View Article

Abstract

Aim: The aim of this review is to highlight the transformative role of nanotechnology in targeted drug delivery systems, emphasizing the advancements in nanoparticle development, including silica-based mesoporous silica nanoparticles (MSNs) and carbon-based materials like carbon nanotubes (CNTs) and graphene oxide (GO). Purpose: This review aims to examine how the unique properties of nanoparticles—such as their small size, large surface area, and functionalization capacity—enable the efficient delivery of therapeutic agents, including proteins, nucleic acids, and chemotherapeutics. Additionally, it explores challenges related to regulatory hurdles, manufacturing scalability, and the clinical translation of nanomedicines. Discussion: Nanoparticles have revolutionized drug delivery by improving the safety and efficacy of therapeutic agents. Silica-based MSNs and carbon nanotubes (CNTs) are particularly effective due to their high drug-loading capacity, controlled release, and ability to encapsulate both hydrophilic and hydrophobic drugs. These nanoparticles enhance therapeutic efficacy while reducing systemic toxicity. Recent developments like mannosylated selenium nanoparticles target immune cells to improve outcomes, and smart nanoparticles responsive to environmental cues offer controlled drug release for increased precision. Despite these advancements, challenges remain in regulatory approval, scalable manufacturing, and standardized safety evaluations, which hinder clinical translation. Conclusion: Nanotechnology has significant potential to revolutionize drug delivery systems, particularly in treating complex diseases like cancer and autoimmune disorders. To realize this potential, overcoming regulatory, manufacturing, and standardization challenges through collaboration between researchers, regulatory bodies, and industry stakeholders is crucial for the successful clinical application of nanomedicines.

Keywords

Nanotechnology, chemotherapeutics, targeted drug delivery systems, environmental.

Introduction

Targeted medication delivery methods have greatly improved thanks to nanotechnology, which also increases the therapeutic agents' safety and effectiveness. The creation of nanomaterials that enhance medication accumulation at particular target areas while reducing systemic toxicity and adverse effects has advanced significantly over the last 20 years. Because of their tiny size, large surface area, and capacity to be functionalized for precise targeting, nanoparticles—from carbon-based materials like carbon nanotubes (CNTs) and graphene oxide (GO) to silica-based nanoparticles (SiNPs)—have become excellent options for drug delivery. Numerous therapeutic substances, including proteins, nucleic acids, and chemotherapeutics, can be designed to be carried by these nanoparticles and then precisely delivered to the target tissue or cells. Because of their superior mechanical, thermal, and electrical characteristics as well as their ease of modification with different biomolecules, carbon-based nanoparticles—like CNTs and GO—have garnered a lot of interest. For instance, CNTs can encapsulate pharmaceuticals that are hydrophilic or hydrophobic, enabling a variety of drug delivery applications. To enhance selectivity for particular cell types, like cancer cells, their surface can be functionalized with targeting ligands (such as aptamers, peptides, or antibodies). This will raise the drug's local concentration at the disease site while lowering off-target effects. Functionalized carbon nanotubes (CNTs) have been shown in studies to minimize systemic toxicity while increasing the therapeutic efficacy of medications such as doxorubicin in cancer therapy1,2.

     
            nanoparticles traveling through the bloodstream..png
       

Figure 1: Nanoparticles in Circulation: Showing nanoparticles traveling through the bloodstream.

Furthermore, graphene oxide (GO) provides a large surface area for drug loading and can be coupled with folic acid or other targeting agents to create a potent platform for targeted drug delivery2,3. Mesoporous silica nanoparticles (MSNs), a type of silica-based nanoparticle, are another material family that has demonstrated significant promise in targeted drug delivery. MSNs have a special mesoporous structure that enables regulated release characteristics and a high drug-loading capacity. Selective targeting of cancer cells or inflammatory tissues is made possible by their easy functionalization with targeting ligands like small compounds or monoclonal antibodies. MSNs are also very biocompatible and biodegradable, which lowers the possibility of long-term harm. When compared to traditional drug delivery methods, MSNs have been shown in numerous studies to be successful in delivering chemotherapeutic drugs, such as paclitaxel, with better therapeutic outcomes and fewer side effects4,5.

Novel approaches, such as the application of mannosylated selenium nanoparticles (MSNs-Se), have opened up new avenues for improving medication administration. Because of its well-known antioxidant qualities, selenium has been added to nanoparticles to boost immune responses and lower oxidative stress, thus increasing the therapeutic index of anticancer medications. Targeted distribution to dendritic cells and macrophages, which are essential for immunological responses, is made easier by mannosylation, the process of binding mannose molecules to the surface of nanoparticles. This approach has been very successful in immune regulation and macrophage-targeted medication delivery for cancer treatment. According to recent research, mannosylated selenium nanoparticles can effectively transport medications like doxorubicin and cisplatin to macrophages, improving drug absorption and suppressing tumors6.

  1. Overview Of Nanotechnology In Drug Delivery:

By manipulating materials at the nanoscale, nanotechnology can produce medication delivery systems that can target certain tissues or cells. Materials display distinct physical, chemical, and biological characteristics at the nanoscale that are not seen in their bulk counterparts. Nanoparticles are especially useful in drug delivery applications because of their high surface area-to-volume ratios, improved permeability, and capacity to be functionalized with targeted molecules. Compared to conventional drug delivery techniques, the design and development of drug delivery systems at the nanoscale enables more accurate control over the release, stability, and distribution of therapeutic substances, resulting in increased efficacy and fewer side effects. Usually between 1 to 1000 nanometers in size, nanoparticles are designed to deliver a variety of therapeutic agents, including proteins, nucleic acids, tiny compounds, and even entire cells, with the benefit of targeted delivery. Among the most widely utilized drug delivery vehicles are nanomaterials, including liposomes, dendrimers, carbon nanotubes (CNTs), and mesoporous silica nanoparticles (MSNs). To enable precision drug administration at the site of action, these nanoparticles can be functionalized with particular ligands, such as antibodies, peptides, or small molecules, that bind preferentially to overexpressed receptors on the surface of target cells, such as tumor cells or immune cells7,8. For instance, the hollow core of carbon nanotubes (CNTs) has been the subject of much research due to its capacity to transport hydrophobic medicines, and its enormous surface area enables functionalization with targeted ligands9. By delivering chemotherapy chemicals directly to tumor cells, CNTs have demonstrated considerable promise in cancer therapy. This reduces systemic toxicity while increasing the effectiveness of medications like doxorubicin. The capacity of liposomes, spherical vesicles made of lipid bilayers, to encapsulate both hydrophilic and hydrophobic substances makes them popular for targeted medication delivery. Tumor-targeting medicines, such as antibodies against certain cancer antigens, can be functionalized into liposomes to enable the selective accumulation of medications at the tumor site, enhancing therapeutic results and minimizing adverse effects10. Nanoparticles can be designed to target particular cells and deliver drugs in a controlled or sustained manner. Mesoporous silica nanoparticles (MSNs), for instance, have garnered interest because of their high drug-loading capacity, adjustable pore diameters, and regulated drug release in response to particular environmental stimuli like temperature or pH. Targeting moieties such as transferrin or folic acid can be functionalized into MSNs to improve medication accumulation in cancer cells or other target organs11. In chronic conditions, such as cancer, infections, and neurological disorders, where prolonged therapeutic action without regular dosage is necessary, this sustained release aspect is especially crucial12. Another fascinating aspect of nanomedicine is the capacity of nanoparticles to penetrate biological barriers, including the blood-brain barrier (BBB). For instance, it has been demonstrated that functionalized nanoparticles containing peptides or antibodies that target particular receptors on the BBB endothelium improve medication delivery to the brain, creating new avenues for the treatment of neurological conditions including Parkinson's and Alzheimer's13. These systems lessen systemic toxicity while improving the bioavailability and therapeutic effectiveness of medications. Higher drug concentrations at the target site and better therapeutic results can result from the substantial improvement of therapeutic agents' solubility, stability, and release kinetics provided by nanoparticle-based drug delivery systems. These systems successfully lessen toxicity and undesirable side effects, which are frequently a significant drawback of traditional drug delivery techniques, by reducing the interaction of the medication with non-target tissues. Liposomal formulations, for instance, have been demonstrated to improve the sustained release and bioavailability of poorly soluble medications while lowering systemic toxicity14 by minimizing the exposure of the drug to healthy tissues. Furthermore, mesoporous silica nanoparticles (MSNs) and carbon nanotubes (CNTs) can be designed to increase drug loading capacity and release pharmaceuticals selectively at specific locations, including tumors, improving the safety and effectiveness of chemotherapy medications15,16. By allowing regulated drug release and targeting particular cells or tissues, dendrimer-based delivery systems further demonstrate how nanomaterials can increase therapeutic efficacy, improving treatment outcomes while reducing side effects17. Furthermore, precise medication delivery to certain cells, tissues, or organs has shown significant promise when using nanoparticles functionalized with targeting ligands like peptides or antibodies. Because cancer cells overexpress folate receptors, for example, folic acid-modified nanoparticles can specifically target these cells, decreasing off-target effects and increasing drug delivery efficiency18. This focused strategy reduces the possibility of harmful effects in healthy tissues while simultaneously improving therapeutic efficacy. Consequently, these systems present the possibility of more efficacious therapies with less adverse effects, especially in the management of cancer and other chronic illnesses19.

