Screening and PKPD modeling Flashcards

1
Q

How experimental PK/PD in vitro data can be translated into clinical decision-making.

A

Understanding PK/PD Relationships: PK deals with the absorption, distribution, metabolism, and excretion (ADME) of a drug in the body, while PD deals with the drug’s effect on the body (e.g., efficacy, toxicity). In vitro studies provide data on drug concentrations and their effects in controlled laboratory settings.

Modeling and Simulation: PK/PD modeling involves mathematical modeling to describe the relationship between drug concentration and its effect. In vitro data can be used to develop these models, which can then be extrapolated to predict PK/PD behavior in vivo.

Translational Research: Bridging the gap between in vitro and in vivo data involves translational research. This includes understanding the physiological differences between laboratory models and humans, such as metabolism, organ function, and drug interactions.

Clinical Trials Design: PK/PD data can inform the design of clinical trials, including dosing regimens, patient selection, and endpoints. For example, PK data can inform dose selection to achieve therapeutic concentrations, while PD data can help determine the optimal endpoint for assessing drug efficacy or toxicity.

Dose Optimization: Using PK/PD data, clinicians can optimize drug dosing regimens based on individual patient characteristics such as age, weight, renal function, and co-morbidities. This personalized approach can maximize therapeutic efficacy while minimizing the risk of adverse effects.

Therapeutic Drug Monitoring (TDM): TDM involves measuring drug concentrations in patients’ blood or tissues to optimize dosing regimens. PK/PD data can guide TDM protocols by defining target drug concentrations associated with optimal therapeutic outcomes.

Risk Assessment: PK/PD data can help assess the risk of adverse drug reactions or drug interactions, guiding clinicians in monitoring and managing patient safety.

Regulatory Decision-Making: Regulatory agencies may use PK/PD data to evaluate the safety and efficacy of new drugs, determine appropriate dosing recommendations, and approve labeling information for clinical use.

Clinical Guidelines: Professional medical societies may incorporate PK/PD data into clinical practice guidelines to assist clinicians in making evidence-based treatment decisions.

Continual Evaluation: As new clinical data becomes available, PK/PD models and clinical decision-making algorithms should be continually evaluated and refined to ensure optimal patient outcomes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

How selected physiological parameters can impact PK/PD.

A

Renal Function: Renal function affects drug clearance, particularly for drugs that are predominantly eliminated through the kidneys. Impaired renal function can lead to decreased drug clearance, resulting in higher drug concentrations and increased risk of toxicity.

Hepatic Function: Hepatic function influences drug metabolism, primarily through the cytochrome P450 enzyme system. Impaired liver function can decrease drug metabolism, prolonging drug half-life and potentially increasing drug toxicity. Conversely, induction of hepatic enzymes can accelerate drug metabolism, reducing drug efficacy.

Body Weight and Composition: Body weight and composition can impact drug distribution and clearance. For lipophilic drugs, which tend to distribute into fatty tissues, higher body fat content can lead to prolonged drug action. Conversely, for hydrophilic drugs, which distribute primarily into lean body mass, variations in body composition can affect drug distribution volume and clearance rates.

Age: Age-related changes in physiology can affect both PK and PD parameters. For example, decreased hepatic and renal function in elderly patients can alter drug metabolism and elimination, leading to increased drug exposure and risk of toxicity. Additionally, age-related changes in receptor sensitivity or organ function may affect drug response and efficacy.

Genetic Variability: Genetic polymorphisms in drug-metabolizing enzymes, transporters, and drug targets can influence individual variability in drug response. Pharmacogenetic factors can affect drug metabolism, efficacy, and toxicity, leading to inter-individual differences in PK/PD profiles.

Drug-Drug Interactions: Concurrent use of multiple drugs can alter PK/PD parameters through drug-drug interactions. Interactions may involve inhibition or induction of drug-metabolizing enzymes or transporters, leading to changes in drug absorption, distribution, metabolism, or elimination.

Disease States: Underlying medical conditions can impact PK/PD parameters through alterations in organ function, blood flow, protein binding, or receptor sensitivity. Disease-related changes in physiology may affect drug absorption, distribution, metabolism, or elimination, leading to variability in drug response and efficacy.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Know which factors are important for the planning of high-throughput drug screening.

A

Choice of model system,

choice of Data collection methods,

What to measure

Assay Development:

Design and optimize robust assays that accurately reflect the biological processes of interest.
Ensure the assays are scalable and amenable to automation.
Target Selection:

Clearly define the molecular target or pathway being investigated.
Prioritize targets based on relevance to the disease of interest and potential for therapeutic intervention.
Compound Libraries:

Curate diverse and representative compound libraries for screening.
Consider including known bioactives, FDA-approved drugs, and structurally diverse compounds.
Automation and Robotics:

Implement automated systems to handle liquid handling, plate reading, and other repetitive tasks.
Utilize robotics to increase throughput and reduce human error.
Data Management:

Establish a robust system for data acquisition, storage, and analysis.
Implement informatics tools to handle large datasets efficiently.
Quality Control and Validation:

Develop quality control measures to ensure the reliability of screening results.
Validate screening assays to ensure reproducibility and consistency.
Dose-Response Analysis:

Perform dose-response analysis for hit confirmation and determination of compound potency.
Assess the concentration-response relationship for identified compounds.
Hit Triage and Confirmation:

Develop criteria for hit selection based on assay performance and compound activity.
Confirm hits using orthogonal assays or additional experiments to eliminate false positives.
Chemoinformatics and Computational Approaches:

Utilize computational methods for virtual screening and prioritization of compounds.
Employ chemoinformatics tools to analyze chemical structures and predict bioactivity.
Ethical and Regulatory Considerations:

Adhere to ethical standards and regulatory guidelines in the handling and testing of compounds.
Consider safety, toxicity, and other relevant factors when selecting compounds for screening.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Know which factors are important for the implementation of high-throughput drug screening.

