Sei sulla pagina 1di 66

Drugs

 /  Medication  List    

Pharmacology  And  Therapeutics  (Spring  Term)  

Drug  Name   Action   Use   Reference  

ATROPINE   Peripherally  Acting  Muscarinic  Antagonist  –  HEART   After  myocardial  infarction  (where  there  is  typically  a  lot  of   P&T  Spring  
RATE   reflex  vagus  activity  which  depresses  the  heart  activity/HR,   Lecture  1  
and  is  therefore  reversed  by  atropine)  
(Antimuscarinic  Drug)    

IPRATROPIUM  (bd,  td)   Peripherally  Acting  Muscarinic  Antagonist  –   The  drugs  used  to  modify  bronchial  function  in  clinical  
BRONCHI   practice.    
TIOTROPIUM  (od)    
(Antimuscarinic  Drug)   Ideally  administered  via  inhalation  because:  

  1. Drug  directed  to  the  target  tissue  


2. By  directly  targeting  bronchial  tissue,  a  lower  dose  is  
required  so  fewer  systemic  effects.  

Clinically  useful  in  conditions  such  as  ASTHMA  and  COPD.  


 

OXYBUTYNIN   Peripherally  Acting  Muscarinic  Antagonist  –   Useful  to  modify  bladder  function.  
BLADDER  
TOLTERODINE   Tolterodine  is  relatively  specific  to  bladder  β2  receptors  than  
(Antimuscarinic  Drug)   other  drugs  (like  atropine).  

  Clinically  useful  in:  

• OVERACTIVE  BLADDERS  
• URINARY  FREQUENCY  
• INCONTINENCE  

The  drug  tightens  the  sphincter  (increased  sympathetic,  


decreased  parasympathetic  control),  so  decreased  leakage.  

However,  these  drugs  do  not  work  on  stress  incontinence  (i.e.  
increased  intra-­‐abdominal  pressure).  
 
TROPICAMIDE   Peripherally  Acting  Muscarinic  Antagonist  –  EYE   GLAUCOMA   P&T  Spring  
Lecture  1  
(Antimuscarinic  Drug)   DILATION  OF  THE  PUPILS  

  Tropicamide  is  a  diagnostic  drug  which  is  designed  for  use  only  
in  the  eye.  
 

ADVERSE  EFFECTS  OF  ALL  ANTIMUSCARINIC  DRUGS  GIVEN  SYSTEMICALLY:  

• Dry  mouth  (most  commonest  symptom)  


• Erectile  dysfunction  
• Bronchodilation  (perhaps  good  side  effect?)  
• Constipation  
• ‘Too  Good’  Sphincter  –  Decreased  detrussor  activity  so  increased  urinary  bladder  retention  
• Dry  Eyes  
• Blurred  vision  
• Increased  intraocular  pressure  
 

HYOSCINE   Centrally  Acting  Muscarinic  Antagonist   Acts  in  the  BRAIN.   P&T  Spring  
Lecture  1  
(Antimuscarinic  Drug)   Hyoscine  is  similar  to  atropine,  but  more  sedating.  Widely  
used  in  travel/sea-­‐sickness  (labyrinthine  sedative).  
   

BENZHEXOL   Centrally  Acting  Muscarinic  Antagonist   Used  as  to  treat  Parkinson’s  Disease  
 
(Antimuscarinic  Drug)  

 
 
   
Angiotensin   Angiotensin  Converting  Enzyme  Inhibitors   These  inhibit  the  somatic  form  of  the  ACE  enzyme,  so  prevent   P&T  Spring  
Converting  Enzyme   the  conversion  of  angiotensin  I  into  angiotensin  II.   Lecture  2  
Inhibitors   (ACE-­‐Inhibitors  /  ACEI)  
Uses:  
(e.g.  ENALAPRIL,    
LISINOPRIL)   • Hypertension  
• Heart  failure  
• Post-­‐myocardial  infarction  
• Diabetic  neuropathy  
• Progressive  renal  insufficiency  
• Patients  at  high  risk  of  cardiovascular  disease.  

Side  Effects:  

• Cough  
• Hypotension  
• Urticaria  /  Angioedema  
• Hyperkalaemia  (Contraindications  –  take  care  if  the  
patient  is,  or  may  be  prescribed,  K+  supplements  or  
K+  sparing  diuretics  which  may  further  increase  blood  
K+  levels)  
• Foetal  Injury  
• Renal  failure  in  patients  with  renal  artery  stenosis  
(secondary  to  fall  in  bp)  
 

Angiotensin  Receptor   Angiotensin  Receptor  Blockers   Acts  as  antagonists  of  the  Type  I  Receptors  (AT1)  receptor  for  
Blockers   angiotensin  II.  
(ARB/AIIA)  
(e.g.  LOSARTAN,   This  prevents  the  renal  and  vascular  actions  of  angiontensin  
IRBESARTAN)   II.  

They  are  widely  used  in  hypertension  as  an  alternative  to  ACEI  
(fewer  side  effects),  and  are  used  in  chronic  heart  failure  
patients  who  cannot  tolerate  ACEI.  

Uses:  

• Similar  to  ACEI  


• Alternative  therapeutic  intervention  
Side  Effects:  

• Hypotension  
• Hyperkalaemia  (Contraindications  –  take  care  if  the  
patient  is,  or  may  be  prescribed,  K+  supplements  or  
K+  sparing  diuretics  which  may  further  increase  blood  
K+  levels)  
• Foetal  Injury  
• Renal  Failure  in  patients  with  renal  artery  stenosis  
(secondary  to  a  fall  in  bp,  meaning  a  reduced  renal  
perfusion)  
 

Direct  Renin   Direct  Renin  Antagonist   • Inhibits  the  enzyme  activity  of  renin,  so  prevents  the   P&T  Spring  
Antagonist   conversion  of  angiotensinogen  into  angiotensin  I   Lecture  2  
• Ultimately  prevents  the  formation  of  angiotensin  II  
(e.g.  ALISKIREN)   • New  class  of  agents  
 
 
   
 

PHENYLALKYLAMINES   Calcium  Channel  Blockers  (CCB)   • Reduce  Ca2+  entry  into  cardiac  and  smooth  muscle   P&T  Spring  
(e.g.  Verapamil)   cells   Lecture  2  
Rate  Slowing  Calcium  Antagonists   • Negative  inotropy  effects  (decrease  contractility)  
• Inhibits  AV  Node  Conduction  
Cardiac  and  Smooth  Muscle  Actions  
Uses:  

• HYPERTENSION  
• ANGINA  
• Treating  Paroxysmal  SVT  (Tachycardia  originating  
above  the  ventricular  tissue)  
• Atrial  Fibrillation  

Unwanted  Actions:  

• Bradycardia  and  AV  Block  


• Worsening  of  Heart  Failure  
• Constipation  

Negative  Ionotropic  Effect:  Verapamil  >  Ditiazem  


 

BENZOTHIAZEPINES   Calcium  Channel  Blockers  (CCB)   • Reduce  Ca2+  entry  into  cardiac  and  smooth  muscle  
(e.g.  Diltiazem)   cells  
Rate  Slowing  Calcium  Antagonist  
Uses:  
Cardiac  and  Smooth  Muscle  Actions  
• HYPERTENSION  
• ANGINA  

Unwanted  Actions:  

• Bradycardia  and  AV  Block  


• Worsening  of  Heart  Failure  
• Constipation  

Negative  Ionotropic  Effect:  Verapamil  >  Ditiazem  


 
DIHYDROPYRIDINES   Calcium  Channel  Blockers  (CCB)   • Inhibits  Ca2+  entry  into  vascular  smooth  muscle  cells  
(e.g.  Amlodipine)  
Non-­‐Rate  Slowing  Calcium  Antagonist   Uses:  

Smooth  Muscle  Actions  Only   • HYPERTENSION  


• ANGINA  (Dihydropyridines  are  preferred  here)  

Unwanted  Actions:  

• Ankle  Oedema  
• Headache/Flushing  
• Palpitations  (Reflex  tachycardia)  
 
 

Beta  Blockers   Competitive  Antagonists  of  Beta-­‐Adrenoceptors   Uses:   P&T  Spring  


Lecture  2  
(β-­‐Adrenoceptor   Atenolol  –  Selective  β1  Blocker   • Angina  
Antagonists)   • Post  Myocardial-­‐Infarction  
Bisoprolol  –  Selective  β1  blocker   • Cardiac  Dysrhythmias  
e.g.  ATENOLOL,   • Chronic  Heart  Failure  
BISOPROLOL,   Propranolol  –  Non-­‐Selective  β-­‐Blocker  
• Hypertension  
PROPRANOLOL   • Also  in:  
o Thyrotoxicosis  
o Glaucoma  
o Anxiety  States  
o Migraine  
o Prophylaxis  
o Benign  Essential  Tumour  

Mechanism  of  Action:  

• Competitive  antagonist  of  Beta  Adrenoceptors  


• Many  clinically  used  agents  show  selectivity  (e.g.  
atenolol  for  B1)  

Use  in  Hypertension:  

• No  longer  1st  line  treatment  


• Mechanism  not  fully  understood,  but  B1  antagonists  
preferred  
• They  do  not  reduce  PVR  

Effects:  

• Reduce  cardiac  output  


• Reduce  renin  release  by  the  kidney  
• May  diminish  NA  release  by  sympathetic  nerves  
• Lipophilic  agents  (e.g.  propranolol)  exert  central  
sympatho-­‐inhibitory  actions.    

Unwanted  Actions:  

Can  be  due  to  either  the  actions  on  β1  and  sometimes  due  to  
β2  in  partial  selectivity.  

• Worsening  of  cardiac  failure  


• Bradycardia  (heart  block)  
• Bronchoconstriction  
• Hypoglycaemia  (in  diabetics  on  insulin)  
• Increased  risk  of  new  onset  of  diabetes  
• Fatigue  
• Cold  extremities  and  worsened  peripheral  artery  
disease  
• Impotence  
• CNS  effects  (lipophilic  agents)  e.g.  nightmares  

ORGANIC  NITRATES     Mechanism  of  Action:   P&T  Spring  


Lecture  2  
(e.g.  glyceryl  trinitrate   • Stimulate  the  release  of  NO  in  smooth  muscle  cells  
(GTN)  and  nicorandil)   (nitrate  based  drugs)  
• Stimulate  guanylate  cyclase  (NICORANDIL)  
• Causes  VASODILATION  

Uses:  

• Angina  
• Acute  and  chronic  heart  failure  
• BP  control  during  anaesthesia  
Effects:  

• Reduces  PRELOAD  (venous  return)  


• Reduces  AFTERLOAD  (peripheral  resistance)  
• Minor  Effects:  Antiplatelet  agents,  coronary  artery  
vasodilators  

Pharmacokinetics:  

• Nitrates  undergo  extensive  first  pass  metabolism  by  


the  liver.  
• GTN  is  often  given  sublingually  for  rapid  angina  relief.  
• Longer  acting  transdermal  patches  available  (e.g.  GTN  
and  isosorbide  mononitrate)  

Unwanted  Effects:  Hypotension,  headaches  and  flushing  


associated  with  vasodilation.  

Excess  Use:  associated  with  tolerance.  


 
 
   
Anti-­‐Arrhythmic  Drugs   Treat:  SUPRAVENTRICULAR  ARRHYTHMIAS   ADENOSINE:   P&T  Spring  
e.g.  adenosine,  amidoarone,  dronedarone     Lecture  3  
• Used  i.v.  to  terminate  supraventricular  
Verapamil  (CCB)   tachyarrhythmias  (SVT)  
• Short-­‐lived  action  (20-­‐30s)  
• Safer  to  use  than  verapamil.  

Mechanism  of  Action  

• Adenosine  is  an  endogenous  mediator  produced  by  


the  metabolism  of  ATP.  
• Acts  on  adenosine  receptor  (A1)  to  hyperpolarise  
cardiac  tissue  and  slow  conduction  through  AV  node.  

Adverse  Effects:  

• Chest  pain  
• Dyspnoea  (shortness  of  breath)  
• Dizziness  
• Nausea  

AMIODARONE  &  DRONEDARONE:  

• Used  in  supraventricular  and  ventricular  


tachyarrhythmias  
• Complex  mechanism  of  action  –  probably  involves  
multiple  ion  channel  block.  

Adverse  Effects:  

• Amiodarone  accumulates  in  the  body  (t1/2  10-­‐100d)  


• Has  a  number  of  important  adverse  effects:  
o Photosensitive  skin  rashes  
o Hypo-­‐  and  Hyper-­‐  thyroidism  
• Pulmonary  fibrosis  
• Corneal  deposits  
• Neurological  and  gastrointestinal  disturbances  
 
• Dronedarone  is  non-­‐iodinated  and  less  toxic  than  
amidarone,  but  less  effective.  
 

Treat:  VENTRICULAR  ARRHYTHMIAS    


e.g.  flecainide,  lidocaine,  

(Amidarone,  Dronedarone)  
 

Treat:  COMPLEX  (e.g.  supraventricular  arrhythmias    


and  ventricular  arrhythmias)  
disopyramide  

DIGOXIN  and  CARDIAC   Cardiac  Glycosides   • Digoxin  slows  ventricular  rate  in  ATRIAL   P&T  Spring  
GLYCOSIDES   FIBRILLATION  and  relieves  the  symptoms  of  CHRONIC   Lecture  3  
HEART  FAILURE.  
 
• Long  t1/2  of  ~40hours  
• Narrow  therapeutic  window  
• An  immune  Fab  (Digibind)  is  available  for  digoxin  
toxicity.  

Mechanism  of  Action:  

• Inhibits  Na-­‐K-­‐ATPase  (Na/K  Pump)  


• This  results  in  the  increased  accumulation  of  
intracellular  Na+,  so  increases  intracellular  Ca2+  (as  
more  Na+  can  be  exchanged  out  of  the  cell  for  Ca2+  
via  the  Na+/Ca2+  exchanger)  

 
• So  POSITIVE  INOTROPIC  EFFECT.  
• Central  vagal  stimulation  causes  reduced  rate  of  
conduction  through  AV  node  

Adverse  Effects:  (Common  and  severe)  

• Dysrhythmias  (e.g.  AV  Conduction  block,  ectopic  


pacemaker  activity)  
 
• N.B.  hypokalaemia  and  hypomagnaesia  (usually  a  
consequence  of  diuretic  use),  lower  the  threshold  for  
digoxin  toxicity.    
 

IVABRADINE     Use:   P&T  Spring  


Lecture  3  
• Treating  angina  in  patients  with  normal  sinus  rhythm.  

Mechanism  of  Action:  

• Blocks  If  Channel  (f-­‐funny)  (an  Na-­‐K  channel  important  


in  the  SA  node)  
• Slows  heart  rate.  

Contraindications:  

• Severe  bradycardia  
• Sick  Sinus  Syndrome  
• 2-­‐3rd  degree  heart  block  
• Cardiogenic  Shock  
• Recent  MI  

Adverse  Effects:  

• Bradycardia  
• First-­‐degree  heart  block  
• Ventricular  and  supraventricular  arrhythmias  
CARDIAC  INOTROPES   Dobutamine  –  β1  adrenoceptor  AGONIST  (with  little   Agents  that  INCREASE  THE  FORCE  OF  CARDIAC    
(e.g.  Dobutamine  and   effect  on  heart  rate)   CONTRACTION    
Milrinone)  
  Used  to  treat  acute  heart  failure  in  some  situations  (e.g.  after  
cardiac  surgery  or  in  cardiogenic/septic  shock).  
Milrinone  –  Phosphodiesterase  inhibitors.    
Have  inotropic  effects  by  inhibiting  breakdown  of   Despite  increasing  cardiac  contractile  function,  so  far  all  
cAMP  in  cardiac  myocytes.   inotropes  have  reduced  survival  in  chronic  heart  failure.  

