Dzinos Medicine
Dzinos Medicine
Dzinos Medicine
2020
DZINO’S
MEDICINE
NYASHA DZINOTYIWEI
A compilation by E. Ziumbwa
Page 1 of 455
History taking
History taking is the collection of information from the patient during an examination. It is
the first thing that takes place when a patient comes for a consultation. The history is a 2-
direcitonal process in which a patient narrates the condition they are feeling while at the
same time the clinician is also enquiring about any other information. The patient’s words
should be preserved (although it may need to be paraphrased to medical language when
recorded by the clinician). Histories should be taken in the correct manner.
Histories are essential in making a diagnosis. The history is what the clinician uses to map his
course of examination & tests. An accurate history therefore suggests the correct diagnosis.
There are a number of components that constitute a good history, and there are other
components of a history that establish a good doctor-patient relationship.
Usually, to establish rapport, the doctor begins by introducing themselves and why they are
there. It is important to maintain a good air in the environment. If the patient is standing
you direct them to the chair and ask them to sit down. You then request the patient’s name
if you are meeting the patient for the first time. You should always show genuine interest in
the welfare of the patient – be attentive to what they are saying. You should also probe the
patient. This allows them to let out all their issues.
Presenting complaint
The presenting complaint is the symptom that the patient presents with that prompts the
patient to seek treatment. The best way to start this part of the conversation is, “what has
been the trouble recently?” The patient will then tell the doctor about their problem. During
the interview, you should not stop the patient. You should let the patient run through and
say everything they need to say, as they may end up mentioning things the doctor didn’t
expect.
It may be necessary to ask the patient questions to test diagnostic hypotheses. For example,
the patient may not have noticed that they experience chest pain each time they exercised
Page 2 of 455
(stable angina). It also helps to give the patient an option of answers to choose from or to
give them a scale. It allows the patient to answer a directed question but with the fullest
idea of what they are trying to say. A patient may be prompted to describe conditions that
they may not necessarily feel if one asks the patient a yes/no question. Furthermore, a
patient may change their descriptions after a more senior clinician asks them the questions.
Throughout the interview, you must prompt the patient to state the problem that bothers
them the most. The patient may feel uncomfortable if they do not state the true presenting
complaint. For example, maybe they have a violent cough, but their main problem is that
they are vomiting blood. You need to invite the patient to mention this condition of theirs.
As you ask the patient these questions, you need to ask yourself:
For each symptom that a patient presents with, there is a set of questions that the clinician
should ask. These questions can be represented by the acronym SOCRATES:
1. Site – where is the symptom? Some conditions cannot be localised. Only a general
picture can be given.
2. Onset – when did the symptom present and how rapidly did it present. Some
conditions such as cardiac arrhythmias have an instantaneous onset.
3. Character – what is the character of the symptom? Is it localised or diffuse? This
question prompts details of the symptom to as specific a description as possible.
4. Radiation. This mainly applies to pain. Sometimes pain spreads to other regions. The
clinician should ask if the pain being experienced is spread to surrounding regions.
Certain patterns of distribution are diagnostic, e.g. herpes zoster.
Page 3 of 455
5. Timing. Find out how the frequency of presentation of the symptom varies during
the day (how often it occurs). If the condition is long-standing it is often helpful to
ask the patient what prompted him to visit the clinician now.
6. Exacerbating & relieving factors. Ask the patient what makes the condition the
patient has better or worse.
7. Severity. Severity of the condition is subjective and it depends on the patient. A
patient’s appraisal of a symptom – especially pain – is relative. It is usually advised to
ask them to give a scale of the pain they are feeling out of 10. The numbers given will
not be universal to everyone, but they guide the clinician on what the patient is
experiencing. Other symptoms can be quantified as they can be measured. However,
the interpretation of the severity differs as one severity level for one person may not
affect the patient as much as another patient.
Systems review
The idea behind a systems review is to create a safety net. The patient may have forgotten
some things or deemed them irrelevant. If you ask them questions directed towards specific
systems, then it allows you to include or discard other possible conditions. The systems
enquired are: the cardiovascular system, respiratory system, GIT, genitourinary system, CNS,
musculoskeletal system and skin. Chronological sequence of events should also be noted,
especially when listing neurological symptoms. It may be important to list important
negative answers (e.g. if a person has diarrhoea, you may want to take note of the fact that
they did not have a fever).
In order to establish a better relationship between the doctor and the patient, it may be
important to ask about the patient’s feelings about their condition, especially if it is a
chronic condition. It also allows the clinician to decipher what else is needed to help the
patient – both medical and non-medical. It shows genuine concern for the patient’s welfare.
Some patients who are chronically ill may have or may have had numerous medical
professionals supervising them. Chronic conditions include diabetes, hypertension, asthma,
Page 4 of 455
epilepsy. Also enquire about a history of TB, and if the patient was tested for HIV. The
clinician needs to find out what this supervision was and how often treatment was received.
You may be able to contact the doctor keeping the patient under surveillance.
Past drug history also includes medicine from the traditional healers. Under this heading,
information about allergies to drugs should be collected too. You should find out what the
reaction was in order to determine if the reaction was an allergic reactions or a side effect.
The clinician may also want to know if the patient takes drugs for recreational use, or if they
used to (it is important to emphasize confidentiality when asking such sensitive questions).
Family history
Asking the patient on their family history with respects to the presenting symptoms may
help pick out some familial forms of conditions. Many diseases are family-linked and most of
them are chronic non-communicable diseases. Drawing a family pedigree may help decipher
the possible condition. This is especially beneficial in this modern age of genetic
engineering.
Social history
A patient’s social life says a lot about what possible conditions they are exposed to. For
example, if a patient is a recreational drinker they may be predisposed to cirrhosis; domestic
violence may lead to a woman repeatedly reporting to the hospital with trauma.
• Upbringing and education level. Education level may explain how much you need to
explain the patient’s condition.
• Diet & exercise. An unbalanced diet and inadequate exercise may predispose the
patient to chronic disease like diabetes and hypertension. They may also cause
atherosclerotic plaques which would be risk factors against myocardial infarction and
stroke.
1
Some patients will not take medication if it changes shape & colour even if it is the same medication.
Page 5 of 455
• Marital status, social support and living conditions. Marital status & social support
is important because it lets the doctor know how strong their relationships at home
are. Knowing these is important in the case of someone suffering from a debilitating
condition which requires family support.
• Travel history. This is a recap of any places the patient went to in the near past. If a
patient from a temperate region travels to a tropical region, he may return with a
highly infectious disease that he would not have encountered before.
• Substance use. The major drugs you need to look for are alcohol and tobacco. Ask
the patient how long they used to smoke and how many cigarettes they took every
day. You need to calculate the pack years to effectively quantify how much the
patient smoked. The pack years are calculated as follows:
𝑛𝑢𝑚𝑏𝑒𝑟 𝑜𝑓 𝑐𝑖𝑔𝑎𝑟𝑒𝑡𝑡𝑒𝑟𝑠 𝑝𝑒𝑟 𝑑𝑎𝑦 × 𝑛𝑢𝑚𝑏𝑒𝑟 𝑜𝑓 𝑦𝑒𝑎𝑟𝑠 𝑠𝑚𝑜𝑘𝑖𝑛𝑔
𝑝𝑎𝑐𝑘 𝑦𝑒𝑎𝑟𝑠 =
20
Alcohol quantity is represented in units, of which a single unit is 10ml of pure alcohol
taken. It is important to know how many units the patient takes in a week and how
many drink-free days the patient has. It is generally recommended that males take
no more than 21 units a week and females no more than 14 units a week, and this
week should have at least 2 drink-free days. A binge drink is described as taking
more than 3 units in a single day for females and more than 4 units for males. The
number of units is calculated as follows:
𝑛𝑢𝑚𝑏𝑒𝑟 𝑜𝑓 𝑢𝑛𝑖𝑡𝑠 = 𝑛𝑢𝑚𝑏𝑒𝑟 𝑜𝑓 𝑙𝑖𝑡𝑟𝑒𝑠 𝑡𝑎𝑘𝑒𝑛 × 𝑝𝑒𝑟𝑐𝑒𝑛𝑡𝑎𝑔𝑒 𝑎𝑙𝑐𝑜ℎ𝑜𝑙
Occupational history
Occupational history is particularly important for some conditions, such as respiratory
conditions that are linked to mining. Ask the following questions (abbreviated as WHACS)
Summarising history
In summarising the history, you list down your positive findings and your negative findings.
The positive findings are the symptoms that the patient presents with and they direct the
clinician towards the probable differentials. The negative findings are important in then
ruling out from the wide pool of differentials that the clinician has compiled.
Page 6 of 455
General examination
The general examination is conducted in order to provide a general description of general
symptoms that are not system specific. It is done to bring to light the presence of general
signs, e.g. anaemia, jaundice, clubbing, etc. The general examination is conducted with a
systematic approach. This systematic approach is followed because it ensures that the
examination is thorough and important aspects are not overlooked. As you learn
examination skills, experience and acquisition of new evidence-based data will help the
clinician to perfect their skills. In the general exam, it is important that a person points out
signs that are obvious to see. Symptoms should not be manufactured.
The most common examination technique carried out is the objective structured clinical
examination (OSCE). Other formats used include the long case and the short case. During
the examination, you may need to check vital signs such as pulse, blood pressure,
respiratory rate and temperature. These may be present on some automated machine, but
they are vital for examination. The most common sequence of examination of a system is as
follows:
1. Inspection. You should be able to observe the patient for any scars, abnormal
pigmentation and for certain movements. Everything should be noted and perhaps
said, as you may find relevance of certain paraphernalia later on during the
examination.
2. Palpation. You should feel for deep structures through the skin. Typical structures
palpated for are the lymph nodes, abdominal organs and growths & masses.
3. Percussion. You should percuss over certain areas for resonance and dullness.
Dullness is normally associated with fluid accumulation among many things.
4. Auscultation. Auscultation allows you to listen for different sounds produced by
different structures. Structures auscultated for include the heart, the lungs and the
abdominal viscera.
1. W – Wash your hands. Hands should be washed before and after the examination.
Good hand-washing techniques should be employed.
2. I – Introduce yourself. Introduce yourself to your patient. Let them know who you
are and what you want to do. You should stand on the right side of the bed as you do
so. You’re not allowed to stand on the left side.
Page 7 of 455
3. P – Permission: ask for permission. Ask them for permission to conduct the exam.
You should let them know that you intend to expose them. In the event that the
genitals are being examined, ask them if they need a chaperone to be present and if
they are comfortable with you touching them.
4. E – Expose your patient. You then expose the part you intend to examine. It may not
be necessary to expose every part of your patient. The patient should be exposed in
a secluded room, or there should be curtains lowered around the bed. This makes
the patient feel more comfortable when they are exposed.
5. R – Recline the bed rest. Depending on the examination, you may need to recline the
bed to various angles. The general exam does not require that a patient lie on a bed
– they can be seated in a chair.
Before you start touching the patient anywhere, you should ask them if they are feeling pain
anywhere, so that you don’t hurt them unwillingly.
After the patient has sat down, you should describe the patient’s appearance: age, stature,
race (this is sensitive) and gender (these days this is sensitive too). You comment on their
posture, state of their face (enlightened or dull), natural breathing and their consciousness.
In addition, you should be prepared to describe their immediate environment. You should
state whatever you see around them, be it catheters, food, empty cans or plastics, dirt,
sputum bowl, vomiting buckets, everything. To do this, you will need to go to the end of the
bed and look at them straight-on. This is the only part of the exam that you’re allowed to do
from any position other than the right side of the bed. This is because from the right side of
the bed you may not be able to see important features on the left side, such as catheters,
which are important to state.
Page 8 of 455
Hand & nail signs
The hand is the first thing to check when conducting a general examination. Examination of
the hands is by looking, feeling and moving. There is no subspecialty of internal medicine in
which examination of the hands is unrewarding. On the hands you check for:
• Shape of nails. The nails change shape during clubbing. To test for clubbing, you
slightly squeeze the nail by the lateral nail folds to bring the nail out. Alternatively,
you ask the patient to appose the dorsa of one set of corresponding fingers (e.g. the
index finger) to each other. The two fingers should form a small diamond-shaped gap
called Schamroth’s sign. This sign disappears when there is clubbing. Clubbing occurs
when megakaryocytes that are normally broken down in the lungs are not, resulting
in these cells becoming lodged in capillaries in the nail beds. Here, they release
growth factors (such as PDGF) and recruit cells & promote proliferation of muscle
cells and fibroblasts. This is what accounts for the increased nail bed bogginess this
develops in any process that disrupts normal pulmonary circulation. This is not the
only process believed to cause clubbing. It is also believed that
There are 5 grades of clubbing:
Grade 1 Nail bed bogginess
Grade 2 Disappearance of Schamroth’s window1
Grade 3 Increased nail curvature
Grade 4 Drumstick appearance of fingers
Grade 5 Osteoarthropathy (wrist involvement)
Clubbing is usually bilateral. Unilateral clubbing is rare, but possible. It can be caused
by: arteriovenous shunts (used for dialysis), bronchial arteriovenous aneurysm and
axillary artery aneurysm.
Clubbing may be familial and therefore physiological in 5-10% of individuals. The
causes of pathological clubbing are:
• Respiratory (70%). The respiratory causes of clubbing are: chronic suppurative
lung conditions (such as bronchiectasis, empyema, cystic fibrosis, lung abscess),
infections (e.g. tuberculosis and HIV), degenerative conditions (e.g. idiopathic
pulmonary fibrosis, asbestosis, sarcoidosis, hypertrophic pulmonary
osteoarthropathy) and neoplasms (lung carcinoma2, mesothelioma, pleural
fibroma, neurogenic diaphragmatic tumours and lymphomas).
• Cardiovascular. The cardiovascular causes are: cyanotic congenital heart disease
(e.g. tetralogy of Fallot, persistent truncus arteriosus), infective endocarditis,
arteriovenous shunts and aneurysms.
• Gastrointestinal. These include: cirrhosis, inflammatory bowel disease and
coeliac disease. Gastrointestinal causes are not very common.
1
This represents loss of the normal nail bed angle. The normal nail bed angle is less than 165°.
2
Small cell carcinoma usually doesn’t cause clubbing.
Page 9 of 455
• Others. These include thyrotoxicosis (causing thyroid acropatchy), pregnancy,
and secondary parathyroidism.
• Pallor of the hands. Pale hands could be an indicator of anaemia. It is often a
sensitive indicator of anaemia.
• Colour. Yellow discolorations on the fingers could indicate that a person has been
smoking for a long period of time. Furthermore, if a person is jaundiced they can
have the characteristic yellowish-orange. Nail colour can also be an indicator of
underlying conditions. Blue nails can be an indicator of cyanosis; red nails could
indicate polycythaemia.
• Size. In some endocrine disorders such as GH excess the hand increases in size.
• Gross distortions in architecture. A person may have muscle wasting in their hands or
a slight tremor, indicating some neurological disorder.
• Temperature. You feel for warmth in the patient’s hands. Absence of warmth may
indicate that blood is not flowing adequately through the patient’s arms. Warm arms
are characteristic of people suffering from chronic obstructive pulmonary disease, as
the increased partial pressure of carbon dioxide causes vasodilation which increases
the flow of blood.
• You can also use the dorsum of the hands to test for oedema.
In addition to all these, the clinician may check for the patient’s pulse. This is done using the
radial pulse. To feel for the radial pulse, you use the middle three fingers, as they are more
accurate in sensing for the pulse.
Neck
The neck is mainly palpated for lymph nodes. The lymph nodes are enlarged when they are
greater than 0.5cm in diameter. When examining lymph nodes, assess if they are attached
to deep structures or they are mobile (those attached to deep structures suggest
malignancy). Also assess their consistency: normal nodes feel soft. Rubbery nodes are found
in Hodgkin’s disease, matted nodes are found in TB and hard nodes are found in metastatic
disease. Some nodes may be tender, as in infectious mononucleosis, dental sepsis, tonsillitis
and other acute bacterial or viral infections.
There are 2 chains of lymph nodes in the neck – there is the anterior chain found in the
anterior triangle of the neck, and there is the posterior chain found in the posterior triangle.
There are also supraclavicular lymph nodes found in the root of the neck and the
submandibular & submental lymph nodes. The lymph nodes are usually swollen when there
is an infection. The axilla (axillary lymph nodes) is a common site of inflamed lymph nodes.
When doing the lymph node examination, first inspect the neck for any visible
lymphadenopathy. Palpate one side at a time bimanually in turn. Assess the lymph nodes
for site, size, consistency, mobility and tenderness, and compare lymph node on both sides.
Page 10 of 455
Cervical lymph nodes are examined from behind. The anterior chain is examined from the
back while the posterior chain is examined from the front. From the front you also examine
the posterior auricular & occipital nodes. Other lymph nodes to examine are the epi-
trochlear & inguinal lymph nodes.
Face
The face is a gateway into the way a person is feeling. When a person enters the doctor’s
room, you can tell what the patient is feeling just by looking at the patient’s face.
Furthermore, during the exam, you can tell if the patient is feeling discomfort when certain
procedures are done on them.
A specific diagnosis can be made by inspecting the face. However, other physical signs need
to be sought in order to complete the diagnosis. There are a number of abnormalities that
need to be looked for in the face:
• Jaundice. This is a yellow discoloration of the skin that is a result of an increased level
of serum bilirubin. Bilirubin is deposited in the skin of people with this elevated level
of bilirubin. Jaundice can be caused by a blockage of the biliary tree, leading to
retention of bilirubin in the circulation. It can also be caused by a large instantaneous
increase in the bilirubin levels, such as haemolysis or hepatitis. Jaundice is often
checked using the sclera. However, the sclera is usually visibly discoloured when the
condition is moderate to severe. Mild jaundice does not usually cause discoloration
because bilirubin is broken down by light and therefore the part of the eye that is
exposed to the sun is not yellow. One would need to lift the patient’s upper eyelid
and ask the patient to look down.
• Cyanosis. This is a condition in which the patient has a reduced oxygen tension in
circulation resulting in a bluish discolouration of the skin & mucous membranes.
Cyanosis can be central or peripheral. It only appears if the levels of deoxygenated
Page 11 of 455
blood rises above 5g/dL, which corresponds to a saturation level of <90%3. Cyanosis
does not occur in anaemic hypoxia because anaemic hypoxia is caused by a drop in
the concentration of haemoglobin in blood not the oxygen tension. There are 2
types of cyanosis:
o Central cyanosis occurs due to deoxygenated blood in the arterial circulation,
and the blue discoloration is seen in tissues with a good circulation such as
the mouth & tongue. The easiest way to check for central cyanosis is to check
the tongue. The presence of central cyanosis should prompt careful
examination of the cardiovascular & respiratory systems. Patients with
polycythaemia can be cyanosed at normal oxygen tensions.
o Peripheral cyanosis is found is tissues where there is reduced circulation. The
tissue then extracts more oxygen from the blood than normal, resulting in
the blood appearing bluer than normal. This is seen in the nails & lips on a
cold day. On a cold day, the tongue is usually spared.
• Pallor. Pallor can occur in the eyes of individuals with anaemia. Anaemia is
noticeable in the inner sclera when the anaemia is severe (<7g/dL). Normally, the
palpebral conjunctiva is nice & red, sharply contrasting the bulbar conjunctiva &
sclera. However, in anaemia, the palpebral conjunctiva is pale and whitish. Facial
pallor can occur in individuals with shock. The patients appear cold & clammy and
are hypotensive.
The waist-hip ratio also predicts health risk. Waist-hip ratio is measured by dividing the
circumference around the waist (the waist is found halfway between the costal margin and
the iliac crest) by the circumference around the hips. If the waist-hip ratio exceeds 1.0 in
men and 0.86 in women, the patients are at increased risk. Waist measurements can also be
conducted. If the waist is greater than 94cm in males and 80cm in females, the patient is at
increased risk while greatly increased risk is found in waists greater than 102cm in men and
88 in females.
The clinician should inspect the patient’s stature & posture, which may be hard if the
patient is lying in bed. You should inspect for limb deformities & missing limbs. You should
3
This depends, however, on the haemoglobin concentration. It occurs at lower saturations in anaemic people
and at higher saturations in people with polycythaemia.
4
This gives an estimate of the body’s surface area.
Page 12 of 455
also observe if the patient’s physique is consistent with the patient’s age. As the patient
walks in, you should examine the patient’s gait.
Hydration status
All doctors must be able to assess the patient’s hydration status, albeit being very difficult.
Excessive dehydration can cause death by acute renal failure. An overhydrated patient may
develop fluid overload and pulmonary oedema. Excessive rehydration with water leads to
reduced sodium levels which could lead to confusion and loss of consciousness (due to
cerebral oedema).
One can also assess for postural hypotension. You measure the patient’s blood pressure
while they are lying down. You then ask the patient to stand, and then measure their blood
pressure again after a minute. If the blood pressure has dropped, it means that the patient
has postural hypotension.
Oedema is another common sign that occurs. Oedema occurs when there is increased
interstitial fluid, and often occurs when there is fluid shift from either the vascular or
intracellular compartments to the interstitial compartments. Interstitial fluid is a result of
the balance between the hydrostatic pressure from the action of the heart and the colloid
oncotic pressure from plasma proteins. Any condition that increases hydrostatic pressure
(e.g. heart failure) or reduces colloid pressure (e.g. nephrotic syndrome & hepatic disease)
causes oedema. It can also be a result of increased fluid shift from the intracellular space, as
seen in malnutrition in children. Often oedema is pitting, and this may not be demonstrated
until body weight has increased by 10-15%.
• Fluid overload:
o Heart failure. This occurs through: renal under-perfusion stimulating the
RAAS system and increasing salt & water retention; renal under-perfusion
leading to increased salt & water reabsorption; and pulmonary oedema from
increased hydrostatic pressure when a patient lies flat.
Page 13 of 455
o Renal disease decreases GFR, thus increasing circulating volume and
therefore the hydrostatic pressure. Reduced GFR also increases tubular
reabsorption of sodium.
o Iatrogenic causes include rapid intravenous fluid replacement.
• Hypoalbuminaemia:
o Nephrotic syndrome. This is due to the massive proteinuria accompanying
the condition.
o Chronic liver disease. This results in reduced protein synthesis by the liver.
1. Start by greeting the patient, introducing yourself and then telling the patient what
you intend to do to them. You also ask for their permission to conduct the test.
Page 14 of 455
2. You then move to the end of the bed and comment on the general appearance of
the patient and the surroundings of the patient. This is called an end-of-the-bed
examination.
3. You then move back to the right side of the bed and check the patient’s hands. You
feel the temperature, check for pallor (anaemia), and examine the patient’s hands
for clubbing, discolorations (jaundice & cyanosis) and contractures.
4. You then palpate the patient’s neck and check for swollen lymph nodes. You must
check the anterior triangle lymph nodes and the posterior triangle lymph nodes.
5. You must then check the mouth & tongue for cyanosis.
6. You must then check the eyes to observe for jaundice.
7. Lastly, you check the feet and confirm if the patient is oedematous. You may want to
comment on the extent of the oedema. A sacral oedema test may help to make a
rough estimate of this assessment.
Page 15 of 455
Management of potassium
derangements
Potassium derangements are common complications of either disease or treatment. The
normal potassium levels in serum is 3.5-5.2mM, and this is the potassium concentration in
ECF. Potassium is an important anion in the body. Its extracellular concentration is
important for maintaining the transmembrane gradient in the body. This is important
particularly in excitable tissues, which can become highly excitable.
The distribution of potassium in the body depends on many factors:
1. The sodium-potassium pump. This is the main structure that maintains the
potassium & sodium gradients across the cell membrane.
2. The pH of plasma. Changes in plasma pH lead to changes in ECF potassium
concentration. Acidosis causes hyperkalaemia while alkalosis causes hypokalaemia.
3. Insulin. Insulin stimulates uptake of potassium by cells. Therefore, changes in insulin
levels influence potassium levels in the body.
4. Epinephrine. This hormone increase uptake of potassium through β1-receptors.
5. Glucose distribution. In patients with diabetes, the extracellular glucose
concentration rises, leading to rises in ECF osmolality. This leads to exit of water
from cells, and this raises the intracellular concentration of potassium. This rise in
concentration promotes exit of potassium ions.
6. RAAS (renin-angiotensin-aldosterone system). This system leads to increased
excretion of potassium in exchange for sodium retention. Aldosterone secretion is
also independently influenced by potassium levels; high levels stimulate its secretion
while low levels abolish it. This aims to maintain potassium levels at a homeostatic
level. However, because aldosterone is also under influence from body fluid volume
status, potassium levels are more likely to be deranged when anything happens to
the body.
7. Dietary intake. The normal dietary potassium intake is usually 80-150mM per day.
8. Extra-renal losses. These are mainly through the GIT. They can result from either
vomiting or diarrhoea. Only about 10% of dietary intake is lost in the GIT.
The major potassium derangements that occur in the body are hyperkalaemia and
hypokalaemia.
Hyperkalaemia
Hyperkalaemia is a condition in which serum potassium levels rise above 5.2mM.
hyperkalaemia is either a result of inappropriate release from cells or inability to excrete.
The causes of hyperkalaemia are:
Page 16 of 455
• Vascular. Transfusion of stored blood & massive blood transfusions can cause
hyperkalaemia.
• Inherited. Some patients have familial pseudo-hyperkalaemia.
• Trauma. Acute kidney injury causes hyperkalaemia. This is one of the commonest
causes. Rhabdomyolysis & tissue necrosis also cause hyperkalaemia due to
widespread release of potassium. Excessive exercise also causes hyperkalaemia.
• Autoimmune diseases.
• Metabolic disorders. Acidosis, diabetic ketoacidosis causes hyperkalaemia.
• Iatrogenic. Drugs can cause hyperkalaemia. They are also amongst the most causes.
A lot of drugs have been implicated: potassium-sparing drugs (e.g. amiloride,
spironolactone), ACE inhibitors, NSAIDs, heparin, cyclosporine.
• Neoplasms. Tumour lysis causes hyperkalaemia. Leukaemia, infectious
mononucleosis and
• Degenerative diseases. Addison’s disease and Gordon’s syndrome cause
hyperkalaemia.
Clinical features
A serum potassium level of above 7mM is a medical emergency.
• Concerning signs include: a fast irregular pulse, chest pain, palpitations and light-
headedness.
• Muscle weakness.
• Kusmaul respiration. This is associated with acidosis.
• Decreased cardiac excitability with eventual asystole (cardiac arrest).
• Hypotension & bradycardia.
• On ECG, you will see (in sequence) tall tented T-waves, reduced P-wave, widened
QRS complex and a sine-wave pattern just before cardiac arrest. The patient may
also be in ventricular fibrillation.
Treatment
The management of severe hyperkalaemia should be as follows:
1. Setting up ECG monitoring & IV access.
2. Protect myocardium. You use calcium gluconate to protect the myocardium from
sudden arrest. You give 10ml of 10% calcium gluconate solution over 5 minutes. The
dose can be repeated after 15 minutes, as the effect is temporary.
3. Drive potassium into cells. This you do in 2 ways.
a. You can use insulin 10IU with 50ml of 50% glucose given over 10-15 minutes.
You need to make regular checks of blood glucose & potassium.
b. You can also administer salbutamol instead. You can give it intravenously at a
dose of 0.5mg in 100ml of 5% dextrose over 15 minutes. This is rarely used,
however. You can also nebulise the salbutamol.
Page 17 of 455
To correct severe acidosis (<6.9), infuse 1.26% sodium bicarbonate instead of the
hypertonic type1.
4. Deplete body potassium. This is done later on over the next 24 hours. You give
polystyrene sulphonate resins, which bind potassium in the gut. They make use of
ion fluxes in the gut to remove potassium from the body and are the only way (save
for dialysis) to remove potassium from the body. They may, however, cause fluid
overload because they contain sodium, and they can cause hypercalcaemia due to
the calcium they contain. You give up to 15g 3-times daily with laxatives, or you can
give 30g followed by an enema.
5. Haemodialysis or peritoneal dialysis. This is done when the above measures fail. At
this point, the hyperkalaemia is said to be refractory. It is usually used in patients
with end-stage kidney disease ESKD.
Hypokalaemia
Hypokalaemia is produced by potassium levels below 3.5mM. The causes of hypokalaemia
are:
• Vascular.
• Infection. Severe diarrhoea can cause hypokalaemia.
• Trauma. The “traumatic” causes include:
o Release of urinary tract obstruction.
o Acute myocardial infarction.
• Autoimmune.
• Metabolic. Correction of megaloblastic anaemia can cause hypokalaemia. Dietary
deficiency also leads to hypokalaemia. Alkalosis also causes hypokalaemia.
• Iatrogenic. Iatrogenic causes include:
o Potassium-wasting diuretics lead to hypokalaemia. These include thiazides &
loop diuretics.
o Excess corticosteroid administration also leads to stimulation of
mineralocorticoid receptors, resulting in aldosterone-like effects.
o Nephrotoxic drugs such as aminoglycosides, amphotericin B and cytotoxic
drugs exert their effects by causing renal injury.
o Excessive insulin administration leads to hypokalaemia by increasing shunting
of potassium intracellularly.
o B-agonists such as salbutamol can also shunt potassium intracellularly.
o Other drugs include carbenoxolone and liquorice2.
o Administration of IV drugs without potassium, e.g. normal saline.
• Neoplastic. Neoplasms that cause hypokalaemia include:
o Acute leukaemia.
o Villous adenoma.
o ACTH-producing tumours.
1
The hypertonic type is associated with volume expansion.
2
Liquorice potentiates renal effects of cortisol.
Page 18 of 455
• Degenerative. Degenerative causes include:
o Hepatic failure.
o Heart failure.
o Kidney & adrenal disease. This includes nephrotic syndrome, Barter’s
syndrome and Conn’s disease. renal tubular acidosis (both types) and renal
tubular damage both cause hypokalaemia.
o Syndromes such as Liddle’s syndrome and Gitelman’s syndrome.
o Pyloric stenosis.
Clinical features
Hypokalaemia is usually asymptomatic, and is only symptomatic when it is severe (<2.5mM).
The clinical features of hypokalaemia are:
Management
The underlying cause should be identified and treated as soon as possible.
Acute hypokalaemia may correct spontaneously. You do the following:
1. Withdraw oral contraceptives & purgatives.
2. Administer oral potassium supplements as slow-release potassium or effervescent
potassium.
3. Intravenous potassium is indicated in conditions such as cardiac arrhythmias, muscle
weakness or severe diabetic ketoacidosis. Do not give more than 20mmol/hour3 and
no more concentrated than 40mmol/minute.
4. Serum magnesium levels must be measured when the hypokalaemia has not been
corrected. They should be measured & corrected.
When a patient has poor renal function, the replacement rate should be less than
2mM/hour. There is also need for hourly monitoring for ECG changes. Potassium ampoules
should be mixed with normal saline, as dextrose will make hypokalaemia worse.
3
You can give more provided that there is ECG monitoring.
Page 19 of 455
Meningitis
Meningitis is inflammation of the meninges, and this is usually a serious infection.
Organisms can reach the meninges in a variety of ways: by extension from the ears,
nasopharynx or sinuses, by direct inoculation from cranial injuries or congenital meningeal
defects, or by haematogenous spread from another primary source. Other non-infectious
causes of meningitis include: malignant meningitis, intrathecal drugs and blood in the
subarachnoid space.
Causative agents
The different meningitis syndromes are:
• Acute bacterial meningitis. This is often caused by acute bacterial infection. The
causative agents include: S. pneumoniae, H. influenzae, N. meningitidis, S. aureus,
group B streptococcal species, L. monocytogenes, Gram-negtaive bacilli (e.g. E. coli),
etc. The pia & arachnoid mater are usually congested with polymorphs and form a
layer of pus. This pus layer may organise and form adhesions, which can complicate
with hydrocephalus & cranial nerve palsies. Cerebral oedema can also develop.
• Tuberculous meningitis. Tuberculous meningitis usually causes chronic meningitis.
There is formation of a thick exudate at the base of the brain, which also results in
cranial nerve palsies. There are also numerous meningeal tubercles. Adhesions are
invariable. There is a predominance of lymphocytes in the CSF. Cerebral oedema is
also present.
• Viral meningitis. The viral causes of meningitis are: Enteroviruses (e.g. ECHO virus &
Coxsackie viruses), poliomyelitis, mumps, herpes simplex, HIV, West Nile virus and
EBV. There is predominantly a lymphocytic inflammatory reaction without pus
formation. There are also no polymorphs or adhesions and there is little or no
cerebral oedema (unless encephalitis develops).
• Fungi. Fungal infections often develop in immunocompromised individuals.
Cryptococcal meningitis is a common infection in HIV infected individuals. Other
causes include Candida, Coccidioides, Histoplasma and Blastomyces. There usually
give rise to chronic meningitis.
• Protozoa. Malaria can cause meningitis.
Risk factors
The risk factors for developing meningitis are:
Page 20 of 455
• Comorbid conditions. These include diabetes, chronic kidney disease, adrenal
insufficiency, hypoparathyroidism and cystic fibrosis. Alcoholism & cirrhosis are also
risk factors.
• Immunosuppression. HIV is a common risk factor in sub-Saharan Africa. Chronic
causes such as tuberculosis & fungal meningitis are common in this population.
• Splenectomy & sickle cell disease. This increases the likelihood of meningitis
secondary to encapsulated organisms. Hyposplenism predisposes to infections.
Thalassemia major also predisposes to meningitis.
• Crowding. This predisposes to meningitis from meningococcal infection, which often
spreads in outbreaks in such populations.
• Introduction of foreign bodies into CNS. Ventriculoperitoneal shunts predispose to
meningitis, as they can get infected. Dural defects caused by trauma or surgery can
also introduce infection.
• Pre-existing infection. Common infections that spread contiguously include rhinitis,
sinusitis, otitis media and mastoiditis. Other infections that predispose to meningitis
include Ludwig’s angina. Bacterial endocarditis also predisposes to meningitis. Any
other pre-existing infection elsewhere, e.g. pulmonary TB, can result in meningitis.
• Malignancy. L. monocytogenes is often found in this population.
Clinical presentation
The presentation of meningitis is as follows:
History
• Photophobia.
• Altered mental status.
• Nausea & vomiting.
• Malaise & rigors.
• Loss of consciousness. This occurs in complicated cases. Some patients can even
present in a coma.
• Seizures. They may be the sole presentation if the patient has been treated with
antibiotics before.
Page 21 of 455
• Travel history – malaria.
• Skull fracture, ear disease or congenital CNS lesion – S. pneumoniae.
• Concomitant otitis media or sinusitis.
• HIV – Cryptococcus or TB. For TB, also screen for symptoms, history of TB infection
and look for history of contact.
Examination
• Pyrexia.
• Nuchal rigidity.
• Altered mental status.
• Signs of extracranial infection.
• Infants: bulging fontanels, irritability, high-pitched cry and hypotonia.
• Lymphadenopathy may be present in TB.
• Cranial nerve palsies. The common nerves affected are CNIII, CNIV, CNVI and CNVII.
They are present in 10-20% of patients.
Complications of meningitis
These include:
• Septic shock. This can degenerate into DIC (which causes haemolytic anaemia).
Shock is common with bacterial infections. However, it can occur with fungal
infections as well, and this is usually more difficult to deal with.
• Seizures. These occur in 30% of adults and 40% of children.
• Cranial nerve dysfunctions. These include hearing impairment.
• Focal signs. This includes paralysis, and is due to cerebral ischemia which results
from vascular inflammation and thrombosis.
• Cerebral oedema. This is common with bacterial meningitis and is an important
cause of death.
• Hydrocephalus. This produces ventriculomegaly. Hydrocephalus can be
communicating or obstructive. Obstructive is due to exudates forming at the
foramina of Magendie & Luschka, while communicating hydrocephalus is due to
exudates forming over the arachnoid granulations.
Investigations
The investigations done for meningitis are:
Page 22 of 455
•
Lumbar puncture. This is indicated for most cases with fever, headache and neck
stiffness, as meningitis needs to be ruled out early1. In a lumbar puncture, you need
to take note of the opening pressure2 (which is directly proportional to morbidity &
mortality), take the 1st tube for biochemistry (glucose & protein), the 2nd for
haematology (white cell count with differential), the 3rd to microbiology &
immunology (for microscopy with Gram stain, ZN stain, India ink, culture [bacterial &
fungal] and Cryptococcal antigen [CrAg] testing) and the 4th tube is held for a repeat
cell count with differential if needed. The findings in the different types of meningitis
are as follows:
Opening White cell Glucose Protein Organisms
pressure count (mg/dL) (mg/dL)
(cm H2O) (cells/µL)
Normal 8-20 0-5 50-75 15-40 None
values lymphocytes
Bacterial 20-30 100-5000 Reduced Elevated (>100) Specific
meningitis (elevated) (very high) (<40) pathogen
Polymorphs identified in
60% of Gram
stains & 80%
of culture
Viral 9-20 10-300 Mostly Normal/slightly Viral isolation,
meningitis (normal) (elevated) normal. elevated. PCR
Lymphocytes Reduced
in LCM &
mumps
Tuberculous 18-30 100-500 (very Reduced Elevated (>100) AFBs, positive
meningitis (elevated) high) (<40) on culture &
Lymphocytes PCR.
Cryptococcal 18-30 10-200 Reduced 50-200 Positive on
meningitis (elevated) (elevated) (elevated) India ink, CrAg
Lymphocytes and culture
Aseptic 9-20 10-300 Normal Normal/slightly Negative on
meningitis (normal) (elevated) elevated workup
Lymphocytes
• Blood studies. An FBC is required, particularly in cases of acute bacterial meningitis.
In bacterial meningitis, there will be leukocytosis with left shift (increase in the
number of blasts). LFTs & U&Es are also required for assessing organ function and
adjusting doses to prevent toxicity. A coagulation profile & platelet count is indicated
1
Lumbar puncture should not be done if there is a suspicion of a space-occupying lesion
2
Normal opening pressure is 8-20cm H2O.
Page 23 of 455
for patients with chronic alcohol use or suspected DIC. Blood cultures should also be
done.
• Neuroimaging. These may show signs of meningeal enhancement, but are not
necessary for diagnosis of meningitis. They may be necessary, however, for
identification of complications such as hydrocephalus, cerebral oedema, infarcts,
brain abscesses and venous sinus thrombosis.
Management
Management needs to be prompt, as delay in management increases the risk of mortality.
Even with optimal management, mortality is about 15%. The management is as follows:
• Acute bacterial meningitis. While waiting for results to return, you should start
empirical treatment. You can start cefotaxime, benzylpenicillin or chloramphenicol
(generally not used). When meningococcal infection has been confirmed (either by
culture or by other clinical features), you give intravenous antibiotics immediately.
Lumbar puncture is unnecessary if clinical features suggest meningococcal infection,
and you can rely on blood cultures alone. In other types of acute bacterial
meningitis, lumbar puncture is indicated. However, if a mass lesion is suspected do a
CT scan first. In adults with pneumococcal meningitis, dexamethasone should be
given with the initial antibiotics. Both pneumococcus & Haemophilus respond to
cefotaxime/ceftriaxone.
• TB meningitis. Treatment with anti-TB drugs should be commenced. You give HRZ
for 2 months (initiation phase) followed by HR for 6-9 months (continuation phase).
Ethambutol has ocular complications. Adjuvant corticosteroids are now
recommended, and you give high dose corticosteroids (e.g. prednisolone 60mg for 3
weeks). Relapses & complications are common.
• Cryptococcal meningitis. This is treated in 3 phases. You start with an intensive
phase of amphotericin B (0.7mg/kg/day) with fluconazole (800mg orally once daily)
for 2 weeks [or fluconazole 1200mg orally daily for 2 weeks if amphotericin B is
contra-indicated], followed by a consolidation phase which consists of fluconazole
alone (800mg orally once daily) for 8 weeks, followed by a maintenance phase which
consists of fluconazole (200mg orally once daily) until CD4+ count is above
200cells/µl for 6 months.
Page 24 of 455
Guillain-Barré syndrome
Guillain-Barré Syndrome (GBS) is a collection of clinical syndromes that manifest as an acute
inflammatory polyradiculopathy. It is the most common acute polyneuropathy, with an
incidence of 3 per 100 000 population. It is a demyelinating disease, and is one cause of
acute flaccid paralysis. It has no racial predilection, but it is more common in males. It has a
bimodal distribution, being common at ages 15-35 and 50-75 years.
Pathophysiology
GBS develops as a result of molecular mimicry. It develops after a preceding infection. The
infections known to cause GBS include:
The infection is often trivial & unidentified, but the disease develops about 1-3 weeks
afterwards. As part of the immune response, the immune system produces antibodies to
liposaccharides on the infectious agents. It is thought that there is homology between these
liposaccharides and cerebral gangliosides, and these homologous molecules are attacked.
This leads to destruction of myelinating cells (predominantly Schwann cells), particularly
when GM1 is attacked.
Pathologic findings include lymphocytic infiltration of spinal roots and peripheral nerves,
followed by macrophage-mediated stripping of myelin. This leads to defective propagation
of nerve impulses (which typically becomes slower). In some patients with severe disease,
there is axonal disruption & loss.
Variants of GBS
The 4 main variants of GBS are:
1
Nearly 40% of patients are seropositive for C. jejuni.
Page 25 of 455
1
lead to disruption of the blood-brain barrier. of patients may actually have
3
hyperreflexia. The prognosis is often quite favourable, and recovery is rapid within
days.
• Acute motor-sensory axonal neuropathy (AMSAN). This is a severe acute illness that
also affects the sensory nerve roots. It also typically affects adults. In AMSAN, there
is usually marked muscle wasting, and recovery is poorer than with AMAN. It is also
associated with C. jejuni. There is marked axonal degeneration with little
demyelination.
• Miller-Fisher syndrome (MFS). This is observed in 5% of all cases of GBS. It consists
of a triad of: ataxia, areflexia and ophthalmoplegia2. Patients have mild limb
weakness, ptosis, facial palsy and/or bulbar palsy. Anti-GQ1b antibodies are
prominent in MFS, and these are found in CNIII, CNIV and CNVI.
• Pure sensory GBS. This is typified by sensory loss, sensory ataxia and areflexia in a
symmetrical widespread pattern. The prognosis is often good.
• Acute pan-autonomic neuropathy. This is the rarest variant. It involves the
sympathetic & parasympathetic systems. Patients develop postural hypotension,
bowel & bladder retention, anhidrosis, decreased salivation & lacrimation and
pupillary abnormalities. Cardiovascular involvement is common, with presence of
arrhythmia. Motor & sensory involvement is lacking. Recovery is gradual & often
incomplete.
Clinical features
The clinical features are as follows:
History
• Muscle weakness. The muscle weakness usually starts in the distal extremities, and it
is proximal muscle weakness. It ascends to affect more proximal limbs, and it is
symmetrical. Patients may be unable to stand or walk, especially when
ophthalmoparesis and/or impaired proprioception are present. Weakness develops
acutely and progresses over a few days.
• Cranial nerve involvement. This is seen in 45-75% of patients. It includes facial droop,
diplopias, dysarthria, dysphagia, and ophthalmoplegia & pupillary disturbances.
2
Acute onset of ophthalmoplegia is a cardinal feature.
Page 26 of 455
• Sensory changes. Most patients complain of paraesthesia or numbness. Paraesthesia
usually starts at the toes or fingertips and progresses upwards, but usually does not
pass the wrist or ankles.
• Pain. This is present in more than 50% of patients. The mechanism of pain is not well
understood. It is most severe in the shoulder girdle, back, buttocks and thighs, and it
is aching & throbbing in nature.
• Autonomic changes. These include tachycardia/bradycardia, facial flushing,
paroxysmal hypertension, orthostatic hypotension, anhidrosis and/or diaphoresis.
There may also be urinary retention & constipation, but these are rarely early or
persistent. Patients can also develop arrhythmias.
• Respiratory involvement. 40% of patients present with any of: dyspnoea on exertion,
shortness of breath, difficulty swallowing and slurred speech. This is due to
weakness of the diaphragm.
2
• Antecedent illness. Up to 3s of patients will remember an antecedent illness 1-3
weeks prior to their current symptoms. Often the illness is acute diarrhoea which
may have been bloody. Also find out about any recent vaccines.
Examination
Investigations
The diagnosis of GBS is often clinical, based on the history & examination. Investigations are
done to rule out other diagnoses and rule out other comorbidities. The investigations are:
1
• Blood tests. LFTs are elevated in as many as 3 of patients. SIADH may occur, leading
to true dilutional hyponatraemia.
• Peripheral neuropathy workup. These include: TFTs, rheumatologic profiles, vitamin
B12 & folate screen, HbA1c, ESR, RPR, serum protein and tests for heavy metals.
• Serum antibodies. These are indicated when the diagnosis of GBS is questionable, or
you want to identify a variant of GBS. Anti-GM1 antibodies are found in AIDS &
Page 27 of 455
AMAN and are closely associated with antecedent C. jejuni infection. Anti-GQ1b
antibodies are found in patients with MFS.
• Lumbar puncture. This will demonstrate increased protein to 1-3g/dL (a
phenomenon called albumin cytoplasmic dissociation) with a normal white cell
count.
• Nerve conduction studies. Abnormalities associated with GBS include: nerve
conduction slowing, prolongation of distal latencies, prolongation/absence of F-
waves, and conduction block/dispersion of responses. Changes should be seen in
more than 2 nerves (such as the arm & leg). You can also use electromyography
(EMG) to detect slowed conduction, and this is more specific.
• Pulmonary function tests. You need to measure the forced vital capacity (FVC) to
assess respiratory status and the need to ventilate. You use the 20-30-40 rule to
determine whether intubation is necessary:
o The FVC is less than 15-20ml/kg (or 1L).
o The maximum inspiratory pressure is less than 30cm H2O.
o The maximum expiratory pressure is less than 40cm H2O.
• Imaging. MRI can reveal nerve root enhancement.
Management
Patients diagnosed with GBS should be admitted into hospital for close monitoring until the
disease has reached plateau or is in reversal. This is because symptoms can rapidly progress.
1
of patients require admission into ICU because of respiratory failure.
3
Supportive management
Page 28 of 455
include frequent passive limb movements, gentle massage and frequent position
changes.
3. Cardiac monitoring. Dysautonomia can produce both hypertension & hypotension.
Hypertension can be treated using a β-blocker or nitroprusside. Hypotension can be
treated using intravenous fluid & supine positioning.
4. DVT prophylaxis. This is done by giving compression stockings and administering
LMW heparin. The compression stockings used are true gradient compression
stockings, which exert their highest pressure at the distal extremities (toes) and the
lowest at the most proximal part of the limb (thigh) to reduce the capacitance
volume of the lower limbs and prevent pooling of blood. They exert a pressure of 30-
40mm Hg. Standard white or TED stockings produce a maximum compression of
18mm Hg and are rarely fitted to provide adequate gradient compression. They are
therefore not advised.
5. Nutrition. Patients require feeding when on mechanical ventilation, whether enteral
or parenteral. This is to ensure that caloric intake is adequate when metabolic needs
are high. You should avoid aspiration pneumonia from developing.
6. Bowel & bladder management. This is usually transient, but is required to prevent
development of other complications.
7. Physiotherapy. About 40% of hospitalised patients require physiotherapy. The aim is
to reduce functional deficits and to target impairments & disabilities resulting from
GBS. Speech therapy helps promote speech and safe swallowing skills for patients
with significant oropharyngeal weakness.
8. Recreational therapy. Recreational activities are beneficial to a patient to promote
growth, development and independence of a long-term hospital patient.
Definitive management
• Intravenous immunoglobulins. You give 0.4g/kg per day for 5 days. It is an easy
intervention to implement. This is safe for use in children, but safety in pregnancy
has not been demonstrated. Intravenous immunoglobulins are also preferred in
haemodynamically unstable patients and in those unable to ambulate
independently.
• Plasma exchange. This is carried out over 10 days, and aims to remove auto-
antibodies, immune complexes and cytotoxic constituents from serum. This has been
shown to reduce recovery time by 50%. This has equal efficacy as intravenous
immunoglobulins.
• Immune adsorption. This is a new alternative treatment being tried.
Outcomes
Page 29 of 455
The spectrum of outcomes from GBS is wide, from complete recovery, to tetraplegia, to
death. In most cases, however, there is peak weakness in 10-14 days followed by recovery
within weeks to months. 80% walk independently at 6 months and 60% gain full motor
function within a year.
The mortality from GBS is 2-12%, and causes of death include ARDS, sepsis, pneumonia,
venous thromboembolic disease and cardiac arrest. Deaths often occur amongst those
patients requiring ventilator support. The leading cause of death amongst the elderly is
arrhythmia. Mortality rates increase with the age of the patient. 15-20% of patients develop
moderate residual deficits, while 1-10% of patients are severely disabled.
Page 30 of 455
Multiple sclerosis (MS)
Multiple sclerosis (MS) is an autoimmune neurological disorder of the CNS. It results in
formation of multiple plaques in the brain. Multiple sclerosis is a common disorder in
temperate regions. In the UK, the incidence is greater than 42 cases per 100 000 population,
and in South-East Scotland the incidence is greater than 200 cases per 100 000 population.
In the US, the incidence is 58-95 cases per 100 000 population. It is rarer amongst black
Africans and Asians. Adult migrants take their risks with them, while children acquire the
risks where they settle. The mean age of onset is 30 years, and it is 3-times more common in
females.
Pathophysiology
MS is an autoimmune disorder leading to inflammatory demyelination of the spinal cord.
The autoimmune process is T-cell mediated and it causes an inflammatory process within
the white matter of the brain & spinal cord. There may also be a humoral component, as
evidenced by presence of elevated IgG levels in the CSF of these patients. The exact cause of
MS is not understood.
One of the earliest processes in MS is the breakdown of the blood-brain barrier, leading to
crossing of leukocytes into neural tissue. These leukocytes acquire adhesive molecules that
allow them to cross this barrier. IL-12 is expressed early in MS lesions. There may also be
reduced activity of Treg-cells, which function in regulating the immune system.
MS produces numerous plaques within the white matter of the brain. These plaques can
form anywhere, but they have a predilection for the optic nerve, the periventricular region,
corpus callosum, brainstem & its cerebellar connections and the cervical spinal cord
(corticospinal tracts & posterior columns). In these lesions, there is destruction of
oligodendrocytes, myelin loss and reactive astrogliosis. In some patients there is aggressive
destruction of the axon. There is also infiltration of neural tissue with leukocytes
(predominantly lymphocytes & macrophages).
Aetiology
The exact cause of MS is unknown, but there are many risk factors:
Page 31 of 455
family member has MS is 7-times higher than in the general population. The only
chromosomal locus that is consistently associated with MS susceptibility is HLA-
DRB1.
• Age. Patients typically present at age 20-40 years. Presentation after 60 is rare,
although diagnosis may be delayed and occur years after initial symptoms.
• Sex. MS is 3-times more common amongst females than males. This ratio is
widening.
• Viral infection. It is believed that viruses may infect individuals, activate self-reactive
T-cells and initiate autoimmune processes. Viral reactivation within infected immune
& nervous cells may be responsible for acute attacks. Viruses associated with this are
EBV and HHV-6. Exposure to these infections during childhood, however, appears to
be protective due to the hygiene hypothesis.
• Vitamin D. There is evidence that MS is more common amongst patients with low
vitamin D and little sunlight exposure.
• Geographic aviation. MS is predominantly a disease of temperate climates. The
incidence is higher in Europe & America and low in Africa & Asia. This, however, may
be related to vitamin D exposure.
Clinical patterns
The clinical patterns of MS are:
• Optic neuritis. Optic neuritis occurs in about 40% of patients. 20% present with optic
neuritis as a first presentation.
• Spinal MS. 55-75% of patients develop spinal cord lesions at some point in their
lives. It is often associated with concomitant brain lesions. Spinal MS has a
Page 32 of 455
predilection for the cervical cord, and it often involves the dorsal & lateral tracts
abutting the subarachnoid space.
• Myelocortical MS. This is a new variant of MS marked by demyelination of the spinal
cord & cerebral cortex, but excluding the cerebral white matter. In these patients,
neurodegeneration can be independent of demyelination.
• Brainstem MS. A relapse affecting the brainstem contains combinations of diplopia,
vertigo, facial numbness, dysarthria and dysphagia. Bilateral inter-nuclear
ophthalmoplegia is pathognomonic of MS.
Clinical presentation
The clinical presentation is as follows:
History
Patients are usually monosymptomatic: they have single complaints. The presenting
features:
• Optic neuritis: pain in eye movement, rapid deterioration in central vision, diplopia
on lateral gaze, hemianopia.
• Sensory spinal cord symptoms: paraesthesia, loss of vibration & proprioception
(leading to useless hand/leg).
• Motor spinal cord symptoms: leg weakness, unsteadiness, falls
• Autonomic spinal cord symptoms: bladder hyperreflexia (frequency & urgency),
constipation, sexual dysfunction, temperature sensitivity1.
• Other symptoms: fatigue, depression, neuropathic pain (particularly in the face),
swallowing difficulties, seizures,
• Higher functions: reduced attention span, concentration, memory and judgement.
Examination
On examination:
1
Worsening of symptoms after an increase in temperature (e.g. hot bath or exercise) is called Uhthoff’s
phenomenon.
Page 33 of 455
findings include anterior uveitis, vitreitis, vascular sheathing, disc & papillary
haemorrhage and compromise of central arterial & venous circulations.
Investigations
These are supposed to support the diagnosis of MS by providing evidence of dissemination
in time & space. The investigations done in MS are:
• MRI scans. An MRI scan of the brain & spinal cord has high sensitivity but low
specificity for MS lesions in the brain. MRI demonstrates areas of demyelination.
