Neoplasia: Molecular Basis of Cancer Flashcards

1
Q

To go from a normal cell to cancer cell, we need to create a clone; what is the first thing we are going to do in order to do this?

A

we create a mutation that is divergent enough from the normal cell to be promoting cancer, but not too divergent that it kills the cell

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

after the initiating mutation, what happens next to make cancer?

A

you add on additional driver mutations

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

What are the four general types of gene classes that are responsible for oncogenesis?

A

proto-oncogenes, tumor-suppressor genes, apoptosis-regulating genes, DNA repair genes

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

What are germline mutations?

A

heritable; early mutations present as the gametes are providing the genetic material for an embryo to form; all cells in offspring carry 1 mutated allele

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

What are somatic mutations?

A

non-heritable; involve an original combination of chromosomes that have no mutation whatsoever; mutation only in cells of affected area

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

How do we make cancer from proto-oncogenes?

A

they gain function and become oncogenes

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

how do we make cancer from tumor-suppressor genes?

A

they lose function (so there is no tumor suppression)

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

how do we make cancer from apoptosis-regulating genes?

A

suppress apoptosis/cell death

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

how do we make cancer from DNA repair genes?

A

they lose function (so if there is DNA damage, they can no longer repair it)

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

What is the difference between driver vs. passenger mutations?

A

driver mutations are causal; passenger mutations contribute to cancer growth, but does not establish cancer

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

What is the genetic evolution of cancer?

A

the first mutation allows it to become a malignant clone, but even more mutations can evolve and there can be quite a heterogenous cell population

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

what are oncogenes?

A

mutated genes that result in excessive cell growth

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

what are oncoproteins?

A

the result of the genetic mutation

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

What is the mode of activation in the proto-oncogene PDGFB? and what occurs when this happens?

A

overexpression; astrocytoma

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

What is the mode of activation for the proto-oncogene ERBB1 (EGFR)? and what occurs when this happens?

A

mutation; adenocarcinoma of the lung

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

what is the mode of activation for the proto-oncogene ERBB2 (HER)? and what occurs when this happens?

A

Amplification; breast carcinoma

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

what is the mode of activation for the proto-oncogene KRAS? and what occurs when this happens?

A

point mutation; colon, lung, and pancreatic tumors

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

What is the mode of activation for the proto-oncogene MYC? and what occurs when this happens?

A

Translocation; Burkitt lymphoma

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

what is the mode of activation for the proto-oncogene NMYC? and what occurs when this happens?

A

Amplification; Neuroblastoma

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

What could be the functional product of an activated proto-oncogene (mutated)?

A

abnormal protein, excessive amount of protein, novel protein,

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

What happens when there is an amplification of growth factor/ growth factor receptor? and what is a common example of this?

A

Her-2/neu (aka ERBB2); this amplification results in too many proteins being expressed

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

Too much Her2 generates what?

A

too many protein receptors, which signals for cancer cells to divide and multiply

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

How can we treat amplification of growth factor/growth factor receptor (e.g. Her-2)?

A

we can treat with a receptor antibody called Herceptin

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

What happens when there are point mutations of KRAS?

A

the system is chronically and dramatically on for the downstream signaling (theres too much downstream signaling)

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

In a normal state, what is the favored state of RAS?

A

it tends to be in the inactivated GDP bound state; will be activated in a pulsatile manner- will be activated when it is supposed to be and then turn off again when it is not being used

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

what happens in the mutated RAS associated with cancer?

A

it defaults to the GTP-bound state- there is activation of the downstream signaling; RAS mutations can cause it to be “stuck” in the GTP-bound state

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

Mutations of RAS that contribute to oncogenesis bind it in a _____________?

A

constitutively active state (constantly active)

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

What is a characteristic feature of the mutations that occur in RAS?

A

they contribute to oncogenesis by binding it in a constitutively active state

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

what is PTEN?

A

a negative regulator of cell signaling- so if cancer wants to have uncontrolled cell growth, it needs to down regulate PTEN

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

What mutation is given to PTEN to allow cancer?

A

a loss of function mutation - so it can no longer provide its normal inhibitory function

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

What cancer is strongly associated with initiating mutations causing loss of function of PTEN?

A

endometrial carcinoma

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

What is BCR-ABL and how is it formed?

A

a hybrid gene; ABL comes from chromosome 9 and joins BCR from chromosome 22

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

what happens when BCR-ABL is formed?

A

there is now a tyrosine kinase that is now going to be extremely active inside the cell (its a non-receptor tyrosine kinase)

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

what happens when the non-receptor tyrosine kinase is upregulated?