ALPHA  BLOCKERS  (e.g.   Alpha  Blockers  –  antagonists  of  α1-­‐adrenoceptors   Alpha  blockers  can  be:   P&T  Spring  
doxazosin  and       Lecture  3  
phenoxybenzamine)   • COMPETITIVE  e.g.  dozazosin  
• IRREVERSIBLE  e.g.  phenoxybenzamine  
and  SYMPATHOLYTICS  
(clonidine,   Used  occasionally  in  combination  with  anti-­‐hypertensives  in  
moxonidine)   resistant  hypertension,  but  routine  use  has  declined  since  
shown  to  be  associated  with  increased  rates  of  chronic  heart  
failure.  

Phenoxybenzamine  –  combined  with  a  beta-­‐blocker,  provides  


long-­‐lasting  alpha  blockade  in  catecholamine-­‐secreting  
tumours  (e.g.  phaeochromacytoma)  

Sympatholytics  

Centrally  acting  anti-­‐hypertensive  agents  e.g.  clonidine  (α2*  


adrenoceptor  agonist)  and  moxonidine  (imadazoline  agonist)  
inhibit  sympathetic  outflow  from  the  brain,  and  occasionally  
used  as  antihypertensive  agents.    

(*α2  is  an  inhibitory  alpha  adrenergic  receptor)  

 
VASOCONSTRICTORS   Sumitriptan  used  in  migrane  treatment   SUMITRIPTAN   P&T  Spring  
Lecture  3  
e.g.  Sumitriptan   • Agonst  at  5HT1D  Receptor  (Serotonin  Receptor)  
• Constricts  some  large  arteries  and  inhibits  trigeminal  
nerve  transmission  
• Used  to  treat  migraine  attacks.  
• Contraindication  in  patients  with  coronary  disease.  

Other  ergot  alkaloids  are  also  used  in  migraine  (usually  act  as  
partial  agonist  of  5HT1  receptors)  
 

ADRENALINE   Endogenous  catecholamine   Produced  by  the  adrenal  gland,  used  in  cardiac  arrest  and   P&T  Spring  
anaphylactic  shock.   Lecture  3  

PROMETHAZONE   Anti-­‐emetic   Acts  centrally  (labyrinth,  NTS  and  vomiting  centres)  to  block   P&T  Spring  
activation  of  the  vomiting  centre.   Lecture  10  
-­‐  ANTI-­‐EMETIC   • Acts  as  a  competitive  antagonist  at  
Histaminergic  (H1),  Muscarinic  Cholinergic   Use  an  anti-­‐emetic  in:  
(M)  and  Dopaminergic  (D2)  Receptors  
• Potency:  H1  >  M  >  D2   • Motion  Sickness  (Prophylaxis,  and  during  onset)  
• Disorders  of  Labyrinth  (e.g.  Meniere’s)  
  • Hyperemesis  Gravidarium  
• Pre  &  Post-­‐Operatively  (sedative  and  anti-­‐muscarinic  
effects  are  also  useful)  

Other  Uses:  

• Relief  of  allergic  symptoms  


• Anaphylactic  emergency  
• Night  sedation  and  insomnia  
Unwanted  Effects:  

• Dizziness  
• Tinnitus  
• Fatigue  
• Sedation  
• Excitation  in  excess  
• Convulsions  (children  more  susceptible)  
• Antimuscarinic  side-­‐effects  

Pharmacokinetics:  

• Oral  administration  
• Onset  of  action  1-­‐2h  
• Maximum  effect  at  around  4h  
• Duration  of  action  –  24h  
 
• Order  of  antagonistic  potency:  D2  >>  H1  >>  Muscarinic  
METACLOPRAMIDE   Primarily  Dopamine  Receptor  Antagonist   Receptors   P&T  Spring  
  Lecture  10  
-­‐  ANTI-­‐EMETIC   Potency:  D2  >>  H1  >>  Muscarinic  Receptors   • Acts  centrally,  especially  at  the  Chemoreceptor  
Trigger  Zone  (CTZ)  
 
• Acts  in  the  Gastrointestinal  Tract:  
o INCREASES  SMOOTH  MUSCLE  MOTILITY  
(from  oesophagus  to  small  intestine)  
o ACCELERATED  GASTRIC  EMPTYING  
o ACCELERATED  TRANSIT  OF  INTESTINAL  
CONTENTS  (from  duodenum  to  ileo-­‐coecal  
valve)  
 
NOTE  –  Care  must  be  taken  with  bioavailability  of  co-­‐
administered  drugs  
e.g.  adsorption  and  hence  effectiveness  of  digoxin  may  be  
reduced.  Nutrient  supply  may  be  compromised  –  especially  
important  in  conditions  such  as  diabetes  mellitus.  
 
 
 
 
Use:  
Used  to  treat  nausea  and  vomiting  associated  with:  
• Uraemia  –  Severe  renal  failure  
• Radiation  Sickness  
• Gastrointestinal  Disorders  
• Cancer  Chemotherapy  (high  doses)  e.g.  cisplatin  
(intractable  vomiting)  
 
Unwanted  Effects:  
• Drowsiness  
• Dizziness  
• Anxiety  
• Extrapyramidal  reactions:  
o Children  more  susceptible  than  adults  
(Parkinsonian-­‐like  syndrome:  rigidity,  tremor  
and  motor  restlessness)  
• Note:  No  Anti-­‐Psychotic  Actions  
• In  the  endocrine  system:  
o Hyperprolactinaemia  
o Galatorrhoea  
o Disorders  of  menstruation  
Pharmacokinetic  Considerations:  
• May  be  administered  orally  –  rapidly  absorbed  
• Extensive  first  pass  metabolism  
• May  also  be  given  intravenously  
• Crosses  BBB  
• Crosses  Placenta  

 
Mode  of  action  
HYOSCINE   Muscarinic  Receptor  Antagonist  (Anti-­‐Muscarinic)   • Order  of  antagonistic  potency:  Muscarinic  >>>  D2  =  H1   P&T  Spring  
Receptors   Lecture  10  
 -­‐  (ANTI-­‐EMETIC)   • Acts  centrally,  especially  in  the  VESTIBULAR  NUCLEI,  
NTS,  VOMITING  CENTRES  to  block  activation  of  
vomiting  centres.  
 
Use  as  an  Anti-­‐Emetic  
• Prevention  of  motion  sickness  
• Has  little  effects  once  nausea/emesis  is  established  
• In  operative  pre-­‐medication  
N.B  Atropine  is  less  effective  
 
Unwanted  Effects:  
• Typical  Anti-­‐Muscarinic  Side-­‐Effects:  
o Drowsiness,  Dry  Mouth  
o Cyclopegia  (Paralysis  of  cilliary  muscles  of  the  
eye)  
o Mydriasis  
o Constipation  (not  usually  at  anti-­‐emetic  
doses)  
 
Pharmacokinetic  Considerations  
• Can  be  administered  orally  (peak  effect  in  1-­‐2  hours)  
• Intravenous  
• Transdermally  
 
 
ONDANSETRON   5HT3  RECEPTOR  ANTAGONIST   Use  as  an  anti-­‐emetic:    
  • Main  use  in  preventing  anti-­‐cancer  drug-­‐induced  
Mode  of  action:   vomiting,  especially  cisplatin  
• Acts  to  BLOCK  TRANSMISSION  IN  VISCERAL   • Radiotherapy-­‐induced  sickness  
AFFERENTS  and  CTZ   • Post-­‐Operative  nausea  and  vomiting  
Unwanted  Effects:  
  • Headache  
• Sensation  of  flushing  and  warmth  
• Increased  large  bowel  transit  time  (constipation)  
Pharmacokinetic  considerations:  
• Administer  Orally  
• Well  absorbed,  excreted  in  the  urine.  
 
 
DIURETIC:   Osmotic  Diuretics  are:   • Clinical  uses:    
o Prevent  ACUTE  renal  failure  (by  increase  H2O  
OSMOTIC  DIURETIC   • Pharmacologically  inert   excretion)  
e.g.  Mannitol   • Filtered  by  the  glomerulus,  but  not   (urine  production  ceases)  
reabsorbed.    
• Increase  the  osmolarity  of  tubular  fluid  (and   o Reduce  INTRA-­‐CRANIAL  PRESSURE  
plasma)  so  reduce  the  osmotic  gradient.     o Reduce  INTRA-­‐OCULAR  PRESSURE  
  o Increase  Plasma  Osmolarity  
Therefore,  they  DECREASE  water  reabsorption  where    
the  nephron  is  freely  permeable  to  water:   • Unwanted  Effects:  
  o Increased  EXTRACELLULAR  FLUID  volume  
• Proximal  Tubule   which  can  lead  to:  
• Descending  Limb  of  Loop  of  Henle   § Hyponatraemia  (associated  with  
• Collecting  Duct   nausea,  vomiting  and  pulmonary  
  oedema)  
This  causes  a  DECREASED  H2O  REABSORPTION  and    
INCREASED  H2O  EXCRETION  
(There  is  also  a  small  increase  in  Na+/Cl-­‐  loss)  

 
  Increased  delivery  of  HCO3-­‐  and  Na+  to  the  distal  tubule,  so  
DIURETIC   Carbonic  anhydrase  inhibitors  are  weak  diuretics.   an  increased  K+  loss.    
They  act  mainly  on  the  PROXIMAL  TUBULE,  to:    
CARBONIC     INCREASED  TUBULAR  FLUID  OSMOLARITY  and  DECREASED  
ANHYDRASE   • Prevent  the  reabsorption  of  HCO3-­‐  and  Na+   H2O  REABSORPTION  IN  THE  COLLECTING  DUCT.  
INHIBITORS     • H2O  reabsorption  is  therefore  reduced.    
  Therefore,  they  INCREASE  URINE  VOLUME  (increased  H2O  
    excretion  –  alkaline  urine  due  to  HCO3-­‐)  and  K+,  Na+  and  HCO3-­‐  
Excretion  
e.g.  Acetazolamine  
 
• Clinical  Uses:  
o Renal  Stones  –  Uric  Acid  
o Metabolic  Alkalosis  –  Increased  HCO3-­‐  loss  
o Decreased  intraocular  pressure  
• Unwanted  Effects:  
o K+  Loss  
o Metabolic  Acidosis  
   
DIURETIC   Loop  Diuretics  are  POWERFUL  Diuretics  that  act  on   Effects  of  Loop  Diuretics  (e.g.  Frusemide)    
the  ascending  limb  of  the  loop  of  Henle.    
LOOP  DIURETICS     Large  increase  in  urine  volume  and  Na+,  Cl-­‐  and  K+  loss  (+Ca2+  
INHIBIT  Na+  and  Cl-­‐  Reabsorption  in  the  ascending   and  Mg2+  Loss)  
  limb  by  30%    
  CLINICAL  USES:  
e.g.  Frusemide  
Ca2+  and  Mg2+  Loss  -­‐  Loss  of  K+  Recycling   • OEDEMA  –  Heart  failure,  pulmonary,  renal,  hepatic  
(Furosemide)  
  and  cerebral  
Therefore,  they  DECREASE  THE  OSMOLARITY  of  the   • MODERATE  HYPERTENSION  –  Piretanide  
medullary  interstitium.   • HYPERCALCAEMIA  
  • HYPERKALAEMIA  
Increase  the  delivery  of  Na+  to  the  distal  tubule,  so    
increased  K+  loss  (due  to  increased  Na+/K+  Exchange)   UNWANTED  EFFECTS:  
  • HYPERVOLAEMIA  
Increased  tubular  fluid  osmolarity  (and  so  decrease   • HYPERTENSION  
the  osmolarity  of  the  medullary  interstitium),  which   • K+  Loss  (Ca2+/Mg2+),  Metabolic  Alkalosis  
leads  to  decreased  H2O  reabsorption  at  the    
collecting  duct.    
 
   
DIURETIC   These  are  moderately  powerful  diuretics,  which  act   Clinical  Uses    
on  the  early  distal  tubule.   • Cardiac  Failure  
THIAZIDES     • Hypertension  (Initially  a  decreased  blood  volume  
Inhibit  Na+  and  Cl-­‐  reabsorption  in  the  early  distal   decreases  –  in  the  long  term,  thiazides  cause  
  tubule  (by  about  5-­‐10%)   vasodilation)  
  • Severe  Resistant  Oedema  
e.g.  Bendrofluazide  
So  there  is  an  increased  delivery  of  Na+  to  the  distal   • Idiopathic  Hypercalciuria  (Stone  Formation)  
(Bendroflymethiazide  
tubules,  so  an  increased  K+  Loss  (as  there  is  an   • Nephrogenic  Diabetes  Insipidus  (Paradoxical)  
increased  Na+/K+  exchange)   bhj  
  Unwanted  Effects  
There  is  an  increased  Mg2+  loss  and  increased  Ca2+   • K+  loss  
reabsorption  (REDUCED  LOSS  OF  CALCIUM)   • Metabolic  Alkalosis  
Increased  tubular  fluid  osmolarity,  so  decreased   • Diabetes  Mellitus  (Inhibits  insulin  secretion)  
water  reabsorption  in  the  collecting  duct.    
 
Thiazide  diuretics  lead  to    moderate  increase  in  urine  
volume  and  Na+,  Cl-­‐  and  K+  loss  (Mg2+  loss)    
 
Classes  of  K+  Sparing  Drugs   Potassium  Sparing  Diuretics  e.g.  Amiloride,  Spironolactone  
DIURETIC   • ALDOSTERONE  RECEPTOR  ANTAGONISTS   • INHIBIT  Na+  REABSORPTION  –  (and  therefore  the    
o e.g.  SPIRONOLACTONE   secretion  of  K+)  in  the  early  distal  tubule  (by  5%)  
POTASSIUM  SPARING      
DIURETICS   • INHIBITORS  of  ALDOSTERONE-­‐SENSITIVE  Na+   • INCREASED  TUBULAR  FLUID  OSMOLARITY  –  so  
CHANNELS   decreased  H2O  reabsorption  in  the  collecting  duct  
e.g.  Amiloride,   o e.g.  amiloride    
Spironolactone     • DECREASED  REABSORPTION  OF  Na+  TO  DISTAL  
TUBULE  –  so  increased  H+  retention  (Na+  /  H+  
exchanger)  
• INCREASED  URIC  ACID  LOSS  
 
SMALL  INCREASE  IN  URINE  VOLUME  and  Na+  Loss  
 
Clinical  Uses:  
• For  use  with  K+  losing  diuretics  –  use  Amiloride  
• Primary  and  Secondary  Hyperaldosteronism  –  use  
spironolactone  
 
Unwanted  Effects:  
• Hyperkalaemia  
• Metabolic  Acidosis  
• Spironolactone  –  Gynaecomastia,  Menstrual  
Disorders,  Testicular  Atrophy  
 
 

TRIPLE  THERAPY:  
ANTI-­‐ULCER  DRUGS  –  ANTIBIOTICS   ‘Triple  Therapy’  is  currently  the  best  practice  in  treating  
 
peptic  ulcer  disease  
  • A  single  antibiotic  is  not  sufficiently  effective  –  partly  
ANTIBIOTICS  +  PPI   Triple  Therapy  -­‐  Example  1   due  to  the  development  of  resistance.  
  •  
(FOR  PEPTIC  ULCERS  –   • METRONIDAZOLE  (active  against  anaerobic   THREE  PROBLEMS  WITH  TRIPLE  THERAPY:  
AGAINST  H.  PYLORI)   bacteria  and  protozoa)  or  AMOXYCILLIN   • COMPLIANCE  
(broad  spectrum  antibiotic)  –  depending  on   • DEVELOPMENT  OF  RESISTANCE  (Vaccinations  may  
pattern  of  local  resistance.   soon  be  available)  
  • ADVERSE  RESPONSE  TO  ALCOHOL  –  especially  with  
• CLARITHROMYCIN  antibiotics  with  a   metronidazole  (interferes  with  alcohol  metabolism)    
macrolide  structure  –  inhibits  translocation    
of  bacterial  tRNA.  
 
• PROTON  PUMP  INHIBITOR  (PPI)  improves  
antibiotic  efficiency  possibly  by  increasing  
gastric  pH  which  improves  stability  and  
absorption.  
 
