4 unit.computer aided notes m.pharmcomputer aided biopharamacutical charecterization

618 views 16 slides Oct 09, 2024
Slide 1
Slide 1 of 16
Slide 1
1
Slide 2
2
Slide 3
3
Slide 4
4
Slide 5
5
Slide 6
6
Slide 7
7
Slide 8
8
Slide 9
9
Slide 10
10
Slide 11
11
Slide 12
12
Slide 13
13
Slide 14
14
Slide 15
15
Slide 16
16

About This Presentation

computer aided drug delivary system notes , m.pharm, 2 sem ,short notes , computer aided biopharmaceutical charecterization,theritical backround,model construction,parameter sencitive analysis,parameter sensitive analysis usefull for ,sensitive analysis, virtual trail, what is one size fit all, beni...


Slide Content

A. Computer aided biopharmaceutical
charecterization:
 Several drugs during their development stages fail due to poor
biopharmaceutical properties. Thus to decrease the cost and time involved
in the drug discovery process and to develop more effective dosage
regimens, computer-aided in silico absorption models are required for
better characterization of biopharmaceutical properties. One of the major
objectives of in silico absorption models is to envisage the drug’s
physicochemical properties virtually.
A growing concern for biopharmaceutical characterization of drugs/pharmaceutical
products increased the interest in development and evaluation of in silico
tools capable of identifying critical factors (i.e. drug physicochemical
properties, dosage form factors) infl uencing drug in vivo performance,
and predicting drug absorption based on the selected data set(s) of input
factors.
Varoious models were developed from pH parition
compartmental model
Due to dynamic interpretation of the processes a drug undergoes in the
GI tract, dynamic models are able to predict both the fraction of dose
absorbed and the rate of drug absorption, and can be related to PK
models to evaluate plasma concentration- time profi les,this is used for drug
development in different stages.by providing more mechanistic interpretation of PK
data, these models can be utilized to explore mechanistic hypotheses and to help defi
ne a formulation strategy.
The decisive advantage of in silico simulation tools is that they require less
investment in resources and time in comparison to in vivo studies. Also, they offer a
potential to screen virtual compounds.
As a consequence, the number of experiments, and concomitant costs and time
required for compound selection and development, is considerably reduced.
 In addition, in silico methods can be applied to predict oral drug absorption when
conventional PK analysis is limited, such as when intravenous data are lacking
due to poor drug solubility and/or if the drug shows nonlinear kinetics.
In recent years, substantial effort has been allocated to develop and promote
dynamic models that represent GI tract physiology in view of drug transit,
dissolution, and absorption. Among these are the Advanced Dissolution,
Absorption and Metabolism (ADAM) model, the Grass model, the GI-Transit-
Absorption (GITA) model, the CAT model, and the Advanced CAT (ACAT)
model. Some of them have been integrated in commercial software packages,
such as GastroPlus™, SimCYP, IDEA
TM(not available),
PK-Sim
®
, IDEA™ (no longer available), Cloe ® PK, Cloe ® HIA, and INTELLIPHARM ®
PKCR
Theoretical background:
The underlying model in GastroPlus™ is the ACAT model an improved version of the
original CAT model described by Yu and Amidon (1999).

 This semi-physiological absorption model is based on the concept of the
Biopharmaceutics Classifi cation System (BCS) and prior knowledge of GI physiology,
and is modeled by a system of coupled linear and nonlinear rate equations used to
simulate the effect of physiological conditions on drug absorption as it transits through
successive GI compartments.
The ACAT model of the human GI tract consists of nine compartments linked in series, each of them
representing a different segment of the GI tract.
In general, the rate of change of dissolved drug concentration in each GI compartment depends on ten
processes:
MODEL CONSTRUCTION:
 Data collection
Parameters optimization
Model validation
Modeling and simulation start from data collection.
Mechanistic absorption models require a number of input parameters,
which can either be experimentally determined or in silico predicted. The
common approach is to use literature reported values as initial inputs.
The inputs parameters include:
Physicochemical parameters(solubility, permeability, logp, pka, diffusion coefficient)
Pharmacokinetic parameters(clearance, volume of distribution, percentage of drug
extracted in the oral cavity)
Formulation parameters(density, particle size,dosage firm)
GI track information
BCS classifiaction
Compartmental equations
Gastro plus calculates regional solubility based on the fraction of drug ionized at each
compartmental pH according to he henderson-hasselbalch equation.

