Cancer Flashcards

You may prefer our related Brainscape-certified flashcards:
1
Q

What are the broad categories of cancer and their nomenclature?

A

Benign (tissue + oma): neither invaded nor metastasised
- Can be encompassed in a capsule

Malignant (carcinoma): capable of metastasis and invading into other tissues
- Metastasis = formation of new tumour colonies (major death causing (>90%))

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

What are the ways that cancer can impact normal function?

A
  • Pressure: pituitary gland/medulla, depressing vital function; blockage of vessels/airways/digestion
  • Erosion of bone = fractures
  • Epithelial ulceration: bleeding from colorectal tumours
  • Competition with normal function: failure of bone marrow
  • Metabolic changes: Cachexia = ‘general wasting’ and production of tumour specific products
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

How can cancer be detected?

A

Clinical symptoms:
- Cachexia
- Indirect: bleeding, blocked colon, pain
- Palpation of breast tissue

Clinical tests:
- X-ray
- Screening (cervical cancer)

Tumour mapping:
- Single cell sequencing
- Spatial transcriptomics (including ssRNAseq): to determine which cell types are where in tumour

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

What are mutagens and how do they increase cancer risk?

A

Initiators which increase genetic instability: directly damaging DNA and increasing mutation rate (higher transformation risk)

Examples:
- UV light (causing thymidine dimers)
- Smoking (benzopyrene, β-naphthylamine)
- Environmental carcinogens (weedkiller; agent orange)
- Toxins produced by infectious agents (aspergillus aflatoxin activated by oxidases)
- Interruption of gene by integrating virus (rare)
- NMU (alkylating: AT-GC mutations)

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

How do components of cigarettes increase transformation risk?

A

Benzopyrene (contains benzene rings):
1. Binds Ahr causing CYP1A1 translation
2. Addition of adducts to guanines caused
3. If not removed by NER then A ➡ T visa versa likely

β-naphthylamine:
1. Normally glycosylated to clear in urine
2. Reactivated by human enzyme glucaronidase
3. Adducts formed

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

Give some specific environmental carcinogens. What are the mechanisms by which they cause disease?

A

Agent Orange (containing TCDD):
- Causes translation of CYP1A1 and increased ROS
- Proliferation and increased DNA damage (both initiator and promotor)

NMU:
- Alkylating agent
- Transfers methyl group causing AT ➡ GC transitions

Aflatoxin produced by aspergillus:
- Alkylating agent
- Adduct formation causing disheveled DNA and interrupting repair mechanisms

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

What are promotors and how do they increase transformation risk?

A

Change cell behaviour, increasing the probability of a mutation of an individual cell or a population of cells (or increasing population size):
- Carcinogens causing inflammation (asbestos, TPA)
- Microbes causing inflammation (aspergillus, TB)
- Infection with a transforming pathogen (HPV, Rous’ SV, EBV)

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

Detail the ways that infection can cause transformation of a cell; give examples:

A

Gene modification: Proto-oncogene protomors: HPV, HSV, EBV
- Carry proto-oncogenes (H-ras; K-ras) with a range of actions
- Discovered by Rous’ experiments using filtered sarcoma homogenate injected into chicken wing – sarcoma developed weeks later

Change cell behaviour (create environment to facilitate survival)
- Loss of contact inhibition (HPV)
- High growth density/growth without attachment
- Immortality
- Growth factor independence/resistance to signals causing stasis of growth/death.

Change to behaviour of cell population:
- Malaria causing B cell proliferation/activation (synergy between EBV and malaria)

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

What are gatekeeper and caretaker mutations?

A

Gatekeeper = involved in growth/division:
- Oncogenes/tumour suppressor genes
- E.g. pRb, p53

Caretaker = involved in maintenance of genome:
- SSBR, NER, BER, NHEJ, MMR

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

What is a classic tumour suppressor gene? Describe an example.

A

Recessive tumour suppressor gene.

Retinoblastoma gene (for Rb protein (pRb)):
- pRb normally inhibits DNA replication at G1/S checkpoint
- When cyclin D builds up it activated cdk4/6
- These phosphorylate pRb, inhibiting it.
Mutations in RB1 gene (inactivating it) or genes overactivating cdk4/cyclin D can cause overphosphorylation (and cancer)

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

What is an abnormal tumour suppressor gene? Describe an example.

