B Pharmacy Sem 7: Novel Drug Delivery Systems
Discover sustained/controlled release, targeted nanocarriers, liposomes, implants, transdermal patches & evaluation methods for advanced drug delivery
Subject 4: Novel Drug Delivery Systems
- Introduction to NDDS: Concepts & Importance
- Sustained & Controlled Release Dosage Forms
- Targeted Drug Delivery Systems
- Vesicular Systems: Liposomes & Niosomes
- Micro- & Nanoparticulate Systems
- Implants & Transdermal Drug Delivery
- Evaluation of Novel Delivery Systems
Unit 1: Introduction to NDDS – Concepts & Importance
Novel Drug Delivery Systems (NDDS) encompass advanced formulations and devices designed to optimize the therapeutic performance of drugs by controlling where, when, and how fast a drug is released. This introduction lays the foundation by defining key terms, explaining the rationale for NDDS development, and highlighting their impact on modern pharmacotherapy.
1. Definitions
Novel Drug Delivery System (NDDS):
A formulation or device engineered to deliver active pharmaceutical ingredients (APIs) in a way that improves efficacy, minimizes side effects, or enhances patient compliance, beyond what conventional dosage forms (tablets, capsules, injections) can achieve.
Key Concepts:
Controlled Release: Precise modulation of drug release rate over time.
Targeted Delivery: Directing drug molecules to specific tissues, cells, or subcellular sites.
Site‑Specific Delivery: Releasing drug at the disease locus to maximize local concentration.
Responsive (Smart) Delivery: Systems that alter release in response to stimuli (pH, temperature, enzymes).
2. Rationale & Advantages
Enhanced Therapeutic Index
Improved Patient Compliance
Less frequent dosing (e.g., once‑daily patches or implants).
Non‑invasive routes (transdermal, nasal, pulmonary) for patients averse to injections.
Overcoming Biological Barriers
Protection of labile drugs (peptides, proteins) from enzymatic degradation.
Enhanced permeation across barriers (intestinal epithelium, blood–brain barrier).
Reduced Variability
Economic & Logistical Benefits
Lower healthcare costs through reduced hospitalizations (e.g., depot formulations).
Simplified supply chain via stable, long‑acting products.
3. Historical Perspective
4. Challenges & Considerations
5. Impact on Therapy
6. Key Exam Tips
Define NDDS and list at least four advantages over conventional dosage forms.
Discuss one historical milestone (e.g., osmotic pump development) and its significance.
Explain key challenges in NDDS development, including regulatory and safety considerations.
Provide examples of therapeutic areas where NDDS have made a major impact (oncology, CNS).
Unit 2: Sustained & Controlled Release Dosage Forms
Sustained and controlled release systems are engineered to release an active pharmaceutical ingredient (API) at predefined rates over extended periods, thereby maintaining therapeutic drug levels and improving patient compliance.
1. Definitions
Sustained Release (SR):
Formulations that prolong drug release beyond that of an immediate‑release dosage form, typically reducing dosing frequency but exhibiting a gradually declining release rate.
Controlled Release (CR):
Systems designed to deliver drug at a constant, predictable rate (often zero‑order kinetics), maintaining plasma concentrations within the therapeutic window for the designated period.
2. Design Principles
Rate‑Controlling Membranes (Reservoir Systems)
Matrix Systems
Hydrophilic Matrices: Hydrate upon contact with gastrointestinal fluids; gel layer controls diffusion and erosion (e.g., HPMC-based tablets).
Hydrophobic Matrices: API dispersed in insoluble polymer (e.g., ethylcellulose); drug release via diffusion through polymer channels.
Osmotic Pumps
Design: Semi‑permeable membrane encloses core with API and osmogen.
Mechanism: Water influx generates osmotic pressure, forcing drug solution out through a delivery orifice at near zero‑order rate (e.g., OROS® systems).
Multiparticulate Systems (Pellets, Beads)
Structure: Small coated units each functioning as a micro‑reservoir or matrix.
Advantage: Uniform GI transit, reduced risk of dose dumping, flexible release profiles via varying coat thickness.
