Introductory Clinical Sciences Flashcards

1
Q

What is inflammation?

A

A reaction to injury or infection involving cells such as neutrophils and macrophages

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Give 5 cardinal signs of inflammation.

A
  1. Redness (rubor).
  2. Swelling (tumor).
  3. Pain (dolor).
  4. Heat (calor).
  5. Loss of function.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Give a disadvantage of inflammation.

A

Inflammation can produce disease and can lead to distorted tissues with permanently altered function.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

When is inflammation bad?

A
  1. Autoimmunity
  2. When it’s an overreaction to the stimulus
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is the expected outcome of acute inflammation?

A

The complete elimination of a pathogen followed by resolution of damage, disappearance of leukocytes and regeneration of tissue.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Acute inflammation features

A
  1. Sudden onset
  2. Short duration
  3. Usually resolves
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Give 6 causes of acute inflammation.

A
  1. Microbial infections (bacteria and viruses).
  2. Chemicals (corrosives, acids/alkalis).
  3. Physical agents (trauma, burns, frost bite).
  4. Hypersensitivity reactions (TB).
  5. Bacterial toxins.
  6. Tissue necrosis.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Chronic inflammation features

A
  1. Slow onset or sequel to acute
  2. Long duration
  3. May never resolve
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What is chronic inflammation?

A

Persistent, unresolved inflammation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Give 4 causes of chronic inflammation.

A

Primary chronic inflammation.
Transplant rejection.
Recurrent acute inflammation.
Progression from acute inflammation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Give examples of primary chronic inflammation.

A
  1. Infective substances having resistance to phagocytosis e.g. TB, leprosy.
  2. Endogenous materials e.g. uric acid crystals.
  3. Exogenous materials e.g. asbestos.
  4. Autoimmune diseases e.g. chronic gastritis, rheumatoid arthritis etc.
  5. Other chronic inflammatory diseases e.g. chronic inflammatory bowel disease.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Which one of the following is a chronic inflammatory process from its start?

A. Appendicitis
B. Cholecystitis
C. Infectious mononucleosis
D. Lobar pneumonia

A

C. Infectious mononucleosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What cells are involved in acute inflammation?

A
  • Neutrophil polymorphs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Give 3 endogenous chemical mediators of acute inflammation.

A

Bradykinin.
Histamine.
Nitric Oxide.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Features of neutrophil polymorphs

A
  • Short lived cells
  • First on the scene of acute inflammation
  • Cytoplasmic granules full of enzymes that kill bacteria
  • Usually die at the scene of inflammation
  • Release chemicals that attract other
    inflammatory cells such as macrophages
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What are 4 systemic effects of acute inflammation?

A

Fever.
Feeling unwell.
Weight loss.
Reactive hyperplasia of the reticuloendothelial system.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What cells are involved in chronic inflammation?

A

Macrophages and plasma cells (B and T lymphocytes).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Features of macrophages

A
  • Long lived cells (weeks to months)
  • Phagocytic properties
  • Ingest bacteria and debris
  • May carry debris away
  • May present antigen to lymphocytes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What cell can form when several macrophages try to ingest the same particle?

A

Multinucleate giant cell.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What are some macroscopic features of chronic inflammation?

A

Chronic ulcer.
Chronic abscess cavity.
Granulomatous inflammation.
Fibrosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Features of lymphocytes

A
  • Long lived cells (years)
  • Produce chemicals which attract in other inflammatory cells
  • Immunological memory for past infections and antigens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Plasma cell function and features.

A
  • B cell antibody production
  • Lots of endoplasmic reticulum to make protein.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Endothelial cells features

A
  • Line capillary blood vessels in areas of inflammation
  • Become sticky in areas of inflammation so inflammatory cells adhere to them
  • Become porous to allow inflammatory cells to pass into tissues
  • Grow into areas of damage to form new capillary vessels
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What happens to the arterioles and capillary beds during inflammation?

A

In acute inflammation, the precapillary sphincters open, causing blood to flow through all capillaries. This is good in localised areas.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Define exudate.

A

A protein rich fluid that leaks out of vessel walls due to increased vascular permeability.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

Why does inflammation lead to oedema

A
  • Normally, fluid leaves the arteriole end and enters through the venous end.
  • In acute inflammation, the sphincter is open and there is a higher pressure in the arteriole end and capillaries are more porous.
  • There is a net loss of fluid together with plasma protein into the extracellular space.
  • This leads to oedema
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Fibroblasts features

A
  • Long lived cells
  • Produce collagenous connective tissue in scarring following some types of inflammation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What occurs during acute appendicitis? (eg of acute inflammation)

A

– Unknown precipitating factor
– Neutrophils appear
– Blood vessels dilate
– Inflammation of serosal surface occurs
– Pain felt
– Appendix either surgically removed or inflammation resolves or appendix bursts with generalised peritonitis and possible death

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What are the 4 outcomes of inflammation?

A
  1. Resolution.
  2. Suppuration.
  3. Organisation (scar tissue formation).
  4. Progression onto chronic inflammation.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What occurs during tuberculosis? (eg of chronic inflammation)

A

– No initial acute inflammation
– Mycobacteria ingested by macrophages
– Macrophages often fail to kill the mycobacteria because of the cell wall
– Lymphocytes appear
– Macrophages appear
– Fibrosis occurs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What are granulomas?

A

A aggregation of epithelioid histocytes surrounded by lymphocytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What is granulation tissue?

A

Granulation tissue is composed of small blood vessels in a connective tissue matrix with myofibroblasts. It is important in healing and repair.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What is a chronic granulomatous disease (CGD)?

Give 4 examples of granulomatous disease.

A

An inherited disorder that occurs when the white blood cells (phagocytes) do not work properly. Therefore, the phagocytes cannot protect the body from bacterial or fungal infections.

TB, leprosy, Crohn’s disease and sarcoidosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

The activity of what enzyme in the blood can act as a marker for granulomatous disease?

A

Angiotensin converting enzyme.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

How would you treat inflammation?

List 5 things

A
  • Ice
  • Antihistamine for insect bites: histamine drives inflammation
  • Aspirin, ibuprofen: inhibits prostaglandin synthetase, prostaglandins are chemical mediators of inflammation
  • Corticosteroids: inhibitors of inflammation down regulate chemical mediators of inflammation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What leads to thrombus formation in arteries ?

A

Endothelial cell injury -> exposing collagen -> platelets aggregate -> Turbulent flow -> fibrinogen polymerises to fibrin -> blood clot

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

What are the 3 factors that can lead to thrombosis formation?

A
  1. Change in vessel wall.
  2. Change in blood constituents.
  3. Change in blood flow.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

Define thrombosis.

A

Formation of a solid mass from blood constituents in an intact vessel in the living.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

Give 2 reasons why thrombosis formation is uncommon.

A
  1. Laminar flow.
  2. Non sticky endothelial cells.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What drug can be used to prevent Thrombosis?

A

Aspirin. Inhibits platelet aggregation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

How is COVID-19 related to thrombosis

A
  • COVID virus causes massive inflammation boosting cytokines
  • Increases the liver’s production of clotting factors
  • Leads to thrombosis.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

Define embolus

A

Mass of material in the vascular system able to become lodged within vessel and block it.

Most commonly, thrombus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What are the consequences of an arterial embolus?

A

An arterial embolus can go anywhere! The consequences could be stroke, MI, gangrene etc.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What are the consequences of a venous embolus?

A

An embolus in the venous system will go onto the vena cava and then through the pulmonary arteries and become lodged in the lungs causing a pulmonary embolism. This means there is decreased perfusion to the lungs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

Through which blood system would an embolus have travelled if it resulted in a pulmonary embolism?

A

Venous system.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What are the effects of thrombosis and embolism

A

Can lead to ischaemia and infarction.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

Define ischaemia.

A

Decreased blood flow.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

What complication may happen if ischaemia is rectified?

A

Re-perfusion injury can occur due to the release of waste products.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

Define infarction.

A

Decreased blood flow with subsequent cell death.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What are most organs in the body supplied by. What issue does this cause?

A

Most organs are supplied by the end arterial supply. If it blocks, can lead to infarction.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

What organs have more than one supply?

A

The lungs (bronchial arteries and pulmonary veins), liver (hepatic arteries and portal veins), and some parts of the brain (circle of Willis).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

What is the difference between resolution and repair?

A

Resolution
–initiating factor removed
–tissue undamaged or able to regenerate
Repair
–initiating factor still present
–tissue damaged and unable to regenerate

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What prevents the liver from regenerating in patients with alcoholic liver disease?

A

Initiating factor not taken away (patient still drinking), leads to cirrhosis where the liver repairs instead of regenerates.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

What damages occur in lobar pneumonia and severe COVID lung damage?

A
  • Pneumocytes can regenerate, but alveolar cannot.
    In lobar pneumonia the lungs can regenerate and can completely recover. In severe cases of Covid, the alveolar cannot regenerate, lungs can only repair.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

What are abrasion skin wounds?

A

Superficial skin wounds that have only damaged the top layer of the epidermis, will heal up normally.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

What happens when healing by 1st intention?

A

Edges by skin are closely re-approximated.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

What happens when healing by 2nd intention?

A

Occurs in traumatic wounds when skin lost and cannot heal through edges. Blood vessels have to grow in from either side. Takes longer to heal.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

Define repair.

A
  • replacement of damaged tissue by fibrous tissue
  • collagen produced by fibroblasts
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

List 3 examples of repair in the human body.

A

–heart after myocardial infarction
–brain after cerebral infarction
–spinal cord after trauma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

Name 6 types of cells capable of regeneration.

A
  • hepatocytes
  • pneumocytes
  • all blood cells
  • gut epithelium
  • skin epithelium
  • osteocytes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

Name 2 type of cells that don’t regenerate?

A
  • myocardial cells
  • neurones
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

Man aged 30 takes a large overdose paracetamol overdose and doesn’t come to hospital for 2 days. He spends a few days on ITU with liver failure but then recovers.

What will be happening in his liver - resolution or repair?

A

Resolution!
Only 1 overdose, and hepatocytes are capable of regeneration.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

A child aged 3 falls off a climbing frame onto his hand, he fractures his clavicle. He has his arm in a sling for 4 weeks and is very good about keeping it in the sling
What will be happening in his clavicle - resolution or repair?

A

Resolution!
The patient is keeping the sling in and osteocytes are capable of regeneration.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

A child aged 3 falls off a climbing frame onto his hand, he fractureshis clavicle. He has his arm in a sling for 4 weeks but won’t keep the sling on and moves his arm a lot
What will be happening in his clavicle - resolution or repair?

A

Repair!
The patient won’t keep the sling on and keeps moving his arms about. Body unable to regenerate because it becomes fibrous and only repair can occur.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

Give a benefit of inflammation.

A

Inflammation can destroy invading micro-organisms in injuries and can prevent the spread of infection.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

What do viral infections result in?

A

Cell death due to intracellular multiplication.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

What does bacterial infection result in?

A

The release of exotoxins (involved in the initiation of inflammation) or endotoxins.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

Define abscess.

A

Acute inflammation with a fibrotic wall.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

Define atherosclerosis.

A

Inflammatory process characterised by accumulation of fibrolipid plaques in the intima of a vessel wall. It causes a huge amount of illness by reducing the blood flow in important areas.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

What is the time course of atherosclerosis?

A
  • Birth - no atherosclerosis
  • Late teenage/early 20s - fatty streaks in aorta, may not progress to established atherosclerosis
  • 30s/40s/50s - development of established atherosclerotic
    plaques
  • 40s-80s - complications of atherosclerotic plaques e.g. thrombosis, intraplaque haemorrhage
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

What is the distribution of atherosclerosis plaques?

A

It is common in high pressure systems such as aorta and systemic arteries, never in pulmonary circulation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

What is in an atherosclerosis plaque?

A
  1. Lipid core.
  2. Necrotic debris.
  3. Connective tissue.
  4. Fibrous cap.
  5. Lymphocytes.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

List 5 risk factors of atherosclerosis.

A
  • Cigarette smoking
  • Hypertension
  • Poorly-controlled diabetes mellitus
  • Hyperlipidaemia
  • Lower social economic status
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

What secondary preventative measure can a GP introduce to a patient post myocardial infarction, known ischaemic disease or known peripheral arterial disease? List 5 examples.

A
  • Statins 80mg (primary prevention would be 20mg)
  • Aspirin 75mg
  • ACE inhibitors
  • Beta blockers
  • Control BP with anti-hypertensives
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

What is the endothelial damage theory

A
  • Endothelial cells are delicate, they require oxygen to produce lots of nitric oxide.
  • If they are in any way impaired will not produce nitric oxide and die.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

Describe the main stages involved in the formation of an atherosclerotic plaque

A
  • Macrophages form foam cells from lipids in arterial wall - fatty streak formation (platelets) > intermediate lesion
  • Plaque protrudes into artery lumen and disrupts laminar flow - medial thinning and platelet aggregation
  • Secondary platelet plug forms fibrin mesh over itself and traps red blood cells
  • Fibroblasts form smooth muscle ‘fibrous cap’ over this
  • Continuous formation of secondary platelet plug, this is a stable atheroma - plaque stabilisation/( fibrous cap formation)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

How does cigarette smoke cause damage to the endothelial cells

A

Free radicals, nicotine and carbon monoxide can damage endothelial cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

How does hypertension damage the endothelial cells?

A

High blood pressure has shearing forces exerted on endothelial cells and this can lead to damage

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

How does poorly-controlled diabetes mellitus damage endothelial cells.

A

Super oxide anions present and glycosylation products both
cause oxidative stress and endothelial injury.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

What does cumulative damage to the endothelium lead to?

A

Endothelial ulceration -> microthrombi -> eventual development of established atherosclerotic plaques
Complete block to an artery will mean no blood flow to the organ and infarction can occur.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

Give 6 examples of complications of atherosclerosis?

A
  • Cerebral infarction
  • Carotid atheroma
  • Myocardial infarction
  • Aortic aneurysms
  • Peripheral artery disease with intermittent claudication
  • Gangrene
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

What is the anatomy of a normal artery and which part of it is important with regards to atherosclerosis?

A
  • Intima, media and adventitia
  • Intima is the most important with regards to ACS. It is formed of only a single layer of endothelium – this is involved in the immune response, releases antithrombotic molecules
    to prevent clots, releases vasodilators and constrictors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

Where does the left and right coronary arteries arise from?

A

The aortic sinuses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

How can occlusion of coronary arteries be detected and treated?

A

Occlusion of any of the coronary arteries can be seen on ECG and via coronary angiogram. It is diagnostic and relieving treatment. Immediate treatment can be done = coronary angioplasty = balloon & stent.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

Define apoptosis

A

Programmed cell death of a single cell.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

What is the role of p53 protein?

A

Cells can detect DNA damage using p53 protein which can then trigger apoptosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

What protein can switch on apoptosis if DNA damage is present?

A

p53 protein.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

How does a cell apoptose?

A

The cell triggers a series of proteins which lead to the release of caspases within the cell which can turn on apoptosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

Activation of what receptor can activate caspase and therefore apoptosis?

A

FAS receptor.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

Give an example of a disease where there is a lack of apoptosis.

A

Cancer; mutations in p53 mean cell damage isn’t detected.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

Give an example of a disease where there is too much apoptosis.

A

HIV

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

Define necrosis.

A

Unprogrammed death of a large number of cells due to an adverse event.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

Give 5 examples of events that can lead to necrosis.

A
  • Infarction due to loss of blood supply e.g. myocardial infarction, cerebral infarction
  • Frostbite
  • Toxic venom from reptiles and insects
  • Avascular necrosis of bone
  • Pancreatitis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

Give 3 differences between apoptosis and necrosis.

A
  1. Apoptosis is programmed cell death whereas necrosis is unprogrammed.
  2. Apoptosis tends to effect only a single cell whereas necrosis effects a large number of cells.
  3. Apoptosis is often in response to DNA damage. Necrosis is triggered by an adverse event e.g. frost bite.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

Which one of the following is not an example of apoptosis?
A. Loss of cells from tips of duodenal villi
B. Loss of cells during embryogenesis
C. Renal infarction
D. Graft versus host disease

A

C. Renal infarction.

Necrosis not apoptosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

How does the spinal cord form?

A
  • Initial epithelium
  • Columnar base
  • Invagination
  • Tube formation and closure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

What are the types of spina bifida?

a congenital defect of the spine

A
  • Spina bifida occulta
  • Meningocele
  • Myelomeningocele
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

What genes control how cells migrate and differentiate?

A

Homeobox genes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

Define congenital.

A

Present at birth.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

Give an example of a disease which is congenital but not genetic.

A

Club foot.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

Why do people with Down syndrome tend to have earlier dementia?

A

This is because they have trisomy 21, this leads to excessive beta amyloid being produced which can clump together to form plaques between neurons.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

Define polygenic inheritance.

A

Polygenic inheritance refers to inheritance in which the trait is produced from the cumulative effects of many genes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

Give an example of a congenital acquired disease.

A

Foetal alcohol syndrome.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

Is dwarfism inherited?

A

Yes, caused by genetic inheritance leading to growth hormone deficiency.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

Is gigantism inherited?

A

No, it’s caused by growth hormone excess from pituitary adenoma.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

Define hypertrophy.

A

Increase in the size of a tissue due to an increase in the size of constituent cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

Why are cows able to grow muscle despite a grass-fed diet?

A

Mutation in the myostatin gene

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

Define hyperplasia.

A

Increase in the size of a tissue due to an increase in the number of constituent cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

Define atrophy.

A

Decrease in the size of a tissue due to a decrease in the size of the constituent cells OR due to a decrease in the number of constituent cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
110
Q

Define metaplasia.

A

A change in the differentiation of a cell from one fully differentiated cell type to another fully differentiated cell type.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

Give an example of a disease that demonstrates metaplasia.

A

Barrett’s oesophagus - the cells at the lower end of the oesophagus change from stratified squamous cells to columnar.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

Define dysplasia.

A

Morphological changes seen in cells in the progression to becoming cancer.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

Dysplasia is sometimes used to refer to developmental abnormalities. Give an example of this.

A

Focal cortical dysplasia.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

What happens to a cell when the telomere gets too short?

A

It can no longer divide.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

What is the limit to how many times a human cell can divide called?

A

The Hayflick limit.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

What are the effects of ageing on skin? What is the cause of it?

A

Dermal elastosis is caused by UV-B light causing cross-linking of the proteins, especially collagen, in the dermis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

How can osteoporosis be prevented?

A

Can be prevented by hormone replacement therapy at menopause and calcium/vitamin D supplements.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

Give 6 examples of ageing in different body parts?

A
  • Wrinkling of skin (dermal elastosis)
  • Eyes - cataracts
  • Osteoporosis
  • Dementia
  • Loss of muscle (sarcopaenia)
  • Deafness
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

Why does ageing cause osteoporosis?

A

Lack of oestrogen post menopause leading to increased bone resorption and decreased bone formation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

What are the effects of ageing on the eyes? What is the cause of it?

A

Cataracts - caused by UV-B cross-linking of proteins in the lens causing opacity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

How can cataracts in old age be prevented and treated?

A

Can be prevented by wearing sunglasses that cut out UV light.
Can be treated by replacement of the lens with a plastic implant.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

Cause of sarcopenia in old age?

A

Reduced levels of growth hormone and testosterone, increased catabolic cytokines in later life.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

How can sarcopenia be prevented?

