• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/257

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

257 Cards in this Set

  • Front
  • Back

What is pharmacology a study of?

The action of drugs

What is a drug?

Any substance or product that is used or intended to be used to modify or explore physiological systems or pathophysiological states for the benefit of the recipient.

What is pharmacodynamics?

How the drugs acts on the body

What are the levels of the biological system can a drug have effects on?

- Biological molecules


- Subcellular structures


- Cells


- Tissues and organs


- Intact organism

What is pharmacokinetics?

How the body deals with drugs

In which 2 ways can a drug produce its effect?

1. Non Specific Effects - related to the nature of the chemical. The act by their physical characteristics.

2. Specific Effects - due primarily to specific chemical interactions. This interaction involves binding to proteins.


What are the 4 main types of proteins?

1. Carrier Molecules

2. Enzymes


3. Ion channels


4. Receptors


What is a carrier molecule?

A carrier molecule binds the transported substance and shuttles it to the other side of the membrane for release

What are the 2 methods a carrier molecule can shuttle a substance across a membrane?

1. Facilitated Diffusion - no energy required. Carrier facilitated equilibrium of transported complex across membrane. It is transported in the direction of the concentration gradient.


2. Active transport - energy required. A substance is transported against its concentration gradient, therefore metabolic energy is required. (Na/K ATPase Pump)

What 3 groups can the targets of enzymes be grouped into?

1. Metabolic Processes - drugs that influence the synthesis of genetic material (DNA) (e.g Zidovudine (AZT))

2. Ion Pumps - substances that are transported against their concentration gradient (eg. Digoxin)


3. Synthesis, degradation and action of hormones and transmitters (eg. Paracetamol, Aspirin)


How do ion channels work?

- An ion channel is generally selective for a particular ion/ionic species


- An ion channel is a pore that allows ions to flow into or out of the cell depending on the ions concentration gradient


- Ion channels can interact with drugs via direct targets (drugs bind to the channel directly to alter its function) or via receptors (linked channels)

What is the primary function of receptors?

The act as a recognition site. Binding of the drug initiates effects.


They work like a 'lock and key' to produce a response

What are the characteristics of drugs receptor interactions?

1. Selectivity - only a narrow range of drugs of a similar type (complementary structure) are effective


2. Sensitivity - low concentrations of drugs produce the effect


3. Specificity - the SAME response elicited by activation of a particular receptor in a cell

What is a receptor agonist?

An agonist is a drug that activates the receptor to produce a response. Increasing the amount of the drug will increase the percentage of the Max response until it reaches its maximum possible value

What is a receptor antagonist?

An antagonist is a drug that binds to the receptor, however it doesn't activate the receptor. it prevents the agonist effect.


- Reversible - increasing the amount of the agonist will restore its response


- Irreversible - increasing the amount of the agonist will never restore its response.

What are the 4 main types of receptors?

1. Ligand-gated channel


2. G-Protein coupled


3. Kinase - linked


4. Intracellular Steroid

Describe ligand-gated channels

- Drug binding leads to opening/closing of the channel


- Na+ channels or Cl- (GABA) channels


- Time: very fast = milliseconds


- Example: benzodiazepines (Valium) -> used to treat insomnia (sedative) and anxiety (anxiolytic)

Describe G-Protein coupled receptors

- These receptors affect the intracellular mechanisms via a G-Protein


- Effects can be on an ion channel or a second messenger mechanism


- Time: fast = seconds


- Examples: muscarinic acetylcholine receptor; agonist carbachol; antagonist atropine


- beta-adrenoceptor; agonist salbutamol; antagonist propanolol

Describe Kinase-linked receptors

- Involves a receptor in the membrane that incorporates a protein kinase domain. Phosphorylates a protein (usually a tyrosine residue) to get a response


- Time: minutes


- Examples: Insulin, Growth factors

Describe Intracellular Steroid receptors

- Drug binds to a receptor in the cytosol then is transported into the nucleus.


- Drug needs to be lipid soluble


- Binds ot DNA to affect gene transcription


- Time: slow = hours


- Examples: oestrogen, testosterone

What is Tubocurarine?

A poison that was used on the tip of arrows to cause paralysis


(binds to receptor, but doesn't cause a response)

What are the 4 main processes of pharmacokinetics?

The drug enters the body:


- Absorption: process of a substance entering he blood/systemic circulation, dependent on the route of administration


- Distribution: the distribution of a substance throughout the tissues/compartments of the body


The drug leaves the body:


- Metabolism: the biotransformation (chemical transformation) of a substance/drug by the body (removal/excretion) - occurs mainly in the liver


- Excretion: the process of removal of the substance/drug from the body (e.g.. excretion by kidneys)

Define bioavailability

Bioavailability its the fraction of the administered dose that reaches the systemic circulation (relatively to an intravenous bolus injection)

What are the main parameters influencing drug absorption?

- route of administration


- blood flow


- drug properties: some drugs are better absorbed than others

What are the main DRUG parameters influencing absorption?

- formulation


- molecular size


- lipid solubility


- ionisation state

What is bioequivalence?

Two drugs are considered bioequivalent of they have similar bioavailabilities and show no clinical differences in either their therapeutic or adverse effects.


Bioequivalence is particularly important for the market authorisation of generic products.

Is aspirin well absorbed from the small intestine?


**ASH**

It is uncharged at a low pH (stomach) so it is absorbed easily, however at a charged pH (blood) it is not so easily absorbed

What factors affect GI absorption?

- pH - dissolution ionisation and degradation of active drug


- volume of GI contents


- proteolytic enzyme action


- bacterial metabolism


- gastric retention time


- intestinal transit time and GI motility


- drug interactions in the GI tract


- blood supply


These also affect bioavailability

Factors affecting bioavailability after GI absorption include?

- first pass hepatic metabolism


- entero-hepatic shunting

what is drug distribution?

the process of reversible transfer of a drug between one location and another once the drug is in the systemic system: more or less crossing of vascular endothelium/lipid membranes

what is drug distribution influenced by?

the affinity of drug for various tissue constituents:


- lipid soluble vs water soluble drugs


- more or less binding to plasma proteins (bound drug isn't active)


- affinity for fat tissues, muscles, etc.

what is the apparent volume of distribution?

- a characteristic of the drug (given value for a drug, obtained from PK analysis of clinical trials)


- depicts where the drug goes into the body (the value range is an indication of the relative distribution amongst plasma/tissues)

What is metabolism?

the biotransformation of an administered drug in the body

why does the body metabolise drugs?

The body metabolises drugs (mainly in the liver) with the aid of enzymes to transform them into a generally more water soluble substance for easier excretion

where does metabolism mainly occur?

liver (main), gut wall, muscles, fat, other tissues

Is the extent of metabolism the same for everyone?

No. Because there are various factors that influence metabolism; such as age, pregnancy, disease condition; from person to person

What are the outcomes of metabolism?

- inactive metabolites (most drugs, easy to eliminate via the kidneys)


- active metabolites (administration of an inactive prodrug; eg. codeine is transformed into morphine/activated in the liver)


- toxic metabolites (transforming drug into a toxic molecules via metabolism by liver enzymes eg. paracetamol; more water soluble so excreted in urine)

What is first pass metabolism and where does it occur?

- First pass metabolism is metabolism of a drug before it reaches the systemic circulation.


- it occurs mainly in the liver, but also in the gut wall, muscles, fat, and other tissues

What routes of administration does first pass metabolism occur by?

- oral administration


- rectal route

what is the main mechanism of drug absorption through lipid membranes?

Passive Diffusion

what are two important drug physiochemical properties that affect absorption through lipid membranes?

- ionisation (related to lipid solubility)


- lipid solubility (ability to cross cell lipid membrane)


(also molecular size and formulation)

What are phase 1 reactions?

- redox reactions


- catalysed by CYP450 enzymes family (oxidation) and others (reduction)


- in phase one, the molecule is oxidised and made more water soluble to make it ready for conjugation

What are phase 2 reactions?

- conjugation reactions


- catalysed by enzyme families specific to each conjugated chemical group


- Phase 2 uses enzymes (transferases) to attach (conjugation) endogenous water soluble molecules on drugs.

Describe the enzymes used in Phase 1 metabolism

- enzymes from the cytochrome P450 family for oxidation


- Main CYP450 enzymes are:


CYP3A4 - metabolises more than 50% drugs


CYP2D6 - known for genetic variations (slow vs high metabolisers eg. codeine)


CYP2C8

Why is the extent of metabolism of codeine (inactive) into morphine (active) variable between individuals?

- because the enzyme CYP2D6 is highly dependent on individuals


- the is one of the main enzymes involved in oxidation of a drug


- known for genetic variations (slow vs high metabolisers eg. codeine)

Describe the enzymes used in Phase 2 metabolism

UGTs - for glucuronidation


GSTs - glutathione conjugation


NATs - acetylation


SULTs - sulfation

what is first order elimination?

- the rate of drug metabolism is not constant, but proportional to the drug plasma concentration


- more drug = more enzymes recruited


- the amount f the drug eliminated by the liver is proportional to the amount of drug getting into the liver


- the most common elimination kinetics (most drugs at most doses)

what is zero order elimination?

- the rate of drug metabolism is constant; not dependent on drug plasma concentration


- more drug = if the enzymes are already at full capacity/saturated, no more enzymes are recruited


- the amount of drug eliminated by the liver is not proportional to the drug getting into the liver


- drug can build up in the body = toxicity, side effects

What is clearance? Where does it mainly occur?

- clearance is the volume of plasma that is cleared of drug per unit of time


- main organ is the kidneys

Why is sodium bicarbonate given in a paracetamol overdose?

- it has an ion trapping effect for enhancing elimination


- aspirin is a weak acid and sodium bicarbonate is a base that displaces the pH towards a basic pH


- aspirin is more soluble so more aspirin is excreted

What are the ANS functions

control of involuntary function


- smooth muscle


- cardiac muscle


- glands

Describe the somatic nervous system

- link between the CNS and effector tissues via motor neurons


- no synapse - the cell body terminal is at the effector tissue


- controls voluntary movement of skeletal muscle


- ACh neurotransmitter; acts on nicotinic receptors in skeletal muscles

What are the neurotransmitters of the ANS

- ACh (cholinergic nerves)


- Noradrenaline (adrenergic nerves)


- others (non-adrenergic, non-cholinergic or non-noradrenergic, non-cholinergic transmitter)

Describe the PNS

- controls what is happening at rest (rest and digest system)


- 2 nerves with a ganglion in the middle - located close to their effector organs


preganglionic = ACh acting on nicotinic receptors


postganglionic = there are differentiations as they act on different receptors for different regions (either ACh muscarinic, peptide peptidergic or NO nitrergic receptors)

Describe the SNS

Fight or flight system


SNS ganglia lie on either side of the vertebral column


preganglionic = ACh to nicotinic receptors


postganglionic = NA to a adrenoceptors; NA to b adrenoceptors; ACh to muscarinic or peptide to peptidergic

describe features of the autonomic nerve terminal

- action potenitals: code used by other neurons to transfer information from one neutron to another


- myelinated


- can be taken to or broken down in the cleft

describe the events involved in neurotransmission (drugs can affect these)

1. action potential propagation


2. substrate availability or transmitter production


3. metabolism of transmitter


4. uptake of transmitter


5. release of transmitter, depletion of transmitter


6. modulate presynaptic receptor


7. metabolism of transmitter


8. effector response

what type of pathway is the parasympathetic nerve pathway?

cholinergic pathway

what are the responses to cholinergic stimulation?