A

Have significant effect on cell when only one copy is mutated:

E.g. p53 TF
- Functions as a tetramer, hence most complexes contain some faulty protein
- Mutated copies have reduced ability to bind to DNA
- Reduces ability to induce apoptosis by activating Bcl2 protein, so damaged cells continue to reproduce

Can be hereditary E.g Familial adenomatous polyposis (APC mutation)

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

What are the types of genetic instability? Give examples of each:

A

Sequence instability = mutation:
- Failure to repair DNA damage E.g. MLH1 mutation causing MMR failure)
- Errors in mitosis E.g. pol epsilon mutation causing defective proof reading domain (high error rate)

Chromosomal instability:
- Failure to repair damage: BRCA1/2 mutations
- Errors in replication such as faulty spindle attachments/kinetochores: aneuploidy results

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

What are the different DNA repair mechanisms? What is BRCA involved in?

A

Excision repair: Nucleotide (NER)/base (BER):
- Deal with chemically modified bases E.g. UV-induced pyrimidine dimers

Mismatch repair (MMR): deals with mismatched bases or small loops:
- Caused by polymerase slip during replication of repeats ➡ deletion/insertion on one strand
- Inability to fix loops leads to shrinkage/expansion of short repeats (=microsatellites)
- MMR mutation in hereditary Lynch syndrome (mutation in MLH1/2)

Repair of strand breaks:
- Single stand break repair (SSBR)
- Homologous recombination (HR): relies on other copy (E.g. sister chromatid) as a template = faithful repair
- Non-homologous end-joining (NHEJ): results in loss of sequence fidelity (no template) often occurs at telomeres

BRCA1/2 are involved in HR causing breast cancer if mutated (can be hereditary).

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

What is the Vogelstein model of colorectal cancer development?

A

Suggest tumorigenesis is a sequence of mutations which together cause cancer (not one).
- Order and identity of acquisition affects speed of cancer formation.

Sequence of events:
1. Normal epithelium
2. Hyperplastic epithelium (due to APC mutation)
3. Adenoma due to DNA hypomethylation (epigenetic changes)
4. Carcinoma formation due to accumulation of mutations (activation of K-ras, loss of TSGs, loss of p53)
5. Invasion/metastasis

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

What is the modern interpretation of the Vogelstein model? Give an exemplar sequence:

A
  1. Epigenetic deregulation (E.g. due to infection)
  2. Disruption of onco/TSGs
  3. Transcriptional dysregulation due to mutation (repair cannot keep up with mutation rate)
  4. Metabolic deregulation
  5. Inflammation and microenvironment formed (immune suppressing and metabolically greedy with vascular growth)

E.g. Removal of proliferation brake (β-catenin) ➡ genetic instability (ras) ➡ immune system evasion mutation (e.g. TGF-β) ➡ removal of apoptosis activator (p53) ➡ carcinoma

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

Which cancers are hereditary/show a strong predisposition trend?

A

Hereditary Colon cancer:
- Adenomatous polyposis coli (APC): APC mutation affecting growth control = 80% of sporadic colorectal cancer
- Lynch syndrome: MLH1 mutation causing genetic instability in MMR gene (non polyposis colon cancer)

Hereditary predisposition to breast cancer:
- E.g. BRCA1/2 gene mutations causing problems in HR strand repair pathway (50-80% risk of breast cancer (compared to 15%)
- Even patients with restored BRCA2 the cancer is still lethal to patient (not purely due to mutation, but also due to genetic instability)

Retinoblastoma – RB1 mutation affecting growth control by allowing free entry to S phase causing rare childhood tumour.

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

Describe the Wnt signalling pathway:

A
  • Wnt binding (to frizzled receptor) inhibits degradation of β-catenin
  • β-cahenin (with TCF4) becomes active transcription factor
  • Promotes proliferation through cell cycle entry (cyclin D, VEGF production)

Mutations (E.g. in wnt receptors) can lead to inappropriate inhibition of degradation

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

Describe the GF tyrosine kinase signalling pathway. How can mutations cause immortality?

A

Epidermal GF binding (to EGF receptor):
- RAS activation = promotes proliferation
- PIP2 to 3 conversion = PIP3 inhibits apoptosis

PIP3 normally broken down by PTEN
- Mutations in PTEN cause cancer
- Mutations can occur in all steps of the pathway

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

Describe the TGF-β signalling pathway:

A
  1. TGF-β binds TGF-β receptor
  2. Activation of SMADs (as TFs)
  3. Inhibition of proliferation (tumour suppressor)

Mutations cause release of proliferation inhibition and correlated with metastasis ability.

