Pharmacology Flashcards
Who discovered Reserpine and when?
Kline in 1954
Who discovered Lithium and when?
In 1949, Cade in Australia discovered the use of lithium compounds in mania
Who discovered chlorpromazine and when?
Delay and Deniker’s team, and Charpentier 1950-1952
What was the first true antidepressant and when it discovered?
The first true antidepressant was discovered in 1952 Iproniazid (anti-TB treatment)
Iproniazid is a monoamine oxidase inhibitor.
Who discovered chlordiazepoxide and when?
Leo Sternbach in 1954
Who discovered the first TCA (what was it?) and when?
Kuhn discovered imipramine in 1958
Who synthesised butyrophenone haloperidol from pethidine and when?
In 1958, Janssen synthesised butyrophenone haloperidol from pethidine.
Who proposed the cheese reaction for MAOI associated hypertension?
1963 Cheese reaction was proposed to be the mechanism for MAOI associated
hypertension by Blackwell.
Who synthesised the first atypical agent?
Janssen synthesized an atypical agent RISPERIDONE in 1989
What was the first SSRI? Who synthesised it?
Carlssen synthesized purpose made SSRI Zimeldine – but this was withdrawn due to the
incidence of hypersensitivity syndrome and demyelinating disease that followed its use
What was the first SSRI on the market?
Fluoxetine
Who discovered Clozpaine and when?
Kane et al. 1988 rediscovered clozapine via a multicentre randomized design comparing
chlorpromazine vs. clozapine in ‘treatment resistant’ schizophrenia. 4% showed response
to chlorpromazine while 30% showed response to clozapine
Aliphatic phenothiazines is the chemical class of which drugs?
Chlorpromazine
Promazine
Triflupromazine
Piperidine derivatives is the chemical class of which drug?
Thioridazine
Piperazine derivatives is the chemical class of which drugs?
Trifluoperazine
Fluphenazine
Perphenazine
Thioridazine
Butyrophenones is the chemical class of which drugs?
Haloperidol
Droperidol
Thioxanthenes is the chemical class of which drugs?
Thiothixene
Flupenthixol
Zuclopenthixol
Dihydroindoles is the chemical class of which drug?
Molindone
Diphenylbutylpiperidine is the chemical class of which drug?
Pimozide (long t1/2)
Dibenzoxapine is the chemical class of which drug?
Loxapine
Benzisoxazole derivative is the chemical class of which drug?
Risperidone
Substituted benzamides is the chemical class of which drugs?
Amisulpride, Sulpiride
Dibenzodiazepine is the chemical class of which drug?
Clozapine
Dibenzothiazepine is the chemical class of which drug?
Quetiapine
Thienobenzodiazepine is the chemical class of which drug?
Olanzapine
Benzisothiazole is the chemical class of which drug?
Ziprasidone
Arylpiperidylindole (quinolone) is the chemical class of which drug?
Aripiprazole
Tertiary amines is the is the chemical class of which drugs?
Imipramine, Amitriptyline, Clomipramine, Dosulepin,
Trimipramine (also Venlafaxine)
The tertiary amines are thought to boost serotonin and noradrenaline.
Properties of secondary amines vs. tertiary amines?
more potent; less sedating; more noradrenergic, less antihistaminic or anticholinergic than tertiary
The secondary amines are said to have a lower side effect profile and to act primarily on noradrenaline
Secondary amines is the chemical class of which drugs?
Desipramine, Amoxapine, Nortriptyline and Protriptyline
also Duloxetine
Hydrazine derivatives is the chemical class of which drugs?
Phenelzine, Isocarboxazid (greater hepatotoxicity than
Tranylcypromine, a non hydrazine compound)
Aminoketone is the chemical class of which drug?
