• INTRODUCTION
  • CONCLUSION
  • ACKNOWLEDGEMENT
  • FUNDING
  • AUTHORS CONTRIBUTIONS
  • CONFLICTS OF INTERESTS
  • REFERENCES
  • Int J App Pharm, Vol 16, Issue 6, 2024, 1-9Review Article

    PHARMACEUTICAL NANOCRYSTALS: AN EXTENSIVE OVERVIEW

    GURUBARAN SIVANATHAN1, SANJAI RAJAGOPAL1, GIRIDHARA MAHADEVASWAMY1, GOWTHAM ANGAMUTHU2, NAGASAMY VENKATESH DHANDAPANI1*

    1Department of Pharmaceutics, JSS College of Pharmacy, Ooty, Nilgiris, Tamil Nadu, India. 2Department of Pharmaceutical Regulatory Affairs, JSS College of Pharmacy, Ooty, Nilgiris, Tamil Nadu, India
    *Corresponding author: Nagasamy Venkatesh Dhandapani; *Email: nagasamyvenkatesh@jssuni.edu.in

    Received: 05 Aug 2024, Revised and Accepted: 17 Sep 2024


    ABSTRACT

    In pharmaceutical development, pharmaceutical nanocrystals sized between 10 and 1000 nanometers have been found to hold promise in improving drug solubility. Since they comprise only the active pharmaceutical ingredient, nanocrystals have dramatically increased surface area-to-volume ratios, ensuring improved in vitro dissolution and solubility profiles.

    In view of their strengths and limitations, different production strategies have been reviewed: methods of size reduction such as wet milling and high-pressure homogenization; the bottom-up approaches of controlled precipitation and supercritical fluid technology; and efficient ways to stabilize nanocrystal formulations aided by excipients like surfactants and polymers.

    Techniques used in this characterization of nanocrystals include size analysis, surface-charge measurement, and assessment of crystalline structure. The routes of administration, such as oral, injectable, inhaled, and topical application, are reviewed alongside commercially successful products and clinical trials.

    This work reviews dynamic regulatory scenarios and current challenges of large-scale production, long-term stability, and nanotoxicity evaluation. In addition, it addresses the emerging trends in nanocrystal technology in the field of personalized medicine, targeted drug delivery, and theranostic approaches associated with how nanocrystals can help optimize the outcome of a patient in drug delivery systems.

    Keywords: Pharmaceutical nanocrystals, Drug delivery, Bioavailability enhancement, Nanotoxicity, Regulatory considerations, Stabilization strategies


    INTRODUCTION

    Nanocrystals are a stupendous development in pharmaceutical drug delivery, especially for poorly water-soluble drugs. These are nanosized particles, normally between 10 and 1000 nm in size, with the most common size being 100-300 nm, made purely of drug substances [1]. The concept came into being in the 1990s to solve poor bioavailability, which had become a significant issue for promising drug candidates [2]. This was very important because it is known that about 40% of approved drugs and 90% of drugs under development suffer from low water solubility [3].

    Nanocrystals' efficiency is based on the fundamental laws of physics. When the size of particles decreases, the surface area-to-volume ratio increases, and, therefore, according to the Noyes-Whitney equation [4], dissolution rates increase. Increased solubility of sub-micrometer particles was explained by the Ostwald-Freundlich equation [5].

    While other nanoparticle formulations contain excipients, nanocrystals contain the active drug substance only. This is a benefit that gives them an advantage over all other drug delivery forms in terms of the highly concentrated amount of a drug to be administered in small volumes, offering one main advantage for high-dose medicinal products [6].

    The methods in nanocrystal production mainly rely on two approaches: one is top-down techniques, in which drug particles of larger size are brought down to the nanosize, and the other is the bottom-up technique, where the formation is from dissolved drug molecules in the form of nanoparticles. Again, each method has its own advantages and limitations [7].

    Three nanocrystal drugs have now reached the market and include the following: Rapamune (sirolimus), Emend (aprepitant), and Tricor (fenofibrate), clearly signifying the practical feasibility of the nanocrystal formulations in the clinical [8].

    Besides dissolution and absorption, nanocrystals show huge potential for both targeted drug delivery and controlled release, new administration routes like transdermal and pulmonary delivery, with enhancing developments increasing their scope of technology in many treatments [9]. With improved research in the field, the role of nanocrystals in drug delivery is expected to be even more significant in the future, especially with poorly soluble drugs.

    Such progress may enable more effective treatments and new indications of old drugs.

    These unique properties, in conjunction with their versatility in drug delivery and administration routes, make nanocrystals the powerful tool needed to combat the challenges from poorly water-soluble drugs. Thus, bioavailability enhancement can go hand in hand with targeted delivery, enabling new approaches for the formulation of drugs and for treatment strategies.

    In a developing field, attention is also focused on the optimization of the production methods, improvement in stability, and extension in the spectrum of applications. Commercial successes with nanocrystal-based products have prepared the ground for the development, implementation, and widespread use of this technology in pharmaceutical research and clinical application.

    Most of the nanocrystal technological developments are definitely going to play a vital role in drug delivery systems in the near future and would definitely change the face of treatments for many diseases and prove beneficial for patients.

    The selections of articles for the present review were searched from specialized databases (Range of years: 2014-2024) such as Elsevier, Pubmed, and Cambridge using the keywords Nanocrystals, Nanotoxicity, and Regulatory consideration. Other selections include articles from Springer Wiley, information from Internet sources, and online published articles from The Lancet Respiratory Medicine, Medscape, and Statpearls.

    Key features and benefits of drug nanocrystals in medicine

    Nanocrystals of drugs offer several advantages during pharmaceutical development, particularly for those drugs with poor water solubility. The biggest benefits are based on increased dissolution rates and enhanced solubility. Since the surface area-to-volume ratio is dramatically increased by rendering drug particles to nanosized levels, this results in increased dissolution rates, explained by the Noyes-Whitney equation. Thus, this will improve the absorption rate and pharmacokinetics. For particles less than 1 μ, this is explained using the Ostwald-Freundlich equation: saturation solubility rises very drastically for nanoparticles below 100 nm [5].

    Such properties thus contribute to much-improved bioavailability, which would be especially useful for drugs in the BCS Class II and Class IV. The bioavailability may be enhanced several-fold over conventional formulations, hence improving therapeutic efficacy while allowing a dose reduction [6, 7].

    Nanocrystal technology decreases the variability in absorption due to food intake and lowers the fed/fasted effect, providing more consistent drug exposure [8]. Nanocrystals in the size range of 50-300 nm offer passive targeting potential in tumors, especially through the so-called enhanced permeability and retention effect, opening perspectives for more effective cancer chemotherapy.

    Such versatility makes nanocrystals administrable by other parenteral, pulmonary, and dermal administration routes other than oral. They have high drug loading capacity with minimal content of stabilizing agents-advantageous for high-dose medications. The physical and chemical stability of nanocrystal preparation is usually good because of its crystalline nature.

    Controlled-release formulations with modulated nanocrystal properties, combined with suitable excipients, may lead to reduced dosing frequency and improved patient compliance. Production costs are often lower, especially for top-down techniques like wet milling or high-pressure homogenization [7-13].

    It has been revealed that nanocrystal preparations provide better content homogeneity, which is particularly useful for low-dose products [14]. Although certain benefits might differ with the drug, preparation method, and application, such characteristics have established nanocrystals as a very promising approach for the development of poorly soluble drugs to achieve enhanced efficacy and improvement in the outcomes of treated patients.

    Production methods for pharmaceutical nanocrystals

    Pharmaceutical techniques for nanocrystal manufacturing can be broadly classified into two classes, each with rather different methodologies, benefits, and shortcomings: top-down and bottom-up.

