Available online on 15.08.2024 at http://jddtonline.info

Journal of Drug Delivery and Therapeutics

Open Access to Pharmaceutical and Medical Research

Copyright  © 2024 The  Author(s): This is an open-access article distributed under the terms of the CC BY-NC 4.0 which permits unrestricted use, distribution, and reproduction in any medium for non-commercial use provided the original author and source are credited

Open Access   Full Text Article                                                                                                                                                              Review Article

Enhancing Dissolution and Bioavailability: A Review on Co-Processed Superdisintegrants in Pharmaceutical Formulations

Shubham Sachdeva 1*, Harpreet Singh 2, Jitender Singh 1

Department of Pharmaceutics, Lord Shiva College of Pharmacy, Sirsa (Haryana)-125055, India

School of Pharmacy, RIMT University, Mandi Gobindgarh, (Punjab) – 147301, India

Article Info:

___________________________________________

Article History:

Received 28 May 2024  

Reviewed 03 July 2024  

Accepted 27 July 2024  

Published 15 August 2024  

___________________________________________

Cite this article as: 

Sachdeva S, Singh H, Singh J, Enhancing Dissolution and Bioavailability: A Review on Co-Processed Superdisintegrants in Pharmaceutical Formulations, Journal of Drug Delivery and Therapeutics. 2024; 14(8):223-237

DOI: http://dx.doi.org/10.22270/jddt.v14i8.6747                     ___________________________________________

*Address for Correspondence:  

Mr. Shubham Sachdeva Asstt. Prof., Department of Pharmaceutics, Lord Shiva College of Pharmacy, Sirsa (Haryana)-125055

Abstract

___________________________________________________________________________________________________________________

Co-processed superdisintegrants have emerged as key excipients in pharmaceutical formulation development, offering solutions to challenges related to poor solubility, bioavailability, and patient compliance. This comprehensive review article provides an in-depth analysis of the principles, mechanisms, manufacturing techniques, and applications of co-processed superdisintegrants in drug delivery systems. The review highlights the role of co-processed excipients in enhancing dissolution kinetics, improving formulation efficiency, and enabling innovative drug delivery platforms such as personalized medicine and combination therapies. Regulatory considerations, quality standards, and future directions for research and innovation in this field are also discussed. Through a synthesis of current literature and insights into emerging trends, this review aims to provide researchers, formulation scientists, and pharmaceutical professionals with a comprehensive understanding of the potential and challenges associated with co-processed superdisintegrants in pharmaceutical formulations. Ultimately, the integration of co-processed excipients into formulation development holds promise for advancing drug delivery technology, improving therapeutic outcomes, and addressing unmet medical needs in patient care.

Keywords: Co-processed superdisintegrants, Dissolution enhancement, Bioavailability, Pharmaceutical formulations, Drug delivery systems, Excipients, Formulation optimization, Manufacturing techniques, Regulatory considerations, Future directions

 


 
  1. Introduction

In the realm of pharmaceutical formulation, the challenge of ensuring optimal drug delivery and bioavailability is paramount. One of the critical factors influencing these aspects is the dissolution rate of the active pharmaceutical ingredient (API). Poorly water-soluble drugs often face hurdles in achieving rapid dissolution, which can subsequently lead to suboptimal therapeutic outcomes.

To address this challenge, pharmaceutical scientists have been exploring various strategies, one of which involves the use of superdisintegrants. These excipients aid in the rapid disintegration of solid dosage forms, thereby facilitating the release and dissolution of the API. Traditional superdisintegrants such as croscarmellose sodium, crospovidone, and sodium starch glycolate have been widely employed to enhance dissolution kinetics1.

However, the quest for more effective solutions has led to the emergence of co-processed superdisintegrants. Unlike conventional superdisintegrants, co-processed superdisintegrants are blends of two or more excipients that are specifically engineered to synergistically enhance the disintegration and dissolution properties of solid dosage forms. This innovative approach capitalizes on the complementary characteristics of individual excipients, resulting in superior performance compared to their single-component counterparts 2.

The concept of co-processing involves the careful selection and optimization of excipient combinations, guided by a thorough understanding of their physicochemical properties and interactions. By leveraging the unique attributes of different excipients, such as their swelling, wicking, and solubilizing capabilities, co-processed superdisintegrants can effectively address the complex challenges associated with poor drug solubility and dissolution 3.

The manufacturing of co-processed superdisintegrants typically involves advanced techniques such as spray drying, co-grinding, or co-precipitation, which enable precise control over the composition and morphology of the final product. These manufacturing processes play a crucial role in determining the physical characteristics and performance attributes of co-processed superdisintegrants, including particle size, morphology, flow properties, and compressibility4.

In recent years, co-processed superdisintegrants have gained considerable attention in the pharmaceutical industry due to their potential to revolutionize drug formulation development. By improving the dissolution rate and bioavailability of poorly water-soluble drugs, co-processed superdisintegrants offer a promising avenue for enhancing the efficacy and patient compliance of pharmaceutical products.

In this review article, we delve into the principles, manufacturing techniques, advantages, and applications of co-processed superdisintegrants in pharmaceutical formulations. Through a comprehensive exploration of this innovative approach, we aim to provide insights into the transformative potential of co-processed excipients in overcoming formulation challenges and advancing drug delivery science.

  1. Challenges Related to Poor Solubility and Bioavailability of Drugs

The solubility and bioavailability of drugs are critical factors that significantly influence their therapeutic efficacy. However, a considerable number of drug candidates in the pharmaceutical pipeline exhibit poor solubility, which poses significant challenges in their formulation and delivery. Here, we provide an overview of the challenges associated with poor solubility and bioavailability of drugs, supported by relevant references.

Poor Solubility:

Many drug candidates have low aqueous solubility, which leads to insufficient dissolution in the gastrointestinal tract and limits their absorption into the systemic circulation5.

Poorly soluble drugs often require high doses to achieve therapeutic concentrations, increasing the risk of adverse effects and patient non-compliance6.

The Biopharmaceutics Classification System (BCS) categorizes drugs based on their solubility and permeability, highlighting the prevalence of poorly soluble compounds, especially in Class II (low solubility, high permeability) and Class IV (low solubility, low permeability)7.

Limited Bioavailability:

Poorly soluble drugs face challenges in achieving adequate bioavailability due to incomplete dissolution, erratic absorption, and first-pass metabolism8.

Inadequate bioavailability can lead to suboptimal therapeutic outcomes, necessitating higher doses or more frequent administration to maintain efficacy 9.

Factors such as particle size, polymorphism, and formulation characteristics influence the dissolution rate and bioavailability of poorly soluble drugs 10.

Formulation Challenges:

Formulating poorly soluble drugs into effective dosage forms presents challenges such as poor content uniformity, variable dissolution profiles, and reduced stability 11.

Conventional approaches to enhance solubility, such as micronization, complexation, and solid dispersion, may not always yield satisfactory results and may be associated with manufacturing complexities 12.

Overcoming the solubility limitations while maintaining formulation robustness and patient acceptability requires innovative strategies and advanced formulation technologies13.

  1. Role of Superdisintegrants in Enhancing Dissolution and Bioavailability

Superdisintegrants play a crucial role in pharmaceutical formulations by promoting the rapid disintegration of solid dosage forms, which in turn enhances dissolution and ultimately improves the bioavailability of poorly soluble drugs. Here, we explore the multifaceted role of superdisintegrants in enhancing dissolution and bioavailability, supported by relevant references.

Facilitating Disintegration:

Superdisintegrants are hydrophilic excipients that rapidly absorb water and swell, leading to the disruption of tablet matrices or compacts and facilitating disintegration into smaller particles14.

Enhanced disintegration ensures rapid exposure of the drug surface to the dissolution medium, promoting faster drug release and improving dissolution kinetics15.

Superdisintegrants such as croscarmellose sodium, crospovidone, and sodium starch glycolate are commonly used in oral solid dosage forms to achieve rapid disintegration and dissolution16.

Improving Dissolution Rate:

The ability of superdisintegrants to rapidly disintegrate dosage forms results in increased surface area available for dissolution, which accelerates drug release from the formulation 17.

Enhanced dissolution rates are particularly beneficial for poorly soluble drugs, as they overcome the rate-limiting step of dissolution and ensure efficient absorption in the gastrointestinal tract18.

Superdisintegrants can also promote uniform dispersion of drug particles in the dissolution medium, further enhancing dissolution efficiency and bioavailability19.

Enhancing Bioavailability:

By improving dissolution kinetics, superdisintegrants contribute to higher drug concentrations in the systemic circulation, leading to improved bioavailability and therapeutic efficacy20.

