Growup Pharma

B Pharmacy Sem 3: Pharmaceutics II

B Pharmacy Sem 3: Pharmaceutics II

 

Table of Contents

Subject : Pharmaceutics II

1. Sterile Product Technology: Parenteral Preparations
2. Aseptic Processing & Validation
3. Ophthalmic & Otic Dosage Forms
4. Inhalation & Pulmonary Drug Delivery Systems
5. Transdermal & Topical Delivery Systems
6. Introduction to Novel Drug Delivery (Nanoparticles, Liposomes)

 

Unit 1: Sterile Product Technology – Parenteral Preparations

This unit covers the design, formulation, sterilization, packaging, and quality control of parenteral (injectable) dosage forms, which must be free of viable microorganisms and pyrogens.


1.1 Classification of Parenteral Dosage Forms

1.1.1 Injectable Solutions

  • Type: True solutions of APIs in water, aqueous co‑solvent systems, or non‑aqueous vehicles.

  • Examples: Normal saline injections, diazepam injectable solution (propylene glycol).

1.1.2 Injectable Suspensions

  • Type: Solid drug particles dispersed in a liquid vehicle.

  • Considerations: Particle size (≤5 µm for IV), wetting agents, suspension stability.

  • Examples: Penicillin G potassium suspension, methylprednisolone acetate.

1.1.3 Emulsions

  • Type: Oil-in-water (O/W) or water-in-oil (W/O) systems to solubilize lipophilic drugs.

  • Stabilizers: Emulsifying agents (lecithin, poloxamers).

  • Example: Propofol injectable emulsion.

1.1.4 Lyophilized (Freeze‑Dried) Powders

  • Purpose: Improves stability of labile drugs.

  • Process: Freezing → primary drying (sublimation) → secondary drying (desorption).

  • Reconstitution: Sterile diluent added prior to administration (e.g., vancomycin HCl).


1.2 Formulation Considerations

1.2.1 Solvent & Vehicle Selection

  • Aqueous vehicles: WFI (Water for Injection), Bacteriostatic Water.

  • Non‑aqueous vehicles: Fixed oils (sesame oil), polyethylene glycol.

  • Co‑solvents: Ethanol, propylene glycol for poorly water‑soluble drugs.

1.2.2 pH & Tonicity Adjustment

  • pH range: Typically 4–8 to minimize pain and tissue irritation.

  • Buffers: Phosphate, citrate to maintain pH.

  • Isotonicity: 0.9% NaCl or 5% dextrose equivalence.

1.2.3 Excipients

  • Preservatives: Parabens, benzyl alcohol (multi‑dose vials only).

  • Antioxidants: Sodium metabisulfite, ascorbic acid.

  • Chelating agents: EDTA to complex trace metals.


1.3 Manufacturing Process & Equipment

1.3.1 Clean‐Room Environment

  • Classifications: ISO 5 (critical zone), ISO 7–8 (background).

  • Airflow: Unidirectional (laminar) flow hoods for aseptic filling.

1.3.2 Sterile Filtration & Filling

  • Filtration: 0.22 µm membrane filters remove microorganisms (for heat‑sensitive solutions).

  • Filling: Automated peristaltic or piston fillers under ISO 5 conditions.

1.3.3 Lyophilization Cycle

  • Freezing: Controlled to avoid eutectic collapse.

  • Primary drying: Chamber pressure <100 mTorr, shelf temperature optimization.

  • Secondary drying: Elevated shelf temperature to remove bound water.


1.4 Sterilization Methods

MethodMechanismTypical Uses
Moist Heat (Autoclaving)Denaturation of proteinsGlassware, aqueous solutions (not heat‑sensitive)
Dry HeatOxidation & protein damageEmpty containers, oils, syringes
Filtration SterilizationPhysical removalHeat‑labile solutions, biologicals
Radiation (γ or e‑beam)DNA damage in microbesPre‑sterilized single‑use devices
Ethylene Oxide GasAlkylation of cell componentsMedical device packaging

1.5 Container Closure Systems

1.5.1 Ampoules

  • Material: Type I borosilicate glass.

