Pharmacology and prescribing 2 Flashcards
what toxicities are tested for in animals during drug development?
carcinogenicity, teratogenicity and organ-specific toxicities
what are adverse drug reactions?
- injuries following administration of a drug or combination of drugs under normal conditions of use
- unwanted or harmful effects that occur in humans after it has been marketed, due to adverse effects not being seen in animals
- suspected to be related to the drug
- has to be noxious and unintended
what are examples of adverse drug reactions that occur during prolonged use of a drug?
- osteoporosis during continued high-dose glucocorticoid therapy
- tardive dyskinesia during continuous use of antipsychotics
what are examples of adverse drug reactions occurring on ending treatment?
- within a few days: tachycardia on abrupt discontinuation of beta-adrenoceptor blockade
- months/years after discontinuation: second malignancies following successful chemotherapy
who suggested classifying ADRs according to DoTS? what is DoTS?
- Aronson and Ferner (2003)
- dose, time course and susceptibility
who suggested the ABCDE approach to classifying ADRs?
Rawlins and Thomson (1985)
what are type A ADRs?
- adverse effects related to the known pharmacological actions of the drug are predictable
- augmented
- related to dose and individual susceptibility
- common
what are examples of type A ADRs?
- postural hypotension with alpha1-adrenoceptor antagonists
- bleeding with anticoagulants
- sedation with anxiolytics
- morphine and constipation
- hypotension and antihypertensive
how serious are type A ADRs?
- often is reversible, and the problem can be dealt with by reducing the dose
- can be serious (e.g. intracerebral bleeding caused by anticoagulants, hypoglycaemic coma from insulin)
- may not be easily reversible (e.g. drug dependence from opiod analgesics)
what is an example of type A ADRs leading to discrete events rather than graded symptoms?
- makes them difficult to detect
- drugs that block COX-2 (coxibs, e.g. rofecoxib, celecoxib, valdecoxib and NSAIDs) increase risk of MI in a dose-dependent manner
when can type B ADRs be predictable?
- if taken in excessive dose (e.g. paracetamol hepatotoxicity or aspirin-induced tinnitus)
- increased susceptibility (e.g. pregnancy)
- predisposing disorder
- mutation in mitochondrial DNA
what are type B ADRs?
- adverse effects unrelated to the known pharmacological action of the drug
- unpredictable
- idiosyncratic
what are type B ADRs often initiated by? what are examples of this?
- chemically reactive metabolite rather than the parent drug
- often immunological in nature
- drug-induced hepatic/renal necrosis, bone marrow suppression, carcinogenesis and disordered fetal development
what are examples of severe type B ADRs?
- aplastic anaemia from chloramphenicol
- anaphylaxis due to penicillin
what is involved in toxicity testing in animals?
- wide range of tests in different species
- long-term administration of the drug
- regular monitoring for abnormalities
- detailed postmortem examination to detect gross or histological abnormalities
what doses are used in toxicity testing?
doses well above the expected therapeutic range
- establishes which tissues or organs are likely targets of toxic effects of the drug
why are recovery studies in toxicity testing done?
to assess whether toxic effects are reversible, and looking for irreversible changes e.g. carcinogenesis or neurodegeneration
what are the ranges of toxic effects caused by a drug?
- can range from negligible to so severe that development of the compound is stopped
- intermediate levels of toxicity are more acceptable in drugs for severe illnesses
when can safety of a drug be determined?
only during use in humans; toxicity can be detected in animals too
what are some tests of hepatic damage and renal function?
hepatic: levels of transaminase enzymes measured in blood plasma or serum
renal function: usually creatinine concentration
how does toxin-induced cell damage/death occur?
- necrosis
- apoptosis is increasingly recognised to be of equal or greater importance than necrosis, especially in chronic toxicity
- covalent or non-covalent mechanisms
- caused by reactive metabolites of the drug
what are potentially cytotoxic, non-covalent processes of drug-induced cell damage/death?
