Clinical pharmacokinetics (PK) is the science of quantifying how drugs are absorbed, distributed, metabolized, and excreted in humans, and applying that knowledge to optimize therapy for individual patients. Its goals are to achieve target drug exposures that maximize efficacy and minimize toxicity, accounting for patient-specific factors such as organ function, age, genetics, comorbidities, and concomitant medications [1–3]. This chapter provides a practice-oriented overview of clinical PK concepts, study designs and analyses, dose optimization strategies, and regulatory and ethical considerations that connect quantitative models to bedside decisions.
What clinical pharmacokinetics does in practice
- Predicts and interprets drug concentrations from dose and patient characteristics.
- Informs dose selection and adjustment in special populations (renal/hepatic impairment, pediatrics, geriatrics, pregnancy, obesity, critical illness).
- Anticipates and manages drug–drug interactions (DDIs) involving enzymes and transporters.
- Links exposure to response using PK/PD indices and therapeutic drug monitoring (TDM).
- Underpins model-informed precision dosing, population PK, and physiologically based PK (PBPK) used in development and clinical care [1,4–6].
Core Concepts and Parameters
ADME and the exposure–response paradigm
- Absorption: entry of drug into systemic circulation.
- Distribution: reversible transfer between blood and tissues.
- Metabolism: chemical transformation, primarily hepatic and intestinal.
- Excretion: removal, mainly via kidney and bile.
The time course of drug exposure is summarized by the concentration–time profile after a dose or at steady state. Key parameters include area under the curve (AUC), maximum concentration (Cmax), time to Cmax (Tmax), clearance (CL), volume of distribution (V), and elimination half-life (t1/2). At steady state under linear kinetics, average concentration (Css,avg) equals dose rate divided by clearance; accumulation depends on half-life and dosing interval [1–3].
Clearance, volume, and half-life
- Clearance (CL) is the volume of plasma cleared of drug per unit time; it governs exposure (AUC = Dose/CL for IV dosing).
- Volume of distribution (V) links amount of drug in the body to plasma concentration; it reflects tissue binding and partitioning.
- Half-life (t1/2) equals 0.693 × V/CL; it informs time to steady state and dosing interval but not exposure by itself [1,2].
Clinical translation:
- A change in CL changes AUC and steady-state concentrations.
- A change in V alters peak–trough fluctuation and time to steady state but not AUC when CL is unchanged.
Bioavailability and first-pass metabolism
Oral bioavailability (F) is the fraction of dose reaching systemic circulation. It is reduced by incomplete absorption, intestinal metabolism, and hepatic first-pass extraction. For oral dosing under linear conditions, AUC = F × Dose/CL. Food and formulation can alter F and Tmax; bioequivalence determinations rely on comparison of AUC and Cmax with prespecified acceptance limits [2,7,8].
Determinants of Absorption
Physicochemical and formulation factors
- Solubility and permeability (Biopharmaceutics Classification System, BCS) guide formulation strategies and predict rate-limiting steps [3,9].
- Particle size, polymorphs, salt forms, and excipients influence dissolution and absorption.
- Gastric pH and motility (altered by disease or drugs like proton pump inhibitors) affect weak acids/bases differently [3].
Food effects and first-pass extraction
Meals can increase, decrease, or delay absorption by altering gastric emptying, bile flow, and luminal solubility. High-fat meals are used in standardized food-effect studies. For high-extraction drugs, first-pass metabolism by intestinal CYP3A and efflux by P-glycoprotein (P-gp) can markedly lower F; inhibitors of these pathways (e.g., certain azoles, macrolides, or grapefruit juice) may increase exposure [2,3,6].
Distribution and Protein Binding
Plasma protein binding and clinical implications
Acidic drugs bind mainly to albumin; basic drugs bind to alpha-1-acid glycoprotein (AAG). Only unbound drug is pharmacologically active and available for distribution and clearance. Changes in albumin (e.g., cirrhosis) or AAG (e.g., inflammation) can alter unbound fraction (fu). For high-extraction drugs, changes in binding often do not change unbound exposure, while for low-extraction drugs, fu and intrinsic clearance together influence unbound concentrations [1,2].
