Cancer Flashcards

1
Q

How does a primary tumor differ from a secondary tumor?

A

Primary: benign, restricted to site of origin, often treatable by surgery, requires blood supply.
Secondary: metastatic, able to degrade ECM, invasive.

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

Define: oncogene.
What are the two categories of oncogenes?

A

A gene containing a gain of function mutation or a mutation that acts in a dominant manner to contribute towards tumor formation.

  1. c-onc (mutated chromosome)
  2. v-onc (virally encoded)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Definition: tumor suppressor gene

A

A gene whose loss of function contributes to tumor formation - often acts recessively or in a haploinsufficient manner.

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

List the 10 hallmarks of cancer

A
  1. Self-sufficiency in growth signals
  2. Insensitivity to anti-growth signals
  3. Evading programmed cell death
  4. Immortality
  5. Sustained angiogenesis
  6. Tissue invasion and metastasis
  7. Deregulated metabolism
  8. Evading the immune system
    [9. Genome instability
  9. Inflammation]
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is the clonal expansion theory of tumor development?

A

If cell N gets a mutation, it will divide to give T1 cells that can divide more frequently than N. If T1 then gets another mutation, the resulting T2 cells will divide more frequently than T1. As this goes on, more cells gain mutations and have growth advantages over all other cells.

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

What are the requirements for mammalian cell proliferation?

A
  1. Nutrients e.g., amino acids
  2. Macromolecular nutrients e.g., insulin
  3. Attachment molecules (sometimes) e.g., fibronectin
  4. Serum i.e., PDGF, EGF
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What are the markers of cell cycle entry?

A
  1. S6 phosphorylation of the ribosome
  2. expression of TFs
  3. Cyclin D expression
  4. DNA synthesis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What is the Warburg effect, and how does it help to explain metabolic changes observed in cancer cells?

A

The Warburg effect is the reliance of cancer cells on aerobic glycolysis to generate energy, rather than oxidative phosphorylation. This increases glucose metabolism.

Perhaps this is because aerobic glycolysis generates an excess of metabolic intermediates that can be diverted into other pathways to make other molecules needed for growth.

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

How does angiogenesis relate to cancer cells? What can tumors do to increase this?

A

Angiogenesis is the process by which new blood vessels are formed from existing vessels.

In cancer, angiogenesis is a necessary step for tumor growth beyond a certain size, as tumors require a blood supply to obtain oxygen and nutrients necessary for their survival and proliferation. Tumors can secrete pro-angiogenic factors, such as vascular endothelial growth factor (VEGF), which stimulate the growth of new blood vessels. These new blood vessels can also provide an avenue for tumor cells to spread to other parts of the body through the bloodstream.

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

How can chronic inflammation cause cancer?

A

Chronic inflammation can lead to DNA damage, genomic instability, and alterations in cell signaling pathways, all of which can contribute to the development of cancer. Inflammatory cells can secrete cytokines and growth factors that can promote cell proliferation and survival, stimulate angiogenesis, and promote tissue remodeling, all of which can contribute to tumor growth and metastasis.

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

What is the epithelial-mesenchymal transition? What causes it? How does it relate to cancer?

A

EMT is the process by which epithelial cells acquire mesenchymal properties, such as the ability to migrate and invade surrounding tissues. This can be triggered by increases in TGFb which results in the repression of epithelial markers and activates mesenchymal markers.

EMT is heavily associated with tumor invasion and formation of secondary tumors. It can also contribute to resistance to chemotherapy.

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

Why are growth factors hard to purify?

A

They’re present in very low concentrations within the body. Tumors are rich sources of them, although they may be mutated versions.

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

What are the differences between the INK4 and Kip families of CDK inhibitors? How are they inhibited?

A

INK4 prevents cyclin binding
Kip inhibits the complex

Mitogens inhibit the actions of p21 and p27 by AKT/PKB phosphorylating them to promote cell-cycle progression.

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

List 4 mutation types by which an oncogenes can be activated. Give an example for each.

A
  1. Point mutation e.g., Ras
  2. Gene amplification/overexpression e.g., estrogen R
  3. Chromosomal translocation e.g., bcr-abl
  4. Association with viruses e.g., SV40 T antigen
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What are the 2 classes of tumor-forming retroviruses? What are their differences? How do they give rise to cancer? Give an example of each.

