Summary Flashcards

1
Q

Give 3 examples of oncogenes and explain their function and pathways they regulate and how they drive cancerogenesis forward

A

RAS genes encode for GTPase proteins that in cancer promote proliferation and surrvuval through the MAPK and P13-AKT signalling pathways.

MYC encodes for transcriptional factors that enhances gene expression of genes that regulate cell cycle progression and metabolism through pathways such as WNT and P13K which promotes cell differentiation, surrvival etc.

HER2 is a growth factor that binds to receptor tyrosine kinases which promotes cell proliferation, surrvival, differentiation etc.

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

Describe 2 tumor supressor genes, their function and how they can promote cancerogenesis and how can we target them in cancer therapy

A

P53 also known as the guardian of the genome regulated the cell cycle in G1/S checkpoint in case of DNA damage which is before DNA replication which inhibits propagation of mutations. It can initiate DNA repair pathways by activating genes, and in case of severe DNA damage it can initiate apoptosis by activating pro-apoptopic proteins such as BAX, BAK and PUMA and repress anti-apoptopic factors such as BCL-2. In cancer is van get loss of function mutations, promoter can become hypermethylated or mutations which alters the amino acid sequence so the protein is faulty or can even be tumorogenic which inhibits it’s tumor supressor functions. Gene therapy to introduce a functional TP53 in cancer cells, small molecules that can reactivate or mimic it’s function to promote cancer cell death and immunotherapy with the function to kill P53 defiecent cells is an option.

PTEN regulates the P13K/AKT pathway by dephosroylating PIP3 which prevents AKT activation and this regulating cell growth, differenitation etc. It can develop loss of function mutations that silences it and theraputic targets include inhibiing the P13/AKT pathway again.

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

What are some stel cell markers and what properties does it contribute with?

A

Stem cell cancer markers include CD44 which is a cell surface glycoprotein that is involved in cell-cell interactions, cell adhesion and migration. It is linked to cancer progression and metastasis. CD133 is a membrane protein that can be used as a marker to identify and isolate cancer stem cells, especially in colorectal, liver and brain cancer. ALDH is an enzyme that is highly expressed in cancer stem cells and correlate with tumorigenic potential and resistance to chemotherapy. Oct4 is a transcription factor that plays a key role in maintain pluripotency and self renewal in stem cells and is often overexpressed in cancer stem cells.

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

Why is cancer stem cells more therapy resistant?

A

During cell stress it can enter a quiescent state which eliminates it as a target for chemotherapy that targets rapidly dividing cells, it overexpresses ABC-transporters which are drug efflux transporters, and it has enhanced DNA repair mechanisms such as homologous recombination and non homologous end joining pathways.

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

How can we target CSC in treatment?

A

Focusing on the signalling pathways Notch, WNT and Hedgehog which they are regulated by to eliminate them.

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

Describe correlation between cancer stem cells (CSC) and circulating tumour cells (CTC). How and where are the CTC detected in a patient?

A

Circulating tumor cells are cells that have shed from the tumor and circulates in the body which can have stem cell proprties or be cancer stem cells. This is of importance since only stem cells can initiate tumors on a new site which means it’s a higher risk of metastasis. They can be detected by immunoaffinity assays which uses antibodies targeted for the CTC or size based filtration as the cells are bigger compared to normal red blood cells.

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

How can we study CSC?

A

We can isolate them based on their cancer stem cells markers and isolate them from the bulk tumor population using flow cytometry. We can also use sphere formation assays which are cell cultures without adhesion and serum free medium as primarly CSC can grow spheres under those conditions. Once isolated they can also be injected in immunosupressed xenograph models such as mice and analyze their tumor initiation ability.

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

What does the telomerase activity look like during normal and cancer cells compared?

A

Normal cells have TERC active always shich is the RNA component of telomerase but inactive TERT gene which encodes the catalytic subunit of telomerase. Cancer cells have both these active always.

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

Give 3 examples of cancer types and specific examples of cancers within that type

A

Carcinoma comes from epithelial cells and examples include adenocarcinoma, squamois cell carcinoma.
Sarcomas originate from mesenchymal cells and examples are osteosarcoma and liposarcoma.
Leukemias comes from hematopoietic stem cells and examples include acute myeloid leukemia and acute lymphoblastic leukemia.

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

How can we detect tumor heterogeneity?

A

Single cell RNA sequencing which looks at gene expression of idnevidual cells within the tumor or between the tumors, and immunohistochemistry which looks at differences in protein expression between tissuesections of the tumor.

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

Describe how methylation occurs

A

Substrate Recognition: DNMTs recognize specific cytosine residues in the DNA sequence, typically within CpG dinucleotides.

Methyl Group Transfer: DNMTs transfer a methyl group from SAM to the 5th carbon position of the cytosine ring, resulting in the formation of 5-methylcytosine.

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

What are the two types of radiation therapy called?

A

External Beam Radiation Therapy (EBRT): This uses high-energy beams (like X-rays or gamma rays) directed at the tumour from outside the body.

