Pharmacology Flashcards
What is the therapeutic window?
range of drug concentrations where a clinically useful effect is exerted, but not a exerting toxic effect
Give examples of drugs with a narrow therapeutic window
- Warfarin
- Aminophylline
- Digoxin
- Aminoglycosides
What pharmacokinetic drug interactions can occur in absorption?
changes in gut motility
- slow down eg. morphine leads to enhanced absorption
- speed up eg. metoclopramide impairs absorption
drugs can bind to other drugs and reduce their absorption
What pharmacokinetic drug interactions can occur in distribution?
compete for a common binding site
displacement of object drug by precipitant drug -> total plasma [object drug] increased transiently
Why is the rise in [object drug] only transient after displacement by a precipitant drug?
increased elimination of object drug due to increased conc
What pharmacokinetic drug interactions can occur in metabolism?
induction or inhibition of CYP450
What pharmacokinetic drug interactions can occur in excretion?
changes in protein binding
- increases/deceases conc free drug. unbound drugs can be excreted
inhibition tubular secretion
- results in increased plasma conc
changes to urine flow/pH
- acidic urine = alkaline drugs eliminated at faster rate
- alkaline urine = acidic drugs eliminated at faster rate
Which drug groups commonly contribute to drug-frug interactions?
- Anticonvulsants
- Anticoagulants
- Antidepressants
- Antibiotics
- Antiarrhythmics
What effects does induction of CYP450 enzymes have?
accelerates drug metabolism
increases toxicity of drugs that have toxic metabolites
more rapid elimination
How does induction of CYP450 occur at a cellular level?
implemented by increased transcription, translation or slower degradation of CYP450
How long does it take for induction of CYP450 to happen?
1-2 weeks
Give examples of drugs that induce CYP450
- Phenytoin
- Carbamazepine
- Barbiturates
- Rifampicin
- Alcohol (chronic)
- Sulphonylureas and St John’s Wort
What effects does inhibition of CYP450 enzymes have?
slows drug metabolism
increases action of drugs
How long does it take for inhibition of CYP450 to happen?
few days
Give examples of drugs that inhibit CYP450
- Omeprazole
- Disulfiram
- Erythromycin
- Valproate
- Isoniazid
- Cimetidine and ciprofloxacin
- Ethanol (acute)
- Sulphonamides
How does cranberry affect CYP450?
inhibits CYP2C9
reduces clearance of warfarin
raises INR and increases risk of haemorrhage
How does grapefruit affect CYP450?
inhibits CYP450
reduces clearance
eg. simvastatin
How does renal disease cause disease-drug interactions?
falling GFR
reduced clearance of renally excreted drugs
How do NSAIDs and ACEi affect the kidney?
reduce GFR
due to decreased blood flow
How does hepatic disease cause disease-drug interactions?
reduced activity CYP450
reduced clearance of hepatically metabolised drugs
longer half lives
How does cardiac disease cause disease-drug interactions?
reduced cardiac output
reduced organ perfusion
reduced hepatic and renal blood flow
reduced blood clearance
How does hypoalbuminaemia cause disease-drug interactions?
higher free drug levels
increased free drug conc
increased clearance
What is an ADR?
an unwanted or harmful reaction
which occurs after administration of a drug or drugs
and is suspected or known to be due to the drug
What is an on-target ADR?
an ADR due to the exaggerated therapeutic effect of the drug
most likely due to increased dosing or due to factors affecting the PK or PD
includes effects on the same receptor type but found in other tissues.
What is an off-target ADR?
an ADR involving the drug interacting with other receptor subtypes secondarily to the one intended for the therapeutic effect.
also occurs with metabolites that can subsequently act as a toxin and Inappropriate immune responses
Under what circumstances are drug interactions more likely to occur?
• polypharmacy
• ignorant, inappropriate or reckless prescribing
• patients at the extremes of age
o renal and hepatic insufficiency
o comorbidities
• multiple medical problems
• use of drugs with a narrow therapeutic index
o greater risk of toxicity
• drugs used ear their minimum effective dose
o greater risk of treatment failure if metabolism increased
What can alter the absorption of a drug into the GI system?
gastrointestinal motility splanchnic blood flow molecular size pH levels presence of active transport systems first pass metabolism
What is first pass metabolism?
any metabolism that occurs before the drug enters the systemic circulation
occurs within gut lumen, gut wall and in the liver
How are drugs metabolised in the gut lumen?
gastric acid
proteolytic enzymes
How is drug absorption affected by the gut wall?