A

it is directly feeding into the signaling pathways creating cellular proliferation

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

What is BCR-ABL known as and what is it associated it?

A

the philadelphia chromosome; commonly associated with CML

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

What does oncogene addiction mean?

A

when tumor genesis is extremely dependent on a particular oncoprotein (e.g. BCR-ABL)

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

Why is knowing particular oncogene addictions important?

A

for treatment- if you are dependent on this mechanism, it is a target for therapy for cancer

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

What is a very effective form of treatment for CML? and why?

A

tyrosin kinase inhibitors (imatinib); because of oncogene addiction

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

Is oncogene addiction very common with different cancers?

A

no; CML is a very specific circumstance where we are able to cut out the supplier with this one little snip

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

What is MYC?

A

the master transcriptional regulator; this gets out of hand with several tumors; you don’t want too much MYC because it leads to cancer

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

What is the most common extracranial solid tumor in children?

A

neuroblastoma

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

There are two ways the cyclins can become carcinogenic. What are these?

A

we can turn on the “on” switch or we could turn off the “off” switch

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

What normally inhibits the cyclin pathway?

A

p16

44
Q

what can germline loss of function of p16 lead to?

A

familial melanomas

45
Q

what cyclin is overexpressed in certain cancers such as mantle cell lymphoma?

A

cyclin D1

46
Q

what are three examples of tumor suppressor genes in normal cells?

A

RB, TP53, and APC

47
Q

what happens when RB, TP53, or APC become mutated?

A

they can no longer function as normal, so they cannot suppress tumors

48
Q

How do RB and TP53 shut down proliferation?

A

by causing senescence and apoptosis

49
Q

What is Knudson’s hypothesis?

A

two mutations involving both alleles of RB are required to produce retinoblastoma; in germline (familial) mutations, only one additional somatic hit is needed; in sporadic cases, a particular cell has to have 2 somatic mutations to knock out both genes

50
Q

germline mutations of RB mean all cells have a pre-existing mutation; these cells are at high risk of what?

A

tumorigenesis by succumbing to a second hit

51
Q

What is RB a key negative regulator of?

A

the G1/S cell cycle transition

52
Q

What does RB exist as in quiescent cells?

A

in an active hypophosphorylated state

53
Q

what does RB exist as in cells passing through the G1/S cell cycle transition?

A

inactive hyperphosphorylated state

54
Q

when RB is hyperphosphorylated, what helps usher in the growth cycle?

A

the cyclins

55
Q

what do high levels of cyclins lead to?

A

hyperphosphorylation of RB and therefore inhibition of RB

56
Q

what happens if you have a loss of function of RB?

A

inappropriate activation of the cell cycle

57
Q

What is TP53 known as?

A

the guardian of the genome

58
Q

What is the most frequently mutated gene in human cancers?

A

TP53

59
Q

what cancers are classically positive for p53 mutations?

A

serous adenomas of the ovary

60
Q

what happens if there is a loss of function mutation in p53?

A

there will be no cell cycle arrest; no DNA repair; no senescence–> leads to malignant tumors

61
Q

We treat cancers with radiation and chemotherapy; these should work by inducing DNA damage, but what happens if the p53 is not working/mutated?

A

these tumors have a higher requirement for therapy

62
Q

What syndrome is associated with the germline TP53 mutation?

A

the Li-fraumeni syndrome

63
Q

what differentiates a germline mutation from a somatic mutation of p53?

A

younger age, diverse tumors, and a family history

64
Q

Besides TP53 and RB, what is another tumor suppressor gene we discussed?

A

APC: adenomatous polyposis coli

65
Q

germline mutations of APC are associated with what?

A

familial adenomatous polyposis- polyps develop early and extensively; colectomy in early adulthood or cancer is consider inevitable by 40 years of age

66
Q

What is special about germline mutations of APC?

A

they can occur de novo; so you cannot rely on a family history

67
Q

APC is a component of what signaling pathway?

A

WNT signaling pathway

68
Q

what is a major function of the APC protein?

A

to hold beta-catenin function in check

69
Q

what happens during WNT signaling?

A

it blocks the formation of the destruction complex- so beta-catenin can translocate from the cytoplasm to the nucleus and cause cell proliferation

70
Q

What happens if there is no APC/ mutated APC?

A

the destruction complex will not form- Beta-catenin will be able to signal cell proliferation

71
Q

What is the Warburg effect?

A

aerobic glycolysis

72
Q

What does autophagy in cancer allow?

