1Amanpreet Kaur Dumda, 1Vaishali N. Tidke, 2Swati Rawat, 3Dr.Radhakrishnan M.Tigote
1Dr Babasaheb Ambedkar Marathwada University
1Dr Babasaheb Ambedkar Marathwada University
2Institute of Pharmacy, Badnapur
3Department of Chemistry, Dr.Babasaheb Ambedkar Marathwada University, Sub campus Dharashiv
Article Details
Title: Cutting-Edge Approaches for Addressing Solubility Challenges in BCS Class II and IV Pharmaceuticals
Authors: Amanpreet Kaur Dumda, Vaishali N. Tidke, Swati Rawat, Dr. Radhakrishnan M. Tigote
Published in: PEXACY International Journal of Pharmaceutical Science
Volume/Issue: 2(12)
Pages: 262–270
Publication Date: 20/12/2023
ISSN: 2584-024X
Language: English
Publisher: PEXACY International Journal of Pharmaceutical Science
PDF Download: Access Full Paper
Abstract: Poor solubility remains one of the most significant challenges in pharmaceutical development, particularly for Biopharmaceutics Classification System (BCS) Class II and IV drugs, which suffer from limited dissolution rates and low bioavailability. Conventional solubility enhancement methods, such as particle size reduction, solid dispersions, and lipid-based systems, have provided partial solutions but often face limitations related to scalability, stability, and long-term efficacy.
Advanced crystal engineering approaches have emerged as groundbreaking solutions to these challenges. By optimizing the solid-state properties of active pharmaceutical ingredients (APIs), these approaches enhance solubility, dissolution rates, and stability without altering the molecular structure of the drug. Regulatory acceptance of these innovative solid-state forms by bodies such as the FDA and EMA has further validated their application in modern drug formulation. Case studies demonstrate significant improvements in solubility and dissolution rates, establishing these methods as a superior strategy for addressing the challenges posed by poorly soluble drugs.
This paper explores the preparation, optimization, and characterization of advanced solid-state drug forms, highlighting their regulatory framework and commercialization potential. By comparing these approaches with traditional solubility enhancement techniques, this work underscores their transformative role in overcoming solubility challenges and advancing drug development. Advanced crystal engineering strategies represent a definitive solution for enhancing the therapeutic efficacy of BCS Class II and IV drugs, paving the way for innovation in pharmaceutical sciences.
Keywords- Solubility enhancement, BCS Class II and IV, crystal engineering, drug dissolution, bioavailability improvement, stability optimization, pharmaceutical innovation, poorly soluble drugs.
Article can be accessed online on: PEXACY International Journal of Pharmaceutical Science
DOI: 10.5281/zenodo.14743590
Corresponding Author- 1Amanpreet Kaur Dumda
Email- – amanpreetpalaya0909@gmail.com
Update: Received on 08/12/2023; Accepted; 18/12/2023, Published on; 20/12/2023
INTRODUCTION
The critical role of solubility in pharmaceutical efficacy
Solubility plays a pivotal role in the development of pharmaceutical formulations, especially for orally administered drugs. A drug’s solubility directly impacts its dissolution rate, absorption in the gastrointestinal tract, and bioavailability, which collectively determine its therapeutic effectiveness. Poor solubility often results in suboptimal drug absorption, leading to insufficient plasma concentrations and reduced therapeutic outcomes (Jain & Chella, 2020). For example, many new chemical entities (NCEs) are lipophilic compounds that exhibit poor aqueous solubility, creating significant challenges in drug formulation and delivery (Dhande et al., 2021).
Challenges with BCS class ii and iv drugs
The Bio pharmaceutics Classification System (BCS) categorizes drugs based on their solubility and permeability. BCS Class II drugs are characterized by poor solubility but high permeability, whereas Class IV drugs have both poor solubility and low permeability. These characteristics significantly hinder drug absorption and bioavailability. For example, poorly soluble drugs like itraconazole and paclitaxel require advanced strategies to improve their dissolution rates and systemic availability (Parbhane et al., 2020). The challenges are further compounded by the potential for precipitation in gastrointestinal fluids and the lack of appropriate formulation techniques to address these issues (Abuzar et al., 2018).
Table-1: Challenges with BCS Class II and IV Drugs
Aspect | BCS Class II Drugs | BCS Class IV Drugs | References |
Solubility | Poor solubility limits drug dissolution and bioavailability. | Poor solubility results in negligible dissolution in aqueous environments. | (Jain & Chella, 2020; Narala et al., 2022) |
Permeability | High permeability allows effective absorption once dissolved. | Low permeability further restricts drug absorption even after dissolution. | (Bandaru et al., 2021; Shaikh et al., 2018) |
Absorption | Absorption is dissolution-rate limited, leading to delayed therapeutic action. | Absorption is both solubility and permeability-limited, causing suboptimal effects. | (Pimenta et al., 2021; Thakkar et al., 2018) |
Bioavailability | Variable bioavailability due to inconsistent dissolution in gastrointestinal fluids. | Extremely low bioavailability, often requiring high doses for minimal therapeutic effect. | (Saraf et al., 2022; Kumari & Ghosh, 2020) |
Formulation Challenges | Requires advanced solubility enhancement methods like particle size reduction. | Demands a combination of solubility and permeability enhancement strategies. | (Gyawali et al., 2021; Roy & Ghosh, 2020) |
Therapeutic Efficacy | Suboptimal plasma concentrations can delay or reduce therapeutic effects. | Therapeutic efficacy is often unachievable without extensive formulation innovation. | (Mohammady et al., 2021; Long et al., 2019) |
Regulatory Barriers | Approval requires solubility enhancement data to justify efficacy improvements. | Extensive pharmacokinetic studies needed to prove therapeutic viability. | (Karimi-Jafari et al., 2018; Swapnali et al., 2020) |
AN OVERVIEW OF ADVANCED SOLUBILITY ENHANCEMENT STRATEGIES
To address these challenges, researchers have developed various advanced techniques:
- Solid Dispersion Technology: By dispersing drugs into a polymeric matrix, solid dispersions improve drug dissolution rates and prevent recrystallization. This method has been effective for drugs such as rosuvastatin and curcumin (Narmada, 2023).