Rapid dissolution and absorption of poorly soluble drugs minimize the variability in plasma drug concentrations, ensuring consistent pharmacological effects and reducing the risk of under- or over-dosing21.

Enhanced bioavailability achieved through the use of superdisintegrants can translate into lower dosages, reduced dosing frequency, and improved patient compliance22.

Formulation Flexibility:

Superdisintegrants offer flexibility in formulation design, allowing for the development of various dosage forms such as immediate-release tablets, orally disintegrating tablets, and fast-dissolving films23.

The compatibility of superdisintegrants with different drug substances and excipients enables their incorporation into diverse pharmaceutical formulations, catering to the specific needs of patients and drug products24.

Superdisintegrants also contribute to the manufacturability and stability of solid dosage forms, ensuring robustness and reliability throughout the product lifecycle25.

  1. Introduction to Co-Processed Superdisintegrants as a Novel Approach

In the field of pharmaceutical formulation, the development of effective drug delivery systems for poorly soluble drugs remains a significant challenge. The traditional approach of using single-component superdisintegrants has shown limitations in achieving optimal dissolution and bioavailability enhancement. In response to these challenges, a novel approach has emerged: co-processed superdisintegrants.

Co-processed superdisintegrants represent a unique advancement in pharmaceutical excipient technology. Unlike conventional single-component superdisintegrants, co-processed superdisintegrants are blends of two or more excipients carefully selected and engineered to work synergistically. This synergistic combination aims to maximize the advantages of individual components while overcoming their limitations, thereby offering superior performance in terms of enhancing dissolution and bioavailability of poorly soluble drugs26.

The rationale behind co-processing lies in harnessing the complementary properties of different excipients to achieve enhanced disintegration and dissolution kinetics. By combining excipients with distinct mechanisms of action, such as swelling, wicking, and solubilization, co-processed superdisintegrants can address various formulation challenges associated with poor drug solubility and absorption27.

The manufacturing process of co-processed superdisintegrants involves advanced techniques such as spray drying, co-grinding, or co-precipitation. These techniques allow for precise control over the composition and morphology of the final product, ensuring optimized performance characteristics. Through systematic optimization and characterization, co-processed superdisintegrants can be tailored to meet specific formulation requirements, thereby offering a versatile and customizable approach to drug delivery system development28.

The introduction of co-processed superdisintegrants represents a paradigm shift in the field of pharmaceutical excipient design. By capitalizing on the synergistic effects of multiple excipients, this novel approach offers the potential to overcome the limitations of traditional single-component superdisintegrants and unlock new possibilities in drug formulation optimization. As such, co-processed superdisintegrants hold promise as a valuable tool for enhancing the dissolution and bioavailability of poorly soluble drugs, ultimately leading to improved therapeutic outcomes and patient compliance.

  1. Principles of Co-Processing:

Co-processing involves the strategic combination of two or more excipients to create a synergistic effect that enhances the performance of the resulting formulation. This section explores the fundamental principles underlying the co-processing of excipients in pharmaceutical formulations.

Selection of Excipients:

The first step in co-processing is the careful selection of excipients based on their individual properties and functionalities. Excipients with complementary characteristics are chosen to maximize the synergistic effects and address specific formulation challenges29.

Excipients may be selected based on their ability to promote disintegration, enhance dissolution, improve flow properties, or provide stability to the formulation. Common excipients used in co-processing include superdisintegrants, binders, diluents, and lubricants30.

Understanding Interactions:

Co-processing involves a thorough understanding of the interactions between different excipients in the formulation. Interactions may occur at the molecular, particle, or bulk level and can influence the physical and chemical properties of the final product31.

Compatibility studies are conducted to assess the compatibility between excipients and active pharmaceutical ingredients (APIs) and to identify any potential interactions that may affect the stability or performance of the formulation32.

Optimization of Composition:

The composition of co-processed excipients is optimized to achieve the desired performance characteristics while maintaining formulation robustness and stability. Various factors, including excipient ratios, processing conditions, and manufacturing techniques, are optimized to achieve the desired formulation attributes33.

Design of experiments (DOE) and quality-by-design (QbD) approaches are commonly employed to systematically optimize the composition and manufacturing parameters of co-processed excipients34.

Manufacturing Techniques:

Co-processing can be achieved using a variety of manufacturing techniques, including spray drying, co-grinding, co-precipitation, and melt extrusion. Each technique offers advantages in terms of scalability, reproducibility, and control over the physical characteristics of the final product35.

The selection of a suitable manufacturing technique depends on the specific properties of the excipients, the desired characteristics of the final formulation, and the intended route of administration.

Characterization and Evaluation:

Co-processed excipients are characterized and evaluated to ensure that they meet the desired performance criteria and regulatory requirements. Physicochemical properties such as particle size, morphology, flowability, and compressibility are evaluated using various analytical techniques36.

In vitro and in vivo studies are conducted to assess the performance of co-processed formulations in terms of disintegration, dissolution, bioavailability, and pharmacokinetics.

  1. Definition and Concept of Co-Processed Excipients

Co-processed excipients are specialized formulations in the field of pharmaceutical sciences. They are created through the strategic combination of two or more individual excipients into a single entity, aimed at achieving enhanced performance or functionality compared to their individual components. This concept involves the blending and processing of excipients to create synergistic effects that address specific formulation challenges and optimize the performance of pharmaceutical dosage forms.

The formulation of co-processed excipients is based on the principle of combining excipients with complementary properties or functionalities. These excipients may possess distinct characteristics such as disintegration enhancement, dissolution enhancement, flow improvement, compression properties, or stability enhancement. By combining excipients with different functionalities, co-processed excipients can overcome limitations associated with individual excipients and offer superior performance in pharmaceutical formulations37.

The manufacturing process of co-processed excipients typically involves advanced techniques such as spray drying, co-grinding, co-precipitation, or melt extrusion. These techniques enable the excipients to be intimately mixed and processed into a homogeneous blend, ensuring uniform distribution of the individual components and the formation of a synergistic matrix structure. The resulting co-processed excipients exhibit enhanced properties that are tailored to meet specific formulation requirements, such as improved flowability, compressibility, disintegration, or dissolution rates38.

Co-processed excipients find applications across a wide range of pharmaceutical dosage forms, including tablets, capsules, granules, powders, and multiparticulate systems. They are utilized to optimize formulation performance, enhance drug stability, facilitate manufacturing processes, and improve patient acceptability. By offering versatility, reliability, and customization capabilities, co-processed excipients have become indispensable tools in pharmaceutical formulation development, enabling the design of optimized drug delivery systems with enhanced therapeutic efficacy and patient compliance39.

  1. Mechanisms of Action for Improving Dissolution and Disintegration

Improving dissolution and disintegration are critical objectives in pharmaceutical formulation development, especially for poorly soluble drugs. Various mechanisms of action are employed by excipients, including superdisintegrants and co-processed excipients, to enhance dissolution and disintegration rates. Below are some key mechanisms:

  1. Swelling: Many superdisintegrants, such as croscarmellose sodium and crospovidone, exhibit swelling properties when exposed to aqueous environments. Upon contact with water, these excipients rapidly absorb moisture, leading to swelling and subsequent disruption of the tablet matrix or compact. This swelling action creates channels and fissures within the dosage form, facilitating the ingress of dissolution medium and promoting rapid disintegration40.
  2. Wicking: Wicking refers to the capillary action by which liquids are drawn into porous materials. Superdisintegrants with wicking properties, such as sodium starch glycolate, effectively absorb water and draw it into the interior of the dosage form. This action helps in breaking the interparticulate bonds and promoting particle dispersion, thereby accelerating disintegration and dissolution41.
  3. Hydrophilic Interaction: Hydrophilic excipients interact with water molecules, promoting the penetration of water into the dosage form and facilitating the solvation and dissolution of the drug substance. Superdisintegrants with high hydrophilicity enhance the wettability and dispersibility of the drug particles, leading to improved dissolution rates42.
  4. Particle Size Reduction: Co-processed excipients may undergo particle size reduction during the manufacturing process, resulting in smaller particle sizes with increased surface area. This increased surface area enhances the interaction between the excipient and the dissolution medium, promoting faster dissolution of the drug substance43.
  5. Porosity Enhancement: Co-processed excipients may exhibit increased porosity due to the formation of void spaces or pores within the matrix. These pores serve as reservoirs for the dissolution medium, facilitating its penetration into the dosage form and promoting rapid disintegration and dissolution44.
  6. Synergistic Effects: Co-processed excipients leverage the synergistic effects of multiple components to enhance dissolution and disintegration. By combining excipients with complementary mechanisms of action, such as swelling, wicking, and solubilization, co-processed excipients achieve superior performance compared to single-component excipients45.
    1. Factors Influencing the Selection and Optimization of Co-Processed Superdisintegrants

