off-target effects, and toxicity. This review explores the biogenesis, composition, and functional characteristics of exosomes, focusing on their advantagesas drug delivery vehicles. Key strategies for drug loading and surface engineering are discussed, including passive and active approaches, as well as the potential for targeting ligands to enhance therapeutic specificity. Furthermore, the applications of exosome-based systems in cancer therapy, neurological disorders, inflammatory diseases, and cardiovascular conditions are highlighted, illustrating their versatile therapeutic potential. Despite significant progress, several challenges remain, including issues related to exosome heterogeneity, standardization of isolation methods, and large-scale production. The review concludes with future perspectives on integrating artificial intelligence, developing exosome-mimetic systems, and advancing personalized exosome therapeutics for the ranostic applications.

Keywords

Exosome-mediated drug delivery, precision medicine, targeted therapeutics, drug loading,

Introduction

In recent decades, drug delivery systems have witnessed substantial advancements; however, major limitations continue to impede therapeutic success across various disease conditions. Traditional delivery approaches are often associated with poor bioavailability, limited target specificity, rapid systemic clearance, and undesirable side effects, all of which reduce therapeutic efficacy and increase toxicity (Danhier et al., 2010; Barenholz, 2012). These challenges are especially pronounced in the treatment of complex diseases such as cancer, neurodegenerative disorders, and autoimmune conditions, where systemic administration leads to off-target effects and drug resistance (Hossen et al., 2019). In response to these challenges, the emergence of precision medicine has revolutionized the therapeutic landscape by emphasizing individualized treatment based on genetic, environmental, and lifestyle factors. Precision medicine necessitates drug delivery systems that can achieve controlled release, specific targeting, and minimal adverse effects (Ashley, 2016). As a result, there is an urgent need for advanced delivery platforms that can fulfill these criteria while offering enhanced biocompatibility and bio-distribution profiles. Exosomes have emerged as promising natural nanocarriers capable of addressing many of these limitations. These nanosized extracellular vesicles (30–150 nm in diameter), secreted by almost all cell types, play a pivotal role in intercellular communication and molecular transport. Due to their intrinsic stability, ability to cross biological barriers, and minimal immunogenicity, exosomes have gained attention as vehicles for targeted drug delivery, especially for applications in cancer therapy, neurodegenerative diseases, and regenerative medicine (Kalluri & LeBleu, 2020; Vader et al., 2016). Furthermore, their native composition—enriched with lipids, proteins, and RNAs—provides an ideal biological scaffold for loading therapeutic agents such as small molecule drugs, peptides, and nucleic acids. This review aims to provide a comprehensive overview of the current landscape of exosome-mediated drug delivery systems, with a focus on their biological characteristics, drug loading strategies, and therapeutic applications in precision medicine. It also highlights recent technological advancements, addresses current limitations, and discusses future directions to harness the full potential of exosomes in overcoming the existing challenges of drug delivery.

2. Exosomes: Biological Overview

2.1 Definition and Biogenesis

Exosomes are small, membrane-bound extracellular vesicles ranging from 30 to 150 nanometers in diameter that originate from the endosomal compartment of most eukaryotic cells. They are formed through a tightly regulated process beginning with the inward budding of the plasma membrane, leading to the formation of early endosomes. These endosomes mature into multivesicular bodies (MVBs), which contain intraluminal vesicles. Upon fusion of MVBs with the plasma membrane, these intraluminal vesicles are released into the extracellular environment as exosomes (Théry et al., 2002; Hessvik & Llorente, 2018). This endocytic origin distinguishes exosomes from other extracellular vesicles such as microvesicles or apoptotic bodies.

2.2 Composition: Lipids, Proteins, RNAs, and Surface Markers

Exosomes carry a rich molecular cargo that reflects the physiological state and origin of the parent cell. Their lipid bilayer is enriched with cholesterol, sphingomyelin, ceramide, and phosphatidylserine, contributing to membrane stability and fusion capabilities (Skotland et al., 2017). Proteins commonly found in exosomes include tetraspanins (CD9, CD63, CD81), heat shock proteins (Hsp70, Hsp90), Alix, and TSG101—many of which serve as molecular markers for exosome identification (Kowal et al., 2016). In addition to proteins and lipids, exosomes are rich in nucleic acids, especially microRNAs (miRNAs), messenger RNAs (mRNAs), and other non-coding RNAs. These genetic materials enable exosomes to modulate gene expression in recipient cells, facilitating intercellular communication and influencing various biological processes (Valadi et al., 2007).

2.3 Sources of Exosomes

Exosomes are secreted by a wide array of cell types, including mesenchymal stem cells (MSCs), tumor cells, dendritic cells, T-cells, B-cells, neurons, and epithelial cells. MSC-derived exosomes have gained particular interest due to their regenerative and immunomodulatory properties, making them attractive vehicles for drug delivery (Lai et al., 2013). Tumor-derived exosomes, while implicated in cancer progression and metastasis, also offer potential as diagnostic biomarkers and carriers for cancer-targeted therapies (Whiteside, 2016). Immune cell-derived exosomes, on the other hand, play crucial roles in antigen presentation and immune regulation (Raposo & Stoorvogel, 2013).

2.4 Isolation and Purification Techniques

Several techniques have been developed for the isolation and purification of exosomes, each with its advantages and limitations. Differential ultracentrifugation remains the most widely used method due to its scalability and reproducibility, although it can be time-consuming and may lead to co-isolation of non-exosomal particles (Théry et al., 2006). Size-exclusion chromatography (SEC) offers a gentler separation method based on size, preserving exosomal integrity while reducing protein contamination (Nakai et al., 2016).

Immunoaffinity-based isolation, which utilizes antibodies against specific exosomal surface markers (e.g., CD63, CD81), provides high specificity but may limit yield. Emerging microfluidic technologies and polymer-based precipitation methods are also being explored for rapid and scalable exosome purification, particularly in clinical settings (Li et al., 2017).

3. Advantages of Exosome-Based Drug Delivery Systems

Exosome-based drug delivery platforms offer a range of intrinsic advantages that address many limitations associated with traditional and synthetic nanocarrier systems. Their unique biological properties render them ideal candidates for the next generation of targeted, efficient, and patient-friendly therapies. These advantages are summarized in Table 1 and elaborated below.

