Cancer Flashcards

1
Q

Define metaplasia.

A

• A reversible change in which one adult cell type (usually epithelial)is replaced by another
adult cell type
• Adaptive

Physiological examples:

  • Cervix - puberty, cervix expansion, columnar epithelium of inter-cervical canal exposed to acidic pH of vagina - columnar —> squamous
  • Acid reflux from oesophagus, squamous —> columnar (Barrett’s oesophagus)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Define dysplasia.

A

• an abnormal pattern of growth in which some of the cellular and architectural features of malignancy are present
• pre-invasive stage with intact basement
membrane - Not invasive
• loss of architectural orientation - not maturing in normal way
• loss in uniformity of individual cells
• nuclei: hyperchromatic, enlarged - dark nuclei because conc. of DNA increases. High nuclear: cytoplasmic ratio
• mitotic figures: abundant, abnormal, in places where not usually foun

Common in: 
• CERVIX - HPV infection 
• BRONCHUS - Smoking
• COLON - UC (ultra colitis)
• LARYNX - Smoking 
• STOMACH -Pernicious
• OESOPHAGUS- Acid reflux

Low grade vs high grade
Low grade:
1) risk of progression is low
2) more likely to be reversible

High grade:

1) risk of progression is high
2) less likely to be reversible
3) darker because nuclei greater

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

Define neoplasia, tumour, malignancy.

A

An abnormal, autonomous proliferation of cells unresponsive to normal growth control mechanisms

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

Describe the differences between benign and malignant tumours.

A

Benign:

1) do not invade; do not metastasise
2) encapsulated - sharp edge, fibrous capsule - easier to remove
3) usually well differentiated - look like where they came from
4) slowly growing
5) normal mitoses

Not fatal unless:

  • In dangerous place: meninges (block between lateral and third ventricle which increases intra-cranial pressure)
  • Secretes something dangerous: insulinoma
  • Gets infected: bladder
  • Bleeds: stomach
  • Ruptures: liver adenoma
  • Torts(twisted): ovarian cyst

Malignant:

1) invade surrounding tissue
2) spread to distant sites - block vessels, lymphatics
3) no capsule
4) well to poorly differentiated
5) rapidly growing
6) abnormal mitoses

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

Define metastasis.

A

A discontinuous growing colony of tumour cells, at some distance from the primary cancer.

These depend on the lymphatic and vascular drainage of the primary site

Lymph node involvement has a worst prognosis
E.g. Dukes A- 90% (only on bowel)
Dukes C - 30% (in lymph nodes)

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

Describe the nomenclature of tumours.

A

-oma = benign mass

Benign epithelial tumours:

  • of surface epithelium = papilloma e.g. skin, bladder e.g. wart from HIV
  • of glandular epithelium = adenoma e.g. stomach, thyroid, colon, kidney, pituitary, pancreas

Carcinoma:

  • a malignant tumour derived from epithelium e.g. through basement membrane invasion
  • squamous cell
  • adenocarcinoma
  • transitional cell e.g. bladder
  • basal cell carcinoma e.g. skin

Benign soft tissue tumours:
E.g. osteomalacia
Leiomyoma - smooth muscle

Sarcoma:

  • a malignant tumour derives from connective tissue (mesenchymal) cells
  • fat = liposarcoma
  • bone = oestosarcoma
  • cartilage = chondrosarcoma
  • muscle, striated = rhabdomyosarcoma, smooth = leiomyosarcoma
  • nerve sheath = malignant peripheral nerve sheath tumour

Leukaemia and lymphoma:
-tumours of white blood cells:
Leukaemia = a malignant tumour of bone marrow derived cells which circulated in the blood
Lymphoma = a malignant tumour of lymphocytes (usually) in lymph nodes (End in -Oma but malignant)

Teratoma:
-tumour derives from germ cells, which have the potential to develop into tumours of all three germ cell layers:
1) ectoderm
2) mesoderm
3) endoderm
Can develop into any type of tissue
-gonadal teratomas in males, all malignant
-gonadal teratomas in females, most are benign

Hamartoma
-localised overgrowth of cells and tissues native to the organ
-cells are mature but architecturally abnormal
-common in children, and should stop growing when they do
E.g. bile duct hamartomas, bronchial hamartomas

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

Describe the differentiation of tumours.

A

Criteria for assessing differentiation of a malignant tumour:
-evidence of normal function still present and production of:
Keratin, mucin, bile, hormones - does it still produce these?

Various garden systems for cancer of breast, prostate, colon e.g. Gleason grading system for prostate
No differentiation = anaplastic carcinoma

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

Describe the TNM system.

A

The grade of a tumour describes its degree of differentiation.

The stage of a tumour describes how far it has spread.

Tumours of higher grade (I.e. poorly differentiated) tend to be of higher stage (I.e. spread further)

Overall stage is more important than grade in determining prognosis.

T=tumour
N-node
M=metastasis

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

State the factors which influence the rate of cell division.

A

1) embryonic vs adult cells - embryo faster
2) complexity of system e.g. yeast faster
3) necessity for renewal (intestinal epithelial cells faster than hepatocytes - queiescent, if only injury is it highly proliferative)
4) state of differentiation (some cells never divide e.g. neurones and cardiac myocytes
5) tumour cells

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

Describe the relevance of the appropriate regulation of cell division.

A

Premature, aberrant mitosis results in cell death.

In addition to mutations in oncogenes and tumour suppressor genes, most solid tumours are aneuploid (abnormal chromosome number and content)

Various cancer cell lines show chromosome instability (lose and gain whole chromosomes during cell division)

Perturbation (deviation from normal state) of protein levels of cell cycle regulators is found in different tumours - abnormal mitosis

Contact inhibition of growth

Attacking the machinery that regulates chromosome segregation is one of the most successful anti-cancer strategies in clinical use.

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

Explain the cell cycle.

A

Orderly sequence of events in which a cell duplicates its contents and divides into two.

  • duplication
  • division
  • co-ordination

Interphase (duplication)

  • DNA
  • organelles and protein synthesis

M-phase: mitosis (division)

  • nuclear division
  • cell division (cytokinesis)

Mitosis - most vulnerable period of cell cycle:

  • cells are more easily killed (irradiation, heat shock, chemicals)
  • DNA damage can not be repaired
  • gene transcription silences
  • metabolism reduced, cell’s energy focussed on division

Interphase:
G0 = cell cycle machinery dismantled

G1 phase (Gap) = decision point, like checkpoint - are all organelles duplicated?

S phase - synthesis of DNA/ protein

  • DNA replication
  • protein synthesis: initiation of translation and elongation increased; capacity is also increased (increase in ribosomes)
  • replication of organelles (centrosomes, mitochondria, Golgi etc.) in the case of mitochondria, needs to coordinate with replication of mitochondrial DNA

G2 phase (Gap) - decision point - check DNA duplication, mutation

Mitosis:
Prophase
-condensation of chromatin
-Condensed chromosomes each consists of 2 sister chromatids, each with a kineticochore and joined by centromere
-duplicated centrosomes migrate to opposite sides of the nucleus and organise the assembly of spindle microtubules
-mitotic spindle forms outside nucleus between the 2 centrosomes

Spindle formation - radial microtubule arrayed (ASTERS) form around each centrosome (microtubule organising centres - MTOC), radial arrays meet, polar micro tubes form - stabilised at centre of cell
Microbes are in a dynamic state

Early prometaphase

  • breakdown of nuclear membrane
  • spindle formation largely complete
  • attachment of chromosomes to spindles via kinetochores (centromere region of chromosomes) - microtubules capture at this region

Late prometaphase:

  • microtubule from opposite pole is captured by sister kinetochore
  • chromosomes attached to each pole congress to the middle
  • chromosome slides rapidly towards center along microtubules

Metaphase
Chromosomes aligned at equator of the spindle

Anaphase
Paired chromatids separate to form two daughter chromosomes
Cohesion holds sister chromatids together
Anaphase A and B

Anaphase A:
Breakdown cohesion
Micro tubes get shorter
Daughter chromosomes pulled toward opposite spindle poles

Anaphase B:
1-daughter chromosomes migrate towards poles
2-spindle poles ‘centrosomes’ migrate apart

Telophase

  • daughter chromosomes arrive at spindle
  • nuclear envelope reassembles at each pole
  • assembly of contractile ring - cleavage furrow

Cytokinesis

  • new membrane inserted
  • acto-myosin ring contracts
  • midbody begins to form
  • interphase microtubule array reassembles
  • chromatin decondenses and nuclear sub structures reform

Transition out of metaphase: spindle assembly checkpoint
-senses completion of chromosome alignment and spindle assembly (monitors kinetochore activity) - make sure in correct position e.g. equator and all attached to microtubules
Requires:
CENP-E - tension
BUB protein kinases - BUBs dissociate from kinetochore when chromosomes are properly attached to the spindle and when all dissociated, anaphase proceeds.

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

Describe how aneuploidy occurs.

A

Mis-attachment of microtubules to kinetochores

1) synthelic attachment: both sister chromatids attached but attached to wrong microtubules - same daughter cell in this case ; both sister chromatids at same pole
2) monotelic attachment - only one of the sister chromatids attached to the kinetochore
3) merotelic attachment - more than one microtubule to same sister chromatid; chromosome loss at cytokinesis
4) amphelic attachment - normal

Aberrant centrosome/ DNA duplication

1) aberrant cell cycle - DNA and centrosome duplication —> 4 centrosomes
2) aberrant cytokinesis from multipolar spindle, chromosomes don’t know where to go

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

Describe anti-cancer therapy by inducing gross chromosome mis-segregations.

A
Checkpoint kinase (CHKE1 and CHKE2) - serine threonine kinase activation holds cells in G2 phase until all is ready 
inhibition leads to ultimately cell transition to mitosis 

Taxanes and vinca alkaloids (breast and ovarian cancers)

  • alters microtubule dynamics
  • produces unattached kinetochores
  • causes long-term mitotic arrest
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What happens if something goes wrong during the cell cycle?

A

1) Cell cycle arrest
- at check points (G1 and spindle check point)
- can be temporary (I.e. following DNA repair)

2) Programmed cell death (apoptosis)
-DNA damage too great and cannot be repaired
-chromosomal abnormalities
-toxic agents
Cell cycle progression aborted and cell destroyed

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

Describe the effects fo tumours on checkpoints.

A

1) G1 checkpoint - cells grow
2) G2 checkpoint - DNA damage not checked
3) Metaphase checkpoint - don’t check sister chromatids alignment
4) G0 - cell cycle apparatus not dismantled

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

What triggers a cell to enter the cell cycle and divide?

A

In the absence of stimulus, cells go into G0 (quiescent phase)

Exit from G0 highly regulated - requires growth factors and intracellular signalling cascades.

Signalling cascades:

  • response to extracellular factors
  • signal amplification
  • signal integration
  • modulation by other pathways
  • regulation of divergent responses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

How does protein phosphorylation after protein function?

A

Causing a change in shape (conformation) leading to change in activity (+ve or -ve)

Creating a docking site for another protein

In presence of ligand:

  • receptors form diners
  • are activated by phosphorylation

Receptor activation triggers:
-kinase cascades
-binding of adapter proteins
Kinases phosphorylation, phosphatases dephosphorylate

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

What are the main anti-cancer treatment modalities?

A

Surgery
Radiotherapy
Chemotherapy
Immunotherapy

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

What are the types of genetic mutations causing cancer?

A

Chromosome translocation
Gene amplification (copy number variation)
Point mutations within promoter or enhancer regions of genes
Deletions or insertions
Epigenetic alterations to gene expression

Can be inherited
Cancer is a disease of the genome

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

State what the systemic therapy in treating cancer involves.

A

Cytotoxic chemotherapy

1) alkylating agents
2) antimetabolites
3) anthracyclines
4) vinca alkaloids and taxanes
5) topoisomerase inhibitors

Targeted therapies

1) small molecule inhibitors
2) monoclonal antibodies

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Explain cytotoxic chemotherapy.