S- salivation


L- lacrimation (tears)


U- urination


D- defecation


G- GI distress


E- emesis (increases secretion, urination, vomiting)

describe the cholinergic nerve terminal

- presynaptic vesicles containing ACh


- Action potential stimulates ACh release


- ACE enzyme and ACh molecule


- nicotinic (ligand) and muscarinic (G-Protein coupled) receptors


- termination of action is predominantly due to the breakdown of ACh

What drug affects cholinergic transmission and why?

botulinum


- prevents the release of ACh from nerve terminals by preventing fusion of the vesicles with the membrane


- causes local muscle weakness/paralysis

Describe cholinergic agonist and antagonist effects on the eye

agonists - lead to constriction of the pupil and allows for accomodation for near vision


antagonists - (atropa belladonna/atropine) cause dilation of the pupil

What are 2 therapeutic uses of muscarinic antagonists

Ipratropium - inhaled - for COPD


- inhibits parasympathetic mediated bronchoconstriction


-AEs include dry mouth, blurred vision, urinary retention


Hyoscine - motion sickness/nausea


- penetrates the BBB, and is a CNS depressant


- AEs include sedation, confusion in the elderly



How is cholinergic transmission terminated?

ACh is hydrolysed by esterase enzymes (AChE), which terminates ACh action.


-drugs that inhibit AChE, will enhance the effect of endogenous ACH because you are blocking the enzyme that breaks it down

What are 2 therapeutic uses of AChE inhibitors?

Donezepil - alzheimers disease


- short acting


- makes ACh more available in the brain


- AEs - nausea, vomiting, insomnia, urinary incontinence (cholinergic)


Neostigmine - myasthenia gravis (autoimmune, skeletal muscle weakness)


- medium acting


- enhances transmission at the NMJ


- AEs - nausea, vomiting, insomnia, urinary incontinence (cholinergic)

Describe irreversible AChE inhibitors

- organophosphates (pesticides, chemical warfare agents)


- can lead to cholinergic crisis, and overstimulation of nicotinic and muscarinic receptors


- Muscarinic (SLUDGE)


- Nicotinic (stimulation of somatic NS, catecholamine release)

how do NMJ blockers work

- They block neuromuscular transmission at the neuromuscular junction, producing paralysis of skeletal muscle


- used adjectively to anaesthesia


- can be depolarising or non-depolarising


depolarising - Suxamethonium (nicotinic receptor agonist)


- causes continuous stimulation and ongoing depolarisation.


- short acting muscle relaxant causing temporary paralysis of affected muscle


non-depolarising - Pancuronium (nicotinic receptor antagonist)


- prevents the binding of ACh, reversed by AChE inhibitors (increase ACh loves)


- used in surgery to obtain skeletal muscle relaxation

What type of receptors are Adrenoceptors?


What are adrenoceptor agonist effects?

G-Protein coupled


• A1 – vasoconstriction of peripheral blood vessels; Dilation (contraction) of pupil; Increased contractility of the heart (inotropic effects)


• A2 – inhibition of transmitter release; Aggregation of platelets; Contraction of smooth muscle


• B1 – increased heart rate (chronotropic effect); Increased contractility of the heart (inotropic effects)


• B2 – relatation of uterus; Glycogenolysis; Dilation of bronchial smooth muscle

What are adrenergic agonist effects?

• Alpha - Vasoconstriction; Pupil dilation; Decreased gut motility and secretions; Glycogen breakdown; Urinary retention; Contraction of the smooth muscle of vas deferens; Contraction of non pregnant uterus; Sweating; Goose flesh


• Beta 1 - Cardiac acceleration; Lips lysis; Decreased gut motility and secretions; Renin release


• Beta 2 - Bronchodilation; Fine skeletal muscle tremor; Vasodilation of blood vessels to skeletal muscles; Glycogen breakdown; Relaxation of the pregnant uterus; Mast cell stabilisation

What are therapeutic uses of adrenergic stimulating drugs?

Directly acting


- adrenaline, noradrenaline, dobutamine, phenylephrine, salbutamol and salmeterol


Indirectly acting


- tricyclic antidepressants, MAOi, cocaine, amphetamine

Describe Adrenaline

- alpha and beta agonist (treatment of anaphylactic shock, cardiac arrest, adjunct to local anaesthetics)


- pharmacokinetics: it is rapidly metabolised, therefore only IM or SC injections


- cardiac - ^HR and force of contraction (B1)


- vascular - constriction of skin blood vessels (a1) and dilation of skeletal muscle blood vessels (B2)


- respiratory - bronchodilation (B2)



Describe Noradrenaline

- alpha and beta agonist (acute hypertension)


- pharmacokinetics: administered IV


- vascular - constriction of blood vessels (a)


- contraindications: caution in patients with atherosclerosis, occlusive vascular disease, hypertension

describe dobutamine

- B1 agonist (used in acute cariogenic shock)


-cardiac - increases force of contraction to increase cardiac output


- pharmacokinetics: administered IV

describe phelylephrine

- a agonist (decongestant)


- vascular - constriction of blood vessels (a)


- pharmacokinetics: nasal decongestion, oral administration


- interactions - MAOi


- AEs - rebound congestion with intranasal; stimulatory effects (insomnia, nervousness) with ora preparation


- contraindicated in hypertension

describe salbutamol and salmeterol

- B2 agonist (short and long acting respectively)


- treat acute bronchoconstriction


- respiratory - bronchial dilation (B2)


- pharmacokinetics - inhaled, salmeterol is usually combined with a steroid anti-inflammatory agent


- AEs - skeletal muscle tremor, palpitations, headache, hyperglycaemia (infrequent)

What are the different sites at which drugs can act to modify neurotransmission?

1. Synthesis of neurotransmitter


• uptake of precursors into terminal


• synthesis of transmitter


2. Storage of transmitter in vesicles


3. Degradation of surplus transmitter in cytoplasm


4. Depolarisation by propagated action potential– influx of Ca2+ in response to depolarisation


5. Release of transmitter by exocytosis– fusion of vesicles to terminal membrane– diffusion to postsynaptic membrane


6. Interaction with postsynaptic receptors


7. Inactivation of neurotransmitter


All steps are potential sites for drug action

describe indirect sympathomimetic agents and their mode of action

Cocaine- blocks the reuptake of catecholamines into presynaptic nerve terminals


Amphetamine- taken up via reuptake pump and displaces catecholamines


Tricyclic Antidepressants- inhibit the reuptake of NA and serotonin into presynaptic terminals


(AE - sedation, dry mouth, constipation = likely due to blocking cholinergic receptors)

Describe monoamine oxidase inhibitors

- metabolises amines such as noradrenaline


- MAO is a mitochondrial enzyme


- located in most tissues, nervous system, liver and GIT


Non-selective - in noradrenergic, dopaminergic and serotonin nerve terminals - antidepressant (phenylzine)


MAO a - in noradrenergic and serotonin nerve terminals - antidepressant (moclobemide)


MAO b - in dopaminergic nerve terminals - parkinsons (selegiline)

Describe Dopamine

- catecholamine, immediate precursor of NA


- release is increased by amphetamine


- deficient in parkinson disease


- major target of antipsychotics


-CNS: mood and motor function

How does the body respond to a drop in arterial pressure/

Mean Arterial Pressure - controls perfusion of organs


A decrease in BP= stimulation of the SNS which results in an increase in HR, SV and constriction of vasculature, to increase the cardiac output


This increases renin release which starts a cascade that produces angiotensin 2 (which helps to retain/^fluid= maintain blood volume and balance pressure)

describe the renin-angiotensin system

- a hormonal system


- the body releases renin which acts on angiotensinogen and forms angiotensin 1, which interacts with ACE which chops off some amino acids to turn it into angiotensin 2 (active)


- it promotes thirst and acts to increase the circulating fluid volume

What is essential hypertension and secondary hypertension

Essential hypertension - increased blood pressure due to an unknown cause, likely a combination of diet, genetics and lifestyle


Secondary hypertension - due to a specific underlying pathology (renal, endocrine etc)

Describe left ventricular hypertrophy


(consequences of hypertension)

Increased afterload on the heart increases cardiac work; the heart hypertrophies in response to this.


Major risk factor for congestive heart failure

Describe Stroke/MI


(consequences of hypertension)

Rupture of cerebral/coronary blood vessels

Describe endothelial damage


(consequences of hypertension)

promotes arteriosclerosis

Describe renal failure


(consequences of hypertension)

impairment of renal blood flow

describe retinal damage


(consequences of hypertension)

-alterations in retinal blood vessels


-occurs in type 2 diabetes

How is hypertension treated?

- antihypertensive drugs to reduce TPR, CO or both


- lifestyle interventions


- treatment on uncomplicated hypertension usually begins with a single drug (usually ACE inhibitor)


- if control is inadequate, addition of second drug usually preferred (vs increasing dose) to achieve good BP control

How do drugs reduce total peripheral resistance?