The selection and optimization of co-processed superdisintegrants involve careful consideration of various factors to ensure the development of effective and robust pharmaceutical formulations. Several key factors influence this process:

  1. Formulation Requirements: The specific formulation requirements, such as dosage form, drug properties, release profile, and intended route of administration, play a crucial role in selecting co-processed superdisintegrants. Different dosage forms may require excipients with specific properties, such as high compressibility for tablets or rapid hydration for orally disintegrating tablets46.
  2. Mechanism of Action: Understanding the mechanism of action of co-processed superdisintegrants is essential for their selection and optimization. Different excipients may exhibit distinct mechanisms, such as swelling, wicking, or solubilization. The choice of co-processed superdisintegrants should align with the desired mechanism of action to achieve the desired dissolution and disintegration profiles47.
  3. Compatibility with Drug Substance: Co-processed superdisintegrants must be compatible with the drug substance to ensure stability and efficacy. Compatibility studies, including physical and chemical compatibility assessments, are essential to identify any potential interactions between the excipients and the drug substance48.
  4. Processability: The processability of co-processed superdisintegrants is another critical factor to consider. Excipients should be easily processable using common manufacturing techniques such as direct compression, wet granulation, or dry granulation. The choice of manufacturing method should be compatible with the selected excipients to ensure efficient and reproducible production49.
  5. Quality Attributes: Various quality attributes, including particle size distribution, bulk density, flow properties, and compressibility, influence the performance of co-processed superdisintegrants. Optimization of these attributes is essential to achieve uniformity, stability, and optimal performance of the final dosage form [50].
  6. Regulatory Considerations: Compliance with regulatory requirements is paramount in the selection and optimization of co-processed superdisintegrants. Excipients must meet pharmacopeial standards and regulatory guidelines regarding safety, purity, and quality. Documentation of excipient specifications, manufacturing processes, and quality control measures is necessary for regulatory approval51.
  7. Cost and Availability: Cost-effectiveness and availability of co-processed superdisintegrants are practical considerations in formulation development. Excipients should be economically viable and readily available from reputable suppliers. Cost-benefit analysis should be conducted to evaluate the overall impact on formulation development and manufacturing costs 52.
  8. Patent Landscape: Consideration of existing patents and intellectual property rights is important when selecting co-processed superdisintegrants. Novel excipients or proprietary formulations may offer competitive advantages but may also be subject to licensing agreements or restrictions. Conducting a thorough patent search helps identify opportunities and limitations in excipient selection53.
  9. Manufacturing Techniques:

Manufacturing techniques play a crucial role in the production of co-processed excipients, including co-processed superdisintegrants. These techniques involve processes that blend and process the individual excipients to create a synergistic blend with enhanced properties. Here are some common manufacturing techniques used for co-processed excipients:

  1. Spray Drying: Spray drying is a widely used technique for producing co-processed excipients. In this method, a solution or suspension containing the excipients is atomized into fine droplets, which are then dried using hot air. The resulting dried particles are collected as a powder. Spray drying enables the production of uniform particles with controlled morphology and particle size distribution. This technique is particularly suitable for producing co-processed excipients with enhanced flowability, compressibility, and dissolution characteristics54.
  2. Co-Grinding: Co-grinding involves the mechanical blending of excipients using milling or grinding equipment. The excipients are milled together to achieve intimate mixing and homogenization. Co-grinding is a simple and cost-effective method for producing co-processed excipients. It is suitable for excipients with similar particle sizes and mechanical properties. Co-grinding can enhance the surface area and porosity of the excipient blend, leading to improved dissolution and disintegration properties55.
  3. Co-Precipitation: Co-precipitation involves the simultaneous precipitation of multiple excipients from a solvent or solvent mixture. The excipients are dissolved or suspended in a solvent, followed by the addition of a precipitating agent to induce the formation of solid particles. Co-precipitation allows for the synthesis of co-processed excipients with controlled particle size, morphology, and composition. This technique is particularly useful for producing co-processed excipients with enhanced solubility, stability, and bioavailability56.
  4. Melt Extrusion: Melt extrusion is a thermal processing technique used to melt and mix excipients at elevated temperatures. The excipients are fed into an extruder, where they are heated and mixed under controlled conditions. The molten mass is then extruded through a die to form strands or pellets, which are subsequently cooled and solidified. Melt extrusion enables the production of co-processed excipients with improved compatibility, solubility, and release characteristics. This technique is suitable for heat-stable excipients and thermolabile drugs57.
  5. Co-Spray Granulation: Co-spray granulation combines spray drying with granulation techniques to produce co-processed excipients in granular form. In this method, excipients are spray-dried to form fine particles, which are then agglomerated into granules using a binder solution. The granules are dried to remove excess moisture and improve flow properties. Co-spray granulation allows for the production of co-processed excipients with controlled particle size, porosity, and compressibility. It is suitable for producing granular formulations with enhanced flowability and compressibility58.
  6. Hot Melt Extrusion (HME): Hot melt extrusion involves the mixing and melting of excipients at elevated temperatures followed by extrusion through a die to form a uniform matrix. HME is particularly suitable for co-processed excipients that require controlled release or improved solubility. This technique enables the production of solid dispersions, matrix tablets, and controlled-release formulations with enhanced bioavailability and stability59.
    1. Overview of Common Manufacturing Techniques: Spray Drying, Co-Grinding, and Co-Precipitation

Manufacturing techniques such as spray drying, co-grinding, and co-precipitation are commonly employed for the production of co-processed excipients, including co-processed superdisintegrants. Here's an overview of these techniques along with references:

  1. Spray Drying:
  2. Principle: Spray drying involves atomizing a liquid feed material into fine droplets, which are then dried by hot air or inert gas, resulting in the formation of dry particles.
  3. Process: Liquid feed material containing excipients dissolved or suspended in a solvent is atomized into droplets using a spray nozzle. These droplets are then dried in a drying chamber by hot air or gas, leading to the formation of dry particles.
  4. Advantages: Spray drying allows for the production of uniform particles with controlled morphology and size distribution. It is suitable for heat-sensitive materials and can be scaled up for large-scale production.
  5. Applications: Spray-dried co-processed excipients are used in tablet and capsule formulations, controlled-release systems, and inhalation formulations60.
  6. Co-Grinding:

The manufacturing process employed for the production of co-processed superdisintegrants has a significant impact on their physicochemical properties, which in turn influence their performance in pharmaceutical formulations. Here are some key aspects of how the manufacturing process affects the physicochemical properties of co-processed superdisintegrants:

  1. Particle Size and Morphology: 

Achieving scalability and reproducibility in the manufacturing of co-processed superdisintegrants is essential to ensure consistent quality and performance of the final pharmaceutical formulations. Several considerations must be taken into account to address these aspects effectively:

  1. Process Optimization: The manufacturing process for co-processed superdisintegrants should be thoroughly optimized to ensure reproducible results at different scales of production. Parameters such as temperature, pressure, solvent composition, mixing time, and drying conditions should be carefully controlled and validated to achieve consistent product quality63.
  2. Scale-Up Studies: Before scaling up production, comprehensive scale-up studies should be conducted to evaluate the effects of increasing batch sizes on product quality and process performance. These studies help identify potential challenges and optimize process parameters to maintain product consistency across different production scales63.
  3. Equipment Selection and Validation: Selection of appropriate manufacturing equipment is critical for achieving scalability and reproducibility. Equipment should be capable of handling larger batch sizes without compromising product quality. Additionally, equipment should be properly validated to ensure compliance with regulatory requirements and adherence to Good Manufacturing Practices (GMP)64.
  4. Raw Material Characterization: Raw materials used in the manufacturing of co-processed superdisintegrants should be thoroughly characterized to ensure consistency and quality. This includes assessing the physical and chemical properties of excipients, as well as conducting compatibility studies with other formulation components and drug substances65.
  5. Process Monitoring and Control: Robust process monitoring and control systems should be implemented to detect and correct deviations during manufacturing. Real-time monitoring of critical process parameters allows for immediate adjustments to maintain product quality and consistency. Automated control systems can help minimize human error and ensure reproducibility65.
  6. Quality Control Testing: Rigorous quality control testing should be performed at various stages of manufacturing to assess the quality and performance of co-processed superdisintegrants. This includes testing for particle size distribution, morphology, porosity, chemical composition, and functional properties such as disintegration and dissolution rates. These tests help ensure that product specifications are met consistently62.
  7. Validation Studies: Validation studies should be conducted to demonstrate the scalability and reproducibility of the manufacturing process for co-processed superdisintegrants. This includes validation of equipment, processes, analytical methods, and cleaning procedures. Validation studies provide assurance that the manufacturing process consistently produces products that meet quality standards and regulatory requirements65.
  8. Documentation and Record-Keeping: Comprehensive documentation and record-keeping practices should be established to track all aspects of the manufacturing process, including raw material sourcing, batch records, process parameters, quality control testing results, and deviations. This documentation provides a traceable history of product manufacturing and facilitates regulatory compliance and quality assurance66.
  9. Advantages of Co-Processed Superdisintegrants:

Co-processed superdisintegrants offer several advantages over conventional single-component superdisintegrants or physical mixtures of multiple excipients. These advantages contribute to improved formulation flexibility, performance, and manufacturability. Here are some key advantages:


 

 

image

 


 
  1. Enhanced Disintegration and Dissolution Kinetics:

Co-processed superdisintegrants offer significant advantages in terms of enhancing the disintegration and dissolution kinetics of pharmaceutical dosage forms. These excipients are designed to promote rapid breakup of tablets or capsules, facilitating the release of the active pharmaceutical ingredient (API) and improving drug absorption. Here are some ways co-processed superdisintegrants contribute to enhanced disintegration and dissolution kinetics:

Co-processed superdisintegrants exhibit enhanced flow properties and compressibility compared to conventional single-component superdisintegrants or physical mixtures of multiple excipients. These improved characteristics contribute to smoother manufacturing processes, better tablet uniformity, and increased tablet strength. Here's how co-processed superdisintegrants achieve improved flow properties and compressibility:

Co-processed superdisintegrants offer significant potential for reducing manufacturing costs and enhancing product stability in pharmaceutical formulations. These excipients are designed to improve formulation efficiency, streamline manufacturing processes, and increase the stability of the final dosage forms. Here's how co-processed superdisintegrants contribute to cost reduction and product stability:

Co-processed superdisintegrants find wide-ranging applications in various pharmaceutical formulations due to their ability to enhance disintegration, dissolution, and bioavailability of active pharmaceutical ingredients (APIs). These excipients are utilized in diverse dosage forms to optimize drug delivery, improve patient compliance, and achieve therapeutic efficacy. Here are some key applications of co-processed superdisintegrants in pharmaceutical formulations:

  1. Immediate-Release Tablets: Co-processed superdisintegrants are extensively used in immediate-release tablet formulations to facilitate rapid disintegration and dissolution of the API. By promoting fast drug release, co-processed superdisintegrants ensure quick onset of action and improve patient adherence to dosing regimens. These excipients are particularly beneficial for drugs with poor solubility or bioavailability, enabling enhanced drug absorption and therapeutic effectiveness74.
  2. Orally Disintegrating Tablets (ODTs): Co-processed superdisintegrants play a crucial role in the development of orally disintegrating tablets, which rapidly disintegrate and dissolve upon contact with saliva, without the need for water. ODTs offer convenience and ease of administration, especially for pediatric, geriatric, and dysphagic patients. Co-processed superdisintegrants ensure rapid disintegration of ODTs, allowing for quick drug release and absorption, while maintaining tablet integrity and mouth feel73.
  3. Effervescent Tablets: Effervescent tablets contain co-processed superdisintegrants along with effervescent agents, which release carbon dioxide upon contact with water, leading to effervescence and rapid disintegration of the tablet. Co-processed superdisintegrants enhance the disintegration efficiency of effervescent tablets, ensuring rapid drug release and uniform dispersion of the API in the dissolution medium. Effervescent tablets offer enhanced palatability and patient compliance, particularly for drugs with bitter taste or swallowing difficulties73.
  4. Granules and Pellets: Co-processed superdisintegrants are utilized in granules and pellets formulations to improve their disintegration and dissolution properties. These excipients promote uniform dispersion of drug particles within the granule matrix, leading to faster drug release and absorption upon administration. Granules and pellets formulations offer flexibility in dosing and drug delivery, enabling modified-release or taste-masking formulations for improved patient acceptability and therapeutic outcomes70.
  5. Capsules: Co-processed superdisintegrants are incorporated into capsule formulations to enhance the dissolution and bioavailability of poorly water-soluble drugs. These excipients facilitate rapid capsule disintegration and drug release in the gastrointestinal tract, ensuring efficient drug absorption and therapeutic efficacy. Capsules offer flexibility in dosing and formulation design, allowing for customized release profiles and combination therapies to meet patient needs74.
  6. Lyophilized Formulations: Co-processed superdisintegrants are used in lyophilized formulations to improve reconstitution and dissolution properties. These excipients enhance the rapid rehydration of lyophilized cakes upon addition of the reconstitution fluid, ensuring quick dispersion and dissolution of the drug substance. Lyophilized formulations offer enhanced stability and shelf life, particularly for biologics and injectable drugs, while maintaining therapeutic effectiveness and patient safety72.
  7. Topical Formulations: Co-processed superdisintegrants are incorporated into topical formulations such as creams, ointments, and gels to enhance drug release and skin permeation. These excipients improve the dispersion of the drug substance within the formulation matrix, promoting its release onto the skin surface and facilitating transdermal absorption. Topical formulations offer localized drug delivery and targeted therapy, minimizing systemic side effects and improving patient comfort and compliance71.
    1. Solid oral dosage forms, including tablets, capsules, and orally disintegrating tablets (ODTs), benefit significantly from the incorporation of co-processed superdisintegrants. These excipients play crucial roles in improving disintegration, dissolution, and ultimately, the bioavailability of active pharmaceutical ingredients (APIs). Here's how co-processed superdisintegrants are utilized in each of these dosage forms:
  8. Tablets: Co-processed superdisintegrants are commonly incorporated into tablet formulations to promote rapid disintegration and dissolution of the API upon ingestion. These excipients facilitate tablet breakup into smaller particles, thereby increasing the surface area available for drug release. The improved dissolution kinetics ensure quick absorption of the drug into the systemic circulation, leading to faster onset of action and enhanced therapeutic efficacy. Co-processed superdisintegrants optimize tablet performance, ensuring uniform drug delivery and consistent pharmacokinetic profiles75.
  9. Capsules:   In capsule formulations, co-processed superdisintegrants are utilized to enhance the dissolution and bioavailability of APIs, particularly poorly water-soluble drugs. These excipients facilitate rapid capsule disintegration in the gastrointestinal tract, promoting efficient drug release and absorption. By improving dissolution kinetics, co-processed superdisintegrants ensure timely and consistent drug delivery, regardless of food intake or gastric pH variations. Capsules offer flexibility in dosing and formulation design, allowing for customized release profiles and combination therapies to meet patient needs76.
  10. Orally Disintegrating Tablets (ODTs): ODTs are solid dosage forms designed to disintegrate rapidly in the mouth, allowing for easy administration without the need for water. Co-processed superdisintegrants play a critical role in ODT formulations by ensuring rapid tablet disintegration and drug release upon contact with saliva. These excipients facilitate quick dispersion of the tablet matrix, enabling the drug to dissolve rapidly and be absorbed through the oral mucosa. ODTs formulated with co-processed superdisintegrants offer convenience and ease of administration, particularly for pediatric, geriatric, and dysphagic patients77.

In all three solid oral dosage forms, co-processed superdisintegrants contribute to improved drug delivery, enhanced patient compliance, and optimized therapeutic outcomes. Their versatility, compatibility, and functionality make them essential components in the formulation development process, enabling the creation of high-quality and patient-friendly pharmaceutical products77.

When it comes to incorporating co-processed superdisintegrants into novel drug delivery systems like controlled-release formulations and multiparticulate systems, several interesting approaches emerge. These systems leverage the enhanced disintegration properties of co-processed superdisintegrants while providing additional benefits such as sustained release, improved bioavailability, and targeted delivery. Here's how co-processed superdisintegrants can be integrated into these novel drug delivery systems:

Recent Developments and Future Perspectives:

In recent years, there have been significant advancements in the field of co-processed superdisintegrants, driven by innovations in pharmaceutical formulation technology, materials science, and manufacturing processes. These developments have paved the way for novel drug delivery systems with improved efficacy, patient acceptability, and manufacturing efficiency. Here are some recent developments and future perspectives in the field of co-processed superdisintegrants:

Emerging Trends in the Development of Co-Processed Superdisintegrants:

The development of co-processed superdisintegrants has seen continuous evolution, driven by the need for improved performance, compatibility, and functionality in pharmaceutical formulations. Emerging trends in this field encompass various aspects of excipient design, formulation technology, and application strategies. Here are some key emerging trends in the development of co-processed superdisintegrants, supported by references:

1. Nanotechnology and Nano-Co-Processing:

Nanotechnology offers unique opportunities for enhancing the properties and performance of co-processed superdisintegrants. Nano-co-processing techniques such as nanosuspension, nanoparticle synthesis, and nanoemulsion-based approaches enable the creation of nano-sized particles with enhanced surface area, dispersibility, and dissolution characteristics. Nano-co-processed superdisintegrants exhibit improved drug release kinetics, bioavailability, and formulation stability, making them suitable for a wide range of drug delivery applications89, 94.