Tissue distribution and special compartments
Volume of distribution reflects tissue binding, partitioning into adipose, and sequestration (e.g., lysosomal trapping of weak bases). Large V is typical for lipophilic drugs; hydrophilic drugs often remain extracellular. Special barriers (blood–brain barrier, placenta) are governed by tight junctions and transporters (e.g., P-gp, BCRP), influencing distribution and clinical effects (e.g., CNS penetration) [1,6,10].
Metabolism, Excretion, and Transporters
Metabolic pathways
- Phase I: oxidative, reductive, hydrolytic reactions—principally via CYP450 isoforms (e.g., CYP3A4/5, CYP2D6, CYP2C19, CYP2C9).
- Phase II: conjugations (e.g., UGTs, SULTs, NATs, GSTs) to increase polarity.
Metabolic capacity varies widely due to genetics, disease, and induction/inhibition by other drugs [1,3,6].
Renal and biliary excretion
Renal clearance comprises glomerular filtration (unbound drug), active secretion, and tubular reabsorption. Biliary excretion contributes for large, polar molecules or conjugates. Transporters (e.g., OAT, OCT, MATE, OATP, P-gp, BCRP, BSEP, MRP) mediate uptake and efflux in liver, kidney, and intestine and are common mechanisms for DDIs [6,10,11].
Transporters and clinically relevant DDIs
Inhibitors or inducers of transporters can significantly alter exposure. Examples include inhibition of OATP1B1/1B3 increasing statin levels, or P-gp inhibition elevating digoxin concentrations. Regulatory guidances outline decision trees for in vitro assessment and when to conduct clinical DDI studies, integrating both enzyme and transporter pathways [6,11–13].
Linear, Nonlinear, and Time-Dependent Pharmacokinetics
Linear kinetics
Dose-proportional increases in AUC and Cmax reflect constant CL and V over the therapeutic range. Most drugs at usual doses exhibit linear PK.
Capacity-limited (nonlinear) kinetics
- Saturable metabolism (e.g., phenytoin) follows Michaelis–Menten behavior: small dose changes can cause disproportionate increases in concentration near the capacity limit [1,2].
- Saturable protein binding (e.g., valproate) increases fu at higher concentrations, altering both distribution and clearance.
- Target-mediated drug disposition (TMDD) for biologics occurs when binding to pharmacologic targets contributes to clearance; clearance may decrease as targets saturate [14,15].
Time-dependent kinetics
- Autoinduction (e.g., carbamazepine) increases clearance over time, reducing concentrations at the same dose.
- Mechanism-based inhibition (e.g., erythromycin on CYP3A) decreases clearance until enzyme is resynthesized [6].
Pharmacogenomics and Interindividual Variability
Genetic polymorphisms affecting PK
Common variants in CYP2D6, CYP2C19, CYP2C9, and transporters like SLCO1B1 influence metabolism or distribution and can necessitate dose changes or alternative therapies. Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines provide genotype-based prescribing recommendations for drugs such as clopidogrel (CYP2C19), codeine (CYP2D6), and simvastatin (SLCO1B1) [16–18]. Genetic effects are one contributor to variability; age, organ function, inflammation, and DDIs often co-exist.
Special Populations
Renal impairment
Reduced glomerular filtration, altered secretion, and changes in protein binding affect both renally cleared and some hepatically cleared drugs (via uremic toxin effects on enzymes/transporters). Regulatory guidance recommends dedicated renal impairment PK studies and dosage adjustment strategies based on exposure–response relationships [12,19]. Estimating kidney function (e.g., eGFR, creatinine clearance) is essential when adjusting doses of drugs with significant renal elimination (e.g., aminoglycosides, direct oral anticoagulants).
