Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

13. Discuss the importance bioinformatics in drug design.

• Bioinformatics tools are essential in identifying and validating biological targets for new
drugs. By analyzing large datasets, including genomic, proteomic, and metabolomic
data, researchers can pinpoint molecules that play critical roles in disease processes.
• Additionally, bioinformatics is integral to pharmacogenomics, which studies how
genetic variations affect individual drug responses, paving the way for personalized
medicine and tailored treatment plans.
• Overall, bioinformatics reduces the time and cost associated with drug development by
minimizing the need for extensive laboratory experiments and focusing efforts on the
most promising candidates.

26.a) What are databases?

Databases are designed to ensure data can be easily accessed, managed, and updated. They
are used to efficiently store, retrieve, and manipulate data, supporting various applications
and ensuring data integrity and security. Databases are managed by database management
systems (DBMS), which provide tools for database creation, querying, updating, and
administration.
Example –
27.b) Write in short about the different databases used in drug
design.

28.c) Discuss the significance of ADME databases in view of drug


design.
The prediction of ADME properties is critical in the drug design process because these
properties are responsible for about 60% of all drug failures in clinical trials. Whereas in the
past, ADME methods were used towards the end of the drug development process, today
ADME is used at the beginning of the process to exclude molecules with weak ADME
properties from the drug development pipeline, resulting in substantial research and
development cost savings.
ADME database also contains the updated & comprehensive data on interactions of substances
with drug metabolising enzymes & drug transporters. It is intended for use in drug research &
development, including drug-drug interactions & ADME studies. The information is provided
by category, drug name, enzyme, reaction & type. It is supported by chemical / metabolite
structures as well as kinetic values mention in the literature.
The contents of the database were collected & organized according to the Human P 450 and
Transporter Metabolism Database. The database is accessible and completely searchable by
keywords or chemical structures. Advanced searches are also obtainable to support
investigational studies on drug-drug interactions. ADME Database contain more than 26,000
substances, several natural products and preparations, in addition to others factors affecting
Drug metabolizing enzymes activity. The data collected from more than 18,000 citations.

The significance of ADME databases in drug design:

• Optimizing Absorption:
o Solubility data helps predict drug dissolution in bodily fluids.
o Permeability information aids in assessing oral bioavailability.
• Predicting Distribution:
o Volume of Distribution (Vd) data helps understand drug spread in the body.
o Plasma protein binding information predicts active drug concentration.
• Understanding Metabolism:
o Data on metabolic pathways and enzymes (e.g., cytochrome P450) predicts
stability and interactions.
o Metabolite profiles assess pharmacological activity and toxicity of
metabolites.
• Evaluating Excretion:
o Information on renal and hepatic clearance helps predict drug elimination.
o Excretory pathways data (urine, bile, feces) aids in understanding drug
elimination routes.
• Enhancing Safety:
o Toxicity prediction data helps identify safety issues early.
o Drug-drug interaction information prevents harmful interactions.
• Facilitating Regulatory Approval:
o Detailed pharmacokinetic data meets regulatory requirements.
o ADME data aids in designing safe and effective clinical trials.
• Supporting Personalized Medicine:
o Genetic variability data predicts individual drug responses.
o Population-specific ADME data helps design effective drugs for diverse
groups.

32.a) What is homology modelling?


Homology modelling is one of the computational structure prediction methods that are used to
determine protein 3D structure from its amino acid sequence. It is considered to be the most
accurate of the computational structure prediction methods. The basic principle behind
homology modelling is that proteins with similar sequences tend to fold into similar structures.
The method relies on the fact that the 3D structure of proteins is often better conserved than
their amino acid sequence. Therefore, proteins with similar sequences are likely to have similar
structures. This method is becoming increasingly important as the number of experimentally
determined structures is limited compared to the number of protein sequences available.

Advantages of Homology Modelling

• Efficiency: Homology modelling is much faster and less expensive than experimental
methods like X-ray crystallography or NMR spectroscopy.
• Feasibility: It allows researchers to predict structures for proteins that are difficult to
crystallize or otherwise experimentally characterize.
• Insight into Function: Knowing the structure of a protein helps in understanding its
function, interactions, and role in biological processes.
• Drug Design: Predicted structures can be used to identify potential drug binding sites
and to design molecules that can interact specifically with the target protein.

