B Pharmacy Sem 5: Medicinal Chemistry II
Subject 1. Medicinal Chemistry II
1. Antihistaminic agents
2. Anti neoplastic agents
3. Anti anginal agents, Diuretics & Anti hypertensive agents
4. Anti arrhythmic drugs, Anti hyperlipidemic agents, Coagulants & Anticoagulants, Drugs used in Congestive Heart Failure
5. Drugs acting on Endocrine system (steroids, sex hormones, oral contraceptives, corticosteroids, thyroid/antithyroid)
6. Antidiabetic agents & Local Anesthetics
Unit 1: Antihistaminic Agents
This unit delves into the chemistry, pharmacology and therapeutic application of antihistamines—drugs that block histamine‑mediated responses primarily at the H₁ receptor. You’ll study histamine biosynthesis and storage, receptor subtypes and tissue distribution, the mechanism of action of H₁ antagonists, structure–activity relationships of first‑ and second‑generation agents, their pharmacokinetic profiles, clinical uses, and adverse effects.
1.1 Introduction to Histamine & Histaminergic Receptors
1.1.1 Histamine Biosynthesis & Storage
Histidine Decarboxylation: formation of histamine from L‑histidine by histidine decarboxylase in mast cells, basophils, and ECL cells.
Storage & Release: stored in granules; released upon IgE‑mediated degranulation or non‑immunologic triggers (physical, chemical stimuli).
1.1.2 Histamine Receptor Subtypes
H₁ Receptors: Gq‑coupled; mediates smooth muscle contraction, vascular permeability, bronchoconstriction; targets of classic antihistamines.
H₂ Receptors: Gs‑coupled; stimulates gastric acid secretion; antagonists used in ulcer disease.
H₃/H₄ Receptors: presynaptic autoreceptor (H₃) modulating neurotransmitter release; H₄ in immune cells—emerging drug targets.
1.2 Mechanism of Action of H₁‑Receptor Antagonists
Competitive Reversible Binding: most antihistamines bind the H₁ receptor without activating it, shifting dose–response curves to the right.
Inverse Agonism: certain agents stabilize the inactive receptor conformation, reducing basal activity.
Additional Effects: many first‑generation drugs cross the blood–brain barrier (sedation), block muscarinic/cholinergic, α‑adrenergic, or serotonergic receptors.
1.3 First‑Generation H₁ Antagonists
1.3.1 Chemical Classes & Representative Drugs
Ethanolamines: diphenhydramine, clemastine
Alkylamines: chlorpheniramine, brompheniramine
Piperazines: hydroxyzine, cyclizine
Phenothiazines (weak H₁ activity): promethazine
1.3.2 Pharmacokinetics & Adverse Effects
PK: rapid absorption; extensive hepatic metabolism (CYP450), variable half‑lives.
Side Effects: sedation, anticholinergic effects (dry mouth, urinary retention), hypotension, potential for overdose in children.
1.3.3 Structure–Activity Relationships
Basic Ethanolamine Skeleton: two aromatic rings separated by an ether linkage to an amine—bulk and lipophilicity drive CNS penetration.
Alkylamine Modification: para‑chlorination increases potency (chlorpheniramine).
1.4 Second‑Generation H₁ Antagonists
1.4.1 Key Drugs & Chemical Features
Loratadine, Desloratadine: tricyclic with polar side chains—minimal CNS entry.
Cetirizine, Levocetirizine: zwitterionic; low BBB penetration.
Fexofenadine: carboxylated metabolite of terfenadine.
1.4.2 Clinical Advantages
Selective Peripheral Action: reduced sedation and anticholinergic effects.
Longer Duration: once‑daily dosing, high compliance.
1.5 Therapeutic Uses & Adverse Reactions
Indications: allergic rhinitis, urticaria, conjunctivitis, pruritus, motion sickness (some first‑gen).
Adverse Effects:
First‑Generation: sedation, cognitive impairment, antimuscarinic symptoms.