Hepatic impairment
Cirrhosis alters enzyme/transporter expression, hepatic blood flow, protein binding, and portal shunting. The net effect depends on drug extraction and binding characteristics. Dose adjustments often differ by Child–Pugh class, but mechanistic understanding and, when possible, direct PK data in hepatic impairment should guide therapy [20].
Pediatrics
Ontogeny of enzymes and transporters, body composition, and organ maturation significantly alter PK in neonates, infants, and children. Allometric scaling and maturation functions are commonly used in pediatric population PK models; dosing must consider developmental stage rather than simply body weight [5,21].
Geriatrics
Aging reduces renal function, may alter hepatic blood flow and enzyme abundance, and changes body composition (increased fat, decreased total body water). Polypharmacy elevates DDI risk. Start low and titrate carefully; monitor closely for toxicity and therapeutic effect [3,22].
Pregnancy and lactation
Pregnancy increases plasma volume, cardiac output, glomerular filtration, and can induce some metabolic pathways (e.g., CYP3A) while reducing others. These changes typically lower concentrations of many drugs, necessitating dose adjustments for some indications (e.g., antiretrovirals, antiepileptics). Placental transfer, fetal exposure, and breast milk excretion require risk–benefit considerations [23].
Obesity and critical illness
Obesity increases V for lipophilic drugs and may alter CL via changes in organ size and perfusion. Choice of dosing scalar (total, lean, or adjusted body weight) depends on the drug’s physicochemical properties and clearance mechanisms. In critical illness, capillary leak, fluid shifts, augmented renal clearance, and altered protein binding can dramatically change PK, particularly for hydrophilic antibiotics—necessitating individualized dosing and monitoring [24,25].
From Exposure to Effect: PK/PD and Therapeutic Drug Monitoring
PK/PD indices and antimicrobial therapy
Antimicrobial efficacy often correlates with specific PK/PD indices:
- Time-dependent killers: fT>MIC (e.g., beta-lactams).
- Concentration-dependent killers: fCmax/MIC (e.g., aminoglycosides).
- Exposure-driven: fAUC/MIC (e.g., fluoroquinolones, vancomycin) [26–28].
These indices guide dosing regimens (e.g., extended infusion for beta-lactams to maximize fT>MIC) and clinical trial targets.
Therapeutic drug monitoring (TDM)
TDM is indicated when there’s a narrow therapeutic window, significant PK variability, poor correlation between dose and effect, and well-defined concentration targets. Common examples include:
- Vancomycin: AUC24/MIC target 400–600 mg·h/L using Bayesian AUC monitoring rather than trough-only strategies [29].
- Aminoglycosides: once-daily high peaks for Cmax/MIC and low troughs to minimize toxicity; monitor especially in renal impairment [30].
- Antiepileptics (e.g., phenytoin): monitor due to nonlinear kinetics and variable protein binding [1–3].
- Immunosuppressants (tacrolimus, cyclosporine): trough targets adjusted for time post-transplant, genetics, and interactions [31].
Effective TDM requires clear sampling strategy, validated assays, clinical context, and, increasingly, Bayesian forecasting tools that integrate prior population models with individual data [5,32].
Design and Analysis of Clinical PK Studies
Study designs
- Single-dose and multiple-dose studies (to steady state) characterize linearity, accumulation, and time-dependence.
- Crossover designs reduce variability for bioavailability/bioequivalence.
- Food-effect studies assess fed vs fasted states.
- Special population studies (renal/hepatic impairment, pediatrics).
- DDI studies informed by in vitro enzyme/transporter data according to regulatory decision trees [6,11–13,19,20].
Sampling strategies range from rich intensive designs to sparse opportunistic sampling in patients; both can be informative when analyzed appropriately.
Noncompartmental analysis (NCA)
NCA estimates exposure and rate metrics directly from concentration–time data without assuming a structural model:
- AUC by trapezoidal rules with extrapolation using terminal slope (λz).