- calcium channel blockers inhibit L-type Ca2+ channels in vascular smooth (and myocardium) to reduce Ca2+ influx


- a reduction in intracellular Ca2+ leads to vasodilation and a reduction in TPR


- they reduce cardiac contractility and heart rate , thus reducing cardiac work


-Dihydropyridines such as nifedipine and amloidipine are calcium channel blockers


- AEs - peripheral oedema, flushing, headache, bradycardia

Describe ACEi action in hypertension

-ACEi reduce Ang.2-induced vasoconstriction, aldosterone release and Na+ retention. (first line hypertension therapy: also heart failure, STEMI, diabetic nephropathy)


- prevents the conversion of Ang.1 to Ang.2


- reduces salt and water retention to target cardiac output and TPR


- AE- cough, first dose and/or orthostatic hypotension, hyperkalaemia, angioedema


- contraindicated in pregnancy




(ARBs have a very similar action and AEs)

How do B Blockers work (B adrenoceptors)

Beta blockers competitively block B receptors to prevent an increase in HR or cardiac output that is generally stimulated by the SNS or adrenaline; to reduce cardiac work


-B-adrenoceptor antagonists antagonise these receptors in the heart, vasculature, kidney, bronchi and pancreas


- B1 - selective for B1 receptors in the heart, and are less selective for B2 receptors at the other organs, but this can be overcome with higher doses


AEs - bradycardia, bronchospasm, fatigue, cold extremities


- they can reduce angina frequency nd decrease adverse cardiac events/mortality

What do thiazide diuretics do?


eg. hydrochorothiazide

they bind to and inhibit the Na+/Cl- cotransporter to reduce Na reabsorption and cause a moderate diuresis (increase in urine). They also increase potassium excretion and can impair glucose tolerance.


AEs - hypotension, dizziness, electrolyte disturbances


- for hypertension and mild heart failure

what is the ABCD of anti-hypertensive drugs?

A - ACE inhibitors (-pril) and AT1R antagonists (-sartan) also in limited quantity a1-adrenoceptor antagonists (prazosin)


B - B adrenoceptor antagonists (-olol)


C - Calcium channel antagonists (dihydropyridines)


D - diuretics (thiazide diuretics)

Describe the pathology of heart failure

- heart failure occurs when the heart cannot maintain end-organ perfusion (reduced cardiac output)


- it results from he inability of the ventricle to fill or eject blood, and can be from a range of causes (hypertension, ischaemic heart disease, cardiomyopathy)


- reduction in cardiac output can be due to systolic/diastolic dysfunction


- may require a variety of different therapeutic approaches

List the goals, strategy and pharmacological therapies for the treatment of heart failure

Goals


- symptomatic relief


- prevent further cardiac dysfunction


- reduce mortality


Strategy


- correction of systematic factors and comorbidities


- lifestyle modifications


- treatment of underlying CVD


Pharmacological Therapy


- ACE inhibitors


- Beta blockers


- Diuretics for fluid overload


- aldosterone antagonists


- Digoxin


- Inotropes for acute HF

How does Digoxin treat heart failure?

Digoxin inhibits the Na/K ATPase pump found on the membrane of the muscle cell.


- Digoxin is a positive inotrope that increases the force of cardiac contraction and slows cardiac conduction.


- Used in heart failure especially with atrial fibrillation/flutter


AE - risk of arrhythmias


- narrow therapeutic range


- polar molecule so predominantly renal excretion


- excretion time - ~36hours with normal renal function


- reduced plasma K+ increases digoxin effects due to reduced competition at the K+ binding site

What is coronary heart disease?


(ischaemic heart disease)

- The most common form of CVD


Acute IHD - can be due to worsening/unstable angina, or sudden occlusion of a coronary artery due to rupture of an atherosclerotic plaque (MI)


Chronic IHD - usually presents as angina (predictable chest pain upon exercise/exertion)


- both are characterised by a lack of blood supply to the myocardium, resulting in inadequate O2 to meet metabolic/tissue demand.

What is atherosclerosis?

A progressive disease of the large arteries characterised by the local accumulate o lipids and fibrous elements together with chronic inflammation.


(fibrous plaques are vulnerable to rupture = thrombosis)

What happens when myocardial O2 is insufficient?


(eg. an atherosclerotic plaque in a coronary vessel)

- limits supply to demanding tissues


- during exercise/stress, there is an increased demand for O2 which cannot be met, and therefore pain occurs


- Can have Stable Angina, Unstable Angina or Myocardial Infarction

Describe Stable Angina

Predictable chest pain with exertions due to underlying narrowing of the coronary vessels by atheroma.


Target: cardiac work (organic nitrated, B Blockers, Ca2+ antagonists)


- underlying atherosclerosis (statins)


- anti-thrombotic therapy (aspirin)

Define unstable angina

Pain occurring with reduced exertion, culminating in pain at rest.


Treatment: anti platelet drugs and organic nitrates

Define myocardial infarction

Sudden occlusion of a coronary vessel leading to death of cardiac tissue due to O2 deprivation. The location and size of the blockage determines the extent of damage


Target: restore myocardial flow (physical or pharmacological methods

What is the pharmacological treatment of IHD

reduce cardiac work


- nitrates


- calcium channel antagonists


- B adrenoceptor antagonists


treatment of underlying atherosclerotic disease


- lipid lowering therapy (statins)


- anti platelet drugs


removal of blockage and restoration of coronary flow

Define Preload and Afterload

Preload


-the degree of tension on the muscle when it begins to contract


(the load on the heart before it begins to contract)


- determined by the end diastolic pressure


Afterload


- the load against which the muscle is working (mainly affected by the resistance downstream - in the artery or the circulation = affected by the pressure in arterial circulation leading forth ventricle)


**Treatment of IHD involves targeting these processes to reduce cardiac work**

Describe the mechanism of organic nitrates

They cause vasodilation (more commonly in the venous than arterial system).


This decreases the venous/arterial pressure.


Which in turn reduces the preload/afterload.


Leading to reduced myocardial work and O2 consumption

Describe GTN (organic nitrates)

- GTN is a prodrug that requires metabolism to release nitric oxide and relax vascular smooth muscle


- given sublingually due to extensive first pass metabolism


- AEs - vasodilatory effects (flushing, headache due to influx of blood in the rain leading to static hypertension, reflex tachycardia, orthostatic HT, fainting, peripheral oedema


- tolerance can develop quickly and wear off quickly, therefore when Pts are reexposed to the nitrates, they will re-experience the AEs, therefore the drug isn't given continually. Pts need to have nitrate free intervals to avoid tolerance and see a clinically beneficial effect

What is the role of cholesterol in the body?

- biosynthesis of steroid hormones


- constituent of cell membranes


- synthesis of bile acids


- absorption of lipid soluble vitamins from GI tract

How is cholesterol transported within the body?

- Lipids and cholesterol are transported in the bloodstream as lipid/protein complexes


(LDLs are highly atherogenic, readily oxidised and cross the endothelial barrier easily)


(HDLs transport cholesterol back to the liver for excretion, they are anti-atherogenic)

How do we lower LDL-C levels

Statins are used to competitively inhibit HMG-coA reductase (rate limiting step in cholesterol biosynthesis). this leads to an increase in LDL-C uptake.


-> reduced mevalonate production


-> reduced hepatic cholesterol synthesis


-> increased LDL receptor synthesis on hepatocytes


-> increased uptake of LDL-C from bloodstream


-> reduced plasma LDL-cholestrol


AEs - myalgia, dizziness, headache

what is haemostasis

A complex and highly regulated process involving:


-vasoconstriction


-formation of a platelet plug


-stabilisation of he platelet plug by fibrin


-fibrinolysis


The major classes of drugs that interfere with haemostats include:


-antiplatelet drugs


-anticoagulants

What is thrombosis

- the formation of a haemostatic plug within the vasculature in the absence of bleeding


Venous thrombi - red clots, erythrocytes and fibrin


Arterial thrombi - white clots, platelet aggregation

What is Virchow's triad?


(thrombosis)

- injury to vessel wall (eg. rupture of atherosclerotic plaque)


- altered blood flow (eg. veins in the legs during prolonged sitting, turbulence)


- increased coagulability of the blood (thrombophilia)

How do anti platelet drugs act?


eg. Aspirin

- Aspirin irreversible acetylates active site of COX1, inhibiting enzyme activity and reducing cyclic endoperoxide production.


- inhibition of COX 1 for the lifespan of the platelet reduce aggregation.


AEs - GIT irritation, increased bleeding time, asymptomatic blood loss

How do anti platelet drugs act?


eg. P2Y12 antagonists

- ADP induces platelet aggregation by activating purinergic receptors (GPCRs)


-P2Y12 antagonists (Clopidogrel) irreversible inhibit P2Y12 receptors to inhibit platelet aggregation


- Clopidogrel is well absorbed orally; prodrug; converted to active metabolite by CYP enzymes


AEs - increased risk of bleeding, diarrhoea, GI ulcer



Describe the coagulation cascade

- it converts soluble fibrinogen (inactive precursor) int he blood to fibrin that stabilises the clot


- components of the coagulation cascade print int he blood as zymogens (inactive precursors = require enzymatic cleavage)


- activation initiate cascade, amplifying signal


- Thrombin (factor 2a) cleaves fibrinogen to form fibrin

How is the formation of trombone controlled?

Formation of thrombin form prothrombin requires interaction with factor Xa (enzyme for prothrombin cleavage).


The interaction of another clotting factor (Va) and activated platelets provides a mining site for the interaction


Platelets are critical for correct localisation

Describe Warfarin

- widely used oral anti-coagulant (orally active, rapidly absorbed)


- it competes with vitamin K for binding at vitamin K reductase (reduces amount of vit.K for carboxylation reaction -> for the production of mature clotting factors)


- difficult to dose and maintain -> narrow therapeutic range, requires dose individualisation


-AE - bleeding


- can be affected by diet

What is selective toxicity?

Selective toxicity is the concept which relies on targeting biochemical processes that differ in some way between the host and int invading organism

Describe prokaryotes and eukaryotes

Prokaryotes


- bacteria


- no nuscleus


- cell wall and membrane


- single chromosome


- no mitochondria


- Ribosomes - 705


Eukaryotes


- mammalian


- nucleus


- no cell wall


- chromosomes (condensed mitochondria


- ribosomes - 805

Define bactericidal

Death of bacteria.


Usually results form drugs that interfere with cel wall synthesis or disrupt the cell membrane


The cell dies in response

Define bacteriostatic

Growth of bacteria is prevented. Body's own immune system takes care of the rest.


Prevent it from growth, but if it is already fully formed, there isn't much of an effect.


Usually drugs that interfere with protein or nucleic acid synthesis

Define Broad and Narrow spectrum

Differences int e cell wall are believed to account for the spectrum of effectiveness of antibiotics


Broad - wide susceptibility, both gram types


Narrow - smaller susceptibility (works on a smaller number of bacteria)

Describe Bacteria Gram positive and negative testing (3 points)

1. a way of identifying different toes of grouping of bacteria


2. allows you to see the cells - when they take up the dye - they wither retain the colour or lose it


3. if they retain the colour they are gram positive, if they lose it they are gram negative

What are the 4 major sites of antibacterial drug action?

- inhibition of DNA replication


- inhibition of cell wall synthesis


- interference of cell membrane


- interference with protein synthesis

Drugs affecting DNA synthesis/replication?