Second‑Generation: headache, dry mouth, rarely QT prolongation (astemizole, terfenadine now withdrawn).
1.6 Key Points for Exams
Define histamine biosynthesis and receptor subtypes, and relate H₁ blockade to pharmacological effects.
Compare mechanism and SAR of first‑ versus second‑generation H₁ antagonists.
List common clinical uses and side‑effect profiles for representative antihistamines.
Explain the concept of inverse agonism at H₁ receptors and its therapeutic relevance.
Unit 2: Anti‑Neoplastic Agents
This unit covers the chemistry, mechanisms of action, pharmacokinetics, therapeutic applications, and safety profiles of major classes of anti‑cancer drugs. Emphasis is placed on how structural features relate to activity (SAR), dosing considerations, and common adverse effects.
2.1 Introduction to Anti‑Neoplastic Therapy
Anti‑neoplastic agents are drugs used to treat malignant tumors by interfering with cell division, DNA synthesis, or signaling pathways essential for cancer cell survival and proliferation. Key concepts include:
Cell Cycle Specificity: Agents that act in particular phases (e.g., S‑phase, M‑phase)
Cell Cycle Nonspecificity: Agents effective irrespective of the cell cycle stage
Therapeutic Index: Narrow—balancing tumor kill against toxicity to normal rapidly dividing cells
Combination Therapy: Employing drugs with different mechanisms to prevent resistance and achieve synergistic cell kill
2.2 Alkylating Agents
These cell cycle–nonspecific drugs form covalent bonds with DNA, leading to cross‑linking, strand breaks, and apoptosis.
2.2.1 Classes & Representative Drugs
Nitrogen Mustards: Cyclophosphamide, Ifosfamide
Nitrosoureas: Carmustine (BCNU), Lomustine (CCNU)
Platinum Compounds: Cisplatin, Carboplatin, Oxaliplatin
Others: Busulfan, Melphalan
2.2.2 Mechanism of Action
Generate electrophilic intermediates that alkylate N7 of guanine
Intrastrand and interstrand cross‑links prevent DNA replication/transcription
2.2.3 Pharmacokinetics & Activation
Cyclophosphamide: prodrug activated by hepatic CYP450 to 4‑hydroxycyclophosphamide
Platinum analogs: aquation in plasma yields active species
2.2.4 Therapeutic Uses
Cyclophosphamide: lymphomas, breast cancer, ovarian cancer
Cisplatin: testicular, ovarian, bladder, lung cancers
2.2.5 Adverse Effects
Myelosuppression (dose‑limiting)
Mucositis, alopecia
Nephrotoxicity (cisplatin), hemorrhagic cystitis (cyclophosphamide), pulmonary fibrosis (bleomycin)
2.3 Antimetabolites
These S‑phase–specific agents mimic natural substrates of DNA/RNA synthesis, causing faulty nucleic acids or inhibiting key enzymes.
2.3.1 Folate Analogues
Methotrexate: competitive DHFR inhibitor → ↓ tetrahydrofolate → impaired thymidine/ purine synthesis
2.3.2 Pyrimidine Analogues
5‑Fluorouracil (5‑FU): converted to FdUMP → thymidylate synthase inhibition
Cytarabine (Ara‑C): DNA chain termination
2.3.3 Purine Analogues
6‑Mercaptopurine (6‑MP): converted to TIMP → inhibits purine synthesis
Fludarabine, Cladribine: incorporate into DNA causing strand breaks
2.3.4 Therapeutic Uses
Methotrexate: leukemias, osteosarcoma, breast cancer
5‑FU: colorectal, breast, head & neck cancers
Cytarabine: acute myeloid leukemia
2.3.5 Adverse Effects
Mucositis, myelosuppression (methotrexate)
Hand–foot syndrome, diarrhea (5‑FU)
Neurotoxicity (fludarabine)
2.4 Plant‑Derived Natural Products
Primarily M‑phase–specific agents that target microtubules or topoisomerases.