- Cmax and Tmax observed.
- CL = Dose/AUC (IV); apparent CL/F for extravascular.
- Vss estimated from AUMC/AUC for IV studies.
NCA is simple and widely used for regulatory bioavailability and BE assessments; careful handling of sampling windows and terminal phase is essential [3,9].
Compartmental and mechanistic modeling
Compartmental models (e.g., one- or two-compartment with first-order or nonlinear processes) provide parameters with physiological interpretation and can simulate alternative regimens. Mechanistic models include TMDD for biologics and PBPK for system-level predictions [5,14,15,33].
Population PK (PopPK) and covariate modeling
PopPK uses nonlinear mixed-effects modeling to quantify typical parameters and interindividual variability, identify covariate effects (e.g., weight, eGFR, genotype), and support individualized dosing:
- Model building and evaluation: diagnostic plots, prediction-corrected visual predictive checks (pcVPC), bootstrapping, and external validation.
- Covariates: pre-specified hypotheses and clinical plausibility prevent overfitting.
- Applications: dose selection, labeling, TDM-informed Bayesian dosing, bridging across populations [4,5,32,33].
Model-informed precision dosing (MIPD)
Bayesian forecasting combines prior PopPK models with patient-specific concentrations and covariates to recommend doses achieving target exposures. Evidence and software ecosystems are expanding, particularly for anti-infectives and immunosuppressants [32].
Physiologically based PK (PBPK)
PBPK models describe absorption, tissue distribution, metabolism, and excretion using anatomical and physiological parameters, enabling:
- Prediction of enzyme/transporter-mediated DDIs across scenarios.
- Extrapolation to special populations (e.g., pediatrics, hepatic impairment, pregnancy).
- Quantitative risk assessment to waive or focus clinical studies, per regulatory guidances [4,34,35].
Drug–Drug Interactions: Assessment and Management
From in vitro to in vivo
Regulatory pathways outline when in vitro inhibition/induction data trigger clinical DDI studies, and how to design them. Static and dynamic models (e.g., basic R-value, mechanistic static models, PBPK) estimate interaction magnitude. Enzymes (CYP3A, 2D6, 2C19, 2C9) and transporters (P-gp, OATP1B1/1B3, BCRP, OCT2, MATE) are key [11–13,34,35].
Clinical strategies
- Avoid strong inhibitors/inducers for narrow therapeutic index (NTI) drugs when possible.
- Adjust doses based on predicted or observed AUC changes.
- Monitor concentrations or pharmacodynamic markers (e.g., INR for warfarin).
- Consider staggered administration or alternative agents to manage transporter-mediated interactions (e.g., statins with OATP inhibitors) [6,11–13].
Therapeutic Proteins and Biologics
Distinct PK characteristics
Monoclonal antibodies and other large molecules have low V (confined to vascular and interstitial spaces), are not renally filtered, and are cleared by proteolysis and target-mediated pathways. Neonatal Fc receptor (FcRn) recycling prolongs IgG half-life. TMDD can cause nonlinear PK at therapeutic concentrations. Immunogenicity can alter clearance and response [14,15,36].
Dosing implications
Body weight or body surface area scaling is common but not universal; fixed dosing may be appropriate when variability is low or exposure–response is flat. PBPK and PopPK inform dose selection across indications and populations. For some biologics, exposure–response supports therapeutic range monitoring of target engagement or biomarkers rather than drug levels [14–16].
Regulatory Perspectives and Labeling
Guidances and expectations
- DDIs: comprehensive evaluation of CYP- and transporter-mediated interactions, with decision frameworks for in vitro and clinical studies (FDA, EMA, ICH M12) [11–13].
- Renal and hepatic impairment: dedicated studies and dosing recommendations [19,20].
- PBPK: recommended content, verification, and application to support regulatory decisions [34,35].