  • Sealing: Flame‑sealed neck to ensure integrity.

1.5.2 Vials

  • Stopper: Rubber or elastomeric; must be chemically compatible and allow needle penetration without coring.

  • Crimp Seal: Aluminum cap to secure stopper.

1.5.3 Prefilled Syringes & Cartridges

  • Advantages: Dose accuracy, reduced contamination risk.

  • Materials: Glass or cyclic olefin polymer; silicone‑lubricated plungers.


1.6 Quality Control Tests

1.6.1 Sterility Testing

  • Method: Direct inoculation or membrane filtration per pharmacopeial <71>.

  • Media: Fluid thioglycollate (anaerobes) and soybean–casein digest (aerobes).

1.6.2 Pyrogen/Endotoxin Testing

  • LAL Test: Limulus amebocyte lysate assay for bacterial endotoxins (<0.25 EU/mL for IV).

  • Rabbit Pyrogen Test: Historical in vivo method (used less frequently).

1.6.3 Particulate Matter

  • Limits: ≤6000 particles ≥10 µm and ≤600 particles ≥25 µm per container for small‑volume injectables.

  • Test: Light obscuration counting.

1.6.4 Physical & Chemical Tests

  • Clarity & Color: Visual inspection under defined illumination.

  • pH & Osmolality: Ensures compatibility with blood.

  • Assay & Degradation Products: HPLC or titrimetric analysis.


1.7 Pharmaceutical Examples

  • Gentamicin Injection: Aqueous solution, autoclaved in sealed ampoules.

  • Ceftriaxone for Injection: Lyophilized powder reconstituted with sterile water.

  • Etomidate Injectable Emulsion: Oil-in-water emulsion sterilized by filtration.


1.8 Key Points for Exams

  • Classify parenteral dosage forms and list one example of each.

  • Explain formulation requirements for pH, tonicity, and excipient selection.

  • Describe the principles and applications of at least two sterilization methods.

  • Outline the sterile-filtration and aseptic-filling process in an ISO 5 environment.

  • List four critical quality‑control tests for parenteral preparations and their acceptance criteria.

 

Unit 2: Aseptic Processing & Validation

This unit delves into the principles and practices that ensure parenteral products remain sterile from formulation through final packaging, and the qualification of facilities, equipment, and processes to guarantee consistent aseptic manufacture.


2.1 Principles of Aseptic Processing

  • Definition: Operations performed under controlled, sterile conditions to prevent microbial contamination of sterile products.

  • Core Elements:

    • Controlled Environment: Cleanrooms with defined airflow, pressurization, temperature, and particulate/microbial limits.

    • Personnel Practices: Gowning, aseptic techniques, and behavior designed to minimize contamination.

    • Sterile Components: Use of sterilized inputs (solutions, containers, closures).

    • Closed Systems: Minimizing open handling of sterile materials; use of barrier isolators and restricted-access barriers.


2.2 Cleanroom Design & Classification

2.2.1 ISO Cleanroom Classes

ISO ClassMaximum Particles ≥0.5 µm per m³Typical Applications
ISO 53,520Critical zones (fill/finish)
ISO 7352,000Background areas
ISO 83,520,000Support operations
  • Air Changes per Hour (ACH): ISO 5 requires ≥240 ACH; lower classes require progressively fewer.

  • Pressure Differentials: Positive pressure in cleaner areas relative to adjacent zones (e.g., +15–20 Pa).

2.2.2 Airflow Patterns

  • Unidirectional (Laminar) Flow: HEPA‑filtered air moving in parallel streams; used in ISO 5 zones.

  • Non‑Unidirectional (Turbulent) Flow: Mixing airflow in ISO 7/8 zones; still HEPA‑filtered but less critical.


2.3 Personnel & Aseptic Technique

2.3.1 Gowning Procedure

  • Sequence: Shoe covers → hair cover → face mask → sterile gown → sterile gloves.

  • Materials: Low‑shedding, lint‑free fabrics; sterile, powder‑free gloves.

2.3.2 Operational Practices

  • Entry/Exit Protocols: Airlocks with sequential gowning and hand‑scrub stations.

