Cancer Flashcards

1
Q

what is our central concept of cancer

A

it is a disease caused by alteration of a cell’s genes

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

why do we say gene changes instead of just mutations

A

these may be any of the following

  1. Mutations, in the most general sense: any kind of alteration of DNA sequence
  2. Epigenetic change, such as aberrant DNA methylation or histone modification
  3. Tumour viruses bringing extra genes into the cell
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

How many critical gene changes are needed for adult cancer

A

> 10

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

what is metastasis

A

formation of new colonies of tumour in other parts of the body, by the of seeding cells into the circulation

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

what is the primary tumour

A

original tumour is called the primary tumour and a metastasis may be called a secondary.

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

what are malignant tumours

A

those capable of
metastasis.

A tumour doesn’t have to have formed any metastases to be
malignant: the actual formation of
metastases is very slow and inefficient
and may not actually have happened yet

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

what are benign tumours

A

Incapable of metastasis (unless subsequent mutation may turn the benign tumour into a malignant one).

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

where do the following cancers often metastasize to:
breast
colorectal

A

breast to lymph nodes and bone

colorectal to liver via HPV

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

why is metastasis important clinically and intellectually

A

most deaths from cancer are caused by metastases;

important intellectually because they are the key mechanisms of pathogenesis
and we don’t understand them.

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

what does a benign tumour look like down a microscope

A

confined to their original site in the body;

they have clearly defined boundaries, and can often be physically separated from surrounding tissue;

often surrounded by a capsule of connective tissue, which can be peeled away

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

what do malignant tumours look like down the microscope

A

ragged edges, infiltrating into
surrounding tissue, and growing there.

Malignant tumours often show other morphological differences, eg abnormalities of nuclear size and shape, and alterations or loss of differentiation;
but ‘invasion’ is the most important.

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

what is the classical idea of development of a colorectal tumour

A

Often a polyp/adenoma precedes the malignant tumour, and the polyp may progress through various degrees of abnormality. Polyps may well be preceded by less visible abnormalities.

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

True or false

Malignant tumours often develop via visible benign precursors

A

true but not all benign can become malignant

eg benign smooth muscle tumour of the uterus rarely if ever turns malignant; the rare invasive tumours in this tissue appear to develop via a different set of mutations.

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

can benign tumours kill

A

yes

for example meningiomas growing in the brain, or hormone-producing benign tumours of the pituitary or adrenal.

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

what are the broad rules for naming a benign tumour

eg

A

tissue name + - oma

e.g.
lipoma = benign fat tumour
Leiomyoma = benign smooth muscle tumour
Papilloma = wart

adenoma [of the colon] = benign glandular lump, including glandular polyp

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

nomenclature of malignant tumours from mesenchyme

A

[name of tissue] sarcomas
E.g.
osteosarcoma (malignant bone tumour),
leiomyosarcoma (malignant smooth muscle tumour)

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

nomenclature for malignant tumours of mesenchyme

A

[name of tissue] carcinoma

e.g. breast carcinoma, colorectal carcinoma

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

give some exceptions to the naming rules of cancer

A

malignant melanoma (‘melanoma’ is not used for benign moles, which are called
nevi (singular nevus))
Neuroblastoma, glioblastoma (malignant neural tumours)

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

how do you name cancers of the haemopoietic system

A

Leukaemias liquid haemopoietic neoplasms

lymphoma solid haemopoietic (usually lymphocytic) neoplasms

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

give examples of how cancer can interfere with normal function (4)

A

Pressure: enlarged prostate obstructing ureter, meningioma on brain

Erosion/destruction e.g. of bone -> fractures and pain

Epithelial ulceration: bleeding from colorectal tumours (picture) -> anaemia

Competition with normal: failure of normal bone marrow in leukaemia

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

what are some of the metabolic effects of cancer

A

general, systemic wasting – ‘cachexia’
specific – tumour specific products, e.g. peptide hormones ACTH, ADH secreted by small cell lung
cancer

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

what causes cachexia

A

Its mechanism, other

than competition for metabolic resources, is not known

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

how do you generally die from liver cancer

A
  • Liver overwhelmed by metastatic colon cancer -> liver failure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

how does leukaemia usually kill you

A

Failure of normal bone marrow -> infection through lack of neutrophils; or haemorrhage through lack
of platelets

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

how can pain from cancer kill you

A

Pain management -> analgesia -> respiratory depression

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

how common are benign tumours and malignant tumours of mesenchyme

A

Benign tumours:
very common in all tissues
e.g. Leiomyoma of uterus, lipoma, wart, mole …

Malignant tumours from mesenchyme:
generally rare but often rapidly lethal, e.g. osteosarcoma (malignant bone tumour)

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

how common are malignant cancers of epithelium

A

Common – the most important cancers

eg breast, colorectal, lung, prostate

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

which cancers have low incidence in the west but high elsewhere

A

Nasopharyngeal (in Chinese populations), Liver

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

how do cancers present

A

Tumour may be visible/palpable:
e.g. melanoma, breast carcinoma
but often indirect, through effect of tumour:
prostate: blockage of ureter
colorectal: obstruction of bowel, anaemia due to bleeding

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

how is colorectal cancer usually first noticed

A

patient is anaemic due to chronic bleeding of the ulcerated tumour, or because s/he is losing weight, or has altered bowel habit due to partial obstruction of the bowel

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

how is breast cancer usually spotted

A

unusual in that the tumour is often detected directly as a lump in the breast, but occasionally the first indication is bone pain or pathological fracture caused by metastatic tumour growing in bone.

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

what is the object, ideal, and draw backs of screening for cancer

A

Object: Find cancer earlier
Ideal: Find before malignant
But: have to be able to do something useful

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

what is the ideal screen for cancer

A

find benign precursors of cancer and remove them even before they turn malignant

eg cervical cancer where benign precursor lesions can be detected by the cervical smear test—cells from the cervix are sampled by brushing—and removed. The fall in cervical cancer in countries that screen strongly suggests that it is very successful

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

How is screening for colorectal cancer usually done

what is a draw back

how could this be improved

A

by detecting blood in stool collected at home—
reasonably convenient but
only able to detect tumours that have
already ulcerated.

Endoscopy for polyps would be
better, detecting the benign precursor, but is much more expensive and much more demanding on the patient.

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

How can breast cancer be detected by test

A

by X-ray, ‘mammography’, slightly earlier than it would be detected by palpation.

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

why is breast cancer screening controversial

A

expensive and demanding for patients, especially as many patients who do not have tumours are recalled for further investigation, and some cancers are discovered and treated unnecessarily

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

why do we not screen for prostate cancer

A

currently we could screen for malignant prostate cancers but screening is considered counterproductive, because the tumours are often so indolent that they may never cause a problem; and surgery has a very high morbidity—incontinence and/or impotence.

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

what are the stages of the cell cycle

A

G0+G1 (diploid state),
S (DNA synthesis),
G2 (tetraploid state, tidying up at the end of S phase and preparing for M),
M (mitosis).

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

How does Rb control the cell cycle

A

e by inhibiting the

initiation of DNA replication, i.e. holding the cycle at the G1/S checkpoint.