Sulphonamides


Use: broad spectrum, bacteriostatic. Infections oft he urinary tract, upper/lower respiratory tract, skin, wound and eye infections


Pharmacokinetics: readily absorbed by GIT, Oraly active, 50-70% bound to plasma proteins


Metabolism: mainly acetylated (destroyed) in liver, excreted in urine


AE - rashes, nausea, vomiting, GIT disturbances


Trimethoprim


Use: Bacteriostatic. Infections of the urinary tract


Pharmacokinetics: rapidily absorbed by GIT, Oraly active, 44% protein bound in plasma


Metabolism: excretion via tubular secretion and glomerular filtration, mainly unmetabolised


AE - rash, pruritus, nausea, vomiting, GIT disturbances (hyperkalaemia with higher doses)


Sulphonamides and Trimethoprim


Use: bactericidal. Infections of the urinary tract and respiratory tract,


Pharmacokinetics: as when given alone


Metabolism: as when given alone


AE - rashes, nausea, vomiting, GIT disturbances (as when given alone)

Drugs affecting bacterial cell wall synthesis?

Penicillins (Beta Lactams) - occasionally cephalosporins


MOA -penicillin binds to cell via penicillin binding proteins and then it inhibits the cross linking process by permanently inactivation a transpeptidase involved in cross linking peptidoglycan chains


Pharmacokinetics: poor to very good GIT absorption, oral absorption capability; eliminated via tubular secretion (urine)


AE - hypersensitivity, possibly alteration in GIT flora and hyper infection


Types:


- Benzylpenicillin (G) (naturally made) for Gram +ve bacteria


- for meningococcal meningitis, pneumococcal, streptococcal, leptospiral infections


-Phenoxymethylpeniillin (V) - less potent, Reasonable GIT absorption - similar spectrum to pen G


-Procaine Penicillin, benzathine penicillin -longer acting, depot form (IM)


-Ampicillin, Amoxycillin - broad spectrum, less GT disturbance

Drugs affecting Bacterial cell membrane

Polymyxins (B)


-MOA - cationic detergent properties and interfere wth phospholipids in the cll membrane (bactericidal)


- disrupt the bacterial cell membrane, prevent it from functioning


-Use - effective against gram +ve bacilli (pseudomonas and coliforms) Toxicity mainly restricts use to gut sterilisation and topical use


-AE - interferes with neuromuscular transmission and toxic to the kidney

Drugs affecting protein synthesis

Tetracyclines (bacteriostatic)


MOA - active transport into susceptible organisms. Concentration in the cell builds up. Blocks the binding of tRNA to A site in ribosome/mRNA complex. Prevents binding, and attachment of the amino acid and therefore prevents synthesis of the protein


Use - broad spectrum gram +ve and -ve. (Rickettsia, Mycoplasma and Chlamydia, Cholera Organisms and in respiratory and urinary infections


Pharmacokinetics - Swift but incomplete GIT absorption. Metal chelator (milk etc will reduce absorption) Excreted in urine and bile unmetabolised


AE - GIT disturbances (nausea, vomiting, diarrhoea), sensitisation to light


Macrolides (bacteriostatic or bactericidal)


MOA - Inhibit protein synthesis. Binds to 50s ribosomal subunit to inhibit translocation of tRNA from A site to P site


Use - Narrow spectrum against gram +ve bacteria. Not effective against gram -ve except Gonorrhoea, mycoplasma pneumonia and legionella


Pharmacokinetics - acid stable, orally active. Penetrate into most tissues except CSF and synovial fluid Inactivated partly in the liver. Mainly excreted in bile. May interfere with other drugs (warfarin)


AE - GIT disturbances, unpleasant but usually not serious. Opportunistic superinfection esp. in GIT and urinary tract.

How do DNA viruses replicate?

-Viral DNA gets into a host nucleus and uses the host enzymes to produce more viral DNA, RNA and Proteins necessary for viral replication


-It can be difficult to treat as it becomes part of the host cell


-Viral genetic information is incorporated into the genetic information of the host cell, then all the mechanics of the host cell can be utilised by the viral cell to produce viral proteinsEg Herpes viruses (HSV), VZV (chicken pox, shingles), EBV (glandular fever)

How do RNA viruses replicate?

Virus binds to the host cell, this enables it to release its genetic information into the host cell. But it can't be utilised as mRNA, so it reverses RNA into DNA via reverse transcriptase. Then it acts like a DNA virus -> is incorporated into the nucleus of the cell then host cell machinery is used and viral proteins are produced

What are the main mechanisms for antiviral therapy?


(inhibition of...(4))

1. penetration of virus into host cell


2. nucleic acid synthesis


3. protease enzyme


4. secretion of virus form host cell

drugs that interfere with the penetration of virus into host cell

Amantadine


MOA - blocks an ion channel involved in the entry process of the virus into host cell. Narrow spectrum. Inhibits penetration of the virus into the cell, via inhibition of an ion channel that is involved in entry to the cell


Pharmacokinetics - Given orally. Excreted unchanged via urine.


Use - Active against influenza A (not B). May be used prophylactically.


AE - Infrequent. Dizziness, insomnia, slurring speech. Not serious.

drugs that interfere with nucleic acid synthesis

Zidovudine (AZT)


MOA - Inhibits viral reverse transcriptase. Terminates viral DNA chain. May also affect cellular mitochondrial DNA polymerase.


Pharmacokinetics - Oral or iv. Metabolised in the liver and 20% excreted unmetabolised in the urine. Probenicid inhibits liver metabolism and urinary excretion.


Use - Used in the treatment of HIV. Delays onset of AIDS. Reduces risk of transmission from mother to fetes. Reduces viral load


AE -Mainly anaemia and neutropenia. Resistance develops with long term use.

drugs that interfere with protease enzyme

Antiviral therapy protease inhibitors


(Ritonavir, Saquinavir)


MOA - Interrupt the way HIV uses healthy cells to produce more virus.


Pharmacokinetics - Oral, usually with a meal.


Use - Used in the treatment of HIV. Increases CD4 (T cell) count. Decrease viral load. Usually in combination with one or more reverse transcriptase inhibitors.


AE - GIT disturbances, nausea, vomiting

drugs that interfere with secretion of virus form host cell

Neuramidase inhibitors


(Zanamivir, Ostelamivir)


MOA - Neuraminidase is important in the penetration of the virus into the host cell and in the release of the virus from the host cell.


Pharmacokinetics - Zanamivir – powder taken via nose (Relenza) Oseltamivir – orally (a prodrug(needs to be metabolised to be activated), hydrolysed in the liver to active form) (Tamiflu)


Use - Used in the treatment of influenza A and B


AE -Diarrhoea and nausea most common

what are they symptoms of malaria?

Respiratory distress


Neurological problems


Anaemia

What are the 4 sites of action for antimalarial therapy?

1. Red Blood Cells. Effective against erythrocytic form. Treat acute attacks. Affect a cure for P. falciparum and P. malaria (insignificant/no exo erythrocytic forms.). Suppress attacks from P. vivax and P. ovale but can have relapse due to hypnozoites).


2. Liver. Radical cure. Effective against parasites in the liver.


3. Blood. Block link between exo erythrocytic and erythrocytic stages. Prevent attacks. Agents given as prophylactic to people travelling in malaria regions. Need to be taken before and after return of travel.


4. Transmission. Effective against gametocytes. Prevent transmission in human population.

Anti malarial therapy -


Chloroquine and Quinine

Chloroquine


MOA - accumulates in parasite, prevents parasite breaking down Hb. Inhibits haem polymerase. Converts toxic breakdown products of Hb into harness by-products. Prevents DNA replication and protein synthesis


Pharmacokinetics - complete absorption form GIT. Extensive distribution. Concentrated in parasitised RBC. In severe cases (IM, SC, slow IV) infusion. Released slowly from tissues, excreted mainly unchanged in urine and metabolised in liver


Use - for all susceptible plasmodia infections and chemoprophylaxis in combination with other drugs


AE - dizziness, burred vision, headache, nausea, vomiting, heart rhythm disturbances


Quinine


MOA - similar to chloroquine


Pharmacokinetics - complete absorption form GIT. Extensive distribution. Concentrated in parasitised RBC. In severe cases (IM, SC, slow IV) infusion. Released slowly from tissues, excreted mainly unchanged in urine and metabolised in liver


Use -for plasmodia infections including those not susceptible to chloroquine. Usually given in combination with other drugs


AE - more toxic than chloroquine. Irritates bowel (nausea, vomiting). Cinchonism syndrome (dizziness, blurred vision, headache, tinnitus) At higher doses (heart rhythm disturbances, hypotension)

Anti malarial therapy


Mefloquine



MOA - Interferes with the parasites ability to use Hb as a nutrient source(blocks transport of Hb metabolites into parasite food vacuole).Formation of a complex with breakdown products of Hb - toxic toparasite.


Pharmacokinetics - Rapid GIT absorption. Long duration


Use - Effective against P. falciparum and P. vivax (erythrocytic stage only).


Mainly used in malaria due to resistant strains.


Resistance to mefloquine has been reported.


Can be used for prophylaxis.


AE - GIT disturbances common. CNS disturbances (giddiness,§ dysphoria and insomnia) transient. Rare – psychoses

Anti malarial therapy


Pyrimethamine and Sulphadoxine

MOA


Pharmacokinetics


Use


AE

Anti malarial therapy


Primaquine and Qinghaosu

MOA


Pharmacokinetics


Use


AE

Define Superficial and Systemic Mycoses

Superficial Mycoses


- Infections of skin, hair and nails (dermaphytes). Superficial candidiasis invasion of mucous membrane


Systemic Mycoses


- Infections of deeper tissues. Often originate in the lung but can spread to many other organs

Anti fungal Drugs


Amphotericin B

MOA


Pharmacokinetics


Use


AE

Anti fungal Drugs


Azalea Derivatives

MOA


Pharmacokinetics


Use


AE

Anti fungal Drugs


Griserulvin

MOA


Pharmacokinetics


Use


AE

Anti fungal Drugs


Terbinafine

MOA


Pharmacokinetics


Use


AE

What are the classical signs of injury/inflammation? (5)

-redness (vasodilatation of pre-capillary arterioles - synergistic effect with other vasodilator (histamine) = increased blood flow)


-warmth (as above)


-swelling (increased vascular permeability - indirect effect via histamine - exudate and oedema)


-pain (sensitisation of afferent C fibres (PGE2) potentiate effects of bradykinin and histamine)


-loss of function

During inflammation, what local mediators are released?

