Summary

This document is a revision guide for medicinal chemistry. It covers topics such as drug discovery, drug metabolism, enzyme inhibition, receptors, and ligands. Key concepts like pharmacokinetics and pharmacodynamics are also discussed.

Full Transcript

Revision Document for AP0415 Medicinal Chemistry Chapter 1: Introduction to Medicinal Chemistry and Drug Discovery 1.1 What is a Drug? Definition: Drugs are compounds that interact with a biological system to produce a biological response. Classification: o Natu...

Revision Document for AP0415 Medicinal Chemistry Chapter 1: Introduction to Medicinal Chemistry and Drug Discovery 1.1 What is a Drug? Definition: Drugs are compounds that interact with a biological system to produce a biological response. Classification: o Natural products: Derived from plants, animals, or minerals (e.g., quinine from Cinchona bark). o Fermentation products: Produced by microorganisms (e.g., penicillin from Penicillium notatum). o Semi-synthetics: Modified natural products (e.g., semi-synthetic penicillins). o Completely synthetic: Man-made compounds (e.g., aspirin). Selective Toxicity: Drugs should be toxic to problem cells (e.g., cancer cells) but not normal cells. 1.2 Drug Discovery Process Stages: 1. Target Identification: Identify a biological target (e.g., protein, enzyme, receptor) involved in a disease. 2. Hit Generation: Identify initial compounds (hits) that interact with the target. 3. Lead Optimization: Improve the properties of lead compounds (e.g., efficacy, safety, pharmacokinetics). 4. Preclinical Development: Test drug candidates in non-human systems for safety and efficacy. 5. Clinical Trials: ▪ Phase I: Safety and dosage in healthy volunteers. ▪ Phase II: Efficacy and side effects in patients. ▪ Phase III: Confirm efficacy and monitor side effects in a larger patient population. 6. Regulatory Approval: Submit a New Drug Application (NDA) to regulatory agencies. 7. Post-Marketing Surveillance: Monitor the drug’s safety and effectiveness after approval. 1.3 Drug Metabolism Purpose: Convert drugs into more polar molecules for easier excretion. Phases: o Phase I: Functionalization reactions (e.g., oxidation, reduction, hydrolysis). o Phase II: Conjugation reactions (e.g., glucuronidation, sulfation, glutathione conjugation). Chapter 2: Drug Targets and Mechanisms of Action 2.1 Drug Targets Types of Drug Targets: o Enzymes: Catalyze biochemical reactions (e.g., ACE inhibitors). o Receptors: Bind ligands to trigger cellular responses (e.g., GPCRs). o Ion Channels: Allow ion flow across membranes (e.g., ligand-gated ion channels). o Nucleic Acids: DNA/RNA (e.g., anticancer drugs). o Transporters: Move molecules across membranes (e.g., neurotransmitter reuptake inhibitors). 2.2 Enzyme Inhibition Types of Inhibitors: o Reversible: ▪ Competitive: Competes with the substrate for the active site. ▪ Non-competitive: Binds to a site other than the active site, altering enzyme shape. ▪ Uncompetitive: Binds only to the enzyme-substrate complex. o Irreversible: Binds covalently to the enzyme, permanently inactivating it. Examples: o Aspirin: Inhibits cyclooxygenase (COX), reducing prostaglandin synthesis. o Sulphonamides: Inhibit dihydropteroate synthase, blocking folate synthesis in bacteria. 2.3 Receptors and Ligands Agonists: Activate receptors, producing a biological response (e.g., morphine activates opioid receptors). Antagonists: Bind to receptors but do not activate them, blocking the action of agonists (e.g., beta-blockers). Chapter 3: Drug Metabolism 3.1 Phase I Metabolism Reactions: o Oxidation: Adds oxygen or removes hydrogen (e.g., cytochrome P450 enzymes). o Reduction: Adds hydrogen or removes oxygen. o Hydrolysis: Breaks bonds using water (e.g., ester or amide hydrolysis). Examples: o Aromatic Hydroxylation: Adds a hydroxyl group to an aromatic ring. o Aliphatic Hydroxylation: Adds a hydroxyl group to an aliphatic chain. o Dealkylation: Removes an alkyl group (e.g., N-dealkylation, O- dealkylation). 