    Top-down approaches mostly go for the reduction of larger drug particles in size and thus are more attractive to the pharmaceutical industry. The most widely used technique includes media milling (wet milling), which, in turn, includes dispersing the drug in a liquid medium with stabilizers and treating it with high-energy milling using particular beads [7]. This represents a large-scale manufacturing method that produces particles below 100 nm and can be used for a variety of drug substances. There is; however, some potential risk for contamination from milling materials, and some probable amorphization of the drug may occur. Alkermes' NanoCrystal ® technology uses this process [15].

    High-pressure homogenization is a process that forces a suspension under high pressure through a narrow gap [11]. This technique allows for both aqueous and nonaqueous media, amenable for mass production, with less contamination risk than media milling. On the other hand, it could require cycles to be repeated and tends to heat up during processing. The DissoCubes ® technology utilizes this process [2].

    Bottom-up approaches form nanocrystals from dissolved drug molecules. In such approaches, finer and more uniform particles can be produced, but there are scaling difficulties. Precipitation methods imply the precipitation of a drug from a supersaturated solution [16]; they present lower energy demands but are associated with challenging particle size control. Variants include sonoprecipitation and evaporative precipitation [17, 18].

    Supercritical fluid methods are primarily based on CO2 as a solvent or antisolvent [19]. They are environment-friendly but require drug solubility in supercritical CO2; at the same time, they have high equipment costs. The microemulsion method is based on templates of nanocrystals made by microemulsions [20]; in this way, small and uniform particles are prepared with low energy input, while the drug loading capacity is limited.

    Table 1: Characterization techniques for pharmaceutical nanocrystals

    Technique Principle Benefits Drawbacks Reference
    Particle size analysis: Dynamic Light Scattering (DLS) Measures Brownian motion of particles Fast, Non-destructive, and Suitable for suspensions Assumes spherical particles and Sensitive to large particles [21]
    Laser Diffraction Measures scattered light intensity at different angles Wide particle size range Assumes spherical particles [22]
    Nanoparticle Tracking Analysis (NTA) Tracks the motion of individual particles in real-time Provides concentration data, and can identify aggregates Low concentration at the bottom end, and Sample dilution required [23]

    Zeta potential measurements:

    Electrophoretic Light Scattering

    Measures particle velocity in an electric field Reflects suspension stability/surface charge Temperature sensitive, pH-dependent [24]
    Morphology studies: Scanning Electron Microscopy (SEM) Scans surface with focused electron beam High-resolution and 3D-like images Sample preparation may affect morphology and requires a vacuum [25]
    Transmission Electron Microscopy (TEM) The electron beam passes through the ultra-thin sample Very high resolution and reveals internal structure Complex sample preparation, 2D projections [26]
    Atomic Force Microscopy (AFM) Mechanical probe scans surface 3D topography can be measured in liquid Slow scanning, tip artifacts [27]

    Crystallinity analysis:

    X-ray diffraction (XRD)

    Measures X-ray diffraction by crystal lattice Confirms crystalline state and Identifies polymorphs Challenging for very small crystals, difficult amorphous content quantification [28]
    Differential Scanning Calorimetry (DSC) Measures heat flow differences during heating/cooling Detects melting point, glass transition, crystallization, and quantify amorphous content Low sensitivity for small samples, Risk of sample degradation during heating [29]
    Surface area analysis: Brunauer-Emmett-Teller (BET) Method Measures gas adsorption on the particle surface Quantifies specific surface area Requires dry samples [30]

    Dissolution testing:

    In vitro Dissolution Studies

    Quantifies drug release over time in simulated conditions Predicts in vivo performance and quality control Choice of dissolution medium, maintaining sink condition [31]
    Chemical composition and purity: High-Performance Liquid Chromatography (HPLC) Separates and quantifies components

    Quantifies drug content and detects impurities,

    High sensitivity and specificity

    Lack of structural information, Limited size range, and Aggregation risk [32]

    Stability studies:

    Accelerated Stability Testing

    Subject samples to stress conditions Predicts long-term stability and Determines shelf life Choice of conditions and Packaging considerations [33]

    Advanced techniques:

    Small-angle X-ray Scattering (SAXS)

    Analyzes X-ray scattering at very low angles Can measure in suspension and Provides size distribution Complex data interpretation [34]
    Raman Spectroscopy Measures inelastic scattering of monochromatic light Non-destructive Can detect polymorphs and crystallinity Weak signal, Fluorescence interference [35]

    The combination of top-down and bottom-up techniques shall optimize the production of nanocrystals. Techniques such as Smart Crystal [8] will give smaller particles, hence reducing processing time and energy consumption.

    The choice of the production method will, therefore be based on the physicochemical properties of the drug, desired characteristics of the particles, scalability needs, and production economics.

    Components of pharmaceutical nanocrystals

    Nanocrystals in the pharmaceutical field are normally 10-1000 nm in dimension and include a new, promising delivery system of drugs. The major constituents of these formulations are surfactants and polymers acting as stabilizers.

    Surfactants are either ionic, like sodium dodecyl sulphate, or non-ionic, like Tween 80 and Pluronic F68. Ionic surfactants provide electrostatic stabilization but are sensitive to changes in electrolyte and pH [36]. Non-ionic surfactants provide steric stabilization, which is much less affected by environmental factors; however, they may need to be used at higher concentrations [37].

    These polymers contribute steric stabilization and additional functionalities. On their own, the cellulose derivatives Hydroxypropyl Methylcellulose (HPMC) and Hydroxypropylcellulose (HPC) demonstrated good nanoparticle stabilization, with HPMC actually improving dissolution rates [7]. Polyvinylpyrrolidone (PVP) showed some extent of stabilization and even a potential inhibition of crystal growth, but it might have an impact on the stability of the formulations due to its hygroscopic nature [14]. Polyethylene plastics (PEG) bring about biocompatibility and extended in vivo circulation; however, its efficacy is molecular weight-dependent [38].

    These components will thus differently participate in the stability, functionality, and bioavailability of pharmaceutical nanocrystals, rendering it a very effective drug delivery system.

    Stabilization strategies for pharmaceutical nanocrystals

    Effective stabilization is one of the most important requisites of nanocrystal formulations for avoidance of aggregation, maintenance of the desired size, and long-term stability. Stabilization approaches basically can be divided into two major types: combination strategies and new-age techniques.

    Combinations of stabilizers often result in increased stability. Polymer-surfactant combinations can provide both steric and electrostatic stability; careful balancing may be required [39]. Combinations like Sodium Dodecyl Sulphate (SDS) with poloxamer 188, and ionic-nonionic surfactant combinations, offer stability over a wide range but may have the disadvantage of competitive adsorption issues [40].

    The newer age stabilization strategies include the technologies of layer-by-layer coating and lipid coating. However, in layer-by-layer coating, the oppositely charged polyelectrolytes are adsorbed successively, providing added functionality of sustained release, but at the cost of complicating the process and causing retardation in dissolution [41]. Although lipid coating may enhance oral absorption and improve targeting, it is believed that it reduces the dissolution rate [10].

    Several factors should be considered to achieve proper stabilization. For electrostatic stabilization, zeta potential should be more than |30 mV| and should be checked periodically [42]. The surfactant concentration should generally be higher than the Critical micelle concentration (CMC), considering the effects of drug solubility [5]. Ostwald ripening can be inhibited by the slightly soluble stabilizers where the effect of those on drug solubility should be monitored properly [43].

    Processing conditions are important since the high energy applied in some processes can degrade or destroy the stabilizers. Checks should be made on the thermal stability after high-pressure homogenization [2]. Long-time stability studies at different storage conditions should be performed in relation to the role of these stabilizers on the glass transition temperature for the dried formulations [12].