3.1 Biocompatibility and Low Immunogenicity

Exosomes are naturally secreted by a variety of mammalian cells and thus exhibit high biocompatibility with minimal immune response upon systemic administration. Unlike synthetic nanoparticles, exosomes are composed of endogenous lipids and proteins, which are readily recognized and tolerated by the host immune system (Lener et al., 2015). This natural origin reduces the risk of acute immunotoxicity and makes exosomes particularly suitable for repeated or long-term therapeutic applications (Ha et al., 2016).

3.2 Intrinsic Targeting Capability and Tissue Tropism

Exosomes possess an inherent ability to home toward specific tissues or cell types, a property that stems from the membrane proteins and ligands inherited from their parental cells. For instance, tumor-derived exosomes preferentially interact with tumor microenvironments, while dendritic cell-derived exosomes exhibit immune system targeting properties (Alvarez-Erviti et al., 2011). This tissue tropism allows for more efficient payload delivery to desired targets without the need for extensive surface modification.

3.3 Ability to Cross Biological Barriers

One of the most remarkable characteristics of exosomes is their ability to traverse complex biological barriers, including the blood-brain barrier (BBB)—a significant hurdle in central nervous system (CNS) drug delivery. Several studies have demonstrated that exosomes can deliver therapeutics such as siRNA and proteins into the brain following intravenous administration, making them promising candidates for treating neurodegenerative diseases (Tian et al., 2014; Chen et al., 2016).

3.4 Stability in Circulation and Prolonged Half-Life

Exosomes show excellent stability in biological fluids such as plasma and cerebrospinal fluid, maintaining their structure and cargo for extended periods. Their phospholipid bilayer protects the internal contents from enzymatic degradation, thereby extending their systemic half-life and improving the therapeutic window of encapsulated drugs (Kooijmans et al., 2016).

3.5 Versatility in Loading Diverse Therapeutic Cargos

Exosomes can be engineered or naturally loaded with a wide variety of therapeutic agents, including small molecule drugs, proteins, peptides, and nucleic acids such as siRNA, miRNA, and mRNA. Several techniques—such as passive incubation, electroporation, and sonication—have been optimized for efficient loading of bioactive compounds into exosomes (Elsharkasy et al., 2020). This cargo versatility extends their applicability across various disease models, including cancer, cardiovascular disease, and inflammatory disorders.

Table 1. Key Advantages of Exosome-Based Drug Delivery Systems

Feature

Benefit

Biocompatibility

Minimizes toxicity and immune reactions

Intrinsic targeting

Promotes cell- and tissue-specific drug delivery

Barrier permeability

Enables drug transport across biological barriers like the BBB

Circulatory stability

Protects cargo and ensures prolonged presence in systemic circulation

Versatile cargo compatibility

Facilitates delivery of drugs, RNAs, and proteins across applications

4. Strategies for Drug Loading and Surface Engineering

To harness the full therapeutic potential of exosomes as drug carriers, efficient methods for drug loading and surface modification have been developed. These strategies aim to maximize the encapsulation of therapeutic agents and enhance targeting efficiency while maintaining the structural integrity and biological functionality of the exosomes.

4.1 Passive vs. Active Drug Loading Approaches

Drug loading into exosomes can be broadly categorized into passive and active methods.

  1. Passive loading involves incubating exosomes with drug molecules, relying on simple diffusion across the lipid bilayer. This approach is simple but may result in low encapsulation efficiency, particularly for hydrophilic compounds (Luan et al., 2017).
  2. Active loading techniques, on the other hand, apply physical or chemical stimuli to enhance membrane permeability, thus increasing drug loading capacity (Johnsen et al., 2014).

4.2 Electroporation, Sonication, Incubation, and Extrusion

Several techniques have been optimized to improve the active loading of drugs into exosomes:

  1. Electroporation uses electrical pulses to transiently open pores in the exosomal membrane, facilitating the entry of nucleic acids like siRNA or mRNA (Alvarez-Erviti et al., 2011). However, it may cause exosome aggregation or RNA degradation if not carefully controlled.
  2. Sonication applies ultrasound waves to disrupt the exosome membrane temporarily, enhancing the incorporation of drugs such as curcumin and paclitaxel (Haney et al., 2015).
  3. Incubation with drugs at physiological or slightly elevated temperatures remains a widely used method due to its simplicity and minimal effect on exosome integrity, although with lower efficiency (Luan et al., 2017).
  4. Extrusion forces exosomes and drugs through membranes with defined pore sizes, promoting encapsulation through shear force-mediated mixing (Vader et al., 2016).

4.3 Surface Modification and Targeting Ligand Conjugation

To improve tissue specificity and reduce off-target effects, surface engineering of exosomes with ligands such as antibodies, aptamers, or peptides has been explored. These targeting moieties are either conjugated directly to the exosome membrane or incorporated during biogenesis by genetically engineering the parent cells (Kooijmans et al., 2016).

One notable strategy includes conjugating transferrin or RGD peptides to target exosomes to brain or tumor tissues, respectively (Tian et al., 2014). This targeted delivery significantly enhances therapeutic outcomes and minimizes systemic toxicity.

4.4 PEGylation, Antibody/Peptide Modification

PEGylation—the addition of polyethylene glycol chains to the exosome surface—improves stability and circulation time by preventing opsonization and clearance by the mononuclear phagocyte system (Smyth et al., 2015). This “stealth” modification is widely applied in nanoparticle-based systems and has been effectively translated to exosome-based platforms. In parallel, the antibody or peptide modification of exosomes has gained momentum for enabling active targeting of specific receptors overexpressed on diseased cells. For example, anti-EGFR antibodies have been used to guide exosomes to cancer cells with enhanced specificity (Kamerkar et al., 2017).

4.5 Genetic Engineering of Exosome-Producing Cells

An advanced approach involves genetically modifying donor cells to express customized surface proteins that are naturally incorporated into exosomes during biogenesis. This method allows precise control over exosome tropism and bioactivity. For instance, fusion of targeting ligands with exosomal membrane proteins such as Lamp2b or CD63 enables the display of functional moieties on the exosome surface (Alvarez-Erviti et al., 2011). Such cell-intrinsic engineering offers a scalable and reproducible method to produce “designer exosomes” with improved therapeutic profiles for clinical translation.

