Drug Disposition Flashcards

Galetin teaching

1
Q

Define pharmacokinetics.

A

How the body responds to drugs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Define pharmacodynamics.

A

The impacts the drug has on the body.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What is the main aim of metabolism?

A

To make the molecule more hydrophilic in preparation for elimination.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Define bioavailability.

A

A measure of the extent of absorption of unchanged administered compound.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is distribution?

A

Reversible transfer of a substance between site of administration and site of measurement.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is metabolism?

A

Irreversible loss of unchanged substance by chemical conversion.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What is elimination?

A

Irreversible loss of unchanged substance from site of measurement.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Name 6 factors that influence pharmacokinetics?

A
Genetics
Age
Size of molecule
Disease state
Concomitant drugs
Environment
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What are the names of two pharmacokinetic models?

A

Single equation, integrated

Kinetic compartmental

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Define rate of change in an equation.

A

Rate of change = rate of absorption - (rate of excretion + rate of metabolism)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What is the main reason that the effectiveness of a drug molecule will eventually plateau?

A

Receptor saturation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Name 6 physiological parameters affecting drug action.

A
Tissue volume
Blood flow
Tissue composition
Intestinal pH
Gut transit time
Enzyme/transporter abundance
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What is absolute bioavailability?

A

Bioavailability assessed with reference to an IV dose

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What is relative bioavailability?

A

Comparison of different formulations of the same drug

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Define bioequivalence.

A

Formulations containing the same dose intended to be interchangeable.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What impact does a fasted state have on delivery in the GIT?

A

Rapid delivery to the upper small intestine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What impact does a fed state have on delivery in the GIT?

A

Can delay gastric emptying by up to 10 hours

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

When should enteric coating be given? Why?

A

Given on a fasted stomach to ensure it reaches the small intestine as quickly as possible.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What impact does gastric bypass surgery have on absorption?

A

Reduces surface area of the stomach and bypasses approximately 75cm on the intestine, main area of drug absorption.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What impact does coeliac disease have on drug delivery?

A

Expression of intestinal CYP3A reduced to 15%.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What impact does chronic kidney disease have on drug delivery?

A

Increased gastric pH and emptying time

Can alter CYP450 expressio

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is transcellular movement?

A

Passive diffusion often via facilitated transport
Continues until equilibrium reached
Increased molecule size reduces permeability

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What is paracellular movement?

A

Transport between epithelial cells mainly via passive diffusion
Important for polar, hydrophilic drugs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What is P-glycoprotein?

