Cancer Flashcards

1
Q

Why is cancer a genetic disease?

A
  • There are a number of environmental factors that cause cancer, but these act through genetic modifications e.g. activation of oncogenes
  • Additionally 5-10% of cancers are due to inherited mutations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What proportion of cancers are attributable to modifiable risk factors?

A
  • 30-40% of cancers are preventable due to modifiable variables such as low fruit/veg intake, lack of physical activity, high BMI, alcohol and tobacco use
  • Smoking is the most important risk factor in terms of cancer mortality
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What is the trend in cancer incidence and mortality in developed countries?

A
  • The incidence of cancer is rising (due to aging population) and the mortality is decreasing
  • This combines to increase the prevalence
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What proportion of cancer deaths occur in low and middle income countries?

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

What is attributable vs avoidable burden?

A
  • The attributable burden can be estimated if the past prevalence of population to the risk factor and their relative risk of association with a cancer disease are known
  • The avoidable burden is the burden of disease averted as a result in exposure reduction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What are the main risk factors for cancer?

A
  1. Tobacco smoking
    - leading cause of cancer death worldwide
  2. Alcohol:
    - 22% of mouth and oropharynx cancers in men and 6-20% of breast cancers in women
  3. Diet, overweight and obesity:
    - Linked with cancers of oesophagus, colo–rectum, breast (19%), endometrium (40%) and kidney
  4. Hepatitis B and C:
    - Cause of hepatocellular cancers
  5. Human papillomavirus:
    - Cause of all cervical cancer, 90% of anal cancer and 40% of cancers of the external genitals
  6. Environmental pollution:
    - Pollution of air, water and soil with carcinogenic chemicals
  7. Occupational carcinogens
    e. g. asbestos
  8. Radiation exposure:
    - X-rays and ionizing radiation
    - UV radiation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What are the 4 strategies to deal with cancer?

A
  1. Prevention:
    30-40% of cancer deaths are preventable
  2. Screening:
    - aims to detect ‘pre-cancerous stage’
    - should meet WHO criteria for screening program
  3. Treatment
  4. Pallative care
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What are the features of a good preventative campaign?

What are some examples?

A
  1. Multi-dimensional approach
  2. Ingenuity
  3. Communication
  4. Patience
  5. Sustainability
  6. Creation of new norms
  7. Persistence

Examples:

  • Sun exposure campaigns
  • Preventing sale of alcohol to minors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What are the WHO criteria for introducing a screening program?

A
  1. Condition should be important heath problem
  2. There should be a recognisable latent or early symptomatic stage
  3. The natural history of the disease should be adequately understood
  4. There should be an accepted treatment for the disease
  5. The test should be acceptable to population
  6. There should be an agreed upon policy as whom to treat as patients
  7. Facilities for diagnosis and treatment should be available
  8. The cost of screening should be economically balanced in relation to possible expenditure on medical care
  9. The screening should be a continuous project
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What are the 6 hallmarks of cancer?

A
  1. Resisting cell death
  2. Sustaining proliferative signalling
  3. Evading growth suppresors
  4. Activating invasion and metastasis
  5. Enabling replicative immortality
  6. Inducing angiogenesis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

How does resisting cell death lead to cancer?

A
  • In normal cells a cellular stressor such as DNA damage will activate tumour suppressor programs e.g. p53 that will initiate the apoptosis (controlled cell death) of the damaged cell
  • In cancer cells a cellular stressors such as DNA damage may occur, but the tumour suppressor gene e.g. p53 may be mutated, or antiapoptotic proteins such as Bcl2 may be overexpressed. This means that apoptosis is not initiated and the cancer cell survives and is allowed to accumulate mutations
    e. g. B-cell lymphomas: a chromosomal translocation resulting in an immunoglobulin promoter being next to the Bcl1 gene leads to the cells becoming resistant to apoptosis and contributes to them becoming malignant
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

How does sustained proliferative signalling lead to cancer?

A
  • In normal cells growth and division are tightly constrained by both extrinsic factors such a growth factor and cytokine stimulation and hormone dependency and cell intrinsic factors such as intracellular signalling pathways and cell cycle regulators
  • In many cancerous cells these proliferative signalling pathways are abberantly upregulated

e.g. B-cell sustained proliferate signalling in lymphomas: B-cells should only proliferate when they receive antigen-dependent proliferative signals. Mutations in B-cell receptor signalling leads to the constitutive activation of the BCR and thus abberant proliferation in B-cell lymphomas

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

How does evading growth suppressors lead to cancer?

