Path - Pharm - Exam 3 Flashcards
How is site crucial to the success of antibiotic therapy and resistance control?
acquiring and maintaining effective drug concentrations at an infection site is crucial
What is the difference between pharmacokinetics (PK) and pharmacodynamics (PD)?
PK – fate of drugs within the body. ADME:
- Absorption
- Distribution
- Metabolism
- Excretion
PD – the effects drugs exert on the body
- mechanisms of drug action
What are the two modes of action of antimicrobial antibiotics?
- Bactericidal
- Kills them, I,e, penicillins etc
- preferred for serious infection where host defense is inadequate
- MBC ~ MIC
2. Bacteriostatic - Inhibition (i.e. stops them from reproducing) without necessarily killing them - tetracyclines etc - useful for most infections - requires immune system assistance - MBC>>MIC
What curve is used to measure drug exposure?
- Area Under Curve (AUC)
- [plasma] vs time
- can find bioavailability and relative levels of exposure to oral vs IV route by comparing them
How can knowing how PK variables (fate of drug within body) influence drug efficacy (‘PD response’) improve antibiotic use?
- better design of dosages and dosing interval
- may help limit selection of drug resistant mirobe strains
- especially important with critically-ill patients, i.e. sepsis
What is the relationship between drug concentration and antimicrobial effect?
Concentration vs bacterial burden
Hill curve – starts high then suddenly declines (Z)
EC50 = halfway between the top and bottom of the curve
Emax = difference between bottom and top of curve
What is MIC and how can it be determined?
Minimum inhibitory concentration – least amount which stops bacteria growing visibly
Calculated in vitro and then used as reference plasma concentration in humans
Agar plate + E test. Plastic strop with gradient of antibiotic concentrations.
‘Zone of Inhibition’ – no bacteria. End – MIC.
What is post-antibiotic effect?
PAE shows if a drug has persistant action even after concentration levels fall below MIC
What is MBC?
Minimal bactericidal concentration
Least amount of antibiotic which KILLS bacteria
How can MIC be used to show the different PK-PD relationships?
- Time dependent PD
T > MIC is Key PK-PD endpoint - Concentration dependent PD
Cmax:MIC , or
AUC > MIC is Key PK-PD endpoint - Mixed PD
AUC : MIC is Key PK-PD endpoint
What is the relevant info about time dependent PD?
T > MIC is Key PK-PD endpoint
- i.e. the time that you have a concentration > MIC
- when [plasma]>MIC, rate and extent of killing is maximal regardless of [].
- use of shorter dosing interval may improve outcomes
Antiobiotics:
- beta lactams
- vancomycin
- macrolides
What is the relevant info about concentration dependent PD?
AUC > MIC is Key PK-PD endpoint
- strong post-antibiotic effect
Antibiotics: Cmax/MIC = aminoglycosides AUC>MIC = - fluoroquinolones - ketolides - macrolides
What is the relevant info about concentration dependent PD?
AUC : MIC is Key PK-PD endpoint
- i.e. killing related to drug concentration and time > MIC
- mainly bacteriostatic
- Ratio of 24 hr AUC to MIC
Antibiotics:
- tetracyclines
- macrolides
How do you decide on the type of PK-PD relationship?
Best R2 graph
What is the MPC?
Mutant prevention concentration (may actually be higher than MIC)
Plasma levels kept > MPC helps suppress resistance
What factors are involved in the extrinsic, intrinsic, and combined pathway of coagulation?
Extrinsic: Starts with damaged Tissue (PT)
VII – VIIa (7)
Intrinsic: Starts with abnormal vessel wall (APTT)
XII – XIIa (12)
XI – XIa (11)
IX – IXa (9)
Common: X – Xa (10) Prothrombin (II) – Thrombin -- fibrinogen (I) – fibrin (TT) Thrombin + fibrinogen = thrombin-fibrin clot
What factor is involved in coagulation factor inactivation?
ATIII (antithrombin III) + thrombin = inactive complex
Protein S and Protein C inactivate factors Va and VIIIa
How are platelets activated and first aggregated?
- contact with damaged arterial wall activated platelets
- activated platelets release thromboxane A2 (TxA2) and ADP
- TxA2 and ADP enhance platelet aggregation and adhesion
Describe the mechanisms of action of warfarin
- Vitamin K is a cofactor for post-translational carboxylation of glutamic acid groups on factors II (prothrombin), VII, IX and X
- Warfarin inhibits vitamin K recycling from KO (inactive) to KH2 (active)
- Target = VKORC1
- Weakly acidic
- Lipophilic – well absorbed orally, therefore long term oral coagulant
- Lipophilic – hepatic clearance CYP2C9
- Variation in CYP2C9 activity and VKORC1 sensitivity – variation in warfarin sensitivity
- > 4 fold variation in dose requirement
Consequences:
- protection from thrombosis
- bleeding
Describe the adverse effects of warfarin
a) Pregnancy
- teratogenic (fetal malformation)
b) Protein S and Protein C hereditable deficiency
- transient hypercoagulable state which can result in skin necrosis
- treat with heparin if known deficiency
c) Circumstances that increase bleeding risk, i.e.
