Table of Content
•Introduction to Pre formulation
• Historical Context and Evolution of Pre formulation
• Fundamental Objectives of Pre formulation
• Physical Characterization and Chemical Stability Aspects
• Biopharmaceutical Properties and Excipient Compatibility Studies
• Advanced Pre formulation Techniques and its Integration with Formulation
Development
• Regulatory Considerations and Future Directions of Pre formulation
• Stability Analysis in Industrial Pharmacy
• Purpose and types of Stability Analysis in Industrial Pharmacy
• Factors Influencing Drug Stability:
• Key Elements to Design Stability Studies
3.
Introduction to Preformulation
Pre formulation is the initial and critical phase in drug
development where scientists study the physical, chemical, and
mechanical properties of a drug substance. This process helps
determine how the drug will behave in different formulations
and ensures optimal stability, efficacy, and bioavailability.
Think of pre formulation as a "background check" for a new drug
molecule before it enters full-scale formulation development. It
helps identify potential challenges early, saving time, money, and
effort in later stages.
4.
Historical Context andEvolution of Pre formulation
Let's take a look at how pre formulation has progressed over time.
1. Early Drug Preparation (Ancient to 19th Century)
• In ancient civilizations (Egyptian, Greek, Chinese, and Indian),
medicines were formulated based on empirical knowledge, often
using natural substances like herbs, minerals, and animal extracts.
• There was little scientific understanding of drug stability, solubility, or
bioavailability. Instead, trial-and-error methods were used to
determine the best way to administer medications.
5.
• During theRenaissance and early modern periods, alchemists and
apothecaries started refining drug preparations, but there was still no
formalized approach to studying drug properties before formulation.
2. Rise of Pharmaceutical Sciences (Late 19th to Early 20th Century)
• With the advent of organic chemistry and microbiology, scientists began
studying drug compounds more systematically.
• Early research focused on basic physicochemical properties, such as
melting points, solubility, and chemical reactivity.
• The rise of industrial-scale drug manufacturing led to the need for better
quality control and reproducibility in drug formulations.
3. Birth of Pre formulation as a Formal Discipline (Mid-20th Century)
6.
• The 1950sand 1970s saw major advancements in drug development due
to improved analytical techniques like chromatography and spectroscopy.
• Scientists realized that understanding a drug’s physicochemical,
mechanical, and biopharmaceutical properties before formulation was
crucial for successful drug development.
• Pharmaceutical companies began establishing pre formulation as a
distinct phase in drug development, focusing on solid-state properties,
solubility enhancement, and stability testing.
4. Modern Pre formulation Science (Late 20th Century to Present)
• With the advent of computational chemistry, molecular modeling, and
high-throughout screening, predictive pre formulation studies have
become more efficient.
7.
• Advanced techniquessuch as differential scanning
calorimetry (DSC), X-ray diffraction (XRD), and nuclear
magnetic resonance (NMR) are now used to characterize
drug substances.
• The focus has shifted toward developing targeted drug
delivery systems, improving bioavailability, and designing
formulations for personalized medicine.
• Regulatory agencies (FDA, EMA, etc.) now emphasize pre
formulation data in new drug applications to ensure
safety and efficacy
8.
Fundamental Objectives ofPre formulation
Pre formulation serves as the foundation for successful drug formulation by
systematically studying the physical, chemical, and biological properties of a
drug candidate. The main objectives of pre formulation include:
1. Understanding Physicochemical Properties
• Solubility & Dissolution Rate: Determines how easily the drug dissolves in
biological fluids, affecting absorption and bioavailability.
• Particle Size & Shape: Influences drug flow properties, dissolution rate,
and stability.
• Polymorphism: Identifies different crystalline forms, which can impact
solubility, stability, and therapeutic effectiveness.
9.
• Hygroscopicity: Assessesthe drug’s ability to absorb moisture, which can
affect stability and shelf life.
2. Ensuring Chemical Stability
• Evaluates degradation pathways (e.g., hydrolysis, oxidation,
photodegradation) to enhance drug stability.
• Helps determine suitable storage conditions and packaging materials.
• Identifies possible interactions with excipients that could affect drug
potency.
