View Article

Abstract

Allopathic medicine, the mainstream system of healthcare, relies extensively on scientifically developed formulations to ensure the safe, effective, and targeted delivery of therapeutic agents. This comprehensive review presents an in-depth analysis of the formulation science underlying allopathic drug products. It explores the entire spectrum—from preformulation studies and excipient selection to advanced drug delivery systems and nanotechnology applications. Key formulation types such as tablets, capsules, injectables, transdermal patches, and novel carriers are discussed, along with their design strategies, challenges, and clinical relevance.The review also highlights the importance of regulatory guidelines, quality assurance, bioavailability enhancement, and the growing role of artificial intelligence and personalized medicine in formulation development. Emerging trends such as green formulations, 3D printing, and decentralized manufacturing are examined for their potential to reshape the future of pharmaceutical development. Environmental and ethical considerations, clinical trials, and global supply chain logistics are also incorporated to provide a holistic view of the field. This paper aims to serve as a detailed academic and practical resource for researchers, formulators, and healthcare professionals. It emphasizes the interdisciplinary nature of modern formulation science and the need for continuous innovation to meet evolving patient needs and global health challenges.

Keywords

Allopathic medicine, pharmaceutical dosage forms, nanotechnology, excipients, drug delivery systems, personalized medicine, artificial intelligence in pharmaceutics, regulatory guidelines, preformulation studies, 3D printing, green pharmaceuticals

Introduction

Allopathy, also known as modern or Western medicine, is a system of medical practice that treats disease symptoms using drugs, surgery, or radiation. This approach is based on evidence and scientific research and is widely practiced across the globe. Allopathic medicines are developed through extensive research, clinical trials, and regulatory approval to ensure efficacy and safety [1]. The term “allopathy” was coined by Samuel Hahnemann in the 19th century to differentiate it from homeopathy [5]. However, today, it refers to the mainstream medicine taught in medical schools and practiced by healthcare professionals [2].

Allopathic formulations include a wide variety of dosage forms such as tablets, capsules, injections, syrups, and topical applications. These formulations are designed to deliver drugs effectively and safely into the body to achieve the desired therapeutic effect [2][4].

Allopathy medicine

The main goals of allopathic formulations are:

  1. To deliver the drug at the right site of action
  2. To maintain adequate drug levels in the blood
  3. To ensure patient compliance through easy administration and minimal side effects [3][4]

This review will provide an in-depth overview of allopathic formulations, their classification, mechanisms of action, manufacturing, quality control, innovations, and future prospects.

HISTORY AND EVOLUTION OF ALLOPATHIC MEDICINES

The roots of allopathic medicine trace back to ancient civilizations such as Egypt, Greece, India, and China, where early practitioners used herbal and mineral substances to treat illnesses. The foundations of modern allopathy, however, were laid during the time of Hippocrates (460–370 BCE), often called the “Father of Medicine,” who emphasized rational observation and diagnosis [6]. In the Middle Ages, medical knowledge in Europe was preserved in monasteries and later advanced through Arab scholars like Avicenna, whose “Canon of Medicine” remained a key medical text for over 600 years [7].

The transformation of allopathic medicine began during the Renaissance, when scientific methods and anatomy studies gained momentum. The invention of the printing press allowed the rapid spread of medical knowledge. The 19th century witnessed significant medical breakthroughs including the discovery of anesthesia, antisepsis by Joseph Lister, and the germ theory of disease by Louis Pasteur [8][9]. The 20th century marked a revolution in modern medicine through the invention of antibiotics (e.g., penicillin by Alexander Fleming), the development of vaccines, and the synthesis of modern pharmaceutical compounds. Regulatory bodies such as the U.S. FDA (1906) and India’s CDSCO were established to ensure drug safety and efficacy [10][11].

Medical education also became standardized, as seen in the Flexner Report of 1910 in the U.S., which reshaped curricula and established scientific foundations in medical schools [12]. Today, allopathy continues to evolve with innovations such as targeted therapies, nanomedicine, pharmacogenomics, and artificial intelligence in diagnostics and drug design [13][14].

CLASSIFICATION OF ALLOPATHIC FORMULATIONS

Allopathic formulations are systematically classified based on their dosage form, route of administration, release characteristics, and intended use. Proper classification ensures the correct application, patient compliance, and therapeutic success.

Based on Dosage Form:

  1. Solid Dosage Forms: Tablets, capsules, powders, granules [15].
  2. Liquid Dosage Forms: Syrups, solutions, suspensions, emulsions [15].
  3. Semi-solid Dosage Forms: Creams, ointments, gels, pastes [16].
  4. Gaseous Dosage Forms: Aerosols, inhalers, sprays [17].

Based on Route of Administration:

  1. Oral – Tablets, capsules, liquids.
  2. Parenteral – Intravenous (IV), intramuscular (IM), subcutaneous (SC) injections.
  3. Topical – Ointments, creams, patches.
  4. Inhalation – Nebulizers, metered dose inhalers.
  5. Rectal/Vaginal – Suppositories, pessaries.

Based on Release Characteristics:

  1. Immediate Release (IR) – Rapid action.
  2. Sustained Release (SR) – Gradual release over time.
  3. Controlled Release (CR) – Predictable, uniform release.
  4. Delayed Release (DR) – Action begins after a specific time or condition [18].

Based on Therapeutic Use:

  1. Analgesics – Pain relief.
  2. Antibiotics – Infection treatment.
  3. Antihypertensives – Blood pressure control.
  4. Antipyretics – Fever control.
  5. Anti-inflammatory agents, antacids, sedatives, etc.

Understanding these classifications allows pharmaceutical scientists to design patient-centric, targeted drug delivery systems that improve outcomes and reduce side effects.

ROUTES OF DRUG ADMINISTRATION

The route of drug administration significantly influences the drug’s absorption, onset of action, bioavailability, and therapeutic effect. The selection depends on the drug’s physicochemical properties, disease condition, and patient factors.

Oral Route

  • Definition: Administration of drugs by mouth.
  • Forms: Tablets, capsules, syrups, suspensions.
  • Advantages: Convenient, safe, economical.
  • Limitations: First-pass metabolism, slower onset [19].

Parenteral Route

  • Definition: Administration by injection, bypassing the GI tract.
  • Types: Intravenous (IV), intramuscular (IM), subcutaneous (SC).
  • Advantages: Rapid onset, suitable for emergencies.
  • Limitations: Invasive, requires trained personnel [20].

Topical Route

  • Definition: Application of drugs directly to the skin or mucous membranes.
  • Forms: Creams, ointments, gels, patches.
  • Advantages: Localized effect, minimal systemic side effects.
  • Limitations: Limited absorption, potential for irritation [21].

Inhalational Route

  • Definition: Delivery of drugs to the lungs through aerosols or gases.
  • Forms: Inhalers, nebulizers.
  • Advantages: Rapid action, especially for respiratory diseases.
  • Limitations: Requires coordination, device dependency [22].

Rectal and Vaginal Routes

  • Definition: Administration through rectum or vagina.
  • Forms: Suppositories, enemas, pessaries.
  • Advantages: Useful when oral route is not possible.
  • Limitations: Variable absorption, discomfort [23].

Other Routes

  • Transdermal: Through skin (patches) – for sustained drug delivery.
  • Sublingual/Buccal: Under the tongue/cheek – for rapid systemic absorption.
  • Ocular/Otic/Nasal: Localized delivery for eyes, ears, and nasal cavities [24].
  • Selection of the appropriate route is essential to achieve therapeutic goals with minimal side effects.

EXCIPIENTS USED IN ALLOPATHIC FORMULATIONS

Excipients are the non-active ingredients added to a drug formulation to aid in manufacturing, stability, administration, and patient acceptability. Although they have no therapeutic effect, they play a critical role in drug delivery and performance.

Classification of Excipients

  • Diluents/Fillers: Add bulk to tablets or capsules (e.g., lactose, microcrystalline cellulose) [25].
  • Binders: Help hold ingredients together in a tablet (e.g., starch, polyvinylpyrrolidone) [26].
  • Disintegrants: Facilitate tablet breakup for absorption (e.g., sodium starch glycolate, crospovidone) [27].
  • Lubricants: Reduce friction during tablet manufacturing (e.g., magnesium stearate, stearic acid) [28].
  • Glidants: Enhance flow properties of powders (e.g., colloidal silicon dioxide) [28].
  • Preservatives: Prevent microbial growth in liquid formulations (e.g., parabens, benzalkonium chloride) [29].
  • Sweeteners and Flavoring Agents: Improve taste (e.g., aspartame, sucrose, menthol) [30].
  • Coloring Agents: Provide distinctive appearance (e.g., titanium dioxide, iron oxides) [30].

Functions and Importance

  • Ensure stability and shelf life.
  • Enhance patient compliance.
  • Aid in drug release and bioavailability.
  • Allow accurate dosing and reproducibility [31].

Selection Criteria for Excipients

  • Compatibility with the active drug.
  • Safety and toxicity profile.
  • Regulatory acceptance (e.g., USP/NF standards).
  • Stability under formulation conditions [32].
  • Proper excipient selection is vital for a successful allopathic formulation.

QUALITY CONTROL AND QUALITY ASSURANCE IN FORMULATIONS

Quality control (QC) and quality assurance (QA) are essential pillars in the pharmaceutical industry to ensure the safety, efficacy, and consistency of allopathic drug formulations.

Quality Control (QC)

  • Definition: Operational techniques and activities used to fulfill quality requirements [33].
  • Activities Include:
  • Testing raw materials, in-process materials, and final products.
  • Stability testing.
  • Validation of analytical methods [34].
  • Objective: To detect and eliminate defects in the finished product.

Quality Assurance (QA)

  • Definition: A wide-ranging concept covering all matters that influence the quality of a product [35].
  • Activities Include:
  • Standard Operating Procedures (SOPs).
  • Documentation and batch records.
  • Internal audits and compliance checks [36].
  • Objective: To prevent defects through planned and systematic activities.

Importance in Allopathic Formulations

  • Ensures product meets regulatory standards (e.g., FDA, WHO).
  • Increases patient safety and confidence.
  • Reduces recalls, failures, and production costs [37].

Regulatory Guidelines

  • Good Manufacturing Practices (GMP) – Enforced by FDA, EMA, and WHO.
  • ICH Guidelines – Provide quality, safety, and efficacy standards internationally.
  • ISO Standards – For quality management systems [38].

Tools and Techniques

  • In-process control (IPC).
  • Validation – Process, cleaning, analytical.
  • Risk Assessment – FMEA, HACCP [39].
  • Maintaining high standards of QC and QA is non-negotiable in modern allopathic pharmaceutical production.

STABILITY TESTING OF ALLOPATHIC FORMULATIONS

Stability testing is a key component in the development and maintenance of pharmaceutical formulations. It ensures that a drug maintains its identity, strength, quality, and purity throughout its shelf life.

Objectives of Stability Testing

  • Determine shelf life and storage conditions.
  • Understand the degradation pathways of the drug.
  • Ensure therapeutic efficacy and patient safety [40].

Types of Stability Testing

  • Long-term Testing: Conducted under recommended storage conditions (e.g., 25°C ± 2°C / 60% RH ± 5%).
  • Accelerated Testing: Simulates extreme conditions to predict long-term stability (e.g., 40°C ± 2°C / 75% RH ± 5%).
  • Intermediate Testing: Performed when accelerated conditions fail [41].

