Multistage carcinoma Flashcards

1
Q

What kind of mutations can be seen in cancer?

A
  • TSG loss
  • oncogene activation
  • mutations that allow cells to grow faster than their neighbours offering them a selective advantage
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

10 hallmarks of cancer

A
  • evading growth suppressors
  • avoiding immune destruction
  • sustaining proliferative signalling
  • replicative immortality
  • tumour-promoting inflammation
  • activating invasion and metastasis
  • inducing/acessing vasculature
  • genome instability + mutations
  • deregulating cellular metabolism
  • resisting cell death
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What kinds of cells are in epithelial tumours?

A
  • cancer cells + stromal cells
  • blood vessels, ,cancer-associated fibroblasts, immune/inflammatory cells + factors
  • blood cancers can also have stromal cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What are 90% of tumours made from?

A

epithelial cells - these cells are more exposed to the environment and damage

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

What are the stages of the cell cycle?

A

Go, G1, S, G2, M

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

What regulates the cell cycle?

A
  • cyclins and CDKs
  • growth factors stimulate the cell to enter from quiescent G0 state to G1
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Which cyclin/CDK is important in inhibition of the G phase of the cell cycle?

A
  • cyclin D CDK 6/4
  • growth factor induced
  • upregulated by growth factors
  • can be overexpressed in cancers
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What is the raf-MEK-ERK pathway?

A
  • growth factors such as GFG, EGF are produced by stromal cells and activate tyrosine kinase receptors
  • these activate ras which phosphorylates raf which phosphorylates MEK1+2 which phosphorylates ERK1+2
  • ERK1+2 can then enter the nucleus and activate cell cycle and survival genes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What kind of mutations can occur in the raf-MEK-ERK pathway that lead to cancer?

A
  • tyrosine kinase receptor activation
  • ras activation (most common in K least in H)
  • b-raf activation - seen in 50% of melanomas
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

How are activating mutations of tyrosine kinase receptors treated in cancer?

A
  • small molecule inhibitors bind intracellularly and prevent target binding to the receptor
  • not selective but cheap
  • Ab inhibitors block receptors specifically but are expensive
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

How are activating mutations of ras treated in cancers?

A
  • difficult to target with small molecule inhibitors
  • cant use Abs as ras is inside the cell
  • recently some ras mutations such as B-raf in melanoma can be inhibied
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

PI3K pathway?

A
  • PI3K is made of 2 subunits
  • p85 interacts with receptors
  • p110 phosphorylates lipids in the membrane
  • growth factors bind receptors and activate PI3K which phosphorylates PIP2 into PIP3
  • PIP3’s negative charge helps it to recruit many proteins such as AKT involved in cell survival
    PTEN can turn PIP3 back to PIP2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What kinds of mutations can happen in the PI3K pathway to cause cancer? How can these be targeted?

A
  • p110 activation -> more Akt and cell survival - seeen in breast and coloreactl and kinase inhibitors can be used but with bad side effects at PI3K is important in many processes
  • Akt activation can be treated with kinase inhibitors
  • PTEN deletion is seen in 30% of tumours highest in prostate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What is angiogenesis?

A
  • budding of new blood vessels off of existing ones
  • endothelial cells respond to angiogenic signals and start to move and divide towards them
  • laminin and collagen degrades the basement membrane
  • seen in wound healing and cancer
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

How is angiogenesis induced?

A
  • as tumours grow or during wound healing, new cells have less access to oxygen (hypoxia) and express VEGF
  • this induces new blood vessels in loops to allow blood to flow through the new tissue
  • angiogenesis can also be activate by ras activating mutations that stimulate the production of VEGF even without hypoxia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Not all tumours will induce angiogenesis? WhY?

A
  • those growing in places with lots of blood vessels may not suffer from hypoxia
  • lung, liver brain etc
17
Q

Other than cancer cells, which other cells might produce VEGF in response to hypoxia?

A
  • stromal and immmune cells within a tumour
18
Q

How do cancer cells invade and metastasize?

