1/80
Looks like no tags are added yet.
Name | Mastery | Learn | Test | Matching | Spaced |
|---|
No study sessions yet.
Describe the key mechanism(s) for cellular transformation of chicken, mouse and/or human cells by RNA tumour viruses
Discuss the broader implications of the discovery that human genomes contained cellular counterparts of viral oncogenes (protooncogenes).
There are various methods of detection and analysis of genome instability. Explain one of these methods and what it measures. What are the implications for cancer diagnosis and treatment?
Avian Leukemia Virus (ALV) induces leukaemia by inserting in the promoter of:
TP53
Myc
Ras
Src
Myc
Anaplasia means:
A well differentiated tumour
A benign lesion
A slow growing tumour
A poorly differentiated tumour
A poorly differentiated tumour
Chromosome segments copied many times and fused head-to-tail in long arrays form:
Double minutes
Homologous deletions
Double chromatids
Homogenously stained regions
Homogenously stained regions
Patient X was diagnosed with breast cancer. What antibody stain would you use to aid diagnosis and direct treatment?
HER2 antibody
P21 antibody
Myc antibody
Ras antibody
HER2 antibody
Which of these characteristics is a unique feature of malignant neoplasms?
Fast growth
Metastasis
Increased nuclear-cytoplasmic ratio
Abnormal mitosis
Metastasis
What are the cellular mechanisms that prevent genomic instability?
DNA repair pathways
Telomere maintenance
Centrosome maintenance
What is the difference between DNA Pol I & III?
DNA Pol I:
Synthesises short stretches of DNA
Only in the lagging strand
5’ to 3’ polymerase activity
5’ to 3’ endonuclease activity (proof-reading function)
3’ to 5’ endonuclease activity (proof-reading function)
Is slow
DNA Pol III:
Main Pol used in replication, synthesis of new DNA strand
Works on both the leading and lagging strands
Has 5’ to 3’ polymerase activity
3’ to 5’ exonuclease activity
Synthesises long stretches of DNA
Very fast
What are the mechanisms of DNA repair? Describe their mechanisms.
Nucleotide excision repair
Base excision repair
Mismatch repair

What are the double stranded DNA repair mechanisms? Describe their mechanisms.
Homologous recombination
Nonhomologous end joining
What is needed to protect chromosome ends?
Telomeres
Why do telomeres shorten with each cell division?
Why is there a D-loop formed at the telomere?
What are telomeres made of?
Repetitive DNA sequences
Do cells have active telomerase?
How does telomerase function?
What is chromatin? What molecules form it? What is its function?
Do chromatin remodeling complexes need energy sources?
How do chromatin remodeling complexes influence chromatin compaction
WHat activities do chromatin remodeling complexes perform within chromatin? Give examples.
Are chromatin remodeling complexes involved in replication?
Yes, chromatin remodelling complexes are involved in DNA replication. They facilitate the process by modulating nucleosome position and composition, which helps replication enzymes access DNA, disassemble nucleosomes ahead of the replication fork, and assemble new chromatin after replication
What do histone post-translational modification do?
Give examples of heterochromatin/euchromatin modifications
Give examples of facultative heterochromatin formation. Which post translational modifications? Which protein complexes are involved?
The most likely epigenetic modification of oncogenes in cancer is:
Histone methylation
DNA hypermethylation
Histone acetylation
DNA acetylation
Histone acetylation
A CpG island:
Is a region of DNA with less than 50 base pairs
Has a GC content higher than 50%
Is rarely seen in prostate cancer
Is frequently acetylates
HAs a GC content higher than 50%
Faults in which DNA repair pathways can induce chromosomal translocations?
MMR and BER
HR and NHEJ
HR and MMR
NER and BER
HR and NHEJ
DNA is coiled around:
Chromatin to form nucleosomes
Genes to form nucleosomes
Protein to form nucleosomes
Chromosomes to form nucleosomes
Protein to form nucleosomes
Which method is not regularly used to detect and analyse genome instability?
