Chapter Overview
Pharmacology, the science of drugs and their interactions with living systems, forms the scientific foundation of rational therapeutics. It encompasses the investigation of the physical and chemical properties of drugs, their biochemical and physiological effects, mechanisms of action, absorption, distribution, metabolism, excretion, and therapeutic and toxic effects. The word pharmacology is derived from the Greek words "pharmakon" meaning drug or poison, and "logos" meaning study.
Definition and Scope of Pharmacology
What is Pharmacology?
Pharmacology is broadly defined as the science of drugs, encompassing their origin, composition, pharmacokinetics, pharmacodynamics, therapeutic uses, and toxicology. According to Rang et al., pharmacology can be defined as "the science of what drugs do to the body and what the body does to drugs" (1). This bidirectional relationship forms the cornerstone of modern pharmacological understanding. Brunton and colleagues in Goodman & Gilman's seminal text expand this definition to include "the science of substances used to prevent, diagnose, and treat disease" (2).
The term pharmacology derives from the Greek words pharmakon (drug, medicine, or poison) and logos (study). This etymological origin highlights the dual nature of drugs as both therapeutic agents and potential toxins, emphasizing Paracelsus's fundamental principle that "the dose makes the poison" (3).
Pharmacology differs from pharmacy, which focuses on the preparation, dispensing, and appropriate use of medications. While pharmacy emphasizes the practical aspects of medication management, pharmacology provides the scientific foundation that explains drug action at molecular, cellular, and systemic levels (4). Similarly, pharmacology is distinct from therapeutics, which represents the art and science of choosing and administering appropriate treatments for specific patients and conditions.
Scope and Integration with Other Sciences
Modern pharmacology is inherently interdisciplinary, drawing from and contributing to multiple scientific fields. As described by Katzung, pharmacology serves as "the bridge between the basic biomedical sciences and clinical medicine" (3). This integration encompasses:
Molecular biology and biochemistry provide insights into drug targets, including receptors, enzymes, ion channels, and transporters. Understanding protein structure-function relationships enables rational drug design and explains mechanisms of drug action at the molecular level (5).
Physiology and pathophysiology inform how drugs modify normal and abnormal body functions. Pharmacological interventions often aim to restore physiological homeostasis disrupted by disease processes. For instance, understanding the renin-angiotensin-aldosterone system is essential for comprehending how ACE inhibitors treat hypertension and heart failure (6).
Genetics and genomics increasingly influence pharmacological practice through pharmacogenetics and pharmacogenomics. These fields explain individual variations in drug response and enable personalized medicine approaches. The Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines now inform dosing decisions for over 20 medications based on genetic markers (7).
Historical Development of Pharmacology
Ancient Foundations and Empirical Observations
The use of natural substances for medicinal purposes predates written history. Archaeological evidence suggests that Neanderthals used medicinal plants as early as 60,000 years ago. The Ebers Papyrus (circa 1550 BCE) from ancient Egypt contains descriptions of over 700 remedies, including opium for pain relief and castor oil as a laxative (8).
Ancient civilizations developed sophisticated pharmacopoeias through empirical observation. The Pen Ts'ao, attributed to Chinese Emperor Shen Nung (circa 2700 BCE), describes 365 drugs. Indian Ayurvedic medicine, documented in the Charaka Samhita and Sushruta Samhita (circa 1000 BCE), details numerous plant-based remedies. Greek physician Dioscorides's De Materia Medica (circa 60 CE) remained the authoritative pharmacological text in Europe for over 1,500 years (9).
The Birth of Scientific Pharmacology
The transformation of pharmacology from empirical practice to experimental science began in the 19th century. François Magendie (1783-1855) pioneered experimental pharmacology by demonstrating that strychnine acts on the spinal cord. His student, Claude Bernard (1813-1878), established fundamental principles of experimental medicine and showed that curare blocks neuromuscular transmission (10).
The isolation of pure active compounds marked a crucial advancement. Friedrich Sertürner isolated morphine from opium in 1805, demonstrating that specific chemicals were responsible for drug effects. This discovery initiated the era of alkaloid chemistry, leading to isolation of quinine (1820), atropine (1833), and cocaine (1859) (11).
Modern Era and Drug Development
Paul Ehrlich (1854-1915) introduced the concept of selective toxicity with his "magic bullet" theory, proposing that drugs could specifically target pathogens without harming host tissues. His development of arsphenamine (Salvarsan) for syphilis in 1909 marked the beginning of chemotherapy (12).