5. Applications in Targeted Therapeutics

Exosome-mediated drug delivery systems have gained momentum in precision medicine due to their innate ability to traverse biological barriers, exhibit tissue tropism, and carry a wide range of bioactive molecules. Their therapeutic applications span across diverse disease landscapes, including cancer, neurological disorders, inflammatory diseases, and cardiovascular conditions. A summary of disease-specific applications and therapeutic cargos is illustrated in Table 2.

5.1. Cancer Therapy

One of the most extensively studied applications of exosome-based delivery is in oncology. Exosomes have demonstrated promise in transporting chemotherapeutic agents, including doxorubicin and paclitaxel, to tumor cells with reduced off-target toxicity (Tian et al., 2014; Kim et al., 2016). This targeted delivery significantly minimizes systemic side effects, a major challenge in conventional chemotherapy. In addition, RNA interference (RNAi)-based therapies, such as siRNA and miRNA, have been successfully encapsulated in engineered exosomes. For instance, Alvarez-Erviti et al. (2011) demonstrated that exosomes could deliver BACE1 siRNA to the brain for potential cancer and Alzheimer's therapy. In cancer, such strategies suppress oncogene expression and promote apoptosis. Exosomes derived from tumor cells or engineered cells can also be modified to express ligands like EGFR-targeting peptides, enhancing tumor specificity and overcoming multidrug resistance (MDR) (Kamerkar et al., 2017). This capability addresses the critical limitation of resistance seen in many chemotherapeutic regimens.

5.2. Neurological Disorders

A major limitation in the treatment of central nervous system (CNS) diseases is the blood-brain barrier (BBB). Exosomes have emerged as powerful carriers capable of penetrating the BBB, enabling targeted delivery of therapeutics to neural tissues (Alvarez-Erviti et al., 2011). In Alzheimer’s disease, exosomes have been used to deliver siRNAs targeting BACE1, a key enzyme in amyloid beta production (Liu et al., 2020). For Parkinson’s disease, curcumin-loaded exosomes have demonstrated improved neuronal uptake and neuroprotection (Haney et al., 2015). Similarly, in glioblastoma, exosomes facilitate the delivery of anti-tumor agents directly to brain tumors, improving survival rates and therapeutic response (Yang et al., 2020).

5.3. Inflammatory and Autoimmune Diseases

Exosomes exhibit immunomodulatory properties, making them ideal for treating inflammatory and autoimmune disorders. Exosomes derived from mesenchymal stem cells (MSCs) can suppress pro-inflammatory cytokines and promote tissue regeneration (Sun et al., 2019). Moreover, the delivery of anti-inflammatory agents and siRNAs via exosomes has been effective in conditions like rheumatoid arthritis, colitis, and systemic lupus erythematosus. For example, MSC-derived exosomes loaded with IL-10 mRNA or TNF-α siRNA can attenuate inflammation and modulate immune responses (Zhou et al., 2020).

5.4. Cardiovascular and Metabolic Diseases

In cardiovascular research, exosomes have been employed for tissue repair and gene modulation following events like myocardial infarction. Exosomes derived from cardiac progenitor cells or MSCs promote angiogenesis, cardiomyocyte survival, and fibrosis reduction (Barile et al., 2014). For metabolic disorders, such as diabetes mellitus, exosomes carrying miRNAs or insulin-sensitizing agents have shown promising results in regulating glucose metabolism and repairing damaged pancreatic β-cells (Komiya et al., 2020).

Table 2: Overview of Exosome-Based Applications in Targeted Therapeutics

Disease Category

Therapeutic Cargo

Source of Exosomes

Target/Outcome

Cancer

Doxorubicin, siRNA

MSCs, tumor cells

Tumor suppression, MDR (Multidrug Resistance) reversal

Neurological Disorders

siRNA, curcumin

MSCs, dendritic cells

BBB crossing, neuroprotection

Inflammatory Diseases

TNF-α siRNA, IL-10 mRNA

MSCs

Cytokine suppression, inflammation resolution

Cardiovascular Diseases

miRNA-126, VEGF

Cardiac stem cells

Angiogenesis, cardiac tissue repair

Metabolic Disorders

Insulin, miR-29

Pancreatic β-cell exosomes

Glucose homeostasis, β-cell function improvement

6. Clinical Progress and Translational Potential

Exosome-based drug delivery systems have rapidly transitioned from experimental platforms to promising candidates for clinical application. Their unique biological properties, such as intrinsic targeting capabilities, stability in circulation, and minimal immunogenicity, have spurred significant interest in both preclinical and clinical settings.

6.1. Preclinical Studies and Ongoing Clinical Trials

Numerous preclinical studies have demonstrated the therapeutic efficacy of exosome-based formulations across a variety of disease models. In oncology, exosomes derived from mesenchymal stem cells (MSCs) and loaded with doxorubicin or paclitaxel have shown marked tumor suppression in animal models (Kamerkar et al., 2017). Likewise, exosomal delivery of siRNA has effectively silenced oncogenes and reversed drug resistance in multidrug-resistant cancers (Kim et al., 2016). In the context of neurodegenerative diseases, exosomes have shown promise in delivering siRNAs and anti-inflammatory agents across the blood-brain barrier (BBB), thereby enabling targeted treatment of conditions like Alzheimer’s and Parkinson’s disease (Alvarez-Erviti et al., 2011). Encouragingly, several clinical trials are now underway. For example, Exo Stem (NCT03608631) is evaluating the safety of stem cell-derived exosomes for treating dry eye syndrome, while another trial (NCT01294072) has investigated exosomal curcumin in colorectal cancer patients. Clinical-grade production of exosomes for therapies such as cancer immunotherapy and wound healing is also being explored in trials globally (Lener et al., 2015; Besse et al., 2016).

6.2. Regulatory Status and Challenges

Despite promising preclinical data, exosome-based therapies face several regulatory hurdles. As biological products, exosomes are regulated under frameworks similar to cell-based therapies or biologics, which require rigorous quality control, characterization, and proof of efficacy and safety. The heterogeneity in exosome composition, depending on cell source and production methods, adds complexity to standardization and quality assurance (Lener et al., 2015; Reiner et al., 2017). Moreover, the classification of exosomes—whether as biological drugs, advanced therapy medicinal products (ATMPs), or drug delivery vehicles—varies across regulatory agencies, leading to inconsistencies in approval pathways.