Triple  Therapy  –  Example  2  
• H2  Receptor  Antagonist  
• Clarithromycin  
• Bismuth  
 

PROTEIN  PUMP  
ANTI-­‐ULCER  DRUGS  –  INHIBITORS   Mechanism  of  Action:  
 
• PPIs  are  irreversible  inhibitors  of  the  H+/K+  ATPase  
INHIBITORS   OF  GASTRIC  ACID  SECRETION   • Inactive  at  neutral  pH  
  • As  it  is  a  weak  base,  it  accumulates  in  the  cannaliculi  
e.g.  OMEPRAZOLE   TREATMENT  OF  GASTRIC  ULCERS   of  parietal  cells:  this  concentrates  its  action  there  and  
  prolongs  its  duration  of  action  (2-­‐3  days)  and  
  Inhibit  basal  and  stimulated  gastric  acid  secretion   minimises  its  effects  on  ion  pumps  elsewhere  in  the  
from  the  parietal  cell  by  >90%   body.  
 
Uses  
• Component  of  Triple  Therapy  
• Peptic  Ulcers  resistant  to  H2  Antagonists  
• Reflux  Oesophagitis  
 
Pharmacokinetics  
• Orally  active  
• Administered  as  enteric-­‐coated  slow-­‐release  
formulations  
 
Unwanted  Effects  –  Rare  
 
HISTAMINE  TYPE  2  
ANTI-­‐ULCER  DRUGS  –  INHIBITORS    
 
• Orally  administered  
(H2)  RECEPTOR   OF  GASTRIC  ACID  SECRETION   • Well  absorbed  
ANTAGONISTS     • Unwanted  effects  are  rare  
Inhibit  gastric  acid  secretion  by  approximately  60%    
e.g.  CIMETIDINE,   and  are  less  effective  at  healing  ulcers  than  PPIs   Relapses  likely  after  withdrawal  of  treatment  
RANITIDINE    

CYTOPROTECTIVE  
ANTI-­‐ULCER  DRUGS  –   Mechanism  of  Action:  
 
• Acquires  a  strong  negative  charge  in  an  acid  
DRUGS   CYTOPROTECTIVE  DRUGS   environment  

e.g.  SULCRAFATE  
  • Binds  to  positively  charged  groups  in  large  molecules  
These  drugs  ENHANCE  MUCOSAL  PROTECTION   (proteins,  glycoproteins)  resulting  in  gel-­‐like  
MECHANISMS  and/or  BUILD  A  PHYSICAL  BARRIER   complexes.  
over  the  ulcer.   • These  coat  and  protect  the  ulcer,  limit  H+  diffusion  
and  pepsin  degradation  of  mucus.  
  • Increases  PG,  mucus  and  HCO3-­‐  secretion  and  reduces  
This  is  a  polymer  containing  aluminium  hydroxide   the  number  of  H.  Pylori  
and  sucrose  octa-­‐sulphate.    
  Side  Effects:  
• Most  of  orally  administered  drug  remains  in  the  
gastrointestinal  tract  
• May  cause  constipation  
• Reduces  absorption  of  some  other  drugs  (e.g.  
antibiotics  and  digoxin)  
 

CYTOPROTECTIVE  
ANTI-­‐ULCER  DRUGS  –    
 
Acts  like  sulcrafate  
DRUGS   CYTOPROTECTIVE  DRUGS    

e.g.  BISMUTH  
  Used  in  triple  therapy  (resistant  cases    
These  drugs  ENHANCE  MUCOSAL  PROTECTION  
CHELATE  
MECHANISMS  and/or  BUILD  A  PHYSICAL  BARRIER  
over  the  ulcer.  
CYTOPROTECTIVE  
ANTI-­‐ULCER  DRUGS  –   Misoprostal  may  be  co-­‐prescribed  with  oral  NSAIDs  (non-­‐
 
steroidal  anti-­‐inflammatory  drugs),  when  used  chronically:  
DRUGS   CYTOPROTECTIVE  DRUGS    

e.g.  MISOPROSTAL  
  • NSAIDs  block  the  COX  enzyme  required  for  PG  
(A  stable  prostaglandin  analogue)   synthesis  from  arachidonic  acid  
  • Therefore,  there  is  a  REDUCTION  in  the  natural  
Mimics  the  action  of  locally  produced  prostaglandins   factors  that  inhibit  gastric  secretion,  and  stimulate  
to  maintain  the  gastroduodenal  mucosal  barrier.   mucus  and  HCO3-­‐  production  
 
  Unwanted  Effects:  
• Diarrhoea,  Abdominal  Cramps,  Uterine  Contractions  
• DO  NOT  USE  IN  PREGNANCY  
 

ANTACIDS  
ANTI-­‐ULCER  DRUGS  –  ANTACIDS    
 
• Primarily  used  for  NON-­‐ULCER  DYSPEPSIA  
  • May  be  effective  in  reducing  duodenal  ulcer  
• Mainly  salts  of  Al3+  and  Mg2+   recurrence  rates  
• Neutralises  acid,  raises  gastric  pH,  reduces    
pepsin  activity  
 

 
  Bile  Acid  Sequestrants:  
LIPID  LOWERING   • Decrease  LDL  to  some  extent    
DRUGS  -­‐     • However,  they  increase  hepatic  synthesis  of  LDL  
• Therefore,  they  are  not  very  effective  
Bile  Acid  Sequestrants    
 
 
 
   
LIPID  LOWERING   Mechanism  of  Action:   Cholesterol  Synthesis  Pathway:    
DRUGS  -­‐       • Geranyl  pyrophosphate  and  Farnesyl  Pyrophosphate  –  
Statins  block  the  HMG-­‐CoA  Reductase  Enzyme   these  are  lipids  involved  in  the  modification  of  
STATINS     proteins  (e.g.  rho  and  ras)  
• These  lipids  can  only  by  synthesised  within  the  cell.  
(e.g.  Rosuvastatin,  
• Statins  inhibit  the  production  of  these  lipids.  
Atorvastatin,  
 
Simvastatin,  
Mechanism  by  which  Statins  reduce  elevated  LDL:  
Pravastatin,  
• Inhibition  of  HMG-­‐CoA  Reductase  and  hence,  
Fluvastatin)    
decrease  cholesterol  synthesis  within  hepatocytes.  
• Increase  the  expression  of  LDL  Receptors  on  
hepatocytes:  
o This  means  more  LDL  Receptors  bind  to,  and  
internalise  more  circulating  LDLs  
 
Summary  of  Statin  Effects:  
• All  statins  are  similar:  they  bring  about  the  same  
reduction  in  risk.  
• There  is  a  strong  correlation  between  LDL  level  and  
risk.  
• Relatively  safe.  
• Improves  survival  ad  reduces  risk  in  everyone.  
• Anti-­‐inflammatory  actions  (same  molecule  
mechanism  of  action,  but  acts  on  different  cell  types)  
• Focus  on  patients  with  high  LDL  and  high  CRP  (these  
are  potentially  the  highest  risk  patients)  
 
 
   
   
LIPID  LOWERING   Mechanism  of  Action:     • PPAR  –  Perioxisome  Proliferator  Activated  Receptors    
DRUGS  -­‐       • Thiazolidinediones  (glitazones)  are  PPAR  gamma  
Activate  PPAR  Alpha  receptors  à  Decrease  in  fatty   activators  (and  are  used  in  diabetes)  
FIBRATES   acids  and  triglycerides   • Anti-­‐inflammatory  action  
• Shown  to  reduce  mortality  (by  1  trial)  
• 2nd  choice  to  statins.  
 
   
LIPID  LOWERING   Decreases  cholesterol,  LDL  and  triglyceride  levels  and   • Other  effects:      
DRUGS  –     increases  HDL  levels   o Anti-­‐Coagulant  
  o Anti-­‐Platelet  
NICOTINIC  ACID   o Anti-­‐Inflammatory  
• Shown  to  reduce  risk  in  trials  
• Side  effects  include:  
o Flushing  and  Hepatic  Effects  
 
   
LIPID  LOWERING   Mechanism  of  action:  inhibits  cholesterol  absorption   • Must  be  activated  by  the  liver,  secreted  into  the  bile    
DRUGS  –       and  reabsorbed.  
• Reduces  cholesterol  levels  by  15-­‐20%  
EZETIMIBE   • May  be  used  in  combination  with  statins  
e.g.  Atorvastatin  +  Ezetimibe  à  Extra  12%  reduction  
in  cholesterol  levels  
• But  –  
o No  effect  on  carotid-­‐intima  media  thickness  
o Unknown  effects  on  events/survival  
 
Not  shown  to  work  very  well  in  practice  
 
   
LIPID  LOWERING   • CETP  inhibitors  increase  HDL  levels  (since    
DRUGS  –     they  cannot  be  broken  down)  
  • However  –  they  increase  blood  pressure  
Cholesterol  Ester   (therefore  increase  mortality,  and  hence  are  
Transfer  Protein   no  longer  used)  
(CETP)  and  reverse    
cholesterol  transport  

 
 
   
ANTICOAGULANTS:   Mechanism  of  Action:   Pharmacokinetics:  
  PREVENTS  activation  of  VITAMIN  K   Binds  strongly  to  plasma  proteins  (99%  bound  to  albumin)    
Warfarin     Results  in  a  small  volume  of  distribution.  
Vitamin  K  required  as  a  cofactor  in  synthesis/post-­‐  
translational  modification  of  Factors  VII,  IX,  X  and  II   Metabolised  by  HEPATIC  MIXED  FUNTION  CYTOCHROME  
(Thrombin)     P450  
   
Administration:   Anticoagulant  activity  is  monitored  by  the  International  
Oral,  absorbed  quickly  from  the  GI  tract.     Normalised  Ratio  (a  measure  of  prothrombin  time)  
Peak  blood  concentration  within  1  hour    
  Adverse  Effects:  
Pharmacological  effects  are  delayed  12-­‐16h,  peak   Haemorrhage  (especially  into  the  brain  or  bowel)  
after  48h  and  lasts  4-­‐5  days.     Teratogenicity  (NOT  GIVEN  in  pregnant  mothers)  
This  is  because  of  the  slow  turnover  of  clotting    
factors.     Reversal  of  Effects:  
  • Low  doses  of  Vitamin  K  
• Fresh  Frozen  Plasma  (FFP)  or  prothrombin  complex  
concentrate  can  be  infused  if  a  rapid  reversal  of  warfarin  
effect  is  needed.  
 
Drug  Interactions:  
 
With  drugs  that  inhibit  cytochrome  p450  
Antibacterial  agents  e.g.  Erythromycin  
Antifungal  agents  e.g.  Fluconazole  
   
With  drugs  that  induce  cytochrome  p450  
Anticonvulsants  e.g.  Phenobarbital  
 
With  drugs  that  inhibit  platelet  function  
E.g.  Aspirin  
 
With  drugs  that  displace  warfarin  from  plasma  proteins  
(binding  globulins)  
E.g.  Aspirin  
 
ANTICOAGULANTS:   Mechanism  of  Action:   Administration  
    • Poorly  absorbed  after  oral  administration    
Heparin  and  Low   Activates  Anti-­‐Thrombin  III   • Therefore,  given  either:  
Molecular  Weight   Anti-­‐Thrombin  III  then  INHIBITS  FACTOR  Xa  and   o SUBCUTANEOUSLY  
Heparin   THROMBIN  (FIIa)  by  binding  to  the  active  serine   o INTRAVENOUSLY  
sites.    
  Pharmacokinetics  
LMWH  has  a  similar  effect  on  Xa,  but  less  of  an  effect   • Immediate  onset  when  given  Intravenously  (I.V.)  
on  thrombin.   Delayed  by  about  1  hour  if  given  subcutaneously  
  (LMWH)  
• Short  Half-­‐Life  
• Heparin  exhibits  saturation  kinetics  (apparent  1/2  
life  increases  with  increasing  dose).  
• Anticoagulant  activity  is  measured.  
 
• LMWH  has  a  longer  half-­‐life,  exhibits  1st  Order  
kinetics.  Its  activity  does  not  require  monitoring.  
 
Adverse  Effects  
• Bleeding  
• Thrombocytopenia  
• Osteoporosis  (associated  with  long  term  therapy  
over  3  months)  
• Hypersensitivity  
o Chills,  fever,  urticaria,  possibly  anaphylaxis  
 
Reversal  of  Effects  
• Stop  I.V.  Heparin  and  LMWH.  
• Give  IV  PROTAMINE  
o Protamine  binds  to  heparin  to  produce  an  
inactive  complex.  
 
ANTIPLATELET  DRUGS:   Mechanism  of  Action:   Administration:  
  Irreversibly  inhibits  COX-­‐1  Enzyme   • Oral    
Aspirin      
Inhibits  the  production  of  TXA2  (Thromboxane  A2)  in   Pharmacokinetics:  
platelets     • Highly  plasma  protein  bound  
   
However,  production  of  PGI2  by  endothelium  is  not   Adverse  Effects:  
severely  affected   • GI  Sensitivity  
   
ANTIPLATELET  DRUGS:   Mechanism  of  Action:   Administration:  
    • Oral    
Clopidogrel   A  pro-­‐drug  which  inhibits  fibrinogen  binding  to    
glycoprotein  IIb/IIIa  Receptors.   Pharmacokinetics:  
• Peak  plasma  concentration  4hours  after  a  single  
  dose  
• Inhibitory  effect  on  platelet  not  seen  until  4  days  
of  regular  dosing.  
 
Adverse  Effects:  
• Bleeding  –  GI  Haemorrhage,  Diarrhoea,  Rash  
• In  some  patients,  neutropenia  
 
ANTIPLATELET  DRUGS:   Mechanism  of  Action:   Administration:  
    • Intravenously  (I.V.)    
Abciximab   Antagonist  of  the  Glycoprotein  IIb/IIIa  Receptor.   Pharmacokinetics:  
  • Binds  rapidly  to  platelets.  
This  is  a  hybrid  murine/human  MONOCLONAL   • Cleared  with  platelets.  
ANTIBODY  which  is  licensed  for  use  in  ACUTE   • Antiplatelet  effect  persists  for  24-­‐48  hours.  
CORONARY  SYNDROMES.   Adverse  Effects:  
  • Bleeding  
Used  in  combination  with  heparin  and  aspirin  to   • May  potentially  be  IMMUNOGENIC  
prevent  ischaemia  is  patients  with  unstable  angina.    
 
FIBRINOLYTIC   Mechanism  of  Action:   Administration:  
(THROMBOLYTIC)     • Intravenous  (I.V.)    
DRUGS   Non-­‐Enzyme  Protein   • 30-­‐60  minutes  infusion.  
     
Streptokinase   Derived  from  culture  of  β-­‐Haemolytic  Streptococci   Pharmacokinetics:  
  • Rapidly  cleared  
Binds  to  plasminogen,  causing  a  conformational   • t1/2  12-­‐18  minutes  
change  –  this  exposes  the  active  site  and  causes    
plasmin  activity.   Adverse  Effects:  
  • Bleeding  
Activated  plasmin  degrades  fibrin.   • May  potentially  be  antigenic  
   
FIBRINOLYTIC   Mechanism  of  Action:   Administration:  
(THROMBOLYTIC)   Is  recombinant  tPA  (Tissue  Plasminogen  Activator)   • Intravenous  (I.V.)  30  minute  infusion    
DRUGS   It  works  better  on  plasminogen  bound  to  fibrin  than    
  on  soluble  plasminogen  in  the  plasma  –  and  is   Pharmacokinetics:  
Alteplase   therefore  said  to  be  CLOT  SENSITIVE.   • Rapidly  cleared  
    • t1/2  12-­‐18  minutes  
It  activates  plasmin  that  then  degrades  fibrin  and    
dissolves  the  clot.   Adverse  Effects:  
  • Bleeding  
 
 
Ibuprofen   Non-­‐Steroidal  Anti-­‐Inflammatory  Drugs   • Typical  non-­‐selective  NSAIDs  
Indomethacin   • Inhibit    cyclo-­‐oxygenase  REVERSIBLY    
  • Inhibit  both  COX-­‐1  and  COX2  
(NSAID)   • Have  anti-­‐inflammatory,  analgesic  and  
antipyretic  actions  

Aspirin   Non-­‐Steroidal  Anti-­‐Inflammatory  Drugs    


    • Serious  side-­‐effects  at  therapeutic  doses    
(NSAID)   • Binds  more  avidly  to  COX-­‐1  than  COX-­‐2   • As  well  as  usual  NSAID  actions,  they  also  reduce  
• Binds  irreversibly  to  COX  enzymes    -­‐  A   platelet  aggregation  
unique  property  among  the  NSAIDs    
o It  acetylates  an  amino  acid  in  the   Aspirin  –  Mechanism  of  Action  
active  site  of  COX  (making  its  actions   • Aspirin  inhibits  TXA2  Production  by  platelets  and  
long  lasting)   prostacyclin  production  by  endothelial  cells  
o Its  actions  can  only  be  reversed  by   (NOTE  –  Prsostacyclin  is  DIFFERENT  to  
the  synthesis  of  new  COX  (as  part  of   Prostaglandin)  
a  continuous  production)    
  • However,  as  platelets  have  no  nucleus,  COX1  is  
not  re-­‐synthesised  and  therefore  TXA2  synthesis  
stops  until  a  new  batch  of  platelets  are  produced.  
 