The 20th century witnessed revolutionary discoveries:
- Alexander Fleming's discovery of penicillin (1928) initiated the antibiotic era
- Gerhard Domagk's sulfonamides (1935) provided the first effective antibacterial chemotherapy
- The development of general anesthetics transformed surgery
- Discovery of neurotransmitters led to psychopharmacology
- Molecular biology enabled targeted therapy and biologics (13)
The Human Genome Project's completion in 2003 ushered in the genomic era of pharmacology, enabling precision medicine approaches based on individual genetic profiles (14).
Major Subdivisions of Pharmacology
Pharmacokinetics: What the Body Does to the Drug
Pharmacokinetics describes the time course of drug absorption, distribution, metabolism, and excretion (ADME). Rowland and Tozer define pharmacokinetics as "what the body does to the drug" encompassing all processes that determine drug concentration at the site of action over time (15).
The four fundamental pharmacokinetic processes are:
Absorption involves drug movement from the site of administration into systemic circulation. Factors affecting absorption include drug formulation, route of administration, drug solubility and stability, and physiological factors such as gastric pH and intestinal motility. Bioavailability (F) represents the fraction of administered dose reaching systemic circulation unchanged (16).
Distribution describes drug movement from systemic circulation to tissues. The extent of distribution depends on drug properties (lipophilicity, molecular size, protein binding) and physiological factors (tissue perfusion, membrane permeability, active transport). The volume of distribution (Vd) relates the amount of drug in the body to plasma concentration (17).
Metabolism (biotransformation) involves chemical modification of drugs, primarily in the liver. Phase I reactions (oxidation, reduction, hydrolysis) often involve cytochrome P450 (CYP) enzymes. Phase II reactions (conjugation with glucuronic acid, sulfate, amino acids) generally increase water solubility for excretion. Genetic polymorphisms in metabolizing enzymes contribute to interindividual variability (18).
Excretion removes drugs and metabolites from the body, primarily via kidneys (glomerular filtration, tubular secretion, reabsorption) and bile. Clearance (CL) quantifies the body's efficiency in drug elimination, while half-life (t½) determines dosing frequency for maintaining therapeutic concentrations (19).
Pharmacodynamics: What the Drug Does to the Body
Pharmacodynamics encompasses drug effects on the body and their mechanisms of action. As defined by Lippincott's Illustrated Reviews, pharmacodynamics describes "the quantitative relationship between drug concentration and pharmacologic effect" (20).
Key pharmacodynamic principles include:
Drug-receptor interactions form the basis of most drug actions. Drugs act as ligands binding to four main receptor superfamilies: ligand-gated ion channels, G-protein-coupled receptors (GPCRs), enzyme-linked receptors, and nuclear receptors. The interaction is characterized by affinity (tendency to bind) and efficacy (ability to activate) (21).
Dose-response relationships quantify the relationship between drug dose/concentration and magnitude of response. Graded dose-response curves show continuous variation in response intensity, while quantal dose-response curves depict population responses to increasing doses. These relationships define important parameters: EC50 (concentration producing 50% maximal effect), therapeutic index (TD50/ED50), and margin of safety (22).
Drug interactions at receptors include:
- Agonists: activate receptors (full agonists produce maximal response; partial agonists produce submaximal response)
- Antagonists: block receptor activation (competitive antagonists can be overcome by increasing agonist concentration; non-competitive antagonists cannot)
- Inverse agonists: reduce constitutive receptor activity (23)
Clinical Pharmacology
Clinical pharmacology applies pharmacological principles to patient care, bridging basic science and clinical practice. Bennett and Brown define clinical pharmacology as "the scientific study of drugs in humans" encompassing both therapeutic and adverse effects (24).
Clinical pharmacology encompasses:
- Rational drug selection and prescribing
- Therapeutic drug monitoring
- Management of drug interactions
- Adverse drug reaction assessment
- Pharmacovigilance
- Clinical trials design and interpretation
- Formulary management
- Medication safety initiatives (25)
Toxicology
Toxicology, the study of adverse effects of chemicals on living organisms, overlaps significantly with pharmacology. Casarett and Doull's text defines toxicology as "the study of the adverse effects of xenobiotics" where xenobiotics include drugs, environmental chemicals, and natural toxins (26).
