ICS Flashcards

1
Q

What are the main cells involved in acute and chronic inflammation?

Also, give examples of acute and chronic inflammations (broadly)

A

Acute inflammation - infections, hypersensitivity
Main cells involved - neutrophils

Chronic inflammation - Autoimmune diseases, recurrent infections
Main cells involved - macrophages (may present antigen to lymphocytes for long lived immunity) and lymphocytes

Endothelial cells and fibroblasts also involved.

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

What is the function of fibroblasts in inflammation?

A

They form collagen in areas of chronic inflammation and help with repair

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

How is damaged tissue repaired? E.g heart after MI

A

Damaged tissue is replaced by fibrous tissue e.g. collagen - produced by fibroblasts

e.g. heart after MI
Brain after cerebral infarction
Spinal cord after trauma

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

What are the 5 cardinal signs of inflammation? And what are their causes?

A

Rubor - Redness (vasodilation of arteries/capillaries near the site of injury)

Dolor - Pain (caused by chemical mediators of inflammation e.g. histamine)

Calor - Heat (vasodilation –> increased blood flow –> heat)

Tumor - Swelling (leakage of plasma fluids –> oedema)

Loss of function (generally caused by swelling and pain)

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

What are the stages of inflammation?

A

1) Increased vessel permeability –> (blood vessels adjacent to the injury first dilate, blood flow increases. The endothelial cells contract to increase the space between them, thus increasing permeability of the vascular barrier)

2) Fluid exudate –> Fluid leaks out from the vascular space into the interstitial space resulting in oedema (tumour)

3) Cellular exudate –> The increased fluid acts as a medium for inflammatory proteins to migrate through. Whole cells (RBC, WBC) leave the vessel as well. (e.g neutrophils leave to go to the site of injury)

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

What is the main cell involved in acute inflammation and what are the 3 stages of migration?

What happens at the site of inflammation?

A
  • Neutrophils

1) Margination - Neutrophils line up against the endothelium

2) Adhesion- Selectins (produced by endothelium) bind to the neutrophils causing them to ROLL along the margin (slowing down)

3) Emigration - Neutrophils interact with CD31 molecules and chemokines, becomes static and move through the wall to the affected area up the chemokine concentration gradient(DIAPEDESIS - movement of leukocytes from the blood vessel to sites of tissue damage) - form of cellular exudate

(Migration of neutrophils to the site of inflammation stimulated by chemokines)

At the site
1) Neutrophils begin phagocytosis
2) Formation of a phagolysosome –> bacteria killing
3) Macrophages clear the debris (and pathogens)

Neutrophils usually die at the site of infection after performing their job. may be cleared by macrophages.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What are the possible outcomes following acute inflammation?

Check with lectures

A

1) Complete resolution - Total repair (back to normal) - RESOLUTION

2) Abscess formation - Localised collection of PUS (neutrophils) surrounded by granulation tissue - this will over time, be replaced by scar tissue- (SUPPORATION)

3) Fibrosis and scar fomation - Occurs in cases of significant inflammation - (ORGANISATION) e.g. liver cirrhosis

4) Chronic inflammation - from a persistent insult/cause (Progression)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What are granulomas?

(what combines to form them in chronic inflammatin)

A

They are aggregates of epitheloid histiocytes (clusters of immune cells)- that form in response to the presence of foreign substances (in chronic inflammation, macrophages and lymphocytes can combine to form a granuloma)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What diseases would you see granulomas in?

What would be a blood marker?

A

Tuberculosis - where central necrosis is present

Leprosy - caesation necrosis

Sarcoidosis, vasculitis, crohn’s disease - where central necrosis is not present

ACE would be a blood marker for Sarcoidosis, crohn’s disease, tuberculosis and other granulomatous diseases

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

What is a thrombus?

A

A solid mass of blood constituents formed in an intact vessel in a living person. (blood clot)

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

What is an embolism?

A

A mass of material travelling in the vascular system which becomes lodged within vessel and blocks it.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What is venous thromboembolism (VTE)

A

A condition that occurs when a blood clot forms in the vein. It encompasses Deep vein thrombosis and pulmonary embolism (DVT can lead to PE) - clot from DVT, typically starting in the leg, lodges in the pulmonary arteries.

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

What is a venous thrombosis and what can it be caused by?

How is it treated?
If not treated, what can it lead to?

A

A condition that occurs when a blood clot forms in the vein. (usually leg)

Caused by:
- Venous stasis (DVT)
- Disease or injury to veins in the leg
- A fracture
- Autoimmune disorders that make it more likely for your blood to clot
- Medicines that increase your risk of clotting.

Treated with anticoagulants (warfarin) (you would also administer low dose subcutaneous heparin and have early mobilisation after operation)
Can lead to a PE if not treated

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

What is an arterial thrombosis and what can it be caused by?

How is it treated?
If not treated, what can it lead to?

A

A condition that occurs when a blood clot forms in the artery.

Caused by:
- Atherogenesis (process of plaque buildup)
- Atherosclerosis - arteries become narrowed by plaque buildup

Treated with antiplatelets (aspirin)

If occurs in coronary arteries - MI
If occurs in a blood vessel in the brain - Stroke (Ischemic stroke)

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

What is ischemia and infarction?

A

The inadequate supply of blood to an area due to the blockage of the blood vessels supplying the area.
- Reduction in blood flow (e.g. TiA)

Infarction
- A reduction in blood flow with subsequent death of cells. (e.g. ischemic stroke)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

How are arterial and venous thrombosis treated? Why are they treated differently?

A

Venous thrombosis treated with anticoagulants. (Warfarin)
- In a venous thrombosis when there is stasis, the level of coagulation factors and thrombin is high – thrombin breaks down fibrinogen into fibrin –> fibrin traps red blood cells.
Anticoagulants target the coagulation cascade, they inhibit the formation and growth of the (fibrin) clot, reducing risk of embolisation.

Arterial thrombosis treated with antiplatelets. (Aspirin)

  • Arterial thrombosis are platelet rich clots which develop at sites of artherosclerotic plaques (as fatty deposits damage the endothelial lining. Antiplatelets are taken to inhibit platelet aggregation and activation.

(Essentially tailored to the type of clot, fibrin rich clot or platelet rich clot)

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

What are the possible fates of thrombi (clots)?

A

Resolution –> The clot degrades and results in normal physiological outcome

Organisation –> Leaves scar tissue

Embolism –> Fragments of thrombi break off and lodge in distal circulation

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

What/Where are the most common arterial and venous thrombus and resulting emboli?

Not sure if the most COMMON but should know this?

A

Arterial emboli
–> Lodges in the systemic circulation (from left heart)
E.g. Due to irregular heart rhythm in Atrial fibrillation, sluggish blood flow can lead to the formation of clots –> Atrial fibrillation thrombi. –> Part of the clot can break loose and lodge in the carotid artery leading to ischemic stroke

Venous emboli
–> Lodges in the pulmonary circulation (from right heart)

e.g. DVT thrombus lodges in the pulmonary artery leading to a PE.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Differences between an arterial and venous ulcer

A

Location
Arterial- usually on the tips of toes and lateral malleolus of ankle
Venous - usually on the medial malleolus and inner calf

Exudate
Arterial - punched out hole (deeper) with little exudate
Venous - less demarcated punched out hole appearance (shallower) with a lot of exudate

Colour
Arterial - pale cool skin (or yellowish-grey base)
Venous - Reddish base

Skin changes
Arterial - hair loss, thickened toenails, weak distal pulse of affected limb
Venous - Varicose veins, itchy skin, eczematous (stasis dermatitis)

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

What is virchow’s triad and what are its components?

A

The formation of a thrombus is dependent on any one of Virchow’s triad being present

1) Abnormal blood flow (stasis/decreased blood flow) - due to e.g. periods of immobility (long flights/being bed bound) –> Most common caused of DVT

2) Abnormal blood components (Hypercoagulability - excessively easy clotting of blood) –> alterations in the constitution of blood caused by smoking, sepsis (reaction to an infection), malignancy/cancer – can be due to genetics also - Gene mutations like in essential thrombocythemia (high number of platelets in blood)

3) Abnormal blood vessel wall (Endothelial injury) –> can be from NICOTINE (smoke), atheroma formation (Fatty substance build up in arteries), inflammatory response, surgery, direct trauma, etc.

atheroma = plaque

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

What are some risk factors of developing a venous thromboembolism?

Need to check with lecture notes

A

Obesity,
hormone replacement therapy, increasing age
injury/trauma
Smoking
Prolonged immobilisation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is atherosclerosis?

A
  • The accumulation of fibrolipid plaques in systemic arteries. It narrows the arteries, reducing blood flow to important areas and thus cause illness e.g. MI of the heart

Take note: not really found in low pressure systems (pulmonary arteries). common in high pressure systems (aorta/systemic arteries)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What are plaques that build up in arteries composed of?

A

Lipid, smooth muscle, macrophages + foam cells (macrophages that ingest LDLs), platelets, calcium, fibroblasts, T lymphocytes

Lipid, necrotic core, connective tissue, fibrous cap.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Describe the formation of an atherosclerosis

Look into lectures about smooth muscle cap.

A

1) Fatty streak –> Precursor turns into plaque (late teenage/early 20s) (consists of lipid laden macrophages and T lymphocytes within the INTIMAL layer of the vessel wall)

2) Lipid accumulation –> Endothelial damage initiates an INFLAMMATORY response. This results in monocytes and macrophages being recruited to phagocytose LDLs to become foam cells, which contribute to plaque formation.

3) Platelet aggregation (due to damage of endothelial lining) –> Accumulated lipids lead to plaque protruding into the artery lumen, causing platelet aggregation

4) Smooth muscle cells contribute to the formation of the fibrous cap over the plaque by releasing fibroblast growth factor producing collagen and elastin. (stabilising the plaque - stable atheroma)
atheroma=plaque

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

What is a risk in an advanced stage of atherosclerosis?

A
  • Unstable atheroma (plaque)

The fibrous cap covering the plaque may weaken or rupture. If it ruptures, exposing the plaque’s contents, it will trigger platelet activation which leads to platelet aggregation at the site of the plaque disruption –> leading to the formation of clots within the arterial lumen –> impedes blood flow –> can lead to acute cardiovascular events (MI, stroke)

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

What are some risk factors for atherosclerosis?

A

Smoking, diabetes, hypertension, obesity, hyperlipidemia increased age, males

(Also risk factors for MI)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Briefly what is the pathogenesis of atherosclerosis?

A

ENDOTHELIAL DAMAGE THEORY

Endothelial cells are delicate and can be damaged by cigarette smoke (free radicals released), shearing forces at arterial divisions, hyperlipidemia (lipids taken up by macrophages, forming foam cells which lead to plaque forming) and glycosylation products

(Where blood flow is turbulent or low at arterial branch points, that area is more susceptible to plaque buildup)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What are the complications of atherosclerosis when there is single and dual arterial supply?

A

If an atherosclerotic plaque completely blocks an artery (which can be due to a rupturing of the plaque leading to the formation of a thrombus), then no blood will flow to the organ supplied by that artery and unless there is second arterial supply e.g. the brain, the organ will infarct (die).

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

Name a few possible complications of atherosclerosis

A
  • Cerebral infarction
  • Myocardial infarction
  • Emboli (when pieces of the atherosclerotic plaque break off and travel downstream to smaller vessels) –> can cause transient ischemic attacks or cerebral infarcts if in the carotid artery (carotid atheroma)
  • Aortic aneurysm (if atherosclerosis occurs in the aorta, it can weaken the wall of the aorta causing it to be less elastic and causing turbulent blood flow.
  • Gangrene (lack of blood flow and lack of oxygen to extremities can lead to necrosis)- body tissue dies (usually starts in the toes)
  • Intermitted claudication (muscle pain due to lack of oxygen)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

State 4-5 differences been apoptosis and necrosis

for point 4 need to check with lectures

A

Apoptosis –> Non inflammatory, programmed cell death
Necrosis –> Inflammatory, traumatic cell death (due to injury, disease initiated, etc)

Apoptosis –> Cell membrane remains intact
Necrosis –> Disrupted cell membrane (loss of membrane integrity)

Apoptosis –> Cell shrinks (nucleus condenses)
Necrosis –> Cell swells (and bursts)

Apoptosis –> Chromoatin is unaltered. Pyknosis (condensation of chromatin) and Karyorrhexis (fragmentation of the nucleus)

Necrosis –> Chromatin is altered - degrades and fragments(could mutate). Pyknosis, karyorrhexis and karyolysis occurs (dissolution of cytoplasm)

Apoptosis –> Energy dependent process (and therefore controlled)
Necrosis –> Energy independent

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

How does a cell apoptose?

A

The cell triggers a series of proteins which lead to the release of enzymes like caspases which autodigest the cell.

Cascade of activated enzymes.

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

What is polygenic inheritance?

A

The inheritance of a trait (e.g. height, skin colour) influenced by more than one gene.

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

How does a cell decide to apoptose?

Why does this not work in cancer cells?

What happens in HIV?

A
  • Via the amount of DNA damage within the cell. p53 is a protein that can detect DNA damage and can then trigger apoptosis.
  • In cancer cells, there is a mutation of the p53 protein so it can no longer detect DNA damage and induce apoptosis.
  • In HIV, the HIV virus can induce apoptosis in CD4 helper cells which reduce their numbers, resulting in an immunodeficient state.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What kind of DNA damage results in apoptosis of cells?

A
  • Single stranded break
  • Double stranded break
  • Base alteration
  • Cross linkage

(other forms of damage to cellular systems –> accumulation of toxic by-products of metabolism, telomere shortening, damage to mitochondrial DNA, free radical generation)

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

Name 3 specific types of ageing in parts of the body

A

Wrinkling of the skin (dermal elastosis) - caused by UV-B light (component of sunlight) cross linking proteins, especially collagen in the dermis - can be reduced by avoiding too much sun exposure/using sun screen.