Parameter sensitive
analysis:
Parameter sensitivity analysis is an essential method for examining
mathematical models of a real-life problem. A detailed parameter sensitivity
analysis gives a broad set of predictions that show how changes in a model
parameter affect relevant model outputs.
understanding how the formulation parameters and/or drug physicochemical
properties affect the predicted PK profiles. This kind of evaluation is performed by
the Parameter Sensitivity Analysis (PSA) feature in GastroPlus™.
there have been two broad categories of sensitivity analysis techniques: local and global. 
Useful for:
Better understanding of the model and its mechanisams.
Does the model behave as expected or not
Identification of influential and non influential model parameters
Steps to conduct sensitivity analysis:
Identification of variables
Define the range of vareable
Calculate the impact
Interpret the reuslts

EMV:expected
monetary model
The various techniques widely applied include;
1.Differential sensitivity analysis
2.One at a time sensitivity measures
3.Factorial analysis method
4.Correlation analysis method
5.Regression analysis method
6.Subjective sensitivity analysis
VIRTUAL TRAIL:
New method of collecting the data safely and
efficaciously from the trail participants,
beginning from statup to execution and follow
up.
VCT take full advantage of technologies (apps,
monitoring devices etc) and online social
engagement platforms to conduct each stage
with comforst of patient.
VCT are not a “ONE SIZE FITS ALL” model
and only a fraction of clinical trails are fully
virtual.


Process overview
Benefits :
Less frequent travels,automated data collection
Cost effective
Single facility
Patient with mobility issue can also participate
Eliminate of source document verification
Maximum enrolment
Decision to termenate further devlopment can be taken faster.
Mim amount of credible, replicable , and evaluable data needed
Improve cost ,convenience and confedentiality
Special conditins like logitudinally,and generate new and relevent questions.
Single study coordination centres under direction of PI” s. PI reviews and
monitors the colleccted data.
Challenges:
Developing trust
Great understanding of digital ecosystem
Patient concerns over sensitive data on internet
Sponsorship/finances
Diseases requiring in house monitoring.
Operational challenges
Technical barriers
Culture barriers(such as concerns over data integrity and fear of technology
failing)


Remote patient monitoring:
ALERE HEALTH PAL: an electronic health record , like microsoft” health vault
MEMS: medication event monitoring system.
Electronic patient reported outcome
(E- PRO”s)
E dairs that rae designed for patient to record and report well specified and labelled
information electronically.
FED VS FASTED STATE:
The presence of food may affect the drug absorption via a variety of
mechanisms: by impacting GI tract physiology( eg. Food induced
changes in gastric emptying time, gastric pH, instestinal fluid
composition, hepatic blood flow),drug solubility and dissolution and drug
permeation.
Eg: gastro plus interpretation of changes in human physiology between
fasted and fed states:
Stomach pH :
Fasted: 1.3
Fed: 4.9

One of the frequent used approaches to asses the effect of food on oral drug absorbtion involves
animal studies.
Howerever, due to the fact that physiological fastors are species dependent , the magnitude of food
effect for given compound across species is usuallu differt that complicating the prediction of food
effects in humans
Considering these models are built based on a prior knoledge of GI physiology in the fasted and fed
stattes,they are able to describe the kinetics of drug such as
Permeabiity
Biorelevent solubility
Ionization constant
Dose
Metabolism and distribution data,etc.
GASTROPULSE uses default physiology parameters,which differ between fasted and fed state.
The food effect for each drug was estimated by comparing AUC or Cmax between fasted ,fed
and or high fat conditions.
Prediction and observed plasma concentration time profile and food effects were compered for
a range of doses to asses the accracy of simulation
The gastroplus was able to corectly predict the observed plasma exposure in fasted ,, fed, and
high fat conditions .
Also applied method was able to accurately distingusih between minor ans significance food
effects.

INVITRO DISSOLUTION:
Test used to qualitatively assure the biological availability of a drug from its
formulation.
Dissolution and drug release tests are invitro tests that Measure the rate and
extent of dissolution or release of the drug substance from a drug product ,
usually in an aqueous medium under specified conditions.
Invitro drug dissolution studies are most often used for monitoring drug product
stability and manufacturing process.
https://www.slideshare.net/slideshow/invitro-dissolution/137541136
FACTORS TO BE CONCEDERED:
Factor relating to the disoolution apparatus
Factors relating to the dissolution fluid
Process parameters.