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

What is a differentiation block? Give two examples of cancers resulting from this.

A

Growth control loss causing build up of progenitor cells with fewer specialised differentiated cells.

Chronic myeloid leukaemia = failure of normal haematopoiesis:
- Too many granulocytes causes loss of neutrophils and monocytes
- Chronic lymphocytic leukaemia = mature B cell differentiation block causing plasma cell proliferation
- Symptoms depend on cell line affected

Expansion of crypts in gut due to APC mutation:
- Paneth cells over-proliferate (>rate of loss of specialised cells)
- Leads to adenomas of non-differentiated cells (reduced absorption)
- Due to β-catenin build up

21
Q

Give some general hallmarks of cancer cells. How are these similar to stem cells?

A
  • Differentiation blocks
  • Immortality (resistance to telomere shortening)
  • Resistance to apoptosis
  • Metabolic changes (Warburg effect; metabolic reprogramming)
  • Angiogenesis
  • Metastasis ability

Share stem cell properties: self-renewal; multipotency; plasticity; niche dependence

22
Q

Describe the apoptosis pathway. How might a cancer cell develop resistance to apoptosis?

A

Damage to p53 gene (a master regulator of stress signalling):
- Inputs: DNA damage; ROS etc…
- Outputs: apoptosis pathway and p21 activation to halt cell cycle (when fixable)

Apoptosis pathway:
1. Activation of p53 gene (inhibits Bcl2 so bax gene disinhibited)
2. Bax translocation and oligomerisation in mt membrane
3. Mitochondrial outer membrane permeabilization (MOMP) and Cyt C leakage
4. Caspase activation
5. Apoptosis

23
Q

Why are cells not immortal? What is the Hayflick limit?

A

Telomere shortening causing cell cycle arrest
- Telomeres = TTAGG repeats at chromosome ends (forming protective D-loop)
- Shorten by 100bps per division
- Must be re-synthesised by telomerase

Hayflick limit: is theoretical maximum of divisions:
- Foetal = 50-70; adult = 30-50; cancer = ∞
- Shown by HeLa cells
- Mice have very long telomeres – causes problems for modelling

24
Q

How might senescence features be gained (resultin gin immortality)?

A

By sequence instability:
- Telomerase reverse transcriptase (TERT) promoter mutation

By chromosomal instability:
- TERT promotor rearrangement (E.g. super enhancer chr 5 mutations causing neuroblastoma

25
Q

How do cancer cells manipulate their metabolic microenvironment to increase survival? What is the Warburg effect?

A

Warburg Effect: tumours preferentially use fermentation (aerobic glycolysis):
- Can uptake huge amount of glucose
- Satisfy biosynthetic demands of increased proliferation

Metabolic reprogramming due to mutations in metabolic genes or signalling pathways
- E.g. MYC mutations (master gene for may metabolic pathways)
- E.g. succinate dehydrogenase (SDH) mutations causing abnormally high [succinate] ➡ leads to multiple enzyme dysregulation and epigenetic markers
- Gain of function mutations start producing oncometabolites (rather than intended product) E.g. R-2-HG which act as useful biomarkers of cancer

26
Q

How do tumours maintain a sufficient blood supply?

A

Angiogenesis – tumours need increased blood supply
- Angiogenic switch may occur pre-malignancy
- Secrete angiogenic factors: VEGF; FGF1/2; IL-8
- Reduce inhibitory factors: Angiostatin; IFN α/β

27
Q

What is the metastatic insufficiency principle? What does it suggest?

A

The discordance between the number of metastasising cells and detection of clinical metastases:
- Circulating tumour cells (CTCs) seen in tissues they do not usually arise in.
- Seed and soil compatibility factors matter (does organ site support or suppress growth of cell type?)
- Mechanical factors relevant (circulation patterns)

28
Q

What are the two main models for the tumour initiation ability of cancer cell offspring?