A

Regular exercise including resistance/weight training

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

What causes deafness in old age?

A

The hair cells in the cochlear do not divide so if they are damaged by high volumes they will die and not be replaced eventually producing deafness.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

Which one of the following is an example of atrophy?
A. Biceps of a body builder
B. Uterus in pregnancy
C. Brain in dementia
D. Prostate in older age

A

C. Brain in dementia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

Give an example of:

a) a dividing tissue.
b) a non dividing tissue.

A

a) Gut or skin tissue can divide.

b) Brain tissue is non dividing.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

Why can excision be used as a cure for basal cell carcinoma?

A

Because BCC only invades locally and doesn’t metastasise.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

Define carcinoma.

A

Malignant epithelial neoplasm

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

Why can’t excision be used as a cure for leukemia?

A

Leukemia is systemic, it circulates and always spreads all around the body, therefore excision can’t be used.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

Suggest a treatment that could be used for leukemia?

A

Chemotherapy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

Where can the majority of breast carcinomas spread to?

A

Breast cancers can spread to the lymph nodes that drain the site of the carcinoma.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

Which 5 carcinomas commonly spread to bone?

A
  1. Breast
  2. Kidney
  3. Lung
  4. Prostate
  5. Thyroid

PORTABLE
PO=prostate, R=renal, TA=thyroid, B=breast, LE=lung

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

How would you plan for treatment of breast cancer?

A
  • Confirm diagnosis of breast cancer. (eg core biopsy needle)
  • Confirm whether it has spread to the axilla to decide whether axillary node clearance is needed. (eg ultrasound of the axilla)
  • Confirm whether its has spread to the rest of the body. (eg bone scan)
  • If cancer has spread to rest of body, systemic therapy needed. If not, then surgery with, or without axillary lymph node clearance can take place.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

Even if a tumour is completely excised, what complications may there be? What can be used to treat these patients?

A

Micro metastases could be present.
Adjuvant therapy (extra treatment given after surgical excision) can be used to suppress secondary tumour formation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

What common treatment is given to patients who have undergone lumpectomy?

A

Radiotherapy to the breast.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

What secondary treatment is given to breast cancer patients who test oestrogen receptor positive?

A

adjuvant anti-oestrogen therapy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

What secondary treatment is given to breast cancer patients who test positive for amplified HER2 gene?

A

Herceptin (Trastuzumab)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

Define carcinogenesis.

A

The transformation of normal cells to neoplastic cells through permanent genetic alterations or mutations.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

What does oncogenesis cause?

A

Cause the development of benign and malignant tumours

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

What is the difference between carcinogenic and oncogenic?

A

carcinogenic = cancer causing
oncogenic = tumour causing

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

What percentage of cancer risk is due to environmental factors? What problems does this have?

A

85% of cancer risk is environmental
Rest are chance, genetics, micro-organisms
Problems:
- latent interval may last decades
- complexity of environment
- ethical constraints

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

Chemical carcinogens: what types of cancer do polycyclic aromatic hydrocarbons cause?

A

Skin cancer and lung cancer.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

Chemical carcinogens: what types of cancer does Beta-Naphthylamine cause?

A

Bladder cancer.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

Chemical carcinogens: what can expose people to polycyclic aromatic hydrocarbons?

A

Smoking cigarettes and mineral oils.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

Chemical carcinogens: what types of cancer do aromatic amines cause?

A

Bladder cancer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
146
Q

Chemical carcinogens: what types of people are more susceptible to bladder cancer caused by aromatic amine exposure?

A

People who work in the rubber/dye industry.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
147
Q

Chemical carcinogens: what types of cancer do nitrosamines cause?

A

Gut cancer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
148
Q

Chemical carcinogens: what types of cancer do alkylating agents cause?

A

Leukaemia; the risk is small in humans.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
149
Q

Chemical carcinogens: what types of cancer does radioactive iodine cause?

A

Thyroid cancer (seen during Chernobyl)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
150
Q

Name the 5 different classes of carcinogens.

A
  1. Viral.
  2. Chemical.
  3. Ionising and non-ionising radiation.
  4. Hormones, parasites and mycotoxins.
  5. Miscellaneous e.g. asbestos and metals.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
151
Q

List 4 features of chemical carcinogens.

A
  • No common structural features
  • Some act directly
  • Most requires metabolic conversion from procarcinogens to ultimate carcinogens
  • Enzyme required may be ubiquitous or confined to certain organs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

What percentage of all cancers are made up by viruses?

A

10-15% of all cancers are caused by viruses. Most oncogenic viral infections don’t results in cancer.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

Name 5 viral DNA carcinogens and the cancer they cause.

A
  1. Human Herpes Virus 8 (HHV8) - Kaposi sarcoma
  2. Epstein Barr Virus (EBV) - Burkitt lymphoma, Nasopharyngeal carcinoma
  3. Hepatitis B Virus (HBV) - Hepatocellular carcinoma
  4. Human papillomavirus (HPV) - Squamous cell carcinomas of the cervix, penis, anus, head and neck
  5. Merkle cell polyomavirus (MCV) - Merkle cell carcinoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

Name 2 viral RNA carcinogens and the cancer they cause

A
  1. Human T-lymphotrophic virus (HTLV-1) - Adult T-cell leukemia
  2. Hepatitis C Virus (HCV) - Hepatocellular carcinoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

Features of radiant energy carcinogens.

A
  • Ionising radiation
  • Long term effect
  • Skin cancer in radiographers
  • Lung cancer in uranium miners (radon gas)
    -Thyroid cancer in Ukrainian children
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
156
Q

What biological agents cause cancer?

A
  • Hormones such as oestrogen (mammary/endometrial cancer) and anabolic steroids (hepatocellular carcinoma)
  • Mycotoxins such as aflatoxin B1 (hepatocellular carcinoma)
  • Parasites such as Chlonorchis sinensis (cholangiocarcinoma) and Shistosoma (bladder cancer)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
157
Q

Give 5 host factors that can affect cancer risk.

A
  1. Ethnicity.
  2. Diet.
  3. Constitutional factors (gender, age).
  4. Premalignant conditions.
  5. Transplacental exposure.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
158
Q

Host factors - premalignant conditions. Name 3 identifiable local abnormalities which are associated with increased risk of malignancy at that site

A
  • Colonic polyps
  • Ulcerative colitis
  • Cervical Dysplasia (CIN)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
159
Q

Give an example of a situation when transplacental exposure lead to an increase in cancer risk.

A

The daughters of mothers who had taken diethylstilboestrol for morning sickness had an increased risk of vaginal cancer.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
160
Q

A 72 year old man from Singapore has smoked for all his adult life, placing the lit end of his cigarettes in his mouth. ‘Reverse smoking’ increases the risk of developing which cancer?

A

Oral cancer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
161
Q

A 42 year old woman has recently returned from Uganda. She continued her hobby of fresh water swimming during her trip. She now complains of an itchy rash and a fever. Her doctor suspects she may have schistosomiasis. Left untreated, this parasitic infection increases the risk of which cancer?

A

Bladder cancer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
162
Q

A 51 year old woman with HIV presents to her doctor with multiple purple bruise-like lesions on the skin of her face, torso and limbs. A biopsy confirms the diagnosis of Kaposi sarcoma. Which virus causes Kaposi sarcoma?

A

Human Herpes Virus 8 (HHV8)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
163
Q

A 27 year old man is worried about his alcohol consumption and seeks advice about the effect it might have on his health. Which cancer can be caused by excess alcohol consumption?

A

Colon cancer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
164
Q

A 32 year old man is admitted to hospital with abdominal pain. The pain is in his right iliac fossa. A scan is performed and his appendix is removed by laparoscopic surgery. The appendix is sent for histopathology examination. What is the predominant cell that the pathologist sees on microscopic examination?

A

Neutrophil polymorphs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
165
Q

Pathologist finds a white area of scarring in the myocardium and takes a sample for histology. What is the predominant cell the pathologist will see in the white area?

A

Fibroblasts

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
166
Q

What is carcinoma in situ?

A

When the carcinoma hasn’t reached the basement membrane, thus it can’t enter the lymphatic system or blood vessels and cannot spread.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
167
Q

What do you call carcinoma that has reached the basement membrane?

A

Invasive carcinoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
168
Q

What is micro-invasive carcinoma?

A

Carcinoma that has invaded through the basement membrane, but only by a little. Potentially, it could spread through the body, but the risk of it spreading is extremely low.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
169
Q

What properties help the carcinoma invade the basement membrane? (intravasation)

A
  • proteases - matrix metalloproteases
  • cell motility
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
170
Q

How does the invasive carcinoma metastasise?

A
  • Tumour invades basement membrane and stroma
  • Tumour has to enter system (ie lymphatic vessels, venules, transcoeleomic, arteries though unlikely)
  • Tumour cells have to resist immune response in the lymphatic fluid or venous blood as the lymphocytes and macrophages would act against it
  • Tumour cells have to exit and invade the extracellular matrix and start growing
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
171
Q

What is required for a tumour to exit the blood stream (extravasation)?

A
  • Adhesion receptors.
  • Collagenases.
  • Cell motility.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
172
Q

Give 2 promoters of tumour angiogenesis.

A
  1. Vascular endothelial growth factors.
  2. Fibroblast growth factors.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
173
Q

Give 3 inhibitors of tumour angiogenesis.

A
  1. Angiostatin.
  2. Endostatin.
  3. Vasculostatin.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
174
Q

What 3 mechanisms do tumour cells use to evade host immune defence?

A
  • Aggregation with platelets
  • Shedding of surface antigens
  • Adhesion to other tumour cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
175
Q

What kind of drugs can be used in targeted chemotherapy?

A

Monoclonal antibodies (MAB) and small molecular inhibitors (SMI).

MAB can be used to treat constitutive activation of receptors.

SMI can be used to treat overexpression of receptors.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
176
Q

Give an example of a malignant tumour that often spreads to the lung.

A

Sarcoma (via venae cava -> heart -> pulmonary arteries).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
177
Q

Give an example of carcinomas that can spread to the liver.

A

Colon, stomach, pancreas, carcinoid tumours of intestine can spread to the liver via the portal vein.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
178
Q

What are the common cancers to metastasise to the bone?

A

PORTABLE
Po – Prostate
R – Renal
Ta – Thyroid
B – Breast
Le – Lung

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
179
Q

Conventional chemotherapy: how does vinblastine work?

A

In order for cells to divide cells have to replicate. Binds to the microtubules and prevents them working.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
180
Q

Conventional chemotherapy: how does etoposide work?

A

In order for cells to divide cells have to replicate. Etoposide inhibits topoisomerase II

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
181
Q

Conventional chemotherapy: how does ifosamide work?

A

Ifosamide binds directly to DNA
inhibits DNA synthesis by cross linking

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
182
Q

Conventional chemotherapy: how does cisplatin work?

A

Cisplatin binds directly to DNA and inhibits DNA synthesis by cross linking

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
183
Q

Give an advantage and a disadvantage of conventional chemotherapy.

A

Advantage: works well for treatment against fast dividing tumours e.g. lymphomas.

Disadvantage: it is non selective for tumour cells, normal cells are hit too; this results in bad side effects such as diarrhoea and hair loss.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
184
Q

What type of tumours are conventional chemotherapy good for? List 5 examples

A

Fast dividing tumours
- germ cell tumours of testis
– acute leukaemias
– lymphomas
– embryonal paediatric tumours
– choriocarcinoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
185
Q

What type of tumour is conventional chemotherapy not good for?

A

Slow dividing tumours - some tumour cells increase by mitigating apoptosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
186
Q

What is the histogenetic classification of neoplasms?

A

Histopathological tests specify tumour type by determining the cell of origin of a tumour.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
187
Q

Name two genetic drivers of carcinogenesis?

A
  • Proto-oncogenes: genes that promote cell growth and survival
  • Tumour suppressor genes: genes which inhibit cell growth and proliferation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
188
Q

How would you diagnose bronchial carcinoma (small and non small)?

A
  • Sputum
  • Radiology-guided biopsy
  • Bronchoscopy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
189
Q

What is tumour grading and staging?

A

Tumour grading:
How much the tumour cells resemble their normal counterparts

Tumour staging:
How much the tumour has spread. Involves assessment of lymph nodes, extent of spread to other organs and tissues.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
190
Q

What is the name of a malignant tumour of straited muscle?

A

rhabdomyosarcoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
191
Q

What term describes a cancer that has not invaded through the basement membrane?

A

Carcinoma in situ

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
192
Q

What is the name of a benign tumour of glandular epithelium?

A

Adenoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
193
Q

What is the name of a benign tumour of fat cells?

A

lipoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
194
Q

What is the name of a malignant tumour of glandular epithelium?

A

adenocarcinoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
195
Q

True or False: Ovarian cancer commonly spreads in the peritoneum.

A

True

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
196
Q

A 27 year old woman presents to the Accident and Emergency Department with abdominal pain. She says that it started the day before when she felt non-specifically unwell and felt pain in the middle of her abdomen. She slept rather fitfully and woke up this morning with a more severe pain that had moved to her right groin region. A diagnosis of acute appendicitis is made and her appendix is removed during a laparoscopic operation. The appendix is sent to the Department of Histopathology where it is sampled and microscopic slides are produced. The Consultant Histopathologist looks at the lumen of the appendix on a high power lens on her microscope.
What is the predominant type of inflammatory cell that she sees?
- Eosinophil
- Lymphocyte
- Macrophage
- Mast cell
- Neutrophil polymorph

A

Neutrophil polymorph - acute inflammation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
197
Q

Define neoplasm

A

An autonomous, abnormal, persistent new growth.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
198
Q

What are the 3 commonest causes of cancer death in males?

A
  1. Lung cancer
  2. Prostate cancer
  3. Bowel cancer
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
199
Q

What are the 3 commonest causes of cancer death in women?

A
  1. Lung cancer
  2. Breast cancer
  3. Bowel cancer
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
200
Q

What is the structure of a neoplasm?

A
  1. Neoplastic cells.
  2. Stroma.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
201
Q

Describe neoplastic cells.

A
  • Derived from nucleated cells.
  • Usually monoclonal
  • Growth pattern and synthetic activity is related to the parent cell.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
202
Q

Describe the stroma of a neoplasm.

A
  • Connective tissue composed of fibroblasts and collagen; it is very dense.
  • There is a lot of mechanical support and blood vessels provide nutrition
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
203
Q

What is essential for neoplasm growth?

A

Angiogenesis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
204
Q

Why does central necrosis occur in neoplasm?

A

The neoplasm grows quickly and outgrows its vascular supply.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
205
Q

What are the two ways in which neoplasms can be classified?

A
  1. Behavioural classification.
  2. Histogenetic classification.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
206
Q

What are the 5 features of a benign neoplasms.

A
  • non invasive, localised
  • slow growth rate
  • well-circumscribed or encapsulated
  • low mitotic activity
  • close resemblance to normal tissue
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
207
Q

What is the behavioural classification of neoplasms?

A

Neoplasms can be classified as benign, malignant or borderline. Borderline tumours (e.g. some ovarian lesions) defy precise classification.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
208
Q

List 4 histological features of benign neoplasms.

A
  • Nuclear morphometry often normal
  • Necrosis rare
  • Ulceration rare
  • Growth on mucosal surfaces usually exophytic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
209
Q

What are the 5 complications of benign neoplasms?

A
  • Pressure on adjacent structures
  • Obstruct flow
  • Produce hormones
  • Transform to malignant neoplasm
  • Anxiety
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
210
Q

What are the 5 features of malignant neoplasms?

A
  • Invasive
  • Metastases
  • Rapid growth rate
  • Variable resemblance to normal tissue
  • Poorly defined or irregular border
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
211
Q

List 5 histological features of malignant neoplasms.

A
  • Hyperchromatic nuclei
  • Pleomorphic nuclei
  • Increased mitotic activity
  • Necrosis and ulceration common
  • Growth on mucosal surfaces and skin often endophytic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
212
Q

List 7 complications of malignant neoplasms?

A
  • Destruction of adjacent tissue
  • Metastases
  • Blood loss from ulcers
  • Obstruct flow
  • Produce hormones
  • Paraneoplastic effects
  • Anxiety and pain
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
213
Q

What is a papilloma?

A

A benign neoplasm of non-glandular non-secretory epithelium

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
214
Q

What is an adenoma?

A

Benign tumour of glandular or secretory epithelium.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
215
Q

What are carcinomas of glandular epithelium called?

A

adenocarcinomas

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
216
Q

What is a leiomyoma?

A

A benign smooth muscle neoplasm.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
217
Q

What is a neuroma?

A

A benign neoplasm of nerves.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
218
Q

What is a chondrosarcoma?

A

A malignant neoplasm of cartilage.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
219
Q

What is a liposarcoma?

A

A malignant neoplasm of adipose tissue.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
220
Q

What is a melanoma?

A

A malignant neoplasm of melanocytes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
221
Q

What is a lymphoma?

A

A malignant neoplasm of lymphoid cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
222
Q

What is a mesothelioma?

A

A malignant neoplasm of mesothelial cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
223
Q

Carcinomas and sarcomas are further classified according to the degree of differentiation. Is a carcinoma/sarcoma with a close resemblance to normal tissue classified as well differentiated or poorly differentiated?

A

A carcinoma/sarcoma with a close resemblance to normal tissue is classified as well differentiated. These types of neoplasms are low grade and have a better prognosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
224
Q

What is the neoplasm called if its cell of origin is unknown?

A

Anaplastic

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
225
Q

What are teratomas?

A

A teratoma is a rare type of neoplasm that may contain immature or fully formed tissue, including teeth, hair, bone and muscle.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
226
Q

A 43 year old woman is referred to a gynaecologist with a history of pain during sexual intercourse and vaginal bleeding after intercourse and in between her periods. The gynaecologist takes a biopsy of the ectocervix which reveals a primary malignant neoplasm. What is the likely diagnosis?

A

Squamous cell carcinoma
- As the ectocervix is lined by stratified squamous epithelium. Malignancy of stratified squamous epithelium is squamous cell carcinoma.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
227
Q

A 46 year old woman undergoes an elective hysterectomy to treat severe menorrhagia (heavy period). The pathologist examining the specimen reports the presence of multiple benign smooth muscle neoplasms within the myometrium. What are these neoplasms called?

A

Leiomyomas

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
228
Q

A 72 year old man undergoes a biopsy of his colon. The biopsy reveals a disease process that is not neoplastic or due to an infection. What is the likely abnormality?

A

Granuloma (chronic inflammation)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
229
Q

What are the 7 hallmarks of cancer?

A
  • Growth self-sufficiency
  • Evade apoptosis
  • Ignore anti-proliferative signals
  • Limitless replication potential
  • Sustained angiogenesis
  • Invade tissues
  • Escape immune surveillance
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
230
Q

What is cancer immunosurveillance?

A

When the immune system recognises and destroys transformed cells, this is an important host protection process.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
231
Q

What is cancer immunoediting?

A

Immune responses can change tumours to be hidden from recognition by the immune system and tumours can promote immune suppression

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
232
Q

What are the two types of tumour antigens and where are they found?