– arachidonic acid metabolites


– kinins


– histamine


– nitric oxide


– cytokines

Describe COX 1 and COX 2

COX-1 (constitutive enzyme, ‘house keeping’ role) produces prostanoids that


–protect gastric mucosa


–cause platelet aggregation


–regulate renal blood flow


COX-2 (upregulated during inflammation)


produces prostanoids that cause


– inflammation


–sensitisation of pain fibres

What are the therapeutic effects of NSAIDS? (3)

Analgesic


Antipyretic


Anti-inflammatory

What are the Adverse Effects of NSAIDS?


(similar profile for all NSAIDS)

GIT bleeding/ulcer formation


Renal dysfunction/nephrotoxicity


Cardiovascular toxicity


Hypersensitivity; precipitation of asthma

What are NSAIDS and what are they indicated for?

NSAIDS are cyclo-oxygenase inhibitors. They reduce inflammation by preventing prostaglandin synthesis. (pain sensitisation and inflammation)


They have opioid sparing effects.


They are indicated for treatment of:


- arthritis


- rheumatic diseases


- pain (mild o moderate)


- gout


- inflammation


- dysmenorrhoea

NSAIDS: Aspirin

Inhibits COX1 and COX2


Use - Mild-moderate pain, Fever, Inflammation, Antiplatelet effects (low doses)


Adverse effect: gastrointestinal irritation, Indigestion, nausea, vomiting, diarrhoea, ulcers, Caused by inhibiting gut-protective prostaglandins (secrete cytoprotective mucus,inhibit gastric acid secretion).


Strategies to overcome this: Buffered preparations, Enteric coated tablets, Co-treatment with proton pump inhibitor or misoprostol (PGE analogue)

Describe the prostaglandin analogue: Misoprostol

– Stable synthetic analogue of PGE that acts on PGE receptors


– Inhibits gastric acid secretion


– Increases mucosal blood flow and secretion of mucus and HCO3-


Use for both ulcer treatment and prevention of NSAID-induced ulcers (co-administration with NSAIDs)


Adverse effects: diarrhoea, abdominal cramping


– Contraindicated in pregnancy (can induce premature labour)

Describe the clinical uses and contraindications of NSAIDS

Acute and chronic inflammation


- rheumatoid arthritis


- osteoarthritis


- gout


Analgesia


- headache


- dysmenorrhoea


- post-operative pain


- Short term: aspirin, ibuprofen


- Chronic pain: naproxen (longer lasting)


- Decrease need for opioid analgesics


Fever


Contra-Indications


- Active peptic ulcer or GIT bleeding


- Allergic reactions to NSAIDS (skin reactions, asthma)


- Renal insufficiency (PGE2 & PGI2 regulate RBF)


- CV disease/increased CVD risk; coagulation disorders

List the non-selective and selective NSAIDS

Non selective


- Aspirin


- Ibuprofen


- Naproxen (long 1/2 life, may have lower risk of CV events)


- Diclofenac (rectal/topical use, may have higher risk of CV events)


Selective


Celecoxib - COX 2 inhibitor for rheumatoid and osteoarthritis


Contraindicated in patients with CVD, increased CVD risk, peptic ulcer, history of GI bleeds, renal impairment

Describe glucocorticoids

GCs are potent anti-inflammatory andimmunosuppressant agents that bind to intracellularglucocorticoid receptors(nuclear receptors) to alter genetranscription. They inhibit expression of COX2 and phospholipase A2 (PLA2),the enzyme that releasesarachidonic acid from themembrane – Reduced synthesis ofprostanoids


Uses - chronic inflammation (RA), Skin Disorders (psoriasis, eczema) Inflammatory bowel disease (ulcerative colitis, crohns) Asthma

Describe Rheumatoid arthritis

- chronic inflammatory condition


- Joint inflammation and damage: Auto-immune reaction, Proliferation of synovium, Erosion of cartilage and bone, Pathogenesis involves IL-1 and TNF-α


Treatment: -Glucocorticoids (reduce symptoms early)


- Disease modifying any-rheumatic drugs (DMARD) - sulfasalazine (1st line, also in IBD), Hydroxychloroquine (1st ling, anti-inflam), Methotrexate (folic acid biosynthesis, AE = bone marrow toxicity and GI damage)


- TNF a antagonists (prevent cytokine from interactive with its receptor = reduce inflammatory mediators)

Describe Gout

Characterized by acute pain,swelling and tenderness injoints.


Due to high concentrations ofuric acid in the blood, resultingin deposition of uric acidcrystals in the joints and otherareas (eg kidneys).


Deposition causes aninflammatory response.


Treatment


Acute - for symptomatic relief = NSAIDS, Corticosteroids and Cholchine(prevent neutrophil invasion)


Long term - rate lowering therapy

Describe Asthma

- Asthma is a chronic obstructivedisease of the airways.


- Symptoms include wheezing, chesttightness, shortness of breath,coughing.


- Characterised by reversiblebronchoconstriction, airwayhypersensitivity and increased mucussecretion.


- Chronic changes include airway wallremodelling, fibrosis, inflammationand epithelial cell apoptosis.


Treatment:


Inhaled bronchodilators - B2 adrenoceptor agonists cause bronchodilation. AE=CV (tachycardia) and muscular (tremor) effects


Inhaled corticosteroids - decrease inflamm. response in airways; AE=hoarse voice, local fungal infections

What is Pain?

- An unpleasant sensory or emotional experience associated with actual orpotential tissue damage, or described in terms of such damage


- Important protective mechanism that warns of injury or potential injury.


- Pain signals are transmitted by small diameter peripheral nerve fibres (C and Aδ)

Describe acute vs chronic pain

- Acute pain usually has sudden onset with an obvious cause.


- Many chronic pain states are associated with hyperalgesia (heightened sensitivity of pain) or allodynia (stimuli that would not normally be perceived as painful is perceived as painful – response to non noxious stimuli)

What is Nociceptive Pain

Nociceptive pain arises from stimulation of nociceptors by injury, inflammation ordisease.


- Somatic nociceptive pain originates in the skin, bones/joints, mucosalsurfaces. Usually well-localised; can be described as sharp/stabbing/shooting.


- Visceral nociceptive pain originates in visceral organs (eg liver). Usually amore diffuse/aching pain that can be accompanied bynausea/vomiting/sweating. Can be referred from other areas in the body.


- C fibres (unmyelinated) tend to transmit diffuse/dull/aching pain; Aδ fibres(myelinated) tend to transmit localised/fast/transient pain.

What is Neurogenic Pain

Neurogenic pain arises from dysfunction in the peripheral or central nervous system due to injury or disease (eg phantom limb pain, diabetic neuropathy, spinal cord injury).


- Often associated with parasthesia, hyperalgesia, allodynia.


- Tends to respond poorly to NSAIDs and opioid analgesics; adjuncts often required.


- Hard to treat pharmacologically

What is specific pain?

Generally predictable


- Psychogenic pain can be due to psychiatric, psychological and/or psychosocial causes


- Distressing despite the lack of obvious somatic source


- Pharmacotherapy alone rarely provides relief.


- Breakthrough pain (incident pain) can occur in patients with chronic pain between regular analgesic administration.


- Postoperative pain

How do we assess pain?

- Assessment involves severity, time course, extent, associations, effects.


- Pain scales can be useful as a gauge over time


- Children can be harder to assess - face pain scales can help them visually interpret their pain

How do we treat pain?

–Treat cause in preference to symptoms


–Use accurate assessment to ensure appropriate analgesic prescription/use


–Keep ‘pain-free’. Sufficient analgesic to prevent pain


–Prevent adverse effects (eg of opioids) rather than treating upon occurrence


–Avoid under-treatment –Stepwise management

Describe Paracetamol

- analgesic and antipyretic


- less GIT irritation, allergic reactions rare - however can have fatal effects at larger doses (liver/kidney damage)


- Thought to reduce pain by inhibition of COX in the CNS.


- Combination therapy of NSAIDS and paracetamol is advantageous as it allows you toobtain the same amount of analgesic effect with lower doses. This allows you toreduce the risk of dose related adverse effects

How is paracetamol metabolised?

- Metabolism reactions are intended to make the drug more water soluble to facilitate excretion from the body. The different pathways are subject to saturation (wherethere enzyme/substrates are overwhelmed by the amount of drug present and cantmetabolise all of it)


- Paracetamol is processed down an alternate metabolic pathway. This requirescofactors such as glutathione, to allow it to be excreted. When the glutathionesupply is exhausted, paracetamol is then metabolised down another pathway thatleads to lipid peroxidisation, reactive oxygen species production and hepatocyte celldeath.


- Alternate pathway req. glutathione to be excreted


- Supplies are limited so when stores are exhausted, derivates are metabolised down a pathway that can lead to liver cell death


- If found acutely, there are options for treatment to neutralise/help with the excretion of paracetamol

Describe opioid mechanism of action

Opioids act at opioid receptors – GPCRs that reduce cAMP production


Three main subtypes


– mu (μ), MOR – most effects via.


– kappa (κ), KOR


– delta (δ), DOR


- Different effects at different receptor subtypes. Degree of analgesia and GIT effects via all three receptors.


- Different types have different localisations and distributions


- Constipation get drugs, also respiratory depression

What are endogenous opioids?

Endogenous opioids include endorphins (family of endogenous analgesics that act at opioid receptors) and enkephalins


Physiological role


–Endogenous analgesics


–Regulation of intestinal motility


–“feel good” hormones / “endorphin rush”


Pharmacology


–Rapidly metabolised, not absorbed from GIT, won’t pass through BBB, not useful as therapeutics

Describe Opioid Analgesics

- Opioid analgesics bind to and activate opioid receptors (peripherally and centrally)to produce analgesia (primary clinical use).


- Variety of administration routes/dosage forms; generally opioids are poorlyabsorbed orally with low bioavailability due to first-pass metabolism.


MOA - Activation of opioid receptors produces analgesia by inhibiting transmission through the spinal cord and activating descending inhibitory pathways. Inhibition of afferents in the periphery.

Describe Morphine

Morphine remains the prototypical opioid analgesic to which all others are compared.


Agonist at MOR.


Clinical effects include analgesia, sedation, euphoria, cough suppression. Used in the treatment of moderate to severe pain.


AEs include reduced GIT motility (constipation common), nausea/vomiting, respiratory depression


High first-pass metabolism and low bioavailability

Describe Codeine

Codeine is used for the treatment of mild pain, and as a cough suppressant.


Codeine is a prodrug – metabolised to morphine by CYP2D6 –Variability in response based on genetics


Codeine has minimal euphoric activity; constipation is a common AE.