- Bioavailability and bioequivalence: study design and acceptance criteria (e.g., 90% CI for test/reference ratio of AUC and Cmax within 80–125% for most drugs) [7,8].
Labels should present clear dose adjustment instructions, DDI management guidance, and, when available, genotype-based recommendations. Increasingly, modeling and simulation findings (PopPK, PBPK) inform labeling for unstudied scenarios [4,6,34].
Putting It All Together at the Bedside
A practical workflow
- Define the therapeutic goal and exposure target (e.g., AUC/MIC).
- Assess patient factors: weight/size metrics, eGFR, hepatic function, age, pregnancy, critical illness, inflammation, genotype.
- Identify DDIs (enzymes and transporters) and co-morbidities.
- Choose initial dose and schedule using established guidelines or models.
- Plan monitoring: which concentrations, when to sample, which assay.
- Adjust dose using observed levels and Bayesian tools where available.
- Document assumptions and reassess as clinical status changes (renal function, concomitant drugs).
Common clinical scenarios
- Phenytoin: nonlinear metabolism necessitates careful titration and measured levels; changes in albumin require corrected or free concentrations [1–3].
- Vancomycin: AUC-guided dosing reduces nephrotoxicity and maintains efficacy; consider augmented renal clearance in the critically ill [24,25,29].
- Direct oral anticoagulants: adjust dose in renal impairment per product labeling; beware P-gp/CYP3A interactions [2,6].
- Simvastatin and SLCO1B1: consider alternative statin or lower dose in reduced-function genotypes and when co-administered with OATP inhibitors [17,18].
Common Pitfalls and How to Avoid Them
Pitfalls
- Equating half-life with exposure: half-life guides dosing interval, not AUC.
- Ignoring unbound concentration changes when protein binding shifts.
- Underestimating transporter-mediated DDIs.
- Applying linear adjustments to drugs with nonlinear kinetics.
- Extrapolating adult doses to children by mg/kg without maturation considerations.
- Relying solely on troughs when AUC is the therapeutic driver.
Avoidance strategies
- Anchor decisions to clearance and target exposures when possible.
- Use exposure metrics that match the pharmacology (AUC, fT>MIC, Cmax).
- Apply regulatory decision trees and evidence-based guidelines for DDIs and special populations.
- Adopt PopPK/PBPK tools and Bayesian dosing when feasible.
- Collaborate with clinical pharmacists and use TDM judiciously [1–6,29,32].
Conclusion
Clinical pharmacokinetics connects the science of drug disposition with the art of therapeutic decision-making. By understanding how clearance, volume, and bioavailability shape exposure—and how patient factors, enzymes, and transporters modify these processes—clinicians can select and adapt doses to achieve defined therapeutic targets. Modern tools such as population PK, PBPK, and Bayesian MIPD extend this precision to diverse patients and complex regimens. A disciplined workflow, informed by regulatory guidance and best-evidence PK/PD principles, improves outcomes and safety across therapies and settings.
References
- Rowland M, Tozer TN, Derendorf H, Hochhaus G. Clinical Pharmacokinetics and Pharmacodynamics: Concepts and Applications. 5th ed. Philadelphia (PA): Wolters Kluwer; 2019.
- Shargel L, Wu-Pong S, Yu AB. Applied Biopharmaceutics & Pharmacokinetics. 7th ed. New York (NY): McGraw-Hill; 2015.
- Benet LZ, Hoener BA. Changes in Plasma Protein Binding Have Little Clinical Relevance. Clin Pharmacol Ther. 2002;71(3):115-121.
- Zhao P, Zhang L, Grillo JA, et al. Applications of Physiologically Based Pharmacokinetic (PBPK) Modeling and Simulation During Regulatory Review. Clin Pharmacol Ther. 2011;89(2):259-267.