3. Examples & Applications
System | Example Product | Use |
---|
Reservoir Tablet | Theophylline SR capsules | Chronic asthma management |
Hydrophilic Matrix | Metformin SR tablets | Type 2 diabetes mellitus |
Osmotic Pump | Nifedipine OROS® tablets | Hypertension control |
Multiparticulate Beads | Pantoprazole DR granules | Gastro‑intestinal acid suppression |
4. Advantages & Limitations
Advantages:
Reduced dosing frequency enhances adherence.
Stable plasma levels minimize peaks and troughs, reducing side‑effects.
Flexibility in tailoring release profiles via polymer selection and coat thickness.
Limitations:
Complex manufacturing with stringent quality controls.
Risk of dose dumping if membrane ruptures or coating defects occur.
Higher cost relative to immediate‑release forms.
Inappropriate for drugs with very short half‑lives (<2 h), narrow therapeutic indices, or those requiring rapid onset.
5. Evaluation Methods
6. Key Exam Tips
Differentiate zero‑order vs. first‑order release kinetics and name a system for each.
Diagram a reservoir tablet, labeling core, rate‑controlling membrane, and delivery orifice.
List three physicochemical properties of APIs that influence suitability for SR/CR (e.g., solubility, dose size, half‑life).
Explain IVOS (in vitro–in vivo) Level A correlation and its regulatory importance.
Shall we move on to Unit 4.3: Targeted Drug Delivery Systems, or would you like any clarifications here?
Unit 3: Targeted Drug Delivery Systems
Targeted Drug Delivery Systems (TDDS) are designed to concentrate a therapeutic agent at its site of action while minimizing systemic exposure, thereby improving efficacy and reducing off‑target side effects.
1. Definition & Rationale
2. Mechanisms of Targeting
Passive Targeting
Enhanced Permeability and Retention (EPR) Effect: Leaky tumor vasculature and poor lymphatic drainage allow nanoparticles (100–200 nm) to accumulate preferentially in tumor tissue.
Macrophage Uptake: Liposomes and particulates are naturally taken up by the reticuloendothelial system (useful for targeting liver or spleen).
Active Targeting
Physical Targeting
Magnetic Targeting: Magnetic nanoparticles guided by external magnetic fields to localize at a specific site.
Ultrasound‑Triggered Release: Microbubbles or echogenic liposomes burst upon ultrasound exposure, releasing their payload at the target.
3. Carrier Systems
Carrier Type | Key Features | Applications |
---|
Liposomes | Biocompatible phospholipid bilayers; modifiable surface (PEGylation) | Doxil® for cancer; Amphotericin B liposomes |
Polymeric Nanoparticles | Controlled size/charge; biodegradable polymers (PLGA) | Paclitaxel‑loaded nanoparticles |
Antibody–Drug Conjugates (ADCs) | Monoclonal antibody linked to cytotoxic drug | Brentuximab vedotin; trastuzumab emtansine |
Micelles | Amphiphilic block copolymers form core–shell structures | Solubilizing poorly soluble drugs |
Dendrimers | Highly branched polymers with surface functional groups | Gene delivery; imaging agents |
4. Design Considerations
5. Evaluation of Targeting Efficiency
In Vitro
Binding assays: Measure ligand–receptor interactions (e.g., ELISA, flow cytometry).
Cellular Uptake Studies: Quantify internalization in target vs. non‑target cells (fluorescence microscopy, quantitative assays).
In Vivo
Biodistribution Studies: Radiolabel or fluorescently label carriers and track organ/tumor accumulation in animal models.
Therapeutic Efficacy: Compare tumor growth inhibition or survival in treated vs. control groups.
Toxicity Assessment: Monitor off‑target organ toxicity (histopathology, serum biomarkers).
6. Advantages & Limitations
Advantages:
Enhanced drug accumulation at disease sites leading to improved outcomes.
Reduced systemic side effects and lower required doses.
Potential for combinatorial therapies (e.g., co‑delivery of drugs and imaging agents).
Limitations:
Heterogeneous tumor vasculature can limit EPR-based accumulation.
Immunogenicity or hypersensitivity reactions to carrier materials.
Complex manufacturing and scale‑up challenges.
High development and regulatory costs.
7. Key Exam Tips
Differentiate passive vs. active vs. physical targeting with one example each.
List four carrier systems used in TDDS and a key feature of each.
Explain why PEGylation enhances circulation time and how it is achieved.
Outline an in vivo study design to assess biodistribution of a targeted nanoparticle.