A

Stochastic model:
- Stem cell has ability to become cancerous
- ALL differentiated daughter cells have ability to become cancerous

Cancer stem cell model:
- Stem cell itself not cancerous
- Only some differentiated cell lines are cancerous (cancer stem cell (CSC) is later in differentiation pathway)
- Evidence: certain leukaemia malignancies have CSCs which share phenotype with tissue specific haematopoietic SCs (express CD34 but NOT CD38 (differentiation marker) suggests like stem cell still)
- Evidence: ‘bottom up transformation’ in APC where stem cells express Lgr5

29
Q

What is the difference between ‘top down’ and ‘bottom up’ transformation? How can

A

Bottom up:
- APC mutation causing expansion of stem cell population
- Neighbouring cells can propagate conditions: Wnt producing Paneth cell next to Lgr5 Wnt responsive stem cell = +ve feedback

Top down transformation:
- Inflammation can allow de-differentiation of cells (increasing probability for CSC)
- E.g. KRAS mutation ➡ NfκB with β-catenin build up due to APC mutation ➡ stem cell signature on committed progenitor cell

30
Q

What is evidence that the immune system is involved in cancer development? How does the immune system change the microenvironment around a tumour?

A

Evidence for immune system involvement:
- Mutations necessary but not sufficient for cancer (50% of normal lung carries KRAS or EGFR mutation but not cancerous)
- Immune system may allow this

Microenvironment changes during inflammation:
- Molecules driving ‘stemness’ produced by immune cells: EGFR mutant lung cells exposed to IL1β ➡ stemness traits induced ➡ becomes CSC ➡ lung cancer

Evidence:
- IL1β antibody given prevents cancer development
- Paracrine signalling involved: basal cell carcinomas only grew at secondary site if stroma transplanted also.

31
Q

What are the main methods of treating cancer?

A
  • Surgery (mainly removal of primary tumours): results in death of normal cells but may not matter for life (mastectomy)
  • Traditional cytotoxic drugs
  • Exploiting cancer cell specific properties (defects in cell checkpoints and high genetic instability)
  • Targeted agents (kinase inhibitors; monoclonal antibodies; bicycles)
  • Upregulating the immune system
32
Q

Give examples of cytotoxic drug mechanisms targeting growth control:

A

Target oncogenes – inhibition of hyperactive tyrosine kinases (require ATP to function):
- Crizotinib treating T-cell lymphoma: binds to mutated NMP-ALK TK transmembrane protein (has become intracellular and permanently activated by loss of TM domain due to chromosome translocation)

Target downstream proteins (E.g. Ras, MAP kinase, PIP3K…)
- Vemurafenib inhibits mutant B-RAF hyperactive TK: stops MAP kinase activation.

Inhibit general growth signals:
- Monoclonal antibodies against HER2 (for amplified breast cancer)
- Anti-EGFR for colorectal cancer (resistance likely)
- Specific to organ: E.g. oestrogen blocked for breast cancer

33
Q

Give examples of cytotoxic drug mechanisms targeting DNA synthesis:

A
  • Alkylate bases
  • Intercalate non-covalently between bases of DNA helix (crosslink strands)
  • Inhibition of topoisomerase
  • Toxic analogues of bases – 5-fluoro-uracil
34
Q

Give examples of cytotoxic drug mechanisms targeting mitosis:

A

Disrupt microtubules, including spindle:
- Vinca alkaloids: bind tubulin preventing polymerisation
- Taxanes (Taxol): induces stable microtubule bundles (interrupts dynamic instability)
- Armed monoclonal antibody brentuximab – binds CD30 and toxin MMAE is endocytosed (binds tubulin disrupting mitosis)

35
Q

Give examples of cytotoxic drug mechanisms increasing genetic instability of cancer cell (until they cannot function):

A

Cis-platin: creates DNA cross-links
- Normally repaired by HR
- If cancer is BRCA2 mutant then causes increased genetic instability (cancer cells = more vulnerable)
- Great success for testicular cancer treatment

Block alternative DNA repair mechanisms (so cancer cells cannot cope with everyday damage) – inducing apoptosis:
- Targeting SSBR using Poly ADP ribose polymerase (PARP) inhibitors
- Meaning ssbreaks become dsbreaks

36
Q

What were the Waldmann experiments? What did they show?

A

Showed the effect of cancer cells being sensitive to irradiation (more than normal):
- p21 defective cells sensitive to irradiation
- WT were not

37
Q

Give an example of how can viruses be used to identify cancer cells:

A

Selective viral replication in cancer cells with p53 mutations:
- Engineered adenovirus H101 normally inhibits p53 using EB1
- EB1 protein removed
- Therefore viral replication only occurs in p53 defective cancer cell
- If virus carries toxin this allows selective delivery to cancer cells

38
Q

How can natural defences be boosted to treat cancer?