- Mould DR, Upton RN. Basic Concepts in Population Modeling, Simulation, and Model-Based Drug Development—Part 2: Introduction to Pharmacokinetic Modeling Methods. CPT Pharmacometrics Syst Pharmacol. 2012;1:e38.
- European Medicines Agency. Guideline on the Investigation of Drug Interactions [Internet]. London (UK): EMA; 2012 [cited 2025 Aug 31]. Available from: https://www.ema.europa.eu/
- US Food and Drug Administration. Bioavailability Studies Submitted in NDAs or INDs — General Considerations: Guidance for Industry [Internet]. Silver Spring (MD): FDA; 2014 [cited 2025 Aug 31]. Available from: https://www.fda.gov/
- European Medicines Agency. Guideline on the Investigation of Bioequivalence [Internet]. London (UK): EMA; 2010 [cited 2025 Aug 31]. Available from: https://www.ema.europa.eu/
- Gabrielsson J, Weiner D. Pharmacokinetic and Pharmacodynamic Data Analysis: Concepts and Applications. 5th ed. Stockholm (SE): Swedish Pharmaceutical Press; 2016.
- Lin JH, Lu AY. Role of Pharmacokinetics and Metabolism in Drug Discovery and Development. Pharmacol Rev. 1997;49(4):403-449.
- US Food and Drug Administration. In Vitro Drug Interaction Studies — Cytochrome P450 Enzyme- and Transporter-Mediated Interactions: Guidance for Industry [Internet]. Silver Spring (MD): FDA; 2020 [cited 2025 Aug 31]. Available from: https://www.fda.gov/
- ICH Harmonised Guideline M12. Drug Interaction Studies [Internet]. Geneva (CH): International Council for Harmonisation; 2024 [cited 2025 Aug 31]. Available from: https://www.ich.org/
- Zhang L, Zhang Y, Huang SM. Scientific and Regulatory Perspectives on Metabolizing Enzyme–Transporter Interplay and Its Role in Drug Interactions. Mol Pharm. 2009;6(6):1766-1781.
- Mould DR, Sweeney K. The Pharmacokinetics and Pharmacodynamics of Monoclonal Antibodies—Mechanistic Modeling Applied to Drug Development. Curr Opin Drug Discov Devel. 2007;10(1):84-96.
- Liu L. Pharmacokinetics of Monoclonal Antibodies and Fc-Fusion Proteins. Prog Mol Biol Transl Sci. 2013;112:101-119.
- Scott SA, Sangkuhl K, Stein CM, et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for CYP2C19 and Clopidogrel. Clin Pharmacol Ther. 2013;94(3):317-323. Update: 2022.
- Crews KR, Gaedigk A, Dunnenberger HM, et al. Clinical Pharmacogenetics Implementation Consortium Guidelines for Codeine Therapy in the Context of CYP2D6 Genotype. Clin Pharmacol Ther. 2012;91(2):321-326. Update: 2021.
- Ramsey LB, Johnson SG, Caudle KE, et al. The Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for SLCO1B1 and Simvastatin-Induced Myopathy. Clin Pharmacol Ther. 2014;96(4):423-428. Update: 2022.
- US Food and Drug Administration. Pharmacokinetics in Patients with Impaired Renal Function — Study Design, Data Analysis, and Impact on Dosing: Guidance for Industry [Internet]. Silver Spring (MD): FDA; 2020 [cited 2025 Aug 31]. Available from: https://www.fda.gov/
- US Food and Drug Administration. Pharmacokinetics in Patients with Impaired Hepatic Function — Study Design, Data Analysis, and Impact on Dosing: Guidance for Industry [Internet]. Silver Spring (MD): FDA; 2023 [cited 2025 Aug 31]. Available from: https://www.fda.gov/
- ICH Harmonised Guideline E11(R1). Clinical Investigation of Medicinal Products in the Pediatric Population [Internet]. Geneva (CH): International Council for Harmonisation; 2017 [cited 2025 Aug 31]. Available from: https://www.ich.org/
- Mangoni AA, Jackson SHD. Age-Related Changes in Pharmacokinetics and Pharmacodynamics: Basic Principles and Practical Applications. Br J Clin Pharmacol. 2004;57(1):6-14.