Toxicological considerations include:
- Acute and chronic toxicity
- Organ-specific toxicity
- Carcinogenicity, mutagenicity, teratogenicity
- Risk assessment and management
- Antidote development
- Forensic applications (27)
Drug Classification and Nomenclature
Classification Systems
Drugs can be classified by multiple systems, each serving different purposes:
Therapeutic classification groups drugs by clinical use (antihypertensives, antibiotics, analgesics). This system, used in the British National Formulary (BNF), facilitates clinical decision-making but may group drugs with different mechanisms together (28).
Pharmacological classification categorizes drugs by mechanism of action (β-blockers, ACE inhibitors, calcium channel blockers). This system, emphasized in Katzung's Basic & Clinical Pharmacology, aids understanding of drug effects and interactions (3).
Chemical classification groups drugs by structure (benzodiazepines, phenothiazines, β-lactams). Structural similarities often predict similar pharmacological properties and cross-reactivity patterns (29).
The Anatomical Therapeutic Chemical (ATC) classification, maintained by WHO, combines anatomical, therapeutic, pharmacological, and chemical criteria in a hierarchical system. This standardized system facilitates international drug utilization studies (30).
Drug Nomenclature
Each drug has multiple names serving different purposes:
Chemical name describes molecular structure using IUPAC nomenclature (e.g., N-acetyl-para-aminophenol). While precise, chemical names are impractical for clinical use (31).
Generic (nonproprietary) name is the official name assigned by nomenclature committees (International Nonproprietary Names by WHO, United States Adopted Names by USAN Council). Generic names are designed to be concise and indicate pharmacological class through standardized stems (e.g., -olol for β-blockers, -pril for ACE inhibitors) (32).
Trade (proprietary/brand) name is the manufacturer's registered trademark. Multiple trade names may exist for the same drug, potentially causing confusion. The Institute for Safe Medication Practices (ISMP) maintains lists of confused drug names to prevent medication errors (33).
Sources of Drugs
Natural Sources
Despite advances in synthetic chemistry, nature remains a prolific source of therapeutic agents. Newman and Cragg's analysis revealed that 50% of approved drugs from 1981-2019 were natural products or derivatives (34).
Plant sources have provided numerous essential medicines:
- Alkaloids: morphine (Papaver somniferum), vincristine (Catharanthus roseus), paclitaxel (Taxus brevifolia)
- Glycosides: digoxin (Digitalis lanata), aspirin precursor salicin (Salix species)
- Terpenoids: artemisinin (Artemisia annua) for malaria (35)
Microbial sources revolutionized medicine:
- Antibiotics: penicillins (Penicillium), streptomycin (Streptomyces), cephalosporins (Acremonium)
- Immunosuppressants: cyclosporine (Tolypocladium inflatum), tacrolimus (Streptomyces tsukubaensis)
- Statins: lovastatin (Aspergillus terreus) (36)
Animal sources provided early protein therapeutics:
- Insulin from porcine/bovine pancreas (before recombinant production)
- Heparin from porcine intestinal mucosa or bovine lung
- Thyroid hormone from desiccated thyroid glands (37)
Marine sources offer unique chemical diversity:
- Ziconotide from cone snail venom for severe pain
- Cytarabine analog from Caribbean sponge for leukemia
- Omega-3 fatty acids from fish oils (38)
Synthetic and Semi-synthetic Drugs
Total synthesis allows production of drugs not found in nature and enables structure-activity relationship studies. Examples include:
- Benzodiazepines for anxiety
- Proton pump inhibitors for acid suppression
- Fluoroquinolone antibiotics (39)
Semi-synthetic modification improves upon natural products:
- Amoxicillin from penicillin (broader spectrum, oral bioavailability)
- Heroin from morphine (historically; now illegal)
- Simvastatin from lovastatin (increased potency) (40)
Biotechnology-Derived Drugs
Recombinant DNA technology enables production of human proteins and antibodies:
- Recombinant proteins: insulin, growth hormone, erythropoietin, interferons
- Monoclonal antibodies: therapeutic antibodies ending in -mab constitute the fastest-growing drug class
- Gene and cell therapies: CAR-T cells, gene replacement vectors
- Vaccines: mRNA vaccines for COVID-19 demonstrated rapid pandemic response capability (41)
Drug Development and Regulation
Drug Discovery and Development Pipeline
The journey from drug discovery to market approval typically spans 10-15 years and costs over $1 billion. DiMasi et al. estimated the average cost of developing an approved drug at $2.87 billion in 2013 dollars (42).