A
  • tumour cells degrade the basement membrane
  • some cells become more invasive and can invade their surroundings when provided with new or existing blood vessels
  • cancer cells enter vessels by degrading their basement membrane
  • some may manage to attach to endothelial cells, leave the blood vessels and invade secondary sites
19
Q

Why is metastasis so rare?

A

epithelial cells aren’t designed to be in the blood so they can die or get broken down or struggle to attach to endothelial cells to leave the vessels at secondary sites

20
Q

What are some common sites of metastasis?

A
  • places with good blood supply such as the lung, liver, brain or bone
  • not the heart or other muscles as contractile force destroys the cells
  • some cancers have places they metastasise to more commonly such as prostate cancer going to the bone- there may be nutrients in the bone that support prostate cancer cells well
21
Q

What are the 3 epithelial cell-cell junctions? Which are most important in cancer invasion and metastasis?

A
  • tight junctions stop movement of fluids such as in the gut
  • gap junctions pass messages between cells
  • adherens junctions are the most important in metastasis and tell cells to stop moving and dividing due to neighbours or can allow cell growth following cell loss or altered signalling
22
Q

What is the structure of an adherens junction?

A

two membranes are held together by E-cadherin. E-cadherin binds Beta catenin binds alpha catenin binds actin
- parallel actin filaments stabilise the junctions and prevent migration

23
Q

What changes in adherens junctions can allow cell migration in cancer?

A
  • E-cadherin gene silenced or deleted
  • E cadherin degraded (done in wounds)
  • inherited mutations - some people have one inactive copy of e-cadherin that makes them more susceptible to breast cancer
  • alpha catenin also seen inactivated in cancers
24
Q

What can happen with HGF in cancer?-

A
  • cross-talk between cancer cells and fibroblasts can cause them to see cancer as a wound and produce HGF
  • high levels of HGF constantly lead to no junctions and lots of migration
24
Q

What is the role of HGF?

A
  • made in the stroma by cells such as fibroblasts during wound healing
  • binds met receptor on epithelial cells and induces loss of adherens junctions and cell migration
  • when HGF levels go back down, junctions reform and cells become quiescent again
24
Q

Other than increased HGF what other mutation can lead to migration of cancer cells?

A
  • met receptor constitutively active
  • induces loss of junctions even in the absence of HGF
  • seen in kidney cancers among others
25
Q

What happens downstream of met receptor activation?

A
  • met activates ras
  • signals through raf which activated PI3K-rac and MERK-ERK to cause loss of junctions and invasion
  • ERKS can activate transcription factor SAIL that binds the E-cadherin promoter and inactivates it
26
Q

How does cell migration occur after loss of E-caderin by ERKs?

A
  • HGF acts through PI3K to activate rac
  • rac activates the WAVE complex and Arp2/3
  • THESE PRODUCE ACTIN POLYMERISATION THAT ALLOWS FOR CELL MOVEMENT
27
Q

What is TGF beta

A
  • EMT transition promoter
  • serine threonine kinase produced by fibroblasts
  • when ligand binds, phosphory,lation of the Smad heterodimer makes it move to the nucleus and induce genes seen in development such as N-caherin
28
Q

What are some roles of TGF-beta and Smad in and out of cancer?

A
  • suppress proliferation of stem cells as they move up the colon crypts
  • act as tumour suppressor genes and SMAD IS often mutated in colon cancer
  • in other tumours, TGFB can act as a tumour promoter show that different tumours use different genes for different things and is not universal
29
Q

What do immune cells in the stroma do in cancer?

A
  • are able to produce a fast immune response
  • enter tumours as they produce factors seen in wounds and produce TGFB that allows tumour cells to invade
30
Q

In brief, how do caner cells evade immune destruction?

A
  • things such as PDL-1, CTLA-4 act on T cell receptors and suppress their proliferation
  • T and B cells in tumours start to become quiescent and no longer recognise cancer cells as foreign