Brachytherapy: This involves placing a radioactive source directly inside or next to the tumour, delivering a high radiation dose to the tumour while sparing surrounding healthy tissues.

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

Name 3 immune parameters that can serve as biomarkers in cancer

A

Tumour-Infiltrating Lymphocytes (TILs): The presence and abundance of TILs in the tumour microenvironment can indicate the immune response against the tumour. Higher TIL levels are often associated with better prognosis and response to immunotherapy.

Programmed Death-Ligand 1 (PD-L1) Expression: PD-L1 expression on tumour cells can suppress immune responses. High PD-L1 levels are used as a biomarker for selecting patients who might benefit from immune checkpoint inhibitors (e.g., anti-PD-1/PD-L1 therapy).

Cytokine Profiles: Levels of cytokines such as interleukins (e.g., IL-6, IL-10) and interferons (e.g., IFN-γ) in the blood or tumour microenvironment can reflect the immune status and inflammation associated with cancer.

IL-6: Pro-inflammatory, Promotes tumourigenesis by stimulating cell proliferation and survival. Involved in angiogenesis and metastasis.

IL-10: Anti-inflammatory, Suppresses immune responses to avoid excessive tissue damage, In cancer, IL-10 may promote tumour immune escape by inhibiting antitumour immunity. Elevated IL-10 levels can correlate with poor prognosis in some cancers.

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

Describe how Microenvironment and cancer work together in cancer development, progression and/or metastasis.

A

Cancer cells interact dynamically with their microenvironment, consisting of stromal cells, immune cells, extracellular matrix (ECM), and signaling molecules, to promote development, progression, and metastasis.

Interaction with Fibroblasts: Cancer-associated fibroblasts (CAFs) secrete growth factors like TGF-β and VEGF, promoting tumour growth, angiogenesis, and invasion. CAFs also remodel the ECM, creating pathways for cancer cell migration and metastasis.

Immune Cell Manipulation: Cancer cells recruit and reprogram immune cells like macrophages into tumour-associated macrophages (TAMs). TAMs release cytokines (e.g., IL-10, IL-6) that suppress anti-tumour immune responses and stimulate angiogenesis, aiding in tumour growth and immune evasion.

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

How is apoptosis regulated by surrvival signals?

A

Survival signals (e.g., PI3K/Akt pathway) can block apoptosis by inhibiting pro-apoptotic proteins like Bax and activating anti-apoptotic proteins like Bcl-2.

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

Name Three different cellular assays used to study apoptosis in cancer.

A

Caspase Activity Assay:
This assay measures the activation of caspases using substrates that release a detectable signal when cleaved by caspases, this assay helps quantify apoptotic cell death.

Annexin V/PI Staining:
This flow cytometry-based assay detects early apoptosis by labeling phosphatidylserine exposure on the outer membrane using Annexin V and late-stage apoptosis or necrosis using PI.

Annexin V+ / PI- cells indicate early apoptosis.

Annexin V+ / PI+ cells suggest late apoptosis or necrosis.

TUNEL Assay

The TUNEL assay labels DNA fragments produced during apoptosis by detecting 3’-OH ends of fragmented DNA. Cells undergoing apoptosis will show a positive signal, allowing the quantification of apoptotic cells in tissue sections or cultures.

17
Q

Name other forms of cell death excluding apoptosis

A

Pyroptosis is driven by the recognition of DAMPs by PRR such as mtDNA, RPS etc and cause pore formation to release pro-inflammatory cytokines which recruit immune cells which enhance anti-tumor immunity but also promotes chronic inflammation which can lead to immune evasion.

Necroptosis is regulated necrosis which is activated by a loss of survival factors, DNA damage (can come from cancer treatments) and viral infections. This causes the necrosome to form and become activated which leads to necroptosis.

Ferroptosis is iron dependent and occurs when there is an accumulation of iron which leads to a chemical reaction that causes oxidative stress cascade and membrane rupture. Cancer cells have higher need for iron so it’s a treatment target since they are therefore more sensitive to this reaction.

18
Q

Describe the mechanism of action of 4 anti cancer medications

A

Alkylating Agents

Form covalent bonds with DNA, causing cross-links and strand breaks

Antimetabolites

Mimic cellular metabolites, disrupting DNA and RNA synthesis

Topoisomerase Inhibitors

Inhibit topoisomerase enzymes, preventing DNA unwinding and replication

Microtubule Inhibitors

Disrupt microtubule dynamics, inhibiting mitosis and cell division

19
Q

Name 4 anti cancer medications

A

Cisplatin 

Methotrexate

Doxorubicin

Paclitaxel

20
Q

Describe 3 ways cancer can metastize?

A

Lymphatic Spread: Cancer cells enter lymphatic vessels and travel to regional lymph nodes, eventually reaching distant organs.

Hematogenous Spread: Cancer cells invade blood vessels and circulate through the bloodstream to distant sites.