• As  endothelial  cells  have  nuclei,  they  can  
therefore  replenish  COX1  and  prostacyclin  
synthesis  continues.  
 
Covalent  binding  of  aspirin  confers  its  anti-­‐platelet  property  
which  is  unique  among  NSAIDs.  
 
    Anti-­‐Platelet  Actions  of  Aspirin  is  due  to:  
• Very  high  degree  of  COX-­‐1  inhibition  which    
effectively  suppresses  TXA2  production  by  
platelets  
• Covalent  binding  which  permanently  inhibits  
platelet  COX-­‐1  
• Relatively  low  capacity  to  inhibit  COX-­‐2  
 
Major  Side-­‐Effects  of  Aspirin  seen  at  therapeutic  doses  are:  
• Gastric  irritation  and  ulceration  
• Bronchospasm  in  sensitive  asthmatics  
• Prolonged  bleeding  times  
• Nephrotoxicity  
 
Side  effects  are  more  likely  with  aspirin  than  other  NSAIDs  
because  it  inhibits  COX  covalently.  
 
Celecoxib   Non-­‐Steroidal  Anti-­‐Inflammatory  Drugs   • Selectively  inhibits  COX-­‐2  
    • Less  effective  on  COX-­‐1  mediated  processes  than    
(NSAID)   conventional  NSAIDs  such  as  ibuprofen  and  
indomethacin  
• Fewer  ulcers  (c.f.  non-­‐selective  NSAIDS)  
• But  not  all  COX2  activity  is  pathological  
• COX2  regulates  –  ovulation,  parturition,  renal  
blood  flow,  blood  pressure  (therefore  COX2  
inhibition  is  not  always  desirable)  
 
COX-­‐2  Inhibitors:  
• Have  a  good  GIT  safety  profile  
• Are  well  tolerated  (but  not  recommended)  for  
patients  with  asthma  
• BUT  have  unwanted  CVS  effects  
o Increased  risk  of  myocardial  infarction  in  5  out  
of  8  trials  when  compared  with  non-­‐selective  
NSAID  
 
Cardiovascular  Effects  of  COX-­‐2  Inhibitors:  
• May  selectively  inhibit  PGI2  production  and  spare  
TXA2  production  leading  to  more  aggregation  
• It  is  not  the  only  mechanism  –  since  MIs  still  occur  
even  in  patients  taking  aspirin.  
• Non-­‐selective  NSAIDS  also  inhibit  COX-­‐2  
 
• There  is  increasing  evidence  that  COX-­‐2  inhibitors  
pose  higher  risk  of  cardiovascular  disease  than  
conventional  NSAIDS  even  though  mechanism  is  
unclear.  
• Debate  over  the  safety  of  the  COX-­‐2  inhibitors  is  
continuing.  
 
 
Paracetamol   • Is  a  good  analgesic  for  mild-­‐to-­‐moderate   Mechanism  of  Action  
  pain   • The  mechanism  of  action  of  paracetamol  is    
Analgesic,  Antipyretic   • Has  ANTI-­‐PYRETIC  ACTION   unclear  –  several  mechanisms  have  been  
• However,  it  does  NOT  HAVE  ANY  ANTI-­‐ postulated.  
INFLAMMATORY  EFFECT    
• Therefore,  it  is  not  an  NSAID   • The  most  likely  mechanism  in  man  is:  
  Paracetamol  acts    to  inhibit  the  peroxidation  of  
PGG2  into  PGH2  (also  catalysed  by  COX)  
 
Side  Effects  of  Paracetamol  
• Paracetamol  is  generally  a  very  safe  drug  
• However,  in  overdose  it  may  cause  IRREVERSIBLE  
LIVER  FAILURE  
o A  reactive,  but  minor  metabolite  (NAPQI)  is  
normally  safely  conjugated  with  glutathione  
o If  glutathione  is  depleted,  the  metabolite  
oxidises  thiol  groups  of  key  hepatic  enzymes  
and  causes  cell  death  
 
Antidote  for  Paracetamol  Poisoning  
• Add  compound  with  –SH  Groups  
• Usually  intravenous  acetylcysteine  
• Occasionally  oral  methionine  
• Far  fewer  successful  suicides  with  paracetamol  
since  purchase  has  been  limited  
 
 
   
Morphine  (OPIATE)   Opiate,  as  it  is  a  direct  derivative  of  the  Poppy  resin.   Administration:  
Oral  –    
40-­‐50%  absorbed  into  the  bloodstream  
Slow  acting  –  may  take  up  to  30  minutes  to  have  an  effect  
 
Can  be  administered  i.v.    
 
Distribution:  
Limited  access  to  the  brain  
 
Largely  ionised  at  physiological  pH  (so  becomes  polar),  and  
diffuses  across  the  lipid  membranes  slowly  
 
A  large  proportion  of  the  administered  dose  does  not  access  
the  brain.  
 
Metabolism:  
Rapid  hepatic  metabolism  (GLUCORONIDATION  at  the  6’  
position)  
 
Morphine  is  converted  to  morphine-­‐6-­‐glucuronide,  but  this  
compound  ‘gives  a  handle’  for  the  kidney  to  easily  clear/filter.  
 
Morphine-­‐6-­‐glucuronide,  however,  is  more  potent  (so  
increased  effect)  and  is  subject  to  ENTEROHEPATIC  
CIRCULATION.  
 
Therefore  morphine-­‐6-­‐glucuronidde  may  be  secreted  into  the  
bile,  and  morphine  may  be  regenerated  in  the  GI  Tract  and  
reabsorbed.  
 
Excretion:  
Urine  
 
Codeine  (Opiate)     Codeine  Pharmacokinetics  
• Structurally  similar  to  morphine  (except  for  the    
methyl  group  in  the  3’  position)  
• Far  less  potent  than  morphine  
• Oral  codeine  is  about  5-­‐10%  the  strength  of  i.v.  
morphine  
 

Heroin  (Semi-­‐   Heroin  


Synthetic  Opioid)   Administration    
• Oral  
 
Distribution  
• Very  LIPID  SOLUBLE    
• Enters  the  brain  quicker  than  morphine  
• Converted  to  morphine  in  cells  
 
Metabolism  
• Metabolised  by  plasma  esterases  
• Heroin  is  broken  down  more  rapidly  than  morphine  
 
Short-­‐acting  (high  abuse  potential  because  the  euphoric  effect  
wears  off  quickly)  
 
Fentanyl  (Opioid)   Opioid  –  Synthetic  compound  that  does  not  generally   Administration  
resemble  morphine  in  structure.   Buccal  (lollipop)  or  Intradermal  (patch)    
 
Absorption  -­‐  50-­‐100%  Absorption  
 
Distribution  
Very  LIPID  SOLUBLE  and  is  more  potent  orally  (a  lot  is  
reabsorbed  the  mouth  and  intranasally,  so  more  gets  into  the  
system).  
 
Enters  the  bloodstream  via  the  mucus  membranes/skin  
 
Metabolism  -­‐  Hepatic  metabolism  (oxidation)  
 
Excretion  -­‐  Excreted  in  the  Urine  
Methadone  (Opioid)   Opioid  –  Synthetic  compound  that  does  not  generally   • Administration  
resemble  morphine  in  structure.   o Oral    
 
• Distribution  
o The  most  lipid  soluble  opioid,  therefore  
dissipates  into  fat  very  quickly.  
o Methadone  is  commonly  used  as  a  
morphine/heroin  substitute  (e.g.  in  weaning  
off  addicts)  because  of  the  fact  that  it  
distributes  into  the  fat  very  quickly.  
 
o So  the  half-­‐life  of  methadone  is  MUCH  
LONGER  (150h),  and  can  be  given  to  
morphine  addicts  as  it  is  released  slowly  
(hence  a  low  constant  background  level  of  
opiates)  
 
o Therefore  long  acting,  with  a  prolonged  
euphoric  effect.  
 
Naloxone     Opioid  Receptor  Antagonist   • Naloxone  (Opioid  Antagonist)  
  • i.v.  administration    
(Opioid  Receptor   • Short  acting  
Antagonist)   • Used  in  opioid  overdose  

 
   
Prednisolone,   Glucocorticoids  –  used  in  the  TREATMENT  OF   The  activation  of  glucocorticoid  receptors  can  lead  to:  
Fluticasone,   INFLAMMATORY  BOWEL  DISEASE  (CD  &  IBD)   • Increase  the  expression  of  anti-­‐inflammatory  genes    
Budesonide     (GCR  acting  as  a  positive  transcription  factor)    
  Derived  from  cortisol   • Decrease  the  expression  of  pro-­‐inflammatory  genes  
(Glucocorticoids)     (GCR  acting  as  a  negative  transcription  factor)  
   
  Glucocorticoids  as  anti-­‐inflammatory  
  Reduce  influx  and  activation  of  pro-­‐inflammatory  cells  
  • Reduce  expression  of  adhesion  molecules  on  
  endothelial  cells  and  leukocytes  
  • Reduce  synthesis  of  some  chemokines  
   
  Reduced  production  of  inflammatory  mediators  (e.g.  IL-­‐2,  IL-­‐
  4,  IFN-­‐γ)  that  normally  cause  vasodilation,  fluid  exudation  
  (swelling),  further  inflammatory  cell  recruitment  and  tissue  
  degradation.  
   
  This  essentially  is  a  reduced  synthesis  of  the  following  
  mediators:  
  • Some  cytokines  and  cytokine  receptors  (e.g.  IL-­‐1  
  and  TNF-­‐α)  
  • Proteolytic  enzymes  (e.g.  elastase)  
  • Enzymes  that  catalyse  mediator  synthesis  (e.g.  
  cyclooxygenase)  
Unwanted  Effects  of  Glucocorticoids:   • Eicosanoids  (e.g.  prostaglandins  and  leukotrienes)  
• Osteoporosis   • Nitric  Oxide  
• Increased  risk  of  gastric  ulceration    
• Suppression  of  the  HPA  (Hypothalamo-­‐ Glucocorticoids  as  immunosuppressives  
Pituitary-­‐Adrenal)  Axis   Glucocorticoids  are  potent  immunosuppressives  which  cause  :  
• Type  II  Diabetes   • Reduction  in  antigen  presentation  
• Hypertension   • Reduction  in  production  of  certain  mediators  (e.g.  IL-­‐
• Susceptibility  to  infection   2,  IL-­‐4  and  IFN-­‐γ)  
• Skin  thinning,  bruising  and  slow  wound   • Reduction  in  cell  proliferation  and  clonal  expansion  
healing    
• Muscle  wasting  and  buffalo  hump  (c.f.   Glucocorticoids  virtually  suppress  all  types  of  inflammation.    
Cushing’s)    
   
 
   
Mesealazine  (5-­‐ASA)   These  are  all  types  of  aminosalicylates  –  ONLY   Mechanisms  of  Anti-­‐Inflammatory  Actions:  
Olsalazine  (2x  5-­‐ASA)   EFFECTIVE  IN  ULCERATIVE  COLITIS.   • Reduce    synthesis  of  eicosanoids    
    • Reduce  free  radical  levels  
(Sulfalazine)   Anti-­‐inflammatory,  but  NOT  IMMUNOSUPPRESSIVE   • Reduce  inflammatory  cytokine  production  
    • Reduce  leukocyte  infiltration  
(Aminosalicylates)   They  are  useful  in  the  treatment  of  active  ulcerative    
colitis  and  for  maintenance  of  remission     Metabolised  by:   Site  of  
  Absorption:  
However,  they  are  ineffective  in  Crohn’s  Disease  
  Mesalazine   (Not  absorbed  as   Small  bowel  and  
  it  is  in  the  most   colon  
basic  form  –  c.f.  
sulfasalazine)  

Olsalazine   Colonic  flora   Colon  

(Sulfasalazine)   Colonic  Flora,   Colon  


Liver  
 
 
Azothioprine      
Immunosuppressive  Agent   Mechanism  of  Immunosuppression    
  • Azothioprine  is  a  PRODRUG  activated  in  vivo  by  gut  
Effective  in  BOTH  CROHN’s  DISEASE  and   flora  to  6-­‐MERCAPTOPURINE  
ULCERATIVE  COLITIS    
  • This  interferes  with  purine  biosynthesis  
Can  be  used  to  induce  remission  in  Crohn’s  Disease   • Interferes  with  DNA  SYNTHESIS  and  CELL  
(Treatment  >17  Weeks)   REPLICATION  
   
May  enable  reduction  of  glucocorticoid  dose  or   • It  impairs:  
postponement  of  colostomy.   o Cell-­‐  and  Antibody-­‐  mediated  immune  
  responses  
Useful  for  maintaining  remission  in  Crohn’s  Disease   o Lymphocyte  proliferation  
and  some  patients  with  Ulcerative  Colitis.   o Mononuclear  cell  infiltration  
  o Synthesis  of  antibodies  
 
• It  enhances:  
o T-­‐Cell  Apoptosis  
 
Unwanted  Effects  
• Bone  marrow  suppression  
• Metabolised  by  xanthine  oxidase  
 
• Care  must  be  taken  to  check  whether  there  is  a  co-­‐
administration  of  drugs  which  INHIBIT  XANTHINE  
OXIDASE  e.g.  allopurinol  which  can  cause  a  build-­‐up  
of  6-­‐Mercaptopurine  leading  to  blood  disorders.  
 
Anti-­‐TNFα   Used  successfully  in  the  treatment  of  Crohn’s  Disease   Mechanism  of  Action  of  Infliximab  (Anti-­‐TNFα)  
  Some  evidence  of  effectiveness  in  Ulcerative  Colitis   • Indicates  that  TNFα  plays  an  important  role  in  the    
INFLIXMAB  (i.v.)       pathogenesis  of  IBD  
Potentially  curative,  rather  than  just  simply  palliative   • Anti-­‐TNFα  reduces  activation  of  TNDα  receptors  in  the  
  gut.  
Successful  in  some  patients  with  refractory  disease   • Production  of  other  cytokines,  infiltration  and  
and  fistulae   activation  of  leukocytes  is  also  reduced.  
   
  • Anti-­‐TNFα  also  binds  to  membrane  associated  TNFα  
• Mediates  complement  activation  and  induces  
cytolysis  of  cells  expressing  TNFα  
• This  promotes  apoptosis  of  activated  T-­‐Cells  
 
Pharmacokinetics  of  Infliximab  (Anti-­‐TNFα)  
• Given  intravenously  
• Very  long  half-­‐life  (9.5  days)  
• Benefits  can  last  for  30  weeks  after  a  single  infusion  
• Most  patients  relapse  after  8-­‐12  weeks  
• Therefore,  it  is  important  to  repeat  infusion  every  8  
weeks.  
 