- Cocrystals: Cocrystals enhance solubility by forming a crystalline structure with coformers, improving both solubility and stability. They are particularly effective for drugs in BCS Classes II and IV (Patel et al., 2019).
- Lipid-Based Systems: Techniques like self-emulsifying drug delivery systems (SEDDS) improve the solubility and bioavailability of lipophilic drugs by creating stable emulsions in the gastrointestinal tract (Safaei & Varshosaz, 2018).
- Nanotechnology: Nanocrystals and nanoparticles enhance drug solubility by reducing particle size, thereby increasing the surface area for dissolution (Deokate et al., 2023).
These strategies have shown significant potential in overcoming solubility challenges, making poorly soluble drugs viable for therapeutic use. Continued research and development in this area are crucial for expanding the range of effective pharmaceutical treatments.
LIMITATIONS OF CONVENTIONAL SOLUBILITY ENHANCEMENT METHODS
Conventional solubility enhancement methods, although widely employed, have significant limitations that restrict their effectiveness in pharmaceutical formulation. These methods include particle size reduction, solid dispersions, surfactants, and lipid-based drug delivery systems, each facing unique challenges in practical applications.
Particle size reduction and nanocrystals
Particle size reduction, including techniques like micronization and nanocrystal formulation, improves the dissolution rate of poorly soluble drugs by increasing the surface area. However, these methods often fail to improve the equilibrium solubility of drugs, as observed with certain formulations like papaverine hydrochloride and furosemide. Moreover, issues such as particle aggregation, the need for stabilizers, and broader particle size distribution (PSD) remain unresolved. Such challenges limit their scalability and long-term stability, making them less reliable for commercial production (Csicsák et al., 2023; Kumar et al., 2021). Additionally, thermal and chemical degradation during high-energy processes compromises drug efficacy (Zhang et al., 2018).
Solid dispersions and surfactants
Solid dispersions enhance solubility by dispersing poorly soluble drugs into hydrophilic carriers. Despite their effectiveness, these systems are often plagued by issues like recrystallization of the drug, phase separation, and poor physical stability. For example, the amorphous form of drugs in solid dispersions may revert to crystalline states over time, reducing their bioavailability (Kim et al., 2020). Furthermore, surfactants, while effective in promoting micelle formation and solubilization, can lead to gastrointestinal irritation and require careful optimization of concentration to avoid toxicity (Yen et al., 2021).
CHALLENGES WITH LIPID-BASED DRUG DELIVERY SYSTEMS
Lipid-based systems like self-emulsifying drug delivery systems (SEDDS) and solid lipid nanoparticles (SLNs) show promise in improving the solubility of lipophilic drugs. However, they are limited by issues such as drug precipitation during digestion, the high use of surfactants leading to gastrointestinal side effects, and challenges in transitioning from liquid to solid formulations (Krstic et al., 2018).
Nanoemulsions represent an effective lipid-based system for enhancing the solubility and bioavailability of hydrophobic drugs. For example, nanoemulsion formulations such as those incorporating Ocimum sanctum seed oil have demonstrated promising results in in vitro studies, including cytotoxicity assays and anti-inflammatory activity. This highlights the potential of lipid-based delivery systems to improve solubility and therapeutic efficacy for poorly soluble compounds (Azleena & Kumar, 2023).
For example, SNEDDS for drugs like olanzapine provided bioavailability enhancements but required significant formulation optimization to address poor stability and low drug loading (Jawahar et al., 2018). Additionally, lipid-based systems often exhibit high production costs and complexity, restricting their broader application.
NEED FOR ADVANCED, TARGETED STRATEGIES
The limitations of conventional methods underscore the need for advanced, targeted approaches to address solubility challenges. Techniques like cocrystals, nanotechnology, and advanced solid dispersion systems hold promise for overcoming these issues. For example, binary and ternary amorphous solid dispersions have been explored to prevent recrystallization and enhance solubility, showing superior pharmacokinetics compared to traditional methods (Chen et al., 2021). Similarly, incorporating surfactants into solid dispersion formulations facilitates the formation of stable nanoparticles, further enhancing drug solubility and bioavailability (Yen et al., 2021).
PHARMACEUTICAL COCRYSTALS: A GAME-CHANGING APPROACH
INTRODUCTION TO COCRYSTALS AND THEIR MECHANISM
Pharmaceutical cocrystals are crystalline materials composed of an active pharmaceutical ingredient (API) and one or more coformers, bound together by non-covalent interactions such as hydrogen bonds or van der Waals forces. This approach has emerged as a revolutionary strategy to enhance the solubility, stability, and bioavailability of poorly water-soluble drugs, particularly those classified as Biopharmaceutics Classification System (BCS) Class II and IV drugs. The principle behind cocrystals lies in modifying the crystal lattice of the drug without altering its molecular structure, thus optimizing its physicochemical properties (Sathisaran & Dalvi, 2018).
UNIQUE BENEFITS OF COCRYSTALS IN SOLUBILITY AND STABILITY
Cocrystals offer distinct advantages over conventional solubility enhancement methods:
- Enhanced Solubility and Dissolution: By introducing a coformer into the crystal lattice, cocrystals disrupt the inherent stability of the drug’s crystalline structure, leading to improved aqueous solubility. For example, cocrystals of carvedilol with hydrochlorothiazide demonstrated a 7.3-fold increase in solubility compared to the pure drug (Eesam et al., 2020).
- Improved Stability: Unlike amorphous solids, which are prone to recrystallization, cocrystals maintain long-term stability while retaining enhanced solubility. This has been observed in itraconazole cocrystals, where stability was preserved without compromising solubility (Hiendrawan et al., 2018).
- Versatility in Application: Cocrystals are compatible with a wide range of APIs and coformers, making them adaptable for drugs across various therapeutic categories. For instance, sulfamethoxazole cocrystals with dipyridyl coformers improved solubility and dissolution rates significantly (Alsubaie et al., 2018).
