Principle of Drug Design Flashcards

(54 cards)

1
Q

What is the primary goal of drug (lead) optimization?

A
  • To enhance a drug’s interaction with its target
    binding site.
  • Increase activity and selectivity.
  • Minimize side effects.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What role does structure-based drug design play in lead optimisation?

A
  • Relies on X-ray crystallography and computer-based
    molecular modelling.
  • Allows researchers to visualize how lead
    compounds and their analogues bind to a target.
  • Guides the rational design or modification of compounds for better fit and function.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Why is the compound with the strongest binding to the target not always the best medicine?

A
  • Pharmacokinetics (absorption, distribution, metabolism, excretion) and toxicity must be considered.
  • A compound’s overall ADME profile and safety might not align with clinical use, despite strong binding affinity.
  • Optimal drugs balance binding interactions with other essential properties to ensure efficacy in patients.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

T/F: The best binder always the best drug?

A

False - drug efficacy depends on more than just target affinity, ADME properties are essential. The drug must also be safe and well tolerated clinically.

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

What are the three principal phases of drug action, and what do they determine?

A
  1. Pharmaceutical Phase – Determines how much of the drug becomes available for absorption (e.g., disintegration, dissolution).
  2. Pharmacokinetic Phase – Determines how much of the drug reaches the site of action (via absorption, distribution, metabolism, excretion).
  3. Pharmacodynamic Phase – Determines how the drug interacts with tissues/receptors to produce a therapeutic effect.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is the significance of the pharmaceutical phase?

A
  • Involves formulation and dosage form (e.g., tablet, capsule).
  • Process of disintegration (breaking apart) and dissolution (dissolving).
  • Directly impacts how much drug is available to be absorbed into the bloodstream.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is the main focus of the pharmacokinetic phase (ADME)?

A

Absorption: Getting from the site of administration into circulation.

Distribution: Traveling through blood to tissues and target sites.

Metabolism: Conversion into active/inactive forms (often in the liver).

Excretion: Removal from the body (commonly via kidneys or bile).

Overall, it determines how much drug ultimately makes it to the target and for how long.

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

How does the pharmacodynamic phase differ from the other two phases?

A

Focuses on the drug’s effect on the body, i.e., the mechanism of action at the receptor or target site.

Encompasses therapeutic effects and side effects based on how strongly and selectively the drug binds to its biological targets.

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

What is the goal of lead optimization in drug design?

A

Improve activity and selectivity at the target.

Enhance pharmacokinetic characteristics (e.g., better absorption, stable metabolism).

Minimize side effects and toxicity.

Ensure the final compound performs well in real-world biological systems (not just in binding assays).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What strategies are commonly used to optimize a lead compound?

A

Variation of substituents on the core structure.

Extension of the molecule (adding new functional groups).

Isosteric or bioisosteric replacement (swapping chemical groups with similar properties).

Simplification of overly complex structures (removing non-essential parts).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What are the major enteral routes of drug administration?

A
  • Oral (by mouth)
  • Rectal (suppository) or vaginal (pessary)
  • Buccal (between gum and cheek) and sublingual (under the tongue)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What are examples of parenteral and other administration routes?

A

Parenteral: Intravenous (IV), intramuscular (IM), subcutaneous (SC)
Other: Ocular (eye drops), nasal/pulmonary (inhaled), transdermal (topical creams, patches)

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

What are the three main categories of dosage forms?

A

Liquid formulations: solutions, suspensions, emulsions

Semisolid formulations: creams, ointments, gels

Solid formulations: tablets, capsules

All require suitable excipients to ensure stability and bioavailability.

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

What is the main objective of the pharmacokinetic phase?

A

Optimizing drug availability at the target site.
Involves absorption, distribution, metabolism, excretion (ADME) to ensure sufficient active drug reaches its site of action.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What are the key “partition steps” a drug undergoes to reach its receptor?

A

Leaving aqueous extracellular fluid
Crossing the lipid membrane
Re-entering an aqueous environment before binding to its target

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What factors influence a drug’s ability to reach its target tissue?

A

Hydrophilic/hydrophobic balance
Ionization state
Molecular size
Chemical & metabolic stability

These properties affect how easily a drug crosses membranes and survives in the body.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What are potential problems related to drug metabolism?

A

Loss of activity: Metabolites might be inactive.

Increased toxicity: Some metabolites can be more harmful.

Variable enzyme activity: Differences in cytochrome P450 and other enzymes among individuals affect drug response.

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

Why is the drug–receptor interaction critical in pharmacodynamics?

A

Determines efficacy (how well it activates or blocks a receptor) and selectivity (specificity for the target).