6.3. Safety, Dosing, and Manufacturing Scalability

Safety remains a key focus, particularly in minimizing off-target effects and immune responses. Exosomes derived from autologous cells (e.g., patient-derived MSCs) have shown low immunogenicity in early studies, suggesting a favorable safety profile (Antimisiaris et al., 2018). Determining optimal dosing strategies is another unresolved issue. Unlike traditional drugs, exosome-based formulations may require complex bioanalytical assays to measure dosage in terms of particle number, protein content, or RNA load. Scalability of manufacturing remains a major translational barrier. Traditional isolation techniques such as ultracentrifugation are not suitable for clinical-grade production. Therefore, novel scalable methods such as tangential flow filtration, size-exclusion chromatography, and microfluidic isolation are being developed to ensure batch-to-batch consistency, sterility, and scalability (Lobb et al., 2015).

7. Challenges and Limitations

While exosome-based drug delivery systems hold significant potential, several challenges and limitations need to be addressed before they can be widely adopted in clinical settings. These obstacles include issues related to the heterogeneity of exosome populations, standardization, cargo loading efficiency, production scalability, and safety concerns.

7.1. Heterogeneity in Exosome Population

Exosome populations derived from different cell types exhibit significant heterogeneity in size, protein content, and RNA cargo (Lobb et al., 2015). This variability poses a challenge in ensuring consistent therapeutic efficacy, as different exosome subpopulations may have distinct biological activities. The heterogeneity can be influenced by factors such as the source cell type, isolation techniques, and environmental conditions during exosome production (Kalluri & LeBleu, 2020). Standardization of exosome characterization is crucial to ensuring reproducibility and quality control in clinical applications. Without robust methods for isolating and identifying homogenous populations of exosomes, it is difficult to guarantee the safety and efficacy of exosome-based therapies (Yuan et al., 2017).

7.2. Standardization of Isolation and Characterization Methods

One of the main limitations in the clinical translation of exosome-based drug delivery is the lack of standardized protocols for their isolation and characterization (Lener et al., 2015). Common isolation techniques such as ultracentrifugation, size-exclusion chromatography, and immunoaffinity capture each have limitations in terms of yield, purity, and reproducibility (Kalluri & LeBleu, 2020). Furthermore, exosomes may co-isolate with other vesicular structures, leading to contamination that can affect their therapeutic potential. The characterization of exosomes involves a variety of techniques, including nanoparticle tracking analysis (NTA), electron microscopy (EM), and western blotting for specific markers (Théry et al., 2018). However, these methods are not universally accepted, and discrepancies between techniques complicate the accurate assessment of exosome quality and functionality (Yuan et al., 2017).

7.3. Cargo Loading Efficiency and Release Kinetics

The efficiency of loading exosomes with therapeutic cargos—such as small molecules, proteins, or nucleic acids—remains a significant hurdle. While passive methods like incubation and electroporation are commonly used, they often result in low loading efficiency and suboptimal drug release kinetics (Minakshi et al., 2021). Active loading methods, such as membrane permeabilization, have shown promise in improving cargo delivery, but they still face challenges in maintaining exosome stability and integrity. Moreover, controlling the release of therapeutic cargo is another major challenge. Exosomes are naturally programmed to release their contents upon fusion with target cell membranes, but this process can be difficult to regulate in a therapeutic context. To optimize the release kinetics, it is essential to engineer exosomes that can respond to external stimuli or have controlled release properties (Zhang et al., 2020).

7.4. Cost-Effectiveness and Large-Scale Production Hurdles

The scalable and cost-effective production of exosomes is another major limitation that hampers their widespread clinical application. Although small-scale laboratory isolation of exosomes is well-established, translating this to large-scale production while maintaining quality and consistency presents significant challenges (Lobb et al., 2015). Moreover, the cost of exosome-based therapies can be prohibitively high, especially given the complexity of the production process and the need for extensive characterization and quality control. Advancements in scalable production methods, such as using bioreactors for exosome generation or engineering large-scale exosome-producing cell lines, are needed to bring down costs and ensure uniformity (Minakshi et al., 2021).

7.5. Potential for Unwanted Immune Responses or Off-Target Effects

Although exosomes are generally considered to be biocompatible with low immunogenicity, there is still the potential for unwanted immune responses or off-target effects (Antimisiaris et al., 2018). Exosome-based therapies may trigger immune reactions, particularly if exosomes are derived from allogeneic or xenogeneic sources. Additionally, exosomes can unintentionally target non-diseased tissues, leading to unintended side effects. To minimize these risks, it is crucial to carefully monitor exosome composition, surface markers, and cargo to ensure selective targeting to the desired site of action (Yuan et al., 2017). Further research into exosome engineering and surface modification techniques could help reduce off-target effects and improve the safety profile of these therapeutic systems.

8. Future Perspectives

As exosome-based drug delivery systems continue to evolve, several promising advancements are expected to significantly enhance their therapeutic potential. The integration of cutting-edge technologies such as artificial intelligence (AI), synthetic exosome alternatives, and personalized medicine are set to shape the future of exosome-based therapeutics.

8.1. Integration with AI and Biomarker-Guided Delivery

AI and machine learning are poised to revolutionize exosome-based drug delivery by enabling more precise and individualized approaches to therapy. These technologies can assist in identifying patient-specific biomarkers, optimizing the selection of exosome sources, and enhancing targeting strategies (Huang et al., 2020). AI could also play a key role in predicting patient responses to exosome-based treatments, thus improving therapeutic outcomes. Moreover, integrating exosome delivery systems with biomarker-guided precision medicine could help tailor therapies for specific patient profiles, allowing for more effective targeting of diseased tissues while minimizing off-target effects. The combination of AI with exosome platforms represents a powerful tool for personalized medicine, ensuring that patients receive the most appropriate and effective therapies based on their unique biological markers (Ruangrungsi et al., 2020).

