UNIT 2 – General Pharmacology: Understanding How Drugs Act and Interact Notes

Every drug that enters the human body sets off a chain of biological events — binding, signaling, and response — that ultimately defines its therapeutic or toxic effects. This fascinating process, studied under pharmacodynamics and drug interaction principles, helps us understand not just what drugs do, but how and why they act.

From receptor theories to clinical drug development, this unit explores the intricate science of drug action, receptor mechanisms, and the safeguards that ensure medicines remain safe and effective for human use.

Download UNIT 2 – General Pharmacology: Understanding How Drugs Act and Interact Notes

Get simplified revision notes for this unit:

⬇️

Download Unit 2 Notes PDF

Pharmacodynamics: The Principles of Drug Action

Pharmacodynamics is the study of how drugs produce their effects on the body. It focuses on mechanisms of action, dose-response relationships, and the factors influencing a drug’s potency and efficacy.

When a drug enters the system, it interacts with specific biological targets such as receptors, enzymes, ion channels, or transporters. These interactions trigger biochemical and physiological responses that lead to therapeutic effects — or, at times, adverse reactions.

Receptor Theories and Classification

Receptors are specialized macromolecules that recognize and bind drugs or endogenous molecules (like hormones and neurotransmitters).

Receptor Theories

The earliest receptor theory — the occupancy theory — proposed that drug response is proportional to the number of receptors occupied. Later models, such as the rate theory and two-state model, emphasized receptor activation and conformational changes rather than mere binding.

Classification of Receptors

Receptors are broadly categorized based on their structure and mechanism of action:

  1. G-Protein–Coupled Receptors (GPCRs):
    These receptors, embedded in cell membranes, activate G-proteins that regulate enzymes and ion channels. Examples include β-adrenergic receptors.

  2. Ion Channel Receptors:
    Found in nerve and muscle cells, these receptors control ion movement (Na⁺, K⁺, Cl⁻, Ca²⁺) across membranes. They are responsible for rapid signaling, as seen with nicotinic acetylcholine receptors.

  3. Transmembrane Enzyme-Linked Receptors:
    These possess intrinsic enzyme activity, often tyrosine kinase, which triggers phosphorylation cascades. Insulin receptors work through this mechanism.

  4. Transmembrane JAK-STAT–Linked Receptors:
    These receptors lack intrinsic enzyme activity but activate Janus Kinase (JAK) enzymes, which in turn phosphorylate STAT proteins to regulate gene expression.

  5. Intracellular or Nuclear Receptors:
    Located in the cytoplasm or nucleus, they respond to lipid-soluble drugs or hormones like steroids and thyroid hormones, directly influencing transcription and protein synthesis.

Regulation of Receptors

Receptor activity is not static. Continuous exposure to drugs can alter their sensitivity — a process known as receptor regulation.

  • Upregulation occurs when receptors increase in number after prolonged absence of stimulation, making cells more sensitive.

  • Downregulation happens after prolonged drug exposure, leading to tolerance and decreased responsiveness.

This adaptability explains phenomena like drug tolerance, where higher doses are needed to produce the same effect over time.

Dose-Response Relationship and Therapeutic Index

The dose-response curve illustrates the relationship between the dose of a drug and the magnitude of its effect.

  • The graded dose-response shows how response intensity increases with dose until a maximum effect (Emax) is reached.

  • The quantal dose-response describes the percentage of individuals showing a particular effect at various doses.

The therapeutic index (TI) — the ratio between the toxic dose (TD₅₀) and effective dose (ED₅₀) — indicates a drug’s safety margin. A high TI means a wide safety range, while a low TI signals the need for close monitoring.

Combined Effects and Factors Modifying Drug Action

When drugs are used together, they may interact in ways that alter their effects:

  • Additive effect: The combined action equals the sum of individual effects (e.g., paracetamol + ibuprofen).

  • Synergistic effect: The combination produces a greater effect than expected (e.g., amoxicillin + clavulanic acid).

  • Antagonistic effect: One drug reduces or blocks the effect of another.

Several factors modify drug action, including age, sex, genetic variation, body weight, disease states, and concurrent medications. These variations explain why the same dose can yield different responses in different individuals.

Adverse Drug Reactions (ADRs)

Not all drug effects are beneficial. Adverse Drug Reactions refer to harmful or unintended responses occurring at normal doses. ADRs are classified as:

  • Type A (Augmented): Predictable, dose-dependent reactions like hypoglycemia from insulin.

  • Type B (Bizarre): Unpredictable reactions such as allergies or idiosyncrasies.

  • Type C (Chronic): Effects due to long-term use (e.g., corticosteroid-induced osteoporosis).

  • Type D (Delayed): Late-onset effects like carcinogenesis.

  • Type E (End-of-use): Withdrawal reactions upon sudden discontinuation.

Monitoring and reporting of ADRs form a vital part of pharmacovigilance — the science of detecting, assessing, and preventing drug-related problems.

Drug Interactions: Pharmacokinetic and Pharmacodynamic

Pharmacokinetic Interactions

These occur when one drug alters the absorption, distribution, metabolism, or excretion of another. For example, antacids can reduce absorption of tetracyclines, and enzyme inducers like rifampicin accelerate drug metabolism.

Pharmacodynamic Interactions

These involve direct or indirect effects at the receptor or cellular level. Combining two CNS depressants, for instance, can cause excessive sedation, while combining a β-blocker with insulin may mask hypoglycemia symptoms.

Drug Discovery and Clinical Evaluation

Developing a new drug is a long, meticulous journey involving several stages.

1. Drug Discovery Phase

Potential drug molecules are identified through screening, molecular modeling, and chemical synthesis. Natural products and genetic research often provide leads.

2. Preclinical Evaluation Phase

Candidate drugs undergo laboratory and animal testing to assess safety, toxicity, pharmacokinetics, and pharmacodynamics before human trials.

3. Clinical Trial Phase

Human testing is conducted in progressive stages:

  • Phase I: Safety and dosage studies in healthy volunteers.

  • Phase II: Efficacy and side effects in small patient groups.

  • Phase III: Large-scale trials to confirm safety and effectiveness.

  • Phase IV: Post-marketing surveillance to detect long-term effects.

4. Pharmacovigilance

Once marketed, continuous monitoring ensures detection of rare or delayed adverse effects. Pharmacovigilance strengthens public health by preventing recurrence of harmful drug events.

Leave a Comment

Your email address will not be published. Required fields are marked *

Scroll to Top