Discovery phase (2-4 years):
- Target identification and validation
- High-throughput screening or rational drug design
- Lead compound identification and optimization
- Preclinical pharmacology and toxicology studies (43)
Preclinical development (1-2 years):
- Animal pharmacokinetics and pharmacodynamics
- Acute and chronic toxicity studies
- Reproductive toxicity and carcinogenicity assessment
- Formulation development
- IND (Investigational New Drug) application preparation (44)
Clinical development follows ICH (International Council for Harmonisation) guidelines:
Phase I (1-2 years, 20-100 subjects): First-in-human studies establish safety, tolerability, pharmacokinetics, and preliminary pharmacodynamics in healthy volunteers or patients (45).
Phase II (2-3 years, 100-300 patients): Proof-of-concept studies evaluate efficacy and dose-response relationships while continuing safety assessment. Phase IIa studies focus on dosing; Phase IIb evaluates efficacy (46).
Phase III (3-4 years, 300-3,000 patients): Pivotal trials compare investigational drug to standard therapy or placebo. These multicenter, randomized controlled trials provide definitive evidence of efficacy and safety for regulatory approval (47).
Regulatory review (1-2 years): Comprehensive data package submitted as NDA (New Drug Application) to FDA or MAA (Marketing Authorization Application) to EMA. Review includes benefit-risk assessment, labeling negotiations, and potential advisory committee evaluation (48).
Phase IV (post-marketing surveillance): Ongoing monitoring detects rare adverse events, evaluates long-term effectiveness, and identifies new indications. Pharmacovigilance systems include spontaneous reporting (FDA's FAERS, WHO's VigiBase) and active surveillance programs (49).
Fundamental Principles of Drug Action
Mechanisms of Drug Action
Drugs produce therapeutic effects through various molecular mechanisms. Golan et al. categorize these into four fundamental mechanisms (50):
Receptor interactions represent the most common mechanism. Drugs act as ligands for physiological receptors, either mimicking (agonists) or blocking (antagonists) endogenous ligands. Examples include β-adrenergic agonists for asthma and H2-receptor antagonists for peptic ulcers (51).
Enzyme interactions involve drugs that inhibit or, less commonly, activate enzymes. Competitive inhibitors (statins inhibiting HMG-CoA reductase), non-competitive inhibitors (aspirin irreversibly inhibiting COX), and allosteric modulators (benzodiazepines enhancing GABA receptor function) exemplify this mechanism (52).
Transporter interactions affect movement of endogenous substances across membranes. SSRIs (selective serotonin reuptake inhibitors) block serotonin transporters, increasing synaptic serotonin. Proton pump inhibitors irreversibly inhibit H+/K+-ATPase in gastric parietal cells (53).
Direct chemical interactions don't involve specific macromolecular targets. Antacids neutralize gastric acid through simple acid-base chemistry. Chelating agents like penicillamine bind metal ions in heavy metal poisoning (54).
Selectivity and Specificity
Selectivity refers to a drug's ability to produce a particular effect relative to other effects. No drug is completely specific; all produce multiple effects at different concentrations. The therapeutic window represents the concentration range producing desired effects without unacceptable toxicity (55).
Factors affecting selectivity include:
- Receptor subtype selectivity (β1 vs β2 adrenergic receptors)
- Tissue distribution (statins concentrate in liver)
- Local administration (inhaled corticosteroids for asthma)
- Prodrug activation at target site (56)
Individual Variation in Drug Response
Pharmacogenetics and Pharmacogenomics
Genetic factors account for 20-95% of variability in drug response. Pharmacogenetics studies how single genes affect drug response, while pharmacogenomics examines genome-wide influences (57).