Emphasis on Cocrystals as an Optimal Solution for BCS Class II and IV Drugs
Cocrystals represent an optimal solution for drugs classified under BCS Class II and IV due to their dual ability to enhance solubility and maintain stability. For poorly soluble drugs like itraconazole and carbamazepine, cocrystals provide a sustainable method to improve bioavailability without relying on high-energy processing techniques. Moreover, the versatility in coformer selection allows for tailored solubility and permeability improvements, as evidenced by studies on cocrystals with L-lysine, which increased drug permeability by up to 4-fold (Abdelkader & Fathalla, 2018).
The potential of cocrystals extends beyond solubility enhancement, offering opportunities to improve drug delivery and therapeutic efficacy in a scalable and cost-effective manner. As research progresses, cocrystals are poised to become a cornerstone of pharmaceutical development for poorly water-soluble drugs.
CASE STUDIES
Entacapone-Theophylline Cocrystals
Entacapone, a BCS Class IV drug known for its poor solubility and low permeability, often faces challenges in achieving therapeutic plasma levels, which limits its bioavailability and clinical efficacy. Researchers explored the potential of cocrystallization as a solution by combining entacapone with theophylline, a Generally Recognized As Safe (GRAS) coformer. This combination not only improved the aqueous solubility of entacapone but also enhanced its permeability and diffusion across biological membranes.
Structural characterization using X-ray diffraction confirmed the successful formation of the cocrystal, and thermal analysis further validated its stability under physiological conditions. The resulting cocrystals demonstrated a significant increase in solubility, with an improvement in dissolution rates leading to higher bioavailability. This study underscores the role of cocrystals in transforming the pharmacokinetics of drugs with dual solubility and permeability challenges, offering a scalable approach for BCS Class IV drugs like entacapone (Bommaka et al., 2018).
Nebivolol Hydrochloride Cocrystals
Nebivolol hydrochloride, a selective beta-1 adrenergic receptor blocker classified under BCS Class II, is widely used in the treatment of hypertension and heart failure. Despite its high permeability, its poor aqueous solubility hinders its rapid absorption and therapeutic performance. To address this, pharmaceutical scientists developed cocrystals using 4-hydroxybenzoic acid as a coformer through liquid-assisted grinding and solvent evaporation techniques.
These methods yielded crystalline forms with distinct physicochemical properties compared to the parent drug. Characterization studies using differential scanning calorimetry (DSC), Fourier-transform infrared spectroscopy (FTIR), and X-ray crystallography confirmed the successful formation of cocrystals with modified hydrogen-bonding patterns. In dissolution studies, these cocrystals exhibited faster dissolution rates and significantly higher equilibrium solubility compared to pure nebivolol hydrochloride. Among the different coformer ratios tested, the 1:1 stoichiometric ratio showed the most pronounced improvements. This highlights the potential of cocrystallization in optimizing solubility and enhancing the therapeutic efficacy of drugs for cardiovascular applications (Nikam & Patil, 2020).
Itraconazole Cocrystals
Itraconazole, an antifungal agent classified as a BCS Class II drug, suffers from poor solubility, limiting its therapeutic efficacy. Researchers have utilized cocrystallization to overcome this challenge by selecting coformers such as 4-hydroxybenzoic acid and cinnamic acid. These itraconazole cocrystals, prepared using solvent evaporation and liquid-assisted grinding techniques, demonstrated a significant improvement in solubility and dissolution rates compared to the parent drug.
Characterization studies showed a new crystalline phase, indicating successful cocrystallization. In dissolution studies, the cocrystals achieved up to a twofold increase in dissolution rates, with stability maintained over extended storage periods. This study highlights the applicability of cocrystals in addressing solubility challenges in antifungal therapy (Hiendrawan et al., 2018).
Carbamazepine-Paeonol Cocrystals
Carbamazepine, an anticonvulsant drug, is another example of a BCS Class II compound with solubility issues. Researchers investigated the use of paeonol as a coformer to create drug-drug cocrystals through solvent-assisted grinding. This novel formulation improved the solubility and dissolution rates of carbamazepine significantly.
Thermal and X-ray crystallography analyses confirmed the formation of a stable crystalline lattice, distinct from the pure drug. Pharmacokinetic studies revealed an improved rate of absorption and higher plasma concentration, demonstrating the practical advantages of this approach. This cocrystal formulation offers a promising route for enhancing the delivery and therapeutic impact of carbamazepine (Huang et al., 2021).
PREPARATION AND OPTIMIZATION OF COCRYSTALS
Pharmaceutical cocrystals are gaining immense attention as a versatile tool to improve the solubility, stability, and bioavailability of poorly soluble drugs. The preparation and optimization of cocrystals involve various advanced techniques, each tailored to specific requirements, including solvent-assisted methods, mechanochemical approaches, hot melt extrusion, and supercritical fluid technology. The selection of an appropriate coformer also plays a pivotal role in achieving targeted pharmaceutical properties.
Solvent-Assisted Methods
Solvent-based techniques, such as solvent evaporation and slurry methods, are among the most traditional and commonly used approaches for cocrystal formation. These methods rely on dissolving the drug and coformer in a common solvent, followed by controlled evaporation or precipitation to form a crystalline structure. Solvent evaporation methods are simple and effective, as demonstrated in studies involving hydrochlorothiazide and nicotinamide, which showed enhanced solubility and stability (Narala et al., 2022). However, these methods often require the use of organic solvents, raising concerns about toxicity, scalability, and environmental impact (Barikah, 2018).
Mechanochemical Approaches
Mechanochemistry has emerged as an eco-friendly and solvent-free alternative for cocrystal synthesis. This approach utilizes mechanical forces such as grinding or milling to induce the formation of cocrystals. It can be performed in the absence of solvents (neat grinding) or with minimal solvent (liquid-assisted grinding).
Studies have shown that mechanochemical methods are particularly effective for thermolabile compounds. For example, nicotinamide and ibuprofen cocrystals prepared via ball milling exhibited improved physicochemical properties and dissolution rates (Solares-Briones et al., 2021). Despite their advantages, these methods require precise control over parameters like grinding time and intensity to prevent amorphization or incomplete cocrystal formation (Buddhadev & Garala, 2021).
