molecular basis of cancer Flashcards

1
Q

name the 6 classic hallmarks of cancer

A

1) Sustaining proliferative signalling – don’t need as much nutrients and growth factors
2) Evading growth suppressors
3) Activating invasion and metastasis
4) Enabling replicative immortality
5) Inducing angiogenesis – development of new blood vessels (recruit other cells)
6) Resisting cell death

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

name the 2 emerging hallmarks of cancer and the 2 enabling characteristics of cancer

A

HALLMARKS
1) Avoiding immune destruction
2) Tumour-promoting inflammation – macrophages infiltrate, make them produce factors to promote growth instead of hindering it
ENABLING
1) Genome instability and mutation – thing that causes cancer
2) Deregulating cellular energetics – cancer cells can change the way they metabolise compared to normal cells – use glycolysis even when oxygen is present undergo ‘metabolic switch’

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

how do cells sustain proliferative signalling?

A

3 main ways:
modulation of growth factor (GF) provision
modulation of GF receptor activity
modulation of intracellular signalling pathways

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

how do cancer cells modulate growth factor (GF) provision?

A

they can:

1) stimulate normal cells in the microenvironment (stroma) to provide cells with GFs – eg macrophages releasing lots of GFs in tumour environment
2) produce their own mitogenic GFs and respond to them through autocrine mechanisms (self-sufficient process)
3) make other cells produce mitogenic GFs through paracrine mechanisms

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

how does GF signalling work?

A

GF binds receptor tyrosine kinase –> dimerisation –> autophosphorylation. Some proteins dock with active receptor and become activated by phosphorylation.
Some proteins act as substrates for receptor kinase and become tyrosine phosphorylated and phosphorylate other proteins (mitogen activated protein kinase pathway)
end result = final protein that phosphorylates and activates a TF = signal transduction in cell

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

how do cancer cells modulate GF receptor activity?

A

many GF receptors are proteins TKs. Overexpression of these allows tumours to respond to low levels of GF that wouldnt usually produce a response

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

name the effects of overexpression of GF receptors (protein TKs) and give examples

A

overexpression may lead to:
ligand independent signalling
structurally altered receptors (eg truncated EGF receptor is constitutively active)

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

give 2 examples of ligand-independent signalling

A

EGF-R is up-regulated in stomach, breast and brain tumours and HER2 is over expressed in breast cancer

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

give 2 examples of truncated proteins that have resulted in the modulation of GF receptor activity and explain how this has happened

A

EGF receptor mutated (with deletion of the EGF receptor part) forms ErbB oncoprotein - no receptor = ligand independent = dimerisation = constitutively active TK
Her2 gene point mutation leads to change in membrane embedded region (Val to Gln) forms Neu oncoprotein => dimerizaton and autophosphorylation (ligand independent) – antibody (herceptin) that stops EGF from binding Her2 no longer works in patient

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

which pathway is frequently modulated to increase the amount of GF that cancer cells can bind?

A

SOS-Ras-Raf-MAPK pathway

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

how is Ras normally activated?

A

Ras binds GTP (changing Ras’s conformation) allowing it to bind and activate Raf. Raf then dissociates (by GTP hydrolysis to GDP), is phosphorylated, activated and continues transducing.
Ras is still bound to GDP. SOS comes along and removes GDP. GTP now binds to Ras, reactivating it.

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

name the mutation in Ras that causes it to become hyperactive. explain why this happens and what the result of this is.

A

point mutation in protein resulting in changing Gly12 to any other a/a.
Locks Ras in hyperactive, permanently on state. GTP can’t hydrolyse to GDP, Ras keeps binding to Raf repetitively. This means pathway is constantly on, leading to uncontrolled growth.

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

what are the effects of mutations in PI3 Kinase and Ras?

A

PI3K can frequently mutate in many cancers, causing it to become more active. This means cells develop survival signals that protect against apoptosis.