2.4.1 Vinca Alkaloids
Vincristine, Vinblastine: bind tubulin → inhibit polymerization → metaphase arrest
2.4.2 Taxanes
Paclitaxel, Docetaxel: stabilize microtubules → prevent depolymerization
2.4.3 Camptothecins
Irinotecan, Topotecan: inhibit topoisomerase I → DNA single‑strand breaks
2.4.4 Therapeutic Uses
Vincristine: pediatric leukemias, lymphomas
Paclitaxel: ovarian, breast, lung cancers
Irinotecan: colorectal cancer
2.4.5 Adverse Effects
Neurotoxicity (vincristine), myelosuppression (vinblastine)
Peripheral neuropathy (taxanes)
Diarrhea (irinotecan), myelosuppression
2.5 Cytotoxic Antibiotics
Nonselective cell cycle–nonspecific agents that intercalate DNA or generate free radicals.
2.5.1 Anthracyclines
Doxorubicin, Daunorubicin: intercalation, topoisomerase II inhibition, free radical formation
2.5.2 Bleomycin
Binds DNA → strand scission via free radicals
2.5.3 Therapeutic Uses
Doxorubicin: breast, lymphomas, sarcomas
Bleomycin: Hodgkin’s lymphoma, germ‑cell tumors
2.5.4 Adverse Effects
Cardiotoxicity (anthracyclines; cumulative dose–dependent)
Pulmonary fibrosis (bleomycin)
2.6 Hormonal & Targeted Therapies
Agents that modulate hormone receptors or specific molecular targets in cancer cells.
2.6.1 Hormonal Agents
Tamoxifen: selective estrogen receptor modulator (SERM) in breast cancer
Anastrozole, Letrozole: aromatase inhibitors in postmenopausal breast cancer
Leuprolide: GnRH agonist for prostate cancer
2.6.2 Targeted Small Molecules & Biologics
Imatinib: BCR‑ABL tyrosine kinase inhibitor in CML
Trastuzumab: anti‑HER2 monoclonal antibody in HER2+ breast cancer
Bevacizumab: anti‑VEGF antibody inhibiting angiogenesis
2.6.3 Adverse Effects
Tamoxifen: risk of endometrial carcinoma, thromboembolism
Imatinib: fluid retention, cytopenias
Monoclonal antibodies: infusion reactions, hypertension (bevacizumab)
2.7 Key Points for Exams
Classify anti‑cancer drugs by mechanism (alkylators, antimetabolites, plant alkaloids, antibiotics, hormones, targeted).
Correlate drug structure/SAR with mechanism (e.g., nitrogen mustard bis‑chloroethyl groups, taxane core).
Describe cell cycle specificity and its relevance to scheduling/combination therapy.
List major toxicities and necessary protective measures (e.g., mesna for cyclophosphamide, dexrazoxane for doxorubicin).
Explain rationale behind targeted therapies and the importance of molecular diagnostics (e.g., BCR‑ABL fusion).
Unit 3: Anti‑Anginal Agents, Diuretics & Anti‑Hypertensive Agents
This comprehensive unit explores three interrelated drug categories crucial for cardiovascular therapy: anti‑anginal drugs (relieving myocardial ischemia), diuretics (modulating fluid balance), and antihypertensives (lowering blood pressure). Emphasis is placed on mechanisms of action, structure–activity relationships, pharmacokinetics, therapeutic applications, and safety considerations.
3.1 Anti‑Anginal Agents
Anti‑anginal drugs improve myocardial oxygen supply–demand balance via vasodilation, reduced preload/afterload, or decreased heart rate and contractility.
3.1.1 Nitrates
Mechanism: Prodrugs converted to NO → ↑ cGMP in vascular smooth muscle → venous (and at high doses, arterial) dilation.
Drugs & SAR:
Glyceryl Trinitrate (GTN): short‑acting, sublingual.
Isosorbide Dinitrate/Mononitrate: longer‑acting; mononitrate more bioavailable.
Ester groups at positions 1,2,3 of glycerol backbone determine onset/duration.