- lipid peroxidation
- generation of toxic ROS
- depletion of reduced glutathione (GSH)
- modification of sulfhydryl groups
what is the process of lipid peroxidation? what is it initiated by?
- initiated either by reactive metabolites or by ROS
- lipid peroxyradicals (ROO•) can produce lipid hydroperoxides (ROOH)
- ROOH produce further lipid peroxyradicals (ROO•)
what are defence mechanisms against lipid peroxidation?
GSH peroxidase and vitamin E
how does lipid peroxidation cause cell damage?
from alteration of membrane permeability or from reactions of the products of lipid peroxidation with proteins
what is the process of ROS production?
reduction of molecular oxygen to superoxide anion may be followed by enzymatic conversion to hydrogen peroxide, hydroperoxy and hydroxyl radicals or singlet oxygen
how does ROS production cause cell damage?
- ROS are cytotoxic, both directly and through lipid peroxidation
- important in excitotoxicity and neurodegeneration
what can protect cells from oxidative stress?
GSH redox cycle
how is GSH depleted? how is it usually maintained and regenerated?
- GSH can be depleted by accumulation of normal oxidative products of cell metabolism or by the action of toxic chemicals
- usually maintained in a redox couple with its disulfide, GSSG
- oxidising species convert GSH to GSSG
- GSH is regenerated by NADPH-dependent GSSG reductase
how can depletion of reduced glutathione damage cells?
- GSH redox cycle protects cells from oxidative stress
- when cellular GSH falls to about 20-30% of normal, cellular defence against toxic compounds is impaired and cell death can result
how can modification of sulfhydryl groups be produced?
- by oxidising species that alter sulfhydryl groups reversibly
- by covalent interaction
what is the role of free sulfhydryl groups?
catalytic activity of many enzymes
what are important targets for sulfhydryl modification by reactive metabolites?
- cytoskeletal protein actin GSH reductase
- Ca2+ transporting ATPases in the plasma membrane and ER
what is the action of the targets of sulfhydryl modification?
maintain cytoplasmic Ca2+ concentrations at about 0.1 mewmol/l in the face of an extracellular Ca2+ concentration of more than 1 mmol/l
what does inactivation of protein actin GSH reductase and Ca2+ transporting ATPases by sulfhydryl modification lead to?
- sustained rise in cell Ca2+
- this compromises cell viability
what are lethal processes that lead to cell death after acute Ca2+ overload caused by modification of sulfhydryl groups?
- activation of degradative enzymes (neutral proteases, phospholipases, endonucleases) and protein kinases
- mitochondrial damage
- cytoskeletal alterations (e.g. modification of association between actin and actin-binding proteins)
what are targets for covalent interactions?
DNA, proteins/peptides, lipids and carbohydrates
what are the roles of mutagenic/non-mutagenic chemicals?
- mutagenic chemicals covalently bond to DNA
- non-mutagenic chemicals form covalent bonds with macromolecules
what are examples of covalent interactions? what can they lead to?
- cholinesterase inhibitor paraoxon binds acetylcholinesterase at the NMJ and causes necrosis of skeletal muscle
- Amanita phalloides (from poisonous toadstool binds actin, and another binds RNA polymerase, interfering with actin depolymerisation and protein synthesis, respectively
- adduct formation between a metabolite of paracetamol and cellular macromolecules
what can covalent binding to proteins and DNA lead to?
- binding to protein can produce an immunogen
- binding to DNA can cause carcinogenesis and teratogenesis
what is liver damage manifested as?
manifested clinically as hepatitis or in laboratory abnormalities (e.g. increased activity of plasma aspartate transaminase, an enzyme released from damaged liver cells)
what is an enzyme that’s released by damaged liver cells?
plasma aspartate transaminase
in which cases of hepatotoxicity have genetic differences in drug metabolism been implicated?
isoniazid, phenytoin
what causes reversible obstructive jaundice?
chlorpromazine and androgens
what is the effect of a toxic dose of paracetamol?
enzymes catalysing the normal conjugation reactions are saturated, and P450 mixed-function oxidases convert the drug to the reactive metabolite N-acetyl-p-benzoquinone imine
what is the reactive metabolite of paracetamol?