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

How is the cell cycle inhibition by Rb relieved

A

when CDK4-Cyclin D1 phosphorylates Rb1

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

What inhibits CDK4-cyclin D

A

p16 (aka CDKN2A) or p21 (aka CDKN1A)

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

what has to happen to Rb to promote cancer

how does this compare to CDK4-cyclin D

A

has to be inactivated, for example by deletion of all or part of the RB1 gene

overactivity mutations of CDK4 or CyclinD1 can keep Rb-1 phosphorylated.

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

which cyclin complex is often overactive in breast cancer

A

CCND1/CyclinD1 gene as a result of gene amplification

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

what has to happen p16/INK4A to cause cancer

A

inactivation (as p16 inhibits CDK4 cyclin D1)

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

define oncogene and tumour suppressor gene

A

Oncogene mutations are overactivity mutations - requires 1 mutation
Tumour Suppressor Gene mutations are loss of function mutations - requires 2 mutations

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

what is an intermediate case of oncogene vs tumour suppressor

A

some tumour suppressor gene
mutations, have a significant effect on the cell when only one copy of the gene is mutated, but losing both copies has a stronger effect. These are usually cases where the mutant protein complexes with the normal protein (eg p53)
It functions as a
tetramer, so mutating half the copies of the gene means that most tetramers are faulty

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

what is clonal expansion

A

A normal cell acquires a mutation/gene change that means that, over time, its progeny compete with neighbouring cells so that they take over more than their normal share of a tissue

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

why is there heterogeneity to a tumour

A

may contain not just the latest clone but also preceding clones and dead-end branches of the evolutionary tree

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

how did Jones et al demonstrate clonal expansion

A

They put a mutation and a
fluorescent marker into a few cells in mouse oesophageal epithelium in vivo; over a year the mutant cells’ progeny took over almost the entire epithelium.

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

what is the evidence for tumours being clones

A

all cells have the same gene changes, except for the most recent ones. Precursor clones, having some of the mutations seen in a tumour, can sometimes be detected in flanking tissue that is superficially normal.

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

How do we know that most cancers have specific defects that make them genetically unstable, and it’s not just that they are cycling so fast they mess up their DNA?

A

individual cases of cancer show different kinds of genetic instability

eg in colon cancers where there are at least two obviously different types of instability - sequence instability vs chromosome instability

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

How many cancers display sequence instability

A

15% - stable chromosomes but unstable sequences

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

poor function of what system leads to colorectal cancer 15% of the time

A

mismatch repair (leading to sequence instability)

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

Is sequence instability the most common cause of colorectal cancer

A

no
, most
cases have a lot of rearranged chromosomes, chromosomal instability or ‘CIN’,
while generally having a near-normal rate of point mutation

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

what kind of mutation leads to CIN

A

mutations in managing or repairing chromosomes

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

What are the different types of DNA repair for single strand breaks

A

NER
BER
MMR

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

What damage does MMR deal with

A

mismatched bases and also small loops that occur where polymerases slip while replicating repeats and add or delete a copy of the repeat, generating a tiny mismatched loop

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

What happens to mismatched loops when MMR machinery is defective

A

slippage loops persist, and a striking effect is

shrinkage or expansion of short repeats known as microsatellites

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

What technique are microsatellites used for

A

forensic DNA fingerprinting

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

Give 2 ‘symptoms’ of faulty MMR

A

microsatellite instability

higher point mutation rate

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

what usually causes the MMR failure in 15% of colon cancers

A

inactivation of MLH1 or MSH2, key components of mismatch repair

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

how does the mutation rate in colon cancers with dodgy MMR compare with the rate in a normal cell

A

roughly hundred-fold increased rate of small mutations including both single base changes (mismatches) and frameshifts caused by elongation or shortening of repeats such as AAAAAAA.

Both TGFbetaRII and Bax suffer such mutations

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

How does inactivation of MLH1 often occur

A

by epigenetic change, methylation of

the DNA of its promoter

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

which repair system is lost in Lynch Sydrome

A

MMR

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

What does HR repair of DSB require

Which proteins are important in this repair system

A

there being 2
copies of the genome: it uses the other copy of the broken sequence—usually the
sister chromatid—as a template to resynthesise the broken bit

BRCA1/2

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

What are the BRCA genes infamous for

A

hereditary breast cancer

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

Which repair system deals with crosslinked DNA

A

HR

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

mutations in which protein account for a few percentage of colon cancers

A

mutations in DNA polymerase epsilon proof-reading domain can cause an extremely high error rate

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

what is a source of chromosome instability other than defects in repair mechanisms

A

defects in mitosis

eg multiple centromeres, in some cancer cells, chromosomes sometimes get left behind or broken during anaphase

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

why does a mutation of p53 lead to instability

A

If DNA is damaged or replication encounters a problem, the cell cycle should be halted at a checkpoint, so defects in checkpoints may leave problems unresolved

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

what is the Vogelstein model of colorectal cancer

A

model of the development of colorectal cancer (revised 2008), trying to relate genes mutated to stages in cancer development. The model is speculative and oversimplified—e.g. only some colorectal cancers have these particular mutations, and the list of mutations is not complete

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

What is the sequence of mutations in the Vogelstein model of colorectal cancer

A

1) APC or beta-catenin;
2) CDC4 or CIN;
3) KRAS or BRAF;
4) PIK3CA or PTEN;
5) p53/TP53 or BAX;
6) SMAD4 or TGF-beta.

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

why are the APC and beta-catenin mutations interchangable in the Vogelstein model of colorectal cancer

A

both may be required for adenoma formation—mutating either gene has much
the same effect so these are alternatives

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

how high is the risk of developing cancer if you have a hereditary predisposition

(not including weak predispositions that increase risk by 1.3x eg)

A

e.g. hereditary predisposition to breast cancer, the affected individual has a high risk, usually at least 50% and often higher, of developing cancer.
The individual inherits one of the mutations required to get a cancer, and so is one step down the road to cancer.

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

What is an inherited mutation predisposing cancer usually in

why does this increase their chances of cancer

A

usually (but not always) in one copy of a tumour suppressor gene rather than an oncogene

In a normal individual—known as a ‘sporadic’ case—2 mutations, in the same cell, one on each copy, would be required to inactivate this tumour suppressor gene.

In the predisposed person, all their cells start with one copy inactivated, but one intact so that cells behave normally, and the second mutation occurs in occasional cells during life to alter the cell’s behaviou

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

Why are hereditary predispositions to cancer important

A

(a) Because they are among the commonest genetic diseases,

(b) They provided an important route to discovery of tumour suppressor genes including APC and RB1.

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

How common are hereditary predispositions to cancer

A

among the commonest potentially-lethal hereditary diseases, i.e. are more common than most of the well-known non-cancer genetic diseases that are potentially lethal.

At least a few percent of cancer is due to such predispositions and the individuals affected may have a very high probability of developing the particular cancer, e.g. BRCA2 mutation gives a lifetime risk of around 40-80% of breast cancer

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

true or false

inhereditary predisposition to cancer mutations are always in genetic instability genes

A

false

can either be in growth control genes (e.g. APC, RB1) or in genetic instability genes (MLH1, MSH2, BRCA2).