A

Assay Robustness and Reproducibility:

Develop assays that are robust, reproducible, and amenable to automation.
Optimize assay conditions to minimize variability and ensure consistent results across multiple screenings.
Automation and Robotics:

Utilize automated liquid handling systems, robotics, and other high-throughput technologies to increase screening efficiency.
Implement robotics for tasks such as compound dispensing, plate handling, and data acquisition to reduce human error and increase throughput.
Assay Miniaturization:

Miniaturize assays to reduce reagent and compound consumption, enabling cost-effective high-throughput screening.
Consider using microplates with smaller well volumes to achieve higher throughput without compromising assay quality.
Compound Libraries:

Curate diverse and representative compound libraries for screening.
Include known bioactives, FDA-approved drugs, and structurally diverse compounds in the library for a comprehensive screening approach.
Quality Control Standards:

Implement rigorous quality control standards to ensure the reliability of screening results.
Include positive and negative controls to monitor assay performance and detect potential issues.
Data Management and Informatics:

Establish a robust data management system to handle large datasets generated during screening.
Implement informatics tools for data analysis, visualization, and interpretation to extract meaningful insights.
High-Throughput Screening Platforms:

Choose appropriate screening platforms, such as fluorescence, luminescence, or absorbance-based methods, based on the assay requirements.
Consider multiplexing and parallelization to increase throughput.
Hit Triage and Confirmation:

Develop criteria for hit selection based on assay performance and compound activity.
Confirm hits using orthogonal assays or additional experiments to eliminate false positives and ensure the validity of screening results.
Hit-to-Lead Optimization:

Establish a process for hit-to-lead optimization to prioritize and refine potential drug candidates.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Know which factors are important for the interpretation of high-throughput drug screening.

A

Assay Validation and Quality Control:

Ensure that the screening assay is well-validated and meets predefined quality control standards.
Perform regular quality control checks to monitor assay performance and identify any issues that may affect data interpretation.
Reproducibility and Robustness:

Assess the reproducibility and robustness of the screening assay. Consistent results across replicate experiments increase confidence in the identified hits.
Address any sources of variability that may impact the reliability of the screening data.
Normalization and Data Transformation:

Normalize raw data to correct for experimental variability, plate effects, and other sources of bias.
Consider data transformation techniques to ensure that the data meet the assumptions of the statistical methods used for analysis.
Hit Selection Criteria:

Define clear and stringent criteria for hit selection based on assay performance and compound activity.
Consider using statistical methods to identify hits that significantly deviate from the baseline.
Dose-Response Analysis:

Perform dose-response analysis for hit confirmation and determination of compound potency.
Evaluate concentration-response relationships to assess the effectiveness and concentration-dependent effects of identified hits.
False Positive and False Negative Analysis:

Investigate potential sources of false positives and false negatives in the screening data.
Use orthogonal assays or validation experiments to confirm hits and eliminate false positives.
Chemical Structure and Mechanism of Action:

Analyze the chemical structures of hits to identify potential scaffolds and chemical series.
Consider the mechanism of action of identified compounds and their relevance to the target or pathway of interest.
Data Integration:

Integrate HTS data with other relevant datasets, such as genomics, transcriptomics, or proteomics, to gain a more comprehensive understanding of compound effects.
Utilize systems biology approaches for a holistic interpretation of drug responses.
Biological Relevance:

Evaluate the biological relevance of identified hits in the context of the disease or biological process being studied.
Consider whether the observed effects are physiologically relevant and biologically meaningful.
Hit-to-Lead Optimization:

Prioritize hits based on their potential for further development.
Consider additional optimization steps, such as medicinal chemistry efforts, to improve compound properties and increase their potential as drug candidates.
Identification of Off-Target Effects:

Assess the potential for off-target effects of identified hits.
Utilize computational tools and databases to predict off-target interactions and evaluate their impact on the interpretation of screening results.
Data Visualization and Reporting:

Use effective data visualization tools to present screening results clearly.
Prepare comprehensive reports that include key findings, statistical analyses, and interpretations for easy communication with stakeholders.
Validation in Relevant Models:

Validate the identified hits in relevant in vivo models or primary patient samples to confirm their efficacy and safety.
Consider the translational potential of hits for further preclinical and clinical development.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Funnel strategy in drug screening

A

Primary assay
= assay used for the screening campaign,
Need to be miniaturized, have excellent throughput
and low cost per data point

Counter assay
= to filter out false positive (technology hitters) or
unselective compounds
Sometimes just like the primary assay but with one key
modification.

dose deteremination

Secondary/orthogonal assays
= assays that look at the same thing than the primary assay
but with a different technology or assay that look at other thing
for instance biological effect
Can be more focused with lower throughput

Hit selection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

the clinical trial timeline

A

Phase II & Phase III
Dose, Efficacy, Toxicity

Phase I
PK, Dose escalation, Toxicity

Pre-clinical test
& Lead optimization
SAR, Drug-like properties, Solubility
Permeability, ADME, Plasma PK
Efficacy, Toxicity

Compound screening
Visual screening, HTS

Target validation

Disease models, Target identification, Target validation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

why is there a need for The need for HTS?

A
  • Financial pressures due to the increasing cost of bringing a drug to market
  • The ‘post-genome era’ à increase in the number of targets of therapeutic interest that
    have unknown small-molecule modulators.
  • Stringent safety requirements by regulatory authorities à substantial stress on the
    research and development functions in pharmaceutical and biotechnology companies
  • Generating new molecular entities against 3 to 4 new targets per year
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

why is HTS important for drug discovery?

A
  • High-throughput screening for novel drug discovery to substitute the traditional “trial
    and error” approach
  • To identify therapeutic targets and validate biological effects
  • HTS involves assaying and screening a large number of biological effectors and
    modulators against designated and exclusive targets
  • HTS is generally favored when little is known of the target,
  • HTS can be paired with other strategies such as computational techniques and fragmentbased
    drug design
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

HTS comprises several steps:

A
  • Target recognition,
  • Compound management
  • Reagent preparation
  • Assay development
  • The screening itself.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

HTS platforms ( types of assay) , how we chose them ?

A

–HTS involves in vitro (cell, Biochemical ) or wholeorganism- based assays
* The most common readouts for
biochemical assays in HTS are optical,
including absorbance, fluorescence,
luminescence, and scintillation.
* The efficiency of data production and
cost per screen are the main
determinants in the choice of the most
suitable readout for a particular screen.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

what is the main reason drugs fail in clinical trials or are withdrawn ?