Fentanyl is a highly potent opioid used for moderate to severe pain, as an adjunct analgesic in general anaesthesia and for breakthrough pain (especially in cancer patients).


Various formulations (SM/IV, patch, ‘lollipop’).

What is drug tolerance

Drug tolerance occurs when the effectiveness of the drug diminishes over time/repeated administration. Higher doses of the drug will be required to produce the same effect.

What is dependence?

Dependence has multiple components


–Physical (development of tolerance). Withdrawal symptoms when ceasing drug administration; reversed by re- administration.


–Psychological (craving). Less evident in patients receiving opioids for analgesia.


Withdrawal symptoms for opioids include anxiety and agitation, shivered and sweating, diarrhoea.


Precipitate withdrawal symptoms when drug use is ceased

What do opioid receptor antagonists do?

Opioid receptor antagonists reverse the effects of opioid receptor agonists (eg respiratory depression).


–Binds all three opioid receptors subtypes; higher affinity for MOR.


–Mostly used for treatment of overdose, parenteral administration.

What is Tramadol

• Tramadol is an agonist at MOR but also inhibits re-uptake of serotonin and noradrenaline


- Antidepressant as it inhibits the reuptake of serotonin

What are the analgesics of choice during pregnancy?

Paracetamol and Codeine


(NSAIDS - adverse fetal effects, prolongs gestation and labour)


(opioid use can leas to physical dependence in fetus)

What is the analgesic of choice for children?

Paracetamol

What needs to be considered prior to analgesic use in the elderly?

–Impaired liver/kidney function may slow metabolism/excretion. Dosage adjustment may be required


- May have reduced tolerance of adverse effects


-Presence of co-morbiditie. May alter drug responsiveness -> Polypharmacy


- NSAIDs in particular require care due to GIT/renal/CV adverse effects.

Briefly describe the endocrine system

System composed of specialised glands


Glands secrete hormones that act on specific targets


Integrates and regulates body functions


Disruptions in these systems result in pathological conditions

What is a hormone

A hormone is a chemical mediator secreted by a cell into the bloodstream where it is transported to a distant target cell and exerts effects at low concentrations.


Hormones act by binding to receptors in the target cell. These receptors can be intracellular or located at the cell surface

List the 3 Chemical classes of hormones

Peptides (insulin)


Amines (Catecholamines = Noradrenaline, adrenaline, dopamine)


(thyroid hormone


Steroid (mineralcorticoid, glucocorticoids)


(oestrogen)

How are peptide hormones produced?

Many peptide hormones are produced as inactive precursors, and require furtherprocessing to produce the active molecule

How are amine hormones produced?

*CHECK PP NOTES*

How are steroid hormones produced?

Produced by the parent hormone cholesterol


In the adrenal cortex – aldosterone and cortisol


In the ovary – estradiol




*CHECK PP NOTES*

How do hormones act?

Peptide and amine hormones cannot enter the cell, and therefore must interact with cell surface receptors


The downstream signalling mechanisms will vary depending on the receptor.


Steroid hormones are lipophilic


They act predominantly at intracellular receptors to alter gene transcription

what are the 3 types of stimuli for hormone release?

Humoral. Hormone release is triggered (or supressed) depending on theblood concentrations of particular ions or nutrients. (eg. glucose and insulin)


2. Neural. Hormone release is controlled by neural input.


3. Hormonal. Hormone release is caused by another hormone. A hormone that controls the release of another hormone is known as a trophic hormone.

List the endocrine and exocrine functions of the pancreas

Endocrine functions


islets of Langerhans


–subpopulation of cells


–secrete hormones


Exocrine functions


–involved in digestion


–secrete digestive enzymes

Describe Insulin

- Polypeptide hormone


- Secreted from β cells in response to increased blood glucose


- Decreases blood glucose levels by increasing glucose uptake into muscle and fat cells


- Regulates metabolism of


-carbohydrates


-fats


-proteins

Define Glycolysis, Glycogenesis, Glycogenolysis and Gluconeogenesis

Glycolysis - glucose metabolism


- the oxidation metabolism of glucose molecules to obtain ATP andpyruvate


- Pyruvate from glycolysis enters the Krebs cycle


Glycogenesis - glucose storage


- the conversion of excess glucose into glycogen as a cellular storagemechanism


Glycogenolysis - glucose release from stores


- the breakdown of glycogen into glucose, which provides a glucose supply for glucose-dependent tissues


Gluconeogenesis - glucose formation


- de novo synthesis of glucose molecules from simple organic compoundseg. the conversion of amino acids in cellular protein to glucose

Describe how insulin is released.

Low glucose = decreases metabolism and low ATP production.


- K+ channel then opens; Channel closes when ATP level is high


- When the channels closed it means that K+ is retained in the cell.


- This depolarises the membrane potential/cell. This triggers voltage activated calcium channels, whichacts as an intracellular signal and triggers exocytosis which will lead to the secretionof insulin


- Increases in glucose levels stimulate insulin secretion from pancreatic β cells

What are the actions/causes of insulin

↑ Glycolysis (glucose metabolism)


↑ Glycogenesis (glucose storage)


↓ Glycogenolysis (glucose release from stores)


↓ Gluconeogenesis (glucose formation)


All of these act to reduce blood glucose levels

Describe type 1 and type 2 diabetes

Type1 –autoimmune destruction of pancreatic beta cells


–insulin deficiency


–also known as: § –juvenile onset§ –insulin-dependent (IDDM)


Type2 –insulin resistance


–also known as § –mature onset § –non insulin-dependent (NIDDM)

What are the adverse effects of insulin

Rare with human insulin


Hypoglycaemia is most common


–Anxiety


–Cold sweating, pallor


–Shakiness, weakness


–Drowsiness, headache, nausea, confusion


If severe may result in:


–coma


–brain damage


Treatment:


–sweet drink/snack


- intravenous glucose (patient unconscious)


–glucagon

For which type of diabetes are oral hypoglycaemic agents used? Why?

Only useful in T2DM, as they require residual insulin secretion from the pancreas


However, many patients with T2DM will require additional treatment with insulins


(metformin, sulphonureas, thiazolidinediones, incretin enhancers)

Describe Metformin

Metformin is usually first-line OHA


MOA not completely understood


Increases glucose uptake and use


Reduces gluconeogenesis


Improves insulin sensitivity and blood lipids


Doesn’t affect insulin release (hypoglycaemia unlikely)


Major AEs primarily GI related

Describe Suplhonureas

Sulphonylureas – requires residual insulin secretion – function – as Sulphonylureas such as glibenclamide stimulate insulin secretion


MOA involves inhibition of ATP-sensitive potassium channels


Blockade of K+ channels causes depolarization and insulin secretion (requires functioning β cells)


AEs include hypoglycaemia (taken with food; care with alcohol consumption required)

Describe Thiazolidindiones

Thiazolidinediones (‘glitazones’) such as pioglitazone and rosiglitazone increaseinsulin sensitivity


Activate PPAR nuclear receptors that regulate expression of genes in glucoseand lipid metabolism.


AEs include peripheral oedema, dizziness, fractures (rare).

Describe Incretin Enhancers

Incretin – peptides – that are broken down and inactivated by peptidases


If you inhibit breakdown of incretin, you end up with higher levels of incretin and stimulate insulin secretion


Incretins are peptides released from the GIT and increase insulin release


DPP-4 is a peptidase that inactivates incretins


‘Gliptins’ such as sitagliptin are DPP-4 inhibitors. They increase the concentration of incretins and therefore increase insulin secretion


AEs include hypoglycaemia, headache and musculoskeletal pain

How is hormone release controlled?

- Hypothalamus releases inhibitory or secretory factors onto the pituitary.


- Anterior pituitary releases tropic hormones


- Tropic hormones act on target gland


- Increase hormone levels in the blood, send signal to Ant. Pituitary and hypothalamus, then the signal can be turned off


- Secretion of hormones is regulated by various feedback loops. Often the hormones themselves are the feedback signal. Each hormone in the pathway canfeed back to suppress hormone secretion by acting earlier in the pathway.Feedback can be long-loop or shortloop.

Describe the pituitary gland

The pituitary gland is located below the hypothalamus and is divided into two parts; the anterior and posterior lobes. The anterior lobe is considered the ‘true; endocrine gland as it is derived from epithelial tissue. It secretes the majority of the glands hormones. The posterior lobe is anatomically an extension of the neural tissue of the hypothalamus. It secretes 2 hormones: ADH and Oxytocin.

List hormones released by the anterior and posterior pituitary

Anterior


- ACTH (regulated production of corticoids


- Growth hormone


Posterior


- vasopressin/ADH (regulate water balance, inhibits diuresis)


- oxytocin (muscles in uterus)

How are adrenal hormones regulated?

- Increased levels of corticosteroids suppress the hypothalamic-pituitary axis andinhibit glucocorticoid release


- This is a major adverse effect for the chronic use of corticosteroid drugs


- HPA function can take a long time to recover after extended corticosteroid therapy


- Sudden withdrawal can be hazardous; tapering of dose is recommended

What is primary hyper secretion


**

An increase in cortisol will feedback and suppress CRH and ACTH


Low levels of upstream hormones


The adrenal gland doesn't respond to the anterior pituitary

Describe hypothyroidism

Can be due to autoimmune, iodine deficiencies, radiation therapy


Clinical signs include bradycardia, fatigue, infertility, lethargy, cold intolerance, weight gain


Can be primary (dysfunction of thyroid gland: low T3, T4; high TSH) or secondary/tertiary


Treated with T4 replacement; AEs usually correspond to hyperthyroidism

describe hyperthyroidism

Excessive thyroid hormone production = thyrotoxicosis


Primary hyperthyroidism characterised by elevated T3/T4 despite decreased TSH


Clinical features include bulging eyes, intolerance to heat, ↑appetite, weight loss, nervousness and irritability, diarrhoea


Treatment options include: Anti-thyroid drugs that inhibit thyroid hormone synthesis –Iodine(suppresses TRH/TSH in large doses); Surgery; Radioactivity iodine therapy

Describe the function of FSH and LH

FSH = follicle stimulating hormone


–Stimulates follicles in preparation for ovulation


–Development of seminiferous tubules and spermatogenesis


LH = Luteinising hormone (aka ICSH = interstitial cell stimulating hormone)


–Promotes maturation of the follicle and formation of corpus lute


–Stimulates spermatogenesis


–Formation of androgens


FSH and LH control the menstrual cycle and stimulate the release of oestrogen and progesterone

Describe the follicular/luteal cycle

- A portion of primary follicles are beginning to develop throughout thecycle, however development beyond the early stages only occurs duringthe follicular phase.


