1 / 47

Rational dosage design

Rational dosage design. Is based on the assumption that there is a target concentration that will produce the desired therapeutic effect

ashleypaul
Download Presentation

Rational dosage design

An Image/Link below is provided (as is) to download presentation Download Policy: Content on the Website is provided to you AS IS for your information and personal use and may not be sold / licensed / shared on other websites without getting consent from its author. Content is provided to you AS IS for your information and personal use only. Download presentation by click this link. While downloading, if for some reason you are not able to download a presentation, the publisher may have deleted the file from their server. During download, if you can't get a presentation, the file might be deleted by the publisher.

E N D

Presentation Transcript


  1. Rational dosage design • Is based on the assumption that there is a target concentration that will produce the desired therapeutic effect • The intensity of a drug's effect is related to its concentration above a minimum effective concentration, whereas the duration of this effect reflects the length of time the drug level is above this value • By considering drug’s PKs, it is possible to individualize the dose regimen to achieve the target concentration

  2. Half-Life (t1/2) • Half-Life (t1/2): • it is the time required for the plasma concentration or the amount of drug in the body to change by one-half (i.e. 50%) • The half-life is a derived parameter that changes as a function of both CL and Vd:

  3. Bioequivalence • Two drug formulations are bioequivalent if they show comparable bioavailability and similar times to achieve peak blood concentrations. • Therapeutic equivalence • Two drug formulations are therapeutically equivalent if they are pharmaceutically equivalent (that is, they have the same dosage form, contain the same active ingredient, and use the same route of administration) with similar clinical and safety profiles. • Clinicaleffectiveness often depends on both the maximum serum drug concentration and the time required (after administration) to reach peak concentration. • Therefore, two drugs that are bioequivalent may not be therapeutically equivalent.

  4. Design and optimization of dosage regimen A. Continuous infusion regimens • Therapy may consist of a single dose of a drug, for example, a sleep inducing agent, such as zolpidem. • More commonly, drugs are continually administered, either as an IV infusion or in oral fixed-dose/fixed-time interval regimens (for example, “one tablet every 4 hours”). • Continuous or repeated administration results in accumulation of the drug until a steady state occurs. • Steady-state concentration is reached when the rate of drug elimination is equal to the rate of drug administration, such that the plasma and tissue levels remain relatively constant.

  5. 1. Plasma concentration of a drug following IV infusion: • With continuous IV infusion, the rate of drug entry into the body is constant. • Most drugs exhibit first-order elimination, that is, a constant fraction of the drug is cleared per unit of time. • Therefore, the rate of drug elimination increases proportionately as the plasma concentration increases. • Following initiation of a continuous IV infusion, the plasma concentration of a drug rises until a steady state (rate of drug elimination equals rate of drug administration) is reached, • At which point the plasma concentration of the drug remains constant.

  6. a. Influence of the rate of infusion on steady-state concentration: • The steady-state plasma concentration (Css) is directly proportional to the infusion rate. • Css is inversely proportional to the clearance of the drug. • Any factor that decreases clearance, such as liver or kidney disease, increases the Css of an infused drug (assuming Vd remains constant). • Factors that increase clearance, such as increased metabolism, decrease the Css.

  7. b. Time required to reach the steady-state drug concentration: • The concentration of a drug rises from zero at the start of the infusion to its ultimate steady-state level, Css. • The rate constant for attainment of steady state is the rate constant for total body elimination of the drug. • Thus, 50% of Css of a drug is observed after the time elapsed, since the infusion, t, is equal to t1/2, where t1/2 (or half-life) is the time required for the drug concentration to change by 50%. • After another half-life, the drug concentration approaches 75% of Css . • The drug concentration is 87.5% of Css at 3 half-lives and 90% at 3.3 half-lives. • Thus, a drug reaches steady state in about four to five half-lives.