Cataracts of the eye - caused by UB-B cross-linking proteins in the lens causing opacity. - Prevented by wearing sunglasses that cut out UV light.

Osteoporosis in post menopausal women - loss of bone matrix is due to a significant decline in estrogen levels which help maintain bone density (increased bone resorption, decreased bone formation). - prevented by hormone replacement therapy and vitamin D supplements.

Deafness - Hair cells in the cochlear do not divide. If they are damaged by high volumes, they will die and not be replaced, thus producing deadness. - Prevented by avoiding high volume sounds throughout life.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What is caseous necrosis?

A

A distinct form of necrosis recognised by its cheese-like crumbly appearance. It is associated with granulomatous infections like tuberculosis - tissues cellular material breakdown (stain for mycobacteria).

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

What are the 3 apoptosis pathways (mechanisms)?

A

Intrinsic, extrinsic and cytotoxic

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

Describe the apoptosis intrinsic pathway.

A

Occurs due to internal stimuli (DNA damage, biochemical stress, lack of growth factors)

  • Pathway modulated by the molecules Bcl-2 (inhibits Bax) and Bax (promotes cytochrome C release)
  • Cytochrome C activates caspases - the executioners of apoptosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

Describe the apoptosis extrinsic pathway

A

Occurs due to external stimuli
- Ligands bind to receptors on the cell surface

Tumour necrosis factor receptor (TNFR) binds to TNF ligand. FAS receptor binds to FAS ligand –> these interactions activate caspases for apoptosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

Describe the apoptosis cytotoxic pathway

A

Cytotoxic T cells release granzyme B and perforin which activate caspases for apoptosis. (perforin creates pores or channels in the target’s cell membrane for granzyme B to enter the cell and activate caspases)

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

Definition of hypertrophy

Give examples

A

Hypertrophy –> Increase in size of an organ caused by an increase in the size of its constituent cells (without an increase in number)

(occurs in organs where cells cannot divide)
Examples –> skeletal muscle in athletes/body builders

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

Definition of hyperplasia
(What is mixed hypertrophy/hyperplasia)?
Examples

A

Hyperplasia –> Increase in size of an organ caused by an increase in the number of its constituent cells(via mitotic replication)

occurs in organs where cells can divide.

e.g. benign prostatic hyperplasia, endometrial hyperplasia (irregular thickening of uterine lining)

Take note - mixed hypertrophy/hyperplasia - is an increase in the size of an organ due to increases in the size and number of its constituent cells (e.g. smooth muscle cells of the uterus during pregnancy)

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

Definition of metaplasia and dysplasia

A

Metaplasia - The change in cell differentiation from one fully differentiated type to another fully differentiated type
e.g. Barrett’s oesophagus (squamous to glandular/columnar), bronchial epithelium from ciliated columnar to squamous epithelium due to continuous cigarette smoke
(Usually due to a consistent change in the environment of an epithelial surface)

Dysplasia - basically ABNORMAL GROWTH/DEVELOPMENT OF CELLS - morphological changes that may be seen in cells in the progression on to development of cancer (dysplastic cells are considered pre cancerous)

Metaplasia –> dysplasia –> carcinoma in situ (pre-invasive stage of cancer development - has not breached membrane) –> invasive carcinoma (malignant, it has the potential to invade and damage surrounding tissues)

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

What is a common pathway with chronic inflammation?

A

Chronic inflammation –> metaplasia –> dysplasia –> carcinoma in situ (hasn’t breached membrane) -> invasive carcinoma

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

Definition of atrophy

Examples

A
  • A decrease in the size of an organ due to a decrease in the size or number of its constituent cells via dying(or both)

Main thing is a decrease in the size of an organ/tissue

Example - Alzheimer’s dementia (cortex becomes smaller), quadriceps muscle following knee injuries or immobilisation

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

What does it mean to have a congenital, inherited or acquired disorder?

A

Congenital - present at birth
Inherited - caused by an inherited genetic abnormality
Acquired - caused by non-genetic environmental factors

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

What is the hayflick limit and why is it a thing?

A

Hayflick limit - The limit to which a human cell can divide.

There is a limit because at each cell division, the telomere region at the end of chromosomes shortens and eventually becomes so short that it is not possible for chromosomes to divide and replicate. (telomere length appears to be paternally inherited)

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

Definition of carcinogenesis and neoplasm

A

Carcinogenesis –> The transformation of normal cells into neoplastic cells (cancer cell) through permanent mutation leading to uncontrolled/abnormal growth (possibly into a tumour subsequently)

Neoplasm –> The Autonomous, abnormal, persistent new growth of cells

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

What does the structure of a neoplasm consist of?

A

Neoplastic cells + stroma.

(Cancer cell)
A type of nucleated cell that has undergone genetic mutations that lead to uncontrolled or abnormal growth. These cells divide and proliferate in an unregulated manner forming a mass of of cells - Tumour. They are monoclonal. (growth pattern and synthetic activity are related to parent cell)

A vascular stroma which provide mechanical support and nutrition for the neoplastic cell grows based on growth factors released by the neoplastic cell. (a connective tissue framework)

(They arise from NUCLEATED cells)

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

What are the characteristics of a neoplastic cell?

A

Autocrine growth stimulation - due to overexpression of growth hormone and mutation of tumour suppression genes. - very fast growth

They can evade apoptosis

They have telomerase - Prevents telomere shortening with each replication

Sustained angiogenesis - They have their own blood supply

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

Difference between benign and malignant tumour (behavioural classification of neoplams- benign, borderline, malignant)

A

Benign - localised and non invasive - (no basement membrane invasion)
Malignant - metastases and invasive - basement membrane invading

Benign - Slow growing due to low mitotic activity
Malignant - Fast growing due to high mitotic activity (hyperdense nucleus)

Benign - Well circumscribed (or encapsulated) (restricted)
Malignant - Poor circumscription (or encapsulation) (poorly defined or irregular border of the neoplasm)

Benign - Exophytic - grows upward and outward from surface epithelium
Malignant - Endophytic - grows inward from surface epithelium

Benign - Rare ulceration (breach in mucosal surface) and necrosis
Malignant- Common ulceration and necrosis

Benign - Nuclear shape often normal
Malignant - Hyperchromatic and pleomorphic nuclei (can take different shapes)

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

What are the 3 main histogenesis (need microscope) classes of tumours? - Describe them slightly

A

Carcinomas- Epithelial tumours. Cancer that starts in cells that make up the skin or tissue lining organs.

Sarcomas - Mesenchymal tumours.
Cancer of soft tissues, connective tissue or bone (invade the LUNGS commonly)

Lymphoid - Cancer of the lymph system or blood cells (lymphoma and leukemia)
lymphoma - malignant neoplasm of lymphoid cells.
(melanoma, mesothelioma –> malignant neoplasms)

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

What are tumours called when they are non/glandular benign/malignant?

A

They are Epithelial tumours

Papilloma - non glandular benign
Carcinoma - non glandular malignant
Adenoma - glandular benign
Adenocarcinoma - glandular malignant

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

What are sarcomas? Give examples

A

Cancer of soft tissue, connective tissue or bone - MESENCHYMAL tumours

Lipoma and liposarcoma - adipocytes

Rhabdomyoma (benign) and rhabdomyosarcoma (malignant)- striated muscle

Leiomyoma and leiomyosarcoma - smooth muscle

Chrondroma and chondrosarcoma - cartilage

Osteoma and osteosarcoma - bone

Angioma - Vascular/blood vessels

Neuroma - Nerves

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

What are lymphoid tumours - give examples

A

Cancer of the lymph system or blood cells

Leukemia and lymphoma (always malignant) –> Blood disease

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

Describe melanoma and mesothelioma

A

Melanoma - melanocyte malignancy (skin cancer)

Mesothelioma - Cancer of the mesothelium, typically pleura of the lungs (associated with ASBESTOS - microscopic particles that can be inhaled - long term exposure can cause cancer)

Both malignant

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

How are tumours graded?(in terms of differentiation to parent cell)

A

They are graded based on their similarity to the parent cell

1) Well differentiated (>75% cells resemble the parent)
2) 10-75%
3) Poorly differentiated (<10% cells resemble parent)

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

State some classes of carcinogens

A
  • agents capable of causing cancer

1) Chemicals - e.g. paints, dues, rubber, soot. (most require metabolic conversion from pro-carcinogen to ultimate carcinogens)
2) Viruses - e.g. HPV (cervical cancer), EBV (burkitt lymphoma) - causes 10-15% of cancer
3) Ionising + non-ionising radiation - UVB (or UVA) (skin cancer) - basal cell carcinoma, squamous cell carcinoma (increased risk of xeroderma pigmentosum)
4) Hormones, parasites, mycotoxins - mycotoxins can be found in food/fungi/mold (aflatoxin - hepatocellular carcinoma)
5) Abestos (fibrous minerals that are microscopic and can be inhaled)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

What are the 3 main methods of tumour spread?

A

1) Haematogenous - Via the bloodstream –> common in cancers that spread to the bones, lungs and liver - breast cancer. prostate cancer
(Carcinomas spread to the bone via blood)

2) Lymphatic –> spread via the lymphatic vessels - they travel in lymphatic fluid to lymph nodes –> seen in breast cancer, melanoma
(carcinomas spread to the lymph nodes that drain the site of the carcinoma)

3) Transcolemic –> Cancer cells spread within body cavities (e.g pleural, peritoneal) - via exudative fluid accumulation (pleural, peritoneal, pericardial effusions)
- they form tumour deposits on the surface of organs and tissues –> ovarian cancer (peritoneal cavity)

A direct metastases is the spread of cancer cells from a primary tumour to nearby tissues of organs. (This is without travelling through the bloodstream or lymphatic system)

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

What are the 5 main metastasis to bone? (Cancers that commonly spread to bone)

A

BLT.KP
Breast, lung, thyroid, kidney, prostate

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

Which path of metastases do sarcomas and carcinomas mostly spread through?

A

Sarcomas - haematogenous

Carcinomas - lymphatic (exceptions- follicular thyroid, RCC, HCC)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

What do the stages of a cancer describe? (TNM)

A

Tumour grading - How differentiated the tumour cell is. - well differentiated resembles its original counterpart. (well differentiated tumours have a better prognosis as they tend to grow and spread more slowly)

Tumour staging - How much the tumour has spread (TNM system)

T - size and extent of invasion of the main tumour
N - number of nearby lymph nodes that are involved (have cancer) - extent of lymph node metastases
M - whether the cancer has metastasised - extent of spread to other parts of the body

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

Which is the exception of histogenesis tumour class that is not measured via the TNM system?

Which system is used to describe the severity of it?

A

Lymphomas –> uses Ann Arbour Staging system (stages 1-4 and A or B)
A - asymptomatic, B - presence of B symptoms
1-4 (extent of disease and degree of metastases)

TNM more for solid tumours

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

What are 2 specific mutations involved in colorectal cancer?

A
  • FAP (familial adenomatous polyposis) –> mutated APC (adenomatous polyposis coli) gene, can result in thousands and millions of colorectal adenomas forming - benign polyps that have potential to become cancerous
  • HNPCC (lynch syndrome) –> mutated MSH2 gene (DNA mismatch repair gene)

Both autosomal dominant

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

What layer of prevention is screening?

What kind of cancers are being screened in the UK and what is the method?

What is the Heel prick test used for and when is it done?

A

Secondary prevention - method of early detection which makes management easy

  • Cancers screen include: Breast (mammogram), Cervical (cervical swab), Colorectal (Fecal occult)
  • Heel prick test done at birth is to test for sickle cell anaemia, cystic fibrosis, hypothyroidism, etc.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

Draw the multipotential hematopoietic stem cell pathway (hemocytoblast)

A

refer to phase 2a ics document

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

What are dendritic cells and what is their origination?

A

Dendritic cell - Antigen presenting cells that initiate the adaptive immune response.

They are mesenchymal in origin and not hematopoietic.

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

Which cells are responsible for innate immunity, parasites, allergy and active immunity?

A

Innate immunity - Neutrophils (short lived 2-3 days) and macrophages (long lived months to years)
Parasites - Eosinophil
Allergy - Basophil
Adaptive immunity - T cells and B cells (plasma cells)

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

What are primary and secondary lymphoid organs (name some examples)?

A

Primary –> Where immune cells originate, mature or develop
- Bone marrow (all cells originate here including T cells) - B cells mature here
- Thymus - development and maturation of T cells (thymic tolerance)

Secondary –> Where immune responses are initiated and coordinated
- Lymph nodes - antigen presenting cells and T/B cell interactions –> filters lymphatic fluid allowing immune cells to encounter and respond to antigens
- Spleen - RBC recycling, bacteria killing - it has lymphocytes and macrophages which can respond to infections

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

What is thymic tolerance and where does it occur?

A

A critical process that occurs in the thymus where T cells are developed to recognise and respond to foreign antigens while remaining tolerant to the body’s own tissues thus preventing autoimmune reactions. (T cell selection)

Positive selection - If the T cells recognise the thymus Major histocompatibility complexes (MHCs 1 and 2), then they are selected for

Negative selection - If the T cells produce an immune response (by recognising self antigens as foreign, they are selected against.)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

What are tertiary lymphoid organs?

A

This is pathological and only found in sites of chronic inflammation

They are specialised collections of immune cells that form in response to chronic inflammation or infection.

They sustain immune responses - allowing the continual combating of chronic inflammation

There are germinal centres within, where lymphocytes undergo rapid proliferation (differentiation and antibody productioin)

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

How are CD8+ and CD4+ cells allocated for and what are their functions?

A

T cells that interact with MHC 1 become CD8+ cells (Cytotoxic T cells) INTRINSIC
–> They function to kill

T cells that interact with MHC 2 become CD4+ cells (Helper T cells)
EXTRINSIC
–> They promote the immune response

Elaborated more in a different flash card

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

What is the function of CD8+ cells?