PK: Extensive first‑pass (GTN); mononitrate oral bioavailability ~100%.
Clinical Uses: Acute angina attacks (GTN), prophylaxis (isosorbides).
Adverse Effects: Headache, orthostatic hypotension, reflex tachycardia, tolerance (requires nitrate‑free interval).
3.1.2 β‑Adrenergic Blockers
Mechanism: ↓ heart rate, contractility → ↓ O₂ demand; some (e.g., propranolol) also reduce BP.
Classes:
Nonselective: Propranolol, Nadolol.
β₁‑Selective: Metoprolol, Atenolol.
With ISA: Pindolol (partial agonists).
SAR: Aryloxypropanolamine core; para‑substituents modulate selectivity.
PK: Variable lipophilicity; lipophilic (propranolol) crosses BBB.
Uses: Chronic stable angina, silent ischemia.
Adverse Effects: Bradycardia, fatigue, bronchospasm (nonselective), erectile dysfunction.
3.1.3 Calcium Channel Blockers (CCBs)
Mechanism: Inhibit L‑type Ca²⁺ channels in vascular smooth muscle and myocardium → ↓ afterload and contractility.
Classes & SAR:
Dihydropyridines: Nifedipine, Amlodipine (vascular selectivity; bulky aryl groups at C4).
Phenylalkylamines: Verapamil (cardioselective).
Benzothiazepines: Diltiazem (intermediate).
PK: High first‑pass; long‑acting preparations available.
Uses: Variant (Prinzmetal’s) angina (CCBs preferred), stable angina adjunct.
Adverse Effects: Dihydropyridines—tachycardia, peripheral edema; verapamil—constipation, heart block; diltiazem—bradycardia.
3.2 Diuretics
Diuretics increase renal excretion of salt and water, reducing blood volume and preload, with secondary effects on vascular tone and BP.
3.2.1 Thiazide & Thiazide‑Like Diuretics
Mechanism: Inhibit Na⁺/Cl⁻ cotransporter (DCT) → modest natriuresis.
Drugs: Hydrochlorothiazide, Chlorthalidone, Indapamide.
SAR: Benzothiadiazine ring; sulfonamide moiety essential for activity.
PK: Onset 2 hrs, duration 12–24 hrs.
Uses: Hypertension (first‑line), mild edema.
Adverse Effects: Hypokalemia, hyperuricemia, hyperglycemia, hyponatremia.
3.2.2 Loop Diuretics
Mechanism: Inhibit Na⁺/K⁺/2Cl⁻ cotransporter (thick ascending limb) → potent diuresis.
Drugs: Furosemide, Bumetanide, Torsemide.
SAR: Sulfonamide or sulfamoyl group plus phenoxyacetic acid (furosemide).
PK: Rapid onset (30 min), duration 6 hrs.
Uses: Acute pulmonary edema, severe edema, resistant hypertension.
Adverse Effects: Hypokalemia, ototoxicity, dehydration, metabolic alkalosis.
3.2.3 Potassium‑Sparing Diuretics
Mechanism & Drugs:
Aldosterone Antagonists: Spironolactone, Eplerenone.
Epithelial Na⁺ Channel Blockers: Amiloride, Triamterene.
PK: Spironolactone prodrug; active metabolites long‑lasting.
Uses: Combined with thiazides/loops to prevent K⁺ loss; heart failure (spironolactone).
Adverse Effects: Hyperkalemia, gynecomastia (spironolactone).
3.3 Antihypertensive Agents
A broad array of drugs targeting different pathways to lower systemic vascular resistance and/or cardiac output.
3.3.1 ACE Inhibitors
Mechanism: Block conversion of angiotensin I → II; ↓ aldosterone, ↓ vasoconstriction.
Drugs & SAR: Captopril (thiol), Enalapril (ester prodrug), Lisinopril (active peptide).
Uses: Hypertension, heart failure, diabetic nephropathy.