Pharmacokinetic variations:
- CYP2D6 polymorphisms affect metabolism of 25% of drugs (poor, intermediate, extensive, ultra-rapid metabolizers)
- TPMT deficiency increases 6-mercaptopurine toxicity
- UGT1A1 variants cause irinotecan toxicity (58)
Pharmacodynamic variations:
- VKORC1 variants affect warfarin sensitivity
- HLA-B*5701 predicts abacavir hypersensitivity
- EGFR mutations predict gefitinib response in lung cancer (59)
Age-Related Factors
Pediatric considerations:
- Immature drug-metabolizing enzymes (reduced glucuronidation in neonates)
- Higher body water content affecting drug distribution
- Developing blood-brain barrier
- Growth and developmental changes requiring dose adjustments (60)
Geriatric considerations:
- Decreased hepatic and renal function
- Altered body composition (increased fat, decreased muscle)
- Polypharmacy and drug interactions
- Increased sensitivity to CNS drugs
- Beers Criteria identify potentially inappropriate medications (61)
Physiological and Pathological Factors
Gender differences affect pharmacokinetics (body composition, hormone influences on metabolism) and pharmacodynamics (QT prolongation risk higher in women) (62).
Pregnancy alters drug disposition through increased cardiac output, expanded plasma volume, altered protein binding, and induced metabolic enzymes. Teratogenic risk varies by gestational age and drug (63).
Disease states modify drug response:
- Hepatic dysfunction reduces drug metabolism
- Renal impairment decreases drug excretion
- Heart failure reduces hepatic blood flow
- Hypoalbuminemia increases free drug concentrations (64)
Conclusion
Pharmacology provides the scientific foundation for rational therapeutics, integrating knowledge from basic sciences to optimize clinical outcomes. Understanding fundamental principles of drug action, disposition, and regulation enables healthcare providers to select appropriate therapies, individualize dosing regimens, anticipate drug interactions, and minimize adverse effects.
The field continues evolving with advances in molecular medicine, genomics, and biotechnology. Precision medicine approaches increasingly allow tailored therapy based on individual patient characteristics. Novel drug modalities including gene therapies, RNA therapeutics, and immunotherapies expand therapeutic possibilities for previously untreatable conditions.
Mastery of pharmacological principles remains essential for all healthcare professionals involved in medication therapy. This knowledge base enables evidence-based prescribing decisions that maximize therapeutic benefit while minimizing risk, ultimately improving patient care and clinical outcomes.
References
- Rang HP, Ritter JM, Flower RJ, Henderson G. Rang & Dale's Pharmacology. 9th ed. Edinburgh: Elsevier; 2020. p. 1-5.
- Brunton LL, Knollmann BC, Hilal-Dandan R. Goodman & Gilman's The Pharmacological Basis of Therapeutics. 14th ed. New York: McGraw-Hill; 2023. p. 1-15.
- Katzung BG, Vanderah TW. Basic & Clinical Pharmacology. 15th ed. New York: McGraw-Hill; 2021. p. 1-12.
- Whalen K, Finkel R, Panavelil TA. Lippincott Illustrated Reviews: Pharmacology. 7th ed. Philadelphia: Wolters Kluwer; 2019. p. 1-10.
- Golan DE, Armstrong EJ, Armstrong AW. Principles of Pharmacology: The Pathophysiologic Basis of Drug Therapy. 4th ed. Philadelphia: Wolters Kluwer; 2017. p. 1-24.
- Ritter JM, Flower RJ, Henderson G, Loke YK, MacEwan D, Rang HP. Rang & Dale's Pharmacology. 9th ed. Edinburgh: Elsevier; 2020. p. 267-290.
- Relling MV, Klein TE. CPIC: Clinical Pharmacogenetics Implementation Consortium of the Pharmacogenomics Research Network. Clin Pharmacol Ther. 2011;89(3):464-467.
- Sneader W. Drug Discovery: A History. Chichester: John Wiley & Sons; 2005. p. 1-15.
- Brunton LL, Knollmann BC, Hilal-Dandan R. Goodman & Gilman's The Pharmacological Basis of Therapeutics. 14th ed. New York: McGraw-Hill; 2023. p. 3-8.
- Bennett PN, Brown MJ, Sharma P. Clinical Pharmacology. 12th ed. Edinburgh: Elsevier; 2022. p. 1-12.
- Holmstedt B, Liljestrand G. Readings in Pharmacology. Oxford: Pergamon Press; 1963. p. 23-45.
- Kaufmann SH. Paul Ehrlich: founder of chemotherapy. Nat Rev Drug Discov. 2008;7(5):373.
- Aminov RI. A brief history of the antibiotic era: lessons learned and challenges for the future. Front Microbiol. 2010;1:134.