A

Cytotoxic T cells –> involved in eliminating target cells

1) They secrete perforin which creates pores in the target cell’s membrane, allowing Granzyme B to enter and induce apoptosis.

2) They express the Fas ligand which bind to Fas receptors on the cell’s surface, activating caspases which are the main executioners of apoptosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

What is the function of CD4+ cells?
T regulatory cells

A

(T cells that interact with MHC 2)Helper T cells –> Promotes immune response
cytokines - signalling molecules

1) T helper 1 cells secrete the cytokine Interferon-y, which activates natural killer cells and macrophages (increases innate immune response)

2) T helper 2 cells secrete the cytokine interleukin 4 (5 and 10), which activates B cells to differentiate into plasma cells (increases adaptive immune response) - they also activate eosinophils and induce b cells to make IgE to promote release of inflammatory mediatory - important in helminth (parasitic) infections and allergies.

-
They also help with development of cytotoxic T cells

3) T regulatory cells also referred to as CD4+ cells
- Suppress immune responses (helps with preventing autoimmunity)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

What is a naive T cell?

A

A T cell that has not encountered an antigen or has not matured

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q
A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

Describe the innate immune system

A

1) Non specific and rapid immune response with no memory involved

2) Neutrophils and macrophages are primarily involved

3) Killing occurs via the complement system: Activation of inflammation, opsonization of pathogens, phagocytosis and killing of target cells, by lysis

4) Includes physical barriers like the skin, mucus and cillia
and chemical barriers like lysozyme in tears and stomach acid, saliva.

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

Describe the 5 main immunoglobulins

A

IgG
- The most abundant
- Key in SECONDARY immune response
- Found in bacterial and viral infections

IgA
- Secreted in breast milk and mucosa
- Defends mucosal surfaces
- Forms dimer (effective in defending mucosal surface)

IgM
- The first antibody produced in response to foreign pathogens
- Forms pentamer

IgE
- Antibody that is produced in response to allergies (type 1 hypersenstivity)

IgD
- Unknown function but presumably B cell activation

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

Describe neutrophil, macrophage, eosinophil, basophil and natural killer cells.

A

Neutrophil
- 70% of all WBCs
- Key mediators of acute inflammation
- Act in hours - days
- They express CD 66 which allows neutrophils to adhere to blood vessel walls and migrate to sites of infection.

Macrophage
- Clear apoptotic debris
- Act in months - years –> can be circulating or resident (e.g. kupffer cells, alveolar macrophages)

Eosinophil
- Contain major basic protein
- Often seen in parasitic infection

Basophil/mast cells (resident version of basophil)
- Induces type 1 hypersensitivity reaction (allergy)
- Via IgE binding –> degranulation –> histamine release (promotes inflammation)

Natural killer cells
- Key role in viral cell killing via secretion of perforin

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

What are the 2 scenarios that can happen when dendritic cells present foreign antigens to helper T cells?

A
  • Stimulation of helper T cell proliferation
  • Stimulation of B cell production and differentiation to form antibodies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

3 differences between innate and adaptive immunity

A

Innate - non specific, rapid response with no memory
Adaptive- specific, slow response with memory involved

Innate - Neutrophils and macrophages primarily involved
Adaptive - T cells and B cells are primarily involved

Innate - Killing via complement system
Adaptive - Killing is antibody mediated

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

Type 1 hypersensitivity

A

Anaphylaxis
- IgE mediated
IgE binds to basophils and mast cells –> upon re-exposure, degranulation of mast cells –> release of histamine which causes vasodilation, bronchoconstriction and increased permeability of vascular endothelium

(urticaria, angio-oedema)

Timing - Immediate (within an hour)
Causes an inflammatory response

Example - Atopy (genetic tendency to develop allergic disease), hayfever, asthma, eczema

Occurs when an individual is sensitised to a specific allergen and upon re-exposure of the allergen, IgE antibodies are produced which trigger the release of histamine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

Type 2 hypersensitivity
(Antibody involved, process, timing, examples)

A

Antigen-antibody complex
Antibody - IgM, IgG
Cytotoxic response

Antibody binds to the antigen (e.g. drug that is combines with a protein) –> antibodies activate the complement system. Degranulation of neutrophils (and release of their antimicrobial contents) –> oxygen radicals are released (respiratory burst) leading to the destruction of the cell.
Timing - hours to days
Examples - Good pastures syndrome, pernicious anemia, rheumatic fever

Basically –> categorised by the binding of IgM or IgG to antigens on the surface of target cells. It triggers an immune response that results in the destruction of the cell via the complement system

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

Type 3 hypersensitivity (Antibody, process, timing, examples)

A

Immune complex formation
Antibody - IgG

Difference between type 2 and 3 - type 2 involves antigens that are cell bound (ABO to RBC) while type 3 involves soluble antigens

The antibody and antigen bind, move somewhere downstream and activate the complement system at the site of deposition.
Timing - 7-21 days
E.g. SLE (Systemic lupus erythematosus), farmer’s lung, malt worker’s lung

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

Type 4 hypersensitivity

A

T cell mediated

When antigens enter the body, it is processed by antigen presenting cells and presented to a Th1cell –> activating the T cell which releases chemokines to recruit macrophages and cytokines e.g. interferon-y to activate them

Timing- Days-weeks
DELAYED response
e.g. tuberculosis, type 1 diabetes mellitus, MS, guillain barre syndrome.

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

What are symptoms that may present with anaphylactic shock (an acute medical emergency)?

A
  • Severe hypotension
  • Tachycardia + dyspnoea
  • Pale
  • Cold extremities
  • Puffed up face + tongue
  • Itching
  • Urticaria (Hives)
  • Central cyanosis (lips are blue)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

How would you manage a person with anaphylactic shock?

A

Airway - are they breathing? Are there any signs of airway obstruction? Secure the airway with intubation PRN.

Breathing - any signs of respiratory distress? Wheezing? SPO2<94%? Provide oxygen if required (salbutamol can help with wheezing)

Circulation - Check pulse and BP. Are they pale or cold? Provide an IV bolus PRN - start CPR if appropriate and needed. (IV bolus improves circulation by rapidly delivering a concentrated dose of a substance into the bloodstream)– saline solution can restore fluid balance and increase blood volume (also addresses dehydration)

Disability - Assess patients level of consciousness and mental state - lie patient flat to improve cerebral perfusion (may need to inject drugs to help e.g. epinephrine) A sitting position may help to make breathing easier

Exposure - Look for visible signs of an allergic reaction e.g. urticaria, flushing, angioedema → remove any triggers.

As soon as symptoms are recognised, administer 500mcg of IM adrenaline.
Consider antihistamine (Chlorphenamine) - not for first line but for skin symptoms,

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

What is immune tolerance?

A

A safeguard mechanism to prevent the production of autoreactive cells (where autoreactive B and T cells infiltrate and attack healthy tissues and organs - producing antibodies with high affinity for self antigens)

Split into

Central tolerance - which occurs in primary lymphoid organs. Thymus for T cells and bone marrow for B cells (where they mature)

Peripheral tolerance - occurs in secondary lymphoid organs e.g. Spleen (in the event faulty T and B cells evade central tolerance)

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

What is autoimmunity? What are the 2 categories?

A

System of immune responses of an organism against its own healthy cells, tissues, organs, etc.

It can either be organ specific or non organ specific.

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

What is organ specific autoimmunity?
Examples

A
  • It affects a main organ
    e.g.
    1) Type 1 diabetes mellitus - affected endocrine pancreas where beta cells producing insulin are destroyed

2) Multiple sclerosis - Oligodendrocytes which myelinate cells of the CNS are attacked

3) Graves disease - Thyroid stimulating hormone receptors are stimulated (by TSH receptor antibodies) –> causing the secretion of thyroid - Hyperthyroidism.

4) Myasthenia gravis - antibodies attack acetylcholine receptors on muscle cells (neuromuscular junction) (interferes with transmission of nerve signals to muscles thus leading to muscle weakness and and fatigue)

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

What is non organ specific autoimmunity?
Examples

A

Autoimmunity having a generalised effect.

E.g. Systemic lupus erythmatosus
- The immune system attacks multiple organs and tissues.
- In SLE, individuals produce anti-DNA antibodies which forms an immune complex with DNA molecules. These circulate and deposit in various tissues and organs, triggering inflammation, leading to tissue damage. (joint inflammation, skin rashes, etc) Also affects lungs, brain, kidney, etc.

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

Immunodeficiency can be acquired or inherited, give examples of both.

A

Inherited (genetic) - Defects in T cells e.g. SCID- severe combined immunodeficiency (caused by adenosine deaminase deficiency, where infants lack the enzyme required for T-cell survival)

Acquired- HIV (Unprotected sex, sharing injection drug equipment)

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

What are some examples of how you may be immunodeficient? (patterns of immunodeficiency)

A
  • In HIV, there is a decrease in helper T cells, increasing your susceptibility to diseases. (present with TB, pneumocystic pneuomonia - alveoli attacked)
  • B cells can be deficient
  • Neutrophil and macrophage deficiency (required for phagocytosis and acute inflammation)
  • Complement deficiency - Low C3 and C4 proteins which are required for activation of the complement cascade (innate immune killing of bacteria) –> Associated with systemic lupus erythmatosus
  • Hyposplenism - Lack of function of the spleen - less RBC cycling, less killing of encapsulated bacteria
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

What are covid 19 vaccines (pfizer and moderna) made of?

A

mRNA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

What are the differences between active and passive immunity?

A

Active
- our own immune system produces immunoglobulins to protect us against a pathogen
- Immunological memory - memory cells produced for long term protection
- Secondary response (upon re-exposure to pathogen)

Passive - The administration of PRE-FORMED immunity from one person or animal to another person
- Immunoglobulin passed to host (only antibody mediated, not cell mediated)
- No memory
- Primary response - immediate and temporary effect
- Short lived

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

What are examples of active natural, passive natural, active artifical and passive artificial immunity?

A

Active natural - Body encounters pathogen and produces memory cells against it (after the infection)

Passive natural - Maternal antibodies/immunoglobulins are passed onto feeding baby in breast milk

Active artificial - Vaccine mimics encountering pathogen and stimulates immunoglobulin production

Passive artificial - Antivenom (injection of immunoglobulin from another organism)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

What are the methods of drug administration?

A

Enteral (via the GI tract)
1) Oral - mouth
2) Suppositories - via the rectum

Paranteral (bypasses the GI tract)
3) Injection
- Intramuscular - injected directly into muscle
- Subcutaneous - drug injected directly under the skin
- Intravenous - injected directly into bloodstream

4) Inhalation - vapour
5) Skin patches/topical - transdermal
6) Eye/ear drops

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

What are the 4 main drug targets?

A

1) Receptors (Majority)
2) Enzymes
3) Transporters
4) Ion channels

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

Where drugs target receptors, the ligands can either be agonists or antagonists. What is the difference in terms of affinity and efficacy with the receptor?

What is the most common type of receptor?

A

Agonists - full affinity (binds well to receptor) and full efficacy (fully activates the receptor)

Antagonists - full affinity and zero efficacy (inactivates the receptor)
(Can competitively or non competitively inhibit receptors)
(irreversible antagonist – won’t come off the receptor and will never become available for an agonist)

Most common receptors are G-protein coupled receptors.

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

What is the definition of efficacy and potency?

A

Efficacy- The maximum effect a drug can produce, regardless of dose (the ability of a drug receptor complex to produce a maximum functional response)

Potency - How much of a drug is needed to elicit a response in the body

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

How is potency and efficacy of a drug/agonist affected by a competitive and non competitive antagonist?

A

Competitive –> Potency is decreased, efficacy is not. Ligand concentration is rate limiting.
Thus, an increase in agonist concentration can overcome this.

Non competitive –> Both potency and efficacy is decreased. Ligand concentration is not rate limiting.

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

Examples of non-selective/selective beta blocker/agonist.

A

Non-selective beta blocker –> binds to all beta receptors (B1 and B2) - Propanolol

Selective beta blocker –> binds to a specific beta receptor (B1) - Atenolol, bisoprolol

Non-selective beta agonist –> B1, B2 - Epinerphrine, isoprenaline (so efficacy and potency for B2 and B1 will be similar)
Selective beta agonist (B2) - Salbutamol (Efficacy is similar but potency for B2 receptors is higher then B1)

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

Describe the function of COX1 and ACE Inhibitors. And what kind of drugs target them.

A

COX-1 - Important function in producing prostaglandins which protect the stomach lining. (it is also important in blood clotting pathways - production of thromboxane A2)

NSAIDS - inhibit COX-1 and COX-2, decreasing prostaglandins production.
Inhibiting COX-1–> decreased prostaglandins–>decreasing gastric mucosal protection, decreasing stomach pH resulting in gastric ulcers (and risk gastrointestinal bleeds). Inhibiting COX-2 –> Decreased prostaglandins –> has an anti-inflammatory effect
(basically both COX-1 and 2 are responsible for producing prostaglandins but for different things)

ACE inhibitors – inhibit the conversion of angiotensin 1 to angiotensin 2 –> results in an antihypertensive response, reduced aldosterone, hyperkalemia (as principal cells no longer secrete as much potassium)

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

How are transporters targeted by drugs?(which pumps/channels do they act on)

A

Proton pump inhibitors e.g omeprazole –> irreversibly inhibits H+ K+ ATPase pumps (decreasing gastric pH)

Diuretics (reduces fluid buildup by promoting the excretion of NaCl and water)
- Loop diuretics act on the symporter in the ascending loop of henle to inhibit the reabsorption of sodium, chloride and potassium (NKCC2) e.g. furosemide

  • Thiazide diuretics inhibit the sodium chloride cotransporter in the DCT (increasing excretion of sodium and potassium)

Spironolactone diuretics - Aldosterone receptor antagonist. A potassium sparing diuretic which acts on ENaC channels of the collecting duct, causing the excretion of sodium and water.