Parameters Evaluated5

  • Appearance (color, clarity)
  • pH
  • Assay (potency)
  • Degradation products
  • Dissolution (for solid dosage forms)
  • Microbial load (for sterile products) [42]

Regulatory Guidelines

  • ICH Q1A(R2): Harmonized guideline for stability testing.
  • WHO Stability Guidelines
  • USFDA, EMA, and other local agencies’ requirements [43].

Packaging Considerations

  • Stability is influenced by the choice of packaging materials (e.g., glass, plastic, blister packs).
  • Packaging must protect the formulation from moisture, light, and temperature fluctuations [44].

Documentation and Reporting

  • Results must be documented in a stability summary report.
  • Data must be submitted to regulatory agencies for product approval.
  • Any deviations must be thoroughly investigated [45].

PACKAGING OF ALLOPATHIC MEDICINES

Packaging plays a vital role in preserving the quality, stability, and identity of pharmaceutical formulations. It provides protection against physical, chemical, and biological degradation, and also delivers product information for patient compliance[46].

Objectives of Pharmaceutical Packaging

  • Protection from environmental factors (moisture, light, air) [47].
  • Prevention of contamination and microbial invasion.
  • Containment of the drug product during transport and storage.
  • Convenience in dispensing and use.
  • Communication of essential drug information to users [48].

Types of Packaging

  • Primary Packaging: Comes in direct contact with the drug (e.g., blister packs, bottles, tubes) [49].
  • Secondary Packaging: Provides additional protection (e.g., cartons, boxes).
  • Tertiary Packaging: Used for bulk handling and transport (e.g., shipping containers).

Packaging Materials

  • Glass: Chemically inert, used for injectable and oral liquids.
  • Plastic: Lightweight and versatile, used for tablets and capsules.
  • Metal: Aluminum foils for blister packs, tubes.
  • Paper and Cardboard: For secondary and tertiary packaging [50].

Advanced Packaging Technologies

  • Child-Resistant Packaging: Prevents accidental ingestion by children.
  • Tamper-Evident Packaging: Indicates any unauthorized access.
  • Unit Dose Packaging: Improves dosage accuracy and hygiene [51].
  • Smart Packaging: Includes sensors, indicators, and QR codes for real-time data [52].

Regulatory Requirements

  • Must comply with guidelines set by regulatory bodies like the FDA, WHO, EMA, and USP.
  • Labeling must include dosage instructions, batch number, expiry date, and storage conditions [53].
  • Packaging materials must be tested for leachability, compatibility, and toxicity [54].

Packaging and Patient Compliance

  • Attractive and easy-to-use packaging improves adherence.
  • Clear labeling reduces the risk of medication errors.
  • Blister packs, calendar packs, and color-coding help patients follow complex regimens [55].

Effective packaging is not just a protective barrier—it is an integral part of the pharmaceutical formulation process that ensures safety, compliance, and efficacy.

REGULATORY GUIDELINES FOR ALLOPATHIC FORMULATIONS

Regulatory guidelines ensure the safety, efficacy, and quality of allopathic formulations. Compliance with these guidelines is mandatory for approval and marketing of pharmaceutical products.

Importance of Regulatory Compliance

  • Guarantees public health protection [56].
  • Standardizes product quality across countries.
  • Ensures ethical and scientific manufacturing practices.
  • Facilitates international trade of pharmaceuticals [57].

Major Regulatory Authorities

  • USFDA (United States Food and Drug Administration) – Oversees drug approval and post-marketing surveillance in the USA.
  • EMA (European Medicines Agency) – Regulates drugs within the EU.
  • CDSCO (Central Drugs Standard Control Organization) – India’s national regulatory body.
  • MHRA (Medicines and Healthcare products Regulatory Agency) – UK regulator.
  • WHO (World Health Organization) – Issues international guidelines and prequalification programs [58].

Key Regulatory Documents

  • Common Technical Document (CTD): Standardized format for regulatory submissions.
  • Investigational New Drug (IND) Application.
  • New Drug Application (NDA).
  • Abbreviated New Drug Application (ANDA) for generics [59].

Regulatory Phases in Drug Development

  1. Preclinical Studies: Animal and lab tests for safety.
  2. Clinical Trials (Phases I–IV): Human testing for safety, efficacy, and adverse effects.
  3. Submission of Regulatory Dossier.
  4. Post-marketing Surveillance (Pharmacovigilance) [60].

ICH Guidelines

  • The International Council for Harmonisation (ICH) creates harmonized global standards.
  • Key areas: Quality (Q), Safety (S), Efficacy (E), and Multidisciplinary (M).
  • Examples:
  • ICH Q8 – Pharmaceutical development.
  • ICH Q9 – Quality risk management.
  • ICH Q10 – Pharmaceutical quality system [61].

GMP and GCP

  • Good Manufacturing Practices (GMP): Ensures proper design, monitoring, and control of manufacturing processes.
  • Good Clinical Practices (GCP): Protects the rights and safety of human subjects in trials [62].

Global Harmonization Challenges

  • Variation in country-specific requirements.
  • Language barriers.
  • Different levels of technical capacity among regulatory agencies.
  • Regulatory guidelines form the backbone of pharmaceutical development and play a critical role in ensuring drug safety, quality, and availability to patients.

RECENT ADVANCES IN ALLOPATHIC FORMULATIONS

Advances in pharmaceutical science have led to the development of more effective, patient-friendly, and targeted allopathic drug formulations. These innovations improve therapeutic outcomes, minimize side effects, and enhance patient compliance.

Nanotechnology-Based Drug Delivery

  • Utilizes nanoparticles, liposomes, and nanoemulsions to enhance bioavailability and target delivery.
  • Examples: Liposomal doxorubicin for cancer therapy, nanocarriers for poorly soluble drugs [63].

Controlled and Sustained Release Systems

  • Deliver drugs over extended periods to maintain steady plasma levels.
  • Use polymeric matrices, osmotic pumps, and microspheres.
  • Improves compliance, reduces dosing frequency, and minimizes peaks and troughs [64].

Orally Disintegrating Tablets (ODTs)

  • Disintegrate rapidly in the mouth without water.
  • Beneficial for pediatric, geriatric, and psychiatric patients.
  • Use of superdisintegrants like crospovidone, sodium starch glycolate [65].

Transdermal Drug Delivery Systems (TDDS)

  • Delivers drugs across the skin for systemic effects.
  • Examples: Nicotine, fentanyl, and estradiol patches.
  • Avoids first-pass metabolism and provides sustained action [66].

3D Printing in Pharmaceuticals

  • Enables precise customization of drug dosage forms.
  • Can produce polypills containing multiple drugs.
  • FDA-approved example: Spritam® (levetiracetam) [67].

Smart Drug Delivery Systems

  • Respond to physiological stimuli such as pH, temperature, or enzymes.
  • Example: Glucose-responsive insulin delivery systems.
  • Improves precision and reduces side effects [68].

Biologics and Biosimilars

  • Complex molecules like monoclonal antibodies, peptides, and vaccines.
  • Require special formulation strategies to maintain stability.
  • Examples: Insulin analogs, adalimumab (biologic), and its biosimilar counterparts [69].

Personalized Medicine and Pharmacogenomics

  • Tailors therapy based on individual genetic profiles.
  • Optimizes drug selection and dosage to improve efficacy and safety.
  • Especially useful in oncology and psychiatry [70].

Artificial Intelligence in Formulation Design

  • AI and machine learning tools optimize formulation parameters and predict outcomes.
  • Speeds up development and reduces costs.
  • Example: Deep learning for predicting solubility and permeability [71].

Modern formulation technologies have revolutionized the allopathic pharmaceutical landscape, leading to better-targeted, safer, and more effective therapies.

CHALLENGES IN FORMULATION DEVELOPMENT

Despite technological advancements, formulation development in allopathy continues to face significant challenges. These hurdles can impact drug stability, efficacy, patient compliance, regulatory approval, and commercial viability.

Poor Solubility and Bioavailability

  • Many new drug candidates are poorly water-soluble, leading to low absorption.
  • Requires complex techniques like solid dispersions, nanocrystals, or lipid-based systems [72].

Stability Concerns

  • Physical, chemical, and microbiological stability must be ensured.
  • Common problems: hydrolysis, oxidation, polymorphic changes, and microbial growth.
  • Needs careful selection of excipients, packaging, and storage conditions [73].

Patient Compliance

  • Bitter taste, large tablet size, or frequent dosing can reduce compliance.
  • Taste-masking, extended-release, and user-friendly dosage forms (e.g., ODTs) are required [74].

Regulatory Complexity

  • Varying regulatory requirements across countries.
  • Extensive documentation and validation needed.
  • Delays in approval due to non-compliance or insufficient data [75].

Manufacturing Challenges

  • Scale-up from lab to industrial production introduces variability.
  • Need for process validation, quality control, and equipment suitability.
  • Risk of cross-contamination and batch failures [76].

High Development Costs

  • Cost of preclinical studies, clinical trials, and regulatory compliance is high.
  • Especially burdensome for small companies.
  • Risk of project failure due to formulation inefficiencies [77].

Drug-Excipient Incompatibility

  • Excipients can react with APIs, leading to degradation or reduced activity.
  • Requires compatibility testing during formulation design [78].

Personalized Medicine Challenges

  • Need for custom formulations based on genetic profiles.
  • Difficult to implement on a large scale.
  • Limited availability of predictive biomarkers [79].

Environmental and Sustainability Concerns

  • Use of organic solvents and non-biodegradable materials poses environmental risks.
  • Push for green chemistry and sustainable manufacturing practices [80].

Overcoming these challenges requires multidisciplinary collaboration, innovative technologies, regulatory expertise, and patient-centric design strategies.

INDUSTRIAL CASE STUDIES IN ALLOPATHIC FORMULATIONS

Industrial case studies provide practical insight into the formulation development of widely used allopathic drugs. These cases highlight formulation challenges, regulatory strategies, innovations, and successful market outcomes.

Case Study: Paracetamol (Acetaminophen) Tablets

  • Widely used analgesic and antipyretic.
  • Challenge: Bitter taste and compressibility.
  • Solution: Use of flavoring agents and granulation techniques to enhance flowability and compressibility.
  • Outcome: Marketed in various formulations including ODTs, effervescent tablets, and suspensions [81].

Case Study: Metformin Hydrochloride Extended-Release (ER)

  • Used in type 2 diabetes.
  • Challenge: High dose and gastrointestinal irritation.
  • Solution: Use of hydrophilic polymer matrices for sustained release.
  • Outcome: Improved patient compliance with once-daily dosing [82].

Case Study: Omeprazole Enteric-Coated Capsules

  • Proton pump inhibitor for gastric acid control.
  • Challenge: Acid-labile drug; degrades in stomach.
  • Solution: Enteric coating using pH-sensitive polymers.
  • Outcome: Enhanced stability and therapeutic efficacy [83].

Case Study: Amoxicillin-Clavulanic Acid Formulation

  • Combination antibiotic with β-lactamase inhibitor.
  • Challenge: Clavulanic acid instability in aqueous medium.
  • Solution: Dry powder formulation with co-packaging to maintain stability.
  • Outcome: Widely used in oral and injectable forms [84].

Case Study: Transdermal Fentanyl Patch

  • Potent opioid analgesic for chronic pain.
  • Challenge: Risk of overdose and skin irritation.
  • Solution: Rate-controlling membrane system with adhesive matrix.
  • Outcome: Long-acting, non-invasive pain relief [85].