A
  • Tumour suppressor programs are commonly inactivated in cancer
    e. g. P53:
  • Integrates signals from cellular stressors to induce cell growth and arrest and allow DNA repair, if DNA cannot be repaired it induces cell death
    e. g. Rb (retinoblastoma protein)
  • Central regulator of cell cycle (regulates the G1 checkpoint which is an important barrier to abnormal proliferation)
  • Inhibited by phosphorylation which is done by cyclin-CDK complexes

e. g. Mantle cell lymphoma:
- Immunoglobulin promoter is translocated next to cyclin D1 (due to t(11;14))
- This leads to overexpression of cyclin D1 which inhibits the action of Rb and thus leads to unconstrained cell proliferation (no G1 checkpoint)

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

How does enabling replicative immortality lead to cancer?

A
  • Normal cells have a replicative mortality ‘Hayflick limit’ that is the number of times the cell will divide before cell division stops ‘replicative senescence’
  • The replicative mortality is due to the loss of genetic material on the lagging side, this is capped with telomeres, which are synthesised by TERT enzymes
  • The TERT enzyme is tightly controlled so the telomeric ends will only be lengthened appropriately (within the Hayflick limit)
  • TERT can become activated in cancers via mutation of its promoter region which leads to the upregulation of the enzyme and thus the overcoming of the Hayflick limit making the cell have replicative immortality
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What are the cancer enablers/emerging hallmarks?

A

Emerging hallmarks:

  1. Deregulating cellular energetics
  2. Avoiding immune destruction

Enabling characteristics:

  1. Genome instability and mutation
  2. Tumour-promoting inflammation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

How does deregulating cellular energetics lead to cancer?

A
  • The deregulation of cellular energetics in cancer cells is known as the Warburg effect:
  • Tumour cells are more likely to undergo glycolysis (anaerobic respiration) if in the presence of sufficient amounts of oxygen

e. g. IDH2 mutation in T-cell lymphoma:
- IDH2 is an enzyme that convert isocitrate into a-ketogluterae in the CAC
- When IDH2 is mutated the CAC cycle is broken and this causes the cancer cell to undergo glycolysis even when there is sufficient methylation

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

How does avoiding immune destruction lead to cancer?

A

Cancer cells are able to avoid immune destruction by:

  • Downregulating MHC expression and reducing costimulatory molecule expression
  • Expressing inhibitory ligands (e.g. PDL1 and PDL2) and immunosuppressive cytokines to exhaust host T-cells

e.g. overexpression of PDL1 in Hodgkins Lymphoma

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

How does genome instability and mutation enable cancer?

A

Mechanisms:

  • Inactivation of tumour suppressor genes regulating DNA repair
  • Telomere loss leading to karyotypic instability
  • Hijacking of physiological mutation prone states e.g. B-cell somatic hypermutation
  • Gives intra-clonal heterogeneity
  • Iatrogenic changes can be evoked by cytotoxic therapy/radiotherapy

Consequences:

  • Tumour clonal proliferation
  • Acquisition of a more aggressive disease phenotype
  • Resistance to therapy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

How does venetoclax therapy work?

A
  • Venetoclax is a Bcl-2 antagonist that binds to the Bcl-2 and allows apoptosis to occur
  • It is able to overcome the ‘evasion of cell death’ hallmark of cancer
  • Used to treat CLL
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

How does Ibrutinib therapy work?

A
  • Ibrutinib is a drug (kinase inhibitor) that targets B-cell receptor signalling
  • Ibrutinib initially pushes lymphocutes out of the lymph nodes where they eventually die
  • When combined with ventoclax, an excellent treatment for relapsed refractory CLL
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What is the difference between instrinsic apoptotic stimuli and extrinsic apoptotic stimuli?

A

Intrinsic apoptotic stimuli:

  • DNA damage
  • Via a mitochondrial pathway
  • When DNA damage is induced, p53 is upregulated which promotes the permeabilisation of the mitochondrial membrane and apoptosis

Extrinsic apoptotic stimuli:

  • Immune mediated deletion
  • Via a death receptor pathway which activates intrinsic and executioner pathways
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Describe the extrinsic apoptotic pathway:

A
  • When cytotoxic lymphocytes e.g. NK cells and CD8+ T cells recognise hazerdous antigens on cells an immune synapse is formed and they will release granzyme and perforin from cytotoxic granules
  • The perforin creates holes in the target cell membrane
  • Granzymes activate caspases to induce apoptosis
  • Involves death receptors: initiation of cell death by Fas ligand binding to Fas receptor which activates caspase 8
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

How do tumours develop resistance against extrinsic apoptosis?