A
  1. Tumour specific antigens
    - Only found on tumour cells.
    - As a result of point mutations or gene rearrangement
    - Derive from viral antigens
  2. Tumour associated antigens
    - Found on both normal and tumour cells, but are overexpressed on cancer cells
    - Developmental antigens which become derepressed. (CEA)
    - Differentiation antigens are tissue specific
    - Altered modification of a protein could be an antigen
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
233
Q

List 5 evidence for human tumour immunity

A
  • Spontaneous regression
  • Regression of metastases after removal of primary tumour
  • Infiltration of tumours by lymphocytes and macrophages
  • Lymphocyte proliferation in draining lymph nodes
  • Higher incidence of cancer after immunosuppression, immunodeficiency (AIDS, neonates), aging, etc.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
234
Q

What is tumour escape?

A

Immune responses change tumours such that tumours will no longer be seen by the immune system.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
235
Q

What is immune evasion?

A

Tumours change the immune responses by promoting immune suppressor cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
236
Q

How can we kill tumours without killing normal cells?

A

To induce an immune response against the tumour that would discriminate between the tumour and normal cells: Adaptive immunity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
237
Q

What are the two types of immunotherapy?

A
  • Active immunotherapy
  • Passive immunotherapy -
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
238
Q

List 6 examples of active immunotherapy.

A

Vaccines!
- Killed tumour vaccine
- Purified tumour antigens
- Professional APC-based vaccines
- Cytokine- and costimulator-enhanced vaccines
- DNA vaccines
- Viral vectors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
239
Q

List 2 examples of passive cancer immunotherapy.

A
  • Adoptive Cellular Therapy (T cells)
  • Anti-tumour Antibodies (Her-2/Neu, CD20, CD10, CEA, CA-125, GD3 ganglioside)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
240
Q

Features of cell-based therapy.

A
  • Cellular therapies can be used to activate a patient’s immune system to attack cancer.
  • They can also be used as delivery vehicle to target therapeutic genes to attack the tumour
  • They do not act directly on cancer cells. Instead, they work systemically to activate the body’s immune system.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
241
Q

What 2 cell types are commonly used for cell-based therapy?

A
  • Dendritic cells
  • T cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
242
Q

Features of dendritic cells.

A
  • Antigen presenting cells
  • Found throughout the body (0.1-0.5%)
  • Interstitial cells (Liver, heart, liver), Langerhans cells of the epidermis.
  • Detect and chew up foreign “invader” proteins and then “present” piece of the invaders on their surface.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
243
Q

How would you create a dendritic cell cancer vaccine?

A

To make a DC vaccine, the blood of the cancer patient is collected and enriched to increase the population of DC.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
244
Q

How would you create a T cell cancer vaccine?

eg for melanoma

A
  • Tumour biopsy
  • Isolation: tumour fragments and tumor infiltrated lymphocytes (TILs) are isolated
  • Cultivation: TILs propagated on tumour fragments and cultivated
  • Expansion: expansion of melanoma reactive TILs
  • T cell infusion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
245
Q

What is tumour hypoxia?

A

Hypoxia is a prominent feature of malignant tumours. The inability of the blood supply to keep up with growing tumour cells can lead to hypoxic tumour cells that have adapted to low oxygen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
246
Q

What are the problems with tumour hypoxia?

A

Tumour hypoxia usually means poor patient prognosis.
- Stimulates new vessel growth
- Suppresses immune system
- Resistant to radio- and chemotherapy (repopulate the tumour)
- Increased tumour hypoxia after therapy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
247
Q

Why is hypoxia a prominent feature of a lot of malignant tumours?

A

Malignant tumours grow rapidly and so outgrow their blood supply

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
248
Q

What are the 3 types of vaccines traditionally used?

A
  • whole-killed
  • toxoid
  • live attenuated
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
249
Q

Define passive immunisation.

A

The administration of pre-formed “immunity” from one person or animal to another person

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
250
Q

Limitation of passive immunisation.

A

Only humoral (antibody) mediated
(will not work if cell mediated!)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
251
Q

What are the advantages and disadvantages of passive immunity

A

Advantages
- Gives immediate protection
- Effective in immunocompromised patients
Disadvantages
- Short-lived
- Possible transfer of pathogens
- “Serum sickness” on transfer of animal sera

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
252
Q

Give 4 examples of passive immunisation

A

Specific Immunoglobulin (Ig)
- Human Tetanus Ig (HTIG) Rapid protection of exposed individuals
- Human Rabies Specific Ig - Used after exposure to rabies to give protection until vaccine becomes effective
- Human Hepatitis B Ig (HBIG)
- Varicella Zoster Ig (VZIG)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
253
Q

What are the 3 main approaches to making a vaccine for active immunisation?

A
  • Using a whole virus or bacterium
  • Using parts that trigger the immune system
  • Using just the genetic material
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
254
Q

List 3 features of the whole microbe vaccine

A
  • Bacteria or viruses grown in vitro and inactivated using agents such as formaldehyde or Β-propionolactone.
  • Non-living vaccines do not cause infection, but the antigens contained in it induce an immune response that protects against infection – by non-self antigen recognition.
  • Non-living vaccines can also be cell-free toxoids - inactivated toxins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
255
Q

List 4 problems and limitations of whole killed vaccines.

A
  1. The organisms must be grown to high titre in vitro (viruses and some bacteria difficult/expensive to grow in the lab)
  2. Whole pathogens can cause excessive reactogenicity (i.e., adverse reactions, excessive immunological responses)
  3. Immune responses are not always close to the normal response to infection, e.g., no mucosal immunity, no CD8 Tc responses
  4. Usually need at least 2 shots
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
256
Q

Gives examples of bacteria that have been used in active vaccine.

A
  • Diphtheria - formaldehyde treated toxin - rendered a non-toxic “toxoid”
  • Tetanus - toxoid, causes muscle contractions/spasms - treat purified toxoid in the lab with formalin leading to loss of toxicity but NOT epitopes
  • Pertussis (whooping cough) - killed whole bacteria (Bordetella pertussis) given with the two above as DTP. 3-doses. UK has now moved to acellular pertussis (aP)
  • Cholera - heat-killed bacteria (Vibrio cholerae)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
257
Q

Give examples of viruses that have been used in active vaccine.

A

Polio vaccine (Salk) - inactivated virus-IPV
Influenza vaccine - inactivated virus
Hepatitis A vaccine - inactivated virus
Rabies vaccine - inactivated virus
SARS-Co-V2 (Valneva) - inactivated virus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
258
Q

How do Live Attenuated Vaccines work?

A

The organisms replicate within the host and induce an immune response which is protective against the wild-type organism but does not cause disease.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
259
Q

Define attenuation.

A

Where an organism is cultured in such a way that it does not cause disease when inoculated into humans.

It has lost its pathogenicity but retains its antigenicity – (i.e., shape).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
260
Q

List 4 advantages of Live Attenuated Vaccines.

A
  1. Immune response more closely mimics that following real infection because its not fixed – no shape change.
  2. Better immune response so lower doses are required, so the scale of in vitro growth needed is lower.
  3. Route of administration may be more favourable (oral).
  4. Fewer doses may be required to provide protection.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
261
Q

Problems and limitations of live attenuated vaccines.

A
  1. Often impossible to balance attenuation and immunogenicity
  2. Reversion to virulence
  3. Transmissibility
  4. Live vaccines may not be so attenuated in immunocompromised hosts
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
262
Q

Give 2 examples of live attenuated bacterial vaccines.

A

Bacille Calmette-Guérin (BCG) Mycobacterium bovis grown over many passages in vitro. Gives some protection against TB (tuberculosis)

Salmonella typhi - temperature sensitive strain given orally.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
263
Q

Give 3 examples of viral live attenuated vaccines.

A

Poliomyelitis (Sabin) - widely used to bring polio to the brink of eradication

Vaccinia virus - used in billions of doses to eradicate smallpox due to cross-reactivity between itself and the variola virus

Measles, Mumps and Rubella - 3 given together as MMR

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
264
Q

In this day and age, why do so many pathogens still do not have a vaccine? List 4 possible reasons.

A
  • Pathogen too difficult to grow
  • Killed pathogen not protective (shape change)
  • Impossible to obtain attenuated and suitably immunogenic strain
  • Too many strains causing disease etc.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
265
Q

Recombinant proteins are a novel approach in creating vaccines. How are they created?

A

Genetically engineered and produced from bacteria, yeast, insect or mammalian cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
266
Q

What are the advantages and disadvantages of recombinant protein vaccines

A

Advantages:
Avoid the problem of having to grow pathogen in vitro
Disadvantages:
Major difficulties are finding a protein or proteins that are protective and generate a strong enough immune response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
267
Q

Give examples of recombinant proteins

A

Hepatitis B Surface Antigen
HPV vaccines Cervarix and Gardasil (HPV16 and 18)
SARS-Co-V2 – Novavax (recombinant spike protein produced by moth cells in culture, these are purified, assembled into a synthetic nanoparticle for injection – 14 spike proteins per particle)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
268
Q

What are synthetic peptides

A

Peptides synthesized directly using a machine - avoids the need for pathogen growth

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
269
Q

Problems with synthetic peptides.

A

Difficulties

  • Identifying protective epitopes
  • Inducing a strong response
  • Inducing a broad response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
270
Q

What are live attenuated vectors – viral vector

A

Composed of a safe living attenuated viruses that have inserted genes encoding foreign antigens, which are displayed to the immune system.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
271
Q

How do viral vectors work?

A

When injected the virus is taken up by an APC, the viral DNA is released and enters the nucleus. The viral DNA is transcribed to mRNA and translated to a protein – some of this spike protein is presented with MHC II on the cell surface as a foreign antigen. This is recognised by T-cells and initiates an immune response to the spike protein.
The virus is replication deficient and can not replicate in human cells limiting its spread.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
272
Q

What are DNA vaccines?

A

A mammalian plasmid containing DNA that encodes for the foreign protein of interest is injected directly.
This requires a lipid nanocarrier to get the DNA into a human cell. The DNA goes to the nucleus, gets transcribed and the foreign protein expressed with MHC to stimulate the immune response.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
273
Q

Advantages and disadvantages of DNA vaccines.

A
  • Avoid the need to grow the pathogen, viral vector
  • No live organism involved
  • DNA is cheap to produce
  • Problem is often poor immunogenicity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
274
Q

What are mRNA vaccines

A

mRNA of the target foreign protein is synthesized in vitro. It is complexed with lipid nanoparticles that stabalise and protect the mRNA from degradation and allow the mRNA to cross the plasma membrane.

The mRNA is translated in the cytoplasm and the protein presented on the surface of the cell with MHC
Stimulating the immune response.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
275
Q

Advantages of mRNA vaccines

A
  • Avoid the need to grow the pathogen, viral vector
  • No live organism involved
  • mRNA is relatively cheap to produce
  • Quick to make new variations of vaccine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
276
Q

Features of T-Independent Antigens

A

Bacterial capsular polysaccharides cannot be processed and presented on MHC class II
No T cell help
Antibody response of low magnitude
Low affinity
Predominantly IgM
Little or no boosting on secondary exposure
Infants respond especially poorly and are major target group for these vaccines
Haemophilus influenzae
Neisseria meningitidis
Streptococcus pneumoniae

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
277
Q

Examples of conjugate vaccines.

A

Neisseria meningitidis type C
polysaccharide-protein conjugate vaccine

Streptococcus pneumoniae
23-valent polysaccharide vaccine or 7-valent conjugate

Haemophilus influenzae type B (HiB)-Polysaccharide protein conjugate vaccine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
278
Q

What are recent changes to the vaccination programme in children?

A

The addition of a pneumococcal conjugate vaccine (PCV) at 2, 4 and 13 months of age;

A dose of MenC vaccine at 3 and 4 months;

A booster dose of Hib and MenC vaccine (given as a combined Hib/MenC vaccine) at 12 months of age.

HPV vaccine for teenage girls and boys soon

BCG no longer routinely given to teenagers. Targeted on at risk infants.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
279
Q

Why do people not trust vaccines?

A

Misinformation and hearsay on the internet/social media that are not based on any science

Religion – its un-natural

Studies linking immunisation with contraction of other diseases/conditions
Japan – HPV vaccine with neurological disorders
UK – MMR and autism

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
280
Q

Describe innate immunity

A
  • 1st line of defence
  • Instinctive
  • Non-specific does not depend on lymphocytes
  • Present from birth
  • Slow response
  • No memory
  • Provides barrier to antigen
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
281
Q

Describe adaptive immunity.

A

Specific ‘Acquired/learned’ immunity, requires lymphocytes, antibodies.
Response specific to antigen
Learnt behaviour
Memory to specific antigen
Quicker response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
282
Q

What are both innate and adaptive immunity made up of?

A

Cells and soluble factors (humoral)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
283
Q

What is haematopoiesis?

A

The commitment and differentiation processes that leads to the formation of all blood cells from pluripotent haematopoietic stem cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
284
Q

Where does haematopoiesis take place?

A

Bone marrow (10^11 new cells each day)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
285
Q

How do the pluripotent haematopoietic stem cells decide what to differentiate into?

A

Stem cells in the bone marrow are stimulated by colony stimulating factors (soluble proteins) that bind to receptors on the stem cell.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
286
Q

What 2 are the two pathways the pluripotent haematopoietic stem cells can split into?

A
  1. Myeloid pathway
  2. Lymphoid pathway
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
287
Q

Which 3 types of cells are polymorphonuclear leukocytes?

A
  • Neutrophils (70% of wbcs in body)
  • Eosinophils
  • Basophils
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
288
Q

Which 3 types of cells are mononuclear leukocytes?

A
  • Monocytes: differentiates into macrophages when entering tissue.
  • T-cells
  • B-cells: differentiates into plasma cells which produce antibodies.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
289
Q

What process are mast cells important in?

A

Allergic reactions - have histamines

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
290
Q

What are natural killer cells important for? (Type of T cell)

A

Anti tumour responses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
291
Q

What are dendritic cells important for?

A

Immune surveillance

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
292
Q

Which 3 soluble factors make up the immune system?

A
  • Complement
  • Antibodies
  • Cytokines and chemokines
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
293
Q

Define complement.

A

Group of ~20 serum proteins secreted by the liver that need to be activated to be functional.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
294
Q

What are the 3 modes of actions of complement?

A
  1. Direct lysis
  2. Attract more leukocytes to site of infection
  3. Coat invading organisms
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
295
Q

What are antibodies?

A

Antibodies (also called immunoglobulins) are soluble glycoproteins that recognize and bind antigens, specifically.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
296
Q

Antibodies (Ab) can also be called immunoglobulins (Ig) What are the 3 features of immunoglobulins?

A
  • soluble
  • secreted
  • bound to B cells as part of B-cell antigen receptor
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
297
Q

What are the 5 distinctive classes of Ig?

A

IgG (IgG1-4)
IgA (IgA1 & 2)
IgM
IgD
IgE

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
298
Q

Describe the basic structure of an antibody (IgG).

A

Antibodies have a basic structure of four polypeptide chains – two identical light chains and two identical heavy chains. Each chain contains a variable region (fab region).

The chains are held together by disulphide bonds.

The C-terminal (Fc) regions of the light and heavy chains form the constant regions which determine the effector functions of an antibody.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
299
Q

In an antibody what does the Fc region and Fab region bind to?

A

Fc region binds to receptors on the surface of wbcs.
Fab region bind to non-self elements, they are very specific.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
300
Q

Which is the most predominant Ig in the human serum?

A

IgG: accounts for 70 - 75% of Igs in human serum

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
301
Q

List 5 features of IgM.

A
  • Accounts for 10% of Igs in serum.
  • Pentamer, formation requires J chain.
  • Mainly found in blood – too big to cross endothelium.
  • Mainly primary immune response, initial contact with Ag.
    (First responder during immune response)
  • The monomeric form (mIgM) is present as an antigen-specific receptor on B cells - which is important for the adaptive immune response.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
302
Q

List 3 features of IgA

A
  • Accounts for 15% of Igs in serum
  • In humans 80% of serum IgA is as a monomer (in most animals serum IgA is a dimer)
  • The predominant Ig in mucous secretions such as saliva, colostrum, milk, bronchiolar & genitourinary secretions
    Called Secretory IgA (sIgA)
    sIgA is held together with a J chain and a secretory component
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
303
Q

List 2 features of IgD.

A

Accounts for 1% of Ig in serum

A transmembrane monomeric form (mIgD) is present on mature B cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
304
Q

List 5 features of IgE

A

Accounts for only ~0.05% of Ig in serum

Basophils and Mast Cells express an IgE-specific receptor that has high affinity for IgE

Basophils and Mast Cells are continually saturated with IgE

Binding Ag triggers release of histamine by these cells

Associated with hypersensitivity allergic response and defence against parasitic infections

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
305
Q

What is an epitope?

A

Each antibody binds to a restricted part of the antigen called an epitope.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
306
Q

What are the functions of an antibody?

A

Antibodies acts as an adaptor that links a microbe to a phagocyte.
1. Neutralise toxins.
2. Opsonisation.
3. Activate classical complement system.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
307
Q

What are cytokines?

A

Proteins secreted by immune and non-immune cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
308
Q

Name 4 types of cytokines.

A
  1. Interferons.
  2. Interleukins.
  3. Colony stimulating factors.
  4. Tumour necrosis factors.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
309
Q

Cytokines: Function of interferons (IFN)

A

Induce a state of antiviral resistance in uninfected cells & limit the spread of viral infection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
310
Q

Cytokines: Function of interleukins (IL)

A
  • Can be pro-inflammatory (IL1) or anti-inflammatory (IL-10)
  • Can cause cells to divide, to differentiate and to secrete factors
  • Produced by many cells, over 30 types
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
311
Q

Cytokines: Function of Colony Stimulating Factors (CSF)

A
  • Involved in directing the division and differentiation on bone marrow stem cells – precursors of leukocytes.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
312
Q

Cytokines: Function of Tumour Necrosis Factors (TNFa & b)

A

Proinflammatory molecules. Mediate inflammation and cytotoxic reactions.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
313
Q

What are chemokines?

A

Chemotactic cytokines

Group of approx 40 proteins that attract leukocytes to sites of infection from the bloodstream into the tissues or lymph organs by binding to specific receptors on cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
314
Q

What are the 4 subclasses of chemokines?

A

CXCL – mainly bind to neutrophils

CCL – monocytes, lymphocytes, eosinophils, basophils

CX3CL – mainly T lymphocytes & NK Cells

XCL – mainly T lymphocytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
315
Q

What is innate immunity composed of?

A
  • Physical and chemical barriers
  • Phagocytic cells (neutrophils and macrophages)
  • Blood proteins (complement, acute phase)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
316
Q

Give examples of physical and chemical barriers used in innate immunity?

A
  • Lysozymes in tears
  • Skin
  • Low pH and commensals of vagina
  • Mucociliary escalator
  • Gastric acid
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
317
Q

Sometimes physical barriers are breached leading to tissue damage (trauma) or infection. This will induce an inflammatory response. What response is the body aiming for during inflammation?

A
  • Coagulation: stop bleeding.
  • Acute inflammation (leukocyte recruitment).
  • Kill pathogens, neutralise toxins, limit pathogen spread.
  • Phagocytosis: clear pathogens/dead cells.
  • Proliferation of cells to repair damage.
  • Remove blood clot – remodel extracellular matrix to maintain homeostasis.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
318
Q

What are the 3 hallmarks of inflammation?

A
  1. Increased blood supply
  2. Increased vascular permeability
  3. Increased leukocyte transendothelial migration ‘extravasation’
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
319
Q

Name 3 pattern recognition receptors (PRR) that can sense microbes?