  • Movement: Slow, deliberate motions; minimal talking; avoid crossing critical airflow paths.

  • Sterile Field Management: Maintain one‑inch‑away rule—keep non‑sterile items at least one inch from critical surfaces.


2.4 Process Validation for Aseptic Manufacturing

2.4.1 Installation Qualification (IQ)

  • Verifies that equipment and utilities (HVAC, isolators, airlocks) are installed per design specifications.

  • Documents: Equipment specifications, calibration records, piping and instrumentation diagrams.

2.4.2 Operational Qualification (OQ)

  • Confirms that each system component operates according to its intended limits.

  • Tests: Airflow velocity mapping, HEPA filter integrity (DOP/PARFOG challenge), differential pressure alarms.

2.4.3 Performance Qualification (PQ)

  • Demonstrates that routine production under worst‑case conditions yields sterile products.

  • Activities: Media fill (process simulation) runs, environmental monitoring, operator challenge studies.


2.5 Media Fill Simulation

  • Objective: To simulate the aseptic filling process using a microbiological growth medium instead of product solution to detect contamination events.

  • Procedure:

    1. Prepare sterile culture medium in all product contact containers and closures.

    2. Run full-scale simulated fill under normal conditions and personnel.

    3. Incubate filled units at 20–25 °C for 7 days, then 30–35 °C for 7 days.

    4. Evaluate turbidity or colony formation; no contamination indicates process control.

  • Acceptance Criteria: Zero positive units for ISO 5 processes; low-level positives may be permitted under tightly controlled investigations.


2.6 Environmental Monitoring

2.6.1 Monitoring Parameters

  • Viable Particulates:

    • Air Sampling: Active (SAS) and passive (settle plates) methods in critical zones.

    • Surface Sampling: Contact plates, swabs, and wipes on work surfaces, equipment, gloves.

  • Non‑Viable Particulates: Continuous particle counters to ensure ISO classification compliance.

2.6.2 Alert & Action Limits

  • Alert Level: Early warning threshold (e.g., 1 CFU/m³ in ISO 5).

  • Action Level: Indicates process may be out of control; triggers root cause investigation and corrective actions.


2.7 Equipment & Process Controls

  • Automated Filling Machines: Validated for precision, accuracy, and sterility assurance.

  • Barrier Technologies: Isolators and Restricted Access Barrier Systems (RABS) to further reduce contamination risk.

  • Cleaning & Sanitization: Validated cleaning agents, schedules, and rinse sampling to confirm removal of residues and bioburden.


2.8 Key Points for Exams

  • Define aseptic processing and list its four core elements.

  • Describe cleanroom classification (ISO 5–8), airflow types, and pressure differentials.

  • Outline the gowning sequence and two critical aseptic techniques.

  • Differentiate IQ, OQ, and PQ in process validation.

  • Explain media fill methodology and acceptance criteria for aseptic filling.

  • List environmental monitoring methods for viable and non‑viable particulates, and define alert/action limits.

Unit 3: Ophthalmic & Otic Dosage Forms

This unit details the formulation, manufacture, and quality control of sterile preparations for administration to the eye (ophthalmic) and ear (otic), emphasizing the specialized requirements for safety, efficacy, and patient comfort.


3.1 General Requirements for Ophthalmic & Otic Preparations

  • Sterility: Must be free of viable microorganisms (terminal sterilization or sterile filtration).

  • Isotonicity: Approximate to tear fluid (~0.9% NaCl, 300 mOsm/kg) to avoid irritation or cell damage.

  • pH & Buffering:

    • Ophthalmic: pH 7.0–7.4 preferred (range 6.5–8.5 acceptable).

    • Otic: pH 5.0–7.5 to match ear canal environment and drug stability.

  • Viscosity & Rheology: Optimized to prolong residence time without blurring vision (ophthalmic) or causing discomfort (otic).

  • Preservatives: Only if multi‑dose; must be non‑toxic to sensitive mucosa (e.g., benzalkonium chloride, chlorobutanol).

  • Packaging: Light‑resistant, hermetically sealed containers with sterile droppers or applicators.