Unit 4: Vesicular Systems – Liposomes & Niosomes
Vesicular systems are spherical carriers with one or more bilayer membranes that encapsulate drugs, enabling both hydrophilic and lipophilic compounds to be delivered effectively. Two principal types—liposomes and niosomes—offer customizable properties for targeted and controlled release applications.
1. Definitions & Structure
Liposomes:
Niosomes:
Definition: Non‑ionic surfactant vesicles analogous to liposomes, where the bilayer is formed from surfactants (e.g., Span, Tween) and cholesterol.
Structure:
2. Preparation Methods
Thin Film Hydration (Bangham Method):
Reverse‑Phase Evaporation:
Ether Injection:
Microfluidization & Sonication:
3. Drug Encapsulation & Release Mechanisms
Hydrophilic Drugs: Localize in the aqueous core; release via diffusion through the bilayer.
Lipophilic Drugs: Partition into the lipid bilayer; release as bilayer degrades or through partitioning into biological membranes.
Release Control: Adjust lipid composition, cholesterol content, and vesicle size to modulate membrane permeability and circulation half‑life.
4. Surface Modification & Targeting
5. Applications
System | Key Drug Examples | Therapeutic Use |
---|
Conventional Liposomes | Doxorubicin (Doxil®) | Cancer chemotherapy; reduced cardiotoxicity |
Stealth Liposomes | PEGylated liposomal amphotericin B | Systemic fungal infections; prolonged half‑life |
Niosomes | Diclofenac niosomal gel | Topical anti‑inflammatory therapy |
Targeted Vesicles | Folate‑grafted liposomes with methotrexate | Folate‑receptor–positive tumors |
6. Advantages & Limitations
Advantages:
Encapsulation of both hydrophilic and lipophilic drugs.
Biocompatible and biodegradable (liposomes).
Reduced systemic toxicity through targeted delivery.
Surface modification enables “stealth” behavior and active targeting.
Limitations:
Physical/chemical stability issues: vesicle aggregation, drug leakage.
High production costs and complex scalability.
Potential for rapid clearance by the reticuloendothelial system if not PEGylated.
Limited loading capacity for certain drugs.
7. Evaluation & Characterization
Vesicle Size & Distribution: Dynamic light scattering (DLS) for mean diameter and polydispersity index.
Zeta Potential: Surface charge measurement indicates colloidal stability.
Encapsulation Efficiency: Quantify the percentage of drug entrapped vs. total drug added.
In Vitro Release: Dialysis or Franz diffusion cells to monitor release kinetics under physiological conditions.
Morphology: Transmission electron microscopy (TEM) or cryo‑TEM to visualize vesicle structure.
8. Key Exam Tips
Compare liposomes vs. niosomes in terms of components, stability, and cost.
Outline one preparation method (e.g., thin film hydration) step by step.
List three factors affecting encapsulation efficiency.
Explain how PEGylation prolongs circulation and prevents opsonization.
Unit 4.5: Micro‑ & Nanoparticulate Systems
Micro‑ and nanoparticulate systems encompass solid particles sized from nanometers (1–1,000 nm) to micrometers (1–100 µm), designed to control release, enhance bioavailability, and target drugs to specific tissues.
1. Definitions
Microspheres / Microcapsules:
Spherical particles (1–100 µm) in which the drug is either dispersed within a polymer matrix (microsphere) or enclosed within a polymer shell (microcapsule).
Nanoparticles:
Particles <1,000 nm, including polymeric nanoparticles, solid lipid nanoparticles (SLNs), and nanocrystals, engineered for enhanced cellular uptake, lymphatic transport, or CNS delivery.
2. Materials & Composition
Polymers:
Biodegradable: PLA, PLGA, PCL—degrade into non‑toxic monomers.
Non‑biodegradable: Ethylcellulose—requires removal or remains as depot.
Lipids:
SLNs: Solid lipid cores (e.g., glyceryl behenate) stabilized by surfactants.
Nanostructured Lipid Carriers (NLCs): Blend of solid and liquid lipids to improve drug loading.
Inorganic:
Mesoporous silica nanoparticles (MSNs): High surface area, tunable pore sizes.
Gold & Iron Oxide: For imaging, hyperthermia, or magnetic targeting.