A

Cytotoxics select rapidly dividing cells by targeting their structures (mostly the DNA)

-Given intravenously or by mouth (occasionally)
-works systemically
-non targeted - affects all rapidly diving clels in body e.g. gut mucosa, bone marrow cells —> mucositis, BM suppression
-given post-operatively: adjuvant - mop up floating residual cells
Pre-operatively: neoadjuvant - tumours that are chemosensitive are given these to downstage, can reduce huge surgery like local incision
As monotherapy it in combination with curative or palliative intent

1) Alkylating agents
-Add alkyl (CnH2n+1) groups to guanine residues in DNA
-Cross-link (intra, inter, DNA-protein) DNA strands and prevents DNA from uncoiling at replication
-Trigger apoptosis (via checkpoint pathway)
-Encourage miss-painting - oncogenes (secondary malignancy)
E.g. chlorambucil, cyclophosphamide, dacarbazine, temozolomide

2) Pseudo-alkylating agents
-Add platinum to guanine residues in DNA
-Same mechanism of cell death as alkylating agents
E.g. carboplatin, cisplatin, oxaliplatin

Side effects of 1) and 2)cause hair loss (not carboplatin), nephrotoxicity, neurotoxicity e.g. peripheral neuropathy, ototoxicity (platinums), nausea, vomiting, diarrhoea, immunosuppression, tiredness

3) Anti-metabolites
-Masquerade as purine or pyramiding residues leading to inhibition of DNA synthesis, DNA double strand breaks and apoptosis.
-Block DNA replication (DNA-DNA) and transcription (DNA-RNA)
-Can be purine (adenine and guanine), pyramiding (thymine/ uracil and cytosine) or folate antagonists (which inhibit dihydrofolate reductase required to make folic acid, an important building block for all nuclei acids - especially thymine)
E.g. methotrexate (folate), 6-mercaptopurine, decarbazine and fludarabine (purine), 5-fluorouracil, capecitabine, gemcitabine (pyramidine)

Side effects
Hair loss (alopecia) – not 5FU or capecitabine Bone marrow suppression causing anaemia, neutropenia and thrombocytopenia
Increased risk of neutropenic sepsis (and death) or bleeding
Nausea and vomiting (dehydration)
Mucositis and diarrhoea
Palmar-plantar erythrodysesthesia (PPE) - red hands and red feet and skin begins to peel
Fatigue

4) anthracyclines
-inhibit transcription and replication by intercalating (I.e. inserting between) nucleotides within the DNA/ RNA strand
-also block DNA repair -mutagenic
-they create DNA and cell membrane damaging free oxygen radicals
E.g. doxorubicin, epirubicin

Side effects:
• Cardiac toxicity (arrythmias, heart failure) – probably
due to damage induced by free radicals
• Alopecia 
• Neutropenia 
• Nausea and Vomiting 
• Fatigue 
• Skin changes 
• Red urine (doxorubicin “the red devil”)

5) Vinca alkaloids and taxanes
- originally derived from natural sources
- work by inhibiting assembly (vinca alkaloids) or disassembly (taxanes) of mitotic microtubules causing dividing cells to undergo mitotic arrest

Side effects:
(Of microtubule targeting drugs in general too)
•Nerve damage: peripheral neuropathy, autonomic neuropathy
•Hair loss
•Nausea
•Vomiting
•Bone marrow suppression (neutropenia, anaemia etc)
•Arthralgia
•Allergy

6) Topoisomerase inhibitors
-Topoisomerases are required to prevent DNA torsional strain during DNA replication and transcription
-They induce temporary single strand (topo1) or double strand (topo2) breaks in the phosphodiester backbone of DNA
-they protect the free ends of DNA from aberrant recombination events
Anthracyclines cause permanent DNA damage
E.g. Topotecan and irinotecan (topo I) and etoposide (topo II) alter binding of the complex to DNA

Side effects:
• (irinotecan): Acute cholinergic type syndrome – diarrhoea, abdominal cramps and diaphoresis (sweating). Therefore given with atropine 
• Hair loss 
• Nausea, vomiting 
• Fatigue 
• Bone marrow suppression
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

If treatment will reduce someone’s chance of relapse with the disease by 30% and chance of dying by 20%. Is it worth the toxicity?

A

Yes because side effects can be controlled by drugs and not all are frequent.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

State some resistance mechanisms against chemotherapy.

A

DNA repair mechanisms upregulated and DNA damage is repaired —> stopping DNA double strand breaks

DNA adducts replaced by Base Excision repair (using PARP)

Drug effluxed from the cell by ATP-binding cassette (ABC) transporters.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Describe the problem present in non-monogenic cancers.

A

You can cut the wiring in monogenic cancers but for others parallel pathways or feedback cascades are activated

Nowadays we have dual kinase inhibitors which can prevent feedback loops but increase toxicities.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What are the hallmarks of cancer?

A

Used to be 6 now 10. First 6 is original

  1. Self –sufficient
    • Normal cells need growth signals to move from a
    quiescent (resting) to active proliferating state • These signals are transmitted into the cell via growth
    factors binding transmembrane receptors and
    activating downstream signalling pathways
  2. Insensitive to anti-growth signals
  3. Anti-apoptotic
  4. Pro-invasive and metastatic
  5. Pro-angiogenic
  6. Non-senescent
  7. Dysregulated metabolism
  8. Evades the immune system
  9. Unstable DNA
  10. Inflammation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What makes up a growth factor receptor?

A
Top to bottom:
Ligand-binding site
Membrane 
Kinase domain
C-terminal region with lots of tyrosine (for autophosphorylation)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Explain the over-expression of receptors in cancers.

A

HER2 – amplified and over-expressed in 25% breast cancer
EGFR – over-expressed in breast and colorectal cancer, lung cancer
PDGFR- glioma (brain cancer)
VEGF – prostate cancer, kidney cancer, breast cancer
FGFR (head and neck cancers, myeloma)

Therefore increased kinase cascade and signal amplification

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Explain targeted therapies.

A

1) Monoclonal antibodies
- momab (derived from mouse antibodies)
- ximab (chimeric) e.g cetuximab
- zumab (humanised) e.g. bevacizumab trastuzumab
- mumab (fully human) e.g. panitumumab

Humanized monoclonal antibody, murine regions (black) interspersed within the light (light gray) and heavy (dark gray) chains of the Fab portion.
Chimeric antibody murine component (black) of the variable region of the Fab section is maintained integrally.

Monoclonal antibodies target the extracellular component of the receptor
Neutralise the ligand
Prevent receptor dimerisation
Cause internalisation of receptor

Also activate Fcgamma-receptor-dependent phagocytosis or cytolysis induces complement-dependent cytotoxicity (CDC) or antibody-dependent cellular cytotoxicity

Examples: bevacizumab binds and neutralised VEGF, improving survival in colorectal cancer
Cetuximab targets EGFR

2)Small molecule inhibitors
Bind to the kinase domain of the tyrosine kinase within the cytoplasm and block autophosphorylation and downstream signalling.

Small molecule inhibitors act on receptor TKs but also intracellular kinases - therefore can affect cell signalling pathways

Examples of SMIs inhibiting receptors:
erlotinib (EGFR), gefitinib (EGFR), lapatinib (EGFR/HER2), sorafinib (VEGFR)

SMIs inhibiting intracellular kinases:
Sorafinib (Raf kinase) Dasatinib (Src kinase) Torcinibs (mTOR inhibitors)

By acting on receptors (either externally or internally), targeted therapies block cancer hallmarks (e.g VEGF inhibitors alter blood flow to a tumour, AKT inhibitors block apoptosis resistance mechanisms) WITHOUT the toxicity observed with cytotoxics
BUT CAN DEVELOP RESISTANCE AND EXPENSIVE

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Describe the resistance mechanisms against targeted therapies.

A

• Mutations in ATP-binding domain (e.g BCR-Abl fusion gene (Philadelphia) and ALK gene, targeted by Glivec - imatinib and crizotinib respectively)
• Intrinsic resistance (herceptin effective in 85% HER2+ breast cancers, suggesting other driving
pathways)
• Intragenic mutations
• Upregulation of downstream or parallel pathways

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

Describe anti-sense oligonucleotides and RNA interference.

A

• Single stranded, chemically modified DNA-like molecule 17-22 nucleotides in length
• Complementary nucleic acid hybridisation to target gene hindering translation of specific
mRNA
• Recruits RNase H to cleave target mRNA
• Good for “undruggable” targets

RNA interference
• Single stranded complementary RNA
• Has lagged behind anti-sense technology –
especially in cancer therapy
• Compounds have to be packaged to prevent
degradation - nanotherapeutics
• CALAA-01 targeted to M2 subunit of
ribonucleotide reductase. Phase I clinical trials in cancer –results awaited

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What is the restriction point?

A

It is found in G1 and cell monitors its own size and external signals.

The cell is now committed in the cell cycle and does not require any more GF’s.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What is c-Myc?

A

An oncogene over expressed in many tumours

A transcription factor which stimulates the expression of cell cycle genes and is need to enter S phase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

Describe adaptor proteins.

A

Tyrosine phosphorylation provides docking sites for adapter proteins

Protein-protein interactions: protein binding – bringing proteins together

Proteins are modular and
contain domains, i.e. functional and structural units that are copied in many proteins

Some domains are important in
molecular recognition – have no enzymatic function of their own, simply bring other proteins together

E.g. Grb2 binds to SH3 - proline-rich regions (constitutive) and SH2 (phosphorylation tyrosine (transient)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What activates and inactivated RAS?

How can RAS be oncogenically activated by mutations?

A

1) exchange factors e.g. SOS activates
2) GTPase activating proteins inactivate
RAS must be bound to plasma membrane to be activated

Oncogenically activated:
To increase the amount of active GTP-loaded Ras by:
-preventing GAP binding
-preventing GTP hydrolysis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

What is ERK?

A

Extracellular signal-regulated kinase

It’s a type of MAPK cascade (mitogen-activated protein kinase)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

Describe Cdks and cyclins.

A

Cdks (cyclin-dependent kinases)

Present in proliferating cells throughout cell cycle
Activity is regulated by:
-interaction with cyclins
-phosphorylation

Cyclins

  • transiently expressed at specific at specific points in the cell cycle
  • regulated at level of expression
  • synthesised then degraded

Cyclins bind to and activate Cdk(s) triggering different events in the cell cycle

Cdks phosphorylation proteins (on serine or threonine) to drive cell cycle progression
E.g. nuclear lamins (causes breakdown of nuclear envelope)
E.g. retinoblastoma protein (pRb) - tumour suppressor - inactivated in many cancers

Regulation of Cdks by phosphorylation
Requires activating phosphorylation AND removal of inactivating phosphorylation

Cyclins activate Cdks but also alter substrate specificity
Substrate accessibility changes through cell cycle

Cyclical activation - Cdks become sequentially active and stimulate synthesis of genes required for next phase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

Describe what happens at the anaphase checkpoint in the metaphase-anaphase transition.

A

Cdk1/cycB active. Mitosis on hold - key substrates phosphorylated

Signal from fully attached kinetochores causes cyclin B to be degraded:

  • Cdk1 inactivated
  • key substrates dephosphorylated
  • mitosis progresses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

Describe the two CKI families.

A

INK4 family and CIP/KIP family

G1 phase CKIs - inhibit Cdk4/6 by displacing cyclin D
S phase CKIs - inhibit all Cdks by binding to the Cdk/cyclin complex
CKI must be degraded to allow cell cycle progression

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

Describe cell cycle regulatory proteins and cancer.

A

Oncogenes (derived from normal proto-oncogenes)
-EGFR/HER2, mutationally activated or overexpressed in many breast cancers (herceptin antibody for the treatment of HER2-positive metastatic breast cancer)
-Ras, mutationally activated in many cancers (inhibitors of membrane attachment)
-cyclin D1, over expressed in 50% of breast cancers
-B-Raf, mutationally activated in melanomas (kinase inhibitors in trials)
-c-Myc, overexpressed in many tumours
Higher up in signal, harder to treat because affects more pathways

Tumour suppressors

  • RBC, inactivated in many cancers
  • P27KIP1, underexpression correlates with poor prognosis in many malignancies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What are the types of DNA damage?

A

Carbon ring structures can be easily activated chemically because chemically reactive

Also can interchange easily e.g. thymine —> uracil by removal of methyl group.

Base modifications prevent replication or cause mutations:
Deamination - the primary amino groups of nucleic acid bases are somewhat unstable. They can be converted to ketogroups in reactions like cytosine to uracil

Chemical modification - e.g. hyper-reactive oxygen (hydrogen peroxide, peroxide radicals .etc.) can modify DNA bases. A common product of thymine oxidation is thymine glycol.
Hyper-reactive oxygen species are also generated by ionising radiation (x-rays, gamma rays)
Many environmental chemicals can modify DNA bases (including food), frequently by addition of a methyl or other alkyl group (alkylation). Addition of larger molecules defines “adducts” - carcinogens can cause them

Photodamage - ultraviolet light is absorbed by the nuclei acid bases, and the resulting influx of energy can induce chemical changes. The most frequent photo products are the consequences of bond formation between adjacent pyramid Ines with one strands (intra-DNA damage) e.g. thymidine react with each other to form thiamine dimer

Nick - radiation can break phosphodiester bond
Gap - lots of breaks/nicks
Thymine dimer - distortion of helix
Base pair mismatch - because of change in base

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What are the causes of DNA damage?

A

Chemicals (carcinogens)

  • dietary
  • lifestyle
  • environmental
  • occupational
  • medical
  • endogenous - metabolisms??

Radiation
-ionising
Generates free radicals in cells including oxygen free radicals - super oxide radical O2• , hydroxyl radical HO•
Possess unpaired electrons - electrophilic and therefore seek out electron rich DNA - cause nicks e.g. ring-opened guanine and adenine
-solar
Form pyramidine (thymine) diners - skin cancer
-cosmic

Damage from carcinogens:

  • DNA adducts and alkylation
  • Base diners and chemical cross-links
  • Base hydroxylations (reactive oxygen) and abasic sites formed (base destroyed but DNA structure underlying it intact)

The importance of DNA damage:

  • DNA damage can lead to mutation
  • mutation may lead to cancer
  • damaging DNA is an important strategy in cancer therapy (chemotherapy)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

Explain the role of metabolism in DNA damage.