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

What are some ion channels targeted by drugs?

A

Calcium channel blockers (muscle + glial cells)- cause vasodilation and decreased contractility. It slows the heart rate - amlodipine.

Local anaesthesia - e.g. Lidocaine blocks Na+ voltage gated channels in nerve cells, preventing the propagation of nerve impulses.

Potassium channel blockers - e.g. Repaglinide, nateglinide lower blood glucose levels by blocking potassium channels to stimulate insulin secretion. –> Type 2 diabetes treatment

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

What is pharmacokinetics?

A

The fate of a chemical substance administered to a living organism

((It examines how drugs are absorbed, distributed, metabolised and excreted in the body. ))(what the body does to the drug)

(Pharmacodynamics - what the drug does to the body)

107
Q

What are the 5 steps of neurotransmission where drugs can disrupt synaptic transmission?

A

1) Manufacture (synthesis)
2) Storage (in vesicles)
3) Release
4) Receptor activation and breakdown of neurotransmitter
5) Reuptake

108
Q

What are the effects of botulinum toxin (botox) and acetylcholinesterase inhibitors at neuromuscular junctions?

A

Botox - Acetylcholine release is inhibited which can lead to paralysis

Acetylcholinesterase inhibitors - Inhibits the enzyme acetylcholinesterase from breaking down acetylcholine, increasing the level and duration of cholinergic stimulation. (can result in cholinergic crisis)

109
Q

What are the symptoms of a cholinergic crisis, when does it happen? (also symptoms of organophosphate poisoning)

A

It happens when there is an increased level and duration of cholinergic stimulation (due to inhibition of acetylcholinesterase enzyme) - basically has an agonist effect. (antagonist effect with TCA drugs will have opposite effect of sludge)

SLUDGE
Salivation
Lacrimation
Urination
Defecation
GI distress
Emesis (vomiting)
—— These are all due to excessive parasympathetic nervous stimulation.

Can also lead to severe muscle weakness, paralysis and respiratory distress.

TREAT WITH ATROPINE (as it crosses BBB can cause delirium, confusion and hallucination)

110
Q

Which cells can regenerate and cannot regenerate?

state 2 each

A

Regenerate - Hepatocytes, pneumocytes, skin epithelium (blood cells, osteocytes)

Cannot regenerate - Myocardial cells, neurones.

111
Q

4 examples of macrophages

A

Kupffer cells, alveolar macrophages, osteoclasts, microglia.

112
Q

What is the function of precapillary sphincters and what happens during sepsis?

A

(Sepsis occurs when inflammation spreads through your entire body)

During inflammation, precapillary sphincters relax which allows for increased blood flow to the site of injury/infection.

During sepsis, BP decreases as blood volume is the same while area for blood flow increases (when all the pre capillary sphincters open)

113
Q

Why is there low blood pressure in sepsis?

A

Sepsis is caused by the body’s extreme response to infection. It leads to widespread inflammation, causing widespread vasodilation. Endothelial cells also become more porous, allowing fluid and proteins to leak from the bloodstream to surrounding tissues –> thus decrease in blood volume.

114
Q

Name 3 organs with dual arterial supply

A

Brain, liver, lungs

115
Q

What is the most common cause of gastritis?

A

Helicobacter.Pylori

116
Q

Body responses - Parasympathetic vs sympathetic

A

Parasympathetic
- Rest and digest
- Pupil constricts
- Bronchoconstriction
- Increased GI motility and secretion
- Bladder contraction (detrusor muscle)
- Penis erect
- Decreased heart rate

Sympathetic
- Fight or flight
- Pupil dilates
- Bronchodilation
- Decreased GI motility and secretion
- Detrusor relaxes
- Penis shoots (ejaculation)
- Increased heart rate

117
Q

Noradrenergic pharmacology

A

Tyrosine –> DOPA –> Dopamine –> Noradrenaline –> Adrenaline

118
Q

What does activation of Alpha 1, alpha 2 beta 1 and beta 2 receptors result in?

A

Alpha 1 - Blood vessels and sphincter –> vasoconstriction, bladder contraction and pupil dilation

(alpha 2 - negative feedback - suppresses noradrenaline (catecholamine) release)- slows heart rate

Beta 1 - Heart. –> Agonism increases inotropy (force of contraction of the heart) and increases BP and renin release.

Beta 2 - Lungs. –> Agonism causes bronchodilation (e.g. SABAs and LABAs)

119
Q

When are dopamine agonists and antagonists used?

Give examples of each.

Where are dopamine receptors found?

A

Dopamine agonist (e.g. bromocriptine) –> used in prolactinoma, acromegaly and early parkinson’s

Dopamine antagonist (e.g. metoclopraminde, antiemetics) –> used for nausea and vomiting.

Haloperidol –> used for psychiatric disorders

Dopamine receptors are mostly found in the nucleus accumbens in the brain.

120
Q

What are some GABA agonists?

A
  • It is the main inhibitory neurotransmitter. (reduces neuronal excitability)

e.g. Benzodiazepines - lorazepam, diazepam –> used for anxiety, sleep disorders, alcohol withdrawal, epileptics

121
Q

What are the 4 pain types?

A

Acute - less than 3 months (nociceptive pain - caused by damage to body tissues)

Chronic - more than 3 months

Cancer

Neuropathic (nerve pain - damage to nerves)

122
Q

What is the difference between A delta and C fibres?

A

(They are the first order neurons that synapse with second order neurons in the dorsal grey horn)
Both carry pain, temperature, touch and pressure information

A - thinly myelinated
C - Unmyelinated

A - larger (1.5mm)
C - Smaller (0.2-1.5mm)

A- Associated with sharp well localised pain that first occurs
C- Associated with lingering, poorly localised sensation that follows the first quick sensation of pain

A - Faster conduction speed (5-40m/s)
C - Slower conduction speed (0.5-2m/s)

123
Q

Explain 3 ways that pain is modulated.

A

1) Gate control theory
- By rubbing the painful site, the non-noxious A beta fibres (activated by light touch, pressure) are activated which inhibit the A delta or C fibres causing pain.

2) Periaqueductal grey - descending pain pathway
- When the PAG receives pain signals from the brain, it sends it to the locus coeruleus which releases noradrenaline throughout the CNS –> reducing the perception of pain.
- Noradrenaline can bind to alpha 2 adrenergic receptors on presynaptic nerve cells to inhibit the release of neurotransmitters like substance P or can hyperpolarise the postsynaptic membrane, thus reducing the transmission of pain signals.

3) Medication
- NSAIDS
- Opioids/analgesics
- Local anaesthesia

124
Q

What are the 3 steps of the pain ladder?

Which medication are used for mild, moderate and severe pain?

A

First step - Mild pain –> Non-opioid analgesic with or without adjuvant therapy

Second step - mild to moderate pain –> Weak opioids (hydrocodone, codeine, tramadol) with/without non opioids, with/without adjuvant therapy

Third step - Moderate to severe pain –> Strong opioid (morphine, oxycodone), with/without non-opioid analgesics, with/without adjuvant therapy

Mild - Paracetamol, ibuprofen
Moderate - Naproxen, tramadol
Severe - Morphine

125
Q

What is the Rawlins and Thompson classification of an adverse drug reaction? (types of reaction)

What should you do if you notice an adverse drug reaction?

A
  • All adverse drug reactions should be reported by MHRA yellow card scheme. (ALL ADRs should be reported for black triangle medicines)

Type ABCDEFG

Type A- Augmented
- Most common type of ADR (80%), predictable (COMMON IN CLINICAL TRIALS)
- Dose dependent (too much of the drug) and reversible upon withdrawing drug
(e.g. Bradycardia with beta blockers)

Bizarre -
- Are there any odd responses? Unpredictable?
- Not dose dependent
(Anaphylaxis with penicillins, bleeding with anticoagulants)

Chronic -
- ADRs that continue after the drug has been stopped
- Related to cumulative dose.
(Heart failure with pioglitazone)

Delayed -
ADRs that becomes apparent some time after use of the drug. (Has the patient used the drug in the past that could be an issue now?)
(Leucopenia with chemotherapy)

End of use - (withdrawal) –>
- ADR develops after the drug has been stopped
- Is the patient withdrawing from the drug?
(Insomnia after stopping benzodiazepine)

Failure of treatment
- Unexpected treatment failure (due to drug-drug interaction or drug-food interactions)
- Poor compliance with administration instructions
(Failure of DOAC due to enzyme inducers like carbamazepine)

Genetic
- Drug causes irreversible damage to genome
(Phocomelia in children of women taking thalidomide)

126
Q

What are the 4 main classifications of pharmakokinetics? What are its main influences?

A

Absorption
The transfer of a drug from the site of administration to the systemic circulation
Route of administration (IV is 100% bioavailability)
Absorption is affected by acidity (which can affect drug ionisation), motility e.g. GI motility and the solubility of drugs

Distribution
How the drug is distributed in the plasma according to its chemical properties (hydrophilicity) and size. Are they able to cross the blood brain barrier?
Affected by protein binding - higher protein binding means longer duration of action but reduced distribution.

Metabolism
The chemical transformation of the drug e.g. by CYP 450 in the liver. \
Increased metabolism of the drug means that more will be excreted and less will enter systemic circulation

Excretion
Increased hydrophilicity allows excretion via urine and faeces
Acids are cleared faster if urine is weakly basic and vice versa

127
Q

What do opioids do?

Name 1 example of a naturally occurring and modified opioid

What are some side effects of opioids?

How many percent of morphine is metabolised by the liver in first pass metabolism?

A

Opioids act on receptors in the CNS (GI tract and respiratory centres also) - to inhibit the transmission of pain signals

Naturally occurring - Morphine, codeine

Modified - diamorphine (heroin)

Take note diamorphine is 2x more potent than morphine

Side effects: Addiction, GI (constipation, nausea, vomiting), Respiratory distress (can reduce the brain’s sensitivity to CO2 levels causing respiratory distress), immune suppression

50% of oral morphine is metabolised by first pass metabolism in the liver.

10mg orally is equivalent to 5mg parenterally so make sure to halve dose if giving parenterally

128
Q

What is the definition of tolerance and dependence in opioid use?

A

Tolerance - There is a reduction in agonist effect over time (slow process) – develops with continuous, repeated exposure to high concentrations of a substance. (body becomes less responsive to the drug –> you need a higher dose to achieve the same effect)

Desensitisation - a rapid short term process where the receptor becomes less responsive to a stimulus. Due to uncoupled receptors, internalised receptors (which are engulfed into endosomes), degradation of receptors

Dependence - Psychological state of craving euphoria

129
Q

What are the 4 categories of anti-thrombotic agents? Name examples- briefly how do they work?

A

Antiplatelets (Inhibit platelet activation and aggregation)
- Aspirin - COX1 inhibition –> reduced thromboxane A2 production –> decreased platelet activation and aggregation –> reduced risk of clots forming.
- Clopidogrel - irreversibly binds to P2Y12 receptors, preventing ADP from binding to platelets –> reduced platelet activation and aggregation

Anticoagulants (interferes with coagulation cascade)
- Heparin - enhances activity of antithrombin 3 –> inhibits clotting factors 2 (thrombin) and 10 –> reduced clotting ability

Warfarin –> Antivitamin K (inhibits vitamin K epoxide reductase which activates vitamin K) –> reduces the production of vitamin K dependent clotting factors –> reduced clotting ability (so you give vitamin K to people on warfarin who bleed excessively)

Direct oral anticoagulants (DOAC) - rivaroxaban, apixaban - inhibits factor 10a –> reducing clotting ability - especially used in DVT and PE

Thrombolytics - e.g. Alteplase - tissue plasminogen activator with activates and converts plasminogen to plasmin that breaks down fibrin, dissolving the clot and restoring blood flow. (used in acute MI and ischemic strokes)

130
Q

Common drug side effects of NSAIDS, ACE-inhibitors, proton pump inhibitors, loop diuretics and thiazides, and spironolactone

A

NSAIDS - gastric ulcers, gastric bleeding

ACE inhibitors - dry cough (due to accumulation of bradykinin), low BP, (they dilate blood vessels which can decrease blood flow to the kidney, resulting in acute kidney injury).

PPI - can increase risk of fractures (interferes with bone remodelling)

Loop diuretics and thiazides - Hypokalemia and dehydration

Spironolactone - hyperkalemia

131
Q

What are some steroid side effects?

Check in lectures

A

CUSHINGOIND MAP

Cataract, Ulcer, striae (abdominal), hypertension, increased infection risk, necrosis of bone, growth restriction, osteoporosis, myopathy, pancreatitis.

132
Q

How would you treat a basal cell carcinoma? (what is it?)

A

A basal cell carcinoma of the skin only invades locally (it does not spread to other parts of the body)

  • You treat it with complete local excision
133
Q

What are some symptoms of leukemia?

A
  • Shortness of breath
  • Fatigue
  • Muscle weakness
  • Night sweats
  • Fever
  • Weight loss
  • Spleen enlargement
134
Q

How would you plan for the treatment of breast cancer?

A

First need to confirm the diagnoses of breast cancer.

Has it spread to the axilla?
If yes - Axillary lymph node clearance is needed
If no - has it spread to the rest of the body?
If yes - systemic chemo therapy needed
If no - surgery with or without axillary lymph node clearance.

Take note that even if a tumour is completely excised, micro metastases may be present. Adjuvant therapy can be used (extra treatment after surgical excision- chemotherapy (good for fast dividing tumours - lymphoma, leukemia, but not slow dividing), radiotherapy, hormone therapy - adjuvant anti-oestrogen therapy - Tamoxifen… Herceptin given for HER2 protein treatment)

You do a mastectomy (removal of entire breast) if the tumour is large, you do a lumpectomy (surgical removal of cancerous tumour and a margin of surrounding tissue but preserving as much breast as possible) if the tumour is small.