Classification:
Closed compartment
Opean compartent(continuous flow through apparatuas)
Dialysis system.
INVITRO IN VIVO CORRELATION:
https://www.slideshare.net/slideshow/in-vitro-in-vivo-correlation-122984972/122984972
The main objective of an IVIVC is to serve as a surrgate for in vivo bioavailability and to support
biowaivers
IVIVC S could also be employed to establish dissolution specifications and to support and or validate
the use of dissolution methods.
Used :
Providing necessary process control
Determing stability of dosage form
Parameters:
Drug con in plasma at each sampling time
Apparent rate constant for eliminating
Biological half life
Urinary excretion rate and amount excereted in urine at infinity
Pharmacokinetic parameters:
1.Mean residence time(mrt)
2.Mean absorption time(MAT)
3.Cmax/AUC
4.The peak occupancy time(po)
5.Multiple dosing
6.Coefficient of variation.
There are 5 correlationlevels:
Level A
Level B
Level C
Level D

A Biowaiver means that in vivo bioavailability and/or
bioequivalence studies may be waived (not considered necessary for
product approval).
The term is applied to a regulatory drug approal process when the dossier
is approved based on evidence of equvalance other than through in vivo
equivalance testing.
Health and human services,US FDA Instigated the BCS with aim of
granting so called biowaivers for SUPACS.
At that time the biowever was only conseder for SUPAC to
pharmaceutical products.
More recently the application of the biowaiver concept has been extended
to approval of certain orally administerd generic products
B. Computer simulation in
pharmacokinetics and pharmacodynamics:
Computer simulations play a crucial role in pharmacokinetics (PK) and
pharmacodynamics (PD) research, offering a powerful tool for modeling
and predicting drug behavior in the body. Here's how simulations are
applied in PK and PD:
Pharmacokinetics (PK):

1. Modeling drug absorption: Simulations help predict how drugs are
absorbed, distributed, metabolized, and eliminated (ADME) in the body.
2. Predicting drug concentrations: Computer models estimate drug
concentrations in plasma, tissues, and organs, allowing researchers to
optimize dosing regimens.
3. Simulating special populations: Simulations help predict drug
behavior in specific populations, such as pediatrics, geriatrics, or patients
with impaired organ function.
4. Drug-drug interactions: Computer simulations assess the potential for
drug-drug interactions and their impact on PK profiles.
Pharmacodynamics (PD):
1. Modeling drug efficacy: Simulations predict the relationship between
drug concentrations and efficacy, enabling researchers to optimize dosing
regimens.
2. Predicting adverse effects: Computer models estimate the likelihood
of adverse effects based on drug concentrations and individual patient
characteristics.
3. Biomarker analysis: Simulations help identify and analyze
biomarkers for drug efficacy and toxicity.
4. Personalized medicine: Computer simulations facilitate personalized
treatment planning by accounting for individual patient characteristics,
such as genetics, physiology, and disease state.
By integrating computer simulations in PK and PD research, scientists
can:
- Reduce the need for extensive animal testing and early-stage clinical
trials
- Accelerate drug development timelines
- Improve drug safety and efficacy
- Enhance personalized medicine approaches
- Optimize dosing regimens and treatment strategies
Some common simulation software used in PK/PD research includes:
- NONMEM (Nonlinear Mixed Effects Modeling)
- Phoenix WinNonlin
- MATLAB
- Simul8
- Berkeley Madonna

These simulations have become essential tools in the pharmaceutical
industry, regulatory agencies, and academic research, driving
advancements in drug development and improving patient outcomes.
WHOLE ORGANISM:
Computer simulations on whole organisms, also known as "whole-organism
modeling"
Whole-organism computer simulations in pharmacokinetics (PK) and
pharmacodynamics (PD) involve creating detailed computational models of
entire organisms to predict how drugs interact with the body. These
simulations integrate data from various sources, such as:
1. Genomics: Genetic information to predict enzyme expression and activity.
2. Proteomics: Protein expression and interaction data to model drug binding
and transport.
3. Metabolomics: Metabolic pathway data to predict drug metabolism and
elimination.

4. Physiomics: Physiological data, such as organ size, blood flow, and tissue
composition.
1.Predictive PK/PD modeling: Simulations predict drug concentrations,
bioavailability, and efficacy in specific populations or individuals.
2. Personalized medicine: Models account for individual genetic,
physiological, and disease characteristics to optimize drug therapy.
3. Drug development: Simulations streamline the development process by
predicting drug behavior, identifying potential issues, and optimizing dosing
regimens.
4. Toxicity prediction: Models forecast potential adverse effects and drug-
drug interactions.
1. PK-Sim: A software platform for PK/PD modeling and simulation.
2. MoBi: A modeling framework for simulating biological processes.
3. OpenPK: An open-source platform for PK/PD modeling.
4. Physiomics: A software tool for modeling physiological systems.
5. WholeCell: A computational platform for simulating entire cells.
6. E-Cell: A software environment for modeling and simulating cellular
processes.
IN whole organism simulation ,whole body systems are usually represented in
one of two ways:
Lumped parameter pk-pd model(population pk-pd model)
Physiological modeling
ORGAN /TISSUE SIMULATIONS:
Isolated tissue computer simulation in pharmacokinetics and dynamics is a
cutting-edge approach that uses computational models to simulate the
behavior of drugs in isolated tissues or organs. This enables researchers to
study the absorption, distribution, metabolism, and excretion (ADME) of drugs
in a controlled and virtual environment.
Applications:
1. Predicting drug behavior: Simulations can predict how drugs will behave
in specific tissues or organs, allowing for more accurate predictions of drug
efficacy and toxicity.
2. Personalized medicine: Simulations can be used to model individual
patient characteristics, enabling personalized drug treatment plans.