N-acetyl-p-benzoquinone imine (NAPBQI)
what are the effects of NAPBQI?
- oxidation of SH groups on cellular Ca2+ ATPases
- lipid peroxidation
- NAPBQI-protein adducts
- GSH depletion -> oxidative stress
- NAPBQI-GSH adduct formation -> GSH depletion -> oxidative stress
all of these processes lead to cell death
what does regeneration of GSH depend on?
- regeneration of GSH from GSSG depends on the availability of cysteine
- intracellular availability of cysteine can be limiting
what can be substitute for cysteine?
acetylcysteine or methionine
what can be used to treat patients with paracetamol poisoning?
acetylcysteine or methionine (substitutes for cysteine which enables regeneration of GSH from GSSG)
what is an example of liver damage being caused by immunological mechanisms?
halothane hepatitis
what are types of causes of adverse effects by NSAIDs on the kidney? give examples
- principal pharmacological action (e.g. inhibition of prostaglandin biosynthesis)
- unrelated to principal pharmacological action (e.g. allergic-type interstitial nephritis)
- unknown whether or not related to principal pharmacological action (e.g. analgesic nephropathy)
what are the adverse effects of NSAIDs on the kidney by principal pharmacological action?
- acute ischaemic renal failure
- sodium retention (leading to or exacerbating hypertension and/or heart failure)
- water retention
- hyporeninaemic hypoaldosteronism (leading to hyperkalaemia)
what are the adverse effects of NSAIDs on the kidney by causes unrelated to principal pharmacological action?
- renal failure
- proteinuria
what are the adverse effects of NSAIDs on the kidney by causes unknown whether or not related to principal pharmacological action?
- papillary necrosis
- chronic renal failure
what are some of the commonest precipitants of acute renal failure?
NSAIDs and ACE inhibitors
what can renal damage cause?
- renal tubular cells are exposed to high concentrations of drugs and metabolites as urine is concentrated
- renal damage can cause papillary and/or tubular necrosis
- inhibition of prostaglandin synthesis by NSAIDs causes vasoconstriction and lowers GFR
how can a drug’s genotoxic potential be assessed?
- carry out a battery of in vitro and in vivo tests for genotoxicity
- usually comprise tests for gene mutation in bacteria, in vitro and in vivo tests for chromosome damage and in vivo tests for reproductive toxicity and carcinogenicity
how can drugs lead to carcinogenesis/mutation?
- chemical agents cause mutation by covalent modification of DNA
- certain mutations result in carcinogenesis, because the affected DNA sequence codes for a protein regulating cell growth
- mutations in proto-oncogenes (regulate cell growth) and tumour suppressor genes (code for products that inhibit transcription of oncogenes) are common
how do most chemical carcinogens act on DNA? what does this lead to?
- most act by modifying bases in DNA, particularly guanine
- O6 and N7 positions of guanine readily combine covalently with reactive metabolites of chemical carcinogens
- substitution at the O6 position is more likely to produce a permanent mutagenic effect
- N7 substitutions are quickly repaired
when is the accessibility of bases in DNA to chemical attack the greatest?
when DNA is in the process of replication (i.e. during cell division)
what are genotoxic carcinogens?
carcinogenic compounds that interact directly with DNA
what are epigenetic carcinogens?
carcinogenic compounds that act at a later stage to increase the likelihood that mutation will result in a tumour
what is the sequence of events in muatgenesis and carcinogenesis?
