Cancer Cell Biology ☺️ Flashcards

1
Q

Cancer

  • what is it
  • what causes it
A

Disease where abnormal cells

  • divide without control
  • invade nearby tissues via blood and LN

Environmental/lifestyle affects genome

  • infection - HPV=> cervical
  • carcinogens - smoking=> lung
  • diet - alcohol

Genetics

  • inherited - BRCA
  • somatic - RAS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

How does RAS work

A

GTPase - can be ON/OFF
GDP bound protein - OFF
GTP bound protein - ON => bind to effector proteins

RAS cannot be switched off in cancer
-most common in pancreatic tumours

RAS activates many pathways needed for cancer cells

  • transcription, cell cycle progression
  • cell survival, growth, migration
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Describe the cell cycle

A
Interphase
-G0/1 = quiescent/organelles replicate
  Rb phosporylated => transition from G1 to S
-S = DNA replication
-G2 = growth of structural elements

Mitosis

  • M = nuclear division and cytokinesis
  • mitotic checkpoints present

If cell cycle too fast, DNA replication more likely to have mistakes

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

How do cyclins affect the cell cycle

A

Cyclins - bind to cyclin dependent kinase => move cell through cell cycle
-regulate the checkpoints between each stage of the cell cycle

Phosphorylates Rb gene => G1 to S

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

How does TGFb affect tumour progression

A

Roles include

  • tumour suppressing responses
  • environmental modifying responses
  • cause changes in phenotype

Cancer cell does not respond to TGFb due to

  • lack of TGFb receptor
  • lose response to TGFb but TGFb still being produced

CAN STILL ACT

  • changes in phenotype of integrins and ecadherins => easier to metastasise
  • stimulate release of angiogenetic factors (VEGF)
  • stimulate connective tissue formation
  • stimulate chemokines and cytokines that make it easier for the metastasised tumour to grow in a distant organ
  • increase immunosuppression against tumour
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is genomic instability

A

Increased tendency of genome alteration during cell division due to

  • increased frequencies of base pair mutations
  • microsatellite instability
  • chromosomal structural variation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Resisting apoptosis

  • when does apoptosis happen
  • how do we get resistance
A

Induced in response to DNA damage or oncogene activation
-mediated by p53

Resistance achieved by

  • loss of p53 signalling (non functioning/too much MDM2/inactivation of DNA damage kinase)
  • upregulation of prosurvival factors like Bcl2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

How does p53 work

A

A gatekeeper gene -directly regulates tumour growth by inhibiting growth/induce apoptosis

If no DNA damage present
-p53 degraded by MDM2

If DNA damage present

  • DNA damage kinase detects this => phosphorylates p53
  • MDM2 inactivated
  • cell cycle arrest/sensence/apoptosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Caretaker/stability genes

  • how does MMR (mismatch repair gene) work
  • what happens if you lose this function
  • examples
A

Correct base mispairs

Accummulation of spontaneous errors
-BRCA1, 2

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

Importance of BRCA1, 2
-significance of BRCA1, 2 mutations

How can we exploit BRCA1, 2 mutations in cancer treatment

A

Caretaker gene - encode proteins that stabilise genome, by repairing breaks

Proteins act as scaffolds that allows for repair
-BRCA1 - recognises double breaks
-BRCA2 - supports repair
BRCA1/2 needs double hit to not function

If BRCA1, 2 ability lost => cells still have PARP activity
-key in DNA single strand break repair

Synthetic lethality
-By using PARP inhibitors => cell can no longer repair DNA damage adequately to survive

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

Enabling replicative immortality
-avoiding telomere shortening

What happens in a normal cell
How does this differ to cancer cells

A

Normal cells

  • telomere repeat sequences
  • polymerase doesn’t transcribe the ends of DNA => lose telomeres instead of useful DNA
  • if telomeres lost => p53 triggered => senescence

Cancer cells
-telomerase lengthens telomeres so cell senesence cannot be achieved

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

Inducing angiogenesis

Activating invasion and metastasis

A

When tumour gets larger, normal vasculature cannot supply the increased metabolic demand
-hypoxia triggers angiogenic switch (VEGF)
Form own vessels to maintain high metabolic rate
-increased pathways to the vasculature