Typical lesions are oval in shape, about 2cm in diameter and often perpendicular to
the lateral ventricles. Acute lesions show gadolinium enhancement for 6-8 weeks. Its
low specificity is due to the differentiating MS plaques from small ischemic lesions
(in the elderly) and other non-inflammatory lesions (in young patients) such as
sarcoidosis, Behçet’s syndrome and vasculitis. There are fewer inflammatory lesions
in the spinal cord that mimic MS, so a spinal MRI is more specific. Things to look for
on MRI include:
o Dissemination of lesions in space, i.e. different anatomical locations.
o Dissemination of lesions in time, i.e. different ages.
• CSF examination. CSF examination is often unnecessary because of the usefulness of
MRI in diagnosis. This shows oligoclonal IgG antibodies in over 90% of cases.
However, these are not specific for MS. The CSF cell count may be elevated.
• Evoked responses. These may help demonstrate clinically silent lesions, such as
visual evoked responses in the optic nerve.
• Blood tests. These are used to exclude other inflammatory lesions, such as
sarcoidosis, SLE and other causes of para-paresis such as vitamin B12 deficiency, HIV,
HTLV-1 and adreno-leukodystrophy. An FBC may show infections, such as Lyme
disease. also screen for thyroid diseases.
Diagnosis of MS
Diagnosis of MS is a clinical one, although clinically you cannot diagnose MS based on a
single episode of symptoms. To make a diagnosis from a single episode, you need to be
supported by evidence from investigations. The diagnosis requires 2 or more episodes
attacking different parts of the CNS.
You can make use of the McDonald criteria for diagnosing MS. The criteria are as follows:
Page 34 of 455
≥2 attacks with objective clinical evidence MRI required to demonstrate dissemination in
of 1 lesion. space
OR
nd
Await 2 clinical attack suggesting different
site.
1 attack with objective clinical evidence MRI to determine dissemination in time
of ≥2 lesions. OR
nd
Wait for 2 clinical attack.
OR
Demonstration of CSF-specific oligoclonal bands
1 attack with objective clinical evidence Dissemination in space demonstrated by MRI
of 1 lesion (also called clinically isolated OR
syndrome. Await a 2nd clinical attack implicating 2nd CNS
site AND dissemination in time demonstrated
by MRI or 2nd attack.
Insidious neurologic progression One year of disease progression and
suggestive of MS dissemination in space demonstrated by:
• One or more T2 lesions in regions
characteristic of MS.
• 2 or more T2 focal lesions in the spinal
cord.
• Positive CSF.
An attack is a neurological disturbance that lasts at least 24 hours. For 2 attacks to be
separate, there needs to be at least 30 days between the onset of 1 attack and the onset of
the other.
Management of MS
There is no cure for MS, so treatment is aimed at minimising progression, preventing
complications and improving the quality of life. Treatment is divided into immunotherapy
for the underlying immune disorder, and supportive therapy to relieve symptoms.
In the acute setting, the main aim is to abolish the immediate inflammatory process and
achieve remission. you need to stabilise acute life-threatening conditions, initiate supportive
care & seizure prophylaxis and monitor for increasing intracranial pressure. The
management of acute relapses includes:
Page 35 of 455
or high-dose oral corticosteroids. You can also use plasma exchange if steroids are
contraindicated or they are ineffective.
2. Identification & control of known precipitants. This includes: aggressive control of
infections, normalising body temperature (using antipyretic agents) in patients with
fever, and providing urine drainage & skin care.
Long-term management of MS
Management of symptoms
• Depression. This is managed using standard depression management. You can use
SSRIs.
• Fatigue. Fatigue is one of the most common complaints. You can use amantadine
100mg orally twice-daily. You can also try methylphenidate and fluoxetine. Non-
pharmacologic interventions include energy conservation, work simplification,
scheduled rest periods and use of cooler garments.
• Pain management. This may be primary or secondary. Primary pain is due to
neuronal dysfunction secondary to the demyelination process. This often produces a
2
Mitoxantrone has a lot of side effects, such as cardiotoxicity and AML.
Page 36 of 455
burning or shooting pain. It is treated by TCAs as 1st-line agents, with carbamazepine,
phenytoin and gabapentin are used as 2nd-line agents. Secondary pain is
musculoskeletal in nature, primarily due to poor posture, poor balance or abnormal
use of muscles or joints.
• Heat intolerance. This can be managed by lifestyle changes. They include: placing
activities during early morning & later afternoon to avoid heat of the day; use of air-
conditioning; avoid saunas, hot tubs and hot shower baths; avoid excessive humidity;
and treat fevers.
• Spasticity. This can be done pharmacologically & non-pharmacologically. Non-
pharmacologic management is through physiotherapy. Pharmacologic management
includes use of baclofen, benzodiazepines, dantrolene, gabapentin, tizadine and
intramuscular botulinum toxin.
Page 37 of 455
Neuromuscular junction
disorders
Neuromuscular junction disorders constitute a type of myopathy that is the result of
pathology at the neuromuscular junction. Examples of neuromuscular junction disorders
include myasthenia gravis, Lambert-Eaton syndrome and botulism.
Myasthenia gravis
Myasthenia gravis is an autoimmune neuromuscular junction disorder that is the result of
autoantibodies against the acetylcholine receptor. The prevalence of myasthenia gravis is
about 4 in 100 000. Under 50 years of age, it is twice more common in females than in
males1, and it has a peak age incidence of around 30 years.
Pathogenesis
1
In males, the opposite is true.
Page 38 of 455
• Anti-AChR antibodies. In 70-80%, the disease is caused by autoantibodies against
the nicotinic ACh receptor. Immune complexes of anti-ACh IgG and complement are
deposited on the postsynaptic membrane. These cause interference with and later
destruction of the ACh receptors. However, the plasma concentration of antibodies
does not necessarily correlate with the severity of the disease. Changes in antibody
levels can be used to monitor treatment efficiency, nonetheless.
• Anti-MuSK antibodies. A second group of antibodies has been identified which react
against muscle-specific receptor tyrosine kinase (anti-MuSK antibodies). These
antibodies have been detected in 40-70% of patients that are seronegative to the
anti-AChR antibodies. However, they are found in 10% of seropositive patients.
• Ocular muscle MG. Another group is the ocular muscle MG group. In 15% of
patients, myasthenia gravis is limited to ocular muscles alone.
The autoantibodies are thought to originate in the hyperplastic germinal centres in the
thymus. Most patients have thymic abnormalities: 70-80% of patients below 40 have thymic
hyperplasia and 10-12% have a thymoma (whose incidence increases with age). The role of
the thymoma in autoimmunity, however, is unknown. Thymic hyperplasia & thymoma is less
common in individuals with anti-AChR-negative disease. Young patients have an increased
association with HLA-B8 & -DR3.
Clinical features
Muscle weakness is the predominant feature of myasthenia gravis. Based on this, there are
2 types of myasthenia: generalised and ocular myasthenia gravis. Generalised myasthenia
gravis involves a range of muscle groups, while ocular myasthenia gravis only involves ocular
muscles. The involved muscle groups in myasthenia gravis are (in order):
• Extra-ocular muscles: ptosis (eyelid muscles, starts out bilateral but may become
unilateral during the course of the day, may switch from one eye to the other),
binocular diplopia (horizontal/vertical)2, weak eye movements (not limited to one
muscle). The pupils are always spared. 50% of patients present with ocular
symptoms.
• Bulbar muscles: fatigable chewing (weakness with prolonged chewing), dysarthria
(oral muscle weakness; palatal muscle weakness causes nasal and/or hypophonic
speech; worsens with prolonged speech), dysphagia (oral muscles, imminent risk of
aspiration leads to aspiration crisis), dysphonia (on counting to 50, the fades; rare
presentation). 15% of patients present with bulbar symptoms.
• Facial muscles: expressionless appearance (lost their smile/myasthenic snarl3 when
smiling; weakness of orbicularis oris muscle), peek sign (after brief opposition of
gentle sustained eye closure, the lids separate to show white sclerae)4.
2
This may start as episodes of blurred vision before the diplopia is apparent.
3
When the patient smiles, the midlip rises while the corners remain down.
Page 39 of 455
• Neck & limb muscles: dropped head syndrome (weight of head overcomes neck
extensors during the course of the day, leading to drooping of head; secondary neck
ache), proximal muscle weakness (arms more often affected than legs). Less than 5%
present with proximal weakness alone. Reflexes preserved, but are fatigable.
Wasting may occur after many years.
• Respiratory muscles: respiratory failure (myasthenia crisis) may occur
spontaneously, or may be precipitated. A respiratory infection is the most common
precipitant. Patients often develop increasing muscle (especially bulbar) weakness.
The warning features include:
o Dyspnoea that occurs/worsens when the patient lies supine.
o Severe dysphagia with difficulty clearing secretions.
o Signs of respiratory muscle weakness (e.g. hypophonia), pausing during
speech to take a breath, tachypnoea, use of accessory muscles of respiration,
and paradoxical abdominal breathing.
o Low baseline vital capacity (<30ml/kg ideal body weight), even if the patient
is not in distress.
Investigations
The investigations are done to confirm the clinical diagnosis. The investigations that can be
done include:
• Antibodies. You can test for & measure anti-AChR and anti-MuSK antibodies. These
antibodies are not found in healthy controls. In generalised myasthenia gravis, anti-
AChR antibodies are detected in 80-90% of cases. In ocular myasthenia gravis,
however, anti-AChR antibodies are found in less than 30%. Antibody titre levels,
however, correlated poorly with disease severity. However, changes in antibody
titres can be used to monitor response to treatment. There is a group of patients
who do not test positive for any of the antibodies; they are said to have seronegative
myasthenia gravis, and they constitute 6-12% of patients.
• Electrophysiological tests. These are an important supplement to immunological
assays. They include:
o Repetitive nerve stimulation (RNS). This is the most frequently used
electrodiagnostic test for myasthenia gravis. The test is performed by placing
an electrode at the endplate region of a muscle and stimulating the motor
nerve of the muscle. The nerve is stimulated 6-10 times at a low frequency
(2-3Hz). The compound muscle action potential amplitude produced after
4
Tests orbicularis oculi muscle.
Page 40 of 455
electrical stimulation is recorded. In normal muscle, the amplitude remains
the same with repetitive nerve stimulation. However, with myasthenia gravis,
the amplitude progressively decreases with the first 4-5 stimuli. An RNS study
is considered positive if there is a greater than 10% decline in amplitude. RNS
studies have a sensitivity of 75% in generalised MG and 50% in ocular MG.
The sensitivity is increased by 5-10% when an exercise protocol5 is used.
o Single fibre electromyography (SFEMG). This test is more technically
demanding. In SFEMG, a specialised needle electrode is inserted into the
muscle to simultaneously record the action potentials of 2 muscle fibres
innervated by the same motor axon. The special electrode has a 25µm
recording window and a low frequency filter set at 500Hz. The needle
electrode measures what is called a jitter, which is the variability in time of
the second action potential relative to the first. Any disorder that reduces the
safety factor of transmission at the neuromuscular junction will produce
increased jitter. Myasthenia gravis produces increased jitter. SFEMG has a
sensitivity of 85-95%. Its specificity is low, but it is highly suggestive of
myasthenia gravis when a standard needle EMG has no abnormalities.
• Imaging. Imaging of the mediastinum is important in evaluation of patients with
myasthenia gravis. You can do a chest CT or MRI to look for thymic abnormalities,
either hyperplasia or thymoma6.
• Beside tests. These are considered an extension of the neurologic examination
rather than lab tests. They include:
o Ice pack test. The ice pack test is used in patients with ptosis. Neuromuscular
transmission increases with cooling of muscle fibres, so when a closed eyelid
is cooled by a bag filled with ice blocks for 2 minutes, the ptosis improves by
more than 2mm.
o Tensilon® test. This test is seldom required and is used only in patients with
obvious ptosis & ophthalmoplegia. In this test, you inject edrophonium 10mg
intravenously (following a 1-2mg test dose). If there is substantial
improvement in weakness within seconds, the test is positive.
• Other antibodies. Other autonomic conditions are more common amongst patients
with myasthenia than those without. Autonomic thyroid disease is the commonest
abnormality (occurring in 3-8%). Screening for thyroid abnormalities is part of the
initial evaluation. Other conditions include SLE & rheumatoid arthritis. Antibodies to
striated muscle indicate the presence of thymoma.
• Spirometry. The forced vital capacity (FVC) & maximum inspiratory effort (MIF) are
the main parameters measured. These tests are measured when there is increasing
5
In an exercise protocol, the RNS study is carried out, then the patient is asked to exercise the muscle
maximally for 30-60 seconds and the study is retaken immediately afterwards. A smaller decrement in
amplitude is seen than that done at rest, reflecting post-exercise & post-activation facilitation.
6
Myasthenia gravis may be considered a paraneoplastic effect of thymoma.
Page 41 of 455
generalised muscle weakness (which may mask the respiratory weakness). FVC is
typically measured every 4 hours. These tests are done to detect impending
myasthenia crisis and are used to determine the need for elective intubation. These
should always be interpreted in context: patients with facial weakness who cannot
make a good seal with the mask may have a falsely low VC.
Management
There are 4 basic therapies for myasthenia gravis. The interventions done for myasthenia
include:
7
For this reason, high-dose corticosteroids are only started in hospitalised patients receiving concurrent
plasmapheresis for myasthenic crisis.
Page 42 of 455
They work quickly but are only short term (a couple of weeks). The therapies include:
o Plasmapheresis. This is plasma exchange, in which you replace the plasma of
the patient with plasma from a donor blood pack or with albumin. This works
by removing the plasma containing anti-ACh/anti-MuSK antibodies and
replacing it with plasma that does not have such antibodies. The beneficial
effects are seen within days but only last weeks. A typical treatment course
involves 5 exchanges over 7-14 days.
o Intravenous immunoglobulins. Intravenous immunoglobulins are pooled
immunoglobulins from many patients. The mechanism of action is uncertain.
The total dose is 2g/kg given over 2-5 days. Side effects include headache,
chills, dizziness and fluid retention, and these are related to the infusion rate.
Acute nephrotoxicity is a potential side effect.
• Surgery. Thymectomy is considered for patients on symptomatic treatment &
chronic immunotherapy because of the potential long-term benefits. Those with
thymomas have a clear need for surgery, but those with non-thymomatous tissue
have a less clear need. Nonetheless thymectomy is encouraged. Thymectomy is
encouraged as soon as the degree of weakness is sufficient to allow surgery. If
bulbar/respiratory symptoms are present, surgery is usually deferred until symptoms
resolve. If they persist, plasmapheresis or intravenous IG is administered 2 weeks
prior to surgery.
• Respiratory support. Elective intubation is done for patients that are developing
acutely-worsening symptoms. Succinylcholine is safe to use in myasthenia gravis, but
increased doses are required. Elective intubation is done if successive VC values are
lower than 20ml/kg of if MIF is less than -30cm H2O. after intubation, positive
pressure mechanical ventilation should be initiated. After intubation, anti-
cholinestrase therapy is temporarily hauled to prevent production of secretions.
Weaning from ventilation should only be initiated when plasmapheresis and/or
intravenous immunoglobulins have been initiated.
Pathogenesis
Page 43 of 455
the presynaptic membrane and release acetylcholine into the synaptic cleft. The
autoantibodies therefore block the action of these channels and this leads to reduced
release of acetylcholine. This causes muscle weakness.
From a series of intracellular muscle recordings, the following features were determined:
In patients with LEMS without small cell carcinoma, 64% have family members with
autoimmune thyroid disease or type 1 diabetes.
Clinical features
You can also do a respiratory examination to look for signs of lung cancer.
Investigations
The diagnosis of LEMS is made based on clinical findings and is supported by investigations.
These include:
8
This is called post-exercise/post-activation facilitation and is a feature of LEMS.
Page 44 of 455
antibodies). These are present in 85-95% of patients, and the test has high
specificity; the tests are not commonly found in other autoimmune diseases.
Nonetheless, the test has also been detected in individuals with other neurologic
paraneoplastic disorders, those with cancer without signs & symptoms and those
with ALS (amyotopic lateral sclerosis). Therefore, the test is not diagnostic in the
absence of clinical signs & symptoms.
• Neurophysiologic testing. Most patients have a characteristic electrophysiological
pattern typical of presynaptic neuromuscular disorders (as opposed to the
postsynaptic neuromuscular disorder of myasthenia gravis).
o Repetitive nerve stimulation. The CMAP of resting muscle in LEMS has
significantly lower baseline amplitude than in normal individuals. Following
high-frequency (>10Hz) RNS or brief maximum isometric contraction, there is
a significant increment in the amplitude (incremental response9).
o Electromyography (EMG). On standard EMG, the motor unit action potentials
are unstable. SFEMG has significant jitter & transmission blocking that is
often improved at higher firing rates.
• Chest X-ray. This can be done to look for lung malignancies. You can also do a chest
CT since it has better resolution.
Treatment
Page 45 of 455
• Immunologic therapies. Immunologic therapies can be used when there is
progressive weakness and a limited response to symptomatic therapies. The agents
include:
o Intravenous immunoglobulin. The mechanism of action of intravenous
immunoglobulin is unknown. You give 1g/kg per day for 2 days. The benefits
of this intervention peak at 2-4 weeks, and their half-life is about 1 month.
Maintenance therapy is achieved with repeat therapy at 4-12 weeks
intervals.
o Immunotherapy. You can use oral prednisolone (1mg/kg per day) ±
azathioprine (corticosteroid-sparing agent).
o Plasmapheresis. LEMS patients, however, do not respond as rapidly to
plasmapheresis as those with myasthenia gravis. Furthermore, the benefits
are short-lived, and therefore it needs to be repeated for it to work for long.
• Surgery. If the small cell carcinoma is small enough, it can be excised surgically.
Page 46 of 455
Stroke
A stroke is a focal neurological deficit that arises from a cerebrovascular origin and lasts for
24 hours or longer, or leads to death. It therefore differs from a transient ischaemic attack
(TIA), which is one that lasts a few hours or minutes and may herald a stroke. The arbitrary
of lasting less than 24 hours is no longer used. Stroke is the 3rd most common cause of death
in developed countries and it is the leading cause of adult disability. The mortality after a
person’s first stroke is about 12% in the United Kingdom.
Pathophysiology
In a stroke, the primary deficit comes from poor blood flow. Typically, the poor blood flow
results in death of brain neurons. Based on this, there are 2 kinds of strokes:
• Ischemic strokes. These are strokes that result from reduced blood flow to part of
the brain. This results first in ischemia followed by infarction of the brain in that
area. Ischaemic strokes account for 80% of all strokes.
• Haemorrhagic strokes. These are strokes that result from bleeding into the brain.
There are 3 kinds of haemorrhagic stroke: there are intra-parenchymal
haemorrhage, subarachnoid haemorrhage and intra-ventricular haemorrhage.
Epidural & subdural haemorrhages are not included in haemorrhagic strokes.
Haemorrhagic strokes account for about 17% of all strokes, and of these 12% are
intracerebral (intra-ventricular & intra-parenchymal) while 5% are subarachnoid.
Other causes of stroke include venous sinus thrombosis, arterial dissection and vasculitis.
Aetiology
The aetiology of strokes depends on the type of stroke.
Ischemic strokes
Ischaemic strokes are a result of reduced blood flow to a section of the brain. This can occur
in 4 main ways:
• Thrombosis. Atheromas can develop in the arteries of the brain, resulting in gradual
occlusion of the blood vessels supplying the brain. Rupture of these atheromas
results in the initiation of the coagulation cascade, and this completely occludes the
blood vessels. The mechanism is similar to that which occurs in unstable angina &
NSTEMI myocardial infarctions.
• Embolism. Emboli that go to the cerebral circulation mainly come from the heart or
the arterial supply to the brain. Sources of emboli from the heart include: left
ventricular aneurysms, atrial fibrillation, atrial flutter, valvular heart diseases
Page 47 of 455
(rheumatic & degenerative), infective vegetations, mural thrombi, patent foramen
ovale (paradoxical embolism).
• Large & small artery disease. Large artery diseases involve the large arteries – the
aortic arch, common & internal carotid arteries and the vertebral arteries. Large
artery diseases include large artery stenosis, aortic, carotid or vertebral artery
dissection, large vessel vasculitides and other diseases. Large artery stenosis acts as a
source of emboli rather than as a source of occlusion. Small arteries include the
branches of the circle of Willis: the anterior, middle and posterior cerebral arteries
and the distal vertebral & basilar arteries. They also include the small penetrating
arteries. Small vessel diseases include lipohyalinosis (which occurs in hypertension
and causes thrombi), fibrinoid degeneration and microatheroma. Sickle cell anaemia
can also cause occlusion of blood vessels in the brain, leading to ischemia.
• Systemic hypoperfusion. This is most commonly due to heart failure, cardiac arrest
and arrhythmias. Myocardial infarction, pulmonary embolism and pericardial
effusion can also cause reduced cardiac output and therefore reduced perfusion to
the brain. The most vulnerable areas are watershed regions, which are the boundary
regions between areas supplied by the main branches of the circle of Willis.
Haemorrhagic stroke
Arterial dissection
These account for 1 in 5 strokes in individuals below 40. They can be a result of trivial neck
trauma or from hyperextension injuries, such as whiplash injuries, osteopathic
manipulation, hair wash in salons or exercise. The majority are in large extracranial vessels.
Blood may seep into the subintimal space, leading to formation of a false aneurysm.
However, thrombosis of this blood can lead to formation of potential embolic material that
1
These are also known as Charcot-Bouchard aneurysms.
Page 48 of 455
may embolise and cause ischemic stroke. Arterial dissection should be suspected by facial
pain, Horner’s syndrome or lower cranial nerve palsy.
Venous stroke
Venous strokes occur in 1% of all strokes. Thrombosis within intracranial venous sinuses or
cortical veins leads to congestion and build-up of pressure within the cerebral circulation.
This leads to cerebral oedema and numerous petechial haemorrhages which coalesce to
form haematomas. Sinus thrombosis leads to reduced reabsorption of CSF, which leads
raised intracranial pressure. There is, however, no difference in the pressure between
various parts of the brain and therefore no hydrocephalus.
Pathophysiology
The pathophysiology of strokes depends on the type of stroke in question.
Ischemic strokes
Ischaemic strokes occur because of a loss of blood supply to an area of the brain. This
initiates an ischemic cascade, which eventually leads to infarction. Brain tissue ceases to
Page 49 of 455
function if deprived of oxygen for 60-90 seconds, and after 3 hours will suffer irreversible
injury. When a blood vessel is blocked, a number of things happen:
1. The brain tissue supplied by the artery downstream from the blockage is deprived of
oxygen and resorts to anaerobic respiration. This leads to reduced ATP production as
well as build of lactic acid. Lactic acid is a cerebral irritant and disrupts the acid-base
balance in the brain.
2. A reduction in high-energy phosphate compounds such as ATP leads to reduced
activity of ion channels, most of which rely on the action of the Na +-K+ ATPase to
function. One major defect is the impaired reuptake of glutamate from neuronal
synapses. This leads to increased glutamate-mediated stimulation, leading to an
influx of calcium into neurons. The calcium influx leads to activation of autolytic
enzymes within cells, and it also causes mitochondrial failure & induction of
apoptosis.
3. Ischemia also causes production of oxygen free radicals. These react with various
cellular components and cause damage. They also mediate apoptosis through redox
signalling.
4. Ischemic injury can also lead to loss of structural integrity of brain tissue & blood
vessels. It leads to release of matrix metalloproteases that break down connective
tissue components such as collagen and hyaluronic acid. This leads to the breakdown
in the integrity of the blood-brain barrier and causes cerebral oedema. Cerebral
oedema also occurs because of impairment of the sodium-potassium pump leading
to an intracellular shift of sodium that causes absorption of water.
In ischaemic strokes, the area of damage follows the vascular territory of the blocked vessel.
For example, an occlusion of a middle cerebral artery will lead to ischemia of the lateral
outermost surface of the frontal, temporal and parietal lobes, leading to impairment of
movement, sensation, speech, neglect, etc. An occipital stroke may lead to homonymous
hemianopia. Generally, the affected regions can be divided into 2 regions: the ischemic core
and the ischaemic penumbra. The ischemic core is the main region that is deprived of blood
and undergoes the most extensive injury. The ischemic core can receive less than
10ml/100mg/minute. The cells in the core are expected to die within a minute of the onset
of the stroke. Zones of decreasing or marginal perfusion around the ischemic core are
known as the ischemic penumbra and receive a blood flow rate of less than
25ml/100mg/min. tissues in this region can remain viable for several hours because of
marginal tissue perfusion. Within hours to days of the stroke, however, there is activation of
genes encoding cytokines that mediate inflammation. These cause inflammatory injury and
further microvascular compromise. The ischaemic penumbra is therefore consumed by
these additional insults, resulting in the penumbra coalescing with the ischemic core.
Page 50 of 455
Ischemic strokes lead to death of brain cells. This is also accompanied by infarction of
astrocytes, oligodendrocytes and microglial cells. The infarcted tissue eventually undergoes
liquefactive necrosis and is removed by macrophages. This produces a cavity filled with CSF
that produces a radiolucent lesion on head CT.
Haemorrhagic strokes
Classification of strokes
Ischemic strokes are classified using the Bamford/Oxford classification. This classification is
purely a clinical classification and allows rapid categorisation of the patient. The subtypes of
stroke in accordance with this classification are:
• Total anterior circulation stroke (TACS). TACS is a stroke that involves occlusion of
both anterior & middle cerebral arteries, leading to infarction of zones supplied by
both vessels. All 3 of the following needs to be present for this diagnosis to be made:
o Unilateral weakness and/or sensory deficit of the face, arm and leg.
o Homonymous hemianopia.
o Higher cerebral dysfunction, e.g. dysphasia or visuospatial disorder.
• Partial anterior circulation stroke (PACS). PACS is a stroke in which only part of the
anterior circulation has been affected. Only 2 of the criteria above need to be
present for these effects to be seen.
• Posterior circulation stroke (POCS). This is a stroke involving occlusion of the
posterior part of the circle of Willis (such as brainstem & cerebellum). Only 1 of the
following need to be present for this diagnosis to be made:
Page 51 of 455
o Cranial nerve palsy & contralateral motor/sensory deficit (called Weber’s
syndrome if it involves cranial nerve III [oculomotor nerve]).
o Bilateral motor/sensory deficit.
o Conjugate eye movement disorder (e.g. horizontal gaze palsy).
o Cerebellar dysfunction (e.g. vertigo, nystagmus, ataxia).
o Isolated homonymous hemianopia (due to cortical blindness – called Anton’s
syndrome).
The syndromes that present as a posterior circulation stroke include
o Lateral medullary syndrome. This is caused by PICA (posterior inferior
cerebellar artery) thrombosis, vertebral artery thrombosis/dissection and
Wallenberg’s syndrome. Lateral medullary syndrome presents with acute
vertigo with cerebellar & other signs.
o Pseudo-bulbar palsy. This is a syndrome in which you have dysfunction of
cranial nerves IX to XII. It differs from a bulbar palsy in that a bulbar involves
the motor nuclei and is therefore a lower motor neuron lesion, whilst a
pseudo-bulbar palsy is an upper motor neuron lesion.
• Lacunar syndrome (LACS). This is a stroke that occurs subcortically and is secondary
to small vessel disease (in the lenticulostriate vessels). There is no loss of higher
cortical function, such as speech. The features of a LACS are:
o Purely sensory stroke.
o Purely motor stroke.
o Sensorimotor stroke.
o Ataxia hemiparesis.
Lacunar strokes are common in hypertensive patients. They generally have a better
prognosis than MCA syndromes.
The most common pattern is occlusion of a branch of the middle cerebral artery, leading to
2
infarction in the internal capsule. This accounts for 90% of all infarcts and 3s of all strokes.
MCA syndromes have a bad prognosis.
Other stroke patterns that do not fit into the Bamford classification include:
Page 52 of 455
• Watershed/borderzone infarction. This occurs at the junctional regions between 2
vascular territories, and is due to prolonged periods of systemic hypoperfusion.
Common features are cortical visual loss, memory loss and intellectual impairment.
Clinical presentation
When a patient comes with what appears to be a stroke, you do the following:
2
Aphasia occurs when the dominant hemisphere is affected.
3
Arterial dissections can occur from trauma.
Page 53 of 455
consciousness by using the Glasgow Coma Scale (this is usually normal, but
may be depressed in venous sinus thrombosis).
o Neurological examination. This to establish for the presence of upper motor
neuron lesions. Strokes initially cause hypotonia & arreflexia, which then
progresses to hypertonia & hyperreflexia with an extensor response. You also
need to examine the cranial nerves, cerebellar function, gait, language and
mental status.
o Cardiovascular system. This comprises the following examinations: ocular
fundi for retinopathy & papilloedema, emboli and haemorrhage; the heart
for irregular rhythm, murmur and gallop; and peripheral vasculature
(palpation of carotid & femoral pulses and auscultation for carotid bruits4).
Also check the patient’s blood pressure for hypertension as well as testing
blood sugar to screen for diabetes.
After you have collected the clinical details from the history, & examination, you then
classify according to the Bamford classification of strokes.
• Investigations. The diagnosis of a stroke is actually a clinical one, based on: sudden
onset of facial weakness; sudden weakness of one or both arms; and difficulty
speaking or slurred speech (dysphasia). Nevertheless, investigations are done to
confirm clinical diagnosis, to distinguish between haemorrhage & ischemia, to look
for underlying causes that could direct therapy and to exclude other causes such as
tumours. The investigations to be done in stroke patients include:
o Blood pressure. The blood pressure on each arm should be measured. A
difference of greater than 20mm Hg should suggest subclavian stenosis.
Blood pressure measurement is important because the majority of stroke
patients have hypertension as an underlying risk factor, and hypertensive
patients may actually develop strokes as an initial presentation.
o Routine blood tests. FBC can be done to check for polycythemia,
thrombophilia and signs of infection.
o Other blood tests. These include coagulation profile, glucose level, electrolyte
levels, ESR, U&Es and LFTs.
o Imaging. CT & MRI scans are indicated during the acute phase of
management mainly to differentiate between ischemic and haemorrhagic
causes. On a CT scan, an infarct will appear as a darkened area, usually of
subcortical matter. These changes, however, are not seen initially in the
immediate period. The early changes, however, include loss of grey-white
differentiation and sulci effacement. A non-contrast CT can be used to
demonstrate a haemorrhage, which is normally whiter. A CT scan with
contrast may be used to check for any aneurysms being the source of the
bleeding. An MRI is more sensitive in detecting infarctions during the early
4
These are detected in carotid artery stenosis.
Page 54 of 455
stages. A diffusion-weighted MRI, however, is even more sensitive in
detecting ischemia, while being as accurate as CT in detecting haemorrhage.
Later on, you can employ vascular imaging modalities such as MR or CT
angiography and carotid Doppler/duplex scanning. MR/CT angiography is
valuable in determining surgically-accessible arterial stenosis (mainly internal
carotid stenosis). These imaging modalities are indicated mainly in patients
with TIA/strokes who are normotensive.
Complications of stroke
The complications of ischemic stroke are:
Differential diagnoses
The differentials for an anterior circulation stroke are:
• Cerebral aneurysms.
• Head injury.
• Hypoglycaemia.
• Intracranial haemorrhage.
• Malignancy, such as low-grade astrocytoma, meningioma or metastatic disease to
the brain.
• Subarachnoid haemorrhage & subdural haematoma.
• Viral encephalitis.
Management of stroke
Management of stroke is divided into the immediate management and long-term
management.
Immediate management
Page 55 of 455
The initial management is to prevent secondary brain injury and to prevent continued
worsening of symptoms.
The diagnosis of a stroke is made based on a quick history – the FAST acronym:
• F – Sudden facial weakness. This can be tested by asking the patient to smile. You
will see facial deviation.
• A – Sudden weakness of one or both arms and/or legs. This can be tested by arm
drift.
• S – Difficulty speaking or slurred speech.
• T – Timing: the sooner the treatment can be started, the better. Also establish the
last known well-time (i.e. the time just before symptoms occured).
1. Admit to multidisciplinary stroke unit. Check BP, pulse rate, respiratory rate,
temperature and random blood glucose level. If the patient is on hormone
replacement therapy, stop it. Also do a swallow test:
For a swallow test, you sit the patient up and you give them 5ml of fluid to drink. If
they do not choke, you give 15ml to drink. If they do not choke, they have passed the
test. If they choke on any of the 2 stages, they have failed the test.
2. Protect the airway. This is to prevent hypoxia. Ensure its patency and take the
oxygen saturation value using an oximeter. Give oxygen to the patient if SpO2 is less
than 94%, although you can consider it if the patient is not hypoxaemic.
3. Support the circulation. Make sure you establish intravenous access to the patient.
4. Blood glucose. Aim for a blood glucose of 4-11mM. If the patient is a diabetic patient,
give insulin per sliding scale. If the patient is hypoglycaemic, give dextrose.
5. Blood pressure. The blood pressure of patients who are candidates for fibrinolytic
therapy should be controlled, as they are at risk of intracerebral haemorrhage after
administration of tissue plasminogen activators. If the blood pressure is below
220
mm Hg, you can observe unless there is other end-organ damage. If the systolic
120
220
blood pressure is greater than 120mm Hg but the diastolic pressure is still below
140mm Hg, give labetalol 10-20mg iv over 1-2 minutes. You can repeat the dose
every 10 minutes until you achieve a 10-15% reduction in blood pressure. Do not
reduce blood pressure too rapidly as you can impair cerebral perfusion. If the
diastolic pressure is above 140mm Hg, give nitroprusside 0.5µg/kg per minute IV
infusion with continuous blood pressure monitoring.
6. ECG. This is done to look for arrhythmias for possible sources of emboli. This is
guided by the regularity of the pulse (an irregularly irregular pulse with a very high
Page 56 of 455
tachycardia is suggestive of atrial fibrillation). This should be done if immediately
available, otherwise it should not postpose imaging.
7. Immediate imaging. This is considered particularly if thrombolysis is considered. You
should always have a CT scan available, as it is faster & cheaper than MRI. Order a
non-contrast CT scan of the brain. Look for any bleeds or infarcts. If bleeds are
absent, start on thrombolysis.
8. Thrombolytic therapy. This is indicated ONLY if the patient has an ischemic stroke
AND they have no contraindications. Thrombolysis is considered if the last known
well-time is less than or equal to 4.5 hours from the time of intervention (albeit
alteplase being licensed within 3 hours of the last known well-time). Give a
recombinant tissue plasminogen activator such as alteplase 0.9mg/kg over 1 hour;
10% is given as bolus over 1 minute, and the rest is given over 60 minutes. Take a CT
scan 24 hours afterwards to rule out haemorrhage after administration. Alteplase is
contraindicated in: patients with major infarcts/haemorrhage on CT, mild/non-
disabling deficits, recent birth, surgery, trauma or artery/venous puncture at
uncompressible site, past CNS bleed, AV malformations or aneurysms, INR greater
than 1.7 (or patient on anticoagulants), thrombocytopaenia (less than 100 × 109
185
cells/L) and BP above 110mm Hg. In contraindicated patients, you can give high-dose
aspirin (300mg daily) or consider a flow-restoration device (such as the Solitaire) in
large vessel strokes. In large vessel occlusion, you can also consider mechanical
thrombectomy.
9. Anti-platelet therapy. This is only initiated when there is proven ischemic stroke. You
start with high-dose aspirin (300mg daily initially, followed by 75mg after several
days).
10. Haemorrhagic strokes. For haemorrhagic strokes, thrombolytic and anti-platelet
therapies are contraindicated. In these cases, definitive management is employed if
the clot is large. You can do urgent neurovascular evacuation if there is deepening
coma or there is coning. If a subarachnoid haemorrhage has occurred, treatment is
mainly supportive unless an aneurysm has been identified. For aneurysms, you place
platinum coils endovascularly.
Long-term management
• Antihypertensive therapy. Recognition & good control of high blood pressure is the
major primary & secondary control intervention. Transient hypertension usually
does not require treatment unless diastolic blood pressure rises above 100mm Hg.
• Antiplatelet therapy. You give long-term aspirin at 75mg daily. You can also give
clopidogrel (75mg) as an alternative to aspirin. If clopidogrel is contraindicated, you
can also give dipyridamole 200mg twice daily.
Page 57 of 455
• Anticoagulants. These are given if there is atrial fibrillation, paroxysmal arrhythmias,
cardiac valve lesions, cardiomyopathies or any other lesion that can throw emboli.
Prior to this, brain haemorrhage must be excluded through a CT scan. You can give
warfarin or heparin, but for long-term anticoagulation you give warfarin.
• Rehabilitation. This is of value during the first few weeks following a stroke to
relieve spasticity, prevent contractures and teach patients to use walking aids.
Baclofen or botulinum toxin is sometimes useful to relax the muscles. Speech &
language therapists have a vital understanding of aphasic patients’ problems &
frustrations. If swallowing is unsafe, you can place a nasogastric tube. Physiotherapy,
occupational therapy and speech therapy have a vital role in assessing & facilitating
the future care pathway. Patients require aids at home that make life easier.
• Anti-depressants. Stroke is particularly devastating to the patient, as it often leads to
loss of work, independence, finances and in some cases spouses.
Outcome
Strokes generally have a poor prognosis. About 25% of patients die within the first year,
with nearly 10% dying within the first month. The mortality is higher with haemorrhagic
strokes than with ischemic strokes. Poor outcome is likely when there is a coma, a defect in
conjugate gaze and hemiplegia. Recurrent strokes are also common, and many patients die
from subsequent myocardial infarction. Of the initial survivors, about 30-40% remain alive
after 3-4 years.
Of note, however, is the great deal of disability that this condition causes. Gradual
1
improvement usually follows, but 3 develop disability requiring institutional care, while
1
another 3 developing some milder form of disability.
Page 58 of 455
Respiratory history
The respiratory system consists of the upper airway, which is also called the conduction
system, and the lower airway, which is also called the respiratory airway. The lungs are the
respiratory structures in the lung. They contain alveoli through which respiration takes
place. In addition to the lungs, there are other supportive structures that allow breathing to
take place. The diaphragm & external intercostal muscles are inspiratory muscles that allow
the chest cavity to increase in capacity and allow air into the lungs.
There are a number of symptoms that serve as indicators of respiratory pathology. The
commonest presenting symptoms that should give a person indication of respiratory
pathology are cough, breathlessness, wheezing and chest pain.
Coughing
Coughing is a forced manoeuvre in which a person breathes against an initially closed glottis
and then rapidly opens it, causing a jolting action of anything obstructing the airway. There
are a number of factors that need to be known when the history is being taken:
Page 59 of 455
structures. Other solid material coughed up may include necrotic tumours
and inhaled foreign bodies.
A dry cough may be produced from reflux of oesophageal contents in GERD causing
acid irritation of the lungs. Dry cough may also be a sign of interstitial lung disease
and a side effect of use of ACE inhibitors. Sometimes, there is a pink frothy secretion
secreted in patients with pulmonary oedema that is confused with sputum.
• Haemoptysis. This is coughing up blood-stained sputum. It is an important sign of
lung cancer. Before haemoptysis is characterised, the doctor needs to determine
whether the blood was coughed up, vomited or suddenly appeared in the mouth
without coughing. If the blood tinge has puss in it, one must suggest that the
condition is infective. The condition must always be investigated. Haemoptysis varies
with amount of blood. Mild haemoptysis may be characterised by no more than
20ml blood in 24 hours. In other conditions, however, there may be more 250ml of
blood produced every 24 hours.
• Sound. Some conditions produce a cough with a characteristic sound. Patients with
severe asthma or chronic obstructive pulmonary disease have a wheezing cough. A
feeble non-explosive bovine cough with hoarseness may be indicative of lung cancer
that invaded the left recurrent laryngeal nerve. A moist cough suggests there are
secretions from the upper airways as a result of bronchial infection & bronchiectasis.
• Duration & frequency. The duration of a cough is important as it tells the clinician
how long the patient has endured the cough as well as how long it took to progress
to the level at which it worried the patient. An acute cough (occurred for only 3
weeks) can be a sign of pneumonia or acute bronchitis. A chronic cough (more than
8 weeks) can be a sign of asthma. The change in character of a chronic cough is
usually a result of the development of a new underlying condition and should be
taken seriously.
Breathlessness
This is also known as dyspnoea. It is the awareness that an abnormal amount of effort is
required for breathing. It can be a result of lack of physical fitness, anxiety and cardiac &
respiratory disease. Dyspnoea can be classified into 4 classes according to the New York
Heart Association (NYHA).
1. Class I. in this class of dyspnoea, there is disease but there is no dyspnoea felt at rest
or during physical work.
2. Class II. In this class of dyspnoea, there is dyspnoea present on moderate exertion.
3. Class III. In this class of dyspnoea, there is dyspnoea present on mild exertion.
4. Class IV. In this class of dyspnoea, there is dyspnoea felt at rest.
Usually it is also helpful to give the extent of exertion that produces dyspnoea.
Page 60 of 455
The nature of the dyspnoea determines what differential diagnosis can be given to it. In
interstitial lung disease, there is dyspnoea that progressively worsens. If dyspnoea is more
rapid in onset, it may be due to acute respiratory infection or pneumonitis. Dyspnoea that
varies from day-to-day and hour-to-hour suggests a diagnosis of asthma.
• If the patient is breathing fine in the upright seated position, but becomes breathless
when they lie in the supine position, the patient has orthopnoea/paroxysmal
nocturnal dyspnoea. It can be caused by left ventricular failure.
• If the patient is breathing okay while lying in bed but become restless when
standing. This is called platypnoea.
Wheezing
This is a high-pitched whistling sound produced by air passing through narrow airways. It is
heard maximally during expiration, although patients may refer to other inspiratory sounds
(rattling sounds from secretions; inspiratory sound of stridor) as wheezing. Asthma & COPD
can cause wheezing, just as infections like bronchiolitis and airway obstruction. Night
wheezing suggests asthma, but wheeze in the morning upon waking up suggests COPD.
Stridor is a sound that is similar to wheezing but is loudest over the trachea and is produced
upon inspiration. It is a sign of obstruction of the upper airway.
Chest pain
Chest pain due to respiratory disease is usually different from that due to myocardial
ischaemia. There are different types of chest pain.
• Pleural pain. Pleural pain is characteristically pleuritic in nature: it is sharp and made
worse by deep inspiration. It may be of sudden onset in patients with lobar
pneumonia, pulmonary embolism & infarction or pneumothorax. It is often
associated with dyspnoea, and if dyspnoea occurs together with chest pain the
patient must seek attention urgently. Common causes are pulmonary embolism,
pneumonia, pneumothorax and fractured ribs.
• Chest pain. This suggests respiratory, cardiac or musculoskeletal disease. Patients
with a chronic cough usually have a feeling of tightness in their chest but rarely
mention it when they come to the doctor. The source of pain depends on the
Page 61 of 455
causative agent, and it usually involves irritation of the intercostal nerves.
Sometimes, the causative agent may involve other non-thoracic structures, such as
the arm (Pancoast’s tumour erodes the first rib and the brachial plexus, causing pain
along the medial side of the arm & Pancoast’s syndrome).
• Mediastinal pain. Mediastinal pain is central, retrosternal and unrelated to cough. It
is caused by irritant dust or infection of the tracheobronchial tree.
Past history
For respiratory history, as with every other history, you should take a past history of the
previous medical conditions that the patient has experienced. Typical conditions to ask
about are tuberculosis, pneumonia and chronic bronchitis. You should also enquire about
abnormal X-rays. Many lung investigations may have been performed on the patient, such
as biopsy, bronchoscopy and spirometry.
Occupational history
Occupational history is of no greater importance in any other system examination than in
the respiratory system. Occupational history is important because a lot of lung conditions
are chronic and they develop over time with long-term exposure to certain gas
environments. There are various occupations in which workers deal with poisonous fumes.
Asbestos is one such example, and almost every asbestos worker is going to suffer from
asbestosis. To make matters worse, workers’ wives who wash the asbestos workers’ clothes
after work also inhale asbestos and are likely to develop asbestosis too. Other dangerous
gases/fumes include coal, sulphur, silver, nitrogen dioxide and so on. Nonetheless, there has
been great effort to minimise such diseases. The most common occupational disease today
is asthma.
To find out about the occupational history, you need to ask the patient where he used to
work and what his occupation was. You also need to find out the duration of the job. You
may also want to ask if he smokes, as smoking has an additive effect on these fumes.
Family history
Some respiratory conditions are familial. A typical familial respiratory condition is cystic
fibrosis. Other examples include α1-atitrypsin deficiency, which causes emphysema. A family
history of infection with tuberculosis is also important.
Social history
The incidence of most respiratory conditions increases in smokers. The prevalence of COPD
and lung cancer is reflective of smoking patterns in the general population. An estimate of
the number of pack years that a person has smoked should be made. A pack year is when a
Page 62 of 455
person smokes one pack of cigarettes (1 pack = 20 cigarettes) per day for an entire year.
Most COPD patients have more than 20 pack years. Stopping smoking before the age of 40
is crucial to improving health. After 40, you lose on average 3 months of life per extra year
you continue smoking. Cigarette smoking may be worsened by the fact than someone works
in an asbestos mine.
Page 63 of 455
Respiratory examination
In a respiratory examination, you need to be aware of the surface anatomy of the various
respiratory organs so as to known what the various findings mean after examination. In
addition, one needs to have an appreciation of the systemic effects of respiratory problems.
This is why, in addition to the chest, one also examines the hands, arms, neck and face.
Before starting the respiratory exam, as with every other exam, one should start with the
WIPER sequence: wash your hands, introduce yourself, gain permission to conduct the
exam, expose the patient and then recline the patient to the appropriate angle. In a
respiratory exam, the patient should be undressed to the waist and the angle at which the
patient’s seat is reclined to 45°. Women should be covered by a towel, gown or some
clothing (bra) when the front of their chest is not being examined. The general sequence of
check points in an exam are that one starts with the hands, then moves on to the arms &
axillae, then to the neck, to the face and finally focuses on the system-specific region. In the
respiratory system, this specific region is the chest.
When you ask the patient to breathe in & out, ask them to do it through their mouth. This is
because if their nose is blocked, they will have difficulty breathing properly. Furthermore,
during auscultation, the sounds produced in the nose may be heard in the chest.
General inspection
The general inspection is done from the end of the bed. Upon inspection, one should start
off by commenting on the patient, so that the examiner knows that your focus is on the
patient. You should comment on the patient’s breathing – are they in distress, are their
nares flaring and is there tracheal tug? This gives a clue as to what possible conditions the
patient is suffering from. The signs to look for on the patient are:
• Dyspnoea. This is breathlessness. You should check if the patient is not breathless
• Breathing pattern. You assess the patient’s breathing pattern. The patient can have
periodic breathing, in which the patient has a cyclically increasing rate & depth of
breathing followed by a decreased respiratory effort & rate. This ends in a period of
apnoea or hypopnoea. This relates to altered sensitivity of the respiratory centres
and a delay in circulation between central chemoreceptors and the lungs. It is called
Cheyne-Stokes breathing. It can occur in normal elderly individuals, but it is frequent
in stroke involving the brainstem and in severe heart failure. Also check for
hyperventilation, which is a common response to anxiety or emotional stress. It
often leads to respiratory alkalosis and reduced carbon dioxide tension in blood.
Hyperventilating patients often complain of an inability to fill their lungs.
Hyperventilation can also be associated with deep sighing respirations (this is called
Page 64 of 455
Küssmaul respiration). This type of breathing is seen people suffering from diabetic
ketoacidosis, acute renal failure, lactic acidosis and salicylate & methanol poisoning.
• Other hints. Patients with COPD may have elevated shoulders with inspiration. This
aids chest expansion, and chronic breathing in this manner may lead to the
development of a barrel-shaped chest. Some patients have pursed lips, which is also
indicative of COPD.
• Use of accessory muscles. The accessory muscles of respiration are the
sternocleidomastoid, platysma, pectoral muscles and the strap muscles of the neck.
These muscles are used for forced breathing and use of accessory muscles is usually
a sign of COPD or severe asthma. Most women make use of their intercostal muscles
more than their diaphragm and therefore their breathing is predominantly thoracic
while men use their diaphragm and therefore their breathing is predominantly
abdominal.
• Stridor. This is a harsh rasping and croaking sound produced on inspiration. This is
often aggravated by coughing and should always be investigated. It is caused by
foreign bodies in the airway as well as tumours. You ask the patient to cough, and
then take a deep breath in & out, and then you listen for the noise.
• Hoarseness. This may be due to damage to the left recurrent laryngeal nerve. This
can be caused by a Pancoast tumour. The left vocal cord cannot adduct to the
midline and therefore a prolonged low-pitched bovine-sounding cough is produced.
• Blood pressure. A diastolic pressure below 60mm Hg is associated with increased
mortality in patients with community acquired pneumonia. In pneumothorax,
hypotension indicates tension with reduced venous return. This raises the risk of
cardiac arrest.
• Skin appearance. Erythema nodosum may indicate acute sarcoidosis. Some skin
nodules may signify metastasised lung cancer.
• Examination of sputum. One should observe the colour, volume and type of sputum.
Furthermore, one should also analyse for the presence of blood streaks in the
sputum of it.
Hands
You then return to the right side of the bed to conduct the hand examination. You ask the
patient to raise their hands to observe for signs. There are a number of signs to look for on
the hand which are relevant to the respiratory examination:
Page 65 of 455
hypertrophic pulmonary osteoarthropathy (HPO). This condition is often a result of
lung carcinoma, and there is pain & swelling of the ankles & wrists due to periosteal
inflammation. It is important to note that clubbing does NOT occur with COPD. To
test for clubbing, you place your thumbs under the pulp of the terminal phalanx and
then attempt to move the nail over the nail bed. If there is a spongy feel in the
movement of the nail, then the nail is clubbed. It is often usually to test 2 fingers on
one hand and the other hand. This helps to validate the clubbing and also establish
that it is bilateral.
• Finger staining. Finger stains are found in cigarette smokers. The staining is caused
by tar, as nicotine is colourless. The amount staining does not depend on the
number of cigarettes smoked. Rather, it depends on the manner in which the
cigarette is held.
• Nails. The nails can develop a yellow stain, called yellow staining syndrome. It is
caused by exudative pleural effusion and lymphoedema. You can also check for
cyanosis in the nails, which will appear bluish.
• Wasting & weakness. Wasting & weakness is usually caused by a lung tumour
infiltrating the lower trunk of the T1 nerve root. This leads to wasting of the small
muscles of the hand.
• Tremor. A tremor is associated with the excessive use of β-agonists like theophylline.
• Asterixis. This is the downward flapping of the hands when they are extended. You
ask your patient to hold out their hands and extend their wrists. If the wrists flap,
then they have carbon dioxide retention. Alternatively, you can ask your patient to
squeeze your index or middle finger for 30-60 seconds. A person with asterixis
cannot do this for more than 30 seconds.
While still at the hand level, you can check the patient’s pulse. Here, you will not be testing
the patient’s pulse rate. Rather, you will be checking for a bounding pulse, which is present
in patients with severe carbon dioxide retention. Furthermore, you can also take this
opportunity to measure the respiratory rate. You count the patient’s breathing cycles over
30 seconds and then multiply the number by 2.
Neck
At the neck, you might need to ask the patient to sit up. Therefore, you may do this when
examining the patient’s back so that you avoid the inconvenience of repeatedly making the
patient sit up (this may waste your time if the patient takes time sitting up). However, you
do this after taking the jugular venous pulse level. The things to look for in the neck are:
• Jugular venous pressure. The jugular venous pressure can be checked while the
patient is still reclined. This is because the jugular venous pulse is sensitive to the
posture of the patient. The JVP is raised in patients with right-sided heart failure.
Also, chronic hypoxia in patients with COPD leads to pulmonary artery
Page 66 of 455
vasoconstriction, leading to pulmonary hypertension and heart failure. This is called
cor pulmonale1. JVP is also raised in patients with tension pneumothorax and severe
acute asthma. In superior vena caval obstruction, there is raised jugular venous
pressure that is not sensitive to the abdomino-jugular reflex. Most of such cases are
lung cancer that obstructs the superior vena cava. Other causes include lymphoma,
thymoma and mediastinal fibrosis.
• Neck lymph nodes. Lymph nodes may enlarge as a result of metastatic lung cancer,
and the first lymph nodes to sense this are the cervical nodes. Localised cervical
lymphadenopathy is a common presenting feature of lymphoma. To palpate the
neck lymph nodes, you ask the patient to sit up and you feel for the scalene nodes
above the first rib next to the insertion of the scalene anterior muscle. You also feel
for the anterior cervical chain going up, all the way to the submandibular,
submental, preauricular & post-auricular nodes. You also feel for the posterior lymph
nodes, which are along the anterior margin of the trapezius muscle. The neck lymph
nodes may have characteristic textures in different pathologies:
o In Hodgkin’s disease, they are rubbery.
o In dental sepsis & tonsillitis, they are tender.
o In TB and metastatic cancer, they are usually matted together to form a large
mass.
o Calcified lymph nodes feel stony.
o Palpable lymph nodes fixed to underlying structures are usually malignant.
• Trachea. The position of the trachea should be assessed, as it can serve as an
important indicator of chest pathologies. This examination is uncomfortable so you
must be gentle and tell the patient that this test is uncomfortable. You must test for
the position of the trachea in the neck just before it enters the chest; whether it is
deviated or not. You push your first finger into the suprasternal notch until the
trachea is felt. You must feel its middle. If the trachea is displaced, you will feel its
edge, and one space to the side of the trachea will have a larger paratracheal space
on one side than the other. Deviation of the trachea is characteristic of a number of
conditions:
o If the trachea deviates to the side of the lung lesion, the patient may have
upper lobe collapse, upper lobe fibrosis and pneumonectomy.
o If the trachea deviates away from the side of the lung lesion, the patient may
have tension pneumothorax or pleural effusion2.
o Upon palpation, one may feel a tracheal mass. This may be a retrosternal
goitre, a lymphoma or a lung cancer.