  1. Types of Nanomaterial used:

3.1 Carbon-based Nanoparticles:

The remarkable qualities of graphene and carbon nanotubes (CNTs), such as their high surface area, mechanical strength, electrical conductivity, and biocompatibility, have drawn a lot of interest in nanomedicine. Because these materials may encapsulate or adsorb a wide range of therapeutic agents, including tiny chemicals, proteins, nucleic acids, and even big living entities like viruses, they are being used more and more for medication delivery. The substantial functionalization made possible by graphene and carbon nanotubes' high surface area makes it possible to attach different targeting ligands, like peptides, antibodies, or small molecules, to accomplish selective drug delivery to particular cells or tissues, like inflammatory or malignant sites20, 21. Because of its water solubility, ease of functionalization, and potential for effective drug loading, graphene oxide (GO), a derivative of graphene, is especially preferred for drug delivery. To better targeting to tumor cells and promote cellular uptake, GO can be modified with a variety of bioactive compounds. This will decrease off-target effects and improve the effectiveness of treatment. Chemotherapeutic drugs like doxorubicin, for instance, have been delivered by GO and have shown better accumulation at tumor locations and decreased systemic toxicity in preclinical models22,23. To further improve medication delivery selectivity, GO's surface can be altered to incorporate functional groups that encourage interactions with cancer cell receptors or other particular biomarkers24. Single-walled (SWCNTs) and multi-walled (MWCNTs) carbon nanotubes (CNTs) are cylindrical structures composed of rolled-up graphene sheets. They are perfect for drug administration because of their high aspect ratio, robust mechanical characteristics, and capacity to pass through cell membranes. Because of their hollow shape, CNTs can hold both hydrophilic and hydrophobic medications, and surface functionalization enhances drug release control. According to recent research, CNTs can be employed to deliver anticancer medications, like doxorubicin and paclitaxel, to specific cancer cells, increasing the therapeutic index of these medications while reducing their adverse effects25,26. Furthermore, to ensure localized and regulated drug release at the disease site, CNTs have been designed to release their payload in response to environmental cues like pH or temperature27. It has been demonstrated that both graphene and carbon nanotubes are naturally biocompatible and low toxicity, particularly when functionalized to lessen aggregation or non-specific attachment to healthy cells. However, studies into the safe and effective removal of these nanomaterials from the body have been spurred by worries over their long-term buildup in tissues28. Graphene and carbon nanotube (CNT)-based medication delivery systems that are biodegradable and excretable through natural metabolic routes are being designed in an attempt to allay these worries29.

3.2 Silica-based Nanoparticles:

Because of their exceptional biocompatibility, high surface area, and adjustable pore size, mesoporous silica nanoparticles (MSNs) are one of the most researched silica-based nanomaterials for drug delivery applications. The structured mesoporous structure of these nanoparticles offers a wide surface area for drug loading and permits controlled drug release. MSNs can be used to encapsulate a range of medicinal substances, from tiny compounds to bigger macromolecules like proteins and nucleic acids, because their pore diameters can usually be regulated between 2 and 50 nm30,31. For medications that need higher dosages to be effective, MSNs' vast surface area and pore capacity allow for significant drug-loading capabilities, which are essential for improving therapeutic efficacy32. MSNs are very useful for enhancing the pharmacokinetics of pharmacological therapy because of their capacity to release medications in a regulated fashion. MSNs can be made to release medications that are encapsulated in response to particular environmental cues, such temperature, ionic strength, or pH. By enabling pH-responsive drug release and enhanced selectivity for cancer cells, this property is especially beneficial for targeting tumor locations, where the extracellular environment is usually more acidic than normal tissues33,34. To improve drug accumulation at the target location and reduce systemic exposure and toxicity, MSNs functionalized with pH-sensitive groups, for example, have been demonstrated to release chemotherapeutic medicines, such as doxorubicin, more efficiently in acidic environments, such as those prevalent in tumors35.

Functionalization of MSNs with targeting ligands, such as peptides, small molecules, or monoclonal antibodies, improves their selectivity and targeting capability in addition to controlled release. Therapeutic drugs can be delivered selectively to particular cells, like cancer cells, which frequently overexpress particular receptors, thanks to this functionalization. For instance, MSNs functionalized with folic acid can enhance the therapeutic index of anticancer drugs by precisely targeting folate receptors on the surface of cancer cells36. Similarly, in order to get beyond obstacles like the blood-brain barrier (BBB) in the treatment of neurological disorders, transferrin-functionalized MSNs have been used for targeted delivery to cells that overexpress the transferrin receptor, such as brain cells and some forms of cancer37. When using MSNs for drug delivery, biocompatibility and biodegradability are important factors to take into account. MSNs' silica structure is generally well-tolerated by biological systems, and it is simple to alter to improve biodegradability and lower the possibility of long-term tissue accumulation. In order to have prolonged therapeutic benefits, MSNs must be stable in the bloodstream and have their circulation time extended. This can be achieved through a variety of surface modifications, such as coating with polymers like polyethylene glycol (PEG) or functionalization with targeted ligands38. Furthermore, MSNs are safer than conventional drug delivery methods since they can be made to break down and release their cargo in response to particular physiological cues39.

3.3 Iron Oxide Nanoparticles:

Because of their special magnetic characteristics, which allow for both magnetic targeting and imaging, iron oxide nanoparticles (IONPs), in particular magnetite (FeO?) and maghemite (FeO?), have garnered a lot of interest in nanomedicine. These nanoparticles are frequently employed to improve drug delivery accuracy by enabling the use of external magnetic fields to guide medications to particular bodily locations. Because IONPs may be controlled by magnetic fields and have ideal circulation durations and cellular uptake, they are commonly manufactured in the nanometer size range (1-100 nm), offering a great degree of control over drug delivery and localization40,41. Drugs can accumulate in the intended location with little diffusion to non-target tissues thanks to IONPs' magnetic targeting capabilities. When IONPs are functionalized with anticancer medications and guided to tumor sites by an external magnetic field, this is especially beneficial for cancer therapy. For example, when used for targeted therapy in cancer cells, superparamagnetic iron oxide nanoparticles (SPIONs), which show no residual magnetization after the magnetic field is removed, can be loaded with chemotherapeutic agents such as doxorubicin or paclitaxel. This greatly increases the therapeutic index by reducing systemic side effects^42,43. IONPs have been thoroughly investigated for their potential uses in magnetic resonance imaging (MRI) in addition to medication delivery. Iron oxide nanoparticles' magnetic characteristics enable them to function as efficient contrast agents in MRIs, enabling high-resolution imaging of organs and tissues. The accuracy and tracking of therapeutic interventions are improved by the real-time tracking and monitoring of drug-loaded nanoparticle distribution. In preclinical and clinical research, for instance, SPIONs have been used to view and monitor the accumulation of drug delivery systems, offering important insights into biodistribution, targeting effectiveness, and release kinetics44,45. Functionalization of iron oxide nanoparticles with targeting ligands, such as antibodies, peptides, or small molecules, enhances the specificity of drug administration in addition to their use in imaging and drug delivery. For instance, IONPs can be coupled to antibodies that target certain tumor antigens or cell surface receptors, allowing for the selective targeting of tumor cells and improving the overall therapeutic result^46. In addition to cancer, this targeting ability can be used for gene delivery, the treatment of infections, and cardiovascular disorders47. The biocompatibility and biodegradability of iron oxide nanoparticles are important advantages. The reticuloendothelial system (RES), especially the liver and spleen, is the main route by which iron oxide particles are safely removed from the body because they are often non-toxic. Because of this, they are a safer substitute for other nanoparticle materials that could eventually build up in the body, including gold or carbon nanotubes. Nonetheless, there is a continuous endeavor to enhance the pharmacokinetics and reduce any possible negative consequences of IONPs, including the potential for immune system activation or iron overload48.

  1. Mechanism of targeted delivery:

4.1 Passive Targeting: Exploiting the EPR Effect for Tumor-Specific Nanoparticle Accumulation:

A popular tactic in the creation of drug delivery systems based on nanoparticles, especially in cancer treatment, is passive targeting. This technique takes advantage of the Enhanced Permeability and Retention (EPR) effect, which is a process whereby the distinct physiological characteristics of the tumor microenvironment enable nanoparticles to preferentially aggregate in tumor tissues. Abnormal, leaky blood vessels with increased inter-endothelial gaps are frequently seen in tumor vasculature. These features make it easier for nanoparticles to enter the tumor tissue from the circulation. Additionally, these capillaries lack the tight control that is normally observed in normal tissues, making them loosely structured, which increases the permeability of nanoparticles49, 50. Furthermore, tumors frequently have impaired lymphatic drainage, which hinders the effective removal of extravasated nanoparticles from the tumor site and causes them to remain there for an extended period of time 51. The EPR effect is a passive process that is mostly caused by anomalies in the structure and function of the tumor vasculature, not by particular interactions between tumor cells and nanoparticles. By extravasating through the endothelial gaps and accumulating in the tumor tissue, nanoparticles that fall within the ideal size range of roughly 10 to 200 nm can take advantage of the tumor's leaky vasculature. Particles in this size range are small enough to flow through the permeable tumor vasculature, but large enough to avoid fast renal clearance, which usually happens with smaller nanoparticles52. Once inside the tumor, the nanoparticles become stuck in the tumor microenvironment due to the absence of effective lymphatic outflow, which improves targeted drug delivery53. The capacity of nanoparticles to move through the tumor vasculature and gather in tumor tissues is crucial to the effectiveness of passive targeting. The size, surface charge, and surface modification of the nanoparticles are some of the variables that affect how effective passive targeting is. Specifically, nanoparticles smaller than 10 nm are quickly eliminated by the kidneys, whereas those larger than 200 nm may encounter more steric resistance and so have trouble extravasating through the tumor vasculature54. Consequently, the capacity of nanoparticles to efficiently utilize the EPR effect depends on their design falling within the ideal size range. Surface changes can further extend the period that nanoparticles circulate in the bloodstream, increasing their capacity to reach and concentrate in tumors. One such alteration is the conjugation of polyethylene glycol (PEG) to the nanoparticle surface, also referred to as PEGylation55. To take use of the EPR effect, several kinds of nanoparticles have been created, such as dendrimers, liposomes, polymeric nanoparticles, and gold nanoparticles. Passive targeting works especially effectively with liposomes, which are lipid-based vesicles that can encapsulate a variety of medicinal substances. They are among the most extensively researched nanoparticle systems for drug administration because of their capacity to encapsulate both hydrophilic and hydrophobic medicines, as well as the possibility of surface modification to extend circulation56. For instance, it is well known that PEGylated liposomes have a longer blood circulation, which increases their ability to accumulate at tumor locations via the EPR effect57. With the benefits of controlled drug release and decreased systemic toxicity, polymeric nanoparticles which are derived from biodegradable polymers such as poly (lactic-co-glycolic acid) (PLGA) have also been thoroughly investigated for drug delivery58. Furthermore, gold nanoparticles have demonstrated potential in utilizing the EPR effect for tumor-targeted drug delivery and imaging due to their ease of functionalization for a variety of applications59. Because of their highly branching, tree-like architectures, dendritic nanoparticles provide extensive surface surfaces for drug loading and can improve drug retention at tumor locations by promoting the EPR effect60. Even with passive targeting's encouraging promise, there are still a number of obstacles to overcome before it may be used in clinical settings. Different cancers or even different parts of the same tumor may accumulate nanoparticles differently due to the heterogeneity of malignancies in terms of vascular permeability and tumor architecture61. Furthermore, the effectiveness of passive targeting may be limited by the high interstitial fluid pressure frequently present in solid tumors, which can prevent nanoparticles from penetrating deeper tumor regions62. Moreover, the effective delivery of therapeutic medicines can be made more difficult by the extracellular matrix (ECM) in tumors, which can function as a barrier to nanoparticle diffusion63. Researchers have concentrated on improving the design of nanoparticles to increase their penetration and retention within the tumor microenvironment in order to overcome these obstacles. To improve the overall efficacy of nanoparticle-based therapeutics, strategies such altering the size, shape, and surface characteristics of nanoparticles as well as combining passive targeting with active targeting techniques have been proposed64. To further improve the release and accumulation of nanoparticles at tumor locations, methods that make use of external stimuli—like light, heat, or pH changes—have also been investigated65.