A

toxicity issues

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Animal models are poor predictors of drug safety in humans ( example )

A
  • Fialuridine anti HBV
  • 5 volunteers died during phase II clinical trials
  • Safe in mice, rats, dogs, monkeys, and woodchucks (doses&raquo_space;»)
  • 2 volunteers survived after receiving liver transplants
    Transporter protein expressed in mitochondria in human hepatocytes à mitochondrial toxicity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

3D cell culture models in drug discovery

A
  • Promising model for studying disease mechanisms
  • Application in drug discovery
  • Potentially recapitulate aspects of in vivo physiology more accurately than 2D systems
  • 3D vs 2D cultures differ in many aspects (proliferation, differentiation, migration,
    oxygen and nutrient gradients, cell-cell and cell-extracellular matrix interactions)
  • àimpact cellular phenotypes and drug responses à high failure rates in early stages of
    phenotypic drug screens if performed only in 2D cultures

Primary Human Hepatocyte (PHH) model preserves hepatic molecular
signature

  • The PHH model maintains
    physiological phenotype at
    transcriptomic and proteomic level
    compared to 2D culture
  • Cells in 2D lost the in vivo
    signature after 24hrs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

A good cellular model for HTS should:

A
  • Recapitulate the physiological and functional characteristics of the organ to be
    studied
  • Show the disease hallmarks
  • Scalable system compatible with HTS
  • In vivo models have poor outcome in drug screening and are not compatible
    with HTS
  • 3D cellular models are a suitable alternative for HTS compared to 2D
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What are the challenges and considerations when using patient-derived material
in drug screening?

A

Heterogeneity of Patient Samples:

Patient samples can exhibit significant biological variability due to differences in genetic backgrounds, disease subtypes, and individual responses to treatment.
Addressing this heterogeneity is essential for obtaining meaningful and reproducible results.
Sample Availability and Collection:

Obtaining an adequate supply of patient-derived material can be challenging, especially for rare diseases or situations where obtaining samples involves invasive procedures.
Proper consent and ethical considerations must be addressed in the collection of patient samples.
Sample Processing and Storage:

Standardized protocols for sample processing and storage are crucial to maintaining sample integrity.
Variability in sample handling can impact assay results, necessitating careful documentation of sample processing steps.
Informed Consent and Ethical Considerations:

Ensure proper informed consent from patients for the use of their biological material in research.
Comply with ethical guidelines and regulations related to human subject research.
Limited Availability of Matched Healthy Controls:

Obtaining appropriate control samples that match the characteristics of patient samples can be challenging.
The lack of well-matched controls may complicate the interpretation of screening results.
Technical Challenges in Assay Development:

Patient-derived samples may present technical challenges due to variations in cell viability, growth rates, and other biological parameters.
Assay development may require optimization to accommodate the specific characteristics of patient samples.
Genetic and Molecular Heterogeneity:

Diseases often exhibit genetic and molecular heterogeneity, which can complicate drug screening efforts.
Molecular profiling of patient samples may be necessary to stratify them into subgroups for more targeted screening.
Disease Progression and Evolution:

Patient-derived samples may represent a snapshot of the disease at a specific point in time.
Longitudinal studies or the use of multiple samples from the same patient over time may be needed to capture disease progression and evolution.
Reproducibility and Standardization:

Achieving reproducibility across experiments and laboratories can be challenging when using patient-derived material.
Standardizing protocols and ensuring rigorous quality control measures are critical.
Data Interpretation and Translation to the Clinic:

Translating findings from patient-derived material to clinical applications requires careful consideration of the relevance and predictive value of screening results.
Addressing the gap between preclinical findings and clinical efficacy is a complex process

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Understand the different techniques/applications of 3D cell culture in drug
screening

A

Spheroid Culture:

Technique: Cells are allowed to aggregate and form three-dimensional spheroids.
Applications: Mimics tissue architecture and promotes cell-cell interactions. Suitable for assessing drug penetration, migration, invasion, and responses in a more physiological context.
Organoids:

Technique: Culturing cells derived from tissues or organs to form complex, three-dimensional structures.
Applications: Replicates tissue architecture, cellular heterogeneity, and organ-specific functions. Used for drug testing, disease modeling, and personalized medicine.
Bioprinting:

Technique: 3D printing of cell-laden biomaterials to create tissue-like structures.
Applications: Enables the precise placement of cells in a 3D spatial arrangement. Useful for constructing tissue models for drug testing and regenerative medicine.
Hydrogel-Based 3D Culture:

Technique: Cells are encapsulated within hydrogel matrices to create a 3D environment.
Applications: Provides a tunable and customizable platform for studying cell behavior, drug responses, and interactions within a biomimetic microenvironment.
Microfluidic Systems:

Technique: Cells are cultured in microscale channels or chambers, allowing for precise control over the cellular microenvironment.
Applications: Facilitates high-throughput screening and real-time monitoring of cellular responses. Enables the study of cell migration, drug diffusion, and cell-cell interactions.
Hanging Drop Method:

Technique: Cells are suspended as small droplets, promoting self-aggregation.
Applications: Suitable for spheroid formation and high-throughput drug screening. Mimics cell-cell interactions and cell morphology found in vivo.
Rotary Cell Culture Systems:

Technique: Cells are cultured in rotating vessels to simulate microgravity conditions.
Applications: Useful for studying the effects of altered gravity on cell behavior. Can be employed in drug screening for space-related research.
Magnetic Levitation:

Technique: Cells are combined with magnetic nanoparticles and levitated in a magnetic field.
Applications: Mimics the cellular microenvironment and allows for the study of cell-cell interactions, drug responses, and migration in a 3D context.
Coculture Systems:

Technique: Different cell types are cultured together to model complex tissue interactions.
Applications: Enables the study of cell communication, paracrine signaling, and interactions between different cell types. Useful for drug screening in a multicellular context.
Tumor-on-a-Chip Models:

Technique: Integrates microfabrication techniques with 3D cell culture to create microscale models of tumors.
Applications: Mimics the tumor microenvironment, vasculature, and drug responses. Useful for studying cancer biology and drug screening.
Extracellular Matrix (ECM) Incorporation:

Technique: Incorporates natural or synthetic ECM components into 3D cultures.
Applications: Enhances cell-matrix interactions, providing a more physiologically relevant environment for drug screening.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What are the advantages of using 3D cell culture models in drug screening

A

Mimicking In Vivo Microenvironment:

3D cell cultures better replicate the complex cellular microenvironment found in living tissues compared to 2D cultures. This includes cell-cell interactions, gradients of nutrients and oxygen, and extracellular matrix (ECM) composition.
Cell-Cell Interactions:

3D models allow for the formation of cell-cell interactions, which are crucial for simulating tissue architecture and function. This is particularly important in understanding complex biological processes and responses to drugs.
Cell-ECM Interactions:

The inclusion of ECM components in 3D cultures enables cells to interact with their surrounding matrix, influencing cell behavior, migration, and signaling pathways. This more accurately reflects in vivo conditions.
Physiological Relevance:

3D cultures provide a more physiologically relevant environment, leading to improved cell differentiation, phenotype maintenance, and overall cellular function. This is critical for studying drug responses in a context that better represents in vivo conditions.
Cellular Heterogeneity:

3D models allow for the maintenance of cellular heterogeneity, preserving the diversity of cell types and their interactions within tissues. This is particularly relevant in diseases where multiple cell types contribute to pathology.
Drug Penetration and Distribution:

3D cultures better mimic drug penetration and distribution within tissues, providing insights into how drugs interact with cells in a 3D spatial arrangement. This is crucial for understanding drug efficacy and toxicity.
Disease Modeling:

3D cell culture models can be tailored to mimic specific disease conditions, allowing for the study of disease progression, pathophysiology, and drug responses. This is particularly valuable for diseases with complex and multifactorial etiologies.
High-Throughput Screening:

Advances in automation and imaging technologies have made it feasible to perform high-throughput screening using 3D cell cultures. This enables the screening of large compound libraries in a more physiologically relevant context.
Prediction of In Vivo Responses:

3D models provide a better correlation with in vivo responses compared to 2D models, improving the predictive value of preclinical studies. This can lead to more accurate assessments of drug safety and efficacy.
Personalized Medicine:

Patient-derived 3D cultures can be used to create personalized models for drug screening, considering individual genetic and molecular characteristics. This approach holds promise for tailoring treatments based on patient-specific responses.
Reduction of Animal Testing:

The use of 3D cell cultures can contribute to the reduction of animal testing by providing more relevant and predictive preclinical models. This aligns with ethical considerations and regulatory trends.
Understanding Tumor Biology:

3D models, such as spheroids and organoids, are valuable for studying tumor biology, including aspects of tumor growth, invasion, and metastasis. These models are particularly relevant in cancer drug discovery.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

why is there a Translational problem in drug development ?

A
  • Despite outstanding technological achievement and
    accumulated knowledge  high attrition rates and failure in
    drug development.
  • Drug development is a complex problem where we try to predict
    drug behavior in humans based on other systems.
  • Knowledge of human physiology and pharmacology is
    fragmented.
  • Need for more basic and translational research and better
    integration of findings from different scientific disciplines
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Need for in-silico PKPD studies ?

A
  • PKPD studies are the core of the drug discovery and
    development process
  • Optimising PKPD properties for human disease is the ultimate goal of
    the drug discovery/development journey
  • Need to establish a quantitative link between experimental
    PK/PD data and our understanding of the biological
    mechanisms that generated the data
    We need a MODEL (PKPD model)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What is a model? - definition

A
  • Model: a simplified representation of a real-world system or
    process
  • A box of relationships that describes a system using the system
    parameters.
  • How do you think your data were generated?
  • Some models
  • Newton’s second law of motion: F = ma
  • Special relativity: E = mc2
  • Fick law :
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Modeling and simulation

A

Modeling

Confirms
knowledge

SEARCH for optimal parameter
values that explains the data
F = 0.5, V = 20L, CL = 2 L/h

Explore
possibilites
Parameters are FIXED to
F = 0.5, V = 20L, CL = 2 L/h

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

do absorption distribution and elimination happen simultaneusly ?

A

yes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Pharmacokinetic parameters - Human

A
  1. Clearance
  2. Volume of distribution
  3. Half-life
  4. Bioavailability
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Clearance (CL)

A
  • It is the rate at which a drug is eliminated from the body = the
    volume of blood cleared of the drug per unit of time.
  • It is the irreversible removal of a drug from the measured matrix
    (blood or plasma)  permanent change.
  • CL unit: volume/time
  • CL is typically constant

CL = Rate of elimination constant(ke) * Vd Vd = Dose/C0
crearance is different for oral versus IV administration

Clearance (CL) – Organ CL
* CL can be measured for each organ
CLsystemic = CLrenal + CLhepatic + CLother

and for one organ its Q*E with :
E=(Cin-Cout)/Cin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

what is Volume of distribution (V)?

A

It is a theoretical concept: The volume in which the amount of
drug in the body would be needed to uniformly distribute and
produce the observed concentration.
* Relates the drug concentration in plasma to the total amount of
drug in the body

or

Reversible partitioning of drug molecules into various tissues from plasma

V =Total amount of drug (A) (mass)/Plasma concentration (C) (mass/volume)

  • At time = 0
  • Drug amount in the body = dose
  • Concentration = C0

Volume of distribution (V)
* Small values of V  Drug distributes in the central compartment
* Large values of V  Central compartment + other
* The volume of distribution can be larger than the plasma volume
(approx. 3L) or the whole body volume.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Half-life

A
  • It is the time for a drug to be reduced by 50%
    ln2/ke or 0,693 *V/CL
  • Determine when reaching the steady state (Css)
  • The Css is reached after 4-5 half-lives
  • Determine the time required to eliminate all or a
    portion of the drug if the treatment is discontinued
  • Determine dosing frequency
  • Longer half-life  longer dosing interval
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Bioavailability (F)

A
  • Percentage or fraction of administrated dose
    that reaches the systemic circulation: F
  • The IV bolus is the reference
    F= AUC oral / AUC IV
  • Amount of absorbed drug = F * Dose
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Prediction of oral absorption

A

Bioavailability (F)
(The extent of absorption)

The absorption rate constant (ka)
(The speed of absorption)

Lag time (only humans)
(The delay in absorption)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

Determination of Fa

A
  • Earlier methods = Ex-vivo
    1. Animal intestine
    2. Endoscopic balloons
     Gives remaining/absorbed drug
    concentration in the intestine.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

Prediction of Fa - Caco2 permeability

A
  • Fa is determined using the Caco2 cell line
  • Derived from a colon carcinoma
  • Ability to differentiate into a monolayer of
    cells
  • Has the enterocytes absorption properties
    with a brush border layer typically found in
    small intestines.
  • Papp: apparent permeability (Caco2 assay)
  • Peff: effective permeability (Human)
  • Tres: transit time in the human small intestine (3h)
  • R: radius of the human small intestine
  • Papp and Peff are closely correlated

Prediction of Fa - Challenges
* Caco2 express P-gp and MRP2
* Passive drug permeability estimation is
affected when the drug is a substrate of P-gp
MDCK (Madin-Darby Canine Kidney) cells
* High Papp Inter-laboratory variability
especially when Peff < 1x10-4 cm/s

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

Prediction of Fg

A
  • No consensus on how to estimate Fg
  • Yang et al model
    Extrapolate from hepatic CL

Prediction of Fg
* Can be estimated from in-vitro assays
* Simplified form: Total fraction of the drug
* Can take into account the unbound (free) fraction of the drug

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What is Fa , Fg , Fh

A

Membrane
penetration (Fa)

Metabolism in the
enterocyte (Fg)

Metabolism
during first pass
through liver (Fh)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

Fh ?