3.2 Phase II Metabolism Conjugation Reactions: o Glucuronidation: Adds glucuronic acid to increase water solubility (e.g., morphine glucuronide). o Sulfation: Adds a sulphate group (e.g., minoxidil sulphate). o Glutathione Conjugation: Detoxifies reactive metabolites (e.g., NAPQI from paracetamol). o Amino Acid Conjugation: Adds glycine or glutamine (e.g., salicyluric acid from aspirin). o Acetylation: Adds an acetyl group (e.g., procainamide acetylation). o Methylation: Adds a methyl group (e.g., catechol-O-methylation). Chapter 4: Enzymes as Drug Targets 4.1 Angiotensin-Converting Enzyme (ACE) Inhibitors Mechanism: Inhibit ACE, preventing the conversion of angiotensin I to angiotensin II, a potent vasoconstrictor. Examples: o Captopril: Contains a thiol group that binds to the zinc ion in ACE. o Enalaprilat: Replaces the thiol group with a carboxylate group, reducing side effects. o Lisinopril: Contains a lysine side chain that binds to additional residues in ACE. 4.2 Acetylcholinesterase (AChE) Inhibitors Mechanism: Inhibit AChE, leading to acetylcholine buildup and overstimulation of the nervous system. Examples: o Nerve Gases (e.g., Sarin): Irreversibly inhibit AChE, causing severe neurological effects. o Organophosphate Insecticides (e.g., Parathion): Inhibit AChE in insects, leading to paralysis and death. Chapter 5: Antibiotics and Drug Resistance 5.1 Antibiotics Mechanisms of Action: o Inhibition of Cell Wall Synthesis: Penicillins, cephalosporins. o Inhibition of Protein Synthesis: Tetracyclines, chloramphenicol. o Inhibition of Nucleic Acid Synthesis: Quinolones, rifamycins. o Disruption of Cell Membrane: Polymyxins. o Inhibition of Metabolic Pathways: Sulphonamides. 5.2 Drug Resistance Causes: o Genetic Mutations: Bacteria develop resistance through mutations. o Horizontal Gene Transfer: Bacteria share resistance genes. o Overuse/Misuse of Antibiotics: Accelerates resistance development. Prevention: o Antimicrobial Stewardship: Responsible use of antibiotics. o Infection Control: Hand hygiene, sanitation, isolation. o Research and Development: New antibiotics, vaccines, and diagnostics. Chapter 6: Pharmacokinetics and Pharmacodynamics 6.1 Pharmacokinetics (ADME) Absorption: Movement of a drug from the administration site to the bloodstream. Distribution: Movement of a drug throughout the body. Metabolism: Chemical alteration of a drug (e.g., Phase I and Phase II reactions). Excretion: Removal of the drug and its metabolites from the body (e.g., via urine or bile). 6.2 Pharmacodynamics Dose-Response Relationship: o ED50: Dose that produces 50% of the maximum effect. o LD50: Dose that is lethal to 50% of the population. o Therapeutic Index: Ratio of LD50 to ED50 (higher values indicate safer drugs). Receptor Binding: o Agonists: Activate receptors. o Antagonists: Block receptors. Chapter 7: Case Studies in Medicinal Chemistry 7.1 Aspirin Mechanism: Inhibits cyclooxygenase (COX), reducing prostaglandin synthesis and inflammation. Metabolism: o Hydrolysis to salicylic acid. o Glucuronidation or glycine conjugation to form salicyluric acid. 7.2 Paracetamol Mechanism: Inhibits prostaglandin synthesis in the CNS, reducing pain and fever. Metabolism: o Glucuronidation (primary metabolite in adults). o Sulfation (primary metabolite in children). o Oxidation to NAPQI (toxic metabolite, detoxified by glutathione). 7.3 Sulphonamides Mechanism: Inhibit dihydropteroate synthase, blocking folate synthesis in bacteria. Selective Toxicity: Bacteria synthesize folate, while humans obtain it from their diet. Chapter 8: Advanced Topics in Medicinal Chemistry 8.1 Bioinformatics and Drug Design Role of Bioinformatics: Combines biology, computer science, and information technology to analyze biological data. Applications: o Target Identification: Identify potential drug targets using genomic and proteomic data. o Virtual Screening: Predict how well compounds bind to targets using computational models. o QSAR (Quantitative Structure-Activity Relationship): Correlate molecular properties with biological activity. 