    1. Active Targeting: Enhancing Specificity in Nanoparticle Delivery:

Modifying nanoparticles with specialized ligands—such as antibodies, peptides, or small molecules—that can attach to receptors that are overexpressed on the surface of target cells—especially tumor cells—is known as active targeting. By using this method, medicinal drugs can be delivered to the intended location more precisely, limiting systemic toxicity and off-target consequences. Targeting ligands can be attached to the surface of nanoparticles to greatly increase their uptake by target cells, frequently by receptor-mediated endocytosis. Monoclonal antibodies, such trastuzumab, which targets HER2 receptors in breast cancer, and peptides that detect integrins or other tumor-associated markers are examples of frequently utilized ligands66,67. Active targeting has emerged as a promising cancer therapeutic technique due to its capacity to directly target cancer cells, opening the door to more localized and effective treatment68,69.

  1. Recent Innovations:

5.1 Mannosylated Selenium Nanoparticles: Targeting Immune Cells for Enhanced Therapeutic Efficacy

With its ability to specifically target immune cells, especially macrophages and dendritic cells, mannosylated selenium nanoparticles (Man-SeNPs) have shown great promise in improving treatment results for a number of illnesses, including autoimmune disorders and cancer. These nanoparticles' surface is coupled with mannose, a sugar molecule that selectively attaches itself to immune cell surface-expressed mannose receptors (MRs). This focused strategy makes it easier for immune cells to absorb the nanoparticles, which improves medication delivery to the targeted cellular populations70,71. Because it can alter immune responses and have lethal effects on tumor cells, selenium, which is well-known for its antioxidant qualities, further enhances the therapeutic efficacy of these nanoparticles72. According to studies, Man-SeNPs can deliver biologics like cytokines or immunomodulatory medicines as well as small-molecule medications with greater selectivity and less off-target toxicity73. They are a flexible tool in both drug administration and immunotherapy applications, and their capacity to elicit immunological responses, especially by activating antigen-presenting cells, shows promise for cancer treatment74.

5.2 Smart Nanoparticles: Stimuli-Responsive Drug Delivery Systems

In order to enable regulated medication release at specified places, smart nanoparticles a sophisticated type of nanocarriers—are made to react dynamically to particular environmental stimuli, such as pH, temperature, or enzymes. The materials used to create these nanoparticles change structurally or chemically in response to the physiological conditions found in the inflammatory tissues or tumor microenvironment. pH-sensitive nanoparticles, for instance, use the acidic environment of tumors or inflammatory tissues to release encapsulated medications only when they get at the target site75. Likewise, when the target temperature threshold is met, temperature-sensitive nanoparticles might react to localized tissue heating or hyperthermia, facilitating the release of therapeutic agents76. By reducing systemic toxicity and enhancing therapeutic efficacy, these responsive systems provide the benefit of spatiotemporal control over drug release. Polymers that can undergo conformational changes in response to environmental stimuli, such as poly(lactic-co-glycolic acid) (PLGA), poly(N-isopropylacrylamide) (PNIPAM), or lipids, are frequently used to create smart nanoparticles77.

To further improve drug delivery specificity and guarantee that the medicine is delivered just at the location of disease, these nanoparticles can also be coupled with targeting ligands78. Smart nanoparticle development has demonstrated encouraging outcomes in chronic inflammation, cancer treatment, and even gene transfer, highlighting its potential in precision medicine79.

  1. Challenges and future direcions:

6.1 Regulatory Hurdles: Navigating the Complexities of Nanomedicine Approval

Nanomedicines' distinct qualities, which set them apart from traditional medications, make the approval procedure extremely difficult. Size, surface charge, and surface chemistry are some of the unique physicochemical properties of nanoparticles that might affect their pharmacokinetics, biodistribution, and toxicity profiles in unpredictable and poorly understood ways. These intricacies necessitate specific testing and regulatory systems, making the assessment of safety and efficacy more difficult. Guidelines for evaluating nanomedicines have been produced by regulatory bodies including the European Medicines Agency (EMA) and the U.S. Food and Drug Administration (FDA), but they are constantly changing due to the quick development of nanotechnology. The FDA, for instance, has established guidelines for preclinical safety testing of nanomaterials, which includes investigations into their potential for long-term impacts, immunogenicity, and toxicity80,81. Moreover, producers seeking certification face additional challenges due to the absence of established techniques for characterizing nanoparticles, specifically with regard to batch-to-batch consistency, stability, and interactions with biological systems^82. Despite the remarkable therapeutic potential of nanomedicines, the lack of clarity surrounding these aspects has caused delays in their commercialization. Furthermore, as nanomedicines' unique qualities may present patients with unanticipated dangers, it is imperative that strong post-marketing surveillance be in place to track the long-term effects of these treatments after they are approved83.

6.2 Manufacturing and Scalability: Overcoming Challenges in Nanomaterial Production:

The creation of economical and repeatable nanomaterial production procedures is one of the main obstacles to the clinical translation of nanomedicines. Complex synthesis methods including solvent evaporation, nanoprecipitation, and emulsion polymerization are frequently used in the creation of nanoparticles, and they can be challenging to scale up while preserving constant quality and performance84,85. Ensuring safety and efficacy requires repeatability across many production batches; however, batch-to-batch variability can be substantial because to the susceptibility of nanoparticles to environmental variables, including temperature, pH, and solvent composition86. Furthermore, it is difficult to maintain the appropriate size, shape, and surface characteristics on a wide scale because even little variations can change the nanomedicine's pharmacokinetics and therapeutic results87. Attempts to scale up production for commercial usage are made more difficult by the absence of standardized manufacturing processes for nanomaterials. Technological developments in nanomanufacturing, including automated processes, microfluidics, and continuous flow synthesis, have demonstrated promise in increasing cost-effectiveness and scalability88.

    1. Clinical Translation: Bridging the Gap Between Lab and Clinic:

It is still very difficult to translate the encouraging outcomes of laboratory-based nanomedicine research into clinical settings. Even while preclinical models of nanoparticle-based medication delivery systems have advanced quickly, many of these developments encounter challenges when they are tested on humans. Important concerns include the possibility for unexpected toxicities, the diversity of nanoparticle activity in complex biological systems, and the distinctions between human physiology and animal models89,90. Furthermore, the clinical translation process is made more difficult by the absence of established techniques for evaluating the efficacy and safety of nanomedicines. For instance, because of variations in the pharmacokinetics of nanoparticles in people or their interactions with the immune system, preclinical success may not always translate into clinical outcomes91. Furthermore, there are major obstacles to speeding up clinical studies due to the constantly changing regulatory criteria for nanomedicines92. More study is required to better understand the long-term behavior of nanoparticles in the human body, improve their design for particular therapeutic targets, and create trustworthy biomarkers for tracking their distribution and activity in clinical settings in order to overcome these obstacles93.