3.2 Ophthalmic Dosage Forms

3.2.1 Eye Drops (Solutions & Suspensions)

  • Solutions:

    • Clear, sterile aqueous vehicles (WFI) with dissolved API.

    • Must be filtered through 0.22 µm membranes.

  • Suspensions:

    • Finely divided drug (mean particle size ≤10 µm) dispersed in vehicle.

    • Require wetting agents (e.g., polysorbate 80) and suspending agents (e.g., methylcellulose).

Formulation Considerations

ParameterTarget Range/AgentRationale
Tonicity0.6–1.8% NaCl equivalentComfort, prevents corneal swelling
pH6.5–8.5Minimize irritation, maintain drug stability
Viscosity15–25 cP (drops)Longer contact time, acceptable blinking
PreservativeBenzalkonium chloride 0.01–0.02%Broad‑spectrum antimicrobial

3.2.2 Ophthalmic Ointments & Gels

  • Ointments:

    • Semi‑solid vehicles (e.g., petrolatum–mineral oil) for prolonged contact.

    • Lower drug release rate; used at night.

  • Gels & In Situ Gelling Systems:

    • Polymers (carbomers, Pluronic) that gel upon contact with tear fluid.

    • Combine ease of instillation with extended retention.

3.2.3 Ophthalmic Inserts & Implantables

  • Soluble Inserts: Cellulose acetate or gelatin rods that dissolve, releasing drug over hours.

  • Contact‑Lens Systems: Drug‑loaded hydrogel lenses for sustained delivery.

  • Implants: Biodegradable polymers (e.g., PLGA) for long‑term therapy (weeks–months).


3.3 Otic Dosage Forms

3.3.1 Ear Drops (Solutions & Suspensions)

  • Solutions: Sterile aqueous or mixed solvent (propylene glycol–water) for antibiotics, antiseptics.

  • Suspensions: Suspended corticosteroids, cerumenolytics; particle size ≤50 µm.

3.3.2 Otic Gels & Powders

  • Gels: Carbomer or cellulose-based to adhere to canal walls and prolong retention.

  • Powders: Boric acid or aluminum acetate powders for dryness and mild antisepsis.


3.4 Manufacturing & Packaging

3.4.1 Aseptic Processing

  • Conducted in ISO 5 laminar‑flow hoods.

  • Components sterilized by filtration or autoclaving before filling.

3.4.2 Fill‑Finish & Container Closure

  • Droppers & Pipettes: Low‑dead‑volume designs to ensure accurate dosing.

  • Multi‑Dose Vials: Incorporate validated preservative; single‑dose units avoid preservatives.

3.4.3 Labeling & Patient Instructions

  • Indicate “For Ophthalmic Use Only” or “For Otic Use Only.”

  • Include directions for instillation angle, number of drops, and avoidance of contamination.


3.5 Quality Control Tests

TestOphthalmicOtic
SterilityMembrane filtration per USP <71>As above
Particulate MatterLight obscuration (≤50 particles/mL ≥10 µm)Visual/obscuration less critical but monitored
pH6.5–8.55.0–7.5
ViscosityBrookfield viscometerMeasured if gel form
Preservative EfficacyUSP <51> antimicrobial effectiveness testIf multi‑dose
Drop Size & Volume~25–50 µL/drop50–100 µL/drop

3.6 Key Points for Exams

  • List two formulation differences between ophthalmic and otic drops.

  • Explain why ophthalmic solutions require smaller particle sizes than otic suspensions.

  • Describe three mechanisms by which in situ gelling systems prolong ocular drug contact.

  • Outline the sterility and particulate tests specific to eye drops.

  • Identify packaging features that prevent contamination and ensure accurate dosing for both dosage forms.

 

 

 

Unit 4: Inhalation & Pulmonary Drug Delivery Systems

This unit covers the science and technology underpinning delivery of drugs via the respiratory tract—including formulation design, device mechanics, aerosol physics, and quality control—to achieve local (e.g., asthma) or systemic (e.g., insulin) therapy through the lungs.


4.1 Introduction & Rationale

  • Advantages:

    • Rapid onset via large absorptive surface and thin alveolar–capillary membrane.