Adverse Effects: Cough (bradykinin), angioedema, hyperkalemia.
3.3.2 Angiotensin II Receptor Blockers (ARBs)
Mechanism: Competitive antagonism at AT₁ receptor.
Drugs: Losartan, Valsartan, Telmisartan.
SAR: Biphenyl tetrazole structure; acidic tetrazole mimics carboxylate.
Uses: Similar to ACE inhibitors, useful if cough intolerable.
Adverse Effects: Hyperkalemia, rare angioedema.
3.3.3 Direct Renin Inhibitor
Drug: Aliskiren.
Mechanism: Binds renin active site → ↓ angiotensin I generation.
Uses: Hypertension; limited by side‑effect profile.
Adverse Effects: Diarrhea, hyperkalemia, hypotension.
3.3.4 α‑Blockers
Mechanism: α₁‑receptor antagonism → vasodilation.
Drugs: Prazosin, Doxazosin.
Uses: Hypertension with benign prostatic hyperplasia (BPH).
Adverse Effects: Orthostatic hypotension, reflex tachycardia.
3.3.5 Central Sympatholytics
Mechanism: ↓ central sympathetic outflow (α₂ agonism or imidazoline).
Drugs: Clonidine, Methyldopa, Moxonidine.
Uses: Resistant hypertension; methyldopa in pregnancy.
Adverse Effects: Sedation, dry mouth, rebound hypertension on withdrawal.
Key Points for Exams
Classify and contrast anti‑anginal mechanisms (nitrate vs. β‑blocker vs. CCB).
Relate diuretic site of action in nephron to efficacy and electrolyte changes.
Understand RAAS inhibition steps (renin, ACE, AT₁ receptor) and clinical implications.
Memorize major adverse effect profiles (tolerance with nitrates, cough with ACE‑I, hypokalemia with thiazides, hyperkalemia with K⁺‑sparing).
Integrate combined regimens (e.g., ACE‑I + thiazide; β‑blocker + nitrate) for optimized cardiovascular care.
Unit 5: Drugs Acting on the Endocrine System
This unit examines hormone‑based therapies—sex steroids, contraceptives, corticosteroids, and thyroid agents—focusing on their chemistry, mechanisms, pharmacokinetics, clinical uses, and safety.
5.1 Sex Steroids
Hormones that regulate reproductive physiology via intracellular nuclear receptors.
5.1.1 Estrogens
Key Drugs: 17β‑Estradiol, Ethinyl Estradiol, Conjugated Equine Estrogens
Mechanism: Bind estrogen receptors (ERα/ERβ) → receptor dimerization → gene transcription (estrogen‑responsive elements)
SAR Highlights: Aromatic A‑ring; 17α‑ethinyl group (↑ oral bioavailability); C3 hydroxyl essential for receptor binding
PK: Extensive first‑pass metabolism; ethinyl substitution prolongs half‑life; available oral, transdermal, vaginal
Uses: Hormone replacement therapy (HRT), menopausal symptom relief, osteoporosis prevention, dysmenorrhea
Adverse Effects: Thromboembolism, endometrial hyperplasia, breast tenderness, nausea
5.1.2 Progestins
Key Drugs: Progesterone, Medroxyprogesterone Acetate, Levonorgestrel, Norethindrone
Mechanism: Bind progesterone receptor → regulate genes for endometrial secretory changes
SAR Highlights: Removal of C19 methyl (19‑nor) increases progestational activity; 17α‑acetate or 17α‑alkyl enhances duration
PK: Variable oral bioavailability; depot injectable formulations (e.g., medroxyprogesterone)
Uses: HRT (opposed cycles), dysfunctional uterine bleeding, part of combination contraceptives
Adverse Effects: Weight gain, mood changes, spotting, decreased bone density (depot forms)
5.1.3 Androgens & Anabolics
Key Drugs: Testosterone, Methyltestosterone, Oxandrolone
Mechanism: Bind androgen receptor → ↑ protein synthesis, male secondary sexual characteristics
SAR Highlights: 17α‑alkylation (methyltestosterone) for oral activity; esterification (testosterone enanthate) for depot use
PK: Oral (hepatotoxic risk), injectable esters, transdermal patches/gels
Uses: Hypogonadism, delayed puberty, muscle wasting syndromes
Adverse Effects: Virilization, acne, hepatic dysfunction, dyslipidemia
5.2 Oral Contraceptives
Combination and progestin‑only pills that prevent pregnancy by endocrine feedback and cervical mucus alteration.