A

Fh = 1 - Eh

(CL= Qh* Eh)

Eh = (CLin - CLout) / CLin

Q: Blood flow
E: Extraction coefficient

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

Bioavailability

A

F = 0.8 * 0.9 *0.7 = 0.5 = 50%
Effective dose = Administrated Dose * F = 100 mg * 0.5 = 50mg

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

Prediction of ka

A
  • In-vitro assays
  • Papp : drug permeability across intestinal mucosa
    (Caco-2)
  • S: absorptive surface area (200 m2)
  • Vc: central volume of distribution
  • ka can be estimated using:
  • Assays (Caco-2)
  • Allometric scaling from other species
  • Averaging of ka from different species
  • In-silico methods
  • It is also possible to predict ka from the physicochemical
    property of drugs, pH, CYP activity, transporter, and blood flow
     (e.g., GastroPlus)

ka=Papp *S/ Vc

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

CL prediction

A

Methods
Hepatic CL:
* In vitro estimation
* Metabolite formation rate
* Substrate depletion
* Allometry can also be used
Renal CL:
* Allometry  recommended for renal CL : glomerular filtration is well correlated
with the body size (Body weight of Body surface area: W/L^2) simple to scale up
to human.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

Prediction of the CLh - The unbound fraction (CLint,u)

A
  • Highly bound drugs
  •  low unbound fraction (fu)
  •  low exposure free drug
  •  low CLint,app
  • CLint,app needs to be corrected for the unbound
    fraction

CLint,u=CLint,app /fu

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

Prediction of the CLh- Scaling to the organ level ( sos)

A
  • CLint unit is volume/min/mg protein or million hepatocytes
  • Need to scale the assay to the organ
  • There is 45 mg of microsomal protein/gram of liver
  • There is 120 million hepatocyte/gram of liver
  • Human Liver weight = 1500 g

Clint,u_scaled=CLint,u=CLint,app /fu*MPPGL * Liver_Weigth

40
Q

Prediction of the CLh - Summary ?

A

In vitro incubation

In vitro apparent intrinsic CL

In vitro unbound apparent intrinsic CL

Organ intrinsic CL (Predicted in vivo CL)

Hepatic CL with blood flow

41
Q

In vitro apparent intrinsic CL ?

A

In vitro apparent intrinsic clearance (CL) refers to the rate at which a drug is metabolized or eliminated by hepatic microsomes or other in vitro systems normalized to the amount of enzyme or protein present. It is an important parameter in drug metabolism studies and is often used to predict in vivo clearance. The intrinsic clearance reflects the intrinsic metabolic capacity of the enzyme system in the absence of any limitation due to blood flow or other physiological factors.

CLapp,int= Vmax/Km

42
Q

Prediction of the CLh – Could it be more complex?

A
  • The model can also consider:
  • Blood
  • Unbound fraction
  • Hepatocyte:
  • Uptake, efflux processes
  • Bile excretion
  • Unbound fraction based on
    physicochemical properties
43
Q

How is Prediction of the renal clearance (CLr) done ?

A
  • Renal clearance : 3 processes
  • Mostly estimated in vivo using allometry
  • Allometry :
  • scale values from lab animals to humans
  • Allometry assumption: the only
    difference in renal clearance between
    species is the size of species.

Allometry assumption: the only difference in renal clearance
between species is the size of species.
* a, b: coefficient and exponent (The scaling (a) and the correction (b) factors).
* For the volume of distribution prediction b tend to be 1.
* For physiological processes b tends to be 0.75.
* Ignores metabolism process, transportation processes,
gastrointestinal physiology, absorption  imprecisions

Y= a * BWb
Clr = a * BWb

44
Q

Estimation of the Vd – Allometric methods ?

A

. Allometric scaling of Vdss
. VDss = a * Weight^b
. a and b can be determined by fitting observed VDss in animals
. Extrapolate to human
. Allometric scaling of unbound VD
VDss,u=VDss/fup

fup= fraction unbound in plasma

Estimation of the Vd - Performances of allometric scaling
* VDss  Overestimation
* VDss,u  Underestimation
* Best result using 3 animal species

45
Q

Estimation of the Vd – PBPK methods ?

A

Estimation of the Vd – PBPK methods
* There are many PBPK methods to estimate the volume of
distribution
* PBPK methods can include:
* Tissue and plasma composition of lipid, water, and plasma proteins
* Lipophilicity of each tissue = ratio of hydrophilic and lipophilic
components
* Lipophilicity of the drug
* Poulin and Theil PBPK model
* Po:w, Dvo:w, Vnl, Vph from literature

46
Q

the principle of in-silico modeling ?

A

takes Four PK parameters:
1. Clearance (CL)
2. Volume of distribution (V)
3. Oral absorption (ka)
4. Bioavailability (F)

and can make a graph out of connecting the dots from different plasma concentrations of the drug in time

47
Q

what do you know about IVIVE ?

A
  • In vitro-in vivo extrapolation (IVIVE) is a crucial concept in translational
    pharmacology that allows the prediction of human pharmacokinetics from
    preclinical data.
  • IVIVE is an essential tool in optimizing drug development and selecting
    promising drug candidates.
  • The utilization of IVIVE and PBPK modeling can help in the development
    of personalized medicine strategies
  • However, it’s important to keep in mind that IVIVE has limitations and it’s
    important to validate predictions with clinical data
  • Future research in the field of IVIVE should focus on improving the
    predictive power of the models and increasing their translational potential
48
Q

What are the ordinary differential equations
(ODE) ?

A

Ordinary differential equations
* Ordinary differential equation (ODE):
* a mathematical equation that describes the rate of change of a
dependent variable (e.g., drug concentration) with respect to an
independent variable (e.g., time).
* Often used in pharmacokinetic modeling to describe the
dynamics of drug absorption, distribution, metabolism, and
elimination in the body.
* ODEs are used to describe the mass balance of a drug within a
particular compartment (such as the gut, liver, or plasma).

dy/dt= f(t,y)

  • The mass balance equation describes the flow of the drug into
    and out of the compartment and the rate at which the drug is
    metabolized or eliminated from the compartment.
  • The solution to the ODE describes the amount/concentration of
    the drug within the compartment over time.
49
Q

One compartment differential equation?