    • Targeted therapy for pulmonary diseases with reduced systemic side effects.

    • Non‑invasive alternative to injections for peptides and vaccines.

  • Anatomical Considerations:

    • Upper vs. Lower Airways: Mouth → trachea → bronchi → bronchioles → alveoli.

    • Deposition Sites: Determined by particle aerodynamic diameter.


4.2 Aerosol Physics & Deposition Mechanisms

4.2.1 Aerodynamic Particle Size

  • Mass Median Aerodynamic Diameter (MMAD): Critical metric; 1–5 µm particles reach lower airways.

  • Geometric Standard Deviation (GSD): Distribution breadth; tighter distribution (GSD < 1.5) improves predictability.

4.2.2 Deposition Mechanisms

  • Inertial Impaction: Particles >5 µm deposit in oropharynx and upper airways.

  • Gravitational Sedimentation: 1–5 µm particles settle in bronchioles and alveoli under gravity.

  • Brownian Diffusion: <1 µm particles deposit by diffusion in alveolar region or are exhaled.


4.3 Formulation Types & Devices

4.3.1 Pressurized Metered‑Dose Inhalers (pMDIs)

  • Propellants: Hydrofluoroalkanes (HFA‑134a, HFA‑227ea).

  • Formulation: Solution or suspension of API + co‑solvent (ethanol) + surfactant.

  • Actuation: Delivers metered plume; coordination with inhalation critical.

4.3.2 Dry Powder Inhalers (DPIs)

  • Formulation: Micronized API blended with larger carrier particles (lactose).

  • Device Types: Single‑dose, multi‑dose reservoir or blister.

  • Patient Effort: Inhalation flow de‑aggregates powder into respirable fraction.

4.3.3 Nebulizers

  • Jet Nebulizers: Compressed air or oxygen creates aerosol via Venturi effect.

  • Ultrasonic Nebulizers: Piezoelectric crystal generates high‑frequency vibration.

  • Mesh Nebulizers: Liquid pushed through micro‑pores; efficient, quiet.

4.3.4 Soft‑Mist Inhalers (SMIs)

  • Mechanism: Spring‑powered piston forces liquid through nozzle to generate fine mist.

  • Advantage: Slower plume velocity, longer aerosol cloud duration for ease of use.


4.4 Excipient Selection & Formulation Considerations

  • Solubility & Stability: API solubility in propellant or suspension media; use of co‑solvents.

  • Surfactants: Poloxamers or lecithin to stabilize suspensions in pMDIs.

  • Carrier Selection: Lactose monohydrate properties (particle size, surface roughness) for DPIs.

  • Hygroscopicity: Minimized to prevent powder agglomeration in DPIs.

  • Osmolarity & pH: For nebulized solutions, matched to pulmonary lining fluid (≈300 mOsm, pH 7.0–7.4).


4.5 Manufacturing & Quality Control

4.5.1 Filling & Assembly

  • pMDIs: Under low‑temperature, high‑pressure filling of propellant–API blend into canisters, crimping with metering valve.

  • DPIs: Blend milling → mixing → capsule or reservoir filling → sealing to protect from moisture.

4.5.2 Critical Quality Attributes

AttributeMethodAcceptance Criteria
Delivered Dose UniformityDose collection apparatus (e.g., Copley)RSD ≤10% across 10 doses
Aerodynamic Particle Size DistributionCascade impactor or laser diffractionMMAD 1–5 µm; FPF (≤5 µm) >30%
Spray Pattern & Plume Geometry (pMDI/SMI)High‑speed photographyConsistent plume angle (e.g., 15°–30°) and shot weight
Moisture Content (DPI)Karl Fischer titration<1% w/w
Microbial Load & Sterility (Nebulizer Solutions)USP <61>/<71>Sterile; bioburden limits as per pharmacopeia

4.5.3 Stability Testing

  • Accelerated Conditions: 40 °C/75% RH for 6 months.

  • Propellant Compatibility: Check valve and canister integrity, spray characteristics over time.


4.6 Pharmacokinetics & Clinical Applications

  • Local Delivery: β₂‑agonists, corticosteroids for asthma/COPD; rapid bronchodilation and anti‑inflammation.