Adverse  Effects  of  Infliximab  (Anti-­‐TNFα)  
• 4x  to  5x  increase  in  incidence  of  Tuberculosis  and  
other  infections  
• Also  risk  of  reactivating  dormant  TB  
• Increased  risk  of  SEPTICAEMIA  –  therefore,  
contraindications  if  abscesses  are  present  
• Worsening  of  heart  failure  
• Increased  risk  of  demyelinating  disease  
• Increased  risk  of  malignancy  
 
• Can  be  immunogenic  (monoclonal  antibody)  –  
therefore  given  with  azathioprine  
• Should  only  be  used  by  specialists  where  adequate  
resuscitation  facilities  are  available  –  due  to  a  RISK  
OF  ANAPHYLAXIS  
• 2-­‐4%  risk  of  serious  side-­‐effect  
 
Infliximab  Summary  
• In  steroid-­‐dependent  patients  infliximab  +  AZA  
doubles  the  number  of  patients  in  steroid-­‐free  
remission  after  1  year  of  treatment,  but  still  only  by  
40%.  
• This  combination  delays  relapse  
• It  is  most  beneficial  in  patients  who:  
o Have  not  taken  thiopurines  before  
o Are  young  (~26  years)  
o Have  colonic  CD  
 
Adalimumab  (sc.)   TNF  Inhibitor   Binds  to  TNFα  and  prevents  activation  
 

Natalizumab   Antibody  against  alpha-­‐4-­‐integrin   • Antibody  against  alpha-­‐4-­‐integrin  


• Cell  adhesion  molecule    
• Evidence  that  it  induces  remission  in  some  patients  
with  Crohn’s  Disease  
• Generally  well  tolerated  
• Rarely  (1:1000)  encephalopathy  if  taken  in  
combination  with  other  drugs.  
 

 
Muscimol   GABAA  Agonist     • This  is  a  selective  GABAA  Receptor  agonist  (i.e.  does  
  not  stimulate  GABAB  receptors  etc.)   Principles  of  
(I.e.  Post-­‐Synaptic  GABA  Receptor)     GABA-­‐ergic  
Cellular  Mechanism  of  Action:   Transmission  
• Binding  of  GABA  (or  GABAR  Agonist)  to  GABAA  
Receptors  causes  an  activation  of  the  Cl-­‐  channel  on  
the  same  post-­‐synaptic  knob  
• This  leads  to  hyperpolarisation  and  an  INHIBITORY  
POST-­‐SYNAPTIC  POTENTIAL  
• Therefore,  this  causes  an  inhibition  of  firing  by  the  
post-­‐synaptic  knob  
 
Bicuculline   GABAA  Antagonist   • Competitive  Antagonsist  
Principles  of  
GABA-­‐ergic  
Transmission  
Picrotoxin   GABAA  Antagonist   • Non-­‐competitive  
• Binds  to  chloride  channel  itself  and  blocks  the  action   Principles  of  
of  GABA  receptors  non-­‐competitively   GABA-­‐ergic  
• Inhibits  hyperpolarisation   Transmission  
 
Convulsants   GABAA  Antagonists   • Not  used  clinically  
Principles  of  
GABA-­‐ergic  
Transmission  
Benzodiazepines  and   GABAA  Antagonists   • Clinically  used  drugs  (see  other  lecture)  
Barbiturates   Principles  of  
GABA-­‐ergic  
Transmission  
 
Baclofen   GABAB  Agonist   Selective  GABAB  Agonist  
(I.e.  Pre-­‐Synaptic  GABA  Receptor)       Principles  of  
Inhibit  neurotransmitter  release  and  function  in  two  ways:   GABA-­‐ergic  
• AUTORECEPTORS  –  (Negative  Feedback  on  the   Transmission  
presynaptic  knob)  
 
• HETERORECEPTORS  –  (Sit  on  neurones  of  other  
terminals  e.g.  Dopaminergic  Neurones,  and  regulate  
other  neurones,  such  as  by  decreasing  dopamine  
release)  
Cellular  Mechanism  of  Action:  
• G-­‐Protein  linked  
• Decreased  Calcium  Conductance  (i.e.  Influx  of  Ca2+  
decreased)  so  decreased  neurotransmitter  release  
(reduce  vesicular  transport)  
• Increased  K+  conductance,  (so  efflux  of  K+  increased)  
and  therefore  more  hyperpolarisation  
 
• Used  as  a  muscle  relaxant  (effects  in  the  spinal  cord)  
• Used  also  as  a  spasmolytic  drug  (Anti-­‐Spastic  Drug)  
 
 
Phaclofen   GABAB  Antagonist   Antagonises  GABAB  Receptor  
Principles  of  
GABA-­‐ergic  
Transmission  
Saclofen   GABAB  Antagonist   Competitive  Antagonist  (most  commonly  used)  
Principles  of  
GABA-­‐ergic  
Transmission  
 
Flumazenil   Competitive  Benzodiazepine  Antagonist   Acts  on  BDZ  Receptor  on  GABAA  Receptor  Protein    
• Benzodiazepine  binding  to  the  BDZ  receptor  leads  to   Anxiolytics,  
enhanced  GABA  Action   Sedatives  and  
• Enhanced  GABA  Binding  to  the  GABA  receptor  protein   Hypnotics  
(reciprocated)  
 
Anaesthetics   These  are  all  barbiturates  producing  such  effects.   • Unwanted  Side  Effects  of  Barbiturates  
Barbiturates   o They  are  not  the  1st  drug  of  choice  due  to   Anxiolytics,  
e.g.  THIOPENTONE   their  unwanted  effects   Sedatives  and  
o Low  Safety  Margins:   Hypnotics  
Anticonvulsants   § Depress  Respiration  
Barbiturates   § Overdosing  is  lethal  (Treated  with   Anxiolytics,  
e.g.  Phenobarbital   forced  alkaline  diuresis  to  promote   Sedatives  and  
  excretion)   Hypnotics  
Anti-­‐spastic   o Alters  natural  sleep  (Decreased  REM  Sleep)  à  
Barbiturates   hangovers/irritability   Anxiolytics,  
e.g.  Diazepam   o Enzyme  inducers   Sedatives  and  
  o Potentiate  the  effect  of  other  CNS   Hypnotics  
depressants  e.g.  Alcohol  
o Development  of  Tolerance  (both  
pharmacokinetics  and  tissue  tolerance)  
 
o Dependence:  Withdrawal  Syndrome  Causes:  
§ Insomnia  
§ Anxiety  
§ Tremor  
§ Convulsions  
§ Death  
 
Sedatives  /  Hypnotics   Used  for  severe  intractable  insomnia  
e.g.  Amobarbital   Half-­‐Life  20-­‐25  Hours   Anxiolytics,  
  Sedatives  and  
Side  effects  –  see  above.   Hypnotics  
Benzodiazepines   All  benzodiazepines  act  at  GABAA  Receptors   Pharmacokinetics:  
Administration:   Anxiolytics,  
• Well  absorbed  P.O.  (Orally)   Sedatives  and  
• Plasma  concentration  peaks  at  around  1  hour   Hypnotics  
(oxazepam  is  slower)  
• I.v.  administration  for  status  epilepticus  (prolonged  
tonic  clonic  seizure  activity  >30minutes)  
 
Absorption:  
• Well  absorbed  following  oral  administration  
 
Distribution:  
• Binds  to  plasma  proteins  strongly  
• Highly  lipid  soluble  –  wide  distribution  
 
Metabolism:  
• Extensive  hepatic  metabolism  (glucoronidation)  
 
Excretion:  
• Excreted  in  the  urine  as  glucoronide  conjugates  
 
Duration  of  action:  
• Varies  greatly  
• Short  Acting  
• Long  Acting  (due  to  slow  metabolism  and/or  active  
metabolites)  
Anxiolytics     Advantages  of  Benzodiazepines:  
  • Wide  margin  of  safety:   Anxiolytics,  
e.g.  Diazepam,     o Overdose  leads  to  prolonged  sleep  which  is   Sedatives  and  
Chloridazepoxide   rousable   Hypnotics  
(Librium),  Nitrazepam,   o No  respiratory  depression    
Oxazepam   o Flumazenil  
  • Mild  effect  on  REM  Sleep  
Sedatives/Hypnotics     • Do  not  induce  liver  enzymes  
e.g.  Temazepam,     Anxiolytics,  
Oxazepam,   Unwanted  Effects  of  Benzodiazepines   Sedatives  and  
Lorazepam,   • Sedation,  Confusion,  Ataxia  (Impaired  manual  skills)   Hypnotics  
Nitrazepam   • Potentiate  the  effects  of  other  CNS  Depressants  
(Alcohols,  BARBs)  
• Tolerance  (Less  than  BARBs,  ‘tissue’  tolerance  only,  so  
no  pharmacokinetic  tolerance)  
• Dependence:  
o Withdrawal  Syndrome  (similar  to  
barbiturates,  but  less  intense)  
o Withdraw  slowly  
• Free  plasma  concentrations  are  increased  by  certain  
drugs  e.g.  aspirin  and  heparin  
 
Other  Sedatives  /     • Liver  à  Trichloroethanol  
Hypnotics   • Mechanism  of  action  –  unknown   Anxiolytics,  
  • Wide  margin  of  safety  (safe  for  use  in  children  and  the   Sedatives  and  
Chloral  Hydrate   elderly)   Hypnotics  
 
Other  Anxiolytics:     • Improves  physical  symptoms  
Propranolol   Tachycardia  –  β1   Anxiolytics,  
Tremor  –  β2   Sedatives  and  
  Hypnotics  
• Commonly  used  to  ‘cure’  stage  fright  
 
Other  Anxiolytics:     • 5HT1A  Agonist  
Buspirone   • Slow  Onset  of  Action  (Days/Weeks)   Anxiolytics,  
• Few  Side-­‐Effects   Sedatives  and  
  Hypnotics  
 
   
L-­‐DOPA   Dopamine  Replacement  Therapy   L-­‐DOPA  Clinical  Uses:  
    • Hypokinesia,  rigidity  &  tremor   Dopaminergic  
(SINAMET,  MADOPAR   DOPA  is  the  precursor  to  dopamine,  and  is  converted   • Start  with  low  doses  of  the  drug,  and  increase  dose   Pathways  and  
–  with  peripheral   to  dopamine  in  the  brain.   until  the  maximum  benefit  is  achieved  without  side-­‐ Anti-­‐Parkinson  
inhibitors)     effects   and  Schziophrenic  
However,  the  enzyme  DOPA  Decarboxylase  is  also   • Effectiveness  of  L-­‐DOPA  declines  with  time,  however   Drugs    
present  in  peripheral  tissues   Side  Effects:  
Therefore,  95%  of  administered  L-­‐DOPA  would  be   • Acute  
metabolised  to  dopamine  in  the  periphery  with   o Nausea  –  prevented  by  doperidone    
major  side  effects  of  nausea  and  vomiting.   (peripheral  acting  antagonist)  
  o Hypotension  
Therefore  L-­‐DOPA  is  commonly  prescribed  with  a   o Psychological  effects  –  Schizophrenia-­‐like  
peripheral  DOPA  decarboxylase  inhibitor   syndrome  with  delusions,  hallucinations,  also  
  confusion,  disorientation  and  nightmares  
Preparations  include:    
• SINAMET  (Carbidopa  +  L-­‐DOPA)    
• MADOPAR  (Benserazide  +  L-­‐DOPA)   • Chronic  
  o Dyskinesias  –  abnormal  movements  of  the  
limbs  and  face.  Can  occur  within  2  years  of  
treatment.  Disappear  if  the  doses  are  
reduced,  but  clinical  symptoms  then  
reappear.  
 
o “On-­‐Off”  Effects  –  Rapid  fluctuation  in  clinical  
states.  Off  periods  may  last  from  minutes  to  
hours.  Occurs  more  with  L-­‐DOPA.  
 
Bromocriptine   Dopamine  Agonists   Dopamine  Agonists:  Actions  
Pergolide     • Act  on  D2  Receptors   Dopaminergic  
Ropinerol   Actions:   • Longer  duration  of  action  than  L-­‐DOPA   Pathways  and  
• Act  on  D2  Receptors   • Smoother  and  more  sustained  response   Anti-­‐Parkinson  
  • Actions  independent  of  dopaminergic  neurons   and  Schziophrenic  
• Incidence  of  dyskinesias  is  less   Drugs  
• Can  be  used  in  conjunction  with  L-­‐DOPA  
 
Adverse  Effects:  
• Common  –  Confusion,  Dizziness,  Nausea/Vomiting,  
Hallucinations  
• Rare  –  Constipation,  Headache,  Dyskinesias  
 
Deprenyl  (Selegiline)   MAO  Inhibitors   Deprenyl  (Selegiline)  
    • Selectively  inhibits  MAO-­‐B  (and  hence  inhibits   Dopaminergic  
dopamine  breakdown)   Pathways  and  
• Predominantly  acts  in  dopaminergic  areas  of  the  CNS.   Anti-­‐Parkinson  
• Actions  are  without  peripheral  side  effects  of  non-­‐ and  Schziophrenic  
selective  MAO-­‐I’s   Drugs  
 
• Clinical  Uses  
o It  can  be  given  alone  in  the  early  stages  of  
Parkinson’s  Disease  
o Alternatively,  it  can  be  given  in  combination  
with  L-­‐DOPA  (reduce  the  dose  of  L-­‐DOPA  by  
30-­‐50%)  
 
• Side  Effects  (Rare)  
o Hypotension  
o Nausea/Vomiting  
o Confusion  
o Agitation  
 
Resagiline   MAO  Inhibitors   • Shown  to  have  neuroprotective  properties  by  
    inhibiting  apoptosis   Dopaminergic  
• Promotes  anti-­‐apoptosis  genes  leading  to  increased   Pathways  and  
cell  survival   Anti-­‐Parkinson  
• Early  clinical  trials  suggest  that  Resagiline  may  slow   and  Schziophrenic  
the  progression  of  Parkinson’s  disease   Drugs  

 
Tolocapone  (CNS  &   COMT  Inhibitors   CNS  Effects  
Peripheral)     Prevents  the  breakdown  of  dopamine  in  the  brain   Dopaminergic  
    Pathways  and  
Entacapone   Peripheral  Effects   Anti-­‐Parkinson  
(Peripheral  only)   COMT  in  the  periphery  converts  L-­‐DOPA  to  3-­‐0-­‐methyl-­‐DOPA   and  Schziophrenic  
  (3-­‐0MD).  3-­‐0MD  and  L-­‐DOPA  compete  for  the  same  transport   Drugs  
system  into  the  brain.  
   
COMT  inhibitors  stop  3-­‐0MD  formation,  thus  increasing  the  
bioavailability  of  L-­‐DOPA,  thus  more  L-­‐DOPA  crosses  into  the  
brain  and  is  converted  to  dopamine  in  the  CNS.  
 
Therefore,  this  reduces  L-­‐DOPA  dosage.  
Neuroleptics   • Mechanism  of  action  –  dopamine  antagonists   Other  Actions/Side  Effects:  
• Site  of  action  –  ‘D2-­‐like’  receptors   • Anti-­‐emetic  effect   Dopaminergic  
• Most  neuroleptics  block  other  receptors  e.g.   • Blocking  dopamine  receptors  in  the  chemoreceptor   Pathways  and  
5-­‐HT,  thus  accounting  for  some  of  their  side   trigger  zone.   Anti-­‐Parkinson  
effects   • Neuroleptic  Phenothiazine  –  effective  at  controlling   and  Schziophrenic  
  vomiting  and  nausea  induced  by  drugs  (e.g.   Drugs  
• Clozapine  is  relatively  non-­‐selective  between   chemotherapy),  renal  failure  
D1  and  D2  receptors,  but  does  have  a  high   • Many  neuroleptics  also  have  a  blocking  action  at    
affinity  for  D4  receptors  that  have  been   histamine  receptors.  
shown  to  be  increased  in  schizophrenia.   • Effective  at  controlling  motion  sickness.  
   