8.2 Personalized Medicine Role of Pharmacogenomics: Study how genetic variations affect drug response. Applications: o Tailor drug treatments to individual patients based on their genetic makeup. o Reduce adverse effects and improve efficacy. Chapter 9: Summary and Key Takeaways 9.1 Key Concepts Drug Discovery: A multi-step process involving target identification, lead optimization, and clinical trials. Drug Metabolism: Converts drugs into more polar molecules for excretion, involving Phase I and Phase II reactions. Enzyme Inhibition: A common mechanism of action for many drugs, including antibiotics and ACE inhibitors. Receptors and Ligands: Agonists activate receptors, while antagonists block them. Pharmacokinetics and Pharmacodynamics: Describe how drugs move through the body (ADME) and how they produce their effects (dose-response relationships). 9.2 Future Directions Combatting Drug Resistance: Develop new antibiotics and promote responsible use of existing ones. Advances in Drug Design: Use computational tools and bioinformatics to accelerate drug discovery. Personalized Medicine: Tailor treatments to individual patients based on genetic and molecular data. Molecules 1. Fundamental Biomolecules A. Carbohydrates Monosaccharides: o Basic units (e.g., glucose, fructose) o D/L configuration based on chiral carbon o Cyclic forms (α/β anomers) Polysaccharides: o Starch (α-1,4 with α-1,6 branches) - energy storage o Cellulose (β-1,4) - structural o Glycogen - animal starch, highly branched B. Nucleic Acids DNA Structure: o Double helix with antiparallel strands o Base pairing: A-T (2 H-bonds), G-C (3 H-bonds) o Phosphodiester backbone (5'→3') RNA Types: o mRNA: carries genetic code o tRNA: brings amino acids (anticodon matches codon) o rRNA: ribosome structure 2. From DNA to Protein A. Central Dogma Flow 1. Replication (DNA → DNA) o High fidelity (error rate 10⁻⁸-10⁻¹⁰) 2. Transcription (DNA → mRNA) o RNA polymerase o Post-transcriptional modifications 3. Translation (mRNA → Protein) o Ribosomes read codons o tRNA delivers amino acids o Peptide bond formation B. Key Processes tRNA Charging: o Aminoacyl-tRNA synthetases attach correct amino acid o Requires ATP Protein Synthesis: o Initiation: Start codon (AUG), Shine-Dalgarno sequence o Elongation: Peptide bonds form (GTP hydrolysis) o Termination: Stop codons (UAA, UAG, UGA) 3. Protein Structure & Function A. Structural Levels 1. Primary: Amino acid sequence 2. Secondary: o α-helices (H-bonds parallel to axis) o β-sheets (parallel/antiparallel) o Turns and loops 3. Tertiary: 3D folding o Stabilized by hydrophobic effect, H-bonds, disulfide bridges 4. Quaternary: Multiple subunits (e.g., hemoglobin) B. Protein Folding Driving Forces: o Hydrophobic collapse (nonpolar residues inside) o Hydrogen bonding o Chaperones assist folding Misfolding Diseases: o Prion diseases (α-helix → β-sheet) o Cystic fibrosis (CFTR mutation) 4. Drug Design Principles A. Structure-Activity Relationships (SAR) Key Concepts: o Bioisosteres: Replace groups while maintaining activity o Lipinski's Rule of 5 (oral drug guidelines) o Partition coefficient (log P) measures hydrophobicity B. QSAR (Quantitative SAR) Parameters: o Hydrophobicity (π constant) o Electronic effects (Hammett σ constant) o Steric factors (Taft Es) Applications: o Optimize drug absorption (e.g., balance hydrophilic/hydrophobic) o Reduce toxicity (e.g., replace toxicophores) 5. Membranes & Drug Transport A. Membrane Structure Phospholipid bilayer (amphipathic) Proteins: integral (transmembrane) vs. peripheral Composition varies (e.g., myelin = 79% lipid, erythrocyte = 44% lipid) B. Drug Absorption Lipinski's Rules: o MW < 500 o ≤5 H-bond donors o ≤10 H-bond acceptors o log P < 5 Polar Drugs: o Pinocytosis (cell "drinking") for large polar molecules o Transport proteins for small polar molecules Summary Sheet Carbohydrates Energy (starch), structure (cellulose) Glycosidic bonds: α (starch) vs β (cellulose) Nucleic Acids DNA: A-T, G-C, double helix RNA: single-stranded, U instead of T Protein Synthesis DNA → mRNA → protein tRNA matches codons, ribosomes make peptide bonds Protein Structure Levels: 1°→4° Folding driven by hydrophobic effect Drug Design Optimize log P, use bioisosteres Follow Lipinski's rules for oral drugs Membranes Lipid bilayer with proteins Drugs need right balance of polar/nonpolar