  • Systemic Delivery: Insulin, calcitonin—avoids needle‑related issues; absorption correlates with particle size and formulation matrix.

  • Dose Scaling: Fine‑tune inhaled dose based on lung deposition efficiency (~10–20% of nominal dose reaches deep lung).


4.7 Key Points for Exams

  • Define MMAD and explain its impact on regional lung deposition.

  • Compare pMDIs vs. DPIs in terms of formulation, patient technique, and advantages.

  • List three deposition mechanisms and the particle size range each governs.

  • Outline the steps and critical parameters in pMDI canister filling.

  • Describe two quality‑control tests for inhalation aerosols and their acceptance criteria.

 

Unit 5: Transdermal & Topical Delivery Systems

This unit explores drug delivery across and into the skin for both local (topical) and systemic (transdermal) therapy, covering skin physiology, formulation strategies, permeation enhancement, product types, manufacturing, and quality control.


5.1 Skin Structure & Barrier Function

5.1.1 Layers of Skin

  • Stratum Corneum: Outermost, keratin‑rich dead cell layer; primary barrier to drug penetration (~10–20 µm thick).

  • Epidermis: Viable cell layers beneath the stratum corneum; limited diffusional resistance.

  • Dermis: Vascularized connective tissue; site of systemic drug uptake.

  • Hypodermis: Fatty layer; minor role in transdermal uptake.

5.1.2 Routes of Penetration

  • Intercellular: Through lipid matrix between corneocytes (major route).

  • Transcellular: Across corneocytes and lipid bilayers (less common).

  • Appendageal: Via hair follicles and sweat ducts (minor, but faster for large molecules).


5.2 Transdermal Drug Delivery Systems (TDDS)

5.2.1 Advantages & Limitations

  • Advantages:

    • Bypasses first‑pass metabolism.

    • Provides controlled, sustained release.

    • Improves patient compliance.

  • Limitations:

    • Only potent, low‑dose drugs with suitable lipophilicity (log P 1–3) and molecular weight (<500 Da).

    • Skin irritation and sensitization risks.

5.2.2 Design Principles

ParameterTarget/StrategyRationale
Drug PropertiesMW < 500 Da; log P 1–3; low melting pointFacilitates skin permeation
Flux (J)Maintain therapeutic flux (µg/cm²·h)Controlled plasma levels
Patch Size & LoadingBalance dose with skin area toleratedMinimize irritation, maximize compliance
Reservoir vs. MatrixReservoir: liquid/semi‑solid drug chamber; Matrix: drug dispersed in polymerMatrix simpler, lower risk of dose dumping

5.2.3 Patch Types

  • Matrix (Drug‑in‑Adhesive): Drug blended into adhesive layer; simplest design.

  • Reservoir: Drug solution or gel in a compartment; rate‑controlling membrane separates drug from skin.

  • Microreservoir: Drug micro‑droplets within polymer matrix combining features.

  • Drug‑in‑Adhesive on Film: Adhesive layer directly contacts skin, on backing film.

5.2.4 Permeation Enhancers

  • Chemical Enhancers: Ethanol, propylene glycol, oleic acid disrupt lipid bilayer.

  • Physical Methods: Iontophoresis (electrical current), sonophoresis (ultrasound), microneedles.


5.3 Topical Dosage Forms

5.3.1 Semisolid Preparations

  • Ointments: Hydrophobic bases (petrolatum, lanolin); occlusive, increase hydration and permeation.

  • Creams: Oil‑in‑water or water‑in‑oil emulsions; non‑greasy, patient‑friendly.

  • Gels: Hydrophilic polymer networks (carbomer, cellulose); rapid release, cooling effect.

  • Pastes: High powder content in ointment base; protective barrier for exuding lesions.

5.3.2 Solutions & Lotions

  • Aqueous or hydroalcoholic solutions for antiseptics, anti‑inflammatories; easy spreadability.

  • Lotions: Low‑viscosity suspensions or emulsions for hairy areas.