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

name 2 effects of growth supressors

A

driving cells out of the cell cycle

inducing entry into a post-mitotic differentiated state

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

what are inhibitory signals and how do they work?

A

similar to GFs theyre generated by signals binding to surface receptors
purpose is to drive cells out of the cell cycle (eg G0) or differentiated state.

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

give an example of an inhibitory signal and its mode of action.

A

TGF-beta binds receptors and stimulates SMADs (proteins) which activate p15, p27 and p21. These proteins inhibit the cell cycle.

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

give 2 ways in which TGF-beta action is blocked

A

down regulation of TGF-beta receptor

mutation of TGF-beta receptor to inactive or less active form

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

describe the 2-hit hypothesis and give an example of it

A

hypothesis: cancers usually require more than 2 mutations to form
eg Rb mutation in one gene and develop mutation in other gene = more likely to develop retinoblastoma cancer

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

what is Rb? what are the effects of the loss of function in Rb?

A

Rb is the gatekeeper of the cell cycle, it decides which cells should(n’t) proceed through the cell cycle.
Loss of function = persistent cell proliferation.

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

with respect to E2F, how does Rb act as a tumour suppressor ?

A

Rb binds E2F (a TF) and stops it from transcribing DNA. The inactivation of E2F halts cell cycle.

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

how does EGF stop Rb from acting as a tumour suppressor?

A

EGF binds either EGF-R or HER2, stimulating the Ras-MAPK pathway. This results in CDK binding cyclin which then phosphorylates Rb. Rb can now not bind to E2F and there is transcription.

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

describe in detail the stages of the cell cycle after stimulation by a GF (include checkpoints).

A

GF stimulates CDK1 & CDK2 and cyclins D1, D2, D3 which push the cell into G1. Gets past checkpoint by phosphorylating Rb which dissociates from E2F = gene transcription.
Because there is gene transcription there is more CDK2/cyclin E which allows the cell to enter S phase. This produces CDK2 and cyclin A.
Second checkpoint where p53 checks for errors in DNA replication, if there is none then cell enters G2 and M phase where CDK1 and cyclin B are present => cell cycle can continue.
Antisignals TGFβ via SMADs upregulate p21 and p27 which bind CDK2/4-cyclin D (inhibits them), therefore can’t phosphorylate Rb.
Cell must avoid use of antisignals which it does by automatically altering/removing Rb (remember Rb plus E2F1 = no transc)

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

how do DNA viruses induce tumours?

A

Rb is inactivated by being complexed with a viral protein. In human cervical tumours this is the E7 protein produced by HPV or the E6 protein which binds p53 and promotes its destruction.

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

do you tumours under or over produce c-myc?

A

overproduce

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

describe the steps of apoptosis

A

trigger
chromatin condenses and cytoplasm shrinks
blebbing
nucleus fragments by caspases, DNA ladderings, cell fragmentation
phagocytosis by macrophages

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

name 4 possible external triggers of apoptosis

A

not enough GFs
hypoxia
activation of death receptors (eg by p53)
loss of adhesion (to other cells)

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

name 3 modulators of apoptosis

A

Bcl-2 family
p53
Mdm2

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

what are the effectors of apoptosis?

A

caspases which act on substrates and activate a proteolytic cascade

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

what the substrates for apoptosis? (ie which parts of the cell are broken down?)

A

many cellular proteins

DNA

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

what is the difference in the extrinsic and intrinsic pathways of apoptosis initiation?

A
extrinsic pathway uses fas ligand and receptor and tumour necrosis factor (TNF) and TNF receptor
intrinsic pathway uses
intracellular signals (p53, Bcl2, Bax)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

describe the extrinsic caspase pathway

A

death ligand (fas/TNF) –> death receptors –> caspases (8 for extrinsic pathway which triggers 3,6,7 which cleave cell and DNA and result in apoptosis) –> cell death

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

describe the intrinsic caspase pathway

A

Apoptotic signal ER stress and DNA damage and cell cycle stress –> activate BAK & BAX –> downregulates Bcl2 –> BAK and BAX makes holes I mitochondria –> cytochrome C leaks out –> Apaf-1 with cytochrome C activate caspase 9 by apoptosome formation –>activates caspases 3,6,7 –> cell death

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

why are p53, MDM2 and Bax unregulated in cancer and Bcl2 upregulated?