- primary carcinogen -> alteration of DNA (mutation)
- secondary carcinogen -> reactive metabolite via metabolising enzymes -> mutation
- co-carcinogen: amplifies mutations
- promotor: amplifies mutations leading to oncogene expression
- oncogene expression -> malignant transformation
what is the Ames test?
- test for mutagenicity
- widely used
- measures the effect of substances on the rate of back-mutation (reversion from mutant to wild-type form) in Salmonella typhimurium
what is back-mutation?
reversion from mutant to wild-type form
what does the Ames test involve?
- growing the mutant form on a medium containing a small amount of histidine, plus the drug to be tested
- after several divisions, the histidine becomes depleted
- only cells that continue dividing are those that have back-mutated to the wild type
- count of colonies following subculture on plates deficient in histidine gives a measure of the mutation rate
- a liver microsomal enzyme preparation for generating reactive metabolites is present
what are some short-term in vitro tests for genotoxic chemicals?
- measurements of mutagenesis in mouse lymphoma cells
- assays for chromosome aberrations and sister chromatid exchanges in Chinese hamster ovary cells
what are the most important groups of therapeutic drugs that increase the risk of cancer?
- drugs that act on DNA (i.e. cytotoxic and immunosuppressant drugs)
- sex hormones
what is teratogenesis?
the production of gross structural malformations during fetal development, in distinction from other kinds of drug-induced fetal damage e.g. growth retardation, dysplasia or asymmetrical limb reduction from vasoconstriction caused by cocaine
what are examples of known teratogens?
thalidomide penicillamine warfarin phenytoin valproate cytotoxic drugs ethanol retinoids ACE inhibitors
what are examples of suspected teratogens?
carbamazepine
tetracycline
what are examples of micellaneous teratogens?
corticosteroids androgens oestrogens stilbestrol aminoglycosides
what is the effect of thalidomide on fetal development?
phocomelia, heart defects, gut atresia
what is the effect of penicillamine on fetal development?
loose skin etc
what is the effect of warfarin on fetal development?
saddle nose, retarded growth, defects of limbs, eyes and CNS
what is the effect of corticosteroids on fetal development?
cleft palate and congenital cataract - rare
what is the effect of androgens and oestrogens on fetal development
masculinsation in female and testicular atrophy in male, respectively
what is the effect of stilbestrol on fetal development?
vaginal adenosis in female fetus and vaginal or cervical cancer
what is the effect of phenytoin on fetal development?
cleft lip/palate, microcephaly, mental retardation
what is the effect of valproate on fetal development?
neural tube defects
what is the effect of carbamazepine on fetal development?
retardation of fetal head growth
what is the effect of cytotoxic drugs on fetal development?
hydrocephalus, cleft palate, neural tube defects etc
what is the effect of aminoglycosides on fetal development?
deafness
what is the effect of tetracycline on fetal development?
staining of bones and teeth, thin tooth enamel, impaired bone growth
what is the effect of ethanol on fetal development?
fetal alcohol syndrome
what is the effect of retinoids on fetal development?
hydrocephalus etc
what is the effect of ACE inhibitors on fetal development?
oligohydramnios, renal failure
what is a key characteristic of a mutant Salmonella typhimurium?
unlike the wild type, it cannot grow without histidine
what are disadvantages of carcinogenicity testing?
- involves chronic dosing of groups of animals
- is expensive and time-consuming
- doesn’t readily detect epigenetic carcinogens
what are the three phases of mammalian fetal development?
- blastocyst formation
- organogenesis
- histogenesis and maturation of function
what are the drug effects on blastocyst formation?
gestation period: 0-16 days
main cellular processes: cell division
affected by: cytotoxic drugs, alcohol
what are the drug effects on organogenesis?
gestation period: 17-60 days
main cellular processes: division, migration, differentiation, death
affected by: teratogens
what are the drug effects on histogenesis and functional maturation?
gestation period: 60 days to term
main cellular processes: division, migration, differentiation, death
affected by: micellaneous drugs (e.g. alcohol, nicotine, antithyroid drugs, steroids) - interfere with supply of nutrients
what are the effects of thalidomide?
producing, at therapeutic dosage, virtually 100% malformed infants when taken in the first 3-6 weeks of gestation
when was thalidomide introduced? what as?