(also even if excised, it may have spread to other parts of the body already)

135
Q

Define carcinogenesis

A

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

Take note: Oncogenesis is tumour causing.

136
Q

Examples of chemical carcinogens

A

Most chemical carcinogens require metabolic conversion from pro-carcinogen to ultimate carcinogens. (depending on where these enzymes are, the carcinogens can be spread?)

Polycyclic aromatic hydrocarbons
- Lung cancer (smoking) and skin cancer (mineral oils)

Aromatic amines
- bladder cancer (rubber/dye workers)

Nitrosamines
- Gut cancer (processed meat)

Alkylating agents
- leukemia (chemotherapy)

137
Q

Name 5 DNA viral carcinogens

and 1 RNA viral carcinogen

A

DNA Viruses
Hepatitis B virus (HBV)
- Hepatocellular carcinoma

Human papillomavirus (HPV) - Squamous cell carcinomas of the (CAPN’H) - Cervix, anus, penis, neck and head

Epstein Barr virus (EBV) - Burkitt lymphoma (uncontrolled growth of B cells)- nasopharyngeal carcinoma

Human herpes virus 8 - kaposi sarcoma (most well known) - Vascular endothelial malignancy (GI tract)

Merkle cell polyomavirus - merkle cell carcinoma

RNA Virus
Hepatitis C virus (HCV)
- Hepatocellular carcinoma

Human T-lymphotrophic virus = Adult T-cell leukemia

138
Q

Give examples of biological agents (hormones, mycotoxins and parasites) of carcinogens

A

Hormones
- Increased estrogen - endometrial/mammary (breast) cancer + nasopharyngeal carcinoma
- Anabolic steroids - hepatocellular carcinoma

Mycotoxins (toxins produced by moulds/fungi)
- Aflatoxin B1 –> hepatocellular carcinoma

Parasites
- Schistosoma - bladder cancer (from infested water)
- Clonorchis sinensis - cholangiocarcinoma - bile duct cancer(from raw fish)

139
Q

What are the 3 main lymph nodes and which area do they drain?

A

Cervical lymph nodes in the neck –> drain lymph from the head and neck

Axillary lymph nodes in the armpits –> drain lymph from the upper limb, breast and trunk above umbilicus

Inguinal lymph nodes in the groin area –> drain lymph from the lower extremity, genitals, (dorsal perineum)

140
Q

How many percent of cancer is environmental?

A

85%

141
Q

What are some host factors that can affect your risk of getting carcinomas?

A
  • Ethnicity (increased oral cancer in india)
  • Diet (excess alcohol, obesity can increase risk of breast and colon cancer) - exercise can reduce risk of colon and breast cancer
  • Constitutional factors : age, gender (incidence increases with age) –> you can inherit familial polyposis coli (chromosome 5) or familial adenomatous polyposis
  • Premalignant lesions (e.g. colonic polyps, ulcerative colitis, undescended testis)
  • Transplacental exposure (e.g. diethylstilboestrol can increase vaginal cancer)

(unprotected sex can increase risk of HPV related cancer (cervis, penis, oropharyngeal)

142
Q

What is infectious mononucleosis? What is it commonly caused by?

A

(most commonly caused by epstein-barr virus) - Also known as Glandular fever.

Symptoms - Swollen lymph nodes in neck and armpits, fatigue, fever, sore throat, swollen liver or spleen.

143
Q

What cancers does excess alcohol increase the risk of?

What cancers does obesity increase the risk of?

What cancers does unprotected sex increase the risk of?

A

Alcohol - Mouth, oesophagus, liver, colon and breast

Obesity - breast, oesophagus and colon

Unprotected sex - Cervix, penis, oropharyngeal

144
Q

What is a neoplasm?

A

A LESION resulting from the autonomous abnormal growth of cells that persist after the initiating stimulus has been removed.

Neoplasia - the autonomous, abnormal, persistent new growth of cells.

(A new growth) (25% of the population develops neoplasms)

145
Q

What is carcinogenesis? Give an example of it in the colon/lung

A

The formation of a cancer where normal cells are transformed to cancer cells.

Normal colon –> DNA methylation and APC gene mutations –> Adenoma –> further mutation –> carcinoma

Normal lung –> (Chronic lung injury) cells get injured –> cells become atypical - atypical adenomatous hyperplasia –> carcinoma in situ –> invasive carcinoma (progressive accumulating of DNA damage)

146
Q

What are the 2 types of genetic drivers of carcinogenesis? Give examples of each

A

Proto-oncogenes - genes which promote cell growth and survival, promoting carcinogenesis e.g. myc.

Tumour suppressor gene - genes which inhibit cell growth and proliferation, playing a role in inhibiting carcinogenesis. e.g. BRCA, P53

147
Q

How would you acquire genetic changes?

A

Chance - sporadic genetic mutations

Hereditary - inherited genetic mutations (lynch syndrome, xeroderma pigmentosa)

Environment - environmental exposures and toxins (smoking, radiation, asbestos)

Micro-organism - oncogenic virus, bacteria, epstein barr virus, human papilloma virus, H pylori.

148
Q

What are the PT, aPTT and TT tests?

And what is INR - international normalised ratio.

Confirm

A

aPTT- (Partial thrombin time) - Measures how long it takes your blood to form a clot. - in intrinsic pathway

PT- (prothrombin time) Measures how long it takes your blood to clot in the extrinsic pathway.

TT- (thrombin time) Measures the time it takes for a fibrin clot to form in the plasma.

INR- international normalised ratio - used to measure the dose of oral anticogulant medication given, most commonly warfarin. –> the ratio of a patients PT compared with a control.

149
Q

What is protein C and S, antithrombin 3, antiplasmin?

What factors prevent platelets from adhering?

A

Proteins C and S produced by the liver. (Protein S is a cofactor of protein C) - They inactivate factors 5 and 8

(protein C is activated when thrombin is bound to endothelial proteins like thrombomodulin)

Antithrombin 3 - anticoagulant - inhibits action of thrombin and some other clotting factors.

Antiplasmin - protein inhibiting the activity of plasmin (antiplasmin maintains the stability of clots once they are formed)

Nitric oxide and prevents platelets from adhering

Prostacyclin decreases thromboxane A2 which in turn prevent platelets from adhering.

150
Q

What is disseminated intravascular coagulation?

A

The paradoxical combination of haemorrhage and thrombosis.

Caused by
- infection
- neoplasm
- major trauma e.g. burns
- liver disease

Uncontrolled activation of clotting factors and the coagulation cascade –> widespread clotting –> body uses up platelets/fibrin/clotting factors –> haemorrhage

From excessive clotting (ischemia, impaired organ perfuion, end-organ damage). –> to excess bleeding (shock, hypotension, increased vascular permeability). 2

151
Q

What is a carcinoma in situ, micro-invasive carcinoma and invasive carcinoma?

A

Carcinoma in situ - (first mutations produce cancer celles) The tumour/abnormal cells have not breached the basement membrane – if you excise it at this stage, there is no where for the carcinoma to spread.

Micro-invasive carcinoma - A tumour that can metastasise to other parts of the body but low chance. e.g.Cervical

Invasive carcinoma - Spreads further than micro-invasive carcinomas, they breach the basement membrane into the extracellular matrix. e.g. invasive ductal breast carcinoma.

152
Q

Describe the process of metastasis (spread of tumour)

A

5 step process
1) The tumour breaches the basement membrane (requires enzymes e.g. proteases and cell mobility)
2) Intravasation - Migration into vessels (usually lymphatic vessels and venules - small thin walled vessels) –> requires collagenase
3) Survival in the circulating system - due to lymphocytes an immune response will be triggered –> tumour cells A) Aggregate with platelets B)Shed surface antigens which lymphocytes pickup or C) Adhere to other tumour cells to resist the immune response
4) Extravasation - (exit of circulating tumour cells) –> Can be done via collagenase or adhesion receptors (allow for rolling and margination of tumour cells and eventual emmigration and diapedesis) into normal tissue
adhering allows them to resist blood flow and exit the blood stream
5) Growth at metastatic site - via growth factors (the tumour cells produce)

Where tumour cells grow bigger than 1-2mm, they need their own blood supply. Tumour cells produce angiogenesis promoters e.g. vascular endothelial growth factor.

153
Q

Where can a tumour that has invaded a venule spread?

Where can tumours of the gut spread? Where can they spread from?

A

Tumour has invaded venule –> Vena cava –> to the heart –> via pulmonary artery to the lungs –> could result in an embolism in a capillary in the lungs (common site for metastases) –> can also invade the pulmonary venules after growing too big and invade the left side of the heart.

Tumours of the gut such as from the colon, stomach, pancreas and intestine can spread to the liver via the portal venous system. (which can spread to the lungs via inferior vena cava)

154
Q

Name and explain 2 types of target therapies used in cancer treatment

A

1) Monoclonal antibodies
- Blocks receptor for growth factor. They block signalling pathways that promote cancer cell growth. – competitive inhibition
(But if there is a mutation so that the receptor is always switched on regardless of binding of growth factor, then monoclonal antibodies won’t help - use small molecular inhibitors) e.g. Cetuximab, herceptin
(inhibit tyrosine kinase activity?)

2) Small molecular inhibitors
- There is no activation of the receptor as the intracytoplasmic domain is blocked. (They bind to the inside of growth factor receptors.) e.g Gleevec

155
Q

What is a hamartoma?

A

A benign mass of organised tissue native to a particular anatomical location e.g. typically found in the lungs, breast, colon.

Growth is related to overall body growth.

156
Q

Which is the most aggressive and advanced thyroid cancer?

A

Anaplastic thyroid cancer

Anaplastic - Where cell type of origin cannot be determined (degree of differentiation can’t be determined)

157
Q

What are the commonest causes cancer deaths in males and females?

A

Males - Lung (commonest), prostate, bowel

Females - Lung, breast, bowel

158
Q

What is a carcinoma?

A

A malignant epithelial neoplasm

159
Q

How can benign neoplasms cause morbidity and mortality? (Pathology)

A
  • Pressure on adjacent structures can cause pressure necrosis
  • If they grow into the lumen of a structure, it can obstruct flow
  • They can produce hormones - e.g. benign tumour in thyroid gland can cause hyperthyroidism
  • They can transform into a malignant neoplasm
  • They can cause anxiety
160
Q

How can malignant neoplasms cause morbidity and mortality? (Pathology)

A
  • Destruction of adjacent tissues
  • Metastases
  • Blood loss from ulcers
  • Obstruct flow
  • Produce hormones
  • Paraneoplastic effects /syndromes - disorders that occur when the immune system has a reaction to a neoplasm e.g lung cancer can cause finger clubbing, small cell lung cancer can cause SIADH (inappropriate ADH secretion)
  • Anxiety and pain
161
Q

State some hallmarks of cancer

A
  • Growth self sufficiency
  • Evade apoptosis
  • Ignore anti-proliferative signals
  • Limitless replication potential
  • Sustained angiogenesis
  • Invades tissues
  • Escape immune surveillance
162
Q

What are some examples of evidence for human tumour immunity?

A

Spontaneous regression: melanoma, lymphoma

Regression of metastases after removal of primary tumour

Infiltration of tumours by lymphocytes and macrophages

Higher incidence of cancer after immunosuppression, immunodeficiency. (AIDS)

163
Q

What is immune surveillance and immunoediting?

A

Dual function - Immunosurveilance and immunoediting.

Immunosurveillance - The immune system’s continuous monitoring and recognition of abnormal or transformed cells (pre-cancerous and cancerous) which could develop into tumours.

Immunoediting- Immune responses can change tumours to be hidden from recognition by the immune system (tumour escape) and tumours can promote immune supression (immune evasion). (So while the immune system protects the body from cancer, it can also drive the development of tumours- helping them to adapt)

Three stages of immunoediting:
- Elimination - cancer cell immunosurveillance - T kymphocytes identify and eliminate tumour cells
- Equilibrium - a phase of tumour dormancy where tumour cells and immunity enter into an equilibrium, keeping tumour expansion in check.
- Escape - the final outgrowth of tumours that have overcome immunological restraints of the equilibrium phase. They are capable of spreading.

164
Q

What are some problems with tumour hypoxia?

A
  • Hypoxia is a prominent feature of malignant tumours
  • When the blood supply is unable to keep up with the growing tumour cells, hypoxic tumour cells can adapt to low oxygen.

Tumour hypoxia is a poor patient prognosis
- It stimulates new vessel growth
- Suppresses immune system
- Resistant to radio and chemotherapy
- Increased tumour hypoxia after therapy

165
Q

Describe whole-killed (inactivated) vaccines along with some problems and limitations

A

Whole-killed - uses the killed version of the germ that causes the disease (killed via chemicals or heat).

The organism must be grown in vitro (which could be expensive)

  • Usually need 2 shots (booster shots)

e.g. Influenza, polio, hepatitis A

166
Q

What are the 3 traditional types of vaccines?

A

Whole-killed (inactivated), toxoid and live attenuated.

167
Q

Describe live-attenuated vaccines. Advantages + limitations.Examples.

A
  • Uses a weakened form of the pathogen that causes the disease (weakened via multiple mutations)
  • Better immune response so lower doses are required
  • Route of administration may be more favourable (oral)
  • Fewer doses are required to provide protection

However

  • Impossible to balance attenuation and immunogenicity
  • Possibility of the pathogen reverting to its virulent form (can cause the disease)
  • Transmissibility - Can be passed to non-vaccinated individuals.
  • May not be safe for immunocompromised individuals. (people with weakened immune system)

E.g. Measles, mumps, rubella, smallpox, chickenpox

(Organisms replicate within the host and induce and immune response which is protecting against the wild-type organism but does not cause the disease)
So attenuation means - an organism is cultured in such a way it does not cause disease when inoculated in humans.