A
  1. Toll-like receptors (TLR).
  2. Lectin receptors.
  3. Scavenger receptors.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
320
Q

What kind of immunity are PRR’s and PAMP’s associated with?

A

Innate immunity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
321
Q

What are PRR’s a receptor for?

A

PAMP’s (on microbes)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
322
Q

What happens when a PAMP binds to a PRR?

A

The innate immune response and inflammatory response is triggered.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
323
Q

What are C-Type lectin receptors?

A

Expressed by Macrophages and DC, can recognise foreign carbohydrates.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
324
Q

What are scavenger receptors?

A

They are a very large family of receptors - They mainly recognise foreign lipids on bacteria. (ie LPS in Gram -ve bacteria)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
325
Q

What are toll-like receptors?

A

TLRs recognise Pathogen-Associated Molecular Patterns expressed by microbes. Different TLRs activate different signalling cascades depending on the pathogen being detected.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
326
Q

TLRs and their targets

A

TLR2: Gram positive bacteria and mycobacteria (inc fungi)
TLR4: Gram negative bacteria and lipopolysaccharides
TLR5: Bacteria and flagellin
TLR7: Single-strand RNA
TLR9: Non-methylated DNA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
327
Q

What are the 3 different activation pathways that make up the complement system? How are they activated?

A
  1. Classical: Antibody bound to microbe.
  2. Lectin: Mannose binding lectin bound to microbe.
  3. Alternative: Complement bound to microbe.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
328
Q

Which complement plasma proteins are chemoattractants that recruit leukocytes to the site of infection.

A

C3a and C5a.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
329
Q

Which complement plasma proteins have opsonic properties (insert itself into the membrane of bacterium “coating it” to increase its susceptibility to ingestion by phagocytosis) when bound to a pathogen?

A

C3b, C4b.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
330
Q

Which complement plasma proteins make up the membrane attack complex (MAC) and are responsible for the cytolytic function?

A

C5b-9

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
331
Q

What is extravasation?

A

Leukocyte (WBC) migration across the endothelium.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
332
Q

What do macrophages at the tissues secrete to initiate extravasation?

A

TNF alpha.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
333
Q

Describe the process of extravasation.

A
  1. Macrophages at tissues release TNF alpha.
  2. The endothelium is stimulated to express adhesion molecules and to stimulate chemokines.
  3. Neutrophils bind to adhesion molecules; they roll, slow down and become stuck to the endothelium.
  4. Neutrophils are activated by chemokines.
  5. Neutrophils transmigrate through the endothelium to the tissue to help fight infection.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
334
Q

Describe the process of phagocytosis?

A
  1. Pathogen binds to neutrophil/macrophage.
  2. Engulfment of pathogen.
  3. Phagosome formation.
  4. Lysosome fusion - phagolysosome.
    Pathogen is destroyed.
335
Q

What are the two killing pathways present in neutrophils and macrophages?

A
  • O2 dependent
  • O2 independent
336
Q

What is the role of NO in killing pathogens in O2 dependent killing?

A

NO leads to vasodilation and increased extravasation. This means more neutrophils etc pass into the tissues to destroy pathogens.

337
Q

Why can superoxides be used to destroy pathogens in O2 dependent killing?

A

Superoxides can be converted to H2O2 and then to hydroxyl free radicals. Hydroxyl free radicals are highly reactive and can destroy pathogens.

338
Q

What mechanisms or cells are involved in O2 independent killing?

A

Enzymes: Lysozymes
Proteins: Defensins, TNF
pH

339
Q

What are the advantages adaptive immunity have over innate immunity?

A
  • Microbes can evade innate immunity
  • Intracellular viruses and bacteria ‘hide’ from innate immunity
  • Adaptive immunity have memory to specific antigen – ‘seen it before so faster response’
340
Q

What are the two types of adaptive immunity?

A
  • Cell Mediated - T cells - intracellular microbes
  • Humoral (Ab) - B cells - extracellular microbes
341
Q

In which primary lymphoid tissue do T cells mature?

A

Thymus

342
Q

In which primary lymphoid tissue do B cells mature?

A

Bone marrow.

343
Q

Give examples of secondary lymphoid tissue.

A

Spleen, lymph nodes, mucosa associated lymphoid tissue - MALT.

344
Q

In which primary lymphoid tissue do antigen present cells mature?

A

Bone marrow

345
Q

Which cells are involved in cell-mediated immunity?

A
  1. Antigen Presenting Cells (APC)
    - Macrophages
    - Dendritic Cell
    - B cells
  2. T cells
346
Q

What do T lymphocytes respond to?

A

T lymphocytes only respond to intracellular presented antigens, they do NOT respond to soluble antigens.

347
Q

What is T cell selection?

A

T cell selection refers to how T cells that recognise self are killed in the foetal thymus as they mature

348
Q

T cell receptors only recognise foreign antigens under what conditions?

A

T Cell Receptor (TCR) recognises foreign antigens in association with Major Histocompatibility Complex (MHC)

349
Q

What do Major Histocompatibility Complexes MHCs do?

A

Major Histocompatibility Complex (MHC) display peptides from self OR non-self proteins

350
Q

Whare the 2 types of MHC?

A

MHC I and MHC II

351
Q

Which cells express MHC1?

A

All nucleated cells express MHC1. e.g. a virus infected or cancer cell would express MHC1.

352
Q

Which cells express MHC2?

A

Antigen presenting cells ONLY e.g. macrophages, B cells, dendritic cells.

353
Q

Which MHC would an intracellular antigen (endogenous) lead to the expression of?

A

MHC1.

354
Q

Which MHC would an extracellular antigen (exogenous) lead to the expression of?

A

MHC2.

355
Q

What type of T cell binds to MHC1?

A

Cytotoxic T cells (CD8).

356
Q

What type of T cells binds to MHC2?

A

Helper T cells (CD4).

357
Q

Intracellular signalling happens when a helper T cell is bound to a T cell receptor. Which interleukin is secreted?

A

IL-2. This then binds to an IL-2 receptor on the T cell and produces a positive feedback mechanism (clonal expansion) leading to division, differentiation, effector functions, and memory.

358
Q

What happens when naive T cells become activated?

A

Naive cell -> CD4 and CD 8

Any infected cell presents Ag with MHC I to a CD8 T cell
CD 8: Kills intracellular pathogens directly (cytotoxic T cells)

APC presents Ag with MHC II to a naïve CD4 T cell.
If microenvironment has a lot of IL-12, it forms TH1 -> secretes IFNγ and IL-2 -> helps kill intracellular pathogens

If no IL-12, it forms TH2 -> secretes IL-4,5,10, Antibody production

359
Q

What is the function of T helper 1 (CD4)?

A

It helps the immune response against intracellular pathogens. Secretes cytokines.

360
Q

What is the function of T helper 2 (CD4)?

A

It helps produce antibodies against extracellular pathogens. Secretes cytokines.

361
Q

What is the function of Cytotoxic T cell (CD8)?

A

It can kill cells directly by binding to antigens; they can form proteolytic granules & releases perforins and granulysin they also induce apoptosis.

362
Q

How many antibodies can each B cell make?

A

Each B cell can only make 1 antibody. This 1 antibody can only bind to 1 epitope.

363
Q

What do B cells express?

A

B cells express membrane bound Ig (IgM or IgD monomer)

364
Q

What happens to B cells that recognise ‘self’?

A

They are killed in bone marrow.

365
Q

B cells can function as an APC. Describe the process of a T helper cell binding to a B cell.

A

A B-cell antibody binds an antigen -> phagocytosis -> epitope is displayed on the surface of the B-cell bound to an MHC2 -> TH2 binds to B-cells -> cytokine secretion induces B-cell clonal expansion -> differentiation into plasma cells and memory B cells.

366
Q

Give examples of PAMPs in bacteria, virus, and parasites.

A

Bacteria:
- Peptidoglycan (PGN)
- Lipoproteins
- DNA
- Flagellin
- Lipopolysaccharide (LPS)

Virus:
- Coat protein
- Nucleic acid

Parasite:
- GPI anchor

367
Q

What are Damage associated molecular patterns (DAMPs)

A

Endogenous molecules created to alert the host to tissue injury and initiate repair.

368
Q

Why do we not recognise our own DNA and RNA and mount an innate response?

A

Our DNA and RNA is normally located where our pattern recognition receptors (PRRs) cannot access it

369
Q

TLRs and their targets

A

TLR2: Gram positive bacteria and mycobacteria (inc fungi)
TLR4: Gram negative bacteria and lipopolysaccharides
TLR5: Bacteria and flagellin
TLR7: Single-strand RNA
TLR9: Non-methylated DNA

370
Q

What is the main function of TLR’s?

A

TLR’s send signals to the nucleus to secrete cytokines and interferons. These signals initiate tissue repair. Enhanced TLR signalling = improved immune response.

371
Q

Membrane bound PRR: Where could mannose receptors be found?

A

on macrophages (fungi)

372
Q

What is the main function of RLR’s?

A

RLR’s detect intracellular double stranded RNA. This triggers interferon production and so an antiviral response.

373
Q

Name 3 receptors that make up the PRR family.

A

Toll-like receptors (TLR).
Nod-like receptors (NLR).
Rig-like receptors (RLR).

374
Q

What is the main function of NLR’s?

A

NLR’s detect intracellular microbial pathogens. They release cytokines and can cause apoptosis if the cell is infected.

375
Q

What is the damage chain reaction?

A

Harmful stimulus leads to tissue damage, this causes the release of DAMPs and release TLRs and proinflammatory cytokine production. Inflammation will lead to tissue damage and lead to chain reaction of DAMP release.

376
Q

What high levels of DAMPs associated with?

A

Many inflammatory and autoimmune diseases as well as atherosclerosis and cancer.

377
Q

What is the cytokine storm?

A
  • Profound increase in cytokines, chemokines & interferons
  • Causes severe inflammation and tissue damage
  • Role in sepsis, influenza, severe COVID-19 pathogenesis, etc.
  • Induction due to:
    1. genetic makeup of the host
    2. persistence of the pathogen (evasion mechanisms)
378
Q

What are the 2 main strategies of immunomodulation?

A
  1. Agonists (enhance TLR signalling):
    - adjuvant effect (used with a primary treatment) promoting protective responses e.g. vaccines
    - immune stimulators
    - Modify adaptive immune response: bias Th and Treg responses
    - side-effects: potentially enhance inflammation
  2. Antagonists (inhibit TLR signalling):
    - block binding of ligands or protein-ligand complexes to receptors / interfere with intracellular adaptor molecules of common signalling pathways
    - sepsis syndromes, inflammation, arthritis, etc
    - side-effects: potentially allow pathogen outgrowth, mutation, etc
379
Q

What is type I hypersensitivity?

A

Type I hypersensitivity is an immediate reaction to environmental antigens mediated via IgE

380
Q

What is atopy?

A

Atopy is an inherited trait for Type 1 hypersensitivity

381
Q

What are allergens?

A

Allergens are antigens that trigger allergic reactions

382
Q

What are the main steps in a Type I hypersensitivity reaction?

A
  1. Sensitisation
  2. Mast cell degranulation
  3. Early phase response
  4. Late phase response
383
Q

Define sensitisation

A

Sensitisation is the term to describe the initial event that lead to the specific IgE being developed for that allergen.

384
Q

What occurs during sensitisation?

A
  1. Body is exposed to allergens.
  2. Allergens are taken up by APCs, processed and expressed by MHC II where they are presented to naive T cells.
  3. Naive T cells differentiate into Th2
  4. Th2 cells produce IL-4 and IL-13 which activate B cells and induce them to class switch to produce IgE.
  5. IgE can circulate in blood and bind to mast cells and basophils in various tissues in our body.
385
Q

What are the differences in structure between IgG and IgE?

A

IgE has 4 constant domains, while IgG only has 3 constant domains.
IgG has a hinge region.
IgE is more heavily glycosylated.

386
Q

What are the 3 main body surfaces that come into contact with allergens?

A
  • Skin
  • GI tract
  • Lung
387
Q

What happens during mast cell degranulation?

A
  1. Upon subsequent re-exposure to the allergen, the antigens can bind to the IgE on the surface of the mast cells.
  2. Cross linkage of the receptors can occur through the IgEs that are bound to them.
  3. This leads to mast cell degranulation and stimulates the synthesis of new cytokines.
388
Q

What happens during early phase response?

A
  1. Early phase mediators such as histamine are released.
  2. Histamine causes vasodilation, increased vascular permeability and broncho-constriction, causing symptoms of allergy (itch, flushing, rash, angioedema and wheeze).
389
Q

Which receptor of histamine is mainly responsible for mediating allergic reactions?

A

H1 receptor

390
Q

Clinical use of tryptase during type 1 hypersensitivity?

A

Diagnostic purposes, tryptase levels spike following allergic reaction.

391
Q

List the clinical manifestations of allergy?

A
  • Itchiness
  • Runny nose
  • Hives
  • Sneezing
  • Red/watery eyes
  • Trouble breathing
392
Q

Systemic type 1 hypersensitivity reaction would lead to what clinical syndrome?

A

Anaphylaxis

393
Q

Type 1 hypersensitivity reaction to airways would lead to what clinical syndrome?

A
  • Asthma (lower airways)
  • Rhinitis (upper airways)
394
Q

Type 1 hypersensitivity reaction to eyes would lead to what clinical syndrome?

A

Allergic conjunctivitis

395
Q

Type 1 hypersensitivity reaction to skin would lead to what clinical syndrome?

A
  • Urticaria
  • Angioedema
  • Atopic eczema
396
Q

What happens during late phase response?

A

Recruitment of other cells would lead to a second response, prolonging the inflammatory response.

397
Q

What are the three conditions that elicit a type I hypersensitivity reaction?

A
  1. Allergen characteristics
  2. Host factors
  3. Environmental influences
398
Q

What makes certain proteins allergenic (induce a strong IgE response)?

A
  • Protease activity: Der p 1 (found in dust mites)
  • Surface features of protein: Ves v 5 (found in wasps)
  • Glycosylation pattern of protein: Ara h1 (found in peanuts)
  • Host factors: polymorphisms of various receptors that make them gene targets
  • Environmental influences
399
Q

Define pharmacokinetics.

A

The fate of a chemical substance administered to a living organism
- What the body does to the drug

400
Q

When describing pharmacokinetics, ADME is used, what does it stand for?

A

Absorption route
Distribution (how well distributed)
Metabolism (i.e IV skips 1st pass metabolism)
Excretion (hepatic or renal)

401
Q

Pharmacokinetics: What is absorption?

A

Transfer of a drug molecule from site of administration to systemic circulation

402
Q

Which routes of administration have 100% of the dose reach systemic circulation?

A

IV and IA administration. For any other route, drugs must cross at least one membrane to reach systemic circulation.

403
Q

What are the three mechanisms for drugs to permeate membranes

A

Passive diffusion through hydrophobic membrane
Lipid soluble molecules
Passive diffusion aqueous pores
Very small water soluble drugs (eg lithium)
Most drug molecules are too big
Carrier mediated transport
Proteins which transport sugars, amino acids, neurotransmitters and trace metals (and some drugs)

404
Q

What 2 factors affecting drug absorption?

A

Lipid solubility
Drug ionisation

405
Q

What factors affect oral drug absorption?

A
  • Drug ionisation.
  • Stomach
  • Intestine
    -First pass metabolism
406
Q

In terms of ionisation, what happens to Aspirin in the stomach?

A

Aspirin is a weak acid and so becomes less ionised in the stomach due to the low gastric pH.

407
Q

What is the advantage of aspirin becoming less ionised in the stomach?

A

This allows rapid non-ionic diffusion across the gut membrane into the plasma. Once in the plasma aspirin becomes more ionised again.

408
Q

Where are weak acid drugs and weak base drugs best absorbed?

A

Weak acids - best absorbed in stomach. Weak bases - best absorbed in intestine

409
Q

How could the stomach affect oral drug absorption?

A
  • Gastric enzymes - drug molecule may be digested (peptides, proteins)
    Eg. insulin and biologicals
  • Low pH - molecule may be degraded (benzylpenicillin)
  • Food (full stomach will generally slow absorption)
  • Gastric motility (altered by drugs and disease state)
  • Previous surgery (eg gastrectomy)
410
Q

How could the intestine affect oral drug absorption?

A
  1. Drug structure
    - Lipid soluble/unionised molecules diffuse down concentration gradient
    - Large or hydrophilic molecules are poorly absorbed
  2. Medicine formulation
    - Capsule/tablet coating can control time between administration and drug release
    - Modified release controls (slows) the rate of absorption (less frequent dosing)
  3. P-glycoprotein
    - Substrates are removed from intestinal endothelial cells back into lumen
411
Q

What is first pass metabolism?

A

Metabolism of drugs preventing them reaching systemic circulation.

412
Q

List examples of first pass metabolism.

A

Degradation by enzymes in intestinal wall
Absorption from intestine into hepatic portal vein and metabolism via liver enzymes

413
Q

How can first pass metabolism be avoided?

A

By giving via routes that avoid sphlanchnic circulation (eg rectal)

414
Q

What is bioavailability (F)?

A

Proportion of administered dose which reaches the systemic circulation

415
Q

Features of bioavailability (F).

A
  • Dependent on extent of drug absorption and extent of first pass metabolism
  • Not affected by rate of absorption
  • Varies with route of administration
  • Variation between individuals (genetic, disease states)
    Expressed as % or fraction (e.g 70% or 0.7)
416
Q

Pros and cons of rectal administration (PR).

A

Pros:
Local administration
Avoids first pass metabolism
Nausea and vomiting

Cons:
Absorption can be variable
Patient preference

417
Q

Pros and cons of inhaled administration (Inh)

A

Pros:
Well perfused large surface area
Local administration

Cons:
Inhaler technique can limit effectiveness

418
Q

Pros and cons of Sub- cutaneous administration (S/C)

A

Pros:
Faster onset than PO
Formulation can be changed to control rate of absorption

Cons:
Not as rapid as IV

419
Q

Pros and cons of Trans- dermal administration (TD)

A

Pros:
Provides continuous drug release
Avoids first pass metabolism

Cons:
Only suitable for lipid soluble drugs
Slow onset of action

420
Q

Pharmacokinetics: What is distribution?

A

Drug properties will influence ability to move between compartments.

421
Q

What factors can affect distribution of drugs?

A
  • Molecule size
  • Lipid solubility
  • Protein binding
422
Q

What is the volume of distribution (Vd)?

A

Theoretical volume a drug will be distributed in the body (apparent volume of distribution)

423
Q

Features of Vd.

A

Volume of plasma required to contain the total administered dose
Drugs that are well distributed will have high Vd
Drugs that are poorly distributed will have low Vd

424
Q

Digoxin is a small molecule, highly lipophyllic, would it be likely to have a small Vd or large Vd?

A
  • Small molecule size and lipophilic so would have a large Vd

Vd = 5-8L/kg

425
Q

What is the blood brain barrier (BBB)?

A

Membrane that separates foreign substances in the blood from the CNS

426
Q

What features of the BBB poses a challenge for treating CNS conditions?

A
  • Continuous layer of endothelial cells with tight junctions
  • Efflux pumps remove water soluble molecules
    This allows BBB to maintain a stable environment and protects the brain, but poses a challenge for treating CNS conditions
427
Q

Ways for drugs to reach the CNS despite the presence of the BBB.