- active peptic ulcer at risk of bleeding
- liver disease with defective clotting factor synthesis
- platelet defects
Be aware of major interactions affecting warfarin activity
- some drugs compete for CYP450 (metabolism in liver)
- some drugs induce CYP450
- amount of vitamin K
- conditions increasing/reducing coagulation factors
- other anticoagulants or antiplatelet drugs
a) Drugs that affect absorption – decreased efficacy
b) CYP2C9 induction – decreased efficacy
c) CYP2C9 inhibition – increased bleeding risk
What are the clinical indications for heparin and warfarin
- DVT
- Pulmonary embolism
- Arterial thrombosis
- Atrial fibrillation
- Thrombosis and embolic stroke
Use Heparin if:
- Venous and arterial thrombosis – it’s more rapid
- pregnant
- known protein S/C deficiency
What are the clinical indications for antiplatelet drugs?
Symptomatic atherosclerosis
Be able to discuss the clinical monitoring of heparin and warfarin
Monitoring Warfarin Therapy:
- take patient’s blood
- activate extrinsic and common pathways with thromboplastin
- measure PT (time to clot = prothrombin time)
- Ratio of PT to normal = INR
- INR 2-3 for most indications
Can reverse with:
- vitamin K
- fresh donor plasma or factor concentrates
Describe the mechanisms of action of heparin
- inactivation of Xa and thrombin
- binds to Lysine on AT III
- increased affinity for activated factors of intrinsic arm
- therefore, accelerated inactivation of these
- prevents conversion of fibrinogen to fribin
- stops clot propagation
Describe the adverse effects of heparin
- bleeding
- immune mediated platelet activation – thrombosis or bleeding
/What are the benefits of low molecular weight heparin?
- more predictable activity and elimination so can be dosed on body-weight basis
- slower elimination, so can be given by subcutaneous bolus (injection)
Name and describe the mode of action of drugs that block thrombin and Xa. Be aware of renal elimination.
Blocking Xa- II can’t be converted to thrombin
- rivaroxaban
- apixaban
Blocking thrombin – it can’t form thrombin-fibrin clot
- dabigatrin
Efficacy and bleeding risk comparable to or better than warfarin.
Renally excreted – caution with elderly and renal impairment
What are the anti-platelet drugs?
- Aspirin
- Clopidogrel
- Dipyridamole
Describe the mechanism of action of aspirin
- acetylation of NH2 terminal serine of cyclooxygenase – irreversible
- permanent loss of TxA2 production
- defective platelet clot formation
- protection from thrombotic disorders
When asprin alone doesn’t work, can protect patients from strokes with dipyridamole.
Explain the important aspects of clopidogrel transformation & its mode of action on platelets
Clopidogrel’s active metabolite blocks platelet ADP receptors irreversibly
Explain the mechanism of action of the thrombolytic drug, tPA
- tPA = tissue plasminogen activator
- protein involved in breakdown of clots
- it catalyzes conversion of plasminogen to plasmin – the major enzyme responsible for clot breakdown
- IV admin
- Relieves blockage of critical vessel (i.e. stroke)
What are the two categories and subcategories of undesirable drug effects?
- Non-deleterious = side effects
- Deleterious
a) pharmacological
b) pathological
c) genotoxic
What is the difference between adverse effects and toxicity?
Adverse effects = unwanted drug effects at normal doses
Toxicity = unwanted effects at supratherapeutic doses
Dose-response relationships in toxicity
- Quantal Dose-Response Curve (DRC) = Dose or [blood] vs % responding
- sigmoidal curve
- different toxic responses can display different dose-response relationships (i.e different curves/slope)
- see diagram in notes
What is the therapeutic window?
A range of doses that produces therapeutic response without causing any significant adverse effect in patients
Usually starts at minimum effective concentration (MEC) and ends at minimum toxic concentration (MTC) or maximum effective concentration.
What is the therapeutic index?
Therapeutic Index = Dose resulting in toxicity /dose giving therapeutic response i.e. ED50 to TD50. ED50 = effective dose at 50% responding TD50 = toxic dose LD50 – Lethal dose
Why do drugs cause toxicity?
a) toxicological bioactivation – i.e. become more toxic during metabolism in liver (especially CYP450). Products = ‘adducts’. EG – paracetamol. Induced hepatotoxicity syndromes.
b) predisposing genetic traits:
- CYP
- genetic deficiencies in CYP may mean they metabolize drugs more slowly
- can ensure that toxic thresholds are exceeded on repeated dosing
- may allow more metabolism via minor pathways to form toxic metabolites
- some patients may have diminished capacity for detoxification
- i.e. GILBERT’s SYNDROME
c) age
What is Gilbert’s syndrome?
- decreased hepatic levels of UGT1A1 – catalyzes the glucuronidation of bilirubin, therefore causes accumulation of bilirubin
- mild hyperbilirubinemia (jaundice)
- effects hepatic clearance of paracetamol, irinotecan (cancer drug) etc
Discuss bioactivation and the liver
- high metabolic activity renders liver vulnerable to reactive metabolited
- drug-induced liver injury (DILI)
- covalent binding to hepatocyte proteins may predict DILI potential