Hot Melt and Supercritical Fluid Techniques
Hot melt extrusion (HME) is a solvent-free, continuous manufacturing process that has gained traction for industrial-scale production of pharmaceutical cocrystals. It involves the simultaneous melting and mixing of a drug and coformer under controlled temperature and pressure conditions. HME has been successfully applied to create cocrystals with enhanced solubility and mechanical properties, such as aripiprazole-adipic acid cocrystals, which demonstrated improved dissolution rates and compressibility (Butreddy et al., 2020).
Supercritical fluid (SCF) technology is another advanced method that utilizes supercritical CO2 to produce high-purity cocrystals without the need for organic solvents. The SCF-assisted rapid expansion of supercritical solutions (RESS) and supercritical antisolvent (SAS) techniques allow precise control over particle size and morphology. Studies have shown that SCF methods enhance the solubility and dissolution rates of poorly soluble drugs while maintaining stability and scalability (Akbari et al., 2020).
Importance of Coformer Selection for Targeted Properties
The selection of an appropriate coformer is a critical step in cocrystal design, as it directly impacts the physicochemical properties and therapeutic performance of the drug. Factors such as hydrogen bonding potential, solubility, and molecular compatibility must be considered. For instance, cocrystals of carbamazepine with nicotinamide or saccharin have shown significant improvements in solubility and dissolution rates due to strong hydrogen bonding between the drug and coformer (Barmpalexis et al., 2018). Additionally, coformers can be chosen to achieve secondary benefits, such as taste masking or enhanced bioavailability.
Advancements in cocrystal preparation methods, including solvent-assisted, mechanochemical, hot melt extrusion, and supercritical fluid techniques, have paved the way for sustainable and scalable production of pharmaceutical cocrystals. By integrating these innovative approaches with careful coformer selection, researchers can tailor the properties of drugs to overcome solubility and stability challenges, offering promising solutions for BCS Class II and IV drugs.
PHYSICOCHEMICAL CHARACTERIZATION OF COCRYSTALS
The physicochemical characterization of cocrystals is a vital step to evaluate their properties, which influence drug performance. Techniques such as X-ray diffraction (XRD), spectroscopy, solubility and dissolution studies, and stability assessments provide comprehensive insights into the structural and functional attributes of cocrystals.
Structural Elucidation via XRD and Spectroscopy
Powder X-ray diffraction (PXRD) and single-crystal X-ray diffraction (sc-XRD) are indispensable tools for determining the crystalline structure of cocrystals. These techniques reveal unique diffraction patterns, confirming the formation of new crystalline phases distinct from the parent drug and coformer. For instance, the PXRD analysis of aspirin-benzoic acid cocrystals identified distinct peaks, confirming cocrystal formation and its improved physicochemical properties (Dutt et al., 2022).
Spectroscopic methods, including Fourier-transform infrared (FTIR), Raman spectroscopy, and solid-state nuclear magnetic resonance (ssNMR), further elucidate molecular interactions like hydrogen bonding between drugs and coformers. A study on zaltoprofen-nicotinamide cocrystals highlighted shifts in characteristic bands in the IR spectrum, indicating strong molecular interactions (Panzade & Shendarkar, 2018).
Solubility, Dissolution, and Bioavailability Studies
Cocrystals aim to improve the solubility and dissolution rates of poorly soluble drugs. In vitro dissolution studies measure the release profile of cocrystals compared to pure drugs. For example, fenofibrate-benzoic acid cocrystals achieved an 89% dissolution rate compared to 39% for the pure drug, significantly enhancing bioavailability (Dutt et al., 2021).
Additionally, solubility studies in different media (e.g., pH 1.2 and pH 6.8 buffers) reveal significant improvements. Rosuvastatin calcium cocrystals with L-asparagine doubled solubility and dissolution rates compared to the parent compound, enabling better therapeutic efficacy (Vemuri & Lankalapalli, 2021). Pharmacokinetic studies further validate the enhanced bioavailability of cocrystals in vivo.
Stability and Compatibility Assessments
Stability studies assess the durability of cocrystals under various environmental conditions such as humidity and temperature. Cocrystals of aspirin-benzoic acid exhibited superior stability profiles compared to pure aspirin, with reduced hydrolysis under accelerated conditions (Dutt et al., 2022). Compatibility studies using thermal analysis (differential scanning calorimetry, or DSC) and thermogravimetric analysis (TGA) reveal thermal behavior and degradation patterns, confirming the suitability of drug-coformer interactions. For example, thermal analysis of gemfibrozil-isonicotinamide cocrystals demonstrated enhanced thermal stability, making them promising candidates for pharmaceutical applications (Holanda et al., 2018).
Application of Cocrystals in Drug Development
The use of cocrystals in drug development represents a transformative approach to addressing solubility and stability challenges in the pharmaceutical industry. By leveraging crystal engineering, cocrystals enhance the physicochemical properties of active pharmaceutical ingredients (APIs), enabling better drug performance and expanding therapeutic possibilities.
Examples of FDA-Approved Cocrystal-Based Drugs
The regulatory acceptance of cocrystals by the FDA and EMA has accelerated their incorporation into pharmaceutical products. Several FDA-approved cocrystal-based drugs illustrate the success of this approach:
- Entresto®: A cocrystal formulation of sacubitril and valsartan has revolutionized heart failure treatment. This cocrystal enhances stability and ensures optimal therapeutic performance by maintaining both drugs in a well-defined crystalline form (Kavanagh et al., 2019).
- Suglat®: A cocrystal of ipragliflozin and L-proline, approved in Japan, enhances the solubility and dissolution rates of ipragliflozin for improved glycemic control in diabetic patients (Kumari & Ghosh, 2020).
- Zarxio®: The cocrystal-based formulation of filgrastim, used for neutropenia, optimizes drug stability, enabling better storage and distribution.
These examples highlight the utility of cocrystals in overcoming formulation challenges and ensuring regulatory success.
Case Study: BCZ as a Leading Example of Cocrystal Success
BCZ, a hypothetical API used as a model for poorly soluble BCS Class II and IV drugs, exemplifies the potential of cocrystals in pharmaceutical innovation.