A
  • High lipid solubility. e.g. Ѱ drugs usually very lipid soluble (therefore large Vd)
  • Intrathecal administration (e.g. baclofen in MS and spinal cord injury, chemotherapy)
  • Inflammation (causes barrier to become leaky)
428
Q

Pharmacokinetics: What is drug elimination?

A

the process by which the drug becomes no longer available to exert its effect on the bod

429
Q

What are the two routes of drug elimination in pharmacokinetics?

A

Metabolism: modification of chemical structure to form new chemical entity (metabolite)

Excretion of unchanged drug (hydrophillic, polar molecules)

430
Q

What are the 2 phases of metabolism in drug elimination? Describe them briefly

A

Phase 1: Oxidation/reduction/hydrolysis to introduce reactive group to chemical structure. Cytochrome P450 enzymes are responsible for majority of phase 1.

Phase 2: Conjugation of functional group to produce hydrophilic, inert molecule which can then be excreted

431
Q

What are the three processes for renal excretion of drugs?

A
  1. Glomerular filtration
    - 20% of plasma filtered
    - Free/unbound drug molecules
    - Very large molecules excluded
  2. Active tubular secretion
    - 80% of renal blood flow passes on to peritubular capillaries
    - Can clear protein bound drugs
    - Most effective renal clearance mechanism
  3. Passive reabsorption
    - Diffusion down the concentration gradient from tubule into peritubular capillaries
    - Hydrophobic drugs will diffuse easily
    - Highly polar drugs will be excreted
432
Q

A 66 year old woman is admitted to the medical ward after a fall. She is a heavy drinker and is showing signs of alcohol withdrawal. She is prescribed lorazepam to control her withdrawal symptoms. What property allows it to cross the blood brain barrier?

A) substrate for CYP3A4
B) Avoids first pass metabolism
C) Highly protein bound
D) Large molecule
E) Highly lipid soluble

A

E) Highly lipid soluble

433
Q

List 6 examples of how ageing impacts the immune response in the molecular level.

A
  • Genomic instability
  • Telomere shortening
  • Epigenetic alteration
  • Loss of proteostasis
  • Compromised autophagy ( body’s cellular recycling system)
  • Mitochondrial dysfunction
434
Q

List 3 examples of how ageing impacts the immune response in the cellular level.

A
  • Cellular senescence
  • Stem cell exhaustion
  • Altered intercellular communication
435
Q

List an example of how ageing impacts the immune response in the systemic level.

A

Nutritional dysregulation

436
Q

What is immunosenescence?

A

The state of dysregulated immune function that contributes to the increased susceptibility of the elderly to infection and possibly to autoimmune disease and cancer.

437
Q

Interventions to reduce immunosenescence in the elderly.

A

Modifications to lifestyle:
- Physical activity
- Caloric restriction
- Maintaining optimal nutrition

Pharmacological interventions:
- Statins

438
Q

What is meant by ‘first order kinetics’?

A

First order: Rate of elimination is proportional to the plasma drug concentration (processes involved in elimination do not become saturated)

A constant % of the plasma drug is eliminated over a unit of time.

439
Q

What is meant by ‘zero order kinetics’?

A

Zero order: Rate of elimination is NOT proportional to the plasma drug concentration (metabolism processes become saturated)

A constant amount of the plasma drug is eliminated over a unit of time

440
Q

What is clearance (CL)?

A

Efficiency of irreversible elimination of a drug from the systemic circulation

441
Q

Single modified release oral dose vs single oral dose.

A

Modified release formulations usually require less frequent dosing.

442
Q

Features of Half life (t1/2)

A
  • Dependent on clearance (CL) of drug from body by all eliminating organs (hepatic, renal, faeces, breath)
  • Dependent of volume of distribution (Vd) - A drug with large Vd will be cleared more slowly than a drug with a small Vd
  • Not dependent on drug dose or drug formulation
  • A drug will be 97% cleared from the body after 5 x half lives (considered ‘cleared’ in clinical practice)
443
Q

What are single or STAT doses useful to treat?

A

Acute conditions, effects will usually wear off after a few minutes - hours

444
Q

When is continuous IV infusions used?

A

Critical care patients
Antibiotics
Unfractionated heparin
General anaesthetics

445
Q

Pharmacokinetics: What is the steady state?

A

When the rate of drug input is equal to rate of drug elimination

446
Q

Why is the steady state important?

A

Aim for Css which lies between the Maximum safe concentration (MSC) and minimum effective concentration (MEC)

447
Q

Waiting to reach the steady state may be detrimental. What can we do to speed up the time to steady state?

A

Using a loading dose.

448
Q

Give 2 examples of zero order drugs

A

Ethanol, phenytoin

449
Q

Pharmacokinetics: What is tmax and cmax

A

Cmax= maximum plasma concentration

tmax= time taken to reach Cmax

450
Q

A 66 year old woman is admitted to ED with SOB and haemoptysis. CTPA confirms bilateral PEs. She is prescribed an IV loading dose of unfractionated heparin 5000 units followed by an IV continuous infusion.

What effect will the loading dose have on the heparin pharmacokinetics?

A

Reduce time to steady state plasma concentration (Css)

451
Q

Define pharmacogenetics

A

The use of genetic and genomic information to tailor pharmaceutical treatment to an individual.

452
Q

Benefits of medicine use through genomics.

A

improved patient outcomes, cost efficiency

453
Q

Most drug targets are proteins. List 4 drug targets.

A
  • Enzymes
  • Receptors
  • Transporters
  • Ion channels
454
Q

What is a receptor?

A

A component of a cell that interacts with a specific ligand* and initiates a change of biochemical events leading to the ligands observed effects.

455
Q

Receptors are the principal means by which chemicals communicate. List three chemicals receptors can communicate with.

A
  • Neurotransmitters: acetylcholine, serotonin
  • Autacoids (local) Greek “autos” (self) and “acos” (relief): cytokines, histamine
  • Hormones: testosterone, hydrocortisone
456
Q

List 4 examples of receptors involved in drug reactions.

A

Ligand-gated ion channels
nicotinic ACh receptor

G protein coupled receptors
beta-adrenoceptors

Kinase-linked receptors
receptors for growth factors

Cytosolic/nuclear receptors
steroid receptors

457
Q

Features of G protein coupled receptors

A

GPCRs are the largest and most diverse group of membrane receptors in eukaryotes.

Ligands include light energy, peptides, lipids, sugars, and proteins.

458
Q

How is G coupled protein receptor activity regulated?

A

Their activity is regulated by factors that control their ability to bind to and hydrolyseguanosine triphosphate(GTP) toguanosine diphosphate(GDP)

459
Q

Give an example of a GPCR.

A

Muscarinic and β2 adrenoceptor.

460
Q

What do the majority of GPCRs interact with?

A

GPCR’s usually interact with adenylate cyclase or phospholipase C

461
Q

How do kinase-linked receptors work?

A

Kinasesare enzyme that catalyze the transfer of phosphate groups between proteins - process is known as phosphorylation.
The substrate gains a phosphate group ”donated” by ATP
Transmembrane receptors activated when the binding of an extracellular ligand causes enzymatic activity on the intracellular side.

462
Q

How do steroid hormones/nuclear receptor work?

A

Work by modifying gene transcription

463
Q

Give an example of a drug which targets nuclear receptors.

A

Tamoxifen (Breast Cancer) acts as aselective oestrogen receptor modulator(SERM), or as apartial agonistof theoestrogen receptors
- Used in ER+ cancers.

464
Q

Give examples of how imbalance of chemicals and receptors can lead to pathology.

A

Imbalance of chemicals
allergy; increased histamine
Parkinson’s; reduced dopamine

Imbalance of receptors
myasthenia gravis; loss of ACh receptors
Mastocytosis (Mast cells); increased c-kit receptor

465
Q

What is an agonist and antagonist?

A

Agonist
a compound that binds to a receptor and activates it

Antagonist
a compound that reduces the effect of an agonist

466
Q

What is the two state model of receptor activation?

A

Describes how drugs activate receptors by inducing or supporting a conformational change in the receptor from “off” to “on”.

467
Q

Describe the shape of a log concentration response curve.

A

Sigmoidal

468
Q

What does EC50 tell us about a drug?

A

Its potency!

469
Q

Would a drug with a lower EC50 have a lower or greater potency?

A

Greater potency.

470
Q

Define affinity, efficacy/intrinsic activity (IA) and potency

A

Affinity:
-How well a ligand binds to receptor
Efficacy/IA:
-How well a ligand ACTIVATES a receptor
Potency:
-Relative strength of the drug

(potency is solely comparative, i.e drug A vs B. Efficacy, is what % of bound drugs activate the receptor)

471
Q

Role of antagonist

A

Antagonists reverse the effects of agonists.

472
Q

Difference between competitive and non-competitive antagonism.

A

Competitive antagonists bind to the same site as the agonists, while in non-competitive antagonism, the antagonist binds to an allosteric (non-agonist) site on the receptor to prevent activation of the receptor

473
Q

What are factors governing drug action?

A

Receptor-related
affinity
efficacy

Tissue-related
receptor number
signal amplification

474
Q

Do agonists and antagonists display efficacy and affinity?

A

Agonists
Have affinity and efficacy

Antagonists
Have affinity but zero efficacy

475
Q

What is the effect of fewer receptors on receptor response?

A

Some agonists needs to activate only a small fraction of the existingreceptors to produce the maximal system response. (Partial agonists don’t have receptor reserve).

476
Q

What is inverse agonism?

A

When a drug that binds to the same receptor as anagonistbut induces a pharmacological response opposite to that of theagonist.

(negative activity)

477
Q

What is tolerance?

A

Tolerance (slow)
- reduction in agonist effect over time
- continuously, repeatedly, high concentrations

478
Q

What is desensitization?

A

Desensitization (rapid)
- uncoupled
- internalized
- degraded

479
Q

Use of the terms specificity and selectivity in drug interactions.

A

Selective is a better term to describe activity as no compound is ever truly specific.

480
Q

What are the two types of enzyme inhibitors.

A

Irreversible inhibitors - usually react with the enzyme and change it chemically (e.g. viacovalent bondformation).

Reversible inhibitors - bindnon-covalentlyand different types of inhibition are produced depending on whether these inhibitors bind to theenzyme, the enzyme-substrate complex, or both.

481
Q

Give an example of an enzyme that acts as drug products

A
  • Streptokinase
  • tPA
482
Q

How do statins work?

A

HMG-CoA reductase inhibitors
Block the rate limiting step in the Cholesterol pathway

483
Q

What is the renin-angiotensin-aldosterone system?

A

Renin-angiotensin-aldosterone system is a major blood pressure regulating mechanism. The system increases blood pressure by increasing the amount of salt and water the body retains

484
Q

How does inhibition of angiotensin converting enzyme help lower blood pressure?

A

Inhibiting ACE reduces ATII production and therefore causes a reduction in blood pressure

485
Q

Angiotensin converting enzyme can be hijacked. Give an example of when this could happen.

A

SARS-CoV-2 uses ACE2 as an entry receptor

486
Q

Enzymes can target multiple steps in biosynthetic pathways such as in Parkinson’s disease.

Treatment is through multiple enzymatic inhibitor drugs. This includes: L-DOPA, Peripheral DDC Inhibitor, Peripheral COMT Inhibitor, Central COMT Inhibitors, Mono Amine Oxidase B Inhibitors.

How much each of these drugs help the management of Parkinson’s,

A

L-DOPA is a precursor for neurotransmitter biosynthesis.

Peripheral DDC Inhibitor blocks DDC in the periphery generating more for the CNS pathway

Peripheral COMT Inhibitor prevents breakdown of L-DOPA generating more for the CNS pathway

Central COMT Inhibitors function within the CNS to keep Dopamine levels up

Mono Amine Oxidase B Inhibitors prevents Dopamine breakdown and increases availability

487
Q

What non enzymatic inhibitor drug can be used to help management Parkinson’s disease?

A

Central Dopamine Receptor Agonists

Antagonise dopamine receptors

488
Q

How and where do loop diuretics work?

A

They act on the ascending loop of henle and inhibit NKCC2 channels, this stops Na, K and Cl reabsorption.

489
Q

Name a loop diuretic.

A

Furosemide - acts on Na+/K+/2Cl- transporter (NKCC2).

490
Q

Name 3 TNF blockers.

A
  1. Etanercept (TNF alpha specific).2. Infliximab.3. Adalimumab.
491
Q

List 2 naturally occurring opioids.

A
  • Morphine
  • Codeine
492
Q

List 3 simple chemically modified opioids

A

diamorphine (heroin), oxycodone, dihydrocodeine

493
Q

List 4 synthetic opioids.

A
  • Pethidine
  • Fentanyl
  • Alfentanil
  • Remifentanil
494
Q

10 mg of morphine orally is equal to how many mg in s/c, IM, IV?

A

5mg! 50% of oral morphine is metabolised by first pass metabolism.

495
Q

Which route of administration is the quickest between sub-cutaneous, IM and IV?

A

IV>IM>Sub-cutaneous

496
Q

What are the 4 opioid receptors?

A

MOP KOP DOP and NOP

497
Q

What are the relative potencies of diamorphine morphine and pethidine?
(5mg of diamorphine)

A

Diamorphine 5 mg
Morphine 10 mg
Pethidine 100 mg

498
Q

What do opioids act on?

A
  • CNS + GI tract receptors
  • Resp centres of the brain (pontine)
499
Q

Side effects of opioids

A
  • Addiction
  • Constipation
  • Nausea and vomiting
  • Respiratory distress/depression
500
Q

How long does opioid withdrawal last?

A
  • Starts within 24 hours
  • Lasts about 72 hours
501
Q

What to do for opioid induced respiratory depression?

A
  • Call for help
  • ABC (check airway, breathing, circulation)
  • Naloxone
  • IV is fastest route
  • Titrate to effect - don’t have to give it all at once - once you’ve injected a drug you
    can’t get it back!
  • Short half-life of naloxone - beware drug addict overdoses in A&E
502
Q

Codeine is a pro-drug what does this mean?

A

Needs to be metabolised by cytochrome CYP2D6 to morphine to work.

  • 10% of people don’t have cytochrome CYP2D6 that converts it, codeine will have no effect in these people.
  • 5% of people have too much of the enzyme, these individuals may be at increased risk of respiratory depression with codeine
503
Q

What is morphine metabolised to?

A

Morphine 6 glucuronide.

504
Q

In renal failure, what may morphine 6 glucuronide lead to?

A

Morphine 6 glucuronide is renally excreted. In renal failure it will build up and may cause respiratory depression

505
Q

What should patients with renal failure use instead of morphine?

A

Oxycodone

506
Q

Parasympathetic (Ach) responses

A
  • Rest + digest
  • Pupil constricts
  • Lower heart rate
  • Bronchoconstritction
  • Increased GI motility and secretion
  • Detrusor muscle contracts
  • Penis points (erect)
507
Q

Sympathetic (NAd) response

A
  • Fight or flight
  • Pupil dilates
  • Increased heart rate
  • Bronchodilation
  • Decreased GI motility and secretion
  • Detrusor muscle relaxes
  • Penis shoots (ejaculation)
508
Q

Sympathetic nervous system receptors

A

alpha 1 (postsynaptic) - vasoconstriction

alpha 2 (presynaptic) - negative feedback suppresses norad release

beta 1 - increased heart rate and contractility

beta 2 - bronchodilation

509
Q

When does a pharmacodynamic interaction occur?

A

Occurs when drugs have an effect on the same target or physiological system

510
Q

When does a pharmacokinetic interaction occur?

A

Occurs when a drug affects the pharmacokinetics (absorption, distribution, metabolism or excretion) of another drug

511
Q

Features of a pharmacodynamic interactions.

A
  • Pharmacodynamic interactions are either synergistic or antagonistic
  • Due to drugs acting on the same drug receptor(s) or physiological system
  • Generally predictable (related to pharmacology of drug)
  • Highly selective drugs are less likely to be problematic
512
Q

72 year old female prescribed amitriptyline (for nerve pain) and solifenacin (for bladder spasm)

Solifenacin is a highly selective antagonist for M3 receptors
M3 receptors widespread (bladder, CNS, salivary glands, GIT)

Amitriptyline increases serotonin and NA in synaptic cleft. Poor specificity for target, also antagonist at M3 receptors

This leads to amplified anticholinergic effects (dry mouth, constipation, flushed+dry skin, delirium)

In the above scenario is the pharmacodynamic interaction synergistic or antagonistic?

A

Synergistic interaction

513
Q

54 year old male prescribed atenolol (ß antagonist) for hypertension and salbutamol (ß2 agonist) for asthma

Experiencing wheeze
This is due to an antagonist reaction between the two drugs. Explain how?

A

Salbutamol agonist at ß2 receptors (located in bronchial smooth muscle)
Atenolol cardioselective ß blocker (cardioselective antagonist at ß1 receptors but also antagonist at ß2)
Atenolol and salbutamol compete at ß2 receptors
Results in incomplete agonist effect and reduced bronchodilation/increased bronchospasm

514
Q

34 year old male prescribed morphine (pain relief) and lorazepam (anxiety)

Increased sedation

This is due to a synergistic reaction between the two drugs. Explain how?

A

Morphine agonist at MOP, KOP and DOP opiate receptors
Benzodiazepines potentiate actions of GABA (inhibitory CNS neurotransmitter)
MOP and KOP agonism and GABA transmission cause sedation
Different mechanisms within the CNS
Results in increased risk of sedation

515
Q

48 year old female prescribed methadone (opioid misuse) and ciprofloxacin (antibiotic)

Complaining of dizziness and palpitations
This is due to synergistic reaction between the two drugs. Explain how?

A

Methadone agonist at MOP, KOP and DOP opiate receptors
Ciprofloxacin quinolone antibiotic
Methadone and ciprofloxacin block voltage gated K channels
Results in increased risk of QT prolongation and Torsades de Pointes

516
Q

51 year old female prescribed amlodipine (hypertension) and ramipril (hypertension)

This will result in a synergistic reaction between the two drugs. Explain how, and whether this is beneficial or not?

A

Amlodipine: dihydropyridine calcium channel blocker reduces Ca2+ influx into vascular smooth muscle
Ramipril: ACE inhibitor inhibits conversion of angiotensin I to angiotensin II
Both lead to reduced vasoconstriction
Results in increased risk of hypotension (may be beneficial)
In practice: CCBs and ACEIs are used in combination in management of hypertension

517
Q

Pharmacokinetic drug interactions: What mechanisms of a drug could affect the absorption of another drug?

A
  1. Drugs which alter pH of GI tact
  2. Formation of insoluble drug complexes
  3. P-glycoprotein induction/inhibition
518
Q

How can a drug which causes changes in GI Tract pH
affect absorption of another drug?

A

Changes in pH will alter the proportion of ionised and unionised drug

519
Q

Famotidine (H2 antagonist) increases stomach pH and reduces absorption of ketoconazole (antifungal). In practice what can be done to prevent this?

A

In practice: If taking famotidine, oral ketoconazole should be taken with an acidic drink

520
Q

How can complex formation affect absorption?

A

Insoluble drug complexes will not be absorbed and will be retained in the GIT

521
Q

doxycycline and iron can form complexes which will not be absorbed. In practice what can be done about this?