A

Efflux transporter found on the apical membrane of enterocytes and biliary canicular membrane of hepatocytes
Active efflux of drugs into intestinal lumen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
What is first pass metabolism?
Metabolism of molecules before reaching systemic circulation. Can occur in the intestine, liver or kidney F= FaFgFh
26
What are the pharmacokinetic parameters affecting IM/SC absorption?
Muscle capillary membrane is more porous than the gut, allowing paracellular absorption Perfusion rate is limited by blood flow
27
What are the pharmacokinetic parameters affecting transdermal absorption?
Permeability rate is limited, even for lipophilic drugs
28
What are the pharmacokinetic parameters affecting pulmonary absorption?
Rapid access too systemic circulation due to surface area | Only 2-10% of aerosol actually deposits in the lungs
29
Name two drugs affecting absorption of paracetamol. Explain why.
Metoclopramide- increases gastric emptying therefore paracetamol reaches SI faster, increasing Cmax and reducing Tmax Anticholinergics- reduce Cmax of paracetamol and increase Tmax however the AUC remains the same
30
When does perfusion limited distribution occur?
When the tissue/cell membrane does not represent a barrier to drug distribution
31
Give 4 examples of highly perfused tissues.
Brain Lungs Liver Kidney
32
Which tissue do lipophilic drugs have particularly high affinity for?
Adipose
33
When is permeability limited distribution most likely to occur?
Polar molecules | Relatively impermeable membranes
34
What are the features of the blood brain barrier relevant for molecule distribution?
Very small pores | Barrier at capillary level
35
What are the features of the muscle/kidney cells relevant for molecule distribution?
Capillaries porous to small molecules | Barrier at cellular level
36
What are the 3 key water volumes within the body?
Plasma water 3L Extracellular water 12L Total body water 40L
37
At equilibrium what is the amount of drug in the body defined by?
A= V x C | Where V= volume of distribution and C= concentration
38
What do acidic drugs show strong affinity to? Give an example.
Plasma proteins | I.e. warfarin
39
What do basic drugs show strong affinity to? How does this impact the volume of distribution? Give an example.
Acidic phospholipids in tissues, thus have a higher volume of distribution. I.e. fluoxetine
40
How do lysosomes affect volume of distribution? Give examples of drugs affected by this.
Sequester cationic amphiphilic drugs Molecule (logP >2, pH 6.5-11) can accumulate and not reach receptor target Reduces activity and off target effects of drugs. I.e. azithromycin, vinblastine
41
Name the three plasma proteins largely responsible for drug binding. What drugs are likely to bind?
Albumin- acidic and neutral drugs such as cyclosporine Alpha-1 acid glycoprotein- basic drugs Globulins- steroids
42
What is Fu?
Fraction unbound | Fu= Cu/C
43
What circumstances may alter fraction of drug unbound?
Pregnancy Liver cirrhosis Renal impairment
44
What is Kp?
Tissue to plasma partition coefficient | VpC= KpVtCt
45
What are the most common drug-drug interactions?
Metabolic DDIs, inhibition or induction of CYP enzymes
46
Describe the effects of an inhibition DDI.
Reduced metabolising rate, increased drug concentration potentiating drug effect.
47
Describe the effects of an induction DDI.
Increased metabolising rate, decreased drug concentration and drug effect.
48
What is Ki?
The inhibitor-enzyme dissociation constant, analogous to Km
49
What are the 3 classifications of CYP inhibition?
Strong 5 fold increase in AUC Moderate 2-5 fold increase Weak 1.25-2 fold increase AUC ratio= AUC(inh)/AUC
50
What impact does grapefruit have on drug metabolism?
Irreversible inhibition of intestinal CYP3A4 by furanocoumarins No effect seen when given IV so minimal effects on hepatic CYP
51
What is auto-induction? Give a drug example.
A drug increases its own metabolism at an enzyme | I.e. Carbamazepine
52
What is the common mechanism of enzyme induction?
Requires complement of cellular processes | Increased transcription and translation, reduced degradation
53
What occurs physiologically upon enzyme induction?
Increased Vmax Altered Km at enzyme level Increased CYP synthesis Pronounced proliferation of endoplasmic reticulum Increased liver size, weight and blood and bile flow
54
Describe the clinical consequences of concomitant administration of ketoconazole and terfenadine.
Ketoconazole is a CYP3A4 inhibitor Terfenadine must be broken down to fexofenadine for action but CYP3A4 Increased levels of terfenadine can cause QT prolongation and severe arrhythmia
55
What are the two major subfamilies of transporters?
ATP-binding cassette | Solute carriers
56
Where are the most clinically relevant transporters expressed?
Epithelia of intestine, liver and kidney | Endothelium of BBB
57
What is OATP?
Organic anion transporting peptide Located on sinusoidal membrane of hepatocytes Mediates uptake of drugs and endogenous compounds