You also measure the crico-sternal length, which is the length between the lower
margin of the cricoid cartilage and the suprasternal notch. It is usually 3
1
Cor pulmonale is a condition in which there is a disorder/dysfunction of the right ventricle due to a problem
in the pulmonary system.
2
At this stage, the condition is usually advanced.
Page 67 of 455
fingerbreadths. If it is smaller than this, then suspect that there has been tracheal
tug. This sign is seen in COPD
Face
The face can be examined while the patient is lying on the bed. The things to look for are:
• Nose. You may want to inspect the inside & outside of the nose. You ask the patient
to tilt their head backward. You may need a nasal speculum & torch. You may find
polyps associated with asthma, a deviated septum associated with nasal blockage
and engorged turbinates which are associated with various allergic conditions.
• Tongue & mouth. You can look at the tongue to observe for central cyanosis (you
may be able to observe this around the mouth). A reddened pharynx & engorged
tonsils may indicate an upper respiratory tract infection. The engorged tonsils may or
may not have pus. You can also look for a reduced velopharyngeal lumen, which is
present in people with sleep apnoea.
• Facial skin. Look at the patient’s face. Smokers have red leathery wrinkled skin.
There may also be facial plethora and peripheral cyanosis if the superior vena cava is
blocked.
• Sinuses. Patients with sinusitis have tenderness on the skin overlying the sinuses.
This is excluded by normal trans-illumination.
• Eyes. Look for signs of Horner’s syndrome, which may be a sign of apical lung
carcinoma (Pancoast’s tumour).
Chest
The chest is the area of interest in the respiratory system. Both the front and the back of the
chest must be examined for signs. The back needs to be examined because most of the
chest’s pathologies are in the back and the lungs’ position in the chest cavity is mainly
posterior, hence examining the back gives a more holistic picture. When examining the
chest you follow 4 procedures: inspection, palpation, percussion and auscultation. When
doing all these procedures, you don’t test the front & back repeatedly between the
procedures. You conduct all procedures for the front, and then you move to the back. While
on the back, you then conduct the neck examinations. With each examination, it is
important to examine both sides.
Inspection
In inspection, you mainly observe. You may need to physically measure the dimensions of
the chest using a tape measure. The things to look for are:
• Shape. When the anteroposterior diameter of the chest is significantly larger than
the lateral diameter, the chest is described as barrel-shaped. Other shapes include:
Page 68 of 455
o Pigeon chest/pectus carinatum. This is an outward bowing of the sternum &
costal cartilages, leading to a localised prominence. It may be a manifestation
of a chronic childhood respiratory illness thought to result from repeated
strong contractions of the diaphragm while the thorax is still pliable. It also
occurs in rickets.
o Funnel chest/pectus excavatum. This is a developmental defect involving a
localised depression of the lower part of the sternum. The problem is usually
an aesthetic one, although in some cases lung capacity may be reduced.
o Kyphosis & scoliosis. Kyphosis is an exaggerated forward curvature of the
spine while scoliosis is lateral bowing of the spine. These conditions may be
idiopathic or secondary to poliomyelitis. Severe thoracic kyphoscoliosis may
reduce lung capacity and increase the work of breathing.
• Symmetry. You should inspect for asymmetrical chest wall expansion anteriorly &
posteriorly. Uneven movement indicates underlying lung disease, such as collapse,
consolidation, lung fibrosis, pleural effusion or pneumothorax. Alternatively, there
could be bilateral reduced movement, indicating a diffuse abnormality such as COPD
and interstitial lung disease. Analysis of expansion of the upper lobes is best done by
observing the patient posteriorly, looking down at the clavicle. Lower lobe expansion
is best observed posteriorly.
• Scars. Scars should be noted on the chest wall, both anteriorly & posteriorly. Scars
from a lobectomy (removal of a lobe of the lung) or pneumonectomy (removal of an
entire lung) leave a long diagonal scar on the back. However, if there are 3 scars 2-
3cm long, it indicates that there was video-assisted thoracoscopic surgery. There
may also be erythema on the skin, indicating radiated skin. That patch of skin may be
thickened too, and there may be small tattoo marks indicating the limits of the
irradiated area. Other non-respiratory-related scars may be present. A scar along the
left side of the sternum may be indicative of open heart surgery. Diagonal scars on
the back close to the costal margin may be indicative of nephrectomy or
adrenectomy.
Palpation
Palpation reveals a number of conditions:
• Chest expansion. To assess evenness of chest expansion, you place your hands
around the patient’s chest. The hands are placed with the fingers extended &
wrapped around the lateral aspects of the chest. The thumbs are only placed
slightly off the chest and they should almost meet in the midline. You then ask the
patient to take a deep breath in & out. The thumbs should separate at least 5cm
during inspiration. This test allows the clinician to determine even inflation as well
as whether there is reduced inflation or not. Lower lung expansion is assessed from
the back in this way. You can also place your arms on the upper front chest to
Page 69 of 455
assess expansion of the upper lobes of the lung, although this can only help you
determine if expansion is uniform.
• Apex beat. The apex of the heart can be shifted in the event that there is a lung
lesion. Displacement of the apex beat towards the side of the lesion may indicate
collapse of the lower lobe of a lung or localised interstitial lung disease. Movement
of the apex beat away from the side of the lesion may indicate pleural effusion or
tension pneumothorax. The displacement of the apex beat and the deviation of the
trachea both indicate a shift in the mediastinum. The apex beat is often impalpable
in a chest that is expanded secondary to COPD.
• Tactile vocal fremitus. This is a palpable vibration that can be felt when a person
speaks. This test is subjective to the person conducting the test, and it is a difficult
sign to interpret. The front & back are both palpated in 2 variable positions to test
the upper & lower lobe. It depends on the recognition of the changes in vibration in
the chest wall. Vocal fremitus is more obvious in men because of their low-pitched
voices. Vocal fremitus is abnormal if it is different between the two sides.
Percussion
Percussion allows you to listen for changes in the pitch of sound as you percuss different
regions of the chest. Percussion is done by placing your left hand over the area being
percussed, with the fingers separate. The middle finger must be firmly placed on the point
to be percussed. The middle finger on the right hand should then beat on the middle
phalanx of the left hand on the chest. This beating action should be done by swinging your
hand about the wrist in a pendulous fashion.
When percussing, you feel for resonance & dullness. Resonance is produced in regions
where there is air, such as in a normal lung, and is therefore the normal sound produced
when a person with normal healthy lungs is percussed. Dullness is a sign that the lung is
filled with something, be it fluid or puss, or merely consolidation. The different sounds are
as follows:
• The supraclavicular fossae, just anterior to the anterior margin of the trapezius
muscle. The apices of the lungs are found here.
Page 70 of 455
• The clavicle (this is usually used as the baseline sound for dullness). The clavicle is
not percussed with the left middle finger overlying it. You just strike the clavicle
directly with the right middle finger.
• The axilla, which is where the lateral aspect of the lungs is found.
• The front of the chest3. In the front of the chest, you percuss down the mid axillary
line.
• The back. On the back, you should make sure the patient roles their scapulae aside
so that you can access the ribcage. You do not move down a straight line. Rather,
you move in a divergent pattern downward. You start off closer to the midline in the
upper chest (to stay clear of the scapulae), and then you move laterally downwards.
The feel of the percussion note is as good as its note. The note is affected by the thickness
of the skin.
There are 2 regions of dullness that are expected. There is liver dullness, which is due to
percussion over the anterior aspect of the liver. This is usually below the right 6th intercostal
space. If there is resonance felt in this region, there is hyperinflation of the lung, and this
may be a sign of emphysema or asthma. There is also cardiac dullness which is felt over the
area where the heart is. If this area is reduced, it may indicate emphysema or asthma.
Auscultation
Most sounds reaching the chest wall are low pitched and are best heard using the bell of the
stethoscope. The diaphragm can be used to detect higher-pitched sounds such as pleural
friction rubs. Stretching the skin & hair underneath the diaphragm may produce a sound
that sounds like crackles. Ask the patient to breathe deeply & calmly. Make sure as you
auscultate, you hear the breath sound from the beginning of inspiration to the end of
expiration. You must auscultate anteriorly in the midclavicular line from the clavicle down to
the 6th rib; laterally from the axilla down to the 8th rib and; posteriorly down to the 11th rib.
There are a number of things to look for when auscultating. You do not just look for breath
sounds. The breath sounds must be assessed based on the following:
• Quality of the breath sounds. There are 2 types of breath sounds based on the
quality of the sound.
o Vesicular sounds. These are the normal breath sounds. These are heard over
nearly all parts of the chest. The sounds heard during inspiration are probably
produced near the part you’re listening to. The sounds of expiration are
produced in the larger airways rather than in the alveoli. Vesicular sounds are
louder and last longer on inspiration than on expiration. In addition, there is
no gap between inspiration & expiration.
3
In women, you should lift the breast. The breast is lifted using the back of the hand.
Page 71 of 455
o Bronchial sounds. These sounds are not normally heard when one is listening
to the lungs. However, they are normal when the trachea is auscultated as
well as the right upper chest (where the right bronchus opens into the
trachea). They are caused by pneumonia, bronchiectasis, lung collapse and
fibrosis. The expiratory sound has a higher intensity & pitch than the
inspiratory sound. They are heard in areas of consolidation, as solid lungs
conduct the turbulent sounds of the larger airways to the peripheral areas
without filtering.
Occasionally, bronchial sounds over a large cavity have exaggerated bronchial
quality.
• Intensity of the breath sounds. The intensity of breath sounds is not an indicator of
the amount of air entering the lungs. Rather, it is a reflection of the intensity with
which air enters the lungs. Breath sounds are reduced in individuals with COPD,
pleural effusion, pneumonia, a large neoplasm and pulmonary collapse. Breath
sounds are older when a person breaths deeply.
• Presence of other sounds. There are various other sounds that can be produced in
addition to the normal breath sounds. These are usually pathological:
o Crackles. These are produced mainly by abnormally large obstruction of the
airways. There are 2 types of crackles. There are coarse crackles which result
from large occluding structures such as puss & fluid and have an unpleasant
gurgling quality. Bronchiectasis is a common cause. There are also fine
crackles which are only heard in interstitial lung fibrosis. They are heard at
the end of inspiration. They are the result of the loss of stability of the
peripheral airways that collapse at the end of expiration. These airways are
then snapped open during deep inspiration, thus causing the characteristic
sounds.
o Wheezes. These are continuous sounds produced by fixed or variable opening
of smaller collapsed airways. They tend to be louder on expiration because
airways dilate on inspiration but narrow on expiration, thereby raising the
resistance against which the air has to flow out. This produces friction which
is converted to sound energy. High-pitched wheezes arise from smaller
bronchi while low-pitched wheezes arise from larger bronchi. They are
caused by conditions such as COPD (low-pitched), asthma (high-pitched).
They are, however, a poor guide to severity of airway obstruction
o Stridor. These sound similar to wheezes and are also caused by airway
obstruction. However, they are louder in the trachea and are always
produced in inspiration (wheezes are heard in both processes but are mainly
heard in expiration).
o Pleural rub. This is a condition caused by pleural irritation and can be
detected by the diaphragm of the stethoscope. When the pleura are irritated,
Page 72 of 455
they become thicker and rub against each other during breathing. The sound
is a continuous grating sound.
• Vocal fremitus. The patient is asked to say something, such as 99 or “nyama
nyama”. Over a normal lung, the low-pitched components of speech are heard with
a booming quality while high-pitched are attenuated. In consolidated lungs, the high-
pitched sounds are heard with a bleating quality – a trait called aegophony.
Page 73 of 455
Pneumonia
Pneumonia is an acute lower respiratory tract illness caused by infection of the lungs. The
infections typically cause inflammation within the lung parenchyma. Pneumonia is a
common condition in a variety of settings.
Pneumonia affects a wide variety of individuals, with different types of pneumonia affecting
different subgroups.
Classification of pneumonia
The different types of pneumonia are:
Community-acquired pneumonia
Page 74 of 455
healthcare workers who acquire pneumonia while working in hospital. It is the 2nd most
common type of hospital-acquired infarction after UTIs. A different spectrum of organisms is
implicated in hospital-acquired pneumonia. The common aetiological agents include K.
pneumoniae, P. aeruginosa, S. aureus (including MRSA), Acinetobacter and anaerobic
bacteria (e.g. Enterobacter).
Aspiration pneumonia
This is the aspiration of gastric contents into the respiratory tract. This is extremely severe
and can be fatal, due to the destructiveness of gastric acid. It can also be a complication of
anaesthesia, particularly during pregnancy. It can also complicate patients unable to protect
their airway, e.g. patients with strokes, myasthenia, bulbar palsies, reduced consciousness.
It also occurs in patients with oesophageal conditions, such as dysphagia, achalasia, reflux
and tracheoesophageal fistula. In the majority of patients, contents enter the right side due
to the right bronchus being wider & more vertical. They usually end up in the middle lobe or
apical and/or posterior segments of the lower lobe.
• Aspiration pneumonia. This is due to infection with oral & pharyngeal bacteria. The
common causes are oral anaerobes, S. pneumoniae, H. influenzae, S. aureus and
Gram-negative bacteria.
• Chemical pneumonitis. This is also called Mendelson syndrome and is due to
aspiration of gastric juices.
• Exogenous lipoid pneumonia. This is caused by aspiration of oils, such as vegetable
or mineral oil.
• Acute respiratory emergency. This is due to inhalation of a foreign body. This may
be complicated by bacterial infection.
PCP is a type of pneumonia caused by infection with Pneumocystis jirovecii, which is a fungal
infection. This infection is common in immunocompromised individuals, particularly those
living with HIV/AIDS. In patients with HIV, it used to be an AIDS-defining illness, and is
commonly associated with a CD4+ count of less than 200cells/mm3. The organism is found in
the air, and it establishes infection in immunocompromised individuals alone (despite being
a normal resident in the respiratory tract of most individuals).
Pneumocystis jirovecii pneumonia is the 2nd commonest opportunistic infection in HIV after
TB.
Pathophysiology
Page 75 of 455
For an organism to cause infection in the respiratory system, it needs to be able to bypass
the defence systems in the respiratory tract. They are physical barriers to infection, such as
the cough reflex and the muco-ciliary apparatus. Immunological barriers include alveolar
macrophages and lymphocytes. When any of these are impaired, the risk of infection
increases. Other factors that increase the risk of infection are pulmonary congestion &
oedema.
1. Congestion. This is when there is dilation of blood vessels within the infected region.
It is due to bacterial infection of the lung tissue. The affected lung is red, heavy and
boggy, and the alveoli are filled with proteinaceous fluid, bacteria and scattered
neutrophils.
2. Red hepatisation. This is when the lung develops consistency similar to the liver. The
alveoli are packed with red cells, neutrophils and fibrin.
3. Grey hepatisation. This lung is dry, grey and firm. The red cells have been broken
down while the fibrinopurulent exudate still remains within the alveoli.
4. Resolution. This usually happens in uncomplicated pneumonia. The exudate are
actively broken down to form granular semi-fluid debris.
The organisation of the exudate produces consolidation, in which the alveoli are clogged up
with fluid and are resistant to distension.
In atypical pneumonia, the infectious reactions are in the interstitium rather than in the
alveoli. This accounts for the more subtle clinical features.
Page 76 of 455
• Extremes of age. Individuals younger than 16 and older than 65 are at risk of
developing pneumonia. This pattern is more typical in individuals that are not
immunocompromised.
• Other respiratory conditions. These include cystic fibrosis, COPD, bronchiectasis and
obstructive lesions such as inhaled foreign bodies or endoluminal cancer.
• Comorbidities. These include HIV, diabetes mellitus, chronic kidney disease,
malnutrition, and recent viral respiratory infection.
• Lifestyle. Cigarette smoking, excess alcohol and intravenous drug use all increase the
risk of pneumonia.
• Iatrogenic. Immunosuppressant therapy (such as prolonged steroids) predisposes to
pneumonia.
Clinical features
The clinical features of pneumonia are:
History
The symptoms often occur acutely, although some conditions such as pneumocystis jirovecii
pneumonia & other fungal pneumonias take a more protracted course. In atypical
pneumonia, the symptoms are not as florid, and sputum is often non-purulent.
Also ask about risk factors: any pre-existing respiratory conditions, history of TB, HIV co-
infection, smoking & alcohol, etc.
Examination
Page 77 of 455
• Palpation: reduced chest expansion, increased tactile fremitus if there is
consolidation; decreased tactile fremitus if there is an effusion.
• Percussion: apple core dullness in affected lung regions. Stoney dull if there is a
concomitant effusion.
• Auscultation: bronchial breath sounds may be present in affected regions. There may
also be fine crackles in consolidated areas. There may be an effusion, with reduced
breath sounds in the affected area. There will be bronchial breathing just above the
effusion.
Complications of pneumonia
The complications of pneumonia are:
• Lung abscess. An acute bacterial pneumonia is one of the causes of a lung abscess.
Often, the abscess cavity is not ventilated and therefore anaerobic bacteria grow in
it. They can be a result of aspiration pneumonia, and they are often single well-
localised lesions. If they occur in the setting of community-acquired pneumonia, they
are most likely due to S. aureus or Klebsiella pneumoniae. TB can also cause a lung
abscess. A lung abscess will present with a productive cough with purulent sputum,
spiking fever & malaise, clubbing, weight loss and normocytic anaemia. Secondary
amyloidosis can occur.
1 1
• Para-pneumonic effusion & empyema. Para-pneumonic effusions complicated 3 to 2
of all cases of community-acquired pneumonia. They are usually simple exudative
effusions. An empyema can develop in this effusion when it becomes infected1. Early
indications of an empyema are a spiking fever and rising/persistently elevated
inflammatory markers. TB can also result in an effusion.
• Organisation & fibrosis. Fibrosis can occur, particularly with conditions that result in
chronic consolidation of the lungs. The lung reduces in volume and becomes less
distendable.
• Sepsis. Pneumonia can act as a focus of infection for sepsis. The sepsis can lay
ground for other infections, such as meningitis, infective endocarditis and arthritis.
• Sputum studies. These are done for patients with productive cough. The sputum
specimen is taken for microscopy, culture and sensitivity. The sputum should be
stained using both Gram & Ziehl-Neelsen staining (to check for TB).
1
Para-pneumonic effusions are the commonest cause of empyema thoracis.
Page 78 of 455
• Blood culture. A positive blood culture in a S. pneumoniae community-acquired
pneumonia indicates more severe disease and poor prognosis. If the pneumonia is
not severe, blood culture is not necessary.
• Blood workup. This includes FBC, U&Es and LFTs. FBC will reveal leukocytosis in S.
pneumoniae pneumonia, and it is usually normal in M. pneumoniae pneumonia. The
FBC shows leukocytosis with an elevation in neutrophil count. Anaemia may be
present, and if M. pneumoniae is the causative organism, haemolytic anaemia should
be ruled out by a direct Coombs test. A CD4+ count is indicated in individuals living
with HIV.
• Bronchoscopy. This is required for patients with PCP as they do not produce much
sputum. The bronchoscopy is done for bronchoalveolar lavage to obtain a specimen
for microscopy. When stained using Giemsa or methenamine silver, you will see
Pneumocystis cysts.
• Chest X-ray. Typical bacterial pneumonias will reveal lobar or multilobar
heterogenous opacification characteristic of consolidation. In the region with
consolidation, air bronchograms will also be present. There may also be cavitation,
although this is more suggestive of TB. An effusion can also be identified. Atypical
pneumonias have variable features, ranging from lobar involvement to bilateral
interstitial disease. In PCP, there may be a normal chest X-ray in early mild disease.
In most PCP patients, there are diffuse bilateral infiltrates extending from the peri-
hilar region. Pleural effusions in PCP are rare.
• CT scan. This is indicated when an underlying bronchogenic carcinoma is suggested.
For PCP patients, lung windows will demonstrate a ground glass appearance with a
background of interlobar septal thickening.
• Arterial blood gases. This is done to check for the oxygen saturation (when oxygen
saturation is below 94% or when there is severe pneumonia), and to check for
respiratory acidosis.
• Serum biochemistry. LDH levels can be measured in patients with suspected PCP.
They are usually elevated (>220U/L) and they reflect the degree of lung injury.
• Pleural fluid. Microscopy can be done to identify any infective processes present.
Cytology is usually done to rule out malignancies. You can also biochemistry studies,
(and you use the Light‘s criteria to determine whether it is exudative or transudative
if the protein level is between 20g/L & 30g/L), and you can culture to determine
whether it is an empyema or not.
Severity of pneumonia
The severity of pneumonia can be quickly determined using the CURB 65 scoring system
Page 79 of 455
R – respiratory rate Tachypnoea of 30bpm or more
90
B – blood pressure Hypotension of less than 60mm Hg
65 – age Age above 65 years
Each parameter scores 1 point. If the patient scores 0-1, they can be treated as an
outpatient. If they score 2, they require hospital therapy. If they score 3 or more, they have
severe pneumonia. Severe pneumonia has a very high mortality (15-40%) and therefore
requires ICU.
Management of pneumonia
The definitive management of pneumonia is tailored for the severity of the patient. The
general management interventions done for most patients are as follows:
2
This is important in COPD patients, as restoring oxygen saturation will lead to abolishment of the hypoxic
response required to maintain alveolar ventilation. This will lead to respiratory failure.
Page 80 of 455
you should not wait for specimen results. The choice of antibiotic regime is as
follows:
Community- Low-risk
acquired • If CURB65 = 0, Home care is appropriate. No need for
pneumonia microbiological tests.
CURB 65 = 0-1 • If CURB65=1, patients only require a short ambulatory
(mild disease; stay. No microbiological tests required unless outbreak
<3% mortality) suspected or there is a Mycoplasma epidemic.
• Give oral medications:
o Amoxicillin 500mg tds.
o Clarithromycin 500mg bd.
o Doxycycline 200mg loading dose then 100mg od.
Course of antibiotics is given for 7 days.
Community- CURB65 2:
acquired • You need to admit and do blood cultures, sputum
pneumonia studies, test for pneumococcus antigen and serology/PCR
CURB 65 = 2 if there is an epidemic.
(moderate • Give oral combination therapy:
disease; 9% o Amoxicillin 500-1000g tds, OR doxycycline 200mg
mortality) loading dose then 100mg od, OR levofloxacin
500mg bd
o Clarithromycin 500mg bd OR erythromycin 500mg
qid
This is administered for suspected/confirmed
pneumococcal infection. It is given for 7 days.
• For confirmed staphylococcal infection, give cloxacillin 1-
2g IV 6-hourly OR clindamycin 600mg IV 3-4 times daily.
Give medication for 14 days, although after day 7
consider changing to oral medication.
• For Klebsiella & other Gram-negative organisms, give
ceftriaxone 1g IV bd + gentamycin 120mg IV bd for 10-14
days.
Community- CURB65 of 3-5
acquired • You need to admit the patient into ICU and take blood
pneumonia culture & sputum samples for microscopy, culture and
CURB 65 = 3-5 sensitivity.
(severe disease; • Antibiotics should be administered as soon as possible:
15-40% o Co-amoxiclav 1.2g IV tds + clarithromycin 500mg
mortality) IV bd3.
3
Consider a fluoroquinolone, e.g. levofloxacin, if Legionnaire’s disease is considered
Page 81 of 455
o Ceftriaxone 2g IV od + clarithromycin 500mg
bd/erythromycin 500mg qid.
o Benzylpenicillin 1.2g qid + erythromycin 500mg
qid
The course of anitbiotics should be taken for 7 days.
• For confirmed staphylococcal infection, give cloxacillin 1-
2g IV 6-hourly OR clindamycin 600mg IV 3-4 times daily.
Give medication for 14 days, although after day 7
consider changing to oral medication.
• For Klebsiella & other Gram-negative organisms, give
ceftriaxone 1g IV bd + gentamycin 120mg IV bd for 10-14
days.
Hospital- Benzylpenicillin 1.5g IV 6-hourly + gentamycin 120mg IV 12-
acquired hourly for 7-10 days.
pneumonia
Aspiration IV cephalosporin + metronidazole.
pneumonia
Pneumocystis •Cotrimoxazole 1920mg (4 tablets) orally tds for 21 days.
jirovecii OR
pneumonia Clindamycin 600mg tds + primaquine 15mg od for 21
days.
• Prednisolone is given if the patient has tachypnoea
and/or cyanosis. Give 40mg bd for 5 days, then 40mg od
for 5 days, then 20mg od for 11 days.
• If patient is on ART, give cotrimoxazole prophylaxis
indefinitely – 960mg od.
5. Thrombophylaxis. This is required for all patients admitted for longer than 12 hours.
You can give subcutaneous LMW heparin, unless it is contraindicated and TED
stockings are fitted.
6. Chest physiotherapy. This is indicated when sputum retention is an issue.
7. Nutritional supplementation. This is particularly assessed in severe disease, and the
assessment is made by a dietician.
8. Surgery. Surgery is mainly indicated for complications.
a. Lung abscess. Lung abscesses need to be drained surgically. This is followed
by antibiotic therapy which is normally guided by culture results. You then
give benzylpenicillin 1.5mg IV 6-hourly + metronidazole 400mg orally tds, for
4-8 weeks4. Alternatively you can use co-amoxiclav 625mg po tds for 4-8
weeks.
4
If the patient ceases to be toxic before 4 weeks are up, continue intravenous benzylpenicillin for about 7 days
then switch to oral amoxicillin and discharge the patient for the remainder of the course.
Page 82 of 455
b. Empyema. For an empyema in the exudative & fibrinopurulent stage, you can
place a large intercostal tube connected to an underwater seal drain. If the
empyema is in the organising stage, you surgically remove the pleura in a
decortication procedure. After inserting the drain, you give benzylpenicillin
2.5MU IV 6-hourly + metronidazole 400mg po tds for 10-14 days. If the
suspected pneumonia was staphylococcal, give cloxacillin 1g 6-hourly +
metronidazole 400mg po tds for 10-14 days. Also institute thrombophylaxis
with heparin or warfarin.
Page 83 of 455
Tuberculosis (TB)
TB is a chronic infectious disease caused by Mycobacterium tuberculosis. It is a disease
characterised by formation of multiple granulomata in various body tissues. TB is the 9th
leading cause of death worldwide, and is the single leading cause of death from a single
infectious agent (including HIV).
There is a very large disease burden across the world. More than 2 billion individuals are
thought to be infected with TB in the world. In 2016, there were 10.4 million new TB cases,
equivalent to 140 cases per 100 000 population. The incidence varies from 10 per 100 000
population in low incidence countries to 150-300 cases per 100 000 population, with some
countries reaching over 500 per 100 000 population. The top 5 countries with the highest
number of cases are (in descending order) India, Indonesia, China, the Philippines and
Pakistan1. 90% of patients were adult, and 65% were male. 10% of patients worldwide were
infected with HIV. However, in countries with high HIV prevalence, TB-HIV co-infection is
often above 50%. In addition, 74% of patients with HIV-TB confection came from Africa. In
Zimbabwe in 2016, the number of incident TB cases was 34 000 (or 208 cases per 100 000
population), of which the number of cases with HIV-TB co-infection was 23 000 (139 cases
per 100 000, or 67.6%).
About 2 million people die every year (1.7 million in 2017), although the mortality rate is
declining by 3% every year. The mortality rate is about 4-times higher amongst those living
with HIV. The mortality rate in Zimbabwe is 7.2 deaths per 100 000 amongst HIV-negative
individuals and 27 cases per 100 000 amongst HIV-positive patients. There were an
estimated 1300 MDR-TB cases amongst notified pulmonary TB cases.
Under the microscope, they appear as thin curved rods. They are hydrophobic.
Mycobacteria contain a complex lipid-rich cell wall that protects the bacteria (acid-fastness,
1
45% of the global burden of TB is carried by China, India and Indonesia alone.
2
Most of these, however, are slow-growing
Page 84 of 455
resistance to detergents & common antibacterial agents, slow growth) and has antigenic
properties. The surface glycolipids constitute 25% of the bacterium’s dry weight. The major
component of the cell wall is mycolic acid. The plasma membrane of the bacterium contains
many proteins, including phosphatidylinositol and lipoarabinomannan (LAM). Mycobacteria
are sensitive to heat & sunlight and are susceptible to formaldehyde, glutaraldehyde and
phenol. They can survive in milk.
Risk factors
The risk factors for developing TB are:
Pathogenesis
M. tuberculosis is an obligate aerobic organism and facultative intracellular organism. It
often gains access into the body through respiratory droplets, and the organism gets
phagocytised, primarily by mononuclear phagocytes such as macrophages. M. tuberculosis is
able to prevent fusion of the phagosome with a lysosome, but allows fusion of the
phagosome with other intracellular vesicles. It also prevents antigenic killing by catabolising
the reactive oxygen species formed in macrophages, and thus prevents macrophage-
mediated killing.
Page 85 of 455
The immune system has a prominent role in limiting spread of infection. In response to
infection, macrophages release IL-12 & TNF-α which increase local inflammation. These
activate T-helper cells which differentiate into TH1- cells with release of a TH1-cytokine profile
(IFN-γ, IL-2, IL-12, TNF-β) & cell-mediated immunity and phagocyte-dependent inflammation
(delayed type hypersensitivity reaction). This is a favourable response for clearance of
infection as it allows granuloma formation & therefore containment of disease. There is also
concurrent activation of a TH2-response (with release of IL-4, IL-5, IL-9, IL-10 and IL-13),
which is a humoral/antibody response. The antibodies include IgE release, and there is also
eosinophil accumulation. This response, however, inhibits phagocyte function and leads to
free bacterial proliferation & disease dissemination.
Clinical syndromes
The majority of individuals exposed to TB do not develop infection; only 10% develop
detectable infections. In infected individuals, the infection can be an overt clinical infection
with signs & symptoms, or there can be latent infection. Clinical infection can be primary
infection or reactivation.
Primary TB
Primary TB describes a first time infection in an individual who has never been infected or
sensitised before. Elderly & immunosuppressed individuals can lose their sensitivity to the
bacterium and therefore get repeated primary infection.
Primary infection is almost always established in the lungs. Once inhaled, the alveolar
macrophages ingest the bacteria and the bacteria proliferate within them. This leads to an
inflammatory reaction leading to neutrophil chemoattraction & release of cytokines. The
usual affected areas are the upper lobes & the upper parts of the lower lobes. The infection
usually begins close to the pleura where there is most air, and leads to the formation of a
Ghon focus in which a granulomatous reaction forms with caseous necrosis. These are
usually peripheral and located close to the pleura & along fissures. The bacteria travel up
the lymphatic channels to the lymph nodes, in which granulomata also form. An involved
hilar lymph node is called a Ghon complex, and if it calcifies it is called a Ranke complex.
Page 86 of 455
The primary infection can be cleared, or go into clinical latency where it hides and reappears
(reactivation) many years later.
Secondary TB/reactivation TB
• Secondary pulmonary TB. This is the most common form, and occurs in 55% of
cases. Because of the pre-existing hypersensitivity, the bacilli initiate a prompt
immune response which walls off the infection. This increases the likelihood of
cavitation. It also means that the lymph nodes are less likely to be involved.
Secondary pulmonary particularly likes to affect the apex of the lung. It may progress
and lead to expansion of the cavity, with erosion into bronchi or blood vessels. With
progressive pulmonary TB, there is invariable pleural involvement, with formation of
a serous pleural effusion3.
• Military TB. Military TB occurs when the bacilli enter the systemic circulation and
seed in different organs across the body. They can even return to the lungs. There
are numerous white foci in affected organs which consist of caseating granulomas.
• Lymphadenitis. This is the most common form of extra-pulmonary disease. It usually
occurs in the cervical region, producing scrofula. The lymphadenopathy tends to be
unifocal, and extra-nodal disease is not usually present. HIV-positive patients,
however, almost always tend to have multifocal disease.
Clinical features
Clinical features are seen in patients with clinically active disease. Latent disease is
asymptomatic. The majority of cases are pulmonary tuberculosis cases. The clinical features
are:
3
This can complicate with a tuberculous empyema or an obliterative fibrous pleuritis.
Page 87 of 455
may be sinus tract formation with a purulent discharge but no erythema (the so-
called cold abscess).
Investigations
The diagnostic investigations done for TB are:
• Chest X-ray. The chest X-ray may be normal in some symptomatic patients. In
others, chest X-ray findings may be suggestive while the patient is asymptomatic.
The chest X-ray findings include: consolidation ± cavitation, fibrosis, calcification
(Ranke complex), hilar lymphadenopathy, and pleural effusion/thickening.
• Microscopy. M. tuberculosis does not take up the Gram stain well. For microscopy,
therefore, you use the Ziehl-Neelsen stain and use it to stain the bacterial sample.
Mycobacteria are resistant to decolorisation by acid during the staining procedure,
and therefore they stain red on a blue background. This test is very sensitive, and has
the advantage of only showing live mycobacteria and not dead organisms. You can
also use an auramine-rhodamine stain which is more sensitive but less specific than
the ZN stain. It requires fluorescent microscopy and highlights bacilli as yellow-
orange on a green background.
• Tuberculin skin test/Mantoux test. This is an immunologic test which makes use of
delayed hypersensitivity which occurs after exposure to TB antigens. You inject TB
antigens (5 tuberculin units of purified protein derivatives, PPDs) intradermally and
then wait for 48-72 hours. Skin test reactivity is defined by the diameter of the area
of induration. If the diameter is greater than 10mm, then the test is positive and is
highly suggestive of TB infection. The test can also be used for patients with
suspected latent TB. However, it also tests positive in patients with immunity to TB
and those that have been vaccinated. In individuals with immunosuppression (HIV,
sarcoidosis, military TB, lymphoma), the indurated area is usually small and so you
can get a false negative. For all these reasons, therefore, it is unreliable to use alone
for diagnosis of TB.
• Culture. Culture is the gold standard for diagnosis of TB. However, because it takes
time to produce results, it is not routinely ordered for diagnosis of every patient. You
can use:
o The LJ (Lowenstein-Jensen) medium, which produces results after 8 weeks.
o The Middlebrook agar.
o The microscopic-observation drug-susceptibility (MODS) assay.
o The Mycobacterium Growth Indicator Tube (MGIT), which produces results
within 2 weeks.
• Molecular tests. The prototypical test used is the GeneXpert® test, which uses a
device and a cartridge to conduct PCR on a sputum specimen. It is fast and therefore
can be used on initial contact. However, it remains positive even after treatment as
it also detects dead bacteria. Molecular tests are also useful in detecting rifampicin
Page 88 of 455
resistance. You can also use the Line Probe Assay (LPA), which is a drug susceptibility
test
• IFN-γ release assays. The test uses immunoassays to measure the amount of IFN-γ
released by sensitised T-cells after introduction of TB antigens. The antigens used
include ESAT-6 (early secreted antigenic target 6) and CFP-10 (culture filtrate
protein). An example of such a test includes Quantiferon-TB gold kit.
• Biopsy. Biopsies can be obtained, particularly for extra-pulmonary cases. The
pathognomonic feature is a central-caseating granuloma.
• Antigen test. You can also do a urine LAM assay, in which you measure LAM
(lipoarabinomannan) levels in urine. LAM is shed from metabolically active or
degenerating bacterial cells. It only tests positive in individuals with active TB
disease.
• FBC. Routine FBC is usually normal in early stages. However, patients may develop
normocytic normochromic anaemia. There may also be a leukocytosis (rich in
lymphocytes) or monocytosis (rarely).
• U&Es. Hyponatraemia can occur due to SIADH or due to adrenal insufficiency.
• CRP. This is raised in up to 85% of cases.
• LFTs. Hypoalbuminaemia and hypergammaglobulinaemia (widened γ-gap) are late
features.
Management
Patients are divided into drug-susceptible & drug-resistant TB cases based on whether their
rifampicin resistance testing results on GeneXpert are positive or negative. All patients that
are diagnosed with TB must be notified & registered in the facility’s TB register.
Drug-susceptible TB
For all cases of drug-susceptible TB, whether primary or reactivation TB, you treat with first-
line antiTB drugs: rifampicin (R), isoniazid (H), pyrazinamide (Z) and ethambutol (E). For
pulmonary TB and most forms of extra-pulmonary TB, you start with an intensive phase of
HRZE for 2 months followed by a continuation phase of HR for 4 months. For meningo-
cerebral and skeletal TB, the continuation phase is extended to 6 months, with the option of
continuing for up to 12 months. The duration of treatment is extended due to the poor
penetration of medicines to these sites.
All first-line drugs are orally administered. Direct observed therapy (DOT) is encouraged for
treatment of tuberculosis. It is defined as treatment supervised by a healthcare professional
or family member. The patient is observed swallowing their medication. DOT achieves
treatment completion rates of over 85%.
Page 89 of 455
The side effects to watch out for include:
Drug-resistant TB
4
This is prevented by co-administering pyridoxine 10mg daily.
5
The duration of the intensive phase depends on culture conversion. The intensive phase is extended to 6
months if culture conversion has not occurred by the 4th month.
Page 90 of 455
According to WHO, there should be at least 5 effective TB medicines in the intensive phase.
They should include pyrazinamide and 4 other 2nd-line agents: 1 from class A, 1 from class B
and 2 from class C.
Adjuvant therapy
• TB meningitis.
• TB pericarditis.
• Patients with paradoxical massive lymphadenopathy.
• Severe hypersensitivity reactions to anti-TB medication.
• Hypoadrenalism.
• Renal tract TB (to prevent scarring).
• TB laryngitis with life-threatening airway obstruction.
Page 91 of 455
The dosing regimens are as follows:
• For TB meningitis, you give prednisolone 60mg orally per day for 4 weeks then taper
off, OR dexamethasone 8-12mg day tapered off over 6-8 weeks (for those that
cannot take prednisolone orally).
• For TB pericarditis, you give prednisolone 60mg orally daily for 4 weeks, then 30mg
orally daily for another 4 weeks then taper off over several weeks.
• For hypersensitivity reactions to anti-TB medicines, you give 20-80mg daily and taper
off over 2-8 weeks.
Monitoring of therapy
Once a patient is started on treatment, he/she must be followed-up regularly. At a
healthcare facility, this is done every 2-4 weeks by a clinician or daily by the DOT nurse. At
community level, this is done daily during the intensive phase and weekly during the
continuation phase by the treatment supporter. All TB patients must be monitored for
adherence, response to treatment and adverse drug events. At the end of treatment, it is
compulsory to assess outcomes of patients.
Adherence to treatment
It is not only the duty of the patient to adhere to treatment, but also of the healthcare
system (particularly the healthcare workers who are treating the patient) to ensure this.
Adherence should therefore be emphasised at every visit.
Response to treatment
Page 92 of 455
• Clinical monitoring. This is done for all patients. It includes retaking the history,
examining the patient, and checking the weight. This may be the only way of
monitoring in situations like extra-pulmonary TB where repeated samples cannot be
taken.
• Bacteriological monitoring. This is done using sputum smear microscopy, as it is
more sensitive in detecting live organisms6. It is the best indicator that treatment is
being taken regularly and is effective. After 2 months of treatment, more than 80%
of new pulmonary bacteriologically-confirmed cases should be smear-negative. After
3 months, the rate should increase to at least 90%. A positive sputum result at the
end of the intensive phase should prompt a review of the supervision & support
provided by the programme and adherence to treatment. Sputum should be
examined at 2, 5 and 6 months.
Repeated use of chest X-rays for monitoring is unnecessary and wasteful of resources. Chest
radiograph changes may take longer to resolve than clinical and laboratory changes.
All patients must be monitored and managed appropriately for adverse drug reactions.
6
GeneXpert® will still test positive even with dead organisms.
Page 93 of 455
Asthma
Asthma is a chronic inflammatory condition characterised by reversible airway obstruction.
It affects 5-8% of the population, and it commonly starts in childhood from ages 3-5 years.
The prevalence of asthma, however, is increasing in many countries, and it is more common
in developed countries. It is thought that the incidence increases as the country adopts a
more westernised lifestyle.
Pathogenesis
Asthma classically has 3 characteristics:
1
These constitute airway remodelling.
Page 94 of 455
• Environmental allergens: grass pollen, domestic pets, fungi (Dermatophagoides
pteronyssinus).
• Occupational sensitizers. These can be divided into low & high molecular weight.
Low molecular weight allergens (e.g. wood dust, bleaches and dyes) do not elicit an
IgE response, while high molecular weight allergens (e.g. platinum salts, acid
anhydrides) do. Low molecular weight allergens bond chemically to epithelial cells
and therefore stimulate them to release cytokines. The allergens also act as haptens.
Some forms of occupational asthma are increased in smokers.
• Cold air & exercise. These cause the fluid film lining the airways to be hyper-
osmolar, leading to degranulation of mast cells & basophils (releasing histamine,
leukotrienes and prostaglandins) and stimulation of neural reflexes.
• Atmospheric pollution and irritants, dusts, vapours and fumes. Irritants directly
irritate the airway, eliciting inflammation in an already hyper-responsive mucosa.
Acute exacerbations increase in areas with summer & winter pollution associated
with climate temperature inversions.
• Diet. Intake of fresh fruits & vegetables is protective. This is believed to be related to
the anti-oxidants in food.
• Drugs. The drugs implicated in asthma include:
o NSAIDs. NSAIDs block production of prostaglandins (particularly PGE2), which
induces increased production of cysteinyl leukotrienes by eosinophils, mast
cells and macrophages. This causes increased inflammatory responses in
patients. The response is more marked with NSAIDs that inhibit COX-1 such
as aspirin, ibuprofen, indomethacin, etc.
o B-blockers. These cause bronchoconstriction by lifting the sympathetic tone
that opposes bronchoconstriction by parasympathetic innervation. This is
through inhibition of β2-receptors that induce relaxation of bronchial smooth
muscle. B1-blockers (e.g. atenolol) can also induce bronchoconstriction.
• Dyspnoea.
• Cough. The cough is usually nocturnal, and may be the only presenting feature in
children. The cough may be productive.
• Wheezes.
Symptoms are usually worse during the night, and attacks vary in frequency & duration.
Other things to ask about include:
• Precipitating factors, e.g. cold air, dust, exercise, specific allergens, infections,
smoking, pollution and drugs (NSAIDs & β-blockers).
Page 95 of 455
• Diurnal variation in symptoms. Marked morning dipping is common.
• Quantify exercise tolerance.
• Quantify disturbed sleep as nights per week.
• Ask about acid reflux. This is present in 40-60% of patients.
• Other atopic conditions such as eczema, hay fever, allergies. Also ask about family
history.
• Any potential irritants at home, such as pets, carpets, feather pillows/duvets.
• Occupation.
Examination
The findings you will get on examination are:
• Mild attack. The patient will present with tachypnoea, audible wheeze, and a hyper-
inflated chest. On percussion, the patient will have a hyper-resonant percussion
note. On auscultation, there is reduced air entry and a widespread polyphonic
wheeze.
• Severe attack. The patient is unable to complete sentences on a single breath. The
pulse rate is above 110bpm and respiratory rate is greater than 25bpm. The wheezes
are more prominent.
• Life-threatening attack. The patient is exhausted and can barely talk. The patient
may be cyanosed, hypoxic and hypercapnic. The patient may also have a
bradycardia.
Investigations
The investigations done in a suspected asthma patient are:
• Peak expiatory flow rate (PEFR). This is used to demonstrate the airflow limitation
that classifies the disease. It can also be used to monitor progress of the condition
after treatment. Chronic asthma is diagnosed on PEFR by a diurnal variation of more
than 20% on more than 3 days a week. PEFR can also classify an acute asthma attack,
although clinical features are used to do this.
PEF predicted value
Mild-moderate 50-75%
Severe 33-50%
Life-threatening <33%
• Spirometry. On spirometry, you can demonstrate the obstructive picture that is
typical of asthma: reduced FEV1/FVC ratio and increased residual volume. You can
also demonstrate the reversibility of asthma by an increase in FEV 1 of greater than
15% after administration of a bronchodilator (β2-agonist) or corticosteroid.
• Blood tests. The patient must have an FBC taken. This may demonstrate elevated
eosinophil count & fraction. You can also take U&E and blood cultures (to rule out
Page 96 of 455
infection). ABG (arterial blood gas analysis) usually demonstrates reduced paO2 &
paCO2 levels (due to hyperventilation)2.
• Sputum tests. These can also help diagnose asthma. Sputum eosinophilia is more
diagnostic of asthma than eosinophilia on FBC. You can also do sputum microscopy,
culture and sensitivity to exclude superimposed infection.
• Chest X-ray. This may show a hyper-inflated chest in an acute episode or in chronic
disease. it is also helpful in excluding a pneumothorax and infection (particularly
pulmonary infiltrates of allergic bronchopulmonary aspergillosis).
• Allergen tests. You can do a skin prick test, in which you aim to identify an offending
allergen based on the history provided. Allergen-specific IgE can then be measured.
A proper allergen provocation test is usually done for research purposes.
• Histamine/methacholine provocation challenges. These are used to demonstrate
airway hyper-responsiveness. They are mainly useful in investigating patients in
whom the main symptom is cough. It should not be performed in patients with poor
lung function (FEV1 < 1.5L) or a history of “brittle” asthma.
Management of asthma
Management of asthma aims to:
• Abolish symptoms.
• Restore normal/best possible lung function.
• Reduce cases & risks of severe attacks.
• Enable normal growth in children.
• Minimise absence from work or school.
Management of asthma is divided into management of chronic asthma and management of
acute asthma.
Management of chronic asthma
Management of chronic asthma is the control of day & night-time symptoms, preventing
occurrence of attacks, no limitations on exercise and normal lung function (FEV1 and/or
PEFR > 80% predicted or best). It consists of the following:
2
A normal or elevated paCO2, however, warrants transfer to the HDU or ICU because of impending failing
respiratory effort.
Page 97 of 455
using a peak flowmeter. Also give specific advice on what to do in an emergency.
Also teach relaxed breathing.
• Pharmacologic therapy. There are many drugs used in asthma, and they serve
different functions. The drugs used in asthma include:
o B2-agonists. These are the most widely used agents in asthma. They work by
stimulating β2-receptors, which cause bronchial smooth muscle relaxation.
Being selective agents, they usually do not stimulate the β1-receptors on the
myocardium. They are divided into short-acting β2-agonists (SABA), such as
salbutamol & terbutaline, and long-acting β2-agonists (LABA), such as
salmeterol & formoterol. The agents used are mainly inhalational agents, and
they are used in different situations. It is not advised to use LABAs for long
periods of time, particularly if they are not working. LABAs are also always
used with corticosteroids.
o Anti-muscarinic agents. These drugs work by blocking parasympathetic M3-
receptors found on bronchial smooth muscle, which normally contracts
under parasympathetic stimulation. These drugs are not encouraged for use
in chronic asthma, but they can be used in acute attacks. They are mainly
used for COPD. They can also be taken as inhaled agents.
o Corticosteroids. These agents are used to inhibit bronchial inflammation
which can cause airway obstruction. They usually work over days. They are
best given inhaled to minimise systemic side effects. However, they can also
be given orally or intravenously depending on the precise indication. After
taking inhaled corticosteroids, the mouth should be rinsed to prevent oral
candidiasis from developing. Inhalational corticosteroids include
beclomethasone, budesonide, fluticasone and mometasone. Asthmatic
patients who smoke are less responsive to corticosteroids. Therefore,
smoking cessation should be offered before corticosteroids are considered.
o Leukotriene receptor antagonists (LTRAs). These drugs act by inhibiting the
cysteinyl leukotriene receptor (LT1). Examples include montelukast,
pranlukast and zafirlukast. They are mainly used in patients who are
uncontrolled on inhaled corticosteroids. However, you cannot predict who is
going to benefit from them, and therefore they are tried for 4 weeks before
the decision to continue or stop them will work.
o Methylxanthenes. These drugs include theophylline, caffeine and
theobromine. They are known to cause bronchodilation, but the mechanism
is not well known. One proposed mechanism is that they inhibit
phosphodiesterase, thus increasing cAMP levels and inhibiting
bronchoconstriction.
o Anti-inflammatory drugs. Sodium cromoglycate used to be used regularly in
asthma treatment. They mainly act on eosinophils, mast cells and epithelial
cells but not lymphocytes.
o Anti-IgE monoclonal antibody. Omalizumab is a recombinant humanised
monoclonal antibody that chelates free IgE and downregulates the number &
Page 98 of 455
activity of mast cells & basophils. It is effective in patients with persistent
allergic asthma. It is given as a subcutaneous injection for 2-4 weeks.
In accordance with the British Thoracic Society guidelines, asthma is treated in a step-wise
approach aimed at stopping symptoms quickly and improving peak expiratory flow. The
steps are as follows:
1. Step 1: Mild intermittent asthma. In this case, PEFR may even be 100% predicted.
Symptoms are occasional and they occur less frequently than once daily. You use
inhaled SABAs such as salbutamol (100-200µg up to 4-times daily). You can also use
short-acting antimuscarinic drugs e.g. ipratropium, but SABAs are preferred. You
move to step 2 if: the patient has more than 2 attacks in a week; the patient has
night symptoms more than once a week; the patient has had an acute exacerbation
(asthma attack) in the last 2 years.
2. Regular preventer therapy. In this case, the PEFR is usually less than 80% predicted.
In this stage, symptoms may even appear daily. You place the patient on an inhaled
SABA (salbutamol 100-200µg up to 4-times daily) combined with an inhaled
corticosteroid (such as beclomethasone 200-800µg daily). Alternatives to inhaled
corticosteroid include leukotriene receptor antagonist, theophylline or sodium
cromoglycate, but these are less effective than corticosteroids.
3. Initial add-on therapy. In this case, you consider using a LABA (e.g. formoterol or
salmeterol 50µg twice daily) in addition to the SABA & inhaled corticosteroid. It is
done for patients with PEFR of 50-80% predicted, and not controlled on regular
preventer therapy. This prevents the need for continuously increasing corticosteroid
doses. If control is better but still inadequate, you continue the LABA and increase
the dose of inhaled corticosteroid. You can also add leukotriene antagonists or
modified release theophylline instead. LABAs should not be continued if they are not
benefiting.
4. Persistent poor control. Persistent poor control is when there are severe symptoms
or symptoms persist despite high-dose inhaled corticosteroids. You consider:
increasing inhaled corticosteroids to up to 2000µg/day; adding leukotriene receptor
antagonist, modified-release theophylline or oral β2-agonist. These are considered in
patients on initial add-on therapy (with PEFR being 50-80% predicted).
5. Regular oral corticosteroids. This is done for patient with PEFR of less than 50%
predicted. You can add oral prednisolone at the lowest possible dose (usually 7.5mg
daily, but target being 5mg). you also continue with high-dose inhaled
corticosteroids, and you refer to the asthma clinic.
Patients can develop an asthma attack, which occurs as a result of lack of adherence, acute
viral infection, or exposure to allergen or triggering drug.
Management of acute asthma
Acute severe asthma is typically indicated by:
Page 99 of 455
• Tachycardia greater than 110bpm. Bradycardia or hypotension is life-threatening.
• PEFR less than 50% predicted.
• Wheezes. A silent chest is life-threatening.
The management is as follows:
1. Oxygen per face mask.
2. High-dose inhaled SABA, such as salbutamol 5mg or terbutaline 10mg. an oxygen-
driven nebuliser is advised in patients with life-threatening asthma.
3. Systemic corticosteroids. Oral prednisolone once daily for 5 days is preferred.
Alternatively, you can use intravenous hydrocortisone or intramuscular
methylprednisolone.
4. Nebulise ipratropium bromide.
5. Magnesium sulphate.
COPD is believed to affect 10-20% of individuals over 40 years of age. This is largely
associated with smoking & environmental exposure.
Aetiology
The risk factors for COPD are:
Pathophysiology
1
Wood & paraffin smoke both cause what is called the hut lung disease.
Patients are classified into 2 groups depending on the prevailing disease process.
Clinical features
The clinical features of COPD are:
History
The symptoms may be worsened by cold, foggy weather and atmospheric pollution. You
should also look for:
Examination
The examination findings that you can get with COPD include:
Investigations
• Lung function tests. The lung function test will reveal an obstructive profile. This will
show a reduced FEV1 of less than 80% the predicted for age, as well as FEV1/FVC
ratio of less than 0.7/70%, with individual values for FEV1 & FVC being reduced as
well. Spirometry findings can be used to grade the severity of COPD:
Gold stage % predicted FEV1
Mild ≥80
Moderate 50-79
Severe 30-49
Very severe <30
• Chest X-ray. On chest X-ray, you will see chest hyperinflation with flattening of the
diaphragm. There will be large central pulmonary arteries with reduced lung
markings in the peripheries. You may be able to see bullae in the lung fields. Chest X-
ray is seldom used for diagnosis and is used for identifying other things
• FBC. On a full blood count, there will an elevated haemoglobin level & haematocrit
• ECG. This will demonstrate right atrial & ventricular enlargement due to pulmonary
hypertension. This results from chronic hypoxia, which causes vasoconstriction of
pulmonary vessels.
• Arterial blood gases. This will demonstrate reduced paO2 with/without hypercapnia
(elevated paCO2).
• A1-antitrypsin deficiency screening. This is done mostly in white patients who
develop symptoms before the age of 45 years with a strong family history of disease.
Management
Management of COPD is composed of 2 aspects. The first aspect is management used for
long-term symptom relief and delaying the progression of disease. The second aspect of
management is the management of acute exacerbations of symptoms.
3
A hyper-resonant chest may be a result of an ensuing spontaneous pneumothorax as a result of a ruptured
bulla.
4
This is the drop in the arterial paO2 due to respiratory depression during sleep.
Long-term management
• Mild & moderate disease. For patients with mild & moderate disease (FEV1 is
greater than or equals to 50% of predicted), you can start with a LABA or LAMA. If a
SAMA had been used, it should be discontinued before the LAMA is introduced. You
can combine a LABA with a corticosteroid, and the combination has been
demonstrated to be of greater benefit than either of the drugs used alone.
