Cancer Flashcards

1
Q

What is metaplasia?

A

A reversible change in which one adult cell type (usually epithelial) is replace by another adult cell type
- Adaptive

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

What is dysplasia?

A
  • An abnormal pattern of growth in which some of the cellular and architectural features of malignancy are present
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

How does dysplasia appear?

A
  • Loss of architectural orientation

- Loss in uniformity of individual cells

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

How do nuclei appear in dysplasia?

A

Hyperchromatic

Enlarged

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

How do mitotic figures appear in dysplasia?

A

Abundant
Abnormal
In places where they are not usually found

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

Where is dysplasia common?

A
  • Cervix- HPV infection
  • Bronchus- Smoking
  • Colon- UC
  • Larynx- Smoking
  • Stomach - Pernicious anaemia
  • Oesophagus- acid reflux
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What is neoplasia/tumour/malignancy?

A

An abnormal autonomous proliferation of cells unresponsive to normal growth control mechanisms

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

How do benign tumours differ from malignant tumours?

A

1) Do not invade/ do not metastasise
2) Encapsulated
3) Usually well differentiated
4) Slowly growin
5) Normal mitoses

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

When can benign tumours be fatal?

A

1) When in a dangerous place e.g. meninges, pituitary
2) Secretes something dangerous: insulinoma
3) Gets infected: bladder
4) Bleeds: stomach
5) Ruptures: liver adenoma
6) Torts (twisted): ovarian cyst

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

What are the characteristics of a malignant tumour?

A

1) Invade surrounding tissues
2) Spread to distant sites
3) No capsule
4) Well to poorly differentiated
5) Rapidly growing
6) Abnormal mitoses

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

What is a metastasis?

A

A discontinuous growing colony of tumour cells, at some distance from the primary cancer

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

What does a metastasis depend on?

A

The lymphatic and vascular drainage of the primary site

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

What is a benign tumour of the surface epithelium?

A

Papilloma

e.g. skin, bladder

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

What is a benign tumour of the glandular epithelium?

A

Adenoma

e.g. stomach, thyroid, colon, kidney, pituitary, pancreas

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

What is a carcinoma?

A

Malignant tumour derived from epithelium

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

What are the different types of carcinomas?

A
  • Squamous cell
  • Adenocarcinoma
  • Transitional cell
  • Basal cell carcinoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What is a sarcoma?

A

A malignant tumour derived from connective tissue (mesenchymal) cells

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

What are the different types of sarcomas?

A
Fat= liposarcoma 
Bone= osteosarcoma 
Cartilage= chondrosarcoma 
Muscle striated= rhabdomyosarcoma 
Muscle smooth= leiomyosarcoma 
Nerve sheath= malignant peripheral nerve sheath tumour
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What are the tumours of white blood cells?

A

Leukaemia

Lymphoma

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

What is leukaemia?

A

A malignant tumour of bone marrow derived cells which circulate in the blood

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

What is lymphoma?

A

A malignant tumour of lymphocytes (usually) in lymph nodes

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

What is teratoma?

A

A tumour derived from germ cells, which have the potential to develop into tumours of all three germ cell layers

1) ectoderm
2) mesoderm
3) endoderm

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

How do gonadal teratomas differ in males and females?

A
Males= all malignant 
Females= most are benign
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What is hamartoma?

A
  • Localised overgrowth of cells and tissues native to the organ
  • Cells are mature but architecturally abnormal
  • Common in children and should stop growing when they do
  • E.g. Bile duct hamartomas, bronchial hamartomas
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What is the criteria for assessing differentiation of a malignant tumour?

A
  • Evidence of normal function still present production of keratin, mucin, bile and hormones
  • Various grading systems, for Ca breast, prostate colon
  • There is no differentiation for anaplastic carcinoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What does the grade of a tumour describe?

A

Its degree of differentiation

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

What does the stage of tumour describe?

A

How far it has spread

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

What is more important (stage or grade) when determining prognosis?

A

Stage is more important.

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

What does the rate of cell division depend on?

A

1) Embryonic vs adult cells
2) Complexity of system
3) Necessity of renewal
4) State of differentiation (some cells never divide i.e. neurons)
5) Presence of tumour cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

Why is appropriate regulation cell division important?

A
  • Premature, aberrant mitosis results in cells death
  • In addition to mutations in oncogenes and tumour suppressor genes, most solid tumours are aneuploid
  • Various cancer cell lines show chromosome instability
  • Perturbation of protein levels of cell cycle regulators is found in different tumours
  • Contact inhibition of growth
  • Attacking the machinery that regulates chromosome segregation is one of the most successful anti-cancer strategies n clinical use
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What is the cell cycle?

A

Orderly sequence of events in which a cell duplicates its contents and divides in two

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

What is the order of the eukaryotic cell cycle?

A
M phase 
Interphase 
G0 
G1 
S 
G2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What occurs during the M phase?

A

Mitosis

  • Nuclear division
  • Cell division (cytokinesis)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What occurs during interphase

A

Duplication

  • DNA
  • Organelles
  • Protein synthesis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

Why is mitosis the most vulnerable period of the cell cycle?

A
  • Cells are more easily killed (irradiation, heat shock, chemicals)
  • DNA damage can not be repaired
  • Gene transcription is silenced
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What occurs during the S phase?

A

Replication for division
- DNA replication
- Protein synthesis: initiation of translation and elongation increased; capacity is also increased
- Replication of organelles (centrosome, mitochondria, Golgi, etc)
In the case of mitochondria, this needs to coordinate with the replication of mitochondrial DNA

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

What is the centrosome?

A
  • Consists of two centrioles (barrels of nine triplet microtubules)
  • Functions: microtubule organising center (MTOC) and mitotic spindle
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What are the six different phases of mitosis?

A
Prophase 
Prometaphase 
Metaphase 
Anaphase 
Telophase 
Cytokinesis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

What occurs during prophase?

A
  • Replicated chromosome condense to avoid breaking
  • Duplicated centrosomes migrate to opposite sides of the nucleus and organise the assembly of spindle microtubules
  • Outside the nucleus, the mitotic spindle assembles between the two centrosomes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What does each condensed chromosome consist of?

A

2 sister chromatids, each with a kinetochore

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

How does the mitotic spindle form?

A
  • Radial microtubule arrays (ASTERS) form around each centrosome (microtubule organising centres MTOC)
  • Radial arrays meet
  • Polar microtubules form
  • Microtubules are in a dynamic state
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What occurs during early prometaphase?

A
  • Breakdown of the nuclear membrane
  • Spindle formation is largely complete
  • Attachment of the chromosome to spindle via kinetochores (centromere region of chromosome)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What occurs during late prometaphase?

A
  • Microtubule from opposite pole is captured by sister kinetochore
  • Chromosomes attached to each pole congress to the middle
  • Chromosome slides rapidly towards centre along microtubules
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What occurs during metaphase?

A

The attached chromosome have undergone active movement and are lined up at the midline of the spindle.
The kinetochores of each sister chromatid are attached to opposite poles of the spindle

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

What occurs during anaphase?

A
  • Paired chromatids separate to form two daughter chromosomes
  • Cohesin holds sister chromatids together
  • Consists of Anaphase A and B
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What occurs during anaphase A?

A
  • Breakdown cohesin
  • Microtubules get shorter
  • Daughter chromosome pulled toward opposite spindle poles
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What occurs during anaphase B

A

1) Daughter chromosomes migrate towards poles

2) Spindle poles (centrosomes) migrate apart

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

What occurs during telophase?

A
  • Daughter chromosome arrive at the spindle
  • Nuclear envelope reassembles at each pole
  • Assembly of contractile ring
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

What is cytokinesis?

A

The stage where the cytoplasm is divided into two daughter cells by the contractile ring of actin and myosin filaments.
Each daughter cell now has its own nucleus

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

What is the Spindle- assembly checkpoint?

A

Controls the transition out of metaphase into anaphase by sensing completion of chromosome alignment and spindle assembly.

  • Achieved by monitoring kinetochore activity
  • Requirements for this include CENP-E and BUB protein kinases
  • BUBs dissociate when chromosomes are properly attaches to the spindle
  • When all has dissociated, anaphase proceeds
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

What is aneuploidy?

A

Abnormal number of chromosomes

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

What are the different ways in which aneuploidy can be caused?

A

1) Mis-attachment of spindle to kinetochores
2) Aberrant mitosis
3) Anti-cancer therapy

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

What are the types of attachment of the spindle to kinetochores?

A

Amphelic attachment= normal: kinetochores do not produce a checkpoint signal
Merotelic attachment= spindle attachment from the same centrosome to both kinetochores
Monotelic attachment= movement of both pairs of chromatids to the same pole
Synthelic attachment= movement of both pairs of chromatids to the same pole, but in this case they are attached to different spindles

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

Why does merotelic attachment have negative consequences?

A

Results in aneuploidy

  • Kinetochores won’t produce a checkpoint signal which indicates something is wrong
  • There is a loss of a chromosome at cytokinesis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

Why can synthelic attachment result in aneuploidy?

A

Both sister chromatids are at the same pole

- These sister chromatids are likely to break and their kinetochores may or may not produce checkpoint signals

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

Why does aberrant mitosis result in aneuploidy?

A

Results in 4 instead of 2 centrosomes being produced

- This leads to a multi-polar spindle with abnormal cytokinesis following. This results in 4 daughter cells

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

Why can anti-cancer therapy induce aneuploidy?

A
  • Drugs have been developed to induce chromosome mis-segregation with the aim of this to lead to apoptosis of the cancer cell
  • This is achieved by either inhibiting attachment-error-correction mechanisms, or inhibiting checkpoint protein kinases
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

Give examples of anti-cancer therapies that can result in aneuploidy

A
  • Taxanes and vinca alkaloids
  • Used in breast and ovarian cancers
  • Produces unattached kinetochores
  • Causes long-term mitotic arrest
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

What might happen if something goes wrong during the cell cycle?

A

1) Cell cycle arrest
- At check points (G1 and spindle check point)
- Can be temporary, allowing time for DNA repair
2) Programmed cell death (apoptosis)
- DNA damage too great and cannot be repaired
- Chromosome abnormalities
- Toxic agents

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

What is the G1/S checkpoint influenced by?

A

Growth factors

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

What is the G2/M checkpoint influenced by?

A

DNA damage

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

What is the Metaphase checkpoint influenced by?

A

Sister chromatid alignment

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

During the cell cycle, what occurs in the absence of stimulus?

A

The cells go into G0 (quiescent phase)
- This is how most differentiated cells in the body exist where they are able to perform specific functions without dividing

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

What does exit from the G0 phase require?

A
  • Is highly regulated
  • Requires growth factors and intracellular signalling cascades
  • This does not happen in tumour cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

How are signalling cascades activated?

A

In response to extracellular ligands that bind to cell surface receptors and trigger an intracellular signal

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

What happens to the intracellular signal that is triggered by a signalling cascade?

A
  • Amplified
  • Integrated
  • Modified by other pathways before diverging to have effects on metabolism, gene expression and the cytoplasm
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

Give an example of an intracellular signalling cascade

A

EGF (epidermal growth factor) and PDGF (platelet-derived growth factor) attach to their respective receptors (receptor protein tyrosine kinase)
- These are found in a monomeric, inactive state

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

During a signalling cascade, what happens in the presence of a ligand?

A
  • Receptors form dimers

- Are activated by phosphorylation of amino acid residues in the kinase domain

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

During a signalling cascade, how does phosphorylation to activate receptors occur?

A

Via the transfer of phosphate from ATP to a hydroxyl group on serine, threonine and tyrosine.

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

During a signalling cascade when the receptor is activated, how does the added phosphate group alter protein function?