    Finally, regulatory considerations are important, and innovative stabilizers likely will require more safety data. Regulatory status, like generally recognized as safe (GRAS) or inclusion in a pharmacopeia, should be a consideration for the selection of the stabilizer [15].

    The right stabilization strategy would hence be selected based on the physicochemical properties of the drug, manufacturing process, route of administration, and target product profile. Optimization typically requires a combination of approaches to achieve both stability and performance.

    This means that effective stabilization of nanocrystal formulations comes out as the result of a highly complex interplay of aspects and techniques. Different facets of such mastering form the ground for the development of stable, efficient, and regulatory-compliant nanocrystal-based drug delivery systems.

    Applications of nanocrystals in drug delivery

    Such pharmacological nanocrystals bear a high degree of versatility in terms of administration routes and a wide spectrum of therapeutic areas. Their applications in drug delivery are manifold and full of promise:

    Oral delivery is the most frequent application and increases the bioavailability of poorly soluble drugs [6, 109]. Immediate-release formulations, for example, Rapamune ® and Emend ®, provide an increased dissolution rate and bioavailability for immunosuppressants and antiemetics respectively [9]. Modified release formulations are possible by a combination of nanocrystals with polymers or matrix systems and lead to controlled release in, for example, the treatment of chronic diseases [10]. Gastroretentive systems provided by mucoadhesive nanocrystal formulations prolong the residence time in the stomach and increase drug absorption [44].

    Based on this, the parenteral administration can be broadly classified into intravenous and intramuscular [8]. The intravenous administration allows the infusion of smaller volumes and avoids toxic cosolvents present in PaxceedTM for metastatic breast cancer [45]. Intramuscular administration facilitates the formation and sustained release of drug depots, just like Invega SustennaR for antipsychotic treatment [46, 108].

    Pulmonary delivery improves the efficacy of inhaled medications [47]. Dry powder inhalers with nanocrystals embedded in microparticles increase lung deposition and dissolution [48]. Nebulized formulations enhance the local concentration and provide for systemic delivery [49].

    Dermal and transdermal applications enhance skin penetration and allow localized drug action [50]. Topical formulations for the treatment of psoriasis, such as Neoral, provide higher skin penetration [51], whereas transdermal delivery techniques allow increased permeation for systemic delivery [52].

    Oculary delivery increases the bioavailability and residence time [53]. Topical ophthalmic formulations increase the corneal permeation as evidenced by the BromSite post-operative inflammation [54]. Intravitreal delivery maintains the level of drugs within the eye and reduces the frequency of injections [55].

    Targeted drug delivery thus can be based upon surface-engineered nanocrystals [56]. Passive targeting is typically exploited for the Electron paramagnetic resonance (EPR) phenomenon for the management of solid tumors with the aid of Theralux™ by IDEXX [57]. By active targeting, we mean that the addition of targeted elements that are conjugated to nano-crystal surfaces produces enhancements in cellular intake [58].

    Combination products have fixed-dose combinations for better compliance and synergistic effects [59, 60] and theranostics combine diagnosis and treatment [61].

    Nanocrystal delivery of a vaccine can increase the immune response to antigens especially subunit vaccines by giving them enhanced stability and increasing cellular uptake [62, 63].

    It is a fledgling area of development that continuously gives rise to new techniques and technologies for formulation, with flexibility across diversified routes of dosing and therapeutic areas.

    Commercial products and clinical studies of pharmaceutical nanocrystals

    Several drugs have been developed using this approach to the point where they are in commercial use demonstrating proof of concept [9] (table 2). There are several nanocrystal-based formulations currently under clinical evaluation (table 3). Since the field is developing and research is presently in the stage of overcoming traditional issues associated with pharmaceutical nanocrystals, newer applications are being looked upon. This drug delivery technology is likely to remain upbeat due to the success of marketed products and encouraging results of several molecules under clinical trials (table 4).

    Table 2: Marketed nanocrystal-based products

    Product name Generic name Manufacturer Use Technology Benefits Reference
    Rapamune® Sirolimus Pfizer (originally Wyeth) Prevents rejection of transplanted organs Elan's NanoCrystal® Improved uptake (tablet form rather than liquid). [64]
    Emend® Aprepitant Merck Treats chemotherapy-induced nausea Elan's NanoCrystal® More food independent, better absorption. [65]
    Tricor® Fenofibrate AbbVie Reduces blood lipids Elan's NanoCrystal® Less food effect, lower doses required. [6]
    Megace ES® Megestrol acetate Par Pharmaceutical Appetite stimulation in AIDS patients Elan’s NanoCrystal® Smaller volume per dose, increased absorption. [66]
    Invega Sustenna® Paliperidone palmitate Janssen Pharmaceuticals Schizophrenia and related disorders Elan's NanoCrystal® Long-acting injectable form improved the drug's behavior in the organism. [67]
    Ryanodex® Dantrolene sodium Eagle Pharmaceuticals Malignant hyperthermia SkyePharma’s IDD-P® Short preparation time, smaller infusion. [2]

    Table 3: Clinical studies and pipeline products

    Product name Developer Intended use Progress Benefits Reference
    SUBA™-Itraconazole Hedgepath Pharmaceuticals Basal cell carcinoma nevus syndrome Complete Phase 2b trial Improved bioavailability of itraconazole for cancer therapy. [68]
    Theralux™ (thymectacin) Celmed BioSciences Solid tumors Phase 2 trial Better tumor targeting and lesser general toxicity. [57]
    Paxceed™ (paclitaxel) Angiotech Pharmaceuticals (now Argon Medical Devices) Rheumatoid arthritis Phase 3 trials (abandoned) Reduced toxicity and ability to be administered orally. [69]
    NKTR-181 Nektar Therapeutics Chronic axial (lower back) pain Phase 3 completed (not FDA approved) Novel opioids with minimal abuse liability. [70]
    Docetaxel-NC Samyang Biopharmaceuticals Various cancers Phase 2 trials Improved solubility potential for oral dosing. [10]

    Table 4: Ongoing research and future prospects

    Research area Focus Potential uses Benefits Reference
    Combination Products Development of nanocrystals with several drugs HIV therapy, cardiovascular diseases Improved patient compliance, potential synergistic effects [71]. [71]
    Targeted Nanocrystals Surface engineering with nanocrystals for targeted delivery Cancer treatment, inflammatory disorders Enhanced efficacy and reduced side effects [72]. [72]
    Nanocrystals for Non-Oral Routes Nanocrystal development for lung, skin, and eye delivery Respiratory, skin, and eye diseases Local delivery minimized systemic exposure [8]. [8]
    Theranostic Nanocrystals Merging of treatment and diagnostic capabilities Cancer therapy, personalized medicine Simultaneous imaging and drug delivery [61]. [61]

    Regulatory considerations for pharmaceutical nanocrystals

    Along with the increasing technological capability comes the related regulatory landscape associated with nanocrystal-based pharmaceuticals. Global regulators develop guidelines to provide for safety and efficacy in the context of such rapid innovation.

    The U. S. Food and Drug Administration has led in the development of a regulatory framework for products based on nanotechnology [73]. Their definition encompasses products from 1 nm to 100 nm, or up to 1000 nm for size-related properties, thus covering pharmaceutical nanocrystals [75]. Material analysis, production process, physical and chemical characteristics, and biological behavior have been pointed out as key issues for the regulation of such materials [76]. Nanocrystal formulations of existing drugs are processed as the 505(b) (2) pathway, while new chemical entities in nanocrystal form will be processed as a New Drug Application [11].

    Several of the various regulatory problems on nanomedicines, including nanocrystals, have been addressed by the European Medicines Agency [74]. These reflection papers refer to the development of nanomedicines in relation to environmental risk assessment and data requirements for nanoparticle products administered intravenously [77]. The key areas of focus that are targeted are physical and chemical characterization, non-clinical studies, clinical development, and quality with manufacturing [78].