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

what are the 2 best known examples of hereditary predisposition to colon cancer

A

Familial Adenomatous Polyposis

Lynch Syndrome

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

What are the other names for familial adenomatous polyposis

how common is it

A

Adenomatous Polyposis Coli or just polyposis coli

1/10,000

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

What happens in familial adenomatous polyposis

A

individuals develop ~1000 polyps
(adenomas) in colon in late adolescence

associated with mutation in tumour suppressor APC gene

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

How common is APC mutation in sporadic colorectal cancer

A

Around 80% of sporadic colorectal cancers also have this mutation, but both copies have to be damaged after birth

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

what proportion of colon cancers are from Lynch Sydrome

What are the mutations in

what do these genes encode

A

1%

(one copy of) MLH1 and MSH2, both encoding components of DNA mismatch repair

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

How common are MLH1 and MSH2 mutations in sporadic cases of colon cancer

A

15% have mutation

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

What is Lynch Sydrome also called

A

old name Hereditary Non-Polyposis Colon Cancer, HNPCC but it afffects other tissues as well

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

How much of breast cancers are in individuals with an inherited predisposition

what genes are often involved in inheritance

A

5%

Perhaps half of these have a mutant copy of BRCA1 or BRCA2.

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

How are BRCA 1 and 2 related

A

unrelated proteins but are both components of DNA strand break repair, and loss of both copies of the genes gives genetic instability.

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

what is the major difference between the genes associated with inherited predisposition to breast rather than colon cancer

A

APC, MSH2 and MLH1 are found in both predisposed and sporadic colon cancers

BRCA1/2 are rarely mutated in non hereditary breast cancer

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

what are BRCA1 and 2 involved in normally

so what happens if they are lost

A

both components of DNA strand break repair, and loss of both copies of the genes gives genetic instability

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

what is retinoblastoma

A

a rare tumour arising in the immature retina, in children under a few
years old. About 40% of cases are hereditary, and on average predisposed individuals
develop 3 tumours (range 0 to many)

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

what cancers does a BRCA2 mutation lead to (3)

A

breast
prostate
ovarian

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

which part of the cell cycle does Rb control

A

g1/s checkpoint

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

What is the Knudson 2 hit hypothesis

A

The idea that typical tumour suppressor genes require two mutations to inactivate both copies

developed from studying retinoblastoma incidence

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

true or false

genetic instability may be separated from growth control

A

true
When cancers with BRCA2 mutations are treated with DNA-crosslinking agents, treatment selects for cells that can repair the lesions, so selects for revertants of the BRCA2 mutation. These revertants have lost genetic instability but go on to kill the patient

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

What are the hallmarks of cancer that are concerned with loss of growth control (5)

what is another key hall mark

A
independence of growth stimulating signals
resistance to growth inhibitory signals 
differentiation block
resistance to apoptosis 
immortality 

genetic instability

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

What are the 4 more controversial hall marks of cancer

A

metabolic changes
metastasis
angiogenesis
evasion of immune response

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

Why is angiogenesis a controversial hallmark of cancer

A

arguably any growth in the body will grow extra blood vessels, not just tumours

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

Which receptor type are usually involved in growth factor pathways

A

RTK

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

name 3 types of signaling pathway in cancers

A

GFs
Wnts
TGF-beta

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

What does the TGF-beta family usually do in cancer

A

generally inhibit proliferation in epithelial cells.

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

Which of the 3 main types of cancer signaling pathways are involved in the Vogelstein model of colorectal cancer

A

pro-proliferation pathways—the Wnt pathway; and two pathways downstream of receptor tyrosine kinase signalling, the KRAS-BRAF MAP kinase pathway and the PI3 kinase-Akt pathway—and a growth-inhibitory pathway—TGF-beta signalling

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

Why is Wnt pathway of particular importance to the colorectal cancers

A

almost all colorectal cancers have a mutation in it, either in APC, beta-catenin, or, more recently discovered, other components such as a Tcf transcription factor

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

What is the most frequent mutation in the Wnt pathway in colorectal cancer

A

APC inactivation

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

What does beta catenin do

A

acting with Tcf transcription factors in the nucleus, drives cell proliferation and/or clonal expansion.

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

How do APC and Wnt relate to beta catenin

A

APC forms part of a complex that normally degrades beta-catenin, and Wnt signalling prevents the degradation.

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

Which type of mutations are required to make the Wnt pathway cancerous and pro-proliferative

A

inactivation of APC or activation of beta-catenin

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

How is activation of beta-catenin usually achieved

A

by preventing degradation, typically by point-mutating the motif on beta-catenin that the degradation machinery recognises.

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

What is the target of Herceptin in breast cancer treatment

A

HER2

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

Name 2 receptors in the ErbB family

A

EGFR

HER2

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

What are the 2 possible downstream pathways from ErbB receptors

A

1) MAPK (signals via RAS and RAF family members)

2) PIP3 (PI3K phosphorylates PIP2 to PIP3, which activates AKT, inhibiting apoptosis)

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

What dephosphylates PIP3

A

PTEN

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

What would have to happen PTEN to cause cancer

A

PTEN is a tumour suppressor (dephosphorylates PIP3 so stops inhibition of apoptosis) so must be deleted

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

What are the alternatives to PI3KCA and PTEN in Vogelstein’s colorectal cancer model

A

KRAS/BRAF

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

How do TGF beta family peptide GFs signal

A

via a transmembrane receptor to the SMAD family (which carry signals to nucleus)

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

How are TGF beta families implicated in cancer

A

Mutations in TGFbetaRII (TGFbeta receptor two), SMAD4 or SMAD2 are quite common in colon cancer.

All are tumour suppressor genes.

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

How does mutating RAF differ from mutating RAS in cancer

A

Mutating RAS has much the same effect as mutating RAF, so both mutations can be regarded as activating the RAS-RAF pathway

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

What is an example of differentiation block in the Vogelstein model

A

inactivation of APC (or activation of beta-catenin).

Mutating APC in the stem cell compartment of the
colon in an animal model blocks differentiation: the stem cell compartment expands and the proliferating cells are no longer able to migrate up the villi.

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

True or false

Leukaemias must arise as in full differentiated lymphocytes

A

false
Leukaemias can arise in stem cells or in fully differentiated lymphocytes (as in chronic B-lymphocytic leukaemia and also myeloma which arises in plasma cells).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

How can chronic and acute leukaemias differ

A

stem cell leukaemias can either have differentiation blocked giving acute
leukaemias, with stem cells in the blood; or retain differentiation, as in chronic myeloid leukaemia, where cells in the blood are differentiated

120
Q

Is there a difference in severity between stem cell and differentiated leukaemias

A

The leukaemias with blocked differentiation are more aggressive than the differentiating ones

121
Q

Name 2 anti apoptotic mutations

A

inactivation of p53 and BAX

122
Q

How do p53 and BAX interact

A

p53 upregulates BAX to promote cell death via MOMP and casp activation

123
Q

Which of BAX’s relatives oppose its effects

A

BCL2

124
Q

How does the PIP3 pathway affect apoptosis

A

acts to reduce apoptosis

125
Q

What did Hayflick show in the 1950s

A

normal human somatic cells in culture would only divide a fixed number of times before entering cycle arrest, a response known as ‘senescence’, and tumour cells escaped this limit to grow indefinitely, they are ‘immortal’. Limited division potential is controlled by telomere length

126
Q

What are telomeres

How are they restored

A

repeat structures at the ends of chromosomes, and in most somatic cells they are shortened at each division

telomerases

127
Q

How do 10-20% of cancers turn on telomerase

How else might they do it

A

point mutation in the promoter

a rearrangement of DNA brings in a promoter from another gene

128
Q

Give an estimate for the hayflick limit

A

50-100 divisions

129
Q

How much do telomeres shorten per cell cycle

A

100bp

130
Q

What is oncogene induced senescence

A

senescence due to activation of some oncogenes, e.g. expressing mutant RAS in otherwise normal cells

131
Q

How can cancers remove stress responses

A

Mutation of p53, with or without mutation of RB1, alleviates stress responses.