Transcoelomic Spread: Cancer cells spread across body cavities, such as the peritoneal cavity, leading to metastasis in organs like the liver and lungs.

21
Q

How can cancer cells avoid immune recognition?

A

Immune Checkpoint Inhibition: Tumour cells express PD-L1 to bind PD-1 on T cells, inhibiting their activity.

Secretion of Immunosuppressive Cytokines: Tumour cells secrete cytokines like TGF-β and IL-10 that suppress immune responses.

Downregulation of Antigen Presentation: Tumour cells decrease the expression of MHC molecules, reducing recognition by cytotoxic T cells.

22
Q

New drug development against cancer, what are your main drug targets and why?

A

Tyrosine Kinases: Essential for signaling pathways that drive cell growth and survival.

Immune Checkpoints: Modulating checkpoints like PD-1/PD-L1 can enhance anti-tumour immunity.

Angiogenesis Pathways: Targeting VEGF pathways can inhibit tumour blood supply.

23
Q

Name and describe the hallmarks of cancer

A

Sustaining Proliferative Signaling

Cancer cells maintain constant signals to divide, bypassing normal growth control.

Evading Growth Suppression

Inactivation of tumour suppressor genes (e.g., TP53, RB1) to avoid growth inhibition.

Resisting Cell Death

Avoiding apoptosis by altering pathways (e.g., overexpressing Bcl-2).

Enabling Replicative Immortality

Maintenance of telomeres, often via telomerase activation, allowing limitless divisions.

Inducing Angiogenesis

Stimulating new blood vessel growth to supply nutrients and oxygen.

Activating Invasion and Metastasis

Gaining the ability to invade tissues and spread to distant sites.

Avoiding Immune Destruction

Evasion of immune surveillance through immune checkpoints or TME alterations.

Deregulating Cellular Energetics

Shifting metabolism to favor glycolysis (Warburg effect), even in oxygen presence.

Genome Instability and Mutation

Increased mutation rates due to defects in DNA repair mechanisms.

Tumour-Promoting Inflammation

Chronic inflammation fosters cancer development and progression.

24
Q

Describe the process of EMT and what it is

A

EMT is the process by which epithelial cells acquire mesenchymal traits, enhancing their migratory and invasive abilities. EMT allows cancer cells to detach, invade surrounding tissues, and enter the bloodstream, facilitating metastasis.

Process:

Loss of Epithelial Markers: Cells downregulate epithelial markers like E-cadherin, weakening cell-cell adhesion.

Gain of Mesenchymal Traits: Cells upregulate mesenchymal markers like N-cadherin and vimentin, enhancing motility and invasiveness.

Cytoskeletal Reorganization: Actin cytoskeleton rearranges, aiding migration.

Migration and Invasion: EMT cells degrade extracellular matrix (ECM) and penetrate tissues, allowing dissemination to secondary sites.

25
Q

Name an mRNA and it’s function in cancer

A

MicroRNA-200 (miR-200) regulates cancer progression by controlling EMT. It inhibits the expression of key EMT transcription factors like ZEB1 and ZEB2, maintaining epithelial characteristics and preventing metastasis.

In Cancer:

Tumour Suppressor: miR-200 prevents metastasis by maintaining cell adhesion and inhibiting cell migration.

Cancer Progression: Downregulation of miR-200 promotes EMT, leading to increased invasiveness and metastatic potential in tumours.

26
Q

Give one example of an mRNA acting on a tumor supressor gene and another on an oncogene

A

miR-21: Oncogenic miRNA that inhibits tumour suppressor genes like PTEN, activating the PI3K/AKT pathway.

let-7: Tumour suppressor miRNA that downregulates oncogenes like RAS, reducing cell proliferation.

27
Q

What are 4 Effects of HIF Activation:

A

Angiogenesis: HIF induces the expression of vascular endothelial growth factor (VEGF), promoting the formation of new blood vessels to supply oxygen to the tumour.

Metabolic Reprogramming: HIF activates glycolytic enzymes, shifting the tumour cell metabolism towards glycolysis (the Warburg effect), even in the presence of oxygen, to support rapid cell growth.

Cell Survival and Proliferation: HIF promotes the expression of genes involved in cell survival, such as erythropoietin (EPO), and regulates the cell cycle to enhance proliferation in low-oxygen environments.

Invasion and Metastasis: HIF can upregulate genes involved in extracellular matrix degradation and cell migration, facilitating cancer cell invasion and metastasis.

28
Q

What is haploinsufficency and name 3 genes that have it

A

Haplo-insufficiency occurs when loss of one gene copy results in insufficient protein activity to maintain normal cellular functions. Examples include:

TP53: Partial loss reduces the cell’s ability to respond to DNA damage, resulting in compromised tumour suppression.

PTEN: Its dosage-dependent role in antagonizing PI3K-Akt signaling is disrupted even with a single-allele loss, leading to increased proliferation and tumour growth.

RB1: A single functional copy may be insufficient to fully suppress the cell cycle in the presence of oncogenic stimuli.