• Drugs  treat  positive  symptoms,  but  not   • Extrapyramidal  side  effects  –  Blockade  of  dopamine  
negative  ones.   receptors  in  the  nigrostriatal  system  can  induce  
• Delayed  effects  –  takes  weeks  to  work.   ‘Parkinson’  like  side-­‐effects  
• Initially  neuroleptics  induce  an  increase  in  DA    
synthesis  and  neuronal  activity.  This  declines   Acute  dyskinesias  –  Related  to  blockade  of  dopamine  
with  time.   receptors  in  the  striatum  which  leads  to  an  increase  in  
  cholinergic  function.  Develop  at  onset  of  treatment,  reversible  
on  drug  withdrawal  or  anti-­‐cholinergic  drugs.  
 
Tardive  dyskinesias  –  Involuntary  movements,  often  involving  
the  face  and  tongue.  
Occur  in  about  20%  of  patients  after  several  months  or  years  
of  therapy.  
• Made  worse  by  drug  withdrawal  or  anti-­‐cholinergics.  
• May  be  related  to  proliferation  in  presynaptic  DA  
receptors  or  drug  toxicity.  
• Incidence  is  less  with  atypical  drugs.  
 
Endocrine  Effects:  
• DA  is  involved  in  the  tuberoinfundibular  system  that  
regulates  prolactin  secretion.  
• Neuroleptics  increase  serum  prolactin  levels  –  this  can  
lead  to  gynaecomastia  (men  and  women)  and  
lactation  (women)  
 
Blockade  of  Muscarinic  Cholinergic  Receptors  
• Leads  to  typical  peripheral  anti-­‐muscarinic  side-­‐
effects  e.g.  blurred  vision,  increased  intraocular  
pressure,  dry  mouth,  constipation,  urinary  retention  
General  Anaesthetics    
Principles  of  
Clinical  setting:  
General  
Anaesthesia  
Desired  effect   Drug  
 
 
Loss  of  consciousness   Induction:  propofol  (i.v.)    
Suppression  of  reflex  responses   Maintenance:  enflurane  (inhalation)    
 
Analgesia   Opioid  (e.g.  i.v.  fentanyl)    
 
Neuromuscular  blocking  drugs  (e.g.    
Muscle  relaxation    
suxamethonium)  
 
Amnesia   Benzodiazepines  (e.g.  i.v.  midazolam)  
   
   
Local  Anaesthetics   Ester:  Cocaine  /    
Amide:  Lidocaine   Principles  of  Local  
  Anaesthesia  
Routes/Methods  of  Administration  
1. Surface  Anaesthesia  
Mucosal  surface  (mouth,  bronchial  tree),  Spray  (powder),  High  concentrations  –  can  lead  to  systemic  toxicity  
   
2. Infiltration  Anaesthesia  
Directly  into  tissues  à  sensory  nerve  terminals,  Used  in  minor  surgery  
Adrenaline  Co-­‐Injection  (NOT  in  EXTREMITIES  i.e.  fingers  or  toes,  as  it  can  cause  ischaemic  damage  due  to  its  
potent  vasoconstrictor  effects  in  extremities)  
 
3. Intravenous  Regional  Anaesthesia  
Intravenous,  distal  to  a  pressure  cuff  (which  is  also  administered)  e.g.  during  limb  surgery  
System  toxicity  can  be  caused  by  premature  cuff  release  (so  the  local  anaesthetic  reaches  systemic  
circulation)  
 
4. Nerve  Block  Anaesthesia  
Close  to  nerve  trunks  (e.g.  dental  nerves),  Widely  used  in  low  doses,  with  slow  onset,    
Co-­‐injected  with  a  vasoconstrictor  
 
5. Spinal  Anaesthesia  
Administered  in  the  sub-­‐arachnoid  space  (spinal  roots),  Used  in  abdominal,  pelvic,  lower  limb  surgery  
Causes  a  decrease  in  blood  pressure;  prolonged  headache  
 
6. Epidural  Anaesthesia  
Fatty  tissue  of  epidural  space  –  spinal  roots,  Uses  similar  to  spinal  anaesthesia  (abdominal,  pelvic,  lower  limb  
surgery)  and  painless  childbirth,  Slower  onset  –  higher  doses  
 
Lidocaine  and  cocaine:  pharmacokinetics  

  Lidocaine  (amide)   Cocaine  (ester)  

Absorption  (across  mucous  membranes)   Good   Good  

Plasma  protein  binding   70%   90%  


Hepatic   Hepatic  and  plasma  
Metabolism  
N-­‐dealkylation   Non-­‐specific  esterases  
Plasma  half-­‐life   2  hours   1  hour  
 
Administration:  
• Cocaine:  Now  only  used  as  a  surface  anaesthetic  (in  ophthalmology)  
• Lidocaine:  may  be  administered  by  any  route  
Metabolism:  
• Cocaine:  Metabolised  rapidly  
• Lidocaine:  Relatively  resistant  to  metabolism  
Unwanted  Effects:  
• Lidocaine:  A  classic  local  anaesthetic;  therefore  most  of  its  side  effects  apply  to  other  local  anaesthetics  
• Cocaine:  exception  to  the  rule  (unwanted  effects  affect  the  CNS  and  CVS)  
Lidocaine  Unwanted  Effects:  
• CNS  –  Paradoxical  Effect  (you  would  expect  it  to  damp  down  CNS  activity,  but  it  stimulates  CNS  activity)  
• CVS  –  Predictable  Effects  (due  to  Na+  Channel  Blockade)  
Cocaine  Unwanted  Effects:  
• CNS  &  CVS  –  Unwanted  effects  occur  due  to  the  sympathetic  actions  of  cocaine  (it  blocks  NA  reuptake)  
 
Lidocaine  and  cocaine:  unwanted  effects  

  Lidocaine   Cocaine  

CNS  stimulation  
CNS   Restlessness  &  confusion   Euphoria  and  excitation  
Tremor  

Myocardial  depression   Increased  CO  


CVS   Vasodilatation   Vasoconstriction  
Reduced  BP   Increased  BP  
 
 
 
 
   
Methotrexate   Alkylating  Agent  (Cytotoxic  Drugs)   Folic  Acid:    
    • An  essential  nutrient   Cytotoxic  Drugs  
Cytotoxic  Drugs  –   Interfere  with  thymidylate  synthesis  (generation  of   • Important  for  nucleotide  synthesis  
ALKYLATING  AGENTS   pyrimidines)    
  Methotrexate:  
Fluorouracil   Alkylating  Agent  (Cytotoxic  Drugs)   • Structurally  similar  to  folic  acid  
    • Can  substitute  for  folate   Cytotoxic  Drugs  
Cytotoxic  Drugs  –   Inhibit  enzymes  in  DNA  synthesis      
ALKYLATING  AGENTS   (e.g.  thymidylate  synthetase)   Fluorouracil:  
  • Characteristic  of  pyrimidines:  structurally  similar  
to  both  uracil  and  thymidine  except  for  the  
fluorine  group  
• Fluorine  makes  the  molecule  unreactive  at  that  
point  –  therefore  it  can  interfere  with  nucleic  
acid  synthesis  
 
Methotrexate  and  Fluorouracil:  
• DHFR  (Dihydrofolate  Reductase)  generates  
tetrahydrofolate  derivatives  
• Folate  metabolites  donate  and  accept  electrons  
and  protons  as  part  of  the  synthetic  pathway  
• Uridine  derivatives  à  Thymidine  derivatives  (by  
the  passage  of  electrons)  
• Thymidylate  synthetase  is  a  key  target  for  
chemotherapy  
Methotrexate:  Blocks  DHFR  Activity  
• It  is  a  folate  mimic  –  DHFR  binds  to  
methotrexate  as  if  it  were  folate,  but  as  
methotrexate  cannot  participate  in  the  reaction,  
it  essentially  BLOCKS  DHFR  
Fluorouracil:  Uridine  and  Thymidine  Analogue  
• Thymidylate  synthetase  binds  to  fluorouracil  
(thinking  it  is  a  uridine)  
 
 
Azothioprine   Alkylating  Agent  (Cytotoxic  Drugs)    
    Cytotoxic  Drugs  
Cytotoxic  Drugs  –   Inhibit  purine  synthesis  
ALKYLATING  AGENTS  
 
Actinomycin  D   Intercalates  with  DNA  and  interferes  with  topoisomerase  II   • Topoisomerase  II  cuts  the  DNA,  allows  it  to  
  unwind,  and  re-­‐joins  it  –  this  allows  proliferation   Cytotoxic  Drugs  
Cytotoxic  Drugs  – to  occur.  
CYTOTOXIC   • Actinomycin  D  inhibits  topoisomerase  II  by  
ANTIBODIES   binding  DNA  in  this  way  
 
DOXORUBICIN   Inhibits  DNA  and  RNA  synthesis     • It  has  the  ability  to  intercalate  DNA  and  inhibit  
    topoisomerase  II   Cytotoxic  Drugs  
Cytotoxic  Drugs  –  
CYTOTOXIC  
ANTIBODIES  
 
BLEOMYCINS   Metal-­‐Chelating-­‐Glycopeptide  antibiotics  that  degrade  DNA   • Chelate  m  etals  and  generate  free  radicals  
    (oxygen  derived)   Cytotoxic  Drugs  
Cytotoxic  Drugs  – • Free  radicals  cause  DNA  strand  breaks  
CYTOTOXIC   • Bleomycins  are  active  against  non-­‐dividing  cells,  
ANTIBODIES   since  they  do  not  rely  on  cell  proliferation  
  • Problems  associated  with  high  damage  to  LUNG  
TISSUE  
• Intravenous  administration  
 
PODOPHYLLOTOXINS   Podophyllotoxins:  e.g.  etoposide   • Inhibit  DNA  synthesis  and  cause  a  cell  cycle  block  
e.g.  etoposide     at  G2   Cytotoxic  Drugs  
   
Cytotoxic  Drugs  –  
PLANT  ALKALOIDS  
 
VINCA  ALKALOIDS  e.g.   Vinca  Alkaloids:  e.g.  vincristine   • Bind  to  tubulin  and  inhibit  its  polymerisation  
Vincristine       into  microtubules  –  this  prevents  spindle  fibre   Cytotoxic  Drugs  
  formation  
Cytotoxic  Drugs  –    
PLANT  ALKALOIDS  
 
HYDROXYUREA     • Inhibits  ribonucleotide  reductase  (involved  in  
  nucleic  acid  synthesis)   Cytotoxic  Drugs  
Cytotoxic  Drugs  –    
MISC.  
 
CISPLATIN     • Interacts  with  DNA  and  causes  guanine  intra-­‐
  strand  cross-­‐links   Cytotoxic  Drugs  
Cytotoxic  Drugs  –    
MISC.  
 
PROCARBAZINE     • Inhibits  DNA  and  RNA  Synthesis  and  interferes  
  with  mitosis  at  interphase   Cytotoxic  Drugs  
Cytotoxic  Drugs  –   • Metabolically  activated  by  cytochrome  P450  and  
MISC.   MAO  à  alkylate  DNA  (at  N7  and  O6  of  guanine)  
   
• Prodrug  –  a  substrate  for  Cytochrome  P450  and  
MAO  
• N-­‐N  à  N=N  (this  activates  it  and  produces  
multiple  metabolites,  including  alkylating  
agents)  
• Alkylating  agents  covalently  bind  to  DNA  to  
produce  bulky  DNA  adducts  
 
Hormones     • Used  for  chemotherapy  but  are  not  technically    
As  Cytotoxic  Drugs   cytotoxic   Cytotoxic  Drugs  
Hormones:   Prednisolone   Fosfestrol   Tamoxifen  
• Can  inhibit  tumours  in  hormone-­‐sensitive  tissues  
(e.g.  prostate,  breast)   Mechanism   Glucocorticoid   Androgen   SERM  
• Gonadotrophin-­‐releasing  hormone  analogues  e.g.  
Goserelin   Leukaemias  &   Prostate   Breast  
  Use  
lymphomas   cancer   cancer  
 
General  Toxic  Effects  
• Myelotoxicity  
• Impaired  wound  healing  
• Depression  of  growth  (children)  
• Sterility  
• Teratogenicity  
• Loss  of  hair  
• Nausea  and  Vomiting  
 
 
 
 
 
Side  Effects:  
On  fast  growing  cells:  
• Inhibit  cell  division  
• Cell-­‐cycle  specific  drugs  affect:  bone  marrow,  GI  
Tract,  Epithelium,  hair  &  nails,  spermatogonia  
 
On  slow  growing  cells:  
• Introduce  DNA  mutations  
• Cell-­‐cycle  independent  drugs  (alkylating  agents)  
cause  secondary  tumours  
 
 
 
Phenytoin  (PHT)   • Blocks  voltage-­‐gated  Na+  channels   Indications:  
  • Partial  Epilepsy    
Anticonvulsant     • Status  Epilepticus  
(Anti-­‐Epileptic)    
Mechanism  of  Action:  
• Blocks  voltage-­‐gated  Na+  Channels  
 
Drug  Level  Monitoring  
• Useful  
 
Elimination  Half-­‐Life  
• 20  Hours  
 
Metabolism  
• Hepatic  metabolism  
• Oxidation  &  hydroxylation,  then  conjugation  
• Potent  hepatic  enzyme  inducer  
 
Active  Metabolites  
• None  
 
Drug  Interactions  
• Complex  
 
Adverse  Drug  Reactions  
• Ataxia  
• Sedation  
• Hypersensitivity  
• Rash  
• Fever  
• Gingival  hypertrophy  
• Folate  deficiency  
• Megaloblastic  anaemia  
• Vitamin  K  deficiency  
• Depression  
• Hirsutism  
• Peripheral  neuropathy  
• Osteomalacia  
• Reduced  bone  density  
• Hypocalcaemia  
• Hepatitis  
• Vasculitis  
• Myopathy  
• Coagulation  defects  
• Bone  marrow  hypoplasia  
 
Carbamazepine   • Blockade  of  voltage-­‐gated  Na+  Channels   Indications:  
  • Partial  and  Secondary  Generalised  Seizures      
Anticonvulsant      
(Anti-­‐Epileptic)   Mechanism  of  Action:  
• Blocks  voltage-­‐gated  Na+  Channels  
 
Drug  Level  Monitoring  
• Useful  
 
Elimination  Half-­‐Life  
• 5-­‐26  hours  (x3  daily  dosing,  unless  SR  
preparations)  
 
Metabolism  
• Hepatic  oxidation  then  conjugation  
• CBZ  is  a  potent  hepatic  enzyme  inducer    
 
Active  Metabolites  
• Carbamezepine  epoxide    
 
Drug  Interactions  
• Complex  drug  interaction  profile    
 
Adverse  Drug  Reactions  
• Hypersensitivity  (rash,  hepatitis,  nephritis)  
• Dose-­‐Related  (Ataxia,  dizziness,  sedation,  
diplopia)  
• Chronic  (Vitamin  K  Deficiency,  depression,  
impotence,  osteomalacia,  hyponatraemia)  
 
Lamotrigine   • Blocks  voltage-­‐gated  Na+  channels   Indications:  
  • Partial  and  generalised  epilepsy  (wide    
Anticonvulsant     spectrum)  
(Anti-­‐Epileptic)    
Mechanism  of  Action:  
• Blocks  voltage-­‐gated  Na+  Channels  
 
Drug  Level  Monitoring  
• N/A  
 
Elimination  Half-­‐Life  
• 29  hours  (monotherapy)  
• 15  hours  (enzyme  inducing  co-­‐medication)  
• 60  hours  (valproate  co-­‐medication)  
 
Metabolism  
• Hepatic  metabolism  
• Glucuronidation  (no  phase  I  metabolism)  
• Does  not  induce  hepatic  enzymes  
 
Active  Metabolites  
• None    
 
Drug  Interactions  
• Valproate  
• Enzyme  inducing  AEDs  
 
 
 
 
Adverse  Drug  Reactions  
• Usually  well  tolerated  
• Rash  (high  incidence;  may  be  severe)  
• Headache  
• Blood  dyscrasias  
• Ataxia  
• Diplopia  
• Dizziness  
• Sedation  
• Insomnia  
• Mood  disturbance  
 