A
  1. Downregulation of Fas receptor (death receptor)
  2. Decoy receptors expressed
  3. Silencing (e.g. methylation) of genes required to transduce death signals
  4. Overespression of negative regulators of death receptor
  5. Overexpression of inhibitor of apoptosis proteins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Describe the intrinsic apoptotic pathway:

A
  • p53 is one of the most important regulators of the intrinsic apoptotic pathway
  • p53 works by protein-protein interactions so it is difficult to devise ways to restore its function
  • p53 is upregulated in the event of DNA mutation/cellular stressors and it upregulates proapoptotic proteins to induce apoptosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Describe apoptosis and co-operative oncogenesis:

A
  • In a normal cell, c-Myc (oncogene) activation would cause the upregulation of pro-apoptotic proteins such as Puma and Noxa
  • c-Myc promotes genome instability and then cooperates with the suppression of tumour suppressor genes such as a mutation in p53 or an overexpression of Bcl2 to form cancer
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What is oncogene addicition?

A
  • Oncogene addiction is when the activation of an oncogene is necessary for the survival and growth of a tumour
    e. g. A tumour that is Myc-driven is Myc oncogene addicted can be targeted with Myc inhibitors
    e. g. CML has an oncogene addiction to BCR-Abl, so switching off this oncogene cures the diseases
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

What is epigenetic regulation?

A
  • Epigenetic regulation refers to the differential expression of genes due to changes that do not affect the DNA sequence
    e. g. chromatin remodelling (histone modification) and DNA methylation
  • Epigenetic mechanisms can be used to reverse cancer phenotypes, e.g. epigenetic silencing of a tumour suppressor gene can be reversed
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Describe DNA methylation and its effect on gene expression:

A
  • DNA methylation (especially at gene promoters) usually occurs on CpG motifs and silences genes by preventing the binding of transcription factors
  • DNA methylation is due to the action of DNA methyltransferases
  • These DNA methyltransferases can be inhibited therapeutically inhibited using DNA hypomethylating agents such as 5-Azacitidine which traps DNMT1 and causes it to be degraded
  • These drugs have been used off-label to treat T-cell lymphoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

How are histones modified and how does this affect gene transcription?

A
  • Histone tails can undergo acetylation, methylation and phosphoryation
  • Overexpression of enzymes that cause these modifications are commonly seen in malignancies
  • Histone deacetylase (HDAC) inhibitors allow global acetylation and reading of genes to occur which can upregulate apoptosis
  • Only really effective in haematologic malignancies
  • BET-bromodomain (reader) inhibitors mimic acetylated lysine residues e.g. JQ1
  • Bromodomain-containing proteins such as BDR4 are known to be of importance in midline carcinoma so inhibiting the reading of these genes can help treat cancers
  • cMyc is an example of a gene regulated by BRD4, using a BET-bromodomain (reader) inhibitor such as JQ1 can treat malignancies driven by abberant cMyc
  • However tumours can develop JQ1 resistance via upregulation of Bcl-2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What are the primary roles of the immune system against tumours?

A
  1. Elimination or suppression of viral infections
    - E.g. HPV elimination prevents cervical cancer
  2. Elimination of pathogen and resolution of inflammation
  3. Immune surveillance:
    - Elimination of tumour cells via detection of tumour specific antigens or molecules induced by cellular stress
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What are the tumour/host immunity dynamics?

A
  • When normal cells become transformed into immortalised cancerous cells via activation of oncogenes e.g. cMyc, Ras or loss of tumour suppressors e.g. p53, Rb- the cell becomes stressed and immunogenic
  • These stressed cells can then be:
  1. Eliminated:
    - The innate and adaptive immune system may remove the cancerous cells before they can cause a malignancy
  2. Equilibrium:
    - The lymphocytes may be able to remove most of the tumour cells but some may survive in a quiescent state due to immunological control
    - The genetic instability of the surviving cells means they may be able to develop more mechanisms the subvert/inhibit the normal immune response
  3. Escape:
    - The tumour cells are able to escape the immune system and develop into clinically apparent cancer
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What is clinical evidence of immunosurveillance?