- in 1957
- as a hypnotic and sedative with a feature of being less hazardous in overdosage than barbituates
- recommended for use in pregnancy
when did suspicion of thalidomide’s teratogenicity arise?
1961
- reports of a sudden increase in the incidence of phocomelia
what are the effects of thalidomide teratogeneisis on different days of gestation?
21-22: malformation of ears and cranial nerve defects
24-27: phocomelia of arms
28-29: phocomelia of arms and legs
30-36: malformation of hands and anorectal stenosis
what are the teratogenic effects of cytotoxic drugs?
- alkylating agents (e.g. chlorambucil and cyclophosphamide) and antimetabolites (e.g. azathioprine and mercaptopurine) cause malformations and often lead to abortion
- folate antagonists (e.g. methotrexate) produce much higher incidence of major malformations
what are retinoids used for?
vitamin A derivatives
what are the teratogenic effects of retinoids/etretinate?
etretinate has marked effects on epidermal differentiation; is a known teratogen and causes a high proportion of serious abnormalities (notably skeletal deformities) in exposed fetuses
what is the mechanism of etretinate? what is an implication of this?
- accumulates in subcutaneous fat and is eliminated extremely slowly, detectable amounts persisting for many months after chronic dosing is discontinued
- women should avoid pregnancy for at least 2 years after treatment
what is an active metabolite of etretinate?
acitretin
- equally teratogenic, but tissue accumulation is less pronounced and elimination may be more rapid
what heavy metals cause teratogenesis?
lead, cadmium and mercury all cause fetal malformation in humans
where does the main evidence for heavy metal teratogenesis come from?
Minimata disease, named after the locality in Japan where an epidemic occurred when the local population ate fish contaminated with methylmercury that had been used as an agricultural fungicide
what does Minimata disease lead to?
impaired brain development in exposed fetuses results in cerebral palsy and mental retardation, often with microcephaly
what is the mechanism for mercury causing teratogenic effects?
mercury/other heavy metals inactivate many enzymes by forming covalent bonds with sulfhyfryl and other groups; this is responsible for developmental abnormalities
what are antiepileptic drugs causing teratogenic effects? how does epilepsy affect fetuses?
- congenital malformations are increased 2-3 fold in babies of epileptic mothers treated with 2+ antiepileptic drugs during first trimester
- phenytoin, valproate, carbamazepine, lamotrigine and topiramate
what teratogenic effects does warfarin have?
- first trimester: associated with nasal hypoplasia and various CNS abnormalities, affecting 25% of exposed babies
- last trimester: must not be used because of the risk of intracranial haemorrhage in baby during delivery
what is a hapten?
a small molecule which, when combined with a larger carrier such as a protein, can elicit the production of antibodies which bind specifically to it
what does the formation of an immunogenic conjugate between a small molecule and an endogenous protein require?
covalent bonding
what are immunological mechanisms of adverse drug reactions?
- reactive metabolites, rather than the drug itself, are responsible. produced during drug oxidation or by photoactivation in the skin
- reactive metabolites can also be produced by action of toxic oxygen metabolites generated by activated leukocytes
- reactive moiety may interact to form an immunogen with nuclear components rather than proteins
- conjugation with a macromolecule is usually essential
what are drugs that cause anaphylactic shock?
- penicillins (75% of anaphylactic deaths)
- enzymes e.g. asparaginase
- therapeutic monoclonal antibodies
- hormones e.g. corticotropin
- heparin
- dextrans
- radiological contrast agents
- vaccines
- other serological products
- local anaesthetics
- antispetic chlorhexidine
what can drug-induced haemotological reactions be caused by?
type II, III, or IV hypersensitivity
what drugs is haemolytic anaemia associated with?
sulfonamides and related drugs, and with an antihypertensive drug, methyldopa
what is agranulocytosis?
complete absence of circulating neutrophils
what are characteristics of drug-induced agranulocytosis?