    The International Council for Harmonisation (ICH) develops internationally applicable guidelines that are important for nanocrystal formulations and includes those on impurities, specifications, pharmaceutical development, quality risk management, and pharmaceutical quality systems [12, 79].

    Nanocrystals-specific regulatory considerations include characterization of particle size by using multiple analytic methods [80], adaptation of methods for dissolution testing [81], long-term stability studies [12], total description and validation of manufacturing processes [13], bioequivalence for generic products [82], safety aspects including the toxicological impact at nanoscale [83].

    Global harmonization efforts in regulation are on their way, such as the International Pharmaceutical Regulators Programme Nanomedicines Working Group [84] or the development of International Organization of Standardization (ISO) standards for the characterization of nanoparticles [85].

    Some of the regulatory challenges that lie ahead include pathways for nanocrystal-based combination products [86], new uses for nanocrystals in targeted delivery and theranostics [61], and frameworks to evaluate long-term safety and potential environmental impact [87].

    In the final analysis, pharmaceutical nanocrystals are still subject to regulatory changes. Broadly speaking, manufacturers should initiate early and several reviews with the regulatory agencies regarding compliance, smoothness, and acceleration of approval procedures. With many nanocrystal-based products coming into the market, it is hoped that the regulatory frameworks will become more mature and focused.

    Key focus areas involve dimension, distribution, morphology, and surface properties of the material. The manufacturing process, homogeneity, and scale-up are quite crucial in ensuring quality and consistency. Physical and chemical properties, including solubility and dissolution rate, have a great impact on the bioavailability of the product and its overall effectiveness. Lastly, the biological behavior of the material, such as pharmacokinetics and biodistribution, is helpful in forecasting therapeutic outcomes and profiles of safety.

    This involves validation of manufacturing processes, identification of critical process parameters, and reproducibility in order to meet regulatory requirements. Extended in vivo bioequivalence studies under various conditions, such as food effect studies, are vital for the establishment of stability and performance of the product in case of various eventualities. Long-term stability studies would also be important, as over a period of time, altered distribution and potential nanotoxicity may be observed. Finally, other critical issues include safety concerns regarding cellular uptake and any possible environmental impacts, which need to be analyzed in detail to minimize risks resulting from the use of the product.

    It is, therefore, expected that with new applications and combination products, and in establishing the long-term safety concerns associated with nanocrystal-based pharmaceuticals, the challenges to regulatory agencies will further increase as the field continues to evolve.

    Challenges and Future Perspectives

    Nanocrystal formulations raise a number of challenges in scale-up, long-term stability, nanotoxicity, and personalized medicine applications. Scale-up problems include a decrease in the uniformity of the particles' size [88], a rise in heat generation [89], and significant investment in equipment is needed [9]. Process parameters mostly vary significantly between the laboratory and industrial scales during translation, so substantial adjustment could be required [14]. New continuous manufacturing, innovative milling techniques [72], and quality-by-design approaches will be future perspectives to overcome these challenges.

    Such issues of long-term stability include particle aggregation [12], Ostwald ripening [91], drug degradation [92], and efficiency of different classes of stabilizers [37]. To deal with these problems, improved methods of stabilization [10] are being developed, and surface engineering strategies [1] are already in use, along with predictive models of stability [93] and new developments in freeze-drying techniques [94].

    It has a nanotoxicity potential due to modified drug distribution [95], unique cellular interactions [96], and unknown long-term effects [87]. Further complicating the assessment of toxicity is a lack of standardized testing protocols [97]. In this respect, future prospects include the development of advanced in vitro models [98], computational prediction methods [99], and genetic toxicity assessments, with the establishment of safety guidelines [101].

    Specialized applications in personalized medicine bring about development challenges for individual-specific formulations [102], rapid small-batch production [15], multi-drug loading [59], and diagnostic/therapeutic function integration [103]. The prospects of this area include additive manufacturing [104], genetic personalization [105], responsive delivery systems [106], and point-of-care production [107].

    Continuous production methods may guarantee more consistency in the overcoming of scale-up issues [90]. New milling technologies are under development for large-scale nanocrystal production, and quality-by-design approaches should guarantee consistent quality across the different scales [36].

    Improvement in long-term stability will need the development of more active and efficient stabilizers, notably multi-component formulations [10]. Surface engineering strategies are currently being investigated to improve stability [1], and predictive models are under development for long-term stability from data of short-term experiments [93].

    It will be very important to establish better testing models that will most closely approximate the in vivo situation to address nanotoxicity concerns [98]. Computational prediction methods [99] and genomic and proteomic techniques [100] are under study for their potential to give insight into the mechanisms of nanotoxicity.

    In this respect, future prospects include 3D printing of bespoke nanocrystal formulations [104], incorporation of genetic information in formulation design [105], and responsive delivery systems that adapt to patient characteristics [106]. Point-of-care production systems are developed to manufacture personalized nanocrystal formulations [107].

    These developments are targeted to overcome the present limitations, thereby opening a broader scope of application for nanocrystal formulations in drug delivery and personalized medicine.

    DISCUSSION

    Pharmaceutical nanocrystals are thus one of the most promising approaches in formulating low-solubility drugs: they enhance bioavailability and steep dose-response relationships, demonstrating reduced food effects on absorption. The successes among commercial products such as Rapamune®, Emend®, and Tricor® have proved the validity of nanocrystal formulations at the clinical level [7]. Nanocrystals have a variety of administration options: oral, parenteral, pulmonary, dermal, and ocular applications, which give totally new prospects for drug delivery [11].

    Manufacturing processes can be segregated into top-down and bottom-up processes. Among them, top-down approaches like media milling and high-pressure homogenization are used industrially in continuous manufacturing because of their scalability issues and homogeneous properties seen in the product [11]. However, it suffers from a lack of control over the particle size when scaled up and heat generation problems [88, 89]. The bottom-up process produces smaller and more homogeneous particles but has dimension control problems in the particles and issues with scale-up.

    Stabilization remains central in nanocrystal development. Several approaches using surfactants, polymers, and layer-by-layer coating have been previously studied [12, 36, 37, 41]. Future studies must, therefore, focus on finding better stabilizers and the use of multiple-component stabilizations for long-term stability.

    The regulatory landscape for nanocrystal-based pharmaceuticals is shifting. Food and Drug Administration (FDA) and European Medicines Agency (EMA) guidelines deal with the development and evaluation of nanomedicine products [73, 74]. Critical concerns include the characterization of particle size, long-term stability demonstration, and nanotoxicity assessment [75, 76, 83]. When this field continues to progress, so will regulatory frameworks with respect to newer applications in the areas of personalized medicine and theranostics.

    Nanocrystal technology contributes to drug delivery and extends medical applications of established and new treatments in a wide range of administration routes.

    CONCLUSION

    Nanocrystal technology has achieved greater potential in addressing the solubility issues associated with poorly aqueous soluble drugs. This technology shall be utilized for various routes of administration to improve the bioavailability and enhanced efficacy of drugs. However, many issues are taken into consideration such as scaling up technology, long-term stability and assessment of toxicity. Apart from these, regulatory frameworks are essential to evaluate and approve nanocrystal-based formulation in the field of drug targeting, combination therapy, and personalized medicine which would be expected to improve patient outcomes for the management and treatment of diseases.

    ACKNOWLEDGEMENT

    The authors would like to thank the Department of Science and Technology-Fund for Improvement of Science and Technology Infrastructure (DST-FIST) and Promotion of University Research and ScientificExcellence (DST-PURSE) and Department of Biotechnology-Boost to University Interdisciplinary Life ScienceDepartments for Education andResearch program (DBT-BUILDER) for the facilities provided in our department.