Bupropion
SSRIs
Citalopram, Paroxetine, Fluoxetine, Sertraline
and Fluvoxamine, S enantiomer of citalopram
- Escitalopram
SNRIs - serotonin and noradrenaline
reuptake inhibitor
Venlafaxine, Milnacipran, Duloxetine,
Sibutramine
NaSSA – Noradrenergic and specific
serotonergic antagonist
Mirtazapine and
Mianseri
NARI - Noradrenaline reuptake inhibitor
Reboxetine
NDDI - Noradrenaline Dopamine DisInhibitor -
Agomelatine
DARI – Dopamine reuptake inhibitor
*norepinephrine and dopamine re-uptake inhibitor
Bupropion
RIMA – reversible inhibitor of Monoamine A
oxidase
Moclobemide
brofaromine
SARI – serotonin antagonist and reuptake
inhibitors
Nefazodone, Trazodone
What is the novel agent Xanomeline?
A treatment option for schizophrenia. It acts via M1/M4
agonism
*also showed a trend toward improving cognitive function in Alzheimer’s
Which condition has ketamine been used in?
(NMDA) receptor antagonist that has shown
rapid antidepressant effects in treatment-resistant depression
What is the novel agent Pomaglumetad methionil?
a metabotropic glutamate receptor 2/3
agonist,
used to treat positive and negative symptom as an add-on therapy in schizophrenia.
What is deliquescence?
sensitivity to environmental moisture to such an extent that drugs turn from crystalline states into pastes or
liquids if left in contact with moist air even for a
short period of time.
Which drug has deliquescence?
Sodium valproate
What’s an active placebo?
An ‘active placebo’ has some activity inherently, but not against the treated condition.
What’s a nocebo?
When a substance administered for placebo effects produces prominent side-effects, it is
known as a ‘nocebo’.
What’s placebo sag?
Placebo sag is a term used to refer to decrease in placebo effect with repeated or chronic
administration of placebo drugs.
What is efficacy paradox?
The placebo effect may be disproportionately large for non-blinded therapies potentially
resulting in what has been called the efficacy paradox.
What do placebos work best for?
Placebos work best for pain, disorders of autonomic sensation, and disorders of factors
under neurohumoral control e.g. nausea, blood pressure, and bronchial asthma.
Placebo rates in psychiatric disorders
Depressive illness: 25 - 60%
Mania: up to 25%
Schizophrenia: 25 - 50%
Panic disorder: up to 70%
What colour tablets do anxiety and depression respond better to?
Anxiety symptoms responded better to green tablets and depressive symptoms responded
better to yellow tablets.
Why do placebos work?
- Natural remission theory
- Measurement regression (to the mean)
- Conditioning theory
- Endogenous opioids
Which medication form is associated with increased response?
Capsules
Phases before drug approval
- Preclinical Animal Studies - Mutagenicity, carcinogenicity and organ system toxicity are studies at this phase
- Human trials – volunteers phase 1 (safety) - tolerability and pharmacokinetics of the
drug are ascertained - Human trials – patients phase 2 (effectiveness) - RCTs
- Human trials – patients phase 3 (superiority or equivalence to standard looking for
comparative efficacy and tolerance profile). Extensive double blind RCT to determine how well does it work and what are the common side effects. - Human trials – post-marketing surveillance phase 4:
Adverse effects detected by post marketing surveillance of psychotropic medications:
- Nefazadone
- Droperidol, Thioridazine
- Sertindole
- Thalidomide (analgesic)
- Nomifensine
- Zimeldine
- Remoxipiride(sulpiride group)
- Mianserin
- MAOIs
- Clozapine
- Nefazadone - Hepatotoxicity
- Droperidol, Thioridazine - prolonged QTc
- Sertindole - Sudden cardiac death
- Thalidomide (analgesic) - Phocomelia
- Nomifensine - Hepatotoxicity
- Zimeldine - Hypersensitivity reactions and Guillain-Barre syndrome
- Remoxipiride(sulpiride group) - Aplastic Anaemia
- Mianserin - Blood dyscrasias
- MAOIs - Cheese reactions
- Clozapine - Agranulocytosis
Non-adherence rates for antipsychotics?