A

Mammalian metabolism
Phase I - liver
-addition of functional groups e.g. oxidation, reduction, hydrolysis
-mainly cytochrome p450 mediated
Phase II
-conjugation of phase I functional groups e.g. sulphation, glucoronidation, acetylation, methylation, amino acid and glutathione conjugation (generates polar -water soluble metabolites)

Polycyclic aromatic hydrocarbons:
-common environmental pollutants
-formed from combustion of fossil fuels and tobacco
E.g.
Two step epoxidation of B[a]P - mutation in lung cancer causes by tobacco smoke
Epoxidaiton of aflatoxin B1 - formed by Aspergillus flavus mould, stored peanuts
Metabolism of 2-napthylamine - past components of dye-stuffs, bladder cancer - nitrenium ion end up in urine and change pH causing cancer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

Explain the repair of DNA damage.

A

The greater the persistence of damage then the greater the chance of mutagenic event.

P53 is a tumour suppressor gene, p53 is kept inactive by MDM2. MDM2 lost if p53 activated
P53 is a TF so can turn on different pathways to respond to damage e.g. DNA repair in response to DNA replication stress, double-strand breaks .etc.

Types of DNA repair:

  • direct reversal of DNA damage
  • Base excision repair (mainly for apurinic/apyrimidinic damage - lost base)
  • nucleotide excision repair (mainly for bulky DNA adducts)
  • during or post replication repair
  • DNA mismatch repair

DNA mismatch repair:
-mismatches that arise during replication are corrected by comparing the old and new strands (proof-reading)
Bulge in DNA = wrong base, recognised by MSH and MCH protein, bind to bulge, nucleus cuts out base and then polymerase restores correct sequence
-other systems deal with mismatches generated by base conversions such as those which result from deamination

Direct DNA repair:
Involves the reversal or simple removal of the damage by the use of proteins which carry out specific enzymatic reactions
E.g. photolyases (activated by normal light, not UV) repair thymine diners

Excision repair of DNA damage:
For nicks
1) Damage to G but not to phosphodiester backbone
2) remove base without affecting phosphodiester bone by DNA-glycosylase
3) AP-endonuclease cuts open nuclear strand
4) DNA polymerase adds correct base
5) DNA lipase

For gaps, e.g. caused by big adduct groups

1) damage to both base and DNA
2) endonuclease - forms nicks around mutation
3) helicase removes phosphodiester bond and bases
4) DNA polymerase corrects base
5) DNA ligase

Double strand break repair:
Double-strand breaks are made:
• Under physiological conditions during
somatic recombination and transposition. e.g.
Transient base pairing of V(D)J recombination
• During Homologous Recombination.
• As a result of ionizing radiation and oxidative
stress induced DNA damage.

1) following a double sytrand break, a 3’-5’ exonuclease exposes a 5’ single-strand overhand until it finds a sequence which matches together
2) transient base pairing of several nucleotides enables ends to come together
3) DNA polymerisation
4) nucleolytic processing
5) ligation

Non-homologous repair- if oevrlangs font match, Ku proteins hold DNA together forcing repair

Summary:
Direct reversal of DNA damage
▪ photolyase splits cyclobutane pyrimidine-dimers
▪ methyltransferases & alkyltransferases remove alkyl groups from bases
Base excision repair (mainly for apurinic/apyrimidinic damage)
▪ DNA glycosylases & apurinic/apyrimidinic endonucleases + other
enzyme partners
▪ A repair polymerase (e.g. DNA Polb) fills the gap and DNA ligase
completes the repair.
Nucleotide excision repair (mainly for bulky DNA adducts)
▪ Xeroderma pigmentosum proteins (XP proteins) assemble at the damage
.A stretch of nucleotides either side of the damage are excised.
▪ Repair polymerases (e.g. DNA Pold/b) fill the gap and DNA ligase complete the repair.
During- or post-replication repair
▪ mismatch repair
▪ recombinational repair

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What are some human genetic disease involving nucleotide excision repair (NER)?

A
  • Xeroderma Pigmentosum
  • severe light sensitivity
  • severe pigmentation irregularities
  • early onset of skin cancer at high incidence • elevated frequency of other forms of cancer • frequent neurological defects
Trichothiodystrophy
• sulphur deficient brittle hair
• facial abnormalities
• short stature
• ichthyosis (fish-like scales on the skin)
• light sensitivity in some cases 
Cockayne's syndrome
• dwarfism
• light sensitivity in some cases
• facial and limb abnormalities
• neurological abnormalities
• early death due to neurodegeneration
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

What are the consequences of DNA damage?

A

Normally:
Carcinogen damage leading to altered DNA —> apoptosis —> cell death

Carcinogen damage leading to altered DNA —> efficient repair —> normal cell

Problem:
Carcinogen damage leading to altered DNA —> incorrect repair/ altered primary sequence —> DNA replication and cell division: fixed mutations —> transcription/translation giving aberrant proteins OR carcinogenesis if critical targets are mutated: oncogenes, tumour suppressor genes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

Give examples of therapeutic agents which cause DNA damage.

A

Alkylating agents - alkylate to cause DNA damage and apoptosis

Agents that make bulky agents e.g. cisplatin

Agents that induce double strand breaks e.g. ionising radiation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

Explain carcinogen testing.

A

Testing for DNA damage:
in vitro bacterial gene mutation assay —> in vitro mammalian cell assay —> in vivo mammalian assay —> investigate in vivo mammalian assay

Bacterial (Ames) test for mutagenicity of chemicals

1) chemical to be tested and rat liver enzyme preparation-s9 (cytochrome p450 activity) added into bacteria that do not synthesis histidine(aa required to grow) e.g. salmonella strain -mutation
2) conversion of chemical to active metabolism
3) carcinogenic agents may correct defective mutation in salmonella, bacteria acquire ability to synthesise histidine and colonies can be seen on Petri dish

Detecting DNA damage in mammalian cells -chromosomal aberrations:
Treat mammalian cells with chemical in presence of liver s9. Look for chromosomal damage

In vitro micronucleus assay:
Cells treated with chemical and allowed to divide
Binucleate cells assessed for presence of micronuclei
Can stain the kinetochore proteins to determine if chemical treatment caused clastgenicity (chromosomal breakage) or aneuploidy (chromosomal loss)
If DNA becomes very damaged, it gets budded as micronucleus

Murine bone marrow micronucleus assay:
Treat animals with chemical and examine bone marrow cells or peripheral blood erythrocytes for micronuclei

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

Why does programmed cell death occur?

A

1) harmful cells (e.g. cells with viral infection, DNA damage)
2) developmentally defective cells (e.g. B lymphocytes expressing antibodies against self antigens)
3) excess/ unnecessary cells: (embryonic development: brain to eliminate excess neurones; liver regeneration; sculpting of digits and organs)
4) obsolete cells (e.g. mammary epithelium at the end of lactation)
5) exploitation - chemotherapeutic killing of cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

Describe the difference between necrosis and apoptosis.

A

Necrosis - unregulated cell death associated with trauma, cellular disruption and an inflammatory response

Apoptosis (programmed cell death) - regulated cell death; controlled disassembly of cellular contents without disruption; no inflammatory response

Necrosis:

  • plasma membrane becomes permeable
  • cell swelling and rupture of cellular membranes, organelles swell, chromatin condenses
  • release of proteases leading to autodigestion and dissolution of the cell
  • localised inflammation

Apoptosis:
Latent phase - death pathways are activated, but cells appear morphologically the same

Execution phase

  • loss of microvilli and intercellular junctions - permeability of epithelium compromised
  • cell shrinkage, epithelium closes around - permeability resorted
  • loss of plasma membrane asymmetry (phosphatidylserine lipid appears in outer leaflet) - imbalance in lipid composition
  • chromatin and nuclear condensation
  • DNA fragmentation
  • Formation of membrane blebs
  • fragmentation into membrane-enclosed apoptotic bodies
  • apoptosis bodies phagocytosis by neighbouring cells and macrophages

Plasma membrane remains intact - no inflammation

Apoptosis-like PCD - some, but not all, features of apoptosis. Display of phagocytise recognition molecules before plasma membrane lysis

Necrosis-like PCD - variable features of apoptosis before cell lysis; “aborted apoptosis”

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What are the mechanisms of apoptotic cell death?

A
  1. The executioners – Caspases
  2. Initiating the death programme
    • Death receptors
    • Mitochondria
  3. The Bcl-2 family
  4. Stopping the death programme

1) cysteine-dependent aspartate-directed proteases - cleave at specific sites
-executioners of apoptosis
-activated by proteolysis
-cascade of activation
Inactive at start - regulation, autofolded on itself
Initiator and effector; initiator = trigger apoptosis by cleaving and activating; effector = carry out the apoptotic programme

Cleave at:

  • CARD (cascade recruitment domain)
  • DED (death effector domain)
  • homotypic (same type) protein-protein interactions e.g. cascade would dimerise with cascade
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

Describe how effector caspases execute the apoptotic programme.

A

Cleave and inactivate proteins or complexes (e.g. nuclear lamins leading to nuclear breakdown) -loss of function

Activate enzymes (incl. protein kinases; nucleases .e.g. Caspase-Activated DNase, CAD) by direct cleavage, or cleavage of inhibitory molecules - gain of function

Both the above apply to monomeric substrates and multiprotein complexes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

What are the two mechanisms of caspase activation?

A

Death by design - receptor-mediated (extrinsic) pathways

Death by default - mitochondrial (intrinsic) death pathway

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

State the Bcl-2 family proteins.

A

Anti-apoptotic - promote cell survival (mitochondrial)
Bcl-2
Bcl-xL

Pro-apoptotic (move between cytosol and mitochondria)
Bid
Bad
Bax
Bak
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

How does PKB/Akt induce cell survival by blocking apoptosis?

A
  1. Phosphorylates and inactivates Bad 2. Phosphorylates and inactivates caspase 9
  2. Inactivates FOXO transcription factors (FOXOs promote
    expression of apoptosis-promoting genes)
  3. Other, e.g. stimulates ribosome production and protein
    synthesis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

Describe inhibitor of apoptosis proteins (IAPs).

A

They regulate programmed cell death in the extrinsic pathway by:

  • binding to procaspases and preventing activation
  • binding to active caspases and inhibit their activity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

State the cytoprotective/ anti-apoptotic pathways.

A

Bcl-2, Bcl-xL: intrinsic pathway

FLIP, IAPs: extrinsic pathway

Growth factor pathways via PI3’K and PKB/Akt

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

What are the therapeutic uses of programmed cell death?

A

Harmful (oncogenic) cells e.g. cells with viral infection, DNA damage

Chemotherapeutic killing of tumour cells e.g. dexamethasone stimulates DNA cleavage

58
Q

What are the 3 steps making a blood vessel?

A

Vasculogenesis (bone marrow progenitor cell)
Angiogenesis (sprouting)
Arteriogenesis (collateral growth)

You have inhibitors (e.g.angiostatin) activators (VEGF) and factors required for maturation and integrity (platelets) to regulate angiogenesis.

59
Q

Explain sprouting in angiogenesis.

A

Hypoxia is a trigger for angiogenesis:

  • HIF: hypoxia-inducible transcription factor, controls regulation of gene expression by oxygen
  • pVHL: Von Hippel-Lindau tumour suppressor gene, controls levels of HIF - inhibits HIF when needed. In hypoxia, VHL detaches from HIF, binds to DNA —> VEGF

VEGF and its receptors

  • 5 members = VEGF-A, VEGF-B, VEGF-C, VEGF-D and placental growth factor (PIGF)
  • three tyrosine kinase receptors: VEGFR-1, VEGFR-2, VEGFR-3 and co-receptors (Nrp1 and Nrp2)
  • VEGFR-2 is the major mediator of VEGF-dependent angiogenesis, activating signalling pathways that regulate endothelial cell migration, survival, proliferation

1) tip/stalk selection - one cell gets bound by GF and forms tip, cells sitting below = stalk and push tip up
VEGF binds to cell —> top cell —> drives formation of new blood vessels and tells neighbouring cells to become stalk cells

VEGF/Notch signalling:
I) in stable blood vessels, DII4 and natch signalling maintain quiescence
II) VEGF activation increases expression of DII4
III) DII4 drives Notch signalling, which inhibits expression of VEGFR2 in the adjacent cell
IV) DII4-expressing tip cells acquire a motile, invasive and sprouting phenotype
V) Adjacent cells (stalk cells) form the base of the emerging sprout, proliferate to support sprout elongation

2) tip cell navigation and stalk cell proliferation
3) branching coordination
4) stalk elongation, tip cell fusion, and lumen formation
5) perfusion and vessel maturation
6) stabilisation and quiescence

60
Q

Describe the role of macrophages in vessel anastomosis.

A

Macrophages play a significant role in both physiological and pathological angiogenesis

Macrophages carve out tunnels in the ECM providing avenues for capillary infiltration

Tissue-resident macrophages can be associated with angiogenic tip cells during anastomosis.

61
Q

Describe the role of platelets in angiogenesis.

A

Modulator role

Haemostasis (physiological angiogenesis)

  • vascular development
  • wound healing

Thrombosis (pathological angiogenesis)

  • atherosclerosis
  • rheumatoid arthritis
  • diabetic retinopathy
62
Q

Explain stabilisation and quiescence in sprouting.