- Collins FS, McKusick VA. Implications of the Human Genome Project for medical science. JAMA. 2001;285(5):540-544.
- Rowland M, Tozer TN. Clinical Pharmacokinetics and Pharmacodynamics: Concepts and Applications. 5th ed. Philadelphia: Wolters Kluwer; 2019. p. 1-18.
- Shargel L, Yu ABC. Applied Biopharmaceutics & Pharmacokinetics. 7th ed. New York: McGraw-Hill; 2016. p. 1-24.
- Winter ME. Basic Clinical Pharmacokinetics. 6th ed. Philadelphia: Wolters Kluwer; 2018. p. 8-31.
- Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol Ther. 2013;138(1):103-141.
- Benet LZ. Clearance concepts: a downdate. Clin Pharmacokinet. 1984;9(6):425-432.
- Whalen K, Finkel R, Panavelil TA. Lippincott Illustrated Reviews: Pharmacology. 7th ed. Philadelphia: Wolters Kluwer; 2019. p. 25-38.
- Kenakin T. A Pharmacology Primer: Techniques for More Effective and Strategic Drug Discovery. 5th ed. San Diego: Academic Press; 2018. p. 1-39.
- Tallarida RJ, Jacob LS. The Dose-Response Relation in Pharmacology. New York: Springer-Verlag; 1979. p. 1-25.
- Neubig RR, Spedding M, Kenakin T, Christopoulos A. International Union of Pharmacology Committee on Receptor Nomenclature and Drug Classification. XXXVIII. Update on terms and symbols in quantitative pharmacology. Pharmacol Rev. 2003;55(4):597-606.
- Bennett PN, Brown MJ, Sharma P. Clinical Pharmacology. 12th ed. Edinburgh: Elsevier; 2022. p. 13-24.
- Aronson JK. A manifesto for clinical pharmacology from principles to practice. Br J Clin Pharmacol. 2010;70(1):3-13.
- Klaassen CD, Watkins JB III. Casarett & Doull's Toxicology: The Basic Science of Poisons. 9th ed. New York: McGraw-Hill; 2019. p. 3-27.
- Hodgson E. A Textbook of Modern Toxicology. 4th ed. Hoboken: John Wiley & Sons; 2010. p. 1-18.
- Joint Formulary Committee. British National Formulary. 82nd ed. London: BMJ Group and Pharmaceutical Press; 2021. p. vii-xii.
- Patrick GL. An Introduction to Medicinal Chemistry. 6th ed. Oxford: Oxford University Press; 2017. p. 1-16.
- WHO Collaborating Centre for Drug Statistics Methodology. Guidelines for ATC Classification and DDD Assignment 2022. Oslo: Norwegian Institute of Public Health; 2021. p. 13-24.
- Trissel LA. Handbook on Injectable Drugs. 20th ed. Bethesda: American Society of Health-System Pharmacists; 2021. p. xi-xx.
- WHO. The Use of Stems in the Selection of International Nonproprietary Names (INN) for Pharmaceutical Substances. Geneva: World Health Organization; 2018. p. 1-8.
- ISMP. List of Confused Drug Names. Horsham: Institute for Safe Medication Practices; 2019. p. 1-11.
- Newman DJ, Cragg GM. Natural products as sources of new drugs over the nearly four decades from 01/1981 to 09/2019. J Nat Prod. 2020;83(3):770-803.
- Heinrich M, Barnes J, Gibbons S, Williamson EM. Fundamentals of Pharmacognosy and Phytotherapy. 3rd ed. Edinburgh: Elsevier; 2018. p. 12-39.
- Demain AL, Sanchez S. Microbial drug discovery: 80 years of progress. J Antibiot. 2009;62(1):5-16.
- Walsh G. Biopharmaceuticals: Biochemistry and Biotechnology. 2nd ed. Chichester: John Wiley & Sons; 2003. p. 1-27.
- Molinski TF, Dalisay DS, Lievens SL, Saludes JP. Drug development from marine natural products. Nat Rev Drug Discov. 2009;8(1):69-85.
- Silverman RB, Holladay MW. The Organic Chemistry of Drug Design and Drug Action. 3rd ed. San Diego: Academic Press; 2014. p. 1-42.
- Wermuth CG, Aldous D, Raboisson P, Rognan D. The Practice of Medicinal Chemistry. 4th ed. San Diego: Academic Press; 2015. p. 1-29.