A
  • The phosphate group is negatively charged
  • It causes a change in conformation leading to a change in activity (+ve or -ve)
  • Creating a docking site for another protein
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

During a signalling cascade, what does activation of a receptor result in?

A
  • Intracellular kinase cascade mediated by adaptor proteins which bind to the newly created docking site
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

What do protein kinase cascades result in?

A

Lead to signal amplification, diversification and opportunity for regulation

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

Why are their protein kinase cascades?

A

Frequently the protein regulated by a kinase is another kinase, and so on

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

How can protein kinase cascades be reversed?

A

By phosphatases

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

How long does G1 last?

A

10 hours

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

How long does the S phase last?

A

9 hours

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

How long does G2 last?

A

4 hours

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

Which stages are interphase?

A

G1
G0
G2

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

What is c-Myc?

A

A transcription factor which stimulates the expression of cell cycle genes
- Has an important role in G1

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

What are they key components of signalling pathways?

A

1) Regulation of enzyme activity by protein phosphorylation (kinases)
2) Adapter proteins
3) Regulation by GTP-binding proteins

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

How does growth factor stimulate signalling pathways?

A

Mitogenic growth factor -> Receptor protein tyrosine kinase -> Small G (GTP-binding) protein (Ras) -> Kinase cascade -> Immediate early genes- control the expression of other genes

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

What does the first 15 minutes of growth factor stimulation of signalling pathways consist of?

A
  • The mitogenic growth factor binding to the cell surface receptor protein tyrosine kinase
  • Phosphorylation of RTPK initiates the kinase cascade which provides additional docking sites and activates Ras
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

What does the Ras binding stimulate?

A

Stimulates and mediates early-response gene expression from the MAPK/ERK cascade e.g. c-Myc expression

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

What does the last hour of growth factor stimulation of signalling pathways involve?

A

Stimulation of expression of delayed-response genes which leads to the activation of the cell-cycle control system

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

What happens following phosphorylation of receptor protein tyrosine kinase?

A

Additional docking sites are formed.

Adapter proteins such as Grb2 dock to the phosphorylated site

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

What happens when Grb2 binds to the phosphorylated site?

A

Recruits inactive Ras protein to the cytosolic surface of the plasma membrane. The inactive Ras protein is associated with a GDP molecule.

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

How do Ras-activating proteins activate Ras?

A

Using exchange factor Sos, which exchanges a phosphate from a GTP molecule associate with the Ras-activating protein for the GDP molecule associated with Ras

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

How is Ras deactivated?

A

G-proteins act as a molecular switch, turning Ras off.

They do this through GTP hydrolysis using GTPase activating proteins (GAP)

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

How is Ras oncogenically activated?

A

Mutations that increase the amount of active GTP-loaded Ras.

The mutations either prevent GAP binding or prevent GTP hydrolysis.

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

What does Ras activate one it is in its active GTP-loaded form?

A

Protein kinase cascade.
Specifically= Extracellular signal-regulated kinase (ERK) cascade
Gernerically= Mitogen-activated protein kinase (MAPK) cascades

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

What are the generic and specific names of the protein kinase cascde

A
Kinase I-
Generic name: MAP-KKK
Specific: Raf 
Kinase 2-
Generic name: MAP-KK 
Specific: MEK 
Kinase 3- 
Generic name: MAP-K 
Specific: ERK
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

What does ERK go on to stimulate?

A

Changes in cell protein and gene expression (eg c-Myc) in order to promote cell division

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

What do mutated Ras and c-Myc act as?

A

Oncogenes

Lead to uncontrolled cell division and tumour proliferation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

What is cell cycle control based on?

A

Cyclin-dependent kinases (Cdks)

- They are present in proliferating cells throughout the cell cycle

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

How is the activity of cyclin-dependent kinases (Cdks) regulated?

A
  • Through their interaction with cyclins

- Their phosphorylation

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

What are cyclins?

A
  • Proteins which activate Cdks
  • Transiently expressed at specific points in the cell cycle
  • Level of expression if highly regulated
  • They are synthesised and then degraded
  • Form cyclin-Cdk complexes of which different complexes trigger different events in the cell cycle
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

What are the different cyclin-Cdk complexes which trigger different events in the cell cycle?

A

S phase: Cdk2/Cyclin A
Mitosis: Cdk1/Cyclin B
G1 progression: Cdk2/Cyclin E

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

What types of genes are activated by the Ras-ERK pathway?

A

c-jun, c-fos, c-myc encoding transcription factors

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

How does mitotic cyclin B interact with Cdk1?

A

They form a complex which acts as the mitosis promoting factor (MPF)

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

How is the mitosis promoting factor (MPF) activated?

A

Requires activating phosphorylation using CAK (cdk activating kinase)
- As well as removal of the inactivating phosphorylation by inhibitory kinase - Wee1

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

What determines whether the mitosis promoting factor becomes activated?

A

The balance between CAK and Wee1

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

Why is mitosis promoting factor activated at the end of interphase?

A

WEE1 is removed at the end of interphase using CDC25 phosphatase.
- There is positive feedback

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

How does mitosis progress?

A

Cdk1/cycB active- then mitosis is on hold and the key substrates are phosphorylated.
A signal from fully attached kinetochores causes cyclin B to be degraded
- Cdk1 inactivated
- Key substrates dephosphorylated
- Mitosis progresses

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

How does growth factor stimulation of signalling pathways promote G0 to G1 transition?

A

c-Myc is an immediate early gene transcription factor.

- c-Myc stimulates transcription of cyclin D which is required for re-entry into the cell cycle from G0

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

How does Cyclin D stimulate progression from G1 into the S phase?

A

Cyclin D activate cdk4 and cdk6 which stimulates cyclin E production which is required for the cell cycle to progress

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

What is the purpose of Cdks

A

Sequentially activated by cyclins and stimulate the synthesis of genes required for the next phase
e.g. CycD/Cdk4/6 stimulates the expression of CycE

  • This gives direction and timing to the cycle
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

How Cdks work?

A

They phosphorylate proteins (or Serine or threonine) to drive cell cycle progression

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

What do MPF target?

A

Nuclear lamins.

The phosphorylation of which causes the breakdown of the nuclear envelope

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

What does Cdk4/6- Cyclin D target?

A

Retinoblastoma protein
The phosphorylation of which inactivates the protein
Releases E2F transcription factor, driving gene transcription of Cyclin E and allowing the progression of the cell cycle from G1 > S phase

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

How can the Cdk5/6-cyclin D complex have negative effects.

A
Retinoplastoma protein (pRB) is a tumour supressor which acts as a break on the cell cycle until it receives the signal from the Cdk4/6-Cyclin D complex 
- Is the tumor suppressor is lost, uncontrolled cell division may occur
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

How Cdk inhibitors (CKIs) regulate Cdks

A

Inhibit them to prevent progression through the cell cycle, which is important in ensuring the integrity of the cell cycle is maintained

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

What are the two familiess of CDk inhibitors (CKIs)

A

INK4

CIP/KIP

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

How does the INK4 family inhibit Cdks

A

Inhibits Cdk4/6 by displacing cyc D

- Arrest G1 at the restriction checkpoint

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

How does the CIP/KIp family inhibt Cdks

A

Inhibits all Cdks especially in the S phase

- Does this by binding to the Cdk/cyc complex

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

What codes for INK4?

A

p16

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

What codes for CIP/KIP?

A

p27

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

How can problems with Cdk inhibitors result in cancers?

A
  • Loss of INK4 inhibition
  • Over-production of cdk4/cyclin D
  • Loss of retinoblastoma protein (highly prevalent in lung cancer)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

How can cell cycle regulatory proteins lead to cancer and act as oncogenes?

A
  • EGFR/HER2= mutationally activated or over-expressed in many breast cancers
  • Ras= mutationally activated in many cancers
  • Cyclin D1
  • B-Raf
  • c-Myc
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

Why may Ras lead to cancer?

A

When mutationally activated.

They are inhibitors of membrane attachment

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

Which tumour suppressors and cell cycle regulatory proteins are inactivated in cancer?

A

Rb

p27KIP1

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

What are the types of genetic mutations that cause cancer?

A
  • Chromosome translocation
  • Gene translocation
  • Gene amplification (copy number variation)
  • Point mutations within promotor or enhancer regions of genes
  • Deletions or insertions
  • Epigenetic alterations to gene expression
  • Can be inherited
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

What are the components of cytotoxic chemotherapy?

A

1) Alkylating agents
2) Antimetabolites
3) Anthracyclines
4) Vinca alkaloids and taxanes
5) Topoisomerase inhibitors

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

How is cytotoxic chemotherapy administered?

A
  • Given intravenously or by mouth
  • Works systemically
  • Non ‘targeted’ affects all rapidly dividing cells in the body
  • Given post-operatively: adjuvant
    Pre-operatively: neoadjuvant
    As monotherapy or in combination
    With curative or palliative intent
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

How do alkylating agents act in cytotoxic chemotherapy?

A
  • Add alkyl groups to guanine residues in DNA
  • Cross-link DNA strands and prevent DNA from uncoiling at replication
  • Trigger apoptosis (at checkpoint pathway
  • Encourage miss-pairing- oncogenic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

What are pseudo-alkylating agents?

A
  • Add platinum to guanine residues in DNA
  • Same mechanism of cell death as alkylating agents
    E.G. Carboplatin, cisplatin, oxaliplatin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

Give examples of alkylating agents

A

Chlorambucil
Cycophosphamide
Dacarbazine
Temozolomide

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

What are the side effects of alkylating and pseudoalkylating agents?

A
  • Cause hair loss ( not carboplatin)
  • Nephrotoxicity
  • Neurotoxicity
  • Ototoxicity (platinums)
  • Nausea
  • Vomiting
  • Diarrhoea
  • Immunosuppression
  • Tiredness
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

How do anti-metabolites act in cytotoxic chemotherapy?

A

Masquerade as purine or pyrimidine residues leading to inhibition of DNA synthesis, DNA double strand breaks and apoptosis

  • Block DNA replication and transcription
  • Can be purine (adenine and guanine), pyrimidine or folate antagonists
  • Examples= methrorextate (folate), 6-mercaptopurine, carbazine and fludarabine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

What are the side effects of anti-metabolites?

A
  • Hair loss (alopecia): not 5FU or capecitabine
  • Bone marrow suppression causing anaemia, neutropenia and thrombocytopenia
  • Increased risk or neutropenic sepsis (and death) or bleeding
  • Nausea and vomiting (dehydration)
  • Mucositis and diarrhoea
  • Palamar-plantar erthrodysethesia (PPE)
  • Fatigue
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

How do anthracyclines work in cytotoxic chemotherapy?

A
  • Inhibit transcription and replication by intercalating (i.e. inserting between) nucleotides within the DNA/RNA strand
  • Also block DNA repair- mutagenic
  • They create DNA and cell membrane damaging free oxygen radicals
  • E.g. doxorubicin, epirubicin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

What are the side effects of anthracyclines?

A
  • Cardiac toxicity (arrhythmias, heart failure)
  • Alopecia
  • Neutropenia
  • Nausea and vomiting
  • Fatigue
  • Skin changes
  • Red urine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

How do vinca alkaloids and taxanes work in cytotoxic chemotherapy?

A

Inhibit assembly (vinca alkaloids or disassembly (taxanes) of mitotic microtubules causing dividing cells to undergo mitotic arrest

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

What are the side effects of microtubule targeting drugs?

A
  • Nerve damage: peripheral neuropathy, autonomic neuropathy
  • Hair loss
  • Nausea
  • Vomiting
  • Bone marrow suppression (neutropenia, anaemia etc)
  • Arthralgia
  • Allergy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

How do topoisomerase inhibitors work in cytotoxic chemotherapy?