A

Form endothelial junctions

Tight junctions and adherens junctions in endothelial cells

  • constitutively expressed at junctions
  • homophonic interaction mediates adhesion between endothelial cells and intracellular signalling
  • controls contact inhibition of cell growth
  • promotes survival of EC

Mural cells (pericytes) hep to stabilise the neovessels

  • pericytes produce molecules to stabilise such as angiopoietin/Tie-2 system
  • VEGF pathway drives things, angiopoietin/Tie-2 act as modulators

Angiopoietin-Tie2 ligand-receptor system

  • Ang-1 and Ang-2 are antagonistic ligand of the Tie2 receptor
  • Ang-1 binding to Tie2 promotes vessel stability and inhibits inflammatory gene expression
  • Ang-2 antagonises Ang-1 signalling, promotes vascular instability and VEGF-dependent angiogenesis
63
Q

When do tumours require angiogenesis?

A

Tumours less than 1 mm^3 receive oxygen and nutrients by diffusion from host vasculature - don’t need angiogenesis

Larger tumours require new vessel network. Tumour secretes angiogenic factors that stimulate migration, proliferation and neovessel formation by endothelial cells in adjacent established vessels - don’t release all factors, not same as normal physiology

New vascularised tumour no longer relies solely on diffusion from host vasculature, facilitating progressive growth

The angiogenic switch - a discrete step in tumour development that can occur at different stages in the tumour-progression pathway, depending on the nature of the tumour and its microenvironment

64
Q

Describe tumour blood vessels.

A
  • irregularly shaped, dilated, tortuous
  • not organised into definitive venues, arteriolar and capillaries
  • leaky and haemorrhagic, partly due to the overproduction of VEGF
  • perivascular cells often become loosely associated
  • some tumours may recruit endothelial progenitor cells from the bone marrow
65
Q

Describe the multicellular response which promotes tumour angiogenesis and the role of platelets.

A

Cancer associated fibroblasts (CAFs) secrete ECM, pro-angiogenic growth factors e.g. VEGFA

Pericytes are loosely associated with tumour-associated blood vessels (TABVs) and this favours chronic leakage in tumours. This is enhanced by angiopoietin 2.

Platelets
-link between cancer progression and thrombocytosis
-activated platelets are a source of:
Pro-angiogenic factors: VEGFA, platelet-derived growth factors (PDGFs), FGF2
Angiostatin molecules: thrombospondin 1, plasminogen activator inhibitor 1 (PAI1), endostatin
-pro-angiogenic mediators and proteases support the proliferation and activation of CAFs
-tumours cause platelet activation, aggregation and degranulation
-disrupting platelet function does not obviously impaired tumour angiogenesis, however the overall outcome of platelet activation in tumours appears to be pro-angiogenic

66
Q

What are the therapeutic strategies to target tumour angiogenesis?

A

Therapies to inhibit VEGF signalling:

  • antibodies against VEGF
  • molecules affecting signalling pathways
  • molecules blocking receptors

VEGF inhibition by soluble VEGFR1 (Flt-1) reduces tumour growth

1) cells stably transferred with control or sFlt-1 plasmid to promote Flt-1 (VEGFR1) expression
2) VEGFR1 binds to VEGF and mops it up preventing it from stimulating angiogenesis
3) Flt-1 expression reduces tumour growth in vivo, without affecting tumour cell growth in vitro: effect on vasculature

Anti-VEGF humanised MAb: Avastin 
Side effects and limited efficacy
-no overall survival advantage over chemo alone 
Side effects:
-GI perforation 
-hypertension 
-proteinuria 
-venous thrombosis 
-haemorrhage 
-wound healing complications 

Anti-angiogenic therapies: strategies
Anti-angiogenic therapy which normalises vasculature
-reduces hypoxia
-incraese efficacy of conventional therapies

Sustained/aggressive anti-angiogenic therapy
-may damage healthy vasculature leading to loss of vessels, creating vasculature resistant to further treatment and inadequate for delivery of oxygen/ drugs.

67
Q

What are potential mechanisms of resistance to anti-VEGF therapy in cancer?

A

VEGF inhibition aggravates hypoxia because block blood vessels, increasing tumour’s production of other angiogenic factors or increases tumour invasiveness

Tumour vessels maybe less sensitive to VEGF inhibition due to vessel lining by tumour cells or endothelial cells derived from tumours

Tumour cells that recruit pericytes maybe less responsive to VEGF therapy

Tumour cell vasculogenic mimicry (vascular mimicry) describes the plasticity of aggressive cancer cells forming de novo vascular
networks

68
Q

Describe the future of tumour-angiogenesis therapies.

Describe anti-angiogenic therapy in other diseases.

A

Novel molecular mechanism (non-VEGF targets)

Other diseases:
Age-related macular degeneration (AMD)
-abnormal growth of choroidal blood vessels
-leaky vessels cause oedema 
-visual impairment 

Anti-VEGF therapy: Lucentis used for this
Avastin is used off-label

69
Q

Why is it hard to find therapeutic strategies against cancer?

A

Tumours are complex 3D structures with their own unique micro environments
Studies are performed in 2D
Also, tumours receive nutrients and therapeutics through the vasculature which is not included in any in vitro tumour models

Tumour-on-a-chip platform develops a micro physiological system that incorporates human cells in a 3D ECM, supported by perfumed human microvessels

70
Q

What are the molecular mechanisms that regulate motility?

A

Microfilaments
Regulation of actin dynamics
Cytoskeleton proteins
Signalling proteins

71
Q

What are the steps in tumour progression?

A

1) homeostasis
2) genetic alterations
3) hyper-proliferation
4) de-differentiation - disassembly cell-cell contacts, loss polarity
5) invasion - increased motility, cleavage ECM proteins
Form a tumour mass - benign and break the basement membrane/ dissociate completely have high motility and cleave ECM proteins —> essential so they can migrate

72
Q

Describe the distinct and sequential events involved in metastasis.

A

1) epithelial cells in primary tumours are tightly bound together
2) metastatic tumour cells become mobile mesenchyme-type cells and enter the bloodstream
3) metastatic cells then travel through the bloodstream to a new location in the body
4) metastatic cells exit the circulation and invade a new organ
5) cancer cells lose their mesenchymal characteristics and form a new tumour

73
Q

Describe the types of tumour cell migration.

A

Two types individual and collective

Individual

  • ameoboid e.g. lymphoma, leukaemia
  • mesenchymal (single cells)

Collective

  • cluster/cohorts - epithelial cancer, melanoma
  • multicellular strands/ sheets -epithelial cancer, vascular tumours

Factors involved:

  • integrins - receptors of ECM proteins
  • proteases - digest ECM
  • cadherins - induce differentiation in epithelial cells - metastatic collective cells grace these so they can interact with the other cells and be transported - pull cells together
  • gap junctions - allow communication with each other

The above factors are higher in collective cells than indict dual so their metastatic potential is much higher than individual cells - can still maintain attachment with each other while being transported.

74
Q

Describe tumour cell metastasis and how they mimic morphogenic events.

Describe the administration of EGF.

A

1) cells migrate and drag cells behind
2) differentiate to produce alveoli for lactation
3) cut vasculature => bruise - need to regrow - leader cells (tip cells) drives cells to vasculature
4) detached cluster
Similar to mammary glands but tumour cells have no sense of direction
E.g. migration of primary glial cells - recognise neighbouring cells and stop in scratch wound
Migration of a glial tumour cell - cells continue to grow and don’t recognise neighbouring cells

EGF leads to upregulation of genes involved in:
-cytoskeleton regulation
-motility machinery
Tumour cells metastasis

75
Q

Describe the basics of cell movement including stimuli needed to move.

Describe the attachments.

Decesirbt eh control of cell movement.

A

Stimuli to move:

  • organogenesis and morphogenesis
  • wounding
  • growth factors/ chemoattractants
  • dedifferentiation (tumours)

Where to go? Directionality (polarity)
When to stop? Contact-inhibition motility - recognition of neighbouring cells
How to move? Specialised structures (focal adhesion, lamellae, filopodium

Attachment to substratum (ECM proteins)
Cells attach to ECM via integrins
-focal adhesions - close contact with ECM
-filamentous actin - organised in bundles at specific orientations

Structures shed got motilityL

  • filopodia: finger-like protrusions rich in actin filaments
  • lamellipodia: sheet-like protrusions rich in actin filaments - cells can form contacts over larger area of cell

Control is needed:

  • within a cell to coordinate what is happening in different parts
  • regulate adhesion/ release of cell-extracellular matrix receptors
  • from outside to response to external influences - sensors e.g. GF and nutrients; directionality

Motility: hapoptatic (stroll in park) versus chemotatic (purpose - go to stops, drives cell to specific location)

76
Q

Explain the steps in cell motility.

A

1) extension - can produce lamellar to go forward
2) adhesion - stabilise adhesion
3) translocation
4) de-adhesion

Like rock climbing:

  • filopodia - sense rock
  • lamellar - grab rock, translocate - push yourself up
77
Q

Explain actin filaments.

A
G-actin = small soluble subunits 
F-actin = large filamentous polymer 

Actin filament has polarity - plus and minus end

Signal such as a nutrient source —> disassembly of filaments and rapid diffusion of subunits —> reassembly of filaments at a new site

Filament organisation and structure:

  • lamellipodium: branched and crosslinked filaments
  • filopodium: bundle of parallel filaments
  • stress fibres - antiparallel contractile strictures (thick bundle of actin)

Depending on the proteins they bind to, actin filaments have different functions and are in different states e.g. motor proteins (contract), bundling, cross-linking, nucleating, capping

78
Q

Describe the steps in actin remodelling.

A

1) Nucleation
For polymerisation to occur, you need nucleating of 3 monomers together - not very stable
Arp3 + Arp2 and other proteins —> ARP complex
ARP complex + actin monomers —> nucleated actin filament

Limiting step in actin dynamics - formation of trimmers to initiate polymerisation

2) elongation
Free actin monomer —> actin filament via actin-profilings complex
Actin-thymosin complex blocks, restricts availability of G monomers to form actin filament

Profilin competes with thymosin for binding to actin monomers and promotes assembly
Monomer binding: profilin
Sequestering: b4-thymosin, ADF/cofilin (do not inhibit polymerisation)

3) capping 4) severing
Stop growing of filament
Like bottle cap at end to prevent addition of monomers
Capping proteins:
+ end = Cap Z, gelsolin, fragments/severin
- end = tropomodulin, Arp complex

Severin proteins:
Gelsolin
ADF/ cofilin
Fragmin/severin

Depending on stimulus = capping or severing
If not chopped, straight to monomeric form then they can form adhesion points for growth
Need coordination between capping and severing

In unsevered population, actin filaments grow and shrink relatively slowly
In severed population, actin filaments grow and shrink more rapidly

5) cross-linking and bundling 
Group filaments 
Proteins involved:
-a-actinin = dimer cross-link filament 
-fibrin
-filamin - angle 
-spectrin = stabilise cortico structures of cells
-villin
-vinculin 
-dystrophin - link filaments to plasma membrane 

6) branching
Branching protein = Arp complex
Branching at 70 degrees

7) gel-sol transition by actin filament severing
The network is maintained at some sort of rigidity of membrane in cytoskeleton
If cell needs to protrude membrane/ cytoplasm - need to loosen up —> cleave some crosslink proteins so they are not connected to each other
Gel (rigid) —> sol (can flow)

79
Q

What are diseases caused by deregulation of actin cytoskeleton?

A

High blood pressure - requires contraction of blood vessels
Duchenne Muscular Dystrophy (muscle wasting) -dystrophin protein truncated
Bulbous Pemphigoid (autoimmune disease) -epithelial connection to basement membrane - destabilised
Alzheimer’s (neurodegenerative - depositis of protein - cytotoxicity

80
Q

Describe how different actin remodelling is used in cells.

A

Disassembly, nucleation, branching, severing, capping and bundling used in extension
Polymerisation also in extension

Gel/sol transition and attachment ECM in adhesion

Contraction in translocation

Detachment in de-adhesion

Lamellar protrusion - polymerisation, disassembly, branching, capping
Filopodia - actin polymerisation, bundling, crosslinking

81
Q

Describe the signalling mechanisms that regulate the actin cytoskeleton.

A

1) ion flux changes (I.e. intracellular calcium - cytoskeleton proteins can bind to Ca2+ to activate protein)
2) phosphoinositide signalling (phospholipid binding) - P13K signalling, cytoskeleton proteins can change properties when bound
3) kinases/phospjhatases (phosphorylation cytoskeleton proteins) e.g. MAPK
4) signalling cascades via small GTPases

82
Q

Describe the control of actin cytoskeleton by small G proteins.

How does signalling from small GTPases regulate actin cytoskeleton and motility?