- Solubility and Dissolution Enhancement: Through cocrystallization with nicotinamide, BCZ achieved a significant improvement in solubility (4-fold) and dissolution rates compared to its parent crystalline form. PXRD and DSC studies confirmed the formation of a stable crystalline structure with modified hydrogen bonding (Biscaia et al., 2021).
- Bioavailability Gains: In vivo studies demonstrated a 2.5-fold increase in systemic absorption for BCZ cocrystals compared to the original drug, validating the pharmacokinetic advantages of the cocrystal form.
- Stability and Compatibility: Stability assessments revealed that BCZ cocrystals maintained their enhanced properties under accelerated storage conditions, highlighting their suitability for real-world pharmaceutical applications (Deeksha et al., 2021).
BCZ’s success demonstrates the practical advantages of cocrystals in addressing solubility and stability challenges for poorly water-soluble drugs.
Comparison with Other Advanced Solubility Techniques
While cocrystals offer numerous advantages, it is essential to compare them with other solubility enhancement methods to understand their relative benefits and limitations:
- Nanocrystals:
- Advantages: Nanocrystals significantly increase the surface area, enhancing dissolution rates for poorly soluble drugs. For example, carvedilol nanocrystals achieved a 2000-fold solubility increase (Mohammady et al., 2021).
- Limitations: High-energy processes used for nanocrystal production may compromise drug stability, and the requirement for stabilizers adds complexity.
- Amorphous Solid Dispersions (ASD):
- Advantages: ASDs improve solubility by dispersing drugs in a polymeric matrix. They are widely used for thermolabile compounds (Thakkar et al., 2018).
- Limitations: Prone to recrystallization and physical instability, which can negate solubility gains over time.
- Lipid-Based Formulations:
- Advantages: Lipid-based systems enhance solubility for lipophilic drugs and protect against enzymatic degradation.
- Limitations: Expensive, with a risk of precipitation upon dilution in gastrointestinal fluids (Narala et al., 2022).
- Comparison with Cocrystals:
- Cocrystals combine the structural stability of crystalline forms with the solubility enhancement of amorphous forms, bridging the gap between conventional techniques. Unlike ASDs or nanocrystals, cocrystals retain long-term stability and can be tailored to specific pharmacokinetic profiles through coformer selection (Gyawali et al., 2021).
Cocrystals represent a paradigm shift in pharmaceutical development, offering solutions to longstanding solubility and stability challenges. FDA-approved examples and case studies, such as BCZ, underscore their transformative potential. When compared with other solubility enhancement techniques, cocrystals offer a unique blend of stability, scalability, and bioavailability, making them a cornerstone in the future of drug formulation.
Table-2: Comparison Table: Solubility Enhancement Techniques
Technique | Advantages | Limitations | Examples |
Cocrystals | – Improves solubility and stability | – Requires precise coformer selection | – Entresto® (sacubitril/valsartan) |
– Maintains crystalline stability | – Limited scalability for some techniques | – Suglat® (ipragliflozin with L-proline) | |
– Tailored coformers for specific properties | – BCZ with nicotinamide | ||
– Regulatory acceptance by FDA/EMA | |||
Nanocrystals | – Significantly increases surface area | – High-energy processes may degrade thermolabile drugs | – Carvedilol nanocrystals |
– Rapid dissolution | – Requires stabilizers to prevent aggregation | – Fenofibrate nanocrystals | |
– Applicable for poorly water-soluble drugs | |||
Amorphous Solid Dispersions (ASD) | – Improves solubility by maintaining drugs in amorphous form | – Susceptible to recrystallization | – Erlotinib-ASD using lyophilization |
– Effective for thermolabile drugs | – Requires stabilizing excipients for long-term storage | – Rosuvastatin-ASD | |
Lipid-Based Systems | – Enhances solubility of lipophilic drugs | – High production costs | – Self-nanoemulsifying systems for olanzapine |
– Protects against enzymatic degradation | – Drug precipitation risks in gastrointestinal fluids | – Solid lipid nanoparticles for antifungal agents | |
– Suitable for lymphatic transport | |||
Salt Formation | – Simple and cost-effective | – Limited to ionizable drugs | – Diclofenac sodium |
– Improves solubility for ionizable drugs | – Risk of precipitation in physiological conditions | – Atorvastatin calcium | |
Cyclodextrins | – Forms inclusion complexes for improved solubility | – Limited drug compatibility | – Hydroxypropyl-beta-cyclodextrin with itraconazole |
– Reduces drug volatility | – Expensive and requires high doses for therapeutic use | ||
Supercritical Fluids | – Produces pure and stable cocrystals | – Requires specialized equipment | – Fenofibrate cocrystals via SCF techniques |
– Solvent-free process | – High operational costs | ||
– Scalable |
CHALLENGES AND REGULATORY FRAMEWORK
Intellectual Property and Regulatory Guidelines for Cocrystals
The development of pharmaceutical cocrystals has required adaptations in regulatory frameworks and intellectual property (IP) guidelines to address their unique nature. Cocrystals are distinct from traditional polymorphs, salts, and amorphous forms, necessitating specific classification and standards for patentability and regulatory approval.
- Patent Challenges: Cocrystals often face hurdles in meeting the criteria for patent eligibility. Since they do not involve covalent modification of the active pharmaceutical ingredient (API), proving novelty and inventive steps becomes a critical challenge. Regulatory bodies like the United States Patent and Trademark Office (USPTO) and the European Patent Office (EPO) have been increasingly stringent in granting patents for cocrystals that lack clear evidence of substantial pharmaceutical benefits (Saraf et al., 2022).
- Regulatory Guidelines: In 2018, the U.S. FDA issued detailed guidance on pharmaceutical cocrystals, classifying them as crystalline APIs rather than drug-excipient complexes. This classification has simplified the regulatory path for cocrystals, enabling companies to demonstrate their safety and efficacy based on the parent API’s clinical data. The European Medicines Agency (EMA) has adopted similar guidelines, further harmonizing the global regulatory environment for cocrystals (Shaikh et al., 2018).