5.2.1 Combination Pills (Estrogen + Progestin)
Formulations: Monophasic, Biphasic, Triphasic dosing to mimic physiologic cycles
Mechanisms:
Suppress hypothalamic GnRH → ↓ FSH/LH → inhibit ovulation
Thicken cervical mucus → resist sperm penetration
Induce endometrial atrophy → prevent implantation
Adverse Effects: Breakthrough bleeding, nausea, headache, ↑ thromboembolic risk
5.2.2 Progestin‑Only Pills (“Mini‑Pill”)
Key Drugs: Norethindrone, Desogestrel
Mechanism: Thickens cervical mucus; less reliably ovulation suppression
Uses: When estrogen contraindicated (e.g., breastfeeding, thrombotic risk)
Adverse Effects: Irregular bleeding, headache, breast tenderness
5.3 Corticosteroids
Mimic adrenal cortex hormones; divided into glucocorticoids (anti‑inflammatory) and mineralocorticoids (electrolyte balance).
5.3.1 Glucocorticoids
Key Drugs: Hydrocortisone, Prednisone, Methylprednisolone, Dexamethasone
Mechanism: Bind glucocorticoid receptor → translocate to nucleus → modulate pro‑ and anti‑inflammatory gene expression
SAR Highlights: 11β‑hydroxyl for activity; 1‑dehydro (prednisone) ↑ potency; 9α‑fluoro (dexamethasone) ↑ anti‑inflammatory vs. mineralocorticoid ratio
PK: Oral, IV, IM, inhaled, topical; variable half‑lives (dexamethasone longest)
Uses: Asthma/COPD exacerbations, autoimmune diseases, transplant rejection prophylaxis
Adverse Effects: Cushingoid features, osteoporosis, adrenal suppression, hyperglycemia, immunosuppression
5.3.2 Mineralocorticoids
Key Drug: Fludrocortisone
Mechanism: Bind mineralocorticoid receptor → ↑ Na⁺ reabsorption and K⁺ excretion in renal distal tubule
Uses: Primary/secondary adrenal insufficiency (Addison’s disease)
Adverse Effects: Hypertension, hypokalemia, edema
5.4 Thyroid & Antithyroid Agents
Regulate thyroid hormone synthesis, storage, and action.
5.4.1 Thyroid Hormones
Key Drugs: Levothyroxine (T₄), Liothyronine (T₃)
Mechanism: T₄ converted to T₃ in peripheral tissues; T₃ binds nuclear thyroid hormone receptors → regulate metabolic gene transcription
PK: Oral levothyroxine half‑life ~7 days; narrow therapeutic index; affected by food, absorption interactions
Uses: Hypothyroidism, myxedema coma (IV T₃/T₄)
Adverse Effects: Signs of hyperthyroidism (tachycardia, weight loss, tremor) if overdosed
5.4.2 Antithyroid Drugs
Thioamides: Methimazole, Propylthiouracil (PTU)
Mechanism: Inhibit thyroid peroxidase → block iodination and coupling of tyrosines; PTU also inhibits peripheral T₄→T₃ conversion
PK: Oral; PTU shorter half‑life, requires multiple daily doses
Uses: Graves’ disease, preoperative preparation for thyroidectomy
Adverse Effects: Agranulocytosis, hepatotoxicity (PTU), rash
Inorganic Iodides: Lugol’s solution, Potassium iodide
Mechanism: Wolff‑Chaikoff effect → transient ↓ hormone release; also reduce gland vascularity pre‑surgery
Uses: Thyrotoxic crisis, preoperative prep
Radioactive Iodine (¹³¹I)
Mechanism: β‑emission → follicular cell destruction
Uses: Definitive therapy for hyperthyroidism
Adverse Effects: Hypothyroidism (common), radiation thyroiditis
Key Points for Exams
Estrogens/Progestins: know receptor signalling, SAR modifications for oral activity, and thrombotic risks.