132
Q

How do viruses eg HPV associated with cervical cancer overcome senescence

A

by binding and inactivating Rb-1 and p53.

133
Q

How common is p53 in human neoplasm

A

It is mutant in around one-third to half of all human neoplasms.

134
Q

Describe levels of p53 levels

A

can be raised in minutes. Protein is constantly being translated then
degraded, so blocking degradation rapidly
raises the level.

135
Q

What did Folkman postulate

A

tumour growth might be limited by the need for angiogenesis, and that a tumour had to produce angiogenic factors

136
Q

How do tumour cells migrate through tissue

A

It used to be assumed that this was a destructive process, involving proteases, but recent videomicroscopy shows cells slipping through pre-existing spaces between cells, along collagen fibres, etc

Cells eventually get into the lymphatics or veins

137
Q

How easily do tumour cells spread once they have reached the circulation

A

cells can circulate surprisingly freely, to distant sites, e.g. from one breast to the other.

138
Q

Give 2 experiments on metastasis

A

Cells injected into capillary beds can be isolated from draining lymphatics: they can cross tissue.

Cells injected at different points, e.g.
left ventricle and tail vein, give much the same eventual
distribution of metastatic colonies.

139
Q

How can we show that the site of metastasis is at least a little bit dependent on an intrinsic property of the primary tumour

A

by selecting variants of a mouse melanoma line that preferentially colonized respectively the brain or the lung independently of where they were injected.

This was done by repeated intravenous injection of cells and recovering colonies from the chosen organ

140
Q

What does the process of metastasis entail

which step(s) are critical

A

escape of cells into vessels, survival in the circulation, escape out of vessels into tissue, then survival and growth.

only survival and growth in a distant site

141
Q

How do we know that the major barrier to metastasis is survival and growth in the distant site

A

video microscopy of fluorescent cells injected into the circulation shows
that even normal cells can circulate freely and exit the circulation quite efficiently

142
Q

True or false

metastasis is very inefficient

A

true

Many cells may be released from a tumour with only a few metastatic colonies forming

143
Q

If only a few cells in tumour were capable of metastasis, what would the secondary tumours look like

how do we know this is wrong

A

highly purified for metastasis-capable cells and would give more metastases if transplanted

this was tested by Weiss who found Cells from metastases were no more metastatic than cells from the primary tumour, so most primary tumour cells are capable of metastasis

144
Q

What is the lecturer’s view of the step from malignancy to metastasis

A

requires little or no additional genetic change; it is first and foremost a rare stochastic process.

145
Q

What do Tcf proteins target

when might you find a mutant version of this

A

Wnt pathway

in colon cancer instead of APC or beta-catenin (itself a transcription factor)

146
Q

What is MYC

What is ERG

A

MYC is an oncogene and transcription factor that seems to powerfully upregulate many genes involved in proliferation and is widely activated.

ERG is a transcription factor that may control differentiation; fused in prostate cancer

147
Q

Name a chromatin modifier involved in cancer

A

MLL, fused to other genes by chromosome translocations in leukaemias, is
a histone methylase

148
Q

Name a complex that modifies chromatin

how is it mutated in cancer

A

BAF - moves nucleosomes

several components of these are mutated, e.g. ARID1A. overall maybe 20% human cancers have mutations in BAF components.

149
Q

What is E-cadherin

how is it involved in cancer

A

a cell adhesion molecule

inactivated in some breast cancers, often by epigenetic change: DNA methylation

150
Q

Give an example of mutations to carbohydrate metabolism leading to cancer

A

mutations in isocitrate dehydrogenase IDH1, or IDH2 an enzyme in the
Krebs cycle. e.g. in some brain tumours
and leukaemias

changes the enzyme specificity to make hydroxyglutarate, which accumulates and interferes with DNA demethylation (which may block differentiation) and histone acylation (which may block DNA damage signalling)

151
Q

What does chromosome translocation involve

A

pieces of two chromosomes are joined to each other, following breakage of the DNA or an accident during DNA replication.

152
Q

What are the possibly sequence level changes that can occur

A

SNV

indel

153
Q

Why do indels usually give truncated proteins

A

because most of

them cause frameshifts and frameshifts usually lead to a stop a few amino acids later

154
Q

What are 4 large scale mutations/ changes that can happen to DNA

A

deletion
duplication
amplification
Chr translocation

155
Q

What is gene fusion

A

when rearrangement of large chunks of DNA creates a new gene by joining two genes together to create a new gene

156
Q

How do we study sequence level DNA changes

A

PCR - amplifying a region of interest—say an exon—and sequencing it on a Sanger sequencer

Increasingly this is replaced by Illumina (originally Solexa) sequencing

157
Q

What is Illumina

A

millions of DNA fragments, are attached to a glass slide and sequenced simultaneously (hence ‘massively-parallel sequencing’)

158
Q

How can structural DNA arrangements be found

A

d by finding sequence fragments that cross rearrangement junctions—e.g. for the BCR-ABL fusion, finding a DNA fragment with sequence from the BCR gene at one end and sequence from the ABL gene at the other end.

159
Q

What is cytogenetics

A

study of chromosomes by microscopy

160
Q

How would you perform a cytogenetic study

A

Cells are arrested in metaphase and their chromosomes spread on a slide and stained. This can show chromosome translocations and some large deletions or inversions.

161
Q

what is the most famous example of a Chr translocation

A

reciprocal translocation between chromosomes 9 and 22 found in most
chronic myeloid leukaemias. The smaller chromosome formed is called the Philadelphia Chromosome.

162
Q

Why can you not study carcinomas by classical cytogenetics

A

it’s difficult to get metaphases and there are so many complicated rearrangements they can’t be identified

163
Q

What is FISH

What is the method

A

fluorescence-in situ hybridization

DNA is labelled with fluorescence and hybridized to metaphase chromosomes. DNA from a chromosome can be labelled, or a small segment of genome can be labelled, e.g. in the example chr12 in red, and a 200kb chunk including the NMYC gene in green. This example shows amplification of N-MYC in a lung tumour.

164
Q

How can FISH be used clinically

A

e.g. to detect amplification of HER2(ERBB2), the

target of Herceptin therapy in breast cancer.

165
Q

How have large deletions/ amplifications been detected

A

by measuring the relative loss or increase in the amount of DNA in different regions of the genome

166
Q

What is RAS

A

a G-Protein that is activated by GTP binding and inactivated by hydrolysis of the GTP to GDP

167
Q

What do mutations to the RAS pathway affect (2)

A

mutations block the hydrolysis, at least some of them by preventing access of GTPase-activating proteins that complete the active site.

activation of BRAF

168
Q

Which particular RAS is important to cancer

A

K-RAS in particular in frequently mutated in a range of human cancers.