(Sodium)  Valproate   • Enhances  GABA  transmission  by  several   Indications:  
  mechanisms   • Partial  or  generalised  epilepsy  (wide  spectrum)    
Anticonvulsant      
(Anti-­‐Epileptic)   Mechanism  of  Action:  
• Enhances  GABA  transmission  by  several  
mechanisms    
 
Drug  Level  Monitoring  
• N/A  
 
Elimination  Half-­‐Life  
• 4-­‐12  hours  
 
Metabolism  
• Hepatic  metabolism  
• Oxidation,  then  conjugation  
• Potent  hepatic  enzyme  inhibitor  
 
Active  Metabolites  
• None    
 
Drug  Interactions  
• Many  
 
 
 
 
 
Adverse  Drug  Reactions  
• Severe  hepatic  toxicity  (especially  in  young  
patients)  
• Pancreatitis  
• Encephalopathy  (ammonia  driven)  
• Drowsiness  
• Tremor  
• Blood  dyscrasias  
• Hair  thinning  and  hair  loss  
• Weight  gain  
• Endocrine  (PCO)  
 
 
Sulphanilamide   Folate  is  required  for  DNA/RNA  Synthesis  in  both  man  and   Pharmacokinetics:  
(P-­‐Aminobenzoic  Acid   bacteria   • Readily  absorbed  in  the  GI  tract.   Anti-­‐Microbial  
Analogue)   Man  has  evolved  specific  uptake  processes  for  transporting   • Maximum  plasma  concentration  is  reached   Drugs  
  folate  into  the  cells.   within  4-­‐6  hours  
(ANTI-­‐MICROBIAL      
DRUG)   Bacteria  have  to  synthesise  folate.  P-­‐aminobenzoic  acid  is   Side  Effects:  
  essential  for  the  synthesis  of  folic  acid  in  bacteria.   Mild/moderate  (do  not  warrant  withdrawal)    
  • Nausea  and  vomiting  
Sulphanilamide  is  a  structural  analogue  of  P-­‐aminobenzoic   • Headache  
acid  that  COMPETES  for  the  enzyme  dihydropteroate  which   • Mental  depression  
is  involved  in  the  synthesis  of  folate.    
  Severe  (warrants  withdrawal)  
They  interfere  with  bacterial  DNA/RNA  synthesis  and  are   • Hepatitis-­‐type  reaction  
bacteriostatic  i.e.  they  arrest  the  growth  of  the  bacteria,  but   • Hypersensitivity  reactions  
do  not  kill  them  –  it  is  then  up  to  the  host  defence  system.  • Bone  marrow  suppression  
   
Note  –  WIDESPREAD  RESISTANCE  
 
Trimethoprim   The  utilisation  of  folate,  in  the  form  of  tetrahydrofolate  as  a   Pharmacokinetics  
(Folate  Antagonists)   co-­‐factor  in  thymidylate  synthesis  is  an  example  of  a   • Oral  administration   Anti-­‐Microbial  
  pathway  in  which  there  is  a  differential  sensitivity  of  human   • Trimethoprim  is  fully  absorbed  from  the  GI  tract   Drugs  
(ANTI-­‐MICROBIAL   and  bacterial  enzymes  to  drugs.   • Widely  distributed  throughout  the  tissues  and  
DRUG)     body  fluids  
This  pathway  is  virtually  identical  in  micro-­‐organisms  and   • Reaches  high  concentrations  in  the  lungs  and  
man,  but  one  of  the  key  enzymes  dihydrofolate  reductase  is   kidneys.  
many  times  more  sensitive  to  particular  analogues  in  either    
man  or  bacteria.    
   
Trimethoprim  is  a  folate  antagonist  that  is  more  sensitive  to   Unwanted  Effects  
the  dihydrofolate  reductase  enzyme  in  BACTERIA  than  in   • Nausea/vomiting  
man  (IC50  [mmol/l]  0.005  bacteria,  260  man)   • Skin  rashes  
  • Hypersensitivity  –  even  the  small  dose  of  
sulphonamide  which  is  used  in  co-­‐trimoxazole  
can  still  cause  serious  hypersensitivity  reactions,  
which  are  not  dose  related.  
 
Clinical  uses  
• Urinary  tract  and  Respiratory  tract  infections  
 
Co-­‐Trimoxazole   Sulphamethazole  and  trimethoprim.   Pharmacokinetics  
(Sequential  Blockade)     • When  given  as  co-­‐trimoxazole,  about  two-­‐thirds   Anti-­‐Microbial  
  • Since  sulphonamides  affect  the  earlier  stage  in  the   of  each  drug  is  protein  bound  and  about  half  of   Drugs  
(ANTI-­‐MICROBIAL   same  metabolic  pathway  i.e.  folate  synthesis,  they   each  is  excreted  within  24  hours.  
DRUG)   potentiate  the  actions  of  trimethoprim.    
  Clinical  Uses  
• When  given  in  combination,  the  drugs  are  effective   • For  infections  with  pneumocystis  carinii  which  
at  one  tenth  or  less  of  what  would  be  needed  if   causes  pneumonia  in  patients  with  AIDS,  co-­‐
each  drug  was  given  on  its  own.   trimoxazole  is  used  in  high  doses.  
   
Penicillin   β-­‐Lactam  antibiotics  e.g.  penicillin  inhibit  the  formation  of   Pharmacokinetics  
(β-­‐Lactam  Antibiotics)   peptidoglycan  and  are  subsequently  bacteriocidal   When  administered  orally  -­‐  Different  penicillins  are   Anti-­‐Microbial  
    absorbed  to  differing  degrees  depending  on  their   Drugs  
(ANTI-­‐MICROBIAL   Method  of  Action:   stability  in  acid  and  their  adsorption  on  to  food.  
DRUG)   • Interfere  with  the  synthesis  of  the  bacterial  wall    
peptidoglycan   The  drugs  are  widely  distributed  by  the  bodily  fluids,  
• Inhibit  the  transpeptidation  enzyme  that  cross  links   passing  into  joints,  pleural  and  pericardial  cavities,  into  
the  peptide  chains  attached  to  the  backbone  of  the   the  bile,  the  saliva  and  the  milk,  and  across  the  placenta.  
peptidoglycan.    
  Being  lipid  insoluble  they  do  not  enter  mammalian  cells.  
 
They  therefore  do  not  readily  cross  the  blood  brain  
barrier,  UNLESS  the  meninges  are  inflamed,  in  which  
case  they  may  reach  effective  therapeutic  
concentrations.  
 
 
 
Elimination  
Elimination  of  most  penicllins  is  mainly  renal  and  occurs  
rapidly  -­‐  90%  tubular  secretion  
 
Unwanted  Effects  
Relatively  free  from  direct  toxic  effects  
Hypersensitivity  reactions  –  breakdown  products  of  
penicillins  combine  with  host  protein  and  become  
antigenic.  
 
Most  common  reactions  are  skin  rashes  and  fever,  acute  
anaphylactic  shock.  
 
A  side  effect  of  broad  spectrum  penicillins  is  effect  on  
the  gut  bacterial  flora  -­‐  in  GI  tract  disturbances.  
 
Resistance  to  β-­‐Lactam  Antibiotics  
1. Production  of  β-­‐Lactamases  by  bacteria  
Genetically  controlled  and  can  be  transferred  
from  one  bacterium  to  another.  
 
Staphylococcal  resistance  via  β-­‐lactamase  
production  has  spread  progressively.  
 
In  developed  countries,  at  least  80%  of  
staphylococci  now  produce  β-­‐Lactamase.  
 
Solution:  Use  β-­‐lactamase  inhibitors  e.g.  
CLAVULANIC  ACID  which  functions  by  covalently  
binding  to  the  enzyme  at,  or  close  to,  its  active  
site.  
 
2. Reduction  in  the  Permeability  of  the  outer  
membrane  
Therefore,  there  is  a  decreased  ability  of  the  
drug  to  penetrate  to  the  target  site.  
 
3. The  occurrence  of  modified  penicillin-­‐binding  
sites  
 
CLAVULANIC  ACID   Functions  by  covalently  binding  to  the  β-­‐Lactamase  enzyme   Reduces  resistance  to  penicillin  
  at,  or  close  to,  its  active  site.   Anti-­‐Microbial  
(β-­‐lactamase   Drugs  
inhibitors)  
 
CEPHALOSPORINS   e.g.  Cephalexin  (oral),  Cefuroxime  and  Cefotaxime    
(β-­‐Lactam  Antibiotics)   (parenteral)   Pharmacokinetics   Anti-­‐Microbial  
    Some  cephalosporins  may  be  given  orally,  but  most  are   Drugs  
  e.g.  cefoperazone,  cefotaxime  (can  cross  BBB)   given  parenterally  –  intramuscular  (i.m.)  or  intravenous  
(ANTI-­‐MICROBIAL     (i.v.)  
DRUG)      
  Mechanism  of  Action   Widely  distributed  in  the  body,  passing  into  the  pleural,  
• Same  as  penicillins,  interfere  with  peptidoglycan   pericardial  and  joint  fluids,  and  across  the  placenta.  
synthesis    
• Resistance  to  this  group  of  drugs  has  increased.   Some  cephalosporins  cross  the  blood  brain  barrier  
• Nearly  all  Gram  –ve  bacteria  have  the  gene   e.g.  cefoperazone,  cefotaxime  (Drug  of  choice  for  
encoding  for  β-­‐lactamase,  which  is  more  active  in   bacterial  meningitis)  
hydrolysing  cephalosporins  than  penicillin.    
  Excretion  (Most  Cephalosporins)  
• Resistance  also  occurs  if  there  is  decreased   • Excretion  is  mostly  via  the  kidney  –  largely  by  
penetration  of  the  drug  –  due  to  alterations  of  the   tubular  secretion  
membrane  proteins  or  mutations  of  the  binding   • But  40%  of  ceftriaxone  and  75%  of  
site  proteins.   cefoperazone  is  eliminated  in  the  bile.  
  • Since  different  β-­‐Lactam  antibiotics  may  bind  to  
• They  are  bactericidal.   different  binding  proteins,  it  may  be  feasible  to  
  combine  two  or  more  of  these  agents  and  
achieve  a  synergistic  action  between  them.  
 
Unwanted  Effects:  
• Hypersensitivity  reactions  (similar  to  those  with  
penicillin)  may  be  seen.  
• Some  cross-­‐reaction  occur  (about  10%  of  
penicillin  sensitive  individuals  will  be  allergic  to  
cephalosporins)  
• Nephrotoxicity  has  been  reported  (especially  
with  cephradine)  
• Diarrhoea  can  occur  with  oral  cephalosporins.  
 
TETRACYCLINES   Tetracyclines  are  broad-­‐spectrum  antibiotics  that  have  a   Pharmacokinetics  
(Drugs  that  inhibit   polycyclic  structure.     • Usually  given  orally   Anti-­‐Microbial  
bacterial  protein     • Can  also  be  given  parenterally   Drugs  
synthesis)   Method  of  Action   • The  absorption  of  most  preparations  from  the  
  • Actively  transported  into  bacteria  and  interrupt   gut  is  irregular  and  incomplete,  and  is  improved  
(ANTI-­‐MICROBIAL   protein  synthesis.   by  the  absence  of  food  
DRUG)   • Competition  with  tRNA  for  the  A-­‐binding  site.   • Since  tetracycline’s  chelate  metal  ions  (e.g.  
  • Bacteriostatic,  not  bactericidal.   iron),  forming  a  non-­‐absorbable  complex,  
  absorption  is  decreased  by  the  presence  of  
Spectrum   MILK,  certain  antacids  and  iron  preparations  
• Very  wide  and  include  Gram  +ve  and  Gram  –ve   • The  drugs  have  wide  distribution  
bacteria,  mycoplasma,  Rickettsia,  Chlamydia,  some   • The  enter  most  fluid  compartments  
spirochaetes  and  some  protozoa  (e.g.  amoebae)    
  Excretion  
• However,  many  strains  have  become  resistant  to   • Excretion  is  both  via  the  bile  and  by  glomerular  
these  agents.   filtration  in  the  kidney  
• The  basis  of  resistance  is  the  development  of   • Most  tetracyclines  will  accumulate  if  renal  
energy-­‐dependent  efflux-­‐mechanisms  which   function  is  impaired.  
transport  the  tetracyclines  OUT  OF  THE   Exception  –  DOXYCYCLINE  (largely  excreted  into  the  
BACTERIUM,  but  alterations  of  the  target  (the   gastrointestinal  tract  via  the  bile).  
bacterial  ribosome)  also  occur.    
  Unwanted  Effects  
  • Most  common  is  gastrointestinal  disturbances,  
due  initially  to  direct  irritation  and  later  by  the  
modification  of  gut  flora  
• Because  they  chelate  calcium,  tetracyclines  are  
deposited  in  GROWING  BONES  and  TEETH,  
causing  staining  and  sometimes  bone  
deformities.  They  should  therefore  not  be  given  
to  children,  pregnant  women  or  nursing  
mothers.  
 
• Phototoxicity  (sensitisation  to  sunlight)  has  been  
seen  –  more  particularly  with  demeclocycline.  
• Minocycline  can  produce  vestibular  
disturbances  (dizziness  and  nausea)  –  the  
frequency  of  which  is  dose  related.  
• High  doses  of  tetracyclines  can  decrease  protein  
synthesis  in  host  cells  –  an  anti-­‐anabolic  effect.  
 
Chloramphenicol   Mechanism  of  Action   Pharmacokinetics  
(Drugs  that  inhibit   • Inhibition  of  protein  synthesis.   • Oral  chloramphenicol  is  rapidly  and  completely   Anti-­‐Microbial  
bacterial  protein   • Chloramphenicol  binds  to  the  50s  Subunit  of  the   absorbed   Drugs  
synthesis)   ribosome  and  inhibits  transpeptidation.   • Reaches  maximum  concentration  in  the  plasma  
    within  2  hours.  
(ANTI-­‐MICROBIAL   Spectrum   • Can  be  given  parenterally  
DRUG)   • Wide  spectrum  of  activity,  including  Gram  –ve  and    
  Gram  +ve  bacteria.   • Widely  distributed  throughout  the  tissues  and  
  • They  are  bacteriostatic  for  most  organisms.   body  fluids  (including  CSF)  
  • In  the  plasma,  30-­‐50%  plasma  protein  bound  
Resistance   • Half-­‐life  is  approximately  2  hours  
• Resistance  is  due  to  the  production  of    
chloramphenicol  acetyl-­‐transferase  and  is  plasmid-­‐ • About  10%  is  excreted  unchanged  in  the  urine  
mediated.   • The  remainder  is  inactivated  in  the  liver.  
  • Metabolites  are  excreted  via  the  kidney  and  the  
• R-­‐plasmids  containing  determinants  for  multiple   bile.  
drug  resistance  for  chloramphenicol,  streptomycin    
and  tetracyclines  etc.  may  be  transferred  from  one   Unwanted  Effects  
bacterial  specie  to  another  by  promiscuous   • The  most  important  unwanted  effect  is  
plasmids.   depression  of  the  bone  marrow  resulting  in  
  pancytopenia  
• Derivatives  of  chloramphenicol  with  the  terminal  –  
OH  on  the  side-­‐chain  replaced  by  fluorine  are  not  as   • Decrease  in  all  blood  cell  elements  –  an  effect  
susceptible  to  acetylation,  and  thus  retain   which  (although  rare)  can  occur  even  with  very  
antibacterial  activity.   low  doses  in  some  individuals.  
   
• Chloramphenicol  should  be  used  with  great  care  
in  new-­‐borns  because  inadequate  inactivation  
and  excretion  of  the  drug  can  result  in  ‘grey  
baby  syndrome’  –  vomiting,  diarrhoea,  
flaccidity,  low  temperature  and  an  ash  grey  
colour.  
 
• This  carries  a  40%  mortality  rate  
 
• Hypersensitivity  reactions  can  occur  
• GI  disturbances  
• Other  alteration  of  the  intestinal  microbial  flora.  
 