A

ODE
* dA/dt represents the rate of change of drug
amount (A) with respect to time (t).
* Dka represents the rate of drug absorption
into the compartment
* A
ke represents the rate of drug elimination
from the compartment
* Hint: what happens in the compartment?
* Drug increasing or decreasing?  define the
amount and singe (+/-)
* At which rate?  defines which rate to use (ka, ke)

  • To solve the ODE and obtain the amount of drug in the compartment (or concentration)
  • numerical integration method:
  • The Euler or the Runge-Kutta method
    1. Needs to define initial conditions
  • At time = 0
  • Dose = D(0) = 100mg
  • Amount in compartment = A(0) = 0 mg
    2. Needs to define the step of integration
  • dt = 1
50
Q

ODE applications?

A

PK/PD
dA/dt = Dk - Ak

Growth models
dN/dt = N*k

Chemical reactions
dA/dt = BKBA - AKAB
dB/dt = AKAB - BKBA

51
Q

what is the framework before working with ODE in PK ?

A
  1. Define mass balance (the amounts of drug and rates) for each
    compartment
  2. If needed, define condition-based changes (e.g., the rate of
    infusion changes at a specific time)
  3. Define equations for the investigated parameter(s) (e.g.,
    Concentration = Amount/Volume)
  4. Define each compartment state at time = 0 (initialize the
    system) (e.g., The drug amount in the central compartment at
    time =0
52
Q

all the different instances of compartments in PK models

A

look at the slides 11-19 now

53
Q

Compartment definition , intercompartmental clearance ?

A
  • Compartments can be defined using inter-compartmental clearance (Q)
     It is NOT THE BLOOD FLOW!!!
  • k12 = Q/V1
  • k21 = Q/V2
54
Q

Types of PD markers ?

A
  • Biomarker
  • Measurable physiological parameters that reflect the activity of the drug
  • T-cells CD4 count (HIV), LDL-cholesterol (statins)
  • Surrogate marker
  • Observed earlier than the clinical outcome, easy to quantify
  • Albumin in liver impairment.
  • Clinical outcome
  • Arterial tension, graft survival, patient survival
55
Q

PD , EC50,Emax , gamma , and effect

A

look at slides 22-24

56
Q

what should be considered Before building a model

A
  • Why build a model?
  • Leaning, understanding
  • Prediction = simulation
  • Which level of details to include/what to describe?
  • Empirical model
  • Semi-mechanistic model
  • Mechanistic model
  • Learning from the data Vs. Confirming the data
  • Learning: model built on the data
  • Confirming: model built before the data and compared to the data.

Empirical Model:

Empirical models are based solely on observed data without explicit consideration of underlying biological mechanisms.
They describe the relationship between drug concentrations and time using mathematical equations or statistical techniques.
These models are useful when little is known about the underlying biological processes or when the data is limited.
Examples include simple exponential decay models (e.g., one- or two-compartment models) or non-linear mixed-effects models.
Semi-Mechanistic Model:

Semi-mechanistic models combine elements of empirical observation with partial consideration of underlying biological mechanisms.
They incorporate both empirical parameters and parameters representing physiological or biochemical processes.
These models provide a balance between simplicity and biological relevance, often capturing important aspects of drug behavior while still being relatively interpretable.
Examples include physiologically-based pharmacokinetic (PBPK) models, which incorporate physiological parameters like blood flow rates and tissue volumes, along with empirical parameters to describe drug disposition.
Mechanistic Model:

Mechanistic models fully incorporate known biological mechanisms underlying drug disposition.
They are often complex and detailed, describing drug behavior based on the underlying physiology and biochemistry of the body.
These models provide a comprehensive understanding of drug kinetics and dynamics, allowing for detailed predictions under various conditions.
Examples include systems pharmacology models, which integrate information about drug targets, pathways, and cellular processes to predict drug effects.

57
Q

how are we Building a PK model?

A

Dataset

Suitable
model

Fit data to
model

Get PK
parameter
estimates

58
Q

what does Fitting data mean ?

A

Dataset
times and Concentrations in plasma (Cp)

Model selection

Starting values:
Ka = 0.8
Ke = 0.1
V = 6
Fitting algorithm: try to find values
for which we have the best fits for
data

Final parameters values
Final Model : Structural model
Ka = 0.6888
Ke = 0.3415
V = 7.21551

59
Q

Building a PK model – what is The error ?

A

Data = Model + Error

Residual Error (RE)

-Variability
from biological
processes
- Error of measurement
- Noise

60
Q

Building a PK model – Model selection

when a change of model is needed ?

A

Different starting values cannont explain it well

-Exploring a different structural model might be an option

61
Q

Building a PK model – General rules ?

A
  • One change at a time
  • Compare models based on metrics:
  • Objective function value (OFV), AIC, BIC
  • Error values
  • Graphical diagnostics
  • Parsimony
  • Physiological plausibility of final estimates :
  • Model fitting is a blind process

A parsimonious model is a model that accomplishes the desired level of explanation or prediction with as few predictor variables as possible.

62
Q

PK/PD modeling tools

A

Commercial, specialized
in PKPD:
NONMEM= Versatile Cost but
Blackbox

Commercial for general
modeling: Matlab=
Flexible but
Costly

berkley madona = better transparency but need fro programming

Open source modeling:
R
Pkfit for R
Tdm for R
nlmixr

Open source
Transparent
Flexible

but

Require learning
programming

63
Q

why is there a Need for in-silico PK/PD tools ?

A
  • In-silico PK/PD studies
  • Less expensive and quicker experiments
  • Provide a more complete understanding of the mechanisms of drug
    action and the factors influencing it Inform the study design for more
    targeted and effective drugs.
  • Easier in-vitro in-vivo extrapolation, identify potential issues earlier 
    fail earlier.
  • Extrapolate from animal models of disease.
  • Offer an integrative framework that combines most of the data
    generated during in vitro assays and in vivo experiments.
  • Can be updated as the project progress and more data are collected
  • Optimize dosing regimens  Reduce the cost and time of drug
    development
64
Q

Pharmacokinetics – why is there a Need for a simplified model ?

A
  • The handling of a drug by the body is complex
  • Absorption, distribution, metabolism, elimination
  • Simplifications are necessary to predict drug behavior in the
    body
  • Model of the body can be applied
  • Compartmental models allow the description of the dynamic
    changes in the drug concentration across time
65
Q

why in Pharmacokinetics there is a Need for a simplified model ?

A
  • The handling of a drug by the body is complex
  • Absorption, distribution, metabolism, elimination
  • Simplifications are necessary to predict drug behavior in the
    body
  • Model of the body can be applied
  • Compartmental models allow the description of the dynamic
    changes in the drug concentration across time.
66
Q

Pharmacokinetics – Individual level variabilities ?

A

Residual Error (RE) Intra-individual (intra-occasion) variability (IOV)

67
Q

Pharmacokinetics – Population level variabilities ?