168
Q

What is a toxoid vaccine? Some examples

A

Toxoid vaccines use an inactivated toxin made by the germ that causes a disease. (immune response is targeted to the toxin instead of the whole germ) e.g.

DIPHTHERIA and TETANUS

169
Q

What are recombinant vaccines?
Examples?
1 advantage and 1 disadvantage

A

These vaccines are genetically engineered (using specific parts) of bacteria, yeast, insect or mammalian cells)

  • Avoids the problem of needing to grow the pathogen in vitro.
  • However it is difficult to find a protein that is protective and can generate a strong enough immune response.
  • e.g. Hepatitis B, HPV, SARS-Co-V2
170
Q

Describe the action of a protein based/recombinant vaccine (SARS-COV-2). Give examples of protein based/recombinant vaccines.

A

Recombinant DNA technology to insert the genetic code of the spike protein into the host cell is used to produce large quantities of the spike protein which is combined with an adjuvant (to enhance immune response) –> the injected spike proteins are engulfed by APCs and broken down into peptides which are presented on the cell surface by major histocompatibility complex to elicit an immune response (activation of T and B cells)

e.g. Novavax, hepatitis B, HPV

171
Q

Describe the action of a viral vector vaccine (SARS-COV-2). Example

A

An adenovirus (common virus with mild respiratory infections) is genetically altered to be non-replicating and to carry the synthesised spike protein gene (Complementary DNA). When injected, the adenovirus vector enters host cells and release the modified genetic material. The viral DNA is transcribed and translated by the cell’s machinery to produce spike proteins. Some spike proteins are broken down and presented as peptides on the cell surface by MHCs which activate T cells. T helper cells activate B cells for antibody production and cytotoxic T cells for the destruction of infected cells, while memory B and T cells contribute to a swift and potent immune response upon re-exposure to SARS-CoV-2, preventing viral binding and facilitating destruction.

e.g. Oxford AstraZeneca

Pro
- Effective in delivering therapeutic genes into target cells

Con
- Pre-existing immunity in individuals due to previous exposure to the virus being used as a vector can limit the effectiveness of the therapy.

172
Q

How do mRNA vaccines work? Advantages?

A

mRNA of the target foreign protein is synthesised in vitro (from a DNA template). It is complexed with lipid nanoparticles that stabilise and protect the mRNA from degradation and allows the mRNA to cross the plasma membrane. The mRNA is translated in the cytoplasm and the protein is presented on the surface of the cell with MHC, stimulating the immune response.

  • Avoids the need to grow the pathogen or viral vector
  • No live organism involved
  • mRNA is relatively cheap to produce

Con
- Requires extremely cold storage conditions

e.g. Pfizer, moderna

173
Q

What are the 8 week immunisations for a baby and how are they administered?

A

Diphtheria, tetanus, pertussis, polio, haemophilus influenzae type b (Hib) and hepatitis B) - One injection,

Meningococcal B - One injection
Rotavirus - One oral application

174
Q

What is a conjugated vaccine?Give example

A

A vaccine which links polysaccharide antigens from a bacteria (bacteria capsule) to a carrier protein which elicits an immune response when injected. (not sure if this is true but the polysaccharide antigens would be weaker antigen compared to the carrier protein- stronger antigen so the immune system has a stronger response to the weaker antigen)

e.g. Haemophilus influenzae type b

175
Q

What are the contents of a blood sample and what is serum?

A

55% plasma (water, electrolytes, proteins, lipids, sugars)

<1% Leukocytes (WBC)

45% erythrocytes, platelets

Serum is plasma without fibrinogen and clotting factors.

176
Q

Which are the 3 polymorphonuclear leukocytes and the 3 mononuclear leukocytes?

A

Polymorphonuclear - Neutrophil, eosinophil, basophil

Mononuclear - Monocyte, T cell and B cell

177
Q

What are the soluble factors that are part of the immune system?

A

Complement - A group of 20 serum proteins secreted by the liver that needs to be activated to be functional. (they circulate in inactive forms)

Antibodies - which bind specifically to antigens (immunoglobulins - 5 distinct classes)

Cytokines - proteins secreted by immune and non-immune cells (e.g. interferon, interleukin

178
Q

What are the 3 modes of action of complement proteins? What are they secreted by?

A

A group of 20 serum proteins secreted by the liver. They need to be activated to be functional.

1) Direct lysis
2) Attract more leukocytes to site of infection (chemoattract)
3) Coat invading organisms (opsonisation)

179
Q

What are some cytokines of the immune system?

A

Interferon alpha and beta - produced by virus infected cells and interferon gamma -produced by activated T helper 1 cells –> Induce an antiviral state in cells =activate natural killer cells, stimulate dendritic cells

Interleukin
- Can be pro-inflammatory (IL1) or anti-inflammatory (IL10)
- IL4 secreted by T helper 2 cells can activate B cells to differentiate into plasma cells (has anti inflammatory properties)

Colony stimulating factors
- Involved in directing the division and differentiation of stem cells in the bone marrow (into leukocytes)

Tumour necrosis factors - alpha and beta
- Mediates inflammation
- Mediates cytotoxic reactions (can induce apoptosis - alpha)

Chemokines -
- Leukocyte chemoattractants (determine the movement and positioning of immune cells to ensure effective and targeted immune response)

180
Q

What is the response when there is tissue damage and bleeding / infection?

A
  • Coagulation - stop bleeding
  • Acute inflammation - Leukocyte recruitment
  • Kill pathogens, neutralise toxins, limit pathogen spread
  • Clear pathogens/dead cells via phagocytosis)
  • Proliferation of cells to repair damage
  • Remove blood clot (remodel extracellular matrix)
  • Re-establish normal structure and function of tissue
181
Q

Definition of inflammation

A

A series of reactions that brings cells and molecules of the immune system to sites of infection or damage.

182
Q

Which are the complement proteins that allow for each of the steps of the complement cascade to occur (other than inflammation)

A

Lyse microbes directly - MAC (Membrane attack complex - final step of complement cascade)

Chemotaxis - C3a and C5a (IMPORTANT)

Opsonisation - C3b (inserts itself into the membrane of bacteria) - IMPORTANT

183
Q

What are the 2 regions of antibodies and what are their roles?

A

Fab regions (the two outgoing portions in a Y) – ANTIGEN RECOGNITION
- Variable in sequence
- Bind different antigens specifically (antigen binding sites)

Fc region (the single bottom port of the letter Y) - ANTIGEN ELIMINATION
- Constant in sequence
- Binds to complement Fc receptors on phagocytes, NK cells

184
Q

What does the structure of an antibody entail?

Try to draw

A

(FC region) Heavy chain - constant region
- Bind to immune cells (Phagocytes, NKC)

(Fab region has both types)
Light chain and heavy chain- variable region
- Has antigen binding sites

185
Q

What are the molecular weights of light chain and heavy chain in an antibody?

What are the two types of light chains?

A

25 kD - light chain (kappa and lambda)

50 kD - heavy chain

186
Q

How do antibodies protect against infection in terms of the site of binding (e.g. Fab and Fc)

A

Fab (antigen recognition) - specific binding
- Neutralise e.g. toxins (IgG and IgA)
- Immobilise motile microbes (IgM)
- Prevent binding to and infection of host cell

Fc (antigen elimination) - enhance innate mechanisms
- Activate complement system (IgM, IgG)
- Phagocytes can bind to Fc receptors (IgG and IgA) - increase opsonisation and enhance phagocytosis
- Mast cells can bind to Fc receptors (IgE) - release of inflammatory mediators
- NK cells can bind to Fc receptors (IgG) - enhanced killing of infected cells

187
Q

What kind of antigens do B cells and T cells recognise?

A

B cells - Soluble, free, native antigens

T cells - cell associated, processed antigens

188
Q

State some key points about MHC proteins (major histocompatibility)

A
  • They are encoded by the major histocompatibility gene complex on chromosome 6 (cluster of genes)
  • They are important in graft rejection (as they play a role in the recognition of foreign tissues- mismatched MHC molecules between donor and recipient can trigger an immune response)
  • They are highly polymorphic (diversity of alleles allowing the recognition of a wide range of pathogens)
  • Also known as HLA (human leukocyte anitgen)
  • They play a crucial role in presenting antigens to T cells and initiating T cell responses.
189
Q

Explain the process of T cell recognition

A

1) In a virus infected cell –> viral proteins are broken down in the cytosol –> in the endoplasmic reticulum, peptides bind to MHC1 molecules –> it is transported to cell surface –> Cytotoxic T cells recognise the peptide bound to MHC1 and are activated to kill the infected cell by inducing apoptosis

2) Macrophages/dendritic cells/B cells internalise foreign material and transported to endosomes–> within endosomes, the antigens are broken down to peptides and the peptide fragments bind to MHC2 molecules which are transported to the cell surface –> helper t cells recognises the peptide bound to MHC2 and are activated –> helps B cells make antibody, produce cytokines that activate/regulate other leucocytes.

190
Q

Function of IL1,2,3,8,10. Interferon-gamma and tumour necrosis factor alpha.

Also what are they secreted by?

A

IL1 - Macrophages, endothelial and epithelial cells –> induces inflammation, fever, activates leukocytes

IL2 - T cells - Stimulates T, B and NK cell growth

IL4 - T helper 2 cell, mast cell -
- Differentiates naive T cells into T helper 2 cell
- Stimulates B cell proliferation and differentiation to plasma cells - antibody production
- Induces IgE production

IL8 - induces neutrophil chemotaxis (slight macrophages chemotaxis also)

IL10 - monocytes, TH2 cells –>
- down regulates TH1 cytokines
- Anti inflammatory

Interferon gamma - TH1 cells, NK cells –> activated macrophages and NK cells, increases MHC2 expression

Tumour necrosis factor alpha - t cells, macrophages, NK cells –> activates neutrophils, can induce apoptosis, can induce cachexia (wasting - loss of muscle and body fat)

191
Q

Which cytokines do TH1, TH2 and TREG cells make?

A

TH1 - Interferon gamma, IL2, TNF-beta

TH2 - Interleukin 4,5,6,10 and 13

TREG - Interleukin 10, transforming growth factor beta.

192
Q

Definition of type 1 hypersensitivity, atopy and allergens

A

Type 1 hypersensitivity - an immediate reaction to environmental antigens mediated by IgE

Atopy - an inherited trait for type 1 hypersensitivity

Allergens - antigens that trigger allergic reactions

193
Q

What are the 4 main types of chemokines and what are the leukocytes they move?

A

Chemokines - group of proteins that direct movement of leukocytes (and other cells) from the bloodstream into tissues or lymph organs (by binding to specific receptors on cells)

CXCL - mainly neutrophils

CCL - Monocytes, lymphocytes, eosinophils, basophils

CX3CL - Mainly T lymphocytes and NKC

XCL - mainly T lymphocytes

194
Q

What are pattern recognition receptors (PRRs)?

Which cells are they expressed by?

A

They are proteins of the innate immune system which play a role in recognising molecular patterns associated with pathogens. These include pathogen associated molecular patterns (PAMPS) and damage associated molecular patterns (DAMPS)- released during tissue damage or stress.

PRRs are expressed in cells such as dendritic cells, macrophages, monocytes and neutrophils.

Upon binding to a specific ligand they initiate signalling cascades that lead to immune responses (inflammatory pathways, production of antibodies, recruitment of immune cells)

195
Q

What are some examples of PAMPS?
Name 2 membrane bound PRRs and 2 cytoplasmic sensor PRRs
(what do the receptors recognise?)

A

Pathogen associated molecular patterns –>
- Bacterial carbohydrates (Mannose)
- Nucleic acid (bacterial DNA)
- Bacterial peptides (flagellin, microtubules, peptidoglycan)

Above no need to know in depth.

Membrane bound PRRs
- Toll like receptors
- C type lectin receptors (recognise particularly mannose)

Cytoplasmic sensor PRRs
- Nod like receptors (- detects microbial infections or cellular stress - upon activation, they lead to the release of pro-inflammatory cytokines (interleukin-1)
- RIG-I-like receptors (primarily involved in detecting viral RNA. When activated, they trigger signalling pathways that lead to the production of interferons and other antiviral molecules)

196
Q

What do Toll like receptors (TLR) 2,4,5,7 and 9 detect?

Which are intracellular which are extracellular?

A

2- Peptidoglycans in gram positive bacteria (components of the cell wall of gram positive bacteria)

4- Lipopolysaccharides (components of outer membrane of gram negative bacteria)

5- Flagellin (for bacterial motility)

7 - Single stranded RNA from bacteria

9 - Non-methylated DNA

Intracellular - 3,7,8,9
Extracellular- 4 and 5

197
Q

What are the 3 activation pathways of the complement cascade?

A

Classical - Antibodies bind to antigens via FAB portion (antigen-antibody complex) –> (complement protein binds to fc portion of the antibodies –> triggers complement cascade (MAC-lysis, chemotaxis via inflammation, opsonisation which enhances phagocytosis)

Alternative - Complement protein C3b can bind directly to the antigen (of microbe) –> complement cascade

Lectin pathway - mannose-binding lectin to mannose (on the surface of bacteria –> complement cascade

All 3 pathways converge at the level of C3 activation, leading to the formation of the membrane attack complex (lyse microbes directly)- final stage of complement cascade

198
Q

What are the 2 killing pathways of microbial killing?(oxygen dependent and oxygen independent)

A

Oxygen dependent - Superoxides are converted to H2O2 then free radicals which can kill the microbes.
- Nitric oxide can also kill microbes directly or can result in vasodilation which increases extravasation of neutrophils

Oxygen independent -
(Enzymes) e.g. lysozyme.
Proteins e.g. defensins and TNF.
pH

199
Q

MHC 1 and 2 What are they coded by? Where are they found?