A

Mouse studies:

  • If a tumour is resected out of a mouse and transplanted into a different mouse of the same strain, the tumour will grow (as there is no immunological memory)
  • If the tumour is transplanted back into the original mouse the tumour will regress/not grow and this mouse will have immunological memory against that tumour antigen

Evidence of micro-metastases:

  • Even after someone is cleared of cancer, micro-metastases may persist that are not clinically evident, but they are controlled by the person’s immune system
  • If an organ with micrometastases is transplanted into someone else, that person will develop cancer as they do not have sufficient immunological surveillance to keep the micro-metastases in equilibrium
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What is a tumour rejection antigen?

A
  • These are antigens that are expressed in immunoprivelaged sites of the body e.g. cancer-testes area
  • These antigens are usually expressed at a low level, but can be upregulated in cancer e.g. PSA is upregulated in prostate cancer
34
Q

Does a tumour being more immunogenic indicate a better or worse prognosis?

A
  • Based on studies on ovarian cancers, tumours with greater infiltrating lymphocytes belonged to patients with improved survival
  • This suggests that the more immunogenic a patient is- the better their chances of responding to surgery and conventional chemotherapy
  • These findings were also seen in studies of colorectal tumours: the greater the immunogenicity, the bettter survival
35
Q

How is mutational burden and immunogenecity correlated?

A
  • The greater the mutational burden (genetic damage) the greater the immunogenecity
  • Immuno-oncology agents that tend to have the most activity in the clinic against genetically complex tumours such as melanomas and lung cancer
36
Q

Describe the cancer immunity cycle:

A
  1. Release of cancer cell antigens (due to cancer cell death)- may be due to cell being hypoxic/necrotic or may be due to chemotherapy
  2. These antigens produced by the cancer cells are picked up, processed and presented by dendritic cells and other APCs
  3. The antigens are presented to T cells which are primed and activated
  4. The activated T-cells are trafficked to the tumour
  5. The T-cells infiltrate the tumours (may be inhibited by VEGF)
  6. The T-cells recognise the cancer cells via the antigens (inhibited by reduced MHC expression)
  7. T-cells cause cancer cell death (inhibited by PDL1)
37
Q

What is immunogenic cancer cell death?

A
  • Immunogenic cancer cell death is cell death that causes an instigation of the cancer immunity cycle
  • Conventional chemotherapy agents that induce immunogenic cell death are:
    1. Anthracyclines (via signalling to dendritic cells with cross-priming of CD8+ T cells)
    2. Cyclophosphamide (via promotion of Th1/Th17 differentiation, reduced suppressor T-cells, expression of type 1 interfons and mobilisation of DCs
    3. Doxorubicin (via upregulation of CRT on tumour cell surface that signals immune system)
  • Chemotherapy is most effective in organisms with a strong immune response (reduced efficacy in immunosuppressed people)
38
Q

How are monoclonal antibodies used as chemotherapeutics?

A

E.g. anti-CD20 (rituximab)

  • Targets CD20 which is on the surface of B-cells in B-cell lymphoma
  • This causes the death of these cells via:
    1. Antibody-dependent cellular cytotoxicity (priming of cell for NK cells)
    2. Direct cytotoxicity (apoptosis induction)
    3. Complement-dependent cytotoxicity (activates complement system)
  • Rituximab significantly improves cure rate in aggressive diffuse B-cell lymphoma from 50% to 80% compared to standard chemotherapy
39
Q

What aspects of the cancer immunity cycle are targeted in chemotherapy?

A

Step 3: the priming and activation of T-cells

  • Via the activation of T-cells with antibodies
    e. g. Anti-CD137 (agonist) binds to CD137 which is an activating receptor on the T-cells

Step 7: the killing of cancer cells
- Anti-PD-L1, Anti-PD-1 and Anti-CTLA-4

e. g. 1 CTLA-4 is an inhibitory ligand that prevents the activation of T-cells via MHC + costimulation, blocking CTLA-4 allows this activation to proceed unchecked
- This is the mechanism of Ipililmumab which works in metastatic melanoma

e. g. 2 Blockage of PD1 interactions: PD1 is a receptor on T-cells that can be ligated with PD-L1 and PD-L2 which prevents the activation of the T-cell
- These ligands are expressed both on the tumour and in the tumour environment which leads to immune cell exhaustion
- Drugs such as nivolumab use this mechanism
- Hodgkins lymphoma is driven by PDL1 overexpression and treatment with nivolumab increased progression free survival rate in relapsed and refractory patients to 84%

40
Q

How are CAR T-Cells used to treat cancer?