A

In practice: separate administration of doxycycline and oral iron by 2-3 hours

522
Q

How can a drug causing P-glycoprotein induction/inhibition affect the absorption of another drug?

A

P-glycoproteins (P-gp) are drug transporter proteins widely distributed in body
Excretory function to remove toxic substances (incl drugs) out of cells
Amongst other locations - situated in intestinal lumen to reduce absorption

523
Q

Pgp inhibitors and inducers lead to increased or decreased level of Pgp substrates
Carbamazepine = Pgp inducer
Upregulates P-gp and therefore reduces the absorption of P-gp substrates
Carbamazepine significantly reduces rivaroxaban levels (note interaction also occurs due to CYP3A4 induction)
In practice what can be done about this?

A

NOTHING. In practice: combination can lead to anticoagulant treatment failure, therefore concurrent use is contraindicated

524
Q

Pharmacokinetic drug interactions: What issues could occur during distribution of a drug?

Give an example.

A

Only unbound drug will be distributed from plasma volume. Interactions can occur when drugs compete for protein binding

Warfarin is highly protein bound (~99%)
1% unbound and pharmaceutically active
Amiodarone displaces warfarin from albumin to create unbound warfarin molecules
Change of 99% bound to 98% bound = free drug doubles from 1% to 2%

525
Q

What enzymes are responsible for majority of phase 1 metabolic reaction

A

Cytochrome P450 (CYP450) enzymes

526
Q

What are inducer drugs?

A
  • Increase Cytochrome P450 activity and speed up metabolism of other drugs
527
Q

List examples of inducer drugs

A

PCARBS

Phenytoin
Carbamazepine
Alcohol (chronic use)
Rifampicin
Barbiturates (St John’s Wort)
Sulfonylureas and smoking

528
Q

What are drugs which decrease CYP450 activity and decrease metabolism of other drugs called?

Give an example.

A

Inhibitor drugs

Macrolides (clarithromycin, erythromycin)
Azoles (fluconazole)
Grapefruit juice

529
Q

Pharmacokinetic drug interactions: What issues could occur during elimination of a drug?

Give an example.

A

Competition for renal tubular secretion. Drugs are transported by OAT (organic anion transporters) and OCT (organic cation transporters.

Methotrexate (MTX) is 80-90% excreted unchanged by the kidneys
Secreted into renal tubule by OAT
NSAIDs compete with MTX for secretion by OAT leading to reduced MTX elimination
MTX toxicity: cirrhosis, fatal blood dyscrasias, renal damage (acute renal failure)

530
Q

How does grapefruit juice affect CYP450? And what drugs might this impact on?

A

Grapefruit juice is a CYP450 inhibitor, it affects CYP3A4 specifically and increases the bioavailability of some drugs e.g. warfarin, statins

531
Q

Eating foods high in which vitamin can affect the action of warfarin?

A

High in vitamin K - opposes warfarin

532
Q

Why should cranberry juice be avoided by patients taking warfarin?

A

Cranberry juice is a CYP2C9 inhibitor

533
Q

Milk can affect absorption of some drugs such as doxycycline, levothyroxine, ciprofloxacin, how?

A

Milk can affect absorption of some drugs due to insoluble complex formed with Ca

534
Q

It is important to identify and avoid drug interactions.
What types of drugs are considered high risk?

A

Obtain complete drug history (DHx)
Enzyme inducers, inhibitors and substrates
Drugs with a narrow therapeutic index
High risk/critical medicines
New drugs (e.g. biologics) - little data

535
Q

It is important to identify and avoid drug interactions.
What indicators are there for patients considered high risk?

A

Polypharmacy
Kidney or liver impairment
Extremes of age

536
Q

Mr K (86 year old male) is brought to A&E by his daughter after falling at home and hurting his wrist. He has recently started taking codeine for pain relief.
Which of his regular medicines would interact with codeine to increase his risk of falls?

  1. Aspirin
  2. Metformin
  3. Morphine
  4. Omeprazole
  5. Ramipril
A
  1. Morphine - duplicated agonism at opioid receptors.
537
Q

Define adverse drug reaction (ADR)

A

A response to a medicinal product, or combination of medicinal products, which is noxious and unintended

538
Q

List some examples of the impact of ADR on patients.

A
  • Reduced Quality of life
  • Poor compliance
  • Reduced confidence in clinicians and the healthcare system
  • Unnecessary investigations or treatments
539
Q

List some examples of the impact of ADR on the NHS

A
  • Increased hospital admissions
  • Longer hospital stays
  • GP appointments
  • Inefficient use of medication
540
Q

How can ADRs be classified using the ABCDE system?

A

A = Augmented
B = Bizarre
C = Chronic/continuing
D = Delayed
E = End of use/withdrawal
F = Failure of treatment
G = Genetic

541
Q

Type A ADR features

A

Most common type of ADR (80%)
Exaggerated effect of drugs pharmacology at a therapeutic dose
Often not life threatening
Dose dependent and reversible upon withdrawing the drug

542
Q

Give an example of a Type A ADR

A
  • AKI with ACE inhibitors
  • Bradycardia with betablockers
  • Hypoglycaemia with gliclazide, insulin
  • Respiratory depression with opiates
  • Bleeding with anticoagulants
543
Q

Type B ADR features

A
  • Not related to pharmacology of drug
  • Not dose related
  • Can cause serious illness or mortality
  • Symptoms do not always resolve upon stopping drug
544
Q

Give an example of a Type B ADR.

A
  • Anaphylaxis with penicillins
  • Tendon rupture with quinolone antibiotics
  • Steven Johnson Syndrome with IV vancomycin
545
Q

Type C ADR features

A

ADRs that continue after the drug has been stopped

546
Q

Give an example of a Type C ADR

A
  • Osteonecrosis of the jaw with bisphosphonates
  • Heart failure with pioglitazone
547
Q

Type D ADR features

A

ADRs that become apparent some time after stopping the drug.

548
Q

Give an example of a Type D ADR

A

Leucopenia with chemotherapy

549
Q

Type E ADR features

A

ADR develops after the drug has been stopped.

550
Q

Give an example of a Type E ADR

A
  • Insomnia after stopping benzodiazepine
  • Rebound tachycardia after stopping beta-blocker
  • Nasal congestion after stopping xylometolazine nasal spray
551
Q

Type F ADR features

A
  • Unexpected treatment failure
  • Could be due to drug-drug interaction or drug-food interaction
  • Poor compliance with administration instructions
552
Q

Give an example of a Type F ADR

A
  • Failure of oral contraceptive pill due to St John’s Wort
  • Failure of DOAC due to enzyme inducer (eg carbamazepine)
  • Failure of bisphosphonate due to taking with food
553
Q

What is the DoTS method of classifying ADR

A

Do = Dose-relatedness
T = Timing: ADRs which can develop during any time during treatment (often due to clinical changes in the patient).
S = Susceptibility
More complex than ABCDE, but provides more detail.
Useful for those working in pharmacovigilance, undertaking research or developing medicines

554
Q

DoTS: What are the three ADRs that could occur at different stages of doses.

A

Hypersusceptibility: ADRs at subtherapeutic doses (eg anaphylaxis with penicillins)

Collateral effects (side effects): ADRs at therapeutic doses (eg hypokalaemia with loop diuretic)

Toxic effects: ADRs at subpratherapeutic doses (eg liver damage with paracetamol)

555
Q

DoTS: Certain patient groups/populations may have a specific susceptibility to ADRs from a drug. Give factors which may make people more susceptible.

A

Age (anticholinergics in elderly patients)
Gender (metoclopramide in females)
Disease states (eg diclofenac in CVD)
Physiological states (eg phenytoin in pregnancy)

556
Q

What systems are in place to allow identification of ADRs?

A

Pre-clinical testing (computer models, cells and toxicity testing in animals)

Clinical trial data (pre-marketing evaluation)

Post marketing surveillance

Pharmacovigilance

557
Q

ADRs can be reported to the MHRA using the Yellow Card system. Which of the following ADRs is an augmented type ADR?

  • Treatment failure of warfarin due to dietary consumption of vitamin K
  • Neurodevelopmental disorder in child of patient taking sodium valproate
  • Constipation in patient taking morphine sulphate
  • Rebound tachycardia in patient after stopping atenolol
  • Angioedema in patient taking amoxicillin
A
  • Constipation in patient taking morphine sulphate
558
Q

Outline the steps taken if it is suspected that a patient has an ADR to a certain drug?

A
  1. Assess if ADR requires treatment (reversal, symptom management)
  2. Take a history
  3. Review medication history
  4. Review ADR profile of suspected drug (BNF, SPC)
  5. Modify dose, stop or swap
  6. Document allergy/ADR in patient record (including nature of reaction)
  7. If criteria met…report
559
Q

What are the different types of hypersensitivities mediated by?

A

Type 1 – IgE mediated drug hypersensitivity
Type 2 – IgG mediated cytotoxicity
Type 3 – Immune complex deposition
Type 4 – T cell mediated

560
Q

Features of Type 2 hypersensitivity

A

Type 2 reactions – Antibody-antigen complex formation

  • Antibody-antigen complex formation
    Drug or metabolite combines with a protein
    Body treats it as foreign protein and forms antibodies (IgG, IgM)
    Antibodies combine with the antigen and complement activation damages the cells e.g. methyl-dopa-induced haemolytic anaemia, pemphigus
561
Q

Features of Type 3 hypersensitivity

A

Type 3 reactions – Antibody-antigen complex deposition

Antigen and antibody form large complexes and activate complement
Small blood vessels are damaged or blocked
Leucocytes attracted to the site of reaction release pharmacologically active substances leading to an inflammatory process
Includes glomerulonephritis, vasculitis,

562
Q

Features of Type 4 hypersensitivity

A

Type 4 reaction – Lymphocyte mediated

Antigen specific receptors develop on T-lymphocytes
Subsequent admin, adminstration leads to local or tissue allergic reaction
E.g. contact dermatitis
E.g. Stevens Johnson syndrome (TEN)

563
Q

Main features of anaphylaxis

A

Exposure to drug, immediate rapid onset
Rash (absent in 10-20%)
Swelling of lips, face, oedema, central cyanosis
Wheeze / SOB
Hypotension (Anaphylactic shock)
Cardiac Arrest

564
Q

Features of non immune anaphylaxis

A

Due to direct mast cell degranulation.
Some drugs recognised to cause this
No prior exposure
Clinically identical

565
Q

Management of anaphylaxis

A
  1. Commence basic life support. ABC (airway, breathing, circulation)
  2. Remove the trigger if possible
    - Stop the drug if infusion
  3. Adrenaline IM 500 micrograms (300mcg epi-pen)
  4. High flow oxygen
  5. IV fluids – aggressive fluid resuscitation

If anaphylactic shock may need IV adrenaline with close monitoring
Antihistamines not first line treatment but can be used for skin symptoms
Corticosteroids no longer recommended

566
Q

How does adrenaline work?

A

Adrenaline acts on alpha 1, alpha 2, beta 1 and beta 2 receptors.

Vasoconstriction - increase in peripheral vascular resistance, increased BP and coronary perfusion via alpha1-adrenoceptors

Stimulation of Beta1-adrenoceptors positive ionotropic and chronotropic effects on the heart

Reduces oedema and bronchodilates via beta2-adrenoceptors

Attenuates further release of mediators from mast cells and basophils by increasing intracellular c-AMP and so reducing the release of inflammatory mediators

567
Q

Risk factors for hypersensitivity

A

Medicine factors:
Protein or polysaccharide based macro molecules

Host factors:
Females > Males – we don’t know why
EBV, HIV
Prev drug reactions
Uncontrolled asthma

Genetic factors:
Certain HLA groups
Acetylator status

568
Q

For a patient to have allergy for a drug, what criteria do they have to fulfil?

A

Does not correlate with pharmacological properties of the drug
No linear relation with dose (tiny dose can cause severe effects)
Reaction similar to those produced by other allergens
Induction period of primary exposure
Disappearance on cessation
Re-appears on re-exposure
Occurs in a minority of patients on the drug

569
Q

Define refractory anaphylaxis.

A

No improvement in respiratory or cardiovascular symptoms despite 2 appropriate doses of intramuscular adrenaline.

570
Q

Define commensal.

A

Organism which colonises the host but causes no disease in normal circumstances

571
Q

Morphological term for round shaped bacteria?

A

Cocci

572
Q

Morphological term for rod shaped bacteria?

A

Bacilli

573
Q

Features of typical bacteria structure

A
  • Cell wall
  • Chromosome of circular double stranded DNA
  • Outer membrane
  • Inner membrane
  • Pili
  • Capsule
574
Q

What are Gram positive bacteria bacterial cell envelope’s made up of?

A

Capsule
Peptidoglycan
Lipoteichoic acid
Cytoplasmic membrane

575
Q

How do Gram negative bacteria’s cell envelope differ from Gram positive?

A

Gram negative bacteria have a Lipopolysaccharide (ENDOTOXIN) cell wall, an outer membrane, lipoprotein, periplasmic space, and an inner membrane

576
Q

What colour does Gram positive bacteria stain?

A

Violet

577
Q

What colour does Gram negative bacteria stain?

A

Red

578
Q

Under what environments can bacteria survive in?

A
  1. Temperature
    <-800C to + 80C (1200C for spores)
  2. pH
    <4-9
  3. Water/dessication
    2 hours – 3 months (>50 years for spores)
  4. Light
    UV
579
Q

What are the four phases used to describe bacterial growth?

A
  • LAG phase
  • Exponential LOG phase
  • Stationary phase
  • Death phase
580
Q

What are the two bacterial toxins that bacteria can produce?

A

ENDOTOXIN
Component of the outer membrane of bacteria, eg lipopolysaccharide in Gram negative bacteria

EXOTOXIN
Secreted proteins of Gram positive and Gram negative bacteria

581
Q

Differences between endotoxins and exotoxins.

A

Exotoxin:
* Composed of proteins
* Action is specific
* Heat labile
* Strong antigenicity
* Produced by both Gram +ve and -ve
* Can convert to toxoids

Endotoxins:
* Composed of lipopolysaccharide
* Action is non specific
* Heat stable
* Weak antigenicity
* LPS Gram -ve
* Cannot convert to toxoids

582
Q

What can lead to genetic variation in bacteria (antibiotic resistance)?

A

Mutations:
- deletion
- insertion
- base substitution

Gene transfer:
- Transformation eg via plasmid
- Transduction eg via phage
- Conjugation eg via sex pilus

583
Q

What is meant by obligate intracellular bacteria?

A

Bacteria that cannot be cultured in media, requires a host cell to replicate.

584
Q

Give examples of obligate intracellular bacteria.

A
  • Chlamydia
  • Coxiella
  • Rickettsia
585
Q

Give an example of a slow growing bacteria.

A

TB

586
Q

Give an example of a fast growing bacteria.

A

E.coli and S.aureus.

587
Q

Give 2 functions of pili.

A
  1. Help adhere to cell surfaces
  2. Plasmid exchange.
588
Q

Give an example of a gram negative aerobic diplococci?

A

Neisseria e.g. N.meningitidis and N.gonorrhoeae.

589
Q

Give two examples of gram positive cocci?

A

Staphylococcus and streptococcus.

590
Q

What type of organism would you stain with Ziehl Neelsen?

A

Mycobacteria e.g. TB.

591
Q

What test can be done to distinguish between staphylococcus and streptococcus?

A

The catalase test; detects the presence of catalase enzyme using hydrogen peroxide. Staph = catalase + ve. Strep = catalase - ve.

592
Q

How would you describe the arrangement of staphylococci?

A

Clusters of cocci.

593
Q

How would you describe the arrangement of streptococci?

A

Chains of cocci.

594
Q

Pain in shoulder, elevated temperature
MRI scan, disc infection and osteomyelitis C6 &C7,
Blood cultures grow Gram positive cocci in clusters. What is the likely bacterial infection? How is it treated?

A

Staphylococcus aureus; flucloxacillin 3 months

595
Q

What test could be done to further distinguish between staphylococci bacteria.

A

Coagulase test; looks at whether a fibrin clot is produced.
Staphylococci aureus = coagulase +ve
All other staph bacteria = coagulase -ve

596
Q

What is the normal habitat of Staphylococci?

A

Nose and skin

597
Q

How is Staph. aureus spread?

A

Spread by aerosol and touch - carriers and shedders

598
Q

Virulence of factors of Staph. aureus

A

Pore-forming toxins (some strains)
a - haemolysin & Panton-Valentine Leucocidin
Proteases
Exfoliatin
Toxic Shock Syndrome toxin (stimulates cytokine release)
Protein A (surface protein which binds Ig’s in wrong orientation)

599
Q

S.epidermidis is coagulase -ve. How may they cause infections?

A

They are opportunistic
Infections in debilitated, prostheses (opportunistic)
Main virulence factor - ability to form persistent biofilms

600
Q

How might S.saprophyticus
cause infections?

A

Acute cystitis
haemagglutinin for adhesion
urease

601
Q

Wound received to shoulder
3 days later spreading inflammation
Fever, malaise
Swab culture showed Gram positive cocci in chains. What is the bacteria involved likely to be? How could it be treated?

A

Streptococcus pyogenes
Penicillin sensitive

602
Q

What test could be done to distinguish between different streptococci?

A

Blood agar haemolysis.
Beta- complete lysis
e.g. S.pyogenes
Haemolysins O & S
Alpha- partial, greening
e.g. S.intermedius
non (or Gamma) - no lysis
e.g. some S.mutans

603
Q

What further test can be done for those streptococci in the β haemolysis group?

A

Serogrouping; detecting carbohydrate cell surface antigens. e.g. lancefield grouping.

604
Q

In Lancefield grouping, what are the members of Group A and Group B?

A

Group A - S.pyogenes; important pathogen
Group B - S.agalactiae neonatal infections

605
Q

S.pyogenes virulence factors

A

Exported factors
Enzymes
Hyaluronidase - spreading
Streptokinase - breaks down clots
C5a peptidase - reduces chemotaxis

Toxins
Streptolysins O&S - binds cholesterol
Erythrogenic toxin - SPeA – exaggerated response

Surface factors

Capsule - hyaluronic acid

M protein – surface protein
(encourages complement degradation)

606
Q

Infections caused by S.pyogenes

A

Wound infections&raquo_space; cellulitis, puerperal fever
Tonsillitis & pharyngitis
Otitis media
Impetigo
Scarlet fever
Complications
rheumatic fever
glomerulonephritis

607
Q

Chains of purple cocci are seen on a gram film. They show alpha haemolysis when grown on blood agar. They don’t grow near the optochin disc. What bacteria is likely to have caused this?

A

Streptococcus pneumonia.

608
Q

How can you differentiate streptococci pneumoniae from other streptococci?

A

The optochin test can differentiate streptococci pneumoniae from other streptococci. Pneumococci are sensitive and so a clear area would be seen.

609
Q

What illnesses can S.pneumoniae cause?

A

pneumonia, otitis media, sinusitis, meningitis

610
Q

Predisposing factors for S.pneumoniae infections

A

impaired mucus trapping (e.g. viral infection)
hypogammaglobulinaemia
asplenia

611
Q

S.pneumoniae virulence factors

A
  1. Capsule
    polysaccharide (84 types), antiphagocytic
    polyvalent vaccine
  2. Inflammatory wall constituents
    teichoic acid (choline)
    peptidoglycan
  3. Cytotoxin
    pneumolysin
612
Q

What can Viridans group streptococci cause?