  1. DISCUSSION:

Drug delivery methods have been completely transformed by nanotechnology, especially in the field of targeted medicines, which has improved safety and efficacy. By precisely delivering therapeutic chemicals to particular cells or tissues, nanoparticles' special qualities such as their small size, large surface area, and capacity for functionalization help to reduce systemic toxicity and adverse effects. Because of their adaptability and simplicity of modification, carbon-based nanoparticles such as graphene oxide (GO) and carbon nanotubes (CNTs) have attracted interest. Numerous therapeutic compounds can be encapsulated in these nanoparticles, enabling targeted delivery to cancer cells. Functionalized carbon nanotubes (CNTs) have demonstrated the potential to cure cancer by increasing the therapeutic efficacy of medications such as doxorubicin while decreasing off-target effects.  Silica-based nanoparticles, particularly mesoporous silica nanoparticles (MSNs), offer special benefits because of their excellent drug-loading capacity and adjustable pore diameters. Their targeting powers are further improved by their capacity to release medications in response to environmental cues, such as pH shifts in tumor microenvironments. Enhancing therapeutic results, functionalization with ligands enables targeted distribution to particular cell types. Another important development is iron oxide nanoparticles (IONPs), which allow magnetic targeting and imaging. Because of their magnetic characteristics, medications can be precisely accumulated at tumor locations, increasing therapeutic efficacy while reducing systemic distribution. Real-time insights into the dynamics of medication distribution are made possible by the capacity to follow these nanoparticles using magnetic resonance imaging (MRI). The potential of nanotechnology to boost immune responses and target immune cells for better therapeutic results is demonstrated by recent developments like mannosylated selenium nanoparticles. A promising approach to controlled medication release that could improve the accuracy of drug delivery systems is the use of smart nanoparticles that react to environmental cues. Notwithstanding these developments, there are still issues, namely with clinical translation, manufacturing scalability, and regulatory approval. The intricacy of producing nanoparticles makes it difficult to achieve uniform quality, and their special qualities call for specialized regulatory frameworks. Furthermore, a better comprehension of the behavior of nanoparticles in biological systems is necessary to convert preclinical achievements into clinical applications.

  1. CONCLUSION :

Drug distribution has seen a revolution because to nanotechnology, which provides creative ways to improve the effectiveness and safety of medicinal substances. Numerous platforms for targeted drug delivery have been made possible by the development of several nanoparticle systems, including as iron oxide nanoparticles, silica-based mesoporous nanoparticles, and carbon-based materials like carbon nanotubes and graphene oxide. High surface area, adjustable release profiles, and the capacity to be functionalized with targeting ligands are some of the special qualities of these nanoparticles that enable targeted administration to particular tissues or cells, especially in oncology. By targeting immune cells and facilitating stimuli-responsive medication release, recent developments like mannosylated selenium nanoparticles and smart nanoparticles demonstrate the potential for significantly improving therapeutic outcomes. These developments not only raise the therapeutic index of currently available medications but also create new therapeutic options for treating complicated illnesses including autoimmune disorders and cancer. The clinical translation of nanomedicines nevertheless faces a number of obstacles, notwithstanding these encouraging advancements. The transition from laboratory research to clinical use is complicated by regulatory barriers, manufacturing scalability, and the requirement for uniform assessment techniques. In order to create strong guidelines and manufacturing methods, researchers, regulatory bodies, and industry stakeholders must work together to address these issues.

  1. Conflict of Interest:None.
  2. Acknowledgement:Not applicable.
  3. Source of Funding:None

 