A
  • Chimeric antigen receptor T cells
  • Genetically engineered to link MHC-independent surface antigen recognition to T-cell activation
  • Contains:
    1. Targeting moiety e.g. anti-CD19
    2. Co-stimulation domains
    3. CD3 signaling domains
  • Used to treat relapsed and refractory B-ALL
  • Side effects include cytokine release syndrome
41
Q

What proportion of cancers worldwide are due to infectious agents?

A
  • 1 in 6 cancers worldwide are due to infectious agents (a higher proportion in developing countries)
  • 3/4 of hepatocellular cancer is due to hep B or hep C
  • 70% of cervical cancers are due to HPV 16 and 18
42
Q

What are the most important infectious agents which cause cancer on a worldwide basis?

A
  1. HPV- causes cervival cancer
  2. Hep B- causes hepatocellular cancer
  3. Hep C- causes hepatocellular cancer
  4. Helicobacter pylori- causes stomach cancer
43
Q

How do infectious agents cause cancer?

A
  1. Disrupting normal immune cell function
    - e.g. human T cell lymphocytic virus causes adult T cell leukemia
  2. Causing chronic inflammation- resulting in cell proliferation, DNA damage and inadequate DNA repair
    e. g. TB causes lung cancer
    e. g. Hepatitis B and C cause liver cancer
  3. Disrupting the function of the immune system
    e. g. HIV is associated with a range of cancers caused by human herpes virus 8 (Kaposi’s sarconoma) and HPV
44
Q

Why is mycobacterium tuberculosis a risk factor for lung cancer?

A
  • Due to chronic inflammatory processes potentially increasing the likelihood of cancer
  • Complex relationship
  • Factors predisposing to TB also predispose to lung cancer, and the conditions can mask eachother
45
Q

How do HBV and HCV cause liver cancer?

A

HBV:

  • Chronic hepatitis B virus (HBV) is the cause of 50% of the world’s hepatocellular carcinoma
  • The most important mode of HBV transmission is perinatal
  • HBV infections interact with exposure to aflotoxin (through consumption of contaminated food) to increase the risk of liver cancer
  • There is a vaccine against HBV given to babies at birth to prevent perinatal transmission

HCV:

  • Hepatitis C is the cause of 20% of the world’s hepatocellular carcinoma
  • More than half of hep C in Australia is due to injecting drug use
  • Causes chronic inflammation in the liver
  • There is no vaccine against it
  • New antiviral treatment can clear the virus in >90% of cases
46
Q

How does HPV cause cervical cancer?

A
  • HPV causes squamous cell carcinoma of the cervix, vulva, penis, anus, oral cavity, larynx and esophagus
  • HPV is an STI
  • Of the 40 types, 15 are considered high risk, 70% of cervical cancer cases are caused by HPV types 16 and 18
  • A small proportion who contract but do not clear the virus will go onto develop a squamous cell carcinoma (takes around 10 years)
  • There is a vaccination against types 6, 11, 16 and 18
  • From December 2017, papsmears are being replaced with HPV tests in Australia that looks for infection with HPV rather than histologically abnormal cells
47
Q

How do Herpes viruses cause cancer?

A

Herpes viruses:

  • Usually self limiting in people with healthy immune systems
  • EBV can cause nasopharyngeal cancer and Burkitt’s lymphoma
  • HHV8 can cause Kaposi’s sarcoma (usually in people with AIDS)
48
Q

How does Helicobacter pylori cause stomach cancer?

A
  • H. pylori is a bacteria present in the stomachs of 40% of people (usually acquired in childhood)
  • Causes 2/3 of stomach ulcers
  • Can cause people to go on to develop gastric cancer (MALT)
  • Early stages of gastric cancer caused by H. pylori may be treated with antibiotics to treat the infection
49
Q

What parasites cause cancer?

A
  1. Schistoma (blood flukes) can cause schistosomiasis and bladder cancer due to chronic inflammation and fibrosis
  2. Liver flukes can cause cholangiocarcinoma (bile duct cancer)
50
Q

What is the key oncogenic signalling pathway?