169
Q

Give the steps of the RAS pathway (4)

A

EGF binds to EGFR
EGFR activates Grb2 and SoS
Grb2 activates RAS, which activates BRAF
BRAF activates MAPK …

170
Q

Which cancers are related to BRAF mutation

A

notably in melanomas (60% cases) and colorectal carcinomas.

171
Q

How are BRAF and RAS mutations related in cancer

A

B-RAF and Ras mutations seem to be alternatives with rather similar effects on the cell.

172
Q

What is the commonest change to BRAF in cancer

A

BRAF V600E

commonest mutation
changes a valine to a glutamic acid at position 600

This is adjacent to an activating phosphorylation site, and the negative charge of the glutamic acid presumably mimics phosphorylation and so activates.

173
Q

What is PI3KCA

A

the catalytic subunit of PI-3Kinase

174
Q

What is the importance of PI3K to cancer

A

PIK3CA is mutated in about 1/3 of breast cancers, the most frequent mutation yet found in breast cancer. One of the Common mutations changes one particular negatively charged amino acid to a positively charged one, clearly a dramatic change.

175
Q

Give an example of a mutation that a) stabilises the active conformation of a protein or b) mimics an activating phosphorylation

Are these oncogenes or tumour suppressors

A

a) RAS
b) BRAF

oncogenes

176
Q

How do the ease of getting an activating mutation in an oncogene compare to deactivating a tumour suppressor

A

oncogene-activating mutations have to be precise mutations at particular amino acids; in contrast inactivating tumour suppressor genes is much easier—anything that interferes with the protein’s function will do.

177
Q

What is PTEN and how is it associated with cancer

A

which reverses the action of PIK3CA—is a known tumour suppressor gene and can also suffer inactivating point mutations or deletions.

178
Q

What kind of sequence change usually inactivates tumour suppressors

A

Indels are thought to be important particularly as inactivators of tumour suppressors,
as they usually truncate.

Many APC mutations are indels.

179
Q

Name 3 proteins inactivated by deletion, leading to cancer

A

Deletions of tumour suppressors are common.

PTEN is often deleted, as are the cellcycle controllers Rb and p16/INK4A.

180
Q

Give an example of duplication in cancer

A

a fusion of BRAF to a neighbouring gene KIAA1549, as a result of tandem duplication of a segment of DNA (in paediatric brain tumours)

181
Q

Give 4 examples of amplification gene mutations in cancer

A

EGFreceptor in brain and ERBB2/HER2 in
around 10-20% breast cancers.

Also Cyclin D1 in breast; and MYC family genes in various cancers

182
Q

True or false

there is only one way to activate an oncogene

A

false
BRAF can be activated by point mutation or gene fusion, and fusion can be by tandem duplication or chromosome translocation.

Even RAS can be fused (rarely)

EGFR and ERBB2 can be via SNV or amplification

183
Q

What is BCR-ABL

A

iconic fusion found in most cases of
chronic myeloid
leukaemia, formed by the reciprocal 9:22 translocation

184
Q

What is ABL

how is it regulated

A

tyrosine kinase

regulatory domain at the N terminus, and, like most tyrosine kinases, activated by dimerisation

185
Q

Why does the BCR-ABL fusion work

what does the fusion do (3)

A

N terminus of the BCR protein naturally forms dimers or oligomerises.

The fusion does several things:
the inhibitory N terminus of ABL is removed;
BCR holds the ABL kinase in dimers or oligomers, activating it;
the BCR promoter may be stronger than ABL’s.

186
Q

What is an important example of fusion in a carcinoma

how common is it

A

TMPRSS2- ERG, found in ~50%
prostate cancers. Since prostate cancer is common, this is the most
prevalent known fusion gene by far.

187
Q

How is the TMPRSS2-ERG fusion gene formed

A

by deletion between the two genes TMPRSS2 and ERG, rather than by chromosome translocation.

188
Q

Give 2 examples of genes inactivated by DNA methylation

A

MLH1 (mismatch repair) and E-cadherin (cell adhesion) respectively in colon cancers and lobular breast cancers

189
Q

How common is epigenetic instability in cancer

what does this mean

A

30-50% colon cancers

analogous to genetic instability, i.e. a high rate of aberrant DNA methylation .

190
Q

Name 2 proteins that can be inhibited by carcinogenic viruses

A

p53 and Rb1

191
Q

How many driver mutations are required for cancer development

A

Vogelstein model suggests 6 - but this is incomplete (so >6, more like 10)

192
Q

What is a good way to estimate the proportion of mutations in a tumour that are selected for

A

compare the relative proportions of mutations that do or do not alter amino acids, since most of the latter will be random

193
Q

What has statistical analysis of tumours revealed about the amount of driver mutations in colon cancers

how does this compare to other adult carcinomas

A

> 10 coding mutations are drivers in colon cancer, and this does not include DNA rearrangements or epigenetic gene changes

other adult carcinomas are similar, but a higher proportion are DNA rearrangements. At least half of these mutations have not been identified yet

194
Q

What is the estimate for the number of driver mutations in breast and ovarian cancers

A

about 8 mutations per tumour are already known in breast and ovarian
cancer, of which 75% are rearrangements

195
Q

How do we find out what mutating the genes does to the cell and tissue?

A

Cells in vitro

Animal models

196
Q

Having found a mutation in cancer, what would we first need to find out

A

first that it actually does contribute to cancer development and isn’t a random mutation; and second, what sort of effect it might have

197
Q

Give an example of finding out the function of a mutation in vitro

A

e.g. the classical ‘transforming’ activity of mutant RAS in fibroblasts—mutant-bearing cells overgrow their neighbours

198
Q

Give an example of using an animal model to find out the effect of a cancerous mutation (for an oncogene)

A

genomic region around the gene CCND1, which encodes CyclinD1 (in Rb1 pathway), is amplified in many breast cancers, but this doesn’t prove CCND1 is an oncogene, because a neighbouring gene
might be the real culprit.
A transgenic mouse was made that expresses a lot of CyclinD1 from the MMTV promoter, which is active specifically in the mammary glands: the mice get mammary hyperplasias that develop into tumours, suggesting CCND1 is indeed the key gene

199
Q

Give an example of using an animal model to find out the effect of a cancerous mutation (for a tumour suppressor)

A

inactivation of APC in the crypts of the colon in mice, mentioned in
previous lecture, which showed that removing APC profoundly alters the differentiation pattern of the crypt, so that the dividing cell compartment expands and the cells are prevented from migrating up the villus and maturing.

Expression of cre recombinase is activated by administration of a drug and recombines between lox sequences to cut out the APC gene.

200
Q

How can you mimic deletion of APC (tumour suppressor) in a mouse model

A

Expression of cre recombinase is activated by administration of a drug and recombines between lox sequences to cut out the APC gene.