Case Study: Insulin Analog Injections

  • Biologic drug for diabetes.
  • Challenge: Protein stability and aggregation.
  • Solution: pH adjustment, stabilizers, and cold-chain maintenance.
  • Outcome: Safe and effective subcutaneous administration [86].

Case Study: 3D Printed Levetiracetam (Spritam®)

  • Antiepileptic drug approved by FDA.
  • Challenge: High dose requiring large tablet.
  • Solution: ZipDose® 3D printing technology to produce porous ODT.
  • Outcome: Faster disintegration and ease of swallowing [87].

Case Study: Liposomal Amphotericin B (AmBisome®)

  • Antifungal drug with nephrotoxicity.
  • Challenge: Reduce toxicity while maintaining efficacy.
  • Solution: Encapsulation in liposomes.
  • Outcome: Reduced renal toxicity and improved delivery [88].

These real-world examples illustrate how formulation challenges are addressed using innovative strategies, leading to improved therapeutic outcomes and commercial success.

INTELLECTUAL PROPERTY RIGHTS IN ALLOPATHIC FORMULATIONS

Intellectual Property Rights (IPRs) play a crucial role in protecting innovative pharmaceutical formulations, encouraging research and development (R&D), and enabling companies to gain competitive advantages.

Types of Intellectual Property Rights

  • Patents: Grant exclusive rights for inventions (new formulations, processes).
  • Trademarks: Protect brand names and logos.
  • Copyrights: Protect written materials, software, and packaging designs.
  • Trade Secrets: Confidential business information not publicly disclosed [89].

Patents in Allopathic Drug Formulations

  • Critical for protecting formulation innovation.
  • Valid for 20 years from the filing date.
  • Types:
    • Composition patents
    • Process patents
    • Method-of-use patents
  • Example: Modified-release formulations often receive patent protection [90].

13.3 Patent Filing Process

  1. Novelty search
  2. Drafting of patent application
  3. Filing with national or international patent offices
  4. Examination and grant
  5. Post-grant opposition or revocation if applicable [91].

International Patent Protection

  • Governed by the Patent Cooperation Treaty (PCT).
  • Allows filing a single application recognized in multiple countries.
  • Important for global pharmaceutical companies [92].

TRIPS Agreement and Its Impact

  • Trade-Related Aspects of Intellectual Property Rights (TRIPS): Agreement under the WTO.
  • Sets minimum standards for IP protection globally.
  • Mandates patent protection for pharmaceuticals in all member countries [93].

Compulsory Licensing and Public Health

  • Allows governments to authorize production of patented drugs without consent during emergencies.
  • Example: India granted compulsory license for a cancer drug to ensure affordability [94].

Patent Cliff and Generic Entry

  • After patent expiry, generics enter the market.
  • Leads to sharp revenue decline for innovator companies.
  • Strategy: Patent evergreening by slight modifications to extend exclusivity [95].

Challenges in IPR Enforcement

  • Patent infringement disputes and litigation.
  • Issues with counterfeit drugs.
  • High cost of maintaining patents in multiple jurisdictions [96].

IPR in India

  • Governed by the Indian Patent Act, 1970, amended in 2005.
  • India recognizes product patents for pharmaceuticals.
  • Patent filing through Controller General of Patents, Designs & Trade Marks (CGPDTM) [97].

IPRs in pharmaceutical formulations are vital for protecting innovation, encouraging investment, and balancing public access to essential medicines.

Ensuring product quality is critical in pharmaceutical formulation to guarantee efficacy, safety, and patient compliance. Quality Control (QC) and Quality Assurance (QA) form the backbone of Good Manufacturing Practices (GMP).

Definitions

  • Quality Control (QC): Analytical testing to ensure product specifications are met.
  • Quality Assurance (QA): Systematic activities ensuring that quality requirements will be fulfilled [98].

Objectives

  • Prevent defects in drug formulation.
  • Ensure consistency in batch production.
  • Comply with regulatory requirements.
  • Ensure safety and efficacy of drug products [99].

Components of Quality Control

  • Raw Material Testing: Identity, purity, and strength checks.
  • In-Process Control: Monitoring during production.
  • Finished Product Testing: Assay, dissolution, sterility, etc.
  • Stability Testing: Product behavior under various conditions [100].

Components of Quality Assurance

  • Documentation and Record Keeping: SOPs, batch records, audits.
  • Validation and Qualification: Equipment and process validation.
  • Deviation and Change Control: Handling out-of-specification results.
  • Self-Inspection and Audits: Ensuring internal compliance [101].

Good Manufacturing Practices (GMP)

  • Mandated by WHO, USFDA, and other regulatory bodies.
  • Covers facility design, personnel hygiene, documentation, training, and quality systems.
  • Ensures pharmaceutical products are consistently produced and controlled [102].

Role of QMS (Quality Management System)

  • Framework that integrates QC and QA.
  • Promotes continual improvement.
  • Examples: ISO 9001, ICH Q10 guidelines [103].

Regulatory Guidelines

  • WHO GMP Guidelines
  • USFDA cGMP (21 CFR Part 210 & 211)
  • EU GMP Annex 15
  • Schedule M (India) [104]

Common Quality Defects and Prevention

  • Tablet capping, lamination, contamination
  • Microbial growth in suspensions or creams
  • Prevented through proper environmental monitoring, process control, and validation [105].

Quality Control and Quality Assurance ensure that pharmaceutical formulations meet international standards and are safe for patient use throughout the product lifecycle.

BIOPHARMACEUTICAL CONSIDERATIONS IN FORMULATION DEVELOPMENT

Biopharmaceutics is the study of how a drug’s physical and chemical properties, dosage form, and route of administration affect its absorption, distribution, metabolism, and excretion (ADME).

Importance in Formulation

Biopharmaceutical studies help optimize drug delivery by enhancing bioavailability, minimizing variability, and improving therapeutic effectiveness [106].

Key Parameters

  • Solubility: Affects drug absorption rate.
  • Permeability: Ability of drug to cross biological membranes.
  • Dissolution Rate: Speed at which drug dissolves in bodily fluids.
  • Stability in GI Tract: Resistance to pH or enzymatic degradation [107].

BCS Classification

Biopharmaceutical Classification System (BCS) classifies drugs into four categories based on solubility and permeability:

  • Class I: High solubility, high permeability (e.g., paracetamol)
  • Class II: Low solubility, high permeability (e.g., ketoconazole)
  • Class III: High solubility, low permeability (e.g., cimetidine)
  • Class IV: Low solubility, low permeability (e.g., hydrochlorothiazide) [108]

Formulation Strategies Based on BCS

  • Class II drugs: Use of solubilizers, particle size reduction, solid dispersions.
  • Class III drugs: Use of permeability enhancers or prodrugs.
  • Class IV drugs: Need advanced drug delivery systems [109].

Bioavailability and Bioequivalence

  • Bioavailability: The rate and extent of drug absorption.
  • Bioequivalence: Comparison between test and reference formulations.
  • Required for generic drug approval [110].

In Vitro–In Vivo Correlation (IVIVC)

  • Establishes predictive relationship between in vitro dissolution and in vivo absorption.
  • Levels A, B, and C defined by FDA.
  • Helps in formulation optimization and reduces need for clinical trials [111].

Role in Modified Release Dosage Forms

  • Understanding drug kinetics is essential to design sustained or controlled release systems.
  • Ensures therapeutic drug levels over time [112].

Biopharmaceutical Tools

  • GastroPlus, Simcyp: Simulation software.
  • Permeation studies: Caco-2 cell lines, PAMPA.
  • In vitro models: Dissolution testing, diffusion cells [113].

Biopharmaceutical considerations are critical in designing effective allopathic drug formulations and meeting regulatory requirements for bioequivalence.

FUTURE TRENDS IN ALLOPATHIC FORMULATION DEVELOPMENT

The landscape of allopathic drug formulation is continuously evolving with the integration of advanced technologies and personalized medicine approaches. Future trends aim to enhance drug efficacy, patient compliance, and manufacturing efficiency.

Personalized Medicine

  • Tailoring drug formulations based on patient’s genetic profile.
  • Pharmacogenomics-guided dosing to minimize adverse effects.
  • Customized drug delivery devices [114].

3D Printing in Pharmaceuticals

  • Enables personalized drug dosing and complex release profiles.
  • Used for creating polypills and multi-layer tablets.
  • FDA-approved 3D-printed drug: Spritam (levetiracetam) [115].

Artificial Intelligence (AI) and Machine Learning

  • Predicts drug stability, solubility, and optimal excipients.
  • Facilitates virtual screening of formulation parameters.
  • AI-integrated Quality by Design (QbD) for faster development [116].

Smart Drug Delivery Systems

  • Use of stimuli-responsive systems (pH, temperature, enzymes).
  • Incorporation of biosensors for real-time drug monitoring.
  • Smart polymers for controlled and targeted release [117].

Nanotechnology

  • Development of nanosuspensions, nanogels, and nanocapsules.
  • Increases solubility, bioavailability, and targeted delivery.
  • Applications in cancer, CNS disorders, and vaccine delivery [118].

Green Chemistry in Formulation

  • Use of eco-friendly solvents and biodegradable excipients.
  • Energy-efficient manufacturing processes.
  • Focus on sustainable pharmaceutical development [119].

Blockchain in Pharmaceutical Supply Chain

  • Enhances traceability, transparency, and security.
  • Prevents counterfeit drugs.
  • Improves regulatory compliance [120].

Continuous Manufacturing

  • Replaces traditional batch processing.
  • Reduces production time and increases consistency.
  • Enables real-time quality monitoring and control [121].

These advancements are expected to redefine the future of allopathic drug formulation, making therapies more effective, safe, and tailored to individual needs.

ENVIRONMENTAL AND ETHICAL CONSIDERATIONS IN ALLOPATHIC FORMULATIONS

Modern pharmaceutical development is increasingly focusing on environmental sustainability and ethical responsibility. These considerations are essential for minimizing the ecological impact and ensuring public trust.

Environmental Impact of Pharmaceutical Manufacturing

  • Waste Generation: Includes solvents, intermediates, and APIs.
  • Water Contamination: Drug residues often found in rivers and lakes.
  • Air Emissions: Volatile organic compounds (VOCs) released during synthesis [122].

Green Chemistry Approaches

  • Use of environmentally benign solvents.
  • Catalytic instead of stoichiometric reagents.
  • Process intensification to reduce energy and materials [123].

Biodegradable Excipients

  • Natural polymers such as chitosan, alginate, and starch-based carriers.
  • Reduce accumulation of non-degradable materials.
  • Safer for patients and the environment [124].

Ethical Considerations

  • Animal Testing: Replaced where possible by in vitro and in silico models.
  • Informed Consent: Required for clinical trials.
  • Patient Privacy: Secure handling of personal health data [125].

Regulatory Compliance

  • Guidelines from US FDA, EMA, and CPCB in India for waste disposal.
  • Environmental risk assessments mandatory for new drugs.
  • Adherence to Good Environmental Practices (GEP) [126].

Corporate Social Responsibility (CSR)

Pharmaceutical firms increasingly engage in CSR initiatives.Support health education, access to essential medicines, and local environmental protection [127].

Sustainable Packaging

  • Recyclable and biodegradable packaging materials.
  • Reduction of single-use plastics.
  • Compliance with eco-labelling standards [128].

Global Initiatives

  • UN Sustainable Development Goals (SDGs): Promote responsible consumption and production.
  • Pharmaceutical Supply Chain Initiative (PSCI): Encourages ethical supply chain management [129].