58
Describe the clinical consequences of concomitant administration of cyclosporine and rosuvastatin.
Cyclosporine inhibits OATP1B1 Rosuvastatin is transported into the liver for metabolism via OATP1B1 Inhibition of this increases Cmax and AUC of rosuvastatin leading to rhabdomyolysis
59
Which drug is responsible for the most relevant P-gp DDIs?
Digoxin
60
What is the role of BCRP in drug uptake?
Attenuates intestinal absorption of poorly permeable drugs and contributes to biliary elimination
61
What drug does turmeric most commonly interact with?
Inhibits intestinal efflux of sulphasalazine.
62
What effect does apple juice have on drug absorption?
Inhibits intestinal uptake of aliskiren and fexofenadine via OPAT2B1
63
Define renal clearance.
CLr= rate of urinary excretion/concentration in plasma
64
What percentage of cardiac output do kidneys receive?
20%
65
What is the average urine flow rate and GFR?
Urine flow= 1-2mL/min | GFR= 120mL/min
66
What is the rate of renal excretion?
Rate of excretion= (rate of filtration + rate of secretion) x (1-fraction resorbed)
67
What is active tubular secretion?
Active uptake of compounds via OAT/OCT paired with efflux transporters
68
Give 4 examples of OAT1 substrates.
Adefovir, oseltamivir, methotrexate, penicillin G
69
Give 2 examples of OCT2 substrates.
Metformin, pindolol
70
What two major factors affect reabsorption in kidneys?
Lipophilicity and ionisation
71
How does urine pH affect renal clearance?
Ranges from 4.5-7.6 Weak acids- CLr is urine pH sensitive, pKa 3-7.5 drug is non-ionised and lipophilic Strong acids- pKa<3 mostly ionised thus minimal absorption Weak bases- CLr is urine pH sensitive at pKa 6-12, drug is non-ionised
72
How does urine flow affect renal clearance?
Flow dependent on CLr occurs when drug is resorbed | If equilibrium is achieved, higher urine volume means higher CLr
73
What occurs when renal resorption rate = secretion rate?
CLr = fu x GFR
74
Which CYP enzymes are present in the kidneys?
CYP3A5 in cortex and medulla | CYP2D6 in PT and loop of Henle
75
Where are carboxylesterases present in the kidneys?
PT and bowman's capsule
76
What affect does CKD have on metabolism and elimination?
``` Reduced CLr and GFR Reduced kidney weight Reduced tubular secretion and transporter expression Reduced CLh Increased gastric emptying time and pH Downregulation of CYP enzymes ```
77
What is clearance?
CL= rate of elimination/concentration in plasma | Remains constant irrespective of dose if drug PK is linear
78
What are the two major sites of metabolism?
Liver and intestine
79
What is the elimination rate constant (k)?
The fractional rate of drug loss | k= Cl/V
80
What is flow rate (Q)?
CL= Q(Ca-Cv)/Ca
81
What is Fh?
Fraction escaping hepatic metabolism | Fh= 1-Eh
82
What are the three classes of hepatic extraction? Give examples.
``` Low extraction (<0.3) diazepam, warfarin, phenytoin Medium extraction (0.3-0.7) quinidine, codeine, cyclosporine High extraction (>0.7) propranolol, verapamil, lidocaine ```
83
What is the typically blood flow through the liver?
1300-1500mL/min
84
What is the additivity of clearance?
The total clearance is the addition of the individual organ CL values.
85
What is unique about the hepatic blood supply?
``` It has a dual supply: Portal vein (1.1L/min) Hepatic artery (0.4L/min) ```
86
What can affect the hepatic flow rate?
Increase- bed rest, thyrotoxicosis, isoprenaline | Decrease- exercise, heart failure, propranolol
87
What can affect plasma protein binding?
Increase- burns, neonate, nephrosis, heart failure | Decrease- stress, cancer, arthritis, crohn's, myalgia
88
What physiological effects does liver cirrhosis have?
``` Decreased liver volume Portal hypertension Renal impairment CLh reduced Impaired albumin synthesis and protein binding ```
89
What parameters are affected by hepatic extraction?
Hepatic clearance only affects drugs with high Eh | Plasma protein binding is important for low Eh drugs
90
What are the structural requirements for biliary excreted molecules?
Active facilitated transport Polar MW >350 Da
91
What is the average bile flow?
0.5-0.8mL/min
92
What is bile clearance?
CLb = (bile flow x drug concentration)/drug concentration in plasma
93
Describe enterohepatic recirculation. Give examples of drugs.
Liver-> common bile duct-> small intestine-> superior mesenteric vein-> portal vein-> liver Occurs with bile salts, rosuvastatin, mycophenolic acid
94
Briefly describe how CYP enzymes are classified.
12 families total, 1-4 important in drug metabolism | >60% homology in subfamilies
95
Which letter/number denotes the family, subfamily and gene? CYP2D6
Family- 2 Subfamily- D Gene- 6
96
Describe the activity of CYP enzymes in disease.
Reduced activity of CYP2D6 and CYP3A4 in cancer and HIV | Downregulation of CYPs mediated by pro-inflammatory mediators
97
What are the major roles of CYP450?
Membrane bound haem containing proteins | Activation of oxidation and oxidation of drug