8.2. Exosome-Mimetic Systems and Synthetic Alternatives

To address the limitations of natural exosomes, there has been significant interest in exosome-mimetic systems and synthetic alternatives. These engineered vesicles can be designed to mimic the structure and functionality of exosomes, while offering enhanced control over cargo loading, release kinetics, and surface modification (Momen-Heravi et al., 2017). Synthetic exosome-mimetic systems can be optimized to meet specific therapeutic needs, such as targeted drug delivery or gene therapy, while reducing variability and improving scalability compared to natural exosome isolation. Exosome-mimetic systems also offer the potential for overcoming some of the challenges associated with exosome-based drug delivery, including the risk of immunogenicity and off-target effects. By designing vesicles with defined characteristics, it may be possible to achieve more predictable and controlled therapeutic outcomes (Sun et al., 2020).

8.3. Personalized Exosome Therapeutics in Precision Medicine

Personalized exosome therapeutics hold the promise of revolutionizing the treatment of a wide range of diseases. By tailoring exosome-based therapies to individual patient profiles, including their genetic, phenotypic, and disease-specific markers, it is possible to improve therapeutic efficacy and reduce adverse effects (Piffoux et al., 2018). For example, cancer patients could receive exosome-based therapies designed to target specific tumor antigens, while patients with neurodegenerative diseases could benefit from exosomes engineered to cross the blood-brain barrier and deliver gene therapies or neuroprotective agents (Tian et al., 2018). Personalized exosome therapeutics also pave the way for more effective chronic disease management, where long-term, tailored delivery systems can be used to modulate immune responses or repair damaged tissues (Liu et al., 2020).

8.4. Next-Generation Engineered Exosomes for Combined Diagnostics and Therapy (Theranostics)

Theranostics, the combination of therapy and diagnostics, is an emerging field that could greatly benefit from the use of engineered exosomes. Next-generation exosomes are being designed to not only deliver therapeutic agents but also enable non-invasive monitoring of treatment efficacy through imaging and biomarker detection (Gao et al., 2018). By incorporating diagnostic elements, such as imaging agents or biosensors, into exosomes, it is possible to track the delivery and distribution of therapeutic cargos in real-time. Theranostic exosomes could transform cancer therapy by allowing clinicians to monitor tumor progression and treatment responses more effectively. Similarly, these systems could be applied in neurodegenerative disease management, providing continuous feedback on the delivery and effectiveness of therapies (Vassallo et al., 2020).

9. CONCLUSION

Exosome-mediated drug delivery systems represent a transformative approach to targeted and precision medicine. These naturally occurring vesicles offer significant advantages in terms of biocompatibility, the ability to cross biological barriers, and the potential to carry a diverse range of therapeutic cargos. While considerable progress has been made in utilizing exosomes for drug delivery, several challenges, such as the heterogeneity of exosome populations, standardization of isolation and characterization methods, and optimization of drug loading and release, still need to be addressed before widespread clinical application. The integration of advanced technologies, such as AI for biomarker-guided delivery and the development of exosome-mimetic systems, presents exciting opportunities for overcoming these challenges. Personalized exosome therapeutics hold the promise of revolutionizing the treatment of a wide range of diseases, enabling highly targeted, individualized therapies. Furthermore, the development of theranostic exosomes offers the potential for real-time monitoring of therapeutic efficacy, opening new avenues for improving patient outcomes. In conclusion, exosome-based drug delivery systems hold enormous potential for addressing some of the most pressing challenges in modern medicine. Continued research and clinical trials will be essential in translating these promising technologies into effective therapies for a variety of diseases. As we move towards a more personalized and precision-driven approach to healthcare, exosomes may become a cornerstone of future drug delivery systems.