• Severe disease. For patients with severe disease (FEV1 less than 50% predicted), you
use a LABA/LAMA in combination with a corticosteroid in a combination inhaler. If
the patient remains breathless, you add a LAMA (e.g. tiotropium) to a LABA to make
it triple therapy. If the patient remains symptomatic you should try steroid trial
(30mg prednisolone po for 2 weeks5), theophylline, roflumilast and mucolytic drugs.
• Long-term oxygen therapy (LTOT). This is indicated when the patient has a paO2 of
less than 7.3kPa (55mm Hg), or less than 8kPa (60mm Hg) with cor pulmonale,
polycythemia, peripheral oedema or nocturnal hypoxia.
• Surgery. Surgery is indicated in patients with recurrent pneumothoraxes and isolated
bullous disease.
• Palliative/end-of-life care. This is indicated for advanced cases.
5
If after 2 weeks, the patient’s FEV1 increases by more than 15%, the patient is steroid responsive and can be
continued on an inhaled corticosteroid.
6
This suggests some other aetiology apart from smoking, such as α1-antitrypsin deficiency.
Aetiology
The causes of atelectasis can be classified according to either obstructive causes or causes
that result in diminished alveolar distension. The causes are:
Pathophysiology
Atelectasis develops through 4 main mechanisms:
Clinical presentation
The clinical features of atelectasis are:
• History. Most symptoms of atelectasis are non-specific and are usually related to the
underlying disorder and the degree of lung volume loss. If present alone, the child
may present with tachypnoea to compensate for decreased tidal volume. If serious
enough, the child may present with grunting to create PEEP (positive end-expiratory
pressure). If a child has underlying cardiopulmonary or neuromuscular disease,
sudden decrease in oxygen saturation may be a sign of atelectasis.
• Examination. Breath sounds may be decreased in the affected region of the lung
(changes may not be perceived, however). If the portion is large enough, dullness on
percussion may be elicited. The anterior chest wall must be auscultated for possible
atelectasis, since the right middle lobe and the lingula of the left lung are best heard
from the anterior chest wall.
Investigations
The investigations to do in a person with atelectasis are:
• Arterial oxygen saturation. This can be done using pulse oximetry or by measuring
arterial blood gases for hypoxaemia.
• Pulmonary function tests. These may detect obstruction, restrictive causes or
decreased respiratory muscle pressures.
• Imaging. A chest X-ray is often the first to reveal atelectasis. It may fail, however, to
detect small areas of lung collapse. CT scanning may help in assessing compression
of the airway or other underlying pathology that may cause collapse. Lung
ultrasound is also accurate & reliable in diagnosing atelectasis, particularly in
neonates.
• Chest physiotherapy. This has been used for children on mechanical ventilation. A 4
step procedure can be employed – it involves: bagging with 100% oxygen;
endotracheal instillation of 0.25-0.5ml/kg sterile saline; bagging with momentary
inspiratory hold; and then release of hold and simultaneous forced exhalation &
vibration. This stimulates coughing, and the phlegm can be suctioned.
• Pharmacotherapy. The medications in use include DNAse, bronchodilators and
surfactant. DNAse has been used successfully in children with cystic fibrosis and
other patients with acute atelectasis. It improves flow properties & clearance of
mucus. Success of the medication, however, depends on the amount of DNA in the
secretions. N-acetylcysteine is also used as a mucolytic.
• Fibre-optic bronchoscopy. This can be used if the degree of lung collapse is severe
and response to therapy is suboptimal. It has both diagnostic & therapeutic value.
Flexible bronchoscopy helps define the nature of intrinsic obstruction and distinguish
it from extrinsic obstruction. You can also employ rigid bronchoscopy to remove
thick mucus plugs, thick secretions and foreign bodies. Surfactant can also be
administered bronchoscopically.
• Treatment of the underlying condition. Children with asthma require oral & inhaled
corticosteroids with frequent inhaled bronchodilators. Antibiotics are not indicated.
Patients with cystic fibrosis, however, require aggressive antibiotic therapy together
with chest physiotherapy and postural drainage. If pain is causing atelectasis,
appropriate pain therapy is indicated.
Pathogenesis
Embolic blood clots mainly originate from thrombi formed in deep systemic veins. Most
blood clots are formed in the iliofemoral veins. It is thought that 50-80% of blood clots in
the iliofemoral veins (iliac, femoral and popliteal veins) originate from below the popliteal
vein and propagate proximally. Thrombi can also form in pelvic & abdominal veins as well as
the axillary vein. Clots can also originate from other sources, such as the right atrium, right
ventricle or septic emboli (from right-sided infective endocarditis).
Clots form as a result of a disturbance of the Virchow’s triad (endothelium, blood flow,
coagulability). This can be due to sluggish flow (which can be generalised or localised to a
precise region), damage to the endothelium and a hypercoagulable state.
Since they vary in size, emboli can lodge in different parts of the pulmonary arterial tree.
Large thrombi obstruct larger proximal vessels and have more devastating effects. Examples
include saddle emboli, which lodge at the junction between the pulmonary trunk and the
pulmonary arteries. Smaller emboli continue travelling downstream and obstruct arterioles.
Examples include showers/microemboli, which are often multiple.
1
This is due to the release of inflammatory mediators.
Risk factors
The risk factors for pulmonary embolism are those for deep vein thrombosis. They are as
follows:
Clinical features
The presentation of pulmonary embolism depends on the size, number and distribution of
emboli. The typical symptoms are:
Pleuritic chest pain & haemoptysis only occur when there has been pulmonary infarction.
Diagnosis
The investigations done for pulmonary embolism are:
Management
2
There may be ST depression instead.
3
This is a type I respiratory failure pattern.
Emergency treatment
Definitive therapy
Prevention
Chest pain
A presenting complaint of chest pain is usually treated as a medical emergency because
ischaemic heart disease normally presents as chest pain (angina pectoris). Chest pain is a
common presentation of patients with heart problems and chest pain ranges from trivial to
life-threatening. The severity of chest pain is normally unrelated to the severity of the heart
disease. Chest pain due to heart disease develops as a result of accumulation of metabolites
released by injured myocardial cells following the obstruction (partial or complete) of a
coronary artery. These metabolites stimulate the cardiac sympathetic nerves. Patients with
heart transplants, therefore, are usually unable to feel angina pain. Patients with diabetes
may be diagnosed with silent infarcts.
When a patient presents with chest pain, it is important to determine whether the pain is of
cardiac origin or not. Non-cardiac origin chest pain may be caused by acute pulmonary
embolism, mediastinal conditions or pneumonia. To help determine the cause of chest pain,
one needs to assess the four cardinal features: duration, location, quality and precipitating
& aggravating factors. When asking a patient about chest pain, it is normal to ask them
about chest discomfort rather than chest pain, because ischaemic heart disease usually
starts by causing discomfort. The pain/discomfort is usually central rather than left-sided.
The pain may radiate to the shoulder, medial arm, neck & jaw and it rarely reaches below
the umbilicus.
Angina pectoris
Angina is the most common cardiac chest pain experienced by patients. It is usually because
of myocardial ischaemia of a coronary artery, by may also be caused by aortic stenosis or
hypertrophic cardiomyopathy which increased cardiac oxygen demand. It is a diffuse pain or
discomfort which is felt in the centre of the chest and lasts about 10 minutes. It is
unaffected by inspiration, twisting or turning. Stable angina is triggered by exercise, cold or
windy weather, walking uphill or carrying a heavy load and exercise following a heavy metal
meal. All these causes result in either an increase in oxygen demand, an increased force of
Angina is relieved by rest and by glycerol trinitrate. In some instances, warming up relieves
symptoms as warming up results in peripheral vasodilation resulting in reduced resistance.
Myocardial infarction
This is a condition in which the heart muscle infarcts (dies) due to the occlusion of an artery.
The condition begins as ischaemia which progresses to infarction. The symptoms of
myocardial infarction are similar to those of angina pectoris, but they are more severe and
last longer. The development of unstable angina (angina developing at rest) usually signifies
the onset of myocardial infarction. In addition to angina, other presenting symptoms include
restlessness, breathlessness and a feeling of impending death. Autonomic stimulation
produces sweating, pallor, nausea, vomiting and diarrhoea. Pain is absent in up to 30% of
patients.
• Pericardial pain. Pericardial pain is caused by inflammation of the pericardial sac and
may coexist with angina (it occurs on the anterior chest wall). This may be secondary
to myocardial infarction, viral infection, after surgery, catheter ablation, angioplasty
or radiotherapy. Pericardial pain is exacerbated by inspiration and movement,
particularly leaning forward. Pericardial pain is a form of pleuritic chest pain.
• Pleuritic pain. This pain is similar to pericardial pain. However, it is caused by either
inflammation of the pleura as a primary problem or secondary to pneumonia and
pulmonary embolism. It is also worsened on inspiration.
• Aortic dissection. Tearing of the intima of the aorta results in blood gushing into the
medial layer, causing tearing. This is called a dissection. An abrupt tearing chest pain
occurs which can radiate to the back depending on the origin & extent of dissection.
It is also associated with profound autonomic stimulation. Predisposing factors
include Marfan’s syndrome & hypertension.
• Massive pulmonary embolism. This causes pain of very sudden onset. The pain may
be retrosternal and associated with collapse, dyspnoea and cyanosis. It is often
pleuritic, but can be confused with angina especially if associated with right
ventricular ischaemia.
• Spontaneous pneumothorax. This produces a sharp localised pain and severe
dyspnoea.
• Gastro-oesophageal reflux disease. This typically causes a sharp angina-like pain
without heartburn. Oesophageal spasm may cause retrosternal chest pain or
discomfort.
Breathlessness (Dyspnoea)
Breathless is another common cardiovascular symptom. It is also a common respiratory,
neuromuscular & metabolic symptom and it can also be caused by toxins and anxiety.
Dyspnoea is normal in people after they exercise. However, it is pathological if it occurs at a
significantly lower threshold than expected.
Dyspnoea due to ischaemic heart disease has similar precipitations to angina. It may also be
associated with extreme fatigue. The relieving symptoms depend on the causative agent. If
the dyspnoea is caused by pulmonary oedema resulting from left heart failure, the patient
prefers to sit upright. Those with dyspnoea coming from pulmonary embolism prefer to lie
flat and may faint if they sit uptight.
Dyspnoea can be graded using the New York Heart Association (NYHA) grading system:
Orthopnoea
This is dyspnoea that occurs when the patient is lying flat on their back, and is relieved when
the patient sits up. This kind of dyspnoea is a sign of heart failure, and lying on one’s back
increases venous return. This increases the volume of blood that the pulmonary circulation
deals with and because there is left ventricular failure, there is increased left atrial pressure,
leading to pulmonary oedema. This is the cause of the breathlessness. Furthermore, sitting
upright redistributes the fluid in the lungs so that the upper regions of the lungs have less
fluid & are more aerated. The severity of the condition can be gauged by the number of
pillows that the patient requires to sleep on in order to relieve the orthopnoea.
Palpitations
This is an unexpected awareness of the heart beating in the chest. The patient usually
experiences a rapid, forceful & irregular heartbeat. They usually occur in sinus rhythm with
anxiety, intermittent irregularities of the heartbeat such as extra beats or dropped beats.
Palpitations are normally associated with cardiac arrhythmias and/or sinus tachycardias.
To investigate this heartbeat, you ask the patient to tap out the pattern of beats they can
feel. If the heartbeat is rapid, it is important to ask the patient if the palpitations are of
sudden or gradual onset & offset. Cardiac arrhythmias are associated with sudden onset &
offset, while sinus tachycardia is associated with gradual onset & offset. It is also beneficial
to ask the patient about the precipitating factors as well as the frequency & duration of the
palpitations. The supraventricular (sinus) tachycardia can be reversed using the Valsava
manoeuvre.
• Postural hypotension. This is a condition in which the systolic blood pressure falls by
more than 20mm Hg upon standing. It is commonly caused by hypovolaemia and
antihypertensive drugs. It is also a symptom of autonomic neuropathy.
• Neurocardiogenic syncope. This is a group of conditions that are caused by
abnormal autonomic reflexes. It occurs in healthy people forced to stand for a long
time in a warm environment or subjected to painful or emotional stimuli. If the
patient is held in an upright position, continued cerebral hypoperfusion precipitates
a cerebral anoxic seizure.
Ankle swelling of cardiac origin is usually symmetrical and is worsened in the evening with
improvement during the night. As heart failure progresses, the oedema progresses from the
ankles to the legs, thighs, genitalia and abdomen.
As with every examination, start with the WIPER sequence: wash your hands, introduce
yourself, gain permission to conduct the examination, expose your patient appropriately
and recline the seat to the appropriate angle. In a cardiovascular examination, you expose
your patient completely up to their waste (in a female patient, you want to delay this until
you are examining the precordium). The bed should be reclined to 45°.
After this, you move on to the examination. The examination of the cardiovascular system
consists of a general examination, arterial pulses, blood pressure, jugular venous pulse and
precordial examination. On examination, you start with inspection from the end of the bed,
followed by a general examination incorporating cardiovascular signs. You examine the
hands, arms, axillae, neck & face. You then move onto the IPPA sequence (inspection,
palpation, percussion & auscultation) although percussion is unnecessary in a cardiovascular
examination.
General inspection
From the end of the bed, you want to comment on the patient first before you comment on
their surroundings. Comment on the general status of the patient: are they in any distress?
Are they lying comfortably on the bed? Are they conscious & fully aware of their
surroundings? Do they have obvious oedema? Do they have an obvious scar on their chest1?
• Comment on whether the patient is cachexic (severe weight loss with muscle wasting).
This may be a sign of a malignant disease, but severe cardiac failure may also cause this
condition (cardiac cachexia). It may be a combination of anorexia, impaired intestinal
absorption and increased levels of inflammatory chemokines such as TNF-α.
• There are other syndromes associated with cardiovascular disease, such as Marfan’s
syndrome, Down’s syndrome and Turner’s syndrome.
After commenting on their general appearance, comment on their surroundings: look for
paraphernalia relevant to the cardiovascular system, such as fluid restriction sign above the
bed, obvious weight chart
1
If they have a scar on their chest indicating a sternotomy, look at their legs for evidence of harvesting of the
saphenous vein.
• Temperature & general feel of the hand. The hand should feel warm, although it may
feel cold in cold weather. However, you should be able to look at the colour of the hand
and also exclude other indications of ischaemia. The hands may be clammy or
diaphoretic. Cold clammy hands must make you suspicious of shock.
• Clubbing. Clubbing is an increase in the soft tissue in the distal part of the fingers & toes.
The true mechanism is unknown, but it is suspected to be a result of release of platelet-
derived growth factor (PDGF) by platelets & extra-medullary megakaryocytes lodged in
the nail beds. This PDGF causes proliferation of fibro-vascular tissue. The most common
cardiovascular condition manifesting as clubbing is cyanotic congenital heart disease and
infective endocarditis. Other conditions that are not as common are bronchial
arteriovenous aneurysm and axillary artery aneurysm2. Clubbing is confirmed by
Schamroth’s sign (the disappearance of the diamond sign between the nails when
corresponding nails are placed together) and an increased interphalangeal depth ratio.
This is the ratio between the distal phalangeal depth (anteroposterior length of the
distal phalanx at the point where the nail “joins” the skin) and the interphalangeal depth
(anteroposterior length at the distal interphalangeal joint). The interphalangeal depth
ratio is ≥1.
• Splinter haemorrhages. These are linear haemorrhages lying parallel to the long axis of
the nail. In the cardiovascular system, they are due to infective endocarditis. They are
also due to trauma (especially in manual workers), scleroderma and systemic lupus
erythematosus. They are thought to be caused by emboli causing blood clots in
capillaries under the nail, thus resulting in haemorrhage. These, however, are only seen
in 15% of cases and are therefore not very sensitive. One or two splinter haemorrhages
are normal and therefore you should also look at the toes to confirm.
• Osler’s nodes. These are red raised tender palpable nodules on the pulp of the fingers or
toes or on the thenar & hypothenar eminences. Their incidence is 5-25%. They are
thought to be a result of underlying immunologic or vasculitic process, and histologic
evidence reveals the likelihood of an embolic process. The condition is also present in
SLE, gonococcal infection and the region distal to an arterial catheter.
• Janeway lesions. These are red non-tender haemorrhagic macules or papules on the
palmar aspect of the hand, especially on the thenar & hypothenar eminences. It is most
commonly found in bacterial endocarditis. Their mechanism is not fully understood,
although it is believed that they are caused by septic micro-emboli deposited in
peripheral sites. It has also been shown that an immunological vasculitic process could
be developing. They appear in 4-10% of patients.
2
These 2 conditions cause unilateral clubbing.
Arterial pulses
Normally, it is easiest to feel the pulse of the radial artery, and this pulse can be
characterised.
• Locating pulses. The radial pulse is located on the distal end of the radius just lateral to
the flexor carpi radialis tendon. The brachial pulse is detected in the antecubital fossa
medial to the tendon for biceps brachii. The carotid pulse can be detected by placing
your fingers between the angle of the jaw and the anterior margin of the
sternocleidomastoid muscle (alternatively, you can tuck your fingers beneath the
sternocleidomastoid muscle just lateral to the trachea). The femoral pulse is felt below
the inguinal ligament midway between the anterior superior iliac spine and the
symphysis pubis. The popliteal pulse is felt at the level of the knee creases. The posterior
tibial artery is felt 2cm below & posterior to the medial malleolus. The dorsalis pedis
artery is felt at the proximal end of the 1st intertarsal space lateral to the tendon of the
flexor hallucis longus.
• Radial pulse. You should make a number of observations based on this pulse. To
measure pulse rate, you count the number of beats you feel in 30 seconds and multiply
by 2 (or count the number of beats in 15 seconds and multiply by 4). Patients may also
have a pulse deficit, which is a pulse that is higher when listened to using a stethoscope
than when counted on pulsation. This may be present in ectopic heart beats or in atrial
fibrillation.
• Rhythm of pulse. The rhythm of the pulse should also be described (regular or irregular)
as well as the character of the pulse. The rhythm may be regularly irregular. There may
be ectopic beats that come after every normal beat (bigeminy) or every 2 normal beats
(trigeminy). There may also be an inspirational sinus arrhythmia in which the heart rate
Neck
In the neck, you look for:
• Jugular venous pressure. The jugular pressure is measured with the patient lying down
on the bed at an angle of 45°. It is measured by placing 2 rulers perpendicular to each
other: one vertically at the sternal notch, another horizontally at the uppermost part of
the waveform. You then read the height on the vertical ruler where it is crossed by the
horizontal ruler.
• Carotid pulse. The carotid pulse is inspected & palpated. If it is visible on inspection, it is
a sign of aortic regurgitation and is called Corrigan’s sign. You then test the volume &
character on both sides.
Face
The inspection of the face involves the inspection of the eyes, face & mouth. There are
different signs to look for. In the eyes, the signs to look for are:
• Malar flush. This refers to rosy cheeks with a blue tinge to them. It is due to dilation
of malar capillaries. It is associated with pulmonary hypertension & low cardiac
output, such as occurs in mitral stenosis. This, however, is now rare.
• High arched palate. A high arched palate is found in Marfan’s syndrome. This
syndrome is associated with congenital heart disease (including aortic regurgitation
secondary to aortic root dilation and mitral regurgitations due to mitral valve
prolapse).
• Dentition. Diseased teeth may be a source of infection which could cause infective
endocarditis.
• Pallor. This is seen in the tongue & floor of the mouth. The tongue appears pink. It is
a sign of anaemia, and the tongue is a sensitive indicator of pallor as it is well
perfused.
• Central cyanosis. Central cyanosis is indicated with a dark purplish tongue. Central
cyanosis is due to reduced oxygenation of arterial blood, resulting from respiratory
disease or from right-to-left shunting of blood in the heart. It may also be due to
heart failure.
Precordium
Inspection
On inspection, you look for scars on the chest. The location of the scar is important as it
gives an indication to the type of procedure carried out.
Skeletal abnormalities such as pectus excavatum and kyphoscoliosis may be present. These
may distort the position of the heart and its apex. You also look for pacemakers or
cardioverter-defibrillator boxes.
Palpation
There are a number of items that are palpated for:
• Apex beat. The apex is located by first placing the whole palm of the hand on the left
side of the chest and then localising the apex using two fingers. When it has been
localised, you determine the intercostal space in which it is found. The apex is only
palpable in about 50% of adults. The apex is normally palpable in the 5 th intercostal
space in the mid-clavicular line3.
The character of the apical beat should be determined also. The normal character
gently lifts the fingers. Other characters include:
o Pressure loaded/heaving. This is a forceful & sustained impulse and may
occur with left ventricular hypertrophy. It occurs in aortic stenosis,
hypertension and hypertrophic cardiac myopathy.
o Volume loaded/thrusting. This is a displaced, diffuse, non-sustained impulse.
It occurs in aortic regurgitation or dilated cardiomyopathy.
3
This is not the anatomical apex. It is a point above. The palpable apex is felt because when the ventricle fills
up, it assumes a spherical shape and this part moves towards the chest wall & can be felt. The true apex is a
point below it and it swings away from the chest wall when the heart fills up.
Auscultation
The heart is auscultated with both the diaphragm and the bell of the stethoscope. Left-sided
murmurs (mitral & aortic) are best heart during expiration while right sided murmurs are
best heard during inspiration. The areas to listen to are:
• Heart sounds. The apex is the first part auscultated in order to determine the
character of the heartbeat. Normally, only the 1st & 2nd heart sounds (S1 & S2) are
heard. You should simultaneously palpate the carotid artery in order to determine
which one is the first heart sound. This allows you to time any murmurs you hear.
Listen for S1 & S2 and determine if S3 & S4 are present. You should do the same for
all the other valve areas.
• Mitral area. This is the apical area. You should listen for any murmurs, which are
louder on expiration. Place the bell of your stethoscope and then ask the patient to
roll over to their left side. This brings the apex – and consequently the mitral valve –
closer to the chest wall, allowing it to be more audible. If the murmur is heard louder
using the bell, the murmur is most likely due to mitral stenosis and it is heard as a
rumbling mid-diastolic murmur. You then adjust the stethoscope and listen using the
While the patient is sitting forwards, auscultate the lung bases to hear for bibasal crackles.
These can signify pulmonary oedema secondary to congestive cardiac failure. You must also
feel for sacral oedema.
• Auscultating the lung bases. Auscultate the lung bases to look for bibasal
crepitations. These signify congestive heart failure. While you do this, you look for
sacral oedema, which indicates extensive oedema if lower limb oedema is present.
• Abdomen. Look for a tender hepatomegaly. This indicates right heart failure. It is
pulsatile in tricuspid regurgitation. Also look for a splenomegaly which may be
present in infective endocarditis. Ascites may occur in severe right heart failure. Also
feel for the pulsations of the abdominal aorta, which may be present in an
abdominal aortic aneurysm.
• Lower limbs. Check for bipedal oedema.
1. Sinus rhythm.
2. Rate.
3. Axis.
4. Different ECG segments & intervals and corrected QT interval (QTc).
5. Chamber size.
6. Bundle branch blocks.
7. Other abnormalities where present.
Sinus rhythm
The sinus rhythm is determined using the rhythm strip. Every ECG should have a strip that
has a single lead recorded for 10 seconds. This strip usually records either lead II or V1. The
rhythm strip is also used to calculate the heart rate. The rhythm strip is used to determine
the presence of a sinus rhythm because it provides a more holistic picture of the conduction
of the heart and is therefore reliable.
A sinus rhythm is one in which the P-wave is always followed by a QRS complex. The
presence of a sinus rhythm means that the source of the heart’s impulses is the SA node,
with normal progression of cardiac impulses to the AV node, His-Purkinje system and
ventricular myocardium.
In addition to establishing the sinus rhythm, it is also important to establish the regularity of
the heartbeat. Check (using the rhythm strip) if the RR intervals are constant. If not, use the
card technique or just count the number of small boxes between successive R waves. If the
R interval is irregular, this is called sinus arrhythmia. This is sometimes normal, particularly
in young people, as heart rate increases with inspiration and decreases with expiration. The
difference, however, between the largest and smallest RR intervals should be 80-120ms
(which is 2-3 small boxes).
There is also need for a delay between the T-wave and the subsequent P-wave. If this delay
disappears, the patient is said to have a premature beat. If this premature beat originates
from the atria, it has a P-wave with a normal duration QRS complex, since it mimics the
normal flow of impulses from the atria to the ventricles. However, if the premature beat is
Rate
The heart rate can be calculated from the ECG as well. Since the ECG has standard
progression of 25cms-1, it means that you can employ the same method of calculating the
heart rate with any ECG. To find the heart rate using the ECG, you can do the following:
1. You can count the number of large squares (5cm×5cm or 0.2s) in the RR interval and
divided 300 by that number. For example, if there are 4 large squares, you divide 300
by 4, giving you 75 beats per minute. This is because there are 300 large boxes in 60
seconds and therefore you use the period (RR interval) to determine with frequency
of heartbeats.
2. You can count the number of small squares in the RR interval, multiply it by 0.04s
and then divided 60 by the answer. For example, in the above example, 4 large
squares have 20 small squares. Therefore 20 × 0.04 = 0.8, then 60 ÷ 0.8 = 75 beats
per minute.
3. If the rhythm is irregular, i.e. the RR interval is not constant, you count the number
of R-waves in the rhythm strip and then multiply it by 6 (since the rhythm strip is 10s
long).
The normal heart rate in adults is 60-100 beats per minute. This is due to the high intrinsic
firing rate of the SA node, which would be higher if it were not under tonic control by the
vagus nerve. Taking the rate is important, particularly in conjunction with establishing the
sinus rhythm, because it helps determine the source of pacing of the heart. A normal heart
rate with a sinus rhythm means that the SA node is the pacemaker of the heart. You can get
a sinus rhythm that is either too fast or too slow, leading to sinus tachycardia or
bradycardia. You can also get a rate that is either high or low that not a sinus rhythm. It
could be due to SA nodal block (nodal rhythm), AV block or other arrhythmias, such as atrial
& ventricular fibrillation, atrial flutter, ventricular & supraventricular tachycardia and other
arrhythmias.
Axis
The axis of the heart is the net vector of ventricular forces doing ventricular depolarisation.
It is generally goes anteriorly & infero-laterally to the left for a number of reasons:
1. The SA node, where the impulses of the cardiac potential in the heart originate, is
located in the right atrium, and impulses are conducted from the right atrium to the
left atrium through the Bachman’s bundle. This gives a net vector of impulses in the
leftward direction.
The heart axis, however, is not a constant figure for everyone, as there are inter-individual
variations. The axis, however, deviates when there is left or right ventricular enlargement,
particularly from hypertrophy. It can also deviate to the right/left if there is a disturbance in
the normal progression of impulses of the heart, as in bundle branch blocks. The normal
heart axis is -30 to 90°:
aVR
(-150°) aVL
(-30°)
Lead I
(0°)
aVF
(90°)
Therefore, an axis greater than 90° is a right axis deviation while an axis that is less than -30°
is left axis deviation. Left axis deviation occurs in left ventricular hypertrophy, left anterior
hemiblock, inferior MI, ventricular tachycardia from left ventricular focus and some types of
Wolff-Parkinson-White syndrome. Causes of right axis deviation include right ventricular
hypertrophy, anterolateral MI, pulmonary embolism, left posterior hemiblock and some
types of Wolff-Parkinson-White syndrome. Left axis deviation, however, ends at -90°, while
right axis deviation ends at ±180°. Between -90° and -180°, the heart is said to have extreme
right axis deviation.
As a general rule of thumb, if both leads I & II are positive, the axis is normal.
• PR interval. This interval is from the beginning of the P-wave to the beginning of the
R-wave. This represents conduction from the atria to the ventricles, and it is usually
0.12-0.2s or 3-5 small boxes. Delay or shortening of this interval implies delay or
shortening1 of conduction from atria to ventricles.
• QRS complex. This interval is from the beginning of the Q-wave to the end of the S-
wave. It represents ventricular depolarisation. The normal duration of the QRS
complex is 0.08-0.12s, which is 2-3 small boxes. If it is wide, you should suspect
bundle branch blocks.
• QT interval. The QT interval is the interval from the beginning of the Q-wave to the
end of the T-wave. The interval varies with the sinus rate, and therefore it is more
reliable to calculate the corrected QT interval. This is found by dividing the
measured QT interval by the square root of the RR-interval2.
𝑄𝑇
𝑄𝑇 𝑐 =
√𝑅𝑅
The normal QTc interval duration is 0.38-0.42s. The QT interval assesses ventricular
conduction (depolarisation and repolarisation). The most common QT anomaly is
QTc segment prolongation.
• ST segment. This is the portion between the end of the QRS complex and the
beginning of the T-wave. In a normal heart, all the cells of the ventricular
myocardium are depolarised by this stage, and therefore the ST segment is
isoelectric at 0mV. Elevation above 1mm implies infarction while depression of
1
This is usually through an accessory pathway.
2
This is called the Bazett’s square root formula. There is also a Fridericia’s cube root formula in which the RR
interval is cube rooted instead.
Chamber size
The size of the cardiac chambers can be ascertained from the ECG, particularly looking at
enlargement of the chambers.
3
In myocarditis/pericarditis, there will ST elevation in all leads except aVR & V1. In these 2 leads, there will be
ST depression instead (since these leads oppose the cardiac axis).
When there is a bundle branch block, there is a prolongation of the QRS complex to more
than 0.12s. The QRS duration may be normal if the block is partial. There are different types
of heart block that produce different patterns:
• Right bundle branch block. The QRS complex is prolonged. Furthermore, there is a
terminal R-wave in lead V1 (also called R prime), and this produces an M-shaped QRS
complex. There is also a prolonged S-wave (also called a slurred S-wave) in leads I &
V6. There is also T-wave inversion in leads V1-V3/4. There is usually no axis deviation.
• Left bundle branch block. This is a complex left bundle block and therefore the QRS
complex is prolonged. There is a broad monophasic R-wave in V6 that has a notch,
producing an M-shape. There are also inverted T-waves in leads I, aVL, V5 and V6.
There is also a deep S-wave in V1, and this is sometimes accompanied by a deep Q-
wave.
• Left anterior fascicular block. These are seen in about 4% of cases of acute
myocardial infarction. The QRS complex is prolonged, but not to the same extent as
left bundle branch block. There is also an abnormal left axis deviation, usually
between -45° and -60°. There is also a tall R-wave with a small Q-wave in leads I &
4
Some divide the left bundle branch into 3, with a left septal fascicle as the extra fascicle.
5
Intrinsicoid deflection is the time it takes from the beginning of the QRS complex to the peak of the R-wave.
It is also known as the R-wave peak time. It reflects the depolarisation vector from the endocardium to the
epicardium.
Aetiology
The organisms that cause infective endocarditis are:
Pathogenesis
Endocarditis is usually the consequence of 2 factors:
1. The presence of bacteraemia in the circulation. This can occur due to patient-specific
reasons (such as intravenous drug use, poor dental hygiene, and soft tissue
infections) or diagnostic/therapeutic factors (dental procedures, intravascular
cannulae, cardiac surgery, permanent pacemakers). There isn’t good evidence that
bacteraemia leads to endocarditis, however.
2. Abnormal endocardium (which facilitates adherence & growth of bacteria). Damaged
endothelium promotes platelet & fibrin deposition, and this allows organisms to
adhere & grow and create an infected vegetation. Pre-existing valvular lesions may
disturb laminar flow. Jet lesions (from septal defects or persistent ductus arteriosus)
result in abnormal cardiac endothelium.
In both forms, the vegetations are friable, bulky and potentially destructive – vegetations
can erode the underlying myocardium and produce a ring abscess, and can also destroy the
chordae tendinae. They are composed of fibrin, inflammatory cells and micro-organisms1.
The vegetations can embolise, and because they are infected they can cause septal infarcts
& mycotic aneurysms. Embolism to other structures can also produce embolic phenomena.
Infective endocarditis can also lead to formation of immune complexes which can be
deposited in blood vessels and cause vasculitis.
Risk factors
The risk factors for infective endocarditis are:
1
This is where they differ with other vegetative diseases, which have sterile vegetations.
2
For this reason, porphylaxis for dental procedures does not make sense.
3
Poorer outcomes are associated with MRSA, and this is often hospital-acquired.
Clinical features
The clinical features of infective endocarditis are:
History
• Fever & rigors. Despite being absent in sub-acute infective endocarditis, this is the
most consistent symptom. The other symptoms are non-specific and not consistent.
• Night sweats.
• Weight loss.
• Fatigue.
• Malaise.
• Flu-like syndrome.
• Cutaneous lesions, which can be painful.
• Symptoms of congestive heart failure: chest pain, breathlessness, fatigue, oedema,
poor exercise tolerance, etc.
Acute infective endocarditis can present with rapidly-developing fever, chills, weakness and
lassitude.
• General: The patient is usually febrile & anaemic. Patients may have clubbing. They
may also have numerous peripheral abscesses of unknown origin.
• CVS: patient may have insignia of infective endocarditis, such as Janeway lesions,
Osler’s nodes and spinter haemorrhages. Splinter haemorrhages & Osler’s nodes are
vasculitic phenomena, while Janeway lesions are embolic phenomena. Any new
murmur must raise suspicion of infective endocarditis. There is often mitral and/or
aortic regurgitation, or valve obstruction producing stenotic murmurs. Signs of left
ventricular failure should be sought, as this is a common cause of death.
• Fundoscopy: look for Roth’s spots, which are small retinal haemorrhages with pale
centres. Patients may also have conjunctival haemorrhages.
Investigations
• Blood cultures. You do 3 sets of blood cultures at different times from different sites
at peaks of fever. You often have to space the timing of the specimens. 85-90% of
cases will be diagnosed from the first 2 sets. 10% are culture-negative.
• Echocardiography. A transthoracic echocardiogram may show the vegetations but
only if they are greater than 2mm. It is sensitive for detecting valvular dysfunction,
ventricular function and abscesses. Trans-oesophageal echocardiography is more
sensitive, and it is better for visualising mitral valve lesions and identifying aortic root
abscesses. It is also more sensitive for assessing prosthetic valves.
• Urinalysis. Haematuria is found in 70% of cases of infective endocarditis and should
be done to rule out glomerulonephritis. Proteinuria can also occur.
• ECG. On an ECG, if a patient has a prolonged PR interval or heart block, suspect
aortic root dilatation.
• Blood tests. FBC may reveal a normocytic normochromic anaemia. There is also a
leukocytosis and thrombocytosis/thrombocytopaenia. On LFTs, there is elevated
ALP. On U&Es, there is elevated urea & creatinine. There is also elevated ESR & CRP.
• Serologic tests. These should be considered for culture-negative cases to diagnose
Coxiella, Chlamydia, Bartonella and Legionella.
• Chest X-ray. This may show cardiomegaly with pulmonary oedema or pulmonary
emboli/abscesses.
Major criteria
Minor criteria
The diagnosis of infective endocarditis is made when there are 2 major criteria met, or there
is 1 major criterion & 3 minor criteria, or 5 minor criteria. If there is 1 major & 2 minor
criteria, or there are 3 minor criteria alone, infective endocarditis is probable and not
definite.
Treatment
Infective endocarditis is managed medically & surgically
Medical management
Surgical management
It is estimated that 33.4 million people across the world have rheumatic heart disease. It is
more common in children & young adults, with a peak age of 5-15 years of age. It is more
common in developing countries: Middle East, Far East, Eastern Europe and South America.
Its incidence is related to hygiene levels and the use of antibiotics.
Pathogenesis
The disease is often initiated by infection with group A streptococci, such as Streptococcus
pyogenes. S. pyogenes can be a normal resident of the oropharynx, and therefore
pharyngitis is a precipitating factor for rheumatic fever. Other infections (such as
streptococcal skin infections) can also cause rheumatic fever. However, rheumatic fever
often develops 2-4 weeks after the primary pharyngeal infection. The risk of developing
rheumatic fever after an acute pharyngitis is 0.3-3%.
In rheumatic fever, it is not the primary disease that causes damage. Rather, it is antibodies
formed against streptococcal antigens that cross react with body antigens and cause
damage. This is an example of molecular mimicry. The antibodies are usually against cell
wall M proteins and streptolysin O. in the different tissues, different proteins are affected.
In the heart, the antibodies react with cardiac myosin & laminin. In the brain, the antibodies
react with brain gangliosides. T-helper 1 & cytokine Th17 appear to be the key mediators of
the disease. There is a suspected genetic susceptibility that accounts for the likelihood of
developing rheumatic fever.
The average duration of an acute rheumatic fever attack is 3 months. A chronic attack
normally lasts 6 months or longer.
Clinical presentation
The clinical information you obtain from a patient with suspected rheumatic fever is:
History
• A history of a sore throat or scarlet fever. Only 35-65% of patients recall this,
however. If antibiotics were taken for the sore throat, the risk is lower.
Examination
• CVS: signs of heart failure, new/changed heart murmurs (usually mitral & aortic
regurgitation), pericardial rub. You can also hear a Carey-Coombs murmur, which is
an apical low-pitched mid-diastolic flow murmur.
• Musculoskeletal: warm, swollen, tender joints in frank arthritis. There may be
arthralgia alone, without other signs of inflammation.
• Neurological examination: patients will appear fidgety, and display spasmodic
unintentional movements. they will have lack of coordination, inability to sustain
tetanic contraction, such as clenched fist. Speech is often unaffected. When you ask
them to extend their arms & wrists, their hands & wrists form a typical eating fork
appearance. Shining light into their eyes produces a hippus, which is alternating
constriction & relaxation of the pupil.
• Skin: patients may have subcutaneous nodules found primarily over bony surfaces &
tendons (on extensor surfaces). Patients may also have erythema marginatum
lesions which tend to demonstrate advancing erythematous margins with clearing of
the centre.
Investigations
• Throat swabs. You can culture these to reveal group A Streptococcus species. This,
however, is usually negative by the time symptoms of rheumatic fever appear.
To diagnose rheumatic fever you need evidence of streptococcal infection with either 2
major criteria or 1 major and 2 minor criteria.
Management
Management of rheumatic fever is as follows:
More than 50% of patients will develop chronic rheumatic heart disease later (10-20 years
later). Recurrence is precipitated by further infections, pregnancy and use of combined oral
contraceptive pills.
Prevention
Prevention of rheumatic fever is divided into primary prevention, secondary and tertiary
prevention.
Primary prevention
Primary prevention aims at preventing the development of rheumatic fever. The measures
include improving housing conditions as well as preventing progression of the causative
infection. Housing conditions are improved by improving the quality of housing as well as
reducing overcrowding and providing primary healthcare facilities. For those that develop
diseases such as streptococcal throat infections. Streptococcal infections need to be
adequately treated within 1 week of onset of symptoms to prevent the development of
rheumatic fever. You can give any one of the following:
• A single dose of benzathine penicillin G 100 000IU for those below 30kg or 200 000IU
for those above 30kg intramuscularly.
• Oral penicillin V 50mg/kg/day qid for 10 days. it should be given before meals and
the course should be completed.
Secondary prevention
Generally there is little secondary prevention available for detection of subclinical disease
and institution of preventive measures.
Tertiary prevention
Once rheumatic fever has occurred, the most important aspect of treatment is to prevent
recurrences as each subsequent attack inflicts more damage on the heart valves and
therefore predisposes to infective endocarditis & heart failure. Adolescents & young adults
are at risk of developing new infections. Prevention is achieved by antibiotic prophylaxis.
• Oral penicillin V 250mg twice daily. This, however, is not very successful in providing
complete protection.
• Oral sulphonamides 0.5-g twice daily or erythromycin 250mg twice daily in patients
that are allergic to penicillin. Patient compliance must be ensured.
Aetiology
The majority of cases of aortic stenosis are a result of disease of the aortic valve. The most
common diseases of the aortic valve that cause stenosis are:
• Calcified aortic valvular disease (CAVD). This is the commonest of aortic stenosis
and occurs mainly in the elderly. It is a result of an inflammatory process involving
macrophages & T-cells, with initial thickening of the sub-endothelium with adjacent
fibrosis. The lesions contain lipoproteins which calcify, thus increasing leaflet
thickness and reducing systolic opening. The risk factors for CAVD are: old age, male
gender, elevated lipoprotein (a) & LDL, hypertension, diabetes and smoking.
• Bicuspid aortic valve. This is a congenital heart disease that occurs in 1-2% of live
births; it is familial in many cases. Patients with CAVD of a bicuspid valve tend to
present earlier than those of a tricuspid one. Bicuspid aortic valve is associated with
coarctation of the aorta, aortic root dilatation and potential aortic dissection. These
patients should therefore receive regular follow-up echocardiography.
• Rheumatic fever. Rheumatic fever can produce progressive fusion, thickening and
calcification of the aortic valve. The aortic valve is affected in 30-40% of cases of
rheumatic fever, and there is usually associated mitral valve disease.
Other causes of aortic valvular disease include: chronic kidney disease, Paget’s disease of
bone, previous radiation exposure and homozygous familial hypercholesterolaemia.
Other forms of aortic stenosis do not involve the aortic valve. They include:
Pathophysiology
1
This is found in William’s syndrome, which is a congenital disease.
Clinical presentation
Patients are typically elderly patients. There are usually no symptoms until the obstruction is
1
moderate to severe (when the orifice is reduced to 3 of its original size). At this stage, the
patient develops exercise-induced syncope, angina and dyspnoea. The angina develops due
to myocardial ischemia. The syncope is a result of hypo-perfusion to the brain secondary to
hypotension. Patients may also present with heart failure.
When symptoms occur, prognosis is generally poor, and patients die within 2-3 years if no
surgical intervention is offered.
Investigations
The investigations to be done in patients with aortic stenosis are:
2
It can also be heard at the left sternal edge.
Management
In patients with aortic stenosis, the presence of symptoms is a good indicator of severity.
Without treatment, prognosis of symptomatic disease is poor. If patients present with
angina or syncope, the mean survival is 2-3 years. Those with heart failure, however, survive
for an average duration of 1 year. If the disease is moderate-to-severe and is treated
medically, mortality can be as high as 50% at 2 years.
3
The normal valve area is 3-4cm2
Asymptomatic patients should be under regular review for assessment of symptoms and
echocardiography.
Classification
The classification of myocarditis according to Lieberman is as follows:
In the acute phases, hearts with myocarditis are flabby with focal haemorrhages. In chronic
myocarditis, the hearts are enlarged & hypertrophied. Histologically, there is an
inflammatory infiltrate in the heart. In viral infections, lymphocytes predominate; in
bacterial infections, PMN leukocytes predominate; and in allergic & hypersensitivity
reactions, eosinophils predominate.
Aetiology
The agents causing myocarditis cause damage by either of 4 mechanisms:
Clinical presentation
Myocarditis may be acute or chronic, and it ranges in presentation from being
asymptomatic to fatigue, palpitations, chest pain and dyspnoea, to fulminant congestive
cardiac failure with/without cardiogenic shock. In viral myocarditis, patients may present
with a recent history of flu-like symptoms: fever, arthralgia, malaise, or pharyngitis,
tonsillitis and upper respiratory tract infection. Suspect myocarditis in a patient with prior
history of heart disease.
On examination, patients may develop signs of heart failure, such as tachycardia, gallop,
raised JVP & distended neck veins, mitral regurgitation and oedema. In those with
concomitant pericarditis, there may be a pericardial rub. The first heart sound (S 1) is often
soft.
1
Myocarditis is found in about 20% of HIV patients at post mortem, but only 10% of these develop symptoms.
2
This often causes toxin-induced heart block.
• ECG. This reveals ST elevation/depression with T-wave inversion. You may also find
atrial arrhythmias or AV block with Chaga’s disease, diphtheric myocarditis or Lyme
disease. You can also get changes mimicking an MI (called a pseudo-infarction
pattern), and these are associated with a poor prognosis. Right bundle branch blocks
are very common in myocarditis.
• Chest X-ray. This may demonstrate heart enlargement depending on the stage &
virulence of the organism.
• Echocardiography. This is done to exclude other causes of heart failure and to
evaluate the degree of cardiac dysfunction. It is also used to distinguish between
fulminant & acute myocarditis by measuring the left ventricular diastolic dimensions
and septal thickness3.
• Cardiac enzymes. Positive troponin I or T will indicate presence of myocarditis. Anti-
myosin scintigraphy (using anti-myosin antibodies) helps identify the presence of
disease.
• Endomyocardial biopsy. This is the criterion standard for diagnosis of myocarditis. It
is reserved for cases of deteriorating acute heart failure of unknown origin that is not
responding to treatment. The procedure, however, has limited sensitivity &
specificity as inflammation can be diffuse or focal, and adverse events can occur in
6% of cases.
• Viral antibodies & PCR. Antibodies to common aetiological viruses can be taken
from serum. Antibody diagnosis, however, relies on identification of acutely rising
titres. You can do PCR studies on the biopsy sample taken from the heart.
Management
Because many cases of myocarditis are not clinically obvious, a high degree of suspicion is
required. Many patients have mild disease and recover with supportive care. Fulminant
cases, however, need to be managed in tertiary care facilities.
3
In fulminant myocarditis, the diastolic dimensions of the left ventricle are normal while the thickness of the
septum is increased. In acute myocarditis, however, the left ventricle’s diastolic dimensions are increased
while the septal thickness is normal.
After management, patients should be encouraged to have adequate bed rest, and should
avoid sporting activities for 6 months. Patients should be monitored every 1-3 months until
they fully recover. Any evidence of residual cardiac dysfunction or remodelling should be
treated as chronic hear failure.
4
Digoxin should be used with caution as it promotes expression of pro-inflammatory cytokines and increases
mortality rate.
Acute pericarditis
This refers to inflammation of the pericardium. Acute pericarditis has numerous aetiologies,
although in most cases the cause is not identified (idiopathic1). The causes of acute
pericarditis are:
1
This, however, is assumed to be viral.
Clinical presentation
• Chest pain. The chest pain is central, sharp and exacerbated by movement,
respiration and leaning forward, and relieved by sitting down. It may also be referred
to the neck & shoulders.
• Fever & malaise. This may be present, particularly in tuberculosis.
On examination, you will most likely find a pericardial friction rub occurring in 3 phases
(triphasic): atrial systole, ventricular systole and ventricular diastole. It may also be biphasic
(to-and-fro). The friction rub is best heard with the diaphragm of the stethoscope placed at
left lower sternal edge at the end of expiration with the patient leaning forward. Features of
pericardial effusion or tamponade may be present.
Investigations
Treatment
Types of effusion
2
About 20% of patients with acute pericarditis go on to develop recurrent idiopathic acute pericarditis. It may
be incessant (occurring during the 6-week weaning period from NSAIDs) or intermittent (occurring after 6
weeks, which is after weaning)
Clinical presentation
The features of cardiac tamponade are important and should be picked on examination. The
triad of symptoms (Beck’s triad) is:
• Hypotension.
• Raised JVP, with a sharp rise & y-descent. This is called Friedreich’s sign. There is also
a Kussmaul’s sign, which is a rise in JVP or neck vein distension during inspiration.
• Muffled heart sounds.
Investigations
• ECG. You can get low-voltage QRS complexes, flattened T-waves and electrical
alternans (alternating QRS morphologies). There may also be sinus tachycardia.
• Chest X-ray. This will show cardiomegaly with a rounded cardiac contour. The
pulmonary veins are not distended.
Treatment
You must avoid giving diuretics, as they cause a further decrease in cardiac output. IV fluids
can be given.
Constrictive pericarditis
Constrictive pericarditis is a condition characterised by a rigid pericardium due to fibrosis &
calcification. It usually develops secondary to acute pericarditis. The causes include:
• Idiopathic.
• TB.
• Haemopericardium.
• Bacterial infection.
• Rheumatic heart disease.
• Open heart surgery.
• Drugs, e.g. dopamine agonists (cabergoline, pergolide).
• Radiation.
• Uraemia.
• MI.
In most cases, the changes do not cause symptoms or interfere with cardiac function.
However, if the changes interfere with diastolic filling, the pericardium is said to have
constrictive pericarditis.
Clinical features
• Features of right ventricular failure: raised JVP with prominent x- & y-descents,
hepatosplenomegaly, ascites and oedema.
• Features of reduced ventricular filling (similar to cardiac tamponade): Kussmaul’s
sign (engorgement of neck veins during inspiration), Friedrich’s sign (raised JVP with
sharp rise & y descent) and pulsus paradoxus (usually absent).
• Features of left heart failure: dyspnoea, cough, orthopnoea, PND. These are less
common.
• Reduced cardiac output: fatigue, hypotension (although BP is usually normal), reflex
tachycardia.
Patients may have a pericardial knock, which is heard early during diastole at the left lower
sternal border and is due to rapid ventricular filling. 30% of cases have atrial fibrillation.
Investigations
• ECG. This shows low-voltage QRS complexes with generalised T-wave flattening or
inversion. There may be atrial fibrillation.
• Chest X-ray. This will reveal a small heart in relation to the ensuing cardiac failure.
Pericardial calcification is present in up to 50% of patients. A lateral X-ray may be
useful in detecting calcifications missed on an anterior film. If there are no
calcifications, an MRI/CT scan will help distinguish this condition from a
cardiomyopathy.
• Echocardiography. This shows a thickened calcified pericardium and small
ventricular cavities with walls of normal thickness. Doppler studies may be useful.
Treatment
The prognosis is best with idiopathic & infectious causes, and it is worst with post-radiation
constrictive pericarditis. Death usually occurs due to heart failure.
Classification
120
An optimal blood pressure is anything below mm Hg. A systolic blood pressure of 120-
80
129mm Hg or a diastolic blood pressure of 80-84mm Hg is still normal, but not within
optimal range.
Aetiology
There are 2 types of hypertension: primary/essential hypertension or secondary/non-
essential hypertension.
Primary/essential hypertension accounts for the majority (80-90%) of cases. In these cases,
the precise aetiology is unknown, but there are a variety of risk factors that predispose
individuals to developing hypertension. The suggested risk factors for essential hypertension
are:
Secondary hypertension is that which results from another cause, where the defect is not
intrinsic to the vasculature. Causes of secondary hypertension include:
1
You need at least 3 of the 5 features to diagnose metabolic syndrome.
Pathophysiology
The precise pathophysiology is unknown. However, in some young patients there is an
increase in cardiac output associated with an increase in pulse rate and increased
catecholamine levels in the circulation. These are known to decrease baroreceptor
sensitivity, allowing it to tolerate higher pressures.
Diagnosing hypertension
When you meet a patient with suspected hypertension, you take their blood pressure while
they are seated. This should be done after the patient has rested for 5 minutes. The
standing blood pressure should be taken in patients who are diabetic and in elderly subjects
140
to rule out orthostatic hypotension. If their blood pressure is below mm Hg, there is no
90
need to refer for treatment. You should, however, encourage the patient to have their
blood pressure monitored routinely at least every 5 years up to the age of 80.
140
If the clinical blood pressure is above mm Hg, you offer ambulatory blood pressure
90
monitoring, since you want to rule out white collar hypertension. White collar hypertension
refers to blood pressures that rise to hypertensive ranges only after clinical visits. Therefore,
you attach an ambulatory pressure monitoring machine and take blood pressure recordings
over 24 hours. In these patients, hypertension is diagnosed by an ambulatory blood
180
If the clinical blood pressure is above 110mm Hg, the patient needs to be started on an
antihypertensive drug immediately, referred and be put on ambulatory blood pressure
monitoring. The blood pressure parameters for 24-hour ambulatory blood pressure remain
the same.
• Fasting blood glucose & fasting lipid profile. This is to screen for diabetes &
dyslipidaemias.
• ECG. ECGs are done to look for left ventricular hypertrophy and to look for
arrhythmias or past MIs.
• Urinalysis. This may help identify renal disease. Look for proteinuria and haematuria,
and assess renal function. Suspicion of phaeochromocytoma should be followed by
measurement of urinary metanephrines and plasma/urinary catecholamines.
• Urea & electrolytes. If urea & creatinine are both elevated, conduct more specific
renal studies, such as creatinine clearance, renal ultrasound and/or renal isotope
scan or angiography. Low serum potassium may indicate an endocrine disease, and
in this case you need to measure, aldosterone, cortisol and renin levels.
• Echocardiography. This is done when left ventricular hypertrophy or coarctation of
the aorta is suspected.
• MRI. This is done if coarctation of the aorta is suspected.
Management of hypertension
Unless the patient is in the malignant phase of hypertension, there should be a period of
reassessment of the patient with repeated blood pressure measurements together with
advice & non-pharmacological interventions. Treatment is often commenced if blood
160
pressures remain elevated above mm Hg over 4-12 weeks, or if the blood pressure
100
140
remains above mm Hg in a patient with an estimated 10-year cardiovascular risk over
90
20%. However, if there are cardiovascular complications, target organ damage (e.g. stroke,
MI, peripheral vascular disease, renal disease) or diabetes mellitus, start treatment over 1-2
weeks regardless of blood pressure.
Lifestyle modification
Lifestyle modification is used for patients in the initial phase in order to reduce blood
pressures non-pharmacologically. The lifestyle interventions are:
Pharmacological treatment
2
ACE inhibitors are normally reno-protective, because they relax the efferent arterioles and reduce the hyper-
filtration that leads to progressive deterioration in chronic renal disease. however, they also impair renal
autoregulation, and this is a problem in patients with bilateral renovascular disease
The choice of drug depends on the age of the patient and the race. Generally the first-line
agents used in hypertension are: angiotensin inhibitors (ACE-inhibitors & AT1-receptors
antagonists), calcium-channel blockers and diuretics (thiazides). In younger (less than 55
years of age) Caucasian individuals, the first choice is usually an angiotensin blocker due to
the high plasma angiotensin levels in these people. However, in older Caucasians and in
black people, you start with calcium channel blockers or diuretics because in older
Caucasians renin levels drop and in black people renin levels are low while fluid levels are
high.