201
Q

Name some things we are actually sure cause cancer

A

smoking tobacco, some important tumour viruses, and some occupational exposures such as asbestos

202
Q

How much of the cancer in england does tobacco cause

A

30% (probs higher in actual fact)

203
Q

true or false

cancer is more common in some populations

A

Incidence of particular cancers varies dramatically—sometimes 50-fold—between
populations (after correcting for age distribution), for example prostate, liver, melanoma. But the total amount of cancer does not seem to vary so much

204
Q

Which group of people were studied to see where the difference in cancer type incidence stems from

What was found

give some more evidence to further support this

A

migrant populations such as Japanese immigrants to the west coast of the United States.

changed from having the Japanese pattern of cancer, a high incidence of stomach cancer, but a low incidence of breast cancer, to the American pattern, which is the other way round, within a generation or two: clearly not primarily genetic

Similar changes have occurred in Japan as it has become westernized.

205
Q

What kinds of environmental factors could affect cancer risk

A

agents that cause mutations in DNA, but could also include lifestyle factors (e.g. reproductive behaviour, nutrition) that affect the rate of ‘spontaneous’, DNA damage such as errors in replication that go uncorrected

206
Q

What is the principal carcinogenic effect of UV light

A

dimerisation of pyrimidines (TT or CT)

207
Q

Which group of people demonstrate the effect of UV light on DNA and the importance of DNA repair mechanisms

A

UV sensitivity of Xeroderma Pigmentosum patients who lack excision repair

208
Q

What are the 5 types of carcinogenic agents

A
UV light
Ionising radiation 
Chemical carcinogens
Minerals (eg asbestos)
infectious agents (eg HPV)
209
Q

name some chemical carcinogens (5)

A

smoke
aflatoxin
Dimethyl nitrosamine (in some meat products)
mustard gas

210
Q

True or false

all carciogens are very reactive

A

false
some are, eg mustard gas
but the most potent chemical carcinogens are more or less inert in the form we are exposed to, but get activated by metabolism

211
Q

Why are the most potent carcinogens not usually reactive without being metabolised

A

Highly reactive chemicals if introduced into the body would react with something else before they could reach the DNA

212
Q

What system are enzymes that activate carcinogens part of

A

e ‘xenobiotic metabolism’ systems that have evolved to render harmless or ‘detoxify’ lipid-soluble molecules that the body needs to get rid of, by making more soluble derivatives of them, that can be excreted

213
Q

What fact is β–napthylamine an important example of

how

A

Mutagenic chemicals tend to show tissue specificity

activated by hydroxylation, but then rapidly made soluble and harmless by addition of glucuronic acid.
However, if the glucuronate is hydrolysed off again, the reactive form is regenerated. β-napthylamine acts primarily on the bladder in man, where the glucuronic acid conjugate is removed by glucuronidase. This bladder specificity made it possible to identify it as a carcinogen of importance in humans.

214
Q

True or false

Mutagenic chemicals often show species-specificity

A

true

probably down to variations in metabolism, either activation or detoxification of the carcinogen, e.g. β-naphthylamine is a potent bladder carcinogen only in species that have glucuronidase in the bladder, such as dog and man, not rodents

215
Q

What is NMU

What feature of carcinogens has it been used to identify

A

nitroso-methyl-urea

Mutagenic chemicals (and radiation) often show stage-specificity

216
Q

how was NMU used to show Mutagenic chemicals (and radiation) often show stage-specificity

A

When the mutagen NMU (nitroso-methyl-urea) was fed to female rats of different ages, the
rats were most susceptible to developing mammary tumours when pubescent, when the mammary epithelium is proliferating.
Similarly breast cancer in human survivors of Japanese atomic bombs occurred mainly in those who were teenagers when exposed

217
Q

Give a reason mutagens may be stage and tissue specific

A

β-naphthylamine suggests that one factor is tissue-specific metabolism, while the mammary cancers suggest that rapid cell proliferation makes the tissue susceptible

218
Q

Give 2 important examples which debunk the idea that carcinogens are man made

A

Aflatoxin B1

Aristolochic acid

219
Q

What is aflatoxin B1`

A

the most powerful carcinogen known to man, at least when assayed in rats, and it is a natural contaminant of peanuts, produced by the fungus Aspergillus flavus, which sometimes grows on peanuts in warm humid conditions.
Like other carcinogens it is activated by metabolism.

220
Q

What is Aristolochic acid

A

from Aristolochia plant species, causing kidney toxicity and kidney and liver cancer, and is present in some foods and
traditional medicines.

221
Q

What is a carcinogenic signature

eg?

A

Sequencing shows that some carcinogens give a characteristic pattern of mutation, and this may help in future to identify exposure.

E.g. many mutations found in melanoma are those expected from UV exposure, predominantly C>T.

222
Q

Name 2carcinogen signatures

A

many mutations found in melanoma are those expected from UV exposure, predominantly C>T.

Aristolochic acid produces a characteristic mutation pattern, found in upper urinary tract carcinomas, predominantly changing A to T at A[C|T]AGG. This ‘signature’ has also been found in a subset of southeast-Asian liver cancers, suggesting it might have been the cause of those cancers.

223
Q

What does asbestos cause

how?

A

inhalation causes mesothelioma, arising from the mesothelial lining of the pleural cavity. Probably, small fibres that penetrate deep into the lung cause chronic inflammation and cell turnover of these cells

doesn’t damage dna but but is a ‘tumour promoter’

224
Q

What are tumour promoters

A

substances that promote the development of tumours without damaging DNA.

225
Q

Why is there a distinction between tumour initiators and promoters

A

Classical experiments painting substances on mouse skin
distinguished ‘initiators’ (mutagens) from ‘promoters

If an initiator was applied, it would give the occasional tumour but far more tumours would appear if, after the initiator, a promoter was repeatedly applied. Promoters only worked after initiators had been applied.
Promoters behaved like drugs.

226
Q

Name a natural tumour promoter

A

the most potent promoters we know are natural

The best known is TPA, which comes from the seeds of Croton tiglium

227
Q

How does TPA promote cancer

A

mimic diacyl glycerol, agonist for protein kinase C.

228
Q

Are the following promoters or initiators of cancer

a) hormones
b) cig smoke
c) asbestos

what links b and c

A

a) promoter
b) both
c) promoter

Chronic irritation/inflammation behaves as a promoter, perhaps accounting for asbestos’ action and the promoter activity of smoke.

229
Q

Give an example of a non chemical environmental/ life style choice that increases the chance of cancer

A

breast cancer
Risk is roughly proportional to the interval between menarche(onset of menstrual cycling) and first pregnancy, for childbearing women - probably related to cell proliferation

230
Q

What are the 2 key things we need to find out when assessing a chemical that may cause cancer

A

(1) Whether a substance is capable of acting as a mutagen or promoter;
(2) How potent it is - how much can we afford to be exposed to.

231
Q

Why do we need to know how potent a carcinogen is

A

bc we cannot eliminate all of them from the environment (that would include removing O2 and tomatoes for example)

232
Q

What are the 3 general ways to identify a carcinogenic agent

A

In humans – epidemiology
In vivo – mice and rats
In vitro – the Ames test and others

233
Q

How were asbestos and β napthylamine discovered to be carcinogenic

does this method of discovery always work?