Informs the pharmacophore—the essential features needed for activity.

Guides Structure-Activity Relationship (SAR) studies to improve therapeutic profiles.

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

What is a pharmacophore, and why is it important?

A

A pharmacophore is the 3D arrangement of molecular features necessary for a drug’s biological activity.

It focuses optimization efforts, ensuring modifications retain or enhance the crucial interactions with the target receptor.

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

What is the primary purpose of Lipinski’s Rule of Five in drug design?

A

It’s a simple “rule of thumb” to gauge whether a molecule has properties compatible with reasonable oral absorption.

Proposed by Chris Lipinski at Pfizer, derived from surveying marketed drugs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What are the key criteria in Lipinski’s Rule of Five?

A

No more than 5 hydrogen bond donors (sum of OH and NH groups).

A molecular weight (MW) below 500 Da.

A log P (lipophilicity measure) below 5.

No more than 10 hydrogen bond acceptors (sum of N and O atoms).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Do Lipinski’s rules ensure a compound will be pharmacologically active?

A

No. They only suggest a molecule is likely to be orally bioavailable.

Activity still depends on target specificity, pharmacodynamics, and other biochemical factors.

22
Q

How do absorption, distribution, and metabolism impact a drug’s effect at its receptor?

A

They determine the amount of drug that actually reaches and remains at the target site.

Poor ADME properties can render even a potent drug ineffective or toxic in vivo.

23
Q

Why is lipophilicity and ionization important for passive membrane crossing?

A

Lipophilic and unionized compounds cross lipid membranes more easily.

Ionized or highly polar drugs may struggle to permeate cell membranes, affecting their bioavailability.