40–60%
*Nonadherent patients with schizophrenia are 3.5 x
more likely to relapse within 2 years
Non-adherence rates for mood stabilisers?
18–56%
Non-adherence rates for antidepressants?
30–97% (median 63%)
Factors that reduce adherence
•Asymptomatic stage of illness •Cognitive deficits •Comorbidity – alcohol and substance misuse •Devaluation of medication effects by the physician •Fear of side-effects. •High frequency of daily doses •Homelessness •Lack of insight (most common cause) •Long duration of illness (chronic diseases) •Oral formulations have poorer adherence than depots •Past history of non-adherence •Polypharmacy •Prophylactic or maintenance treatments •Psychopathology of hostility, suspiciousness and disorganization
Factors that improve adherence
- Presence of family support
- Liquid or sublingual forms
- High enthusiasm fromclinican
- Good patient-clinician relationship
- Continued access to clincian
Factors that have no influence on adherence
- Age at illness onset
- Age at first hospitalization
- Sex
- Socioeconomic status,
- Marital status
- Ethnicity
What are the four main categories of the health belief model of adherence?
- Benefits
- Costs
- Susceptibility
- Secondary benefits of medication and adherence.
What are some methods of improving adherence?
Psychoeducational programmes: Cognitive-based interventions Behaviour-modification interventions Motivational interviewing Compliance therapy
What are pharmacokinetics?
the time course and disposition of drugs in the body (what the body does to the drug)
*affected by the method of administration
What processes are involved in pharmacokinetics?
ADME; Absorption, Distribution, Metabolism,
and Elimination
Which factors affect the rate of absorption?
- The form of the drug
- The rate of blood flow at the site of administration
- Solubility of the drug which depends on the pH of the drug, size of particles in the formulation and the pKa of the drug (pKa is the pH at which precisely half of the drug is in its ionised form)
What is first-pass effect?
metabolism by liver and gut
mucosa - drugs absorbed from the gut undergo extensive metabolism before entering the systemic circulation
What are the mechanisms of absorption of drugs from the GI tract?
- Active transport 2. Passive
diffusion (most common mechanism) 3. Pore filtration
Which factors influence absorption of drugs from GI tract?
Intestinal motility
Gastric emptying
Gastric and intestinal pH
Intestinal microflora e.g. Chlorpromazine is sulfated in the gut which reduces its absorption
Area available for absorption
Integrity of blood flow
Presence or absence of food (food delays gastric emptying)
What is dissolution rate dependent on?
- Size of drug particle
- Solubility of the drug
- Properties of intestinal fluid (e.g. p H)
How quickly does absorption happen with IM administration?
10-30 minutes (avoids most of the first pass
metabolism)
What factors influence the rate of absorption of drugs administered intramuscularly?
Blood flow and aqueous solubility.
Lipid soluble drugs are rapidly absorbed; drugs with a relative low molecular weight are better absorbed. Increased muscle blood flow e.g. after muscular exercise increases the rate of absorption
Which method of administration gives 100% bioavailability?
IV
Which factors affect permeation?
Lipophilicity
Concentration gradient - either simple or facilitated diffusion
Surface area and vascularity
Which is more water soluble the ionised or non-ionised form of a drug?
The ionized form is more water-soluble than the nonionized form
Is the renal clearance higher or lower for ionised drugs?
Renal clearance is higher for ionised drug (drug is “trapped” in the glomerular filtrate and does not get reabsorbed)
What is distribution of a drug?
to ‘where’ in the body it can be found.
Some drugs are confined to the body fluids only, but others accumulate in particular tissues.
*Distribution of a drug leads to a fall in
the plasma concentration (central to peripheral shift) and is most rapid after intravenous
administration
Which factors influence Drug distribution?