Contraceptives: mechanisms beyond ovulation suppression; monophasic vs. multiphasic formulations.
Glucocorticoids vs. Mineralocorticoids: structural changes that alter potency and specificity; major side‑effect profiles.
Thyroid Therapy: conversion of T₄→T₃, interactions affecting levothyroxine absorption; antithyroid drug monitoring (CBC, LFTs).
Adverse Effects: recognize Cushingoid features, hyperglycemia, osteoporosis, agranulocytosis, and thyroid storm management.
Unit 6: Antidiabetic Agents & Local Anesthetics
6.1 Antidiabetic Agents
6.1.1 Insulin & Insulin Analogues
Definition: Exogenous preparations of human insulin or its molecular variants designed to replace or supplement endogenous insulin in diabetes.
Key Points:
Mechanism: Binds the insulin receptor tyrosine kinase → promotes glucose uptake into muscle and fat (GLUT4 translocation), inhibits hepatic gluconeogenesis, and stimulates glycogen/lipid synthesis.
Types & Profiles:
Rapid‑Acting (Lispro, Aspart, Glulisine): Onset 10–20 min; duration 3–5 h. Ideal for covering mealtime glucose spikes.
Short‑Acting (Regular): Onset 30–60 min; duration 6–8 h. Requires injection 30 min before meals.
Intermediate‑Acting (NPH): Onset 1–2 h; duration 12–18 h. Provides basal insulin when dosed twice daily.
Long‑Acting (Glargine, Detemir): Onset ~1 h; duration up to 24 h. Mimics basal insulin secretion with once‑daily dosing.
Clinical Use: Type 1 diabetes (basal–bolus regimens), advanced type 2 diabetes.
Adverse Effects: Hypoglycemia (most common), weight gain, lipodystrophy at injection sites.
6.1.2 Sulfonylureas & Meglitinides
Definition: Oral insulin secretagogues that close pancreatic β‑cell K<sub>ATP</sub> channels to trigger insulin release.
Key Points:
Mechanism (Shared): Block ATP‑sensitive K⁺ channels on β‑cells → membrane depolarization → Ca²⁺ influx → insulin exocytosis.
Sulfonylureas (e.g., Glibenclamide, Glipizide, Glimepiride):
Duration: Long‑acting; typically once‑daily dosing.
Use: First‐ or second‐line add‑on in type 2 diabetes with residual β‑cell function.
Risks: Prolonged hypoglycemia (especially in the elderly), weight gain, possible secondary failure over time.
Meglitinides (e.g., Repaglinide, Nateglinide):
Duration: Very short; peak effect within 1 h, duration 4 h.
Use: Target postprandial glucose; administer immediately before meals.
Risks: Hypoglycemia (less severe than sulfonylureas), modest weight gain.
6.1.3 Biguanides
Metformin
Definition: A first‑line oral agent that reduces hepatic glucose production and improves peripheral insulin sensitivity.
Mechanism: Activates AMP‑activated protein kinase (AMPK) → inhibits gluconeogenic enzymes, increases GLUT4‑mediated glucose uptake in muscle.
Benefits: Lowers A1C by ~1–2%; weight neutral or modest loss; cardiovascular protection.
Adverse Effects: Gastrointestinal upset (nausea, diarrhea), rare lactic acidosis (contraindicated in renal impairment).
6.1.4 Thiazolidinediones (Glitazones)
Pioglitazone, Rosiglitazone
Definition: Insulin sensitizers that bind PPARγ nuclear receptors to modulate gene transcription in adipose tissue.