A

doesn’t really help with growth, but it does help with survival

73
Q

How do cancer calls evade cell death?

A

they mutate/down regulate TP53 and they upregulate BCL-2

74
Q

What cell death pathway is important for killing cancer cells?

A

the intrinsic apoptotic pathway

75
Q

What is the opposite of fertility?

A

senescence

76
Q

What determines senescence?

A

telomeres

77
Q

What do stem cells have that prevent senescence?

A

telomerase- an enzyme that protects their telomeres

78
Q

What is the relationship with cancer and telomeres?

A

cancer also uses telomerase so that it will never be unable to replicate

79
Q

What are cancer stem cells?

A

like normal stem cells- they can self renew- give rise to heterogenous populations of daughter cells and proliferate extensively

80
Q

Solid tumors cannot grow more than a few mm unless they induce what?

A

angiogenesis

81
Q

Tumors set up their vascular system how?

A

by switching the tumor microenvironment from antiangiogenic to proangiogenic (an angiogenic switch)

82
Q

What is a proangiogenic factor?

A

HIF1

83
Q

what is the effect of HIF1?

A

VEGF and FGF

84
Q

how could you treat cancer when thinking about angiogenesis?

A

block VEGF- this would block the process of angiogenesis from occurring

85
Q

What are the three major steps in invasion and metastasis?

A

Getting through the basement membrane, getting into the vessels, getting out of the vessels

86
Q

How does the tumor initiate getting through the basement membrane?

A

there will be dissociation of tumor cells from each other

87
Q

how do the tumors cells dissociate from each other?

A

an epithelial to mesenchymal transition

88
Q

what are the hallmarks of epithelium?

A

in epithelial tumors, there are cadherins that are responsible for binding these cells to each other

89
Q

what occurs during the epithelial to mesenchymal transition?

A

the silencing of the E-cadherins

90
Q

After the epithelial to mesenchymal transition, what occurs next?

A

the degradation of the ECM basement membrane and connective tissue

91
Q

how do cancer cells accomplish the degradation of the ECM basement membrane and connective tissue?

A

with enzymes called Matrix Metalloproteinases

92
Q

what occurs after the degradation of the ECM basement membrane and connective tissue?

A

attachment and locomotion and invasion into the vessel

93
Q

What is the major cell responsible for the immune defense against tumors (tumor antigens)?

A

CD8+ Cytotoxic T lymphocytes

94
Q

Although we have CD8+ CTLs to help defend against tumor cells, some tumor cells have a way around this. What do they do?

A

antigen loss (failure to produce tumor antigen) and class I MHC-deficient tumor cells–> both lead to lack of recognition by t cell; tumor cells can also produce immunosuppressive proteins or expression of inhibitory cell surface proteins –> leads to inhibition of t cell activation

95
Q

What is an example of an immunosuppressive protein or expression of inhibitory cell surface proteins that tumor cells present to t cells to inhibit their activation?

A

PD-1 Ligand

96
Q

How can we treat tumor cells with the over expression of inhibitory cell surface proteins?

A

by using PD-1/PD-1 Ligand antibodies–> immune checkpoint inhibitors

97
Q

what does failure of the mismatch repair lead to?

A

unstable, persistent microsatellites in the DNA (microsatellite instability)

98
Q

What is Lynch syndrome?

A

germline loss of function mutations in a mismatch repair gene (remember that failure of the mismatch reapir leads to microsatellite instability)

99
Q

what do lynch syndromes include?

A

colorectal, stomach, pancreas, ovary and uterus, prostate gland, and the urinary tract

100
Q

How might DNA damage (like strand breaks) be repaired?

A

by the homologous recombination repair (HRR)

101
Q

what are the responsible genes for HRR?

A

BRCA-1 and BRCA2

102
Q

what is a treatment for acute promyelocytic leukemia?

A

all trans retinoic acid (since the PML/RAR complex doesn’t “like” retinoic acid anymore

103
Q

What are the 2 major mechanisms of epigentics?

A

DNA methylation and Histone modification

104
Q

what is the role of miRNA (micro RNA)?

A

to regulate RNA

105
Q

what happens if we down regulate miRNAs?

A

the translation of RNA will go up (and this includes oncogenic RNA)

106
Q

when is an example of when cancer utilizes down regulation of miRNAs?

A

with BCL-2s–> miRNAs are in part responsible for keeping BCL-2 in check; so when you have downregulation of the miRNAs for BCL2, you will have upregulation of the actual BCL2 molecule