A
  • Topoisomerases are required to prevent DNA torsional strain during DNA replication and transcription
  • Induce temporary single strand (topo1) or double strand (topo2) breaks in the phosphodiester backbone of DNA
  • They protect the free ends of DNA from aberrant recombination events
  • Drugs such as anthacyclines have anti-toposomerase effects through their action on DNA
  • Specific topoisomerase inhibitors include Tpotecan and irinotecan (topo I) and etoposide (topo II) alter binding of the complex to DNA and allow permanent DNA breaks)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

What are the side effects of topoisomerase inhibitors?

A
  • Irinotecan- Acute cholinergic type syndrome- diarrhoea, abdominal cramps and diaphoresis (sweating). - Therefore given with atropine
  • Hair loss
  • Nausea, vomiting
  • Fatigue
  • Bone marrow suppression
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

How can a resistant cell survive?

A
  • Drug effluxed from the cell by ATP-binding cassette (ABC) transporters
  • DNA adducts replaced by Base Excision repair (using PARP)
  • DNA repair mechanisms are up-regulated and DNA damage is repaired so the DNA double strand does not break
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

What are the ten hallmarks of a cancer cell?

A

1) Self-sufficient
2) Insensitive to anti-growth signals
3) Anti-apoptoctic
4) Pro-invasive and metastatic
5) Pro-angiogenic
6) Non-senscent
7) Dysregulated metabolism
8) Evades the immune system
9) Unstable DNA
10) Inflammation

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

What receptors are over-expressed in cancers?

A

HER2: amplified and over-expressed in 25% breast cancer
EGFR: over-expressed in breast cancer and colorectal cancer
PDGFR: glioma (brain cancer

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

What ligands are over-expressed in cancers?

A

VEGF: prostate cancer, kidney cancer, breast cancer

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

What are the different monoclonal antibodies?

A
  • momab ( derived from mouse antibodies)
  • ximab (chimeric) e.g. cetumixab
  • zumab (humanised) e.g. bevacizumab, trastuzumab
  • mumab (fully human) e.g. panitumumab
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

What do monoclonal antibodies target?

A

The extracellular component of the receptor

  • They neutralise the ligand
  • Prevent receptor dimerisation
  • Cause internalisation of receptor
  • mAbs also activate Fcy-receptor-dependent phagocytosis or cytolysis induced complement-dependent cytotoxicity (CDC) or antibody-dependent cellular cytotoxicity (ADCC)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

Which monoclonal antibodies are used in oncology?

A
  • Bevacizumab binds and neutralises VEGF. Improves survival colorectal cancer
  • Cetuximab targets EGFR
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

What is the mechanism of small molecule inhibitors (SMIs)?

A

Bind to the kinase domain of the tyrosine kinase within the cytoplasm and block autophosphorylation and downstream signalling
- Also act on intracellular kinases therefore can affect cell signalling pathways

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

What is Glivec?

A

A small molecule inhibitor that targets the ATP binding region within the kinase domain

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

Give examples of SMIs inhibiting receptors

A

Erlotinib (EGFR)
Gefitinib (EGFR)
Lapatinib (EGFR/HER2)
Sorafinib (VEGFR)

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

Give examples of SMIs inhibiting intracellular kinases

A

Sorafinib (Raf kinase)
Dasatinib (Src kinase)
Torcinib (mTOR inhibitors)

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

What are the advantages of SMIs

A
  • Can target TKs without an extracellular domain or which are constitutively activated (ligand independent)
  • Pleiotropic tagets (useful in heterogenic tumours/cross talk)
  • Oral administration
  • Good tissue penetration
  • Cheap
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
148
Q

What is a disadvantage of SMIs and monoclonal antibodies?

A

Resistance

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

What are the different resistance mechanisms to targeted therapies?

A
  • Mutations in ATP-binding domain (e.g. BCR-Abl fusion genes and ALK gene, targeted by Glivec and crizotinib respectively)
  • Intrinsic resistance (herceptin effective in 85% HER2 & breast cancers, suggesting other driving pathways)
  • Intragenic mutations
  • Upregulation of downstream or parallel pathways
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
150
Q

What are anti-sense oligonucleotides?

A
  • Single stranded, chemically modified DNA like molecule. 17-22 nucleotides in length
  • Complementary nucleic acid hybridisation to target gene hindering translation of specific mRNA
  • Recruits RNASe H to cleave target mRNA
  • Good for ‘undruggable’ targets
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
151
Q

What is cell behaviour?

A

The term used to describe the way cells interact with their external environment and their reactions to this, particularly proliferative and motile responses of cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

What external influences are detected by cells?

A
  • Chemical: hormones, growth factors, ion concentrations, ECM, molecules on other cells, nutrients and dissolve gas (O2/CO2) cells
  • Physical: mechanical stresses, temperature, the topography or layout of the ECM and other cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

What is cell-spreading dependent on?

A

Gravity dependent

Energy is required to modulate cell adhesion and the cytoskeleton during spreading

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

What is the importance of cell- ECM adhesion?

A
  • In suspension, cells do not significantly synthesise protein or DNA
  • Cells require to be attached to ECM (and a degree of spreading is required) to begin protein synthesis and proliferation (DNA synthesis)
  • Attachment to ECM may be required for survival (e.g. epithelia, endothelia)
  • i.e. anchorage dependence
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

What are cell-ECM adhesion molecules?

A
  • Cells have receptors on their cell surface which bind specifically to ECM molecules
  • These molecules are often linked at their cytoplasmic domains, to the cytoskeleton
  • This arrangement means that there is mechanical continuity between ECM and the cell interior
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
156
Q

What are integrins?

A

Heterodimer complexes of alpha and beta subunits that associate extracellularly by their ‘head’ regions. Each of the ‘tail’ regions spans the plasma membrane.

  • Recognise short, specific, peptide sequences
  • More than 20 combinations of alpha/beta known
  • Each combination specifically binds a particular peptide sequence
  • Such peptide sequences found in more than one ECM molecule e.g. RGD found in fibronectin, virtonectin, fibrinogen plus others
  • Linked via actin binding proteins to the actin cytoskeleton
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
157
Q

What do integrin complexes cluster to form?

A

Focal adhesions
Hemidesmosome (alpha6Beta4- linked to the cytokeratin network)
- These clusters are involved in signal transduction
- Integrins also bind to specific adhesion molecules to some cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
158
Q

How can ECM receptors (e.g. integrins) act to transduce signals?

A
  • ECM binding to an integrin complex can stimulate the complex to produce a signals inside the cell i.e. ‘outside-in’ integrin signalling
  • A signal generated inside the cell can act on an integrin complex to alter the affinity of an integrin i.e. inside out integrin signalling (e.g. in inflammation or blood-clotting, switching on adhesion of circulating leukocytes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
159
Q

What is outside- in signalling

A
  • A cell can receive information about its surroundings from its adhesion to ECM
  • E.g. the composition of the ECM will determine which integrin complexes bind and which signals it receives
  • This can alter the phenotype of the cell
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
160
Q

Describe the density-dependence of cell division

A
  • When cells in culture form a confluent monolayer, they cease proliferating and slow down many other metabolic activities.
    This used to be known as contact inhibition of cell division
  • Another set of experiments have now suggested that it is competition for external growth factors and not cell-cell contact responsible. This is known as the density dependence of cell division.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
161
Q

What is the ERK MAP kinase Cascade?

A
  • The pathway by which growth factors induce gene expression and cell proliferation is known as the ERK MAP kinase cascade
  • The signal starts when a growth factor binds to the receptor on the cell surface and ends when the DNA in the nucleus expresses a protein and produces some change in the cell, such as cell division.
  • The proteins included are MAPK which communicate by adding phosphate groups to a neighbouring protein- acts as an on or off switch
  • The extracellular growth factor binds to the membrane ligand. This allows Ras to swap its GDP for a GTP. It can now activate MAP3K, which activates MAP2K which activates MAPK. MAPK can now activate a transcription factor, such as myc
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
162
Q

What is proliferation dependent on?

A

Density (e.g. growth factor)

Anchorage (e.g. ECM)

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

What is the mechanism of anchorage dependence?

A
  • Growth factor receptors and integrin signalling complexes can each activate identical signalling pathways (e.g. MAPK)
  • Individually, this activation is weak and/or transient
  • Together, activation is strong and sustained
  • The separate signalling pathways act synergistically
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
164
Q

What are the types of contact interactions between cells?

A

Short term: transient interactions between cells which do not form stable cell-cell junctions
Long-term: stable interactions resulting in formation of cell-cell junctions

165
Q

What is the cell-cell contact between non-epithelial cells?

A
  • Contact inhibition of locomotion
  • When non-epithelial cells collide they do not form stable cell-cell contacts
  • They repel one another by paralysing motility at the contact site, promoting the formation of a motile front at another at another site on the cell and moving off in the opposite direction
  • The contact inhibition of locomotion is responsible for preventing multilayering of cells in culture and in vivo
166
Q

What are the long-term cell-cell contacts?

A
  • Upon contact, some cell types strongly adhere and form specific cell-cell junctions (adherens junctions, desmosomes, tight junctions, gap junctions)
  • This is true of epithelial cells and endothelial cells, which form layers and neurones forming synapses
167
Q

How are cell-cell junctions arranged in epithelia?

A

As continuous belts (zonula) or as discrete spots (macula)

168
Q

What causes contact-induced spreading of epithelial cells?

A

Contact between epithelial cells leads to the mutation induction of spreading.
The total spread area of the contacted cells is greater than that of the sum of the two separated cells
- This could result in a stable monolayer

169
Q

How does cell-cell adhesion affect cell proliferation?

A

The formation of cell-cell junctions occurs when the MAP Kinase pathway is active, there is increase p27kIP1. It results in low proliferation

170
Q

Give examples of SMIs inhibiting receptors

A

Erlotinib (EGFR)
Gefitinib (EGFR)
Lapatinib (EGFR/HER2)
Sorafinib (VEGFR)

171
Q

Give examples of SMIs inhibiting intracellular kinases

A

Sorafinib (Raf kinase)
Dasatinib (Src kinase)
Torcinib (mTOR inhibitors)

172
Q

What are the advantages of monoclonal antibodies?

A
  • High target specificity
  • Cause ADCC, complement mediated cytotoxicity and apoptosis infuction
  • Can be radiolabelled
  • Cause target receptor internalisation
  • Long half-life (lower dosing frequency)
  • Good for haematological malignancies
  • Liked by regulatory authorities (FDA)
173
Q

What external factors can influence cell division?

A

Growth factors, cell-cell adhesion and cell-ECM adhesion

174
Q

How are adherens junctions mediated?

A
  • By cadherins
  • Molecules that span the plasma membrane and associated with identical molecules on adjacent cells
  • Cadherin in the transmembrane homophilic cell adhesion molecule (Ca++ dependent) that associated with beta-catenin intracellular
  • Beta-catenin then associates with an alpha-catenin molecule which associates with an actin filament
175
Q

Is Beta-catenin the link between cell-cell adhesion and proliferation?

A
  • When bound to cadherin, Beta-catenin is not available for LEF-1 binding
  • In adenomatous polyposis coli (APC), the ATP gene-product is involved in the degradation of the junction-associated molecule beta-catenin
  • Results in an overgrowth of the colon epithelium which results in thousands of polyps
176
Q

What are the dynamics of beta-catenin?

A
  • APC part of a complex that phosphorylates Beta-catenin. When it does so, it targets it for degradation. If beta-catenin hangs around in the cytoplasm, it gets targeted with LEF-1, goes into the nucleus and switches on proliferation
177
Q

What is the mechanism for contact inhibition of proliferation?