  8. The sole determinant of the rate that a drug achieves steady state is the half-life (t1/2) of the drug, and this rate is influenced only by factors that affect the half-life. • The rate of approach to steady state is not affected by the rate of drug infusion. • When the infusion is stopped, the plasma concentration of a drug declines (washes out) to zero with the same time course observed in approaching the steady state.

  9. B. Fixed-dose/fixed-time regimens • Fixed doses of IV or oral medications given at fixed intervals result in time-dependent fluctuations in the circulating level of drug. • 1. Multiple IV injections: • When a drug is given repeatedly at regular intervals, the plasma concentration increases until a steady state is reached. Because most drugs are given at inter-vals shorter than five half-lives and are eliminated exponentially with`time, some drug from the first dose remains in the body when the second dose is administered, some from the second dose remains when the third dose is given, and so forth. • Therefore, the drug accumulates until, within the dosing interval, the rate of drug elimination equals the rate of drug administration and a steady state is achieved.

  10. a. Effect of dosing frequency: • With repeated administration at regular intervals, the plasma concentration of a drug oscillates about a mean. • Using smaller doses at shorter intervals reduces the amplitude of fluctuations in drug concentration. • the Css is affected by neither the dosing frequency (assuming the same total daily dose is administered) nor the rate at which the steady state is approached.

  11. 2. Multiple oral administrations: • Most drugs that are administered on an outpatient basis are oral medications taken at a specific dose one, two, or three times daily. • In contrast to IV injection, orally administered drugs may be absorbed slowly, and the plasma concentration of the drug is influenced by both the rate of absorption and the rate of elimination

  12. C. Optimization of dose • The goal of drug therapy is to achieve and maintain concentrations within a therapeutic response window while minimizing toxicity and/or side effects. With careful titration, most drugs can achieve this goal. • If the therapeutic window of the drug is small (for`example, digoxin, warfarin, and cyclosporine), extra caution should be taken in selecting a dosage regimen, and monitoring of drug levels may help ensure attainment of the therapeutic range. • Drug regimens are administered as a maintenance dose and may require a loading dose if rapid effects are warranted. • For drugs with a defined therapeutic range, drug concentrations are subsequently measured, and the dosage and frequency are then adjusted to obtain the desired levels.

  13. 1. Maintenance dose: • Drugs are generally administered to maintain a Css within the therapeutic window. It takes four to five half-lives for a drug to achieve Css. • To achieve a given concentration, the rate of administration and the rate of elimination of the drug are important. • The dosing rate can be determined by knowing the target concentration in plasma (Cp), clearance (CL) of the drug from the systemic circulation, and the fraction (F) absorbed (bioavailability):

  14. 2. Loading dose: • Sometimes rapid obtainment of desired plasma levels is needed (for example, in serious infections or arrhythmias). • Therefore, a “loading dose” of drug is administered to achieve the desired plasma level rapidly, followed by a maintenance dose to maintain the steady state. • In general, the loading dose can be calculated as: • For IV infusion, the bioavailability is 100%, and the equation becomes

  15. Loading doses can be given as a single dose or a series of doses. • Disadvantages of loading doses include increased risk of drug toxicity and a longer time for the plasma concentration to fall if excess levels occur. • A loading dose is most useful for drugs that have a relatively long half-life. • Without an initial loading dose, these drugs would take a long time to reach a therapeutic value that corresponds to the steady-state level.

  16. 3. Dose adjustment: • The amount of a drug administered for a given condition is estimated based on an “average patient.” • This approach overlooks interpatient variability in pharmacokinetic parameters such as clearance and Vd, which are quite significant in some cases. • Knowledge of pharmacokinetic principles is useful in adjusting dosages to optimize therapy for a given patient. • Monitoring drug therapy and correlating it with clinical benefits provides another tool to individualize therapy.

  17. When determining a dosage adjustment, Vd can be used to calculate the amount of drug needed to achieve a desired plasma concentration. • Suppose the concentration of digoxin in the plasma is C1 and the desired target concentration is C2, a higher concentration. • The following calculation can be used to determine how much additional digoxin should be administered to bring the level from C1 to C2.