- usually delayed 2-12 weeks after beginning drug treatment but may then be sudden in onset
- often presents with mouth ulcers, a severe sore throat or other infection
what drugs are associated with agranulocytosis?
NSAIDs (especially phenylbutazone, carbimazole and clozapine) and sulfonamides and related drugs (e.g. thiazides and sulfonylureas)
what drugs is thrombocytopenia caused by?
- reduction in platelet numbers
- can be caused by type II reactions to quinine, heparin and thizide diuretics
what is aplastic anaemia?
anaemia with associated agranulocytosis and thrombocytopenia
what is aplastic anaemia caused by?
drugs (notably chloramphenicol) can suppress all three haemopoietic cell lineages
what is a reactive metabolite of halothane? what is its action?
trifluoracetylchloride
- couples to a macromolecule to form an immunogen
how can cells affected by allergic liver damage be killed?
- halothane-protein antigens can be expressed on the surface of hepatocytes
- destruction of the cells occurs by type II hypersensitivity reactions involving killer T cells, and type III reactions can contribute
what are examples of other hypersensitivity reactions caused by drugs?
- caused by type IV hypersensitivity
- rashes can be antibody mediated but are usually cell mediated; range from mild reuptions to fatal exfoliation
- Stevens-Johnson syndrome is a severe generalised rash that extends into the alimentary tract
are what is a side effect? how does it differ from ADRs?
- an unintended effect of a drug related to its pharmacological properties
- can include unexpected benefits of treatment
- can describe minor and predictable ADRs
- side effects can be beneficial, e.g. PDE5 inhibitors improve urinary flow
what are the types of effects caused by ADRs?
toxic effects (beyond therapeutic range) collateral effects (therapeutic range) hypersusceptibility effects (below therapeutic range)
what are examples of toxic effects from ADRs? what can they be caused by?
- nephrotoxicity with high doses of aminoglycosides e.g. gentamycin
- dysarthria and ataxia with lithium toxicity
- cerebellar signs and symptoms
- caused by too high dose, reduced drug excretion by impaired renal or hepatic function or by interaction with other drugs
what are examples of collateral effects caused by ADRs?
- beta blockers causing bronchoconstriction
- broad spectrum antibiotics causing c. diff. and pseudomembranous colitis
- caused by standard therapeutic doses
what are examples of hypersusceptibility reactions caused by ADRs? what can they be caused by?
- anaphylaxis and penicillin
- sub therapeutic doses
what are symptoms of mild and severe ADRs?
mild: nausea, drowsiness, itching rash
severe: respiratory depression, neutropenia, catastrophic haemorrhage, anaphylaxis
what is an example of a time independent reaction (ADRs)?
- occur at any time during treatment
- e.g. INR increase when erythromycin is administered with warfarin
what are types of time dependent reactions due to ADRs? give examples for each
- rapid reactions (red man syndrome due to histamine release with rapid administration of vancomycin)
- first dose reactions (hypotension and ACE inhibitors)
- early reactions (nitrate induced headache)
- intermediate reactions (delayed immunological reactions e.g. Stevens-Johnson syndrome with carbamazepine)
- late reactions (adverse effects of corticosteroids, seizures on withdrawal of long term benzodiazepine
- delayed reaction (thalidomide and phocomelia)
what are type C ADRs? give examples
chronic
- osteoporosis and steroids
- analgesic nephropathy
- steroids and iatrogenic Cushing’s syndrome
- colonic dysfunction due to laxatives
what are type D ADRs? give examples
delayed
- malignancies after immunosuppression
- teratogenesis
- carcinogenesis e.g. cyclophosphamide and bladder cancer
what are type E ADRs? give examples
end of treatment/abrupt drug withdrawal
- opiate withdrawal syndrome
- glucocorticoid abruptly withdrawn leads to adrenocortical insufficiency
- withdrawal seizures when anti-convulsants are stopped
what are type F ADRs?
failure of therapy
- failure of OCP in presence of enzyme inducer
what are patient risk factors for ADRs?