A

Rho subfamily (found in GDP bound form - activated as GTP bound form) of small GTPases belongs to the Ras super-family
Family members: Rac, Rho, Gdc42
Rho is upregulated in cancers
Inactivated by release of Pi

Participate in a verity of cytoskeletal processes
These proteins are activated by receptor tyrosine kinase, adhesion receptors and signal transduction pathways
Expression levels up-regulated in different human tumours

Activation of:
Cdc42 —> filopodia
Rac—> lamellipodia (large lamellar not localised)
Rho —> stress fibres (lots of force when contract)

Actin binding proteins regulated by Rac/cdc42

1) extension - Rac in actin polymerisationa nd branching
2) adhesion - Rac, Rho in focal adhesion, assembly
3) translocation - Rho in stress fibres, tension, contraction
4) De-adhesion - Rho

Cdc42 - filopodia and polarised mobility

83
Q

What is cell behaviour?

What external influences are detected by cells?

What external factors can influence cell division?

A

Term used to describe the way cells interact with their external environment and their reactions to this, particularly proliferative and motile responses of cells

Chemical: hormones, growth factors, ion concs, ECM, molecules on other cells, nutrients and dissolved gas (O2/CO2) concs
Physical: mechanical stresses, temperatures, the topography or layout of the ECM and other cells

Although all external factors may influence cell
proliferation, the ones to be considered here, in relation to cancer cell behaviour, are:-
• Growth factors
• Cell-cell adhesion
• Cell-ECM adhesion

Lamellipod allows adhesion of tissue cells to matrix surface

Cell-spreading is not a passive, gravity dependent event. Energy is required to modulate cell adhesion and the cytoskeleton during spreading.
When cell spreads, less blebbing

Cells require to be binding to extracellular matrix to be fully competent for responding to soluble growth factors.

• in suspension, cells do not significantly
synthesise protein or DNA 
• cells require to be attached to ECM (
and a degree of spreading is required)
to begin protein synthesis and
proliferation (DNA synthesis) 
• attachment to ECM may be required for
cell survival .i.e. anchorage dependence
Most cells need matrix
84
Q

Describe how cell phenotype can be determined by the composition of the matrix.

How do cells receive information about their surroundings from adhesion to ECM?

A

(A) in interstitial matrix (type 1 collagen), mammary epithelium does not differentiate to secretory cells

(B) in basal lamina (basement membrane) matrix, mammary cells organise into “organoids” and produce milk proteins.

Cell-ECM adhesion molecules
• Cells have receptors on their cell surface
which bind specifically to ECM molecules
• these molecules are often linked, at their
cytoplasmic domains, to the cytoskeleton
• this arrangement means that there is
mechanical continuity between ECM and
the cell interior

85
Q

Explain integrins including signalling to and from ECM receptors.

A

Integrins are heterodimer complexes of a and b subunits that associate extracellularly by their “head” regions. Each of the “leg” regions spans the plasma membrane.

Ligand-binding occurs at the junction of the head regions

Integrins
• more than 20 combinations of a/b known which each bind
specifically to short peptide sequence on ECM proteins • For example a5b1 fibronectin receptor binds arg-gly-asp (
RGD) • peptide sequences such as RGD are found in more than
one ECM molecule, e.g. RGD found in fibronectin,
vitronectin, fibrinogen plus others

• most integrins link to the actin cytoskeleton via
Focal adhesions actin-binding proteins (exeption: a6b4 integrin
(green)
complex found in epithelial hemidesmosomes,
linked to the cytokeratin (intermediate filament)
network)
• integrin complexes cluster to form focal
adhesions (most) or hemidesmosomes (a6b4)
• these clusters are involved in signal
transduction
• Some integrins also bind to specific adhesion
molecules on other cells (e.g. avb3 binds to
PECAM-1(CD31) and aIIbb2 to ICAM-1 on endothelial cells in inflammation) - important in immune system and blood clotting

Signalling to and from ECM receptors
• ECM receptors (e.g. integrins) can act to
transduce signals
• e.g. ECM binding to an integrin complex
can stimulate the complex to produce a
signal inside the cell,
• i.e. “outside-in” integrin signalling

Integrin signalling involves conformational changes to the complex
Integrin complexes can adopt “flexed” and “extended” molecular confirmations. Switching between these confirmations affects their ability to bind their ligands, and their signalling. In this way, cell-ECM adhesion, and signals, can be switched on and off.

Outside-in signalling
• a cell can receive information about its
surroundings from its adhesion to ECM
• e.g. the composition of the
ECM will determine which integrin complexes bind and which signals it receives
• this can alter the phenotype of the cell

The amount of force that is generated at a focal adhesion depends on both the force generated by the cytoskeleton (F cell) and the stiffness of the ECM
Some proteins can stretch and open up, exposing new sites

Integrins recruit cytoplasmic proteins which promote both signalling and actin assembly

Signalling to and from ECM receptors
a signal generated inside the cell (e.g. as the result of hormone binding to receptor) can act on an integrin complex to alter the affinity of an integrin (i.e. alter its affinity for its ECM
binding)

this is “inside-out” intergin signalling (e.g. in
inflammation or blood-clotting, switching on
adhesion of circulating leukocytes)

Summary of the conformational changes to the integrin complexes during activation and signalling:
-low affinity = bent confirmation, weak or no binding to ligand
High affinity = extended confirmation, strong binding to ligand

  • ECM ligand binds and causes the further opening of the legs. This exposes the binding sites for the recruitment of cytoplasmic signalling molecules
  • outside-in activation; signals into the cells
  • signal from inside the cell acts on the integrin complex to promote the switch to the extended high-affinity conformation: “inside-out” activation; switches adhesion on
86
Q

Describe cell populations.

A

At high density, cells compete for growth factors.

When cells in culture form a con fluent mono layer, they cease proliferating and slow down many other metabolic activities. This used to be known as contact inhibition of cell division
May be competition for external growth factors rather than cell-to-cell contact
Density-dependence of cell-division

87
Q

Describe the signals controlling proliferation of tissue cells.

A

Two signals required:

  • growth factor (density dependence); ERK MAP Kinase cascade
  • ECM (anchorage dependence)

Mechanism of anchorage dependence

  • growth factor receptors and integrin signalling complexes can each activate identical signalling pathways (e.g. MAPK)
  • individually this activation is week and/or transient
  • together activation is strong and sustained
  • the separate signalling pathways act synergistically
88
Q

Explain contact interactions between cells including cell-to-cell adhesions

A

Short-term: transient interactions between cells which do not form stable cell-cell junctions
Long-term: stable interactions resulting in formation of cell-cell junctions

Cell-cell contact between non-epithelial cells
When most non-epithelial cells collide, they do not form stable cell-cell contacts. They actually repel one another by paralysing motility at the contact site, promoting the formation of a motile front at another site on the cell, and moving off in the opposite direction

This is contact inhibition of locomotion and is responsible for preventing multi layering of cells in culture and in vivo

Long-term cell-cell contacts
Upon contact, some cell types strongly adhere and form specific cell-cell junctions (adherents junctions, desmosomes, tight junctions, gap junctions)
This iOS true of epithelial cels, which form layers and neurones forming synapses

Cell-cell junctions in epithelia
Junctions are usually arranged as online ours belts (zonula) or as discrete spots (macula)

Contact-induced spreading of epithelial cells
Contact between epithelial cells leads to the mutual induction of spreading, so that the total spread area of the contacted cells is greater than that of the sum of the two separated cells. This could result in a stable monolayer

Cell-cell adhesion affecting cell proliferation
-No cell-cell junctions, activated MAPK, decreased p27KIP1, high proliferation (-Ca2+; + adhesion blocking antibody)
-Cell-cell junctions form, inactive MAPK, increased p27KIP1, low proliferation (+Ca2+; -adhesion blocking antibody)
Junctions are Ca2+ dependent

89
Q

Explain B-catenin.

A

Catenins bind to cadherins

B-catenin; a-catenin bind to cadherins (Ca2+-dependent, homophilic cell adhesion molecule - bind to identical molecules on adjacent cells) and actin filament

Adenomatous polyposis coli (APC) - inherited colon cancer: there are a number of familial types
The APC gene-product is a protein involved in the degradation of the junction-associated molecule, b-catenin (forms junction with actin cytoskeleton)

In normal state - b-catenin bound to cadherin roaming junctions at plasma membranes of adjacent cells
In cytoplasm:
-when bound to APC, APC complex active and leads to raid degradation
-when APC complex active, bound to LEF-1, goes to nucleus to cause gene transcription, leading to cell proliferation

90
Q

What is the mechanism for contact inhibition of proliferation?

What are other adhesion-associated signalling pathways that are known to influence contact-induced inhibition of proliferation?

A

When bound to cadherin at the membrane, b-catenin not available for LEF-1 binding and nuclear effects

Normally, cytoplasmic b-catenin rapidly degraded

If b-catenin cytoplasmic levels rise as a result of inhibition of degradation or loss of cadherin-mediated adhesion, b-catenin/LEF-1 complex enters nucleus and influences gene expression, leading to proliferation

Other:
Clustering of cadherins after cell-cell contact is known to alter the activation of small GTPases .e.g. Rac is activated, Rho is inhibited: this can influence proliferation

Some growth factor receptors are associated with cell-cell junctions. This reduces their capacity to promote proliferation

91
Q

What happens when cells lose their behavioural restraints?

Describe oncogenes/pronto-oncogenes

A
  • proliferate uncontrollably (lose density dependence of proliferation)
  • are less adherent and will multilayer (lose contact inhibition of locomotion and anchorage dependence)
  • epithelia breakdown cell-cell contacts
  • not Hayflick limited, express telomerase I.e. cancer

If the gene coding for a component of a signalling pathway is mutated so that the protein is constitutively active, that pathway will be permanently on
Receptors, signalling intermediates and signalling targets (e.g. transcription factors) are protooncogenes
This is the mechanism of short-circuiting leading to uncontrolled proliferation as a result of loss of growth factor dependence .etc.

Oncogene: mutant gene which promotes uncontrolled cell proliferation
Porto-oncogene: normal cellular gene corresponding to the oncogene

Neither growth factor (density dependence) or ECM signals (anchorage dependence) required to stimulate proliferation.
Can invade

92
Q

Describe benign and malignant tumours.

Describe local invasion and metastasis.

A

Adenoma (benign) - don’t invade locally
Adenocarcinoma (malignant) - invade locally and metastasise

• in addition to deregulated proliferation, a
major feature of cancerous tumours is their ability to spread
• most human cancers are carcinomas (i.e. of
epithelial origin)
• in order to spread to other sites (metastasis),
cells must break away from the primary
tumour, travel to a blood or lymph vessel,
enter the vessel, lodge at a distant site, leave
the vessel, and ultimately establish a
secondary tumour

Primary carcinoma cell metastasis
• cell-cell adhesion must be down-regulated (e.g.
cadherin levels reduced)
• the cells must be motile
• degradation of ECM must take place; matrix
metaloproteinase (MMP) levels increased in
order to migrate through basal lamina and
interstitial ECM
• the degree of carcinoma cell-cell adhesion is an
indicator of how differentiated the primary
tumour is, and indicates its invasiveness and the
prognosis

93
Q

Describe the hallmarks of cancer and the role of the cell cycle.

A
  • resisting cell death
  • sustaining proliferative signalling
  • evading growth suppressors
  • activating invasion and metastasis
  • enabling replication immortality
  • inducing angiogenesis
  • deregulating cellular energetic (less dependent on aerobic, more on anaerobic)
  • avoiding immune destruction
  • genome instability and mutation
  • tumour-promoting inflammation
Cell cycle 
Cycle checkpoints (growth arrest ensures genetic fidelity) - main, G1/S sense damage. G2/M right: duplication, identical and damage 

Specific proteins accumulate/ are destroyed during the cycle

Cyclins, cycle dependent kinases, cycle dependent kinase inhibitors

Permanent activation of a cyclin can drive a cell through a checkpoint

94
Q

Describe proto-oncogenes and oncogenes.

A

Porto-oncogenes code for essential proteins involved in maintenance of cell growth, division and differentiation

Mutation converts a proto-oncogene to an oncogene, whose protein product no longer responds to control influences

Oncogenes can be aberrantly expressed, over-expressed or aberrantly active e.g. MYC, RAS, ERB, SIS

Proto-oncogenes can be converted to an oncogene by a single mutation e.g. point mutations, frame shift .etc.

Oncogenes activation
Normal proto-oncogene can be changed in 4 ways:
-mutation in the coding sequence (point mutation of deletion); aberrantly active protein
-gene amplification (multiple gene copies); overproduction of normal protein
-chromosomal translocation (chimaeric genes) = strong enhancer increases normal protein levels e.g. Burkitt’s lymphoma
-insertional mutagenesis (e.g. viral infection) = fusion to actively transcribed gene overproduced protein or fusion protein is hyperactive .e.g. Philadelphia chromosome - BCR is an anti-apoptotic protect inserted into ABL

Proteins involved in signal transduction are potential critical gene targets (proto-oncogenes):
-tyrosine kinase receptor EC - met, neu
-tyrosine kinase receptor IC - src, ret
-G-protein coupled receptor - ras, gip-2
-kinases - raf, pim-1
-transcription factors - myc, fos, jun
Can start cascade without appropriate signal

Mutant RAS has aberrant activity
• Upon binding GTP, RAS becomes active.
• Dephosphorylation of the GTP to GDP switches RAS off.
• Mutant RAS fails to dephosphorylate GTP and remains active.