- Hemodynamic factors - cardiac output, regional blood flow. Organs with the highest blood perfusions such as the brain, kidneys, and liver receive the highest distribution
- Plasma protein binding
- Permeability factors- higher the lipid solubility of the drug, the greater its rate of entry into cells
- Blood-brain barrier
- Blood- CSF barrier
What are the two compartments the body is divided
into?
Central compartment - plasma
Peripheral compartment - fat and other tissues, which vary with age, sex and weight.
Which drugs are highly protein bound?
95 - 99% –> diazepam, chlorpromazine, amitriptyline and imipramine
90 - 95% –> Phenytoin, valproate and clomipramine
What can occur with drugs that are highly protein bound?
They are prone to interactions mediated b ythis mechanism
e.g. Diazepam displaces phenytoin from plasma proteins, resulting in an increased plasma concentration of free phenytoin and an increased risk of adverse effects.
*The effects of protein displacement are usually not of clinical significance, as the metabolism of the affected drug increases in parallel with the free drug concentration.
What are the plasma proteins responsible for binding to drugs?
acidic drugs - albumin.
alkaline drugs - α1-acid glycoprotein
*most psychotropic drugs are basic
Which factors can affect the permeability of the BBB?
fever, head injury, hypoxia, hypercapnia, retroviruses, inflammation, vasculitis, hypertension, cerebral irradiation and aging
What determined the ability of a drug to pass blood brain barrier?
molecular size, lipid solubility and ionic status (Unionized molecules that are freely available and less protein bound are transported across the barrier)
Which areas lack BBB?
Circumventricular organs - subfornical organ, area postrema of the medulla and the median eminence.
What is bioavailability of a drug?
how much of an administered drug reaches its target - affected by absorption, distribution and elimination
What is metabolism of a drug?
process that renders drug less lipid-soluble and more water-soluble so that products are more easily excreted from the body
What happens during phase 1 of metabolism?
oxidation, reduction and hydrolysis
What happens during phase 2 of metabolism?
conjugation - to produce a water soluble conjugate
Where can conjugated drugs be excreted through?
renal excretion if relative molecular mass < 300
bile excretion if relative molecular mass > 300
Most psychotherapeutic drugs are oxidized by which enzyme system?
Hepatic cytochrome P-450 enzyme system
Between 5and 10% of Caucasians lack which P-450 enzyme and are poor metabolisers of corresponding substrates?
CYP2D6
Up to 15-20% of East Asians are poor metabolisers of substrates of which P-450 enzymes?
CYP2C19
Which psychotropic medications are metabolised by CYP2D6?
All TCAs, fluoxetine, paroxetine, trazodone, nefazodone, valproate, antipsychotics, risperidone, carbamazepine
Which psychotropic medications inhibit CYP2D6?
Paroxetine, to some extent fluoxetine, antipsychotics, amitriptyline and clomipramine
Which psychotropic medications are metabolised by CYP3A4?
Clomipramine, fluvoxamine, mirtazapine, nefazodone, Carbamazepine, most benzodiazepines.
Which psychotropic medications stimulate CYP3A4?
Carbamazepine and barbiturates
Which psychotropic medications inhibit CYP3A4?
Calcium channel blockers, fluoxetine and nefazodone.
What does smoking do to CYP1A2?
Inhibits
What does Fluvoxamine do to plasma Clozapine concentrations?
Increases plasma clozapine concentrations (by inhibiting CYP system)
*Clozapine levels may be increased 10-fold by the addition of fluvoxamine, which can induce seizures.
What does Fluoxetine do to plasma TCA levels?
Fluoxetine increases plasma tricyclic antidepressants via 2D6 and 2C19
What does carbamazepine do to the plasma concentration of the oral contraceptive?
Decreases the plasma concentration of several drugs including contraceptive pills.
Which drugs undergo autoinduction?
Carbamazepine and phenobarbitone
How does carbamazepine undergo autoinduction?