- gender (F>M)
- elderly
- neonates
- polypharmacy (21% 5 or more drugs)
- genetic predisposition
- hypersensitivity/allergies
- hepatic/renal impairment
- adherence problems
what are drug risk factors for ADRs?
- steep dose-response curve
- low therapeutic index
- commonly causes ADRs
what are causes for ADRs? give examples
- pharmaceutical variation (eosinophilia-myalgia syndrome with L-tryptophan)
- receptor abnormality (malignant hyperthermia with general anaesthetics)
- abnormal biological system unmasked with drug (primaquine induced haemolysis in patients deficient in glucose 6-phosphate dehydrogenase)
- abnormalities in drug metabolism (isoniazid induced peripheral neuropathy in people deficient in N-acetyl transferase)
- immunological (penicillin induced anaphylaxis)
- drug-drug interactions (increased incidence of hepatitis when isoniazid is prescribed with rifampicin)
- multifactorial (halothane hepatitis)
what is idiosyncrasy?
inherent abnormal response to a drug
- rare but serious
what can idiosyncrasy be caused by? give examples
genetic abnormality/enzyme deficiency
- X linked enzyme deficiency
- G6PD deficiency + primaquine leads to haemolysis and haemolytic
anaemia
abnormal receptor activity
- malignant hyperpyrexia with general anaesthetics
- huge rise in Ca2+ conc. -> increased muscle contraction -> increased metabolic activity -> increased body temp
when should we suspect an ADR?
- symptoms soon after a new drug is started
- symptoms after a dosage increase
- symptoms disappear when the drug is stopped
- symptoms reappear when the drug is restarted
what are the most common drugs to have ADRs?
- antibiotics
- antineoplastics
- cardiovascular drugs
- hypoglycaemics
- NSAIDs
- CNS drugs
what are the most common systems to be affected by ADRs? give examples of effects
GI, renal, haemorrhagic, metabolic, endocrine, dermatologic
what are common symptoms of ADRs?
- dystonic reactions, serotonin syndrome, neuroleptic malignant syndrome
- thyroid abnormalities
- pulmonary fibrosis
- torsades de pointes
- liver failure
- bone marrow aplasia
- GI bleeding
- avascular necrosis
- rhabdomyolysis
- skin rashes
- Stevens-Johnsons syndrome
- toxic epideral necrolysis
- photosensitivity
what are common ADRs?
confusion nausea balance problems diarrhea constipation hypotension
can ADRs be avoided?
- 30-50% are preventable
- drug interactions
- inappropriate medication
- unnecessary medication
what is the MHRA? what does it do?
Medicines and Healthcare products Regulatory Agency
- responsible for approving medicines and devices for use
- watch over medicines and devices and take actions (e.g. drug withdrawal) to protect the public promptly if there is a problem
what is the yellow card scheme?
- introduced in 1964
- world’s first ADR reporting scheme
- collects spontaneous reports
- collects suspected ADRs
- is a voluntary reporting scheme
what are strengths of the yellow card scheme?
- acts as an early warning system for identification of previously unrecognised reactions
- provides information about factors which predispose patients to ADRs
- allows comparisons of ADR profiles between produces within the same therapeutic class
- continual safety monitoring of a product throughout its life span
- works rapidly, easily accessible, cheap
what are weaknesses of the yellow card scheme?
- cannot provide estimates of risk as true number of cases is underestimated and total number of patients exposed is unknown
- relies on ADR being recognised
- not all ADRs are reported (10% of serious reactions are)
- may be stimulated by promotion and publicity
- reporting high for newly marketed drugs and falls off over time
what are reasons for underreporting of ADRs?
ignorance diffidence fear lethargy guilt ambition complacency
why should we report ADRs?