These practices ensure that allopathic drug formulation remains aligned with global sustainability and ethical benchmarks.

CLINICAL TRIALS AND FORMULATION ASSESSMENT

Clinical trials are a crucial step in evaluating the safety, efficacy, and acceptability of allopathic formulations. These trials ensure that the formulation performs as expected in human subjects before regulatory approval.

Preclinical Formulation Evaluation

  • Conducted on animals or using in vitro methods.
  • Assesses stability, bioavailability, and toxicity.
  • Guides formulation refinement before human trials [130].

Phases of Clinical Trials

  • Phase I: Tests safety and dosage in healthy volunteers.
  • Phase II: Evaluates efficacy and side effects in patients.
  • Phase III: Large-scale trials for confirming efficacy and monitoring adverse reactions.
  • Phase IV: Post-marketing surveillance and long-term safety data [131].

Bioavailability and Bioequivalence Studies

  • Measure the rate and extent of drug absorption.
  • Required for generic formulations.
  • Uses parameters like Cmax, Tmax, and AUC [132].

Patient Compliance and Acceptability

  • Taste, size, dosing frequency, and side effects affect adherence.
  • Trials often include patient feedback surveys.
  • Reformulation may be required based on results [133].

Formulation Stability Studies

  • Evaluate physical, chemical, microbiological, and therapeutic stability.
  • Conducted under ICH guidelines for various storage conditions.
  • Determines shelf life and packaging needs [134].

Regulatory Guidelines for Trials

  • Governed by ICH-GCP, CDSCO (India), US FDA, and EMA.
  • Ethical committee approval and informed consent are mandatory.
  • Data integrity and transparency must be ensured [135].

Role of Clinical Pharmacologists

  • Ensure rational formulation design and dosing.
  • Monitor pharmacokinetics and pharmacodynamics in trials.
  • Support interpretation of clinical outcomes [136].

These assessments ensure that the formulation meets regulatory standards and provides maximum benefit to the patients with minimal risks.

ROLE OF NANOTECHNOLOGY IN ALLOPATHIC FORMULATIONS

Nanotechnology has revolutionized drug delivery systems by improving bioavailability, targeting, and controlled release. It enables the development of novel allopathic formulations with enhanced therapeutic profiles.

Introduction to Nanotechnology in Pharmaceutics

  • Deals with particles sized 1–100 nm.
  • Nanocarriers include liposomes, dendrimers, nanoparticles, and nanoemulsions.
  • Offers site-specific drug delivery and reduced toxicity [137].

Types of Nanocarriers

  • Liposomes: Phospholipid vesicles for hydrophilic and hydrophobic drugs.
  • Polymeric Nanoparticles: Biodegradable carriers for sustained release.
  • Solid Lipid Nanoparticles (SLNs): Enhance stability of sensitive drugs.
  • Dendrimers: Branched nanostructures for precise delivery [138].

Advantages of Nanotechnology-Based Formulations

  • Improved drug solubility and absorption.
  • Enhanced permeability across biological membranes.
  • Reduced dosing frequency and side effects.
  • Better patient compliance [139].

Challenges in Nanotechnology-Based Formulations

  • Complex and expensive manufacturing.
  • Potential cytotoxicity and immunogenicity.
  • Regulatory hurdles due to lack of standardized guidelines [140].

Clinical Applications

  • Oncology: Targeted delivery of anticancer agents.
  • Neurology: Brain targeting via nanoparticulate systems.
  • Anti-infective therapy: Enhanced delivery of antibiotics and antivirals [141].

Regulatory and Quality Control Aspects

  • Need for characterization of size, shape, surface charge, and entrapment efficiency.
  • US FDA and EMA provide evolving guidelines for nanopharmaceuticals.
  • Requires preclinical safety and toxicology evaluation [142].

Future Perspectives

  • Personalized nanomedicine based on genetic profiling.
  • Smart nanocarriers responding to physiological triggers.
  • Integration with AI for design and optimization [143].

Nanotechnology offers vast potential in formulating next-generation allopathic medicines that are safer, more effective, and patient-friendly.

PERSONALIZED MEDICINE AND FORMULATION TAILORING

Personalized medicine aims to deliver treatments tailored to individual patient characteristics such as genetics, environment, and lifestyle. In allopathic formulation development, this approach opens new avenues for precision and efficacy.

Introduction to Personalized Medicine

  • Focuses on patient-specific therapy based on biomarkers and genomics.
  • Minimizes adverse effects and enhances therapeutic outcomes [144].

Pharmacogenomics in Formulation Design

  • Understanding genetic variations (e.g., CYP450 enzymes) guides drug choice and dose.
  • Formulations are adjusted to match metabolic profiles [145].

Role of Companion Diagnostics

  • Diagnostic tools used alongside formulations.
  • Help identify responders and non-responders to certain drugs.
  • Support decision-making for personalized treatments [146].

Custom Dosage Forms

  • 3D printing of tablets: Allows dose variation per patient.
  • Modular drug delivery systems: Tailored release patterns.
  • Multi-drug layering: Combines therapies based on patient needs [147].

Benefits of Formulation Tailoring

  • Improved efficacy and patient compliance.
  • Reduction in trial-and-error prescribing.
  • Better outcomes in chronic and complex diseases [148].

Regulatory and Ethical Challenges

  • High cost and complexity of implementation.
  • Need for patient data privacy and ethical use.
  • Limited standardized guidelines for customized products [149].

Future Outlook

  • Integration of AI and big data in formulation design.
  • Development of biosensors and wearable drug delivery systems.
  • Greater collaboration among pharma, diagnostics, and IT sectors [150].

Personalized medicine is shifting the paradigm in allopathic formulation from a one-size-fits-all to a precision-based strategy, optimizing both drug safety and effectiveness.

FUTURE PROSPECTS

The future of allopathic formulation development is evolving rapidly due to advancements in technology, personalized medicine, regulatory frameworks, and global healthcare demands. This chapter summarizes emerging trends, key opportunities, and the overall significance of innovations in formulation science.

Integration of Digital Health

  • Incorporation of wearable devices and digital monitoring for real-time drug delivery feedback.
  • Smart formulations responsive to physiological changes [151].

Expansion of Personalized Formulations

  • Wider adoption of 3D-printed and genetically tailored medicines.
  • Use of big data to predict formulation needs per patient [152].

Green and Sustainable Formulations

  • Use of eco-friendly excipients and solvents.
  • Biodegradable packaging and energy-efficient manufacturing [153].

AI and Automation in Manufacturing

  • Predictive analytics for scale-up and process control.
  • Robotic systems for continuous and flexible production [154].

Globalization and Decentralized Production

  • Shift towards local production using modular, portable facilities.
  • Ensures access in low-resource and remote settings [155].

CONCLUSIONS

The field of allopathic formulation has significantly evolved, driven by advancements in pharmaceutical sciences, regulatory insights, and patient-centric innovations. Over the past few decades, the transition from traditional dosage forms to highly specialized, targeted, and intelligent drug delivery systems have redefined therapeutic strategies. This comprehensive review outlined the critical components of formulation development, including preformulation studies, excipient selection, nanotechnology applications, regulatory aspects, and modern trends such as AI-driven optimization and personalized medicine. Formulators today are not only focused on drug efficacy and safety but also on improving patient compliance, manufacturing scalability, and environmental sustainability. The integration of advanced technologies such as artificial intelligence, 3D printing, and continuous manufacturing is paving the way for faster and more precise development cycles. Meanwhile, global health challenges emphasize the need for adaptable, cost-effective, and accessible formulations. Moving forward, collaborative research, multidisciplinary expertise, and policy support will be essential to overcoming the challenges of drug resistance, variable bioavailability, and global accessibility. Allopathic formulations will continue to be a cornerstone of modern medicine, and ongoing innovations will ensure their relevance in an ever-changing healthcare landscape. The future holds promise for safer, more effective, and patient-specific therapeutic solutions that cater to diverse medical needs.