Mechanism: ↑ adipocyte differentiation, ↑ GLUT4 expression, redistribute lipids away from muscle/liver.
Benefits: Improve insulin sensitivity; durable glycemic control.
Risks: Weight gain, fluid retention/edema, risk of heart failure exacerbation, increased fracture risk.
6.1.5 α‑Glucosidase Inhibitors
Acarbose, Miglitol
Definition: Oral agents that delay carbohydrate absorption by inhibiting brush‑border α‑glucosidases in the small intestine.
Mechanism: Slows breakdown of complex carbohydrates → blunts postprandial glucose rise.
Adverse Effects: Flatulence, bloating, diarrhea due to undigested sugars in colon.
6.1.6 Incretin‑Based Therapies
DPP‑4 Inhibitors (Sitagliptin, Saxagliptin):
Definition: Oral drugs that prevent degradation of GLP‑1 and GIP.
Mechanism: ↑ endogenous incretin levels → glucose‑dependent insulin secretion, ↓ glucagon.
Adverse Effects: Nasopharyngitis, headache; rare pancreatitis.
GLP‑1 Receptor Agonists (Exenatide, Liraglutide):
Definition: Injectable peptides that mimic GLP‑1.
Benefits: Enhance insulin release, suppress glucagon, slow gastric emptying, promote satiety and weight loss.
Adverse Effects: Nausea, risk of pancreatitis.
6.1.7 SGLT2 Inhibitors
Dapagliflozin, Empagliflozin
Definition: Oral agents that lower renal glucose reabsorption.
Mechanism: Inhibit sodium‑glucose cotransporter 2 in proximal tubule → increased urinary glucose excretion.
Benefits: Weight loss, blood pressure reduction, proven cardiovascular and renal protection.
Adverse Effects: Genitourinary infections, volume depletion, rare euglycemic ketoacidosis.
6.2 Local Anesthetics
6.2.1 Chemical Classification
Esters (Procaine, Tetracaine, Benzocaine):
Metabolized rapidly by plasma pseudocholinesterases → PABA byproducts (allergenic potential).
Amides (Lidocaine, Bupivacaine, Mepivacaine, Prilocaine):
Metabolized hepatically by CYP450 → more stable; low allergy risk.
6.2.2 Mechanism of Action & SAR
Mechanism: Uncharged form diffuses into nerve axon → protonation→ cation binds intracellular domain of voltage‑gated Na⁺ channels → prevents Na⁺ influx → no action potential propagation.
SAR Elements:
Aromatic Ring: Lipophilicity → potency & duration.
Linkage (Ester vs. Amide): Metabolic stability and allergenicity.
Tertiary Amine: pKa determines fraction of unionized drug at physiologic pH → onset speed.
6.2.3 Pharmacokinetics & Administration
Onset: Faster when pKa ≈ physiological pH (7.4).
Duration: Proportional to lipid solubility & protein binding.
Epinephrine Co‑administration: Vasoconstricts local vessels → prolongs effect, reduces systemic absorption.
Routes: Topical, infiltration, peripheral nerve blocks, epidural/spinal anesthesia.
6.2.4 Toxicity & Management
Local Reactions: Tissue irritation; transient neurologic symptoms (TNS) post‐spinal lidocaine.
Systemic Toxicity:
CNS: Initial excitation (tremors, seizures) → depression (coma, respiratory arrest).
Cardiovascular: Hypotension, arrhythmias, cardiac arrest.
Treatment: Supportive care; intravenous lipid emulsion therapy in severe cardiotoxicity.
Key Takeaways for Exams
Antidiabetics: Match each class to its mechanism, dosing profile, and principal adverse effects.
Insulin Regimens: Understand onset/duration curves for basal–bolus planning.
Local Anesthetics: Differentiate esters vs. amides by metabolism, allergy risk, and relate pKa/lipophilicity to clinical onset/duration.
Toxicity Management: Recognize hypoglycemia signs and local anesthetic systemic toxicity protocols.