- Leader B, Baca QJ, Golan DE. Protein therapeutics: a summary and pharmacological classification. Nat Rev Drug Discov. 2008;7(1):21-39.
- DiMasi JA, Grabowski HG, Hansen RW. Innovation in the pharmaceutical industry: new estimates of R&D costs. J Health Econ. 2016;47:20-33.
- Hill RG, Richards D. Drug Discovery and Development: Technology in Transition. 3rd ed. Edinburgh: Elsevier; 2021. p. 1-22.
- ICH. Guidance on Nonclinical Safety Studies for the Conduct of Human Clinical Trials and Marketing Authorization for Pharmaceuticals M3(R2). Geneva: International Council for Harmonisation; 2009. p. 1-11.
- Marchetti S, Schellens JH. The impact of FDA and EMEA guidelines on drug development in relation to Phase 0 trials. Br J Cancer. 2007;97(5):577-581.
- Morgan P, Van Der Graaf PH, Arrowsmith J, et al. Can the flow of medicines be improved? Fundamental pharmacokinetic and pharmacological principles toward improving Phase II survival. Drug Discov Today. 2012;17(9-10):419-424.
- Friedman LM, Furberg CD, DeMets DL, Reboussin DM, Granger CB. Fundamentals of Clinical Trials. 5th ed. New York: Springer; 2015. p. 1-23.
- FDA. Guidance for Industry: Providing Clinical Evidence of Effectiveness for Human Drug and Biological Products. Silver Spring: US Food and Drug Administration; 1998. p. 1-20.
- Pitts PJ, Le Louet H. Advancing drug safety through prospective pharmacovigilance. Ther Innov Regul Sci. 2018;52(4):400-402.
- Golan DE, Armstrong EJ, Armstrong AW. Principles of Pharmacology: The Pathophysiologic Basis of Drug Therapy. 4th ed. Philadelphia: Wolters Kluwer; 2017. p. 3-24.
- Kenakin T. Principles: receptor theory in pharmacology. Trends Pharmacol Sci. 2004;25(4):186-192.
- Copeland RA. Enzymes: A Practical Introduction to Structure, Mechanism, and Data Analysis. 2nd ed. New York: Wiley-VCH; 2000. p. 266-304.
- Giacomini KM, Huang SM, Tweedie DJ, et al. Membrane transporters in drug development. Nat Rev Drug Discov. 2010;9(3):215-236.
- Flora SJ, Pachauri V. Chelation in metal intoxication. Int J Environ Res Public Health. 2010;7(7):2745-2788.
- Muller PY, Milton MN. The determination and interpretation of the therapeutic index in drug development. Nat Rev Drug Discov. 2012;11(10):751-761.
- Rautio J, Meanwell NA, Di L, Hageman MJ. The expanding role of prodrugs in contemporary drug design and development. Nat Rev Drug Discov. 2018;17(8):559-587.
- Evans WE, Relling MV. Pharmacogenomics: translating functional genomics into rational therapeutics. Science. 1999;286(5439):487-491.
- Sim SC, Ingelman-Sundberg M. The Human Cytochrome P450 (CYP) Allele Nomenclature website: a peer-reviewed database of CYP variants and their associated effects. Hum Genomics. 2010;4(4):278-281.
- Pirmohamed M. Pharmacogenetics and pharmacogenomics. Br J Clin Pharmacol. 2001;52(4):345-347.
- Kearns GL, Abdel-Rahman SM, Alander SW, et al. Developmental pharmacology--drug disposition, action, and therapy in infants and children. N Engl J Med. 2003;349(12):1157-1167.
- American Geriatrics Society 2019 Beers Criteria Update Expert Panel. American Geriatrics Society 2019 Updated AGS Beers Criteria for Potentially Inappropriate Medication Use in Older Adults. J Am Geriatr Soc. 2019;67(4):674-694.
- Soldin OP, Mattison DR. Sex differences in pharmacokinetics and pharmacodynamics. Clin Pharmacokinet. 2009;48(3):143-157.
- Feghali M, Venkataramanan R, Caritis S. Pharmacokinetics of drugs in pregnancy. Semin Perinatol. 2015;39(7):512-519.
- Verbeeck RK. Pharmacokinetics and dosage adjustment in patients with hepatic dysfunction. Eur J Clin Pharmacol. 2008;64(12):1147-1161.