    FUNDING

    Nil

    AUTHORS CONTRIBUTIONS

    Gurubaran Sivanathan: Literature review, Data curation, Writing-original draft, and Evaluation; Sanjai Rajagopal: Literature review, Data curation, and Writing-original draft; Giridhara Mahadevaswamy: Writing-original draft, Conceptualization, Critical Evaluation; Gowtham Angamuthu: Writing-original draft, Conceptualization, Critical Evaluation; Nagasamy Venkatesh Dhandapani: Review and editing, Supervision, Evaluation, Visualization.

    CONFLICTS OF INTERESTS

    The authors declare no conflict of interest

    REFERENCES

    1. Muller RH, Gohla S, Keck CM. State of the art of nanocrystals special features production, nanotoxicology aspects and intracellular delivery. Eur J Pharm Biopharm. 2011 May;78(1):1-9. doi: 10.1016/j.ejpb.2011.01.007, PMID 21266197.

    2. Keck CM, Muller RH. Drug nanocrystals of poorly soluble drugs produced by high pressure homogenisation. Eur J Pharm Biopharm. 2006 Jan;62(1):3-16. doi: 10.1016/j.ejpb.2005.05.009, PMID 16129588.

    3. Loftsson T, Brewster ME. Pharmaceutical applications of cyclodextrins: basic science and product development. J Pharm Pharmacol. 2010 Nov;62(11):1607-21. doi: 10.1111/j.2042-7158.2010.01030.x, PMID 21039545.

    4. Noyes AA, Whitney WR. The rate of solution of solid substances in their own solutions. J Am Chem Soc. 1897 Dec;19(12):930-4. doi: 10.1021/ja02086a003.

    5. Kesisoglou F, Panmai S, WU Y. Nanosizing oral formulation development and biopharmaceutical evaluation. Adv Drug Deliv Rev. 2007 Jul 30;59(7):631-44. doi: 10.1016/j.addr.2007.05.003, PMID 17601629.

    6. Junghanns JU, Muller RH. Nanocrystal technology drug delivery and clinical applications. Int J Nanomedicine. 2008;3(3):295-309. doi: 10.2147/ijn.s595, PMID 18990939, PMCID PMC2626933.

    7. Peltonen L, Hirvonen J. Pharmaceutical nanocrystals by nanomilling: critical process parameters particle fracturing and stabilization methods. J Pharm Pharmacol. 2010 Nov;62(11):1569-79. doi: 10.1111/j.2042-7158.2010.01022.x, PMID 21039542.

    8. Shegokar R, Muller RH. Nanocrystals: industrially feasible multifunctional formulation technology for poorly soluble actives. Int J Pharm. 2010 Oct 31;399(1-2):129-39. doi: 10.1016/j.ijpharm.2010.07.044, PMID 20674732.

    9. Moschwitzer JP. Drug nanocrystals in the commercial pharmaceutical development process. Int J Pharm. 2013 Aug 30;453(1):142-56. doi: 10.1016/j.ijpharm.2012.09.034, PMID 23000841.

    10. Gao L, Liu G, MA J, Wang X, Zhou L, LI X. Drug nanocrystals: in vivo performances. J Control Release. 2012 Jun 28;160(3):418-30. doi: 10.1016/j.jconrel.2012.03.013, PMID 22465393.

    11. Muller RH, Keck CM. Twenty years of drug nanocrystals: where are we and where do we go? Eur J Pharm Biopharm. 2012 Jan;80(1):1-3. doi: 10.1016/j.ejpb.2011.09.012, PMID 21971369.

    12. Wu L, Zhang J, Watanabe W. Physical and chemical stability of drug nanoparticles. Adv Drug Deliv Rev. 2011 May 30;63(6):456-69. doi: 10.1016/j.addr.2011.02.001, PMID 21315781.

    13. Junyaprasert VB, Morakul B. Nanocrystals for enhancement of oral bioavailability of poorly water-soluble drugs. Asian J Pharm Sci. 2015 Feb 1;10(1):13-23. doi: 10.1016/j.ajps.2014.08.005.

    14. Van Eerdenbrugh B, Van den Mooter G, Augustijns P. Top-down production of drug nanocrystals: nanosuspension stabilization, miniaturization and transformation into solid products. Int J Pharm. 2008 Nov 19;364(1):64-75. doi: 10.1016/j.ijpharm.2008.07.023, PMID 18721869.

    15. Merisko Liversidge E, Liversidge GG. Nanosizing for oral and parenteral drug delivery: a perspective on formulating poorly water-soluble compounds using wet media milling technology. Adv Drug Deliv Rev. 2011 May 30;63(6):427-40. doi: 10.1016/j.addr.2010.12.007, PMID 21223990.

    16. Zhang HX, Wang JX, Zhang ZB, LE Y, Shen ZG, Chen JF. Micronization of atorvastatin calcium by antisolvent precipitation process. Int J Pharm. 2009 Jun 5;374(1-2):106-13. doi: 10.1016/j.ijpharm.2009.02.015, PMID 19446766.

    17. Thorat AA, Dalvi SV. Liquid antisolvent precipitation and stabilization of nanoparticles of poorly water soluble drugs in aqueous suspensions: recent developments and future perspective. Chem Eng J. 2012 Feb 1;181-182:1-34. doi: 10.1016/j.cej.2011.12.044.

    18. Zhang JY, Shen ZG, Zhong J, HU TT, Chen JF, MA ZQ. Preparation of amorphous cefuroxime axetil nanoparticles by controlled nanoprecipitation method without surfactants. Int J Pharm. 2006 Oct 12;323(1-2):153-60. doi: 10.1016/j.ijpharm.2006.05.048, PMID 16828244.

    19. Chattopadhyay P, Gupta RB. Production of griseofulvin nanoparticles using supercritical CO(2) antisolvent with enhanced mass transfer. Int J Pharm. 2001 Oct 9;228(1-2):19-31. doi: 10.1016/s0378-5173(01)00803-1, PMID 11576765.

    20. Kakran M, Sahoo NG, LI L, Judeh Z. Fabrication of quercetin nanoparticles by anti-solvent precipitation method for enhanced dissolution. Powder Technol. 2012 Jun 1;223:59-64. doi: 10.1016/j.powtec.2011.08.021.

    21. Bhattacharjee S. DLS and zeta potential-what they are and what they are not? J Control Release. 2016 Aug 10;235:337-51. doi: 10.1016/j.jconrel.2016.06.017, PMID 27297779.

    22. Shekunov BY, Chattopadhyay P, Tong HH, Chow AH. Particle size analysis in pharmaceutics: principles methods and applications. Pharm Res. 2007 Feb;24(2):203-27. doi: 10.1007/s11095-006-9146-7, PMID 17191094.

    23. Filipe V, Hawe A, Jiskoot W. Critical evaluation of nanoparticle tracking analysis (NTA) by nanosight for the measurement of nanoparticles and protein aggregates. Pharm Res. 2010 May;27(5):796-810. doi: 10.1007/s11095-010-0073-2, PMID 20204471, PMCID PMC2852530.

    24. Clogston JD, Patri AK. Zeta potential measurement. Methods Mol Biol. 2011;697:63-70. doi: 10.1007/978-1-60327-198-1_6, PMID 21116954.

    25. Klang V, Valenta C, Matsko NB. Electron microscopy of pharmaceutical systems. Micron. 2013 Jan;44:45-74. doi: 10.1016/j.micron.2012.07.008, PMID 22921788.

    26. Hondow N, Brydson R, Wang P, Holton MD, Brown MR, Rees P. Quantitative characterization of nanoparticle agglomeration within biological media. J Nanopart Res. 2012 Jul;14(7):1-5. doi: 10.1007/s11051-012-0977-3.