98
What are the binding properties of CYP450 enzymes?
Substrate binding at protein active site Haem ligand for oxygen and CO binding CO binding has absorption spectrum max at 450nm
99
Describe the 6 stages of the CYP cycle?
1- substrate binds to active site, enzyme conformation changes 2- transfer of electron (NADPH-> NADP+) via flavoprotein-1 3- binding of oxygen to harm 4- transfer of second electron, reducing dioxygen to negatively charged peroxy group (NADPH-> NADP+) 5- peroxy group protonated twice forming highly reactive Fe 6- substrate reacts with Fe species, releases hydroxylated product, water molecule returns to distal haem position
100
Describe the preference for substrates of CYP2D6? Give an example.
Arylalkylamines (basic) with a site of oxidation 5-7 atoms from protonated nitrogen. i.e. ecstasy
101
Describe the preference for substrates of CYP2C9? Give an example.
Neutral or acidic molecules with oxidation site 5-8A from H-bond donor heteroatom Generally amphipathic with lipophilic region at site of hydroxylation Hydrophilic area around H-bond forming region i.e. tolbutamide
102
Describe the preference for substrates of CYP3A4? Give an example.
Neutral or basic molecules Lipophilic and bulky Site of oxidation often basic nitrogen i.e. testosterone or midazolam
103
Describe the multimodal distribution of CYP2D6.
7% of caucasians are poor metabolisers, lacking CYP2D6 gene | 2% of Swedish, 10% of Spanish and 8% of Italians are ultra-metabolisers, have gene duplication
104
What is the role of phase I metabolism?
Expose functional groups via oxidation at C, N or S atom
105
What is phase 1 hydroxylation?
Aromatic oxidation to form phenols i.e. lignocaine | Aliphatic oxidation to form alcohols i.e. pentobarbitone or tolbutamide
106
What is phase 1 cleavage?
O-dealkylation to form phenols i.e. codeine | N-dealkylation to form amines i.e. theophylline
107
What is phase 1 reduction?
Nitro-reduction i.e. chloramphenicol, nitrazepam | Azo-reduction i.e. prontosil
108
What is phase 1 hydrolysis?
Catalysed by carboxylesterases Ester hydrolysis i.e. oseltamivir Amine hydrolysis i.e. procainamide
109
What is phase II metabolism?
Conjugation of phase I metabolites for renal and biliary excretion via efflux transporters
110
What is glucuronidation?
Occurs via UDO-glucuronosyltransferases on luminal side of the endoplasmic reticulum Glycoproteins (UGT) catalyse addition of glucuronic acid
111
What are the two key paediatric clinical differences in glucuronidation?
``` Morphine glucuronidation (UGT2B1) is deficient in young adults UGT1A1 reaches adult levels at 3-6 months, congenital jaundice due increased bilirubin ```
112
What is Gilbert's syndrome?
Unconjugated hyperbilirubinaemia in adults due to deficiency of UGT1A1
113
What are the three most abundant glucuronidation enzymes in healthy kidneys and tumour tissues?
UGT1A9 UGT2B7 UGT1A6
114
How does CKD affect glucuronidation?
Inhibition of UGT enzymes by uremic toxins
115
What is phase II sulphation?
Occurs mainly via cytosolic enzymes, activated by ATP cofactor PAPs SULT enzymes present in liver and small intestine Metabolism of drugs such as paracetamol and salbutamol
116
What is glycine conjugation?
Phase II reaction limited to acids | Drug becomes activated as acyl coenzyme A intermediate
117
What is acetylation?
Phase II reaction limited to amines | Formation of N-acetyl conjugates via acetyl CoA
118
What is phase III metabolism?
Prolongs drug effect via action of glucuronidase of sulphates enzymes reversing phase II metabolism
119
Describe the 3 major metabolites of diazepam.
Temazepam and oxazepam are rapidly metabolised and barely detectable in the plasma but marketed as short acting Nordazepam has a longer half life and can cause local toxicity at site of formation i.e. liver injury
120
What is benzopyrene?
A major polycyclic aromatic hydrocarbon in cigarette smoke | Forms a reactive metabolite in the lung causing irritation
121
What is glutathione conjugation?
Protective mechanism | Electrophilic phase I metabolic interacts with nucleophilic glutathione tripeptide
122
What occurs in paracetamol overdose?
Electrophilic metabolite forms (NAPQI) which leads to liver cell death N-acetylcystiene is a well tolerated nucleophile that can be given as an antidote.
123
What is a type A adverse drug reaction?
Reversible reaction that is dose related | Causes 80% of ADRs
124
What is a type B adverse drug reaction?
Irreversible, idiosyncratic ADR with a genetic basis, often associated with the liver
125
What is a type C adverse drug reaction?
Irreversible chemical reaction with tissue macromolecule often leading to necrosis
126
What is a type D adverse drug reaction?
Irreversible ADR that is chronic, response is often delayed
127
Under what circumstances, according to the FDA, should a drug metabolite be evaluated as an enzyme inhibitor?
Less polar and present at >25% of parent systemic exposure | More polar and present at >100%