P-glycoprotein efflux pumps drugs out of enterocyte and back into the lumen
Define bioavailability
the amount of drug which reaches the systemic circulation in an unchanged form (once overcome all barriers to its absorption) relative to that if it was administered via IV.
what is bioavailability affected by?
drug preparation intestinal motility, food (lipid-soluble or water-soluble), age, first-pass metabolism.
therefore it varies between patients
What factors affect the distribution of a drug through the body?
lipophilic/hydrophilic
protein binding
also:
disease states
regional blood flow
specific receptor sites in tissues
If a drug is lipophilic, how does this affect it’s distribution?
it will move out of the plasma into tissues with a higher lipid content
reaches all body compartments and accumulate in fat
If a drug is lipid-insoluble, how does this affect it’s distribution?
the drug is mainly confined to the plasma an interstitial fluid
If a drug is not lipid soluble or is highly plasma bound, what does this mean for it’s volume of distribution and plasma concentration?
low Vd
therefore plasma concentration remains high
If the drug can diffuse out of the plasma compartments and pass into other fluid compartments, what does this mean for it’s volume of distribution and plasma concentration?
high Vd
plasma conc falls
If a drug is highly bound by tissue proteins, what does this mean for it’s volume of distribution and plasma concentration?
Vd becomes very high
large proportion of drug removed from fluid compartments
low plasma conc
Why are acute increases in Vd seen in sepsis?
increased vascular permeability
Describe Phase 1 drug metabolism in the liver
dependent on CYP450
catabolic
produces products that are chemically active
Describe Phase 2 drug metabolism in the liver
anabolic
involve conjugation
less pharmacologically active and less lipid-soluble products
What factors determine the rate of renal excretion of drugs?
GFR
passive tubular secretion
active tubular secretion
renal blood flow
plasma protein binding
tubular urinary pH
Why would a drug need to be monitored?
- Zero-order kinetics
- Long half life
- Narrow therapeutic window
- High risk of drug-drug interactions
- Known toxic effects
How can steady state (CpSS) be calculated?
CpSS = dose rate / clearance
How can a loading dose be calculated?
Loading Dose (DoseL) = Vd x CpSS
How are acidic drugs actively secreted in the proximal tubule?
OAT
transports in negatively charged anionic form
can transport against electrochemical gradient
How are basic drugs actively secreted in the proximal tubule?
OCT
in their protonated cationic form
only down a electrochemical gradient
State some of the actions of insulin
- Activates GLUT4 to relocate to the cell membrane, to increase uptake of glucose into cells
- Inhibits glycogen breakdown and enhances uptake by liver and adipose tissue
- Stimulates fatty acid synthesis for transport as lipoproteins and inhibits lipolysis in adipocytes
- Inhibits proteolysis to reduce amino acid levels in the blood
What is the difference between type 1 and type 2 diabetes?
Type 1 = absolute insulin deficiency from destruction of β-cells in the pancreas; = “insulin-dependent diabetes”
Type 2 diabetes is due to insulin resistance and progressive insulin deficiency
What lifestyle changes can act to control diabetes a first?
limiting fat intake increasing proportion of complex carbohydrates reduction in alcohol smoking cessation increase exercise
Give the name of a biguanide drug
metformin
Describe the mechanism of action of metformin
- increasing cells sensitivity to insulin
- reduces hepatic gluconeogenesis – primary effect
- increased glucose intake and utilisation in skeletal muscle
- reduced carbohydrate absorption from the intestine
- increased fa oxidation
- reduced circulating LDLs and VLDLs
Describe the pharmacokinetics of metformin
oral administration half-life 2-3 hours no binding to plasma proteins no metabolism renal elimination
Why is metformin the first line therapy for type 2 diabetes?
can reduce HbA1c by 2%
does not stimulate appetite
weight neutral
What are metformin’s ADRs?
Gi disturbances
interferes with absorption of vitamin B12 in long term
Why I smetformin contraindicated in patients with compromised HRH function or respiratory disease?
risk of lactic acidosis
reduced tissue perfusion
Give an example of a sulphonylurea
gliclazide
Describe the mechanism of action of sulphonylureas
bind to and antagonise β-cells’ K+-ATP channel activity,
increases K+ concentration within the cell
leads to depolarisation.
increases Ca2+ ion entry into the cell
increases insulin release from β-cells.