- important for patient safety
- to identify ADRs not identified in clinical trials
- to identify new ADRs ASAP
- to compare drugs in the same therapeutic class
- to identify ADRs in at risk groups
what should be reported on a yellow card?
- all suspected reactions for: herbal medicines and black triangle drugs
- all serious suspected reactions for: established drugs, vaccines, contrast media and drug interactions
what does the black triangle mean?
indicates that a medicine is undergoing additional monitoring
what types of preparation are black triangles assigned to?
- contains a new active substance; new medicines or vaccines authorised on or after Jan 2011
- is a biological medicine, e.g. a vaccine or a medicine derived from plamsa (blood)
- has been given conditional approval/approved under exceptional circumstances
- the company that markets the medicine is required to carry out additional studies
what is a serious reaction?
- fatal
- life threatening
- disabling or incapacitating
- results in hospitalisation
- prolongs hospitalisation
who can report on a yellow card?
doctors, dentists, coroners, pharmacists, nurses, midwives, health visitors, radiographers, optometrists, patients
what information should be included on a yellow card?
- suspected drug
- suspected reaction
- patient details
- reporter details
- additional useful information
what is hypersensitivity?
objectively reproducible symptoms or signs, initiated by exposure to a defined stimulus at a dose tolerated by normal subjects and may be caused by immunologic (allergic) and non-immunologic mechanisms
what is immediate and delayed drug hypersensitivity?
immediate < 1hr (utricarial rash, anaphylaxis)
delayed > 1hr (other rashes, hepatitis, cytopenias)
what is type 1 hypersensitivity?
acute anaphylaxis
- prior exposure to the antigen/drug
- IgE antibodies formed after exposure to molecule
- IgE becomes attached to mast cells or leucocytes, expressed as cell surface receptors
- reexposure causes mast cell degranulation and release of substances e.g. histamine, prostaglandins, leukotrienes, PAF
what are the signs of anaphylaxis?
- occurs within minutes and lasts 1-2 hours
- vasodilation
- increased vascular permeability
- bronchoconstriction
- utricaria
- angioedema
what are type 2 hypersensitivity reactions?
antibody dependent cytotoxicity
- drug or metabolite combines with a protein
- body treats it as a foreign protein and forms antibodies (IgG, IgM)
- antibodies combine with the antigen and complement activation damages the cells e.g. methyl-dopa-induced haemolytic anaemia, pemphigus
what are type 3 hypersensitivity reactions?
immune complex mediated
- antigen and antibody form large complexes and activate complement
- small blood vessels are damaged or blocked
- leucocytes attracted to site of reaction release pharmacologically active substances leading to an inflammatory process
- includes glomerulonephritis, vasculitis
what are type 4 hypersensitivity reactions?
lymphocyte mediated
- antigen specific receptors develop on T-lymphocytes
- subsequent administration leads to local or tissue allergic reaction
- contact dermatitis, Stevens-Johnson syndrome
what are drugs commonly implicated in hypersensitivity reactions?
aspirin penicillins and cephalosporins sulfonamides nitrofurans anticonvulsants griseofulvin allopurinol
what is non immune anaphylaxis?
- previously called anaphylactoid reactions
- due to direct mast cell degranulation
- some drugs recognised to cause this
- no prior exposure
- clinically identical to the immunological mechanism
what is the management of anaphylaxis?
- commence basic life support, ABC
- stop drug if infusion
- adrenaline IM 500micrograms (300mcg epipen)
- high flow oxygen
- IV fluids
- IV antihistamine (chlorphenamine 10mg)
- IV hydrocortisone (100-200mg)
what antihistamine and hydrocortisone doses are given?
IV chlorphenamine 10mg
IV hydrocortisone 100-200mg