REFERENCES

  1. Brown KJ, Downs CT. Seasonal patterns in body temperature of free-living rock hyrax (Procavia capensis). Comparative Biochemistry and Physiology Part A: Molecular & Integrative Physiology. 2006 Jan 1;143(1):42-9.
  2. Chun AL. Nanomaterials: Mercury mop. Nature Nanotechnology. 2008 Jul 11.
  3. Han XM, Zheng KW, Wang RL, Yue SF, Chen J, Zhao ZW, Song F, Su Y, Ma Q. Functionalization and optimization-strategy of graphene oxide-based nanomaterials for gene and drug delivery. American Journal of Translational Research. 2020;12(5):1515.
  4. Rani R, Malik P, Dhania S, Mukherjee TK. Recent advances in mesoporous silica nanoparticle-mediated drug delivery for breast cancer treatment. Pharmaceutics. 2023 Jan 9;15(1):227.
  5. Hwang SR, Chakraborty K, An JM, Mondal J, Yoon HY, Lee YK. Pharmaceutical aspects of nanocarriers for smart anticancer therapy. Pharmaceutics. 2021 Nov 5;13(11):1875.
  6. Rastegari E, Hsiao YJ, Lai WY, Lai YH, Yang TC, Chen SJ, Huang PI, Chiou SH, Mou CY, Chien Y. An update on mesoporous silica nanoparticle applications in nanomedicine. Pharmaceutics. 2021 Jul 12;13(7):1067.
  7. Bjarnsholt T, Ciofu O, Molin S, Givskov M, Høiby N. Applying insights from biofilm biology to drug development—can a new approach be developed?. Nature reviews Drug discovery. 2013 Oct;12(10):791-808.
  8. Singh R, Lillard Jr JW. Nanoparticle-based targeted drug delivery. Experimental and molecular pathology. 2009 Jun 1;86(3):215-23.
  9. Xing H, Hwang K, Lu Y. Recent developments of liposomes as nanocarriers for theranostic applications. Theranostics. 2016;6(9):1336.
  10. Darban SA, Nikoofal-Sahlabadi S, Amiri N, Kiamanesh N, Mehrabian A, Zendehbad B, Gholizadeh Z, Jaafari MR. Targeting the leptin receptor: To evaluate therapeutic efficacy and anti-tumor effects of Doxil, in vitro and in vivo in mice bearing C26 colon carcinoma tumor. Colloids and Surfaces B: Biointerfaces. 2018 Apr 1;164:107-15.
  11. Zhang L, Gu FX, Chan JM, Wang AZ, Langer RS, Farokhzad OC. Nanoparticles in medicine: therapeutic applications and developments. Clinical pharmacology & therapeutics. 2008 May;83(5):761-9.
  12. Choudhari M, Hejmady S, Saha RN, Damle S, Singhvi G, Alexander A, Kesharwani P, Dubey SK. Evolving new-age strategies to transport therapeutics across the blood-brain-barrier. International Journal of Pharmaceutics. 2021 Apr 15;599:120351.
  13. Kreuter J. Nanoparticulate systems for brain delivery of drugs. Advanced drug delivery reviews. 2001 Mar 23;47(1):65-81.
  14. Bianco A, Kostarelos K, Prato M. Applications of carbon nanotubes in drug delivery. Curr Opin Chem Biol. 2005;9(6):674–9. doi:10.1016/j.cbpa.2005.10.020.
  15. Chun AL. Nanomaterials: Mercury mop. Nature Nanotechnology. 2008 Jul 11.
  16. Han XM, Zheng KW, Wang RL, Yue SF, Chen J, Zhao ZW, Song F, Su Y, Ma Q. Functionalization and optimization-strategy of graphene oxide-based nanomaterials for gene and drug delivery. American Journal of Translational Research. 2020;12(5):1515.
  17. Kumar P, Huo P, Zhang R, Liu B. Antibacterial properties of graphene-based nanomaterials. Nanomaterials. 2019 May 13;9(5):737.
  18. Zhang L, Gu FX, Chan JM, Wang AZ, Langer RS, Farokhzad OC. Nanoparticles in medicine: therapeutic applications and developments. Clinical pharmacology & therapeutics. 2008 May;83(5):761-9.
  19. Prasad RD, Desai CB, Shrivastav OP, Charmode N, Prasad SR, Samant A, Mirajkar R, Banga S, Shaikh VS, Padvi MN, Patil S. A Critical Review on Design and Development of Carbonaceous Materials for Veterinary Medicine. ES Food & Agroforestry. 2022 Sep 8;9:15-38.
  20. Kumar N, Chamoli P, Misra M, Manoj MK, Sharma A. Advanced metal and carbon nanostructures for medical, drug delivery and bio-imaging applications. Nanoscale. 2022;14(11):3987-4017.
  21. D?ugosz O, Szostak K, Staro? A, Pulit-Prociak J, Banach M. Methods for reducing the toxicity of metal and metal oxide NPs as biomedicine. Materials. 2020 Jan 8;13(2):279.
  22. Rane BR, Patil VL, Mhatre NR, Padave AP. Recent advances in mesoporous silica nanoparticles research. Mesoporous Silica Nanoparticles: Drug Delivery, Catalysis and Sensing Applications. 2024 Nov 18:187.
  23. Inam H, Sprio S, Tavoni M, Abbas Z, Pupilli F, Tampieri A. Magnetic hydroxyapatite nanoparticles in regenerative medicine and nanomedicine. International Journal of Molecular Sciences. 2024 Feb 28;25(5):2809.
  24. Rani R, Malik P, Dhania S, Mukherjee TK. Recent advances in mesoporous silica nanoparticle-mediated drug delivery for breast cancer treatment. Pharmaceutics. 2023 Jan 9;15(1):227.
  25. Kim K, Bou-Ghannam S, Kameishi S, Oka M, Grainger DW, Okano T. Allogeneic mesenchymal stem cell sheet therapy: A new frontier in drug delivery systems. Journal of Controlled Release. 2021 Feb 10;330:696-704.
  26. Zhang L, Gu FX, Chan JM, Wang AZ, Langer RS, Farokhzad OC. Nanoparticles in medicine: therapeutic applications and developments. Clinical pharmacology & therapeutics. 2008 May;83(5):761-9.
  27. Li Z, Zhang Y, Feng N. Mesoporous silica nanoparticles: synthesis, classification, drug loading, pharmacokinetics, biocompatibility, and application in drug delivery. Expert opinion on drug delivery. 2019 Mar 4;16(3):219-37.
  28. Nam L, Coll C, Erthal LC, De la Torre C, Serrano D, Martínez-Máñez R, Santos-Martínez MJ, Ruiz-Hernández E. Drug delivery nanosystems for the localized treatment of glioblastoma multiforme. Materials. 2018 May 11;11(5):779.
  29. Varma LT, Singh N, Gorain B, Choudhury H, Tambuwala MM, Kesharwani P, Shukla R. Recent advances in self-assembled nanoparticles for drug delivery. Current Drug Delivery. 2020 May 1;17(4):279-91.
  30. Li R, Peng F, Cai J, Yang D, Zhang P. Redox dual-stimuli responsive drug delivery systems for improving tumor-targeting ability and reducing adverse side effects. Asian journal of pharmaceutical sciences. 2020 May 1;15(3):311-25.
  31. Muheem A, Baboota S, Ali J. An in-depth analysis of novel combinatorial drug therapy via nanocarriers against HIV/AIDS infection and their clinical perspectives: a systematic review. Expert Opinion on Drug Delivery. 2021 Aug 3;18(8):1025-46.
  32. Cepraga C. Two-photon chromophore-polymer conjugates grafted onto gold nanoparticles as fluorescent probes for bioimaging and photodynamic therapy applications (Doctoral dissertation, INSA de Lyon).
  33. Trombino S, Cassano R. Special Issue on designing hydrogels for controlled drug delivery: Guest Editors’ Introduction. Pharmaceutics. 2020 Jan 10;12(1):57.
  34. Gupta AK, Gupta M. Synthesis and surface engineering of iron oxide nanoparticles for biomedical applications. biomaterials. 2005 Jun 1;26(18):3995-4021.
  35. Shen Z, Wu A, Chen X. Iron oxide nanoparticle based contrast agents for magnetic resonance imaging. Molecular pharmaceutics. 2017 May 1;14(5):1352-64.
  36. Campos EA, Pinto DV, Oliveira JI, Mattos ED, Dutra RD. Synthesis, characterization and applications of iron oxide nanoparticles-a short review. Journal of Aerospace Technology and Management. 2015;7(3):267-76.
  37. Javed Y, Hussain MI, Yaseen M, Asif M. Gold–Iron Oxide Nanohybrids: Characterization and Biomedical Applications. Hybrid Nanocomposites. 2019 Mar 11:285-332.
  38. Spernath A, Aserin A, Ziserman L, Danino D, Garti N. Phosphatidylcholine embedded microemulsions: Physical properties and improved Caco-2 cell permeability. Journal of controlled release. 2007 Jun 22;119(3):279-90.
  39. Gupta AK, Gupta M. Synthesis and surface engineering of iron oxide nanoparticles for biomedical applications. biomaterials. 2005 Jun 1;26(18):3995-4021.
  40. Laurent S, Forge D, Port M, Roch A, Robic C, Vander Elst L, Muller RN. Magnetic iron oxide nanoparticles: synthesis, stabilization, vectorization, physicochemical characterizations, and biological applications. Chemical reviews. 2008 Jun 11;108(6):2064-110.
  41. Gaspar MM, Calado S, Pereira J, Ferronha H, Correia I, Castro H, Tomás AM, Cruz ME. Targeted delivery of paromomycin in murine infectious diseases through association to nano lipid systems. Nanomedicine: Nanotechnology, Biology and Medicine. 2015 Oct 1;11(7):1851-60.
  42. Debnath S, Deb K, Saha B, Das R. X-ray diffraction analysis for the determination of elastic properties of zinc-doped manganese spinel ferrite nanocrystals (Mn0. 75Zn0. 25Fe2O4), along with the determination of ionic radii, bond lengths, and hopping lengths. Journal of Physics and Chemistry of Solids. 2019 Nov 1;134:105-14.
  43. Spernath A, Aserin A, Ziserman L, Danino D, Garti N. Phosphatidylcholine embedded microemulsions: Physical properties and improved Caco-2 cell permeability. Journal of controlled release. 2007 Jun 22;119(3):279-90.