Describe the pharmacokinetics of sulphonylureas
oral
bind to albumin - 99% affinity
metabolised in liver
excreted by kidneys
Why are sulphonylureas contraindicated in pregnancy?
can cross placenta and enter breast milk
Describe the ADRs of sulphonyureas
hypoglycaemia
stimulate appetite
weight gain
Give the names of two thiazolidinediones
Rosiglatazone
pioglatizone
Describe the mechanism of action of thiazolidinediones
agonistically bind to nuclear hormone receptor site the peroxisome proliferator-activated receptor-γ or PPAR- γ (mainly found in adipose tissue, but also in muscle and liver)
Activation of PPARγ regulates the transcription of several insulin responsive genes
results in increased insulin sensitivity in adipose tissue, liver, and skeletal muscle
There is differentiation of adipocytes, increased lipogenesis and increased uptake of fatty acids and glucose. It also promotes amiloride sensitive sodium ion reabsorption, which leads to fluid retention.
Describe the pharmacokinetics of thiazolidinediones
oral rapidly absorbed 99% albumin affinity metabolised by CYP450 pioglatizone eliminated in bile rosiglatoazobe renal elimination
Describe the ADRs of thiazolidinediones
liver toxicity
weight gain - increase in subcutaneous fat
fluid retention
osteopenia and increased fracture risk
State the mechanism of action of acarbose
reversibly Inhibits intestinal α glucosidase.
delays carbohydrate absorption from gut
reducing increase in blood glucose after a meal.
Describe the pharmacokinetics of acarbose
oral
poorly absorbed
metabolised by intestinal bacteria
some of the metabolites are absorbed and excreted into the urine
Describe the ADRs of acarbose
flatulence, loose stools and diarrhoea, abdominal pain and bloating
Describe the mechanism of action of Glucagon-like Peptide 1 (GLP1) Analogues
lowers blood glucose after a meal by:
increasing insulin secretion, -> increases peripheral glucose uptake
supressing glucagon secretion -> reduced hepatic glucose output
slowing gastric emptying.
So:
They reduce food intake and are associated with weight loss. They reduce hepatic fat accumulation.
Describe the pharmacokinetics of Glucagon-like Peptide 1 (GLP1) Analogues
SC twice daily
Describe the ADRs of Glucagon-like Peptide 1 (GLP1) Analogues
hypoglycaemia
gastrointestinal effects
Describe the mechanism of action of Dipeptyl-peptidase 4 (DPP4) Inhibitors
competitively inhibit DPP-4 - normally responsible for breakinf down GLP-1
increases endogenous GLP-1 concentration after eating.
GLP-1 stimulates insulin secretion
weight neutral
Describe the pharmacokinetics of Dipeptyl-peptidase 4 (DPP4) Inhibitors
oral
well absorbed
Saxagliptin metabolised by CYP450 to active metabolite
renal excretion
Describe the ADRs of Dipeptyl-peptidase 4 (DPP4) Inhibitors
nasopharyngitis
headache
Although infrequent, pancreatitis
How is gradation of glycaemic control achieved using exogenous insulins?
variation in pharmaceutical preparation
single amino acid substitutions
Describe a ‘pre-mixed’ insulin regime
injected twice a day, with morning and evening meals
mixture of short and long acting insulin
Describe a Basal Bolus insulin Regimen
intermediate/long lasting insulin to provide a basal level that extends overnight.
supplemented with short acting insulin injected with meals to provide acute response,
This involves injecting around 5 times a day yet does provide good flexibility and better control
Describe the action of short-acting insulin
onsets around 30min after injected
peak around 2-5h after injections
can be given just before a meal.
Describe the action of intermediate-acting insulin
onsets around 2h after injected
peak around 4-10h after injection
can be given as basal control
Describe the action of long-acting insulin
onsets around 5h after injected
peak around 8-30h after injection
can be given as basal control
Describe the treatment steps in type 2 diabetes
lifestyle
metformin
HbA1c >7% = sulphonylureas
HbA1c >7.5% = TZDs/insulin
What needs to be monitored during diabetes treatment?
glycaemic levels dietary habit HbA1c renal function hepatic function neurological function cardiovascular function
State an anti-obesity lipase inhibitor drug
Orlistat
Describe the mechanism of action of orlistat
inhibits gastric and pancreatic lipase
decreases the breakdown of dietary fat
decreases fat absorption by about 30%. T
The loss of calories from decreased absorption of fat is the main cause of weight loss.