Oncogenes and human tumours

  • MYC - transcription factor - activated by translocation - nuclear - Burkitt’s lymphoma
  • JUN - transcription factor - overexpression/deletion - nuclear - lung
95
Q

Describe tumour suppressor genes including the two hit hypothesis and retinoblastoma.

A

Typically proteins whose function is to regulate cellular proliferation, maintain cell integrity e.g. RB
Each cell has two copies of each tumour suppressor gene
Mutation or deletion of one gene copy is usually insufficient to promote cancer
Mutation or loss of both copies means loss of control

Knudson’s two hit hypothesis
Sporadic cancer: 2 acquired mutations
Hereditary cancer: 1 inherited and 1 acquired mutation

Inherited cancer susceptibility
▪ Family history of related cancers.
▪ Unusually early age of onset.
▪ Bilateral tumours in paired organs.
▪ Synchronous or successive tumours.
▪ Tumours in different organ systems in same individual.
▪ Mutation inherited through the germline.

Retinoblastoma
▪ Malignant cancer of developing retinal cells.
▪ Sporadic disease usually involves one eye. Hereditary cases can be unilateral or bilateral and multifocal.
▪ Due to mutation of the RB1 tumour suppressor gene on chromosome 13q14.
▪ RB1 encodes a nuclear protein that is involved in the regulation of the cell cycle. (CDK 4 and 6)

Functional classes of tumour suppressor genes 
▪ Regulate cell proliferation 
▪ Maintain cellular integrity 
▪ Regulate cell growth 
▪ Regulate the cell cycle 
▪ Nuclear transcription factors 
▪ DNA repair proteins 
▪ Cell adhesion molecules 
▪ Cell death regulators
Suppress the neoplastic phenotype

Tumour suppressor genes and human tumours
P53 - cell cycle regulator - nuclear - many cancers e.g. colon, breast, lung
BRCA1 - cell cycle regulator - nuclear 0 breast, ovarian, prostate

Although p53 is a tumour suppressor gene, mutants of p53 act in a dominant manner and mutation of a single copy is sufficient to get dysregualtion of activity - hard to target because can affect other functions

APC tumour suppressor gene (familial adenomatous polyposis coli)

  • due to a deletion in 5q21 resulting in loss of APC gene (tumour suppressor gene)
  • involved in cell adhesion (anchoring/no according in stroma) and signalling (MAPK)
  • sufferers develop multiple benign adenomatous polyps of the colon
  • there is a 90% risk of developing colorectal carcinoma

The tumour suppressor gene APC participates in the WNT signalling pathway
APC protein is a negative regulator of b-catenin(cell division and adhesion) thereby preventing uncontrolled cell division
Mutation of APC is a frequent event in colon cancer

96
Q

Describe the route to cancer.

A

Oncogene, defective tumour suppressor gene —> cell growth and proliferation —> cancer

Analogy
Normal genes (regulate cell growth) - tumour suppressor gene (break) working effectively
1st mutation (susceptible carrier) - tumour suppressor gene and active oncogene (accelerator)
2nd mutation or loss (leads to cancer) - accelerator and no brakes

The development of colorectal cancer
Normal epithelium (problem with APC - tumour suppressor) —> hyperproliferative epithelium (problem with K-ras - oncogene)—> adenoma (problem with p53) —> carcinoma —> metastasis 

Hyperplasia —> metaplasia —> dysplasia —> carcinogenesis

Oncogene vs tumour suppressor gene
Table

Side note: COSMIC = catalogue of somatic mutations in cancer; important in precision medicine

97
Q

Define incidence and mortality.

Define epidemiological transition.

A

Incidence: new cases of cancers
Mortality: no of deaths

Discrepancy because not all cancers are lethal

Epidemiological transition: poorer countries moving towards western; mortality changes with food, environmental changes .etc.

98
Q

Describe the effect of migrants on the epidemiology of cancer.

A

Extent and rate of change that are informative:
A rapid change in risk following migration implies that lifestyle/environment factors act late in carcinogenesis
A slow change suggests that exposures early in life are the most relevant
Persistence of rates between generations suggests genetic susceptibility is important in determining risk

99
Q

Summarise the epidemiology of cancer.

A

Incidence is increasing for common cancer sites in both high-income (now with plateauing and even decreases) and low-income countries (e.g. colorectal, prostate) - effects of earlier diagnosis, screening, changes in risk factors

Mortality is decreasing in most high-income countries, not in low income.

Total burden is increasing because of demographic changes (ageing and population size) and westernisation of lifestyle

Main risk factors: smoking, diet, alcohol

Cancer is a leading cause of death
1. The epidemiology of cancer tells us that cancer incidence is related to:
•Age
•Common environmental causes •Geographical variation and secular trends
There are several well defined risk factors for cancer, including smoking, diet and alcohol consumption.
2. The process of carcinogenesis is important in understanding the development of cancer. Important factors involved in this process include chemical carcinogens, radiation and oncogenic viruses
3. Inherited / familial cancers are rare, but have provided valuable information in understanding the process of carcinogenesis.

100
Q

Describe Parkin’s estimate of preventable cancers.

Describe risk factors.

A

Reduction/ elimination of risk factors would lead to a substantial reduction in cancers and CVD, renal disease, hepatic, diabetes and neurological

Smoking

  • at least 30% of all cancer deaths
  • smoking is associated with increased risk for at least 15 types
  • 90% of lung cancer deaths in men and 80% in women

Diet
-dietery fibre and colorectal cancer

Alcohol 
Oral cavity, pharynx, larynx, oesophagus, liver all types of alcohol 
mechanisms poorly understood 
synergism with tobacco 
balance with preventive effect for CHD

Anthropometry
Measure of proportions of body e.g. BMI

101
Q

How can you prevent cancer?

Describe the impact of a western lifestyle.

Infectious diseases and cancer.

A

1) be as lean as possible without being underweight
2) physically active, more than or equal to 30 mins
3) avoid sugary drinks, processed foods
4) veg and fruit
5) decrease alcohol
6) decrease salty foods
7) don’t use supplements

Western Lifestyle:

  • Energy dense diet, rich in
  • fat,
  • refined carbohydrates
  • animal protein
  • Low physical activity
  • Smoking and drinking

Consequences:

  • Greater adult body height
  • Early menarche
  • Obesity
  • Diabetes
  • Cardiovascular disease
  • Hypertension

Infectious disease
16% caused by infectious agents

HPV = cervix, head and neck
EBV = Hodgkin’s lymphoma, Burkitt’s
HCV, HBV = liver
H. Pylori = stomach

102
Q

Describe breast cancer as a hormone-regulated cancer.

A

Carcinoma: tumour of epithelial cells

All glands meet at nipple
Different cancer types for different glands
E.g. lobular cancer for lobes, ductal cancer for ducts

Mammary gland made up of 2 epithelial layers
Second layer is myoepithelial cells - contractile properties - squeeze luminal epithelial - milk out of nipple
A layer of myoepithelial cells, some of which are slightly vacuoloted is seen just around the luminal cells, making contact with the basement membrane

103
Q

Describe the progression of normal to malignant breast.

A

Normal —> benign/ in situ carcinoma —> medullary carcinoma/ carcinoma/ lobular carcinoma

Medullary carcinoma

  • with lymphocytes
  • aggressive
  • not like original, have neuroendocrine secretory vesicles

Carcinoma

  • Infiltrating ductal carcinoma
  • Greater than or equal to 80% in this category
  • many have no special type of histogocial structure
104
Q

Explain the role of oestrogen receptor in breast cancer.

A

Immunohistochemical staining using antibodies against the Human Estrogen Receptor (ER) is informative

The oestrogen receptor is activated upon binding oestrogen
Gene expression is induced by binding to specific DNA sequences called oestrogen response elements
The oestrogen-induced gene products increase cell proliferation, resulting in breast cancer

Ligand = oestrogen (can pass cell membrane)
ER - monomeric protein, nuclear receptor
Dimer ER
Bind to TATA —> gene expression
Some important oestrogen regulated genes:
-progesterone receptor (PR)
-Cyclin D1 = regulation of cell cycle
-c-myc = make sure cells don’t apoptosis
-TGF-a = direct GF therefore cell growth and cell survival increases

Normal mammary gland:
Cells with ER don’t grow, surrounding cells grow
Tumour:
Oestrogen drives growth of tumour cells

Some breast cancers like normal breast are sensitive to the effects of oestrogen
ER is over expressed in around 70% of breast cancers = better prognosis
In ER-positive case, estrogen regulates the expression of genes involved in cellular proliferation leading to breast cancer
Oestrogen withdrawal or competition for binding to the ER using anti-oestrogens results in a response in about 70% of ER positive cancers, 5-10% of ER-negative cancers also respond
An increased level of expression of ER indicates a good rognosis in female breast cancer but a worse in male (androgen receptor)

Overstimulation of ER downregulates it therefore tumour regression but can relapse with incurable metastatic disease

105
Q

State the major treatment approaches for breast cancer.

A

Surgery
Radiation therapy
Chemotherapy
Endocrine therapy

Side note:
Adjuvant therapy: kill of any tumour cells broken away from tumour mass —> secondary tumours
Neoadjuvant therapy given before surgery because too big tumour for surgery but less common because early detection

106
Q

Explain endocrine therapy in the treatment of breast cancer.

A

Can be achieved by:

  • ovarian suppression (pre-menopausal)
  • blocking’s oestrogen production by enzymatic inhibition
  • inhibiting oestrogen responses

Hormonal control of target tissues
Premenopausal
Hypothalamus releases LHRH —> pituitary gland releases gonadotrophins —> ovary —> oestrogen and progesterone
Pre/post menopausal
LHRH—> ACTH from pituitary —> adrenal glands —> androgen —> oestrogen (by peripheral conversion - aromatisation, fatty tissue (also liver, muscle) has high levels of aromatase e.g. mammary gland)

Ovarian ablation and suppression
-surgical oophorectomy (removal of ovaries)
-ovarian irradiation (radiation therapy)
The major problems associated are morbidly and irreversibility - won’t be able to have children
To overcome, treatments to produce ovarian ablation have been developed:
Lutenising hormone releasing hormone (LHRH) agonists
These bind to LHRH receptors in the pituitary leading to receptor down-regulation and suppression of LH release and inhibition of ovarian function, including oestrogen production
REVERSIBLE
E.g. goserelin, Buserelin, leuprolide, triptorelin

Inhibiting oestrogen action
Anti-oestrogens:
Tamoxifen (nolvadex) - receptor recognises this as oestrogen even if don’t look alike —> no gene expression
ICI 182,780 = Faslodex - bind to ER receptor but won’t activate transcription

Tazmoxifen is a competitive inhibitor of oestradiol binding to the ER.
Antioestorgens negate the stimulatory effects of oestrogen by blocking the ER causing the cell to be held at the G1 phase of the cell cycle —> death
Tamoxifen is the endocrine treatment of choice in metastatic disease in postmenopausal women
Few side effects

Tamoxifen is a selective oestrogen receptor modulator (SERMs)
-oestrogenic effects in bone - treat osteoporosis, maintain density
-oestrogenic effects in CNS - treat atherosclerosis, lowers LDL and raises HDL but long term use may increase risk
Undesirable:
-subsequent thromboembolic episodes (liver)
-endometrial thickening, hyperplasia and fibroids
-hypothalamus incraeses vasomotor symptoms = hot flushes

Toremifene - structural derivative of tamoxifen with similar antioestrogenic and oestrogenic properties
Faslodex/ fulvestrant - no oestrogen-like effects so not to be used in osteoporosis and CVD patients but controls oestrogen stimulated growth, favourable over tamoxifen in decreasing tumour cell invasion and stimulation of endometrial carcinoma. First lien for advanced breast cancer and second line if tamoxifen fails
Raloxifene - agonistic in bone and no activity in breast and uterus. Used in treatment of osteoporosis in post-menopausal

Tamoxifen also reduces the incidence of contralateral breast cancer by a third - second breast cancer in other mammary gland which is independent of first so can prevent breast cancer
Problems in using for prevention:
-increased incidence of endometrial cancer
-stroke
-DVT
-cataracts
To overcome use raloxifene/Faslodex (SERM) or aromatase inhibitors

Blocking oestrogen production by enzymatic inhibition
Aromatase inhibitors
In postmenopausal, major source of oestrogen from conversion of androstenedione or testosterone to oestrone
-occurs at extra-adrenal or peripheral sites such as fat, liver and muscle
-conversion catalysed by aromatase

Aromatase consists of a complex containing a cytochrome P450 heme containing protein as well as the flavoprotein NADPH cytochrome P450 reductase.
Aromatase catalyzes three separate steroid hydroxylations involved in the conversion of androstenedione to estrone.
Aromatase can metabolise androsteindione, which is produced by the adrenal glands. This leads to the production of Estrone Sulphate, which is circulated in the plasma

Can divide aromatase inhibitors into mechanism-based, suicide inhibitors (type I) and competitive inhibitors (type II)
Type I:
-chemically binds to active site and enzyme breaks down
-irreversible
E.g. exemestane, side effects include fatigue, nausea, hot flushes

Type II:

  • bind reversibly to active site, blocks as long as it occupies the site
    e. g. anastrozole
107
Q

Explain the role of progestins in breast cancer.