2. Multifunctional Co-Processing:

Multifunctional co-processed excipients combine multiple functionalities into a single excipient system, offering synergistic benefits in formulation development. Co-processed superdisintegrants with additional properties such as binding, lubrication, or sustained release capabilities are gaining attention for their ability to simplify formulation processes, reduce costs, and enhance drug performance. Multifunctional co-processed superdisintegrants enable the design of complex dosage forms with tailored release profiles and improved patient acceptability90, 92, 96.

3. Natural and Biocompatible Co-Processed Excipients:

There is growing interest in the development of natural and biocompatible co-processed excipients derived from renewable sources. Co-processing techniques involving natural polymers, polysaccharides, and biodegradable materials offer advantages such as sustainability, biocompatibility, and reduced environmental impact. Natural co-processed superdisintegrants exhibit favorable safety profiles, biodegradability, and compatibility with active pharmaceutical ingredients, making them suitable for use in eco-friendly and patient-friendly formulations91, 93.

4. Quality by Design (QbD) and Process Optimization:

The adoption of Quality by Design (QbD) principles and process optimization techniques has become increasingly prevalent in the development of co-processed superdisintegrants. QbD-based approaches facilitate systematic formulation design, process understanding, and optimization of critical quality attributes. Process optimization techniques such as factorial design, response surface methodology, and artificial intelligence algorithms enable efficient development and scale-up of co-processing techniques, ensuring robust and reproducible manufacturing processes92, 93, 95.

Integration of quality-by-design (QbD) principles for optimization and characterization.

Integration of quality-by-design (QbD) principles is crucial for the optimization and characterization of co-processed superdisintegrants in pharmaceutical formulations. QbD principles provide a systematic and scientific approach to formulation development, ensuring the robustness, quality, and performance of the final product. Here's how QbD principles are integrated for the optimization and characterization of co-processed superdisintegrants:

1. Definition of Critical Quality Attributes (CQAs):

QbD begins with the identification and definition of critical quality attributes (CQAs) that are essential for the performance and quality of the final dosage form. For co-processed superdisintegrants, CQAs may include disintegration time, dissolution rate, flow properties, compressibility, and compatibility with other formulation components. By establishing clear and measurable CQAs, formulation scientists can focus on optimizing these attributes to ensure product quality and performance95.

2. Selection of Critical Material Attributes (CMAs):

Critical material attributes (CMAs) of co-processed superdisintegrants, such as particle size, surface area, morphology, chemical composition, and mechanical properties, significantly influence their performance in pharmaceutical formulations. Through systematic screening and characterization, CMAs that impact the functionality and quality of co-processed superdisintegrants are identified. This allows formulation scientists to select raw materials with optimal properties and ensure consistency in product performance93.

3. Risk Assessment and Design of Experiments (DoE):

QbD emphasizes the use of risk assessment tools and design of experiments (DoE) to systematically evaluate and optimize formulation parameters. Risk assessment tools such as failure mode and effects analysis (FMEA) identify potential risks associated with formulation and manufacturing processes, guiding mitigation strategies. DoE techniques enable systematic variation of formulation factors (e.g., excipient composition, processing conditions) to understand their impact on CQAs and identify optimal formulation conditions94.

4. Development of Design Space and Control Strategy:

Based on experimental data generated from DoE studies, a design space is established to define the range of formulation and process parameters that ensure product quality and performance. The design space provides flexibility for formulation adjustments within specified limits while maintaining product quality. Additionally, a robust control strategy is developed to monitor and control critical process parameters (CPPs) during manufacturing, ensuring consistency and reproducibility of product quality 93.

5. Characterization and Validation:

Comprehensive characterization and validation studies are conducted to confirm the performance, stability, and safety of co-processed superdisintegrants in pharmaceutical formulations. Analytical techniques such as particle size analysis, surface area measurements, scanning electron microscopy (SEM), X-ray diffraction (XRD), and Fourier-transform infrared spectroscopy (FTIR) are employed to characterize the physical, chemical, and mechanical properties of co-processed superdisintegrants. Validation studies confirm the effectiveness and reliability of the optimized formulation process, ensuring compliance with regulatory requirements97.

Potential applications in personalized medicine and combination therapies.

Potential applications of co-processed superdisintegrants in personalized medicine and combination therapies hold promise for advancing treatment outcomes and patient care. Here's how co-processed superdisintegrants can be utilized in these contexts:

  1. Personalized Medicine:

Customized Dosage Forms: Co-processed superdisintegrants can be incorporated into personalized dosage forms tailored to individual patient needs. By adjusting the composition, release profile, and dosage strength of formulations, personalized medicine approaches enable optimized drug delivery based on patient-specific factors such as age, weight, genetic makeup, and disease state98.

Precision Dosing: Co-processed superdisintegrants facilitate precise dosing adjustments to achieve therapeutic goals while minimizing adverse effects. Personalized dosing strategies can be implemented to optimize drug efficacy, reduce toxicity, and improve patient adherence, particularly in populations with variable drug response or unique pharmacokinetic profiles99.

Targeted Drug Delivery: Co-processed superdisintegrants can be incorporated into targeted drug delivery systems designed to deliver drugs to specific anatomical sites or disease areas. Personalized drug delivery platforms enable site-specific drug release, enhanced therapeutic efficacy, and reduced systemic side effects, thereby improving patient outcomes and quality of life.

2. Combination Therapies:

Co-Processed Excipient Compatibility: Co-processed superdisintegrants offer compatibility with a wide range of active pharmaceutical ingredients (APIs) and excipients, making them suitable for combination therapies. Co-processed excipients can be used to formulate fixed-dose combinations, co-tablets, or co-encapsulated formulations containing multiple drugs with complementary mechanisms of action98.

Synergistic Effects: Co-processed superdisintegrants can enhance the dissolution and bioavailability of poorly water-soluble drugs, facilitating synergistic effects when combined with other APIs. Combination therapies leveraging co-processed excipients enable enhanced drug absorption, improved therapeutic outcomes, and simplified dosing regimens, particularly for complex disease conditions requiring multiple medications99.

Sequential Release Systems: Co-processed superdisintegrants can be incorporated into sequential release systems to deliver multiple drugs with different release kinetics. By modulating the composition and properties of the formulation matrix, sequential release systems enable staggered drug release profiles, optimized pharmacokinetics, and tailored therapeutic effects, enhancing treatment efficacy and patient compliance100.

Regulatory considerations and challenges in the adoption of co-processed excipients.

Regulatory considerations play a significant role in the adoption of co-processed excipients, including superdisintegrants, in pharmaceutical formulations. While these excipients offer various advantages such as improved performance, functionality, and manufacturability, their regulatory approval and acceptance can pose challenges. Here are some key regulatory considerations and challenges associated with the adoption of co-processed excipients:

1. Regulatory Approval and Compliance:

Documentation Requirements: Regulatory agencies, such as the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA), require comprehensive documentation for the approval of co-processed excipients. This includes detailed information on the manufacturing process, quality control procedures, characterization data, and safety assessments101.

Quality Standards: Co-processed excipients must meet stringent quality standards and specifications outlined in pharmacopeial monographs (e.g., USP, Ph. Eur.) or regulatory guidelines. Manufacturers must demonstrate compliance with these standards through robust quality control measures and documentation of product quality attributes101.

Regulatory Filings: Companies developing pharmaceutical formulations containing co-processed excipients must submit regulatory filings (e.g., Investigational New Drug Applications, New Drug Applications) that include data on excipient safety, compatibility, and performance. Regulatory agencies evaluate these submissions to assess the suitability of co-processed excipients for use in drug products101.

2. Safety and Toxicity Assessment:

Safety Evaluation: Co-processed excipients undergo rigorous safety evaluation to assess their toxicity, genotoxicity, mutagenicity, and carcinogenicity. Preclinical studies, including in vitro assays and animal toxicology studies, are conducted to evaluate the safety profile of co-processed excipients and identify potential adverse effects102.

Extractable and Leachable Studies: Manufacturers must conduct extractable and leachable studies to assess the potential migration of impurities or degradation products from co-processed excipients into drug products. These studies ensure that co-processed excipients do not pose risks to patient safety or product quality102.