Describe the pharmacokinetics of orlistat
oral
minimal systemic absorption
mainly excreted in the faeces
Describe the ADRs of orlistat
oily spotting, flatulence with discharge, fecal urgency, increased defecation Pancreatitis liver injury interferes with the absorption of fat-soluble vitamins and β-carotene, as well as some drugs
State the names of some short-acting insulin drugs
Actrapid,
Humulin S,
Novorapid
State the names of some intermediate-acting and long-acting insulin drugs
Insulin Glargine,
Isophane insulin
What is the method of administration of insulin?
Subcutaneous injection
What levels should cholesterol, LDLs and HDLs be at?
Total cholesterol 5.0mmol/L or below
Fasting LDL 3mmol/L or below
HDL 1.2mmol/L or above
Give two examples of a statins
Simvastatin
Atorvastain
What is the mechanism of action of statins?
Inhibit HMG-CoA reductase in hepatocytes
Decreases hepatic cholesterol synthesis
Up regulates LDL receptor synthesis
Increases LDL clearance from plasma into hepatocytes
Reduce LDL
Give an example of a cholesterol lipase inhibitor
Ezetimibe
Describe the mechanism of action of ezetimibe
Blocks cholesterol transport protein NPC1L1 in brush border of intestine
Inhibits intestinal cholesterol uptake
Reduces dietary cholesterol reaching liver
Up regulates LDL transporter expression
Reduces circulating Cholesterol levels
NB does not affect absorption of fat soluble vitamins, TAGs or bile acids
Describe the pharmacokinetics of cholesterol lipase inhibitors
Metabolised in brush border to active metabolite
Repeated enterohepatic circulation
Slowly eliminated
Describe ADRs of cholesterol lipase inhibitors
Abdominal pain
Diarrhoea
Head aches
Give an example of a fibrate
Bezafibrate
Describe the mechanism of action of fibrates
Stimulate PPAR-alpha (Peroxisome proliferator-activated receptor-alpha)
Increases production of lipoprotein lipase
Increases hepatic LDL uptake
Lowers TAGs
Slightly Lowers LDL and raises HDL
Give examples of the ADRs of fibrates
Rhabdomyolysis resulting in AKI
GI upsets
Rash
Pruritis
What is the mechanism of action of nicotinic acid?
Inhibits hepatic VLDL secretion
Reduces circulating triglyceride and LDL
Raises HDL
What are the ADRs of nicotinic acid?
Flushing
Palpitations
GI disturbance
What levels should cholesterol, LDLs and HDLs be at?
Total cholesterol 5.0mmol/L or below
Fasting LDL 3mmol/L or below
HDL 1.2mmol/L or above
Give two examples of a statins
Simvastatin
Atorvastain
What is the mechanism of action of statins?
Inhibit HMG-CoA reductase in hepatocytes
Decreases hepatic cholesterol synthesis
Up regulates LDL receptor synthesis
Increases LDL clearance from plasma into hepatocytes
Reduce LDL
Give an example of a cholesterol lipase inhibitor
Ezetimibe
Describe the mechanism of action of ezetimibe
Blocks cholesterol transport protein NPC1L1 in brush border of intestine
Inhibits intestinal cholesterol uptake
Reduces dietary cholesterol reaching liver
Up regulates LDL transporter expression
Reduces circulating Cholesterol levels
NB does not affect absorption of fat soluble vitamins, TAGs or bile acids
Describe the pharmacokinetics of cholesterol lipase inhibitors
Metabolised in brush border to active metabolite
Repeated enterohepatic circulation
Slowly eliminated
Describe ADRs of cholesterol lipase inhibitors
Abdominal pain
Diarrhoea
Head aches
Give an example of a fibrate
Bezafibrate
Describe the mechanism of action of fibrates
Stimulate PPAR-alpha (Peroxisome proliferator-activated receptor-alpha)
Increases production of lipoprotein lipase
Increases hepatic LDL uptake
Lowers TAGs
Slightly Lowers LDL and raises HDL
Give examples of the ADRs of fibrates
Rhabdomyolysis resulting in AKI
GI upsets
Rash
Pruritis
What is the mechanism of action of nicotinic acid?
Inhibits hepatic VLDL secretion
Reduces circulating triglyceride and LDL
Raises HDL
What are the ADRs of nicotinic acid?