  44. Arruebo M, Fernández-Pacheco R, Ibarra MR, Santamaría J. Magnetic nanoparticles for drug delivery. Nano today. 2007 Jun 1;2(3):22-32.
  45. Selvarajan V, Obuobi S, Ee PL. Silica nanoparticles—a versatile tool for the treatment of bacterial infections. Frontiers in Chemistry. 2020 Jul 15;8:602.
  46. Wust P, Hildebrandt B, Sreenivasa G, Rau B, Gellermann J, Riess H, Felix R, Schlag PM. Hyperthermia in combined treatment of cancer. The lancet oncology. 2002 Aug 1;3(8):487-97.
  47. Dang Y, Guan J. Nanoparticle-based drug delivery systems for cancer therapy. Smart Materials in Medicine. 2020 Jan 1;1:10-9.
  48. Vaiserman A, Koliada A, Zayachkivska A, Lushchak O. Nanodelivery of natural antioxidants: an anti-aging perspective. Frontiers in bioengineering and biotechnology. 2020 Jan 10;7:447.
  49. Maeda H. The 35th anniversary of the discovery of EPR effect: a new wave of nanomedicines for tumor-targeted drug delivery—personal remarks and future prospects. Journal of personalized medicine. 2021 Mar 22;11(3):229.
  50. Jain RK. Vascular and interstitial barriers to delivery of therapeutic agents in tumors. Cancer and Metastasis Reviews. 1990 Nov;9:253-66.
  51. Fang J, Nakamura H, Maeda H. The EPR effect: unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect. Advanced drug delivery reviews. 2011 Mar 18;63(3):136-51.
  52. Upponi JR, Torchilin VP. Passive vs. active targeting: an update of the epr role in drug delivery to tumors. InNano-Oncologicals: New Targeting and Delivery Approaches 2014 Sep 6 (pp. 3-45). Cham: Springer International Publishing.
  53. Allen TM, Cullis PR. Liposomal drug delivery systems: from concept to clinical applications. Advanced drug delivery reviews. 2013 Jan 1;65(1):36-48.
  54. Yan J, Wang Y, Jia Y, Liu S, Tian C, Pan W, Liu X, Wang H. Co-delivery of docetaxel and curcumin prodrug via dual-targeted nanoparticles with synergistic antitumor activity against prostate cancer. Biomedicine & Pharmacotherapy. 2017 Apr 1;88:374-83.
  55. Barenholz Y. Liposome application: problems and prospects. Current opinion in colloid & interface science. 2001 Feb 1;6(1):66-77.
  56. Danhier F, Ansorena E, Silva JM, Coco R, Le Breton A, Préat V. PLGA-based nanoparticles: an overview of biomedical applications. Journal of controlled release. 2012 Jul 20;161(2):505-22.
  57. HUANG HC, RAMOS J, GRANDHI TS, POTTA T, REGE K. Gold nanoparticles in cancer imaging and therapeutics. Nano Life. 2010;1(03n04):289-307.
  58. Karimi M, Ghasemi A, Zangabad PS, Rahighi R, Basri SM, Mirshekari H, Amiri M, Pishabad ZS, Aslani A, Bozorgomid M, Ghosh D. Smart micro/nanoparticles in stimulus-responsive drug/gene delivery systems. Chemical Society Reviews. 2016;45(5):1457-501.
  59. Khawar IA, Kim JH, Kuh HJ. Improving drug delivery to solid tumors: priming the tumor microenvironment. Journal of Controlled Release. 2015 Mar 10;201:78-89.
  60. Jain RK. Delivery of molecular medicine to solid tumors. Science. 1996 Feb 23;271(5252):1079-80.
  61. Ferrari M. Cancer nanotechnology: opportunities and challenges. Nature reviews cancer. 2005 Mar 1;5(3):161-71.
  62. Liang C, Xu L, Song G, Liu Z. Emerging nanomedicine approaches fighting tumor metastasis: animal models, metastasis-targeted drug delivery, phototherapy, and immunotherapy. Chemical Society Reviews. 2016;45(22):6250-69.
  63. Hong L, Li W, Li Y, Yin S. Nanoparticle-based drug delivery systems targeting cancer cell surfaces. RSC advances. 2023;13(31):21365-82.
  64. Menilli L, Milani C, Reddi E, Moret F. Overview of nanoparticle-based approaches for the combination of Photodynamic Therapy (PDT) and chemotherapy at the preclinical stage. Cancers. 2022 Sep 14;14(18):4462.
  65. Yu C, Li L, Hu P, Yang Y, Wei W, Deng X, Wang L, Tay FR, Ma J. Recent advances in stimulus?responsive nanocarriers for gene therapy. Advanced Science. 2021 Jul;8(14):2100540.
  66. Allen TM, Cullis PR. Liposomal drug delivery systems: from concept to clinical applications. Advanced drug delivery reviews. 2013 Jan 1;65(1):36-48.
  67. Jain RK. Vascular and interstitial barriers to delivery of therapeutic agents in tumors. Cancer and Metastasis Reviews. 1990 Nov;9:253-66.
  68. M Rabanel J, Aoun V, Elkin I, Mokhtar M, Hildgen P. Drug-loaded nanocarriers: passive targeting and crossing of biological barriers. Current medicinal chemistry. 2012 Jul 1;19(19):3070-102.
  69. Danhier F, Ansorena E, Silva JM, Coco R, Le Breton A, Préat V. PLGA-based nanoparticles: an overview of biomedical applications. Journal of controlled release. 2012 Jul 20;161(2):505-22.
  70. Fotooh Abadi L, Damiri F, Zehravi M, Joshi R, Pai R, Berrada M, Massoud EE, Rahman MH, Rojekar S, Cavalu S. Novel nanotechnology-based approaches for targeting HIV reservoirs. Polymers. 2022 Jul 29;14(15):3090.
  71. Tavakolidakhrabadi N, Ding WY, Saleem MA, Welsh GI, May C. Gene Therapy and kidney diseases. Molecular Therapy Methods & Clinical Development. 2024 Sep 6.
  72. Vahab SA, KI A, Kumar VS. Exploring chitosan nanoparticles for enhanced therapy in neurological disorders: a comprehensive review. Naunyn-Schmiedeberg's Archives of Pharmacology. 2024 Oct 8:1-7.
  73. Khatua R, Bhar B, Dey S, Jaiswal C, Victoria J, Mandal BB. Advances in engineered nanosystems: immunomodulatory interactions for therapeutic applications. Nanoscale. 2024.
  74. Barchi Jr JJ. Glycoconjugate Nanoparticle-Based Systems in Cancer Immunotherapy: Novel Designs and Recent Updates. Frontiers in Immunology. 2022 Mar 30;13:852147
  75. Allen TM, Cullis PR. Liposomal drug delivery systems: from concept to clinical applications. Advanced drug delivery reviews. 2013 Jan 1;65(1):36-48.
  76. Yang W, Wang M, Ma L, Li H, Huang L. Synthesis and characterization of biotin modified cholesteryl pullulan as a novel anticancer drug carrier. Carbohydrate polymers. 2014 Jan 2;99:720-7.
  77. Li Z, Tan S, Li S, Shen Q, Wang K. Cancer drug delivery in the nano era: An overview and perspectives. Oncology reports. 2017 Aug 1;38(2):611-24.
  78. Pattni BS, Torchilin VP. Targeted drug delivery systems: Strategies and challenges. Targeted drug delivery: Concepts and design. 2015:3-8
  79. Bennet D, Kim S. Polymer nanoparticles for smart drug delivery. Application of nanotechnology in drug delivery. 2014 Jul 25;8.
  80. Miller J. Beyond biotechnology: FDA regulation of nanomedicine. Colum. Sci. & Tech. L. Rev.. 2002;4:1.
  81. Hwang SR, Chakraborty K, An JM, Mondal J, Yoon HY, Lee YK. Pharmaceutical aspects of nanocarriers for smart anticancer therapy. Pharmaceutics. 2021 Nov 5;13(11):1875.
  82. Moghimi SM, Hunter AC, Murray JC. Nanomedicine: current status and future prospects. The FASEB journal. 2005 Mar;19(3):311-30.
  83. Henn JG, Aguirre TA, Nugent M, Moura DJ. Cancer nanomedicine: Recent developments in drug delivery systems and strategies to overcome eventual barriers to achieve a better outcome. Journal of Drug Delivery Science and Technology. 2023 Dec 9:105254.
  84. Fang J, Islam W, Maeda H. Exploiting the dynamics of the EPR effect and strategies to improve the therapeutic effects of nanomedicines by using EPR effect enhancers. Advanced drug delivery reviews. 2020 Jan 1;157:142-60.
  85. Moghimi SM, Hunter AC, Murray JC. Nanomedicine: current status and future prospects. The FASEB journal. 2005 Mar;19(3):311-30.
  86. Lee ES, Gao Z, Bae YH. Recent progress in tumor pH targeting nanotechnology. Journal of Controlled Release. 2008 Dec 18;132(3):164-70.
  87. Fu Z, Xiang J. Aptamer-functionalized nanoparticles in targeted delivery and cancer therapy. International Journal of Molecular Sciences. 2020 Nov 30;21(23):9123.
  88. Osouli-Bostanabad K, Puliga S, Serrano DR, Bucchi A, Halbert G, Lalatsa A. Microfluidic manufacture of lipid-based nanomedicines. Pharmaceutics. 2022 Sep 14;14(9):1940.
  89. Ferrari M. Cancer nanotechnology: opportunities and challenges. Nature reviews cancer. 2005 Mar 1;5(3):161-71.
  90. Jain RK. Delivery of molecular medicine to solid tumors. Science. 1996 Feb 23;271(5252):1079-80.
  91. Allen TM, Cullis PR. Liposomal drug delivery systems: from concept to clinical applications. Advanced drug delivery reviews. 2013 Jan 1;65(1):36-48.
  92. Bawa R. FDA and nanotech: baby steps lead to regulatory uncertainty. Bio?Nanotechnology: A Revolution in Food, Biomedical and Health Sciences. 2013 Jan 18:720-32.
  93. Eloy JO, Petrilli R, Trevizan LN, Chorilli M. Immunoliposomes: A review on functionalization strategies and targets for drug delivery. Colloids and Surfaces B: Biointerfaces. 2017 Nov 1;159:454-67