3. Enhancing Bioavailability
• Determines lipophilicity (log P value), which affects drug absorption and
10.
• Guides theselection of appropriate formulation strategies (e.g., salt
formation, nanoparticles, lipid-based delivery).
4. Selecting Suitable Drug Formulation Approaches
• Identifies the best excipients to enhance stability, solubility, and drug
release.
• Guides the choice of dosage form (tablets, capsules, injections, etc.) based
on drug properties.
• Helps in designing controlled or targeted drug delivery systems.
5. Supporting Regulatory & Manufacturing Requirements
• Provides essential data for regulatory approvals by agencies like the FDA or
EMA.
11.
Physical Characterization andChemical Stability
Aspects of Pre formulation
Pre formulation studies focus on understanding the physical and chemical
properties of a drug substance to ensure successful formulation development.
These studies help in optimizing drug stability, bioavailability, and overall
performance.
1. Physical Characterization
Physical characterization evaluates the solid-state properties of the drug, which
impact formulation, processing, and drug performance.
Key Physical Properties Studied in Pre formulation:
a) Particle Size and Shape d) Crystallinity and Amorphous Nature
b) Polymorphism e) Flow Properties and Compressibility
12.
a) Particle Sizeand Shape
• Affects dissolution rate, bioavailability, and uniformity in drug
formulations.
• Smaller particle sizes generally lead to faster dissolution and
absorption.
• Larger or irregularly shaped particles may cause flowability issues
during manufacturing.
b) Polymorphism
• Many drugs exist in multiple crystalline forms (polymorphs), which
have different solubilities and stabilities.
• Some polymorphs may be more bioavailable, while others may be
more stable.
13.
c) Hygroscopicity
• Describesthe drug’s tendency to absorb moisture from the
environment.
• High hygroscopicity can lead to instability, degradation, or poor flow
properties.
• Measured using techniques like Dynamic Vapor Sorption (DVS).
d) Crystallinity and Amorphous Nature
• Crystalline drugs have high stability but may have low solubility.
• Amorphous drugs dissolve faster but are often less stable.
• X-ray Diffraction (XRD) and Differential Scanning Calorimetry (DSC)
help characterize these properties.
14.
e) Flow Propertiesand Compressibility
• Important for tablet and capsule formulations.
• Poor flow properties can lead to manufacturing issues like inconsistent
drug dosing.
• Measured using angle of repose, bulk density, and Carr’s index.
2. Chemical Stability Aspects
Chemical stability studies help identify degradation pathways and develop
strategies to improve drug stability.
Key Chemical Stability Parameters in Pre
formulation
a) Hydrolysis (Water-Induced Degradation) d) Temperature Sensitivity
b) Oxidation e) Drug-Excipient Compatibility
15.
a) Hydrolysis (Water-InducedDegradation)
• Common in ester, amide, and lactone drugs (e.g., aspirin).
• Affected by humidity, pH, and formulation composition.
• Controlled by using anhydrous formulations, pH buffers, or protective
coatings.
b) Oxidation
• Drugs with hydroxyl, thiol, or aromatic amine groups are prone to oxidation.
• Oxygen, light, and temperature can lead to degradation in these.
c) Photodegradation (Light-Induced Instability)
Certain drugs which degrade under UV or visible light exposure in these.
16.
d) Temperature Sensitivity
•High temperatures can lead to chemical degradation or polymorphic
transformations.
• Conducted using accelerated stability testing (e.g., 40°C and 75% relative
humidity for 6 months).
e) Drug-Excipient Compatibility
• Some excipients may react with the drug, affecting its stability and efficacy.
• Studied using Fourier Transform Infrared Spectroscopy (FTIR), Differential
Scanning Calorimetry (DSC), and High-Performance Liquid Chromatography
(HPLC).
17.
Biopharmaceutical Properties andExcipient
Compatibility Studies
Pre formulation is a critical phase in drug development, where the
physicochemical and biopharmaceutical properties of a drug candidate are
assessed to optimize its formulation. A key aspect of pre formulation is
studying the compatibility of the drug with excipients to ensure stability,
efficacy, and manufacturability. Biopharmaceutical properties influence
drug absorption, distribution, metabolism, and excretion (ADME)
Biopharmaceutical Properties in Pre formulation
Key properties include:
18.