    27. Surwase SA, Boetker JP, Saville D, Boyd BJ, Gordon KC, Peltonen L. Indomethacin: new polymorphs of an old drug. Mol Pharm. 2013 Dec 2;10(12):4472-80. doi: 10.1021/mp400299a, PMID 24025118.

    28. Nigmatullin R, Lovitt R, Wright C, Linder M, Nakari Setala T, Gama M. Atomic force microscopy study of cellulose surface interaction controlled by cellulose binding domains. Colloids Surf B Biointerfaces. 2004 May 15;35(2):125-35. doi: 10.1016/j.colsurfb.2004.02.013, PMID 15261045.

    29. Wiley J. Principles and applications of thermal analysis; 2008. doi: 10.1002/9780470697702.

    30. Sing KS. Reporting physisorption data for gas/solid systems with special reference to the determination of surface area and porosity (Recommendations 1984). Pure Appl Chem. 1985 Jan 1;57(4):603-19. doi: 10.1351/pac198557040603.

    31. Dokoumetzidis A, Macheras P. A century of dissolution research: from noyes and whitney to the biopharmaceutics classification system. Int J Pharm. 2006 Sep 14;321(1-2):1-11. doi: 10.1016/j.ijpharm.2006.07.011, PMID 16920290.

    32. Kazakevich YV, Lobrutto R. HPLC for pharmaceutical scientists. John Wiley & Sons; 2007 Feb 16. doi: 10.1002/0470087951.

    33. Bajaj S, Singla D, Sakhuja N. Stability testing of pharmaceutical products. J App Pharm Sci. 2012 Mar 30:2(3):129-38. doi: 10.7324/JAPS.2012.2322.

    34. Stuhrmann HB. Small angle scattering of X-rays. Prog Cryst Growth Char. 1989 Jan 1;18:1-19. doi: 10.1016/0146-3535(89)90023-3.

    35. Paudel A, Raijada D, Rantanen J. Raman spectroscopy in pharmaceutical product design. Adv Drug Deliv Rev. 2015 Jul 15;89:3-20. doi: 10.1016/j.addr.2015.04.003, PMID 25868453.

    36. Verma S, Kumar S, Gokhale R, Burgess DJ. Physical stability of nanosuspensions: investigation of the role of stabilizers on ostwald ripening. International Journal of Pharmaceutics. 2011;406(1-2):145-52. doi: 10.1016/j.ijpharm.2010.12.027.

    37. Tuomela A, Hirvonen J, Peltonen L. Stabilizing agents for drug nanocrystals: effect on bioavailability. Pharmaceutics. 2016 May 20;8(2):16. doi: 10.3390/pharmaceutics8020016, PMID 27213435, PMCID PMC4932479.

    38. Gigliobianco MR, Casadidio C, Censi R, Di Martino P. Nanocrystals of poorly soluble drugs: drug bioavailability and physicochemical stability. Pharmaceutics. 2018 Aug 21;10(3):134. doi: 10.3390/pharmaceutics10030134, PMID 30134537, PMCID PMC6161002.

    39. Cerdeira AM, Mazzotti M, Gander B. Miconazole nanosuspensions: influence of formulation variables on particle size reduction and physical stability. Int J Pharm. 2010 Aug 30;396(1-2):210-8. doi: 10.1016/j.ijpharm.2010.06.020, PMID 20600732.

    40. Rachmawati H, Al Shaal L, Muller RH, Keck CM. Development of curcumin nanocrystal: physical aspects. J Pharm Sci. 2013 Jan;102(1):204-14. doi: 10.1002/jps.23335, PMID 23047816.

    41. Elbakry A, Wurster EC, Zaky A, Liebl R, Schindler E, Bauer Kreisel P. Layer by layer coated gold nanoparticles: size dependent delivery of DNA into cells. Small. 2012 Dec 21;8(24):3847-56. doi: 10.1002/smll.201201112, PMID 22911477.

    42. Muller RH, Jacobs C. Buparvaquone mucoadhesive nanosuspension: preparation optimisation and long-term stability. Int J Pharm. 2002 Apr 26;237(1-2):151-61. doi: 10.1016/s0378-5173(02)00040-6, PMID 11955813.

    43. Ghosh I, Bose S, Vippagunta R, Harmon F. Nanosuspension for improving the bioavailability of a poorly soluble drug and screening of stabilizing agents to inhibit crystal growth. Int J Pharm. 2011 May 16;409(1-2):260-8. doi: 10.1016/j.ijpharm.2011.02.051, PMID 21371540.

    44. Pawar VK, Singh Y, Meher JG, Gupta S, Chourasia MK. Engineered nanocrystal technology: in-vivo fate targeting and applications in drug delivery. J Control Release. 2014 Jun 10;183:51-66. doi: 10.1016/j.jconrel.2014.03.030, PMID 24667572.

    45. LU Y, LI Y, WU W. Injected nanocrystals for targeted drug delivery. Acta Pharm Sin B. 2016 Mar;6(2):106-13. doi: 10.1016/j.apsb.2015.11.005, PMID 27006893, PMCID PMC4788714.

    46. Park K. Controlled drug delivery systems: past forward and future back. J Control Release. 2014 Sep 28;190:3-8. doi: 10.1016/j.jconrel.2014.03.054, PMID 24794901, PMCID PMC4142099.

    47. Chiang PC, HU Y, Blom JD, Thompson DC. Evaluating the suitability of using rat models for preclinical efficacy and side effects with inhaled corticosteroid nanosuspension formulations. Nanoscale Res Lett. 2010 Apr 10;5(6):1010-9. doi: 10.1007/s11671-010-9597-y, PMID 20672144, PMCID PMC2893943.

    48. Pilcer G, Amighi K. Formulation strategy and use of excipients in pulmonary drug delivery. International Journal of Pharmaceutics. 2010;392(1-2):1-19. doi: 10.1016/j.ijpharm.2010.03.017.

    49. Patel A, Patel M, Yang X, Mitra AK. Recent advances in protein and peptide drug delivery: a special emphasis on polymeric nanoparticles. Protein Pept Lett. 2014;21(11):1102-20. doi: 10.2174/0929866521666140807114240, PMID 25106908, PMCID PMC4407643.

    50. Pireddu R, Caddeo C, Valenti D, Marongiu F, Scano A, Ennas G. Diclofenac acid nanocrystals as an effective strategy to reduce in vivo skin inflammation by improving dermal drug bioavailability. Colloids Surf B Biointerfaces. 2016 Jul 1;143:64-70. doi: 10.1016/j.colsurfb.2016.03.026, PMID 26998867.

    51. Muller RH, Shegokar R, Keck CM. 20 y of lipid nanoparticles (SLN and NLC): present state of development and industrial applications. Curr Drug Discov Technol. 2011 Sep;8(3):207-27. doi: 10.2174/157016311796799062, PMID 21291409.

    52. Patel MN, Lakkadwala S, Majrad MS, Injeti ER, Gollmer SM, Shah ZA. Characterization and evaluation of 5-fluorouracil-loaded solid lipid nanoparticles prepared via a temperature-modulated solidification technique. AAPS Pharm Sci Tech. 2014 Dec;15(6):1498-508. doi: 10.1208/s12249-014-0168-x, PMID 25035070, PMCID PMC4245423.

    53. Sharma OP, Patel V, Mehta T. Nanocrystal for ocular drug delivery: hope or hype. Drug Deliv and Transl Res. 2016 Aug;6(4):399-413. doi: 10.1007/s13346-016-0292-0, PMID 27165145.

    54. Tuomela A, Liu P, Puranen J, Ronkko S, Laaksonen T, Kalesnykas G. Brinzolamide nanocrystal formulations for ophthalmic delivery: reduction of elevated intraocular pressure in vivo. Int J Pharm. 2014 Jun 5;467(1-2):34-41. doi: 10.1016/j.ijpharm.2014.03.048, PMID 24680962.