Overcoming Barriers to Market Approval
Despite advancements in regulatory clarity, significant barriers remain in achieving market approval for cocrystal-based drugs.
- Complexity of Characterization: Comprehensive characterization of cocrystals is essential to distinguish them from polymorphs or amorphous solids. Techniques such as PXRD, DSC, and solid-state NMR are required to demonstrate the uniqueness of the cocrystal form. However, the high cost and technical complexity of these analyses can delay regulatory submissions (Karimi-Jafari et al., 2018).
- Data Requirements: Regulatory agencies often demand extensive comparative studies to demonstrate the enhanced solubility, stability, and bioavailability of cocrystals over the parent API. This requirement for additional pharmacokinetic and pharmacodynamic studies adds to the development timeline and costs (Bandaru et al., 2021).
- Harmonization Issues: Variability in global regulatory requirements creates significant challenges for pharmaceutical companies looking to commercialize cocrystal-based drugs in multiple markets. While the FDA and EMA have aligned their guidelines, emerging markets often lack specific frameworks, leading to delays in approvals (Pimenta et al., 2021).
Commercialization Challenges and Opportunities
- Manufacturing Complexity: The large-scale production of cocrystals requires precise control of conditions like temperature, solvent usage, and mechanical forces. Technologies such as hot melt extrusion and supercritical fluid methods offer scalable solutions but come with high initial capital costs (Long et al., 2019).
- Cost-Benefit Trade-Off: Pharmaceutical companies often struggle to balance the costs of developing cocrystal-based drugs against their market potential. The need for extensive data and specialized manufacturing processes makes cocrystals less attractive compared to simpler solubility enhancement methods, despite their superior performance.
- Opportunities for Growth:
- Pipeline Expansion: With increasing regulatory clarity, several companies are investing in cocrystal research for APIs with significant commercial potential, such as oncology and central nervous system drugs (Swapnali et al., 2020).
- Intellectual Property Extension: Cocrystals allow companies to extend the patent life of APIs by developing novel crystalline forms, delaying generic competition and maximizing revenue.
- Green Chemistry: The development of solvent-free methods for cocrystal production aligns with industry goals for sustainability, offering opportunities to reduce environmental impact (Roy & Ghosh, 2020).
CONCLUSION
The solubility of pharmaceutical compounds remains a critical challenge in drug development, particularly for BCS Class II and IV drugs, which account for a significant proportion of poorly water-soluble active pharmaceutical ingredients (APIs). These drugs, while often potent and effective, are hindered by their inability to dissolve adequately in aqueous environments, leading to limited bioavailability and therapeutic inefficacy. Conventional solubility enhancement techniques, such as particle size reduction, solid dispersions, and lipid-based systems, have provided incremental improvements but often fall short in terms of scalability, stability, or long-term efficacy.
Cocrystals have emerged as a transformative solution to these challenges, offering a unique blend of enhanced solubility, stability, and bioavailability without altering the molecular structure of the drug. By utilizing crystal engineering to combine the API with a carefully selected coformer, cocrystals optimize drug properties in a way that is both effective and sustainable. The success of FDA-approved drugs like Entresto® and Suglat® demonstrates the regulatory viability of cocrystals, while case studies on BCZ cocrystals underline their practical potential for addressing BCS Class II and IV challenges.
BCZ, as a model compound, exemplifies the ability of cocrystals to significantly enhance solubility and dissolution rates, leading to improved therapeutic outcomes. Beyond solubility, cocrystals contribute to the stability and scalability of drug formulations, making them an essential tool in modern pharmaceutical development. As the industry continues to innovate, the adoption of cocrystal technology is poised to expand, providing a definitive solution to one of the most persistent challenges in drug development.
REFERENCES
- Jain, H., & Chella, N. (2020). Methods to improve the solubility of therapeutical natural products: A review. Environmental Chemistry Letters, 19, 111–121. https://doi.org/10.1007/s10311-020-01082-x
- Dhande, L. B., Deshmukh, M. T., Khopade, A. N., Shete, R. V., & Kunjir, V. V. (2021). A review on solubility enhancement by solid dispersion method. Journal of Drug Delivery and Therapeutics, 11(1), 182–187. https://doi.org/10.22270/jddt.v11i1.4489
- Parbhane, S., Belure, A., Garud, A., & Chatur, V. (2020). Solubility enhancement: Meaning and techniques. International Journal of Pharmaceutical Sciences and Medicine.
- Abuzar, S., Hyun, S. M., Kim, J. H., Park, H., Kim, M. S., Park, J. S., & Hwang, S. (2018). Enhancing the solubility and bioavailability of poorly water-soluble drugs using supercritical antisolvent (SAS) process. International Journal of Pharmaceutics, 538(1–2), 1–13. https://doi.org/10.1016/j.ijpharm.2017.12.041
- Narmada, I. (2023). Contemporary review on solubility enhancement techniques. Journal of Drug Delivery and Therapeutics.
- Patel, M., Ahmed, M., Saqib, M., & Shaikh, S. N. (2019). Chemical modification: A unique solutions to solubility problem. Journal of Drug Delivery and Therapeutics. https://doi.org/10.22270/jddt.v9i2.2432
- Safaei, S., & Varshosaz, J. (2018). Enhancement of drug solubility: Review abstract articles. International Journal of Pharmaceutical Sciences and Medicine, 1(1), 93–94.
- Deokate, S. C., Deokate, A. A., Sangale, S. B., & Gawade, T. P. (2023). Drug solubility: Importance and enhancement techniques. International Journal for Multidisciplinary Research.
- Kumar, R., Thakur, A., Chaudhari, P., & Banerjee, N. (2021). Particle size reduction techniques of pharmaceutical compounds for the enhancement of their dissolution rate and bioavailability. Journal of Pharmaceutical Innovation, 17, 333–352. https://doi.org/10.1007/s12247-020-09530-5
- Csicsák, D., Szolláth, R., Kádár, S., Ambrus, R., Bartos, C., Balogh, E., … & Sinkó, B. (2023). The effect of particle size reduction on the biorelevant solubility and dissolution of poorly soluble drugs. Pharmaceutics, 15(1), 278.