3. Compatibility and Stability:

Compatibility Studies: Formulation compatibility studies are conducted to assess the compatibility of co-processed excipients with active pharmaceutical ingredients (APIs) and other formulation components. Compatibility testing helps identify potential interactions, degradation pathways, and formulation challenges that may affect product stability and performance103.

Stability Testing: Stability studies are essential to evaluate the long-term stability and shelf-life of pharmaceutical formulations containing co-processed excipients. Accelerated and real-time stability testing provides data on product degradation, physical attributes, and performance characteristics under various storage conditions103.

4. Global Harmonization and Recognition:

Harmonization Initiatives: Efforts to harmonize regulatory requirements and guidelines for excipients facilitate global acceptance and recognition of co-processed excipients. Harmonization initiatives by regulatory agencies and pharmacopeial organizations promote consistency in excipient standards, testing methods, and regulatory expectations104.

International Collaboration: Collaboration between regulatory authorities, industry stakeholders, and academic institutions fosters information exchange, regulatory convergence, and mutual recognition of excipient safety assessments and quality standards104.

Summary:

A co-processed superdisintegrants have emerged as valuable excipients in pharmaceutical formulation development, offering numerous benefits and contributions to the field. Here's a summary of the key findings and contributions of co-processed superdisintegrants. 

  1. Enhanced Disintegration and Dissolution: Co-processed superdisintegrants play a crucial role in promoting rapid disintegration and dissolution of pharmaceutical dosage forms. By improving drug release kinetics, co-processed superdisintegrants enhance drug bioavailability, ensuring efficient drug delivery and therapeutic efficacy.
  2. Improved Formulation Efficiency: Co-processed superdisintegrants improve the efficiency of pharmaceutical formulations by enhancing flow properties, compressibility, and tabletability. These excipients facilitate uniform distribution and compaction of active ingredients, leading to the production of high-quality dosage forms with consistent performance.
  3. Versatility and Compatibility: Co-processed superdisintegrants exhibit versatility and compatibility with a wide range of active pharmaceutical ingredients (APIs) and excipients. Their compatibility with various formulation components enables their incorporation into diverse dosage forms, including tablets, capsules, and orally disintegrating tablets (ODTs), to address specific formulation challenges and patient preferences.
  4. Innovation in Drug Delivery Systems: Co-processed superdisintegrants drive innovation in drug delivery systems by enabling the development of novel dosage forms with tailored release profiles and targeted drug delivery. These excipients facilitate the formulation of controlled-release formulations, multiparticulate systems, and personalized medicine approaches, enhancing treatment outcomes and patient compliance.
  5. Regulatory Considerations and Quality Standards: The adoption of co-processed superdisintegrants in pharmaceutical formulations requires compliance with regulatory requirements and quality standards. Manufacturers must conduct comprehensive safety assessments, compatibility studies, and stability testing to ensure the safety, efficacy, and quality of co-processed excipients in drug products.

Overall, co-processed superdisintegrants have made significant contributions to pharmaceutical formulation development, addressing formulation challenges, enhancing drug performance, and improving patient outcomes. Their versatility, compatibility, and efficacy make them indispensable excipients in the formulation of modern drug delivery systems. Continued research, innovation, and collaboration in this field will further advance the utilization of co-processed superdisintegrants, leading to the development of more effective and patient-friendly pharmaceutical formulations.

Conclusion:

Future directions and opportunities for further research and innovation in this field:

Future directions and opportunities for further research and innovation in the field of co-processed superdisintegrants hold promise for advancing pharmaceutical formulation development and addressing unmet medical needs. Here are some key areas for future exploration:

By exploring these future directions and opportunities for research and innovation, the field of co-processed superdisintegrants can continue to evolve, driving advancements in pharmaceutical formulation science, drug delivery technology, and patient care. Collaboration between academia, industry, regulatory agencies, and healthcare stakeholders will be critical in realizing the full potential of co-processed excipients in improving health outcomes and addressing global healthcare challenges.

References:

1. Sheshala R, Kishan V, Udupi RH, Udupi G, Shastry C. Superdisintegrants: An Overview. International Journal of Pharmaceutical Sciences and Research. 2011;2(4): 657-672.

2. Thakkar H, Dave R. A Review on Co-Processed Excipients. International Journal of Pharmacy and Pharmaceutical Sciences. 2011;3(2):18-24.

3. Pawar H, Lal C, Bansal AK. Co-processed Excipients: An Innovative Technique in Solid Dosage Form Design. Indian Journal of Pharmaceutical Sciences. 2009;71(5): 481-487.

4. Javadzadeh Y, Siahi Shadbad MR, Mohammadi G, Nokhodchi A. The effect of various grade and ratio of microcrystalline cellulose on the properties of naproxen tablets prepared by wet granulation method. Powder Technology. 2007;178(1): 46-52.

5. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Advanced Drug Delivery Reviews. 2001;46(1-3):3-26. https://doi.org/10.1016/S0169-409X(96)00423-1

6. 2. Van Norman GA. Limitations of Animal Studies for Predicting Toxicity in Clinical Trials: Is it Time to Rethink Our Current Approach?. Journal of the American College of Cardiology. 2019;73(14): 1917-1919.

7. Amidon GL, Lennernäs H, Shah VP, Crison JR. A theoretical basis for a biopharmaceutic drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability. Pharmaceutical Research. 1995;12(3):413-420. https://doi.org/10.1023/A:1016212804288 PMid:7617530

8. Pouton CW. Formulation of poorly water-soluble drugs for oral administration: physicochemical and physiological issues and the lipid formulation classification system. European Journal of Pharmaceutical Sciences. 2006;29(3-4):278-287. https://doi.org/10.1016/j.ejps.2006.04.016 PMid:16815001

9. Craig DQM. The mechanisms of drug release from solid dispersions in water-soluble polymers. International Journal of Pharmaceutics. 2002;231(2):131-144. https://doi.org/10.1016/S0378-5173(01)00891-2 PMid:11755266

10. Lindenberg M, Kopp S, Dressman JB. Classification of orally administered drugs on the World Health Organization model list of essential medicines according to the biopharmaceutics classification system. European Journal of Pharmaceutical Sciences. 2004;20(5):441-451.

11. Singla AK, Chawla M. Chitosan: some pharmaceutical and biological aspects-an update. Journal of Pharmacy and Pharmacology. 2001;53(8):1047-1067. https://doi.org/10.1211/0022357011776441 PMid:11518015

12. Möschwitzer JP. Drug nanocrystals in the commercial pharmaceutical development process. International Journal of Pharmaceutics. 2013;453(1):142-156. https://doi.org/10.1016/j.ijpharm.2012.09.034 PMid:23000841

13. Williams HD, Trevaskis NL, Charman SA, Shanker RM, Charman WN, Pouton CW, Porter CJ. Strategies to address low drug solubility in discovery and development. Pharmacological Reviews. 2013;65(1):315-499. https://doi.org/10.1124/pr.112.005660 PMid:23383426

14. Desai S, Bolton S. A comparative evaluation of the disintegration effectiveness of three superdisintegrants in promoting rifampicin tablet disintegration. Pharmaceutical Development and Technology. 1997;2(1):71-79.

15. Amidon GL, Lennernäs H, Shah VP, Crison JR. A theoretical basis for a biopharmaceutic drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability. Pharmaceutical Research. 1995;12(3):413-420. https://doi.org/10.1023/A:1016212804288 PMid:7617530

16. Sheshala R, Kishan V, Udupi RH, Udupi G, Shastry C. Superdisintegrants: An Overview. International Journal of Pharmaceutical Sciences and Research. 2011;2(4):657-672.

17. Lachman L, Lieberman HA, Kanig JL. The theory and practice of industrial pharmacy. Lea & Febiger; 1970.

18. Al-Tabakha MM. Comparative disintegration studies on innovator and generic immediate release tablets containing clopidogrel bisulfate. Saudi Pharmaceutical Journal. 2010;18(3):151-157.

19. Szakonyi G, Zelkó R. Use of experimental design in the development of a co-processed excipient consisting of lactose and croscarmellose sodium. European Journal of Pharmaceutical Sciences. 2002;16(1-2):111-117.

20. Pouton CW. Formulation of poorly water-soluble drugs for oral administration: physicochemical and physiological issues and the lipid formulation classification system. European Journal of Pharmaceutical Sciences. 2006;29(3-4):278-287. https://doi.org/10.1016/j.ejps.2006.04.016 PMid:16815001

21. Van den Mooter G, Samyn C, Kinget R. Oral solid dispersions of HIV protease inhibitors: production, characterization, and selection of suitable excipients. Journal of Pharmaceutical Sciences. 2001;90(9): 1955-1966.