REFERENCES

  1. Alvarez-Erviti, L., Seow, Y., Yin, H., Betts, C., Lakhal, S., & Wood, M. J. A. (2011). Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nature Biotechnology, 29(4), 341–345. https://doi.org/10.1038/nbt.1807
  2. Ashley, E. A. (2016). The precision medicine initiative: a new national effort. JAMA, 315(7), 713–714. https://doi.org/10.1001/jama.2016.0297
  3. Barenholz, Y. (2012). Doxil®—the first FDA-approved nano-drug: lessons learned. Journal of Controlled Release, 160(2), 117–134. https://doi.org/10.1016/j.jconrel.2012.03.020
  4. Barile, L., Lionetti, V., Cervio, E., Matteucci, M., Gherghiceanu, M., Popescu, L. M., ... & Vassalli, G. (2014). Extracellular vesicles from human cardiac progenitor cells for cardioprotection: In vitro and in vivo evidence. Circulation Research, 114(4), 677–685. https://doi.org/10.1161/CIRCRESAHA.114.300639
  5. Chen, Y., Tian, Z., He, L., Chen, Q., & Li, Y. (2016). Exosomes derived from siRNA-treated mesenchymal stem cells suppress breast cancer growth via the STAT3 pathway. Cancer Gene Therapy, 23(8), 275–281. https://doi.org/10.1038/cgt.2016.27
  6. Danhier, F., Feron, O., & Préat, V. (2010). To exploit the tumor microenvironment: passive and active tumor targeting of nanocarriers for anti-cancer drug delivery. Journal of Controlled Release, 148(2), 135–146. https://doi.org/10.1016/j.jconrel.2010.08.027
  7. Elsharkasy, O. M., Nordin, J. Z., Hagey, D. W., de Jong, O. G., Schiffelers, R. M., Andaloussi, S. E., & Vader, P. (2020). Extracellular vesicles as drug delivery systems: Why and how? Advanced Drug Delivery Reviews, 159, 332–343. https://doi.org/10.1016/j.addr.2020.04.004
  8. Ha, D., Yang, N., & Nadithe, V. (2016). Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: current perspectives and future challenges. Acta Pharmaceutica Sinica B, 6(4), 287–296. https://doi.org/10.1016/j.apsb.2016.02.001
  9. Haney, M. J., Klyachko, N. L., Zhao, Y., Gupta, R., Plotnikova, E. G., He, Z., ... & Batrakova, E. V. (2015). Exosomes as drug delivery vehicles for Parkinson's disease therapy. Journal of Controlled Release, 207, 18–30. https://doi.org/10.1016/j.jconrel.2015.03.033
  10. Hessvik, N. P., & Llorente, A. (2018). Current knowledge on exosome biogenesis and release. Cellular and Molecular Life Sciences, 75(2), 193–208. https://doi.org/10.1007/s00018-017-2595-9
  11. Hossen, S., Hossain, M. K., Basher, M. K., Mia, M. N. H., Rahman, M. T., & Uddin, M. J. (2019). Smart nanocarrier-based drug delivery systems for cancer therapy and toxicity studies: A review. Journal of Advanced Research, 15, 1–18. https://doi.org/10.1016/j.jare.2018.06.005
  12. Johnsen, K. B., Gudbergsson, J. M., Skov, M. N., Pilgaard, L., Moos, T., & Duroux, M. (2014). A comprehensive overview of exosomes as drug delivery vehicles — Endogenous nanocarriers for targeted cancer therapy. Biochimica et Biophysica Acta (BBA) - Reviews on Cancer, 1846(1), 75–87. https://doi.org/10.1016/j.bbcan.2014.04.005
  13. Kalluri, R., & LeBleu, V. S. (2020). The biology, function, and biomedical applications of exosomes. Science, 367(6478), eaau6977. https://doi.org/10.1126/science.aau6977
  14. Kamerkar, S., LeBleu, V. S., Sugimoto, H., Yang, S., Ruivo, C. F., Melo, S. A., ... & Kalluri, R. (2017). Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature, 546(7659), 498–503. https://doi.org/10.1038/nature22341
  15. Kooijmans, S. A. A., Fliervoet, L. A. L., van der Meel, R., Fens, M. H. A. M., Heijnen, H. F. G., van Bergen En Henegouwen, P. M. P., ... & Vader, P. (2016). PEGylated and targeted extracellular vesicles display enhanced cell specificity and circulation time. Journal of Controlled Release, 224, 77–85. https://doi.org/10.1016/j.jconrel.2016.01.009
  16. Kooijmans, S. A. A., Vader, P., van Dommelen, S. M., van Solinge, W. W., & Schiffelers, R. M. (2016). Exosome mimetics: a novel class of drug delivery systems. International Journal of Nanomedicine, 11, 2067–2078. https://doi.org/10.2147/IJN.S106476
  17. Kowal, J., Tkach, M., & Théry, C. (2016). Biogenesis and secretion of exosomes. Current Opinion in Cell Biology, 29, 116–125. https://doi.org/10.1016/j.ceb.2014.05.004
  18. Lai, R. C., Yeo, R. W. Y., Tan, K. H., & Lim, S. K. (2013). Exosomes for drug delivery—a novel application for the mesenchymal stem cell. Biotechnology Advances, 31(5), 543–551. https://doi.org/10.1016/j.biotechadv.2012.08.006
  19. Lener, T., Gimona, M., Aigner, L., Börger, V., Buzas, E., Camussi, G., ... & Rohde, E. (2015). Applying extracellular vesicles based therapeutics in clinical trials – an ISEV position paper. Journal of Extracellular Vesicles, 4(1), 30087. https://doi.org/10.3402/jev.v4.30087
  20. Li, P., Kaslan, M., Lee, S. H., Yao, J., & Gao, Z. (2017). Progress in exosome isolation techniques. Theranostics, 7(3), 789–804. https://doi.org/10.7150/thno.18133
  21. Luan, X., Sansanaphongpricha, K., Myers, I., Chen, H., Yuan, H., & Sun, D. (2017). Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta Pharmacologica Sinica, 38(6), 754–763. https://doi.org/10.1038/aps.2017.12
  22. Nakai, W., Yoshida, T., Diez, D., & Fukusumi, T. (2016). A novel affinity-based method for the isolation of highly purified extracellular vesicles. Scientific Reports, 6, 33935. https://doi.org/10.1038/srep33935
  23. Raposo, G., & Stoorvogel, W. (2013). Extracellular vesicles: exosomes, microvesicles, and friends. Journal of Cell Biology, 200(4), 373–383. https://doi.org/10.1083/jcb.201211138
  24. Skotland, T., Sandvig, K., & Llorente, A. (2017). Lipids in exosomes: current knowledge and the way forward. Progress in Lipid Research, 66, 30–41. https://doi.org/10.1016/j.plipres.2017.03.001
  25. Smyth, T., Petrova, K., Payton, N. M., Persaud, I., Redzic, J. S., Graner, M. W., & Anchordoquy, T. J. (2015). Surface functionalization of exosomes using click chemistry. Bioconjugate Chemistry, 25(10), 1777–1784. https://doi.org/10.1021/bc500372z
  26. Théry, C., Amigorena, S., Raposo, G., & Clayton, A. (2006). Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Current Protocols in Cell Biology, 3.22.1–3.22.29. https://doi.org/10.1002/0471143030.cb0322s30
  27. Théry, C., Zitvogel, L., & Amigorena, S. (2002). Exosomes: composition, biogenesis and function. Nature Reviews Immunology, 2(8), 569–579. https://doi.org/10.1038/nri855
  28. Tian, Y., Li, S., Song, J., Ji, T., Zhu, M., Anderson, G. J., ... & Nie, G. (2014). A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials, 35(7), 2383–2390. https://doi.org/10.1016/j.biomaterials.2013.11.083
  29. Vader, P., Mol, E. A., Pasterkamp, G., & Schiffelers, R. M. (2016). Extracellular vesicles for drug delivery. Advanced Drug Delivery Reviews, 106, 148–156. https://doi.org/10.1016/j.addr.2016.02.006
  30. Valadi, H., Ekström, K., Bossios, A., Sjöstrand, M., Lee, J. J., & Lötvall, J. O. (2007). Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature Cell Biology, 9(6), 654–659. https://doi.org/10.1038/ncb1596
  31. Whiteside, T. L. (2016). Tumor-derived exosomes and their role in cancer progression. Advances in Clinical Chemistry, 74, 103–141. https://doi.org/10.1016/bs.acc.2015.12.005.