A

Inter-individual variability

68
Q

what do we need to summarize everything in a model?

A
  • Describe pharmacokinetics on the population level
  • Describe pharmacokinetics on the individual level
  • Describe the residual error on every individual
  • Describe the variability between individuals

Population pharmacokinetic modeling
(PopPK)

69
Q

what are the 3 main Population modeling - Approaches ?

A

Naive Pooling
* Two-Stage
* Nonlinear Mixed Effect Modeling (NLME)

70
Q

Naïve Pooling ?

A
  • Create a PK profile from the combination of all data points (population)
  • Ignore
  • Inter-individual variability
  • Intra-individual variability (Residual error)
71
Q

Two-Stage approach ?

A
  • Require complete PK profiles of your population (Rich data)
    1.Estimate separately each individual PK parameters
  • One model per individual
    2.Determine the mean and the variability of each PK parameter* Require rich sampling
  • Estimate separately each individual PK parameters
  • Estimate the mean of the population PK parameters
  • Variabilities are inflated (Poor separation IIV - RE)
72
Q

when does the Classical two-stage approach work and when does it not work?

A

Works well to get an estimate of the average concentration curve, i.e. in a
”typical” subject
Problematic:
* Different PK models (1-compartment, 2-compartment, etc.) are fitted to
different subjects. Cannot calculate relevant average PK parameters.
* If other statistics than average values are needed, (e.g., variances)
* Difficult with inhomogeneous sampling, (e.g., different sampling times in
different subjects).

73
Q

Nonlinear Mixed Effect
approach (NLME)

A

Population modeling – Nonlinear Mixed Effect
approach (NLME)
* Fit ONE model to all the data from all individual
* WHILE retaining the notion of individuals
* Estimate the mean and variabilities (RE + IIV) of the PK parameters

  • Based on assumptions
  • Variability in the measurement of a population
  • Errors in measurement of a population

Come from a distribution
(e.g., Normal)

74
Q

Residual error

A

Residual error ( epsilon ) = Cobs – Cpred(i)

75
Q

in practice how do we come to the CL final for example ?

A

CLi=CLpop+IIV
V i= Vpop+IIV

kai go ana vrw to CLfinal prosthetw se auta to RE

76
Q

Covariate model ?

A
  • Explain the variability between subjects
  • Reduces the variability
  • Quantification of the PK parameters – covariates relationship
  • Allow controlling the safety and efficacy of the drug
  • Allow the individualization of doses
  • Covaraite can be
  • Continious
  • Discrete

Definition of Covariates: Covariates are patient-specific factors that may influence drug PK parameters. These factors can include demographic characteristics (e.g., age, sex, body weight), physiological variables (e.g., renal function, hepatic function), genetic factors (e.g., genetic polymorphisms), and clinical variables (e.g., disease status, concomitant medications).

Identification of Covariates: Covariates are identified through statistical analysis of the population PK data. This typically involves evaluating the relationship between individual PK parameters (e.g., clearance, volume of distribution) and potential covariates using regression analysis or other statistical methods.

Modeling Approach: In covariate modeling, the relationship between PK parameters and covariates is described using mathematical equations within the population PK model. These equations allow for the estimation of how each covariate influences the individual PK parameters for each patient in the population.

Parameterization: Covariate modeling involves parameterization of the covariate effects on PK parameters. This may include fixed effects parameters (e.g., slope and intercept for linear relationships) and random effects parameters (e.g., variability in covariate effects between individuals).

77
Q

Case – Tamoxifen PopPK model ?

A
  • Goals :
  • Create a PopPK model for tamoxifen and 3 of its metabolites
  • Quantify the variability in each of the PK parameters
  • Identify covariates that affect the PK of tamoxifen
  • Age, weight, gender,….
  • The effect of genetic polymorphisms on drug PK
  • CYP2C19
  • CYP2D6
  • Simulate different treatment regimen
78
Q

modling in the case of tamoxifen can lead to ..?
While simulations lead to …?

A

Individualisation
of the
treatment

Improvement
of the dosing
regimen

79
Q

which are the Population model components ?

A

Mechanistic model
Model structure

Statistical model
Statistical description of
* Data variability
* Error

Covariate model
Factors affecting
the data
variability

80
Q

Applications of PopPK ?

A
  • Drug development: Prediction of the optimal dose for certain
    populations (e.g., renal impairment, pediatric)
  • In combination with PD (PopPKPD) define a therapeutic window
  • Simulation of different dosing regimens (chance of efficacy vs
    the risk of toxicity)
  • Covariate identification/analyses
  • Precision medicine: Dose individualization using Bayesian
    forecasting.
81
Q

which is the predominant technique and the
most flexibl?

A

Non-linear mixed-effects modeling

82
Q

Future directions in PopPK ?

A

Future research in PopPK should focus on the identification of new
sources of variability and the development of more sophisticated models to
improve the predictability of drug response.

83
Q

User
why Traditional PK is a Top-down approach ?

A

Data-Driven: Traditional PK modeling begins with empirical data obtained from in vivo studies, such as clinical trials or preclinical experiments. These data consist of observed drug concentrations at various time points following drug administration.

Curve Fitting: The observed drug concentration-time data are analyzed using mathematical techniques to fit mathematical models to the data. These models describe the relationship between drug concentration and time, typically using compartmental or non-compartmental models.

Parameter Estimation: The parameters of the PK model, such as clearance rate, volume of distribution, and absorption rate constants, are estimated by fitting the model to the observed data. These parameters represent the underlying physiological and pharmacokinetic processes governing drug disposition in the body.

Extrapolation and Prediction: Once the PK model parameters are estimated, they can be used to extrapolate drug behavior beyond the observed data points. This allows for prediction of drug concentrations under different dosing regimens, in different patient populations, or in response to drug-drug interactions.

Sensitivity Analysis: Traditional PK modeling often involves sensitivity analysis to assess the impact of model parameters on drug exposure. This helps identify key factors influencing PK variability and informs decision-making in drug development and dosing optimization.

Limited Mechanistic Insight: While traditional PK models provide a quantitative description of drug concentration-time profiles, they may lack mechanistic insight into the underlying physiological processes governing drug disposition. Instead, they rely on empirical relationships derived from observed data.

Application in Clinical Practice: Traditional PK modeling is widely used in clinical pharmacology to guide dosing regimens, optimize drug therapy, and predict drug exposure in patients. It provides valuable information for drug labeling, dosing recommendations, and therapeutic drug monitoring.

84
Q

why PBPK is a Bottom-up approach ?