Single cell sequencing
Karyotyping
Fluorescence in situ hybridisation (FISH)
Bisulfite sequencing
Bisulfite sequencing
Inhibitory phosphorylation of Cyclin B occurs via:
CAK
Wee1
Chk1
Cdc25
Wee1
Which statement about pRb is most correct?
pRb is a potent oncogene
pRb binds Bcl2 to promote apoptosis
pRb binds the E2F transcription factor to prevent S phase
pRb binds the transcription factor E2F to stimulate S phase
pRb binds the E2F transcription factor to prevent S phase
p21, the protein activated by p53 after DNA damage is:
A ubiquitin ligase
a CDK inhibitor
A phosphatase
A MDM2 inhibitor
a CDK inhibitor
Spindle checkpoint defects can result in:
Chromosome bridges
Telomere lengthening
G1 phase
DNA replication
Chromosome bridges
BH3 mimetics such as venetoclax kill cancer cells by:
Inhibiting Myc
Inhibiting MDM2
Inhibiting Bcl-2
Inhibiting p27
Inhibiting Bcl-2
Which of the following is not a direct cause of oncogenesis in humans:
HTLV-1
Merkel Cell Polyomavirus
Human Immunodeficiency Virus
Epstein-Barr Virus
Human Immunodeficiency Virus
Childhood malaria contributes to development of EBV related lymphomas in which of the following ways:
Impairment of host immunity following malaria infection
Increased susceptibility to EBV infection following malaria infection
Increased genomic instability following malaria infection
Increased lymphocyte production in response to malaria infection
Increased genomic instability following malaria infection
Which of the following is not a factor in the contribution of HIV infection to viral oncogenesis in humans:
Impairment of host immunity following HIV infection
Increased dissemination of malignant cells following HIV infection
Increased susceptibility to infection with oncogenic viruses following HIV
infection
Increased inflammatory factors following HIV infection
Increased dissemination of malignant cells following HIV infection
Describe examples where viral modulation of host pathways contributes to oncogenesis.
Describe the cancers associated with EBV. What factors might contribute to the regional variation in the cancers caused by this virus?
As cells age Telomeres:
Shorten
Lengthen
Replicate
Fuse
Shorten
Ectopic expression of which protein induces a cellular senescence phenotype?
pRb
CycD
Myc
p16 (INK4A)
p16(INK4A)
Multiple molecular changes are required for cancer initiation and progression.
What was the tumour type that suggested cancer evolves over a long time?
What sequence of cellular and genetic changes are associated with evolution of this cancer?
What is clonal evolution? Discuss in context with acquired chemotherapy resistance.
Discuss the evidence for the “mutator phenotype” and whether it might be a necessary step for tumourigenesis.
Describe the key principles of stem cell fate regulation within the stem cell compartment.
Why do you think an understanding of the stem cell-niche interaction might be important for improving cancer treatment?
Although cancer cell dissemination can start early during tumour progression, most cells leaving a tumour fail to colonise distant organs and instead succumb to various stresses.” What are these stresses?
Eliminated once entering the blood stream due to sheer stress within microvessels, cell-death induced by loss of attachment to a substrate (anoikis), and lysis by natural killer cells.
Stresses encountered following extravasation into the host tissue, including reduced bioavailability of nutrients, oxidative stress, and immune attack by tissue resident
immune cells (i.e. tissue resident macrophages and natural killer cells). Bone-marrow derived, and peripheral immune cells are also recruited to eliminate the newly seeded
metastases. The host tissue is a hostile place!
A large array of gene expression changes is required to promote metastasis. Do these changes occur in the primary tumour or once the disseminated cancer cell has seeded a distant tissue?
Both. Gene expression changes in the primary tumour prime the cells for metastasis, while other gene expression changes develop in the host tissue to allow the cells to adapt to the new environment.
New mutations are not required to promote metastasis. What evidence exists to support this claim?
The mutation patterns and overall mutational burden are almost indistinguishable between primary and metastatic cancers. However certain clones with particular genetic
alterations will dominate in metastatic lesions, which can be explained by the monoclonal metastasis model as described in L17 (slide 17)
After the primary tumour is removed by surgical resection, the patient may develop clinically detectable metastases years later. How is this possible if circulating tumour cells are eliminated from the blood stream after 24 hours?
Micrometastases which form before the removal of the primary tumour, can enter proliferative quiescence before colonising the tissue as a macrometastases.
Alternatively, a macrometastases may fail to grow to a clinically detectable size due to factors in the host stroma or insufficient vascularisation.