1. Solubility &Dissolution Rate
• Determines the drug's bioavailability.
• Poorly soluble drugs require solubility enhancement techniques like salt
formation, nanotechnology, or amorphous solid dispersions.
2. Permeability
• Assesses drug absorption across biological membranes (e.g., intestinal walls).
• Evaluated using techniques like Caco-2 cell assays or PAMPA (Parallel Artificial
Membrane Permeability Assay).
3. Partition Coefficient (Log P) & Distribution Coefficient (Log D)
Log P: Measures lipophilicity (affects drug permeability & distribution) ,While Log D:
Adjusted for pH dependency, relevant for ionizable drugs.
19.
4. pKa (IonizationConstant)
• Influences solubility, permeability, and stability.
• Weak acids/bases change ionization based on pH, affecting drug behaviour in
the body.
5. Stability & Degradation Kinetics
• Evaluates drug stability under different conditions (pH, temperature, light,
humidity).
• Determines shelf-life and formulation strategies.
Excipient Compatibility Studies
Excipients are inactive ingredients used to enhance drug formulation. However, drug-
excipient interactions can lead to instability, affecting drug performance.
20.
1. Importance ofExcipient Compatibility Studies
• Prevents degradation due to interactions (e.g., oxidation, hydrolysis).
• Ensures uniformity and stability of the final formulation.
• Avoids potential toxic byproducts.
2. Methods for Drug-Excipient Compatibility Studies
a. Differential Scanning Calorimetry (DSC)-Detects physical or chemical
interactions through changes in thermal properties.
b. Thermogravimetric Analysis (TGA)-Assesses weight loss under heat, indicating
possible degradation.
21.
c. Fourier-Transform InfraredSpectroscopy (FTIR)-Identifies
molecular interactions between drug and excipients (e.g., hydrogen
bonding, oxidation).
d. X-Ray Powder Diffraction (XRPD)-Determines changes in drug
crystallinity due to excipients.
e. High-Performance Liquid Chromatography (HPLC)-Quantifies
degradation products over time.
f. Stress Testing & Forced Degradation Studies-Evaluates
compatibility under extreme conditions (pH variations, humidity,
UV light).
22.
Advanced Pre formulationTechniques and its
Integration with Formulation Development
Pre formulation studies play a crucial role in drug development by
assessing the physicochemical, biopharmaceutical, and stability
properties of a drug candidate. Advanced pre formulation techniques
provide deeper insights into drug behavior, enabling better
formulation design, improved drug stability, and enhanced
bioavailability. Integrating these techniques with formulation
development ensures a rational approach to developing effective and
stable pharmaceutical products.
23.
Advanced Pre formulationTechniques
1. Solid-State Characterization-Understanding the solid-state properties of a drug helps in
optimizing its stability and performance in formulation.
a. X-Ray Powder Diffraction (XRPD)-Determines the crystallinity of a drug (amorphous vs.
crystalline).also helps identify polymorphic forms, which impact solubility and
bioavailability.
b. Differential Scanning Calorimetry (DSC)-Measures thermal transitions such as melting
point, glass transition, and polymorphic transitions and Detects drug-excipient
interactions that might cause instability.
c. Thermogravimetric Analysis (TGA)-Assesses moisture content and thermal degradation
properties and Helps in selecting appropriate drying techniques for formulation.
d. Hot-Stage Microscopy (HSM) -Visualizes phase transitions and drug-excipient
interactions at elevated temperatures.
24.
2. Drug-Excipient CompatibilityStudies
Advanced techniques ensure the stability of the drug in the presence of excipients.
a. Fourier-Transform Infrared Spectroscopy (FTIR)
• Identifies chemical interactions between drug and excipients.
• Detects potential degradation mechanisms (oxidation, hydrolysis).
b. Isothermal Microcalorimetry (IMC)
• Measures heat changes during drug-excipient interaction.
• Useful for detecting incompatibility at an early stage.
c. High-Performance Liquid Chromatography (HPLC)
• Monitors the degradation kinetics of drug-excipient mixtures.