A

MHC 1 –> Human leukocyte antigen (HLA) - A, B and C
- Found on the surface of all nucleated cells (relevant to graft rejection)

MHC2 –> coded by HLA DP, DQ and DR
- Primarily found on the surface of antigen presenting cells (dendritic cells, macrophages, B cells)

200
Q

What is the process of T cell recognition and activation?

A

Recognition
- T cells bind to specific antigens presented by MHC on the surface of APC. (MHC1 - cytotoxic , MHC2 - helper)

Division
- Activated T cells undergo CLONAL EXPANSION - proliferation of T cells.

Differentiation
- Helper T cells can differentiate into TH1 or TH2 cells
- Cytotoxic T cells –> effector cytotoxic T lymphocytes
DIFFERENTIATION IS INFLUENCED BY CYTOKINES

Effector functions
- Differentiated T cells perform their functions.
- Cytotoxic T cells release perforin and granzymes
- Helper T cells release cytokines (e.g. interferon and interleukin) to influence activity of other immune cells

Memory
- Some T cells survive and become memory T cells which are long lived and can quickly respond upon re-exposure to the same antigen.

201
Q

In which scenario would T helper cells differentiate to TH1 cells or TH2 cells?

A

When there is a high concentration of IL-12 in the surrounding environment, naive CD4+ T cells will differentiate into TH1 cells.

Low concentration of IL-12 –> TH2 cells.

202
Q

What are the 3 antigen presenting cells?

A

Dendritic cells, macrophages and B cells

203
Q

What is a cytokine storm?

A

Systemic inflammatory syndromes which involve an elevated level of circulating cytokines and chemokines and immune cell hyperactivation.

  • Can lead to widespread inflammation throughout the body
  • Can cause tissue damage
  • Organ failure
  • Associated with severe illnesses and viral infections like SARS

(triggered by pathogens, cancers, autoimmune conditions, etc)

204
Q

How many percent of each class of immunoglobulin is found in the blood?

A

IgG - 70-75%
IgM - 10%
IgA - 15%
IgD - 1%
IgE - 0.05%

205
Q

Examples of Pattern recognition receptors mutations (2)

A

Non functioning mutations in the NOD2 gene can lead to Crohn’s disease

Gain of function mutations in TLR7 is associated with SLE

206
Q

Describe B cell activation

A

B cell activation
- They become activated upon binding to a specific antigen.

Clonal expansion and differentiation
- B cells go to lymph nodes where they undergo CLONAL EXPANSION (proliferate) and differentiate into plasma cells.

Antibody production
- Plasma cells generally produce antibodies mainly of IgM (primary response)class.
- Over time B cells undergo class switching to produce antibodies of IgG class (secondary response)

Memory
- Some B cells persist after the immune response and become memory cells for long lasting immunity.

207
Q

What are the 3 mechanisms for drug permeation across cell membranes?

A

1) Passive distribution through hydrophobic membranes (Lipid soluble molecules)

2) Passive distribution through aqueous pores (very small water soluble drugs) - most are too big

3) Carrier mediated transport
- Proteins which transport sugars, amino acids, neurotransmitters, etc.

208
Q

What are the factors affecting drug absorption?

A

1) Drug ionisation
Ionised drugs have poor lipid solubility and are poorly absorbed.

2) Lipid solubility

209
Q

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

A

Weak acids are better absorbed in the stomach. Because the stomach is an acidic environment, weak acids are more likely to remain in their non-ionised form (good lipid solubility) - thus well absorbed

Weak bases are better absorbed in the intestine because the small intestine has a more alkaline environment allowing weak bases to remain in their non-ionised form (good lipid solubility –> well absorbed)

210
Q

What are factors affecting oral drug absorption in the stomach? and in the intestines

A
  • Gastric enzymes (drug molecules may be digested) - e.g. peptides and proteins like insulin
  • Low pH - molecules may be degraded
  • Food (full stomach can slow absorption)
  • Gastric motility
  • Previous surgery

Intestine
Drug structure - Large hydrophilic molecules are poorly absorbed + lipid soluble/unionised molecules diffuse down concentration gradient.

Medicine formulation - capsule or tablet coatings can control the timing of drug release. (modified release can control the rate of absorption)

P glycoprotein causes substrates/drugs to be removed from the intestinal endothelial cells back into the lumen.

211
Q

What is first pass metabolism?

A

Metabolism of drugs preventing them from reaching systemic circulation.

  • Degradation by enzymes in intestinal wall
  • Absorption into hepatic portal vein and metabolism in the liver.
    (Avoided via rectal route of administration or IV administration)
212
Q

What is the definition of bioavailability?

A

The proportion of administered drug that reaches the systemic circulation.

213
Q

What is bioavailability dependent on?

A
  • The extent of drug absorption and extent of first pass metabolism
  • Route of administration
  • Variation between individuals - genetic, disease states
214
Q

Pros and cons of administration - rectal, inhaled, subcutaneous, transdermal

A

Rectal
- Avoids first pass metabolism

  • Absorption can be variable
  • Patient preference (may not want)

Inhaled
- Well perfused (large surface area)

  • Inhaler technique can limit effectiveness

Subcutaneous
- Faster onset then oral administration

  • Not as rapid as IV

Transdermal (skin patch)
- Provides continuous drug release
- Avoids first pass metabolism

  • Only suitable for lipid soluble drugs
  • Slow onset of action
215
Q

What factors can affect the distribution of drugs?

What is volume of distribution?

A
  • Size (small - larger distribution)
  • Lipid solubility (Lipophilic - larger distribution)
  • Protein binding (Increased protein binding - less distribution)

Volume of distribution - theoretical volume a drug will be distributed in the body (drugs that distribute well have high Vd)

216
Q

3 ways drugs can enter the CNS?

A
  • High lipid solubility –> can pass through BBB
  • Intrathecal administration - chemotherapy
  • Inflammation (sepsis can cause BBB to become leaky)
217
Q

How do certain diseases states - renal impairment, sepsis, hypoalbuminaemia affect pharmacokinetics of drugs?

Obesity and older age

A

Renal impairment
- Reduced kidney function can lead to accumulation and toxicity of renally cleared drugs (as kidneys excrete drugs and drug metabolites)

Sepsis
- Can lead to increased permeability of blood vessels (leaky blood vessels) –> allows drugs to move between blood stream and tissues WITH GREATER PENETRATION OF BBB

Liver impairment: Hypoalbuminemia
- In conditions of liver impairment like cirrhosis or acute liver failure, the synthesis of albumin may be reduced –> many drugs bind to albumin so when albumin levels are low, the amount of unbound drugs increase –> leading them to distribute more extensively into tissues

Obesity: caution dosing drugs with a small Vd (meaning it is not well distributed to fat)

Older age: change in body composition (decrease in lean body mass, increase in body fat), leads to smaller Vd of water-soluble drugs (tends to stay in systemic circulation), therefore higher plasma conc which can be toxic

218
Q

What are hallmarks of ageing (on a systemic, cellular and molecular level)

A

Systemic
- Nutritional dysregulation (reduced nutrient sensing can lead to metabolic dysfunction)

Cellular level
- Cellular senescence (cells lose the ability to divide and undergo functional changes–> senescent cells can accumulate in tissues over time and contribute to inflammation and tissue dysfunction) - due to telomere shortening and dna damage
- Stem cell exhaustion (stem cells undergo a decline in their ability to proliferate and differentiate, impairing tissue renewal)

Molecular level
- Telomere shortening - leads to cellular senescence (telomeres gradually shorten with aging)
- Compromised autophagy (a process responsible for the degradation and recycling of damaged cellular components –> reduced autophagy contributes to the accumulation of damaged proteins and organelles within cells.
- Loss of proteostasis –> process regulating proteins within the cell –> leads to accumulation of misfolded or aggregated proteins
- Epigenetic alteration – DNA methylation can affect gene expression and contribute to age related phenotypes
- Mitochondrial dysfunction - dysfunction (due to accumulation of ROS) can lead to increased oxidative stress

219
Q

What happens during immunosenescence? (innate response, adaptive response and inflammaging)

A

Immunosenescence - dysregulated immune function which contributes to increased susceptibility of the elderly to infection.

Innate response
- Decreased MHC 2 expression –> impairs the activation of T helper cells
- Decreased phagocytosis - diminished ability to clear pathogens leads to increased susceptibility to infections.
- Decreased interferon gamma (released by TH1 cells) –> lack of activation of macrophages and NKC

Adaptive response
- Decline in naive T cells
- Decreased antibody production

Inflammaging - chronic, low grade inflammation in aging
- Increased inflammatory cytokines due to mitochondrial dysfunction, cellular senescence and impaired autophagy

220
Q

What is efferocytosis? What happens when it is impaired?

A

The process whereby dying or dead cells are cleared away by macrophages.This is essential for preventing inflammation and resolving the immune response.

Macrophages engulf the cells and form a phagosome –> which fuses with lysosomes to form a phagolysosome –> cellular debris is degraded by lysosomal enzymes –> macrophage releases anti-inflammatory signals and pro-resolution cytokines.

However when efferocytosis is impaired/inefficient, apoptotic cells may undergo secondary necrosis which leads to the release of histotoxic neutrophil contents –> tissue damage.
Also when macrophages sense the presence of uncleared apoptotic cells, they release pro-inflammatory cytokines–> can lead to chronic inflammation

221
Q

What are age related alterations in innate immune cells (neutrophil, NK cells, monocytes, dendritic cells?)

A

Neutrophils and monocytes - Decreased chemotaxis and phagocytosis. Decreased superoxide generation and NET generation

NK cells - increased numbers, decreased cytotoxicity

Dendritic cells - Decreased recruitment to lymphoid organs, phagocytosis, decreased antigen presentation.

222
Q

What is the difference between transient and persistent Senescence-Associated Secretory phenotype?

A

Transient SASP - Development, repair, regeneration, tumour suppression

Persistent SASP - Age-related diseases, chronic inflammation, tumour progression

Transient
- Antifibrotic, anti inflammatory, Senescent cell clearance, tissue repair

Persistent
- Profibrotic, pro-inflammatory, senescent cell accumulation, tissue dysfunction

Transient – Beneficial as it is a temporary response to acute stress which contributes to tissue repair, clearance of senescent cells and supports the healing process

Persistent - Detrimental as it can lead to sustained inflammation, tissue dysfunction and fibrosis –> chronic state may contribute to the development and progression of age related diseases. (heart failure, atherosclerosis, COPD, Parkinson’s, alzheimer’s, benign prostatic hyperplasia, osteoarthritis, type 2 diabetes)

223
Q

How does ageing affect the thymus, bone marrow, spleen + lymph nodes and lungs?

A

Thymus involution - decreased thymic output of mature T cells

Bone marrow - decreased B cell maturation
- Impaired haematopoiesis due to increased adiposity of bone marrow

Spleen + lymph nodes - Reduced number and size of follicles. Reduced B cell migration into follicles (i’m assuming follicles is lymph nodes)

Lungs - increased infiltration of pro-inflammatory cells leading to lung tissue damage

224
Q

Differences in immune cells between people who are considered elderly and people who can live to an old age (centenarians)

A

CD8+
Elderly - decrease in proliferation ability and cytotoxicity of CD8+ T cells.
Centenarians - Highly differentiated CD8+ T cells

B cell
Elderly - decrease in naive b cells
Centenarians - increase in naive B cells (but decrease in number of B cells)

NK cells
Elderly - impaired cytotoxicity
Centenarians - maintained cytotoxicity

Neutrophil
Elderly - Decreases in phagocytic ability, adhesion and chemotaxis
Centenarians - increase in neutrophil chemotaxis and microbial capacity

Inflammatory mediators
Elderly - increased inflammatory molecules
Centenarians - increased anti inflammatory molecules.

225
Q

What are some intervention strategies to reduce immunosenescence inflammaging?

A

Modification of lifestyle
- Physical activity
- Caloric restriction
- Maintain optimal nutrition

Pharmacological interventions
- Statins
- IL7 therapy - -which promotes the survival, proliferation and maturation of T cells.
- Caloric restriction mimics

(possible anti inflammatory drugs, cell replacement therapy, gene therapy against aging related diseases)

This reduces the risk of infections, chronic diseases and autoimmunity leading to improved health and reduced mortality

226
Q

What is first order kinetic and zero order kinetics (drugs)?

(why do you need to be cautious when administering zero order drugs)?

A

First order - Rate of elimination is proportional to the plasma drug concentration (a constant % of the drug)

Zero order - A constant amount of the plasma drug is eliminated over a unit of time (as elimination is irrespective of plasma concentration and small increases in dose may cause large increases in plasma concentration , caution is needed when adjusting doses - dose changes should be small)

227
Q

What is Cmax, Tmax and clearance (when it comes to drug plasma concentration)

When is a drug considered cleared?

A

Cmax - Maximum plasma concentration

Tmax.- time taken to get to Cmax (slower absorption - increased Tmax)

Clearance - rate of drug elimination by all eliminating organs (rate of drug elimination/drug plasma concentration) - efficiency of irreversible elimination of the drug from the body
- Drug is considered cleared after 5 half lives

T 1/2 AKA half life is the time taken for plasma drug concentration to fall 50%

228
Q

Describe the drug plasma concentration-time relationship for IV and oral medication

A

Single oral dose
- Drug is absorbed in the GI tract but must undergo first-pass metabolism in the liver before entering the systemic circulation.
- The time taken for the drug to reach its peak concentration in the plasma (Cmax) varies based on drug’s solubility, presence of food in the stomach, etc. Bioavailability would be less than 100% also.
(Graph is –> an increase till C max, and then a decrease (drug elimination)

Single IV dose
- Drug directly injected into bloodstream bypassing the absorption phase and first-pass metabolism
- Administration is a rapid and complete delivery thus graph - plasma concentration is highest at the beginning and slowly declines over time.
- 100% bioavailability

229
Q

In what ways can drugs and metabolites be excreted?