A
  • When bound to cadherin at the membrane, beta catenin is not available for LEF-1 binding and nuclear effects
  • Normally, cytoplasmic beta-catenin rapidly degraded
  • If beta-catenin cytoplasmic levels rise as a result of inhibition of degradation or loss of cadherin-mediated adhesion, beta-catenin/lef-1 complex enters nucleus and influences gene expression, leading to proliferation
178
Q

What other adhesion-associated signalling pathways are known to influence contact-induced inhibition of proliferation?

A
  • Clustering of cadherins after cell-cell contact is known to alter the activation of small GTPases e.g. Rac is activated, Rho is inhibited: this can influence proliferation
  • Some growth factor receptors are associated with cell-cell junctions
179
Q

In what ways can cells lose their social skills?

A
  • Proliferate uncontrollably (lose density dependence of proliferation)
  • Are less adherant and will multilayer (lose contact inhibition of locomotion and anchorage dependence)
  • Epithelia breakdown cell-cell contacts
  • Not Hayflick limited, express telomerase i.e. cancer
180
Q

What is the mechanism of short-circuiting leading to uncontrolled proliferation?

A
  • If a gene coding for a component of a signalling pathway is mutated so that the protein is constitutively active, that pathway will be permanently ‘on’
  • Receptors, signalling intermediates and signalling targets (e.g. transcription factors) are proto-oncogenes
  • This mechanism arises as a result of loss of growth factor dependence
181
Q

What oncogene?

A

Mutant gene with promotes uncontrolled cell proliferation

182
Q

What is a proto-oncogene?

A

Normal cellular gene corresponding to the oncogene

183
Q

What proportion of cancers is Ras mutated in?

A

30%

184
Q

How does a primary carcinoma cell metastasise?

A
  • Cell-cell adhesion must be down-regulated (e.g. cadherin levels reduced)
  • The cells must be motile
  • Degradation of ECM must take place: matrix metalloproteinase (MMP) levels increased in order to migrate through basal lamina and interstitial ECM
  • The degree of carcinoma cell-cell adhesion is an indicator of how differentiated the primary tumour is and indicates its invasiveness and the prognosis
185
Q

What molecular mechanisms regulate motility?

A
  • Microfilaments
  • Regulation of actin dynamics
  • Cytoskeletal proteins
  • Signalling proteins
186
Q

What are the steps in tumours progression?

A

1) Homeostasis
2) Genetic alterations
3) Hyper-proliferation
4) De-differentiation
- Disassembly cell-cell contacts
- Loss polarity
5) Invasion
- Increased motility
- Cleavage ECM proteins

187
Q

What acts as stimuli for cells to move?

A
  • Organogenesis and morphogenesis
  • Wounding
  • Growth factors/chemoattractants
  • Dedifferentiation (tumours)
188
Q

Where does a cell go when it is stimulated to move?

A

It changes shape to achieve polarity and will therefore have a directionality to the movement

189
Q

When does the cell know when to stop moving and how to move?

A

When to stop: contact-inhibition motility

How to move: specialised structures (focal adhesion, lamellae, filopodium)

190
Q

How does the cell attach to ECM proteins

A
  • Attachment to substratum is important for cell movements
  • Focal adhesions are sites at which the cell attaches to proteins which make the ECM
  • Filamentous actin lies beneath the membrane, it hooks the focal adhesions to the cytoskeletons via integrins
  • Integrins interact with various cytoskeletal proteins to form a plaque on the intracellular side
191
Q

What are filopodia?

A

Finger like protrusions that are rich in actin

  • Structures used for cell motility
  • Protrude from the cell to sense where they want to attach and direction of movement
  • Vinuncilin is the protein that overlies these protrusions
192
Q

What are lamellipodia?

A

Sheet like protrusions that are rich in actin filaments

  • Structures used for motility
  • Attach to the substratum and when they move back, the structures are called ruffles
193
Q

Why is control of cell movement needed?

A
  • Within a cell to coordinate what is happening in different parts
  • Regulate adhesion/release of cell-extracellular matrix receptors
  • From outside to respond to external influences
194
Q

What are the different types of motility?

A

Hapoptatic: movement with no direction
Chemotactic: movement in which the cell senses a stimulus and goes towards it

195
Q

How do cells regulate their shape changes by regulating their actin cytoskeleton?

A
  • Actin can be found as small soluble globular monomers (G-actin) or in a large polymerised filamentous polymer (F-actin)
  • If there is a signal like a nutrient source at one end of the cell the F-actin at the other end will disassemble and the subunits will diffuse tot he side with the signal. There is reassembly of the subunits at the new site and this facilitates movement
196
Q

How does the arrangement of actin filaments allow a variety of structure to be made?

A

Lamellipodium: has branched and crosslinked filaments
Filopodium: bundle of parallel filaments
Stress fibers: has antiparallel arrangement which forms contractile bundles

197
Q

What is the purpose of the motor proteins and in the remodelling of actin filaments?

A

Motor: They provide contraction.

Other proteins bind to stabilise the filaments

198
Q

What is actin dynamics?

A

The cycle between G-actin and F-actin

- Remodelling of actin filaments is driven by different binding proteins

199
Q

What is the nucleation step in actin dynamics?

A

The rate limiting step in actin dynamics- it involves the formation of trimers to initiate polymerisation

  • Arp 2 & 3 (actin-related proteins) are similar to actin, and form a complex on which the trimers of actin can attach (on the minus end)
  • Elongation of the filament can then occur from the Arp2/3 complex
200
Q

What is the elongation step in actin dynamics?

A
  • Brings monomers (usually sequestered) to the filament
  • Profilin binds to the monomers and help elongate the filament
  • Thymosin is a sequestering protein which prevents polymerisation
  • There is a balance between these two proteins to regulate filament growth, as profilin competes with thymosin for binding to actin monomers and promote assembly
201
Q

What is the purpose of branching in actin dynamics?

A

Branched filaments form a precise 70 degree angle.

Arp complex binds to the filament to provide nucleation a a different site

202
Q

What is the purpose of capping proteins in actin dynamics?

A
  • Prevent further growth of the filament
  • Capping proteins include Cap Z, Gelsolin, Fragmin and Severin at the plus end. Tropomodulin and Arp are proteins at the minus end
  • Can be used to regulate directionality of growth
203
Q

What proteins are involved in cross-linking and bundling of actin dynamics?

A
alpha-actinin 
fimbrin 
filamin 
spectrin 
villin 
vinculin
204
Q

What is the purpose of servering in actin dynamics?

A
  • Used to prevent low rate of growth/shrinking of very long filaments
  • In unsevered population actin filaments grow and shrink relatively slowly
  • In severed population actin filaments grow and shrink more rapidly
  • Severing proteins include: gelsolin, ADF/cofilin, fragmin/severin
  • E.g., severin is used in the gel-sol transition
205
Q

What are activities of actin during cell movement?

A

Disassemble, nucleation, branching, severing, capping and bundling

206
Q

What actin activities are involved during lamellae protrusion?

A

Polymerisation, disassembly, branching, capping
- During protrusion there is net filament assembly at the leading edge, and net filament disassembly behind the leading edge

207
Q

What actin activities are involved in the formation of filopodia?

A

Actin polymerisation, bundling and cross-linking to form the tight parallel bundles

208
Q

Which signalling mechanisms regulate the actin cytoskeleton?

A

1) Ion flux changes i.e. intracellular calcium
2) Phosphoinositide signalling (phospholipid binding)
3) Kinases/phosphatases (phosphorylation cytoskeletal proteins)
4) Signalling cascades via small GTPases

209
Q

How is the actin cytoskeleton controlled by small G proteins?

A
  • Rho subfamily of small GTPases belongs to the Ras super family
    Family members: Rac, Rho, Cdc43 best known
  • Participate in a variety of cytoskeletal processes
  • These proteins are activated by receptor tyrosine kinase, adhesion receptors and signal transduction pathways
  • Expression levels are up-regulated in different human tumours
210
Q

How do small GTPases participate in cell migration?

A
  • Rac is an important protein for lamellar protrusion
  • It controls the actin branching
  • Rho is important in the cell contraction
  • Rho is also important in the retraction of the cell to the rear ie. the arrangement of stress fibres and tension
  • Both rac, rho and CDC42 control how the cells form focal adhesions
  • Rho gives a very strong adhesion whereas cdc43 has a more docile adhesion
  • Cdc42 regulates filopodia, polarised motility and actin polymerisation
211
Q

Why is programmed cell death important?

A

To remove-

1) Harmful cells (e.g. cells with viral infection, DNA damage)
2) Developmentally defective cells (e.g. B lymphocytes expressing antibodies against self antigens)
3) Excess/unnecessary cells (embryonic development e.g. brain to eliminate excess neurons, liver regeneration, sculpting of digits and organs)
4) Obsolete cells (e.g. mammary epithelium at the end of lactation)
5) Exploitation- chemotherapeutic killing of cells

212
Q

What is necrosis and apoptosis?

A

Necrosis: unregulated cell death associated with trauma, cellular disruption and an inflammatory response
Apoptosis: regulated cell death; controlled disassembly of cellular contents without disruption: no inflammatory response

213
Q

What happens during necrosis?

A
  • Plasma membrane becomes permeable
  • Cell swelling and rupture of cellular membranes
  • Release of proteases leading to autodigestion and dissolution of the cell
  • Localised inflammation
214
Q

What happens during the latent phase of apoptosis?

A

Death pathways are activated, but cells appear morphologically the same

215
Q

What happens during the execution phase of apoptosis?

A
  • Loss of microvilli and intercellular junctions
  • Cell shrinkage
  • Loss of plasma membrane asymmetry (phospatidylserine lipid appears in outer leaflet)
  • Chromatin and nuclear condensation
  • DNA fragmentation
  • Formation of membrane blebds
  • Fragmentation into membrane-enclosed apoptotic bodies
  • There is no inflammation as plasma membrane remains intact
216
Q

What is the result of DNA modification in apoptosis?

A
  • Fragmentation of DNA ladders (seen in agarose gel)

- Formation of more ‘ends’ (which can be labelled fluorescently in a TUNEL asasay)

217
Q

What are apoptosis-like PCD and necrosis-like PCD?

A

Apoptosis-like PCD: some, but not all, features of apoptosis. Display of phagocytic recognition molecules before plasma membrane lysis
Necrosis-like PCD: variable features of apoptosis before cell lysis: aborted apoptosis

218
Q

What are the mechanisms of apoptotic cell death?

A

1) The executioners- Caspases
2) Initiating the cell death programme
- death receptors
- mitochondria
3) The Bcl-2 family

219
Q

What are caspases?

A

Cysteine-dependent aspartate-directed proteases

  • Executioners of apoptosis
  • Activated by proteolysis
  • Cascade of activation
220
Q

What are the classes of caspases?

A
Effector caspases (3,6 and 7) 
Initiator caspases ( 2,8,9 and 10) 
- Initiator caspases have subunits either called CARD or DED 
CARD- Caspase Recruitment Domain 
DED- Death Effector Domain
221
Q

How do caspases maturation?

A
  • Procaspases (zymogens) are single chain polypeptides. To be activated they undergo proteolytic cleavage to form a large and small subunit
  • These cleavages are done by the caspases themselves
  • Cleavage of the inactive procaspase precursor is followed by folding of 2 large and 2 small chains to form an active L2S2 heterotretamer
222
Q

What do caspase cascades consist of?

A
  • Amplification
  • Divergent responses
  • Regulation
  • Initiator caspases trigger apoptosis by cleaving and activating
  • Effector caspases carry out the apoptotic programme
223
Q

How do effect caspases execute the apoptotic programme?