After this, you add a β-blocker, α-blocker or other diuretic. By this time, a specialist should
be sought.
The drugs used in both hypertensive urgency and hypertensive emergency are:
• Labetalol 20mg IV infusion stat over 2 minutes, then 10-80mg IV infusion every 10
minutes until the desired blood pressure is attained. You can also give 2mg per
minute through continuous intravenous infusion.
• Hydralazine 6.25-25mg PRN until desired blood pressure is attained.
In patients with encephalopathy or stroke, the target blood pressure should be attained
over 4 hours.
Coronary artery syndrome is characterised by dull retrosternal chest pain with various
characteristics. The relation of the chest pain to exertion & rest is important in determining
the type of condition being experienced.
Development of atherosclerosis
In the majority of cases of coronary artery disease, atherosclerosis is the underlying cause.
This is a slow progressive degenerative process which involves gradual obstruction of the
arterial lumen by an atheroma. This atheroma is formed from lipids, macrophages and
smooth muscle, and atheromas predominantly occur in large- & medium-sized blood
vessels.
1. Vascular injury. Chronic vascular injury plays a critical role in the initiation of
atheroma. This can come about by mechanical shear stress (e.g. morbid
hypertension), biochemical abnormalities (elevated cholesterol, diabetes mellitus),
immunologic factors (e.g. free radicals from smoking) and inflammation (infections –
C. pneumoniae). The vascular endothelium is responsible for homoestasis, and
therefore disturbance in the endothelium predisposes to atheroma. Endothelial
injury leads to increased permeability, leukocyte adhesion and thrombosis. However,
Risk factors
The risk factors for coronary artery disease are divided into modifiable and non-modifiable.
The non-modifiable risk factors are:
• Age. Coronary artery disease increases with age. It is rare in childhood, except in
familial hyperlipidaemias. Atheromatous lesions in the elderly are often
accompanied by calcification.
• Gender. Men have a higher incidence of coronary artery disease than
premenopausal women. After menopause, however, the incidence of coronary
artery disease approaches that of men. This is thought to be due to the loss of the
protective effect of oestrogen.
• Smoking. The risk of developing CAD is directly related to the number of cigarettes
smoked per day. 20% of deaths in men & 17% in women are attributable to smoking.
Stopping smoking actually decreases the cardiovascular risk, such that after 10 years
the risk declines to normal.
• Diet. Diets that are high in fats with low antioxidant levels1 are associated with
increased incidence of ischemic heart disease. It is estimated that up to 30% of cases
of CAD are attributable to the diet. Beneficial dietary changes include reduction in
fats, reduction in salt intake, an increase in carbohydrates, and an increase in fruit &
vegetable intake by 50%.
• Weight. 5% of deaths in men and 6% of deaths in women are due to obesity (BMI of
greater than 30kg/m2). The adverse effects of weight gain are more significant when
the weight is mostly concentrated in the abdomen (central obesity).
• Exercise. Reducing weight by diet & exercise reduces incidence of coronary artery
disease as well as insulin resistance. The physical activity needs to be regular &
aerobic. Adults must participate in a minimum of 30 minutes a day at least 5-times a
week.
• Hypertension. Both systolic & diastolic hypertension is associated with an increased
risk. Drug treatment & lifestyle modification can effectively lower cardiovascular risk.
• Hyperlipidaemia. High LDL & low HDL cholesterol are associated with increased
cardiovascular risk. Familial hypercholesterolaemia is associated. All patients at risk
of cardiovascular disease should have their fasting lipid profile taken. A cholesterol
target of less than 5.0mM is suggested for prevention of CAD.
• Diabetes mellitus. Diabetes substantially increases the risk of CAD. Men with type 2
diabetes have 2-4 fold increased risk of CAD.
• Alcohol. Moderate alcohol intake is associated with low risk of CAD. Excessive intake,
however, is associated with increased risk.
• Drugs. Some drugs, such as corticosteroids, increase the risk of atherosclerosis.
Spectrum of diseases
The spectrum of diseases constituting coronary artery disease occurs on a continuum:
1
Supplementation with antioxidants has been shown to be unhelpful.
Unstable angina, NSTEMI and STEMI all make up a clinical entity called acute coronary
syndrome.
Stable angina
Stable angina (also known as classical/exertional angina) is a condition that results from
temporary ischemia that develops when there is increased activity by the heart. It is
characterised by typical chest pain: central/retrosternal chest pain that is precipitated by
exertion and relieved by rest. At times the chest pain is atypical, in which case there are only
2 of the 3 features. Stable angina is caused by the presence of an atheroma in the coronary
circulation, causing ischemia when the oxygen demand of the heart rises (such as during
exercise). There is usually no progression of the chest pain symptoms as well as frequency &
severity of attacks.
• Unstable angina. This is a sharp chest pain that occurs at rest resulting from
ischemia, but is not due to myocardial infarction.
• Non-ST elevation myocardial infarction (NSTEMI). In this condition, there is
evidence of myocardial necrosis (MI) but the ST segment is not elevated. Other
features are present instead: ST segment depression, T-wave inversion or a normal
tracing.
• ST elevation myocardial infarction (STEMI). This is a myocardial infarction in which
the ST segment is elevated. There may also be a new-onset LBBB (left bundle branch
block)
The common mechanism for ACS is rupture or erosion of the fibrous cap of the coronary
artery plaque. This leads to initiation of thrombosis in the vessel which leads to total
occlusion of the blood vessel. The risk of rupture of a plaque depends on the presence of a
rich lipid pool within the plaque and a thin fibrous cap. After occlusion of the supplying
blood vessel, myocardial necrosis can occur within 15-30 minutes. The sub-endocardium is
initially affected, but with continued ischemia this extends to the sub-epicardium, thus
producing Q-waves.
Clinical presentation
The presentation of patients with coronary artery disease is as follows:
History
The history that is important is that of retrosternal chest pain that radiates to the left axilla,
shoulder, neck and jaw and sometimes to the epigastrium. The features are the ones that
differ:
Examination
Investigations
The investigations done for a patient who presents with acute retrosternal chest pain are as
follows:
2
Stress ECG, however, has a sensitivity of 67% & specificity of 72%.
3
If the initial troponin assay is negative it should be repeated 6-12 hours after admission.
The UK NICE guidelines recommend assessing the likelihood of coronary artery disease in
order to determine choice of diagnostic method & intervention. The likelihood depends on
the risk stratification of the patient in terms of:
• High & low risk stratification based on the presence of any of: diabetes, smoking,
hyperlipidaemia (total cholesterol > 6.47mM).
• Sex. Males have higher risk.
• Type of chest pain: non-anginal, atypical anginal or typical angina.
The likelihood is expressed as a percentage. If a patient has known CAD and has typical pain,
no further investigations are indicated. However, if the pain is atypical, you can do exercise
testing or functional imaging.
If a patient has unknown CAD, then you use the patient’s likelihood percentages:
Management of CAD
The management of CAD depends on the patient’s presentation:
Stable angina
Patients should be informed of their condition, and that the mortality from disease is low
(<2%). The patient is managed as follows:
4
NICE recommends that >70% of angioplasties be accompanied by stenting.
Some patients remain symptomatic even after all these interventions. These patients need
enrolment into a pain management program.
These patients must be managed as a medical emergency, but they are managed medically
until symptoms settle. You need to also take a brief history & examination, and then take an
ECG during pain.
1. Oxygen. You give oxygen if SaO2 is less than 90%, if there is pulmonary oedema, or
continuing myocardial ischemia. Hyperoxia should be avoided.
2. Nitrates. Nitrates are used to relieve pain. You can give 0.5mg glyceryl nitrate
sublingually as required.
3. Morphine. This is given if the pain does not resolve with nitrates alone.
4. Aspirin. You give 75-150mg once daily. You also add another antiplatelet agent, such
as clopidogrel, prasugrel and ticagrelor.
5. Oral β-blocker. These drugs reduce myocardial ischemia by inhibiting circulating
catecholamines. You can give atenolol 25-100mg once daily or metoprolol 50mg
twice daily. If this is contraindicated (e.g. in asthma, COPD, left ventricular failure,
bradycardia or coronary spasm) you can give non-dihydropyridine calcium channel
blockers.
6. Anticoagulation. You give fondaparinux 2.5mg once daily subcutaneously, or LMW
heparin 1mg/kg twice daily subcutaneously. You can also use rivaroxaban.
After doing this, the next phase of management depends on whether the patient is a high-
risk patient or not:
High-risk Low-risk
Criteria: Criteria:
• Rise in troponin • No recurrence of chest pain.
5
Bivalirudin is a direct thrombin inhibitor.
Management of STEMI
Management of STEMI entails supportive management (oxygen, morphine for pain and
nitrates), promoting reperfusion (aspirin, PCI and thrombolytics) and to reduce re-occlusion
& embolization (anticoagulants). Ultimately, the aim is to reduce mortality. In the initial
phases, these patients are also managed as a medical emergency. In these patients you take
a brief history, do a quick general examination and take a 12-lead ECG. On admission you
take bloods for U&Es, FBC, troponin, lipid profile and glucose, and you order a chest X-ray.
6
This is done within 72 hours of onset of symptoms.
7
This low saturation helps maintain the hypoxic drive which keeps these patients alive.
8
This should not be given if there is left ventricular failure.
9
The dihydropyridine drugs (nifedipine, amlodipine, etc.) have no benefit.
The patient also needs to be informed that they are a life-long patient and that they need
regular checkups to ensure that no other changes are occurring. They are also at risk of
repeat MIs.
Aetiology
The main causes of heart failure are hypertension, dilated cardiomyopathy, ischemic heart
disease, rheumatic heart disease and congenital heart diseases. The full list of causes
includes:
• Congenital. Some congenital heart disease can cause heart failure, whether early in
life or later on. Common congenital malformations include ASD, VSD and patent
ductus arteriosus. Tetralogy of Fallot is one exception.
• Infections. Infections that cause myocarditis can predispose to heart failure. These
include Coxsackie B virus and Chagas disease. Another infectious cause is infective
endocarditis. Infections causing IE can be bacterial (viridans streptococcal species, S.
aureus, Gram-negative organisms) or fungal (Candida).
• Structural anomalies. These include valvular heart diseases (both stenosis &
regurgitation). Dilated, hypertrophic and restrictive cardiomyopathies also cause
heart failure. Pericardial diseases (constrictive pericarditis & pericardial effusion
and/or tamponade) also cause heart failure.
• Conduction defects. Cardiac arrhythmias can cause heart failure. They include atrial
fibrillation, bradycardia, heart block, supraventricular tachycardia and ventricular
tachycardia.
• Hyperdynamic circulation. These include anaemia, thyrotoxicosis,
haemochromatosis, Paget’s diseases.
• Others. Other causes include diabetes, acute & chronic kidney disease (causing
cardio-renal syndrome), dyslipidaemias and some drugs (e.g. ACE inhibitor & AT 1-
receptor blockers in some patients).
• Systolic vs. diastolic heart failure. Systolic heart failure is the inability of the heart to
contract normally. This results in a reduced cardiac output from a reduced ejection
fraction (<45%). Conditions that do this are those that destroy the myocardium, such
Pathophysiology
The cardiac output is the product of the stroke volume and the heart rate:
𝐶𝑂 = 𝑆𝑉 × 𝐻𝑅
Heart failure in most cases is caused by a decrease in cardiac output, which is caused by
either a drop in the stroke volume or a drop in the heart rate. The stroke volume is
determined by ventricular filling (preload), cardiac muscle contractility and peripheral
vascular resistance (afterload). Any condition affecting preload, contractility, afterload and
heart rate subsequently leads to cardiac failure.
1
This manifests as hyponatraemia, which is an ominous prognostic indicator.
The symptoms of heart failure depend on which side of the heart is failing. In right heart
failure, there will be symptoms of fluid overload in the systemic veins. These symptoms are:
• Peripheral oedema.
• Ascites.
• Facial engorgement.
• Pulsations in neck & face (in tricuspid regurgitation).
• Fatigue.
In left ventricular failure, the symptoms are mostly pulmonary as the pulmonary veins
become engorged. They include:
• Dyspnoea on exertion.
• Orthopnoea.
• Paroxysmal nocturnal dyspnoea.
• Fatigue.
• Nocturnal cough with pink frothy sputum.
A combination of both will result in congestive cardiac failure. In both cases, there is often
poor exercise tolerance.
The symptoms of heart failure can be classified using the New York Heart Association
(NYHA) classification, based on severity of symptoms.
Examination
Investigations
• Chest X-ray. This is used to assess for signs of heart failure. These can be abbreviated
by ABCDE:
o A – Alveolar oedema. This is demonstrated by the presence of bat’s wing
sign.
o B – Kerley B lines. These signify interstitial oedema. There may also be fluid in
the fissures.
o C – Cardiomegaly. Cardiomegaly can be suspected if the cardiac shadow is
1
more than 2 the transthoracic diameter in a well-inspired PA film.
o D – Dilated upper lobe vessels. This is due to upper lobe diversion of blood
secondary to pulmonary congestion.
o E – Pleural effusion. This is secondary to fluid overload.
• ECG. On an ECG, you look for signs of ischemia, MI, arrhythmias or left ventricular
hypertrophy (e.g. hypertension). It is rare to get a completely normal ECG in chronic
heart failure.
• Natriuretic peptides (ANP & BNP). These are elevated in heart failure. Their
measurement is a good indicator of the presence of failure in a patient. The normal
level of natriuretic peptides is less than 100pg/mL.
• Echocardiography. This is the key investigation in heart failure. It is often done when
the other tests – chest X-ray, ECG and natriuretic peptides – are positive and
suggestive of heart failure. In addition, an echocardiogram will demonstrate the
presence of left ventricular dysfunction and possibly the cause of failure. You can use
an echocardiograph to determine: cardiac chamber dimensions, systolic & diastolic
function, regional wall motion abnormalities, valvular heart diseases and
cardiomyopathies.
• Blood tests. You can do FBC (to rule out anaemia & active infection), U&E, LFTs and
cardiac enzymes (if there is suspicion of an MI).
Normal Abnormal
Take echocardiogram
Heart failure unlikely
Normal Abnormal
• Aetiology.
• Degree of failure.
• Precipitating factors.
• Type of cardiac
dysfunction.
Choose treatment
• Lifestyle advice.
• Treat the cause.
• Pharmacological therapy.
• Surgical intervention.
• Hospice care.
Lifestyle advice
• Stop smoking. This may predispose to other cardiovascular events which may
worsen heart failure and the prognosis. If necessary, an anti-smoking clinic can be
used.
• Stop alcohol. It has a negative inotropic effect on the heart.
• Weight control. Obesity is associated with poor prognosis. You need to maintain a
certain body mass index.
• Diet. Large meals should be avoided, and a weight-reduction diet may also be
instituted. Salt restriction is necessary to reduce solute load – less salt should be
added to food and placed on the eating table. Fluid restriction is necessary in severe
heart failure. Patients are encouraged to eat foods rich in omega-3 polyunsaturated
fatty acids, such as fish.
Examples of causative factors for heart failure include arrhythmias & valvular heart diseases.
Until these are corrected, heart failure may continue to recur in patients.
Pharmacologic therapy
1. Diuretics. Diuretics can reduce the risk of worsening heart failure & death. They are
mostly used for symptomatic relief, and they work by relieving fluid overload
(through promoting renal excretion of fluid). You can use loop diuretics such as
furosemide 40-80mg orally 1-2 times daily, or bumetanide 1-2mg orally once daily.
You can also use thiazide diuretics (metozalone 5-20mg once daily) for patients with
mild failure & good renal function2. You can add a thiazide diuretic if the patient has
severe heart failure and the oedema is refractory to loop diuretics. Diuretics are
mostly potassium-wasting and therefore they pose the risk of hypokalaemia and
renal impairment.
2. ACE inhibitors. These drugs act by inhibiting formation of aldosterone II, resulting in
vasodilation and reduced sodium & water retention. ACE inhibitors have been shown
to improve symptoms & reduce mortality in all classes of heart failure. They also
prevent cardiac remodelling (by inhibiting the RAAS pathway). The typical drug used
is enalapril 5-20mg once daily. The main adverse effects are cough, hypotension,
hyperkalaemia and renal dysfunction (which occurs in renal artery stenosis,
pregnancy and patients with previous angioedema).
Angiotensin II receptor inhibitors are indicated in patients intolerant to ACE
inhibitors (the cough), and they also inhibit the RAAS system. They include losartan,
valsartan and candesartan. Losartan is given at a dose of 50-100mg once daily.
Candesartan can be given at a dose of 4mg once daily.
3. B-blockers. B-blockers have been shown to decrease mortality in heart failure due to
left ventricular dysfunction. They also inhibit cardiac remodelling by blocking the
cardiac effects of chronic activation of the sympathetic nervous system. Examples of
β-blockers used in heart failure include carvedilol (3.125-25mg twice daily),
bisoprolol, and nebivolol. Carvedilol & bisoprolol are indicated for any grade of heart
2
Good renal function is a must.
Surgery
• Interventional surgery. This is indicated for specific causes of heart failure. You can
do myocardial revascularisation, although this procedure’s role in heart failure is
unclear. Patients with angina & left ventricular dysfunction are at highest risk of
mortality from surgery, but stand to benefit most from surgical intervention.
3
Amiodarone was not shown to be of any benefit in heart failure patients with atrial fibrillation.
1. Stage A – high-risk patient with no symptoms. You start with risk factor reduction
(cessation of smoking & alcohol, controlling diet, control exercise) and patient &
family education. You should also treat comorbid conditions, such as dyslipidaemias,
hypertension and diabetes.
2. Stage B – structural heart disease with no symptoms. In these patients, you start on
ACE inhibitors (enalapril 5-20mg once daily) or AT1-receptor antagonists (losartan 50-
100mg daily). You can also give β-blockers (carvedilol 3.125-25mg twice daily)4.
These drugs are known to slow down remodelling of the heart, and this is the
primary aim in asymptomatic patients.
3. Stage C – structural heart disease with previous or current symptoms. In these
patients, symptom relief is one of the priorities. All patients should be on a
combination of an ACE inhibitor/AT1-receptor blocker and a β-blocker. You can also
add digoxin & diuretics. In these patients, you need to watch for hypokalaemia, and
be prepared to administer potassium chloride to the patient. If a bundle branch
block is present or the failure is unresponsive for 3 months, consider cardiac
resynchronisation therapy (a pacemaker).
4. Stage D – refractory symptoms requiring specialist interventions. You can use
inotropic agents. These are generally not recommended for heart failure, but are
indicated for intractable heart failure. You can use bipyrimidines (such as amrinone
& milrinone), levosimendan and β-agonists such as dobutamine. If these do not
work, consider a VAD while working towards transplantation, otherwise hospice
care.
4
B-blockers are started after ACE-inhibitors because ACE inhibitors provide rapid haemodynamic benefit,
while β-blockers have delayed onset.
The causes of acute decompensation can be abbreviated by the acronym HEART FAILURE:
Drugs such as digoxin, NSAIDs and β-blockers can tip patients into failure.
You diagnose acute heart failure using an ECG, echocardiogram, a chest X-ray, FBC & U&E,
blood glucose, and blood cardiac enzymes & plasma BNP.
Treatment of acute heart failure aims to relieve symptoms, reduce mortality and reduce
hospital stay. Patients must be managed in a high-care area with regular temperature, blood
pressure and heart rate monitoring.
• Bradycardias. The heart rate is below 60 beats/min during the day or below 50
beats/min during the night.
• Tachycardias. The heart rate is above 100 beats/min.
Bradycardias
Bradycardias may be due to failure of impulse formation or failure of impulse conduction
from the atria to the ventricles. The different bradycardias are:
Sinus bradycardia
This is a pulse rate of less than 60beats/min during the day or less than 50beats/min during
the night. The common causes of sinus bradycardia are:
Extrinsic: Intrinsic:
• Hypothermia, hypothyroidism, cholestatic • SA nodal acute
jaundice, raised intracranial pressure. ischemia/infarction.
• Drugs – β-blockers, digitalis and other • Chronic degenerative disorders
arrhythmic drugs. of the atrium/SA node.
• Neurally-mediated syndromes.
• Sick sinus syndrome (also known as sinoatrial disease). This is usually caused by
idiopathic fibrosis of the SA node, but the fibrosis can be secondary to ischemic heart
disease, cardiomyopathy or myocarditis. The syndrome is characterised by episodes
of sinus bradycardia or sinus arrest. They also commonly experience paroxysmal
atrial tachy-arrhythmias called the tachy-brady syndrome.
• Neurally-mediated syndromes. These are due to a reflex called the Bezold-Jarisch
which results in bradycardia & reflex peripheral vasodilation. These syndromes
present as syncope or pre-syncope. They include: carotid sinus syndrome, neuro-
cardiogenic syncope and postural orthostatic tachycardia syndrome (POTS).
Sinus bradycardia is treated by first identifying any extrinsic causes and removing them.
Other treatment options include:
1. Temporary pacing. This is done in acute extrinsic conditions until the offending
agent is removed. For chronic degenerative conditions, this is put in place until a
permanent pacemaker is put in place.
2. Permanent pacemaker. Chronic symptomatic sick sinus syndrome requires a
permanent pacemaker. These devices are also beneficial in patients with malignant
neuro-cardiogenic syncope.
AV heart block
A blockage of conduction of impulses in the AV node or the His bundle results in AV block.
There are 3 main types of heart block.
Bundle branch block occurs when there is a block in one of the bundle branches of the His-
Purkinje system rather than the whole system. Various disturbances can occur. The
disturbances that can occur are:
• Bundle branch conduction delay. This produces slight widening of the QRS complex.
It is also known as incomplete bundle branch block.
• Complete bundle branch block. This produces a widened QRS complex. The ECG
features depend on the branch that is blocked.
o Right bundle branch block. This produces late activation of the right ventricle
and therefore produces deep S-waves in leads II & V6 and an additional late
R-wave (called rsR’) in lead V1.
o Left bundle branch block. This produces late activation of the left ventricle. It
results in deep S-waves in lead V1 and tall wide, notched or M-shaped R-
waves in leads I & V6. There are also abnormal Q-waves.
• Hemiblock. In a hemiblock, there is a block in a single division of the fascicles into
which the left bundle branch divides. There is a change in the axis of the heart.
o When the anterior division is blocked, the left ventricle is activated from
infection to superior and therefore there is left axis deviation.
o When the posterior division is blocked, the left ventricle is activated from
inferiorly to superiorly and therefore there is right axis deviation.
• Bifascicular block. This is a combination of block of any of the following: the right
bundle branch, anterior division of left bundle branch, and posterior division of left
bundle branch. In this type of block, only one fascicle remains.
Bundle branch blocks are usually asymptomatic. Right bundle branch block may cause wide
but physiologic splitting of the 2nd heart sound.
Tachycardias
1
A junctional/narrow complex escape rhythm means that the new dominant pacemaker activity is coming
from the AV node. The junctional escape rhythm operates at a rate of 40-60bpm. On an ECG, the P-waves may
or may not be present and the QRS is of normal morphology & duration.
2
A ventricular/broad complex escape rhythm means that the new dominant pacemaker activity is coming
from the ventricular myocytes themselves. The ventricular escape rhythm operates at a rate of 15-40 beats per
minute. On an ECG, the QRS complex is widened.
• Anti-arrhythmic drugs.
• Ablative techniques.
• Device therapy.
Anti-arrhythmic drugs
Anti-arrhythmic drugs are drugs that are used to treat arrhythmias. They bind avidly to
activated or inactivated channels, but poorly to resting ones. Therefore, they act on highly
active tissue. Anti-arrhythmic drugs are divided into 4 classes according to the Vaughan-
Williams classification based on the part of the cardiac potential they affect.
• Class 1 drugs. These are membrane-depressing drugs that block sodium channels.
They all slow the entry of sodium channels and therefore increase the duration
phase 0. Class 1 drugs are further divided into 3 groups based on their effects on the
length of the action potential, their reaction kinetics and their potency.
3
Their effect on ventricular ectopics, however, is lower than that of class 1 drugs.
4
Sotalol is also a β-blocker.
5
This is due to its iodine content. Dronedarone carries a lower risk and therefore can be used to avoid this side
effect.
Ablation therapy
1
• Paroxysmal SVT, of which AVNRT accounts for more than 2 of cases.
• Accessory pathways (present in 30% of all SVTs), such as WPW syndrome. In these
patients, RFA is indicated as 1st-line therapy if they have a pre-excited ECG & are
symptomatic. In those that conduct retrograde from the ventricles, RFA is offered if
simple methods (e.g. AV nodal slowing) do not work. RFA can also be offered
prophylactically in asymptomatic patients.
• Atrial flutter (in which the focus is in the right atrium). Ablation of a typical flutter is
effective in 90-95% of cases, and it is used for cases of flutter that are not responsive
to medical therapy.
• Atrial fibrillation (potential for pulmonary vein ablation).
• Normal heart ventricular tachycardia (focus in right ventricular outflow tract or
infero-septal left ventricle near apex)6.
To perform ablation, you first place 3 or 4 electrode catheters into the heart chambers to
record & pace from various sites. Pacing is used to trigger the arrhythmia and study the
tachycardia mechanism. Radiofrequency energy (low-frequency, high voltage) is then
passed into this focus, and it produces small homogeneous necrotic lesions approximately
5-7mm in diameter and 3-5mm in depth.
ICDs are used to recognise ventricular tachycardias & ventricular fibrillation and
automatically pacing or administer a shock to the heart. This causes cardioversion to sinus
rhythm. This is an important intervention in preventing sudden cardiac death from life-
threatening ventricular tachycardias in at-risk patients. There is also a considerable
6
The majority of ventricular tachycardias are due to scarring from MI and cannot be ablated.
Modern ICDs are only a little larger than pacemakers and are placed in the chest in the
pectoral position. They have lithium batteries to power over 100 shocks at 30J. ICD shocks
are painful if the patient is conscious. Ventricular tachycardia may be terminated by
overdrive pacing the heart, and this is painless.
Sinus tachycardia
A sinus tachycardia can be a normal finding if there is physical or emotional stress. However,
if the tachycardia is persistent & out of proportion with the level of stress, then it is termed
inappropriate sinus tachycardia. It is predominantly found in young women and is
uncommon in health professionals. Sinus tachycardia due to intrinsic sinus node
abnormalities is extremely rare.
A sinus tachycardia is generally a secondary phenomenon, and the root cause needs to be
investigated. The causes include:
Acute: Chronic:
• Exercise. • Pregnancy.
• Emotion. • Anaemia.
• Pain. • Hyperthyroidism.
• Fever & infection. • Catecholamine excess (as in
• Acute heart failure. phaeochromocytoma).
• Acute PE.
• Hypovolaemia.
You can use β-blockers or ivabradine to slow down the heart rate if it is excessively high.
Atrial tachycardias
Atrial tachycardias arise from the atrial myocardium. They include atrial fibrillation, atrial
flutter, atrial tachycardia and atrial ectopic beats. The causes of atrial tachycardias can be
classified into cardiac & non-cardiac:
Cardiac Non-cardiac
Systemic causes: Endocrine:
• Hypertension • Thyrotoxicosis.
Atrial fibrillation
Atrial fibrillation is a condition in which there is a chaotic irregular atrial rhythm with
intermittent AV response, resulting in a very high atrial rhythm (300-600 bpm) that differs
from the ventricular rhythm. Atrial fibrillation is the most commonly-encountered cardiac
arrhythmia, occurring in 1-2% of the general population and 5-10% of patients older than 75
years. In younger patients it can occur in a paroxysmal form. The classification of atrial
fibrillation is as follows:
Pathophysiology
Atrial fibrillation seems to require an initiating event and a permissive atrial substrate. It is
thought that in atrial fibrillation there is an autonomic focus of impulses in the heart. The
pulmonary vein (that containing cardiac muscle) seems to be the most common source of
1
Most episodes last less than 24 hours.
3 forms of atrial remodelling cause during the progression of atrial fibrillation have been
described.
Because of these remodelling changes, most patients with paroxysmal AF will progress to
persistent or permanent AF.
Clinical features
On examination, the key feature is an irregularly irregular pulse that is tachycardic. Findings
include:
• ECG: this will reveal absence of discrete P-waves, which are replaced by irregular F-
waves (flutter waves). The ventricular rate is irregular.
• Blood studies: FBC (anaemia, infection), U&Es (electrolyte imbalances), TFTs
(thyrotoxicosis),
• Echocardiography: to estimate chamber size, identify valvular heart lesions and
ascertain ventricular function. It also helps identify thrombi.
Management
The management of atrial fibrillation centres on rate control, anticoagulation and rhythm
control for those who are symptomatic. It divided into management of new-onset AF,
paroxysmal AF and persistent AF.
Acute management
1. Oxygen.
2. Ventricular rate control. This is achieved by AV node blockers. You can use β-blockers
& non-dihydropyridine calcium channel blockers as 1st-line agents, or you can use
digoxin in heart failure patients. If patients are unresponsive to these agents, you
can use amiodarone.
3. Treat the provoking cause. This includes thyrotoxicosis, alcohol toxicity, chest
infection, etc.
4. Anticoagulation. This is done to prevent further growth of thrombi. It is often
necessary to anticoagulate prior to cardioversion, as cardioversion can dislodge
emboli. Anticoagulate using heparin, warfarin (aiming for INR 2-3) or dabigatran
150mg twice daily for 3 weeks.
5. Cardioversion. This is usually achieved by direct current shock, which achieves sinus
rhythm in 80% of patients. Biphasic defibrillation is more effective than conventional
monophasic defibrillation. Prior to electric cardioversion you need to ensure that the
patient is full anticoagulated. If cardioversion is urgently required without having
time to wait for anticoagulation, you can do transoesophageal echocardiography to
rule out the presence of significant emboli. You can also cardiovert medically using
amiodarone, flecainide and propafenone.
Long-term management
Atrial flutter
This is an arrhythmia in which the heart produces organised atrial rhythms. The atrial rate is
often in the range of 240-400beats/min, and often every second atrial beat conducts to the
ventricles producing a ventricular rate of about 150bpm. There is usually some degree of AV
nodal block. 30% of patients have coronary artery disease, 30% have hypertensive heart
disease, and 30% have no identifiable heart pathology.
Pathophysiology
The majority of cases of atrial flutter occur as a result of a macro-re-entrant right atrial
circuit around the tricuspid annulus. The wave travels down the lateral wall of the right
atrium, down through the Eustachian isthmus (which is an area of slow conduction between
the tricuspid valve annulus and the ostium of the inferior vena cava & coronary sinus) and
up the inter-atrial septum. This is called typical/isthmus-dependent atrial flutter and is also
referred to as a counter-clockwise atrial flutter. Atrial flutter can also occur in the opposite
direction (atypical flutter).
Clinical presentation
Symptoms of atrial flutter are related to the degree of AV block. They are a result of reduced
cardiac output as a result of the rapid ventricular rate. The symptoms include:
• Palpitations.
• Fatigue/poor exercise tolerance.
• Mild dyspnoea.
• Presyncope.
Patients can easily degenerate into atrial fibrillation or sinus rhythm. Chronic stable atrial
flutter is rare. If there is a history of pre-excitation syndrome (e.g. Wolff-Parkinson-White
syndrome) there is a risk of 1:1 conduction, which can lead to ventricular fibrillation.
On examination, determine the vital signs (pulse rate, respiratory rate, blood pressure, SpO 2
and temperature) to determine the urgency of restoring sinus rhythm.
Investigations
• ECG. This will demonstrate saw-tooth flutter (F) waves. In a typical flutter, the F-
waves are inverted in leads II, III and AVF, positive in lead V 1 and are negative in lead
V6. In an atypical flutter, the opposite occurs. A clue to pre-excitation syndrome is a
very short PR interval with no delta wave.
Management
Treatment approaches for atrial flutter are similar to those for acute atrial fibrillation.
• Cardioversion. This is the treatment of choice for an acute episode. Patients who
have had atrial flutter for more than 1-2 days need to be anticoagulated for 3 weeks
prior to cardioversion. Atrial flutter generally requires less energy than atrial
fibrillation. Cardioversion can also be achieved by using class Ic & III drugs.
• Rate control. Rate control may alleviate symptoms while awaiting cardioversion.
However, it is more difficult for atrial flutter than for atrial fibrillation. You can use
non-dihydropyridine calcium channel blockers & β-blockers, although negative
inotropic effects & hypotension are considerable concerns.
• Ablation. This is indicated for repeated episodes. You can use catheter ablation or
radiofrequency ablation. This creates a line of conduction block in the Eustachian
isthmus which prevents the re-entry circuit. This offers almost definite cure in
individuals in whom the atrial flutter is the only problem. However, atrial fibrillation
can occur later.
Atrial tachycardia
This is a rare type of tachycardia, accounting for less than 1% of all cases of cardiac
arrhythmias. It may be idiopathic or it may be related to structural heart disease. It can also
occur after open surgery for congenital heart diseases (causing macro-re-entrant circuits). It
can also occur as a result of digitalis poisoning. The mechanisms of atrial tachycardia are:
On an ECG, you will see abnormal P-waves in front of QRS complexes. There may also be a
concomitant right bundle branch block particularly in digitalis toxicity.
Treatment options include cardioversion, anti-arrhythmic drug therapy (to maintain sinus
rhythm) and AV nodal slowing agents (to slow the rate). In some cases you can use ablative
techniques. In cases due to digoxin toxicity you need to stop digoxin and maintain
potassium at 4-5mM.
The junctional tachycardias are re-entry tachycardias where the AV node is an essential
component of the re-entry circuit. This may be induced by premature atrial or ventricular
beats. Other extrinsic triggers include hyperthyroidism and stimulants (caffeine, drugs and
alcohol).
Classification
Clinical features
• Sudden onset & termination of palpitations. This occurs in greater than 96% of
patients. These may be spontaneous or precipitated by movements or certain states.
These may be terminated by Valsaver manoeuvre. This is often associated with
dizziness.
• Cardiovascular compromise. This occurs in patients that do not have a good
functional reserve. The symptoms include fatigue, shortness of breath, chest pain
and syncope.
On physical examination, patients are often distressed, and tachycardia may be the only
finding. In AVNRT, you can find prominent jugular venous pulsations. Patients may also
present with decompensated heart failure.
Investigations
The tests done are generally to rule out other differentials for symptoms as well as
identifying other causes.
• Cardiac enzymes. These are done to rule out acute coronary syndrome.
• U&Es. Electrolyte abnormalities can cause paroxysmal SVTs.
• FBC. This is used to assess whether anaemia is contributing to the tachycardia or
ischemia.
• TFTs to rule out hyperthyroidism.
• Digoxin levels for patients on digoxin.
Management
Acute management
In an emergency setting, the distinction between AVNRT & AVRT is not important as both
respond to the same treatment. Patients may be haemodynamically stable, and the only
problem they have is the on-going palpitations & dizziness. Other patients, however, may be
haemodynamically unstable, as indicated by hypotension, pulmonary oedema, chest pain
with ischemia or be unstable in general. These patients require emergency treatment. The
interventions include:
Long-term management
This is a ventricular tachycardia that is sustained for at least 5 consecutive beats but lasts no
more than 30s. It can be found in 6% of patients with normal hearts, and it is documented in
60-80% of patients with heart disease.
1
This is called Galavardin’s tachycardia.
On an ECG, the QRS complex is broad (>120ms) with a bizarre morphology and there is no
preceding P-waves. There is often a complete compensatory pause following the ectopic
because the AV node & ventricle are refractory. Early R-on-T ventricular beats (in which the
R-wave of the ectopic beat coincides with the upstroke/peak of the previous T-wave) may
induce ventricular fibrillation in patients with heart disease, particularly following MI.
Ventricular ectopic/premature beats are usually treated only when they are symptomatic.
Usually reassurance & β-blocker therapy is indicated. If the ectopics are very frequent, left
ventricular dysfunction may develop and if they come from a single focus (especially in the
right ventricle), catheter ablation can be very effective.
• Monomorphic VTs. In this form there are identical complexes with uniform
morphology. This is the most common type. They typically result from
intraventricular re-entry circuits. Potential causes include:
o Idiopathic.
o Chronic infarct scarring.
o Acute ischemia/MI.
o Cardiomyopathies.
o Myocarditis.
o Arrythmogenic right ventricular dysplasia.
o Drugs (e.g. cocaine).
Clinical features
Patients develop presyncope, syncope, hypotension and cardiac arrest. In some patients,
this condition may be well-tolerated. There are often clinical signs of atrioventricular
dissociation, such as cannon a-waves in the JVP and variable intensity of the first heart
sound.
On ECG
On the ECG you will see a tachycardia with widened QRS complexes. AV dissociation will
result in visible P waves which march through the tachycardia and cause capture beats
(normal-morphology QRS complexes caused by conduction through the normal conduction
pathway from the atria to the AV node) and fusion beats (QRS complexes that are
intermediate between widened and capture beats).
VT SVT
Broad complexes + RBBB morphology
Lead V1 Monophasic R, QR or RS waves. Triphasic RSR
Lead V6 R-wave smaller than S-wave. Triphasic R wave.
QS/QR R-wave greater than S-wave.
Monophasic R-wave
Broad complexes + LBBB morphology
Lead V1/2 R-wave > 30ms.
>60s to nadir of S-wave.
Notched S-wave.
Lead V6 Presence of any Q, QR or QS No Q-wave
Treatment
After stabilising the patient, a full history & examination needs to be taken and the
diagnosis needs to be made backed by investigations. Maintenance cardioversion using
amiodarone may be required. To prevent recurrent VTs, you can use implantable automatic
defibrillators. In refractory cases, radiofrequency VT ablation may be tried.
Ventricular fibrillation
This is a chaotic ventricular activation with no mechanical effect. The condition is usually
provoked by an ectopic ventricular beat. The patient is pulseless, becomes rapidly
unconscious and develops cardiac arrest. It is usually a terminal event unless advanced
cardiac life support (ACLS) procedures are promptly initiated to maintain ventilation &
cardiac output, and electrical defibrillation is carried out. It is the most frequent cause of
death. On ECG, you will see shapeless rapid oscillations, and there is hint of organised
complexes.
To treat these patients, ACLS is needed. If the attack happened within 1-2 days of an acute
MI, prophylactic therapy is unlikely to be necessary. If the attack was not linked to a recent
MI, the long-term risks of recurrent cardiac arrests & sudden death are high. Implantable
cardioverter-defibrillators are the 1st-line therapy in the management of these patients.
Brugada syndrome
This condition accounts for some patients with idiopathic ventricular fibrillation who have
no evidence of causal structural cardiac disease. It is common in young male adults in SE
2
Even if you’re not sure of the diagnosis between ventricular & supraventricular tachycardias, treat the
disorder as a VT.
The condition can present with sudden death during sleep, resuscitated cardiac arrest and
syncope. The patient may also be asymptomatic and diagnosed incidentally or during family
assessment. The only successful treatment is an implantable cardioverter defibrillator. Use
of β-blockers is not useful and may actually be harmful.
QT syndromes
QT syndromes include long QT syndrome and short QT syndrome.
Long QT syndrome
Clinical features
Patients with long QT syndrome develop syncope & palpitations secondary to a polymorphic
ventricular tachycardia. They usually terminate spontaneously, but may evolve into
ventricular fibrillation.
On ECG, the most notable feature is a prolonged QT segment, which is more notable
between spells of tachycardia or immediately before the onset of tachycardia. It may evolve
into Torsades de pointes (twisting of the points), which is characterised by rapid irregular
sharp complexes that rotate around the baseline, changing their axis & amplitude.
Treatment
1. Treat the cause: correct electrolyte imbalances, stop any causative drugs.
2. Maintain the heart rate. This is done with atrial/ventricular pacing.
3. Magnesium sulphate. You give 2g IV over 10 minutes. Alternatively, you can use
isoprenaline3.
Short QT syndrome
There are 5 types of short QT syndromes described. They are caused by genetic
abnormalities leading to faster repolarisation. Ventricular arrhythmias & sudden death can
occur. Implantable cardioverter defibrillator is the best treatment.
3
Isoprenaline is contraindicated in congenital long QT syndrome.
DVTs are common in hospitalised patients; they occur in 25-50% of surgical patients and
many non-surgical patients. They are often undetected; 65% of below-knee DVTs are
asymptomatic. Axillary vein thrombosis may also occur, sometimes related to trauma.
Risk factors
The risk factors for developing DVTs are related to anything that disrupts the Virchow’s
triangle. It consists of: endothelium, blood flow and coagulability. In other words, condition
that increase risk of DVT formation are those that disrupt the endothelium, interrupt/slow
blood flow or increase the coagulability of blood (by increasing its constituents/making it
more viscous, or by activating clotting factors).
• Calf pain.
• Swelling & redness.
• Engorgement of superficial veins.
• Mild fever.
• Pitting oedema may be present.
• Cyanotic discoloration of the limb. This can occur with complete occlusion of a large
vein. It is usually accompanied by severe oedema.
• Pulmonary embolism. This can occur from any deep venous thrombosis. It is most
common, however, with iliofemoral thrombosis.
Investigations
The tests done in investigating a DVT are:
• D-dimer levels. D-dimers are fibrin degradation products that signify breakdown of
an already-made fibrin clot. Their levels signify the presence of secondary
haemostatic activity. High D-dimer levels would point towards the presence of
thromboembolic phenomena. Nonetheless, they are not specific, and they are
elevated in infections, pregnancy, malignancy and post-operative period. A negative
result, however, is highly suggestive of absence of disease.
• Doppler ultrasound. This is an ultrasound test that is done to assess the patency of
blood vessels. It can be used to rule out occlusions as a definitive test. The Doppler
ultrasound has a sensitivity & specificity of over 90%. If the Doppler ultrasound is
negative, do a repeat ultrasound after 1 week to pick out early but propagating
DVTs.
• Thrombophilia testing. This should be done in a patient before commencing
anticoagulant therapy if there are no predisposing factors, in recurrent DVTs or when
a family history is present.
• Screening for underlying malignancy. Do FBCs, LFTs, serum calcium, chest X-ray and
CT scan of the pelvis and/or abdomen.
Treatment
The primary aim of treatment is to stop the progression of the thrombus as well as prevent
pulmonary embolism. Anticoagulation is therefore indicated for patients with above- &
below-knee thrombi. Bed rest is advised until the patient is fully anticoagulated. Afterwards,
they should be allowed to mobilise and given a pressure stocking to give graduated pressure
on the leg.
The duration of warfarin treatment is debatable. 3 months is the recommended period, but
4 weeks is long enough if a definite risk factor has been present. Cancer patients should
receive LMWH for 6 months.
You can also use an inferior vena cava filter if the patient is actively bleeding or when
anticoagulants fail to reduce the risk of pulmonary embolism. Thrombolytic therapy may be
employed for thrombi in large veins, but there is the risk of bleeding.
Prevention of DVT
DVTs can be prevented. Because a large number of thrombi develop in hospitalised patients,
we can significantly prevent them by employing measures to prevent their development.
The principles are:
• All patients should be assessed for presence & risk of DVT on admission and 24 hours
after admission or when their condition changes.
• Use the Wells score to assess for the likelihood of developing a DVT.
Wells score
Clinical features Score
Active cancer with treatment in the last 6 months, or palliative. +1
Paralysis, paresis or recent plaster immobilisation of leg. +1
Recently bed-ridden for more than 3 days, or major surgery in last 12 weeks. +1
Local tenderness along distribution of deep venous system. +1
2
Calf swelling of greater than 3cm compared with asymptomatic leg. +1
Pitting oedema. +1
Collateral superficial veins. +1
Previously documented DVT. +1
Alternative diagnosis to DVT as likely as DVT. -2
If the patient’s Wells score is 1 or below, a DVT is unlikely. The patient should have a D-
dimer test done and excluded if the D-dimer level is low. If D-dimer test is positive, do a
Doppler ultrasound test to confirm or exclude a DVT. If the patient’s Wells score is 2 or
more, do a D-dimer test & Doppler ultrasound. If both are negative, the DVT is excluded. If
1
In this case, heparin is stopped when INR is in the target range.
2
This is measured 10cm below tibial tuberosity.
• Hypovolaemic shock.
• Cardiogenic shock.
• Septic shock.
• Anaphylactic shock.
• Neurogenic shock.
• Spinal shock.
Mechanism of shock
The blood pressure is a product of cardiac output and peripheral vascular resistance. Cardiac
output is determined by the stroke volume & heart rate, while peripheral resistance is
determined by arterial vascular tone. Therefore there are 2 ways in which shock can
develop:
• Shock can develop when the cardiac output decreases. It can be a result of a
reduction in the circulating volume, as is the case in hypovolaemic shock or in
obstructive shock. It can also be a result of pump failure (cardiogenic shock).
• Shock can also develop as a result of reduced peripheral vascular resistance. This is
the case in septic shock, anaphylactic shock and neurogenic & spinal shock.
Hypovolaemic shock
Hypovolaemic shock is the common type of shock around. It is a result of reduced
circulatory volume, and it is usually serious when it overwhelms the cardiovascular
compensatory mechanisms. Causes of hypovolaemic shock can be divided into exogenous
and endogenous. Exogenous causes are ones in which fluid leaves the body through the skin
or a visceral compartment. They include haemorrhage and ECF losses from burns or from
vomiting & diarrhoea. Endogenous causes include 3rd-spacing.
Pathophysiology
In response to acute fluid or blood loss, the following happens:
1
This indicates narrowing of the pulse pressure.
Investigations
The investigations to be done include:
• Blood pressure.
• Oxygen saturation monitoring.
• FBC (anaemia, infection), cross-matching, U&E (renal function: rule out AKI), blood
glucose and blood cultures for screening for infection.
• Urine output measurement.
Management
In the initial setting, the most important thing is to identify the source of bleeding and
control bleeding. This is in the event that the cause of shock is haemorrhagic. You should
also raise the legs to increase venous return.
The management of a hypovolaemic patient in resuscitation is indicated for patients in
stages 2 going forward. The management is as follows:
1. Insert 2 wide-bore cannulas (e.g. 14-gauge cannulas; grey).
2. Take blood samples for FBC & cross-matching.
3. Run 10-20ml/kg of crystalloid (2L in an average adult) in the patient over 10 minutes.
4. Have 2 units prepared for possible blood transfusion.
5. Insert a urinary catheter to monitor urine output.
6. Insert an NGT tube to decompress stomach.
The subsequent management depends on the response to resuscitation:
• Rapid responders. These are patients whose haemodynamic status is restored after
initial resuscitation. The patients are usually those in stage 1 shock.
• Transient responders. These are patients who initially respond after resuscitation,
but when the bolus infusion is slowed to maintenance their vitals & perfusion
deteriorate. The patients either need more fluid or have on-going losses. They need
blood and they need surgical control of haemorrhage.
• Non-responders. These are patients who do not respond to initial resuscitation. They
need immediate surgical referral to locate & stop the source of bleeding. Blood
transfusion is withheld until the bleeding source is stopped.
Cardiogenic shock
2
Β-blockers & ACE inhibitors usually cause cardiogenic shock in the setting of acute coronary syndrome.
• ECG. This can be done in the immediate setting as a patient comes in. it helps
identify features linked to the aetiology: abnormal heart rhythm (arrhythmias), acute
MI (ST elevation, deep Q-waves) and signs suggestive of cardiomyopathy (left
ventricular enlargement).
• Echocardiography. This will show the calibre of heart muscles and their function. It
may show poor ventricular function, rupture of interventricular septum, an
obstructed outflow tract or cardiomyopathy.
• Biopsy. This is done for patients in whom cardiomyopathy is suspected.
• Cardiac index. This relates the cardiac output from the left ventricle to the patient’s
total body surface area. It relates cardiac performance to the size of the individual.
𝐶𝑂
𝐶𝐼 =
𝑇𝐵𝑆𝐴
The normal range is 2.6-4.2L/min. A cardiac index of less than 2.2L/min is suggestive
of cardiogenic shock.
• Blood investigations. FBC can be used to exclude anaemia, infection and
coagulopathy3. Cardiac enzymes can be measured for patients with anaemia. Arterial
blood gas measurement can be done to assess the overall acid-base balance of the
patient as well as degree of oxygenation of the patient.
• Filling pressure. This can be measured using a Swan-Ganz catheter. It measures the
filling pressure of the heart. The normal filling pressure is 8-10mm Hg.
Management
Cardiogenic shock is a medical emergency, and patients require prompt admission into an
intensive care unit. The majority of patients should be treated as though they have a
myocardial infarction. Treatment is as follows:
1. Oxygen & ventilator support. In patients that are not able to breathe properly, you
may need to ventilate them. Positive pressure ventilation is not advised because it
may compromise venous return & preload to the heart. Oxygen should be titrated to
maintain oxygen saturations of 94-98%. The target may be lowered to 88-92% in
patients with COPD.
2. Analgesia. You can give diamorphine (heroine) 1.25-5mg stat intravenously.
diamorphine also helps ease anxiety.
3. Fluid administration. Fluid can be given unless pulmonary oedema is present. This is
to optimise filling of the heart, provided the patient is under-filled. The patient is
given 100ml intravenously every 15 minutes. You should aim for a mean arterial
pressure of 70mm Hg and a central venous pressure of 8-10mm Hg.
3
This may be low due to sepsis.
• Abdominal pain.
• Abdominal distension.
• Loss of appetite & weight change.
• Nausea & vomiting.
• Heartburn.
• Diarrhoea & faecal incontinence.
• Constipation.
• Bloating.
• Postprandial fullness/early satiety.
• Jaundice & pruritus.
• Dark urine.
• Pale stool.
Abdominal pain
Abdominal pain is a common presentation in abdominal disease. When a patient presents
with abdominal pain, you need to enquire about the exact site of the pain and its character,
timing/pattern, severity, radiations, aggravating factors. A correct description of the pain
will usually give a clue to the exact illness that a patient has. Abdominal pain is mainly a
result irritation of the peritoneum. There are 3 types of abdominal pain.
• Parietal pain. Parietal pain is a result of irritation of the parietal peritoneum. Parietal
peritoneum has numerous receptors, such as pain, temperature, stretch and others.
Parietal pain is conducted by somatic nerve fibres that have pin-point distribution in
the abdomen. It is therefore sharp & well localised. It is also aggravated by
movement. Peritonitis is a cause of parietal pain, and in this case is it associated with
tenderness on light palpation and rebound tenderness.
The onset of abdominal pain is a good indicator of the cause of the pain. Sudden onset of
pain suggests intestinal obstruction/perforation, ruptured abdominal aortic aneurysm or
mesenteric infarction (these would be further supported by other comorbid conditions,
such as hypertension, peripheral vascular disease, heart failure or atrial fibrillation). Sudden
onset conditions are therefore those conditions in which there is mechanical obstruction or
damage to abdominal viscera.
Pain radiating from the right hypochondrium to the shoulder or inter-scapular region
suggests diaphragmatic irritation. There are various other radiations. The combination of
severe back & abdominal pain may suggest ruptured or disseminated abdominal aortic
aneurysm.
Some of the common pain patterns that are produced abdominal pathology are:
• Peptic ulcer disease. This produces a dull & burning pain in the epigastrium
(described as a gnawing pain) that radiates into the back. It is typically episodic and
may occur at night, and each episode lasts for 30 minutes to 2 hours. It is relieved to
some degree by food & antacids, although it is not always related to meals. It is
1
The nerves will fuse with the nerves from structures that arise from the same dermomyotome as the viscous
in question.
2
The appendix, however, has a spectrum of pain symptoms that are unique to it.
Pathophysiology of diarrhoea
Diarrhoea is caused by an imbalance between absorption & secretion of fluid into the GIT.
Normally, when food passes through the GIT, there is secretion of large amounts of water
into the GIT in the form of digestive secretions. This water is then reabsorbed in the
intestines, and enterally-taken water is also absorbed. Any process that increases secretion
and impairs absorption leads to diarrhoea.
The location of the pathologic process of the diarrhoea is also important in determining the
cause. Small intestinal diarrhoea is associated with large amounts of watery diarrhoea
associated with abdominal cramps. Fever and weight loss are rare features. Large intestinal
diarrhoea is associated with frequent low-volume toilet visits associated with colicky
abdominal pain, urgency1, tenesmus2 and incontinence. Fever and bloody stool are
common.
Aetiology
The causes of diarrhoea can be classified into infectious & non-infectious.
Infections
Bacteria are not the most common causes of diarrhoea, but they are the most common
causes of severe diarrhoea with complications. The bacterial causes of acute diarrhoea are:
• E. coli. E. coli possesses a wide range of virulence factors, which are either
endotoxins or exotoxins. E. coli is a normal GIT commensal, and infection occurs due
to acquisition of virulence factors. Majority of infections arise endogenously. There
are 5 major subgroups of E. coli. These are:
o Entero-toxigenic E. coli (ETEC). ETEC is one of the most common forms,
accounting for 650 million cases per year. It is acquired by ingesting
contaminated food or water. It produces secretory small intestinal diarrhoea.
ETEC produces both heat-stable & heat-labile toxins, and the heat-labile toxin
1 is structurally similar (has an α-subunit and 5 β-subunits) & has similar
actions to the cholera toxin. The heat-labile toxin binds to the cholera
receptor and the α-subunit leads to activation of a Gs-protein which leads to
an increase in intracellular cAMP. The heat-stable toxin binds to a
transmembrane guanylyl cyclase receptor which leads to an increase in
cGMP. Both these toxins lead to secretion of chloride ions and inhibition of
sodium & chloride reabsorption. The diarrhoea is profuse & watery.
1
Faecal urgency is the inability to postpone urges to defecate.
2
Tenismus is the continuous/recurrent sensation of wanting to evacuate bowel.
Viruses are the most common causes of diarrhoea, although the majority are self-limiting.