  18. Pharmacovigilance (PV)

  19. Pharmacovigilance (PV) • PVis concerned withdetection, assessment & preventionof adverse reactions to drugs (ADRs) or any drug-related problems 28

  20. Drug-Related Problems • Lack of efficacy • Manufacturing defects • Medication errors • Drug misuse and abuse • Overdose • Contamination • Counterfeit products 29

  21. Recently, the concerns of PV have been widened to include: • Herbal • Traditional and complementary medicines • Blood products • Biologicals • Medical devices • Vaccines 30

  22. Why Pharmacovigilance? • The root of pharmacovigilance: Pharmaco (Greek)= Drug Vigilance (Latin)= to keep awake or alert • Because information collected during pre-marketing phase are incomplete with regard to possible ADR • Tests in animals are insufficiently predictive of human safety

  23. Why Pharmacovigilance? 32 • In clinical trials: • Patients are limited in number • Conditions of use differ from those in clinical practice • Duration of trials is limited

  24. Why Pharmacovigilance? Post-marketing surveillance by companies is therefore essential • Information about rare adverse reactions, chronic toxicity, use in special groups (children, elderly or pregnant women) or drug interactions is often incomplete or not available

  25. Why Pharmacoviglance? • Pre-marketing clinical trialsdo not have : • Statistical power to detect rare ADRs • To identify delayed ADRs • To identify effects from long-term exposure PV plays a prominent role in establishing safety profile of marketed drugs 34

  26. 35

  27. Definition of ADR • An ADRis defined according to definition of WHO“any response to a drug which is noxious, unintended& thatoccurs at dosesused in man for prophylaxis, diagnosis, or therapy of diseases’’ 36

  28. Epidemiology of ADRs ADRs represent a significant cause ofmorbidity & mortality Many ADRs aremild, sometimesserious& cancause death U.S, ADRs caused100 000deaths per year, 4th & 6thleading cause of death About 50% of ADRs are preventable 37

  29. Importance of ADRs Prolonglength of stay in hospitals Increasecostsof patientcare Commonest cause ofdrug withdrawal from market: Reductil (Sibutramine) 2010 Valdecoxib (Bextra) 2005 Rofecoxib (Vioxx) 2004 38

  30. ADRs is considered serious if: Causesdeath of patient Life-threatening Prolonginpatient hospitalisation Causessignificant or persist disability Congenital abnormality 39

  31. Risk Factors predisposing to ADRs • Age • Long duration of treatment • Polypharmacy • Liver, kidney diseases

  32. Causes of ADRs • Patient • Drug • Prescriber • Environmental factors

  33. Causes of ADRs 1. The patient: - Age (over 60 or under one month) - Genetic factors - Previous history of ADR - Hepatic or renal diseases 42

  34. Causes of ADRs 2. The drug - Narrow therapeutic index, e.g. warfarin, digoxin - Antimicrobials have a tendency to cause allergy - Ingredients of a formulation, e.g. colouring, flavouring 43

  35. Drugs most commonly causing ADRs • Warfarin • Diuretics • Digoxin • Antibacterials • Steroids • Antihypertensives Anticancer drugs Immunomodulators Analgescis

  36. Why report suspected ADRs? • Documentation of ADRs in patients’ records is often poor • Physicians fear that reporting of ADR may put them at risk • Under-reportingis common phenomenon 45

  37. Reporting Methods Spontaneous reporting: (Voluntary) • Doctors, nurses & pharmacists are supplied with forms to record suspected ADRs • Reporting ADRs to National Pharmacovigilance Centre • In UK, this is called ‘Yellow Card system’

  38. Prevention of ADRs • Taking a drug history • Reduce number of prescribed drugs • Remembering that certain patients (elderly, those with liver or renal diseases) more susceptible to ADRs

More Related