Flushing
Palpitations
GI disturbance
What serum levels need to be checked when taking statins and why?
transaminase - risk of liver failure
creatine kinase - risk of Myopathy and rhabdomyolysis
Describe the effects of drug interactions between statins and CYP inducers
decrease the plasma levels of statin
Describe the effects of drug interactions between statins and CYP inhibitors
increase the risk of myopathy and other side effects
How are statin used in primary prevention?
primary prevention of arterial disease in patients who are at high risk because of elevated serum cholesterol.
How are statin used in secondary prevention?
secondary prevention of MI and stroke in those who already have symptomatic atherosclerosis.
Describe the transport of oestrogens in the blood
most bound to albumin or sex-hormone binding globulin
2% travels as free hormone
How does oestrogen bind ot oestrogen receptors?
travels across cell membranes
binds to nuclear receptors - ERα and ERβ
What effect does oestrogen binding to its receptor cause?
the oestrogen receptors form dimers once oestrogen binds
activated steroid-receptor complex interacts with nuclear chromatin
initiates hormone-specific RNA synthesis.
results in the synthesis of specific proteins that mediate physiologic functions.
what effect does oestrogen have on progesterone?
Oestrogens induce PR expression in endometrial cells, so act to condition the endometrium
Describe the ligand-receptor interactions of progesterone
Progesterone binds to the dimeric progesterone receptor, PR,
induces secretory changes the endometrium
What are the effects of progesterone in females?
promotes the development of a secretory endometrium
high levels of progesterone inhibit the production of gonadotropin and, therefore, prevent further ovulation.
If conception takes place, progesterone continues to be secreted, maintaining the endometrium and reducing uterine contractions.
increases bone density
fluid retention
What are the side effects of progesterone?
- Weight gain
- Fluid retention
- Acne
- Nausea and vomiting
- Irritability
- Lack of concentration
headache
depression
increased risk breast cancer
What are the systemic effects of oestrogen in females?
- Mild anabolic
- Sodium and water retention
- Raise HDL, lower LDL
- Decrease bone reabsorption
- Improve blood coagulability
What are the side effects of oestrogen?
- Nausea and vomiting
- Water retention and peripheral oedema
- Risk of thromboembolism and MI
- Impaired glucose tolerance
- Endometrial hyperplasia and cancer
breast tenderness
Describe the pharmacokinetics of oestrogen
low oral bioavailability due to first pass metabolism
hydroxylated in liver by CYP450 , then subsequently glucuronidated or sulfated.
excreted into bile and then reabsorbed through the enterohepatic circulation
Inactive products are excreted in urine
Describe the pharmacokinetics of progesterone
rapid absorption orally
metabolised in liver by CYP450 - glucaronidation
short half life
excreted primarily by the kidney
What is the COCP?
oestrogens in combination with progesterone
Describe the mechanism of action of the COCP
Oestrogen inhibits secretion of FSH by –ve feedback at the anterior pituitary.
This supresses follicular development in the ovary.
Progesterone inhibits secretion of LH and therefore prevents ovulation.
What is the difference between monophasic and triphasic COCP?
mono = dose of oestrogen and progesterone is constant in the active tablets
tri = attempt to mimic the natural female cycle and most contain a constant dose of estrogen with increasing doses of progestin given over three successive 7-day periods
How should COCP be taken?
The combined pill is usually taken for 21 days followed by 7 placebo pill days. This causes a withdrawal bleed.
What is the POP?
progesterone only pill
what is the mechanism of action of POP?
cause cervical mucus thickening
preventing sperm movement into uterus
In which patients is POP commonly used?
offered to women for whom the COCP is contraindicated
e.g. risk factors for venous thromboemboli, smokers, or hypertensive.
How is POP taken?
at the same time each day, every day
What are the problems with POP?
less effective than COCP
may produce irregular menstrual cycles and spotting more frequently
What are the ADRs of COCP?
increased risk of DVT raising blood pressure increased risk of gall stones, decrease glucose tolerance, nausea, weight gain (due to anabolic state or fluid retention) increased risk of breast cancer
Describe the Drug interactions of COCP
CYP450 inducers (increase plasma clearance) and inhibitors (decrease plasma clearance)
Name some CYP450 inducers
PCBRAS Phenytoin, Carbamazapine, Barbiturates, Rifampicin/rifabutin, Alcohol, Sulphonylureas/St John’s Wort
why is the dose of oestrogen kept to a minimum?
avoid excess risk of thromboembolism
How do broad-spectrum antibiotics affect plasma concentration of the COCP?
reduce efficacy of COCP
the effect they have on intestinal flora will reduce their re-uptake into the circulation (by lowering enterohepatic recycling)
What is the menopause?
cessation of menstrual periods
What is the perimenopause?
time shortly before the occurrence of the menopause
Why are sex steroids given in menopause?
to reduce the symptoms of the menopause
and to prevent osteoporosis
What is ERT?
just oestrogen given in menopause
what is HRT?