A

p53, MDM2, Bax normally stimulate apoptosis – downregulated/mutated
Bcl2 normally inhibits apoptosis – upregulated

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

how does Bcl2 (the protein) inhibit apoptosis?

A

Bcl2 inhibits apoptosis by binding and inactivating pro-apoptotic proteins BAX and BAK, in the mitochondrial membrane.
BAX and BAK (pro-apoptotic) cause release of cytochrome c which activates caspases leading to apoptosis

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

what is the effect of the joint overexpression of c-myc and Bcl2? Give some experimental evidence for this

A

Fibroblasts overexpressing c-myc were grown in culture
In low serum (introduces element of stress) the c-myc expressing cells show high proliferation but also high apoptosis
Increased apoptosis could be abolished by overexpression of Bcl2

36
Q

what happens at the G1 checkpoint with p53?

A

Following DNA damage p53 stops cell cycle
p53 stimulated p21
p21 binds cdk cyclin
Cell cycle arrest
Allows time for DNA repair
If damage too extensive/irreparable p53 triggers apoptosis via Bax

37
Q

what happens at the G2 checkpoint with p53?

A

Chromosomes not properly replicated p53 activates Bax expression and apoptosis

38
Q

how is replicative immortality enabled?

A

normal cells proliferate until a certain number of doublings, however p53- and Rb- (knockout genes) keep dividing (and divide quicker) until enters a second state called ‘crisis’ – massive cell death (1 in 10^7 cells survives and can continue to grow) – now divide without a limit – a state of immortalization

39
Q

define a telomere

A

Simple sequence DNA repeats at end of chromosomes

40
Q

describe human telomeres

A

Human telomeres: 250-1500 copies (6-12kb) of the sequence TTAGGG

41
Q

what is telomerase?

A

Specialised DNA pol that adds telomere repeats

42
Q

why is telomerase upregulated in cancer cells?

A

The telomeres are therefore kept at a length above a critical threshold – allows for unlimited cell multiplication

43
Q

define angiogenesis

A

sprouting of new vessels from the pre-existing ones within the normal tissues

44
Q

how do tumours stimulate angiogenesis?

A

tumours produce GFs, the concentration gradient of which blood vessels follow, leading to the tumour.

45
Q

tumours alter their microenvironment as they grow. name 3 of these changes that would stimulate angiogenesis

A

low pH
low concentration of nutrients
decrease in oxygen tension (hypoxia).

46
Q

which 2 oncogenes are involved in triggering the angiogenic switch? which GF is upregulated from this?

A

RAS and myc upregulate the GF Vascular Endothelial GF (VEGF)

47
Q

describe tumour-induced sprouting angiogenesis

A

angiogenic factors are released
Activation of endothelial cells – uses receptor TKs
Endothelial cells produce matrix degrading proteins = degradation of basement membrane
Release of cell-matrix interactions
Invasion and migration of endothelial cells through basement membrane – tip cells migrate to tumour
Proliferation of endothelial cells
Lumen formation and capillary formation
Recruitment of pericytes/blood vessel stabilisation/maturation

48
Q

how does hypoxia result in the upregulation of VEGF (vascular endothelial GF)?

A

Low oxygen tension happens as tumours grow faster than their surroundings
Results in translocation of HIF-1α and HIF-1β to the nucleus where they bind together and act as a TF for the hypoxia response elements in their promoters – eg upregulating VEGF

49
Q

how does VEGF (vascular endothelial GF) activate the endothelial cells in angiogenesis?