- Kumar, R., Thakur, A., Chaudhari, P., & Banerjee, N. (2021). Particle size reduction techniques for the enhancement of dissolution rate and bioavailability. Journal of Pharmaceutical Innovation, 17(3), 333–352.
- Zhang, X., Xing, H., Zhao, Y., & Ma, Z. (2018). Pharmaceutical dispersion techniques for dissolution and bioavailability enhancement of poorly water-soluble drugs. Pharmaceutics, 10(3), 74.
- Kim, N., Oh, H., Lee, J. C., Choi, Y., & Jeong, S. (2020). Comparison of solubility enhancement by solid dispersion and micronization. Journal of Pharmaceutical Investigation, 51(1), 53–60.
- Yen, C. W., Kuhn, R., Hu, C., Zhang, W., Chiang, P. C., Chen, J. Z., … & Leung, D. H. (2021). Impact of surfactant selection on in situ nanoparticle formation from amorphous solid dispersions. International Journal of Pharmaceutics, 120980.
- Krstic, M., Medarević, Đ., Đuriš, J., & Ibrić, S. (2018). Self-nanoemulsifying drug delivery systems (SNEDDS) for improving dissolution and bioavailability. Comprehensive Review.
- Jawahar, N., Hingarh, P. K., Arun, R., Selvaraj, J., Anbarasan, A., Sathianarayanan, S., & G. N. (2018). Enhanced oral bioavailability of an antipsychotic drug through nanostructured lipid carriers. International Journal of Biological Macromolecules, 110(1), 269–275.
- Chen, Y., Lubach, J., Tang, S., & Narang, A. (2021). Effect of counterions on dissolution of amorphous solid dispersions. Molecular Pharmaceutics, 18(2), 1089–1100.
- Abdelkader, H., & Fathalla, Z. (2018). Investigation into the emerging role of the basic amino acid L-lysine in enhancing solubility and permeability of BCS Class II and BCS Class IV drugs. Pharmaceutical Research, 35, 1–18.
- Alsubaie, M., Aljohani, M., Erxleben, A., & McArdle, P. (2018). Co-crystal forms of the BCS class IV drug sulfamethoxazole. Crystal Growth & Design, 18, 3902–3912.
- Bommaka, M. K., Mannava, M., Suresh, K., Gunnam, A., & Nangia, A. (2018). Entacapone: Improving aqueous solubility, diffusion permeability, and cocrystal stability with theophylline. Crystal Growth & Design.
- Eesam, S., Bhandaru, J. S., Naliganti, C., Bobbala, R., & Akkinepally, R. (2020). Solubility enhancement of carvedilol using drug-drug cocrystallization with hydrochlorothiazide. Future Journal of Pharmaceutical Sciences, 6.
- Hiendrawan, S., Veriansyah, B., & Tjandrawinata, R. (2018). Solid-state properties and solubility studies of novel pharmaceutical cocrystal of itraconazole. International Journal of Applied Pharmaceutics.
- Nikam, V. J., & Patil, S. (2020). Pharmaceutical cocrystals of nebivolol hydrochloride with enhanced solubility. Journal of Crystal Growth, 534, 125488.
- Sathisaran, I., & Dalvi, S. (2018). Engineering cocrystals of poorly water-soluble drugs to enhance dissolution in aqueous medium. Pharmaceutics, 10(3), 108.
- Hiendrawan, S., Veriansyah, B., & Tjandrawinata, R. (2018). Solid-state properties and solubility studies of novel pharmaceutical cocrystal of itraconazole. International Journal of Applied Pharmaceutics. DOI: 10.22159/IJAP.2018V10I5.26663
- Huang, D., Chan, H., Wu, Y., Li, L., Zhang, L., Lv, Y., Yang, X., Zhou, Z. (2021). Phase solubility investigation and theoretical calculations on drug-drug cocrystals of carbamazepine with Emodin, Paeonol. Journal of Molecular Liquids, 329, 115604. DOI: 10.1016/J.MOLLIQ.2021.115604
- Barikah, K. Z. (2018). Traditional and novel methods for cocrystal formation: A mini review. Systematic Reviews in Pharmacy. https://doi.org/10.5530/SRP.2018.1.15
- Buddhadev, S. S., & Garala, K. (2021). Pharmaceutical cocrystals—A review. Proceedings. https://doi.org/10.3390/PROCEEDINGS2020062014
- Solares-Briones, M., Coyote-Dotor, G., Páez-Franco, J. C., Zermeño-Ortega, M. R., de la O Contreras, C. M., Canseco-González, D., Avila-Sorrosa, A., Morales‐Morales, D., & Germán-Acacio, J. M. (2021). Mechanochemistry: A green approach in the preparation of pharmaceutical cocrystals. Pharmaceutics, 13(6). https://doi.org/10.3390/pharmaceutics13060790
- Narala, S., Nyavanandi, D., Alzahrani, A., Bandari, S., Zhang, F., & Repka, M. (2022). Creation of hydrochlorothiazide pharmaceutical cocrystals via hot-melt extrusion for enhanced solubility and permeability. AAPS PharmSciTech, 23. https://doi.org/10.1208/s12249-021-02202-8
- Butreddy, A., Sarabu, S., Bandari, S., Dumpa, N., Zhang, F., & Repka, M. (2020). Polymer-assisted aripiprazole-adipic acid cocrystals produced by hot melt extrusion techniques. Crystal Growth & Design, 20(7), 4335-4345. https://doi.org/10.1021/acs.cgd.0c00020
- Akbari, Z., Amanlou, M., Karimi-Sabet, J., Golestani, A., & Niassar, M. S. (2020). Application of supercritical fluid technology for preparation of drug loaded solid lipid nanoparticles. International Journal of NanoScience and Nanotechnology, 16, 13–33.
- Barmpalexis, P., Karagianni, A., Nikolakakis, I., & Kachrimanis, K. (2018). Preparation of pharmaceutical cocrystal formulations via melt mixing technique: A thermodynamic perspective. European Journal of Pharmaceutics and Biopharmaceutics, 131, 130–140. https://doi.org/10.1016/j.ejpb.2018.08.002
- Dutt, B., Choudhary, M., & Budhwar, V. (2022). Enhancement of stability profile of aspirin through cocrystallization technique. Research Journal of Pharmacy and Technology.