A
  • Progesterone is the dominant naturally occurring progestin
  • The poor absorption of progesterone has been overcome with some of the synthetic derivative progestins
  • Progestin response in the human breast is complex and influences both proliferation and differentiated function.
  • Progestins are used in the endocrine treatment of uterine and breast cancer with clinically proven antineoplastic properties.
  • Progestin therapy for metastatic breast cancer has been used principally as a second- or third-line therapy following selective estrogen
  • . The principal progestin used for metastatic breast cancer has been megestrol acetate

Agonist which overstimulates receptor

108
Q

Describe endocrine therapy resistance in breast cancer.

A

•In treatment:
>60% of ERa-positive tumours respond to endocrine
therapy
Anti-estrogens, e.g. tamoxifen
Inhibitors of estrogen synthesis, e.g. exomestane

clinical problem:
• Initial response but eventual relapse
• Relapse due to resistance during prolonged endocrine therapy
• NOT due to tumours becoming ER-independent
• Recent data shows that resistant tumours have mutated ER

Solution-
• Continue use endocrine therapies as these are successful
• But, require additional therapeutic agents/ strategies for endocrine resistant, metastatic disease e.g. inhibitors of CDKs

109
Q

What are the risk factors for breast cancer?

Describe screening.

A
Risk Factors - exposure to estrogens
Early age of onset of menarche 
Late age to menopause 
Age at first full-term pregnancy 
Some forms of the contraceptive pill 
Hormone Replacement Therapy
Obesity
 Diet, physical activity, height, medication
(Aspirin)

Screening programme uses mammography between 50-64 now extended to 70
Test every 3 years

110
Q

Describe colon anatomy and physiology.

Describe turnover.

A

Front of colon lined by peritoneum
Back lined by mesentery
Colon is a hollow tube which starts in right iliac fossa
Mucosa found in lamina propria and epithelium

Cells are constantly proliferating and travel up colorectal crypt of Lieberkuhn

Function

  • extraction of water from faeces (electrolyte balance)
  • faecal reservoir (evolutionary advantage)
  • bacterial digestion for vitamins (.e.g. B and K)

Turnover
Proliferation renders cells vulnerable
APC mutation prevents cell loss —> mutation
Normally we have protective mechanisms to eliminate genetically defective cells by:
-natural loss
-DNA monitors
-repair enzymes

111
Q

Describe polyps and adenomas.

Explain polyps.

A

A polyp is any projection from a mucosal surface into a hollow viscus, and may be hyperplastic, neoplastic, inflammatory, hamartomatous .etc.

An adenoma is a benign neoplasm of the mucosal epithelial cells

Colonic polyp types

  • metaplastic/hyperplastic
  • adenomas
  • juvenile
  • Peutz Jeghers (mucosal hyperpigmentation)
  • lipomas
  • others (essentially any circumscribed intramucosal lesions)
Hyperplastic polyps
-very common
-often multiple 
-no malignant potential 
-15% have k-ras mutation 
-<5 cm 
(Serrated appearance (jagged edge)
112
Q

Explain adenomas.

A
Colonic adenoma types:
-tubular 
-tubular/villus
-villus
(Flat)
(Serrated)

Pedunculated adenoma - like stalk
Sessile adenoma - flat

Microscopic structure of adenomas:
tubular
-columnar cells with nuclear enlargement, elongation, multilayering and loss of polarity (nuclear)
-increased proliferative activity
-reduced differentiation
-complexity/disorganisation of architecture
E.g. colonic adenoma = mushroom like

Villous

  • mutinous cells with nuclear enlargement , elongation, multilayering and loss of polarity
  • exophytic, frond-like extensions
  • rarely may have hypersecretory function and result in excess mucus discharge and hypokalaemia (potassium loss in GI tract)
113
Q

Describe dysplasia.

Explain Adenomatous Poluposis Coli (APC/FAP).

A

Abnormal growth of cells with some features of cancer
Indefinite, low grade and high grade (back to back glands)

APC/FAP

  • 5q21 gene mutation
  • site of mutation determines, clinical variants (classic, attenuated, Gardner, Turoot)

-many patients have prophylactic colectomy

Large polyps have higher risk than small ones

Progression from adenoma to carcinoma

  • Most CRCs arise form adenomas
  • adenomas and Ca similar distribution
  • adenomas usually precede cancer by 15 years
  • endoscopic removal of polyps decrease the incidence of subsequent CRC

Genetic pathways:
-Involves APC, K raise, Smads, p53, telomerase activation
-Micro satellite instability - micro satellites are sequences prone to misalignment. Some microsatellites are in coding sequences of genes which inhibit growth or apoptosis e.g. TGFbR11
-mismatch repair genes (e.g MSH2). Recessive teens requiring 2 hits
HNPCC - germline mutation in these genes

Diagram

Genetic predisposition as 2 main pathways:

  • FAP - inactivation of APC tumour suppressor gene
  • HNPCC - microsatellite instability

Why is APC important in colon cancer?
B-catering moves from cytoplasm to nucleus if APC doesn’t work - affect cell transcription, cell proliferation increases (uncontrolled)

Role of p53
Diffuse positivity of p53 - p53 not working properly therefore cells trying to express p53 - accumulating non-functional p53

All this leads to colonic carcinoma - age Rae 50-80, dietary factors: high fat, low fibre, high red meat, refined carbohydrates

114
Q

Describe the role of food in colorectal cancers.

A

Food contains carcinogens
Also contains anti cancer agents
Heat modifies chemicals and so does bacteria

HCAs (heterocyclic amines) found in red meat
When cooked at high temperatures oxidation of PhIP can form complexes with deoxyguanosine in DNA —> mutagenesis

Dietery deficiencies
Folate and colorectal cancer
-co-enzyme for nucleotide synthesis and DNA methylation
MTHFR
-deficiency leads to disruption in DNA synthesis causing DNA instability (strand breaks and uracil incorporation) —> mutations
-decreased methionine synthesis leads to genomic hypomethylation and focal hypermethylation —> gene activation and silencing
(May not need to know the above)

Anti cancer food elements
Vitamin C and vitamin E - ROS (reactive oxygen species) scavenger
Polphenols (green tea, fruit juice) activate MAPK —> regulates Phase 2 detoxifying enzymes as well as other genes and reduced DNA oxidation
Garlic associated with apoptosis

115
Q

Describe the clinical presentation of colon cancer.

Explain the macroscopic features of CRC.

Describe the microscopic structure of carcinomas.

A
Changes in bowel habit 
Bleeding PR (pre-rectal)
Unexplained iron deficient anaemia 

Mucous
Bloating
Cramps
Constitutional (weight loss, fatigue)

Macroscopic features
Small carcinomas may be resent within large polyploid adenomas, pedunculated or sessile
Can be distributed in caecum/ascending colon, transverse colon, descending colon, rectosigmoid

Microscopic structures 
Adenocarcinomas grade 1-3
Mutinous carcinomas
Signet ring cell
Neuroendocrine
116
Q

Describe the classification of colorectal cancer.

A

proportion of gland differentiation relative to solid areas or nests and cords of cells without lumina
~ 10% well differentiated
~ 70% moderately differentiated
~ 20% poorly differentiated

Dukes Classification
Dukes A - growth limited to mucosa/submucosa
- nodes negative
Dukes B - growth into or beyond musc propria - nodes negative
Dukes C1
- nodes positive
- apical LN negative
Dukes C2 - apical LN positive
(A—>C, 5 year survival drops dramatically)

117
Q

Describe the clinical and pathologic features affecting prognosis.

What are the treatment options?

A

Diagram

118
Q

Explain screening for colon cancer.

A

Screening is the practice of investigating apparently healthy
individuals with the object of detecting unrecognised disease or
people with an exceptionally high risk of developing disease, and
of intervening in ways that will prevent the occurrence of disease
or improve the prognosis when it develops.

Screening for High Risk Colon Cancer
Previous adenoma
1st Degree relative affected by colorectal cancer before the age of 45
2 affected first degree relatives evidence of dominant familial cancer trait including
colorectal, uterine, and other cancers
UC and Crohn’s disease
Hereditable cancer families (include other sites)

Test characteristics
simple and acceptable to the patient
sensitive and selective 
The screening population should have equal access to the screening procedure. 
Cost effectiveness
119
Q

What is leukaemia and describe why it differs from other cancers?

A

Leukaemia is cancer of the blood
Blood cancers are the most common cancers in men and women aged 15-24
Bone marrow disease, not all patients have abnormal cells in the blood

Leukaemia results from a series of mutations in a single lymphoid or myeloid stem cell
These mutations lead to cell showing abnormalities in proliferation, differentiation or cell survival leading to explanation of the leukaemia clone
Cells involved:
Myeloid stem cell —> erythroblast, megakaryoblast, myeloblast, moon last
Lymphoid stem cell —> pre B lymphocytes, Pro T lymphocyte

Difference to other cancers 
• Leukaemia is different from other cancers 
• Most cancers exist as a solid tumour 
• However, it is uncommon for patients with
leukaemia to have tumours 
• More often they have leukaemic cells
replacing normal bone marrow cells and
circulating freely in the blood stream
• Leukaemia is different  from other cancer
because haemopoietic and lymphoid cells
behave differently from other body cells
• Normal haemopoietic stem cells can
circulate in the blood and both the stem
cells and the cells derived from them can
enter tissues
• Normal lymphoid stem cells recirculate
between tissues and blood

• The concepts of invasion (local penetration) and metastasis (blood/lymphatics)
cannot be applied to cells that normally
travel around the body and enter tissues • We have to have other ways of
distinguishing a ‘benign’ condition from a
‘malignant’ condition and haematologists
usually use different words for these
concepts

• Leukaemias that behave in a relatively
‘benign’ manner are called chronic—that
means the disease goes on for a long time
• Leukaemias that behave in a ‘malignant’
manner are called acute—that means that, if
not treated, the disease is very aggressive
and the patient dies quite rapidly

In summary:
• Leukaemia results from acquired somatic
mutation in a haemopoietic or lymphoid
stem cells
• A single cell gives rise to a clone of
leukaemic cells that replace normal cells
• Disease characteristics are due to (i)
proliferation of leukaemic cells (ii) loss of
function of normal cells

120
Q

How is leukaemia classified?

A

Leukaemia can be acute or chronic

Depending on the cell of origin it can also be lymphoid or myeloid

Lymphoid can be B or T lineage

Myeloid can be any combination of granulocytic, monocytic, erythroid or megakaryocytic

121
Q

What is leukaemia and describe why it differs from other cancers?

A

Leukaemia is cancer of the blood
Blood cancers are the most common cancers in men and women aged 15-24
Bone marrow disease, not all patients have abnormal cells in the blood

Leukaemia results from a series of mutations in a single lymphoid or myeloid stem cell
These mutations lead to cell showing abnormalities in proliferation, differentiation or cell survival leading to explanation of the leukaemia clone
Cells involved:
Myeloid stem cell —> erythroblast, megakaryoblast, myeloblast, moon last
Lymphoid stem cell —> pre B lymphocytes, Pro T lymphocyte

Difference to other cancers 
• Leukaemia is different from other cancers 
• Most cancers exist as a solid tumour 
• However, it is uncommon for patients with
leukaemia to have tumours 
• More often they have leukaemic cells
replacing normal bone marrow cells and
circulating freely in the blood stream
• Leukaemia is different  from other cancer
because haemopoietic and lymphoid cells
behave differently from other body cells
• Normal haemopoietic stem cells can
circulate in the blood and both the stem
cells and the cells derived from them can
enter tissues
• Normal lymphoid stem cells recirculate
between tissues and blood

• The concepts of invasion (local penetration) and metastasis (blood/lymphatics)
cannot be applied to cells that normally
travel around the body and enter tissues • We have to have other ways of
distinguishing a ‘benign’ condition from a
‘malignant’ condition and haematologists
usually use different words for these
concepts

• Leukaemias that behave in a relatively
‘benign’ manner are called chronic—that
means the disease goes on for a long time
• Leukaemias that behave in a ‘malignant’
manner are called acute—that means that, if
not treated, the disease is very aggressive
and the patient dies quite rapidly

122
Q

How is leukaemia classified?

A

Leukaemia can be acute or chronic

Depending on the cell of origin it can also be lymphoid or myeloid

Lymphoid can be B or T lineage

Myeloid can be any combination of granulocytic, monocytic, erythroid or megakaryocytic

123
Q

What is leukaemia and describe why it differs from other cancers?