A
  • Cleave and inactivate proteins or complexes (e.g. nuclear lamins leading to nuclear breakdown)
  • Activate enzymes (include protein kinases: nucleases e.g. Caspase Activated DNase, CAD) by direct cleavage, or cleavage or inhibitory molecules
224
Q

What are the mechanisms of caspase activation?

A

1) Death by deign- receptor-mediated (extrinsic) pathways

2) Death by default- mitochrondrial (intrinsic) death pathway

225
Q

What do death receptors consist of and how are they activated?

A

Extracellular cysteine-rich domain
Single transcellular domain
Cytoplasmic tail with ‘death domain’
- They are activated when they encounter secreted or transmembrane trimeric ligands (TNF or Fas) called DEATH LIGANDS

226
Q

What are the adaptor proteins in the death by design pathway?

A
  • FADD- a positive regulator which promotes cell death
    = The two domains of FADD are DED (death effector domain) and DD (death domain)
  • FLIP- a negative regulator which inhibits the death pathway
  • FLIP contains two DED domains
227
Q

What is the death receptor signalling process?

A

1) Death ligand binds to the death receptor (i.e. Fas receptor engaged by Fas ligand)
2) Death receptor undergoes trimerisation which brings the 3 cytoplasmic DD domains together
3) Trimerised death domains recruit the positive adaptor protein by its own DD
4) Binding of FADD causes recruitment and oligomerisation of procaspase 8
5) Binding of caspase 8 leads to formation of a DISC (death inducing signalling complex)
6) DISC formation results in cross-activation of procaspase8 where they cleave each other within the complex. Active caspase 8 is release where it cleaves effector caspases to execute the death programme

228
Q

What inhibits death receptor activate of caspase 8?

A

FLIP

  • It has caspase homology in DED domain but no proteolytic activity therefore it competes with the procaspase
  • Competes for binding to receptor tails
  • Incorporates into receptor/procaspase complexes and interferes with transcleavage
229
Q

What happens during mitochondrial regulation of apoptosis?

A
  • Loss of mitochondrial membrane potential
  • Release of cytochrome c
  • Release of other apoptosis inducing factors
  • Formation of the apoptosome complex
230
Q

What is the apoptosome complex?

A

’ Wheel of death’- Contains APAF-1, cycotchrome c, ATP and procaspase 9

  • Each APAF-1 in the heptameric apoptosome can potentially bind a procaspase 9
  • Oligomerisation brings multiple procaspase9s close together, resulting in cleavage, activation and release as active caspase 9 tetramer, which initiates a caspase cascade leading to apoptosis
  • Procaspase 9 binds to CARD
231
Q

What links the receptor mediated and mitochondria mediated death pathways?

A

A protein called Bid

- Caspase 8 cleaved Bid which enhances the release of mitochondrial proteins, thus engaged the intrinsic pathway

232
Q

Why does apoptosis require energy?

A
  • The apoptosome requires ATP

- Energy levels in the cell may determine whether death is by necrosis or apoptosis

233
Q

What is the Bcl-2 Family?

A

Proteins that are intrinsic modulators of apoptosis
- 3 groups, all of which contain the BH3 domain
- Members of the family are in two categories:-
Anti apoptotic proteins: localised to mitochrondrial membrane and inhibit apoptosis (Bcl-2, Bcl-xL)
Pro-apopotic proteins: move between the cytosol and the mitochondrial membrane promoting apoptotis (Bid, Bad, Bax, Bak)

234
Q

What is the role of the PI3’-Kinase signalling pathway in cell cycle and apoptosis regulation?

A

PI3’-K is a lipid kinase involved in growth control and cell survival

  • Activate kinase PKB/Akt which is anti-apoptotic
  • PKB phosphorylates the BAD and causes it to inactivate leading to cell survival
  • When growth factors are absent, PkB fails to come to the cell membrane so Bad is dephosphorylated and is released from its heterodimer.
  • Bad will displace Bcl-2/-cL from Bax/Bak leading to apoptosis
235
Q

What is the role of PBK/Akt?

A

1) Phosphorylates and inactivates Bad
2) Phosphorylates and inactivates caspase 9
3) Inactivates FOXO transcription factors (FOXOs promote expression of apoptosis-promoting genes)
4) Other e.g. stimulates ribosome production and protein synthesis

236
Q

What is PTEN?

A

A lipid phosphatase and extrinsic regulator which counteracts PI3-K signalling. It reduces the regulation of cell survival and promotes apoptosis

237
Q

What are IAPs?

A

Inhibitors of apoptosis proteins

  • They regulate programmed cell death
  • Bind to procaspases and prevent their activation. Bind to active caspases and inhibit their activity
238
Q

What are the characteristics of the cancer cell phenotype?

A
  • Disregard of signals to stop proliferating
  • Disregard of signals to differentiate
  • Capacity of sustained proliferation
  • Evasion of apoptosis
  • Ability to invade
  • Ability to promote angiogenesis
239
Q

What are pro-oncogenes?

A

They code for essential proteins involved in the maintenance of cell growth division and differentiation
- Can be converted into oncogene due to mutation

240
Q

What will a point mutation or deletion of a proto oncogene lead to?

A

Aberrantly active protein

241
Q

What will gene amplification of a proto-oncogene lead to?

A

Overproduction of normal protein

242
Q

What is the result of chromosomal translocation (chimaeric genes) or insertional mutagenesis of proto-oncogenes?

A
  • Strong enhancer increases normal protein levels e.g. Burkitt’s lymphoma
  • Fusion to actively transcribed gene overproduces protein or fusion protein is hyperactive e.g. Philadelphia chromosome
243
Q

What are the consequences of mutant RAS?

A
  • Upon binding GTP, Ras becomes active
  • Dephosphorylation of the GTP to GDP switches RAS off
  • Mutant RAS fails to dephosphorylate GTP and remains active
244
Q

What tumours are associated with the SRC oncogene?

A

Breast, colon, lung

- Cytoplasmic location

245
Q

What tumours are associated with the MYC oncogene?

A

Burkitt’s lymphoma

246
Q

What tumours are associated with the JUN oncogene?

A

Lung

247
Q

What tumours are associated with the Ha-RAS oncogene?

A

Bladder

248
Q

What tumours are associated with the Ki-RAS oncogene?

A

Colon

Lung

249
Q

Why is mutation or deletion of one gene copy usually insufficient to promote cancer?

A
  • Typically proteins whose function it is to regulate proliferation, maintain cell integrity
  • Each cell has two copies of each tumour suppressor gene
  • Mutation or loss of both copies means loss of control
250
Q

What features contribute to inherited cancer susceptibility?

A
  • Family history of related cancers
  • Unusually early age of onset
  • Bilateral tumours in paired organs
  • Synchronous or successive tumours
  • Tumours in different organ systems in the same individual
  • Mutation inherited through the germline
251
Q

What is retinoblastoma?

A
  • Malignant cancer of developing retinal cells
  • Sporadic disease that usually involved one eye
  • Hereditary cases can be unilateral, bilateral and multifocal
252
Q

What causes retinoblastoma?

A

Mutation of the RB1 tumour suppressor gene on chromosome 13q14
- RB1 encodes a nuclear protein that is involved in the regulation of the cell cycle

253
Q

What are the functional classes of tumour suppressor genes?

A
  • Regulate cell proliferation
  • Maintain cellular integrity
  • Regulate cell growth
  • Regulate the cell cycle
  • Nuclear transcription factors
  • DNA repair proteins
  • Cell adhesion molecule
  • Cell death regulators
  • They suppress the neoplastic phenotype
254
Q

What tumours are associated with the BRCA1 tumour suppressor gene?

A

Breast
Ovarian
Prostate

255
Q

What tumours are associated with the PTEN tumour suppressor gene?

A

Prostate

Glioblastoma

256
Q

What tumours are associated with the APC tumour suppressor gene?

A

Colon

257
Q

Why is a mutation in a single copy of the P53 tumour suppressor gene enough to get dysregulation?

A

Mutants act in a dominant manner

258
Q

What is Familial adenomatous polyposis coli?

A
  • Due to a deletion in 5q21 resulting in the loss of the APC tumour suppressor gene
  • Involved in cell adhesion and signalling
  • Sufferers develop multiple benign adenomatous polyps of the colon
  • 90% risk of developing colorectal carcinoma
259
Q

How does the APC tumour suppressor gene work?

A
  • Participates in the WNT signalling pathway

- APC protein helps control the activity of beta-catenin and thereby prevents uncontrolled growth

260
Q

What steps lead to the development of colorectal cancer

A
  • Damage to the APC
  • Hyperproliferation of the epithelium (loss of tumour suppressor function)
  • DNA hypomethylation is epigenetic modification of the epithelial cell. Combins with the action of a mutated RAS gene (oncogene) this will push the polyps to develop into an adenoma
  • Mutation of p53 will result in a carcinoma
  • Matastases
261
Q

What are the differences between oncogenes and tumour suppressor genes?

A
Oncogene:
- Gene active in a tumour 
- Specific translocation/point mutations 
- Mutations rarely hereditary 
- Dominant at cell level 
- Broad tissue specificity 
- Leukaemia and lymphoma
Tumour suppressor gene:
- Gene inactive in a tumour 
- Deletions or mutations 
- Mutations can be inherited 
- Recessive at cell level 
- Considerable tumour specificity 
- Solid tumours
262
Q

What steps are needed to make a blood vessel?

A
  • Vasculogenesis (bone marrow progenitor cell)
  • Angiogenesis (sprouting)
  • Arteriogenesis (collateral growth)
263
Q

What are the inhibitors of angiogenesis?

A
- Thrombospondin-1
The statins:
- Angiostatin 
- Endostatin 
- Canstatin 
- Turnstatin
264
Q

What are the activators of angiogenesis?

A
  • VEGFs
  • FGFs
  • PDGFB
  • EGF
  • LPA
265
Q

What is the initial stimulus for angiogenesis?

A

Hypoxia

  • It is sensed in cells by the transcription factor HIF (hypoxia-inducible factor) which leads to the expression of hypoxia-inducible genes
  • Encodes VEGF which is the signal to initiate blood vessel formation via endothelium activation
266
Q

What is pVHL?

A

von Hippel-Lindau tumour suppressor gene

- Controls levels of HIF

267
Q

What is vascular endothelial growth factor (VEGF) and its receptors?

A
  • Family of 5 members: VEGF-A, VEGF-B, VEGF-C, VEGF-D and placental growth factor (PIGF)
  • Three tyrosine kinase receptors: VEGF receptor (VEGFR)-1 VEGFR-2 and VEGFR-3: an co-receptors neuropilin Nrp1 and Nrp2
  • VEGFR-2 is the major mediator of VEGF-dependent angiogenesis, activating signalling pathways that regulate endothelial cell migration, survival, proliferation
268
Q

What happens in sprouting angiogenesis?

A

Specialised endothelial tip cells lead the outgrowth of blood-vessel sprouts towards gradients of VEGF

269
Q

What is the canonical notch signalling pathway?

A
  • Notch receptors and ligands are membrane-bound proteins that associate through their extracellular domains
  • The intracellular domain of Notch (NICD) translocates to the nucleus and binds to the transcription factor RBP-J
270
Q

What is involved in tip cell selection?

A

1) In stable blood vessel DII4 and Notch signalling maintain quiescence
2) VEGF activation increases expression of DII4
3) DII4 drives Notch signalling which inhibits expression of VEGFR2 in the adjacent cell
4) DII4-expressing tip cells acquire a motile invasive and sprouting phenotype
5) Adjacent cells (Stalk cells) form the base of the emerging sprout, proliferate to support sprout elongation

271
Q

How do macrophages participate in vessel anastomosis?