Carbamazepine is metabolised by CYP2D6, synthesis of which in turn is induced by carbamazepine
As a result of this autoinduction, the rate of metabolism of carbamazepine (and other P450 substrates) gradually increases over the first several weeks of treatment.
Which enzyme do smoking and caffeine affect?
Smoking and caffeine affect glucuronidation reaction via UGT enzyme and CYP1A2
- caffeine competitively inhibits CYP1A2
- Polyaromatic Hydrocarbons (PAH)in cigarettes induce CYP1A2
What are aripiprazole and risperidone metabolised by?
CYP2D6 and CYP3A
What are Quetiapine and ziprasidone metabolised by?
mainly metabolized by CYP3A
What are Olanzapine and Clozapine metabolised by?
CYP1A2 and UGTs
What’s the effect of caffeine on Clozapine/olanzapine?
increases levels
What’s the effect of caffeine on Clozapine/olanzapine?
decreases levels
What are the major routes of drug excretion?
urine, faeces and bile
*may also be excreted in sweat, sebum, tears, saliva and breast milk
What are the factors that influence excretion?
- Increased age (decreases excretion)
- Reduction in renal blood flow e.g. dehydration
- Renal impairment leading to decreased renal function
- Alterations in re-absorption: urine pH
Which drugs undergo renal elimination without significant liver breakdown?
lithium, amisulpride, sulpiride, gabapentin, acamprosate and amantadine.
What’s the half life of a drug?
The time taken for the plasma concentration of a drug to halve
What is first order kinetics?
a constant fraction of drug is cleared per unit time
(the higher the amounts of a drug present, the faster the elimination)
e.g. 100mg/ml (2hours) -> 50mg/ml (2hours) -> 25mg/ml (2hours) -> 12.5mg/ml
What is zero-order kinetics?
a constant amount, not a fraction, of the drug is cleared per unit time
e.g. 100mg (2hours) -> 80mg (2hours) -> 60mg (2hours) -> 40mg
How long does it take drug to reach the steady plasma level?
4-5 half lives
What is the median toxic dose?
dose at which 50% of patients experience a specific toxic effect
What is the median effective dose?
dose at which 50% of patients have a specified therapeutic effect.
What is the therapeutic index?
measure of the toxicity or safety of a drug
ratio of the median toxic dose to median effective dose
Changes in body composition in the elderly?
Increase in total body fat
Decrease in total muscle mass (lean body mass)
Decrease in total body water
What’s the effect of changes in body composition in the elderly?
Larger volume of distribution and longer half-life of lipophilic chemicals because of their increased sequestration in fat. e.g. benzodiazepines excretion is slower in the elderly
Changes in plasma protein in the elderly?
Decrease in plasma protein binding capacity
What’s the effect of changes in plasma protein in the elderly?
15-25% decrease in protein binding due to higher proteinuria and lesser plasma protein synthesis by the liver.
Changes in the liver in the elderly?
Decreased hepatic blood flow occurs.
What’s the effect of changes in the liver in the elderly?
After 80, CYP system declines.
Changes in the kidney in the elderly?
Decreases in renal blood flow have been approximated at 10% per decade beginning after the fourth decade - leads to reduced creatinine clearance and GFR.
What’s the effect of changes in the kidney in the elderly?
More frequent toxicity of renally eliminated agents(e.g. lithium).
Changes in the GI tract in the elderly?
- GI blood flow is diminished
- Gastric pH is increased as acidity drops
What’s the effect of changes in the GI tract in the elderly?
- Slower but nearly equal absorption of oral administered drugs
- Decreased gastric first pass metabolism noted
A reduction in the gastric wall content of dopa decarboxylase leads to a 3-fold increase in the concentration of levodopa in the elderly
Changes in the brain receptors in the elderly?
- Decreased number of brain acetylcholine postsynaptic receptors
- choline acetyltransferase is diminished
- brain acetylcholinesterase decreased
What’s the effect of changes in the brain receptors in the elderly?