Applications of Homology Modelling:

• Homology modelling is important for the prediction of protein structures that are
difficult to obtain experimentally using techniques like X-ray crystallography or NMR.
• Homology modelling is used to predict the functional properties of proteins with
unknown structures by modelling the protein structure and comparing it with known
structures of related proteins.
• Homology modelling is also useful in studying the evolution of protein families by
comparing the structures of related proteins from different species to identify the
changes that have occurred during evolution.
• Homology modelling can be used to identify potential drug targets by predicting the
structure of a protein to understand its function and identify potential sites for drug
binding. This information can be used to design new drugs or optimize existing ones.

33.b) How homology modelling helps in predicting protein


structures?

Steps in Homology Modelling:

1. Template Identification:

• Sequence Alignment: The target protein sequence is aligned with


sequences of known structures in databases like the Protein Data Bank
(PDB) to find the best template.
• Selection of Templates: One or more templates with high sequence
similarity to the target protein are selected.

2. Alignment Correction:

• Refinement of Alignment: The initial sequence alignment is refined to


ensure the best possible match between the target and template sequences,
particularly in regions critical for the structure.

3. Model Building:

• Core Region Modelling: The regions of the target protein that align well
with the template are modelled directly.
• Loop Modelling: Regions where the target and template do not align well,
such as loops, are modelled using specialized techniques.
• Side Chain Modelling: Side chains of amino acids are added and adjusted
to optimize their fit in the structure.

4. Model Refinement:

• Energy Minimization: The initial model is refined using computational


techniques to relieve any steric clashes and optimize the geometry.
• Molecular Dynamics: Further refinement may involve simulating the
physical movements of atoms to achieve a more accurate structure.

5. Model Validation:

• Quality Assessment: The final model is evaluated using various tools to


check for structural correctness, such as Ramachandran plots, which show
the distribution of phi and psi angles, and other criteria like bond lengths
and angles.

34.c) How the target is identified in drug design?

Ans: Steps to Identify a Target in Drug Design

1. **Disease Understanding**:

- Study disease mechanisms and biological pathways.

- Identify disease-associated biomarkers.

2. **Target Selection**:

- Review scientific literature for potential targets.

- Use genomics and proteomics to find differentially expressed or mutated proteins.

- Apply bioinformatics tools for target prediction and validation.

3. **Target Validation**:

- Perform genetic validation using RNAi or CRISPR.

- Conduct chemical biology studies with small molecules.

- Execute functional studies through biochemical and cellular assays.


4. **Target Characterization**:

- Determine 3D structures via X-ray crystallography, NMR, or cryo-electron microscopy.

- Develop binding assays to measure drug-target interactions.

- Analyze pathways to understand target's role and potential side effects.

5. **Drugability Assessment**:

- Evaluate target for drugability (suitable binding sites, modifiability).

- Screen chemical libraries for lead compounds.

OR

38.a) Enlist the various database’s used in drug design.


39.b) Discuss the significance of cheminformatics in drug design.
49.How bioinformatics helps in drug design?
50.Elaborate the applications of ADME databases in Drug Design.

Applications of ADME Databases in Drug Design

1. **Predicting Drug Absorption**:

- Provides permeability and solubility data.

- Models intestinal absorption for better bioavailability predictions.

2. **Understanding Drug Distribution**:

- Offers volume of distribution (Vd) information.

- Provides plasma protein binding data.

3. **Metabolism Prediction**:

- Details metabolic pathways and involved enzymes.

- Predicts drug-drug interactions.

4. **Excretion and Elimination**:

- Provides renal and hepatic excretion data.

- Supplies half-life information for dosing.

5. **Toxicity Prediction**:

- Identifies adverse drug reactions (ADRs).

- Predicts off-target effects and toxicological risks.

6. **Optimization of Lead Compounds**:

- Correlates chemical structure with ADME properties.

- Assists in high-throughput screening for optimal pharmacokinetics.

7. **Regulatory Compliance and Documentation**:

- Supports regulatory submissions with required ADME data.

- Informs clinical trial design with preclinical pharmacokinetic predictions.

OR

You might also like