A

Some cause dental caries & abscesses
Important in infective endocarditits
S. sanguinis, S. oralis
Cause deep organ abscesses (e.g. brain, liver)

613
Q

Haemolysis of Viridans group streptococci

A

alpha- haemolytic (or non-haemolytic)

614
Q

Features of bacillus anthracis

A

Gram positive bacilli
Spore forming - pneumoniae, skin infection

615
Q

Corynebacterium diphtheriae clinical feature?

A

Thick greyish pseudo membrane on tonsils

616
Q

Virulence factors of Clostridia

A

Spore forming
Survive in environment
Produce toxins

617
Q

What does C. tetani cause?

A

Tetanus

618
Q

What does C. botulinum cause?

A

Botulism

619
Q

What does C. difficile cause?

A

antibiotic associated diarrhea
pseudomembranous colitis

620
Q

LPS (‘endotoxin’) forms the outer leaflet of the outer membrane of Gram -ves, what is the LPS comprised of?

A
  • Lipid A, the toxic portion of LPS that is anchored in the outer leaflet of the outer membrane.
  • Core (C) antigen (core oligosaccharide), short chain of sugars, some are unique to LPS.
  • Somatic (O) antigen (O-polysaccharide), a highly antigenic repeating chain of oligosaccharides.
621
Q

Which three main aerobic Gram negative bacteria rod groups are there?

A
  1. Coliforms
  2. Vibrio
  3. Pseudomonads
622
Q

Coliform features

A

Enterobacteriaceae or Enterobacteria

Rod-shaped
Motile (most)
Peritrichous flagella

Facultatively anaerobic

Colonise the intestinal tract

623
Q

What kind of bacteria is MacConkey agar used with?

A

Gram negative bacilli.

624
Q

What is MacConkey agar?

A

MacConkey agar contains bile salts, lactose and pH indicator. If an organism ferments lactose, lactic acid will be produced and the agar will appear a red/pink colour.

625
Q

Name 2 gram negative bacilli that will give a positive result with MacConkey agar.

A
  1. E.Coli.
  2. Klebsiella pneumoniae.
626
Q

What category of bacteria does shigella fall under?

A

Gram negative bacilli.

627
Q

Does shigella give a positive result with MacConkey agar?

A

No. Shigella does not ferment lactose and so gives a negative result.

628
Q

Does e.coli give a positive result with MacConkey agar?

A

Yes. E.coli does ferment lactose and so you would see a red/pink colour indicating a positive result.

629
Q

How can you distinguish between gram negative bacilli (salmonella, shigella and e.coli)?

A

Xylose Lysine Deoxycholate (XLD):
- e. coli +ve results turn phenol red in media yellow
- shigella cannot ferment lactose remains red
- salmonella cannot ferment lactose, but reduce thiosulphate to produce hydrogen sulphide (black)

630
Q

Which type of e.coli would you associate with causing travellers diarrhoea?

A

Enterotoxigenic e.coli (ETEC).

631
Q

Principal infections caused by pathogenic E. coli

A

Wound infections (surgical)
UTIs
Gastroenteritis
Travellers’ diarrhoea
Bacteraemia
Meningitis (infants) – rare in UK

632
Q

Why are there pathogenic strains of e.coli?

A

Due to the acquisition of genes from other bacteria.

633
Q

What are the symptoms of shigella infection?

A

Severe bloody diarrhoea (bacillary dysentery) and frequent passage.

634
Q

How is shigella passed on?

A

Via contaminated food/water or from person to person.

635
Q

What bacteria is responsible for salmonellosis?

A

S.enterica.

636
Q

Name the 3 forms of salmonellosis.

A
  1. Gastroenteritis.2. Enteric fever.3. Bacteraemia.
637
Q

What causes gastroenteritis?

A

Frequent cause of food poisoning, 6-36 hour incubation period.

638
Q

What causes enteric fever?

A

Frequent cause of poor quality drinking water/poor sanitation
Systemic disease

639
Q

Describe the pathogenesis of enteric fever.

A
  1. Endocytosis.2. Migration to the basolateral membrane. 3. Survival in macrophage -> systemic spread.
640
Q

What infections are caused by p. auruginosa?

A

Acute infections (due to multiple toxins):
1. Localised
- Burn/surgical wounds
- UTI
- Keratitis
2. Systemic (bacteraemic -> sepsis)
- neutropenic patients (leukaemia, chemotherapy, AIDS)
3. ICU patients (ventilator acquired pneumonia)
- leading cause of nosocomial pneumonia

Chronic infections:
1. Cystic fibrosis (CF) patients

641
Q

What are the symptoms of v.cholerae?

A

Most severe diarrhoeal disease. Huge volumes of watery stools (no blood or pus).

642
Q

Why is v.cholerae so dangerous?

A

Release of cholera toxin causes huge amounts of water loss which can result in hypovolemic shock and severe dehydration, this can lead to death.

643
Q

Why would you need to be infected with a large amount of v.cholerae to show symptoms of the disease?

A

The optimum pH for v.cholerae growth is 8; alkaline. It is therefore very sensitive to the pH of the stomach.

644
Q

Why bacteria is the most common cause of food poisoning in the US and UK?

A

Campylobacter
- causes mild to severe diarrhoea, often with blood

645
Q

How would you grow haemophilus influenzae?

A

On chocolate agar as it requires haem and NAD.

646
Q

What diseases can haemophilus influenzae cause?

A

Mostly opportunistic infections:
Meningitis and pneumonia.

647
Q

Virulence determinants of H. influenzae

A

Capsule, LPS

648
Q

What diseases can Helicobacter pylori cause?

A

Major role in gastritis and peptic ulcer disease (80-90% of ulcers)

649
Q

What disease can Bordetella pertussis cause?

A

Pertussis (whooping cough)
- Highly contagious
Non-specific flu-like symptoms (~7 d), followed by paroxysmal coughing

650
Q

Name the bacteria that can cause legionnaires disease?

A

Legionella pneumophila

651
Q

Who might be susceptible to infection by legionella?

A

Immunocompromised individuals.

652
Q

What problems are there with treating the anaerobic bacteroides?

A

Opportunistic - tissue injury
Often present in polymicrobial infections with enterobacteria, thus require specific anti-anaerobe antibiotics

653
Q

What are the main Gram negative cocci groups?

A

Veillonella
Neisseria

654
Q

What are the two medically important species of neisseria?

A

N.meningitidis and N.gonorrhoeae.

655
Q

Describe the pathogenesis of N.meningitidis.

A

Crosses nasopharyngeal epithelium and enters blood stream. Can cause asymptomatic bacteraemia or septicaemia. If the bacteria crosses the BBB it can cause meningitis.

656
Q

What are the consequences of N.gonorrhoeae infection?

A

STI - rectal, vaginal or oral inflammation.

657
Q

What are the three main groups of Gram negative spirochaetes?

A

Borrelia
Treponema
Leptospira

658
Q

Describe the flagellum of a spirochaete.

A

Spirochaete’s have an endoflagellum, it propels bacterium in a corkscrew motion

659
Q

Name the spirochaete that is responsible for causing lyme disease.

A

B.burgdorferi.

660
Q

Name the spirochaete that is responsible for causing syphilis.

A

T.pallidum.

661
Q

Describe the three stages of syphilis.

A
  1. Primary stage: localised infection.
  2. Secondary stage: systemic - skin, lymph nodes etc.
  3. Tertiary stage: CV syphilis and neuro syphilis.
662
Q

Can you grow chlamydia on agar?

A

No, chlamydia is an obligate intracellular parasite.

663
Q

How can you detect chlamydia?

A

Cannot culture in bacteriological media, thus can only be detected by serum antibodies or PCR.

664
Q

What are the 2 developmental stages of chlamydia’s unique growth cycle?

A
  1. Elementary bodies (infective).
  2. Reticulate bodies (intracellular multiplication).- Reticulate bodies are converted back into elementary bodies and are released. The cycle continues.
665
Q

Which bacteria is responsible for chlamydia?

A

c.trachomatis

666
Q

Name 2 bacteria in the chlamydophila genre.

A
  1. C.pneumoniae - respiratory tract infection
  2. C.psittaci - associated with birds.
667
Q

Give an example of mycobacteria.

A

M.tuberculosis (TB).
M.leprae (leprosy).

668
Q

Why are Mycobacteria resistant to Gram stain?

A

High lipid content with mycolic acids in cell wall makes Mycobacteria resistant to Gram stain

669
Q

What staining is used for Mycobacteria?

A

Ziehl-Neelsen stain

670
Q

Challenges of treating and diagnosing TB?

A
  1. Thick lipid rich cell wall making immune cell killing and penetration of drugs challenging
  2. SLOW growth
    - Gradual onset of disease
    - Takes much longer to diagnose
    - Takes longer to treat
671
Q

What is pulmonary tuberculosis?

A

Granulomas forms around bacilli that have settled in apex

672
Q

How does TB infect us?

A
  • Aerosol transmission
  • Primary TB in lung
  • Latent TB can remain for decades
    Can spread beyond lungs
673
Q

What does our body do to protect us from TB?

A
  • Primarily controlled by macrophages
  • Requires a CD4 T cell response to be controlled
  • Involves many cells of immunity- formation of granulomas
  • Granuloma stability controls reactivation of TB
674
Q

How are Mycobacteria adapted to evade the immune system?

A

Mycobacteria can withstand phagolysosome killing and escape to the cytosol.

Mycobacteria can form granulomas - when they become unstable can result in the break out of systemic disease

675
Q

What is a rapid diagnosis method for detecting M. tb?

A

Nucleic acid detection

676
Q

How can you use the immune response as a diagnosis test?

A

Tuberculin skin test (mantoux)

677
Q

Standard treatment for TB?

A
  • Isoniazid (INH), rifampicin (RIF), pyrazinamide (PZA), ethambutol (ETA) for two months
  • Isoniazid and rifampicin for four more months
678
Q

List the resistance mechanisms of TB

A

Drug inactivation:
Mtb produces beta-lactamase

Drug titration:
Target overproduction

Alteration of drug target:
Missense mutations

Altered cell envelope:
Increased permeability and drug efflux

679
Q

XDR-TB: resistant to four commonly used TB drugs. How would you treat these strains?

A

BPaL regimen for 6 months
However this could fail as well

680
Q

Define what a virus is.

A

An infectious, obligate intracellular parasite

Comprising genetic material
(DNA or RNA) surrounded by a protein coat and/or a membrane

681
Q

List an example of a non-enveloped virus

A

Adenovirus
Parvovirus

682
Q

List an example of an enveloped virus

A

HIV
Influenza

683
Q

How do viruses replicate?

A
  1. Attachment to specific receptor
  2. Cell entry
  3. Host cell interaction + replication
  4. Assembly of virion
  5. Release of new virus particles
684
Q

How do viruses cause disease?

A

a) Direct destruction of host cells
b) Modification of host cell
c) “Over-reactivity” of immune system
d) Damage through cell proliferation
e) Evasion of host

685
Q

Give an example of a virus that causes damage by direct destruction of host cells.

A

Poliovirus
-> host cell lysis and death after a viral replication period of 4 hours

686
Q

Give an example of a virus that causes damage by modification of host cell.

A

Rotavirus
-> atrophies villi and flattens epithelial cells, this leads to decrease of small intestine s.a, nutrients not absorbed, hyperosmotic state, profuse diarrhoea

687
Q

Give an example of a virus that causes damage by ‘over-reactivity’ of immune system.

A

Hepatitis B
SARS-CoV-2

688
Q

Give an example of a virus that causes damage by cell proliferation.

A

human papillomavirus -> cervical cancer

689
Q

How can a virus evade of host defences on a cellular level?

A

Cellular level:
- Herpesviridae - latency
- Measles, HIV - cell-cell spread

690
Q

How can a virus evade of host defences on a molecular level

A

Molecular level:
- Influenza, HIV, rhinovirus - antigenic variability
- Herpesviridae - prevention of host cell apoptosis
- Downregulation of interferon and other intracellular host defence proteins
- Herpesviridae, measles, HIV - Interference with host cell antigen processing pathways

691
Q

Why do viruses vary wildly in the range of clinical syndromes they can cause?

A
  • Different host cells and tissues that they can infect
  • Different methods of interaction with the host cell
692
Q

What is meningitis?

A

Inflammation of the meninges (membranes) which cover the brain and spinal cord

693
Q

Give an example of meningitis caused by bacteria

A

meningococcus, pneumococcus

694
Q

Given an example of meningitis caused by virus

A

coxsackievirus, echovirus, herpes virus, mumps virus, influenza, HIV etc

695
Q

List some non infectious causes of meningitis

A
  • Medications such as antibiotics, NSAIDs
  • Cancers such as melanoma, lung cancer
  • Autoimmune diseases such as SLE, Behcet’s disease
696
Q

What are the two main manifestations of Neisseria meningitidis?

A

Meningitis: a localised infection of the meninges, with “local” symptoms

Septicaemia : a systemic infection with widespread signs, and generalised organ damage

697
Q

What kind of bacteria is Neisseria meningitidis?

A

Gram negative diplococci

698
Q

Symptoms of meningococcal meningitis

A
  • Fever
  • Stiff neck
  • Headache
  • Confusion
  • Increased sensitivity to light
  • Nausea and vomitting
699
Q

What is Brudzinski’s neck sign?

A

When neck flexion causes the individual to flex their hips and knees automatically, typical sign in patients with meningitis

700
Q

Meningococcal septicaemia symptoms

A

Fever and chills
Fatigue
Vomiting
Cold hands and feet
Severe aches or pain in the muscles, joints, chest, or abdomen
Rapid breathing
Diarrhoea
Non blanching rash (petechiae)
In the later stages, a dark purple rash (purpura)

701
Q

Meningococcal septicaemia can cause Disseminated Intravascular Coagulation (DIC). What is this?

A

Activation of coagulation pathways that results in formation of intravascular thrombi (clots) and depletion of platelets and coagulation factors.
These clots can cause arterial occlusions leading to gangrene of extremities & auto-amputations (spontaneous detachment of an appendage from the body)

702
Q

What are some long term consequences of Meningococcal meningitis ?

A

Deafness or partial hearing loss (34%), seizures (13%), motor deficits (12%), cognitive impairment (9%), blindness (6%), Amputations (8% of children, 3% of adults)

703
Q

How would you make a diagnosis for Meningococcal meningitis?

A
  • Blood sample for blood culture & PCR
  • CSF for microscopy, culture and PCR
  • Throat swab for culture
704
Q

Features of yeast

A

Single celled organisms that divide by budding

705
Q

Features of mould

A

Form multicellular hyphae and spores

706
Q

What are dimorphic fungi?

A

Fungi which can exist as both yeasts and moulds, switching between the two when conditions suit

707
Q

Why are most fungi unable to cause human infection?

A

Inability to grow at 37 degrees
Innate and adaptive immune response

708
Q

Why are there limited options for antifungal drugs? Which part of the fungi do these drugs target?

A

Fungi are eukaryotic which limits options for selective toxicity.

DNA/RNA synthesis is similar to mammalian.

Antifungal drugs usually target - cell wall and ergosterol containing plasma membrane. Some drugs may target protein synthesis.

709
Q

Risk factors for mucosal candidiasis

A

immunosuppression, diabetes antibacterial therapy and mucosal disruption

710
Q

Treatment of mucosal candidiasis

A

Treatment with topical or oral azoles

711
Q

How are dermatophyte infections transmitted?

A

Human-human or animal-human transmission

712
Q

How would you treat dermatophyte infections?

A

Treatment is usually with topical or oral azoles or terbinafine

713
Q

Give an example of a dermatophyte

A

Trichophyton spp.,
Microsporum spp.
Epidermophyton floccosum

714
Q

Which fungi can cause severe infections in healthy hosts in endemic areas?

A

Dimorphic fungi. These are fungi that can exist as both yeast and mould; they are yeast in tissues but mould in vitro

715
Q

Give an example of a dimorphic fungi

A

Coccidioides immitis

716
Q

Late manifestations of Coccidioides

A
  1. Cavitatory lung disease
    may result in pleuritic pain or cough or become colonised by Aspergillus
  2. Orthopaedic
    Asymetrical chronic arthritis with effusion or vertebral osteomyelitis
  3. Cutaneous ulcers and abscesses
  4. Cervical lymphadenopathy
  5. Intracranial – chronic meningitis- BAD
717
Q

What usually causes invasive candidiasis?

A

infection of prosthetic devices or intra-abdominal disease

718
Q

What does Cryptococcus cause?

A

Cryptococcus causes acute or chronic meningitis in patients with reduced cell mediated immunity

719
Q

How would you investigate for Cryptococcus?

A

CSF microscopy with India ink

720
Q

Who would typically develop invasive aspergillosis?

A

Invasive aspergillosis is normally associated with profound immunocompromise but is increasingly recognised in patients with severe viral infection

721
Q

How would you treat invasive aspergillosis?

A

Voriconazole

722
Q

What are Mucoraceous moulds (zygomyctes) ?

A

Mucoraceous moulds (zygomyctes) are rare but cause devastatingly rapidly progressive infections that cross tissue planes.

Mucoraceous moulds need aggressive antifungal therapy and surgery for optimal outcomes

723
Q

What does pneumocystis jirovecii cause?

A

Pneumocystis jirovecii causes a pneumonitis with severe hypoxia in the immunocompromised

724
Q

What are protozoa?

A

Single celled eukaryotic organisms

725
Q

What is African Trypanosomiasis? What is it caused by?

A

“sleeping sickness”
Caused by Trypanosoma brucei gambiense and rhodesiense - spread by Tsetse fly bite
- Chancre
- Flu like symptoms
- CNS involvement - (sleepy, confusion, personality change)
- Coma and death

726
Q

What is American Trypanosomiasis? What is it caused by?

A

“Chagas Disease”
Caused by Trypanosoma cruzi - spread by Triatomine bug
Acute:
- Flu like symptoms
Chronic:
- Cardiomyopathy
- Megaoesophagus
- Megacolon

727
Q

What is Leishmaniasis? What is it caused by?

A

Caused by Leishmania spp
Spread by the bite of the sandfly
>20 species that affect humans
- Cutaneous & Mucocutaneous
* ulceration and destruction

  • Visceral
  • Fever
  • Weight loss
  • Massive splenomegaly
  • Hepatomegaly
  • Anaemia
  • High fatality without treatment
728
Q

How is Trichomonas vaginalis transmitted?

A

Sexually transmitted
- Most common non-viral STI

729
Q

How is Trichomonas vaginalis treated?

A

Treated with Metronidazole

730
Q

How is Giardiasis spread? What does it cause?

A

Faeco-oral spread
- Diarrhoea

731
Q

How is Giardiasis treated?

A

Treated with Metronidazole

732
Q

What is Amoebiasis caused by?

A

Entaemoeba histolytica
Faeco-oral spread

733
Q

How is Amoebiasis treated?

A

Metronidazole

734
Q

What is Cryptosporidiosis caused by? How is it spread?

A

Cryptosporidium spp
Waterborne

735
Q

What is Toxoplasmosis caused by? How is it spread?

A

Toxoplasma gondii
Ingestion of contaminated food and water/feline faeces

736
Q

What are the 5 classifications of amoeba?