REFERENCES

  1. World Health Organization. WHO Drug Information. [Online].
  2. Rang HP, Dale MM, Ritter JM, Flower RJ. Rang and Dale’s Pharmacology. Elsevier.
  3. Kumar V, Abbas AK, Aster JC. Robbins and Cotran Pathologic Basis of Disease. Elsevier.
  4. Tripathi KD. Essentials of Medical Pharmacology. Jaypee Brothers.
  5. Hahnemann S. Organon of Medicine. Boericke & Tafel.
  6. Nutton V. Ancient Medicine. 2nd ed. Routledge; 2013.
  7. Pormann PE, Savage-Smith E. Medieval Islamic Medicine. Edinburgh University Press; 2007.
  8. Porter R. The Greatest Benefit to Mankind: A Medical History of Humanity. Harper Collins; 1999.
  9. Bynum WF. Science and the Practice of Medicine in the Nineteenth Century. Cambridge University Press; 1994.
  10. Weatherall MW. In Search of a Cure: A History of Pharmaceutical Discovery. Oxford University Press; 1990.
  11. Central Drugs Standard Control Organization. About CDSCO. Available from: https://cdsco.gov.in
  12. Flexner A. Medical Education in the United States and Canada. Carnegie Foundation; 1910.
  13. Ginsburg GS, Phillips KA. Precision Medicine: From Science to Value. Health Affairs. 2018;37(5):694–701.
  14. Rajan JV, Ramanathan M. Artificial Intelligence in Drug Discovery and Development. Trends Pharmacol Sci. 2021;42(10):813–827.
  15. Aulton ME, Taylor KMG. Aulton’s Pharmaceutics: The Design and Manufacture of Medicines. 5th ed. Elsevier; 2018.
  16. Allen LV. Ansel’s Pharmaceutical Dosage Forms and Drug Delivery Systems. 12th ed. Wolters Kluwer; 2021.
  17. Hickey AJ. Inhalation Aerosols: Physical and Biological Basis for Therapy. 3rd ed. CRC Press; 2019.
  18. Brahmankar HA, Jaiswal SB. Biopharmaceutics and Pharmacokinetics: A Treatise. 2nd ed. Vallabh Prakashan; 2009.
  19. Shargel L, Yu AB. Applied Biopharmaceutics and Pharmacokinetics. 7th ed. McGraw-Hill; 2016.
  20. Florence AT, Attwood D. Physicochemical Principles of Pharmacy. 6th ed. Pharmaceutical Press; 2015.
  21. Walters KA, Roberts MS. Dermatological and Transdermal Formulations. CRC Press; 2002.
  22. Labiris NR, Dolovich MB. Pulmonary drug delivery. Part I: Physiological factors. Clin Pharmacokinet. 2003;42(6):529–545.
  23. Brunton LL, Hilal-Dandan R, Knollmann BC. Goodman & Gilman’s: The Pharmacological Basis of Therapeutics. 13th ed. McGraw-Hill; 2018.
  24. Allen LV. Remington: The Science and Practice of Pharmacy. 22nd ed. Pharmaceutical Press; 2013.
  25. Aulton ME, Taylor KMG. Aulton’s Pharmaceutics: The Design and Manufacture of Medicines. 5th ed. Elsevier; 2018.
  26. Allen LV, Popovich NG, Ansel HC. Ansel’s Pharmaceutical Dosage Forms and Drug Delivery Systems. 10th ed. Wolters Kluwer; 2014.
  27. Kibbe AH. Handbook of Pharmaceutical Excipients. 6th ed. Pharmaceutical Press; 2009.
  28. Rowe RC, Sheskey PJ, Quinn ME. Handbook of Pharmaceutical Excipients. 7th ed. Pharmaceutical Press; 2012.
  29. Moldenhauer J. Pharmaceutical Cleanroom Technology. CRC Press; 2011. 30. Lachman L, Lieberman HA, Kanig JL. The Theory and Practice of Industrial Pharmacy. 3rd ed. Lea & Febiger; 1986.
  30. Mahato RI. Pharmaceutical Dosage Forms and Drug Delivery. 2nd ed. CRC Press; 2011.
  31. ICH Harmonised Tripartite Guideline. Stability Testing of New Drug Substances and Products Q1A(R2), ICH Steering Committee; 2003.
  32. WHO. Quality assurance of pharmaceuticals: a compendium of guidelines and related materials. WHO; 2007.
  33. United States Pharmacopeia (USP). General Chapters: <1225> Validation of Compendial Procedures.
  34. ICH Q10: Pharmaceutical Quality System. International Council for Harmonisation; 2008.
  35. FDA. Guidance for Industry: Quality Systems Approach to Pharmaceutical CGMP Regulations; 2006.
  36. EMA. Guidelines on Good Manufacturing Practice specific to Advanced Therapy Medicinal Products. European Medicines Agency; 2017.
  37. ISO 9001:2015 – Quality management systems – Requirements. International Organization for Standardization; 2015.
  38. Sandle T. Pharmaceutical Microbiology: Essentials for Quality Assurance and Quality Control. Elsevier; 2015.
  39. ICH Q1A(R2): Stability Testing of New Drug Substances and Products. ICH Steering Committee; 2003.
  40. FDA. Guidance for Industry: Stability Testing of Drug Substances and Drug Products; 2003.
  41. Aulton ME, Taylor KMG. Pharmaceutics: The Science of Dosage Form Design. 5th ed. Elsevier; 2018.
  42. WHO. Stability testing of active pharmaceutical ingredients and finished pharmaceutical products. WHO Technical Report Series; No. 1010, 2018.
  43. Collett JH, Aulton ME. Pharmaceutical Packaging Technology. Taylor & Francis; 2007. 45. Nash RA, Wachter AH. Pharmaceutical Process Validation. 3rd ed. Marcel Dekker; 2003.
  44. Allen LV. Remington: The Science and Practice of Pharmacy. 22nd ed. Pharmaceutical Press; 2013.
  45.  Aulton ME, Taylor KMG. Aulton’s Pharmaceutics: The Design and Manufacture of Medicines. 5th ed. Elsevier; 2018.
  46.  Rowe RC, Sheskey PJ, Quinn ME. Handbook of Pharmaceutical Excipients. 7th ed. Pharmaceutical Press; 2012.
  47. Felton LA. Remington Essentials of Pharmaceutics. Pharmaceutical Press; 2013.
  48.  United States Pharmacopeia (USP). <1> Injections; <661> Containers.
  49. Smart Packaging Market Report. Allied Market Research; 2020.
  50. FDA. Guidance for Industry: Container Closure Systems for Packaging Human Drugs and Biologics; 1999.
  51. WHO. Guidelines on packaging for pharmaceutical products. WHO Technical Report Series, No. 902, 2002. 55. EMA. Guidelines on the packaging of medicinal products for human use. European Medicines Agency; 2017.
  52. USFDA. Drug Development and Approval Process. Available at: https://www.fda.gov
  53.  EMA. Overview of regulatory responsibilities. European Medicines Agency. 58. WHO. Regulatory system strengthening. Available at: https://www.who.int 59. ICH. The Common Technical Document (CTD). https://www.ich.org
  54. USFDA. New Drug Application (NDA) process. https://www.fda.gov/drugs 61. ICH Guidelines Q8–Q10. International Council for Harmonisation. https://www.ich.org
  55. WHO. GMP and GCP standards. WHO Technical Report Series.
  56. Barenholz Y. Doxil®—the first FDA-approved nano-drug: lessons learned. J Control Release. 2012.
  57. Siepmann J, Siepmann F. Modeling of drug release from delivery systems based on hydroxypropyl methylcellulose (HPMC). Adv Drug Deliv Rev. 2012. 65. Fu Y, Yang S, Jeong SH, Kimura S, Park K. Orally fast disintegrating tablets: developments, technologies, and optimization. Drug Dev Ind Pharm. 2004.
  58. Prausnitz MR, Langer R. Transdermal drug delivery. Nat Biotechnol. 2008. 67. Goyanes A, et al. 3D printing of medicines: engineering novel oral drug products. Curr Pharm Des. 2015.
  59. Zhang Y, et al. Glucose-sensitive insulin release systems: synthesis and performance. J Biomed Mater Res. 2017.
  60. Schellekens H. Biosimilar therapeutics—what do we need to consider? NDT Plus. 2009. 70. Johnson JA. Pharmacogenetics in clinical practice: how far have we come and where are we going? Pharmacogenomics J. 2013. 71. Stokes JM, et al. A deep learning approach to antibiotic discovery. Cell. 2020.
  61. Lipinski CA. Poor aqueous solubility—an industry wide problem in drug discovery. Adv Drug Deliv Rev. 2002.
  62. Waterman KC, Adami RC. Accelerated aging: prediction of chemical stability of pharmaceuticals. Int J Pharm. 2005.
  63. Arya A, Chandra A, Sharma V, Pathak K. Fast dissolving oral films: an innovative drug delivery system and dosage form. Int J ChemTech Res. 2010. 75. International Conference on Harmonisation. Regulatory considerations for pharmaceutical development.
  64. Garcia CV, et al. Pharmaceutical manufacturing process validation: quality and regulatory perspectives. J Pharm Sci. 2018.
  65.  DiMasi JA, Grabowski HG, Hansen RW. Innovation in the pharmaceutical industry: new estimates of R&D costs. J Health Econ. 2016.
  66. Bhatt DA, Pethe AM. Drug-excipient compatibility screening. Asian J Pharm. 2010.
  67. Ginsburg GS, Phillips KA. Precision medicine: from science to value. Health Aff. 2018.
  68. Kümmerer K, Dionysiou DD, Olsson O, Fatta-Kassinos D. A path to clean pharmaceuticals and chemical manufacturing. Green Chem. 2018.
  69. Patel DM, Patel MM. Optimization of fast dissolving tablets of paracetamol using simple lattice design. Int J Pharm Sci. 2009.
  70. Ramprakash V, Varghese L. Design and evaluation of sustained release matrix tablets of metformin hydrochloride. Int J PharmTech Res. 2010.
  71. Shrivastava AR, et al. Formulation and evaluation of enteric-coated tablets of omeprazole. Indian J Pharm Sci. 2007.
  72. Ganesh M, et al. Formulation and evaluation of co-amoxiclav dry syrup. Int J Pharm Sci Res. 2012.
  73. Guo JH. Transdermal drug delivery systems for fentanyl. Pharm Tech. 1994. 86. Vajo Z, Duckworth WC. Genetically engineered insulin analogs: diabetes in the new millennium. Pharmacol Rev. 2000.
  74. Aprecia Pharmaceuticals. Spritam® (levetiracetam) tablets: FDA approval announcement. 2015. 88. Adler-Moore JP, Proffitt RT. AmBisome: liposomal formulation, structure, and stability. J Liposome Res. 1993.
  75. WIPO. Types of Intellectual Property. World Intellectual Property Organization.
  76. Gassmann O, Reepmeyer G. Organizing pharmaceutical innovation: from science-based knowledge creators to drug-oriented knowledge brokers. Creat Innov Manage. 2005.
  77. Kumar V, et al. Patent filing procedure in India. Int J Res Pharm Sci. 2012.
  78. WIPO. Patent Cooperation Treaty (PCT). Overview.
  79. World Trade Organization. TRIPS Agreement.
  80. Basheer S. India’s first compulsory license: Natco v. Bayer. SpicyIP Blog. 2012.
  81. Kapczynski A, et al. Polymorph patents and the law. N Engl J Med. 2012.
  82. Singh A. IPR challenges in pharma industry. J Pharm Sci Res. 2016.
  83. Indian Patent Office. Manual of Patent Office Practice and Procedure. Govt. Of India.