A
  • Macrophages have been shown to carve out tunnels in the extra cellular matrix thereby providing avenues for subsequent capillary infiltration
  • Tissue resident macrophages were shown to be associated with angiogenic tip cells during anastomosis
272
Q

What is the purpose of VE-cadherin?

A

Essential for vessel stabilisation and quiescence

  • Constitutively expressed at junctions
  • Homophilic interaction mediates adhesion between endothelial cells and intracellular signalling
  • Controls contact inhibition of cell growth
  • Promotes survival of EC
273
Q

What is the purpose of mural cells?

A
  • Help stabilise the neovessels
274
Q

What is involved in vessel stabilisation?

A

Pericytes

Mural cells

275
Q

How do signalling pathways control stability?

A
  • The Angiopoietin-Tie2 ligand-receptor system?
  • Ang-1 and Ang-2 are antagonistic ligands of the Tie2 receptor
  • Ang-1 binding to the Tie2 promotes vessel stability and inhibits inflammatory gene expression
  • Ang-2 antagonises Ang-1 signalling, promotes vascular instability and VEGF-dependent angiogenesis
276
Q

Describe tumour angiogenesis and neovasculature

A
  • Tumours less than 1mm3 receive oxygen and nutrients by diffusion from host vasculature
  • Larger tumours require new vessel network. Tumour secretes angiogenic factors that stimulate migration, proliferation and neovessel formation by endothelial cells in adjacent established vessels
  • Newly vascularised tumour no longer relies solely on diffusion from host vasculature which facilitates progressive growth
277
Q

What is the angiogenic switch?

A

A discrete step in tumour development that can occur at different stages in the tumour-progression pathway, depending on the nature of the tumour and its microenvironment

278
Q

What are the characteristics of tumour blood vessels?

A
  • Irregularly shapes, dilated, tortuous
  • Not organised into definitive venules, arteriole and capillaries
  • Leaky and hemorrhagic, partly due to overproduction of VEGF
  • Perivascular cells often become loosely associated
  • Some tumours may recruit endothelial progenitor cells from the bone marrow
279
Q

How does anti-angiogenic therapy normalise the vasculature?

A
  • Reduced hypoxia

- Increases efficacy of conventional therapies

280
Q

What are the problems with sustained/aggressive anti-angiogenic therapy?

A

May damage healthy vasculature leading to loss of vessels, creating vasculature resistant to further treatment and inadequate for deliver of oxygen/drugs

281
Q

What is Avastin

A

Monoclonal antibody which inhibits VEGF

282
Q

What are the side effects of Avastin therapy for cancer?

A
  • GI perforation
  • Hypertension
  • Proteinuria
  • Venous thrombosis
  • Haemorrhage
  • Would healing complications
283
Q

How does therapeutic angiogenesis help coronary artery disease and peripheral artery disease?

A

Promotes neo-vascularisation to prevent ischaemic damage

284
Q

What types of damage can occur to DNA as a result of carcinogens?

A
  • Base dimers and chemical cross links
  • Base hydroxylations and abasic sites formed
  • DNA adducts and alkylation
  • Double and single strand breaks
285
Q

What does mammalian metabolism consist of?

A

Phase I:
- Addition of functional groups e.g. oxidations, reductions, hydrolysis
- Mainly cytochrome P540-mediated
Phase II:
- Conjugation of Phase I functional groups e.g. sulfation, glucuronidation, acetylation, methylation, amino acid and glutathion conjugation
- Generates polar (water soluble) metabolites

286
Q

What are polycyclic aromatic hydrocarbons?

A
  • Common environmental pollutants
  • Formed from combustion of fossil fuels
  • Formed from combustion of tobacco
287
Q

What is aflatoxin B1?

A
  • Formed by Aspergillus flavus mould
  • Common on poorly stored grains and peanuts
  • Aflatoxin B1 is a potent human liver carcinogen, especially in Africa and Far-East
288
Q

How is 2-napthylamine metabolised?

A
  • CYP1A2 converts the amine to a hydroxylamine. This is toxic and will damage cells and DNA
  • A phase II enzyme detoxifies the substrate by adding a sugar molecule to it. However in the bladder, the new metabolite is labile in the acid pH of the urine. This generate s a positively charged amino group that is a highly DNA-reactive electrophile
  • DNA is particular electron-rich because of all the nitrogenous bases. The new metabolite reacts with the DNA, forming DNA adducts
289
Q

How does UV radiation act as a carcinogen?

A
  • High energy and attacks DNA
  • Cross links thymidine to form pyrimidine dimers
  • Skin cancer
290
Q

How does ionising radiation act as a carcinogen?

A
  • Generates free radicals in cells
  • Includes oxygen free radicles (super oxide radical, hydroxyl radical)
  • Possess unpaired electrons (electrophilic and therefore seek out electron-rich DNA)
  • Radicals attack DNA leading to double and single strand breaks often in apurinic and apyrimidinic sites
  • Radials cause base modifications such as ring-opened guanine and adenine, thymine and cytosine glycols and 8-hydroxyadenine & 8-hydroxyguanine (mutagenic)
291
Q

What are the different types of DNA repair?

A

1) Direct reversal of DNA damage
2) Base excision repair
3) Nucleotide excision repair
4) During-or post replication repair

292
Q

What happens during direct reversal of DNA damage?

A
  • Photolyase splits cyclobutane pyrimidine dimers

- Methyltransferases and alkyltransferases remove alkyl groups from bases

293
Q

What happens during base excision repair?

A
  • Mainly for apurinic/apyrimidinic damage
  • DNA glycoslyases and apurinic/apyrimidinic endonucleases and other enzyme partners
  • A repair polymerase fills the gap and DNA ligase completes the repair
294
Q

What happens during nucleotide excision repair?

A
  • Mainly for bulky DNA adducts
  • Xeroderma pigmentosum proteins assemble at the damage. A stretch of nucleotides either side of the damage are excised
  • Repair polymerases fill the gap and DNA ligase completes the repair
295
Q

What are the two pathways of excision repair?

A
  • Base excision repair pathway: the chain remains intact

- Nucleotide excision repair pathway: a stretch of DNA is removed therefore the chain does not remain in tact

296
Q

What is the Ames test?

A

A crude bacterial test looking at potential mutagenicity of chemicals, ie the ability of the chemical to cause DNA damage

297
Q

How can chromosomal aberrations be detected in mammalian cells?

A

Treat mammalian cells with chemical in presence of liver s9.

Look for chromosomal damage

298
Q

How is cytokinesis blocked in an in vitro micronucleus assay?

A

Treat with cytochalasin-B

299
Q

What is the function of the colon?

A
  • Extraction of water from faeces (electrolyte balance)
  • Faecal reservoir (evolutionary advantage)
  • Bacterial digestion for vitamins (e.g. B and K)
300
Q

What is the number of cells that die in the colon?

A

2-5 million die per minute

  • Proliferation renders cells vulnerable
  • APC mutations prevent cell loss
301
Q

What is a polyp?

A

Any projection from a mucosal surface into a hollow viscus and may be hyperplastic, neoplastic, inflammatory, hamartomatous

302
Q

What is an adenoma?

A

A benign neoplasm of the mucosal epithelial cells

303
Q

What are the different types of colonic polyps?

A
  • Metaplastic/hyperplastic (no malignant potential and 15% have k-ras mutation)
  • Adenomas
  • Juvenile
  • Peutz Jeghers
  • Lipomas
  • Others
304
Q

What are the different types of colonic adenomas?

A
  • Tubular (90%)
  • Tubulovillous (10%)
  • Villous
  • Flat
  • Serrated
305
Q

What is the microscopic structure of a tubular adenoma?

A
  • Columnar cells with nuclear enlargement, elongation, multilayering and loss of polarity
  • Increased proliferative activity
  • Reduced differentiation
  • Complexity/disorganisation of architecture
306
Q

What is the microscopic structure of villous adenomas?

A
  • Mucinous cells with nuclear enlargement, elongation, multilayering and loss of polarity
  • Exophytic, frond-like extensions
  • Rarely may have hypersecretory function and result in excess mucous discharge and hypokalaemia
307
Q

What is colonic dysplasia?

A

Abnormal growth of cells with some features of cancer. Requires subjective analysis, may be indefinite, low grade and high grade

308
Q

What is Adenomatous polyposis coli (APC/FAP)?

A

A disease caused by a 5q21 mutation.

  • Site of mutation determines clinical variants
  • Many patients will have a prophylactic colectomy
309
Q

From where do most colonic carcinomas arise?

A

From adenomas

  • There is a residual adenoma in 10 to 30% of carcinomas
  • Adenomas and carcinomas have similar distribution and adenomas usually precede cancer by 15 years
310
Q

What genetic pathways are involved in the progression from adenomas to carcinomas?

A
  • The adenoma carcinoma sequence: Involves APC, K-ras, p53 and telomerase activation
  • Microsatellite instability: Microsatellites are repeat sequences prone to misalignment. Some microsatellites are in coding sequences of genes which inhibit growth or apoptosis e.g. TGFbR11.
    Mismatch repair genes are are recessive genes requiring 2 hits
311
Q

What are the two main pathways of genetic predisposition to colonic carcinoma

A

FAP: inactivation of APC tumour suppressor genes
HNPCC: micro-satellite instability

312
Q

What is the most common mutation in colon cancer?

A

Inactivation of APC

When APC does not an inactivating mutation, beta catenin does

313
Q

What dietary factors contribute to colonic carcinoma?

A

High fat
Low fibre
High red meat
Refined carbohydrates

314
Q

How does food contribute to carcinoma?

A
  • Contains 5,000-10,000 bioactive chemicals
  • Food also contains carcinogens and anti-cancer agents
  • Heat modified chemicals further and bacteria can modify food residues
  • Heteocyclic amines (HCAs) are generated when meat is cooked at high temperatures and these may cause mutations
315
Q

Why are dietary deficiencies significant in colorectal cancer?

A
  • Folates are important, as a co-enzyme is needed for nucleotide synthesis and DNA methylation
  • MTHFR deficiency leads to a disruption in DNA synthesis causing DNA instability (strand breaks and uracil incorporation) leading to mutations.
    Decreased methionine synthesis leads to genomic hypomethylation and focal hypermethylation leading to gene activation and silencing
316
Q

What are the various anti-cancer food elements?

A
  • Vitamin C: ROS scavenger
  • Vitamin E: ROS scavenger
  • Isothiocyanates (cruciferous veg)
  • Polyphenols (green tea, fruit juice): activate MAPK- regulates Phase2 detoxifying enzymes as well as other genes and reduces DNA oxidation
317
Q

What is the clinical presentation of colorectal cancer?

A
  • Altered bowel habit
  • Rectal bleeding
  • Unexplained Fe deficiency- anaemia
  • Mucous
  • Bloating
  • Cramps ‘colic’
  • Constitutional weight loss, fatigue
318
Q

What are the macroscopic features of colonic carcinomas?

A
  • Small carcinomas may present with large polypoid adenomas, pedunculated or sessile
  • Caecum/ascending colon: 22%
  • Transverse colon- 11%
  • Descending colon- 6%
  • Rectosigmoid- 55%
319
Q

What is grading?

A

The proportion of gland differentiation relative to solid areas of nests and cord of cells without lumina

320
Q

What is the Dukes Classification?

A
Dukes A: 
- Growth limited to wall 
- Nodes negative 
Dukes B
- Growth beyond musc propria 
- Nodes negative 
Dukes C1: 
- Nodes postive 
- Apical LN negative 
Dukes C2: 
- Apical LN positive
321
Q

How do clinical features affect prognosis?

A
  • Diagnosis in asymptomatic patients: Improved prognosis
  • Rectal bleeding presenting as symptoms: Improved prognosis
  • Bowel obstruction/perforation: Diminished prognosis
  • Tumour location: Colon better than rectum, Left colon better than right colon
  • Age
322
Q

How do pathological features affect prognosis?