A

Class I:
- Rapid formation of tumors
- Tumors arise from multiple founder cells
- Rare
Encode constitutively active versions of proteins involved in GF signaling.
e.g., Avian leukosis virus (ALV) which captured the tyrosine-kinase Src

Class II:
- Slow formation of tumors
- Not all infected get tumors
- Tumors arise from a single founder cell
- Common
Integration of gene next to a cellular gene that’s transcriptionally active.
e.g., MHC leukemia virus

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

How might avian leukosis virus have picked up the Src gene?

A

ALV undergoes reverse transcription and integrates somewhere into the genome. This may be random or via hot spots, and at one time integrated into the scr gene. A fusion RNA was then transcribed that contained v-src and was packaged into the viral capsid. Viral capsids can’t tolerate change in length of information (they will lose some of the viral genome to replace it with src).

The particle will then infect another cell, but will now carry the v-src protein and have high levels of expression (strong promoters and enhancers) resulting in overexpression of the protein. These also mutate very rapidly and so the src will start to pick up mutations that lead to activation of the protein. These mutations will be selected for so that invasion of the next cell will result in overexpression of an activated src protein, resulting in a progressively more aggressive cancerous phenotype.

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

List the ways that oncogenes can be identified.

A
  1. Class I retroviruses: assay for the ability of the virus to transform cells in culture and look at the genome sequence.
  2. Class II retroviruses: identify site of viral insertion and clone sites either side.
  3. Isolation from tumor-derived DNA.
  4. Identify commonly occurring rearrangements and breakpoints in chromosomes.
  5. Identify regions of genome amplification.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

List 3 ways in which a chromosomal rearrangement can give rise to cancer.

A
  1. Movement of gene to be under a strong promoter.
  2. Changing of 3’UTR structure so that miRNA cannot bind to degrade it.
  3. Creation of a novel fusion protein.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What is the Philadelphia chromosome?

A

A fusion of DNA from chromosomes 22 and 9. This creates the BCR-ABL protein that displays high protein tyrosine kinase activity with unregulated expression that activates other proteins involved in the cell cycle. As a result, this chromosome is associated with leukemia.

20
Q

Give an example of an oncogene at each of the following levels of action:
- Growth factor
- Growth factor receptor
- Transducer of GF responses
- Transcription factors

How are each of these acting in an oncogenic manner?

A

GF:
- v-sis/c-sis
- overexpression results in autocrine signaling

GFR:
- EGF/PDGF/VEGF receptors
- constitutive activation or overexpression

Transducer:
- Ras (GTPase), Src
- Locked in GTP-bound state; lack of inhibitory domain

TF:
- myc, fos, jun
- overexpression, mutations in inhibitors

21
Q

How is c-myc associated with cancer development? What is the role of Mxd/Max?

A

The c-myc oncogene encode a TF that is normally tightly regulated with low levels of expression. This is deregulated in cancer cells, leading to increased proliferation.

In normal cells, Myc forms heterodimers with Max that leads to proliferation. In the absence of Myc, Max binds Mxd to repress transcription. This balance is destroyed in cancer.

22
Q

What is an E-box and how is it associated with oncogenic c-myc?

A

Enhancer boxes are consensus motifs that activate or repress transcription.

When c-myc binds these it activates gene expression, while Mxd binding represses gene expression. Hence, increasing myc expression alongside E box overexpression leads to an even greater amplification in transcription.

23
Q

How can signal inhibitors be used in cancer treatment? What are the benefits of selective targets over universal targets?

A

Signal inhibitors are a class of drugs that target specific cellular signaling pathways that are involved in cancer development and progression. By inhibiting these pathways, signal inhibitors can block the growth and survival of cancer cells and prevent the spread of the tumor.

  • Selective targets minimize the number of normal cells being affected.
24
Q

Explain Gleevec’s mechanism of action. How is it specific?

A

Gleevec inhibits Bcr-Abl, acting as a kinase inhibitor. Crystal structures suggest it locks the kinase in an inactive conformation.

This protein is unique to cancer cells, so Gleevec will only inhibit the fusion protein and not the WT proteins in normal cells.