Viral causes of diarrhoea are:
• Rotavirus. Rotavirus is the most common cause of diarrhoea amongst children under
5 years. The virus is stable in extreme pH environments (3.5-10), detergents and
repeated freezing & thawing. Rotavirus possesses a NSP4 protein which acts like a
toxin by inhibiting calcium influx into enterocytes, releasing neuronal activators and
neuronal alteration of water reabsorption. The virus is spread by faecal-oral route
through person-to-person contact. Patients present with vomiting, profuse
diarrhoea, fever and dehydration.
• Noroviruses. The prototypical Norovirus is the Norwalk virus. The Norwalk virus is a
positive-sense single-strand RNA virus with a naked capsid. The norovirus strains that
infect humans can only infect humans. These viruses are often spread during
outbreaks from a common source of contamination. They cause acute onset
diarrhoea with nausea & vomiting. Bloody stool does not occur.
Non-infectious
Clinical assessment
When assessing a patient who presents with diarrhoea, you need to ask the following
questions:
• Duration. Acute diarrhoea is that which develops over less than 14 days. If it is acute,
suspect infectious causes such as bacteria or viruses and some parasites. If it is
chronic (longer than 14 days), suspect other non-infectious causes as well.
Management of diarrhoea
Management of diarrhoea is as follows:
3
Presence of white cells excludes ETEC, Vibrio and viruses.
4
Campylobacter does not grow on traditional culture media. It requires Skirrow media.
5
This may be a consequence of malabsorption syndromes.
6
In severe disease, use vancomycin 125mg 6-hourly orally.
Although both conditions have a worldwide prevalence, the highest rates have been
reported in Europe & North America. The prevalence of Crohn’s disease is 25-100 cases per
100 000 population. The incidence is 4-10 cases per 100 000 population. The prevalence of
Crohn’s disease is lower in Hispanics & Asians compared to white people. Prevalence rates
in populations also change after migration. About 25% of patients are diagnosed before they
turn 18, and there is increasing evidence that disease commencing in youth is more
extensive & aggressive that adult onset disease. The prevalence of ulcerative colitis is 6-15
cases per 100 000 population. This prevalence has remained static over time.
Risk factors
The precise aetiology of IBD is unknown. However, it is known to be the result of
interactions between several factors:
1
This, however, is associated with extra-intestinal manifestations of Crohn’s disease.
Pathogenesis
Inflammatory bowel disease occurs when there is an inappropriate immune response
mounted against luminal antigens, such as bacteria. Bacterial ligands interact with the
innate & acquired mucosal immune system via Toll-like receptors. There are deficiencies in
the clearance of invading bacteria. This may allow inappropriate activation of the acquired
immune system.
In addition, special bacteria have distinct immunologic effects mediated by dendritic cells
which sample bacteria from the intestinal lumen and direct the differentiation of naïve T-
cells into effector & regulator populations. There is an imbalance in the relative numbers of
intestinal homing effectors (Th1 & Th17) and regulatory T-cell populations. This disturbs the
normal tolerance to the luminal antigenic load.
Pathology
The pathologic features of the 2 conditions differ:
Crohn’s disease
Crohn’s disease may affect any part of the GIT, from the mouth to the anus. It does,
however, have a particular tendency to affect the ileum & ascending colon (ileocolonic
disease; 50% of cases). It can involve one small area in the gut, or it can involve multiple
isolated areas, with regions or normal gut mucosa in between lesions (called skip lesions). It
can also involve the whole colon (total colitis) without involving small bowel, thus mimicking
ulcerative colitis.
Macroscopically, the involved bowel is usually thickened & narrowed. Deep ulcers & fissures
produce a cobblestone appearance on endoscopy. Fistulae & abscesses may develop, and
these represent penetrating disease. An early feature seen is aphthoid ulceration, which
later progresses into larger deeper ulcers. Microscopically, the inflammation extends
throughout all layers of the mucosa. There is an increase in chronic inflammatory cells and
lymphoid hyperplasia. In 50-60% of patients, there are granulomas which are non-caseating
epithelioid cell aggregates with Langhan’s giant cells.
Ulcerative colitis
Ulcerative colitis is mainly limited to the colon & rectum. It can involve the rectum alone
(proctitis) or it can extend proximally to involve the colon. Unlike Crohn’s disease, ulcerative
colitis forms one single extensive lesion which always begins in the rectum and ascends
proximally. It can therefore simply be a proctitis (in the rectum), extend to the descending
colon (left colitis) or involve the whole large intestine (total colitis). In a few patients, there
is also inflammation of the distal ileum, a phenomenon called backwash ileitis.
Macroscopically, the mucosa looks reddened & inflamed, and it bleeds easily. In severe
disease, there is extensive ulceration with the adjacent mucosa appearing as an
inflammatory polyp (pseudopolyp). Microscopically, the inflammatory infiltrate & the
ulceration are limited to the lamina propria. There are also crypt abscesses & goblet cells
depletion. Granulomas are rare.
Extra-GI manifestations
The extra-gastrointestinal manifestations can be seen with both diseases, but they are more
likely in Crohn’s disease than in ulcerative colitis:
Clinical features
The clinical features of inflammatory bowel disease depend on the actual disease present:
Crohn’s disease
Ulcerative colitis
Complications
The complications of Crohn’s disease are:
• Anal strictures.
• Anal fistulae.
• Perianal disease, leading to formation of tags.
• Toxic megacolon. On a plain abdominal X-ray, the patient will have dilated thin-
walled colon loops with mucosal islands. It is a serious disease, with impending
perforation & mortality reaching 15-25%. Urgent surgery is required in these
patients.
• Venous thrombosis.
• Colonic cancer.
Investigations
The investigations done for inflammatory bowel disease are:
2
This may also cause psoas irritation.
3
These are less than with Crohn’s disease.
Medical management
Patients with good prognosis disease – older age at diagnosis, no perianal disease, limited
ulceration at index investigation, non-smoker – may not require maintenance therapy.
Those with poor prognosis disease or those that have a flare after withdrawal of induction
therapy require maintenance therapy. This can be achieved with long-term
immunosuppressive therapy:
Surgical management
Approximately 80% of patients will require surgery at some point in their lives. Nonetheless,
surgery should be avoided as much as possible. Minimal resections should be done, as
recurrences are common if prophylactic maintenance therapy is not used. The indications
for surgery are:
• In patients with small bowel disease, the stricture can be widened, while others
require resectioning & anastomosis.
• When Crohn’s disease involves the entire colon and the rectum is spared, a subtotal
colectomy with ileorectal anastomosis is conducted. An eventual recurrence rate of
2
60-70% is expected, although 3s of these patients retain functional rectum for at
least 10 years.
• If the whole colon & rectum are involved, a panproctocolectomy is done (whole
colon + rectum) with end ileostomy. The patient then needs to have an ileostomy
bag attached. Patients with Crohn’s disease are not suitable for a pouch operation.
Medical management
• Mild ulcerative colitis. Aminosalicylates acting topically in the colonic mucosa form
the mainstay of treatment. The active moiety is 5-aminosalicylic acid (5-ASA), and for
it to be delivered effectively, it is combined with sulfapyridine (to form sulfasalazine),
4-aminobenzol-β-alanine (balsalazide) or to 5-ASA itself (olsalazine) it can also be
given as it is (mesalazine). The mode of action of 5-ASA is unknown, although it may
involve modification of PPAR-γ signalling. Steroids may help with induction of
remission, and these can be tapered down if improvement is seen within the first 2
weeks.
Surgical management
Surgery is required at some stage in about 20% of patients. The indications for surgery are:
4
This can complicate with “pouchitis”, which is characterised by diarrhoea, bleeding, fever, and exacerbation
of extra-colonic manifestations. It is treated with antibiotics.
Pathogenesis
Cirrhosis is a result of necrosis of hepatocytes. Necrosis initiates repair, which is initially
through primary intention with reconstitution of hepatocytes. However, conditions leading
to repeated inflammation of the liver, or chronic low-grade smouldering inflammation,
result in healing by secondary intention.
Fibrosis is initiated by activation of stellate cells, which are also known as Ito cells and are a
store of vitamin A (retinoids). In the early stages, stellate cells are activated to transform by
growth factors such as PDGF & TGF-β1. These growth factors are secreted by inflammatory
cells, hepatocytes, Kupffer cells and the sinusoidal epithelium. The stellate cells are
transformed into collagen-forming cells, which form collagen I & III fibres which are then
deposited in the space of Disse. These fibres are normally metabolised by matrix
metalloproteases (MMPs), but are inhibited by tissue inhibitors of metalloproteases
(TIMPs). Deposition of collagen in the space of Disse leads to loss of fenestrations in the
sinusoidal capillaries in the liver, leading to capillarisation of hepatic sinusoids. The fibrosis
in the space of Disse cause constriction of hepatic sinusoids, and this contributes to portal
hypertension.
Within the increasingly fibrous extracellular matrix, islands of regenerating cells form. The
cellular islands are separated by fibrous septa. There is loss of the classical lobular
architecture in these cellular islands. The regenerating islands of cells form nodules, which
can be macronodules or micronodules. Macronodular cirrhosis (>3mm) occurs in chronic
viral hepatitis, and is typified by large nodules of variable size with some normal acini in the
larger nodules. Micronodular cirrhosis (<3mm) is seen with chronic alcoholism and biliary
tract disease.
• Dehydration.
Aetiology
The main causes of cirrhosis are:
• Viral hepatitis. HBV & HCV chronic hepatitis are the viral causes of cirrhosis. Cirrhosis
is more likely to develop in HBV & HCV co-infection, as well as HBV & HDV co-
infection. Viral hepatitis is the leading cause of cirrhosis in the developing world
where the prevalence of these viruses is high.
• Alcohol. Chronic alcoholism leads to repeated hepatocyte damage, which leads to
chronic inflammation. Chronic alcoholism is the leading cause of cirrhosis in
developed countries where the prevalence of viral hepatitis is low.
• Non-alcoholic fatty liver disease. 1% of cases of non-alcoholic fatty liver disease
eventually evolve to cirrhosis. In non-alcoholic fatty liver disease, oxidative stress
leads to lipid accumulation in the liver. Fatty infiltration leads to inflammation, which
leads to fibrosis & cirrhosis. Non-alcoholic fatty liver disease affects 3-6% of the
population in the US.
Clinical presentation
The majority of patients are asymptomatic and have no complaints. The ones that do,
however, either have signs of chronic liver disease or have features of decompensated liver
disease.
• Jaundice.
• Abdominal swelling due to ascites. Patients may also develop ankle swelling.
• Confusion (due to encephalopathy).
Complications of cirrhosis
The complications of liver cirrhosis are:
Portal hypertension
Portal hypertension is an elevation of the blood pressure within the portal vein. The normal
portal venous pressure is 5-8mm Hg, with only a small gradient across the liver to the portal
vein. When there is blockage of outflow through the portal vein, the portal pressure rises. If
it rises above 10-12mm Hg, collaterals occur within the systemic venous circulation. The
main sites of collaterals are: the lower oesophagus, the rectum, the left renal vein,
diaphragm, retroperitoneum and the anterior umbilical wall. The causes of portal
hypertension can be classified into pre-hepatic, hepatic and post-hepatic.
Variceal haemorrhage
Ascites
• Sodium & water retention results from peripheral vasodilation and consequent
reduction in the effective blood volume. This activates the RAAS system, thus
promoting sodium & water retention. Urinary sodium excretion rarely exceeds
5mmol/24h.
• Portal hypertension exerts a local hydrostatic pressure that leads to increased
production of lymph by the liver & blood vessels and increased transudation of fluid
with little protein into the peritoneum.
• Low serum albumin results from chronic liver disease. This reduces the oncotic
pressure.
The abdominal swelling develops over many weeks or days. It can be precipitated by a high
salt diet, splanchnic vein thrombosis or a new hepatocellular carcinoma. Abdominal pain &
discomfort are common (if abdominal pain is more severe, suspect spontaneous bacterial
peritonitis). On examination, you can demonstrate shifting dullness & a fluid thrill.
To investigate ascites, you aspirate ascitic fluid (through an ascitic tap) and conduct
investigations on it. You do the following:
1
Schistosomiasis is the most common underlying cause of non-cirrhotic variceal bleeding in the world.
Porto-systemic/hepatic encephalopathy
This is a chronic neuro-psychiatric syndrome that results from diversion of blood from the
liver and directly into the systemic circulation. It is thought that the toxic metabolites in
portal blood that are normally processed by the liver enter the systemic circulation and
reach the brain. The causative toxins include ammonia, free fatty acids, mercaptans and
false neurotransmitters. Another theory is that GABA produced by the GIT crosses the extra-
permeable blood vessels of individuals with cirrhosis, leading to generation of IPSPs
(inhibitory post-synaptic potentials).
Patients typically have elevated free venous ammonia. CT/MRI of the brain is important in
ruling out intracranial lesions.
Hepatorenal syndrome
Hepatorenal syndrome is a condition in which there is cirrhosis, ascites and renal failure. It is
the result of massive splanchnic & systemic vasodilation but with renal vasoconstriction. The
initiating factor is production of nitric oxide which leads to hypotension. This activates the
RAAS system and leads to elevated levels of angiotensin II, which vasoconstricts the renal
vasculature. There is increased pre-glomerular resistance, leading to shunting of blood away
from the renal cortex. This reduces the GFR, with salt & water retention. This leads to
reduced urine output with a low sodium concentration. Hepatorenal syndrome is
precipitated by over-vigorous diuretic therapy, NSAIDs, diarrhoea, paracentesis and
infection.
• Type 1 HRS. This is a rapidly progressive deterioration in circulatory & renal function,
with a median survival of 2 weeks. It is often triggered by other pathology.
• Type 2 HRS. This is a more steady deterioration in circulatory & renal function.
Trans-jugular intrahepatic porto-systemic shunting is the best option for most.
Hepatocellular carcinoma
This is a major cause of mortality amongst patients with cirrhosis. It evolves faster in
patients with viral hepatitis involve multiple viruses (HBV, HCV and HDV). HCC develops in
10-25% of patients with cirrhosis. It occurs in about 3% of patients per year.
Cholangiosarcoma occurs in 10% of patients with primary sclerosing cholangitis.
• Serum albumin & prothrombin time. The best indicators of liver function are serum
albumin & prothrombin time. The outlook is poor when serum albumin drops below
28g/L. Prothrombin time increases as the severity of liver dysfunction progresses.
• Liver biochemistry. Bilirubin levels are normal or slightly increased. There is a slight
elevation in serum transaminases & ALP levels. These are grossly elevated in
decompensated cirrhosis.
• Urea & electrolytes. A true hyponatraemia develops because of a defect in free
water clearance or excessive diuretic therapy. An elevated creatinine level above
130µM is a bad prognostic indicator.
• Viral markers. For HBV, you can test for the presence of HBsAg. For HCV, you do RT-
PCR.
• Serum auto-antibodies. This is done when autoimmune diseases are suspected.
• Iron indices & ferritin. This is done to check for haemochromatosis.
• Copper & ceruloplasmin2. This is done to check for Wilson’s disease.
• A1-antitrypsin1.
Grading of cirrhosis
2
These should always be measured if the cirrhotic patients are young.
You then add the score that the patient has in each parameter. Patients are then stratified
according to their Child scores.
Management
Cirrhosis is irreversible, and management of cirrhosis is mainly to prevent progression of
fibrosis and to manage complications.
Long-term management
• Lifestyle modification. Patients should abstain from alcohol. Patients should also
take a balanced diet and restrict salt intake. Patients should avoid NSAIDs, opioids
and sedatives. Regular exercise is encouraged.
• Pharmacologic management. Colestyramine may help with pruritus. You can use
ursodeoxycholic acid for patients with primary biliary cirrhosis, which normalises
LFTs but does not alter progression of condition. You can treat autoimmune hepatitis
using azathioprine & prednisolone. Supplementation of vitamin D is important for
patients at risk of osteoporosis. Vaccination against hepatitis A is also important.
• Liver transplant. This is the only definitive treatment for cirrhosis. It significantly
increases 5-year survival from 20% to 70%. For acute hepatitis, liver transplantation
is indicated for patients with fulminant hepatic failure of any cause, including acute
viral hepatitis. For chronic liver disease, it is indicated for cirrhosis that is no longer
Treating complications
1
Budd-Chiari syndrome is a condition in which the hepatic vein is blocked. This syndrome is independent of
the cause of the blockage, although most blockages are a result of thrombosis. It causes prominent ascites,
hepatomegaly and abdominal pain.
• Full blood count (FBC). This is to identify any anaemia, thrombocytopenia and
leukopenia.
• Liver function tests (LFTs). The liver is the only site of albumin synthesis, and
therefore hypoalbuminaemia can serve as an indicator of liver dysfunction. However,
this is not a reliable test as albumin levels in circulation fluctuate naturally in
response to other non-hepatic causes. Prothrombin time, rather, is used to assess
liver function as it is more sensitive to liver function.
• Other tests assessing the cause of cirrhosis include hepatitis serology and ferritin &
ceruloplasmin levels.
• Ultrasound. These reveal features suggestive of cirrhosis & portal hypertension such
as a nodular liver, splenomegaly and collateral circulation.
• CT scanning & MRI scans. These are useful when the ultrasound evaluation is
inconclusive.
2
All these are complications of portal hypertension.
Treatment of the portal hypertension itself can be achieved by using β-blockers to lower the
portal pressure.
Physiology of anaemia
The major physiologic mechanisms relevant to anaemic patients are: the degree to which
oxygen delivery to tissues is adequate, and compensatory mechanisms for maintaining
oxygen delivery that will not become overwhelmed or deleterious. Oxygen delivery is
determined by the product of cardiac output & arterial oxygen content:
Anaemia in the postoperative setting has effects on the overall mortality of patients. Based
on a study on postoperative mortality in relation to haemoglobin levels, the mortality rates
were as follows:
• Blood loss. Transfusions should be considered when there has been a massive acute
bleed. It is usually indicated when there has been blood loss of more than 15% of
total blood volume in an otherwise healthy individual. This can be secondary to
trauma or major surgery. It can also be given prophylactically.
• Bone marrow failure. Causes include post-chemotherapy, some drugs, leukaemias
and chronic illnesses (e.g. CKD).
• Red blood cell disorders. These may be inherited (e.g. homozygous β-thalassemia,
red cell aplasia, sickle cell anaemia) or acquired (myelofibrosis, myelodysplasia,
selected use in renal disease).
• Neonates. Exchange transfusions are usually done for neonates. The indications
include: haemolytic disease of the new-born, neonatal jaundice, meningococcal
septicaemia, malaria.
Historically, the target for transfusion was 10g/dL. However, this has since been associated
with higher levels of morbidity & mortality. Transfusion is usually necessary when the
haemoglobin level falls below 7g/dL.
• Whole blood. Whole blood contains blood that is physiologic, i.e. with quantities
that are in the same proportions as in normally-occurring blood. It is blood that is
taken as is and is delivered un-separated. It is now rarely available in civilian practice
1
You can also thaw it at room temperature, but it takes longer. Higher temperatures will destroy clotting
factors & proteins.
2
Usually 6 packs of cryoprecipitate are required per patient. The packs may be separate or pooled into 1 large
pack.
3
Do not store it at 2-6°C.
4
Platelets should never be placed in the fridge.
5
When non-group-specific blood is being transfused, red cell concentrates (packed cells) are preferred
because plasma has been removed.
6
This may indicate that the blood has been contaminated, allowed to freeze or has become warm.
Complications of transfusions
The complications of transfusions are:
7
The filter removes clots and small clumps of platelets & white cells that develop during blood collection &
storage.
8
Blood is only to be warmed in a blood warmer and not in a water bath.
9
In adults, this is 5-6 litres (or 70ml/kg). In infants, it is 85ml/kg.
10
If none of these is available, give cryoprecipitate instead. It also contains factor VIII & fibrinogen.
11
There is little citrate in red cell concentrates & red cell suspensions.
Vitamin B12 is released from food by acid & pepsin, and then becomes complexed to an R-
binder in the stomach1. In the duodenum, B12 is released from the R-binder pancreatic
enzymes and binds to intrinsic factor (IF) released from gastric parietal cells. The IF-B12
complex is then carried down to the terminal ileum where it is absorbed. This complex binds
to a complex called cubillin, and the new complex binds to another protein called
amnionless which facilitates the uptake of the IF-B12-cu complex into the enterocytes.
Successful absorption of vitamin B12 therefore depends on the presence of:
The complex is then broken down and B12 is released into the circulation. In the circulation,
it is transported bound to plasma transcobalamin II (TCII) [70-90%] & haptocorrin. It is then
transported to the liver & bone marrow, where it is stored. On an average diet, a human
being will absorb 5-25µg of vitamin B12 every day. However, only 1-5µg is used daily by the
body. The rest of it is stored in the liver & bone marrow. The body has total stores of 2-5mg
of vitamin B12.
1
This protein is related to plasma transcobalamin I (TCI), a salivary protein.
Pathophysiology
The major consequence of vitamin B12 deficiency is megaloblastic erythropoiesis. Vitamin
B12 deficiency leads to a rate of DNA synthesis that is too slow to keep with cell growth &
division. Megaloblasts are erythroblasts with delayed nuclear maturation because of
defective DNA synthesis. Megaloblasts are large and have large immature nuclei. The
nuclear chromatin is more dispersed than normal. On occasion, giant metamyelocytes are
also seen in megaloblastic anaemia. It is also suggested that methionine deficiency may
contribute to the slowing of DNA synthesis.
The cause of anaemia can be addressed by the kinetic (quantitative) & morphological
(qualitative) approach. There is ineffective erythropoiesis, and that contributes to ultimately
reduced red cell output (largely by increased apoptosis), as reflected by the reduced
reticulocyte count. There is also increased intramedullary haemolysis of red cells before
they are released into the circulation. Therefore, there is intense erythroid hyperplasia but
relative reticulocytopenia. Megakaryocytosis is also affected: there is an increased number
of megakaryocytes, but they have diminished ploidy and therefore they have a reduced
capacity to produce adequate platelets.
For anaemia to develop in B12 deficiency, there needs to be either depletion of body stores
or inhibition of the utilisation of the vitamin. The body has 2-5mg of cobalamin stores, but
uses 1-5µg per day. Therefore, it takes years for a person who stops taking vitamin B 12 to
develop vitamin B12 deficiency symptoms simply from not eating enough.
Aetiology
2
Homocysteine levels will accumulate because methionine is used to form SAM (S-adenosine methionine),
which acts as a methyl donor for many other reactions, such as methylation of cytosine.
• Low dietary intake. This is more common in vegans & vegetarians who preferentially
do not eat meat. It is uncommon in people who skip meat every now and then.
• Pernicious anaemia. This is an autoimmune condition that is caused by a reaction to
intrinsic factor. This leads to the inhibition of absorption of vitamin B 12 in the gut. It
is a form of atrophic gastritis. There are many mechanisms as to how the antibodies
inhibit the interaction.
o There could be production of anti-parietal antibodies that attack parietal cells
of the gastric glands, leading to atrophy of the gastric mucosa. These are
found in 90% of patients.
o There could be production of blocking antibodies that interfere with the
interaction between intrinsic factor and vitamin B12.
o There could be production of binding antibodies that bind to the IF-B12
complex and interfere with the absorption of the complex.
It occurs in 1 in 8000 individuals in the UK. It is more common amongst women, and
it usually presents in the elderly. It also has an association with other autoimmune
disorders.
• Previous gastric surgery. This reduces the available gastric glands for the secretion
of acid and production of intrinsic factor. This reduces the body’s capacity to absorb
vitamin B12.
• Ilial surgery. Vitamin B12 is absorbed in the terminal ileum. Therefore, if you get ileal
resection surgery, you are likely to impair the capacity for absorption by reducing its
surface area. This forms part of the short bowel syndrome.
• Pancreatic insufficiency. Pancreatic insufficiency means that the pancreatic enzymes
that normally cleave the complex between vitamin B12 & R-binder cannot be cleaved.
• Crohn’s disease. Crohn’s disease also affects the terminal ileum and impairs its
absorptive capabilities.
• Drugs. Prolonged use of proton pump inhibitors leads to vitamin B12 deficiency. This
is due to inhibition of production of acid, which is vital for the liberation of B12 from
food and is subsequent binding to intrinsic factor. Cancer drugs such as
methotrexate, mercaptopurine and cytosine arabinoside impair the absorption of
the vitamin.
• Fish tapeworm. The fish tapeworm Diphyllobothrium latum is known to cause
vitamin B12 deficiency. This infection, however, is not common.
• Malabsorption syndromes. Tropical sprue & coeliac disease3 can cause vitamin B12
deficiency.
• Bacterial overgrowth in small intestine. Any condition leading to bacterial
overgrowth in the small intestine can cause vitamin B12 deficiency.
3
Coeliac disease often does not cause vitamin B12 deficiency because often the terminal ileum is spared.
The neurologic features of disease can occur without anaemia. They include:
Investigations
The investigations that are done in vitamin B12 deficiency include:
• FBC. This will reveal a macrocytic anaemia with an Hb of less than 12.5g/dL and MCV
greater than 100fL (unless there is a concurrent cause of microcytosis). The red cells
demonstrate an increased red cell distribution width (RDW), which shows the
significant difference in size between red cells. In severe cases, there is a
4
This vitamin B12 contains 58Co.
Management
The treatment of vitamin B12 deficiency requires 2 things: treating the cause, and replacing
vitamin B12. The cause of vitamin B12 deficiency needs to be treated if possible – many of the
causes can be treated in some way.
Vitamin B12 levels also need to be replaced. This can be done in a number of ways:
Clinical improvement may occur within 48hours. Reticulocytosis can be seen in 2-3 days.
Maximum improvement of polyneuropathy & other neurologic features can take 6-12
months. Longstanding spinal cord damage is mostly irreversible. Iron deficiency often
develops in the first few months following therapy and if this happens supplements should
be given. Hypokalaemia can develop during the first few months due to rapid utilisation
during production of new red cells. It may be profound in those that are severely anaemic at
the beginning of treatment.
Aetiology
Anaphylaxis is the result of widespread mast cell degranulation. This can be caused by an
immunologic reaction or non-immunologic reaction.
Immunologic anaphylaxis
Non-immunologic anaphylaxis
This has been implicated for various drugs. Some of the potential mechanisms include:
1
Cremophor EL (now known as Kolliphor EL) is a solvent formed from mixing castor oil with ethylene oxide. It
is usually mixed with many drugs, such as paclitaxel, propofol, miconazole, diazepam, vitamin K and nelfinavir
(protease inhibitor).
Pathophysiology
Anaphylaxis occurs as a result of widespread systemic degranulation of mast cells &
basophils, releasing histamine into the circulation. The mediators of anaphylaxis include:
• Mast cells & basophils. Mast cells and basophils degranulate release a number of
mediators, which include:
o Histamine. Histamine is a known vasodilator, and it also mediates other effects
similar to those of an allergic reaction. Localised release is associated with
urticaria. Widespread histamine release leads to increased vascular permeability
& vasodilation and is not associated with urticaria. Histamine acts on H1 & H2
receptors alike. H1 actions include tachycardia, pruritus, rhinorrhoea and
bronchospasm. Combined H1 & H2 actions include flushing, hypotension and
headache. The binding of histamine to H1-receptors leads to PLC-mediated
calcium mobilisation, leading to increased nitric oxide synthase and increased
nitric oxide levels. This mediates increased vascular permeability & vasodilation.
o Tryptase. Tryptase is a protease that is relatively specific to mast cells, but
basophils & myeloid precursors contain a small amount. Tryptase can activate
complement & coagulation pathways, as well as the kallikrein-kinin contact
system. Potential consequences include hypotension, angioedema, clotting and
clot lysis (with possible DIC). Orally-ingested allergens trigger release of lower
levels of tryptase than parenterally-administered allergens2, and apart from food
allergies, tryptase levels correlate with disease severity.
o Platelet-activating factor (PAF). Serum PAF correlates directly with the severity
of anaphylaxis. Serum PAF hydroxylase, however, correlates inversely with the
severity of anaphylaxis.
2
The opposite, however, is true about antigen-specific IgE antibodies: IgE antibody release is higher with
orally-ingested allergens.
Organ effects
In humans, the predominant shock organs are the heart, vasculature and the lungs.
Fatalities are divided between cardiovascular collapse and respiratory arrest.
3
DIC accounts for 8% of deaths from anaphylaxis.
Clinical presentation
The classic presentation of anaphylaxis comprises of the following:
Criterion 1: Acute onset of illness (minutes to hours) involving skin & mucosal tissue (e.g.
generalised hives, pruritus or flushing, swollen lips, tongue and uvula) AND:
1. Respiratory compromise – dyspnoea, wheezing, stridor, reduced PEF (in older
children & adults), hypoxaemia.
2. Reduced blood pressure or associated symptoms of end organ dysfunction, e.g.
hypotonia, collapse, syncope, incontinence.
Criterion 2: 2 or more of the following that occur rapidly (minutes to hours) following
exposure to a likely allergen for that patient.
1. Involvement of skin or mucosal tissues – generalised hives, itchiness, and swollen
lips, tongue and uvula.
2. Respiratory compromise – dyspnoea, wheeze, stridor, reduced PEF in older children
& adults, hypoxaemia.
3. Reduced BP or associated symptoms – hypotonia, collapse, syncope, incontinence.
4. Persistent GIT symptoms – crampy abdominal pain, nausea & vomiting.
Criterion 3: Reduced BP after exposure to a known allergen for that patient.
• Infants & children: age-specific low systolic BP, or >30% decrease in systolic BP.
o 1-12 months: 70mm Hg.
Differentials
The differential diagnoses for anaphylaxis include:
Management
Anaphylaxis is a medical emergency and management should be initiated immediately
before tests are conducted.
Emergency management
It is managed as follows:
1. Airway support. You must secure the airway by giving 100% oxygen at 6-8L/min and
intubating the patient if respiratory obstruction is imminent. Before, administering
oxygen, ensure that the airway is free. Cricothyrotomy may be necessary.
2. Epinephrine. You must give 0.5mg of epinephrine (or 0.5ml of 1:1000)
intramuscularly into the mid-anterolateral thigh. Also establish venous access. If
symptoms are severe, an intravenous infusion should be prepared. Repeat every 5
minutes if shock persists (as guided by BP, pulse and respiratory function).
3. Remove allergen. At times, this is a drug that is being infused; you can stop infusion.
You must raise the legs to help restore circulation.
4. Intravenous fluids. Give a bolus of 1-2L in adults (20ml/kg in children) of normal
saline as a rapid infusion. Titrate against blood pressure. Be wary of massive
Post-emergency intervention
4
This is administered only for urticaria & itching alone.
5
The ratio of total-to-mature tryptase in anaphylaxis is typically <10 and it is lower with higher tryptase levels.
in other conditions, the ratio is often greater than 20.
Pathogenesis
The majority of patients present with pulmonary manifestations, and it is thought this is the
initial lesion. The initial lesion is a CD4+ T-cell alveolitis, followed by development of a non-
caseating granuloma. The granuloma has a tightly-packed central area composed of
macrophages, epithelioid cells and multinucleated giant cells, and these are surrounded by
lymphocytes, monocytes, mast cells and fibroblasts. The sarcoid granuloma can resolve
without sequelae or undergo fibrosis and lead to interstitial fibrosis.
There is depressed cell-mediated immunity and reduced reactivity to tuberculin skin test.
There is also an overall lymphopaenia: circulating T-cells are decreased, but there is an
increase in circulating B-cells. Bronchoalveolar lavage (BAL) shows a great increase in the
number of cells. Lymphocytes & alveolar macrophages are increased, but the percentage of
alveolar macrophages in the BAL is decreased. All these changes (decreased circulating
lymphocytes, changes in delayed-type hypersensitivity and increased lymphocytes on BAL)
reflect sequestration of lymphocytes in the lung. There is no evidence suggesting that these
patients suffer from an overall decrease in immunity.
Possible aetiology
The exact antigenic stimulus that causes sarcoidosis is unknown. However, several
associations have been made:
• Occupational exposures. Beryllium & its salts have been shown to produces
granulomata similar to those produced by sarcoidosis.
• Infections. M. tuberculosis has been isolated in many patients with sarcoidosis. Other
implicated infections include fungi and EBV.
• Family history. The risk of developing sarcoidosis is higher if you have a 1st-degree
relative who is suffering from sarcoidosis. The risk is higher if you’re Caucasian than
if you’re black (OR 18.0 vs. 2.8).
• HLA. Sarcoidosis is associated with HLA-DRB1 & HLA-DQB1 alleles.
Clinical manifestations
Investigations
The investigations done for sarcoidosis include:
4
There is no effect on fertility.
Management
Patients with stage 1 disease do not require treatment as most (85%) resolve on their own.
50% of patients with stage 2 disease also resolve spontaneously on their own. Persisting
infiltration visible on chest X-ray with normal lung function tests requires close follow-up.
Patients with acute sarcoid syndromes generally have a good prognosis and simply require
bed rest and NSAIDs.
• Corticosteroids. You start with high-dose prednisolone (30-40mg po od) for 4-6
weeks, then decrease to 15mg for 6-12 months according to clinical status. A few
patients relapse and may require another course or long-term therapy. Severe
persistent erythema nodosum or uveoparotid fever will respond rapidly to a 2-week
course of prednisolone 5-15mg po od. In more severe disease, intravenous
methylprednisolone can be used.
• Immunosuppressant therapy. You can use methotrexate, hydroxychloroquine,
cyclophosphamide or ciclosporin. You can try anti-TNF-α agents, such as infliximab,
adalimumab or etanercept.
Prognosis
Prognosis is poorer in black patients, where death rates up to 10% are recorded. Causes of
death include respiratory failure, cor pulmonale, myocardial sarcoidosis and renal damage.
Patients can also die from secondary aspergillomas developing in damaged lung tissue (also
aggravated by long-term immunosuppressant therapy). 20% of patients on steroid therapy
respond. The rest are unlikely to respond to steroids.
Aetiology
Systemic lupus erythematosus is an autoimmune disease resulting from development of
antibodies against self-antigens. The implicated antigens in the disease are nuclear antigens,
and this is why the disease is mostly not organ specific. The trigger initiating development of
autoantibodies in SLE is unknown. The proposed triggers are:
Pathogenesis
There are many proposed mechanisms of development of these antibodies:
• Joints & muscles. 90% of patients have polyarthralgia or arthritis that is symmetrical
and mainly affects the small joints. There is usually no joint erosion or deformity1 in
1
In rare cases, you can get major joint deformity – a phenomenon called Jaccoud’s arthropathy. This,
however, is not associated with joint erosion.
• Proteins & complement. 80% of patients have high levels of α2-microglobulin & γ-
globulins (particularly IgG). Patients may also have hypoalbuminemia. There are low
Management
SLE management needs to be discussed with the patient. You need to explain to the patient
that they have the disease for life, and that it is a chronic disease with remissions &
relapses. The aims of treatment are to avoid stimuli that may trigger exacerbation, to
control autoantibody production by immunosuppression and to limit organ damage, all with
the aim of optimising quality of life in the patient. You do need to advise the patient on the
effects of the disease on his/her appearance and debility due to fatigue. Flare-up of activity
often follows: exposure to sunlight, infections, pregnancy, surgery and stress. Therefore,
patients need to avoid exposure.
SLE is a relapsing & remitting disease and therefore immunosuppressive therapy is variable.
In mild disease, corticosteroids form the basis of treatment. Mild disease mainly comprises
of skin & joint symptoms. High dose aspirin (or any other NSAID) is used to treat arthralgia,
while skin/mucosal involvement is treated with topical steroids & chloroquine2.
In severe SLE, cytotoxic agents are used. Azathioprine is used in persistent disease to spare
the use of corticosteroids. Most patients can be controlled on a combination of
azathioprine, steroids & antimalarial agents. Cyclophosphamide used to be used to treat
vasculitis, cerebral SLE and severe lupus nephritis. Nowadays, cyclophosphamide is being
replaced by mycophenolate mofetil. Newer agents include rituximab (anti-CD20 monoclonal
antibody).
The activity of the disease is monitored by measuring the DNA antibody level, serum C3 &
C4 and CRP levels3.
Prognosis
The effects of the disease may be due to the disease process itself or from the complications
of the disease. SLE has a range of outcomes, from mild to fatal. If left untreated, it is likely to
cause renal failure & CNS lupus. As a result of corticosteroid use, patients may develop
atherosclerosis, infection and cancer. Overall, the 10-year survival rate for SLE is good –
90%.
In pregnancy, SLE raises the risk of spontaneous abortions, intrauterine death, pre-
eclampsia, IUGR and preterm birth. Fertility is usually normal except in severe disease.
Patients with anti-ss A or B have a 2% risk of giving birth of babies with neonatal lupus
syndrome. This presents with rash, hepatitis and foetal heart block. Women with SLE are
advised not to fall pregnant until the disease has been quiescent for at least 6 months.
2
Chloroquine is associated with renal toxicity and therefore low doses should be used
3
CRP level indicate infection, as it rarely rises due to SLE alone.
The majority of insulin produced by the pancreas is carried to the liver, which is the prime
target organ. 50% of circulating insulin is extracted by the liver, with the C-peptide levels
being left relatively constant. The remainder of the insulin “survives” the liver and circulates
to the rest of the organs, and is extracted by the kidneys. In the liver, insulin binds to insulin
receptors, which are receptors linked to the tyrosine kinase signalling pathway. In the liver
and in many other tissues, insulin stimulates the insertion of vesicles containing GLUT4
channels and allows entry of glucose into the cell. It also stimulates glycolysis & glycogenesis
and inhibits glycogenolysis & gluconeogenesis.
Blood glucose levels are usually kept within the range of 3.5-8.0mM in normal people
despite the varying demands of food, fasting and exercise. The majority of carbohydrate
homeostasis is maintained in the liver, which stores excess glucose as glycogen and breaks
down glycogen whenever glucose is needed. The liver can also synthesis glucose from amino
acids and other 3-carbon compounds e.g. glycerol. About 200g of glucose is produced each
day: 90% from the liver through glycogen breakdown & hepatic gluconeogenesis, and 10%
from renal gluconeogenesis. The brain is the major consumer of glucose. It has negligible
1
The C-peptide can be measured to estimate the endogenous insulin production.
Primary diabetes mellitus is a result of a defect in glucose homeostasis that is not a result of
other disease states. It is a result of either decreased insulin production or increased insulin
resistance. Ultimately, there is some degree of β-cell dysfunction. There are 2 main types of
primary diabetes mellitus.
• Type I diabetes mellitus. This is a disease caused by absolute insulin deficiency which
usually involves destruction of β-cells. It is also called insulin-dependent diabetes
mellitus. Diabetes manifests when there is less than 20% of β-islet cells left. The
highest incidence of type I diabetes is in Finland & other Northern European
countries. There are many possible causes, and there are 2 main sub-classifications
of the disease.
o Type Ia. This is an autoimmune disease in which there is immune-mediated
destruction of β-cells. Many antigenic targets have been identified: GAD
(glutamic acid decarboxylase)2; 1A2 (tyrosine phosphatase); ZnT8 (zinc
transporter 8); and insulin. There is a strong genetic link with this condition,
but concordance in monozygotic twins is only about 45%. It is also associated
with other autoimmune diseases, such as autoimmune thyroid disease &
coeliac disease.
o Type Ib. This is an idiopathic disease in which the cause of islet cell
destruction is unknown. The disease has no associated autoantibodies.
There is increased genetic susceptibility, but the condition is not inherited. Genetic
susceptibility to type I diabetes mellitus is polygenic, and the greatest contribution
comes from HLA region. More than 90% of patients carry HLA-DR3-DQ2, -DR4-DQ8
or both. Other implicated genes include: the insulin gene itself (INS; chromosome
2
Antibodies against this antigen are associated with Stiffman syndrome.
3
This octate is important because the efficacy of oral hypoglycaemic treatment is measured by the number of
elements of the octate that the agent in question corrects.
4
In poor countries, diabetes is a disease of the rich, while in rich countries it is a disease of the poor (mainly
associated with obesity).
5
Usually, there also needs to be impaired glucose tolerance.
Secondary diabetes mellitus develops from another disease process that causes chronic
hyperglycaemia in a person with normal β-cells and without intrinsic insulin resistance. The
conditions are divided into:
• Family history. A large proportion of people living with diabetes report a positive
family history of diabetes. There is 45% concordance in monozygotic twins for type I
diabetes, and if one child has diabetes the chances of developing type I diabetes is
about 6%. With type II diabetes, however, the chances are higher: there is greater
than 50% concordance in monozygotic twins, and the risk to non-identical twins &
siblings is around 25%.
• Obesity. Obesity (BMI of greater than 30kg/m2) is a key risk factor particularly for
type II diabetes. Obesity leads to increased levels of free fatty acids, and this is
believed to induce insulin resistance all over the body. Obesity increases the
incidence of type II diabetes mellitus by 80-100 times. The inhabitants of affluent
countries gain 1g of body weight every day between the ages 25-55 years, and this is
because of a more sedentary lifestyle with little exercise rather than increased food
intake.
• Ethnicity. The incidence of diabetes is highest amongst black people, South-East
Asians and people from the Pacific islands.
• Pre-existing disease. The diseases associated with diabetes are hypertension,
coronary heart disease and dyslipidaemias.
Clinical presentations
The presentation of diabetes mellitus depends on the type of diabetes mellitus. There are
different features of the disease & patients that should make the clinician wary of the type
of diabetes being dealt with:
• Asymptomatic. This is often the case in type II diabetes, and it is detected as a high
blood sugar on routine medical check-up or after hospitalisation for some other
condition unrelated to diabetes.
• Symptoms of insulin deficiency. Patients typically present with polydipsia, polyuria,
polyphagia and weight loss. Polyuria develops due to the osmotic diuresis induced by
glucose excess in blood. Polyphagia develops largely due to the insulin deficiency,
which relieves insulin’s satiety effects. Polydipsia is a result of hypovolaemia caused
by polyuria. Weight loss results from the fluid depletion and the accelerated
breakdown of fat & muscle due to insulin deficiency. These symptoms are mainly
found in type I diabetics, and they usually develop within days or weeks of onset of
insulin deficiency. In type II diabetics, the symptoms are usually not as prominent,
especially weight loss. Patients may also present with ketones in their urine.
• Symptoms of complications. At times patients present with complications of
diabetes. This occurs either because patients overlook their initial symptoms, or
because there are no initial symptoms for the patient to pay attention to. This is
often the case for patients with type II diabetes. Patients may present with diabetic
ketoacidosis or hyperosmolar hyperglycaemic coma, which are direct complications
of hyperglycaemia. Other complications involve end-organ damage, and include
retinopathy, nephropathy (chronic kidney disease), polyneuropathy, erectile
dysfunction, etc.
• Random blood sugar (RBS) and Fasting blood sugar (FBS). These are point-of-care
tests that directly measure glucose tests. They are conducted using a small
automated glucometer device, and the test can be done in any setting. The nature of
the test and the interpretation of the results is what is important. In an RBS, you can
conduct the test at any time, although the test is less reliable because it does not
standardise the condition of the patient when the test is taken. It is therefore not
advised to be taken alone. An FBS is conducted after a patient has fasted for at least
12 hours, so it can be taken in the morning after the patient is advised to not eat
anything after 8pm. It is more standard as everyone in whom an FBS is done has
been fasted for that period. It requires, however, compliance with instructions. If the
FBS is too high, the patient has impaired fasting glucose.
• 2 raised venous glucose (FBS > 7.0mM, RBS > 11.1mM or OGTT-2h value > 7.8mM) in
an asymptomatic patient on 2 separate occasions that are 2 weeks apart.
• A single raised venous glucose (RBS > 11.1mM or FBS > 5.6mM) in a patient
presenting with symptoms.
• HbA1C value greater than 6.5% (or 48mM). A negative result, however, does not
exclude diabetes mellitus. The test is not conducted in pregnant women, patients
with type I diabetes and children.
Complications
The complications of diabetes can be divided into acute and chronic. Acute complications of
diabetes are:
Acute Chronic
• Diabetic Vascular:
ketoacidosis. • Macrovascular complications, e.g. atherosclerosis, leading
• Hyperglycaemic to coronary heart disease, stroke, renal artery stenosis,
hyperosmolar etc.
coma. • Microvascular complications, e.g. diabetic foot, diabetic
retinopathy.
Non-vascular:
• Dermopathy.
• Infections.
• Complications of dental procedures.
6
Do not rely on urinary ketones. They stay positive even after DKA has resolved.
7
This little insulin is sufficient to inhibit ketone body production.
8
These are obtained from U&Es.
• Dietary modification.
• Lifestyle changes.
• Pharmacotherapy.
• Monitoring, follow-up & review.
• Screening for complications.
• Psychosocial aspects of diabetes.
Dietary modification
The diet recommended for diabetic patients is no different from that considered healthy for
everyone. The dietary requirements for diabetic patients are:
1
The glycaemic index is defined as the AUC (area under the curve) of the 2-hour blood glucose response curve
after eating a particular test food following a 12-hour fast. The AUC of the test food is divided by the AUC
produced of a standard food item (glucose or white bread, thus producing 2 different definitions) and then
multiplied by 100. Foods with carbohydrates that break down easily an are readily released have a high
glycaemic index, while those that are more complex & break down slowly have a low glycaemic index.
When prescribing a diet, you need to look at the patient’s pre-existing diet and prescribe a
diet with the least possible interference with the patient’s lifestyle. Meals must not be
missed.
Lifestyle changes
The lifestyle changes the patient must employ are:
• Weight control. This is important for ensuring that you do not worsen any pre-
existing insulin resistance.
• Smoking cessation.
• Exercise. This is encouraged for all patients. Any increase in exercise is welcome, but
participation in a more formal exercise regimen is advised. Both aerobic & resistance
training improves insulin sensitivity & metabolic control in type I & type II diabetes.
They should be wary of the risk of hypoglycaemia 6-12 hours after exertion, though.
Always take a snack before & after sports.
• Alcohol. Alcohol is not recommended for diabetic patients.
Pharmacologic intervention
The drugs used for diabetes are divided into definitive medication and supportive. Definitive
medication aims at reducing blood glucose levels. There are 2 broad groups: injectable
insulin agents and oral antidiabetic agents.
Injectable insulin
This a broad group of insulin analogues used for insulin replacement therapy. There are 4
main classes of insulin drugs:
• Rapid-acting insulin. These include insulin lispro, insulin aspart and insulin glulisine.
This type of insulin is mainly used in continuous infusion insulin pumps. They are
taken just before meals.
• Biguanides. The main biguanide used for diabetes is metformin. Metformin acts
mainly in the liver to reduce insulin resistance. It is known to activate AMP kinase,
but its precise mechanism of action is not clear. It reduces hepatic glucose output
and increases insulin sensitivity. It does not affect insulin secretion or induce
hypoglycaemia, and it does not cause weight gain. It is therefore safe to use in
overweight patients. It also reduces cardiovascular risk. Metformin, however, causes
severe lactic acidosis in patients with renal or hepatic disease. It also causes
anorexia, diarrhoea and epigastric discomfort.
• Sulfonylureas. The drugs in this class include glibenclamide, tolbutamide, glipizide
and chlorpropramide. The drugs act as insulin secretagogues, and they close ATP-
sensitive potassium channels, thus increasing insulin release from the pancreas.
Their glucose-lowering effects are good during the first 1-3 years. However, their
function declines as β-cell mass declines. Glibenclamide is the one that is mostly
used, although tolbutamide is preferred in the elderly due to its short half-life.
Sulfonylureas predispose to hypoglycaemia. They should also be used with care in
patients with liver disease.
The treatment regimens depend on the type of diabetes the patient has:
Type 1 diabetes
For type 1 diabetes, insulin is the mainstay of therapy. It is always indicated for patients who
have had ketoacidosis. It is vital to educate the patient on how to self-inject insulin and to
self-adjust doses in the light of exercise, finger-prick glucose (glucometer) and caloric intake.
Finger-prick glucose before meals helps determine the long-acting insulin doses, while
finger-prick glucose after meals determines the short-acting insulin doses. Target glucose
levels are 4-7mM before meals and 4-10mM after meals.
Patients are advised to take a multiple injection dosing regimen containing a short-acting
and intermediate/long-acting insulin. It allows the insulin & food to go in at roughly the
same time. Examples of regimens include:
Type II diabetes
For type II diabetes, oral agents form the mainstay of treatment. However, in certain
situations, insulin therapy is added or substituted for oral agents. The indications for starting
insulin in type II diabetes are:
2
Also give this dose if the patient is on high-dose corticosteroids or has a known high degree of insulin
resistance.
If the patient is of normal weight, you give a combination of metformin (500-850mg 2-3
times daily) and glibenclamide (2.5mg once/twice daily). If poorly controlled despite strict
adherence to diet, you either change to insulin therapy, or you add it to the current therapy.
Monitoring
Diabetic patients are monitored as follows:
33
The minimum level detected on urine dipstick is 10-20mg/mol.
Thyroid hormones are synthesised from thyroglobulin stored in the follicles. The protein
undergoes iodination of tyrosine residues whilst in the colloid, and a large amount of the
protein is iodinated when iodine in the circulation is plentiful. Secretion is stimulated by TSH
(thyroid-stimulating hormone), which is an anterior pituitary hormone that is secreted
under the influence of TRH (thyrotropin-releasing hormone) released by the hypothalamus.
The thyroid hormones exert negative feedback on TSH & TRH secretion. TSH binds to
receptors on thyrocytes which stimulate the cleavage of iodothyronine residues from
thyroglobulin, and these are further processed to form triodothyronine (T3) &
tetraiodothyronine/thyroxine (T4). 90-93% of thyroid hormones in the circulation are T4
while 7-10% is T3.
Thyroid hormones travel bound mainly to thyroid-binding globulin (TBG). Other plasma
proteins involved in thyroid hormone transport are transthyretin (thyroid-binding pre-
albumin) and albumin. T4 has a higher affinity for plasma proteins than T3, and therefore T3
has higher free plasma levels. They have a very long half-life: T3 has a half-life of 1 day, while
T4 has a half-life of 6 days.
Thyroid hormones act in virtually all tissues of the body. They act on nuclear receptors. T 3 is
the active form and T4 is converted to T3 for the hormone to act properly. A considerable
amount of T4 is converted to reverse T3 (rT3), which is metabolically inert.
• T3 & T4 levels. This measures the amounts of each hormone in the circulation. You
can measure either free or total thyroid hormone levels in the circulation. The free
hormone measurement is more reliable as total hormone levels are affected by the
concentration of plasma proteins, particularly TBG1. The normal free T3
concentration is 3.5-7.5pmol/L, and the normal free T4 concentration is 10-25pmol/L.
thyroid hormone levels are elevated in primary hyperthyroidism, and decreased in
primary hypothyroidism.
• TSH. TSH measurement provides the single most specific, sensitive and reliable test
of thyroid status in clinically overt & subclinical thyroid dysfunction. It helps
distinguish the source of dysfunction when there are symptoms. In primary
hyperthyroidism, TSH levels are decreased while in primary hypothyroidism, TSH is
raised. In disease secondary to pituitary or hypothalamic dysfunction, TSH levels
alone are inadequate for diagnosis. The normal TSH level is 0.3-3.5mU/L.
• Thyroid antibodies. These are usually measured when autoimmune disease is
suspected. You can measure anti-thyroid peroxidase antibodies, anti-thyroglobulin
antibodies and anti-TRH/TSH receptor antibodies.
Usually, free T4 & TSH levels are the only ones required to diagnose thyroid disease.
Free T4 TSH
1
The affinities of albumin & transthyretin have little effect on the binding of T4.
They are some problems with interpretation of thyroid hormone levels in some patients.
They include:
1. Pregnancy. Pregnancy leads to increased TBG levels, thus distorting total T3 & T4
levels. Furthermore, the normal ranges for free T4 & TBG change over the course of
pregnancy. These are oestrogen-mediated changes.
2. Oral contraceptives. Combined oral contraceptives contain oestrogen, which lead to
changes similar to pregnancy: elevated TBG & total T4.
3. Serious acute & chronic illness. Illness can cause: reduced concentration & altered
affinity of binding proteins; decreased peripheral conversion of T4 to T3, with
increased rT3; and reduced TSH production. Systemically ill patients can therefore
have low total & free thyroid hormone levels with normal or low basal TSH levels.
This is called the sick euthyroid syndrome. The changes are mediated by IL-1 & IL-6.
4. Drugs. Amiodarone reduces conversion of T4 to T3, and therefore T4 levels can be
above normal in euthyroid patients. Amiodarone can also cause overt hyper- &
hypothyroidism. Many other drugs interfere with thyroid function.
Hyperthyroidism
Hyperthyroidism is an excess of thyroid hormones. It may or may not have clinical
manifestations, and when clinical manifestations are present it is called thyrotoxicosis.
Hyperthyroidism is very common, affecting 2-5% of all women during their lifetime. It is 5-
times more common in females than males and is most common between the ages of 20 &
40 years.
Aetiology
Nearly all cases are primary (>99%) – pituitary causes are very rare. The causes of
hyperthyroidism are:
2
The majority of follicular adenomas are cold – i.e. they do not secrete thyroid hormones.
Clinical features
Investigations
• FBC. There may be a mild normocytic anaemia and mild neutropenia in Graves’
disease.
• Elevated LFTs.
• Raised ESR.
• Hypercalcaemia.
• Radio-isotope scan. This can be used for a thyroid mass.
Management
Hypothyroidism
3
B-blockers can be used alone if the hyperthyroidism is self-limiting, e.g. sub-acute thyroiditis.
Aetiology
4
Hypothyroidism mostly affects women 6-times more than males.
Clinical features
• B – Bradycardia.
• R – Reflexes: slow-relaxing reflexes.
• A – Ataxia, which is cerebellar.
• D – Dry thin skin & hair.
• Y – Yawning & drowsiness. Even coma.
• C – Cold hands, hypothermia.
• A – Ascites. This may be associated with non-pitting oedema (myxoedema5) on the
lips, hands and feet. There may also be a pericardial and/or pleural effusion.