A

VEGF acts as a ligand, binding to a receptor TK (VEGFR2) on the surface of endothelial cells
Receptor dimerises, autophosphorylates and triggers a signalling pathway via Grb2
Grb2 binds SOS – activates RAS and the MAPK signalling pathway
MAPK = upregulation of:
Matrix breakdown/proliferation/migration/permeability/tube formation

50
Q

in order for endothelial cells to migrate cell-cell and cell-matrix contacts must be broken. Which proteins are involved in this?

A

MMPs (Matrix metalloproteinases) and TIMPs (tissue inhibitors of metalloproteinases). The balance of these 2 control it. In cancer MMPs are overexpressed and released

51
Q

what are integrins?

A

transmembrane glycoprotein heterodimers

52
Q

which combinations of α and β subunits in integrin bind fibronectin/fibrinogen and laminin/collagen? what is the significance of this?

A

α5β1 binds fibronectin (more temporary) and fibrinogen
α2β1 binds laminin (present in more stable cells) and collagen
significance: α2β1 switches to α5β1 during angiogenesis

53
Q

what happens to integrins when GFs bind to endothelial cells?

A

Down regulation of integrins for basement membrane

Upregulation of integrins for extracellular matrix

54
Q

describe how endothelial cells migrate and invade the basement membrane then form form a capillary and lumen. how is this structure stabilised?

A

MMPs degrade the basement membrane allowing cells to invade through it. Endothelial cells migrate along VEGF conc grad. Then endothelial cells enter the cell cycle so new vessels can be formed. The endothelial cells then differentiate and the cell vacuoles join to form a lumen. Endothelial cells then lay down a basement membrane, followed by pericytes (contractile cells) and smooth muscle.

55
Q

how do tumour vessels differ from normal vessels?

A

increased number of vessels
More proliferating endothelial cells
Decreased endothelial cell-cell adhesion
Leaky vessels
Decreased vessel stability: decreased association of mural (pericytes & smooth muscle) cells with endothelial cells
Loss of close association of basement membrane with endothelial cells

56
Q

why is tumour angiogenesis important?

A

angiogenesis results in more O2 and nutrients reaching the tumour. The leaky vessels means more cancer cells can escape.

57
Q

define metastasis

A

the process by which a tumour cell leaves the primary tumour to create a secondary tumour.

58
Q

how do cancer cells travel around the body?

A

by the blood (and the lymph and by local invasion but we don’t learn about these 2)

59
Q

describe the steps in the invasion-metastasis cascade

A

loss of cellular adhesion
increase in motility and invasiveness
intravasation into blood and lymphatic vessels
transit and survival through circulartion
extravasion into new tissue
formation of small tumour nodules (micrometastases)
growth into macroscopic tumours

60
Q

what is e-cadherin

A

cell-cell adhesion molecule that helps to assemble epithelial cells into sheets and to maintain quiescence

61
Q

is e-cadherin downregulated or upregulated in cancers? why is this?

A

frequently downregulated because increased expression of e-cadherin antagonises invasiveness

62
Q

what is the epithelial-mesenchymal transition?

A

loss of the epithelial phenotype and gain of the mesenchymal phenotype

63
Q

name some features of the epithelial-mesenchymal transition. how is this regulatory process controlled?

A

increased migration, invasion and resistance to apoptosis. Associated with loss of e-cadherin and upregulation of matrix degrading enzymes.
carcinoma cells become invasive
regulated by TFs (snail, slug, twist)

64
Q

why don’t all malignant cells metastasise?

A
Cells may invade and circulate 
Cells may be killed en route (sheer stress/immune response)
New site of growth not suitable:
incorrect receptors
metabolic factors
Failure of angiogenesis
Inefficient process
65
Q

what are the characteristics of epithelial cells?

A

‘cobblestone’ cells
non-motile
non-invasive

66
Q

what are the characteristics of mesenchymal cells?