(Herceptin is a monoclonal antibody against HER2 EGFR, seen in breast cancer).

25
Q

How is rapamycin used to treat cancer? What was its original use?

A

Rapamycin inhibits mTOR, a protein kinase that regulates cell growth and survival.

It was originally used as an immunosuppressor for organ transplants, but this means it can cause immunosuppression in cancer patients.

26
Q

What is Knudson’s two-hit hypothesis?

A

Explains the genetic basis of cancer development.

Cancer arises when two genetic events occur in the cell. The first hit is typically a germ-line mutation that’s present in all cells. The second hit is a somatic mutation that occurs in specific cells, leading to a loss of function in a TSG.

This explains why some cancers are hereditary and why cancer risk increases with age.

27
Q

List the ways a tumor suppressor gene can be identified.

A
  1. Identify genes in families with a propensity for certain tumor types.
  2. Clone genes that reverse cell transformation.
  3. Identify proteins that interact with immortalizing oncogenes.
28
Q

Give an example of a TSG associated with the following functions:
- Linking DNA damage to cell cycle arrest
- DNA repair
- Inhibition of cell cycle
- Antagonist of GF signaling

A
  1. p53
  2. BRCA2
  3. Rb, INK4
  4. PTEN
29
Q

What is the function of PTEN?

A

Inhibits PI3K/AKT/mTOR signaling by removing a phosphate group from PIP3 which is produced by PI3K.

30
Q

What is the function of pRB? How is it controlled?

A

Retinoblastoma protein is a TSG that regulates the G1/S checkpoint, forcing some cells to enter the G0 state. When active, it binds and inhibits E2F.

It’s highly controlled during the cell cycle through phosphorylation. Hyperphosphorylation prevents it from binding E2F, also triggering positive feedback by CDK2 (activated by E2F) to phosphorylate Rb even more.

31
Q

What are the stages to apoptosis?

A
  1. Initiation by internal or external stimuli
  2. Activation of caspases that results in execution of the cell
  3. Phagocytosis
  4. Degradation
32
Q

What are the specific proteins involved in apoptosis in C. elegans? How are these regulated?

A

CED-3: caspase
CED-4: pro-apoptotic protein
CED-9: inhibits CED-4 (anti-apoptotic)
EGL-1: inhibits CED-9 (pro-apoptotic)

33
Q

Describe the core cell death pathway in Drosophila. How is it regulated?

A
  1. Mitochondria releases pro-apoptotic proteins such as cytochrome c.
  2. Cytochrome c activates the APAF-1 homolog, DAPAF-1 to form the apoptosome.
  3. Apoptosome recruits and activates the initiator caspase Dronc.
  4. Dronc activates Drice, leading to apoptosis.

DIAPs regulate this pathway via their multiple BIR domains that allow for direct binding to initiator and effector caspases.

DIAPs can be inhibited by pro-apoptotic factors that target them for Ub-mediated degradation.

34
Q

How is apoptosis induced extrinsically in mammals? What are the two potential outcomes from this?

A
  1. Fas ligand binds Fas receptor and induces FasR oligomerization
  2. Recruits FADD (Fas-associated death domain)
  3. FADD activates caspase-8 (initiator)
  4. Caspase-8 activates caspase-3 (effector)

Alternatively, caspase-8 can activate Bid, a pro-apoptotic protein that promotes mitochondrial release of pro-apoptotic factors to amplify the apoptotic signal.

35
Q

Describe the BCL-2 family of proteins. What are their roles? Give examples.

A

The BCL-2 family of proteins is a group of regulatory proteins that play a central role in regulating programmed cell death, or apoptosis, in mammals. The family includes both pro-apoptotic and anti-apoptotic members that interact with each other to control the intrinsic apoptotic pathway.

Anti-apoptotic: Bcl-2, CED-9
Pro-apoptotic: Bax, Bak, EGL-1

36
Q

How are BCL-2 proteins involved in mitochondrial permeabilization?

A

Bax and Bak induce MOMP by acting as holins: bacteriophage generators of large membrane holes.

37
Q

What is autophagy? What is it triggered by? Give a basic overview of the process.

A

Autophagy is a cellular process in which cells break down and recycle their own components via autophagosome formation, in order to maintain cellular homeostasis and provide energy during times of nutrient scarcity or stress.