• Provides quantitative stability data for formulation design.
25.
3. Advanced Solubilityand Permeability Assessment
Enhancing drug solubility and permeability is critical for optimizing bioavailability.
a. Solubility Enhancement Techniques
• pH Adjustment & Salt Formation – Converts poorly soluble drugs into ionizable
forms.
• Cocrystals & Solid Dispersions – Improves dissolution rate by altering crystalline
properties.
• Lipid-Based Formulations – Enhances solubility for lipophilic drugs.
b. Parallel Artificial Membrane Permeability Assay (PAMPA)
• A high-throughput method to predict passive diffusion permeability.
• Assists in selecting appropriate permeability enhancers.
26.
4. Particle Engineeringfor Improved Drug Performance
Particle properties influence drug dissolution, stability, and manufacturability.
a. Nanocrystal Technology
• Reduces particle size to nanoscale, increasing surface area and solubility.
• Improves bioavailability of poorly water-soluble drugs.
b. Spray Drying and Freeze Drying
• Enhances drug stability and dissolution characteristics.
• Useful for preparing amorphous solid dispersions.
c. Supercritical Fluid Technology
• Produces micronized or nano-sized drug particles with enhanced
dissolution rates.
27.
Integration of AdvancedPre formulation with
Formulation Development
1. Rational Excipient Selection
• Based on drug solubility, stability, and compatibility data.
• Ensures formulation stability and enhances drug performance.
2. Formulation Design Optimization
• Utilizing Quality by Design (QbD) principles to systematically study drug-excipient
interactions.
• Optimizing excipient ratios for better dissolution and controlled release.
28.
3. Predictive Modellingand Simulation
• In silico modelling predicts drug behaviour in various formulations.
• Computational tools simulate drug absorption and pharmacokinetics.
4. Stability and Scale-Up Considerations
• Ensuring formulation robustness under different storage and processing
conditions.
• Using advanced stability-indicating assays to optimize shelf-life.
29.
Regulatory Considerations andFuture Directions
of Pre formulation
• Pre formulation plays a vital role in pharmaceutical development by
ensuring a drug’s stability, bioavailability, and manufacturability. Regulatory
agencies like the FDA (Food and Drug Administration)and ICH
(International Council for Harmonization) provide guidelines to ensure
pre formulation studies support the safety, efficacy, and quality of drug
products. As pharmaceutical science evolves, future directions in pre
formulation include AI-driven predictive modeling, nanotechnology, and
personalized medicine.
30.
Regulatory Considerations inPre formulation
Regulatory agencies require thorough pre formulation studies to establish the
quality, safety, and effectiveness of a drug before moving into clinical trials
and commercial production. Key regulatory considerations include:
1. Physicochemical Characterization Compliance
• ICH Q6A & Q6B Guidelines require detailed physicochemical characterization
of drug substances, including:
• Polymorphism (ICH Q6A)
• Solubility & Dissolution
• pKa, Log P, and Permeability
• Hygroscopicity & Stability
31.
2. Drug-Excipient CompatibilityStudies
• ICH Q8 (Pharmaceutical Development) emphasizes excipient compatibility
testing to prevent degradation.
• Regulatory authorities require the use of DSC, FTIR, and HPLC to detect
incompatibilities.
3. Stability Testing Requirements
• ICH Q1A(R2) Stability Testing Guidelines mandate stability testing under different
conditions:
• Long-term storage (25°C/60% RH)
• Accelerated conditions (40°C/75% RH)
• Photostability testing (ICH Q1B)
• Helps determine shelf life and degradation pathways.
32.
4. Biopharmaceutical ClassificationSystem (BCS) & Bioavailability
Requirements
• The FDA’s BCS-based Biowaiver Guidance categorizes drugs into four classes:
• Class I (High solubility, High permeability) – Eligible for biowaiver (no in
vivo bioequivalence testing needed).
• Class II (Low solubility, High permeability) – Requires solubility
enhancement strategies.
• Class III (High solubility, Low permeability) – Needs permeability
enhancement techniques.