  84. Sharma PP. How to Practice GMPs. 5th Ed. Vandana Publications.
  85. Makhija R, et al. Concept of quality control in pharma. Int J Pharm Sci Rev Res. 2010. 100. ICH Q1A(R2). Stability Testing of New Drug Substances and Products.
  86. WHO. Quality assurance of pharmaceuticals: a compendium of guidelines and related materials. Vol. 2.
  87. USFDA. Guidance for Industry: GMP for Pharmaceuticals. 21 CFR Part 210 & 211. 103. ICH Q10. Pharmaceutical Quality System. International Conference on Harmonisation. 104. CDSCO. Schedule M – Good Manufacturing Practices for Pharmaceuticals in India.
  88. Patel M, et al. A review on quality defects in tablets and their control. Pharma Times. 2013.
  89. Amidon GL, et al. A theoretical basis for a biopharmaceutic drug classification. Pharm Res. 1995.
  90. Dressman JB, Reppas C. In vitro–in vivo correlations. Eur J Pharm Sci. 2000.
  91. FDA. Guidance for Industry: Waiver of In Vivo Bioavailability and Bioequivalence Studies for Immediate-Release Solid Oral Dosage Forms Based on a BCS.
  92. Shargel L, Yu ABC. Applied Biopharmaceutics and Pharmacokinetics. 6th ed.
  93. WHO. Bioavailability and Bioequivalence Guidelines.
  94. FDA. Guidance for Industry: Dissolution Testing of Immediate Release Solid Oral Dosage Forms.
  95. Robinson JR, Eriksen S. Theoretical considerations of controlled drug delivery. J Pharm Sci. 1971.
  96. Sugano K. Current status of in vitro permeability studies. Int J Pharm. 2009.
  97. Hamburg MA, Collins FS. The path to personalized medicine. N Engl J Med. 2010.
  98. Alhnan MA, et al. Emergence of 3D printed dosage forms: Opportunities and challenges. Adv Drug Deliv Rev. 2016.
  99.  Mak KK, Pichika MR. Artificial intelligence in drug development: Present status and future prospects. Drug Discov Today. 2019.
  100. Raza K, et al. Smart drug delivery system: A novel approach. Drug Deliv. 2014.
  101. Zhang L, et al. Nanoparticles in medicine: Therapeutic applications and developments. Clin Pharmacol Ther. 2008.
  102.  Anastas PT, Warner JC. Green Chemistry: Theory and Practice. Oxford University Press, 1998.
  103. Boehm E, et al. Blockchain technology in the pharmaceutical industry. J Pharm Sci. 2020.
  104. Lee SL, et al. Modernizing pharmaceutical manufacturing: from batch to continuous production. J Pharm Innov. 2015.
  105. Kümmerer K. Pharmaceuticals in the Environment: Sources, Fate, Effects. Springer, 2008.
  106. Sheldon RA. Green chemistry and resource efficiency. Green Chem. 2016.
  107. Varma AJ, et al. Polysaccharides: Natural biodegradable polymers. Carbohydr Polym. 2004.
  108. European Medicines Agency. Ethical considerations for clinical trials. 2018.
  109. Central Pollution Control Board (CPCB) India. Guidelines for Management of Waste from Pharmaceutical Industry. 2020.
  110. Pharmaceutical Research and Manufacturers of America (PhRMA). Corporate Responsibility Report. 2019.
  111. Patel M, et al. Pharmaceutical packaging: Current trends and future. Pharm Sci. 2018. 129. Pharmaceutical Supply Chain Initiative (PSCI). Principles for Responsible Supply Chain Management.
  112. Gad SC. Preclinical development handbook: ADME and biopharmaceutical properties.      Wiley, 2008.
  113. U.S. Food and Drug Administration (FDA). The Drug Development Process. 2021.
  114. Shargel L, et al. Applied Biopharmaceutics and Pharmacokinetics. McGraw Hill, 2012. 133. Saini TR, et al. Patient compliance and pharmaceutical design. Indian J Pharm Sci. 2011.
  115. ICH Q1A(R2). Stability Testing of New Drug Substances and Products. 2003.
  116. Central Drugs Standard Control Organization (CDSCO). Guidelines for Clinical Trials. 2020.
  117. Sharma HL, Sharma KK. Principles of Pharmacology. Paras Medical Publisher, 2017.
  118. Sahoo SK, Labhasetwar V. Nanotech approaches to drug delivery and imaging. Drug Discov Today. 2003.
  119. Torchilin VP. Multifunctional nanocarriers. Adv Drug Deliv Rev. 2006.
  120. Singh R, Lillard JW Jr. Nanoparticle-based targeted drug delivery. Exp Mol Pathol. 2009.
  121. Barenholz Y. Doxil® — the first FDA-approved nano-drug. J Control Release. 2012. 141. Zhang L, et al. Nanoparticles in medicine: therapeutic applications and developments. Clin Pharmacol Ther. 2008.
  122. European Medicines Agency (EMA). Reflection paper on nanotechnology-based medicinal products. 2021.
  123. Patra JK, et al. Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnol. 2018.
  124. Ginsburg GS, Willard HF. Genomic and personalized medicine: foundations and applications. Transl Res. 2009.
  125. Evans WE, Relling MV. Pharmacogenomics: translating functional genomics into rational therapeutics. Science. 1999.
  126. Simon RM, et al. Clinical trial designs for evaluating the medical utility of prognostic and predictive biomarkers in oncology. Per Med. 2009.
  127. Khaled SA, et al. 3D printing of pharmaceuticals. Int J Pharm. 2014.
  128. Spear BB, Heath-Chiozzi M, Huff J. Clinical application of pharmacogenetics. Trends Mol Med. 2001.
  129. McCarthy JJ, et al. The ethical challenges of personalized medicine. Drug Discov Today. 2013.
  130. Topol EJ. The Creative Destruction of Medicine. Basic Books, 2012.
  131. Galliher PM, et al. Digital health integration in drug delivery systems. J Control Release. 2020.
  132. Norman J, Madurawe RD, Moore CM, et al. A new chapter in pharmaceutical manufacturing: 3D-printed drug products. Adv Drug Deliv Rev. 2017.
  133. Thakur VK, Thakur MK. Eco-friendly green composites: science and engineering. Springer. 2017.
  134. Lee SL, et al. Modernizing pharmaceutical manufacturing: from batch to continuous production. J Pharm Innov. 2015.
  135. Prausnitz, M. R., & Langer, R. (2008). Transdermal drug delivery. Nature Biotechnology, 26(11), 1261–1268.
  136. Kunta, J. R., Goskonda, V. R., Brotherton, H. O., Khan, M. A., & Reddy, I. K. (1997). Effects of I-menthol and penetration enhancers on transdermal delivery of nicorandil. Drug Development and Industrial Pharmacy, 23(10), 973–978. https://doi.org/10.3109/03639049709082311
  137. Zhou, Y., Yu, B., Li, Y., Hu, Y., Cheng, H., Yuan, H., & Xu, Q. (2010). Paclitaxel-loaded nanostructured lipid carriers: Preparation, pharmacokinetics and distribution in vivo. Journal of Nanoscience and Nanotechnology, 10(11), 6489–6494. https://doi.org/10.1166/jnn.2010.2765
  138. Kim, D., Jeong, Y. I., & Choi, C. (2008). Self-assembled nanoparticles based on glycol chitosan bearing retinoic acid for anticancer drug delivery. European Journal of Pharmaceutics and Biopharmaceutics, 69(3), 1040–1050. https://doi.org/10.1016/j.ejpb.2008.01.020
  139. Zhang, Y., Wang, Y., Ma, G., Wang, Z., Wang, X., & Su, Z. (2012). Preparation of a novel hydrogel from chitosan-guanidinylated polyethylenimine and its antibacterial activity. International Journal of Biological Macromolecules, 50(4), 910–915. https://doi.org/10.1016/j.ijbiomac.2011.12.033
  140. Zhan, W., Liu, L., Wei, Y., Teng, L., Xu, Z., Zhang, X., & Wang, X. (2013). Preparation and evaluation of vincristine sulfate liposomes with high entrapment efficiency. Asian Journal of Pharmaceutical Sciences, 8(1), 37–43. https://doi.org/10.1016/j.ajps.2013.07.006
  141. Yang, S. C., Lu, L. F., Cai, Y., Zhu, J. B., Liang, B. W., & Yang, C. Z. (1999). Body distribution in mice of intravenously injected camptothecin solid lipid nanoparticles and targeting effect on brain. Journal of Controlled Release, 59(3), 299–307. https://doi.org/10.1016/S0168-3659(99)00027-3
  142. Qiu, L., Qiao, M., Chen, Q., Tian, C., Long, M., Wang, M., & et al. (2005). Sustained-release drug delivery system based on a novel chitosan derivative: In vitro and in vivo evaluation. International Journal of Pharmaceutics, 303(1–2), 137–144. https://doi.org/10.1016/j.ijpharm.2005.06.036
  143. Soppimath, K. S., Kulkarni, A. R., Rudzinski, W. E., & Aminabhavi, T. M. (2001). Microspheres as floating drug delivery systems to increase gastric retention of drugs: Overview of recent advances. Drug Development and Industrial Pharmacy, 27(6), 507–515. https://doi.org/10.1081/DDC-100104114
  144. Prego, C., Torres, D., & Alonso, M. J. (2005). The potential of chitosan for the oral administration of peptides. Expert Opinion on Drug Delivery, 2(5), 843–854. https://doi.org/10.1517/17425247.2.5.843
  145. Mao, S., Sun, W., & Kissel, T. (2010). Chitosan-based formulations for delivery of DNA and siRNA. Advanced Drug Delivery Reviews, 62(1), 12–27. https://doi.org/10.1016/j.addr.2009.08.004
  146. Vllasaliu, D., Exposito-Harris, R., Heras, A., Casettari, L., Garnett, M. C., Illum, L., & et al. (2010). Tight junction modulation by chitosan nanoparticles: Comparison with chitosan solution. International Journal of Pharmaceutics, 400(1–2), 183–193. https://doi.org/10.1016/j.ijpharm.2010.08.029
  147. Bowman, K., & Leong, K. W. (2006). Chitosan nanoparticles for oral drug and gene delivery. International Journal of Nanomedicine, 1(2), 117–128.
  148. Aulton ME, Taylor KMG. Aulton’s Pharmaceutics: The Design and Manufacture of Medicines. 5th ed. Elsevier; 2018.
  149. Allen LV. Ansel’s Pharmaceutical Dosage Forms and Drug Delivery Systems. 12th ed. Wolters Kluwer; 2021.
  150. Hickey AJ. Inhalation Aerosols: Physical and Biological Basis for Therapy. 3rd ed. CRC Press; 2019.
  151. Brahmankar HA, Jaiswal SB. Biopharmaceutics and Pharmacokinetics: A Treatise. 2nd ed. Vallabh Prakashan; 2009.
  152. Shargel L, Yu AB. Applied Biopharmaceutics and Pharmacokinetics. 7th ed. McGraw-Hill; 2016.
  153. Florence AT, Attwood D. Physicochemical Principles of Pharmacy. 6th ed. Pharmaceutical Press; 2015.
  154. Walters KA, Roberts MS. Dermatological and Transdermal Formulations. CRC Press; 2002.
  155. Labiris NR, Dolovich MB. Pulmonary drug delivery. Part I: Physiological factors. Clin Pharmacokinet. 2003;42(6):529–545.