A

bc a particular set of people developed a particular type of tumours

no: only works well where a specific group of people is exposed to a strong carcinogen, and the cancer is relatively specific to those people—dyestuffs workers were exposed to β-napthylamine and got bladder cancer, which is relatively uncommon; construction workers inhaled asbestos and got mesothelioma, which is almost unique to asbestos-exposed individuals

234
Q

Why did epidemiology work to discover the carcinogenic nature of tobacco smoke

A

easy to distinguish people heavily exposed (smokers) from those who are only lightly exposed (non-smokers), and the effect is large (5-10X risk even in earliest studies) and distinctive—squamous lung cancers are rare in non-smoking populations.

235
Q

Name 2 things that are strongly associated with liver cancer

what is the increased risk factor

A

HBV and aflatoxin

combined risk factor of around 50x

236
Q

What is the technique for testing carcinogens on animals

A

o expose animals to as high a dose as possible for as long as possible and look for tumours. Whether this works is debatable.

237
Q

What did experiments on rats and mice for carcinogens reveal

what does this mean for the amount of cancerous agents we are exposed to

what is the precise difficulty with this method

A

of 800 compounds that had been tested, 65% gave a significant increase in tumours in at least one organ and species.

Extrapolating to 60,000 compounds we are exposed to, 40,000 would be ‘carcinogenic’
but on average each would only be responsible for 1/40,000th of human cancer and so unimportant.

many compounds can be carcinogens sometimes, but the effective dose and hence the risk they pose is unknown.

238
Q

How do the results of animal models used to find carcinogens compare between mice and rats

A

2/3 concordance

239
Q

Give 4 advantages of in vitro testing for carcinogens compared to animal models

What is a key disadvantage and how can we overcome it

A

more humane, cheaper, quicker and more statistically robust

only limited ability to reveal carcinogens that require metabolic activation, though a rat liver extract can be added to try and provide this.

240
Q

which organism is used in the Ames test

A

salmonella strains which carry mutations in genes involved in histidine synthesis. These strains are auxotrophic mutants, i.e. they require histidine for growth, but cannot produce it.

241
Q

What does the Ames test analyze

A

the ability of a substance to cause a mutation

tests the capability of the tested substance in creating mutations that result in a return to a “prototrophic” state, so that the cells can grow on a histidine-free medium.

a positive result means the substance is mutagenic and thus potentially carcinogenic

242
Q

Give some sources of ionising radiation (5)

A

X rays

γ rays,

neutrons,

β particles (free e-),

α particles, (charged helium nuclei)

243
Q

How does radiation damage DNA

A

by producing free radicals and ions as it passes through tissue; these then react with DNA and alter the structure of bases or cause strand breaks

244
Q

How is exposure to radiation measured

what units are used

A

as the amount of energy absorbed
per unit of tissue:

Gray (Gy)
(the Gray supersedes the rad: 1 Gray = 100 rads), which is joules absorbed per Kg
tissue.

245
Q

What does damage by alpha particles look like

A

Alpha particles leave dense tracks of ions and radicals (high LET) so that if they pass by a DNA helix they are likely to cause double-strand breaks or multiple chemical changes, which may be difficult to repair reliably.

246
Q

What does damage by gamma rays look like

A

leave scattered ions and radicals (low LET) and so will usually only damage one residue on one strand of a DNA molecule, which is usually repairable.

247
Q

What is the radiation in Gray usually multiplied by

A

Quality Factor for the particular type of radiation, to give an approximate measure of biological damage or dose equivalent measured in Sieverts (Sv).

248
Q

What are the Quality factors for different types of radiation

A

1 for X-rays and gamma rays and ranges up to 20 for alpha particles

249
Q

What is the average UK exposure to radiation

A

2.5 mSv/year of which 1/8th medical, 1/100th cosmic rays from long-haul flights, ½ radon escaping from rocks.

250
Q

HOw can we estimate the effects of radiation doses we experiencce (eg 0.01 Gy)

A

extrapolate back from high doses such as 3 to 5 Gy, where we can measure cancer in people exposed.

eg from the 120,000 survivors of the atomic bombs dropped on Hiroshima and Nagasaki in 1945

251
Q

What data can we use to extrapolate from to estimate the radiation dose-cancer relationship (other than rfom the atom bombs)

A

people with Ankylosing spondylitis were treated with X-rays in 1935-1954, and increasingly we have data for uranium miners etc.

252
Q

Give an example of a virus directly causing cancer`

A

inactivation of p53/ Rb by the E6/7 proteins of HPVs

253
Q

Name a parasite associated with cancer

A

Schistosomes and bladder cancer

254
Q

Name a bacteria associated with cancer

A

Helicobacter pylori association with gastric adenocarcinoma and MALT lymphoma (lymphoma associated with intestinal mucosa).

255
Q

What are the strong epidemiological links between H pylori and cancer (2)

A

eradicating H pylori reduces the incidence of gastric adenocarcinoma;
and H pylori infection induces gastric cancer in animal models.

256
Q

Which types of HPV are particularly associated with cancer

A

`HPV types 16 and 18, where chronic infection creates a high risk of cervical cancer.

257
Q

name a virus common in young Western adults (typically in uni) that can cause cancer

A

Epstein Barr virus (typically transmitted by kissing)

can cause cancer in the immunosuppressed

258
Q

What cells does Epstein Barr virus infect

who is this particularly dangerous for 92)

A

B cells and nasopharyngeal
epithelium.

particularly in ethnic Chinese populations, infection can cause
nasopharyngeal carcinoma;

in malaria-endemic areas infected children may develop the aggressive B-cell lymphoma Burkitt’s Lymphoma.

259
Q

What does HBV act synergistically with to result in cancer

A

Chronic HBV infection gives a high relative risk for primary liver cancer, and in at least some populations acts synergistically with exposure to aflatoxins.

260
Q

What is Kaposi’s sarcoma

who does it develop in

A

seems to develop in immune-suppressed or elderly people infected with HHV8.

It came to prominence as a common problem in AIDS/HIV patients

261
Q

What is HTLV-1

A

a T-cell tropic retrovirus, which causes leukaemias and lymphomas in a small proportion of infected individuals

262
Q

Where is HTLV-1 endemic

A

particularly in SW Japan, the Caribbean and parts of Africa and South America, where up to 10% of the population may be infected

263
Q

How can we make a cancer specific therapy (5)

A

kill normal tissue along with the tumour if it is unnecessary

target mutations or hallmarks

target things that cancers are more vulnerable to eg they proliferate more

treat the viral etc infection

exploit natural defenses

264
Q

What is the current course for cancer treatment typically

A

primary tumour removed surgically followed/ preceded by cytotoxic drugs or radiotherapy

265
Q

What is our first cancer specific therapy

A

Breast and prostate patients may receive hormone therapy, which inhibits growth of both normal and cancerous tissue

266
Q

Why is it hard to use targetted drugs to treat cancer on a broad scale

A

mutation information is not generally available

267
Q

When are cytotoxic drugs usually reserved for

A

when metastasis becomes evident

268
Q

What are the different ways cytotoxic drugs can damage DNA (4)

what are ways to interfere with DNA synthesis in a less direct way (2)

A

alkylate bases, intercalate non-covalently
between bases of the DNA helix, crosslink strands, or be toxic analogues of bases (e.g.
5-fluoro-uracil) that interfere with DNA synthesis.