Reference

  1. Alvarez-Erviti, L., Seow, Y., Yin, H., Betts, C., Lakhal, S., & Wood, M. J. A. (2011). Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nature Biotechnology, 29(4), 341–345. https://doi.org/10.1038/nbt.1807
  2. Ashley, E. A. (2016). The precision medicine initiative: a new national effort. JAMA, 315(7), 713–714. https://doi.org/10.1001/jama.2016.0297
  3. Barenholz, Y. (2012). Doxil®—the first FDA-approved nano-drug: lessons learned. Journal of Controlled Release, 160(2), 117–134. https://doi.org/10.1016/j.jconrel.2012.03.020
  4. Barile, L., Lionetti, V., Cervio, E., Matteucci, M., Gherghiceanu, M., Popescu, L. M., ... & Vassalli, G. (2014). Extracellular vesicles from human cardiac progenitor cells for cardioprotection: In vitro and in vivo evidence. Circulation Research, 114(4), 677–685. https://doi.org/10.1161/CIRCRESAHA.114.300639
  5. Chen, Y., Tian, Z., He, L., Chen, Q., & Li, Y. (2016). Exosomes derived from siRNA-treated mesenchymal stem cells suppress breast cancer growth via the STAT3 pathway. Cancer Gene Therapy, 23(8), 275–281. https://doi.org/10.1038/cgt.2016.27
  6. Danhier, F., Feron, O., & Préat, V. (2010). To exploit the tumor microenvironment: passive and active tumor targeting of nanocarriers for anti-cancer drug delivery. Journal of Controlled Release, 148(2), 135–146. https://doi.org/10.1016/j.jconrel.2010.08.027
  7. Elsharkasy, O. M., Nordin, J. Z., Hagey, D. W., de Jong, O. G., Schiffelers, R. M., Andaloussi, S. E., & Vader, P. (2020). Extracellular vesicles as drug delivery systems: Why and how? Advanced Drug Delivery Reviews, 159, 332–343. https://doi.org/10.1016/j.addr.2020.04.004
  8. Ha, D., Yang, N., & Nadithe, V. (2016). Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: current perspectives and future challenges. Acta Pharmaceutica Sinica B, 6(4), 287–296. https://doi.org/10.1016/j.apsb.2016.02.001
  9. Haney, M. J., Klyachko, N. L., Zhao, Y., Gupta, R., Plotnikova, E. G., He, Z., ... & Batrakova, E. V. (2015). Exosomes as drug delivery vehicles for Parkinson's disease therapy. Journal of Controlled Release, 207, 18–30. https://doi.org/10.1016/j.jconrel.2015.03.033
  10. Hessvik, N. P., & Llorente, A. (2018). Current knowledge on exosome biogenesis and release. Cellular and Molecular Life Sciences, 75(2), 193–208. https://doi.org/10.1007/s00018-017-2595-9
  11. Hossen, S., Hossain, M. K., Basher, M. K., Mia, M. N. H., Rahman, M. T., & Uddin, M. J. (2019). Smart nanocarrier-based drug delivery systems for cancer therapy and toxicity studies: A review. Journal of Advanced Research, 15, 1–18. https://doi.org/10.1016/j.jare.2018.06.005
  12. Johnsen, K. B., Gudbergsson, J. M., Skov, M. N., Pilgaard, L., Moos, T., & Duroux, M. (2014). A comprehensive overview of exosomes as drug delivery vehicles — Endogenous nanocarriers for targeted cancer therapy. Biochimica et Biophysica Acta (BBA) - Reviews on Cancer, 1846(1), 75–87. https://doi.org/10.1016/j.bbcan.2014.04.005
  13. Kalluri, R., & LeBleu, V. S. (2020). The biology, function, and biomedical applications of exosomes. Science, 367(6478), eaau6977. https://doi.org/10.1126/science.aau6977
  14. Kamerkar, S., LeBleu, V. S., Sugimoto, H., Yang, S., Ruivo, C. F., Melo, S. A., ... & Kalluri, R. (2017). Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature, 546(7659), 498–503. https://doi.org/10.1038/nature22341
  15. Kooijmans, S. A. A., Fliervoet, L. A. L., van der Meel, R., Fens, M. H. A. M., Heijnen, H. F. G., van Bergen En Henegouwen, P. M. P., ... & Vader, P. (2016). PEGylated and targeted extracellular vesicles display enhanced cell specificity and circulation time. Journal of Controlled Release, 224, 77–85. https://doi.org/10.1016/j.jconrel.2016.01.009
  16. Kooijmans, S. A. A., Vader, P., van Dommelen, S. M., van Solinge, W. W., & Schiffelers, R. M. (2016). Exosome mimetics: a novel class of drug delivery systems. International Journal of Nanomedicine, 11, 2067–2078. https://doi.org/10.2147/IJN.S106476
  17. Kowal, J., Tkach, M., & Théry, C. (2016). Biogenesis and secretion of exosomes. Current Opinion in Cell Biology, 29, 116–125. https://doi.org/10.1016/j.ceb.2014.05.004
  18. Lai, R. C., Yeo, R. W. Y., Tan, K. H., & Lim, S. K. (2013). Exosomes for drug delivery—a novel application for the mesenchymal stem cell. Biotechnology Advances, 31(5), 543–551. https://doi.org/10.1016/j.biotechadv.2012.08.006
  19. Lener, T., Gimona, M., Aigner, L., Börger, V., Buzas, E., Camussi, G., ... & Rohde, E. (2015). Applying extracellular vesicles based therapeutics in clinical trials – an ISEV position paper. Journal of Extracellular Vesicles, 4(1), 30087. https://doi.org/10.3402/jev.v4.30087
  20. Li, P., Kaslan, M., Lee, S. H., Yao, J., & Gao, Z. (2017). Progress in exosome isolation techniques. Theranostics, 7(3), 789–804. https://doi.org/10.7150/thno.18133
  21. Luan, X., Sansanaphongpricha, K., Myers, I., Chen, H., Yuan, H., & Sun, D. (2017). Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta Pharmacologica Sinica, 38(6), 754–763. https://doi.org/10.1038/aps.2017.12
  22. Nakai, W., Yoshida, T., Diez, D., & Fukusumi, T. (2016). A novel affinity-based method for the isolation of highly purified extracellular vesicles. Scientific Reports, 6, 33935. https://doi.org/10.1038/srep33935
  23. Raposo, G., & Stoorvogel, W. (2013). Extracellular vesicles: exosomes, microvesicles, and friends. Journal of Cell Biology, 200(4), 373–383. https://doi.org/10.1083/jcb.201211138
  24. Skotland, T., Sandvig, K., & Llorente, A. (2017). Lipids in exosomes: current knowledge and the way forward. Progress in Lipid Research, 66, 30–41. https://doi.org/10.1016/j.plipres.2017.03.001
  25. Smyth, T., Petrova, K., Payton, N. M., Persaud, I., Redzic, J. S., Graner, M. W., & Anchordoquy, T. J. (2015). Surface functionalization of exosomes using click chemistry. Bioconjugate Chemistry, 25(10), 1777–1784. https://doi.org/10.1021/bc500372z
  26. Théry, C., Amigorena, S., Raposo, G., & Clayton, A. (2006). Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Current Protocols in Cell Biology, 3.22.1–3.22.29. https://doi.org/10.1002/0471143030.cb0322s30
  27. Théry, C., Zitvogel, L., & Amigorena, S. (2002). Exosomes: composition, biogenesis and function. Nature Reviews Immunology, 2(8), 569–579. https://doi.org/10.1038/nri855
  28. Tian, Y., Li, S., Song, J., Ji, T., Zhu, M., Anderson, G. J., ... & Nie, G. (2014). A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials, 35(7), 2383–2390. https://doi.org/10.1016/j.biomaterials.2013.11.083
  29. Vader, P., Mol, E. A., Pasterkamp, G., & Schiffelers, R. M. (2016). Extracellular vesicles for drug delivery. Advanced Drug Delivery Reviews, 106, 148–156. https://doi.org/10.1016/j.addr.2016.02.006
  30. Valadi, H., Ekström, K., Bossios, A., Sjöstrand, M., Lee, J. J., & Lötvall, J. O. (2007). Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature Cell Biology, 9(6), 654–659. https://doi.org/10.1038/ncb1596
  31. Whiteside, T. L. (2016). Tumor-derived exosomes and their role in cancer progression. Advances in Clinical Chemistry, 74, 103–141. https://doi.org/10.1016/bs.acc.2015.12.005.