Reference

  1. World Health Organization. WHO Drug Information. [Online].
  2. Rang HP, Dale MM, Ritter JM, Flower RJ. Rang and Dale’s Pharmacology. Elsevier.
  3. Kumar V, Abbas AK, Aster JC. Robbins and Cotran Pathologic Basis of Disease. Elsevier.
  4. Tripathi KD. Essentials of Medical Pharmacology. Jaypee Brothers.
  5. Hahnemann S. Organon of Medicine. Boericke & Tafel.
  6. Nutton V. Ancient Medicine. 2nd ed. Routledge; 2013.
  7. Pormann PE, Savage-Smith E. Medieval Islamic Medicine. Edinburgh University Press; 2007.
  8. Porter R. The Greatest Benefit to Mankind: A Medical History of Humanity. Harper Collins; 1999.
  9. Bynum WF. Science and the Practice of Medicine in the Nineteenth Century. Cambridge University Press; 1994.
  10. Weatherall MW. In Search of a Cure: A History of Pharmaceutical Discovery. Oxford University Press; 1990.
  11. Central Drugs Standard Control Organization. About CDSCO. Available from: https://cdsco.gov.in
  12. Flexner A. Medical Education in the United States and Canada. Carnegie Foundation; 1910.
  13. Ginsburg GS, Phillips KA. Precision Medicine: From Science to Value. Health Affairs. 2018;37(5):694–701.
  14. Rajan JV, Ramanathan M. Artificial Intelligence in Drug Discovery and Development. Trends Pharmacol Sci. 2021;42(10):813–827.
  15. Aulton ME, Taylor KMG. Aulton’s Pharmaceutics: The Design and Manufacture of Medicines. 5th ed. Elsevier; 2018.
  16. Allen LV. Ansel’s Pharmaceutical Dosage Forms and Drug Delivery Systems. 12th ed. Wolters Kluwer; 2021.
  17. Hickey AJ. Inhalation Aerosols: Physical and Biological Basis for Therapy. 3rd ed. CRC Press; 2019.
  18. Brahmankar HA, Jaiswal SB. Biopharmaceutics and Pharmacokinetics: A Treatise. 2nd ed. Vallabh Prakashan; 2009.
  19. Shargel L, Yu AB. Applied Biopharmaceutics and Pharmacokinetics. 7th ed. McGraw-Hill; 2016.
  20. Florence AT, Attwood D. Physicochemical Principles of Pharmacy. 6th ed. Pharmaceutical Press; 2015.
  21. Walters KA, Roberts MS. Dermatological and Transdermal Formulations. CRC Press; 2002.
  22. Labiris NR, Dolovich MB. Pulmonary drug delivery. Part I: Physiological factors. Clin Pharmacokinet. 2003;42(6):529–545.
  23. Brunton LL, Hilal-Dandan R, Knollmann BC. Goodman & Gilman’s: The Pharmacological Basis of Therapeutics. 13th ed. McGraw-Hill; 2018.
  24. Allen LV. Remington: The Science and Practice of Pharmacy. 22nd ed. Pharmaceutical Press; 2013.
  25. Aulton ME, Taylor KMG. Aulton’s Pharmaceutics: The Design and Manufacture of Medicines. 5th ed. Elsevier; 2018.
  26. Allen LV, Popovich NG, Ansel HC. Ansel’s Pharmaceutical Dosage Forms and Drug Delivery Systems. 10th ed. Wolters Kluwer; 2014.
  27. Kibbe AH. Handbook of Pharmaceutical Excipients. 6th ed. Pharmaceutical Press; 2009.
  28. Rowe RC, Sheskey PJ, Quinn ME. Handbook of Pharmaceutical Excipients. 7th ed. Pharmaceutical Press; 2012.
  29. Moldenhauer J. Pharmaceutical Cleanroom Technology. CRC Press; 2011. 30. Lachman L, Lieberman HA, Kanig JL. The Theory and Practice of Industrial Pharmacy. 3rd ed. Lea & Febiger; 1986.
  30. Mahato RI. Pharmaceutical Dosage Forms and Drug Delivery. 2nd ed. CRC Press; 2011.
  31. ICH Harmonised Tripartite Guideline. Stability Testing of New Drug Substances and Products Q1A(R2), ICH Steering Committee; 2003.
  32. WHO. Quality assurance of pharmaceuticals: a compendium of guidelines and related materials. WHO; 2007.
  33. United States Pharmacopeia (USP). General Chapters: <1225> Validation of Compendial Procedures.
  34. ICH Q10: Pharmaceutical Quality System. International Council for Harmonisation; 2008.
  35. FDA. Guidance for Industry: Quality Systems Approach to Pharmaceutical CGMP Regulations; 2006.
  36. EMA. Guidelines on Good Manufacturing Practice specific to Advanced Therapy Medicinal Products. European Medicines Agency; 2017.
  37. ISO 9001:2015 – Quality management systems – Requirements. International Organization for Standardization; 2015.
  38. Sandle T. Pharmaceutical Microbiology: Essentials for Quality Assurance and Quality Control. Elsevier; 2015.
  39. ICH Q1A(R2): Stability Testing of New Drug Substances and Products. ICH Steering Committee; 2003.
  40. FDA. Guidance for Industry: Stability Testing of Drug Substances and Drug Products; 2003.
  41. Aulton ME, Taylor KMG. Pharmaceutics: The Science of Dosage Form Design. 5th ed. Elsevier; 2018.
  42. WHO. Stability testing of active pharmaceutical ingredients and finished pharmaceutical products. WHO Technical Report Series; No. 1010, 2018.
  43. Collett JH, Aulton ME. Pharmaceutical Packaging Technology. Taylor & Francis; 2007. 45. Nash RA, Wachter AH. Pharmaceutical Process Validation. 3rd ed. Marcel Dekker; 2003.
  44. Allen LV. Remington: The Science and Practice of Pharmacy. 22nd ed. Pharmaceutical Press; 2013.
  45.  Aulton ME, Taylor KMG. Aulton’s Pharmaceutics: The Design and Manufacture of Medicines. 5th ed. Elsevier; 2018.
  46.  Rowe RC, Sheskey PJ, Quinn ME. Handbook of Pharmaceutical Excipients. 7th ed. Pharmaceutical Press; 2012.
  47. Felton LA. Remington Essentials of Pharmaceutics. Pharmaceutical Press; 2013.
  48.  United States Pharmacopeia (USP). <1> Injections; <661> Containers.
  49. Smart Packaging Market Report. Allied Market Research; 2020.
  50. FDA. Guidance for Industry: Container Closure Systems for Packaging Human Drugs and Biologics; 1999.
  51. WHO. Guidelines on packaging for pharmaceutical products. WHO Technical Report Series, No. 902, 2002. 55. EMA. Guidelines on the packaging of medicinal products for human use. European Medicines Agency; 2017.
  52. USFDA. Drug Development and Approval Process. Available at: https://www.fda.gov
  53.  EMA. Overview of regulatory responsibilities. European Medicines Agency. 58. WHO. Regulatory system strengthening. Available at: https://www.who.int 59. ICH. The Common Technical Document (CTD). https://www.ich.org
  54. USFDA. New Drug Application (NDA) process. https://www.fda.gov/drugs 61. ICH Guidelines Q8–Q10. International Council for Harmonisation. https://www.ich.org
  55. WHO. GMP and GCP standards. WHO Technical Report Series.
  56. Barenholz Y. Doxil®—the first FDA-approved nano-drug: lessons learned. J Control Release. 2012.
  57. Siepmann J, Siepmann F. Modeling of drug release from delivery systems based on hydroxypropyl methylcellulose (HPMC). Adv Drug Deliv Rev. 2012. 65. Fu Y, Yang S, Jeong SH, Kimura S, Park K. Orally fast disintegrating tablets: developments, technologies, and optimization. Drug Dev Ind Pharm. 2004.
  58. Prausnitz MR, Langer R. Transdermal drug delivery. Nat Biotechnol. 2008. 67. Goyanes A, et al. 3D printing of medicines: engineering novel oral drug products. Curr Pharm Des. 2015.
  59. Zhang Y, et al. Glucose-sensitive insulin release systems: synthesis and performance. J Biomed Mater Res. 2017.
  60. Schellekens H. Biosimilar therapeutics—what do we need to consider? NDT Plus. 2009. 70. Johnson JA. Pharmacogenetics in clinical practice: how far have we come and where are we going? Pharmacogenomics J. 2013. 71. Stokes JM, et al. A deep learning approach to antibiotic discovery. Cell. 2020.
  61. Lipinski CA. Poor aqueous solubility—an industry wide problem in drug discovery. Adv Drug Deliv Rev. 2002.
  62. Waterman KC, Adami RC. Accelerated aging: prediction of chemical stability of pharmaceuticals. Int J Pharm. 2005.
  63. Arya A, Chandra A, Sharma V, Pathak K. Fast dissolving oral films: an innovative drug delivery system and dosage form. Int J ChemTech Res. 2010. 75. International Conference on Harmonisation. Regulatory considerations for pharmaceutical development.
  64. Garcia CV, et al. Pharmaceutical manufacturing process validation: quality and regulatory perspectives. J Pharm Sci. 2018.
  65.  DiMasi JA, Grabowski HG, Hansen RW. Innovation in the pharmaceutical industry: new estimates of R&D costs. J Health Econ. 2016.
  66. Bhatt DA, Pethe AM. Drug-excipient compatibility screening. Asian J Pharm. 2010.
  67. Ginsburg GS, Phillips KA. Precision medicine: from science to value. Health Aff. 2018.
  68. Kümmerer K, Dionysiou DD, Olsson O, Fatta-Kassinos D. A path to clean pharmaceuticals and chemical manufacturing. Green Chem. 2018.
  69. Patel DM, Patel MM. Optimization of fast dissolving tablets of paracetamol using simple lattice design. Int J Pharm Sci. 2009.
  70. Ramprakash V, Varghese L. Design and evaluation of sustained release matrix tablets of metformin hydrochloride. Int J PharmTech Res. 2010.
  71. Shrivastava AR, et al. Formulation and evaluation of enteric-coated tablets of omeprazole. Indian J Pharm Sci. 2007.
  72. Ganesh M, et al. Formulation and evaluation of co-amoxiclav dry syrup. Int J Pharm Sci Res. 2012.
  73. Guo JH. Transdermal drug delivery systems for fentanyl. Pharm Tech. 1994. 86. Vajo Z, Duckworth WC. Genetically engineered insulin analogs: diabetes in the new millennium. Pharmacol Rev. 2000.
  74. Aprecia Pharmaceuticals. Spritam® (levetiracetam) tablets: FDA approval announcement. 2015. 88. Adler-Moore JP, Proffitt RT. AmBisome: liposomal formulation, structure, and stability. J Liposome Res. 1993.
  75. WIPO. Types of Intellectual Property. World Intellectual Property Organization.
  76. Gassmann O, Reepmeyer G. Organizing pharmaceutical innovation: from science-based knowledge creators to drug-oriented knowledge brokers. Creat Innov Manage. 2005.
  77. Kumar V, et al. Patent filing procedure in India. Int J Res Pharm Sci. 2012.
  78. WIPO. Patent Cooperation Treaty (PCT). Overview.
  79. World Trade Organization. TRIPS Agreement.
  80. Basheer S. India’s first compulsory license: Natco v. Bayer. SpicyIP Blog. 2012.
  81. Kapczynski A, et al. Polymorph patents and the law. N Engl J Med. 2012.
  82. Singh A. IPR challenges in pharma industry. J Pharm Sci Res. 2016.
  83. Indian Patent Office. Manual of Patent Office Practice and Procedure. Govt. Of India.
  84. Sharma PP. How to Practice GMPs. 5th Ed. Vandana Publications.
  85. Makhija R, et al. Concept of quality control in pharma. Int J Pharm Sci Rev Res. 2010. 100. ICH Q1A(R2). Stability Testing of New Drug Substances and Products.
  86. WHO. Quality assurance of pharmaceuticals: a compendium of guidelines and related materials. Vol. 2.
  87. USFDA. Guidance for Industry: GMP for Pharmaceuticals. 21 CFR Part 210 & 211. 103. ICH Q10. Pharmaceutical Quality System. International Conference on Harmonisation. 104. CDSCO. Schedule M – Good Manufacturing Practices for Pharmaceuticals in India.
  88. Patel M, et al. A review on quality defects in tablets and their control. Pharma Times. 2013.
  89. Amidon GL, et al. A theoretical basis for a biopharmaceutic drug classification. Pharm Res. 1995.
  90. Dressman JB, Reppas C. In vitro–in vivo correlations. Eur J Pharm Sci. 2000.
  91. FDA. Guidance for Industry: Waiver of In Vivo Bioavailability and Bioequivalence Studies for Immediate-Release Solid Oral Dosage Forms Based on a BCS.
  92. Shargel L, Yu ABC. Applied Biopharmaceutics and Pharmacokinetics. 6th ed.
  93. WHO. Bioavailability and Bioequivalence Guidelines.
  94. FDA. Guidance for Industry: Dissolution Testing of Immediate Release Solid Oral Dosage Forms.
  95. Robinson JR, Eriksen S. Theoretical considerations of controlled drug delivery. J Pharm Sci. 1971.
  96. Sugano K. Current status of in vitro permeability studies. Int J Pharm. 2009.
  97. Hamburg MA, Collins FS. The path to personalized medicine. N Engl J Med. 2010.
  98. Alhnan MA, et al. Emergence of 3D printed dosage forms: Opportunities and challenges. Adv Drug Deliv Rev. 2016.
  99.  Mak KK, Pichika MR. Artificial intelligence in drug development: Present status and future prospects. Drug Discov Today. 2019.
  100. Raza K, et al. Smart drug delivery system: A novel approach. Drug Deliv. 2014.
  101. Zhang L, et al. Nanoparticles in medicine: Therapeutic applications and developments. Clin Pharmacol Ther. 2008.
  102.  Anastas PT, Warner JC. Green Chemistry: Theory and Practice. Oxford University Press, 1998.
  103. Boehm E, et al. Blockchain technology in the pharmaceutical industry. J Pharm Sci. 2020.
  104. Lee SL, et al. Modernizing pharmaceutical manufacturing: from batch to continuous production. J Pharm Innov. 2015.
  105. Kümmerer K. Pharmaceuticals in the Environment: Sources, Fate, Effects. Springer, 2008.
  106. Sheldon RA. Green chemistry and resource efficiency. Green Chem. 2016.
  107. Varma AJ, et al. Polysaccharides: Natural biodegradable polymers. Carbohydr Polym. 2004.
  108. European Medicines Agency. Ethical considerations for clinical trials. 2018.
  109. Central Pollution Control Board (CPCB) India. Guidelines for Management of Waste from Pharmaceutical Industry. 2020.
  110. Pharmaceutical Research and Manufacturers of America (PhRMA). Corporate Responsibility Report. 2019.
  111. Patel M, et al. Pharmaceutical packaging: Current trends and future. Pharm Sci. 2018. 129. Pharmaceutical Supply Chain Initiative (PSCI). Principles for Responsible Supply Chain Management.
  112. Gad SC. Preclinical development handbook: ADME and biopharmaceutical properties.      Wiley, 2008.
  113. U.S. Food and Drug Administration (FDA). The Drug Development Process. 2021.
  114. Shargel L, et al. Applied Biopharmaceutics and Pharmacokinetics. McGraw Hill, 2012. 133. Saini TR, et al. Patient compliance and pharmaceutical design. Indian J Pharm Sci. 2011.
  115. ICH Q1A(R2). Stability Testing of New Drug Substances and Products. 2003.
  116. Central Drugs Standard Control Organization (CDSCO). Guidelines for Clinical Trials. 2020.
  117. Sharma HL, Sharma KK. Principles of Pharmacology. Paras Medical Publisher, 2017.
  118. Sahoo SK, Labhasetwar V. Nanotech approaches to drug delivery and imaging. Drug Discov Today. 2003.
  119. Torchilin VP. Multifunctional nanocarriers. Adv Drug Deliv Rev. 2006.
  120. Singh R, Lillard JW Jr. Nanoparticle-based targeted drug delivery. Exp Mol Pathol. 2009.
  121. Barenholz Y. Doxil® — the first FDA-approved nano-drug. J Control Release. 2012. 141. Zhang L, et al. Nanoparticles in medicine: therapeutic applications and developments. Clin Pharmacol Ther. 2008.
  122. European Medicines Agency (EMA). Reflection paper on nanotechnology-based medicinal products. 2021.
  123. Patra JK, et al. Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnol. 2018.
  124. Ginsburg GS, Willard HF. Genomic and personalized medicine: foundations and applications. Transl Res. 2009.
  125. Evans WE, Relling MV. Pharmacogenomics: translating functional genomics into rational therapeutics. Science. 1999.
  126. Simon RM, et al. Clinical trial designs for evaluating the medical utility of prognostic and predictive biomarkers in oncology. Per Med. 2009.
  127. Khaled SA, et al. 3D printing of pharmaceuticals. Int J Pharm. 2014.
  128. Spear BB, Heath-Chiozzi M, Huff J. Clinical application of pharmacogenetics. Trends Mol Med. 2001.
  129. McCarthy JJ, et al. The ethical challenges of personalized medicine. Drug Discov Today. 2013.
  130. Topol EJ. The Creative Destruction of Medicine. Basic Books, 2012.
  131. Galliher PM, et al. Digital health integration in drug delivery systems. J Control Release. 2020.
  132. Norman J, Madurawe RD, Moore CM, et al. A new chapter in pharmaceutical manufacturing: 3D-printed drug products. Adv Drug Deliv Rev. 2017.
  133. Thakur VK, Thakur MK. Eco-friendly green composites: science and engineering. Springer. 2017.
  134. Lee SL, et al. Modernizing pharmaceutical manufacturing: from batch to continuous production. J Pharm Innov. 2015.
  135. Prausnitz, M. R., & Langer, R. (2008). Transdermal drug delivery. Nature Biotechnology, 26(11), 1261–1268.
  136. Kunta, J. R., Goskonda, V. R., Brotherton, H. O., Khan, M. A., & Reddy, I. K. (1997). Effects of I-menthol and penetration enhancers on transdermal delivery of nicorandil. Drug Development and Industrial Pharmacy, 23(10), 973–978. https://doi.org/10.3109/03639049709082311
  137. Zhou, Y., Yu, B., Li, Y., Hu, Y., Cheng, H., Yuan, H., & Xu, Q. (2010). Paclitaxel-loaded nanostructured lipid carriers: Preparation, pharmacokinetics and distribution in vivo. Journal of Nanoscience and Nanotechnology, 10(11), 6489–6494. https://doi.org/10.1166/jnn.2010.2765
  138. Kim, D., Jeong, Y. I., & Choi, C. (2008). Self-assembled nanoparticles based on glycol chitosan bearing retinoic acid for anticancer drug delivery. European Journal of Pharmaceutics and Biopharmaceutics, 69(3), 1040–1050. https://doi.org/10.1016/j.ejpb.2008.01.020
  139. Zhang, Y., Wang, Y., Ma, G., Wang, Z., Wang, X., & Su, Z. (2012). Preparation of a novel hydrogel from chitosan-guanidinylated polyethylenimine and its antibacterial activity. International Journal of Biological Macromolecules, 50(4), 910–915. https://doi.org/10.1016/j.ijbiomac.2011.12.033
  140. Zhan, W., Liu, L., Wei, Y., Teng, L., Xu, Z., Zhang, X., & Wang, X. (2013). Preparation and evaluation of vincristine sulfate liposomes with high entrapment efficiency. Asian Journal of Pharmaceutical Sciences, 8(1), 37–43. https://doi.org/10.1016/j.ajps.2013.07.006
  141. Yang, S. C., Lu, L. F., Cai, Y., Zhu, J. B., Liang, B. W., & Yang, C. Z. (1999). Body distribution in mice of intravenously injected camptothecin solid lipid nanoparticles and targeting effect on brain. Journal of Controlled Release, 59(3), 299–307. https://doi.org/10.1016/S0168-3659(99)00027-3
  142. Qiu, L., Qiao, M., Chen, Q., Tian, C., Long, M., Wang, M., & et al. (2005). Sustained-release drug delivery system based on a novel chitosan derivative: In vitro and in vivo evaluation. International Journal of Pharmaceutics, 303(1–2), 137–144. https://doi.org/10.1016/j.ijpharm.2005.06.036
  143. Soppimath, K. S., Kulkarni, A. R., Rudzinski, W. E., & Aminabhavi, T. M. (2001). Microspheres as floating drug delivery systems to increase gastric retention of drugs: Overview of recent advances. Drug Development and Industrial Pharmacy, 27(6), 507–515. https://doi.org/10.1081/DDC-100104114
  144. Prego, C., Torres, D., & Alonso, M. J. (2005). The potential of chitosan for the oral administration of peptides. Expert Opinion on Drug Delivery, 2(5), 843–854. https://doi.org/10.1517/17425247.2.5.843
  145. Mao, S., Sun, W., & Kissel, T. (2010). Chitosan-based formulations for delivery of DNA and siRNA. Advanced Drug Delivery Reviews, 62(1), 12–27. https://doi.org/10.1016/j.addr.2009.08.004
  146. Vllasaliu, D., Exposito-Harris, R., Heras, A., Casettari, L., Garnett, M. C., Illum, L., & et al. (2010). Tight junction modulation by chitosan nanoparticles: Comparison with chitosan solution. International Journal of Pharmaceutics, 400(1–2), 183–193. https://doi.org/10.1016/j.ijpharm.2010.08.029
  147. Bowman, K., & Leong, K. W. (2006). Chitosan nanoparticles for oral drug and gene delivery. International Journal of Nanomedicine, 1(2), 117–128.
  148. Aulton ME, Taylor KMG. Aulton’s Pharmaceutics: The Design and Manufacture of Medicines. 5th ed. Elsevier; 2018.
  149. Allen LV. Ansel’s Pharmaceutical Dosage Forms and Drug Delivery Systems. 12th ed. Wolters Kluwer; 2021.
  150. Hickey AJ. Inhalation Aerosols: Physical and Biological Basis for Therapy. 3rd ed. CRC Press; 2019.
  151. Brahmankar HA, Jaiswal SB. Biopharmaceutics and Pharmacokinetics: A Treatise. 2nd ed. Vallabh Prakashan; 2009.
  152. Shargel L, Yu AB. Applied Biopharmaceutics and Pharmacokinetics. 7th ed. McGraw-Hill; 2016.
  153. Florence AT, Attwood D. Physicochemical Principles of Pharmacy. 6th ed. Pharmaceutical Press; 2015.
  154. Walters KA, Roberts MS. Dermatological and Transdermal Formulations. CRC Press; 2002.
  155. Labiris NR, Dolovich MB. Pulmonary drug delivery. Part I: Physiological factors. Clin Pharmacokinet. 2003;42(6):529–545.