- Hebert MF, Easterling TR, Kirby B, et al. Effects of Pregnancy on CYP3A and P-glycoprotein Activities as Measured by Disposition of Midazolam and Digoxin. Clin Pharmacol Ther. 2008;84(2):248-253.
- Udy AA, Roberts JA, Lipman J. Implications of Augmented Renal Clearance in Critically Ill Patients. Nat Rev Nephrol. 2011;7(9):539-543.
- Roberts JA, Abdul-Aziz MH, Lipman J, et al. Individualised Antibiotic Dosing for Patients Who Are Critically Ill: Challenges and Potential Solutions. Lancet Infect Dis. 2014;14(6):498-509.
- Craig WA. Pharmacokinetic/Pharmacodynamic Parameters: Rationale for Antibacterial Dosing of Mice and Men. Clin Infect Dis. 1998;26(1):1-10.
- Drusano GL. Antimicrobial Pharmacodynamics: Critical Interactions of ‘Bug and Drug’. Nat Rev Microbiol. 2004;2(4):289-300.
- Mouton JW, Brown DFJ, Apfalter P, et al. The Role of Pharmacokinetics/Pharmacodynamics in Setting Clinical MIC Breakpoints: The EUCAST Approach. Clin Microbiol Infect. 2012;18(3):E37-E45.
- Rybak MJ, Le J, Lodise TP, et al. Therapeutic Monitoring of Vancomycin for Serious Methicillin-Resistant Staphylococcus aureus Infections: A Revised Consensus Guideline and Review by ASHP/PIDS/SIDP/IDSA. Am J Health Syst Pharm. 2020;77(11):835-864.
- Nicolau DP. Pharmacodynamic Optimization of Beta-Lactams in the Patient Care Setting. Crit Care. 2008;12(Suppl 4):S2. [As context for aminoglycosides and beta-lactams dosing]
- Staatz CE, Tett SE. Clinical Pharmacokinetics and Pharmacodynamics of Tacrolimus in Solid Organ Transplantation. Clin Pharmacokinet. 2004;43(10):623-653.
- Neely MN, van Guilder MG, Yamada WM, et al. Accurately Achieving Target Concentrations in Children Through Individualized Dosing of Vancomycin. Ther Drug Monit. 2014;36(1):73-80.
- Ette EI, Williams PJ, editors. Pharmacometrics: The Science of Quantitative Pharmacology. Hoboken (NJ): John Wiley & Sons; 2007.
- US Food and Drug Administration. Physiologically Based Pharmacokinetic Analyses — Format and Content: Guidance for Industry [Internet]. Silver Spring (MD): FDA; 2018 [cited 2025 Aug 31]. Available from: https://www.fda.gov/
- European Medicines Agency. Guideline on the Qualification and Reporting of Physiologically Based Pharmacokinetic (PBPK) Modelling and Simulation [Internet]. London (UK): EMA; 2018 [cited 2025 Aug 31]. Available from: https://www.ema.europa.eu/
- US Food and Drug Administration. Clinical Pharmacology Considerations for Biopharmaceutical Products [Internet]. Silver Spring (MD): FDA; 2014 [cited 2025 Aug 31]. Available from: https://www.fda.gov/
[…] and development, Souces of Drugs, Dosage forms, Routes of drug administration, Pharmacokinetics, Clinical Pharmacokinetics, Pharmacodynamics, Drug interactions and Adverse drug reactions (ADRs). Understanding these […]
[…] Clinical Pharmacokinetics […]
[…] tailored to individual patient needs. Three key pharmacokinetic parameters must be comprehended: bioavailability (F), volume of distribution (Vd), and clearance (CL). The first two have been previously […]