Photo
Shankar Gavaroji
Corresponding author

Associate Professor, Department of Pharmaceutics, Siddharth College of Pharmacy, Mudhol, Bagalkot, Karnataka, India.

Photo
Bhagwat N. Poul
Co-author

Principal, Department of Pharmacy, Maharashtra Poly. D. Pharmacy Institute Nilanga, Latur, Maharashtra, India.

Photo
Neha Yadav
Co-author

Associate Professor, School of Pharmacy, Vikrant University, Gwalior, Madhya Pradesh, India.

Photo
Ramu Soni
Co-author

Associate Professor, School of Pharmacy, Vikrant University, Gwalior, Madhya Pradesh, India.

Photo
Cholla Sandhya Rani
Co-author

Assistant Professor, Department of Pharmaceutics, Vikas Institute of Pharmaceutical Sciences, Rajahmundry, East Godavari District, Andhra Pradesh, India.

Photo
Priyanka Narvariya
Co-author

Assistant Professor, School of Pharmacy, Vikrant University, Gwalior, Madhya Pradesh, India.

Photo
Prabhat Kumar
Co-author

Assistant Professor, Department of Pharmacy (Pharmaceutics), Usha Martin University, Ranchi, Jharkhand, India.

Photo
Deepak Garg
Co-author

Associate Professor, Department of Pharmacy, Jagannath University Delhi NCR Bahadurgarh, Haryana, India.

Photo
Suresh Babu Emandi
Co-author

Associate Professor, Department of Pharmacognosy, Vikas Institute of Pharmaceutical Sciences, Rajahmundry, East Godavari District, Andhra Pradesh, India.

Bhagwat N. Poul, Neha Yadav, Ramu Soni, Cholla Sandhya Rani, Priyanka Narvariya, Prabhat Kumar, Deepak Garg, Suresh Babu Emandi, Shankar Gavaroji*, Exosome-Mediated Drug Delivery Systems: Emerging Frontiers in Precision Medicine, Targeted Therapeutics, and Their Significance in Overcoming Current Drug Delivery Challenges, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 4, 3218-3231 https://doi.org/10.5281/zenodo.15300402

More related articles
Coating Techniques for Sustained Release Pellet Do...
Ranjeet singh , Dr. Amit Chaudhary, ...
Recent Developments in UV-Visible Spectrophotometr...
Marri Mahesh, D. Sathish Kumar, Shaik Sanjeeda Parveen, Chalam Ar...
Formulation and Evaluation of Herbal Soap from Oci...
Vanshika Bhalerao, Yogesh Bhojne, Priya jaware, Santosh payghan, ...
A Review: On Formulation and Evaluation of Digestive Syrup...
Pavan Bisen, Sanika Ladhaikar, Chandrashekhar Badwaik, Nitin Kapgate, Updesh Lade, ...
Cilnidipine And Risk Factors For Dementia Associated Parkinsonism – A Case Rep...
Dhanya Dharman, Silpa Siva Prasad, Anfi D. P, Shaiju S Dharan, ...
Andrographolide Modulates Giardia Gene Expression at Transcriptomic and Proteomi...
Sandipan Ganguly, Arjun Ghosh, Tapas Haldar, Sanjib Kumar Sardar, ...
Related Articles
Invasomes: Potential Nanocarrier System for Transdermal Delivery of Drug Molecul...
Hucche Bhimashankar, Kazi wasim, Mohite Chakradhar, Kolsure Shripal, ...
Preparation and Formulation of Adulsa Cough Syrup...
Hanuman Idhole , Swati Aswani, Dr M .D. Kitukale , ...
Pigment Power: Exploring Botanicals for Vitiligo ...
Saurabh Gadadhe , Nikita Bubera , Shraddha Gawand , Ayush parve , Satya palingi , Dr. Shrutika Patil...
More related articles
Recent Developments in UV-Visible Spectrophotometry for Sodium Benzoate Analysis...
Marri Mahesh, D. Sathish Kumar, Shaik Sanjeeda Parveen, Chalam Arju, ...
Formulation and Evaluation of Herbal Soap from Ocimum Sanctum and Aloe Vera ...
Vanshika Bhalerao, Yogesh Bhojne, Priya jaware, Santosh payghan, ...
Recent Developments in UV-Visible Spectrophotometry for Sodium Benzoate Analysis...
Marri Mahesh, D. Sathish Kumar, Shaik Sanjeeda Parveen, Chalam Arju, ...
Formulation and Evaluation of Herbal Soap from Ocimum Sanctum and Aloe Vera ...
Vanshika Bhalerao, Yogesh Bhojne, Priya jaware, Santosh payghan, ...