24
Why should drugs generally have the lowest possible log P?
Minimizes nonspecific binding and toxicity. Increases ease of formulation and bioavailability. Reduces issues with accumulation in lipid-rich tissues.
25
Why is stereospecificity critical in rational drug design?
Biological targets (enzymes, receptors) may prefer one enantiomer over another. Different stereoisomers can exhibit varying levels of efficacy, affinity, or toxicity. Optimizing stereochemistry is essential to develop safer, more effective drugs.
26
What is a typical model for rational drug design?
Find a lead compound. Study Structure-Activity Relationships (SAR) and modify the structure. Identify the pharmacophore (crucial binding features). Generate new analogues. Evaluate and refine to achieve optimum therapy (efficacy, safety, stability).
27
What is a “drug target” (sometimes called a drug receptor)?
A structure or macromolecule associated with a disease that a drug binds to. Can be on the cell surface or inside the cell (e.g., proteins, nucleic acids).
28
Which types of macromolecules can act as drug targets?
Enzymes Receptors Carrier proteins Structural proteins Nucleic acids Lipids Carbohydrates
29
What is meant by an “agonist” at a receptor?
A drug that mimics the natural ligand (e.g., neurotransmitter or hormone) Activates the receptor, producing a biological response.
30
How does an “antagonist” work at a receptor?
It binds to the receptor but doesn’t activate it. Blocks the binding of the natural messenger or prevents receptor activation. May bind at the active site or an allosteric site, causing structural changes that affect signalling.
31
What are possible outcomes of binding an allosteric site on a receptor?
Inhibition: Changing the receptor conformation to reduce natural messenger binding. Potentiation: Enhancing receptor affinity for the natural ligand.
32
What are the main categories of enzyme inhibitors?
Competitive inhibitors: Compete with substrate for the active site. Non-competitive inhibitors: Bind to an allosteric site, changing enzyme conformation. Reversible inhibitors: Bind temporarily and can dissociate. Irreversible inhibitors: Form stable covalent bonds, permanently inactivating the enzyme. Transition state mimics: Resemble high-energy intermediate, binding tightly to the active site.
33
Name three families of membrane-bound receptors and their general roles.
G-protein coupled receptors (GPCRs): Activate intracellular G-proteins to trigger signalling cascades. Ligand-gated ion channels (ionotropic receptors): Open an ion channel upon ligand binding for fast cellular responses. Kinase-linked receptors: Typically stimulate phosphorylation events within the cell to regulate gene expression and other processes.
34
What are some non-receptor, non-enzyme targets for drugs?
Structural proteins (e.g., microtubules targeted by vincristine) Nucleic acids (e.g., intercalators, alkylating agents, antisense strategies) Lipids (e.g., anesthetics that alter membrane fluidity) Carbohydrates on the cell surface (e.g., drugs affecting cell recognition or adhesion)
35
Which mechanisms of action are most common among first-in-class small-molecule NMEs?
They often work by inhibiting enzymes or modulating receptors. Novel mechanisms can lead to entirely new therapeutic classes.
36
What basic requirements must a drug meet for successful receptor binding?
Correct binding groups (functional groups that can interact with the target). Proper spatial arrangement (groups positioned correctly for binding). Appropriate molecular size to fit the binding site. Must form the right types of interactions (e.g., hydrogen bonds, hydrophobic interactions) based on its functional groups and the receptor’s binding site.
37
Which factors primarily influence a drug’s binding and overall action?
Molecular structure (shape and functional groups). Isomerism (the arrangement of atoms in space). Functional groups (which govern binding interactions). Rigidity or flexibility (more rigid structures may bind in a specific conformation, while flexible ones adapt to multiple conformations).
38
What are the two main approaches to drug design?
Receptor-based (target-based): Begins with a known target structure (e.g., via X-ray crystallography). Ligand-based: Relies on known active compounds (ligands) and their structure-activity relationships (SAR) to guide optimization.
39
What is a pharmacophore, and why is it important?
An abstract description of molecular features necessary for recognition by a biological macromolecule. Summarizes the key binding groups (e.g., H-bond acceptors, hydrophobic regions, aromatic rings) required for biological activity.
40
Which functional groups are typically highlighted in a pharmacophore model?
Hydrophobic regions (often depicted in one color). Aromatic rings. H-bond acceptors and donors. Any other group known to be essential for receptor binding.
41
What are the two major aspects of drug optimization?
Optimizing access to the target (Pharmacokinetics) – Ensuring the drug has suitable ADME properties. Optimizing target interactions (Pharmacodynamics) – Enhancing binding affinity, specificity, and functional activity at the receptor.
42
Why might modifying the structure of a lead compound be necessary?
To improve binding interactions and selectivity. To optimize pharmacokinetic properties (absorption, distribution, metabolism, excretion). To reduce toxicity or side effects.
43
Can you give an example of a critical functional group for analgesic activity?
A free phenolic OH group is often crucial for certain analgesics (e.g., in opioid structures), highlighting the importance of specific functional groups in maintaining activity.
44
What are the essential binding groups in the analgesic’s pharmacophore, and how do they interact with the receptor?
Phenolic –OH: Forms hydrogen bonds. Aromatic ring: Engages in van der Waals interactions. N-methyl group: Participates in ionic bonding. These groups are arranged in a T-shaped conformation, crucial for receptor recognition.
45
Why does the mirror-image (enantiomer) of this analgesic lack activity?
The reversed stereochemistry prevents correct alignment of key binding groups with the receptor’s binding sites. Only one enantiomer fits the required pharmacophore arrangement, so the mirror image cannot fully engage the receptor.
46
What is drug extension, and why is it used in lead optimization?
Drug extension adds extra binding groups to the original structure. This strategy probes for additional binding sites on the receptor surface. The goal is to enhance drug–receptor interactions and potentially improve potency or selectivity.
47
How does changing the length or size of substituents help in drug optimization?
Filling additional hydrophobic pockets within the binding site can enhance potency. Adjusting substituents can introduce selectivity for one target over another. Increases or decreases in substituent size/length can refine the fit between drug and receptor.
48
What is “extension” in drug optimization, and why is it used?
Involves adding extra functional groups to the lead structure. Explores additional binding regions in the target site. Aims to improve drug–target interactions and potentially boost specificity or potency.
49
How does modifying the chain connecting two binding groups help optimize a drug?
Adjusting chain length maximizes each group’s interaction with its binding region. Correct length/angle can enhance affinity and selectivity. Too long or too short a chain may reduce overall binding strength.
50
What are the benefits of modifying or adding rings in a lead compound’s structure?
Novel scaffolds can lead to new drug classes with improved properties. Ring fusion can enhance receptor binding or increase specificity to avoid off-target effects. Changing ring size or substituents can optimize hydrophobic or electronic interactions.
51
What role do isosteres play in drug optimization?
Isosteric replacement swaps functional groups with ones having similar size and electronic properties. Helps retain or enhance activity while potentially improving ADME or reducing toxicity. Often used to overcome metabolic liabilities or patent restrictions.
52
Why might a drug designer simplify a lead compound, and what are the risks?
Removing non-essential groups lowers complexity and cost of synthesis. Can eliminate unnecessary stereocenters or structural elements not needed for activity. Risk: Oversimplification can lead to excessive molecular flexibility, decreasing activity and selectivity.
53
What are conformational blockers, and why introduce them into a lead structure?
Bulky substituents or ring constraints added to limit conformations. Encourages the molecule to adopt the active binding conformation. Reduces entropy cost upon binding, potentially improving potency and selectivity.