• R – Round puffy face. There may also be a double chin & obesity.
• D – Defeated demeanour (depression).
• I – Immobile with/without bowel ileus.
• C – Congestive cardiac failure.
Other features include: neuropathy, myopathy and goitre. Young children with cretinism
demonstrate growth retardation.
5
Myxoedema is caused by accumulation of mucopolysaccharides in subcutaneous tissues.
• Myxoedemic coma. This is when patients with hypothyroidism present with a coma.
It is a very rare phenomenon and the affected patients are often elderly patients.
Hypothermia is often present and patients often have cardiac failure, pericardial
effusion, hypoventilation, hypoglycaemia and hyponatraemia. Treatment is as
follows:
1. Most physicians would advise treating the condition with oral T 3 given at a
dose of 2.5-5µg, and then increasing the dose gradually. Large doses should
not be used.
2. Oxygen per face mask. Ventilation can be given if necessary.
3. Gradual rewarming.
4. Hydrocortisone 100mg intravenously every 8 hours.
5. Glucose infusion.
• Myxoedemic madness. Depression is common in hypothyroidism, and in the elderly
patients may become frankly demented or psychotic. It may be associated with
striking delusions. This may occur shortly after commencing T4 treatment.
• Pregnancy. In pregnancy, hypothyroidism can cause eclampsia, anaemia,
prematurity, low birth weight, still birth and postpartum haemorrhage.
Diagnosis
Management
Clinical improvement is not seen until at least 2 weeks of treatment. Full resolution of
symptoms may take 6 months. Also screen for the development of other autoimmune
disorders.
Causes
The causes of Cushing’s syndrome are classified in two ways. One way of classifying the
causes is to classify them into ACTH-dependent and ACTH-independent causes.
1
The zona glomerulosa is preserved as its function is not entirely dependent on ACTH production.
Clinical features
The clinical features of hypercortisolism are:
Investigations
The investigations for hypercortisolism are divided into diagnostic & supportive
investigations. Random cortisol level measurements are of no value as they are affected by
the time of day, illness and stress (even from venepuncture) all influence cortisol levels. The
diagnostic investigations are:
• U&Es. This may reveal hypokalaemia due to mineralocorticoid actions. In this case,
all diuretics must be stopped.
Treatment
Cushing’s syndrome can lead to death from hypertension, MI, infection and heart failure.
Management of Cushing’s syndrome therefore relies on both lowering the cortisol levels
and offering definitive management of symptoms.
1. Stop all corticosteroids. Since majority of cases are due to iatrogenic use of steroids,
stopping them should be the first step in treatment.
2. Reduce cortisol production. Cortisol levels can be lowered using medical
management. This, however, is not curative. Rather, it is used to prevent the
continued effects of high cortisol levels. You can use metyrapone (an 11β-
hydroxylase inhibitor) at a dose of 750-4000g daily divided into 3-4 doses. You can
also use ketoconazole 200mg 3-times daily – it is synergistic to metyrapone. Other
drugs include aminoglutethimide (blocks conversion of cholesterol to pregnenolone)
1g daily, and trilostane (3β-17-hydroxysteroid dehydrogenase inhibitor).
3. Surgical intervention. Surgical intervention can be employed on the pituitary gland
or the adrenal gland for pituitary or adrenal adenomas & carcinomas respectively.
The surgical interventions for pituitary adenomas are:
The prognosis of Cushing’s syndrome after treatment is generally good, although many of
the effects remain. These include myopathy, diabetes mellitus, obesity, hypertension,
menstrual irregularity, osteoporosis and subtle mood changes.
Pathophysiology
Adrenocortical insufficiency can be divided into primary and secondary. Primary
adrenocortical insufficiency is when the cause of insufficiency is due to a primary disease
process arising from the adrenal cortex. This is further divided into acute and chronic.
1
Neisseria-mediated sepsis is caused by an endotoxin.
Clinical features
The symptomatology of Addison’s disease is often vague & non-specific – it is called “the
unforgiving master of non-specificity & disguise”. For this reason, it is often diagnosed late.
Investigations
Investigations into adrenocortical insufficiency are urgent. If the patient is seriously
hypotensive, he/she must get hydrocortisone 100mg intravenously followed by intravenous
normal saline. This must be done, however, after taking a blood sample for cortisol
measurement. Single cortisol measurements, however, are of no use. Nonetheless, when
done you should suspect Addison’s disease when the cortisol level is below 100nmol/L
during the day. A cortisol level above 550nmol/L makes the diagnosis unlikely.
• Short ACTH stimulation tests. In this test, you try to assess the patient’s response to
tetracosactide (synthetic ACTH). You measure the baseline plasma cortisol, and then
you measure it again 2 hours after administering tetracosactide 250µg
intramuscularly. In normal individuals, the resulting cortisol level is usually above
550nmol/L. An absent or impaired cortisol response is diagnostic of adrenocortical
insufficiency. However, this test does not distinguish between Addison’s disease and
ACTH deficiency and iatrogenic suppression by steroids. Steroid drugs interfere with
the assay.
• 9am plasma ACTH level. This is usually high in primary adrenocortical suppression
(above 80ng/L). It is low in secondary causes.
• Antibodies. You can measure antibodies against 21-hydroxylase. These are positive
in autoimmune adrenalitis in 80% of cases.
• U&Es. These are used to assess for electrolyte imbalances. The classical picture is a
hyponatraemia, hyperkalaemia and a high urea level. However, these can be normal.
• Blood glucose. This may be low.
• Chest & abdominal X-ray. This is done to detect any evidence of past TB, such as
cavities, upper zone fibrosis and adrenal calcifications.
3
Postural hypotension & dehydration are spared in secondary adrenocortical insufficiency because ACTH
deficiency does not cause mineralocorticoid deficiency.
The main deficiencies are of salt, steroid and glucose. For management, you do the
following:
1. Give 1L normal saline over 30-60 minutes with 100mg intravenous hydrocortisone
bolus.
2. The patient will subsequently require several litres of normal saline over the
following 24 hours, coupled with 100mg of hydrocortisone intramuscularly every 6-
hours.
3. Glucose must be infused if there is hypoglycaemia.
4. Oral replacement medication is then started, unless the patient is unable to take oral
medication. Initially, you give hydrocortisone 20mg 8-hourly, and then reduce to 20-
40mg in divided doses over a few days.
The majority of patients are going to require treatment for life. Therefore, their education
requires the following:
Patients must come for annual follow-up for BP & U&E measurement. You should screen for
the development of other autoimmune diseases.
Diagnosis of HIV
HIV diagnosis is included under HIV testing services (HTS), which encompass HIV testing,
counselling, and linkage to appropriate HIV prevention, treatment and care services. HTS is
guided by 6 core principles: consent, confidentiality, counselling, correct results, comfort for
the mother in labour and connection-linkage to care & prevention services. Delivery of HTS
can be done in facilities (clinics, hospitals, etc.), in the community (home-based testing) and
self-testing. Self-testing is now encouraged for first-time testers as some form of “screening
test”. Once a person tests positive, they need to be linked to a formal health facility, and if
they test positive there they can be linked to treatment & care.
Routine testing for HIV in children above 18 months, adolescents and adults makes use of
antibody tests which are both rapid tests. The 1st test that is conducted makes use of HIV
Determine® or SD Bioline®. The 2nd test is conducted using the Chembio or the First
Response test. The way in which HIV is confirmed is as follows:
• If the 1st test comes out negative, the patient is reported negative but encouraged to
return after 3 months for a retest.
• If the 1st test comes out positive, you conduct the 2nd test.
o If the 2nd test comes out positive, the patient is reported positive and
directed to treatment & care.
o If the 2nd test comes out negative, the results are discordant and then you
repeat the tests concurrently.
• if after repeating the 2 tests:
o The tests come out negative – you report the patient as negative.
o The tests come out positive – you report the patient as positive.
o The tests are discordant – you do a 3rd test (INSTI)
• If after doing the 3rd test:
o The result is negative, you report the patient HIV negative.
o The result is positive, the test is inconclusive – the patient is to return after
14 days for a retest.
The effectiveness of ART relies on the efficacy of the regimen and adherence on the part of
the patient. The efficacy of ART depends on: potency of regimen chosen, minimal adverse
events, reduced pill burden, and accessibility & affordability of the medicines. Adherence
ensures that the medication is maintained at effective concentrations in the body so as to
suppress viral replication.
1
These are more pronounced with lopinavir.
The old regimen comprised of 2 NRTIs and 1 NNRTI, as having at least 2 drugs from different
classes improves efficacy of treatment. The common regimen used was tenofovir 300mg,
lamivudine 300mg and efavirenz 600mg (TDF/3TC/EFV), and efavirenz was now being rolled
out at 400mg due to the dose having less side effects. However, efavirenz is being replaced
by dolutegravir (DTG) 50mg due to the superior side effect profile. Tenofovir can be
exchanged for zidovudine, but zidovudine can cause fatal agranulocytosis as a side effect.
Tenofovir itself can cause renal disease (acute & chronic) and therefore it is replaced when
this happens. Efavirenz can be exchanged for nevirapine (particularly in the case of
neuropsychiatric side effects), although nevirapine causes rash and hepatotoxicity
particularly in individuals with higher CD4+ counts (>200 in men, >250 in women).
In patients starting on nevirapine in place of efavirenz, you start with a starter pack for 2
weeks. In the starter pack, tenofovir 300mg & lamivudine 300mg are given in the morning
while nevirapine 200mg is taken in the evening. After the starter pack is taken and no
adverse events develop, you step up to the maintenance dose. You then give tenofovir
300mg & lamivudine 300mg in the morning, and then give 2 dose of nevirapine; one 200mg
dose in the morning and another 200mg in the evening. If the patient had been on efavirenz
before, there is no need for the starter pack – you can start with the maintenance dose.
When you give zidovudine (300mg) in place of tenofovir, you give lamivudine at a dose of
150mg instead of 300mg. furthermore, zidolam (AZT/3TC) is given as a twice daily dose.
Second-line regimen
Second-line treatment should be considered only after assessing for adherence & failure,
and in consultation with a specialist team. There is no need for resistance testing because
NNRTIs have a low barrier to resistance and so the virus can easily become resistant to them
with single mutations. The barrier to resistance with PIs, however, is much lower.
Second-line treatment consists of 2 NRTIs and 1 PI standing in for the NNRTI. The usual PIs
used are ritonavir-boosted (with 100mg) lopinavir (LPV/r) 400mg or atazanavir (ATV/r). If
tenofovir was used in the 1st-line regimen, you replace it with zidovudine or abacavir 600mg.
When there is HIV & TB co-infection and the patient is receiving rifampicin, you can either
replace rifampicin with rifabutin, or you can double the dose of the PI. If patients have co-
infection with HBV, you need to always include tenofovir & lamivudine. ATV/r is taken once
daily while LPV/r is taken twice daily.
Third-line regimen
Those who require third-line treatment need confirmation of virus resistance prior to
initiation, because PIs have a high barrier to resistance and therefore if there is treatment
failure, it is more likely due to adherence issues or inappropriate dosing rather than
resistance.
Initiation of ART
ART is initiated in patients with conclusive positive test results for HIV. ART initiation is a
cooperative process that also involves the patient, and it requires the patient to be in a
sound state of mind. Simply rolling out medication will not achieve the desired effects for
both parties. There are many aims of ART:
Early treatment initiation is associated with clinical & HIV prevention benefits and early
initiation is associated with lower morbidity & mortality from opportunistic infections even
in patients with apparently good virological control. Untreated HIV also increases risk of
non-communicable diseases such as cardiovascular disease, cancer, renal disease, hepatic
disease and neurocognitive disorders. Prior to initiation of ART, however, it is important to
retest patients, even those with prior confirmed HIV tests.
1. History & examination. These need to be taken in full, and should include a detailed
social history.
2. Assessing readiness for ART. You need to discuss many issues with the patient.
These issues include:
a. The ARV regimen. The patient needs to be informed of the regimen they are
starting on. In the majority of cases, patients start on TDF/3TC/EFV, and this
can come as a fixed dose combination with all 3 medications taken once a
day.
b. The side effects of treatment. tenofovir
c. The requirement for monitoring.
d. The importance of adherence. Adherence is important, and patients must
continue taking ART for them to remain well. This is, however, improved
giving enough information about HIV, medications, expected adverse events,
3. Medical criteria for starting ART. All patients with confirmed HIV infection are
eligible for commencement of ART regardless of the WHO clinical stage.
• The patient has Cryptococcal meningitis. In this case, ART is deferred for a month.
• The patient needs further psychosocial counselling.
• The patient has TB. In this case, ART is deferred for at least 2 weeks.
• The patient needs further information on HIV & AIDS.
• The patient is terminally ill and unable to swallow oral medication. Palliative care is
then offered to this patient.
Monitoring of treatment
The components of monitoring of treatment include:
• Treatment adherence. At every visit you need to ensure that the patient is taking
their medication. There needs to be a 95% adherence rate, which comes to a
defaulting rate of about 1 in every 30 days. You need to ask about adherence the
right way, i.e. ask how often they miss medication, and this is more likely to get you
more detail. Adherence is improved by providing accurate & adequate information
on the disease and possible side effects of medication. Also encourage them to bring
medication containers to each visit.
• Frequency of visits. Patients can be seen once every 2-3 months. If a patient is
stable, they do not need to be seen by a clinician. A stable patient is one in whom
the viral load is suppressed (<1000 copies/mL), there are no OIs and the patient has
been on ART for at least 6 months. If viral load is unavailable, the last CD4+ must
have been greater than 200cells/mm3. There are 3 types of visits:
o Clinical visit. This is a scheduled visit where the clinician makes a thorough
assessment and reviews monitoring blood tests. A stable patient should have
1 clinical visit every 6 months.
o Refill visit. This is a visit for collection of prescription medication (ARVs). This
does not require the presence of a clinician.
2
In HBV confection, tenofovir & lamivudine are to be included at all times.
Treatment failure
Treatment failure is defined clinically, virologically and immunologically.
Before switching ART regimens, you need to ensure that the patient has been adherent to
medication. Adherence is affected by:
• Failure of adherence, which occurs in 90% of cases. This must be probed, as it may
be due to certain stresses in the patient’s life. Adolescents are the ones who are
more likely to experience this.
• Inadequate dosing. This is common in patients on TDF/3TC who switch to AZT/3TC
who are not informed that they are changing from a once-daily dosing regimen to a
twice-daily dosing regimen.
• Hyperemesis following dosing. This may be the case in pregnant women, and
therefore pregnant women need to be asked about hyperemesis. Hyperemesis
reduces the effective dose that is working in the patient.
• Sexual reinfection. This happens when 2 HIV-infected individuals have unprotected
intercourse. There is a high likelihood of a resistant strain being transmitted to the
patient which then fails to respond to treatment.
Parasitology
There are 5 Plasmodium species – P. falciparum, P. vivax, P. ovale, P. malariae and P.
knowlesi. The most virulent organism amongst these is P. falciparum, and it is also the most
common cause of death from malaria (the others rarely cause severe malaria). P. falciparum
is most commonly found in sub-Saharan Africa, while P. vivax is the most common form
found in Asia1. Plasmodium species are spread by female mosquitoes. Depending on the
region in question, there are different mosquito species that spread the infection. In
Zimbabwe, female Anopheles mosquitoes are the most common cause of spread of
infection. 2 species of Anopheles mosquito found in Zimbabwe are A. gambiae & A.
fenustus.
The life cycle of malaria consists of 3 phases – the sporogonic cycle, the exo-erythrocytic
cycle and the erythrocytic cycle:
The exo-erythrocytic cycle occurs in the liver.
1. Human infection occurs after a mosquito has deposited the infectious agents (called
sporozoites) into the circulation through its saliva.
2. The sporozoites are carried to the parenchymal cells of the liver, which they infect. In
the liver, the sporozoites can follow one of 2 routes:
a. They can reproduce asexually as merozoites and form what is called a
schizont. All malaria species can do this.
b. They can enter a dormant phase and form what is called a hypnozoite. P.
vivax & P. ovale can enter these forms. These can cause relapse of infection
months or years later.
3. The schizonts rupture, releasing merozoites into the circulation, thus allowing the
erythrocytic phase to occur.
The erythrocytic phase occurs in red blood cells.
1
P. vivax is the most prevalent parasite on the globe.
Pathophysiology
The clinical effects of malaria infection are a result of haemolysis, cytokine release and
impairment of the microcirculation.
• Travel to an endemic area. People who live in non-endemic areas are not protected
against malaria infection because of lack of immunity, and therefore are more likely
to develop infection when exposed.
• Age. In endemic areas, infections are more common in children aged 2-5 years, and
these are more likely to be severe. This does not go to say that adults from these
areas do not develop severe disease.
• Pregnant women. Pregnant women are prone to severe disease. many factors
contribute, such as sequestering of parasites in the placenta, hypoglycaemia,
anaemia, etc. The major consequences are severe anaemia in the mother and low
birth weight infants.
• Lacking the Duffy antigen. Lacking the Duffy antigen on the membrane of red cells
has been shown to be protective against developing P. vivax malaria infection.
• Haemoglobinopathies. Patients with sickle cell anaemia are protected from
developing malaria. Furthermore, patients with the sickle cell trait (heterozygous for
the sickle cell allele) are less likely to develop malaria than patients without the trait
(homozygous normal)3.
• HIV. HIV decreases T-cell mediated immunity to infection. Therefore, in patients who
are immunocompromised, malaria is more likely to progress to becoming a severe
infection. Malaria has also been shown to enhance the course of HIV infection
(enhanced viral replication & high viraemia).
• Splenic sequestration. The size of the spleen has been seen to correlate to the
number of antibodies produced. This means that children with larger spleens are
2
Rosettes formed in blood groups A & B are larger, tighter and stronger than those formed in blood group O.
3
This accounts for the persistence of these alleles in West Africa and other sickle cell & malaria endemic areas.
• Fever. The fever may be continuous or erratic, but it later becomes cyclical, occurring
every 48 hours (tertian fever) or 72 hours (quartan fever). There are generally 3
phases of the fever paroxysm: shivering, hot stage (temperature about 41°C
associated with rigors) and sweating stage.
• General malaise.
• Headache.
• Nausea & vomiting.
• diarrhoea
Other things to look for in the history:
• General: febrile patient, who is pale, but with no rash and no lymphadenopathy.
• Abdomen: hepatosplenomegaly. This may be tender.
• Nutritional status. This is important for children.
Clinical syndromes
The clinical syndromes produced by different Plasmodium infections are:
Complications of malaria
The complications of malaria are mostly related to its microvascular occlusive phenomena
secondary to rosetting of red blood cells. Other mechanisms include direct toxic effects of
haemoglobin (particularly with renal disease). The complications of malaria are:
Investigations
The diagnostic investigations done for malaria are:
• Rapid diagnostic test (RDT). This is a quick easy-to-conduct point-of-care test that
tests for malaria antigens. The antigen detected by RDTs is the histidine-rich protein
2 (HRP2), although some tests can also detect Plasmodial lactate dehydrogenase
(PLDH) and aldolase. The only drawback of the RDT is that it can only detect
falciparum malaria and not the other species.
• Peripheral smear microscopy. This is the gold standard for diagnosis of malaria, and
it makes use of Giemsa staining of a thick & thin smear. It is often done in tertiary
institutions. However, it requires expertise in determining aetiology, and it is time
consuming when case loads are high. The thick smear is used for determining
parasitaemia (quantitative/sensitivity testing) while the thin smear is used for
speciation (qualitative/specificity testing). At least 3 films should be examined before
malaria is declared likely. Parasitaemia above 10% in adults suggests severe
infection. In children, it is above 5% in non-endemic areas and 7% in endemic areas.
Generally, RDTs are done as first line for all suspected malaria cases. However, microscopy is
used for: all patients admitted for malaria (to improve follow-up); confirming co-infections;
those with a recent travel history to areas where other species are reported; suspected
treatment failure; and patients who have received treatment within the preceding 2 weeks.
Other optimisation tests conducted include:
• Full blood count. This is to check for anaemia and thrombocytopaenia. Anaemia is
diagnosed by a haemoglobin level below 5.5g/dL for patients coming from endemic
areas or below 7.5g/dL for patients coming from non-endemic areas. Leukocytosis
can also be suggestive of superimposed infection.
• Urea & electrolytes. This is used to screen for renal impairment. Renal impairment
can also be screened for using urinalysis.
• Liver function tests. You first want to check for bilirubin levels, and if there is
hyperbilirubinaemia, establish whether it is conjugated or unconjugated
hyperbilirubinaemia. Patients with hepatic injury can also get a transaminitis.
• Arterial blood gases. This is used to check for metabolic acidosis. Acidosis is defined
by a pH of less than 7.25 with a plasma bicarbonate below 15mM.
• Chest radiography. Pulmonary oedema can be seen on a chest radiograph.
• Blood glucose measurement. This can be done using POCT glucometer strips.
Hypoglycaemia is indicated by a glucometer reading below 3.4mM.
• Blood culture. This is done to rule out septicaemia.
4
If the stat dose is vomited out, repeat it. If vomiting persists, treat as severe malaria.
5
The combination therapy, not artesunate alone.
6
Phenobarbitone requires monitoring for respiratory depression, which may require ventilator support.
7
10% dextrose is preferred to ameliorate hypoglycaemia.
Hepatitis A
Hepatitis A is caused by the hepatitis A virus (HAV). It is the most common viral hepatitis
occurring worldwide, often occurring in epidemics. It causes about 40% of cases of acute
hepatitis. In western countries, however, its incidence is declining. It is mainly spread
through the faecal-oral route, and it. The disease mainly affects children & young adults.
HAV belongs to the family of Picornaviridae viruses, to which enteroviruses, rhinovirus and
cardiovirus belong – they are all spread by the faecal-oral route. HAV belongs to the
hepatovirus genus. HAV is a small unenveloped positive-sense single-strand RNA virus, and
it has a diameter of 27nm. It is taken up by the GIT and then transported to the liver, where
it replicates slowly.
Pathogenesis
The viral infection itself does not produce cytological changes that are toxic to cells. Rather,
it is the immune reaction that causes disease. IgM antibodies can be detected with the
onset of symptoms. Histologically, there is a portal & periportal lymphocytic infiltrate, with
periportal necrosis.
Clinical features
Patients initially present with non-specific features such as nausea & vomiting, anorexia and
develop distaste for cigarettes. This is the prodromal phase. Jaundice usually develops later
after 1-2 weeks (it is common in adults but is rare in children) and it is usually preceded by
dark urine and pale stool1. This is called the icteric phase. In the icteric phase, there may
also be abdominal pain, itching, arthralgia and a skin rash. If nausea, vomiting or any mental
confusion persists, the patient requires assessment at a hospital.
On examination, patients will be jaundiced once they are in the icteric phase. Hepatomegaly
is common, and splenomegaly occurs in 10% of cases. You should always remember to look
for features of chronic liver disease and rule out features of decompensation.
Complications
The complications of acute HAV infection include arthritis, vasculitis, myocarditis and AKI.
Chronic liver disease does not occur.
Investigations
• Liver function tests (LFTs). There is a transaminitis which begins prior to the
appearance of jaundice, but still occurs 22-40 days after exposure. Nonetheless, AST
levels peak 1-2 days after onset of jaundice and may rise above 500U/L.
Transaminase levels return to normal after 5-20 weeks.
• Serological tests. IgM rises from day 25 post infection, and indicates recent
infection. IgG persists for life.
• Faecal tests. You can do direct RT-PCR on faeces, and this tests positive sooner than
serological tests.
• Other blood tests. FBC will reveal a relative lymphocytosis. There may also be
anaemia (rare) from either Coombs’ test-positive haemolytic anaemia or from
aplastic anaemia. PT is prolonged in severe cases, and ESR is raised.
1
This is due to intrahepatic cholestasis.
Management
Treatment of acute HAV hepatitis is supportive. Admission is usually not necessary, unless in
fulminant cases in which you administer interferon-α.
You can immunise susceptible patients using Havrix Monodose®, which is an inactive
protein derived from HAV. You give 1ml intramuscularly, and this give immunity for 1 year
unless a booster is given at 6 months (which prolongs immunity to 20 years). You can give
Havrix Monodose Junior® to children aged 1-15 years. The vaccine is administered to people
travelling to endemic regions, patients with chronic liver disease, haemophilia patients and
workers who frequently contact hepatitis cases. If exposure will be in less than 2 weeks, you
give normal human immunoglobulins (0.02ml/kg intramuscularly).
Hepatitis D
Hepatitis D is caused by the hepatitis D virus (HDV). Due to defects in HDV, it requires HBV
for its infection, replication and spread and therefore exists as co-infection. About 15 million
people worldwide are living with HDV. HDV is spread through blood, semen and vaginal
secretions. However, it can only establish infection and replicate in individuals with
concurrent HBV infection.
Hepatitis D virus
The virion infects hepatocytes using the envelop HBsAg. HDV is peculiar, however, in that it
utilises the host cell’s RNA polymerase II to make its mRNA. Replication of the delta agent
results in cytotoxicity & liver damage. It is the direct cytopathic effect of HDV that leads to
damage.
Clinical syndromes
• Co-infection. Patients can get infected with both HDV & HBV at the same time. This
is clinically indistinguishable at the time. However, a biphasic transaminitis may be
HDV leads to a more complicated HBV infection: it hastens the progression to cirrhosis,
increases the incidence of fulminant hepatitis and increases development of hepatocellular
carcinoma.
Diagnosis
HDV can be detected by measuring HDV-RNA in serum, the delta antigen and anti-HDV
antibodies. You can order HDV tests in a patient who is HBsAg positive.
Treatment
There is no definitive treatment available for HDV. There is no enzyme in HDV that can be
blocked by drugs, as HDV predominantly depends on the host enzyme machinery to
replicate. You can try to control the HBV, and pegylated interferon-αs been shown to have
some anti-HDV activity. Liver transplantation may be needed in some cases.
Hepatitis E
Hepatitis E is caused by Hepatitis E virus, which is an enteral virus. It is common in
developing countries, and it occurs in epidemics in India, China, Pakistan, Nepal, Burma,
North Africa and Mexico. It is more common in men than women. In the UK, it is commoner
than hepatitis A.
Hepatitis E is its own unique virus, although it resembles caliciviruses in its size (27-34nm)
and structure. It is an RNA virus. It is transmitted through the faeco-oral route, and 30% of
dogs, pigs and rodents are carriers of the virus. Epidemics can occur in developing countries.
In developed countries, it occurs in sporadic cases, especially in individuals with a travel
history to a developing country or those in contact with farm animals.
It follows a course similar to that of hepatitis A: it has an incubation period of 15-50 days
and usually causes mild disease with an abrupt onset. However, symptoms appear later
than in hepatitis A. It has a mortality of 1-2% from fulminant hepatitis2. However, it can be
severe during pregnancy – its mortality in these patients is 20%. It can be diagnosed using
ELISA for IgG & IgM antibodies. It is treated by supportive treatment.
2
Hepatitis A has a mortality of less than 0.5%.
HBV is a very contagious virus. It is spread by sexual transmission, contact with body fluids,
blood products and intravenous drug use. The risk factors for development of hepatitis B
infection are:
Hepatitis B virus
HBV is a double-stranded enveloped DNA1 virus belonging to the Hepadnaviridae family. It
has an average diameter of 42nm. There are 8 serotypes, and each serotype is distributed in
a distinct geographical region. These serotypes have differences in their rate of
seroconversion as well as chronicity. The virion (also called a Dane particle) has a viral
envelope, which contains a surface antigen (HBsAg), and a capsid formed by the core
antigen (HBcAg). Within the capsid there is also an e antigen (HBeAg) which is sequentially
similar to HBcAg but is processed differently by the cell. The virion includes a protein kinase
and a reverse transcriptase.
The replication of HBV is special in that it replicates through an RNA intermediate and it
release HBsAg particles as decoy particles. The mechanism through which it infects
hepatocytes is not well understood. The viral DNA genome is delivered to the nucleus. The
viral genome does not incorporate into the hepatocyte DNA during replication because it
1
This is the only hepatitis virus that has a DNA genome.
Pathogenesis
Replication of HBV within hepatocytes leads to expression of the different HBV antigens.
• The surface antigen HBsAg is released into the serum of infected people, most as
HbsAg-rich particles that are not complete virions. These particles are more
numerous than the actual virion particles. These particles are also very
immunogenic. Antibodies developed against HBsAg (called anti-HBs) imply the
patient has been exposed either to HBV or to the vaccine.
• The core antigen HBcAg is incorporated into the viral capsid. Antibodies developed
against HBcAg (called anti-HBc) imply that there is minute persistence of the virus in
the circulation. The IgM anti-HBc implies acute infection or reactivation, whereas IgG
anti-HBc implies chronic infection. The activity of disease cannot be understood
using this antibody alone, however.
• The e antigen HBeAg is released during active replication of the virus. It therefore
indicates active infection that is replicating. The presence of this antigen also
indicates high infectivity. Antibodies to HBeAg (called anti-HBe) indicate
seroconversion with HBV, or a non-replicative state in a patient with negative HBV-
DNA.
The disease is not a result of any destructive effects of the virus, which itself is not
destructive. Rather, it is the resulting inflammatory reaction that causes disease. HBV-
specific cytotoxic T-cells recognise the viral antigen via MHC class I molecules expressed on
infected cells. Regulatory T-cells, however, limit the inflammatory reaction, and this allows
the HBV genome to persist and develop into a chronic infection.
• Replicative phase. The replicative phase is characterised by active viral replication &
hepatic inflammation. Both HBsAg & HBeAg are present in the circulation and the
patient is HBV-DNA positive. During this stage, the patient is very infectious, and
there is significant hepatocyte damage leading to a transaminitis.
2
However, it can be integrated into the host genome, and this has been found in hepatocellular carcinoma.
Clinical course
The course of infection is as follows:
• Immune-tolerant phase. This stage is the initial stage and it lasts 2-4 weeks (or 3-45
days)3. During this stage, slow active replication continues but seroconversion has
not occurred, and there is no hepatocellular damage. Active replication is indicated
by the presence of HBsAg & HBeAg, and the absence of seroconversion is indicated
by the absence of anti-HBe. HBV-DNA levels are very high (above 200 000IU/mL).
The absence of hepatocellular damage is indicated by normal ALT levels and minimal
histological disease.
• Immune active/clearance. It is during this stage that active disease occurs. There is
widespread destruction of hepatocytes. Patients develop positive HBeAg & HBsAg
due to widespread replication of the virus. Hepatocellular damage is signified by
elevated ALT levels and necro-inflammatory activity on histology. The duration of
this stage is usually 3-4 weeks in acute infection. In patients with chronic infection,
this stage may persist for as long as 10 years. This stage can resolve completely, or
progress to chronic infection. Seroconversion can occur at this stage.
• Inactive chronic hepatitis/immune-control phase. In these patients, there is chronic
infection leading to chronic liver disease. Patients seroconverted in this stage, and
therefore have positive anti-HBe in their blood. HBeAg clears slowly, and once it
clears it can remit. The host targets infected hepatocytes and HBV. HBV-DNA is low
or undetectable. ALT levels are within normal range. HBsAg & anti-HBc can be
detected in serum. This is associated with a low risk of complications, such as
cirrhosis & hepatocellular carcinoma.
• Chronic HBeAg-negative hepatitis/immune-escape mutant. In this stage, there is
negative HBeAg without anti-HBe. It results from a mutation in the pre-core or basal
core promoter gene, resulting in the production of virions without HBeAg. ALT levels
are abnormal, and this indicates background destruction of hepatocytes. HBV-DNA
levels are abnormal or fluctuating. Older patients are at risk of progressive disease,
with rapid progression to cirrhosis.
• Occult hepatitis B. In this form, only HBcAb can be detected. HBsAg and HBeAg are
both undetectable. This indicates hidden infection that reactivates when the patient
3
In neonates born with HBV, this stage lasts very long.
Clinical features
HBV hepatitis can be acute or chronic. Acute infection ranges from subclinical infection to
fulminant hepatitis. Acute hepatitis does not occur in individuals with congenital HBV
infection. Chronic hepatitis occurs in 5-10% of patients.
• History. The symptoms are of insidious onset in the majority of cases. Symptoms
include fever, malaise and anorexia followed by nausea, vomiting, abdominal pain &
discomfort and chills. The classical icteric symptoms of liver damage – jaundice, dark
urine and pale stools – follow thereafter. In fulminant hepatitis, patients have more
severe symptoms, such as ascites & bleeding. Patients may present with more
serious features, such as encephalopathy (confusion, coma,) and coagulopathy.
Chronic hepatitis is often asymptomatic and is detected when there is
decompensation. When they decompensate, they develop symptoms similar to
those of acute hepatitis, but with other features of portal hypertension.
In addition to symptoms, other questions need to be asked, such as history of
hepatitis, HBV vaccination status, HIV status, family history of HCC, and substance
use (especially alcohol and IV drugs).
• Examination. The features on examination are:
o General: low-grade fever, jaundice, palmar erythema, signs of chronic liver
disease (muscle wasting, Dupuytren’s contracture, spider naevi, palmar
erythema, peripheral oedema, gynaecomastia), signs of liver failure (liver
flap, fetor hepaticus, confusion).
o Abdomen: distended abdominal veins (caput medusa), ascites, tender
hepatomegaly (liver smooth in acute hepatitis, nodular in hepatocellular
carcinoma)4, splenomegaly.
4
In cirrhosis, it is small & shrunken.
Investigations
Prior to doing other tests, you first test for HBsAg & anti-HBs. If this is positive, you do:
• Hepatitis B profile. A hepatitis B profile includes: HBsAg, HBcAg and HBeAg. In acute
infection, where HBsAg can be cleared quickly, IgM anti-HBc is diagnostic.
5
If they are in copies per mL, you divide this value by 5 to get the corresponding value in IU/ml. for example,
200 000 copies/ml = 40 000IU/ml.
• Blood tests. You should also do an FBC and U&E to rule out anaemia of chronic
disease and renal impairment.
• HCV screening. You also need to screen for HCV, as HBV-HCV co-infection carriers a
poorer prognosis and has a more rapid progression.
• Imaging. You need to do an ultrasound of the liver to rule out malignancy as the
primary priority prior to biopsy. Once a lesion is suspicious of HCC, you take a triple-
phase high-resolution CT scan with contrast of the liver.
Management
Treatment of confirmed HBV depends on the stage of disease. Patients that require
treatment are those in the immune-reactive stage and those with chronic hepatitis. Those
with occult hepatitis only require treatment when they are undergoing immunosuppressive
therapy. The majority of patients recover completely, with fulminant hepatitis occurring in
1% of patients. The goals of treatment are to prevent progression to cirrhosis, liver failure
and hepatocellular carcinoma.
For acute hepatitis, treatment is mainly symptomatic, as most people will clear the
infection. Patients should, however, have their HBV markers monitored. If HBsAg persists
for more than 12 weeks, the patient should take antivirals. For chronic hepatitis, treatment
is indicated when there is chronic active infection, e.g. abnormal ALT levels and abnormal
HBV-DNA levels with/without positive HBeAg. Therefore, treatment is indicated for patients
with reactivation (decompensated chronic disease) or chronic HBeAg-negative disease.
6
In patients with decompensated cirrhosis, this flare can lead to further deterioration.
7
Orthotopic liver transplantation is the removal of the native liver and its replacement with a donor liver in the
exact same anatomic position as the original liver.
Monitoring treatment
Monitoring treatment of HBV comprises of: monitoring of treatment response, monitoring
of disease progression, monitoring for tenofovir & entecavir toxicity, and monitoring for
hepatocellular carcinoma.
Monitoring is as follows:
• Annual monitoring. You should do HBV-DNA, HBeAg9, HBsAg and ALT levels. These
are done for patients that do not meet the criteria for treatment. In those without
cirrhosis at baseline, you need to do non-invasive tests to assess for likelihood of
development of cirrhosis.
• More frequent monitoring. this depends on whether patients are on treatment or
not:
o Patients who do not meet the criteria for treatment: you monitor more
frequently if patients have intermittently abnormal ALT or HBV-DNA levels.
More frequent monitoring is also indicated in patients co-infected with HIV.
In these patients, the aim is to identify a change in clinical status, such as an
APRI score > 2, development of HCC, and a rise in ALT or HBV-DNA.
o Patients on treatment or following treatment discontinuation: you monitor in:
patients with more advanced disease (compensated/decompensated
cirrhosis); first year of treatment to assess adherence & treatment response;
patients in whom adherence is an issue; HIV-positive patients; patients after
discontinuation of treatment.
• Monitoring for tenofovir & entecavir toxicity. Tenofovir causes proximal tubular
dysfunction, which ranges in severity from mild renal tubular dysfunction &
hyperphosphataemia to classical Fanconi syndrome & impaired GFR. It also causes
osteoporosis, severe hepatomegaly with steatosis and lactic acidosis. Risk factors for
tenofovir toxicity include low CD4 count, low body weight and underlying renal
dysfunction. To monitor for tenofovir toxicity, you need to measure baseline renal
function and assess for baseline risk of renal dysfunction, and then monitor renal
8
The majority of vertical infections occur perinatally.
9
If the patients initially tested HBeAg, you must conduct anti-HBe.
Prevention of HBV
HBV can be prevented by:
Hepatitis C virus
Hepatitis C virus (HCV) belongs to the Flaviviridae family of viruses, to which yellow fever
virus, Dengue virus, West Nile virus, etc. belong to. These viruses are enveloped viruses that
have a positive-sense single-stranded RNA genome. It has a diameter of 50nm. The viral
envelop consists of glycoproteins, with one E protein (E1) folding over another (E2). The
viruses infect hepatocytes, but can also infect B-lymphocytes.
The virus enters cells by means of receptor-mediated endocytosis. The receptor it binds on
is CD-81 (tetraspanin), through which it interacts using E2. The virions can also coat
themselves with LDLs or VLDLs and uses these lipoproteins’ receptors to enter hepatocytes.
The viral envelop then fuses with that of the endosome upon acidification and it releases
the virus into the cytosol. The viral genome is then translated into a polyprotein which is
then cleaved to form non-structural early proteins (NS2, NS3, NS4, NS5A and NS5B) and
structural proteins. The non-structural proteins work as helicase (NS3), protease (NS2 &
NS3) and RNA-dependent RNA polymerase (NS5B) enzymes. The structural proteins include
the C- (core protein) and the E-proteins (envelop proteins E1 & E2). After virion assembly,
the virus assembles and buds into the endoplasmic reticulum and becomes associated with
the cell.
Pathogenesis
HCV proteins inhibit apoptosis & IFN-α action by binding to the TNF receptor and to protein
kinase R. These actions prevent death of hepatocytes. The ability of HCV to remain cell
associated & prevent host cell death both help the virus persist in the body.
Cell-mediated immunity accounts for both resolution of infection and tissue damage. The
inflammatory reaction can exhaust CD8+ cytotoxic T-cells to prevent resolution of infection.
1
More than hepatitis B.
Aetiology
The majority of infections are acquired through infected blood. The risk factors & methods
of spread are:
• Transfusion of infected blood. This was once the leading mode of spread of the virus
prior to identification of the virus and screening of blood products.
• Use of non-sterile needles. This is currently the leading mode of spread in developed
1
countries. It is thought that 3 of young IV drug users in America are infected with
HCV.
• Occupational exposure. Healthcare workers are particularly prone to contracting HCV
through needle stick injuries. Needle stick injuries pose a 3% risk of transmission of
the virus.
• Sexual transmission. This mode of transmission, however, is very rare and
contributes to a very small number of new cases. A higher rate of transmission,
however, occurs in homosexual men.
• HIV. HIV increases the likelihood of contracting HIV. More than 90% of HIV-positive
people who are also intravenous drug users are also infected with HCV. In contrast,
only 4% of those that acquire HIV through sexual intercourse also have HCV.
• Male gender.
• Alcohol intake.
• Non-alcoholic fatty liver disease.
• Concurrent HBV infection.
• Diabetes.
Clinical features
The clinical features of HCV are:
• Acute viral hepatitis. These patients are mostly asymptomatic, with about 10%
presenting with a mild flu-like illness with jaundice and a transaminitis. There are
also prominent extra-hepatic features, such as arthralgia & arthritis, paraesthesia,
myalgia, pruritus, sicca syndrome (Sjögren’s syndrome) and sensory neuropathy.
15% of patients clear the infection within 6 months.
• Chronic persistent infection. This occurs in the majority (70%) of patients. Chronic
active hepatitis develops within 10-15 years, and cirrhosis develops in 20% of chronic
cases after 20 years. This may be exacerbated by alcohol, certain medications and
other hepatitis viruses. This syndrome is often asymptomatic and is detected
through routine biochemical testing. If symptomatic, the commonest presentation is
fatigue, and it may be accompanied by nausea, vomiting, weight loss and anorexia.
These symptoms do not correlate with disease activity.
Patients with cirrhosis can present with decompensation, which causes synthetic
dysfunction & portal hypertension. They can present with ankle swelling, abdominal
distension, melena, hematemesis and mental status changes.
• Rapid onset cirrhosis. In 15% of patients, hepatitis C rapidly progresses to cirrhosis.
Examination
Most patients with HCV infection have no findings on physical examination. Patients with
extra-hepatic features, however, may have signs such as porphyria cutanea tarda or
necrotising vasculitis. The findings of acute/acute-on-chronic hepatitis are:
Complications
The complications of HCV infection are:
• Cirrhosis. The risk of cirrhosis after 20 years of infection is 15-30%. This is initially
compensated, with decompensation occurring later leading to variceal
Investigations
The investigations done for hepatitis C are:
• Liver function tests. In acute infection, the LFTs are usually deranged. ALT levels are
usually elevated more than AST levels such that AST:ALT ration is less than 1.
However, this ratio is reversed in the setting of cirrhosis. In chronic infection, ALT
levels are normal unless there is decompensation.
• Antibody tests. Testing for anti-HCV antibodies confirms previous exposure. These
antibodies are not very useful in an acute infection as antibodies appear about 8
weeks after infection. The test is very useful in patients with chronic infection. False-
negative tests, however, can occur in patients with compromised immune systems
such as those with HIV, renal failure or HCV-induced cryoglobulinaemia. There is also
a rapid antibody test called the OraQuick HCV Rapid Antibody Test, which is used for
patients at risk for hepatitis.
• HCV-RNA assays. This is the gold standard for detecting HCV in an infected
individual. You can use RT-PCR, branched-chain DNA assay (a signal amplification
technique) and other related techniques. HCV-RNA assays are divided into
quantitative assays and qualitative assays:
o Qualitative assay. This is used to detect the presence of HCV in the
circulation.
o Quantitative assay. This is used to determine the amount of HCV in blood. It
makes use of RT-PCR & branched-chain DNA assay. The quantitative assay
2
HCC from HCV infection is associated more with cirrhosis than HBV because HBV is integrates with the host
genome and is more likely to cause carcinogenesis without preceding fibrosis. HCV, however, first causes
fibrosis which leads to chronic regenerative activity which eventually leads to HCC.
Pharmacological intervention
3
Opioid substitution therapy is supplying illicit drug users with a replacement drug for opioid drug users.
Examples include methadone & buprenorphine.
Mycology
Microscopically, Cryptococcus organisms are spherical encapsulated yeast-like organisms
that multiply by budding. Gem tubes, hyphae and pseudo-hyphae are usually absent from
these organisms. The capsule is made out of polysaccharide, and it may be up to 5-times the
diameter of the yeast cell.
There are many serotypes of Cryptococcus: C. neoformans includes A (C. neoformans var
grubii), D (C. neoformans var neoformans), AD and C, while C. gatti includes B & C. The
majority of infections are caused by C. neoformans serotype A (var grubii), which typically
affects immunocompromised individuals. C. neoformans is found as a ubiquitous saprophyte
of the soil, particularly that which is enriched by bird droppings. The organism is found in
high concentrations in pigeon droppings, although the birds themselves do not get infected
by the fungus. In moist excreta, the fungus can persist for up to 2 years. However, in a
saprobic environment (rich in organic matter but lacking oxygen), the fungus can remain un-
encapsulated and is generally non-virulent.
Cryptococcus organisms are mainly spread by inhaling aerosolised cells from the
environment. This accounts for the initial colonisation of the respiratory system.
Subsequent dissemination of infection from there can occur in certain situations.
Pathophysiology
When the aerosolised Cryptococcus fungi are inhaled, they travel to the alveoli, where they
must survive the neutral/alkaline pH here. An enzyme called glucosylceramide synthase
1
The most common C. gatti strain that causes meningitis is C. tetragatti.
The host response to Cryptococcal infection includes both cellular & humoral components.
NK cells participate in the early killing of Cryptococcal cells and in ADCC (antibody-
dependent cell-mediated cytotoxicity). Anti-Cryptococcal antibodies & soluble anti-
Cryptococcal factors also play a role in macrophage- & lymphocyte-mediated immune
response.
C. neoformans can cause pneumonia and meningitis. Patients with pneumonia can develop
poorly-defined lung masses, pulmonary nodules and pleural effusions in rare cases. The
majority of these patients are immunocompetent. Meningitis is common in the setting of
AIDS. It can also cause limited cutaneous disease, although this is rare. Infection causes
little/no necrosis in other organs except in advanced disease. This is primarily due to organ
distortion secondary to increasing fungal burden.
Risk factors
The risk factors for Cryptococcal infection are:
• HIV/AIDS. This is the most common risk factor in countries where HIV prevalence
rates are very high. In these patients, infection usually develops when the CD4 + is
very low (below 100 cell/mm3). C. neoformans is the most commonly isolated
organism in these patients. In immunocompetent patients, C. gatti is isolated in 70-
80% of cases.
Cryptococcal meningitis accounts for 15% of deaths from HIV. There were 223 100
cases of crytpococcal meningitis amongst HIV-infected individuals in 2014, and 181
100 of those patients died.
• Immunocompromise from other conditions. This can be due to steroid use and use of
immunosuppressing agents for cancer therapy or other conditions.
• Age. Cryptococcal infection can occur in individuals of any age, and the mean age of
incidence is younger in HIV-infected individuals.
• Sex. Cryptococcal meningitis is more common.
• Underlying pulmonary disease. This may promote the initial colonisation & infection
by the fungus.
• Malignancies. Malignancies can also lead to immunosuppression, which predisposes
to Cryptococcal infection.
Pulmonary cryptococcosis
• History. Patients present with fever, malaise, cough with scant sputum and pleuritic
chest pain. Haemoptysis is rare. The history is often a chronic rather than acute one.
• Examination. Less than 15% of patients show a picture of pneumonia. Cryptococcus
isolated from sputum is often due to colonisation rather than true infection,
especially in patients with COPD or bronchiectasis.
Cryptococcal meningitis
• History. The most common symptoms are a chronic history of headache, nausea &
vomiting, altered mental status (personality changes, confusion, lethargy and coma).
Fever & neck stiffness are less common since they are due to an aggressive
inflammatory response (typically found in patients with high CD4 + counts).
• Examination. Patients will be ill-looking, have altered mental status and neck
stiffness. Patients with cryptococcosis present with focal neurologic defects.
Cutaneous manifestations
Investigations
The investigations done in cryptococcosis are:
• CSF studies. CSF should be taken to screen for Cryptococcal meningitis. You need to
test for the Cryptococcal antigen (CrAg). High opening pressures above 25cm H2O
have poor prognosis and serial CSF taps need to be done to maintain opening
Management
The management of Cryptococcal infection is as follows:
Cryptococcal meningitis
Cryptococcal meningitis leads to death within 2 years in all untreated cases. It is treated as
follows:
• Induction phase. This is done in first 2 weeks. You can start with a combination of
amphotericin B deoxycholate 1mg/kg/d intravenously and fluconazole 1200mg (or
12mg/kg for children & adolescents up to a maximum of 800mg) once daily orally.
Alternatively, you can start with high dose fluconazole (1200mg orally daily or
12mg/kg for children & adolescents) and flucytosine 100mg/kg/day divided into 4
doses for 2 weeks. You can also give amphotericin B 1mg/kg/day & flucytosine
100mg/kg/day divided into 4 doses for 1 week, followed by 1 week of fluconazole
1200mg daily for adults and 12mg/kg/day up to a maximum of 800mg for children &
adolescents.
• Consolidation phase. This is administered for 8 weeks. You give fluconazole 800mg
once daily orally for adults and 6-12mg/kg/day up to a maximum of 800mg.
• Maintenance phase. This is administered until the CD4+ count is above 200
cells/mm3 for 6 months. It is only indicated in patients living with HIV and not
2
Prior to tapping, however, you need to take a CT/MRI scan of the head to exclude space-occupying lesions.
• Therapeutic CSF tapping (keeping CSF opening pressure below 20cm H2O – you
remove 10-30ml of CSF). There is no need to take a CSF tap at the end of the
induction phase.
• Immediate initiation of ART is not recommended. It should be deferred for 4-6 weeks
after initiation of antifungal treatment.
Pulmonary cryptococcosis
Cryptococcoma
Classification
Kaposi sarcoma is classified into 4 types:
• Classic/sporadic KS. This is an indolent form and is most common in men (by 10-15
times) in the eastern Mediterranean & Jewish communities. It is usually limited to
skin. As many as 30% of patients eventually develop secondary malignancy,
particularly non-Hodgkin’s lymphoma.
• Endemic KS. This form is more common in sub-Saharan Africa amongst men. This
form shows extensive cutaneous lesions with lymph node involvement. Oedema is a
prominent feature. Visceral involvement is rare. The risk of developing endemic KS is
increased by rarely/never wearing shoes in areas with volcanic clay soils (linked to
chronic lymphatic obstruction by fine soil particles).
• Epidemic/AIDS-related KS. This is the most common form found in the world due to
the AIDS pandemic. KS is 500-times more common in HIV-positive people than in the
general population. It is also more common in gay men. KS usually manifests in
individuals with a low CD4+ count and is one of the AIDS-defining illnesses. Survival is
also improved with ARVs. Epidemic KS is very aggressive, and usually has visceral
involvement.
• Immunosuppression-related/iatrogenic KS. This form is found amongst patients on
immunosuppressive therapy, such as organ transplant patients or patients with
autoimmune disease. Just like the epidemic form, it follows an aggressive course
with visceral involvement. The average time for development of KS in these men is
15-30 months. Sirolimus, used to prevent transplant rejection, appears to be
protective against development of KS.
Cigarette smoking is protective against KS. Relative affluence is also associated with
increased risk endemic & epidemic KS.
Pathogenesis
95-98% of cases of KS have been associated with infection by HHV-8 (also known as the
Kaposi sarcoma virus, KSV). It underlies all types of KS. The virus is thought to be spread via
saliva, not sexual contact – intercourse is thought to be a mere surrogate for other close
contact. The virus can also be transmitted through solid organ donation. The HHV-8 genome
has also been identified in Castleman’s disease, some lymphomas and leiomyosarcomas. A
The cell of origin is still unknown, although a pluripotent mesenchymal progenitor cell has
been suggested. The cells develop into spindle cells, which proliferate on a background of
reticular fibres, collagen and mononuclear cells including macrophages, lymphocytes and
plasma cells. The lesions may only involve the reticular dermis (patch stage) or involve the
whole dermis (plaque & nodular stages). The tumour nodules are also believed to arise from
multiple independent foci rather than being metastatic from a single focus. This, however, is
applicable to less aggressive cutaneous KS and not the aggressive visceral forms.
Clinical presentation
History
Physical examination
• General: ill-looking depending on extent of disease. The patient will have often
bilateral oedema which may be pitting or non-pitting. Areas where lymphoedema
once developed will become hyperpigmented & woody hard. In active KS, the woody
hard areas are usually warm to touch. Patients often have lymphadenopathy, and all
Investigations
Staging of KS
No precise staging system has been accepted universally. Nonetheless, there are many
classifications that are used for KS:
This is also known as the TIS staging which is based on 3 parameters: tumour, immunologic
status and systemic illness:
T – tumour
T0 (good Confined to skin and/or lymph nodes, and demonstrates minimal oral disease
risk) (with flat rather than raised lesions).
T1 (poor Widespread KS, as evidenced by:
risk) • Oedema due to tumour.
Mitsuyasu classification
Treatment of KS
There is no treatment that is able to eradicate HHV-8 and therefore there is no cure for KS.
The purpose of therapy is to alleviate symptoms, slow disease progression and prevent
relapses.
Medical therapy
• ART. ARV therapy can be tried in HIV-infected individuals with cutaneous disease
only. The response to ART ranges from 30-80% depending on the stage of disease
and the amount of treatment. However, poor-risk patients on ART may not respond
Local therapy
Monitoring
Since no cure for KS is available, patients require life-long monitoring. Patients are at risk of
relapse even if they achieve remission. Patients should have routine physical examination
every 3-4 months to check for any new lesions, oedema and signs & symptoms suggestive of
visceral involvement. Patients living with HIV should also be monitored using an FBC,
metabolic profile, CD4+ count and viral load. HIV control is important in minimising relapse
The distribution of Schistosomiasis depends on the species in question, and this is due to the
distribution of snail species in the world:
• S. haematobium is mostly found in North Africa, sub-Saharan Africa, the Middle East,
Turkey and India.
• S. mansoni is the most widely distributed Schistosoma species. It is found in some
parts of tropical & subtropical sub-Saharan African, the Middle East, Saudi Arabia,
South America (Brazil, Suriname, and Venezuela) and the Caribbean.
• S. japonicum is regarded as an Oriental blood fluke: it is found in China, Japan, the
Phillipines, Thailand and Sulawesi (Indonesia)
• S. intercalatum is found in Central & West Africa.
• S. mekongi is restricted to Laos & Cambodia.
• S. guineensis is found the rainforests north of Congo.
In endemic areas, the infection is acquired in childhood and the prevalence peaks at 15-20
years. In adults, the prevalence does not change significantly, but the parasite intensity
decreases drastically.