    55. Mandal A, Bisht R, Rupenthal ID, Mitra AK. Polymeric micelles for ocular drug delivery: from structural frameworks to recent preclinical studies. J Control Release. 2017 Feb 28;248:96-116. doi: 10.1016/j.jconrel.2017.01.012, PMID 28087407, PMCID PMC5319397.

    56. Yadollahi R, Vasilev K, Simovic S. Nanosuspension technologies for delivery of poorly soluble drugs. J Nanomater. 2015;2015(1):216375. doi: 10.1155/2015/216375.

    57. Hollis CP, Weiss HL, Leggas M, Evers BM, Gemeinhart RA, LI T. Biodistribution and bioimaging studies of hybrid paclitaxel nanocrystals: lessons learned of the EPR effect and image-guided drug delivery. J Control Release. 2013 Nov 28;172(1):12-21. doi: 10.1016/j.jconrel.2013.06.039, PMID 23920039, PMCID PMC3886194.

    58. Gao W, Chen Y, Zhang Y, Zhang Q, Zhang L. Nanoparticle-based local antimicrobial drug delivery. Adv Drug Deliv Rev. 2018 Mar 1;127:46-57. doi: 10.1016/j.addr.2017.09.015, PMID 28939377, PMCID PMC5860926.

    59. Miao X, Yang W, Feng T, Lin J, Huang P. Drug nanocrystals for cancer therapy. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2018 May;10(3):e1499. doi: 10.1002/wnan.1499, PMID 29044971.

    60. Cavalli R, Argenziano M, Vigna E, Giustetto P, Torres E, Aime S. Preparation and in vitro characterization of chitosan nanobubbles as theranostic agents. Colloids Surf B Biointerfaces. 2015 May 1;129:39-46. doi: 10.1016/j.colsurfb.2015.03.023, PMID 25819364.

    61. Bobo D, Robinson KJ, Islam J, Thurecht KJ, Corrie SR. Nanoparticle-based medicines: a review of FDA approved materials and clinical trials to date. Pharm Res. 2016 Oct;33(10):2373-87. doi: 10.1007/s11095-016-1958-5, PMID 27299311.

    62. Zhao L, Seth A, Wibowo N, Zhao CX, Mitter N, Yu C. Nanoparticle vaccines. Vaccine. 2014 Jan 9;32(3):327-37. doi: 10.1016/j.vaccine.2013.11.069, PMID 24295808.

    63. Cordeiro AS, Alonso MJ. Recent advances in vaccine delivery. Pharm Pat Anal. 2016;5(1):49-73. doi: 10.4155/ppa.15.38, PMID 26667309.

    64. Simamora P, Alvarez JM, Yalkowsky SH. Solubilization of rapamycin. Int J Pharm. 2001 Feb 1;213(1-2):25-9. doi: 10.1016/s0378-5173(00)00617-7, PMID 11165091.

    65. WU Y, Loper A, Landis E, Hettrick L, Novak L, Lynn K. The role of biopharmaceutics in the development of a clinical nanoparticle formulation of MK-0869: a beagle dog model predicts improved bioavailability and diminished food effect on absorption in human. Int J Pharm. 2004 Nov 5;285(1-2):135-46. doi: 10.1016/j.ijpharm.2004.08.001, PMID 15488686.

    66. Fakes MG, Vakkalagadda BJ, Qian F, Desikan S, Gandhi RB, Lai C. Enhancement of oral bioavailability of an HIV attachment inhibitor by nanosizing and amorphous formulation approaches. International Journal of Pharmaceutics. 2009;370(1-2):167-74. doi: 10.1016/j.ijpharm.2008.11.018.

    67. Citrome L. Paliperidone palmitate review of the efficacy safety and cost of a new second generation depot antipsychotic medication. Int J Clin Pract. 2010 Jan;64(2):216-39. doi: 10.1111/j.1742-1241.2009.02240.x, PMID 19886879.

    68. Sohn GK, Kwon GP, Bailey Healy I, Mirza A, Sarin K, Oro A. Topical itraconazole for the treatment of basal cell carcinoma in patients with basal cell nevus syndrome or high frequency basal cell carcinomas: a phase 2, open-label, placebo-controlled trial. JAMA Dermatol. 2019 Sep 1;155(9):1078-80. doi: 10.1001/jamadermatol.2019.1541, PMID 31339515, PMCID PMC6659355.

    69. Kawabata Y, Wada K, Nakatani M, Yamada S, Onoue S. Formulation design for poorly water soluble drugs based on biopharmaceutics classification system: basic approaches and practical applications. Int J Pharm. 2011 Nov 25;420(1):1-10. doi: 10.1016/j.ijpharm.2011.08.032, PMID 21884771.

    70. Markman JL, Rekechenetskiy A, Holler E, Ljubimova JY. Nanomedicine therapeutic approaches to overcome cancer drug resistance. Adv Drug Deliv Rev. 2013 Nov;65(13-14):1866-79. doi: 10.1016/j.addr.2013.09.019, PMID 24120656, PMCID PMC5812459.

    71. Kumar B, Jalodia K, Kumar P, Gautam HK. Recent advances in nanoparticle-mediated drug delivery. J Drug Deliv Sci Technol. 2017 Oct 1;41:260-8. doi: 10.1016/j.jddst.2017.07.019.

    72. Peltonen L, Hirvonen J. Drug nanocrystals versatile option for formulation of poorly soluble materials. Int J Pharm. 2018 Feb 15;537(1-2):73-83. doi: 10.1016/j.ijpharm.2017.12.005, PMID 29262301.

    73. FDA. Considering whether an FDA-regulated product involves the application of nanotechnology. Guid Ind FDA-2010-D; 2017.

    74. European Medicines Agency. Reflection paper on the data requirements for intravenous liposomal products developed with reference to an innovator liposomal product. EMA/CHMP/806058/2009/Rev. Vol. 02; 2015.

    75. Tyner KM, Zou P, Yang X, Zhang H, Cruz CN, Lee SL. Product quality for nanomaterials: current U.S. experience and perspective. WIREs Nanomed Nanobiotechnol. 2015 Sep-Oct;7(5):640-54. doi: 10.1002/wnan.1338, PMID 25641690.

    76. Buckley ST, Frank KJ, Fricker G, Brandl M. Biopharmaceutical classification of poorly soluble drugs with respect to enabling formulations. Eur J Pharm Sci. 2013 Sep 27;50(1):8-16. doi: 10.1016/j.ejps.2013.04.002, PMID 23583787.

    77. Reflection paper on nanotechnology-based medicinal products for human use. European Medicines Agency; 2006.

    78. Ehmann F, Sakai Kato K, Duncan R, Perez DE LA Ossa DH, Pita R, Vidal JM. Next-generation nanomedicines and nanosimilars: EU regulators initiatives relating to the development and evaluation of nanomedicines. Nanomedicine. 2013 May;8(5):849-56. doi: 10.2217/nnm.13.68, PMID 23656268.

    79. International Council for Harmonisation: [ICH guidelines]; 2021. Available from: https://www.ich.org/page/ich-guidelines.

    80. Colombo S, Cun D, Remaut K, Bunker M, Zhang J, Martin Bertelsen B. Mechanistic profiling of the siRNA delivery dynamics of lipid polymer hybrid nanoparticles. J Control Release. 2015 Mar 10;201:22-31. doi: 10.1016/j.jconrel.2014.12.026, PMID 25540904.

    81. Siewert M, Dressman J, Brown CK, Shah VP, FIP, AAPS. FIP/AAPS guidelines to dissolution/in vitro release testing of novel/special dosage forms. AAPS Pharm Sci Tech. 2003;4(1):E7. doi: 10.1208/pt040107, PMID 12916916, PMCID PMC2750303.