A
  • Depth of bowel wall penetration: Increased penetration diminished prognosis
  • Number of regional lymph nodes involved: 1-4 nodes better than >4 nodes
  • Degree of differentiation: Well >poorly differentiation
  • Mucinous or signet ring cell: Diminished prognosis
    Venous invasion:
  • Diminished prognosis
  • Lymphatic invasion: Diminished prognosis
    Perineural invasion: Diminished prognosis
    Local inflammation and immunological reaction: Improved prognosis
323
Q

What are the treatment options available for colorectal cancer?

A

Surgery, with concurrent treatment with 5FU, leucovorin, metastatectomy, chemotherapy and palliative care

324
Q

How are patients selected for screening for high risk colon cancer?

A
  • Previous adenoma
  • 1st degree relative affected by colorectal cancer before the age of 45
  • 2 affected first degree relatives
  • Evidence of dominant familial cancer trait including colorectal uterine and other cancers
  • UC and Crohn’s disease
  • Hereditable cancer families
325
Q

What is screening?

A

The practice of investigating apparently healthy individuals with the object of detecting unrecognised disease or people with an exceptionally high risk of developing disease, and of intervening in ways that will prevent the occurrence of the disease or improve the prognosis when it develops

326
Q

What is the criteria for a screening programme?

A

1) Importance of the disease- condition should be important in respect to the seriousness and/or frequency
2) The natural history of the disease must be known in order to
- identify where screening can take place
- enable the effects of any intervention to be assessed

327
Q

What are the characteristics of screening for colorectal cancer?

A
  • Simple and acceptable to the patients
  • Sensitive and selective
  • Screening population should have equal access to screening procedure
  • Cost effectiveness
328
Q

What is FOB?

A

Fecal Occult Blood Test

  • Most people ages 60-69 are screened ever 2 years and positive are referred for colonoscopy
  • It is relatively insensitive and produces some false positives
329
Q

What are the different types of skin cancer?

A

1) Keratinocyte derives
e. g. basal cell carcinoma, squamous cell carinoma aka non melanoma skin cancer (NMSC)
2) Melanocyte derived
e. g. malignant melanoma
3) Vasculature derived
e. g. Kaposi’s sarcoma, angiosarcoma
4) Lymphocyte derived
e. g. Mycosis fungoides

330
Q

What are the causes of skin cancer?

A

Accumulation of genetic mutations lead to uncontrolled cell proliferation
Genetic syndromes: Gorlin’s syndrome
Xeroderma pigmentosum
Viral infections: HHV8 in Kaposi’s sarcoma
HYPV in SCC
UV light: BCC, SCC, Malignant melanoma
Immunosuppression: drugs, HIV, old age, leukaemia

331
Q

Why is sunlight needed?

A
  • Essential for photosynthesis (plants)
  • Infrared spectra provide warmth
  • Effect on human mood
  • Stimulates the production of vitamin D in the skin
332
Q

What is the most important wavelength in skin carcinogens?

A

UVB (280-315)

333
Q

Which wavelength is a major cause of skin ageing?

A

UVA (315-400)

- Contributes to skin carcinogenesis

334
Q

What is the action of UVB

A
  • Directly induces abnormalities in DNA eg mutations
  • UVB induces photoproducts (mutations)
  • Affects pyrimidines ie Cytosine (C) and Thyme (T) bases
  • Usually repaired quickly by nucleotide excision repair
335
Q

How does UVA contribute to skin carcinogenesis?

A
  • DNA forming cyclobutane butane pyrimidine dimers but less efficiently than UVB
  • Free radicals which damage DNA and cell membrane
336
Q

What is the consequence of UV induced skin carcinogenesis?

A

UV damage to DNA leads to mutations in specific genes

  • Cell division
  • DNA reapir
  • Cell cycle arrest
337
Q

How is UV induced DNA damage repaired?

A
  • Photoproducts are removed by a process called nucleotide excision repair
  • Xeroderma pigmentosum: genetic condition with defective nucleotide excision repair
338
Q

What mutations cause cancer?

A

1) Mutations that stimulate uncontrolled cell proliferations e.g. abolishing control of the normal cell cycle (p53 gene)
2) Mutations that alter responses to growth stimulating/repressing factors
3) Mutations that inhibit programmed cell death (apoptosis)

339
Q

How is sunburn caused?

A
  • UV leads to keratinocyte cell apoptosis
  • Sun burn cells are apoptotic cells in UV overexposed skin
  • Apoptosis removes UV damaged cells in the skin which might otherwise become cancer cells
340
Q

What are the immunomodulatory effects of UV light?

A
  • UVA and UVB effect the expression of genes involved in skin immunity (Depletes Langerhans cells in the epidermis)
  • Reduced skin immunocompetence and immunosurveillance (Basis for UV phototherapy for e.g. psoriasis)
  • Further increases the cancer causing potential of sun exposure
341
Q

What are the Fitzpatrick phototypes?

A

Host response to UV is determined by genetic influences especially skin phototype

1) Always burns, never tans
2) Usually burns, sometimes tans
3) Sometimes burns, usually tans
4) Never burns, always tans
5) Moderate constitutive pigmentation- Asian
6) Marked constitutive pigmentation- Afrocarribean

342
Q

What is melanin?

A
  • Melanin pigmentation is responsible for skin colour
  • Produced by melanocytes within the basal layer of the epidermis
  • Skin colour depends on the amount and type of melanin produced, not the density of melanocytes (which is fairly constant)
  • Melanin dictates skin sensitivity to UV damage
343
Q

What two types of melanin are formed?

A

Eumelanin- brown or black
Phaeomelanin- yellowish or reddish brown
- Melanin is formed from tyrosine via a series of enzymes

344
Q

What is MCR1?

A

Melanocortin 1 receptor

  • Leading role in pigmentation following UV exposure, regulating changes in the skin and hair pigmentation phenotype
  • In response to UV, it promotes a switch from phaeomelanin to eumelanin and therefore has a key role in tanning
  • > 20 gene polymorphisms
345
Q

What is malignant melanoma?

A
  • Malignant tumour of melanocytes: melanocytes become abnormal, atypical cells and architecture
  • Caused by UV exposure and genetic factors
  • Risk of metastasis
346
Q

What is lentigo maligna?

A

(melanoma in situ)

  • Proliferation of malignant melanocytes within the epidermis
  • Risk of metastasis
  • Irregular shape
  • Light and dark brown colours
  • Size is usually >2 cm
347
Q

How does malignant melanoma superficially spread?

A
  • Lateral proliferation of malignant melanocytes
  • Invade basement membrane
  • Risk of metastasis
348
Q

How is superificial spreading of malignant diagnosed?

A

ABCD rule

  • Asymmetry
  • Border irregular
  • Colour variation (dark brown-black)
  • Diameter >0.7 mm and increasing
  • Erythema
349
Q

How does nodular malignant melanoma proliferate?

A
  • Vertical proliferation of malignant melanocytes
    (no previous horizontal growth)
  • Risk of metastasis
350
Q

How does nodular melanoma arise within a superficial spreading melanoma?

A
  • Downward proliferation of malignant melanocytes
  • Following previous horizontal growth
  • Nodule developing within irregular plaque- prognosis will become worse
351
Q

What are the different types of malignant melanoma?

A
  • Superficial spreading
  • Nodular
  • Lentigo maligna melanoma
  • Acral lentignous
  • Amelanotic
352
Q

How is prognosis of melanoma established?

A

Breslow thickness

- Measurement from granular layer to bottom of tumour

353
Q

What are the risk factors for the development of melanoma?

A
  • Genetic markers (e.g. CDKN2A mutations
  • Family history of dysplastic nevi or melanoma
  • Ultraviolet irradiation
  • Sunburns during childhood
  • Intermittent burning exposure in unacclimatised fair skin
  • Number (>50) and size (>5 mm) of melanocytic nevi
  • Congenital nevi
  • Number of atypical nevi (>5)
  • Atypical/dysplastic nevus syndrom
  • Personal history of melanoma
  • High socioeconomic status
  • Skin type I,II
  • Equatorial latitudes
  • DNA repair defects (e.g. xeroderma pigmentosum)
  • Immunosuppression
354
Q

What is squamous cell carcinoma?

A
  • Malignant tumour of keratinocytes
  • Caused by UV exposure, HPV, Immunosuppression, May occur in scars or scarring processes
  • Risk of metastasis
355
Q

What is basal cell carcinoma?

A
  • Malignant tumour arising from basal layer of epidermis
  • Caused by sun exposure, genetics
  • Slow growing
  • Invades tissue, but does not metastasise
  • Common on face
356
Q

What is epidermodysplasia veruciformis?

A

Rare autosome recessive condition

- Predisposition to HPV induced warts and SCCs

357
Q

Why is the incidence of breast cancer rising whereas the mortality is falling?

A
  • Early diagnosis
  • Chemo/radiotherapies
  • Hormonal therapies
358
Q

What is the cellular organisation of the mammary gland?

A

A layer of myoepithelial cells, some of which are slightly vacuolated, is just seen around the luminal cells, making contact with the basement membrane

359
Q

What are the major histological types of invasive breast cancer?

A
  • Infiltrating ductal carcinoma (IDC) many of which feature no special type of histological structure, account for almost 80% of breast cancers
  • Ductal carcinoma in situ (DCIS)
  • Infiltrating lobular carcinoma (ILC)
  • Tubular carcinoma
  • Medullary carcinoma
  • Mucinous carcinoma
  • Inflammatory breast cancer
360
Q

What is used in immunohistochemical staining?

A

Antibodies against the human oestrogen receptor

- About 80% of breast cancers are oestrogen-receptor positive

361
Q

How does oestrogen receptor act as a growth factor?

A
  • The receptor is activated upon oestrogen binding
  • Exists in the cytoplasm, bound to hsp90. Oestrogen crosses the cell membrane, and once bound to the nuclear/oestrogen receptor, displaces hsp90
  • Receptor dimerises, and translocates to the nucleus where it binds to DNA sequences calls oestrogen response elements
  • This increases the expression of oestrogen-induced gene products which increase cell proliferation, inhibit apoptosis and may result in breast cancer
362
Q

What is the role of the oestrogen receptor in breast cancer?

A
  • Some breast cancers like normal breast, are sensitive to the effects of oestrogen
  • One third of premenopausal women with advanced breast cancer will respond to oophorectomy
  • The receptor is over expressed in 50% of breast cancers. Presence is indicative of a better prognosis
363
Q

What are treatment approaches for breast cancer?

A
  • Surgery
  • Radiation therapy
  • Chemotherapy
  • Endocrine therapy
364
Q

How does endocrine therapy treat breast cancer?

A
  • Ovarian suppression
  • Blocking oestrogen production by enzymatic inhibition
  • Inhibiting oestrogen responses
365
Q

How are target tissue controlled hormonally?

A
  • The ovaries in premenopausal women make the majority of oestrogen, under the control of pituitary gonadotrophins, which is under the control of the LHRH from the hypothalamas
  • Aromatisation of androgens is also a source of peripheral oestrogen
366
Q

What is ovarian ablation?

A
  • The ovary is the major source of oestrogen biosynthesis in pre-menopausal women
  • Ovarian ablation aims to eliminate this source.
  • It can be carried out by surgical oophorectomy, ovarian irradiation
  • Major problems with this procedure are morbidity and irreversibility
367
Q

How can reversible and reliable medical ovarian ablation be achieved?

A
  • Using lutenising hormone releasing hormone agonists
  • LHRH agonists binds to LHRH receptors in the pituitary leading to receptor down-regulation and suppression of LH release and inhibition of ovarian function, including oestrogen production
368
Q

Give examples of LHRH agonists?