Triggers:
- nutrient scarcity
- lack of growth factors
- imbalances
- cellular damage

  1. Phagophore forms around the cytoplasmic material being targeted for degradation.
  2. Elongation of the phagophore to form the double-membraned autophagosome.
  3. Autophagosome fuses with a lysosome to form an autolysosome.
  4. The breakdown of materials releases molecules that can be recycled.
38
Q

What is the role of TORC1 in autophagy?

A

TORC1 integrates multiple cellular signals to inhibit autophagy. TORC1 is inhibited during nutrient deprivation or other forms of cellular stress.

39
Q

How is autophagy involved in early vs late stage cancers?

A

Early:
- Decreases genomic instability
- Promotes senescence
- Promotes oxidative phosphorylation
Acts as a tumor suppressor mechanism.

Late:
- Provides nutrients and energy during stress
- Promotes cell survival in harsh environments, such as hypoxic or acidic regions of tumors.
- Inhibits apoptosis
Promotes tumor growth and resistance to therapies. `

40
Q

What is the function of p53? How is it related to SV40 and cancer? Give an example of a human disease that can result from the inheritance of a mutant p53 gene.

A

p53 acts as a tumor suppressor by detecting DNA damage and initiating apoptosis and cell cycle arrest.

SV40 is a virus that can cause tumors in animals. It often does this by inactivating p53 via a large T antigen protein that binds p53’s DNA binding domain. This allows cells infected with SV40 to continue dividing and proliferating even if there’s DNA damage or other abnormalities.

Experiments have shown that p53 with a point mutation is more dangerous than a complete p53 knock-out. This is associated with Li-Fraumeni syndrome.

41
Q

How does p53 stop the cell cycle?

A

It induces the expression of a CDKI (p21-Kip1).

42
Q

Give the domain structure of the p53 protein. Which domain contains the most mutations associated with cancer?

A

Transactivation domain: binds transcriptional co-activators
SH3 domain: protein-protein interactions
***DNA BD: p53-DNA binding
Tetramerization domain
C-terminal regulatory: contains a nuclear localization signal

43
Q

How does MDM2 affect p53 function? How does p19ARF fit into this?

A

MDM2 functions as an E3 ubiquitin ligase that targets p53 for degradation. Disruption of this interaction by various stresses leads to p53 stabilization and stalling of the cell cycle.

MDM2 is also one of the first genes that p53 activates, forming feedback control.

p19ARF inhibits MDM2 activity, thus acting as a tumor suppressor and promoting p53 activation.

44
Q

How is p53 activated by the following:
- oncogenes
- DNA damage
- metabolic stress

A

Oncogenes:
p19ARF binds MDM2 and prevents it from destabilizing p53.

DNA damage:
phosphorylated by ATM protein kinase in response to double-stranded breaks

Metabolic stress:
glucose depletion activates AMPK which phosphorylates p53 to promote survival in the absence of glucose

45
Q

Give examples of genes regulated by p53 that are involved in cell cycle inhibition and apoptosis.

A

CCI:
1. p21 - inhibitor of G1 CDKs
2. 14.3.3.o - this binds and sequesters CDC25

Apoptosis:
1. Genes that generate ROS for mitochondrial degradation
2. PUMA and NoxA (requires acetylation of p53)
[PUMA inhibited BCL2 and NoxA activates Bax and Bak to form holins]

46
Q

What are nutlins? What other methods can be used to target p53 to treat cancer?

A

Small molecule inhibitors that target and inhibit the interaction between p53 and MDM2, hence promoting p53-induced apoptosis of cancer cells.

Other:
- reactivate WT p53
- restore binding of mutant p53
- targeting p53 downstream effectors e.g., BCL-2 inhibitors

47
Q

How do p53 and pRB interact with one another?

A
  1. p53-dependent G1/S arrest via p21 involves the inhibition of pRB phosphorylation from CDKs.
  2. Deregulated E2F activity induces p19ARF and p53-dependent apoptosis (pRB is a negative regulator of E2F)
  3. p16IN4A and p19ARF are encoded by alternative reading frames at the same locus:
    p16INK4A = blocks pRB phosphorylation
    p19ARF stabilizes p53