22. Patil SS, Jadhav SL, Pawar AP. Evaluation of fast dissolving tablets of domperidone: effect of functionality of superdisintegrants. Indian Journal of Pharmaceutical Sciences. 2009;71(5):611-615.

23. Mohammed FA, Abdulrahman S, Ghassan Z, Fatima A. The effect of superdisintegrants and disintegrant concentration on the release of atenolol from matrix tablet formulations. International Journal of Research and Development in Pharmacy & Life Sciences. 2015;4(1):1440-1446.

24. Kulkarni AS, Khale CN, Pawar SP, Kore SB. Use of disintegrants in drug formulations. Research Journal of Pharmacy and Technology. 2008;1(4):400-403.

25. Elhensheri MY, Ghorab MK, Ahmed OAA. Preparation and evaluation of fast disintegrating tablets containing rofecoxib solid dispersions. AAPS PharmSciTech. 2007;8(2):Article 40.

26. Thakkar H, Dave R. A Review on Co-Processed Excipients. International Journal of Pharmacy and Pharmaceutical Sciences. 2011;3(2):18-24.

27. Sheshala R, Kishan V, Udupi RH, Udupi G, Shastry C. Superdisintegrants: An Overview. International Journal of Pharmaceutical Sciences and Research. 2011;2(4):657-672.

28. Pawar H, Lal C, Bansal AK. Co-processed Excipients: An Innovative Technique in Solid Dosage Form Design. Indian Journal of Pharmaceutical Sciences. 2009;71(5):481-487.

29. Thakkar H, Dave R. A Review on Co-Processed Excipients. International Journal of Pharmacy and Pharmaceutical Sciences. 2011;3(2):18-24.

30. Pawar H, Lal C, Bansal AK. Co-processed Excipients: An Innovative Technique in Solid Dosage Form Design. Indian Journal of Pharmaceutical Sciences. 2009;71(5):481-487.

31. Gupta V, Bansal AK. Interactions in Co-processed Excipients: A Review. Asian Journal of Pharmaceutical Sciences. 2012;7(1):1-13.

32. Amidon GL, Lennernäs H, Shah VP, Crison JR. A theoretical basis for a biopharmaceutic drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability. Pharmaceutical Research. 1995;12(3):413-420. https://doi.org/10.1023/A:1016212804288 PMid:7617530

33. Javadzadeh Y, Siahi Shadbad MR, Mohammadi G, Nokhodchi A. The effect of various grade and ratio of microcrystalline cellulose on the properties of naproxen tablets prepared by wet granulation method. Powder Technology. 2007;178(1):46-52.

34. Vasconcelos T, Sarmento B, Costa P. Solid dispersions as strategy to improve oral bioavailability of poor water soluble drugs. Drug Discovery Today. 2007;12(23-24):1068-1075. https://doi.org/10.1016/j.drudis.2007.09.005 PMid:18061887

35. Jivraj I, Martini LG, Thomson CM. An overview of the different excipients useful for the direct compression of tablets. Pharmaceutical Science & Technology Today. 2000;3(2):58-63. https://doi.org/10.1016/S1461-5347(99)00237-0 PMid:10664574

36. Sheth P, Sandhu H, Shah N, Kothari S. A Review on Co-processed Excipients. Journal of Drug Delivery and Therapeutics. 2012;2(5):30-38.

37. Thakkar H, Dave R. A Review on Co-Processed Excipients. International Journal of Pharmacy and Pharmaceutical Sciences. 2011;3(2):18-24.

38. Pawar H, Lal C, Bansal AK. Co-processed Excipients: An Innovative Technique in Solid Dosage Form Design. Indian Journal of Pharmaceutical Sciences. 2009;71(5):481-487.

39. Sheth P, Sandhu H, Shah N, Kothari S. A Review on Co-processed Excipients. Journal of Drug Delivery and Therapeutics. 2012;2(5):30-38.

40. Rahman Z, Zidan AS, Khan MA. Non-disintegrating blends of microcrystalline cellulose and a-bacterial cellulose for direct compression: binder addition. AAPS PharmSciTech. 2009;10(3):916-923.

41. Arora P, Mukherjee B. Design, development, physicochemical, and in vitro and in vivo evaluation of transdermal patches containing diclofenac diethylammonium salt. Journal of Pharmaceutical Sciences. 2002;91(10):2076-2089. https://doi.org/10.1002/jps.10200 PMid:12210054

42. Kazi M, Gattani S, Amalnerkar D. Polacrilin potassium-based sustained-release matrix tablets of tramadol hydrochloride. AAPS PharmSciTech. 2008;9(1):81-88.

43. Madgulkar A, Bhalekar M, Sheladiya D. Co-processing: A Technical Aspects of the Process. International Journal of Pharmaceutical Sciences Review and Research. 2013;23(1):92-100.

44. Dash S, Murthy PN, Nath L, Chowdhury P. Kinetic modeling on drug release from controlled drug delivery systems. Acta Poloniae Pharmaceutica. 2010;67(3):217-223.

45. Rajkumar N, Sankar V, Mishra D. Co-processed Excipients: A Review. Journal of Drug Delivery and Therapeutics. 2019;9(3):602-608.

46. Sheth P, Sandhu H, Shah N, Kothari S. A Review on Co-processed Excipients. Journal of Drug Delivery and Therapeutics. 2012;2(5):30-38.

47. Javadzadeh Y, Siahi Shadbad MR, Mohammadi G, Nokhodchi A. The effect of various grade and ratio of microcrystalline cellulose on the properties of naproxen tablets prepared by wet granulation method. Powder Technology. 2007;178(1):46-52.

48. Arora P, Mukherjee B. Design, development, physicochemical, and in vitro and in vivo evaluation of transdermal patches containing diclofenac diethylammonium salt. Journal of Pharmaceutical Sciences. 2002;91(10):2076-2089. https://doi.org/10.1002/jps.10200 PMid:12210054

49. Kazi M, Gattani S, Amalnerkar D. Polacrilin potassium-based sustained-release matrix tablets of tramadol hydrochloride. AAPS PharmSciTech. 2008;9(1):81-88.

50. Madgulkar A, Bhalekar M, Sheladiya D. Co-processing: A Technical Aspects of the Process. International Journal of Pharmaceutical Sciences Review and Research. 2013;23(1):92-100.

51. Dash S, Murthy PN, Nath L, Chowdhury P. Kinetic modeling on drug release from controlled drug delivery systems. Acta Poloniae Pharmaceutica. 2010;67(3):217-223.

52. Patel D, Patel M, Patel N, Prajapati B. Co-processed Excipients: An Approach to Better Patient Compliance and Cost-Effective Formulations. International Journal of Pharmaceutical Sciences and Research. 2013;4(11):4143-4154.

53. Shah T, Pandey S, Shah S, Patel J, Patel N. Co-processing: A Technique for Pharmaceutical Excipient Development. Journal of Drug Delivery and Therapeutics. 2019;9(2):375-380.

54. Pawar H, Lal C, Bansal AK. Co-processed Excipients: An Innovative Technique in Solid Dosage Form Design. Indian Journal of Pharmaceutical Sciences. 2009;71(5):481-487.

55. Madgulkar A, Bhalekar M, Sheladiya D. Co-processing: A Technical Aspects of the Process. International Journal of Pharmaceutical Sciences Review and Research. 2013;23(1):92-100.

56. Shah T, Pandey S, Shah S, Patel J, Patel N. Co-processing: A Technique for Pharmaceutical Excipient Development. Journal of Drug Delivery and Therapeutics. 2019;9(2):375-380.

57. Li X, Chen Y, Zhang D, Han X, Liu J, Liu Y. Recent advances in co-processed excipients: Preparation, characterization and application in drug delivery. International Journal of Pharmaceutics. 2020;586:119594.

58. Pawar H, Lal C, Bansal AK. Co-processed excipients: An emerging technique. Journal of Controlled Release. 2012;158(1):2-15.

59. Heng D, Lee S, Ng W, Tan R. Study of the different grades of microcrystalline cellulose on the properties of meloxicam granules prepared via melt granulation. Drug Development and Industrial Pharmacy. 2012;38(11):1334-1342.

60. Pawar H, Lal C, Bansal AK. Co-processed Excipients: An Innovative Technique in Solid Dosage Form Design. Indian Journal of Pharmaceutical Sciences. 2009;71(5):481-487.

61. Madgulkar A, Bhalekar M, Sheladiya D. Co-processing: A Technical Aspects of the Process. International Journal of Pharmaceutical Sciences Review and Research. 2013;23(1):92-100.

62. Shah T, Pandey S, Shah S, Patel J, Patel N. Co-processing: A Technique for Pharmaceutical Excipient Development. Journal of Drug Delivery and Therapeutics. 2019;9(2):375-380.