A

Physiological Basis: PBPK models are built on a foundation of physiological principles. They incorporate detailed knowledge of organ physiology, blood flow rates, tissue volumes, tissue composition, and other relevant factors that influence the pharmacokinetics (PK) of drugs.

Anatomical Considerations: PBPK models take into account the anatomical structure of the body, including the distribution of organs and tissues, their sizes, and their spatial relationships. This anatomical detail allows for more accurate representation of drug distribution within the body.

Mechanistic Representation: PBPK models strive to mechanistically represent the processes involved in drug absorption, distribution, metabolism, and excretion (ADME) within the body. This involves incorporating physiological processes such as passive diffusion, active transport, metabolism by enzymes, and elimination by organs.

Integration of Data: PBPK models integrate available data on drug properties (e.g., physicochemical properties, binding affinity, metabolic pathways) with physiological parameters to simulate drug behavior in the body. This integration allows for a comprehensive understanding of how drugs interact with the body.

Parameterization: PBPK models require detailed parameterization based on physiological and drug-specific data. Parameters may include tissue permeability coefficients, enzyme kinetic parameters, protein binding constants, and organ blood flow rates. These parameters are often obtained from experimental data or literature sources.

Validation: PBPK models are validated by comparing model predictions to observed drug concentrations in clinical studies or experimental data. This validation process ensures that the model accurately captures the PK behavior of the drug in humans under various conditions.

Application in Drug Development: PBPK models are used in various stages of drug development, from early drug discovery to regulatory submissions. They are valuable tools for predicting human PK, optimizing dosing regimens, assessing drug-drug interactions, and extrapolating findings across different populations.

85
Q

What is the problem with traditional PK modeling ?

Limitations of the traditional PK modeling?

A
  • Traditional PK analysis rarely take into
    account the knowledge about
    physiological and biological processes.
  • They are restricted to empirical or simple
    compartmental kinetic models.

that is why PBPK -* Not a new concept (1973)
* PBPK was/still used a lot in
environmental toxicology.

  • Based on empirical data
  • May not accurately predict drug behavior in certain populations
    or situations
  • May not take into account differences in physiological parameters such
    as organ size or enzyme activity.
  • Not very suitable for testing a mechanistic hypothesis.
  • Direct extrapolation of PK parameters is not straight-forward
86
Q

why is there an increase in PBPK interest ?

A
  • Availability of user-friendly software
  • Increase in computational power
  • Better connectivity between PBPK to IVIVE
  • Early prediction of ADME properties before in-human trials
  • More In-vitro data is available.
87
Q

what are the Principles of PBPK?

A
  • Uses physiological parameters + mathematical equations
  • Uses the same compartmental approach as traditional PKPD
    models.
  • Uses the ODE to describe the system dynamically.
  • Can integrate physicochemical, biological, physiological, and
    anatomical drug properties related to in vitro-assay, animal, and
    human studies.
     More mechanistic and physiological insights.
88
Q

what are the assumptions we are making Distribution in Tissue in PBPK ?

A

Assumptions:
* Tissue is a well-stirred single
compartment
* Diffusion is instantaneous
* Diffusion is driven by the concentration
gradient
between blood and tissue
* CTu and CvTissue are in equilibrium

89
Q

what is different about the Lung model?

A

Venous blood enters the lungs and
arterial blood leaves the lungs

90
Q

which are the components of a PBPK model?

A
  1. Physiological parameters
    * Blood flow values (Q)
    * Tissues volume (V)
  2. Drug parameters
    * Tissue to plasma partition coefficient (Kp)
    * Protein-binding data
    * Intrinsic clearances
  3. Model structure
    * Number of tissues and their arrangement
    * Systemic circulation (Arterial + venous blood)
91
Q

what are the Advantages of joining PBPK and IVIVE ?

A
  1. Ability to separate drug data from systems data
  2. Prediction of PK and PD of drugs on levels of
  3. Organ
  4. Individual
  5. Population
  6. PBPK rely on IVIVE but not exclusively
    * PBPK get also data from human studies/trials
     reduces the overall risk by balancing the data input
92
Q

PBPK Vs Traditional PK ?

A
  • Traditional PK  Top-down approach
  • PBPK  Bottom-up approach
  • PBPK  Middle out approach: optimization parameters of PBPK model
    as more clinical data become available.

Traditional PK
CL,V,Bioavailability

Use of average/typical person
Need for covariates to personalise

ka, k12, k21, …
POP-PK is needed to define the covariate effect on
the different rates

Sources of variability:

Values are rarely reflective of the drug alone
Data reflect properties of the system and trial
conditions

Covariates : food, age, gender, race, polymorphisms,
comedications, comorbidities, …

PBPK

CL
Sum of affinity, efficacy, enzymes, transporters,
blood flow

V
Sum of physical volumes, red cell volumes. Take into
account drug uptake, the composition of organs

Bioavailability
Formulation, physical properties : solubility,
dissolution, permeability/effect of transporters.

ka, k12, k21, …
Transfer between organs and tissues. Take into
account fluid flows, drug bindings, passive and
active transport (efflux/influx)
Data for the drug, the in vitro, and in vivo systems
are separated.

Sources of variability:
Assays related, analytical methods, correction
factors,…

93
Q

PBPK modeling tools ?

A

Commercial specialised in
PBPK

GastroPlus
SimCyp
PKSim

GUI that takes care of
complex tasks
Cost
Non-transparent
Not-flexible

Commercial for general
modeling

Matlab
Berkley Madonna
Flexible
Better transparency
Cost
Need for programming

Open source modeling R, Python Open source
Transparent
Flexible
Require learning
programming

Open source tools
HTTK package (R) Open source
Transparent
GUI
Need some learning

94
Q

Applications of PBPK ? SOS

A
  1. Prediction of toxicity
  2. Prediction of first-in-human dosage requirements
  3. Prediction of drug ADME in different populations
    * Dosing recommendations + Safety
  4. Evaluating the effect of disease or other physiological changes on drug
    ADME
  5. Understanding the mechanisms of drug action
    * Identify potential drug targets
  6. Assessing the potential for drug-drug interactions
  7. Optimizing drug development and dosing
  8. Improving personalized medicine
95
Q

Limitations of PBPK ?

A
  1. Complexity
  2. Limited data
  3. Assumptions and simplifications
  4. Limited predictive power
  5. Validation
  6. Lack of standardization
96
Q

in the ADME process which are thing mainly dictated by physicochemical properties of the compound and which are active processes ?

A

Solubility, membrane permeability, plama protein binding , Vd

Transporters and metabolic enzymes