Must be able to

  • alter cell adhesion
  • increase motility - downregulation of ecadherin (no longer attached to other cells) and changes in type of integrin (detach cell from BM)
  • degrade matrix with metalloproteinases - form a pathway to the vessels
  • intravasate, survive in the circulation, arrest, extravasate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Describe locations and patterns of metastasis

A

Different primary cancers have specific patterns and locations of metastasis

  • prostate => bone
  • lung => v early mets to liver

Seed and soil hypothesis
-if a local/distant organ secretes a cytokine/chemokine that the tumour has a receptor to, the tumour will preferentiatlly migrate to that organ

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

How do cancer cells avoid immune destruction

A

Tumour cells can dowregulate MHC => reduced tumour antigen presentation

Tumour cells produce chemokines that

  • attract Tregs
  • TGFb converts CD4 => Tregs

Tumour cells express PDL1 => inhibit programmed cell death to suppress Tcell response

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

Describe tumour promoting inflammatory processes

  • macrophages
  • describe the relationship between the 2
A

Tumour microenvironment promotes emergence of M2 macrophages

Functions

  • produce immunosuppresive molecules
  • secrete growth factors => tumour cell proliferation
  • secrete angiogenetic factors => promote angiogenesis
  • support invasion

Costimulatory relationship => may aid intravasation
M2 secretes EGF needed by cancer cells
Cancer cells secrete CSF1 needed by M2

M1-kill tumour cells
M2-promote antiinflammatory functions => promote tumour progression

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

Types of genetic alterations

A

Chromosomal imbalance
Single gene - tumour suppressors and oncogenes
Epigenetic abnormalities

17
Q

Somatic mutations vs germline mutations

Driver mutations vs passenger mutations

A

Somatic

  • in non germline tissues
  • cannot be inherited

Germline

  • in egg/sperm
  • can be inherited => all cells in offspirng affected

Driver - positively selected due to growth advantage

  • causally implicated in oncogenesis
  • often 2-8 driver mutations

Passenger - not selected for
-does not contribute to cancer

18
Q

Protooncogenes => oncogenes

Tumour suppressors => non functional tumour suppressors

A
Regulate cell proliferation
Gain of function dominant mutation => oncogenes that promoted proliferation
-only 1 copy needed to be faulty
-Her2 - breast
-KRas - colorectal
-BRAF - melanoma

Inhibit tumour formation
Loss of function recessive mutation => cell proliferation increases
-both copies must be faulty (2hit hypothesis)
-TP53 - LiFraumeni
-RB1 - retinoblastoma

19
Q

Common hereditary cancer syndromes

  • HBOC
  • LiFraumeni
  • Lynch
  • FAP
  • MEN2
  • Familial retinoblastoma
  • phaechromocytoma syndrome
A

HBOC - breast, ovarian
LFS (SBLA) - sarcoma, breast, lymph/leukemia, adrenals
Lynch syndrome (MMR genes) - colorectal
FAP (APC gene) - colorectal
MEN2 (RET gene) - medullary thyroid
Familial retinoblastoma (RB1 gene)
Phaechromocytoma syndrome (SDHx gene) - phaechormocytoma

20
Q

Red flags suggestive of hereditary cancer

A

Multiple tumours, associations between different tumours
Inheritance pattern
Young age, ethnicity (BRCA1, 2 in Ashkenazi)

21
Q

Lynch syndrome

  • pathophysiology
  • possible treatments
A

Germline mutation in MMR genes - microsatellite instability => colorectal, ovarian cancer

Leads to formation of neoantigens => stimulate antitumour response of host
Tumour upregulates PD1 => avoid immune surveillance

PD1inhibitors target this to restore immune function

22
Q

EGFR in sporadic non small cell lung cancer

  • function of gene normally
  • what happens when it mutates
A

Epidermal growth factor receptor - regulates signal pathways to control cellular proliferation

Becomes oncogene with driver mutations

Tyrosine kinase inhibitors can block this pathway to reduce signalling