A
  • Receptor tyrosine kinases (RTKs)
  • RTKs are cell surface receptors that have an extracellular ligand binding domain and an intracellular domain that includes a TK domain which catalyses the transfer of phosphate from ATP to tyrosine residues
51
Q

What are the 5 key steps through which RTKs are able to activate oncogenic signalling in cancerous cells?

A
  1. Ligand binding and phosphorylation
  2. Phosphorylation of TK (domain)
  3. Catalytic activity of TK (domain) upregulated
  4. Recruitment of adaptor proteins
  5. Binding of signaling proteins/oncogenes to activated RTKs
  • All of these lead to the downstream activation of key oncogenic pathways
52
Q

What are some examples of RTK mutations that are implicated in cancers?

A
  1. EGFR mutations
    - seen in non-small cell lung cancer
  2. BRAF mutations
    - seen in melanoma
53
Q

How are mutations of EGFR implicated in NSCLC?

A
  • The EGFR pathway has a critical role in many malignancies including NSCLC
  • Binding of the EGF ligand causes the receptor to dimerase and activate the TK domain
  • The activation of the TK domain leads to the binding of adaptor proteins (e.g. Grb2)
  • Binding of the adaptor proteins allows for the binding of many key oncogenic molecules e.g. Ras and the activation of multiple oncogenic pathways such as the Raf-MEK-ERK pathway and the PI3K-Akt pathway
  • The upregulation of these pathways leads to proliferation, cell survival, angiogenesis, metastasis, invasion etc.
54
Q

How is EGFR targeted in cancers?

A
  • Antibodies are targeted to the extracellular domain of EGFR e.g. cetuximab and herceptin
  • Antibodies are targeted to the ligand (EGF) e.g. Bevacizumab
  • Small molecule inhibitors of the internal TK domain have been used to treat NSCLC e.g. geftinib
55
Q

What are EGFR sensitizing mutations?

A
  • Mutations in the TK catalytic domain (exon 19 or 21 deletions) can occur
  • Exon 21 deletions are much more susceptible to treatment with TK inhibitors such as Geftinab
  • The presence of sensitizing mutations in exon 19 or 21 of the TK domain confers sensitivity to EGFR inhibitors
56
Q

What are the associated toxicities with EGFR inhibitors?

A
  • EGFR is highly expressed in many tissues in the body including the skin
  • EGFR inhibitors can present as toxicities in many tissues (not tumour specific)
  • Often presents as a pimply outbreak around the facial area
57
Q

How does EGFR inhibitor resistance occur?

A
  • The vast majority of patients with NSCLC (even those with sensitizing exon 19 or 21 mutations) develop resistance to EGFR inhibitors
  • This often involves a mutation known as T790M in the ATP binding pocket of the enzyme
  • This mutation confers EGFR activity that is not affected by small molecule inhibitors so the oncogenic Raf-MEK-ERK and PI3K-Akt pathways remain switched on
  • New drugs (AZD9291) target the T790M mutation in EGFR binding and will soon be prescribed for refractory patients with EGFR mutated NSCLC
58
Q

Describe BRAF oncogenic mutations in metastatic melanoma:

A
  • Mutations in BRAF oncogene has emerged as a therapeutic target for the treatment of advanced melanoma
  • 66% of melanoma harbours a BRAF V600E mutation
  • This causes the abberant upregulation of the Raf-MEK-ERK pathway which leads to tumour growth
59
Q

How are BRAF mutations targeted in metastatic melanomas?

A
  • Standard chemotherapy has a very poor effect on BRAF V600E mutated metastatic melanoma
  • BRAF inhibitors such as Vemurafenib or Dabrafenib inhibit tumour growth in cancers with the V600E BRAF mutation (standard of care)
60
Q

How does BRAF inhibitor resistance arise?

A
  • Many patients that initially respond well to BRAF inhibitors (Vemurafenib) and then develop a resistance and the cancer begins to progress again
  • BRAF inhibitor resistance is mediated through an upregulation of N-Ras
  • Upregulation of N-Ras causes the activation of c-Raf which then activates the MEK-ERK pathway (bypassing BRAF)
  • It is difficult to target c-Raf, so therapies that target downstream molecules in the MEK-ERK pathway are being investigated
    e. g. BRAF + MEK inhibition- better (now standard of care)
61
Q

Describe the toxicity of BRAF inhibitors?