3. Preparation Techniques
Emulsion‑Solvent Evaporation
Salting‑Out / Precipitation
High‑Pressure Homogenization
Spray Drying
Supercritical Fluid Technology
4. Drug Loading & Release Mechanisms
5. Applications
System | Drug Example | Therapeutic Area |
---|
PLGA Microspheres | Lupron Depot (leuprolide) | Prostate cancer, endometriosis |
Solid Lipid Nanoparticles | Tacrolimus SLNs | Dermal delivery, immunosuppression |
Nanocrystals | Itraconazole nanocrystals | Oral bioavailability enhancement |
Mesoporous Silica NPs | Camptothecin-loaded MSNs | Targeted cancer therapy |
6. Advantages & Limitations
Advantages:
Controlled release and sustained plasma levels.
Enhanced solubility and bioavailability of poorly water‑soluble drugs.
Targeted delivery via size‑dependent tissue penetration or ligand attachment.
Limitations:
Complex manufacturing and scale‑up challenges.
Potential cytotoxicity of certain nanomaterials.
Regulatory uncertainties for novel inorganic carriers.
Physical stability concerns: aggregation, sedimentation.
7. Characterization Techniques
Particle Size & Distribution: Dynamic light scattering (DLS), laser diffraction.
Morphology: Scanning/transmission electron microscopy (SEM/TEM).
Surface Charge: Zeta potential to predict colloidal stability.
Thermal Properties: DSC/TGA to assess polymer crystallinity and drug–polymer interactions.
In Vitro Release: Dialysis, sample-and-separate methods under physiological conditions.
8. Key Exam Tips
Differentiate microspheres vs. microcapsules vs. nanoparticles with definitions and size ranges.
List three preparation methods and a key advantage of each (e.g., supercritical fluids for narrow size).
Explain how polymer selection (e.g., PLA vs. PLGA) affects degradation rate and release kinetics.
Describe one application that leverages the enhanced permeability of nanoparticles in cancer therapy.
Unit 4.6: Implants & Transdermal Drug Delivery
This unit examines two delivery modalities—implantable devices for long‑term, controlled dosing and transdermal systems that offer non‑invasive, sustained drug administration through the skin.
1. Definitions & Rationale
Implants:
Sterile, often biodegradable or non‑biodegradable devices placed subcutaneously or intramuscularly to release drugs over weeks to years.
Transdermal Drug Delivery Systems (TDDS):
Patch‑based formulations that deliver drugs across the skin into systemic circulation, providing sustained plasma levels without gastrointestinal involvement.
Rationale:
Improve adherence by reducing dosing frequency.
Bypass first‑pass metabolism, enhancing bioavailability.
Provide steady drug levels, minimizing peaks/troughs associated with oral dosing.
2. Implantable Systems
2.1 Types of Implants
Biodegradable Implants
Polymers (PLGA, PLA) gradually degrade, releasing drug and eliminating the need for removal.
Example: Leuprolide‑loaded PLGA rods (Lupron Depot) for prostate cancer.
Non‑biodegradable Implants
Constructed from inert materials (silicone, ethylene vinyl acetate); require surgical removal after drug depletion.
Example: Norplant® levonorgestrel rods for contraception (now replaced by more modern systems).
2.2 Design & Release Mechanisms
Matrix Implants: Drug uniformly dispersed in polymer matrix; release via diffusion and polymer erosion.
Reservoir Implants: Core containing drug surrounded by rate‑controlling membrane; achieve near zero‑order kinetics.
2.3 Key Considerations
Biocompatibility: Implant materials must not provoke immune reactions.
Mechanical Strength: Must withstand bodily movements without fracturing.
Sterility: Manufactured and implanted under aseptic conditions to prevent infection.
Removal (if non‑biodegradable): Surgical retrieval procedure planning.
2.4 Applications
Oncology: Depot chemotherapy or hormone therapy (e.g., goserelin implants).
Endocrinology: Growth hormone or insulin implants for chronic conditions.
Cardiology: Anticoagulant or antiarrhythmic implants for sustained therapy.
3. Transdermal Drug Delivery Systems
3.1 System Components
Backing Layer: Protects patch from environment.
Drug Reservoir or Matrix: Contains the API, with or without a rate‑controlling membrane.
Adhesive Layer: Ensures patch remains in contact with skin.
Release Liner: Peel‑off film protecting the drug layer before application.
3.2 Release Mechanisms
Matrix Patches: API uniformly dispersed in adhesive or polymer matrix; released by diffusion through skin.