Anticholinergic side effects more pronounced leading to increased frequency of delirium on polypharmacy.
Pharmacokinetic changes in neonates
- higher proportion of total body water and extracellular body water
- lower proportion of adipose tissue
- glomerular filtration rate is lower in those aged less than 3-5 months
- lower gastric acidity and have an increased gastric emptying time
- more permeable blood—brain barrier
- microsomal enzyme activity in the liver is lower in those than 2 months
- lower plasma concentration of albumin
Pharmacokinetic changes in pregnancy
- Delayed gastric emptying
- Decreased GIT motility
- Increased volume of distribution (5%)
- Decreased drug-binding capacity
- Decreased albumin level
- Induced liver metabolic pathway
- Increased GFR & renal clearance.
Pharmacokinetic changes in renal impairment (benzos)
- Benzodiazepines should be used with caution
- half-life of diazepam remains unchanged in end-stage renal disease, but its metabolite, desmethyldiazepam, may accumulate, causing excessive sedation
- The half-life of lorazepam is increased from 8–25 hours in healthy adults to 32–72 hours in end-stage renal disease
- At a low level of renal function, lorazepam dosage should be reduced by 50% to avoid excessive sedation
Pharmacokinetic changes in renal impairment (antidepressants)
- Imipramine and amitriptyline can be given at their usual dosage
- Dosage of fluoxetine and fluvoxamine does not have to be reduced
- Half normal dose is used for citalopram in patients with renal impairment or in elderly
- Half-life of paroxetine is increased with severe renal impairment, requiring dosage reduction
- Sertraline is not recommended in renal impairment
Pharmacokinetic changes in renal impairment (antipsychotics)
- Haloperidol does not require a dose reduction in renal impairment unless excessive sedation or hypotension occurs -Amisulpride is renally excreted almost exclusively so relatively contraindicated in renal failure
- Risperidone and its active metabolite 9-hydroxy-risperidone are substantially excreted in the urine so in renal impairment the elimination half-life is prolonged
Active metabolite of Imipramine
desipramine
Active metabolite of Amitriptyline
nortriptyline
Active metabolite of Trazodone, nefazodone
mCPP
Active metabolite of Fluoxetine
norfluoxetine
Active metabolite of Sertraline
desmethylsertraline
Which drugs inhibit TCA metabolism and therefore increase TCA plasma concentrations?
Quinidine, cimetidine, fluoxetine, paroxetine, phenothiazines, disulfiram, methylphenidate
Which drugs induce TCA metabolism and therefore decrease TCA plasma concentrations?
Smoking, phenytoin, carbamazepine, OC pills and barbiturates
What are the effects of TCAs on warfarin?
TCAs increase warfarin levels so there is a high risk of bleeding
What are the effects of TCAS on clonidine?
TCAs reduce clonidine levels and can cause Hypertensive crisis
What are the effects of TCAs on MOAIs?
- Synergistic serotonergic enhancement esp. with clomipramine
- TCAs reduce tyramine entry via monoamine reuptake channels
- There is a higher risk of serotonin syndrome but lower risk of cheese reaction
What are the effects of TCAS on ldopa?
TCAs reduce absorption of l-dopa and so lower l-dopa efficacy in Parkinsonism
What are the effects of TCAS on morphine?
Amitriptyline and clomipramine decrease the metabolism through UDP glucuronyl transferase interaction leading to increased opioid toxicity
What are the effects of TCAS on Phenothiazines?
Mutual inhibition of metabolism -> both antipsychotic and TCA levels increase
Which SSRI is the least protien bound?
Escitalopram (56%)
What’s the half-life of norfluoxetine?
4–16 days
What’s the half-life of fluoxetine?
4-6 days
Which SSRIs do not have active metabolites?
Fluvoxamine and paroxetine
Which is the most selective SSRI?
Citalopram