- Follicles that have developed sufficiently to respond to FSH are recruitedwhen FSH levels rise.


- Recruited follicles undergo rapid enlargement and development, andbegin producing oestrogen.


- The ‘dominant’ follicle that develops into a mature follicle usually hasthe most FSH receptors. - Follicle rupture is facilitated by enzymes that digest the connectivetissue in the follicular wall.


- Developing follicles that failed to reach maturation degenerate.

Describe Oral Contraceptives

-Provides exogenous hormones


-Exploit the negative feedback mechanisms and suppress the production of FSH and LH from the anterior pituitary


-Prevents the development of the follicle (FSH) and prevents ovulation (LH)


-Additional effects: thicken cervical mucous – increase the physical barrier


- Adverse effects: menstrual related and emotional, fluid retention, weight gain o Increase the chance of thrombotic events



How does the combined oral contraceptive work?

The combined oral contraceptive (COC) pill is extremely effective and commonly used to prevent pregnancy


Combination of an oestrogen (usuallyethinylestradiol) and a progestogen


Oestrogen acts via negative feedback on theanterior pituitary to suppress FSH release and follicle development


Progestogeninhibits LH surge and ovulation; also thicken cervical mucus


Also alter theendometrium to reduce susceptibility to implantation

How does the Progesterone only contraceptives work?

Progestogen-only contraceptives thicken cervical mucus and alter endometrium


Suppress LH surge, may inhibit ovulation –Variability based on route ofadministration; oral forms suppress ovulation in < 50% of women; depot andimplant reliably suppress ovulation (AMH 2016).


Preferred contraceptivesduring breast-feeding as no effect on lactation; safer in women with CV riskfactors (vs COC)


Can be administered by depot injection or implant


High oral dose can be used foremergency contraception within 72 (preferably) to 96 hours

What is Mifepristone and how does it work?



- Block the action of endogenous progesterone


- Partial agonist at progestogen receptors


–Blocks action of endogenousprogesterone


–Breakdown of the uterine lining


–Cervical softening anddilatation


–Release of prostaglandins


–↑contractile effects of prostaglandin


Used in combination with a prostaglandin (eg misoprostol) to terminatepregnancy in the first or second trimester

Describe hormone replacement therapy

- Daily low doses of oestrogen or oestrogen combined with progesterone.


- Treat the symptoms of menopause (hot flushes, nausea, insomnia, vaginitis, palpitations, fatigue, depression) – due to decreased oestrogen


-Provide symptomatic relief


-Prevent complications of post menopause (osteoporosis)

What are the symptoms of schizophrenia

Positive symptoms


–Auditory hallucinations


–Delusions (often paranoid)


–Abnormal experiences (wild trains of thought, irrational conclusions)


Negative symptoms


–Loss of motivation


–Lack of self-care


–Social withdrawal


–Blunted (flat) mood (anhedonia)


–Diminished speech


Cognitive function


–Deficits in executive (planning) function


–Deficits in attention


–Deficits in working memory

Describe the aetiology of schizophrenia

Genetic


- It appears that multiple “susceptibility genes” are involved in creating apredisposition to develop the disorder. There is an increased risk ofsuffering from schizophrenia if a sibling or a parent has a schizophrenicdisorder.


Environmental


- Perinatal complications: maternal complications, nutritional inadequacy, urban birth Stressful life events, drug abuse

Describe the pathology of schizophrenia

- Increased ventricles


- Decreased hippocampus size


- Hypofrontality - lower metabolic activity in the frontal regions of the brain


- Decreased neuronal #, and disorientation and abnormal grouping of neurons

What are typical antipsychotics?

- Typical antipsychotics are effective in alleviating positive symptoms (hallucinations and delusions) in a proportion of patients with schizophrenia.


- Typical antipsychotics have high affinity for dopamine receptors


- AE - extrapyramidal symptoms (acute parkinsonian symptoms = rigidity, akinesia, akathisia (restlessness) dystonia, and chronic, usually delayed onset, tar dive dyskinesia


Antidopaminergic effects (through the tuberoinfundibular system) • -hyperprolactinaemia, galactorrhea, sexual dysfunction


Anticholinergic effects -dry mouth, blurred vision, constipation, urinary retention


Antiadrenergic effects • -sedation, hypotension


Anti-histamine Anti-serotonin • -weight gain, sedation

What are atypical antipsychotics?

eg. Clozapine


- Affinity for D2 and 5HT2A receptors


- Treats positive symptoms and negative symptoms


- Very low incident EPS


- Side effects include constipation, drooling, muscle stiffness, sedation, tremors and weight gain


- Principally used in treating treatment-resistant schizophrenia

What is depression

One or more distinct periods with dysphoric mood or pervasive loss of interest or pleasure.


1. Increase or decrease in appetite or weight.


2. Excessive or insufficient sleep


3. Low energy, tiredness, fatigued Pharmacology and Toxicology


4. Psychomotor agitation or retardation


5. Loss of interest or pleasure in usual activities


6. Feelings of self-reproach, guilt


7. Decreased ability to think or concentrate


8. Recurrent thoughts of death or suicide


Duration of features for at least 2 weeks.

What are the 2 causes of depression

Exogenous (reactive)


- Life events that may cause a depressed mood such as bereavement,employment issues, relationship problems


- Medications (e.g.contraceptives, statins)


- Non-psychiatric illness (e.g. chronic pain,cardiovascular disease, Parkinson’s disease)


Endogenous


- Absence of external causes

Describe the neuroplasticity theory of depression

Neuroplasticity is the changing of neurons and the organization of their networks, and so their function, by experience. Some brain structures (e.g. thehippocampus and prefrontal cortex) are reduced in size in depressive disorder.Neuronal loss is also observed in these regions. There is evidence that someantidepressant medications promote neurogenesis.

Describe the genetic theory of depression

Stressful life events can influence the onset and course of depression However not all people who encounter a stressful life experience succumb to itsdepressogenic effect. Predisposition stress theories of depression predict thatindividuals' sensitivity to stressful events depends on their genetic makeup

Describe the Monoamine theory of depression

- Suggests that clinical depression results from low levels of the monoamines, serotonin and noradrenaline, in the brain. All antidepressants raise levels of these two neurotransmitters.


- Many areas of the brain appear to be involved in depression. However, it is not clear if the changes in these areas cause depression or if the disturbance occurs as a result of the aetiology of psychiatric disorders.

Describe the classes of antidepressants

Tricyclic Antidepressants- inhibit the reuptake of NA and serotonin into presynaptic terminals


(AE - sedation, dry mouth, constipation = likely due to blocking cholinergic receptors)


MAOi - MAOA inhibitors bind to and inhibit MAOA, preventing monoamine degradation.


(AE - postural hypotension, occasional psychosis, tremor, sexual dysfunction, anticholinergic effect)


Selective Serotonin/Noradrenaline Reuptake inhibitors (SSRIs/SNRIs) (fluoxetine)


- restore the levels of 5-HT in the synaptic cleft by binding at the 5-HT re-uptake transporter preventing the re-uptake and subsequent degradation of 5-HT


(AE - emergent anxiety and dizziness (5-HT2), nausea, headaches (5-HT3), sexual dysfunction (5-HT2), hypertension

What are anticholinergic effects

Sedation, blurred vision, urinary retention, constipation, dry mouth

Describe bipolar disorder

The affected person jumps between states of mania, depression and normalmood.


(Mania - inflated self esteem, decreased sleep, unusual talkativeness, racing thoughts, distractibility, increased goal directed activity, unusual activity = have a high potential for painful consequences)

What is Lithium used in treatment for?

Lithium salts are mood stabilizers used in the treatment of bipolar disorder since, unlike most other mood altering drugs, they counteract both mania and depression.


MOA - Inhibition dopamine release –Enhancement serotonin release –Decreased formation of second messengers


AE - nausea, vomiting, diarrhoea, weight gain, fatigue, tremor, headache, polyuria


Toxicity - very dangerous; early signs = exacerbation of normal AEs


Later signs - coarse tremor, ataxia, poor coordination, confusion, disorientation, convulsions, coma, death

Describe the polypharmacy for bipolar disorder

Patients are not just on one type of medication:


- mood stabilizers for aggression and violence


- benzodiazepines for agitation, anxiety, and insomnia


- antipsychotics for psychosis

Describe anxiety

Feelings of apprehension, agitation, uncertainty and fear


Extreme -> autonomic responses including rapid heart rate, dry mouth, sweatypalms, insomnia, loss of appetite, muscle tremor, diarrhoea


- subjective responses include feelings of fear, nervousness, excessiveworry


- escape behaviour, avoidance behaviour, freezing

What are anxiolytics used for?

Anxiety disorders


Sleep disorders


Seizure disorders


Pre operative medication


Muscle spasms


CNS depressant withdrawal

Describe benzodiazepines

Most widely used anxiolytics and hypnotics


MOA - Bind to distinct ‘benzodiazepine regulatory sites’ on GABAA receptors - Increase receptors affinity for GABA, and thus enhance the neuroinhibitory actions of GABA


Children - More sensitive to CNS depressant drugs •BNZ only really used for night terrors and sleep walking


Elderly - Usually take a variety of medications so there could be druginteractions, altered pharmacokinetics; Risk of falls due to increased sensitivity to CNS effects

Describe Beta Blockers

β-adrenoceptor antagonists e.g. propanolol


Reduce peripheral manifestations of anxiety such as tremor, sweating and tachycardia


No effect on the CNS

Depression is a progressive neurodegenerative disorder characterised by what?

- Chronic personality disintegration


- Confusion


- Deterioration of mental capacity

Describe how dementia occurs.


(features observed)

- Plaques develop in the brain – clumps of protein – contain amyloid


- Stop neurones from talking to each other


- Fibrous tangles (aggregates of Tau) don't know what causes it or what comes first


- Separation of the meninges and shrinkage of the cerebral cortex as the brain degenerates


- Shrinkage of the hippocampus (memory) bilateral holes in the brain


- Evidence of increased Alzheimer's disease and plaque development in the obese population


- Changes in acetylcholine – important in memory

List the cognitive deficits present in Alzheimers

- memory impairment


- Aphasia - problems with language (receptive and expressive)


- Apraxia - inability to carry out purposeful movements even though there is no motor or sensory impairment


- Agnosia - failure to recognise, especially people


(decreased need for sleep)

In patients with Alzheimers, what deficits are seen in the cortex and hippocampus?