A

Liquids - small, polar molecules –> Urine, bile, sweat, tears, breast milk

Solids - Large molecules: Faeces

Gas -(volatiles)- expired air

230
Q

What is the half life of a drug determined by?

A

1) Clearance
- If clearance by organs (hepatic, renal, faeces, breath) is high, the drug is quickly eliminated (shorter half life)
(in organ dysfunction, drug half life will be increased)

2) Volume of distribution
- A drug with a larger volume of distribution will have a longer half life

Age - half life of drugs are longer in elderly

Disease states can influence half life also

Protein binding - drugs with high affinity for albumin will have longer half lives.

231
Q

What is steady state in the context of repeated dosing?

How long does it take to get to Css

When would continuous IV infusion be used?

If you reduce the dose of the drug by 50%, how does it affect Css?

A

It is when the rate of drug input is equal to the rate of drug elimination (Continuous IV infusion) - can google for a picture of the graph but essentially plasma concentration of the drug increases till it plateaus which is termed Css - drug plasma concentration at steady state. (The time to Css will be 5 half lives)

Continuous IV infusion used for
- Critical care patients
- Administering of antibiotics
- Unfractionated heparin (anticoagulant)
- General anaesthetics

(infusion rate can be titrated up and down to achieve desired therapeutic effect)

If you reduce the dose of the drug by 50%, the time taken to reach Css doesn’t change but there is a 50% reduction in Css.

(if clearance of a drug is reduced (longer half life), time to Css (half life x 5) increases so you give a lower dose so that Css is reached faster)

232
Q

What is a loading dose?

A
  • It is a higher initial dose of a drug given at the beginning of treatment to rapidly achieve the desired steady state (therapeutic concentration) –> which is within the therapeutic window (between maximum safe concentration (MSC) and maximum effective concentration (MEC)
233
Q

What is pharmacogenomics?

A

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

(patient’s genome is used to identify most appropriate treatment and dose)

234
Q

How can genetic variants alter drugs?

A
  • Variations in drug receptors can affect the interaction between drugs and their targets
  • Variations in efficacy of the drug depending on the individual
  • It can increase the incidence of adverse drug reactions
  • Genetic variations in drug metabolising enzymes can affect the efficiency of drug clearance
235
Q
A
236
Q

What are the 4 receptors that are targeted by drugs?

A

LIgand gated ion channels (nicotinic ACh receptor)
- Ion channels are membrane proteins that allow ions to pass through channel pores which cause the cell to undergo a shift in electric charge distribution (charge determined by influx of cation or efflux of anion)

G protein coupled receptors (beta adrenoreceptors)

Kinase linked receptors (receptors for growth factors)

Nuclear (cytosolic) receptors - (steroid receptors)

237
Q

What are G protein coupled receptors and how do they work?

A
  • Beta adrenoreceptors
    Largest most diverse group of membrane proteins - targets by >30% of drugs
  • Activity is regulated by factors that control their ability to bind to an hydrolyse GTP to GDP
    = On Ligand binding (peptides, lipids, sugars) to the extracellular domain, the receptor interacts with G protein to dissociate GDP and causes GTP to bind to adenyl cyclase (or PLC/phospholipase C) –> ATP is then converted to cAMP which activate protein kinases.
238
Q

What are kinase linked receptors? How do they work?

A

(receptors for growth factors)

Kinases are enzymes which catalyse the phosphorylation of proteins.
- When a signal dimer (pairing of two molecules) binds to the receptor, it results in the activation of kinases –> tyrosine molecules are phosphorylated –> intracellular signalling molecules bind to the phosphorylated tyrosine molecules initiating a cascade of intracellular signalling events (cell growth, differentiation).

239
Q

What are nuclear receptors?
How do they work?

A
  • Steroid hormone receptors
  • They function as transcription factors, modifying gene transcription
  • When a ligand binds, the nuclear receptor is activated and it binds to specific DNA sequences via zinc fingers –> this results in the modulation of expression of target genes (transcription of the gene) which can lead to cellular responses like differentiation.
240
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”

241
Q

Describe the efficacy of full agonist, partial agonist.

What is an inverse agonist?

A

Full agonist - full efficacy (100% response)

Partial agonist - less efficacious than a full agonist (maximum efficacy will be less than 100%- no matter the dose)

Inverse agonist - A drug that binds to the same receptor as an agonist induces a pharmacological response opposite of the agonist.

(An antagonist is something that blocks or reduces the action of an agonist but inverse agonists not only block the activity, they induce an opposite effect)

242
Q

What are the factors affecting drug action?

A

Receptor related - Affinity and efficacy

Tissue related - Number of receptors and signal amplification (same receptor and agonist but tissue can have different amplification responses)

243
Q

How long would opioid withdrawal typically last for? And when does it start after stopping treatment?

A

Starts within 24 hours and lasts 72 hours.

244
Q

What is the treatment for opioid induced respiratory depression?

Why do opioids cause respiratory depression?

A

Treatment for opioid induced respiratory distress –> Naloxone IV (opioid antagonist)

Opioids can inhibit the activity of the brainstem, the area where the respiratory centres are (which are responsible for breathing) - decreases the sensitivity of brainstem to CO2 levels and so respiratory rate decreases

245
Q

Why does codeine require CYP2D6?

A

Codeine is a prodrug and needs to be metabolised by CYP2D6 to morphine in order to work.

In populations where CYP2D6 activity is decreased, codeine will have a reduced effect

In populations where CYP2D6 is overactive, they may be at increased risk of respiratory depression

(Tramadol also)

246
Q

What would you do to dosage of medicine in order to suit a patient?

A

Titrate the dose of the medication to suit the patient

247
Q

Name some beta blockers and their effects

Name some alpha blockers and their effect

A

Beta blcoker
- ACE inhibitor
- Anti platelets
- Statin

  • Blocks the effect of the sympathetic nervous system on the heart
  • Decreases rate and contractility
  • Decreased workload for the cardiac muscle
  • Reduced incidence of heart failure.

Alpha blocker
- Doxazosin, tamsulosin
Used for hypertension

Take note: that too much beta blocker will not only block beta 1 receptors but can also start to block beta 2 receptors resulting in bronchoconstriction (dangerous in people with asthma)`

248
Q

Which of the following drug - action pairings are correct?
Metaraminol - treats hypertension
Tamsulosin - treats Benign Prostatic Hyperplasia
Salbutamol - directly acts on beta 1 receptors
Carvedilol - safe for use in patients with asthma
Isoprenaline - reduces obesity

A

A - false, its an alpha 1 agonist so treats hypotension by causing vasoconstriction
B - True, its action as an alpha 1 blocker relaxes the smooth muscle of the bladder wall
C - False, salbutamol is a beta 2 receptor agonist (2 lungs!)
D - False, beta blockers, especially non-selective ones, can exacerbate asthma
E - False, Isoprenaline is a non-selective alpha and beta agonist. Beta 3 receptor is involved in lipolysis but B2AR agonist drugs for obesity are still in research stage

249
Q

What are the functions of M1,M2,M3 receptors and nicotinic receptors?

A

M1 receptor (CNS and periphery) - Increases gut motility and secretions in the gut, also CNS effects (cognition, learning, memory)

(M1,M4,M5 - brain + spinal cord (CNS))

M2 receptor (mostly in heart) - Reduced contractility and bradycardia

M3 receptor (mainly smooth muscles and glands) - vasodilation, bronchoconstriction, pupil constriction, salivary glands, bladder.

Nicotinic receptors - Primary role in synaptic transmission in the CNS and at neuromuscular junction (some vasodilatory effects)

250
Q

Agonists and antagonists of Nictotinic and muscarinic receptors

A

Nicotinic antagonist - Trimethaphan (rarely used. for hypertensive crisis)

Nicotinic agonist - Nicotine

Muscarinic antagonists - Atropine (crosses BBB), Glycopyrrolate (doesn’t cross BBB), Hyoscine, Ipratropium (topical effect in lungs)

Muscarinic agonists - Pilocarpine, muscarine (found in poisonous mushrooms)

AGONISTS OF BOTH RECEPTORS - Acetylcholine, Neostigmine (reversible acetylcholinesterase inhibitor), organophosphates (irreversible acetylcholinesterase inhibitor)

251
Q

Name non-selective/selective agonists/antagonists for alpha 1, alpha 2, beta 1 and beta 2 receptors

Sympathetic system
- for now just know briefly

A

Non selective alpha antagonists - Phenoxybenzamine, phentolamine

Selective alpha 1 antagonists - Prazosin, doxazosin

Selective alpha 1 agonists - phenylephrine, metaraminol

Selective alpha 2 agonists - Clonidine, dexmedetomidine

Non Selective alpha agonists - noradrenaline

Non selective beta antagonists - Propanolol, carvedilol

Selective B1 antagonists - Metoprolol, atenolol

Selective B1 agonist - Dobutamine
Selective B2 agonist - Salbutamol

non selective beta agonist - isoprenaline

ADRENALINE IS AN AGONIST OF ALL

252
Q

What are the 5 steps in drug development?

A
  • Lead compound identification
  • Pre-clinical research
  • Filing for regulatory status
  • Clinical trials on humans
  • Marketing the drug
253
Q

Identify the two types of drug-drug interactions (pharmacokinetic and pharmacodynamic) and explain how they differ

A

Pharmacodynamic interactions - Occurs when one drug affects the pharmacological response of another drug by acting on the same drug receptors or physiological system.
This can either have a SYNERGISTIC effect or ANTAGONISTIC effect. (Could result in adverse reactions)
- Highly selective drugs are less likely to be problematic

Pharmacokinetic interactions - Occurs when one drug affects the absorption, distribution, metabolism or excretion of another drug.

Simply, pharmacokinetic interactions involve changes in how the body absorbs, distributes, metabolises or eliminates a drug. Pharmacodynamic interactions influence how a drug functions at its target site, altering its effect within the body.

254
Q

Name Cytochromes that are significant for drug metabolism

A

CYP 450 - majority of phase 1 metabolic reactions

CYP 3A4

CYP 2D6

CYP2C9

CYP1A2

255
Q

Explain pharmacokinetic drug-drug interactions in the 4 stages.

A

Absorption
- A drug could increase stomach pH reducing the absorption of another drug (famotidine increases stomach pH, decreasing absorption of ketoconazole)
- 2 drugs that form an insoluble drug complex (when taken together) will not be absorbed (doxycycline and oral iron)
- P glycoproteins throughout the body functions to remove toxic substances including drugs from the body. Some drugs may upregulate P glycoproteins therefore reducing absorption of drugs. (Carbemazepine a PGP inducer can reduce rivaroxaban levels)

Distribution
- Amiodarone could displace warfarin from albumin creating unbound warfarin molecules –> increasing risk of bleeding (as it is an anticoagulant)

Metabolism
- Drugs that are either enzyme inducers or inhibitors could have effects on another drug.

Enzyme inducers –> increase expression of enzymes (e.g. CYP) –> leading to increased inactive metabolites which are excreted –>reduced levels of drug (subtherapeutic, treatment failure) (Rifampicin and CYP3A4)

Enzyme inhibitors –> decrease expression of enzymes (e.g. CYP) –> decreased inactive metabolites which are excreted –> increased levels of drug in the blood (toxicity, adverse drug reactions) (Clarithromycin and CYP3A4)

Excretion
- 2 drugs could compete for elimination from the body via organic anion transporters (or OCT) resulting in reduced elimination of 1 drug –> toxicity (e.g. methotrexate and NSAIDS)

256
Q

Definition of adverse drug reaction

A

A response to a medicinal product or combination of medicinal products which is noxious and unintended.

257
Q

Examples of drug-food interactions

A

Grapfruit juice is a CYP3A4 inhibitor (avoided by patients taking warfarin, statins)

Milk can affect absorption of some drugs due to insoluble complex formed with calcium (doxycycline, levothyroxine, etc)

Eating foods high in vitamin K (kale, spinach, broccoli) will oppose the action of warfarin

Cranberry juice is a CYP2C9 inhibitor (avoided by patients taking warfarin)

258
Q

What is the DoTS method of classifying ADRs?

A

D- Dose related
- Hyper susceptibility - (ADRs at subtherapeutic dose - anaphylaxis with penicillin
- Collateral effects (side effects) - ADRs at therapeutic dose
- Toxic effects - ADRs at subpratherapeutic doses (liver damage with paracetamol)

T- Timing
- ADRs which develop during any time during the treatment.
(Rapid (drug given too quickly), first dose, early (occurs early on in treatment but resolve as treatment progresses), intermediate (occurs after some delay), late (risk increases with prolonged/repeated exposure, delayed - some time after exposure or after drug withdrawal)

S- Susceptibility
- Certain patient groups may have a specific susceptibility to ADRs
- Age
- Gender
- Disease states (e.g. CVD)
- Physiological states (pregnancy)

259
Q

Risk factors for hypersensitivity

A
  • Females are at higher risk than males
  • EBV, HIV
  • Previous drug reactions
  • Uncontrolled asthma
  • Certain HLA groups
260
Q

Clinical criteria for allergy to a drug

A
  • Disappearance on cessation
  • Re-appears on re-exposure
  • Occurs in a minority of patients on the drug
261
Q

What are the 4 compartments that a drug can distribute through in the body?

A

Plasma, interstitial fluid, intracellular fluid and fat.

262
Q

How would you identify and avoid drug interactions?

Which patients would be at high risk of drug-drug interactions?

A
  • Obtain a complete drug history
  • Look out for high risk drugs with a narrow therapeutic index/window
  • Look out for new drugs

Patients may be at high risk of drug interactions due to:
- Polypharmacy
- Kidney or liver impairment
- Extremes of age

263
Q
A