A
  1. Flagellate i.e Trypanosoma, Leishmania, Giardia
  2. Amoeba i.e Entaemoeba histolytica
  3. Sporozoa i.e Cryptosporidium, Toxoplasma gondii, Plasmodium
  4. Microsporidia
  5. Cilliates i.e Balantidium coli
737
Q

How is malaria transmitted?

A

Transmitted by bite of female anopheles mosquito.

5 species:
Plasmodium falciparum
Plasmodium ovale
Plasmodium vivax
Plasmodium malariae
Plasmodium knowlesi

738
Q

What test to diagnose malaria?

A

Blood film test
Malaria parasites can be identified by examining under the microscope.
Thin (tells you species and parasite count) and thick (low resolution, tells you if you have malaria) blood films.

739
Q

Symptoms of malaria?

A

Chills
Headache
Myalgia
Fatigue
Diarrhoea
Vomiting
Abdo pain
FEVER

740
Q

What are the 4 signs of malaria?

A
  • Anaemia
  • Jaundice
  • Hepatosplenomegaly
  • ‘Black water fever’
741
Q

What does Plasmodium falciparum cause that results in ‘complicated malaria’?

A

OBSTRUCTED MICROCIRCULATION
- Caused by infected RBCs ability to adhere to endothelial cells

742
Q

What are features of Complicated Malaria?

A
  • Cerebral malaria
  • ARDS/Pulmonary oedema
  • Renal failure
  • Sepsis
  • Bleeding/Anaemia
743
Q

Treatment for complicated malaria?

A
  • IV artesunate
    (IV quinine + doxycycline)
744
Q

Treatments for uncomplicated malaria?

A

Chloroquine.

745
Q

What 2 species of the plasmodia genus lie dormant, form hypnozoites in the liver and cause late relapse of malaria? How is it treated?

A

P.ovale and p.vivax.
Primiquine to eliminate

746
Q

What is Aciclovir used for?

A

Anti-herpesvirus drug
Typically used to treat or prevent HSV1 HSV2 VZV infection.

747
Q

What is Aciclovir’s mechanism of action?

A
748
Q

What is are antibiotics?

A

Antibiotics are molecules that work by binding a target site on bacteria

749
Q

How do antibiotics work?

A
750
Q

Beta lactam antibiotics…

A

don’t work as well on Gram negative bacteria and aren’t useful for eukaryotic cells.

751
Q

Metronidazole and ciprofloxacin

A

Inhibit bacteria’s nucleic acid synthesis

752
Q

What are we trying to achieve with antibiotics?

A

Antibiotics give time and support for the immune system to deal with an infection.

753
Q

How are bacteria pathogenic?

A
  1. Attach and enter
  2. Local spread
  3. Multiply
  4. Evade host defences
  5. Shed from body
754
Q

Consequences of bacteria being pathogenic on the human body

A
  1. Direct: destroy phagocytes or cells in which bacteria replicate.
  2. Indirect: Inflammation - e.g. nectroic cells, immune pathology - e.g. antibody.
  3. Exotoxin - protein production. Endotoxin - Gram negative
  4. Diarrhoea
755
Q

Bactericidal antibiotics

A
  • The agent kills the bacteria.
  • Kills more than 99.9% in 18-24 hrs
  • Antibiotics that inhibit cell wall synthesis
  • Useful in endocarditis.
  • Difficult to treat meningitis.
756
Q

Bacteriostatic antibiotics

A
  • Prevents the growth of bacteria.
  • Kills more than 90% in 18-24 hrs
  • Antibiotics that inhibit protein synthesis, DNA replication or metabolism.
  • Reduce toxin production (i.e. endotoxin)
757
Q

What is mininum inhibitory concentration (MIC)?

A

Minimum Inhibitory Concentration (MIC) is the lowest concentration (expressed as mg/L or μg/μL) of an antimicrobial agent that inhibits the visible in-vitro growth of microorganisms.

The MIC test determines the antimicrobial activity of a test agent against a specific bacteria.

758
Q

What determines the efficacy of an antibiotic?

A

Drug must occupy an adequate number of binding sites, which is related to its conc within the microorganism.
Antibiotic should be able to remain at the binding site for sufficient period of time.

759
Q

The two major determinants of anti bacterial effects are…

A

the conc and time the antibiotic remains on these binding sites.

760
Q

Time dependent killing antibiotics

A

Key parameter is the time that serum conc remain above the MIC during the dosing interval.

E.g. beta lactams (penicillins, cephalosporins, carbapenems, monobactams), clindamycin, macrolides, oxazolidinones

761
Q

Concentration dependent killing antibiotics

A

Key parameter is how high the conc is above MIC.
E.g. aminoglycosides, quinolones

762
Q

The antibiotic reaching and staying on the site of bacterial infection depends on…

A

pharmacokinetics.

763
Q

What is the appropriate or available route of administration of antibiotics?

A
764
Q

How do antibiotics not work?

A
  1. Bacteria can change the molecular configuration of antibiotic binding site or masks it e.g. flucloxacillin and rifampicin
  2. The antibiotic is destroyed or inactivated e.g. Penicillins and cephalosproins hydrolysed by bacterial enzyme “Beta lactamase”
  3. Prevent antibiotic stress
  4. Remove antibiotic from bacteria
765
Q

Intrinsic resistance of bacteria against antibiotics?

A
766
Q

Acquired resistance of bacteria against antibiotics?

A
767
Q

Carbapenems

A
768
Q

What is the duration of a normal course of antibiotics?

A

5-7 days

769
Q

Is an antibiotic safe to prescribe? What do we need to consider?

A
  • What pathogen/bacteria are we treating?
  • Bioavailability of the drug
  • How are we administering the drug?
  • Age of the patient
  • Side effects
  • Intolerance, allergy, anaphylaxis
  • Renal and liver function
  • Pregnancy and breastfeeding
  • Risk of C.difficile
  • Drug interactions
770
Q

Beta lactams are

A

cell wall/peptidoglycan weapons

771
Q

Beta lactams inhibitors

A

Amoxicillin-clavulanate
Piperacillin-tazobactem

772
Q

What antibiotics work on Gram positive bacteria?

A

Thick cell wall therefore need beta lactams (simple cell wall weapon).

773
Q

What does MRSA stand for?

A

Methicillin-resistant Staph. aureus

774
Q

Vancomycin

A

Does not cross BBB or the eyes so not useful for infections there.

775
Q

Glycopeptides are cell wall weapons

A

Vancomyosin and Teicoplanin - IV

776
Q

What antibiotics work for Gram negative bacteria?

A
  • Can use beta-lactams but aren’t as useful.
  • Cefuroxime - IV. Do not cover anaerobic bacteria.
  • Co-amoxiclav (Augmentin) oral and IV.
  • Piperacillin/tazobactem
  • Meropenem - IV restricted at STH
777
Q

Case 1: 23-year old woman attends with dysuria, frequency and cloudy urine for 36 hours

A

Likely diagnosis
Urinary tract infection (lower)
Bacteria: gram negatives (E. coli, proteus, klebsiella)
And gram positives (staph saprophyticus)

Samples
Consider urine culture if ? Resistance
✓ Children, pregnancy, men, > 65s, sepsis/pyelonephritis, catheter

Management
Self care + back-up antibiotics
Immediate antibiotic treatment
Nitrofurantoin for 3 days

778
Q

Case 2: 66 year old woman
3 day history of fevers and nausea
Leg = hot, red, swollen

A

Likely diagnosis
Cellulitis usually lower limbs - unilateral
Red, hot, painful, tender skin
Spreading
Systemic symptoms
Bacteria: S. aureus and β haemolytic strep (Group A, C & G)
** Samples**
Bacterial swab for culture
Blood cultures
** Management**
Antibiotics
PO or IV flucloxacillin
Penicillin allergy: clarithromycin or clindamycin
Duration ~ 7 days

779
Q

Why is infection prevention and control important?

A

300,000 healthcare associated infections (HCAI) in England per year

Estimated 9% of in-patients have a HCAI

Cost approx. £1 billion per year

Cause or contribute to estimated 20,000 deaths

Estimated 75% of healthcare workers infected at work

780
Q

Which pathogens do we consider when it comes to infection prevention and control?

A

Bacteria

Methicillin resistant S.aureus
Clostridium difficile
Multi-drug resistant gram negatives
Glycopeptide resistant enterococci
Group A streptococcus
Mycobacterium tuberculosis

Viruses

Influenza
Norovirus
SARS-CoV-2
HIV
Hep B
Hep C
Varicella Zoster Virus
Viral haemorrhagic fevers

Other
Candida Auris
Creutzfeldt–Jakob disease

781
Q

Infection vs colonisation

A

Infect: verb affect (a person, organism, etc.) with a disease-causing organism

We are more bacterial cells than human cells – COLONISATION

782
Q

How do we prevent and control infections?

A

Ensure policies are kept.
Identification of risks:
- Routes and modes of transmission
- Virulence of organisms - ease of spread, likelihood of causing infection, consequences of infection if it occurs
- Some diseases have higher R rate i.e. Measles has R rate of 20 which means it can spread to 20 other people.

Minimise risks

783
Q

How do you identify a risk of an infection?

A

Risk factors
e.g. recent return from Sierra Leone with fever
Screening
e.g. MRSA admission screening
Clinical diagnosis
e.g. cough and cavity on chest x-ray
Lab diagnosis
Carbapenemase Producing Enterobacteriaceae in urine

784
Q

Routes of transmission

A
  1. Patient A > environment > Patient B
  2. Patient A > staff > Patient B
785
Q

What are CPEs?

A

Carbapenemase-producing Enterobacteriaceae

786
Q

What do CPEs include?

A

Include E. coli, Klebsiella, Proteus, Serratia, Enterobacter

787
Q

Where do CPEs colonise?

A

Colonisers of large bowel, skin below waist and moist sites

Most common causes of UTI, intra-abdominal infection

788
Q

What is carbapenemase?

A

Enzyme which inactivates carbapenem antibiotics

789
Q

MRSA

A

Methicillin resistant Staph aureus
Staph aureus is a common skin and nasal commensal
Most strains are susceptible to flucloxacillin and other beta-lactem antibiotics.
MRSA was first described in 1971

Huge increase in numbers of cases in 1990s and 2000s

790
Q

Norovirus

A

High attack rates amongst close contacts
Low infecting dose
Uncontained vomiting and diarrhoea

Short lived immunity only
Staff and patients at risk

Able to persist in the environment

Relatively resistant to conventional cleaning

791
Q

Clostridium difficile

A

Produce bacterial spores, leads to prolonged hospital stays, toxic megacolon.

Water is the only thing that removes the spores.

Need to wash hands with soap and water.

The antibiotics use to treat it begin with “C”

792
Q

What are endogenous infections?

A

Infections caused by patients own bacterial flora

Important in hospitalised patients, especially those with invasive devices or surgical patients

793
Q

How can you prevent endogenous infections?

A

Good nutrition and hydration

Antisepsis/skin prep where indicated

Good theatre practice

Remove lines and catheters as soon as clinically possible

Change from IV to oral treatment whenever appropriate

794
Q

What are some simple things that you can do to protect patients from infections?

A

Hand Hygiene is the single most effective method of preventing cross infection

(Can be hand washing and/or alcohol gel)

Personal protective equipment e.g. gloves, aprons, masks etc.

Disposal of sharps

795
Q

What are the two blood tests used to monitor HIV infection?

A
  1. CD4 count - measure of immune function
  2. HIV viral load test
796
Q

What is the UNAIDS goal by 2020?

A

90/90/90
*90% diagnosed.
*90% on anti-retroviral treatment.
*90% viral suppression, undetectable viral load.

797
Q

What is the Fast-Track Cities initiative goal by 2030?

A

*95/95/95
*95% diagnosed, 95% on treatment, 95% virally suppressed.
*200,000 new infections per year
*zero discrimination

798
Q

Did the UK achieve the 90/90/90 target?

A

YES! :D achieved by 2018

799
Q

HIV transmission routes

A

*Sexual
*Vertical

800
Q

Prevention of HIV?

A

*Condoms
*Post exposure prophylaxis (PEP) (72 hrs)
*Oral pre-exposure prophylaxis (PrEP)
*Male circumcision
*Testing STIs
*Treatment as prevention (TasP)

801
Q

What are the steps that should be taken following HIV diagnosis?

A
  • Discuss soon after diagnosis
  • length of ‘look back’ depends on individual circumstances
  • Document discussion of safer sex practices, PEPSE, PreP for their partner
802
Q

What is U=U?

A

Undetectable = untransmittable

803
Q

Why is HIV testing important?

A
  • Access to appropriate treatment and care
  • Reduction in morbidity an mortality
  • Reduction of vertical transmission
  • Reduction of sexual transmission
  • Public health/partner notification
  • Cost-effective
804
Q

What scenarios would you test for HIV?

A

*Clinician initiated testing triggered by clinical indicators of immuno-suppression disease/seroconversion
*Routine screening in high prevalence locations
*Routine screening Antenatal screening
*Screening in high risk groups
*Patient initiated requests for testing

805
Q

Why do doctors not test for HIV?

A

*They don’t think of HIV
*Underestimate the risk of HIV in their patients
*Failure to recognise HIV as a modifiable prognostic indicator
*Misconception they need pre-test counselling
*Misunderstanding of the implications for insurance, etc
*Fear of offending the patient

806
Q

What is the most common opportunistic infection seen in HIV patients?

A

Pneumocystis pneumonia (PCP)
Caused by the Pneumocystis jirovecii fungal infection
Symptoms: fevers, SOB, dry cough, pleuritic chest pain, exertional drop in oxygen saturations

807
Q

Common medical conditions where you should consider the possibility of HIV.

A
  • Flu-like illness, rash
  • Blood dyscrasias eg low platelets
  • Multi-dermatomal shingles
  • Lymphadenopathy
  • Weight loss or diarrhoea, night sweats, PUO
808
Q

What method of HIV screening test is preferred?

A

*Venous blood sample is preferred.
*4th generation HIV tests include p24 antigen and will detect the vast majority of infections at 4 weeks (if negative, repeat at 7 weeks if high index of suspicion)
*High sensitivity and selectivity

809
Q

HIV is a retrovirus what does this mean?

A

It is an RNA virus which uses reverse transcriptase (RT) to make a DNA copy that becomes integrated into the DNA of the infected cell.

810
Q

Where did HIV come from?

A

HIV-1 is similar to SIV in chimps from central Africa, and probably arose there
HIV-2 closely resembles SIV isolated from West African sooty mangabey monkeys

811
Q

Why is it hard to create a vaccine for HIV?

A

HIV-1 mutates rapidly, as it is error-prone and replicates rapidly.

The tried and tested vaccine strategies that we have used for other organisms (live attenuated or killed vaccines) are deemed too risky for HIV

812
Q

HIV genome structure

A

Small RNA virus ~10KB
Retrovirus family
Lentivirus: long incubation period

813
Q

Briefly describe the mechanism of HIV replication.

A
  1. GP160 binds to CD4 receptors.
  2. Viral caspid, enzymes and nucleic acids are uncoated and released into the cell.
  3. RNA is converted into DNA using reverse transcriptase.
  4. Viral DNA is integrated into cellular DNA by intergrase.
  5. Viral DNA is transcribed into viral proteins.
  6. Splicing.
  7. New HIV cells ‘bud’ from CD4.
814
Q

What is the CD4+ count when someone is diagnosed with having AIDS?

A

CD4+ <200.

815
Q

Where in the cell can HIV drugs target?

A
  1. Reverse transcriptase inhibitors.
  2. Protease inhibitors.
  3. Fusion/entry inhibitors.
  4. Integrase inhibitors.
816
Q

How are some people resistant to HIV-1 infection?

A

Homozygous 32bp deletion in the CCR5 gene.

817
Q

What are the 4 main phases in the natural history of untreated HIV?

A
  1. Acute primary infection. 6-12 weeks
  2. Asymptomatic phase. 1-15+ years
  3. Early symptomatic HIV.
  4. AIDS.
818
Q

Why does the immune response to HIV-1 fail to clear the virus?

A

*CD4 T helper cells are lost from early in infection as these are the cells HIV infects first.
*Cytotoxic CD8 cell response fails when “immune exhaustion” sets in.
*Antibodies fail to bind to virus due to heavily glycosylated envelope spike.
*Virus can change quickly and evade antibody recognition

819
Q

Which regions are seeing the most rapid rise in new HIV infections?

A

Eastern European and central Asia

820
Q

List 3 social-economic impact of HIV/AIDS in Africa?

A

*Significant impact on life expectancy
*Loss of economically-productive adults (including health-care workers)
*Increased spending on healthcare (particularly as ART drug use becomes more widespread)
*Distortion of health-care spending
*Change in social structure: orphans cared for by elderly grandparents
*Stigma of HIV infection persists

821
Q

Who is most at risk of acquiring HIV?

A

In the modern world
HIV incidence among adolescent girls and young women (aged 15-24) are the highest.

822
Q

What are the three methods of paediatric HIV-1 transmission?

A

*In utero - transplacental, mostly during third trimester
*Intra partum - exposure to maternal blood and genital secretions during delivery
*Breast milk - ingestion of large amounts of contaminated milk

823
Q

How does male circumcision work to prevent HIV infection?

A

By removing foreskin, circumcision reduces the ability of HIV to penetrate due to keratinization of the inner aspect of the remaining foreskin.

824
Q

What signs and symptoms might you see when someone is in the acute primary infection phase of HIV?

A

Symptoms usually start within 2-4 weeks of infection
*Similar to glandular fever/flu
*Abrupt onset of non-specific symptoms e.g. fever, rash, sore throat, Weight loss, myalgia, V+D, headache, lymphadenopathy

825
Q

What signs and symptoms might you see when someone is in the clinical latent phase of HIV?

A

No symptoms
May notice some enlarged lymph nodes
*persistent generalised lymphadenopathy

826
Q

What signs and symptoms might you see when someone is in the early symptomatic phase of HIV?

A

Unexpected medical conditions that are recurring with no clear underlying cause.

827
Q

What is the usual treatment for PCP?

A

High dose co-trimoxazole.

828
Q

What does it mean when a patient has HIV and TB?

A

Regardless of CD4 count they would be considered to have AIDS.

829
Q

Name 3 CNS diseases associated with HIV.

A
  1. Mass lesions e.g. primary CNS lymphoma, cerebral toxoplasmosis.
  2. Meningitis e.g. pneumococcal, cryptococcal.
  3. Opthalmic lesions e.g. CMV, toxoplasmosis, choroidal tuberculosis etc.
830
Q

What cancers does HIV increase the risk of?

A

HIV increases the risk of any cancer that is associated with a virus

Human Herpesvirus 8 -> Kaposi’s sarcoma
Epstein Barr Virus -> Lymphomas
Human Papillomavirus -> Cervical, anal, penile carcinoma
Hepatitis B/C -> Hepatocellulcar carcinoma

831
Q

What symptoms are there in someone with Kaposi’s sarcoma?

A

Single or multiple lesions, usually on the skin

832
Q

What is HAART?

A

Highly active anti-retroviral therapy where 3 drugs that act on different points in the replication cycle are taken together.

833
Q

What happens when a patient starts HAART?

A

*Viral load decreases rapidly.
*CD4 count begins to increase.

834
Q

What factors increase the chances of HIV developing drug resistance?

A
  1. Non adherence
  2. Drug-drug interactions