• Class IV (Low solubility, Low permeability) – High risk, needs extensive
formulation optimization.
33.
5. Good ManufacturingPractice (GMP) & Quality by Design (QbD)
• ICH Q8-Q11 Guidelines encourage the application of QbD principles to pre
formulation:
• Identifying Critical Quality Attributes (CQA).
• Using Design of Experiments (DoE) for formulation optimization.
• Implementing Process Analytical Technology (PAT) to monitor drug
properties in real-time.
Future Directions in Pre formulation
• With advancements in pharmaceutical technology, pre formulation is
evolving to include AI-driven predictive analytics, 3D printing,
34.
1. AI andMachine Learning in Pre formulation
• Predictive modelling for solubility, stability, and excipient
compatibility.
• AI-assisted molecular docking to design optimal drug formulations.
• Automated formulation optimization using big data and
algorithms.
2. Nanotechnology for Drug Delivery
• Nanocrystals & Liposomes for enhancing bioavailability.
• Polymeric Nanoparticles for controlled drug release.
• Lipid-based nanoparticles (SLNs & NLCs) for improved drug
35.
3. 3D Printingin Drug Formulation
• Allows personalized drug formulations with tailored dose, release profile,
and combination therapies.
• Approved 3D-printed drug: Spritam® (levetiracetam) for epilepsy, with fast
disintegration properties.
4. Biologics and Peptide Drug Pre formulation
• Pre formulation for monoclonal antibodies (mAbs) focuses on stability &
aggregation prevention.
• Peptide drugs require advanced techniques like lyophilization and
PEGylation for stability.
36.
5. Personalized Medicine& Gene Therapy Pre
formulation
• Advances in genomics and AI-driven drug design are
shifting drug development toward patient-specific
formulations.
• mRNA-based formulations (e.g., COVID-19 vaccines)
use lipid nanoparticles for stability and delivery.
37.
Stability Analysis inIndustrial Pharmacy
Stability studies of pharmaceutical products
may be expressed as the time during which the
pharmaceutical products retain its physical,
chemical, microbiological, pharmacokinetic
properties and characteristics throughout the
shelf life from the time of manufacture.
The shelf-life prediction is a major role for the
pharmaceutical product development of all the
dosage forms and also it is utilized to
determine the particular storage conditions
and to suggest
label instructions.
38.
Purpose of StabilityAnalysis in Industrial Pharmacy
Stability analysis in industrial
pharmacy is a critical process
used to determine how
environmental factors such
as temperature, humidity,
and light affect the quality,
safety, and efficacy of
pharmaceutical products
over time. The key purposes
of stability analysis include :
Regulatory Compliance
Ensuring Drug Efficacy and Safety
Determining Shelf Life and Expiry Date
Optimizing Storage Conditions
Formulation Development and
Optimization
39.
Types of StabilityStudies in Pharmaceuticals
1. Based on Duration and Storage Conditions
Long-Term
Stability
Studies:
Accelerated
Stability Studies:
Intermediate
Stability Testing:
These studies involve storing
samples of the drug product
under recommended storage
conditions for an extended
period, typically at room
temperature. Long-term
stability studies help
determine the product’s shelf
life and expiry date.
Samples are subjected to elevated
temperature and humidity
conditions for a shorter
duration to simulate
the effects of long-term storage
in a shorter time frame.
Accelerated studies provide
information on potential
degradation pathways and help
predict shelf life.
These studies are conducted at
conditions between long-term
and accelerated conditions. They
provide additional insights into
the product’s stability profile,
especially when accelerated
conditions alone may not be
sufficient.
40.
2. Based onStress Factors Applied :
These studies intentionally
subject the drug product to harsh
conditions, such as high
temperature, humidity, or
extreme pH, to accelerate
degradation and identify
potential degradation products
and pathways.
Photo-stability studies assess the
product’s susceptibility to
degradation caused by exposure
to light. This is particularly
important for products sensitive
to light, as degradation due to
photolysis can affect product
quality.
(a) Stress Testing (Forced Degradation
Studies):
(b) Photostability Testing
41.
3. Based onthe Product’s Usage:
(a) In-Use Stability Testing (b) Microbiological Stability Testing
•Purpose: Determines how long a product
remains stable after opening or mixing.