AMINOGLYCOSIDES   Method  of  action   Pharmacokinetics  
e.g.  GENTAMICIN   • The  aminoglycosides  inhibit  bacterial  protein   • The  aminoglycosides  are  polycations  and  highly   Anti-­‐Microbial  
  synthesis  by  binding  to  the  30s  Subunit  of  the   polar  –  hence  are  not  absorbed  in  the  GI  tract   Drugs  
(Drugs  that  inhibit   ribosome   • Given  intramuscularly  (i.m.)  or  intravenously  
bacterial  protein   • This  causes  an  alteration  in  the  codon:anticodon   (i.v.).  
synthesis)   recognition  and  results  in  a  misreading  of  mRNA,   • Binding  to  plasma  proteins  is  minimal.  
  and  the  production  of  defective  bacterial  proteins    
(ANTI-­‐MICROBIAL     • They  do  not  enter  cells  and  do  not  cross  the  
DRUG)   • However,  this  action  does  not  entirely  explain  their   blood  brain  barrier  into  the  CNS.  
  rapid  lethality  –  so  it  is  possible  there  may  be  a   • Plasma  half-­‐life  is  2-­‐3  hours.  
second  target.   • Elimination  is  virtually  entirely  by  glomerular  
  filtration  in  the  kidney.  
• Their  penetration  through  the  cell  membrane  of  the   • Tissue  concentrations  increase  during  
bacterium  depends  on  an  oxygen-­‐dependent  active   treatment,  and  can  reach  toxic  levels  after  about  
transport  system,  which  chloramphenicol  can   a  week  of  unmodified  dosage.  
block.    
• Their  effect  is  bactericidal  and  is  enhanced  by   Unwanted  Effects:  
agents  that  interfere  with  cell  wall  synthesis.   • Ototoxicity  –  progressive  damage  to  and  
  destruction  of  the  sensory  cells  in  the  cochlea  
Resistance   and  vestibular  organs  of  the  ear.  
• Resistance  to  aminoglycosides  is  becoming  a    
problem,  and  may  be  due  to  a  number  of  factors  –   • Nephrotoxicity  –  damage  to  the  kidney  tubules.  
the  most  important  is  inactivation  by  microbial   o Can  be  reversed  if  the  use  of  the  drug  is  
enzymes.   stopped.  
• Other  mechanisms  of  resistance  include:   o Since  elimination  of  these  drugs  is  
o Failure  of  penetration  (overcome   almost  entirely  renal,  their  nephrotoxic  
concomitant  use  of  penicillin  and/or   action  can  impair  their  own  excretion  
vanocomycin  which  synergies  with   and  a  vicious  cycle  can  be  set  up.  
aminoglycosides)     o Plasma  concentrations  must  be  
o Lack  of  binding  of  the  drug  due  to   monitored  regularly.  
mutations  that  alter  the  binding-­‐site  on  the    
30S  subunit.  
 
Spectrum  
• The  aminoglycosides  are  effective  against  many  
aerobic  Gram  –ve  and  some  Gram  +ve  bacteria  
• They  may  be  given  together  with  penicillin  in  
infections  caused  by  Streptococcus,  Listeria  or  
Pseudomonas  aeruginosa  
 
ISONIAZID   Mechanism  of  Action:   Pharmacokinetics  
  • The  antibacterial  activity  of  isoniazid  is  limited  to   • Readily  absorbed  from  the  GI  tract,  or  after   Anti-­‐Microbial  
(Antimycobacterial   mycobacteria.   parenteral  injection   Drugs  
agent)   • It  is  bacteriostatic  on  resting  organisms,  and  can  kill   • Widely  distributed  throughout  the  tissues  and  
  dividing  bacteria  (bactericidal  effect).   body  fluids,  and  CSF.  
(ANTI-­‐MICROBIAL      
DRUG)   • It  passes    freely  into  mammalian  cells  and  therefore   • Importantly  –  penetrates  well  into  NECROTIC  
  effective  against  intracellular  organisms   TUBERCULOUS  LESION  
• The  mechanism  of  action  is  not  fully  understood  –   • Metabolism,  involves  largely  acetylation,  
some  evidence  suggests  inhibition  of  the  synthesis   depends  on  genetic  factors  that  determine  
of  mycolicacids  (important  constituents  of  the  cell   whether  a  person  is  a  slow  (t½=3hours)  or  rapid  
wall  and  peculiar  to  mycobacterium)   (t½=1.5hours)  acetylator  of  the  drug.  
  • Slow  acetylators  have  a  better  therapeutic  
response.  
 
RIFAMPICIN   Mechanism  of  Action:   Pharmacokinetics  
  • Binds  to  and  inhibits  DNA-­‐dependent  RNA   • Rifampicin  is  given  orally.   Anti-­‐Microbial  
(Antimycobacterial   polymerase  in  prokaryotic,  but  not  eukaryotic  cells.   • Widely  distributed  in  the  tissues  and  body   Drugs  
agent)   • It  is  one  of  the  most  active  anti-­‐tuberculosis  agents   fluids  
  known.    
(ANTI-­‐MICROBIAL   • It  is  also  active  against  most  other  Gram  +ve   • Excreted  partly  in  the  urine  and  partly  in  the  
DRUG)   bacteria  as  well  as  many  Gram  -­‐ve  species.   bile-­‐  some  of  it  undergoing  enterohepatic  
  • It  enters  phagocytic  cells  and  can  kill  intracellular   cycling.  
micro-­‐organisms.    
  • There  is  progressive  metabolism  of  the  drug  by  
deacetylation  during  its  repeated  passage  
through  the  liver.  
 
 
• The  metabolite  retains  antibacterial  activity  but  
is  less  well  absorbed  from  the  GI  tract.  
 
Unwanted  effects  
• Infrequent,  occurring  in  fewer  than  4%  of  
individuals  
• E.g.  skin  eruptions,  fever,  GI  tract  disturbances  
 
 
PYRAZINAMIDE   Mechanism  of  Action   Pharmacokinetics  
  • Pyrazinamide  is  inactive  at  neutral  pH,  but   • Oral  administration  -­‐  the  drug  is  well  absorbed   Anti-­‐Microbial  
(Antimycobacterial   tuberculostatic  at  acidic  pH.   after  oral  administration     Drugs  
agent)   • It  is  effective  against  the  intracellular  organism  in   • Widely  distributed,  penetrating  well  into  the  
  macrophages,  since  after  phagocytosis  the   meninges  
(ANTI-­‐MICROBIAL   organism  with  be  contained  in  phagolysosomes,  in   • Excreted  through  the  kidneys  (mainly  
DRUG)   which  the  pH  is  low.   glomerular  filtration)  
     
Unwanted  Effects  
• Arthralgia  (associated  with  high  concentrations  
of  plasma  urates)  
• GI  tract  upsets  
• Malaise  
• Fever  
 
NYSTATIN   Nystatin  is  a  polyene  macrolide.    
    There  is  virtually  no  absorption  from  the  mucous   Anti-­‐Microbial  
(Anti-­‐Fungal  Agent)   Mechanism  of  Action   membranes  of  the  body,  or  from  the  skin,  and  its  use  is   Drugs  
  • Binds  to  cell  membrane  and  interferes  with   limited  to  fungal  infections  of  the  SKIN  and  GI  TRACT.  
(ANTI-­‐MICROBIAL   permeability  and  transport  functions.    
DRUG)     Unwanted  Effects  
  • It  forms  a  pore  in  the  membrane  –  the  hydrophilic   • Rare  -­‐  Limited  to  nausea  and  vomiting  when  high  
core  of  the  molecule  creating  a  transmembrane  ion   doses  are  taken  by  mouth  
channel   • Very  rare  -­‐  Rash  
   
• Nystatin  has  a  selective  action,  binding  avidly  to  the  
membranes  of  fungi  and  some  protozoa,  and  less  
avidly  to  mammalian  cells,  and  not  at  all  to  bacteria.  
 
• The  relative  specificity  for  fungi  may  be  due  to  the  
drug’s  greater  avidity  for  ergosterol  (fungal  
membrane  sterol)  than  for  cholesterol  (the  main  
sterol  in  the  plasma  membrane  in  animal  cells).  
 
• It  is  effective  against  most  fungi  and  yeast  
 
 
 
MICONAZOLE   Miconazole  belongs  to  the  azole  group  of  synthetic   Pharmacokinetics  
  antimycotic  agents,  with  a  broad  spectrum  of  activity.   • Miconazole  is  given  by  intravenous  infusion  for   Anti-­‐Microbial  
(Anti-­‐Fungal  Agent)     systemic  infections   Drugs  
  Mechanism  of  Action   • Given  orally  for  infections  of  the  GI  tract.  
(ANTI-­‐MICROBIAL   • Azoles  block  the  synthesis  of  ergosterol  (the  main   • Short  plasma  half-­‐life.  
DRUG)   sterol  in  the  fungal  cell  membrane)  by:    
  o Interacting  with  the  enxyme  necessary  for   Unwanted  Effects  
conversion  of  lanosterol  to  ergosterol.   • Relatively  infrequent  (most  commonly  being  GI  
  Tract  disturbances,  blood  dyscrasias)  
• The  resulting  depletion  of  ergosterol  alters  the    
fluidity  of  the  membrane  and  this  interferes  with    
the  action  of  the  membrane  associated  enzymes.  
 
• The  overall  effect  is  an  inhibition  of  replication.  
• A  further  repercussion  is  the  inhibition  of  the  
transformation  of  candida  yeast  cells  into  hyphae  
(the  invasive  and  pathogenic  form  of  the  parasite)  
 
ACYCLOVIR   • Acyclovir  is  a  guanosine  derivative  with  a  high   Herpes  simplex  is  more  sensitive  to  acyclovir  than  other  
  specificity  for  herpes  simplex.   herpes  viruses  which  cause  glandular  fever  or  shingles.   Anti-­‐Microbial  
(Anti-­‐Viral  Agent)   •     Drugs  
  Mechanism  of  Action   Acyclovir  has  a  small,  but  reproducible  effect  against  
(ANTI-­‐MICROBIAL   • Acyclovir  is  converted  to  the  monophosphate  by   cytomegalovirus  (CMV)  which  can  cause  glandular  fever  
DRUG)   thymidine  kinase  –  the  virus  specific  form  of  this   in  adults,  or  severe  disease  e.g.  retinis,  resulting  in  
  enzyme  is  much  more  effective  at  carrying  out  the   blindness  in  individuals  with  AIDS.  
phosphorylation  than  the  host  cell’s  thymidine    
kinase.   Resistance  
• The  monophosphate-­‐form  is  subsequently   • Resistance  due  to  changes  in  the  viral  genes  
converted  to  triphosphate  by  the  host  cell  kinases.   coding  for  thymidine  kinase  or  DNA  polymerase  
  has  been  reported  
It  is  therefore  only  adequately  activated  in  infected  cells.   • Acyclovir-­‐resistant  herpes  simplex  virus  has  been  
  the  cause  of  pneumonia  and  encephalitis  in  
• Acyclovir  triphosphate  inhibits  viral  DNA-­‐ immunocompromised  patients.  
polymerase,  terminating  the  chain  reaction.    
• It  is  30  times  more  potent  against  the  herpes  virus   Pharmacokinetics  
enzyme  than  the  host  enzyme.   • Acyclovir  can  be  given  orally,  i.v.  or  topically  
  • When  given  orally,  only  about  20%  of  the  dose  is  
• Acyclovir  triphosphate  is  fairly  rapidly  broken  down   absorbed  
within  the  host  cells  by  cellular  phosphatases.   • Peak  plasma  concentrations  reached  in  1-­‐2  
  hours  
• i.v.  infusion  results  in  plasma  concentration  10-­‐  
to  20-­‐  fold  higher  
• The  drug  is  widely  distributed,  reaching  
concentrations  in  the  CSF  which  are  50%  of  
those  in  the  plasma  
 
Excretion  
• Excretion  is  in  the  kidneys,  partly  by  glomerular  
filtration  and  partly  by  tubular  secretion  
 
Side  Effects  
• Local  inflammation  can  occur  during  
intravenous  injection  if  there  is  extravasation  of  
the  solution  (as  it  is  very  alkaline)  
• Renal  dysfunction  has  been  reported  when  
acyclovir  is  given  intravenously  –  slow  infusion  
reduces  the  risk.  
• Nausea  
• Headache  
 
ZIDOVUDINE   Mechanism  of  Action   Pharmacokinetics  
(AZIDOTHYMIDINE,   • Zidovudine  is  an  analogue  of  thymidine   • Given  Orally   Anti-­‐Microbial  
AZT)   • In  retroviruses,  such  as  the  HIV  Virus,  it  is  an  active   • Bioavailability  of  zidovudine  is  60-­‐80%  due  to   Drugs  
  inhibitor  of  reverse  transcriptase  enzyme.   first  pass  metabolism  
(Anti-­‐Viral  Agent)     • Peak  plasma  concentration  occurs  at  30  minutes  
  • It  is  phosphorylated  by  cellular  enzymes  to  the   • It  can  also  be  given  intravenously  
(ANTI-­‐MICROBIAL   triphosphate  form,  which  competes  with  the    
DRUG)   equivalent  cellular  triphosphates  which  are   • There  is  little  plasma  protein  binding  so  there  
  essential  substrates  for  the  formation  of  proviral   are  no  drug  interactions  due  to  the  displacement  
DNA  by  viral  reverse  transcriptase  (viral  RNA-­‐ by  other  drugs.  
dependant  DNA  polymerase)     • Zidovudine  enters  mammalian  cells  by  passive  
• Its  incorporation  into  the  growing  viral  DNA  strand   diffusion  –  unlike  most  other  nucleotides  which  
results  in  chain  termination   require  active  uptake.  
   
• Mammalian  alpha  DNA  polymerase  is  relatively   • The  drug  passes  into  the  CSF  and  the  brain  
resistant  to  the  effect.  However,  gamma  DNA   • Most  of  the  drug  is  metabolised  to  inactive  
polymerase  in  the  host  cell  mitochondrion  is  fairly   glucoronide  in  the  liver  –  only  20%  of  the  active  
sensitive  to  the  compound,  and  this  may  be  the   form  being  excreted  in  the  urine.    
basis  of  unwanted  effects.    
   
Resistance   Uses:  
• In  most  patients  the  therapeutic  response  to   In  patients  with  AIDS  
zidovudine  wanes  with  long-­‐term  use,  particularly   • It  reduces  the  incidence  of  opportunistic  
in  late-­‐stage  disease.     infection  (such  as  Pneumocystis  Carnii  
• It  is  known  that  the  virus  develops  resistance  to  the   Pneumonia)  
drug  due  to  mutations  resulting  in  amino  acid   • Stabilises  weight  
substitutions  in  the  viral  reverse  transcriptase  and   • Reverses  HIV-­‐Associated  thrombocytopenia  
that  these  genetic  changes  accumulate   • Stabilises  HIV-­‐associated  dementia  
progressively.     • Reduces  viral  load  
   
• Thus,  the  virus  is  a  constantly  moving  target.     If  given  to  HIV  +ve  individuals  before  the  onset  of  AIDS  
• Resistant  strains  can  be  transferred  between   • In  combination  with  other  drugs,  can  
individuals.     dramatically  prolong  the  life  expectancy  
• Other  factors  which  could  underlie  the  loss  of    
efficacy  of  the  drug  are:   In  HIV  +ve  mothers  
o Decreased  activation  of  zidovudine  to  the   • Reduces  the  risk  of  transmission  of  the  virus  to  
triphosphate   the  foetus  by  66%  
o Increased  virus  load  due  to  reduction  in    
immune  mechanisms     In  subjects  who  have  been  accidentally  exposed  to  HIV  
Increased  virulence  of  the  pathogen   • E.g.  hospital  worker,  rape  victim,  condom  
problems  etc.  
 
Unwanted  Effects  
• Common  unwanted  effects:  
o Anaemia  
o Neutropenia  
• Uncommon  Effects:  
o GI  tract  disturbances  
o Skin  rash  
o Insomnia  
o Fever  
o Headache  
o Abnormalities  of  liver  function  
o Myopathy  (particularly)  
 
• Confusion,  anxiety,  depression,  and  a  flu-­‐like  
syndrome  also  reported.  
 
 
 

Potrebbero piacerti anche