1. Choline acetyltransferase activity (ChAT)


2. Acetylcholine content


3. Choline transport


4. Nicotinic Acetylcholine receptors

Describe the treatments for Alzheimers

Acetylcholinesterase inhibitors


- donepezil, galantamine, rivals timing


- Efficacy: some improvement of cognition in mild cases


- Side effects: nausea and vomiting


NMDA antagonist


- Memantine


- Glutamate antagonist


- Treatment of moderate to severe cases


- Efficacy: moderately efficacious in the treatment of moderate to severe


- Side effects: CNS effects - confusion, dizziness, drowsiness


Polypharmacy


- Often patients are not just on one type of medication:


- antidepressants for depression or obsessive-compulsive symptoms,


- mood stabilizers for aggression and violence


- benzodiazepines for agitation, anxiety, and insomnia


- antipsychotics for psychosis

Describe Parkinsons Disease and the symptoms

- Progressive debilitating disorder


- Occurs between 50 and 80 years


- Dopamine-deficient state of the extrapyramidal motor system,particularly a loss of dopaminergic neurons in the nigrostriatalpathway Decrease in dopamine (inhibitory) leading to adopamine/acetylcholine (excitatory) imbalance


- Tremors at rest Bradykinesia – slowing of all voluntary movements


- Forward flexion of the trunk


- Muscle rigidity

Describe Levodopa and how it treats Parkinsons

- Decarboxylated to dopamine within surviving nigrostriatal fibres or in other monaminergic neurons to provide some restoration of nigrostriatal activity.


- Prescribed with carbidopa (a dopamine decarboxylase inhibitor which decreases the production of dopamine in the periphery and reduces levodopa dosage)


- Tries to make dopamine from the receptors that are still there but there isn't any improvement in the condition of the Brain; however over time the neurones degrade/degenerate


Pro - improves the slowness of movement, rigidity and tremor


AE - Anxiety, dyskinesias, confusion, hypotension, moodchanges, nausea, Effectiveness wears off after chronic use, chronic use = other movement alterations unrelated to Parkinsons


Interactions: –Anticonvulsants and neuroleptics (decreaselevodopa effects by increasing metabolism) –Antihypertensives (increased risk hypotension) –MAO-A inhibitors (hypertensive crisis)

Describe the other therapeutics used for Parkinsons

Dopamine agonists (Pramipexole, Apomorphine, Pergolide)


- improve motor function and improve the off time (when dopamine stops working)


- Similar AE to L-Dopa


Selegiline


- Selectively Blocks MAO-B - prevents dopamine from being degraded resulting in more dopamine


AE - metabolised to amphetamine ->excitement, anxiety and insomnia


Entacapone


- blocks COMT, doesn't cross the BBB


- prevents the breakdown of L-Dopa


- AE - dyskinesia (^ voluntary movement) and some GIT problems

Describe epilepsy

- Group of chronic disorders characterised by sporadic recurrent episodes of convulsive seizures


- Seizures associate with episodic high-frequency discharge by a group of neurones in the brain


- Site of primary discharge (focus) and extent of spread determine kind of seizure


- There are around 40 different types of seizures.


- Abnormal activity during and following a seizure can be detected by electroencephalogram (EEG) recordings

Describe the different types of seizures

Simple Partial


- no loss of awareness


- motor symptoms, altered hearing, smell, taste, sight, tactile perception


- laboured speech/ inability to speak


Complex Partial


- change in awareness/behaviour


- Sudden and inexplainable feelings of fear, anger, sadness, happiness or nausea


- Aura, automatism (chewing, swallowing movements)


- Motor symptoms


Tonic Clonic


- contracture of the whole musculature causing spasm followed by a series of violent synchronous jerks


- Respiration stops; defecation, urination, salivation occurs, Face becomes blue (important clinical symptom) Patient can stayunconscious for a few minutes followed by recovery


EEG = continuous high frequency activity in tonic phase, intermittent discharge in clonic phase


Absence


- everything stops abruptly – unaware, stare vacantly – then recover abruptly


- children


- EEG - rhythmic discharge


Status Epilepticus


- continuous seizure, longer than 30mins or 2 consecutive without recovery of consciousness


- In humans sustained seizures can cause selective neuronal loss invulnerable regions (eg. hippocampus and cortex)


- The extent of the neuronal injury is closely related to the duration ofthe seizure, therefore Status Epilepticus requires rapid control


(Drug overdose, low glucose, head trauma)

Describe pharmacological treatment for epilepsy


(Sodium Channel inhibition)

Carbamazepine


MOA - –Prevents repetitive neuronal discharge by blocking voltagedependent and use-dependent sodium channels. This stops it from returning to resting stage ad prevents the generation of action potentials = reduced membrane excitability


Indication - simple and complex patrol, tonic-clonic


AE - –Drowsiness, ataxia, dizziness, blurred vision,nausea, vomiting, rash, dry mouth


Pharmacokinetics - absorbed orally, t1/2 of 30hrs, mainly metabolised by the liver by CYP3A4


Interactions - oral contraceptives, anticonvulsants, warfarin, antipsychotics, corticosteroids, benzodiazepines


- interacts with grapefruit, affects the plasma


Phenytoin


MOA - –Prevents repetitive neuronal discharge by blocking voltagedependent and use-dependent sodium channels. This stops it from returning to resting stage ad prevents the generation of action potentials = reduced membrane excitability


Indication - simple and complex partial, tonic-clonic, status epilepticus


AE - –Nausea, vomiting, insomnia, agitation, sedation, ataxia, confusion, behavioral disturbances, teratogenic


Pharmacokinetics - absorbed orally, mainly metabolised by the liver by CYP3A4


Interactions - lowers blood concentration of anti-epileptics, chronic alcohol abuse lowers phenytoin concentration


- diabetes = increases risk of hyperglycaemia

Describe pharmacological treatment for epilepsy(Calcium Channel and GABA inhibition)

Calcium Channel


Ethosuximide


MOA - Reduces low threshold voltage-dependent calcium conductance in neurons


Indications - absence seizures


AE - Anorexia, nausea, vomiting, drowsiness, ataxia


Pharmacokinetics - well absorbed orally, t1/2 60hrs


Interactions - other anti-epileptic drugs (valproate)


GABA


Benzodiazepines


MOA - Potentiates the inhibitory effects of GABAacting on GABAa receptors


Indications - all seizures including status epileptics and febrile


AE - Main side effect is sedation, however may also include memory loss, ataxia


Pharmacokinetics - generally not suited for long term use due to sedative and tolerance effects


Valproate


MOA - Makes GABA more available by inhibiting the GABE transaminase


Indications - generalised and partial seizures


AE - Nausea, vomiting, increased appetite (weight gain), ataxia, baldness,teratogen, liver damage


Pharmacokinetics - well absorbed orally, t1/2 15hrs; metabolised by P450 enzymes,


-teratogen


Interactions - clearance impaired by aspirin, caffeine and antibiotics (lead to increased toxicity, hepatotoxicity)


Gabapentin


- Used with other drugs to treat Partial and some generalised seizures


- Few lasting side effects, some sleepiness and dizziness


Elderly - Interactions with warfarin, anti-hypertensive, anti-depressants; Ageing population hepatic and renal systems may be reduced


Pregnancy - Several anti-epileptics induce hepatic enzyme CYP3A4 (increaseoral contraceptive metabolism); Teratogenic effects are produced; Risk benefit assessment





Describe ADHD

- People with ADHD show a persistent pattern of inattention and/or hyperactivity-impulsivity that interferes with functioning or development


Genetics - twin studies show heritability


Environment - Low birth weight, maternal infection, premature birth


Pathology


- General reduction of brain volume


- Significant global thinning of the cortex, corpus callosum volumetric reductions


- Frontal lobe dysfunction


- Reported abnormalities in the functioning of dopaminergic, adrenergic, serotoninergic and cholinergic pathways

Describe ADHD Treatment

Methylphenidate (ritalin)


Noradrenaline and dopamine reuptake inhibitor


- Psychostimulant


- Stimulates the release of dopamine and noradrenaline into the synapse


- Increases or maintains alertness, improves attention


- Stimulate brain changes involved in learning and memory


- Side effects: nausea, loss of appetite, anxiety, insomnia, irritability, drymouth, tachycardia, palpitations, tremor


Dextroamphetamine


- Psychostimulant o


- Enhances dopamine and noradrenaline neurotransmission


- Side effects: headache, nausea, loss of appetite, anxiety, insomnia, dry mouth, tachycardia, palpitations

Describe Autism and Asbergers

- No medication recommendations listed in the AMH


- antidepressants may be prescribed for anxiety


- antipsychotics to treat severe behavioural problems


- ADHD medications may be prescribed for hyperactivity

What are the 4 forms of drug abuse?

Experimental abuse - use drugs in an exploratory way, may accept or reject to take drug action


Recreational abuse - social contexts


Compulsive drug abuse - irrational abuse of drug


Polydrug abuse - depressants and stimulants

list the stages of addiction

1. acute reinforcement/social drug taking


2. escalating/compulsive use/binging


3. dependence


4. withdrawal


5. protracted withdrawal


6. recovery


(stages 4 and 6 can relapse back to stage 2)

What are the 3 most important predictors of abuse in relation to personality?

1. rebelliousness


2. tolerance of deviance


3. low school performance

Describe the pharmacological aetiology of addiction

- Drugs of abuse act on different chemicals/pathways in the brain, however they can cause certain common effects after both acute and chronic exposure


- Drugs are all acutely rewarding, but chronically in vulnerable individuals can lead to addiction


- All drugs upon withdrawal, produce similar negative emotional symptoms, a prolonger period of sensitisation toward drug related environmental cues


- this contributes to drug craving and relapse, even after long periods of abstinence

What is the mesolimbic pathway important for?

-memory


-motivating behaviours

list different parts of the brain and what they play an important role in.

Prefrontal cortex - decision making


Amygdala- emotion


Hippocampus - memory


Nucleus Accumbens - reward centre


Ventral tegmental area - dopamine tracts

Describe the acute effects of alcohol

- causes progressive and continuous depression from the forebrain to hindbrain (enhances GABAergic inhibition)


- vasodilation = hypothermia


- stimulation gastric juice and salivary secretions = vomiting


- lowers ADH = diuresis


- uncoordination = accidents/injury


- lowers blood sugar = possible seizures


- disinhibition, lack of emotional control, memory loss

Describe the chronic effects of alcohol

brain = dementia, korsakoffs syndrome, cerebellar degeneration


neurological = neuropathy


muscle = chronic myopathy (wasting of proximal limb muscles)


liver = cirrhosis


pancreas/GIT = abdominal pain, fat malabsorption, diabetes


CV system = hypertension, CVD


Malignancy = cancer of mouth, larynx, pharynx, oesophagus


foetal alcohol syndrome


Psyche = hallucinations, depression, anxiety