A
  • When BRAF inhibitors are used alone, a common side effect is the development of SSCs
  • This is due to the upregulation of the RAF-MEK-ERK pathway vi the mutation of KRAS in these cancers
  • Combining BRAF and MEK inhibitors results in less SSCs than BRAF inhibitors alone
62
Q

What is the difference between angiogenesis and vasculogenesis?

A

Angiogenesis:
- Formation of new blood vessels from pre-existing blood vessels

Vasculogenesis:
- Formation of new blood vessels where there are no pre-existing blood vessels

63
Q

What are the key concepts of tumour angiogenesis?

A
  • Driven by hypoxia
  • Balance of pro-angiogenic (e.g. TGF-B, VEGF, PDGF) and anti- angiogenic (e.g. angiostatin, TIMP-2, endostatin) factors produced by tumour and stromal cells
  • Endothelial cells are critical
  • Variety of mechanisms
  • Tumour vessels have abnormal structure and function: disorganised, dilated, uneven, poor contractility
64
Q

What are the key steps in tumour angiogenesis?

A
  1. Hypoxia occurs
  2. Tumour cells releae pro-angiogenic factors (ligands) including VEGF, MMPs (to degrade basemement membrane) and angiopoietin 2
  3. Pro-angiogenic factors bind cell-surface receptors and activate endothelial cells and cause them to migrate towards the tumour
  4. Integrins maintain endothelial cell viability nd increase endothelial cell sensitivity to mitogenic factors e.g. VEGF and b-FGF
  5. The endothelial cells deposit new basememt membrane and secrete PDGF to attract stomal cells to stabilise the new vessel
65
Q

What are the features of the blood vessels produced by tumour angiogenesis?

A
  • The vessels produced by tumour angiogenesis are abnormal meaning they have:
    1. Disorganised vasculature
    2. Toturous, dilated and uneven vessels
    3. Lack of smooth muscle (poor contractility)
    4. Chaotic and variable flow

This leads to:

  1. Hypoxia- which leads to more angiogenesis (positive feedback loop)
  2. Poor drug delivery- difficult to deliver chemotherapeutic drugs
  3. Drug extravasion- due to leaky vessels
  4. Tumour cells entering circulation (circulating tumour cells = CTCs)
66
Q

What are the key pathways underlying tumour angiogenesis?

A
  • Mainly VEGF- A Binding:
  • VEGF receptors are all receptor tyrosine kinases
  • Binding of VEGF-A to VEGFR1 and VEGFR2 induces angiogensis
  • Hypoxia produces HIF-a (hypoxia inducible factor 1-a) which stimualtes the production of VEGF-a and other angiogenic factors
67
Q

How is VEGF/VEGFR targeted in therapies?

A
  • Anti-VEGF antibodies have anti-cancer effects by preventing angiogenesis and restricting the blood supply of a cancer e.g. Avastin
  • Small molecule inhibitors are used to inhibit VEGFR e.g. Sutinitib
  • These drugs are used as a standard of care in kidney and colorectal cancer
68
Q

How does resistance to anti-VEGF therapy occur?

A
  • Resistance to anti-VEGF therapy is essentially inevitable
  • It is adaptive and intrinsic

Adaptive:

  • Upregulation of other pro-angiogenic factors such as EGF, FGF, PDGF etc.
  • Upreguation of pro-malignant signalling pathways

Intrinsic:

  • VEGF- independent angiogenesis
  • Cancer-like stem cells
69
Q

What is metastasis?

A
  • The process by which cancer leaves the primary tumour and travels to a distant site via the blood stream or lymphatic vessels and develops into a secondary tumour
70
Q

What are they key steps in metastasis?

A
  1. Invasion:
    - Cancer cells invade by disrupting ECM and basement membrane
    - This is done using MMPs and other proteolysis
  2. Migration:
    - Cells migrate through reducing cell-cell adhesion (downregulate E-cadherin) and enhancing cell matrix adhesion (using integrins)
    - For this to occur there must be epithelial-mesenchymal transition (EMT): involves cells changing from epithelial phenotype to more mesenchymal phenotype involving loss of cytokeratin expression, E-cadherin expression and polarity and a GAIN in MMP expression, integrin expression
    - The Catenin pathway as well as HGF and FGF are very important for EMT
  3. Circulation:
    - Metastatic tropism: cancers preferentially hone to certain organs
  4. Extravasion
    - Involves invading basement membrane, undergoing MET (gaining E-cadherin and cytokeratin) and attaching to other epithelial cells
  5. Colonisation
    - Often involves angiogenesis
71
Q

What are the different forms of heterogeneity seen in cancer?