3. Drug development: Simulations can streamline the drug development
process by identifying potential issues earlier, reducing the need for animal
testing and clinical trials.
4. Dose optimization: Simulations can help optimize drug dosing regimens
for maximum efficacy and minimal toxicity.
5. Mechanism-based modeling: Simulations can help researchers
understand the underlying mechanisms of drug action and interactions.
Types of simulations:
1. Physiologically based pharmacokinetic (PBPK) modeling: Simulates
the behavior of drugs in the body, taking into account physiological processes.
2. Compartmental modeling: Simulates the behavior of drugs in specific
tissues or organs, using mathematical compartments.
3. Agent-based modeling: Simulates the behavior of individual molecules or
cells, allowing for a more detailed understanding of drug interactions.
BTEX- blood tissue exchanges model,because the increased level detail
and temporal resolution certeinly makes the good mixing and
uniformity hypothesis at the basis of lumped parameter models
less tenable.

Computer simulation of proteins and genes in pharmacokinetics and dynamics is a cutting-edge
approach that uses computational models to study the behavior of proteins and genes in relation to
drug absorption, distribution, metabolism, and excretion (ADME). This enables researchers to:
1. Predict protein-ligand interactions: Simulate how drugs bind to proteins, affecting their
pharmacokinetics and dynamics.
2. Model gene expression: Study how genes regulate protein production, influencing drug response.
3. Analyze protein-protein interactions: Investigate how proteins interact with each other, impacting
drug efficacy and toxicity.
4. Simulate metabolic pathways: Model how drugs are metabolized by proteins, affecting their
pharmacokinetics.
5. Investigate pharmacogenomics: Study how genetic variations affect drug response and
pharmacokinetics.
Types of simulations:
1. Molecular dynamics (MD) simulations: Study protein-ligand interactions and protein dynamics.
2. Quantum mechanics/molecular mechanics (QM/MM) simulations: Model protein-ligand
interactions at the atomic level.
3. Docking simulations: Predict how drugs bind to proteins.
4. Gene regulatory network modeling: Simulate how genes regulate protein production.
5. Physiologically based pharmacokinetic (PBPK) modeling: Integrate protein and gene simulations
with physiological processes.
C. Computer in clinical development
clinical data collection:
Clinical Data collection and management:
It has
Before data collection
Data collaction
Data management
After data collection
By using electronic based system for collection and management.
The advantage provide for quick acces of upto data for feed back to
appropriate stakeholders which allows them to make timely critical
decisions and enables then to easily monitor protocols compliance,
enrollment rates and performance metrics of participating sites. It
involves:
Centeralized systems
Distribution systems
Wireless systems

PDF basedsystems
Web based systems
Direct systems
E Clinical softwres are used(oracle clinical v4i, DatalabsXC,
CTrialmaster, clinplus data management
Process during data collection:
Data quality assurance
Treatment dispensing
Handling unexpected events
Data transfermation
Process after data collection:
Data lockout
Data retention
Data archiving
Data sharing
Data management :
The data management process includes a wide range of tasks and
procedures, such as: Collecting, processing, and validating data.
designing case report forms,
collecting data,
tracking forms,
entering data,
validating data for errors,
resolving discrepancies,
medical coding, and
locking the database
Regulation of computer systems:
1. Good Laboratory Practice (GLP): Ensures data quality and integrity in
laboratory research.
2. Good Manufacturing Practice (GMP): Regulates the production and
quality control of pharmaceuticals.

3. Title 21 CFR Part 11: Sets standards for electronic records and signatures
in pharmaceutical research.
4. European Medicines Agency (EMA) guidelines: Provides guidance on the
use of computer simulations in drug development.
5. US Food and Drug Administration (FDA) guidelines: Regulates the use of
computer simulations in drug development and approval.
6. International Conference on Harmonisation (ICH) guidelines: Provides
harmonized guidelines for drug development, including computer simulations.
7. ISO 27001: Sets standards for information security management in
computer systems.
8. IEEE standards: Provides guidelines for computer simulations, modeling,
and analysis.
9. American Society for Clinical Pharmacology and Therapeutics (ASCPT)
guidelines: Provides guidance on the use of computer simulations in clinical
pharmacology.
10. Pharmacokinetic-Pharmacodynamic (PK-PD) modeling guidelines:
Regulates the use of computer simulations in PK-PD modeling.