    82. Moschwitzer J, Achleitner G, Pomper H, Muller RH. Development of an intravenously injectable chemically stable aqueous omeprazole formulation using nanosuspension technology. Eur J Pharm Biopharm. 2004 Nov;58(3):615-9. doi: 10.1016/j.ejpb.2004.03.022, PMID 15451536.

    83. Oberdorster G. Safety assessment for nanotechnology and nanomedicine: concepts of nanotoxicology. J Intern Med. 2010 Jan;267(1):89-105. doi: 10.1111/j.1365-2796.2009.02187.x, PMID 20059646.

    84. International Pharmaceutical Regulators Programme. Nanomedicines working group; 2021. Available from: https://www.iprp.global/working-group/nanomedicines.

    85. International Organization for Standardization. ISO/TC 229 nanotechnologies; 2021. Available from: https://www.iso.org/committee/381983.html.

    86. Ventola CL. Progress in nanomedicine: approved and investigational nanodrugs. PT. 2017 Dec;42(12):742-55. PMID 29234213, PMCID PMC5720487.

    87. Keck CM, Muller RH. Nanotoxicological classification system (NCS)-a guide for the risk benefit assessment of nanoparticulate drug delivery systems. Eur J Pharm Biopharm. 2013 Aug;84(3):445-8. doi: 10.1016/j.ejpb.2013.01.001, PMID 23333302.

    88. Muller RH, Keck CM. Twenty years of drug nanocrystals: where are we and where do we go? Eur J Pharm Biopharm. 2012 Jan;80(1):1-3. doi: 10.1016/j.ejpb.2011.09.012, PMID 21971369.

    89. r113

    90. Thrix

    91. This is a duplicate of reference 11.

    92. Chin WW, Parmentier J, Widzinski M, Tan EH, Gokhale R. A brief literature and patent review of nanosuspensions to a final drug product. J Pharm Sci. 2014 Oct;103(10):2980-99. doi: 10.1002/jps.24098, PMID 25099918.

    93. Plakkot S, DE Matas M, York P, Saunders M, Sulaiman B. Comminution of ibuprofen to produce nano particles for rapid dissolution. Int J Pharm. 2011 Aug 30;415(1-2):307-14. doi: 10.1016/j.ijpharm.2011.06.002, PMID 21683776.

    94. Kakran M, Shegokar R, Sahoo NG, Shaal LA, LI L, Muller RH. Fabrication of quercetin nanocrystals: comparison of different methods. Eur J Pharm Biopharm. 2012 Jan;80(1):113-21. doi: 10.1016/j.ejpb.2011.08.006, PMID 21896330.

    95. Sarnes A, Kovalainen M, Hakkinen MR, Laaksonen T, Laru J, Kiesvaara J. Nanocrystal based per oral itraconazole delivery: superior in vitro dissolution enhancement versus Sporanox® is not realized in in vivo drug absorption. J Control Release. 2014 Apr 28;180:109-16. doi: 10.1016/j.jconrel.2014.02.016, PMID 24566254.

    96. Shah DA, Murdande SB, Dave RH. A review: pharmaceutical and pharmacokinetic aspect of nanocrystalline suspensions. J Pharm Sci. 2016 Jan;105(1):10-24. doi: 10.1002/jps.24694, PMID 26580860.

    97. Chaubal MV, Popescu C. Conversion of nanosuspensions into dry powders by spray drying: a case study. Pharm Res. 2008 Oct;25(10):2302-8. doi: 10.1007/s11095-008-9625-0, PMID 18509597.

    98. Oberdorster G, Oberdorster E, Oberdorster J. Nanotoxicology: an emerging discipline evolving from studies of ultrafine particles. Environ Health Perspect. 2005 Jul;113(7):823-39. doi: 10.1289/ehp.7339. PMID 16002369, PMCID PMC1257642.

    99. Nel A, Xia T, Madler L, LI N. Toxic potential of materials at the nanolevel. Science. 2006 Feb 3;311(5761):622-7. doi: 10.1126/science.1114397, PMID 16456071.

    100. Fadeel B, Fornara A, Toprak MS, Bhattacharya K. Keeping it real: the importance of material characterization in nanotoxicology. Biochem Biophys Res Commun. 2015 Dec 18;468(3):498-503. doi: 10.1016/j.bbrc.2015.06.178, PMID 26187673.

    101. Mahmoudi M, Hofmann H, Rothen Rutishauser B, Petri Fink A. Assessing the in vitro and in vivo toxicity of superparamagnetic iron oxide nanoparticles. Chem Rev. 2012 Apr 11;112(4):2323-38. doi: 10.1021/cr2002596, PMID 22216932.

    102. Winkler DA, Mombelli E, Pietroiusti A, Tran L, Worth A, Fadeel B. Applying quantitative structure-activity relationship approaches to nanotoxicology: current status and future potential. Toxicology. 2013 Nov 8;313(1):15-23. doi: 10.1016/j.tox.2012.11.005, PMID 23165187.

    103. Fadeel B, Farcal L, Hardy B, Vazquez Campos S, Hristozov D, Marcomini A. Advanced tools for the safety assessment of nanomaterials. Nature Nanotech. 2018 Jul;13(7):537-43. doi: 10.1038/s41565-018-0185-0, PMID 29980781.

    104. Sainz V, Conniot J, Matos AI, Peres C, Zupanoio E, Moura L. Regulatory aspects on nanomedicines. Biochem Biophys Res Commun. 2015 Dec 18;468(3):504-10. doi: 10.1016/j.bbrc.2015.08.023, PMID 26260323.

    105. Patel VF, Liu F, Brown MB. Advances in oral transmucosal drug delivery. J Control Release. 2011 Jul 30;153(2):106-16. doi: 10.1016/j.jconrel.2011.01.027, PMID 21300115.

    106. Muthu MS, Leong DT, Mei L, Feng SS. Nanotheranostics application and further development of nanomedicine strategies for advanced theranostics. Theranostics. 2014 Mar 26;4(6):660-77. doi: 10.7150/thno.8698, PMID 24723986, PMCID PMC3982135.

    107. Norman J, Madurawe RD, Moore CM, Khan MA, Khairuzzaman A. A new chapter in pharmaceutical manufacturing: 3D-printed drug products. Adv Drug Deliv Rev. 2017 Jan 1;108:39-50. doi: 10.1016/j.addr.2016.03.001, PMID 27001902.

    108. Patel M, Joshi G, Sawant KK. Nanotechnology in oral drug delivery: salient aspects state of art and applications. In: Functional bionanomaterials: from biomolecules to nanoparticles; 2020. p. 165-84. doi: 10.1007/978-3-030-41464-1_8.

    109. Mura S, Nicolas J, Couvreur P. Stimuli-responsive nanocarriers for drug delivery. Nature Mater. 2013 Nov;12(11):991-1003. doi: 10.1038/nmat3776, PMID 24150417.

    110. Prasad LK, Smyth H. 3D printing technologies for drug delivery: a review. Drug Dev Ind Pharm. 2016;42(7):1019-31. doi: 10.3109/03639045.2015.1120743, PMID 26625986.

    111. Saritha D, Chandra Bose PS, Osmani RA, Iriventi P, Kanna S, Ravi G. Development optimization and in vitro characterization of haloperidol nanocrystals using 32 factorial design. Int J App Pharm. 2024;16(3):187-94. doi: 10.22159/ijap.2024v16i3.50412.

    112. Abdellatif MM, Ahmed SM, EL Nabarawi MA, Teaima M. Nano delivery systems for enhancing oral bioavailability of drugs. Int J App Pharm. 2023;15(1):13-9. doi: 10.22159/ijap.2023v15i1.46758.