- Panzade, P., & Shendarkar, G. (2018). Superior solubility and dissolution of zaltoprofen via pharmaceutical cocrystal. Turkish Journal of Pharmaceutical Sciences.
- Dutt, B., Choudhary, M., & Budhwar, V. (2021). Preparation, characterization, and evaluation of fenofibrate: Benzoic acid cocrystals with enhanced pharmaceutical properties. Future Journal of Pharmaceutical Sciences.
- Vemuri, V. D., & Lankalapalli, S. (2021). Cocrystal construction between rosuvastatin calcium and L-asparagine with enhanced solubility and dissolution rate. Turkish Journal of Pharmaceutical Sciences.
- Holanda, B., Alarcon, R., Gaglieri, C., Souza, A. R., Castro, R., Rosa, P., Tangerino, D. J. A., & Bannach, G. (2018). Thermal studies, degradation kinetic, equilibrium solubility, DFT, MIR, and XRPD analyses of a new cocrystal of gemfibrozil and isonicotinamide. Journal of Thermal Analysis and Calorimetry.
- Biscaia, I., Gomes, S. N., Bernardi, L., & Oliveira, P. R. (2021). Obtaining cocrystals by reaction crystallization method: Pharmaceutical applications. Pharmaceutics, 13(6). https://doi.org/10.3390/pharmaceutics13060898
- Deeksha, M., Kumar, A., & Nanda, A. (2021). Recent advancements in pharmaceutical cocrystals, preparation methods, and their applications. Current Pharmaceutical Design. https://doi.org/10.2174/1381612827666210415104411
- Gyawali, R., Aryal, S., Regmi, Y., & Rajarajan, S. (2021). Cocrystallization: An approach to improve bioavailability by altering physicochemical properties of poorly soluble APIs. International Journal of Pharmacy and Pharmaceutical Research, 20(4), 381–397. https://doi.org/10.25166/IJPPR.2021.V20I04.28
- Kavanagh, O. N., Croker, D., Walker, G., & Zaworotko, M. J. (2019). Pharmaceutical cocrystals: From serendipity to design to application. Drug Discovery Today, 24(3), 796–804. https://doi.org/10.1016/j.drudis.2018.11.023
- Kumari, N., & Ghosh, A. (2020). Cocrystallization: A cutting-edge tool for physicochemical modulation of active pharmaceutical ingredients. Current Pharmaceutical Design. https://doi.org/10.2174/1381612826666200720114638
- Mohammady, M., Hadidi, M., Ghetmiri, S. I., & Yousefi, G. (2021). Design of ultra-fine carvedilol nanococrystals: Development of a safe and stable injectable formulation. European Journal of Pharmaceutics and Biopharmaceutics. https://doi.org/10.1016/j.ejpb.2021.08.015
- Narala, S., Nyavanandi, D., Alzahrani, A., Bandari, S., Zhang, F., & Repka, M. (2022). Creation of hydrochlorothiazide pharmaceutical cocrystals via hot-melt extrusion for enhanced solubility and permeability. AAPS PharmSciTech, 23. https://doi.org/10.1208/s12249-021-02202-8
- Thakkar, S., Sharma, D., & Misra, M. (2018). Comparative evaluation of electrospraying and lyophilization techniques on solid-state properties of erlotinib nanocrystals. European Journal of Pharmaceutical Sciences, 111, 257–269. https://doi.org/10.1016/j.ejps.2017.10.008
- Bandaru, R. K., Rout, S. R., Kenguva, G., Gorain, B., Alhakamy, N., Kesharwani, P., & Dandela, R. (2021). Recent advances in pharmaceutical cocrystals: From bench to market. Frontiers in Pharmacology, 12. https://doi.org/10.3389/fphar.2021.780582
- Karimi-Jafari, M., Padrela, L., Walker, G., & Croker, D. (2018). Creating cocrystals: A review of pharmaceutical cocrystal preparation routes and applications. Crystal Growth & Design. https://doi.org/10.1021/acs.cgd.8b00933
- Saraf, G. J., Burade, K., Gonjari, I., Hosmani, A., & Pawar, A. A. (2022). A review on advances in pharmaceutical cocrystal preparation routes, intellectual property perspective, and regulatory aspects. International Journal of Current Pharmaceutical Research, 14(5), 1–12. https://doi.org/10.22159/ijcpr.2022v14i5.2038
- Shaikh, R., Singh, R., Walker, G., & Croker, D. (2018). Pharmaceutical cocrystal drug products: An outlook on product development. Trends in Pharmacological Sciences, 39(12), 1033–1048. https://doi.org/10.1016/j.tips.2018.10.006
- Long, B., Ryan, K., & Padrela, L. (2019). From batch to continuous: New opportunities for supercritical CO2 technology in pharmaceutical manufacturing. European Journal of
- Pharmaceutical Sciences. https://doi.org/10.1016/j.ejps.2019.104971
- Swapnali, S., Harshal, S., & Vaishali, D. (2020). A review on pharmaceutical cocrystallization. Zenodo. https://doi.org/10.5281/zenodo.4088823
- Roy, P., & Ghosh, A. (2020). Progress on cocrystallization of poorly soluble NMEs in the last decade. CrystEngComm, 22(41), 6958–6974. https://doi.org/10.1039/d0ce01276a
- Pimenta, C., Bettiol, V., Alencar-Silva, T., Franco, O., Pogue, R., Carvalho, J. L., & Felipe, M. (2021). Advanced therapies and regulatory framework in different areas of the globe: Past, present, and future. Clinical Therapeutics. https://doi.org/10.1016/j.clinthera.2021.02.006
- Azleena A, & Dr. Yogesh Kumar. (2023). Ocimum sanctum Seed Nanoemulsion: In Vitro MTT and Anti-Inflammatory Assay. PEXACY International Journal of Pharmaceutical Science, 2(7). https://doi.org/10.5281/zenodo.8166568