Why could this be better than ERT?
oestrogen + progesterone.
Reduces risk of endometrial carcinoma
Why is history of a hysterectomy important when prescribing HRT?
For women who have an intact uterus, a progestogen is always included with the estrogen therapy, because the combination reduces the risk of endometrial carcinoma associated with unopposed estrogen.
For women who have undergone a hysterectomy, unopposed estrogen therapy is recommended because progestins may unfavourably alter the beneficial effects of estrogen on lipid parameters
Describe the side-effects of HRT
increased risk of stroke
increases HDLs
increased risk venous thromboembolism
decreased resorption of bone
What is the difference between bacteriostatic and bacteriacidal drugs?
Bacteriostatic drugs arrest the growth and replication of bacteria, limiting the spread of infection
Bactericidal drugs kill bacteria
What is an MIC?
Minimum inhibitory concentration
lowest concentration of antibiotic that inhibits bacterial growth
What is MBC?
Minimum bactericidal concentration
lowest concentration of antimicrobial agent that results in a 99.9 percent decline in colony count after overnight broth dilution incubations
Which antibiotics affect DNA synthesis?
quinolones
folic acid antagonists
Which antibiotics affect protein synthesis?
aminoglycosides
macrolides
tetracyclines
Which antibiotics affect cell wall synthesis?
beta-lactams - (penicillins and cephalosporins)
glycopeptides
Give some examples of penicillins
Penicillin,
Amoxicillin,
Flucloxacillin,
Co-amoxiclav
Describe the mechanism of action of penicillins
bind to penicillin binding proteins in peptidoglycan cell wall
inhibit transpeptidation enzyme that cross-links peptidoglycan chains
inactivates inhibitor of autolytic enzymes
cell lysis occurs
bacteriacidal
Describe the antibacterial spectrum of penicillins
gram positive microorganisms have cell walls that are easily traversed by penicillins so are susceptible
Gram-negative microorganisms have an outer LPS envelope surrounding the cell wall that acts as a barrier to the water-soluble penicillins.
BUT gram-negative bacteria have proteins inserted in the lipopolysaccharide layer that act as water-filled channels (called porins) to permit transmembrane entry
How is the GI absorption of penicillins affected by food?
most incompletely absorebed
BUT amoxicillin almost completely absorbed
absorption decreased by food in the stomach, because gastric emptying time is lengthened, and the drugs are destroyed in the acidic environment
administered 30 to 60 minutes before meals or 2 to 3 hours postprandial.
How are penicillins excreted?
organic acid (tubular) secretory system glomerular filtration.
State the ADRs of penicillins
hypersensitivity
diarrhoea
nephritis
neurotoxicity
Name some cephalosporins
Cefaclor,
Cefotaxime,
Aztreonam
Describe the mechanism of action of cephalosporins
bind to penicillin binding proteins in peptidoglycan cell wall
inhibit transpeptidation enzyme that cross-links peptidoglycan chains
inactivates inhibitor of autolytic enzymes
cell lysis occurs
bacteriacidal
How are cephalosporins administered?
most IV or IM
poor oral absorption
Which cephalosporins are readily able to cross the BBB?
ceftriaxone
cefotaxime
therefore used in meningitis
How are cephalosporins excreted?
tubular secretion and/or glomerular filtration
Describe the ADRs of cephalosporins
hypersensitivity reactions.
Nephrotoxicity
Diarrhoea common, leading to C. diff.