Photo
Vishal Jadhav
Corresponding author

Department Of Pharmacognosy, Ashvin College of Pharmacy, Manchi Hills

Photo
Ashok Veer
Co-author

Department Of Pharmacognosy, Ashvin College of Pharmacy, Manchi Hills

Photo
Yuvraj Mhase
Co-author

Department Of Pharmacognosy, Ashvin College of Pharmacy, Manchi Hills

Photo
Dipak Ahire
Co-author

Department Of Pharmacognosy, Ashvin College of Pharmacy, Manchi Hills

Vishal Jadhav, Ashok Veer, Yuvraj Mhase, Dipak Ahire, Comprehensive Review of Allopathic Formulations: Composition, Classification, Mechanisms, and Innovations, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 8, 366-391. https://doi.org/10.5281/zenodo.16737800

More related articles
Comparative Study of Bronchodilators in the Manage...
Dr. Parveen Shaik, Arkari Rishitha, Ayeesha Nazneen, Badagouni So...
Multifarious Effects Of Benidipine In Vitro And In...
Vishnupriya S., Dhinesh M., Anjitha P. S. , Eniya A. A., ...
Formulation and Evaluation of Herbal Mouthwash ...
Pallavi Wagh, Samiksha Wankhade, Aditi Tikait, Dr. Swati Deshmukh...
A Review Article On The Artificial Intelligence [Ai] In Tumour Detection [Oncolo...
Padala Ramesh, Patnala Vaishnavi Gayathri, Mamindla Chandrika, Daravath Pawan Kalyan, ...
A Review on Solid Lipid Nanoparticles for Topical Delivery of Anti-Inflammatory ...
Manjusha Bandi, Dr. Nitalikar Manojkumar, Dr. Gadad Anand, ...
Related Articles
Consideration With Patient Approach and Physician Prescription for Sale and Mark...
Anushka Chormale, S. S. Navale, Dr. N. B. Chougule, Alisha Dhandhore, Atul Bhole, ...
Comparative Study of Bronchodilators in the Management of Asthma...
Dr. Parveen Shaik, Arkari Rishitha, Ayeesha Nazneen, Badagouni Somnath, Baru Bhanu Vara Prasad, Basa...
More related articles
Comparative Study of Bronchodilators in the Management of Asthma...
Dr. Parveen Shaik, Arkari Rishitha, Ayeesha Nazneen, Badagouni Somnath, Baru Bhanu Vara Prasad, Basa...
Multifarious Effects Of Benidipine In Vitro And In Vivo A Review...
Vishnupriya S., Dhinesh M., Anjitha P. S. , Eniya A. A., ...
Formulation and Evaluation of Herbal Mouthwash ...
Pallavi Wagh, Samiksha Wankhade, Aditi Tikait, Dr. Swati Deshmukh, ...
Comparative Study of Bronchodilators in the Management of Asthma...
Dr. Parveen Shaik, Arkari Rishitha, Ayeesha Nazneen, Badagouni Somnath, Baru Bhanu Vara Prasad, Basa...
Multifarious Effects Of Benidipine In Vitro And In Vivo A Review...
Vishnupriya S., Dhinesh M., Anjitha P. S. , Eniya A. A., ...
Formulation and Evaluation of Herbal Mouthwash ...
Pallavi Wagh, Samiksha Wankhade, Aditi Tikait, Dr. Swati Deshmukh, ...