Evidence of metastatic dormancy – reported cases of organ transplant recipients who have developed metastases after receiving organs from donors who had been deemed cured of melanoma
On page 37, under the subheading Developing therapies for micro- and macrometastases the authors make the statement “While targeting the early steps of tumour invasion and survival in the circulation are, in our opinion, unlikely to be effective at preventing or treating metastasis, targeting micrometastatic cells could prevent metastasis”. Why do you think the authors have drawn this conclusion?
Patients presenting with stage IV cancer have overt metastatic disease and therefore interfering with further seeding is likely to be futile in halting disease progression.
In patients that do not present with clinical mets, the primary tumour is usually surgically excised prior to treatment with systemic or targeted therapies, thereby quickly eliminating the source of circulating tumour cells. Patients that develop distant recurrence following primary tumour removal, indicates that micrometastases had formed prior to surgery.
EMT-inducing transcription factors:
Are specific to cancer
Have non-overlapping functions in activating/repressing gene expression
Can induce the expression of other EMT transcription factors
Can act in isolation to execute the entire EMT program
Can induce the expression of other EMT transcription factors
Local invasion does not involve:
Disruption of extracellular matrix (ECM) by proteases
An increase in cell motility
An ability to undergo epithelial to mesenchymal transition (EMT)
Avoiding interaction with surrounding stromal cells
Avoiding interaction with surrounding stromal cells
Metastasis requires:
A cell with the primary tumour acquiring new mutation, which favours the metastatic process
The ability of a cell to maintain a mesenchymal phenotype at the site of seeding
Clonal selection of pre-existing mutations present in the primary tumour
A complete epithelial to mesenchymal transition to occur
Clonal selection of pre-existing mutations present in the primary tumour
Which statement about the actin cytoskeleton is correct?
Filopodia and lamellipodia are actin-based structures, but invadopodia are not
Rho GTPases remodel the actin cytoskeleton by activating actin-binding proteins
The only function of the actin cytoskeleton is to help cells move
Filopodia are composed of branched actin filaments
Rho GTPases remodel the actin cytoskeleton by activating actin-binding proteins
Epithelial to mesenchymal transition is commonly associated with:
A static actin cytoskeleton
The induction of E-cadherin expression
The acquisition of stemness
The formation of invadopodia in neoplastic cells which secrete MMPs
The acquisition of stemness
List two cell types of the immune system that can kill
cancer cells by the secretion of cytotoxic granules,
and the mechanism of action. What are the
implications for cancer treatment?
A patient has a mysterious B cell cancer. This patient also carries genetic deficiencies such that their B cells do not generate antibodies and their NK cells are defective in antibody-dependent cell-mediated cytotoxicity. Discuss TWO immunotherapies and evaluate their efficacy in the context of this patient’s genetics.
Which one of the following cell types secretes antibodies?
B cells
Macrophages
Dendritic cells
T cells
B cells
Which of the following statements about immunotherapy is correct?
CAR-T cell therapy targets immune checkpoint proteins.
Immunotherapy is used as frequently as chemotherapy
Immunotherapy is affordable for all cancer patients
Immunotherapies can induce innate and adaptive immune responses
Immunotherapies can induce innate and adaptive immune responses
Which of the following statements does NOT support the idea that the immune system monitors and eliminates cancer cells?
Mice injected with live tumour cells develop tumours whereas mice injected with dead tumour cells do not develop tumours
Humans whose tumours have high levels of tumour-infiltrating lymphocytes survive longer than those whose tumours have low levels of tumour-infiltrating lymphocytes
Mice lacking perforin have an elevated incidence of spontaneous tumours compared to control mice expressing perforin
Immunocompromised patients experience a higher incidence of cancers
Mice injected with live tumour cells develop tumours whereas mice injected with dead tumour cells don’t develop tumours
Which statement about innate immunity is most correct?
Innate immunity provides rapid and early responses
Innate immunity generates immunological memory that can prevent re-infection by the same microorganism
Innate immunity is triggered by the generation of antigen-specific effector cells with high specificity
T helper cells are a type of innate immune cells
Innate immunity provides and early responses
Which of the following statements about CAR-T cells is most correct?