Reservoir Patches: Drug stored in a liquid reservoir separated by a semi‑permeable membrane that controls release rate.
3.3 Enhancers & Devices
Chemical Enhancers: Compounds (e.g., ethanol, oleic acid) that disrupt stratum corneum lipids to increase permeability.
Physical Enhancers: Microneedles or iontophoresis that create micro‑channels or use electrical current to drive molecules.
3.4 Common Products & Drugs
Drug | Patch Type | Indication |
---|
Nicotine | Matrix | Smoking cessation |
Fentanyl | Reservoir | Chronic pain management |
Scopolamine | Reservoir | Motion sickness prevention |
Clonidine | Matrix | Hypertension control |
3.5 Advantages & Limitations
Advantages:
Non‑invasive and self‑administered.
Bypass GI tract, reducing GI side effects and first‑pass loss.
Controlled, sustained delivery for days to weeks.
Limitations:
Skin irritation or sensitization at application site.
Limited to potent drugs with low daily dose requirements.
Variability due to skin condition, age, and site of application.
4. Evaluation & Testing
In Vitro Skin Permeation Studies:
Adhesion Testing:
Skin Irritation & Sensitization:
In Vivo Pharmacokinetic Studies:
5. Key Exam Tips
Differentiate biodegradable vs. non‑biodegradable implants with examples.
Diagram a transdermal patch, labeling each layer.
List two chemical and two physical penetration enhancers.
Explain the concept of steady‑state flux and how it relates to patch design.
Unit 4.7: Evaluation of Novel Delivery Systems
Evaluation of NDDS is critical to confirm that the system delivers the drug safely, effectively, and reproducibly. This unit outlines the in vitro, in vivo, and analytical methods used to characterize and validate novel delivery platforms.
1. In Vitro Evaluation
Release Kinetics Studies
Purpose: Quantify drug release profile under simulated physiological conditions.
Methods:
USP Apparatus I/II (Basket/Paddle): For tablets, microspheres.
Franz Diffusion Cells: For transdermal patches, vesicular systems.
Data Analysis: Fit cumulative release vs. time data to kinetic models (zero‑order, first‑order, Higuchi, Korsmeyer–Peppas) to elucidate release mechanisms.
Particle/Vesicle Characterization
Size & Distribution: Dynamic Light Scattering (DLS) for mean diameter and polydispersity index.
Morphology: Electron microscopy (SEM, TEM) to visualize shape and surface.
Surface Charge: Zeta potential measurements to predict colloidal stability.
Encapsulation Efficiency & Drug Loading
Encapsulation Efficiency (EE%):
EE%=Total drug addedAmount of drug encapsulated×100
Drug Loading (%): Ratio of drug weight to total particle weight.
Physicochemical Stability
Mechanical & Adhesion Testing
2. In Vivo Evaluation
Pharmacokinetic (PK) Studies
Biodistribution & Targeting Efficiency
Efficacy & Pharmacodynamic (PD) Studies
Disease Models: Animal models (e.g., tumor xenografts, inflammation models) to assess therapeutic outcomes.
Endpoints: Tumor volume reduction, symptom scores, biomarker changes.
Toxicity & Safety Assessment
Acute & Chronic Toxicity: Monitor animal weight, organ histopathology, serum chemistry.
Immunogenicity: Evaluate antibody responses to carriers, complement activation.
3. Analytical & Regulatory Considerations
Method Validation
Analytical assays (HPLC, LC–MS/MS) must be validated for accuracy, precision, specificity, linearity, LOD/LOQ per ICH Q2(R1).
Quality by Design (QbD) & Critical Quality Attributes (CQAs)
Scale‑Up & Process Validation
Documentation for Regulatory Submission
Compile comprehensive CMC data: formulation composition, manufacturing process, in vitro/in vivo correlation (IVIVC), stability studies.
4. Establishing In Vitro–In Vivo Correlation (IVIVC)
Levels of IVIVC
Applications of IVIVC
Modeling & Simulation
5. Key Exam Tips
List at least four in vitro tests and their purposes (e.g., DLS for size, Franz cell for release).
Describe a Level A IVIVC and its regulatory significance.
Outline an in vivo study design to assess both PK and biodistribution of a nanoparticle.
Explain why method validation is critical for NDDS analytical assays (tie to ICH Q2).