A

Increasing CD8 cell toxicity:
- CD8 have effect (thymectomy patients have higher incidence of cancer)
- Blocking inhibitors of CD8: such as CTLA-4 and PDL-1 antibody
- Patients with more mutations in tumour respond better

Target specific cell receptor
- E.g. CD20 on B cell surface of lymphoma cells inducing complement and antibody cell cytotoxicity
- Rituximab targets CD20

Bispecific T cell Engagers (BiTEs):
- Bring together T cell and cancer cells
- Recognition of CD3 on T cell with CD20 on B cell lymphoma cells

Chimeric Antigen Receptor T cells (CART)
- CD19 on B cells
- Effective but very expensive with considerable side-effects (cytokine storms particularly IL-6) and poor tumour infiltration

39
Q

How might resistance to a cancer therapy develop?

A
  • Multi-drug resistance due to upregulation of efflux pumps (similar to bacterial mechanism)
  • Reversion mutations after a frameshift which restores activity
  • Point mutations preventing binding of drugs to their target site (E.g. mutation in ATP binding pocket of TK stopping crizotinib binding due to increased steric hindrance)

Emergence of variants in RAS rendering upstream treatments moot
- Such as Anti-EGFR; BRAF inhibitor

40
Q

How does EBV cause cancer? How can malaria and EBV work together to cause cancer?

A

EBV causes high proliferation rate:
- c-myc upregulated due to oncogene rearrangement
- Upregulates cyclins
- Also causes anti-apoptotic function

Malaria causes B-cell proliferation which increases chance of oncogene rearrangement.

41
Q

How does KSC cause cancer?

A

Lymphatic sinus endothelial cells (LSECs) infected = vasculature tumour:
- LANA-1 protein highly expressed
- Acts as oncogene to maintain latency by inhibiting signalling pathways (PIP3/JAK-STAT…)
- Induces proliferation (via p53)

42
Q

How does UV light lead to positive feedback of mutations in keratinocytes?

A

Healthy:T-cells and NK cells remove keratinocytes with abnormal antigens

UV damages keratinocyte DNA :
- Induces IL-10 secretion
- IL-10 inhibits function of antigen-presenting cells (E.g. Langerhans cells/T-cells)
- Future UV caused mutations unnoticed

43
Q

What is the difference between a lymphoma and a leukaemia?

A

Leukaemia = liquid (of blood/BM)

Lymphoma = solid or nodal proliferation of lymphocytes

44
Q

Why is a immunohistochemical positive test for CD10 and Bcl-2 on one cell a problem? Or both light chain κ and λ?

A

Lymphocytes should NEVER express both.
- CD10 expressed on immature B cells (those cycling between dark and light zones following antigen exposure)
- Bcl-2 inhibited in mutated B cells to cause apoptosis (select against)
- Bcl-2 expressing cells will not undergo apoptosis (even if damaged)

No cell should be both light chain κ and λ (only produce one)

45
Q

How might a PCR study help to identify a lymphoma from a benign mass?

A

Clonality study: have all the cells gone through clonal amplification?

  • PCR should show different lengths of chains (due to somatic hypermutation and sequence diversity) in Gaussian distribution.
  • Normal response needs distribution of chains
  • Lymphoma shows a single spike (monoclonal) = abnormal
46
Q

What are the three ‘principles’ of lymphomagenesis? Name an example of each.

A

Antigen stimulation:
- Chronic infection causing clonal expansion leading to lymphoma
- H.Pylori (CagA/VacA)/chlamydia

Translocation:
- Mistake due to VDJ recombination and somatic hypermutation
- Burkitt’s lymphoma
- Follicular lymphoma

Point mutation:
- Hairy cell leukaemia

47
Q

What are the causes Follicular lymphoma and Hairy cell leukaemia?

A

Follicular lymphoma
- chr 14–>18
- Translocating MLL2 meaning CD10 and bcl-2 expressed

Hairy cell leukaemia:
- BRAF mutation (GF/ras activation no longer needed to cause proliferation)
- Vemurafenib inhibits BRAF (treats well)

48
Q

How can Immunotherapy be used to treat cancer? What are the problems with this?

A

Repressing immune checkpoints:
- CTLA-4 antibody: stops CD86 binding to CTLA-4 causing repression of T cell activity
- Anti PD-1 (programmed death) ligand to reduce induced death of T cells following activation
- Can cause autoimmune problems

Adoptive T-cell therapies:
- CAR-T cells = antigen domain with receptor for cancer cell
- Good for blood cancers