Cytokine release syndrome is a side effect of CAR-T cell therapy
CAR-T cell therapy is standard care for all breast cancer patients
Resistance to CAR-T cell therapy never develops
CAR-T cells require co-stimulation by dendritic cells
Cytokine release syndrome is a side effect off CAR-T cell therapy
The drug discovery and pre-clinical testing phases of a therapeutic designed to
be used to treat cancer is performed to:
A: Evaluate how well the therapy acts on the molecular target/process
B: Refine a large list of molecules down to a handful (‘hit to lead’)
C: Identify whether the therapy has the desired effect in animal models of cancer
D: All of the above
D
Targeted therapies are designed to:
A: Remove or reduce the amount of tumour tissue
B: Target specific proteins/molecular interactions in cancer cells to block/modify their activity
C: Kill cancer cells and the surrounding stromal cells
D: Activate the immune system to kill cancer cells
B
You are in charge of a team of cancer therapeutics researchers in a small biotech
company, and you have been given the task of developing a new drug.
The target you are required to develop the drug for is a highly novel receptor tyrosine
kinase called NOVR.
NOVR is:
• Structurally similar to the Epidermal Growth Factor Receptor (EGFR)
• Activated after stimulation with a ligand (NOV ligand)
• Located in the cell membrane (N-terminus is located extracellularly)
• Activated after ligand stimulation, where the intracellular tyrosine kinase domain
becomes phosphorylated which leads to MAPK pathway activation
Genomic analysis of tumours isolated from breast cancer patients shows that NOVR
expression is highly upregulated, however, the gene itself is not found to be
mutated in breast tumours
Given what we know about NOVR, which approach do you think is the most appropriate
for drug development?
A: Rational drug discovery
B: High-throughput screening
A
You are a junior group leader who has an interest in identifying a novel small molecule therapeutic which will lead to the activation of double-stranded DNA breaks (DSBs) in cancer cells, as a means to induce cytotoxicity and cancer cell death.
A hard-working PhD student in the lab has developed a phenotypic assay which is able to measure the amount of DSBs in cancer cells.
Which approach do you think may be the most appropriate for drug development?
A: Rational drug discovery
B: High-throughput screening
B
Targeted therapies are designed to:
debulk tumour tissue
kill all cells
activate B cells
target specific proteins
Target specific proteins
What is the target of the treatment successfully used in melonoma?
B-RAF
AKT
Rb
MYC
B- RAF
Clinical trials designed to test a small number of human volunteers/patients to identify a safe dose of the drug, route of administration and how the drug affects the body are:
Phase II
Phase IV
Phase III
Phase I
Phase I
Which situation might require a rational drug screen approach?
The aim is to test whether approved therapeutics can be repurposed for other diseases
Molecular target is unknown
Multiple pathways need to be targeted
Knowledge of lead compound mechanism of action is required from the start
Knowledge of lead compound mechanism of action is required from the start
Which treatment can most readily be used for precision medicine?
Tyrosine kinase inhibitors
Chemotherapy
Surgery
Hormone therapy
Tyrosine Kinase Inhibitors
Why is the lymphatic system important for cancer cell invasion?
How do Tissue Inhibitors of Metalloproteinase (TIMPs) impact angiogenesis in normal cells and how they are dysregulated in cancer?
What are the changes that occur in tumour-associated vasculature and how do they contribute to tumour progression?
Loss of function mutations in which of the following genes has been linked to anti-PD-1 resistance?
B2M
KRAS
EGFR
KMT2D
B2M
Which one of the following is a T-cell inhibitory receptor?
LAG-3
CD28
OX40
PD-L1
LAG-3
What tumour characteristic can indicate a favourable response to anti-PD-1 therapy?
Large tumour size
High mutational burden
Presence of tumour associated macrophages
T cell exhaustion phenotype
High mutational burden
Which event might prevent a T-cell from effectively responding to a tumour antigen?
Increase in granzyme production
Loss of PD-1 expression
Physical association with the tumour cell
Upregulation of CTLA-4
Upregulation of CTLA-4
Which of the following statements about anti PD-1 therapy is most correct?
Immune related side effects are more common with anti-PD-1 therapy than anti-CTLA-4 therapy
DNA mismatch repair proficient tumours are more likely to respond to anti-PD-1 therapy than tumours with DNA mismatch repair deficiency
The tumour mutational burden is negatively correlated with the objective response rate in patients treated with anti-PD-1 therapy
PD-L1 immunohistochemistry is performed routinely in metastatic lung squamous cell carcinoma
PD-L1 immunohistochemistry is performed routinely in metastatic lung squamous cell carcinoma