A

Goserelin
Buserelin
Leuprolide
Triptorelin

369
Q

What is Tamoxifen?

A

Anti-oestrogen used be inhibit oestrogen action

  • Most commonly used due to its demonstrated efficacy and low incidence side effects
  • Competitive inhibitor of oestradiol binding to the ER
  • Endocrine treatment of choice for metastatic disease in postmenopausal patients
  • Is a SERM- selective oestrogen receptor modulator
370
Q

How is Tamoxifen administered?

A

As a pro-drug: Tamoxifen Citrate which is metabolised in the GI tract to generate 4-hydroxymoxifen (active component) and endoxifen
- Tamoxifen citrate is metabolised in the gut to form the active drug

371
Q

How do anti-oestrogens negate the stimulatory effects of oestrogen?

A

Blocking the ER which causes the cell to be held at the G1 phase of the cell cycle

372
Q

What effects does tamoxifen have on bone?

A
  • Oestrogen is important to maintain bone in premenopausal women. After menopause, hormone replacement therapy is often recommended to prevent the development of osteoporosis. The long-term of an anti-oestrogen has the potential to precipitate premature osteoporosis
  • Tamoxifen has oestrogenic effect in bone
373
Q

What effect does tamoxifen have in the cardiovascular system?

A

Oestrogen lowers LDL cholesterol levels and raises HDL levels. Following menopause, women are at the same risk for coronary heart disease as me. Long term administration of an antioestrogen could produce a population at risk for premature coronary heart disease
- Tamoxifen has oestrogenic effects in the cardiovascular system

374
Q

What are the undesirable effects of tamoxifen?

A

Endometrial thickening, hyperplasia and fibroids following several years of therapy

375
Q

How does tamoxifen effect the breast, liver and heart and bone?

A

Breast= reduces breast cancer
Liver and heart= Lowers cholesterol, reduces atherosclerosis and heart attacks
Bone= maintains density to help prevent bone loss

376
Q

What are the negative effects of tamoxifen on the hypothalamus, eye, liver and uterus?

A
Hypothalamus= Increases vasomotor symptoms 
Eye= Increases cataracts 
Liver= Increases thromboembolism 
Uterus= Promotes endometrial cancer fibroids, polyps and vaginal discharge
377
Q

What additional drugs have been developed to target breast cancer?

A
  • Toremifene: structural derivative of tamoxifen
  • ICI 182 780: pure anti-oestrogen
  • Raloxifene: anti-tumour agent with no activity in breast or uterus but protective in bone therefore used for osteoporosis
378
Q

What problems are used in using Tamoxifen in prevention of breast cancer?

A
  • Increase incidence of endometrial cancer
  • Stroke
  • Deep vein thrombosis
  • Cataracts
379
Q

What does aromatase consist of?

A

A complex containing a cytochrome P450 heme containing protein and a NADPH cytochrome p450 reductase

  • The enzyme complex catalyses 3 separate steroid hydroxylation involved in the conversion of androstenedione to estrone
  • Aromatase can metabolise androsteinidione which is produced by the adrenal glands. This leads to the production of estrone sulphate which is circulated in the plasma
380
Q

How are aromatase inhibitors used in breast cancer?

A

In postmenopausal women, the major source of oestrogen derives not from the ovaries but from the conversion of the adrenal hormones androstenedione and to a lesser extent- testosterone to estrone

  • This enzymatic conversion occurs at extra-adrenal or peripheral sites such as fat, liver and muscle
  • This conversion is catalysed by the aromatase enzyme complex
381
Q

How are aromatase inhibitors classified?

A

1) Suicide inhibitors- destroy active site
- Initially compete with the natural substrate for active site but then on binding they form covalent bonds which results in irreversible inactivation of aromatase
2) Competitive inhibitors- bind reversible to the active site. These are more clinically useful as have fewer side effects

382
Q

Give an example of a suicide aromatase inhibitor

A

Exemestane
- Single dose administration reveals a major reduction in plasma oestrogen with only mild side effects reported, including hot flushes, nausea and fatigue

383
Q

Give an example of a competitive aromatase inhibitor

A

Anastrozole

384
Q

What is the role of progestins in breast cancer?

A
  • Progesterone- the dominant naturally occurring progestin
  • Progestins are used in the endocrine treatment of uterine and breast cancer with clinically proven antineoplastic properties
  • Progestin therapy for metastatic breast cancer has been used principally as a second or third-line therapy following selective oestrogen
  • The principle progestin used for metastatic breast cancer has been megestrol acetate
385
Q

What are the risk factors of breast cancer?

A
  • Early age of onset of menarche
  • Late age to menopause
  • Age at first full term pregnancy
  • Some forms of the contraceptive pill
  • Hormone replacement therapy
  • Obesity
  • Diet, physical activity, height, medication (aspirin)
386
Q

What does leukaemia result from?

A
  • Results from a series of mutations in a single lymphoid or myeloid stem cell
  • These mutations lead the progeny of that cell to show abnormalities in proliferation, differentiation or cell survival which leads to steady expansion of the leukaemic clone
387
Q

How does leukaemia differ from other cancers?

A
  • Most cancers exist as a solid tumour
  • It is uncommon for patients with leukaemia to have tumours
  • More often they have leukaemic cells replacing normal bone marrow cells and circulating freely in the blood stream
  • Leukaemia is different because haemopoietic and lymphoid cells behave differently from other body cells
  • Normal haemopoietic stem cells can circulate in the blood and both the stem cells and the cells derived from them can enter tissues
  • Normal lymphoid stem cells recirculate between tissues and blood
388
Q

How are leukaemias described?

A
  • Leukaemias that behave regularly ‘benign’ are called chronic- the disease goes on for a long time
  • Leukaemias that behave in a ‘malignant’ manner are called acute- meaning if they are not treated the disease is very aggressive and the patients dies quite rapidly
389
Q

How is leukaemia classified?

A
  • Depending on the cell of origin, it can be lymphoid or myeloid
  • Lymphoid can be B or T lineage
  • Myeloid can be any combination of granulocytic, monocytic, erythroid or megakaryocytic
  • Examples:
    Acute lymphoblastic leukaemia (ALL)
    Acute myeloid leukaemia (AML)
    Chronic lymphocytic leukaemia (CLL)
    Chronic myeloid leukaemia (CML
390
Q

Why do people get leukaemia?

A
  • Loss of function of a tumour-suppressor gene (deletion or mutation)
  • Tendency to increased chromosome breaks, likelihood of leukaemia increases
  • If the cell cannot repair DNA normally, an error may persist whereas in a normal person the defect would be repaired
391
Q

What leukaemogenic mutations result in leukaemia?

A
  • Mutation in a known proto-oncogene
  • Creation of a novel gene e.g. a chimaeric or fusion gene
  • Dysregulation of a gene when translocation brings it under the influence of the promoter or enhancer of another gene
392
Q

What inherited or constitutional abnormalities can contribute to leukaemogenesis?

A
  • Down’s syndrome
  • Chromosomal fragility syndromes
  • Defects in DNA repair
  • Inherited defects of tumour-suppressor genes
393
Q

What are the identifiable causes of leukaemogenic mutations?

A
  • Irradation
  • Anti-cancer drugs
  • Cigarette smoking
  • Chemicals: benzene
394
Q

What happens in Acute myeloid leukaemia?

A

1) Cells continue to proliferate but they no longer mature so there is
- A build of the most immature cells- myeloblasts (blast cells) in the bone marrow with spread into the blood
- A failure of production of normal functioning end cells- neutrophils, monocytes, erythrocytes, platelets
2) In AML the responsibly mutations usually affect transcription factors so the transcription of multiple genes in affected

395
Q

What happens in Chronic myeloid leukaemia?

A
  • The responsible mutations usually affect a gene encoding a protein in the signalling pathway between a cell surface receptor and the nucleus
  • The protein encoded may be either a membrane receptor or a cytoplasmic protein
  • Cell kinetics and function are not as seriously affected as in AML
  • The cell becomes independent of external signals, there are alterations in the interaction with the stroma and there is reduced apoptosis so that cells survive longer and the leukaemic clone expands progressively
396
Q

With regards to to the production of end cells, how does AML and CML differ?

A
AML= Failure of production of end cells 
CML= Increased production of end cells
397
Q

What is the difference between acute and chronic lymphoid leukaemias?

A
  • Acute lymphoblastic leukaemia has an increase in very immature cells (lymphobalasts) with a failure of these to develop into mature T and B cells
  • Chronic lymphoid leukaemias: The leukaemic cells are mature although are abnormal T or B cells
398
Q

What characteristics does accumulation of abnormal cells in leukaemia cause?

A
  • Leucocytosis
  • Bone pain (if leukaemia is acute)
  • Hepatomegaly
  • Splenomegaly
  • Lymphadenopathy (if lymphoid)
  • Thymic enlargement (if T lymphoid)
  • Skin infiltration
399
Q

What characteristics do the metabolic effects of leukaemic cell proliferation in leukaemia cause?

A
  • Hyperuricaemia
  • Renal failure
  • Weight loss
  • Low grade fever
  • Sweating
400
Q

What characteristics does the crowding out of normal cells in leukaemia cause?

A
  • Anaemia
  • Neutropenia
  • Thrombocytopenia
401
Q

What is a standard feature of chronic lymphoid leukaemia?

A

Loss of normal immune function as a result of loss of normal T cell and B cell function

402
Q

What is acute lymphoblastic leukaemia largely a disease of?

A

Children

  • Suggests that is may result from delayed exposure to a common pathogen
  • Family size, new towns, socio-economic class, early social interactions, variations between countries
403
Q

What are the clinical features of acute lymphoblastic leukaemia that result from the accumulation of abnormal cells?

A
  • Bone pain
  • Hepatomegaly
  • Splenomegaly
  • Lymphadenopathy
  • Thymic enlargment
  • Testicular enlargement
404
Q

What are the clinical features of acute lymphoblastic leukaemia that result from the crowding out of normal cells?

A
Caused by anaemia: 
- Fatigue
- Lethargy 
- Pallor
- Breathlessness 
Caused by neutropenia: 
- Fever and other features of infection 
Caused by thrombocytopenia:
- Bruising 
- Petechiae 
- Bleeding
405
Q

What are the haematological features of acute lymphoblastic leukaemia?

A
  • Leucocytosis with lymphoblasts in the blood
  • Anaemia (normocytic, normochromic)
  • Neutropenia
  • Thrombocytopenia
  • Replacement of normal bone marrow cells by lymphoblasts
406
Q

What are the investigations for acute lymphoblastic leukaemia?

A
  • Blood count and film
  • Check of liver and renal function and uric acid
  • Bone marrow aspirate
  • Cytogenetic/molecular analysis
  • Chest X-ray
407
Q

Why is cytogenic and genetic analysis useful in acute lymphoblastic leukaemia?

A
  • Cytogenic/molecular genetic analysis is useful for managing the individual patient as it gives more information about prognosis
  • It advances the knowledge of leukaemia as it has permitted the discovery of leukaemogenic mechanisms
  • Hyperdiploidy reveals a good prognosis
  • Recipricol translocation between chromosome 4 & 11 is a poor prognosis
408
Q

What are the leukamogenic mechanisms of acute lymphoblastic leukaemia?

A
  • Formation of a fusion gene (translocation)
  • Dysregulation of proto-oncogene by juxtaposition of it to the promoter of another gene e.g. T-cell receptor gene
  • Point mutation in a proto-oncogene
409
Q

How is acute lymphoblastic leukaemia treated?

A

1) Supportive
- Red cells
- Platelets
- Antibiotics
2) Systemic chemotherapy
3) Intrathecal chemotherapy