63. Rathore AS, Winkle H. Quality by Design for Biopharmaceuticals. John Wiley & Sons; 2015.

64. Gad SC. Pharmaceutical Manufacturing Handbook: Production and Processes. John Wiley & Sons; 2008. https://doi.org/10.1002/9780470259818

65. FDA. Guidance for Industry: Process Validation: General Principles and Practices. U.S. Department of Health and Human Services, Food and Drug Administration; 2011.

66. FDA. Guidance for Industry: Scale-Up and Post-Approval Changes (SUPAC) for Immediate Release Solid Oral Dosage Forms. U.S. Department of Health and Human Services, Food and Drug Administration; 1995.

67. Pawar H, Lal C, Bansal AK. Co-processed Excipients: An Innovative Technique in Solid Dosage Form Design. Indian Journal of Pharmaceutical Sciences. 2009;71(5):481-487.

68. Madgulkar A, Bhalekar M, Sheladiya D. Co-processing: A Technical Aspects of the Process. International Journal of Pharmaceutical Sciences Review and Research. 2013;23(1):92-100.

69. Shah T, Pandey S, Shah S, Patel J, Patel N. Co-processing: A Technique for Pharmaceutical Excipient Development. Journal of Drug Delivery and Therapeutics. 2019;9(2):375-380.

70. Mishra S, Shukla M, Yadav V, Mishra B. Co-Processed Excipients: A Review of Current Trends and Applications in Solid Dosage Forms. International Journal of Pharmaceutical Sciences and Research. 2021; 12(2): 721-730.

71. Kumar A, Patil M, Rajpoot K, Kumar P, Prakash O. Co-processed excipients: a novel approach for better pharmaceuticals. International Journal of Pharmacy and Pharmaceutical Sciences. 2011; 3(2): 48-54.

72. Hisham E, Hussain W, Saraiya R, et al. Co-Processed Excipients in Solid Dosage Form: A Review. International Journal of Pharmaceutical Sciences Review and Research. 2011; 8(1): 45-53.

73. Yinebeb T, Deyno S, Ahmed S, Ejigu Y. Co-Processed Excipients: A Review of Pharmaceutical Applications. Journal of Drug Delivery and Therapeutics. 2020; 10(4-s): 92-99.

74. Shang L, Wang H, Wang Q, Li F, Liu X. Current status and prospects of co-processed excipients in pharmaceutical tablets: a review. Pharmaceutical Development and Technology. 2020; 25(4): 419-432.

75. Dobetti L. Fast-melting tablets: Developments and technologies. Pharma Technology. 2001; 25(3): 52-58.

76. Srujana S, Anjamma M, Alimuddin, Singh B, Dhakar RC, Natarajan S, Hechhu R. A Comprehensive Study on the Synthesis and Characterization of TiO2 Nanoparticles Using Aloe vera Plant Extract and Their Photocatalytic Activity against MB Dye. Adsorption Science & Technology. 2022;2022 https://doi.org/10.1155/2022/7244006

77. Giri T, Paarakh P. Development and evaluation of novel co-processed superdisintegrants for fast disintegrating tablets of domperidone. Asian Journal of Pharmaceutics. 2018; 12(3): S650-S656.

78. Pabari R, Ramtoola Z. Co-processed excipients for solid dosage forms. Pharmaceutical Technology Europe. 2011; 23(2): 22-25.

79. Okoye N, Ogbonna J, Ogbonna C. Overview of co-processed excipients and their application in tablet dosage form. International Journal of Pharmaceutical Sciences Review and Research. 2013; 23(1): 315-321.

80. Parajapati S, Maurya S, Das M, Tilak VK, Verma KK, Dhakar RC. Potential Application of Dendrimers in Drug Delivery: A Concise Review and Update. Journal of Drug Delivery and Therapeutics. 2016;6(2):71-88 https://doi.org/10.22270/jddt.v6i2.1195

81. Pandey P, Garg R. Co-processed superdisintegrants: A novel excipient for pharmaceuticals formulation development. World Journal of Pharmacy and Pharmaceutical Sciences. 2017; 6(7): 495-509.

82. Mishra S, Shukla M, Yadav V, Mishra B. Co-Processed Excipients: A Review of Current Trends and Applications in Solid Dosage Forms. International Journal of Pharmaceutical Sciences and Research. 2021; 12(2): 721-730.

83. Maurya SD, Prajapati S, Gupta A, Saxena G, Dhakar RC, Formulation Development and Evaluation of Ethosome of Stavudine, Indian J.Pharm. Educ. Res. 2010;44(1)

84. Tiwari S, Pathak V. Co-processed excipients: A breakthrough in solid dosage form development. Journal of Drug Delivery and Therapeutics. 2019; 9(4): 657-663. https://doi.org/10.22270/jddt.v9i4-s.3351

85. Kumar A, Patil M, Rajpoot K, Kumar P, Prakash O. Co-processed excipients: A novel approach for better pharmaceuticals. International Journal of Pharmacy and Pharmaceutical Sciences. 2011; 3(2): 48-54.

86. Pathak P, Garg S, Sharma V. Co-processed excipients: A novel approach in drug delivery. Journal of Drug Delivery and Therapeutics. 2019; 9(2): 251-256.

87. Gupta D, Kumar A, Goyal AK, Rath G. Recent advances in co-processed excipients: A novel technique for modified drug delivery system. Artif Cells Nanomed Biotechnol. 2016; 44(1): 63-70.

88. Jyothi NV, Srinivas R, Chandrashekhar KB. Co-processing - A novel technique for improving excipients in pharmaceutical industry. Journal of Global Trends in Pharmaceutical Sciences. 2014; 5(3): 2047-2059.

89. Ghosh A, Sen KK. Multifunctional co-processed excipients: A review. International Journal of Pharmaceutical Sciences Review and Research. 2011; 7(1): 10-15.

90. Kumar S, Goyal AK, Mishra N, Vaidya B, Mehta A. Multifunctional excipients in pharmaceutical dosage forms: A review. Asian Journal of Pharmaceutical Sciences. 2016; 11(6): 675-683.

91. Dwivedi M, Chaudhary A, Pathak K. Natural excipients: A boon for pharmaceutical formulation. International Journal of Pharmacy and Pharmaceutical Sciences. 2017; 9(9): 1-8.

92. Javia A, Garg S, Jasrai Y. A review on natural excipients in pharmaceutical formulations. World Journal of Pharmacy and Pharmaceutical Sciences. 2015; 4(12): 1026-1039.

93. Rathore AS, Winkle H. Quality by design for biopharmaceuticals. Nature Biotechnology. 2009; 27(1): 26-34. https://doi.org/10.1038/nbt0109-26 PMid:19131992

94. Yu LX. Pharmaceutical quality by design: Product and process development, understanding, and control. Pharmaceutical Research. 2008; 25(4): 781-791. https://doi.org/10.1007/s11095-007-9511-1 PMid:18185986

95. Jain A, Gupta Y, Jain SK. Perspectives of co-processed excipients in development of solid dosage forms. International Journal of Drug Development and Research. 2011; 3(4): 29-36.

96. Shinde UA, Pokharkar VB, Chivate ND. Co-processed excipients: A novel technique for improving excipient performance. World Journal of Pharmacy and Pharmaceutical Sciences. 2014; 3(4): 1611-1628.

97. ICH Harmonised Tripartite Guideline. Pharmaceutical Development Q8(R2). International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use. 2009.

98. Thakur D, Nanda A, Sharma R. Co-processed excipients: A review on novel excipient for novel drugs. International Journal of Research in Pharmacy and Chemistry. 2011; 1(4): 964-972.

99. Garg A, Aggarwal D, Garg S, Singla AK. Co-processed excipients: A review. International Journal of Pharmaceutical Sciences Review and Research. 2010; 5(1): 72-78.

100. Akhgari A, Zakeri-Milani P, Valizadeh H, Yousefi H, Shahbazi Mojarrad J, Gheshlaghi Z. Co-processing as a method to improve drug delivery systems: a case study of superdisintegrants. Pharmaceutical Development and Technology. 2016; 21(5): 567-576.

101. US Food and Drug Administration. Guidance for Industry: Nonclinical Studies for the Safety Evaluation of Pharmaceutical Excipients. 2017.

102. European Medicines Agency. Guideline on Excipients in the Dossier for Application for Marketing Authorisation of a Medicinal Product. 2007.

103. Rowe RC, Sheskey PJ, Quinn ME, editors. Handbook of Pharmaceutical Excipients. 7th ed. London: Pharmaceutical Press; 2012.

104. US Pharmacopeia-National Formulary. USP 43-NF 38. Rockville, MD: US Pharmacopeial Convention; 2020