A

elongated
motile
invasive

67
Q

describe the Warburg Effect

A

normal cells favour glycolysis with no O2 present and oxidative phosphorylation when O2 is present. Cancer cells favour aerobic glycolysis when O2 present and not present. This produces lactate from pyruvate.

68
Q

which oncogenes are associated with glycolysis? how do tumours compensate using the anaerobic glycolytic pathway?

A

Ras and myc involved in glycolysis in tumours.
cancer cells compensate the overuse of glycolysis by up-regulating glucose transporters (GLUT1). hypoxia also favours increased expression of glucose transporters.

69
Q

why do cancer cells favour glycolysis?

A

diversion of glycolytic intermediates into bio-synthetic pathways generating nucleosides and a/a facilitates macromolecular biosynthesis needed for proliferation
eg glutamine used for C & N sources (for tumour growth)
Similar metabolic changes are found in rapidly dividing embryonic tissues (suggests role in active cell proliferation)

70
Q

how does the immune system eliminate the majority of cancer cells?

A

Tumours expressing mutant proteins eg receptors recognised
Tumours expressing mutant antibodies
Triggers cytotoxic T cells and natural killer cells

71
Q

what is the impact of having tumours heavily infiltrated with T-killer cells and natural killer cells?

A

the more heavily infiltrated a patient is with TKCs and NKs the better the prognosis.

72
Q

what can happen when a patient undergoes organ donation and acquires an organ with cancer?

A

because the patient is immunosuppressed (for them to not reject the organ) the cancer in the donated organ can spread into the patient.

73
Q

what is PDL1?

A

programmed death ligand - maintains immune homeostasis

74
Q

how is PDL1 (programmed death ligand) used in cancer?

A

PDL1 can help cancer cells evade immune surveillance

75
Q

what is the impact of genome instability in cancer?

A

increased tendancy of the genome to acquire mutations when various processes involved in maintaining and replicating the genome are dysfunctional => cancers

76
Q

what are BRCA1 and 2?

A

Tumour suppressor genes that play a role in cellular DNA repair

77
Q

what is the impact of a mutation in either BRCA1 or BRCA2?

A

increase in the risk of breast cancer 6-14x

78
Q

what is the effect of the immune inflammatory response in tumour cells?

A

can either:
enhance tumourigenesis and progression
OR
eradicate tumour cells

79
Q

how do inflammatory cells help tumours?

A

supply GFs to sustain proliferative signalling, survival, angiogenesis, invasion, metastasis and EMT
release of reactive oxygen species that are mutagenic

80
Q

name the 6 types of cancer therapy and briefly describe them

A

Surgery
Radiotherapy – causes double strand breaks in DNA
Cytotoxic chemotherapy – highly toxic for rapidly dividing cells
Endocrine therapy – for hormone dependent tumours
Immunotherapy
Biological (targeted) therapy

81
Q

what is neoadjuvant therapy?

A

chemotherapy before surgery to enable surgery eg decreasing tumour size

82
Q

what is adjuvant therapy?

A

chemotherapy to eradicate any residual microscopic cancer by treating patients after their primary surgery

83
Q

how do cytotoxic drugs work?

A

they inhibit proliferation and the cell cycle by interfering with nucleotide synthesis and DNA replication or mitosis.
induce cell death by apoptosis

84
Q

which molecules regulate the cell cycle?

A

cyclin-dependent kinases bound to cyclins

85
Q

name the G1/S checkpoint

A

phosphorylation of Rb is gradual until the cell reaches the correct size. then Rb can’t bind E2F (TF) and the correct genes are transcribed - continuing the cell cycle. mutation in Rb means its always phosphorylated and the cell keeps growing

86
Q

which CDKs are used when in the cell cycle?

A

CDK2 for all stages apart from M (CDK1) and there is CDK4 in G1

87
Q

which cyclins are used when in the cell cycle?

A

cyclin a - S
cyclin b - G2/M
cyclins D1,2,3 - early G1
cyclin E - late G1