5.3.3 Transdermal Patches vs. Topical Formulations

FeatureTransdermal PatchTopical Cream/Gel
PurposeSystemic drug deliveryLocal skin/site-specific action
Contact Duration24 h or longerMinutes to hours
Dose ControlMetered release by designLess precise; depends on patient use

5.4 Manufacturing & Packaging

5.4.1 Patch Fabrication

  • Coating/Printing: Adhesive/drug matrix coated onto backing film.

  • Lamination: Layering backing, drug matrix, release liner.

  • Cutting/Die‑Punching: Defined patch shapes.

  • Packaging: Aluminum pouches for moisture and light protection.

5.4.2 Semisolid Production

  • Homogenization: Uniform drug dispersion in base using high‑shear mixers.

  • Emulsification: Controlled temperature and stirring for O/W or W/O creams.

  • Filling: Tubes or jars under hygienic conditions.


5.5 Quality Control Tests

TestTDDSTopical Semisolids
Content UniformitySingle‑unit assay (HPLC)Sampling from multiple sites in batch
In Vitro ReleaseFranz diffusion cell; receptor mediumFranz cell with synthetic membranes
Adhesion & TackPeel‑strength test; shear‑adhesionNot applicable
Skin Irritation (In Vitro/In Vivo)3D skin models; patch test in volunteersPatch test; Draize test in animals
Viscosity & RheologyBrookfield viscometer; spreadability
Microbial LimitsBioburden per USP <61>; preservative efficacy <51>Bioburden and preservative efficacy

5.6 Pharmaceutical Examples

  • Transdermal:

    • Nicotine Patch: Matrix system for smoking cessation.

    • Fentanyl Patch: Reservoir system for chronic pain.

  • Topical:

    • Hydrocortisone Cream: Emulsion for dermatitis.

    • Diclofenac Gel: Carbomer gel for musculoskeletal pain.


5.7 Key Points for Exams

  • Contrast matrix vs. reservoir patch designs and list one advantage of each.

  • Describe two chemical permeation enhancers and their mode of action.

  • Explain why the stratum corneum is the rate‑limiting barrier in transdermal delivery.

  • Outline the Franz diffusion cell setup and the parameters measured.

  • List three semisolid topical dosage forms and one key QC test for each.

 

Unit 6: Introduction to Novel Drug Delivery (Nanoparticles, Liposomes)

This unit introduces cutting‑edge particulate carriers—polymeric nanoparticles and liposomes—for targeted, controlled, and enhanced drug delivery, covering their design, preparation, characterization, applications, and quality considerations.


6.1 Rationale & Overview

  • Limitations of Conventional Systems: Poor solubility, rapid clearance, lack of targeting, systemic side effects.

  • Advantages of Nanocarriers:

    • Enhanced Solubility: Encapsulation of hydrophobic drugs.

    • Controlled Release: Sustained delivery via matrix diffusion or lipid bilayer release.

    • Targeting: Passive (EPR effect) and active (ligand‑mediated) targeting to tissues or cells.

    • Reduced Toxicity: Lower off‑target exposure.


6.2 Polymeric Nanoparticles

6.2.1 Types & Composition

TypeCompositionCharacteristics
NanocapsulesDrug core + polymeric shell (e.g., PLGA)Reservoir release, high encapsulation
NanospheresDrug dispersed throughout polymer matrixDiffusion‑controlled release
DendrimersBranched, tree‑like polymers (e.g., PAMAM)Precise size, surface functionality
Polymeric MicellesAmphiphilic block‑copolymers (e.g., PEG‑PLA)Self‑assembled core–shell carriers

6.2.2 Preparation Methods

  • Emulsion‑Solvent Evaporation: Polymer + drug in organic solvent emulsified in water → solvent removal → nanoparticles.

  • Nanoprecipitation: Polymer + drug dissolved in water‑miscible solvent added to water → spontaneous nanoparticle formation.

  • Ionic Gelation: Polyelectrolyte (e.g., chitosan) crosslinked with multivalent counterions (e.g., TPP) trapping drug.