1. Humans, who serve as the definitive host, in which sexual reproduction takes place.
2. Snails, which serve as the intermediate host, in which asexual reproduction takes
place.
1. The eggs of the schistosomes are laid in either urine or faeces (depending on the
species)1. The eggs are round with spines. S. haematobium has a terminal spine
while S. mansoni has a lateral spine.
2. In water, the eggs hatch and release larvae called miracidia (sing. miracidium).
3. The miracidia penetrate snails and turn into sporocysts. These multiply in the snail,
and this is the asexual reproduction stage.
4. The snail then release mature larvae called cercariae (sing. cercaria). Cercariae have
a head connected to a forked tail, which allows them to swim in water.
5. Cercariae penetrate human skin and enter the circulation. Here, they lose their tails
and become schistosomulae (sing. schistosomula).
6. After developing, the schistosomulae travel in the circulation until they get to the
portal vein. In the portal vein, the schistosomulae develop into adult schistosomes.
Adult schistosomes have an elongate cylindrical structure, and like other flukes they
have an oral sucker (opens into an incomplete digestive system) and a ventral sucker
(for attachment). They are not hermaphroditic, and they have definitive sexes. The
males are wider than the females and have a slit called the gynaecophoric canal, in
which the female enters. This is where copulation takes place.
7. After development, the adult schistosomulae then travel to their final destinations.
They can travel against the flow of blood to the mesenteric veins (small intestines for
S. japonium & S. mekongi; large intestines for S. mansoni & S. intercalatum). They
can also travel to the vesical plexus (S. haematobium). In their definitive positions,
the females can lay as many as 300-3000 eggs per day for 4-35 years. The eggs travel
in the circulation to other sites. They can also traverse the intestinal/bladder mucosa
and reach the lumen (through release of enzymes by larvae inside the eggs), where
they are excreted in urine or faeces.
Pathogenesis
Adult worms develop an elaborate defence system against host resistance. They coat
themselves with substances that the host recognises as self and consequently they do very
little damage to the host. Only when they migrate to the spinal cord or the brain do they
cause damage.
The eggs, however, are responsible for causing disease. The disease is the result of the
mounting of an immune response to the eggs laid by the females. Eggs invade local tissues
and release toxins & enzymes which mediate a TH2-mediated immune response.
Inflammation & granuloma formation occurs around these eggs, and if the reaction is
chronic enough can lead to fibrosis & scarring of the affected tissue. The eggs of S. mansoni,
S. japonicum, S. intercalatum and S. mekongi tend to penetrate the intestinal wall or travel
1
Urine for S.haematobium; faeces for the other species.
The reaction to infection will depend on where the patient comes from. People from
endemic areas are more likely to develop chronic infection associated with high burden of
infection. People from non-endemic areas generally have acute infection.
Clinical syndromes
Most people infected with Schistosomiasis are asymptomatic. Symptoms are more common
in travellers from non-endemic areas, particularly the acute symptoms. The clinical
syndromes of bilharzia can be divided into 3 stages:
1. 1st stage: Swimmer’s itch. The first stage coincides with the invasion of the human
host by fresh water cercariae. It causes a Swimmer’s itch, which is a localised
dermatitis that can lead to pruritic popular or urticarial rash. It usually occurs within
1 day of exposure and can last up to a week. The reaction is mediated by a
hypersensitivity immune reaction and prior exposure results in a more rapid
response and more severe symptoms. This is more with S. japonicum.
2. 2nd stage: Katayama fever. The second stage coincides with the first 2 weeks of egg-
laying. Katayama fever is a systemic hypersensitivity reaction to the migrating
parasites. This occurs within 2-8 weeks of exposure. It is more common with S.
mansoni & S. japonicum infections. These are also more common with patients from
non-endemic areas. The symptoms of Katayama fever are a sudden onset of fever &
chills, myalgia, arthralgias, dry cough, diarrhoea and headache. In children,
symptoms of encephalitis & cardiac involvement may be present. It often mimics
serum sickness. Lymphadenopathy & hepatosplenomegaly are prominent features
on examination. There is also leukocytosis with eosinophilia. Symptoms usually
resolve over a few weeks, but neck stiffness & coma can occur; it may even be fatal.
The diagnosis relies upon clinical findings & identifying potential exposure. Proper
diagnostic tests, such as egg detection & antibody tests are usually negative, and this
complicates the situation.
3. 3rd stage: Chronic infection. The third stage coincides with the host’s reaction to
eggs deposited in various tissues. The host responds to eggs laid by an intense
inflammatory reaction, with granuloma formation & fibrosis. Chronic infection is
more common in individuals coming from endemic areas in whom the acute stages
of disease are usually not seen. The severity of disease depends on the number of
eggs deposited in the tissues. The precise reaction depends on the tissue affected as
well as the species involved. The clinical syndromes are:
a. Urinary schistosomiasis. Urinary schistosomiasis is caused by S. haematobium
infections. The adult worms are usually located in the vesical plexus. The
worms lay eggs which go and lodge in the distal ureters and bladder, inducing
When symptoms are present, enquire about where the patient comes from and exposure to
fresh water (in patients from endemic areas). Also ask about recent travel history in patients
from non-endemic areas, particularly when they present with acute presentations.
2
Heart failure in bilharzia can result from rare myocardial granulomatous involvement.
• FBC. A routine FBC may reveal eosinophilia in infected patients (>0.4 × 109 cells/L).
The degree of eosinophilia correlates with stage, duration and intensity of infection.
1 2
- s of infected patients have peripheral eosinophilia, and this is frequently marked
3 3
early during infection. Eosinophilia is also a prominent feature in Katayama fever.
Features of iron deficiency anaemia may be present in patients who have had
chronic blood loss. Thrombocytopaenia may be a feature in patients with
hepatosplenic schistosomiasis, and this is a result of splenic sequestration.
Treatment
All patients with evidence of infection should be treated, since adult worms can live for
years and cause complications. The aim of treatment in endemic areas is to decrease the
worm load and therefore minimise the chronic effects of egg deposition. It may not always
be possible to completely eradicate adult worms, and re-infection is common. Nonetheless,
a 90% reduction in egg output has been achieved in mass treatment programmes, and in
light infections with no risk of re-exposure the drugs are usually curative. Treatment of
schistosomiasis leads to reversal of chronic effects. Reversal of periportal fibrosis & portal
vein thickening has been documented. However, late-stage fibrosis & associated
complications (oesophageal varices & cor pulmonale) are irreversible.
3
Treatment therefore partly relies on the strength of the host immune system.
Patients with oesophageal varices benefit from propranolol and from sclerotherapy or shunt
procedures.
Neisseria gonorrhoeae
N. gonorrhoeae belongs to the Neisseria genus, which is an aerobic Gram-negative coccus
typically arranged in pairs (diplococcic). The outer surface of N. gonorrhoeae is not covered
with a polysaccharide capsule, but has a negative charge. The organism has fastidious
growth requirements (grows at 35-37°C in carbon dioxide-enriched air), and therefore
cannot be cultured in just any culture medium. The organism is an intracellular organism.
N. gonorrhoeae has multiple antigens on its outer surface. It has pilli, which extend from the
cytoplasmic membrane through the outer membrane. These pilli mediate a number of
functions, such as attachment to the host cells, transfer of genetic material and motility.
These pilli contain pilli proteins (called pilins). There are also porin proteins, which are
integral outer membrane proteins that form pores & channels for transport of nutrients into
cells and waste products out of cells. One important porin protein (PorB) is important as it
interferes with neutrophil degranulation and thus protects the bacterium from destruction
within the phagosome. PorB also facilitates epithelial invasion, as well as resistance to
complement-mediated serum killing. Iron is also important for the survival of Neisseria
organisms. They can compete & bind host cell transferrin. There are also Opa proteins
(opacity proteins), which are proteins that facilitate binding to epithelial cells as well as cell-
to-cell signalling. They are responsible for producing opaque colonies when grown in
culture1. A major antigen in the cell wall is the LOS (lipo-oligosaccharide) protein, which
possesses endotoxin activity (through a lipid A moiety).
Pathogenesis
N. gonorrhoeae is primarily spread by sexual contact (oral or genital; also includes
homosexuals). It can also be spread vertically from mother to child during vaginal delivery.
The risk of contracting infection is higher in women who have sex with infected men (50%)
1
Opaque colonies are predominantly found in local disease, while translucent colonies are found in PID &
disseminated disease.
N. gonorrhoeae can infect any columnar epithelium, such as the urethra, cervix, rectum,
pharynx and conjunctiva. The organisms attach & penetrate mucosal cells through the use
of their pilli. Following attachment, the organism is phagocytosed by a process called
parasite-directed endocytosis. Here, they multiply and then pass into the sub-epithelial
space where infection is established. In this sub-epithelial space, the gonococci are
phagocytised and internalised in phagosomes. Gonococcal LOS stimulates release of TNF-α
which is responsible for most of the symptoms. Antibodies are formed against pilin, Opa and
LOS proteins.
Normal human beings are able to evade infection through activation of complement and
deposition of terminal complement complexes on the cell surface. However, some strains
are serum-resistant and dissemination is therefore possible.
Risk factors
The risk factors of gonorrhoea infection are those that increase acquisition (through sexual
contact) as well as those that increase progression. They include:
Clinical manifestations
Up to 50% of women & 10% of men are asymptomatic. The clinical manifestations depend
on the location of the infection:
• Local infection. In the majority of cases, patients present with the clinical disease
gonorrhoea, which is a genital disease.
o Gonorrhoea in men. In men, the infection is commonly restricted to the
urethra. The most common syndrome is that of anterior urethritis causing
dysuria & discharge. This comes after a 2-5 day incubation period.
Complications include epididymitis, prostatitis and peri-urethral abscess.
Diagnosis of gonorrhoea
2
PID is associated with development of peri-hepatic adhesions, called Fitz-Hugh-Curtis syndrome.
• Microscopy. You can view your specimen under the microscope using a Gram-stain.
You will see Gram-negative diplococci. Microscopy is very sensitive (90%) and
specific (96%) for identification of organisms on clinical specimens from symptomatic
men. From asymptomatic men, however, the sensitivity is ≤60%. It is also relatively
insensitive to detection of gonococcal cervicitis, both symptomatic & asymptomatic
forms. Microscopy is also useful for examining for gonococcal infection in joint
aspirates, but not from skin lesions, anorectal infections or pharyngitis. Microscopy
can be used to examine CSF.
• Culture. Culture on selective media has a sensitivity of 95%. N. gonorrhoeae grows
on chocolate agar in an atmosphere enriched with 5% carbon dioxide. An example of
a modified medium is the Thayer-Martin medium. For collection of the specimen,
the endocervix must be properly exposed, although you can also use rectal
specimens.
• Nucleic acid amplification tests. These have been developed for direct detection of
N. gonorrhoea organisms in clinical specimens. They are sensitive & specific, and
they are rapid. They also have the advantage of being able to be combined with
Chlamydia nucleic acid amplification tests.
• Antigen detection. Antigen testing is less sensitive than nucleic acid amplification
tests & culture.
• Blood culture. In patients with disseminated disease, blood cultures are positive
during the first week of disease. In addition, special handling is required, as some
supplements present in the blood culture medium are toxic to Neisseria.
Treatment
Treatment of gonorrhoea is divided into pharmacologic & no-pharmacologic:
Pharmacologic treatment
• Patients who have a positive test result for gonorrhoea on microscopy, culture or
NAAT.
• People who recently had sexual contact with an infected person.
For uncomplicated cases, you can treat using single dose ceftriaxone 500mg IM. You can
also give single-dose amoxicillin 3g together with probenecid 1g, ciprofloxacin 500mg or
ofloxacin 400mg. Longer courses of antibiotics are indicated for complicated infections
After treatment is complete, a follow-up assessment & culture should be done after 72
hours.
Non-pharmacologic treatment
You need to trace contacts of the patient so that the sexual contacts may be treated for
infection as well. You also need to encourage the patient to engage in safe sexual practices:
promote sexual negotiation skills and purge abusive relationships.
Neonatal gonorrhoea
Infants with ophthalmia neonatorum should be hospitalised and observed for response to
therapy and for disseminated disease. Treatment includes a single dose of ceftriaxone 25-
50mg/kg (not to exceed 125mg) IV/IM. Treatment is also indicated for infants of
symptomatic mothers. Topical antibiotic therapy alone is inadequate, and unnecessary
when systemic antibiotics are being given. The eyes should be irrigated frequently with
saline until the discharge clears. Septic arthritis is treated with ceftriaxone 25-50mg/kg for 7
days, and the duration should be extended to 10-14 days if meningitis is present.
The most effective way of preventing new-born infections is to diagnose & treat these
infections in pregnant women. Prophylactic eye antibiotic therapy can be given to at-risk
babies: you can use 1% silver nitrate, 1% tetracycline or 0.5% erythromycin eye ointment.
3
There may be need for joint drainage.
Treponema pallidum
T. pallidum belongs to the bacterial order of Spirochaetales, which is a group of thin, helical
Gram-negative bacteria. The organism has pointed straight ends, and has 3 periplasmic
flagella. T. pallidum cannot be cultured extracellularly, because it is an intracellular
organism. T. pallidum lacks the TCA cycle, and therefore relies on the host cell for synthesis
of all purines, pyrimidines and most amino acids. The bacteria are micro-
aerophilic/anaerobic and are therefore extremely sensitive to oxygen toxicity.
The bacterium has lipoproteins anchored to the inner surface of the cell membrane and
none of them are exposed to the outer membrane. It therefore lacks any system-specific
antigens and therefore successfully evades the immune system. The bacteria are able to
resist phagocytosis by coating themselves with host fibronectin, allowing interaction with
host cells. Most investigators believe that the tissue destruction & lesions found in syphilis
are due to the host immune response. The bacterium contains hyaluronidase which
facilitates perivascular infiltration.
Human beings are the only natural host. T. pallidum in its acquired form is spread through
close sexual contact with an unprotected person. It can also be acquired through vertical
transmission from mother to child (congenital form). The organism enters the host through
breaches in the squamous or columnar epithelium. Primary infection of non-genital sites is
rare. The bacterium is not highly contagious: the risk of acquiring infection after a single
sexual contact is estimated to be 30%. However, this only occurs during the early stages of
disease.
Clinical course
Acquired syphilis follows a course of 3 stages:
• Primary syphilis. This is the earliest stage of the disease. The bacterium has a latency
period of 10-90 days (21 days on average), after which a papule develops on the site
of inoculation. This papule ulcerates, leaving a hard/firm painless ulcer with elevated
borders. Histologically, the lesion reveals an endarteritis & periarteritis, and the ulcer
1
This stage is benign in terms of response to treatment rather than clinical manifestations.
2
Tabes dorsalis is due to demyelination of dorsal roots leading to a complex de-afferentiation syndrome.
Diagnosis of syphilis
The diagnosis of syphilis is made as follows:
3
This is fatal within 3 years if left untreated.
4
Yaws is caused by T. pallidum pertenue, bejel is caused by T. pallidum endemicum and pinta is caused by T.
carateum. Syphilis is caused T. pallidum pallidum.
Neurosyphilis is diagnosed by the presence of high antibody titres & neurologic signs. The
antibody test is troublesome. VDRL is highly specific for detection of T. pallidum in CSF
specimens, but is not sensitive. Therefore, a positive test result is diagnostic but a negative
test result does not rule out the disease. In contrast, FTA-Abs has high sensitivity but low
specificity (because treponemal antibodies passively cross from blood to CSF). A positive
FTA-Abs result is not particularly diagnostic, but a negative result rules out infection. T.
pallidum is not amenable to in vitro culture.
Treatment
Syphilis is treated medically using antibiotics. You use long-acting benzathine penicillin (X-
pen or penicillin G) 600mg im od, and you can use doxycycline 100mg b.d. or erythromycin
500mg q.i.d. (or azithromycin) as alternatives. The duration of treatment depends on the
stage of disease.
• Early stage disease (primary & secondary syphilis) should be treated for 10 days.
• Late stage disease should be treated for 2 weeks. Established neurologic disease is
arrested but not reversed by penicillin. If the patient is HIV positive, neurosyphilis
may not be reversed by penicillin alone.
5
It can be, however, found in other diseases caused by spirochaetes such as Lyme disease, leptospirosis and
relapsing fever.
Chlamydia trachomatis
Genital chlamydia is caused by Chlamydia trachomatis. There are 2 other organisms in the
species that cause human disease: C. pneumoniae, which is responsible for respiratory
infections, and C. psittaci, which mainly causes pneumonia.
Chlamydia species are small Gram-negative bacilli which are obligate intracellular organism.
The organisms lack the rigid peptidoglycan found in the cell walls of many other bacteria,
but they have a central dense core surrounded by a cytoplasmic membrane and a double-
layered outer membrane. The cell wall contains a lipopolysaccharide which has weak
endotoxin activity. Chlamydia organisms have a unique developmental cycle, alternating
between metabolically-inactive but infectious forms (called elementary bodies, EB) and
metabolically-active non-infectious forms (called reticulate bodies, RB). The EBs bind to
host cells & enter them, where they transform into RBs within 6-8 hours. They utilise host
ATP for their energy requirements & replication. After replication, the RBs then reorganise
into small EBs which are released after the cell ruptures.
In the outer membrane, there is a protein called the major outer membrane protein
(MOMP), which is a major structural component that is unique for each species. Variable
regions in the gene encoding this protein are found in C. trachomatis and are responsible for
the 18 variants – called serovars – of the organism. C. trachomatis is divided into 2 biovars
(trachoma and lymphogranuloma venereum, LGV) into which all the serovars are grouped.
Serovars Disease
A, B, Ba, C Trachoma
D-K Urogenital disease
L1, L2, L2a, L2b, L3 Lymphogranuloma venereum
The range of cells that C. trachomatis can infect is limited to non-ciliated columnar, cuboidal
and transitional epithelium. This is found on the mucous membranes of the urethra,
endocervix, endometrium, fallopian tubes, anorectum, respiratory tract and conjunctivae.
The LGV serovars are more invasive as they replicate in mononuclear phagocytes.
C. trachomatis gains access to the body through minute abrasions or lacerations in the
mucosal barrier involved. This is mainly through:
Studies of the natural history of chlamydia are scarce. The incubation period of the disease
is 7-14 days. From a study done, spontaneous resolution of infection was seen in only 18%.
The majority of patients persist as asymptomatic carriers.
Clinical syndromes
The clinical syndromes associated with chlamydia are:
1
Chlamydia accounts for 19-31% of cases of non-gonococcal urethritis (NGU) – it is the commonest cause.
2
Frozen pelvis is a condition in which the pelvic organs are distorted & tethered to each other due to
adhesions.
Diagnosis of chlamydia
The diagnosis of chlamydia is made on the basis of a characteristic clinical picture, positive
screening test or when another indication suggests presence of chlamydia (e.g. PID). It can
be diagnosed: based on cytologic, serologic or culture findings; direct detection of the
antigen in clinical specimens; and use of nucleic acid-based tests. Specimens must be taken
from involved sites rather than just pus or a vaginal exudate3; high vaginal or endocervical
swabs should be taken in women. In men, you take first-catch urine4. In those that engage in
receptive anal intercourse, you do a rectal swab. You can also do a conjunctival swab.
• Nucleic acid-based tests. You can use a nucleic acid probe test or a nucleic acid
amplification test (NAAT). This is now the diagnostic test of choice for Chlamydia
infection. Care must be taken to monitor for the presence of inhibitors to the
amplification reaction (e.g. urine). The drawbacks of using NAAT are: same-day
results cannot be obtained; and it is too expensive to use in resource-limited
settings.
• Culture. This remains the most specific method of detecting chlamydia, but is still
insensitive when compared to NAAT (70-85% when a single endocervical smear is
taken). The culture medium includes in-vitro cell lines that normal Chlamydia
organisms infect. The culture is then stained with iodine- or specific fluorescin-
labelled antibodies targeted against inclusion bodies. The sensitivity of culture is
affected if inadequate specimen is taken or if viability is lost during transport.
Culture is the gold standard of isolation of chlamydia in neonatal infection.
• Serologic tests. This is of limited value in urogenital chlamydia as it cannot
differentiate between current & past infection5. Demonstration of a significant
increase in antibody levels can be used, albeit the increase not being demonstrable
for a month or longer. Antibody tests can be used for LGV, however, and this can be
through complement fixation (CF), micro-immunofluorescence (MIF) and enzyme
immunoassay (EIA). CF is directed against species-specific LPS (lipopolysaccharide)
and therefore a 4-fold increase/single titre ≥ 1:256 is highly suggestive of LGV.
• Antigen detection. You can use direct immunofluorescence staining, or you can use
ELISA. The tests target either chlamydial MOMP or LPS. However, because antigenic
3
Pus & exudates usually contain a small number of living cells in which C. trachomatis organisms can be found.
4
The patient must not have voided within the last 2 hours.
5
IgM antibodies may not be detected in adolescents & young adults with active infection, and this limits their
usefulness. They can be measured, however, in infants with chlamydia pneumonitis.
Treatment
The goals of treatment of chlamydia are:
Treatment of chlamydia
The bacterial EBs are generally resistant to antibiotics, while the active MBs are not.
Therefore, the antibiotic chosen should have good intracellular penetration and a relatively
long half-life. In general, chlamydia is sensitive to tetracyclines & macrolides – doxycycline &
azithromycin are used. You can also use long-acting fluoroquinolones (ofloxacin &
levofloxacin) but they require a full week’s therapy and they are relatively more expensive.
6
Administration of erythromycin to neonates predisposes to development of infantile hypertrophic pyloric
stenosis.
Counselling
Pathogenesis
Sepsis is thought to arise from triggering of the innate immune response & inflammatory
cascade due to recognition of pathogen-associated molecular patterns (PAMPs) on
pathogens. These molecular patterns are seen on pathogens, and for septicaemia to occur,
there needs to be systemic dissemination of either an endotoxin or an exotoxin. An
endotoxin is a lipopolysaccharide derived from the cell wall of a Gram-negative bacterium.
Gram-positive organisms also have molecules similar to lipopolysaccharide on their cell
walls, but they act in a different way to activate inflammation. An exotoxin is an antigenic
protein produced & secreted by bacteria. Exotoxins are predominantly produced by Gram-
positive organisms such as Staphylococcus & Streptococcus, but Pseudomonas organisms
also produce exotoxins. Accordingly, gram-positive organisms generally cause sepsis by
release of exotoxins, creating toxaemia. Gram-negative organisms, however, usually
produce sepsis by causing bacteraemia. At times, there is no haematogenous spread of the
infectious agent. In this case, the immune response to the localised infection spreads
beyond the confines of the infection and causes a systemic inflammatory response.
Sepsis & other complications are the result of release of inflammatory mediators such as IL-
6, TNF-α and other substances from inflammatory cells. The processes involved include:
Stages of sepsis
The spectrum of conditions that sepsis goes through is as follows:
1. Sepsis.
2. Severe sepsis.
3. Septic shock.
4. Multi-organ dysfunction syndrome.
Sepsis
Sepsis is defined by the presence of SIRS and a concurrent infection. SIRS (systemic
inflammatory response syndrome) is a condition of disseminated inflammation that is
characterised by any 2 of:
Severe sepsis
Severe sepsis is defined by sepsis (infection + SIRS) in the presence of organ hypoperfusion,
dysfunction or hypotension. The features suggestive of severe sepsis include:
1
The paO2/FiO2 is also called the Horowitz index. In normal individuals, this index yields a value of 350-450mm
Hg.
2
This is a tissue perfusion variable rather than a sign of organ dysfunction.
Septic shock
This is sepsis with arterial hypotension despite adequate fluid resuscitation, or the
requirement of inotropes/vasopressors to maintain blood pressure. This is often the result
of an inflammation-related fall in peripheral vascular resistance, but it may also be related in
part to cardiogenic dysfunction. It is a form of distributive shock. The hypoxia induced by the
shock may be further worsened by reduced mitochondrial respiration at tissue level.
Shock can also be classified as refractory shock, in which the shock does not respond to
conventional therapy – IV fluids or inotropes – within an hour.
1. Shock. Periods of relative/total ischemia are common to all inciting events of MODS.
Ischemia can be due to regional or global perfusion deficits. The path of organ
dysfunction to organ failure depends on the severity of perfusion deficits, the
passage of time to adequate resuscitation, and the functional reserve capacity of the
organ involved.
2. Period of active resuscitation. If resuscitation is rapid & effective, the sequence of
events precipitating MODS may not occur. However, in many cases despite adequate
management, the syndrome progresses4.
3. Systemic inflammatory response. If resuscitation fails, there is widespread cellular
damage with pan-endothelial dysfunction. This is seen with increased microvascular
permeability with exudative oedema. This period is a period of hyper-metabolism
and widespread activation of the immune response, thus causing SIRS. When
associated with infection, this is called sepsis. Mortality at this stage rises to 25-40%.
4. Organ failure. During this stage, there’s increasing organ dysfunction, failure and
death. The appearance of organ dysfunction causes the mortality chances to leap to
3
Sepsis is therefore just one of the causes.
4
This suggests a genetic component.
• Respiratory system (lungs). In the majority of patients, the lungs are the first organs
to fail. The lungs are pivotal in the development of MODS, as they appear to
generate inflammatory mediators and/or allow inflammatory mediators to persist in
the circulation (through decreased capacity to clear them). This leads to persistent
endothelial dysfunction. Lung dysfunction occurs on a spectrum, from minor
pathology (designated acute lung injury. ALI) to massive alterations in the lung
(adult respiratory distress syndrome, ARDS). ARDS is defined by:
o Respiratory distress.
o Stiff lungs.
o Hypoxia despite high levels of supplemental oxygen (paO2/FiO2
<40KpA/300mm Hg for ALI and paO2/FiO2 <26.6kPa/200mm Hg for ARDS).
o Bilateral infiltrates on chest X-ray.
o Pulmonary capillary wedge pressure of less than 18mm Hg (or no clinical
evidence of increased left atrial pressure)5.
In ARSD, there is widespread endothelial damage to pulmonary capillaries. These
become leaky and cause interstitial oedema & fibrosis.
• Gastrointestinal system. The gut is particularly vulnerable to the processes occurring
in MODS. It is believed that sepsis in the absence of a focus of infection is due to
colonisation of the normally sterile upper GIT, with translocation of bacteria & toxins
from the gut into the circulation. The gut wall is also involved in the systemic disease
process, particularly due to redistribution of blood from the splanchnic circulation to
muscles. This predisposes the gut to ischemia & membrane reperfusion injury. This
allows translocation of bacteria & toxins into the portal circulation. For this reason,
the gut has been termed the motor of multiple organ failure. GIT complications
include GI failure (inability to tolerate enteral feeding; paralytic ileus), ischaemic
colitis, acalculous cholecystitis, pancreatitis and GIT haemorrhage.
• Liver. Normally, the hepatic reticuloendothelial system is supposed to be able to
clear overspill of gut luminal toxic products into the portal circulation. In MODS,
however, there is reduced clearance and therefore the spillage of toxins is washed
into the pulmonary microcirculation. This leads to activation of pulmonary
macrophages, leading to increased destruction of pulmonary architecture.
• Cardiovascular system. Normally, when the body is subjected to an insult or stress,
the cells of the body adjust by extracting more oxygen from blood. However, in SIRS
leading up to MODS, the tissues are unable to extract more oxygen from blood and
5
This means the pulmonary oedema is non-cardiac in origin.
Presentation
The presentation of a patient with sepsis is as follows:
• History. The patient will present with fever, chills, dyspnoea, cool extremities,
fatigue, malaise and anxiety. The patient may also be confused.
• Examination. The vitals will be abnormal: fever, tachypnoea, tachycardia and
hypotension. The patient may have local signs of infection.
• Investigations. These are mainly blood investigations.
o FBC will show anaemia and/or leucocytosis. Thrombocytopaenia can occur in
DIC.
o U&Es to check for renal dysfunction and to use as a baseline to assess for
development of AKI.
o Liver enzymes to rule out liver injury.
Management of sepsis
Sepsis is a medical emergency and should be treated in an ICU/HDU. Management of sepsis
is divided into treatment of infection and supportive therapy.
1. Oxygen. Supportive oxygen should be given, and this may require intubation &
ventilation. Oxygen must be increased to a point where oxygen consumption no
longer rises or to levels where serum markers of anaerobic respiration (e.g. lactate)
start falling. Ventilation is instituted using a lung-protective regimen – low tidal
volume (6mL/kg) & limited plateau pressure (<30cm H2O). this reduces mortality.
2. Cardiovascular support. You need to monitor, restore and maintain haemodynamic
function. Insert a central venous & arterial catheter. Maintain central venous
pressure at 8-12mm Hg with crystalloids or colloids. Vasoactive agents should be
used to maintain mean arterial pressure at 65-90mm Hg. You can use inotropes,
such as norepinephrine.
3. Treat the infection. The infection is treated by identifying the focus of infection,
testing for the appropriate microorganism & its sensitivity and instituting empirical
antibiotic therapy until sensitivity results return. Start with a broad-spectrum
antibiotic, with additional drugs depending on patient risk factors (e.g.
immunosuppression), suspected aetiology and local microbial susceptibilities.
4. Corticosteroids. Start IV hydrocortisone in septic shock patients who are
unresponsive to fluids & vasopressors.
5. Controlling hyperglycaemia. Hyperglycaemia is controlled by giving insulin. This
reduces infectious complications.
6. Nutrition. Early enteral feeding is indicated to preserve the function of the intestinal
mucosal barrier. There is a significant increase in amino acid oxidation. There is need
to supply metabolic support by providing adequate calories to maintain nitrogen
balance. Excessive glucose may cause fatty liver, hyperosmolality, hyperglycaemia
and increased CO2 production (which increases the excretory load to the liver and
may exacerbate the lung injury). The requirements are as follows:
Acute kidney injury is a medical emergency. It affects 18% of all hospital admissions and is
an independent risk factor for mortality.
Classification of AKI
AKI is classified using the RIFLE criteria, in an attempt to distinguish between AKI and CKD
(chronic kidney disease). The Acute Kidney Injury Network (AKIN) proposed a modification
to the original RIFLE criteria proposed by the Acute Dialysis Quality Initiative group. The
AKIN modification has been adopted by KDIGO.
Causes of AKI
The causes of AKI are classified into pre-renal, intrinsic and post-renal.
Pre-renal causes account for 40-70% of causes and are therefore the most common. AKI in
these cases results from hypoperfusion of the kidneys. This is caused by:
• Hypovolaemia/hypovolaemic shock.
• Hypotension from other causes, particularly sepsis.
• Cardiogenic shock & cardiorenal syndrome.
• Vascular disease, e.g. renal artery stenosis. This is made worse by concurrent
administration of ACE-inhibitors and NSAIDs1.
A drop in renal perfusion leads to pre-renal uraemia and parenchymal kidney damage.
These are better established by evidence of blood/fluid loss, sepsis or cardiac disease. It
corrected by re-establishing perfusion to the kidney. On examination, you may find postural
hypotension, a low jugular venous pressure and a low-volume tachycardia.
Intrinsic causes
Intrinsic causes result from intra-renal pathology. They account for 10-50% of cases. The
causes of intrinsic renal disease are classified into:
• Glomerular. Glomerular diseases are mainly classified into primary and secondary
glomerulopathies. The primary glomerulopathies are divided into nephrotic
syndrome and nephritic syndrome. Nephrotic syndrome results from increased
leakiness of the glomerular basement membrane, leading to increased protein loss.
The clinical syndromes include: minimal change disease, focal sclerosing
glomerulosclerosis (FSGS), membranous glomerulonephritis and membrano-
proliferative glomerulonephritis. Nephritic syndrome is a result of damage to the
glomerular endothelium & mesangium, resulting in haematuria, increased leakiness,
reduced filtration function, uraemia and hypertension. The clinical syndromes
include post-streptococcal acute glomerulonephritis and IgA nephropathy.
Secondary causes of glomerular disease include autoimmune diseases (such as SLE),
drugs, and infections.
• Tubular. Acute tubular necrosis (ATN) is the leading cause of intrinsic AKI. It can
result from renal ischemia, or from direct toxic effects of drugs and toxins (e.g.
contrast, haemoglobin and myoglobin). ATN is thought to develop through:
o Intra-renal microvascular vasoconstriction. This occurs as a result of
vasoconstrictive agents such as endothelin, adenosine, thromboxane A2 and
leukotrienes. Sympathetic nerves also induce vasoconstriction. There is also
reduced response to nitric oxide, prostaglandin E2, acetylcholine and
bradykinin.
1
ACE inhibitors & NSAIDs impair renal autoregulation, meaning that the kidneys cannot maintain GFR when
there is reduced renal blood flow due to renal artery stenosis.
Postrenal causes
Postrenal causes are a result of obstruction of the urinary tract. Obstruction can occur at
any point along the urinary tract, from the calyces to the urethral meatus. Obstruction
accounts for 10-25% of cases of AKI. The causes of obstruction are classified into:
• Luminal. These are causes that enter the lumen and occlude it form here. Examples
include renal stones, blood clots and sloughed papillae.
• Mural. These are things that develop within the walls of the urinary tract and cause
the wall to collapse and obliterate the lumen. They include strictures, BPH, and
primary malignancies (of the ureter, bladder and prostate).
• Extramural. These are things that do not form the urinary tract that grow and cause
collapse of the lumen through pressure effects. They include other pelvic
malignancies (especially cervical) and retroperitoneal fibrosis.
History
• Risk factors, e.g. comorbid conditions (e.g CKD), previous renal disease, recent fluid
intake & losses (most important), new drugs and chemotherapy.
• Systemic symptoms, such as rash, joint pain and fever.
• Other system-specific symptoms, such as cough & haemoptysis, GI symptoms or
urinary symptoms.
Examination
You should do a full system examination. Look for signs of dehydration, palpable kidneys
and/or bladder, renal angle tenderness, abdominal & pelvic masses, renal bruits and rashes.
Investigations
One of the aims of investigation is to distinguish between pre-renal and intrinsic renal AKI.
This is done by using:
• Blood tests. You can measure FBC (normocytic normochromic anaemia suggests
CKD), U&Es (hyponatraemia may be present in fluid overload; hyperkalaemia
common in oliguric phase of ATN while hypokalaemia common in polyuric phase),
arterial blood gases (acidosis common unless there is concurrent vomiting), clotting
profile, and inflammatory markers (CRP & ESR).
• Urinalysis. Presence of red cells & red cell casts is suggestive of nephritic syndrome.
Massive proteinuria (>3.5g in 24 hours) is suggestive of nephrotic syndrome. The
presence of glucose, phosphates, bicarbonate and hyaline casts is suggestive of
acute tubular necrosis. The presence of white cell casts is suggestive of acute
interstitial nephritis.
• Imaging. A renal ultrasound is indicated in suspected obstruction, and when stones
are suspected, it is necessary to do a CT-IVU. In elderly men, suspect prostatic
enlargement and therefore do a transrectal ultrasound. Chronic kidney disease
should be suspected if the kidneys are small.
Management of AKI
The management of AKI is as follows:
1. Assess volume status. Look for signs of either fluid overload (hypertension, raised
JVP, lung crepitations, peripheral oedema and gallop rhythm) or hypovolaemia
(reduced urine output, reduced JVP, hypotension, tachycardia and reduced tissue
turgor).
2. Aim for euvolaemia. If there is a difficult balance between overload &
hypovolaemia, consider titrating fluid input hourly by matching the previous hour’s
fluid output + 25ml/h (insensible losses). This usually requires intensive care nursing
and is difficult to do outside the HDU setting. If euvolaemic, review balance daily –
match the input to the output + 500ml (for insensible losses). Avoid potassium-
containing fluids, unless the patient is hypokalaemic.
3. Stop nephrotoxic drugs. These include NSAIDs2, ACE inhibitors2, aminoglycosides and
amphotericin B. If the patient is on metformin, stop metformin if serum creatinine
goes above 150µM. Adjust the dose of renally-excreted drugs.
4. Nutritional adjustment. Dietary sodium & potassium restrictions should be made.
Aim for normal caloric intake, and increase in catabolic states such as sepsis & burns.
Dietary protein restriction is controversial. In order to avoid haemodialysis, dietary
protein restriction is placed at 0.5mg/kg/day or 40mg/day. This poses the risk of
negative nitrogen balance, despite efforts to avoid nitrogen metabolism by a high
calorie diet. Catabolic patients require more protein. Enteral food is preferred to
parenteral food. Vitamin supplements are usually supplied.
2
This is if the patient has renal artery stenosis.
Aetiology
20% of cases of CKD are idiopathic and they present late with small shrunken kidneys and an
uninformative biopsy. Otherwise, the causes of CKD can be classified into:
• Congenital & inherited diseases. These include both structural diseases and
metabolic disorders. Structural disorders include polycystic kidney diseases,
medullary cystic disease, tuberous sclerosis and congenital obstructive nephropathy.
The metabolic disorders include oxalosis and cystinosis.
• Glomerulonephritis. These can be classified into primary or secondary depending on
whether they are result of systemic disease or not. The primary causes include focal
segmental glomerulosclerosis (FSGS) and IgA nephropathy. Secondary glomerular
diseases include SLE, amyloidosis, Wegener’s granulomatosis, haemolytic uraemic
syndrome and systemic sclerosis. CKD can also be caused by malaria.
• Vascular diseases. Vascular diseases such as renovascular diseases and small- &
medium-vessel vasculitides cause chronic kidney diseases. Atherosclerosis can also
cause chronic kidney disease.
• Tubulointerstitial diseases. These can be due to drugs or immunological process.
Patients can also have reflux nephropathy, tuberculosis, nephrocalcinosis, multiple
myeloma and renal papillary necrosis
• Urinary tract obstruction. This can be due to calculus disease, prostatic disease,
pelvic tumours, retroperitoneal fibrosis and schistosomiasis.
• Other chronic disease. Diabetes is the leading cause of CKD in the developed world –
it causes diabetic glomerulosclerosis. Other causes include hypertension
(hypertensive nephrosclerosis) and malignancies.
Pathophysiology
Each kidney has about 1 million nephrons in it, and the kidney has a very large functional
reserve. This means that it can maintain normal renal function even when it loses 50% of its
Furthermore, developing AKI (acute kidney injury) is a risk factor for developing chronic
kidney disease in the future.
Most cases of CKD are acquired rather than genetic. However, some genetic syndromes are
associated with CKD. These include polycystic kidney disease, Alport syndrome and atypical
HUS (haemolytic uremic syndrome).
Classification of CKD
CKD is broadly divided into 5 stages that are described by the GFR and the pathological
process in the kidney. This is the KDIGO (Kidney Disease: Improving Global Outcomes)
classification.
1. proteinuria (>30g/24hr).
The suffix P can be added to the stage of renal disease if there is significant proteinuria
defined as an albumin-creatinine ratio of more than 65mg/mmol or protein-creatinine ratio
of more than 100mg/mmol.
Complications of CKD
The complications of CKD are:
1
Pruritus is also a result of hyperphosphataemia & hyperparathyroidism.
2
Therefore, patients who are persistently acidotic are more likely to have osteomalacia and low-turnover
disease.
• History. Ask about any history of hypertension, diabetes, ischemic heart disease or
other systemic disease such as SLE. Also ask about previous UTIs and lower urinary
tract symptoms. Check for a drug history (particularly nephrotoxic drugs) and family
history. Also screen for malignancies in the patient.
• Symptoms. The symptoms are:
o Anorexia.
o Malaise.
o Polyuria & nocturia, in earlier stages and in tubulointerstitial disease.
o Itching.
o Nausea, vomiting & diarrhoea.
o Paraesthesia (due to poly-neuropathy. This may also be due to
hypocalcaemia).
o Restless leg syndrome.
o Bone pain (due to metabolic bone disease).
o Tetany, due to hypocalcaemia.
o Anasarca (generalised body oedema, including pulmonary oedema).
o Anaemic symptoms.
o Amenorrhoea in women; erectile dysfunction in men.
• Examination. The features on examination are either suggestive of uraemia, fluid
overload, aetiology or complications.
o Short stature due to stunting (if CKD was in childhood).
o Elevated blood pressures.
o Pallor. This results from anaemia.
o Increased photosensitive pigmentation.
o Scratch marks (uraemic pruritus).
Severe depression of GFR can cause oliguria. Always ask about it.
Investigations
The laboratory investigations done for a patient suspected to have CKD include:
• Blood tests. Patients may present with a normochromic normocytic anaemia. They
also have decreased/increased calcium levels, raised phosphate levels, raised ALP
(due to renal osteodystrophy) and raised PTH in stage 3 or higher CKD.
• Urine tests. Urinalysis is done to look for haematuria and proteinuria, which may
indicate glomerular disease. Urine microscopy is used to look for white cells
(infections) and casts (tubular cells – active disease; red cells – glomerulonephritis).
You can also measure urine electrolytes (this is not very helpful) & urine osmolality,
and you can conduct urine electrophoresis.
• Serology. Serological tests can be done to screen for numerous diseases, such as
autoimmune diseases (SLE, scleroderma, Wegener’s granulomatosis and
Goodpasture’s syndrome) and infections (post-streptococcus, hepatitis B & C and
HIV).
• Imaging. Ultrasound scans can be done to check the size, anatomy &
corticomedullary differentiation of the kidney. The kidneys are usually small in CKD.
CT scans can be done to diagnose retroperitoneal fibrosis and some causes of urinary
tract obstruction. It may also demonstrate cortical scarring. MRI scans are indicated
in renovascular disease.
• Renal biopsy. This is indicated in patients with unexplained CKD and normal sized
kidneys. If rapidly progressive glomerulonephritis is suspected, you should do the
biopsy as soon as possible.
Management
Renal replacement therapy is mainly indicated for end-stage kidney disease, in which the
GFR falls below 15ml/min/1.73m2. This is a chronic indication. For acute indications, dialysis
is usually used for certain indications, and the indications for dialysis can be abbreviated
using AEIOU:
Haemodialysis
Haemodialysis involves pumping blood through an array of semi-permeable membranes
which bring blood in close contact with the dialysis fluid (called the dialysate) and allow
exchange of substances between the 2 fluids. It is the most effective way of achieving
biochemical improvement, and hence it is used even in acute emergencies.
Basic principles
1
High sodium causes thirst & hypertension.
The dialysate flows in a countercurrent fashion (similar to that found in the loop of Henle).
This countercurrent allows blood solutes to always be at a higher concentration than those
of the dialysate and therefore allow constant diffusion from blood to the dialysate
throughout the interaction between the 2 solutions.
Administering dialysis
2
High calcium causes hypercalcaemia, while low calcium (combined with poor compliance to medication with
oral calcium & vitamin D) will lead to hyperparathyroidism.
1. Pre-dialysis workup. Prior to dialysis a number of tests are taken, including HIV,
hepatitis B & C viruses, and pre-dialysis haemoglobin concentration. The body
weight must be measured before & after dialysis, and the dry weight is recorded.
The transmembrane pressure in subsequent dialysis sessions is adjusted to achieve
excess fluid removal equal to that by which they exceed their dry weight.
2. Anticoagulation. All patients have to be anticoagulated, usually using heparin.
3. Monitoring. This is done to detect and treat complications. The observations made
include: blood pressure, temperature, ECG, post-dialysis haemoglobin.
4. Frequency & duration. An adult of average size usually receives 4-5 hours of
treatment 3 times a week. Twice weekly dialysis is acceptable if the patient has
adequate residual renal function.
Complications
Adequacy of dialysis
The only true measure of the adequacy of haemodialysis is patient morbidity & mortality. In
most cases, dialysis is empirical as the size, number and nature of uraemic toxins in
unknown. Symptoms of under-dialysis include: fatigue, itching, insomnia, restless legs and
peripheral neuropathy.
Adequacy can be assessed by computerised calculation of urea kinetics. This requires the
measurement of: residual renal urea clearance, rate of rise of urea concentration between
3
This is a condition caused by over-rapid correction of uraemia. It is characterised by nausea, vomiting,
restless legs, headache, hypertension and myoclonic jerking. In severe cases, seizures & coma result.
4
This is due to reduced clearance of β2-microglobulin. This protein polymerises and forms amyloid deposits
which can compress the median nerve (carpal tunnel syndrome) or cause a dialysis arthropathy.
5
This predisposes to renal cell carcinoma.
• Urea reduction ratio (URR). A urea reduction ratio of 65% per dialysis sessions is
deemed adequate.
• Equilibrated urea clearance. This found by:
𝐾𝑡
𝑢𝑟𝑒𝑎 𝑐𝑙𝑒𝑎𝑟𝑎𝑛𝑐𝑒 =
𝑣
K = dialyser clearance of urea
t = duration of dialysis
v = urea distribution volume estimated as total body water
A Kt/V value of 1.0-1.2 is deemed adequate for a well-nourished patient who is
dialysed 3-times a week.
Haemofiltration
Haemofiltration is the removal of plasma water & dissolved constituents by convective flow
across a high-flux semi-permeable and replacing with a solution of desired biochemical
composition. Large & small solutes move across the barrier at almost the same rate. The
ultra-filtrate is replaced with an equivalent amount of fluid so there is less haemodynamic
instability. Lactate is used as a buffer in this case because rapid infusion of acetate causes
vasodilation, while bicarbonate may cause precipitation of calcium bicarbonate.
Haemofiltration is used for critically ill patients with haemodynamic instability. It can be
used for AKI and CKD. Its major drawback is the need to exchange large amounts to achieve
adequate small molecule removal. It needs 22L exchange 3-times a week for maintenance
therapy and 1L per hour for acute kidney injury. It is therefore unsuitable for long-term
therapy in the majority of patients. The costs are also high.
Peritoneal dialysis
In peritoneal dialysis, the peritoneum is used as the dialysis membrane. It avoids the need
for extra-corporeal circulation. Dialysis is achieved through diffusion (driven by
concentration gradient of solutes) and convection (driven by osmotic & hydrostatic
gradients of water). The filtration barrier in peritoneal dialysis includes the capillary walls &
interstitium6. The mesothelium of the peritoneum is of much less significant as a transport
hindrance.
Procedure
6
The capillary wall is a barrier for large solutes & fluid, while the interstitium is a barrier for transport of small
solutes.
Osmotic removal of excess water & solutes is achieved using a hypertonic dialysate.
Complications
No consensus on the optimal degree of removal of urea & other waste products has been
reached. However, there is a general agreement that the minimum dose delivered should
not be less than 1.7. Weekly Kt/v of 2 or more coupled with creatinine clearance of at least
60L per week is recommended.
Renal transplantation
This is the best form of renal replacement therapy, and it offers the potential for almost
complete rehabilitation in ESKD. It has a significant survival advantage as well. However, the
transplant involves major surgery, long-term immunosuppression and a number of potential
complications. The patient must also be physically & psychologically suitable for the
transplant.
The supply of donor organs is greatly exceeded by demand and donor organs must
therefore be used optimally. 80% of grafts survive 5-10 years in the best centres, while 50%
survive for 10-30 years. The half-life of renal allografts is still 13-16 years. The 3 commonest
7
This is a potentially fatal complication. If this complication appears, CAPD should be abandoned.
Procedure
The donor kidney is anastomosed onto the iliac vessels of the recipient, while the donor
ureter is implanted into the bladder. Unless the donor is genetically identical to the
recipient, immunosuppressive therapy is required for as long as the transplant remains in
place (to prevent rejection).
8
This is particularly useful in patients with diabetes.
9
Azathioprine
Complications
The absolute contraindications to renal transplantation are: active infection, cancer within
the last 5 years, and severe comorbidity. HIV is no longer an absolute contraindication.
Patient assessment
Rheumatologic patients are assessed as follows:
History
In the history of the patient, you need to find out the following information about the joint:
• Symptoms. The common presenting symptoms that could point towards joint
problems are:
o Pain. This is divided into: usage pain, in which pain worse on use and relieved
by rest; rest pain, which the pain is worsened by rest and improved by
movement; and night/bone pain, which is pain mostly felt at night and poorly
related to movement. Also ask abour diurnal variations.
o Stiffness. This is a subjective feeling of the inability to move freely. Ask about
severity in the morning and after inactivity.
o Swelling. This could be cause fluid, bone or soft tissue
o Loss of function. The joint could have reduced range of movement or be
deformed. This is usually better assessed during examination.
o Warmth and redness. These are signs of inflammation.
Ask about the pattern & progression of joint involvement and symmetry.
Ask about the duration of symptoms and if the patient has had any prior episodes.
• Extra-articular symptoms. These include: rashes, photosensitivity, Raynaud’s
phenomenon (pain, pallor and eventual gangrene of digits), dry eyes/mouth, red
eyes (iritis), diarrhoea, urethritis, nodules, mouth ulcers, genital ulcers,
constitutional symptoms (fever, night sweats and weight loss) and symptoms of
heart failure.
• History of trauma.
• Age & sex. These have an important bearing on disease epidemiology and
likelihoods. Also ask about ethnicity.
• Extent of disability. This can be graded according to WHO as follows:
Examination
You need to be observant as the patient walks in: look at their gait and look for disabilities.
There is a rapid examination of the joints done to look at all joints in general, and there’s a
detailed examination done for all joints.
Gait:
Ask the patient to walk a few steps forwards & backwards:
• Look for smoothness of walking.
• Assess the arm swing: whether it is adequate or not.
• Assess the stride length.
• Normal heel strike & toe off.
Rapid examination of the upper limbs:
• Raise arms sideways right up to the ear (abduct arms slowly) – [screens for shoulder
or rotator cuff problems].
• Hold arms out forwards & straight (elbows extended), fingers spread out, and then
rotate your palms to face up then down – [fixed flexion at the elbows indicates an
elbow problem]. Examine hands for swelling, wasting and deformities.
• Place hands together in prayer position (pray hands) with elbows apart – [flexion
1
Stresses usually cause flair ups of rheumatic diseases.
The detailed examinations for the individual joints follow a similar regimen – look, feel and
move.
LOOK
You first need to observe the patient without touching them. At first you look at the
patient’s joint while it is not moving at all (inspection at rest) and then inspect it as the
patient moves the limb (inspection during movement).
• At rest. At rest, you need to not if there is: symmetry & attitude of limbs, swelling,
visible deformity, skin changes, rash, muscle wasting, signs of inflammation (redness)
and scars.
• During movement. You look for:
o Restriction of movement: whether it is restricted in a single plane or in
multiple planes.
2
You can also do the Schober’s test, in which you measure out a line along the spine between a point 5cm
below the level of the posterior iliac spine and 10cm above it. If upon flexion the line does not lengthen by at
least 5cm, there is reduced lumbar flexion, such as in ankylosing spondylitis.
FEEL
• Tenderness: check to see if the limb is tender at rest. Palpate along the joint line as
well as around the joint (peri-articular). Also check for tenderness on passive
movement.
• Swelling: feel the swelling to establish whether it is fluctuant (fluid), soft (soft
tissue), rubbery (synovium) or hard (bone).
• Warmth: this usually signifies inflammation, whether infectious or not.
• Crepitus: this can be coarse or fine, and they may be loud or soft & only heard by
auscultation.
• Stability: this is established by passively moving the joint. Check the ranges of
movement.
• Restriction of movement: check to see if the patient has full ranges of movement.
MOVE
This is assessed by determining features during active movement (carried out by patient) &
passive movement (carried out by examiner). Movements are assessed during looking &
feeling.
Different findings of joint disease point to different things. One important thing is the
distribution of affected joints (the number of joint):
Common conditions
• Septic arthritis. This is an infection of the joint space. It usually arises from
introduction of infection into the joint space by direct injury or through blood-borne
transmission from an infected site. The risk factors for contracting septic arthritis
include:
o Chronically inflamed & damaged joints.
o Immunosuppression – AIDS, immunosuppressive therapy.
o Extremes of age – infants & elderly.
o Alcohol abusers.
o Those with joint prostheses.
In the majority of patients, S. aureus is the cause. Other causes include: N.
gonorrhoeae, H. influenzae, Gram-negative organisms (both aerobic & anaerobic),
Streptococcus species and TB. In young previously fit patients, the joint is red, hot,
swollen and agonisingly painful, and the joint is held immobile by muscle spasm. In
the young & old, immunocompromised and in RA patients, the picture is less
dramatic, and so an index of suspicion should be suspected. Septic arthritis is a
medical emergency because it causes rapid destruction of the joint. You do a joint
tap and take the fluid for MCS (microscopy, culture and sensitivity) and you take
blood samples for culture and for FBC. In the acute setting, X-rays have no role. You
can also take skin & throat swabs and urine samples to identify the source of
infection. In the initial phase of treatment, you immobilise the joint and then start
physiotherapy early. You first administer empirical therapy:
flucloxacillin/erythromycin/clindamycin + gentamycin/fusidic acid. You then change
to definitive therapy when culture & sensitivity results come out. Drainage of the
joint & arthroscopic joint washouts is helpful in relieving pain. In the case of infected
prostheses, you remove the prosthesis and fill the joint with a spacer impregnated
with antibiotics.
Specific types of septic arthritis include:
Infectious At-risk Features of disease Diagnosis Treatment
organism population
N. Most common Fever, characteristic Blood culture in Oral penicillin,
gonorrho cause in young, pustular eruption on early stage. Joint ciprofloxacin
eae previously fit distal limbs, fluid may be or doxycycline
adults: polyarthralgia, sterile with for 2 weeks
commoner in tenosynuvitis positive genital with joint
women & gay tract culture. immobilisatio
men. n & joint rest.
TB 1% of TB cases; in Insidious course; forms Culture & biopsy Treat with
adults usually caseating granulomas of synovium. HRZE for 2
from secondary in cartilage & adjacent Chest X-ray months and
3
This is known as Poncet’s disease.
4
Gout is less likely in women of reproductive age because oestrogen enhances urate excretion.
5
In pseudo-gout, the crystals are positively birefringent. Pseudo-gout is caused by deposition of calcium
pyrophosphate crystals.