Name some tetracyclines
Tetracycline,
Doxycycline,
Oxytetracycline
Describe the mechanism of action of tetracyclines
enter into organisms by passive diffusion or energy dependant transport
organism accumulates drug
binds to 30S subunit of bacterial ribosome, blocking accses of amino-acyl tRNA
prevents translation
protein synthesis inhibited
bacteriostatic
Describe the antibacterial spectrum of tetracyclines
broad spectrum
gram +ve
gram -ve
Mycoplasma
Chlamydia
Describe the pharmacokinetics of tetracyclines
oral well absorbed (especially doxycycline) cross placental barrier metabolised in liver excreted in bile enterohepatic circulation enter urine by glomerular filtration
What affects the absorption of tetracyclines in the gut?
form complexes with metal ions, so absorption decreased in presence of: dairy products iron preparations antacids
Can tetracyclines be given to pregnant or breast feeding women?
no
cross placenta and enter breast milk
deposited in fetal bone and teeth
Describe the ADRs of tetracyclines
irritation gastric mucosa depostioni n frug and bones liver failure phototoxicity - increased risk of burning in sunlight vertigo
Name some aminoglycosides
gentamicin
neomycin
Describe the mechanism of action of aminoglycosides
orgs allow drug to enter cell
binds to 30S subunit before ribosome formation
interferes with assembly of functional ribosome
protein synthesis cannot be initiated
bactericidal
Describe the antibacterial spectrum of aminoglycosides
broad spectrum
some gram +ve
aerobic gram –ve
Describe the pharmacokinetics of aminoglycosides
IV or intrathecally
high conc in renal cortec and endolymph and perilymph of inner ear
no metabolism in host
excreted in urine by glomerular filtration
Describe the ADRs of aminoglycosides
ototoxicity
nephrotoxicity
paralysis
Can aminoglycosides be given to pregnant women?
no
crosses placenta
accumulates in fetal plasma and amniotic fluid
What needs to be monitored when taking aminoglycosides?
plasma concentration
Name some macrolides
Erythromycin,
Clarithromycin
Describe the mechanism of action of macrolides
irreversibly bind to 50s subunit
inhibiting translocation in protein synthesis
bacteriostatic
Describe the antibacterial spectrum of macrolides
erythromycin = similar to penicillins (gram +ve and gram -ve)
clarithromycin = similar to erythromycin, but also H influenae, Chlamydia and H pylori
Describe the pharmacokinetics of macrolides
oral or IV
good GI absorption
erythromycin = excreted in active form in bile and partial enterohepatic circulation. inactive form by kidneys
clarithromycin eliminated by kidneys
Describe the drug interactions of macrolides
inhibit CYP450
Describe the ADRs of macrolides
GI disturbance
jaundice
ototoxicity
Describe the mechanism of action of vancomycin
inhibits synthesis bacterial cell wall phospholipids and peptidoglycan polymerisation
Describe the antibacterial spectrum of vancomycin
gram +ve orgs
MRSA
Enterococci
Describe the pharmacokinetics of vancomycin
IV infusion
(oral in C. diff)
eliminated by glomerular filtration
Describe the ADRs of vancomycin
fever rashes chills phlebitis ototoxicity nephrotoxicity
What determines bacterial killing?
time
concentration above the MIC
What is time-dependant killing?
major killing affect of an antibiotic depends on how long the drug is bound to the receptor
What is concentration-dependant killing?
major killing affect of an antibiotic depends on their concentration
Describe the steps of Influenza virus replication?
- Binding and adsorption, facilitated by haemagglutinin glycoprotein surface molecule
- endocytosis into the cell. ATP-driven entry into the endosome. Protons gain entry into the virus via an M2-ion channel. The fall in pH within the virus allows for the viral coat of the nucleocapsid to breakdown.
- The viral RNA is synthesised and replicated to form viral proteins. The virus is then assembled within the cell
- virus is released from the cell via budding. Neuraminidase is a glycoprotein antigen found in the viral membrane and enables the virus to leave the host cell.
What is the role of the Influenza protein haemagglutinin?
facilitates adhesion of virus to host cell
What is the role of the Influenza protein Neuraminidase?
Neuraminidase is a transmembrane viral protein which enables newly formed virions to escape from their host cell.
As the newly formed virus egresses from its host cell many re-attach to the sialic acid membrane glycoprotein residues on the cell membrane.
To get released and infect other host cells they need to break this bond. This is carried out by the viral neuraminidase.
What is the role of the Influenza M2 ion channel??
allows H+ ions to enter the endosome, causing the viral coat of the nucleocapsid to break down
Describe the differences between the Influenza virus classes
A - multiple host species, greatest severity, undergoes antigenic drift and shift
B - no animal host, low severity
C - common cold like
Name some M2 ion channel blockers
amantadine
rimantadine