Reference

  1. Brown KJ, Downs CT. Seasonal patterns in body temperature of free-living rock hyrax (Procavia capensis). Comparative Biochemistry and Physiology Part A: Molecular & Integrative Physiology. 2006 Jan 1;143(1):42-9.
  2. Chun AL. Nanomaterials: Mercury mop. Nature Nanotechnology. 2008 Jul 11.
  3. Han XM, Zheng KW, Wang RL, Yue SF, Chen J, Zhao ZW, Song F, Su Y, Ma Q. Functionalization and optimization-strategy of graphene oxide-based nanomaterials for gene and drug delivery. American Journal of Translational Research. 2020;12(5):1515.
  4. Rani R, Malik P, Dhania S, Mukherjee TK. Recent advances in mesoporous silica nanoparticle-mediated drug delivery for breast cancer treatment. Pharmaceutics. 2023 Jan 9;15(1):227.
  5. Hwang SR, Chakraborty K, An JM, Mondal J, Yoon HY, Lee YK. Pharmaceutical aspects of nanocarriers for smart anticancer therapy. Pharmaceutics. 2021 Nov 5;13(11):1875.
  6. Rastegari E, Hsiao YJ, Lai WY, Lai YH, Yang TC, Chen SJ, Huang PI, Chiou SH, Mou CY, Chien Y. An update on mesoporous silica nanoparticle applications in nanomedicine. Pharmaceutics. 2021 Jul 12;13(7):1067.
  7. Bjarnsholt T, Ciofu O, Molin S, Givskov M, Høiby N. Applying insights from biofilm biology to drug development—can a new approach be developed?. Nature reviews Drug discovery. 2013 Oct;12(10):791-808.
  8. Singh R, Lillard Jr JW. Nanoparticle-based targeted drug delivery. Experimental and molecular pathology. 2009 Jun 1;86(3):215-23.
  9. Xing H, Hwang K, Lu Y. Recent developments of liposomes as nanocarriers for theranostic applications. Theranostics. 2016;6(9):1336.
  10. Darban SA, Nikoofal-Sahlabadi S, Amiri N, Kiamanesh N, Mehrabian A, Zendehbad B, Gholizadeh Z, Jaafari MR. Targeting the leptin receptor: To evaluate therapeutic efficacy and anti-tumor effects of Doxil, in vitro and in vivo in mice bearing C26 colon carcinoma tumor. Colloids and Surfaces B: Biointerfaces. 2018 Apr 1;164:107-15.
  11. Zhang L, Gu FX, Chan JM, Wang AZ, Langer RS, Farokhzad OC. Nanoparticles in medicine: therapeutic applications and developments. Clinical pharmacology & therapeutics. 2008 May;83(5):761-9.
  12. Choudhari M, Hejmady S, Saha RN, Damle S, Singhvi G, Alexander A, Kesharwani P, Dubey SK. Evolving new-age strategies to transport therapeutics across the blood-brain-barrier. International Journal of Pharmaceutics. 2021 Apr 15;599:120351.
  13. Kreuter J. Nanoparticulate systems for brain delivery of drugs. Advanced drug delivery reviews. 2001 Mar 23;47(1):65-81.
  14. Bianco A, Kostarelos K, Prato M. Applications of carbon nanotubes in drug delivery. Curr Opin Chem Biol. 2005;9(6):674–9. doi:10.1016/j.cbpa.2005.10.020.
  15. Chun AL. Nanomaterials: Mercury mop. Nature Nanotechnology. 2008 Jul 11.
  16. Han XM, Zheng KW, Wang RL, Yue SF, Chen J, Zhao ZW, Song F, Su Y, Ma Q. Functionalization and optimization-strategy of graphene oxide-based nanomaterials for gene and drug delivery. American Journal of Translational Research. 2020;12(5):1515.
  17. Kumar P, Huo P, Zhang R, Liu B. Antibacterial properties of graphene-based nanomaterials. Nanomaterials. 2019 May 13;9(5):737.
  18. Zhang L, Gu FX, Chan JM, Wang AZ, Langer RS, Farokhzad OC. Nanoparticles in medicine: therapeutic applications and developments. Clinical pharmacology & therapeutics. 2008 May;83(5):761-9.
  19. Prasad RD, Desai CB, Shrivastav OP, Charmode N, Prasad SR, Samant A, Mirajkar R, Banga S, Shaikh VS, Padvi MN, Patil S. A Critical Review on Design and Development of Carbonaceous Materials for Veterinary Medicine. ES Food & Agroforestry. 2022 Sep 8;9:15-38.
  20. Kumar N, Chamoli P, Misra M, Manoj MK, Sharma A. Advanced metal and carbon nanostructures for medical, drug delivery and bio-imaging applications. Nanoscale. 2022;14(11):3987-4017.
  21. D?ugosz O, Szostak K, Staro? A, Pulit-Prociak J, Banach M. Methods for reducing the toxicity of metal and metal oxide NPs as biomedicine. Materials. 2020 Jan 8;13(2):279.
  22. Rane BR, Patil VL, Mhatre NR, Padave AP. Recent advances in mesoporous silica nanoparticles research. Mesoporous Silica Nanoparticles: Drug Delivery, Catalysis and Sensing Applications. 2024 Nov 18:187.
  23. Inam H, Sprio S, Tavoni M, Abbas Z, Pupilli F, Tampieri A. Magnetic hydroxyapatite nanoparticles in regenerative medicine and nanomedicine. International Journal of Molecular Sciences. 2024 Feb 28;25(5):2809.
  24. Rani R, Malik P, Dhania S, Mukherjee TK. Recent advances in mesoporous silica nanoparticle-mediated drug delivery for breast cancer treatment. Pharmaceutics. 2023 Jan 9;15(1):227.
  25. Kim K, Bou-Ghannam S, Kameishi S, Oka M, Grainger DW, Okano T. Allogeneic mesenchymal stem cell sheet therapy: A new frontier in drug delivery systems. Journal of Controlled Release. 2021 Feb 10;330:696-704.
  26. Zhang L, Gu FX, Chan JM, Wang AZ, Langer RS, Farokhzad OC. Nanoparticles in medicine: therapeutic applications and developments. Clinical pharmacology & therapeutics. 2008 May;83(5):761-9.
  27. Li Z, Zhang Y, Feng N. Mesoporous silica nanoparticles: synthesis, classification, drug loading, pharmacokinetics, biocompatibility, and application in drug delivery. Expert opinion on drug delivery. 2019 Mar 4;16(3):219-37.
  28. Nam L, Coll C, Erthal LC, De la Torre C, Serrano D, Martínez-Máñez R, Santos-Martínez MJ, Ruiz-Hernández E. Drug delivery nanosystems for the localized treatment of glioblastoma multiforme. Materials. 2018 May 11;11(5):779.
  29. Varma LT, Singh N, Gorain B, Choudhury H, Tambuwala MM, Kesharwani P, Shukla R. Recent advances in self-assembled nanoparticles for drug delivery. Current Drug Delivery. 2020 May 1;17(4):279-91.
  30. Li R, Peng F, Cai J, Yang D, Zhang P. Redox dual-stimuli responsive drug delivery systems for improving tumor-targeting ability and reducing adverse side effects. Asian journal of pharmaceutical sciences. 2020 May 1;15(3):311-25.
  31. Muheem A, Baboota S, Ali J. An in-depth analysis of novel combinatorial drug therapy via nanocarriers against HIV/AIDS infection and their clinical perspectives: a systematic review. Expert Opinion on Drug Delivery. 2021 Aug 3;18(8):1025-46.
  32. Cepraga C. Two-photon chromophore-polymer conjugates grafted onto gold nanoparticles as fluorescent probes for bioimaging and photodynamic therapy applications (Doctoral dissertation, INSA de Lyon).
  33. Trombino S, Cassano R. Special Issue on designing hydrogels for controlled drug delivery: Guest Editors’ Introduction. Pharmaceutics. 2020 Jan 10;12(1):57.
  34. Gupta AK, Gupta M. Synthesis and surface engineering of iron oxide nanoparticles for biomedical applications. biomaterials. 2005 Jun 1;26(18):3995-4021.
  35. Shen Z, Wu A, Chen X. Iron oxide nanoparticle based contrast agents for magnetic resonance imaging. Molecular pharmaceutics. 2017 May 1;14(5):1352-64.
  36. Campos EA, Pinto DV, Oliveira JI, Mattos ED, Dutra RD. Synthesis, characterization and applications of iron oxide nanoparticles-a short review. Journal of Aerospace Technology and Management. 2015;7(3):267-76.
  37. Javed Y, Hussain MI, Yaseen M, Asif M. Gold–Iron Oxide Nanohybrids: Characterization and Biomedical Applications. Hybrid Nanocomposites. 2019 Mar 11:285-332.
  38. Spernath A, Aserin A, Ziserman L, Danino D, Garti N. Phosphatidylcholine embedded microemulsions: Physical properties and improved Caco-2 cell permeability. Journal of controlled release. 2007 Jun 22;119(3):279-90.
  39. Gupta AK, Gupta M. Synthesis and surface engineering of iron oxide nanoparticles for biomedical applications. biomaterials. 2005 Jun 1;26(18):3995-4021.
  40. Laurent S, Forge D, Port M, Roch A, Robic C, Vander Elst L, Muller RN. Magnetic iron oxide nanoparticles: synthesis, stabilization, vectorization, physicochemical characterizations, and biological applications. Chemical reviews. 2008 Jun 11;108(6):2064-110.
  41. Gaspar MM, Calado S, Pereira J, Ferronha H, Correia I, Castro H, Tomás AM, Cruz ME. Targeted delivery of paromomycin in murine infectious diseases through association to nano lipid systems. Nanomedicine: Nanotechnology, Biology and Medicine. 2015 Oct 1;11(7):1851-60.
  42. Debnath S, Deb K, Saha B, Das R. X-ray diffraction analysis for the determination of elastic properties of zinc-doped manganese spinel ferrite nanocrystals (Mn0. 75Zn0. 25Fe2O4), along with the determination of ionic radii, bond lengths, and hopping lengths. Journal of Physics and Chemistry of Solids. 2019 Nov 1;134:105-14.
  43. Spernath A, Aserin A, Ziserman L, Danino D, Garti N. Phosphatidylcholine embedded microemulsions: Physical properties and improved Caco-2 cell permeability. Journal of controlled release. 2007 Jun 22;119(3):279-90.
  44. Arruebo M, Fernández-Pacheco R, Ibarra MR, Santamaría J. Magnetic nanoparticles for drug delivery. Nano today. 2007 Jun 1;2(3):22-32.
  45. Selvarajan V, Obuobi S, Ee PL. Silica nanoparticles—a versatile tool for the treatment of bacterial infections. Frontiers in Chemistry. 2020 Jul 15;8:602.
  46. Wust P, Hildebrandt B, Sreenivasa G, Rau B, Gellermann J, Riess H, Felix R, Schlag PM. Hyperthermia in combined treatment of cancer. The lancet oncology. 2002 Aug 1;3(8):487-97.
  47. Dang Y, Guan J. Nanoparticle-based drug delivery systems for cancer therapy. Smart Materials in Medicine. 2020 Jan 1;1:10-9.
  48. Vaiserman A, Koliada A, Zayachkivska A, Lushchak O. Nanodelivery of natural antioxidants: an anti-aging perspective. Frontiers in bioengineering and biotechnology. 2020 Jan 10;7:447.
  49. Maeda H. The 35th anniversary of the discovery of EPR effect: a new wave of nanomedicines for tumor-targeted drug delivery—personal remarks and future prospects. Journal of personalized medicine. 2021 Mar 22;11(3):229.
  50. Jain RK. Vascular and interstitial barriers to delivery of therapeutic agents in tumors. Cancer and Metastasis Reviews. 1990 Nov;9:253-66.
  51. Fang J, Nakamura H, Maeda H. The EPR effect: unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect. Advanced drug delivery reviews. 2011 Mar 18;63(3):136-51.
  52. Upponi JR, Torchilin VP. Passive vs. active targeting: an update of the epr role in drug delivery to tumors. InNano-Oncologicals: New Targeting and Delivery Approaches 2014 Sep 6 (pp. 3-45). Cham: Springer International Publishing.
  53. Allen TM, Cullis PR. Liposomal drug delivery systems: from concept to clinical applications. Advanced drug delivery reviews. 2013 Jan 1;65(1):36-48.
  54. Yan J, Wang Y, Jia Y, Liu S, Tian C, Pan W, Liu X, Wang H. Co-delivery of docetaxel and curcumin prodrug via dual-targeted nanoparticles with synergistic antitumor activity against prostate cancer. Biomedicine & Pharmacotherapy. 2017 Apr 1;88:374-83.
  55. Barenholz Y. Liposome application: problems and prospects. Current opinion in colloid & interface science. 2001 Feb 1;6(1):66-77.
  56. Danhier F, Ansorena E, Silva JM, Coco R, Le Breton A, Préat V. PLGA-based nanoparticles: an overview of biomedical applications. Journal of controlled release. 2012 Jul 20;161(2):505-22.
  57. HUANG HC, RAMOS J, GRANDHI TS, POTTA T, REGE K. Gold nanoparticles in cancer imaging and therapeutics. Nano Life. 2010;1(03n04):289-307.
  58. Karimi M, Ghasemi A, Zangabad PS, Rahighi R, Basri SM, Mirshekari H, Amiri M, Pishabad ZS, Aslani A, Bozorgomid M, Ghosh D. Smart micro/nanoparticles in stimulus-responsive drug/gene delivery systems. Chemical Society Reviews. 2016;45(5):1457-501.
  59. Khawar IA, Kim JH, Kuh HJ. Improving drug delivery to solid tumors: priming the tumor microenvironment. Journal of Controlled Release. 2015 Mar 10;201:78-89.
  60. Jain RK. Delivery of molecular medicine to solid tumors. Science. 1996 Feb 23;271(5252):1079-80.
  61. Ferrari M. Cancer nanotechnology: opportunities and challenges. Nature reviews cancer. 2005 Mar 1;5(3):161-71.
  62. Liang C, Xu L, Song G, Liu Z. Emerging nanomedicine approaches fighting tumor metastasis: animal models, metastasis-targeted drug delivery, phototherapy, and immunotherapy. Chemical Society Reviews. 2016;45(22):6250-69.
  63. Hong L, Li W, Li Y, Yin S. Nanoparticle-based drug delivery systems targeting cancer cell surfaces. RSC advances. 2023;13(31):21365-82.
  64. Menilli L, Milani C, Reddi E, Moret F. Overview of nanoparticle-based approaches for the combination of Photodynamic Therapy (PDT) and chemotherapy at the preclinical stage. Cancers. 2022 Sep 14;14(18):4462.
  65. Yu C, Li L, Hu P, Yang Y, Wei W, Deng X, Wang L, Tay FR, Ma J. Recent advances in stimulus?responsive nanocarriers for gene therapy. Advanced Science. 2021 Jul;8(14):2100540.
  66. Allen TM, Cullis PR. Liposomal drug delivery systems: from concept to clinical applications. Advanced drug delivery reviews. 2013 Jan 1;65(1):36-48.
  67. Jain RK. Vascular and interstitial barriers to delivery of therapeutic agents in tumors. Cancer and Metastasis Reviews. 1990 Nov;9:253-66.
  68. M Rabanel J, Aoun V, Elkin I, Mokhtar M, Hildgen P. Drug-loaded nanocarriers: passive targeting and crossing of biological barriers. Current medicinal chemistry. 2012 Jul 1;19(19):3070-102.
  69. Danhier F, Ansorena E, Silva JM, Coco R, Le Breton A, Préat V. PLGA-based nanoparticles: an overview of biomedical applications. Journal of controlled release. 2012 Jul 20;161(2):505-22.
  70. Fotooh Abadi L, Damiri F, Zehravi M, Joshi R, Pai R, Berrada M, Massoud EE, Rahman MH, Rojekar S, Cavalu S. Novel nanotechnology-based approaches for targeting HIV reservoirs. Polymers. 2022 Jul 29;14(15):3090.
  71. Tavakolidakhrabadi N, Ding WY, Saleem MA, Welsh GI, May C. Gene Therapy and kidney diseases. Molecular Therapy Methods & Clinical Development. 2024 Sep 6.
  72. Vahab SA, KI A, Kumar VS. Exploring chitosan nanoparticles for enhanced therapy in neurological disorders: a comprehensive review. Naunyn-Schmiedeberg's Archives of Pharmacology. 2024 Oct 8:1-7.
  73. Khatua R, Bhar B, Dey S, Jaiswal C, Victoria J, Mandal BB. Advances in engineered nanosystems: immunomodulatory interactions for therapeutic applications. Nanoscale. 2024.
  74. Barchi Jr JJ. Glycoconjugate Nanoparticle-Based Systems in Cancer Immunotherapy: Novel Designs and Recent Updates. Frontiers in Immunology. 2022 Mar 30;13:852147
  75. Allen TM, Cullis PR. Liposomal drug delivery systems: from concept to clinical applications. Advanced drug delivery reviews. 2013 Jan 1;65(1):36-48.
  76. Yang W, Wang M, Ma L, Li H, Huang L. Synthesis and characterization of biotin modified cholesteryl pullulan as a novel anticancer drug carrier. Carbohydrate polymers. 2014 Jan 2;99:720-7.
  77. Li Z, Tan S, Li S, Shen Q, Wang K. Cancer drug delivery in the nano era: An overview and perspectives. Oncology reports. 2017 Aug 1;38(2):611-24.
  78. Pattni BS, Torchilin VP. Targeted drug delivery systems: Strategies and challenges. Targeted drug delivery: Concepts and design. 2015:3-8
  79. Bennet D, Kim S. Polymer nanoparticles for smart drug delivery. Application of nanotechnology in drug delivery. 2014 Jul 25;8.
  80. Miller J. Beyond biotechnology: FDA regulation of nanomedicine. Colum. Sci. & Tech. L. Rev.. 2002;4:1.
  81. Hwang SR, Chakraborty K, An JM, Mondal J, Yoon HY, Lee YK. Pharmaceutical aspects of nanocarriers for smart anticancer therapy. Pharmaceutics. 2021 Nov 5;13(11):1875.
  82. Moghimi SM, Hunter AC, Murray JC. Nanomedicine: current status and future prospects. The FASEB journal. 2005 Mar;19(3):311-30.
  83. Henn JG, Aguirre TA, Nugent M, Moura DJ. Cancer nanomedicine: Recent developments in drug delivery systems and strategies to overcome eventual barriers to achieve a better outcome. Journal of Drug Delivery Science and Technology. 2023 Dec 9:105254.
  84. Fang J, Islam W, Maeda H. Exploiting the dynamics of the EPR effect and strategies to improve the therapeutic effects of nanomedicines by using EPR effect enhancers. Advanced drug delivery reviews. 2020 Jan 1;157:142-60.
  85. Moghimi SM, Hunter AC, Murray JC. Nanomedicine: current status and future prospects. The FASEB journal. 2005 Mar;19(3):311-30.
  86. Lee ES, Gao Z, Bae YH. Recent progress in tumor pH targeting nanotechnology. Journal of Controlled Release. 2008 Dec 18;132(3):164-70.
  87. Fu Z, Xiang J. Aptamer-functionalized nanoparticles in targeted delivery and cancer therapy. International Journal of Molecular Sciences. 2020 Nov 30;21(23):9123.
  88. Osouli-Bostanabad K, Puliga S, Serrano DR, Bucchi A, Halbert G, Lalatsa A. Microfluidic manufacture of lipid-based nanomedicines. Pharmaceutics. 2022 Sep 14;14(9):1940.
  89. Ferrari M. Cancer nanotechnology: opportunities and challenges. Nature reviews cancer. 2005 Mar 1;5(3):161-71.
  90. Jain RK. Delivery of molecular medicine to solid tumors. Science. 1996 Feb 23;271(5252):1079-80.
  91. Allen TM, Cullis PR. Liposomal drug delivery systems: from concept to clinical applications. Advanced drug delivery reviews. 2013 Jan 1;65(1):36-48.
  92. Bawa R. FDA and nanotech: baby steps lead to regulatory uncertainty. Bio?Nanotechnology: A Revolution in Food, Biomedical and Health Sciences. 2013 Jan 18:720-32.
  93. Eloy JO, Petrilli R, Trevizan LN, Chorilli M. Immunoliposomes: A review on functionalization strategies and targets for drug delivery. Colloids and Surfaces B: Biointerfaces. 2017 Nov 1;159:454-67