6.2.3 Characterization

ParameterMethodAcceptance/Target
Particle Size & PDIDynamic Light Scattering (DLS)50–300 nm; PDI ≤0.2
Zeta PotentialLaser Doppler Electrophoresis±20 mV to ±40 mV for stability
MorphologyTransmission/Scanning Electron MicroscopySpherical, uniform
Encapsulation EfficiencyCentrifugation + HPLC assay of supernatant≥70% commonly
In Vitro ReleaseDialysis bag method; sampling over timeControlled release profile (e.g., 20% at 1 h, 80% at 24 h)

6.2.4 Applications

  • Cancer Therapy: Doxorubicin‑loaded PLGA nanoparticles for EPR‑mediated tumor targeting.

  • Vaccines: Antigen‑adsorbed chitosan nanoparticles enhancing mucosal immunity.

  • Gene Delivery: PAMAM dendrimers complexed with siRNA for cellular uptake.


6.3 Liposomes

6.3.1 Structure & Classification

  • Unilamellar Vesicles (ULV): Single phospholipid bilayer (Small: 20–100 nm; Large: 100–1000 nm).

  • Multilamellar Vesicles (MLV): Multiple concentric bilayers (0.5–10 μm).

  • Stealth (PEGylated) Liposomes: Surface‑grafted PEG to evade RES uptake.

  • Targeted Liposomes: Surface ligands (antibodies, peptides) for active targeting.

6.3.2 Composition

  • Phospholipids: Phosphatidylcholine, phosphatidylserine.

  • Cholesterol: Modulates bilayer fluidity and stability.

  • Charge Inducers: Phosphatidic acid (negative), stearylamine (positive).

6.3.3 Preparation Techniques

  • Thin‑Film Hydration: Lipid film hydrated with aqueous drug solution → MLVs → size reduction by sonication/extrusion.

  • Reverse‑Phase Evaporation: Water‑in‑oil emulsion formed then solvent removed → large aqueous core.

  • Ethanol Injection: Lipids in ethanol injected into aqueous phase → spontaneous ULV formation.

6.3.4 Characterization

ParameterMethodAcceptance/Target
Vesicle Size & PDIDLS100–200 nm for stealth liposomes; PDI ≤0.2
LamellarityCryo‑TEMSingle vs. multiple bilayers
Encapsulation EfficiencySeparation (gel filtration) + assayHydrophilic drugs ~30–50%; lipophilic ~90%
Surface ChargeZeta potentialSlightly negative (−10 to −30 mV) for stability
In Vitro ReleaseDialysis or diffusion cellSustained release over 24–72 h

6.3.5 Applications

  • Doxil®: PEGylated liposomal doxorubicin for reduced cardiotoxicity and enhanced tumor accumulation.

  • Visudyne®: Verteporfin liposomes for photodynamic therapy in macular degeneration.

  • AmBisome®: Amphotericin B liposomes for systemic fungal infections with reduced nephrotoxicity.


6.4 Regulatory & Stability Considerations

  • Sterilization:

    • Nanoparticles: Sterile filtration (0.22 µm) or aseptic processing.

    • Liposomes: Filtration for small vesicles; gamma irradiation sometimes used.

  • Stability:

    • Aggregation/Aggregation: Controlled by surface charge, cryoprotectants (trehalose) for lyophilization.

    • Oxidation/Hydrolysis: Phospholipid peroxidation prevented by antioxidants (α‑tocopherol) and pH control.

  • Regulatory Guidelines: ICH Q8–Q11 (pharmaceutical development, quality), USP <1131> Nanoparticles, EMA reflection papers.


6.5 Key Points for Exams

  1. Compare nanocapsules vs. nanospheres in structure and drug release.

  2. Outline the thin‑film hydration method for liposome preparation and subsequent size reduction steps.

  3. List four characterization parameters common to both nanoparticles and liposomes, with target ranges.

  4. Describe the EPR effect and its role in passive tumor targeting by nanoparticles.

  5. Explain the purpose of PEGylation on liposomes and its impact on pharmacokinetics.

0 0 votes
Article Rating
Subscribe
Notify of
guest
0 Comments
Oldest
Newest Most Voted
Inline Feedbacks
View all comments
Scroll to Top