A
  • Cancers (especially solid cancers) are highly heterogeneic
  1. Interpatient heterogeneity:
    - When two patients have the same phenotype of cancer e.g. prostate cancer, but their cancers have different genomic profiles
  2. Intratumoural heterogeneity:
    - Different clones within a primary tumour
    - Therefore more difficult to treat
  3. Intermetastatic heterogeneity:
    - Where one metastsis arises from one clone in a primary tumour, and a different metastasis arises from a different clone and thus has a different genetic profile
  4. Intrametastic heterogeneity:
    - Multiple clones present within 1 metastatic lesion
72
Q

What are the types of genetic alterations that lead to cancer?

A
  1. Single base substitutions
    - Most common form of mutation seen
  2. Indels (small insertion or deletion of a few nucleotides)
  3. Rearrangements e.g. translocations
  4. Amplifications (results in large copy number of a gene)
  5. Deletions (homozygous)
73
Q

What are 6 somatic genetic alterations that can be therapeutically exploited in cancer?

A
  1. BRAF mutation in melanoma
  2. Androgen receptor ampification/mutation in prostate cancer
  3. EML4:ALK rearrangement in lung adenocarcinoma (translocation)
  4. EGFR mutation in NSCLC
  5. HER2 mutation in breast cancer (amplification)
  6. K-RAS in colon cancer (mutation)
74
Q

What is the difference between genotyping and sequencing?

A

Sequencing:

  • Involves reading all the data of a library to detect any sequence variant in the gene(s) evaluated
  • Is more challenging, but you may be able to identify new abberations

Genotyping:

  • Detects only known variants that have been selected for analysis (you are specifically looking for certain mutations)
  • Can get better sequencing, but may miss other somatic mutations
75
Q

How does next-generation sequencing work?

A
  1. DNA is extracted from the sample
  2. Adapters bind to the ends of the sequences
  3. The adapters then bind the DNA fragments to a platform e.g. cell
  4. The DNA fragments are then amplified
  5. The DNA sequence is then sequenced using fluroescently tagged nucleotides

Types of NGS include:

  • Whole genome sequencing
  • Whole exome sequencing
    3. Targeted gene sequencing
    4. RNA sequencing
76
Q

How are driver and passenger genes differentiated in NGS?

A

Driving genes: those that change the development and progression of the cancer

Passenger genes: those that are altered, but are not driving cancer progression and growth

  • Differentiated using a COSMIC data base, gene is a driver if: 20% of the mutations for the gene are recurrent and missense
  • 20% of mutations are recorded for that gene as inactivating
  • Only 125 genes have been identified using the 20/20 rule
77
Q

How were BRAF mutations identified in melanoma?

A
  • In the early 2000s it was identified that BRAF mutations were present in 60% of melanomas
  • Specifically the V600E mutation
  • Can be treated with BRAF inhibitors
78
Q

How are KRAS mutations targeted in colorectal cancer?

A
  • It was seen that mutations in the KRAS oncogenes are found in 30-50% of colorectal cancers
  • RAS activates many downstream pathways e.g. Raf-MEK-ERK
  • Anti-RAS drugs cannot be developed, but anti-EGFR drugs have been developed and have MILD benefits in patients with no KRAS mutation (improves quality of life)
  • Therefore KRAS mutation is used as a screening for whether a patient can receive anti-EGFR therapy
79
Q

How is EML-4-ALK mutated in lung adenocarcinoma and how is this treated?

A
  • The EML-4-K inversion mutation is found in 5% of lung adenocarcinoma
  • ALK inhibitors such as crizotinib significantly improve outcomes in patients with this mutation
80
Q

How is the HER2 mutation targeted in the treatment of breast cancer?

A
  • HER2 is a part of the EGFR family of RTKs and is amplified in 15% of breast cancers
  • This amplificaiton leads to the overactivation of the RAS-Raf-MEK-ERK pathway and PI3K-Akt pathway and lead to more aggressive cancer
  • Monoclonal antibody blockage of the HER2 receptor such as herceptin has significantly improved outcomes for people with HER2 mutations
81
Q

What translocation correlates with the abberant upregulation of BCl2 in diffuse large cell lymphoma?

A

t (14;18)

82
Q

What translocation correlates with the abberant upregulation of Cyclin D1 in Mantle cell lymphoma?

A

t (11;14)