Photo
Aditya Suryawanshi
Corresponding author

Department of Pharmacy Practice, SGMSPM’s Sharadchandra Pawar College of Pharmacy, Dumbarwadi (Otur), Tal- Junnar, Dist.- Pune, Maharashtra, India, 410504.

Photo
Sujata Shendage
Co-author

Department of Pharmacy Practice, SGMSPM’s Sharadchandra Pawar College of Pharmacy, Dumbarwadi (Otur), Tal- Junnar, Dist.- Pune, Maharashtra, India, 410504.

Photo
Sanket Kadhane
Co-author

Department of Pharmacy Practice, SGMSPM’s Sharadchandra Pawar College of Pharmacy, Dumbarwadi (Otur), Tal- Junnar, Dist.- Pune, Maharashtra, India, 410504.

Photo
Shreeyash Pathare
Co-author

Department of Pharmacy Practice, SGMSPM’s Sharadchandra Pawar College of Pharmacy, Dumbarwadi (Otur), Tal- Junnar, Dist.- Pune, Maharashtra, India, 410504.

Aditya Suryawanshi*, Sujata Shendage, Sanket Kadhane, Shreeyash Pathare Advancements in Targeted Drug Delivery Systems: Nanotechnology in Pharmacy, Int. J. of Pharm. Sci., 2024, Vol 2, Issue 11, 1233-1249. https://doi.org/10.5281/zenodo.14214265

More related articles
Innovative Horizons In Pain Relief: A Review On Th...
Rahila, Shafnaz Abdul Rahman, Rubayyath.K, Digi Davis.C, Neeshma....
Revitalizing Pharmacy Education: Leveraging Freshe...
S. G. Santhoshkumar, M. Jeyarahavi, S. Jerlin Epsiba, M. Manoj Gu...
Panoptic Review On Psoriasis:Treatments, Types, An...
Pradnya Sandip Shinde , Prof.Pallavi R Kaple, Dr Prof.Sonia Singh...
Melatonin Loaded Nanocarriers...
Raslamol K., Saranya K. G., Amayasree Suresh, Archa Ani, A. S. Athira, Febiya Joy, Gayathri Prasad, ...
Phytochemical Analysis and Potential Anthelmintic activity of Chenopodium ambros...
Manodeep Chakraborty, Rahul Pradhan , Praghya sharma , Nikki kumari Gupta, Samir Chouhan, Nowsad SK,...
Polymers Used In Floating Drugs Delivery System-A Review ...
Ram prasad khoteja, Aarti Kori , Shivanand Patil , ...
Related Articles
Molecular Docking And ADMET Predictions Of Substituted Cordyheptapeptide Derivat...
Shivanand Kolageri, Kavya Pujari, Laxmi Math, Ambika Chavan, Pavitra Patil, Pallavi Bhairagond, Vidy...
Development And Validation Of UV Spectroscopy Method For Simultaneous Estimation...
Shilpa Rathwa, Chainesh N Shah, Badawal Rajnandani, Pooja J. Vyas, Bhoomika Malete, Umesh Upadhyay, ...
Review On Hydrogels: Advances, Challenges, And Applications in Biomedicine...
Vaishanavi Vane, Adinath Sangle, Dr. Megha Salve, ...
Innovative Horizons In Pain Relief: A Review On The Promise Of NSAID Prodrugs...
Rahila, Shafnaz Abdul Rahman, Rubayyath.K, Digi Davis.C, Neeshma.K, Ramsiya.K, Razana Binth Yoosuf P...
More related articles
Innovative Horizons In Pain Relief: A Review On The Promise Of NSAID Prodrugs...
Rahila, Shafnaz Abdul Rahman, Rubayyath.K, Digi Davis.C, Neeshma.K, Ramsiya.K, Razana Binth Yoosuf P...
Revitalizing Pharmacy Education: Leveraging Freshers' Insights To Level Up Pedag...
S. G. Santhoshkumar, M. Jeyarahavi, S. Jerlin Epsiba, M. Manoj Gupta, Niranjani, T. Vishnu, ...
Panoptic Review On Psoriasis:Treatments, Types, And In-Depth Insights...
Pradnya Sandip Shinde , Prof.Pallavi R Kaple, Dr Prof.Sonia Singh, Dr.Nikhil Sutar, Manuja S Zade, P...
Innovative Horizons In Pain Relief: A Review On The Promise Of NSAID Prodrugs...
Rahila, Shafnaz Abdul Rahman, Rubayyath.K, Digi Davis.C, Neeshma.K, Ramsiya.K, Razana Binth Yoosuf P...
Revitalizing Pharmacy Education: Leveraging Freshers' Insights To Level Up Pedag...
S. G. Santhoshkumar, M. Jeyarahavi, S. Jerlin Epsiba, M. Manoj Gupta, Niranjani, T. Vishnu, ...
Panoptic Review On Psoriasis:Treatments, Types, And In-Depth Insights...
Pradnya Sandip Shinde , Prof.Pallavi R Kaple, Dr Prof.Sonia Singh, Dr.Nikhil Sutar, Manuja S Zade, P...