topo inhibitors and drugs that interfere with mitosis (eg Taxol and Vinca alkaloids)

269
Q

Name 2 cancers which can be ‘cured’

A

childhood leukaemia (ALL) and teratomas in young males

treatment can lead to 20 years disease free

270
Q

When was there the breakthrough with testicular cancer

A

1975 with the introduction of cis-platin (crosslinks purine bases within and between strands)

271
Q

Why is cisplatin more effective in cancer than normal tissue

what is another drug that exploits cancer in a similar way

A

the crosslinks it creates are usually healed by HR, which requires BRCA2 (mutated in some cancers)

thus it exploits the genetic instability of the cancer

Taxanes target the mitotic spindle, which also seems to be defective in some cancers (lagging chromosomes)

272
Q

What is the Waldmann experiment

A

made cancer cells that were defective in the p21 that, downstream of p53, arrests the cell cycle in response to DNA damage.

grew them as grafts on mice and X-irradiated them. X-radiation cured
several checkpoint-defective tumours but no wild-type controls.
The checkpoint-defective cells did not arrest when irradiated and presumably died of mitotic catastrophes, while the wild-type cells underwent cycle arrest and then recovered.

273
Q

How are drugs being designed to exploit the genetic instability of cancers

A

If a DNA repair pathway is defective, cells may be killed by blocking alternative repair routes, leaving the cancer cell unable to deal even with everyday spontaneous DNA damage.

eg PARP inhibitors

274
Q

How do PARP inhibitors work

A

targets SSB repair pathway, which normally requires PARP

the PARP inhibitors mean SSBs accumulate and halt DNA replication bc at the replication fork the SSB becomes a DSB

DSB usually repaired by HR, so eg BRCA2-defective cells, they cannot repair so die while normal cells survive

275
Q

How does resistance to PARP inhibitors emerge

A

reverse mutation which restores BRCA2

276
Q

What is Glivec

A

AKA Imatinib

inhibits RTK produced by BCR-ABL fusion gene

277
Q

True or false

Imatinib is only effective against CML stemming from BCR ABL fusion

A

false

not specific for Bcr-Abl, but inhibits several tyrosine kinases including Abl (and is even used to treat cancers with activation of other kinases)

278
Q

How can cancers become immune to Imatinib

how can this be treated

A

point mutation

Second-generation drugs that bypass this resistance have been developed.

279
Q

Name some small molecule drugs / antibodies which have been developed to inhibit certain steps of the pathways

A

Herceptin, the monoclonal antibody to HER2 used in HER2-amplified breast cancer. Anti-BRAF and anti-MEK small-molecule drugs are in use.

There is no anti-RAS drug yet approved.

280
Q

How do anti-EGFR encounter resistance

A

E.g. treating colorectal cancers with anti-EGFR often results in emergence of variant, resistant tumours that have acquired mutations in genes such as RAS or RAF.

281
Q

How can BRAF V600E be treated

does resistance develop?

A

The BRAF inhibitor vemurafenib targets the ATP-binding
domain in BRAF melanomas

yes - eg by acquisition of RAS mutations or amplification of the mutant BRAF.

282
Q

What are oncolytic viruses

A

viruses that t infect and kill cancers, by lysis or exciting an immune
response

eg H101 (an engineered adenovirus)

283
Q

Why are adenoviruses well suited to be designed to be oncolytic

A

Adenoviruses prepare infected

cells for virus replication by expressing proteins that inactivate Rb1 (cell cycle control) and p53

284
Q

How are adenoviruses engineered to be oncolytic

how are they administered

what else is in development

what other viruses could be used in the future

A

have deletions in the E1B_55k gene,
which inactivates p53.
The idea is that without this E1b protein, the virus will only replicate in p53- mutant cancers.

It is injected directly into the tumour.

Derivatives of Herpes viruses, Vaccinia and VSV are in development.

285
Q

What are some of the earliest reasons for thinking the immune system protects against cancer

A

Early experiments grafting tumours from one animal to another led to the discovery of graft rejection, but this was rejection of allogeneic tissue, not the actual tumour, and led to development of inbred laboratory mice

286
Q

What did Burnet suggest about tumour graft rejection

why did his opinion carry a lot of weight

A

proposed, in an attempt to explain why graft rejection occurred (long before we understood MHC), that the true role of cellular immunity was to reject tumours. This was the concept of ‘immune surveillance’ against tumours

he proposed the clonal selection of B cells

287
Q

Some experiments have found tumour specific antigens in mouse models. what were these

A

turned out to be special cases such as env glycoproteins of retroviruses, or mutant MHC alleles, not relevant to most cancers.

288
Q

How does cancer incidence change in the immunosuppressed

A

careful studies of immunosuppressed humans and mice show no major increase in cancer,
except where the cancer has a viral aetiology .
(e.g. Kaposi’s sarcoma caused by Human
Herpesvirus 8 and lymphomas caused by Epstein-Barr Virus)

289
Q

If studies of the immunosuppressed show no major increase in cancer, what does this suggest about the role of the immune system in cancer

A

If the acquired immune system played a major role in controlling cancers, immune suppressed people would get a lot more cancer.

It may detect new proteins on cancers, but if so doesn’t do much.

290
Q

Why does contemporary immunology theory would not necessarily predict a response to tumour cells?

Is this backed up experimentally?

A

Expression of novel (mutant) proteins on a cell would probably induce peripheral tolerance.

DNA sequencing shows that mutant proteins are not selected against: (i) normal cells have many mutations that alter proteins (ii) in cancers overall there is no detectable selection against mutations that create new peptides.

291
Q

Where does most excitement centre in cancer immunotherapy

A
checkpoint inhibitors
(monoclonal antibodies that block signals that hold back cytotoxic T cells)
292
Q

which molecules are involved in downregulating cytotoxic T cells and how is this important for cancer treatment

A
by CTLA-4, PD-L1 and PD-1:
CTLA4 binds to 
B7, displacing co-stimulator CD28; 
PD1 and PD-L1
bind to each other to provide inhibitory signals 

Monoclonal antibodies to these molecules are licensed.

293
Q

Has immunotherapy made any difference to cancer treatment

A

some patients, with advanced metastases, who have failed conventional therapy, show dramatic regression of tumour and improvement in health; and secondly, some go on being healthy for at least many months. Resistance seems to develop more slowly.

294
Q

Why must immunotherapy be treated with caution

A

Only some patients with certain kinds of tumour respond, and treatment is very toxic, giving inflammation and autoimmunity that can be life-threatening

295
Q

What were some of the results from early studies of anti-CTLA4 in melanoma

A

only about 11%
of patients did better at 3 years, while treatment had to be stopped because of toxicity in 50% of all patients; 1% died of immune disease! It is said that these side-effects can now be managed better.

296
Q

Which cancers tend to respond to immunotherapy

A

hose with the most mutations and hence the highest numbers of alien peptides.

Lung and melanoma are two of the most mutated cancers, and response in these clearly correlates with number of mutations per tumour

297
Q

Why is it that cancers with the most mutations are affected most by immunotherapy

A

perhaps there are T cells that recognise mutant peptides but they are mostly tolerised.
If there are enough such antigens and tolerance is downregulated, an effective anti-cancer response may emerge, but
together with some inflammation and autoimmunity.