•Conditions: Real-world conditions based on
storage recommendations.
•Duration: Hours to weeks, depending on
product type.
•Example: Checking how long an insulin pen
remains stable after first use.
• Purpose: Ensures sterility and prevents
microbial contamination in multi-dose
formulations.
• Conditions: Tested under microbiological
challenge conditions.
• Example: Testing preservative efficacy in
multi-dose eye drops
INTERNAL FACTORS:
a.) Chemical
Structure
Thechemical structure of a drug plays a fundamental role in its
stability. Certain molecular arrangements are more prone to degradation
pathways, such as hydrolysis, oxidation, and photodegradation.
Structural modifications can impact a drug’s susceptibility to these
processes.
B.) pH Sensitivity
The pH of a drug’s environment can significantly affect its stability.
Some drugs are sensitive to changes in pH, which can influence their
solubility, chemical reactivity, and degradation rates.
45.
C.) Interaction with
Excipients
Theexcipients used in drug formulations can interact with the active
pharmaceutical ingredient (API) and impact its stability. Incompatibilities
between the API and excipients can lead to degradation or changes in
physical properties.
D.) Microbial
Contamination
•Water-based formulations are prone to microbial growth, leading to spoilage.
•Preservatives like benzalkonium chloride are added to prevent contamination.
•Example: Eye drops require antimicrobial preservatives to maintain sterility.
46.
External factors
1. Temperature
Temperatureis a critical factor affecting drug stability. High temperatures
can accelerate degradation pathways, while low temperatures can impact
solubility and physical characteristics of drugs.
2. Humidity
Humidity levels can impact the stability of drugs, particularly those
sensitive to moisture. Moisture can trigger hydrolysis reactions and
promote microbial growth in formulations.
47.
3. Light
Light, especiallyUV and visible light, can induce photodegradation of drugs.
Photo-sensitive compounds can undergo structural changes when exposed
to light, leading to loss of potency and the formation of degradation
products.
4. Oxygen
Oxygen exposure can lead to oxidative degradation, where drugs lose
electrons and undergo chemical changes. Packaging materials and storage
conditions play a crucial role in minimizing oxygen exposure.
48.
A well-designed stability
studyprotocol should include the following information:
1. Number of batches
2. Containers and Closures
3. Orientation of storage of
containers
4. Sampling time points
5. Test storage conditions
6. Test parameters.
49.
Key Elements toDesign Stability Studies:
The objectives of the study shall be
defined within guidelines concerning
with respect to regulations provided by
FDA, EMA, or ICH. Knowledge and
adherence to these guidelines are
essential in defining conditions
regarding how long a study should be
conducted, any testing parameters, and
what the acceptance criteria are to be to
be compliant with regulations
The stressors that may impact the
stability of the drug are temperature
differences, humidity, exposure to
light, and conditions related to
transportation. The appropriate test
conditions can be identified using
ICH guidelines, which classify
different climatic zones.
1. Regulatory Compliance: 2. Selection of Test Conditions:
50.
Regulatory Guidelines forStability Testing in India
CDSCO is India’s regulatory authority under the Ministry of Health and Family Welfare. It
follows Schedule M of the Drugs and Cosmetics Act, 1940, which outlines Good
Manufacturing Practices (GMP) for pharmaceutical products. Stability testing is
mandatory for New Drug Applications (NDA) and Abbreviated New Drug Applications
(ANDA) before approval.
(a) Central Drugs Standard Control Organization
(CDSCO)
51.
c. ICH StabilityGuidelines Followed in
India
India, as part of the ICH Global Cooperation Group (ICH-GCG), follows these guidelines:
•ICH Q1A (R2): Stability Testing of New Drug Substances and Products.
•ICH Q1B: Photostability Testing.
•ICH Q1E: Evaluation of Stability Data
(b) Indian Pharmacopoeia (IP) Guidelines
The Indian Pharmacopoeia Commission (IPC) provides stability testing protocols for drugs
marketed in India. IP standards align with ICH Q1A (R2) guidelines for new drugs and WHO
recommendations for generic medicines.