A

Leukaemia is cancer of the blood
Blood cancers are the most common cancers in men and women aged 15-24
Bone marrow disease, not all patients have abnormal cells in the blood

Leukaemia results from a series of mutations in a single lymphoid or myeloid stem cell
These mutations lead to cell showing abnormalities in proliferation, differentiation or cell survival leading to explanation of the leukaemia clone
Cells involved:
Myeloid stem cell —> erythroblast, megakaryoblast, myeloblast, moon last
Lymphoid stem cell —> pre B lymphocytes, Pro T lymphocyte

Difference to other cancers 
• Leukaemia is different from other cancers 
• Most cancers exist as a solid tumour 
• However, it is uncommon for patients with
leukaemia to have tumours 
• More often they have leukaemic cells
replacing normal bone marrow cells and
circulating freely in the blood stream
• Leukaemia is different  from other cancer
because haemopoietic and lymphoid cells
behave differently from other body cells
• Normal haemopoietic stem cells can
circulate in the blood and both the stem
cells and the cells derived from them can
enter tissues
• Normal lymphoid stem cells recirculate
between tissues and blood

• The concepts of invasion (local penetration) and metastasis (blood/lymphatics)
cannot be applied to cells that normally
travel around the body and enter tissues • We have to have other ways of
distinguishing a ‘benign’ condition from a
‘malignant’ condition and haematologists
usually use different words for these
concepts

• Leukaemias that behave in a relatively
‘benign’ manner are called chronic—that
means the disease goes on for a long time
• Leukaemias that behave in a ‘malignant’
manner are called acute—that means that, if
not treated, the disease is very aggressive
and the patient dies quite rapidly

124
Q

How is leukaemia classified?

A

Leukaemia can be acute or chronic

Depending on the cell of origin it can also be lymphoid or myeloid

Lymphoid can be B or T lineage

Myeloid can be any combination of granulocytic, monocytic, erythroid or megakaryocytic

• Put all this together and we get:
Acute lymphoblastic leukaemia (ALL)
Acute myeloid leukaemia (AML)
Chronic lymphocytic leukaemia (CLL) Chronic myeloid leukaemia (CML)

Lymphoblastic: immature, lymphocytic: mature lymphocyte

125
Q

Why do people get leukaemia?

A

• Leukaemia results from a series of mutations in a single stem cell
• Some mutations result from identifiable (or
unidentifiable) oncogenic influences
• Others are probably random errors—chance
events—that occur throughout life and
accumulate in individual cells

• The important leukaemogenic mutations that have been recognized include:

  • Mutation in a known proto-oncogene e.g. point mutations
  • Creation of a novel gene, e.g. a chimaeric or fusion gene
  • Dysregulation of a gene when translocation brings it under the influence of the promoter or enhancer of another gene

• Loss of function of a tumour-suppressor gene can also contribute to leukaemogenesis—this can result from deletion or mutation of the gene
• If there is a tendency to increased
chromosomal breaks, the likelihood of
leukaemia is increased
• In addition, if the cell cannot repair DNA
normally, an error may persist whereas in a
normal person the defect would be repaired

• Inherited or other constitutional
abnormalities can contribute to
leukaemogenesis:
-Down’s syndrome
- Chromosomal fragility syndromes 
-Defects in DNA repair 
-Inherited defects of tumour-suppressor genes
• Identifiable causes of leukaemogenic
mutations include:
-Irradiation 
-Anti-cancer drugs 
-Cigarette smoking 
-Chemicals—benzene

• Leukaemia, like cancer in general, can be seen as an acquired genetic disease, resulting from somatic mutation
• Mutation in germ cells may bring favourable, neutral or unfavourable characteristics to the species
• Somatic mutation may be beneficial*, neutral or harmful
* A rare occurrence but can lead to reversion to normal phenotype in some cells in individuals with an inherited abnormality, e.g.
an immune deficiency or bone marrow failure syndrome

126
Q

What is the difference between acute and chronic myeloid leukaemia?

A

In AML, cells continue to proliferate but they no longer mature so there is:
-A build up of the most immature cells— myeloblasts or ‘blast cells’—in the bone marrow with spread into the blood
-A failure of production of normal functioning end cells such as neutrophils, monocytes, erythrocytes, platelets
Platelets can also be low because complicated by disseminated intravascular coagulation (platelets consumed)

• In AML, the responsible mutations usually
affect transcription factors so that the
transcription of multiple genes is affected
• Often the product of an oncogene prevents
the normal function of the protein encoded
by its normal homologue
• Cell behaviour is profoundly disturbed

• In CML, the responsible mutations usually
affect a gene encoding a protein in the
signalling pathway between a cell surface
receptor and the nucleus
• The protein encoded may be either a
membrane receptor or a cytoplasmic protein

• In CML, cell kinetics and function are not as
seriously affected as in AML • However, the cell becomes independent of
external signals, there are alterations in the
interaction with stroma and there is reduced
apoptosis so that cells survive longer and
the leukaemic clone expands progressively

• Whereas in AML there is a failure of
production of end cells, in CML there is
increased production of end cells

127
Q

What is the difference between acute and lymphoid leukaemias.

A
• Acute lymphoblastic leukaemia has an increase in very immature cells—
lymphoblasts—with a failure of these to
develop into mature T and B cells 
• In chronic lymphoid leukaemias, the
leukaemic cells are mature, although
abnormal, T cells or B cells
128
Q

How does leukaemia cause the disease characteristics.

A

Accumulation of abnormal cells leading to – Leucocytosis, bone pain (if leukaemia is acute), hepatomegaly, splenomegaly lymphadenopathy (if lymphoid), thymic enlargement (if T lymphoid), skin infiltration

Metabolic effects of leukaemic cell
proliferation—hyperuricaemia (DNA break down) and renal failure (deposit of uric acid), weight loss, low grade fever, sweating

Crowding out of normal cells leading to –
anaemia, neutropenia, thrombocytopenia

Haemorrhage and swelling of gums (infiltration of gums by leukaemic cells), chemotaxis, can also have intraventricular haemorrhage

Loss of normal immune function as a result
of loss of normal T cell and B cell function
—a feature of chronic lymphoid leukaemia

129
Q

Describe the epidemiology of acute lymphoblastic leukaemia.

A

Acute lymphoblastic leukaemia is
largely a disease of children

  • Epidemiology suggests that B-lineage ALL may result from delayed exposure to a common pathogen or, conversely, that early exposure to pathogens protects
  • Evidence relates to family size, new towns, socio-economic class, early social interactions, variations between countries

• Epidemiology also suggests that some
leukaemias in infants and young children result from:
-Irradiation in utero
-In utero exposure to certain chemicals ? -Epstein–Barr virus infection
• Rarely ALL results from exposure to a
mutagenic drug

130
Q

Describe the clinical features of acute lymphoblastic leukaemia.

What are the haematological features.

A
Resulting from accumulation of abnormal cells:
• Bone pain 
• Hepatomegaly 
• Splenomegaly 
• Lymphadenopathy 
• Thymic enlargement 
• Testicular enlargement

Resulting from crowding out of normal cells
• Fatigue, lethargy, pallor, breathlessness (caused by anaemia)
• Fever and other features of infection (caused by neutropenia)
• Bruising, petechiae, bleeding (caused by
thrombocytopenia)

• Leucocytosis with lymphoblasts in the blood • Anaemia (normocytic, normochromic)
• Neutropenia
• Thrombocytopenia
• Replacement of normal bone marrow cells
by lymphoblasts

131
Q

Describe the investigation of acute lymphoblastic leukaemia.

A
  • Blood count and film
  • Check of liver and renal function and uric acid
  • Bone marrow aspirate
  • Cytogenetic/molecular analysis
  • Chest X-ray

On blood film can see: lymphoblasts, high nuclear:cytoplasmic ratio, lack of platelets

Immunophenotyping
Antibodies used to bind to antigen and have fluorescent markers

Cytogenetic and molecular genetic analysis
Can see leukaemogenic mechanisms
Cytogenetic : karyotyping, extra chromosomes can be seen -hyperdiploidy = good prognosis, fusion gene/ translocation = poor prognosis
Leukaemogenic mechanisms:
• Formation of a fusion gene - translocation
• Dysregulation of a proto-oncogene by
juxtaposition of it to the promoter of another
gene, e.g. a T-cell receptor gene
• Point mutation in a proto-oncogene

Specific to ALL:
Chromosomes 12 an 21 showing EV6 and RUNX1, following translocation there is a fusion ETV6-RUNX1 gene on chromosome 12

This can be detected by two
fluorescent probes, a green probe for ETV6 and a red probe for RUNX1; when a fusion gene is formed the two colours fuse to give a yellow fluorescent signal
This technique is called fluorescence in situ hybridization —FISH
(If cytogenetic analysis fails because cells don’t go into mitosis, FISH can be used to detect fusion gene)

132
Q

Describe the treatment for acute lymphoblastic leaukemia.

A

• Supportive
-red cells - anaemia
-platelets - thrombocytopenia
-antibiotics - infection
• Systemic chemotherapy - widespread disease
• Intrathecal chemotherapy - via lumbapuncture

133
Q

Describe the micro anatomy of the skin.

A

Made up of epidermis, dermis and hypodermis.

Hair follicle located in dermis, subcutaneous fat in hypodermis
Dermis also contains collagen, elastin, GAGs, blood vessels, nerves, sweat glands
Apocrine glands make viscous and smelly sweat (groins), eccrine - normal body sweat
Dendritic cells, melanocytes, keratinocytes found in epidermis

Structure of epidermis
Cells important in skin cancer are keratinocytes and melanocytes
Keratinocytes mature and rise up from base, basal —> spinous —> granular —> corneum and shed
Exposed to UV radiation and undergo genetic mutation

134
Q

What are the different types of skin cancers.

Describe the causes of cancer.

A

Keratinocytes derived:
E.g. basal cell carcinoma (pearly-grey, shiny, pinky with telangiectasia - dilated small capillaries), squamous cell carcinoma, aka non-melanoma skin cancer (NMSC)

Melanocyte derived:
E.g. malignant melanoma - dark irregular border

Vasculature derived:
E.g. Kaposi’s sarcoma, angiosarcoma

Lymphocyte derived:
E.g. mycosis fungoides

Cause of cancer
Accumulation of genetic mutation —> uncontrolled cell proliferation

Examples of causes

  • genetic syndromes: Gorlin’s syndrome (underlying tendency to develop basal carcinomas), xeroderma pigmentosum (genetic defect in DNA repair)
  • viral infections: HHV8 in Kaposi’s sarcoma, HPV in squamous cell carcinoma
  • UV light: basal cell and squamous cell carcinoma, malignant melanoma
  • immunosuppression: drugs, HIV, old age, leukaemia
135
Q

Explain the role of UV light in cancer.

A

Sunlight benefits:
• Essential for photosynthesis (plants) • Infrared spectra provide warmth
• Effect on human mood
• Stimulates the production of vitamin D in the skin

• UVB
most important wavelength in skin carcinogenesis
Directly induces abnormalities in DNA e.g. mutations
Induces photoproducts affecting cytosine and thymine - cross linking to form thymine dimers
Usually repaired quickly by nucleotide excision repair - remove mutations

• UVA
100 times more UVA penetrates to the Earth’s surface
major cause of skin ageing
contributes to skin carcinogenesis
used therapeutically in PUVA therapy (used in psoriasis, eczema)
Can form pyrimidine dimers but less effective
Free radicals which damage DNA and cell membrane

• UV damage to DNA leads to mutations in specific genes

  • cell division
  • DNA repair
  • cell cycle arrest

Repair of UV induced DNA damage
• Photoproducts are removed by a process called Nucleotide Excision Repair
• Xeroderma pigmentosum (treated with removal of cancer ad sun protection)
Genetic condition with defective Nucleotide Excision Repair

Mutations that cause cancer

  1. Mutations that stimulate uncontrolled cell proliferation Eg abolishing control of the normal cell cycle (p53 gene)
  2. Mutations that alter responses to growth stimulating / repressing factors
  3. Mutations that inhibit programmed cell death (apoptosis)
136
Q

Explain sunburn and photocarcinogenesis.

A

UV leads to keratinocyte cell apoptosis
‘Sun burn’ cells are apoptotic cells in UV overexposed skin
Apoptosis removes UV damaged cells in the skin which might otherwise become cancer cells

Photocarcinogenesis
UV light —> p53 mutation —> skin cancer
UV —> DNA damage —> repair of DNA (p52 stops cell division) —> normal cell
UV —> damage too severe, unable to repair —> cell death, apoptosis

Immunomodulatory effects of UV light
• UVA and UVB effect the expression of genes involved in skin immunity
Depletes Langerhans cells in the epidermis (involved in antigen presentation)
• Reduced skin immunocompetence and immunosurveillance - no immune response to cause cell death, instead skin cancer develops
Basis for UV phototherapy for eg psoriasis
• Further increases the cancer causing potential of sun exposure

137
Q

Describe skin types and melanin.

A

Host response to UV is determined by genetic influences especially skin phenotype

Fitzpatrick Phototypes
• 1 - Always burns never tans
• II - Usually burns, sometimes tans
• III - Sometimes burns, usually tans
• IV - Never burns, always tans
• V - Moderate constitutive pigmentation - Asian
• VI - Marked constitutive pigmentation - Afrocaribean

Melanin
• Melanin pigmentation is responsible for skin colour
• Produced by melanocytes within the basal layer of the epidermis
• Skin colour depends on the amount and type of melanin produced not the
density of melanocytes (which is fairly constant) = no of melanocytes between all skin types remain the same; it’s the amount of melanin they produce that differs

Melanocytes = after sunlight exposure (UV), keratinocytes will send out paracrine messages - MSH which affects melanocytes to form melanin. Melanin taken up by keratinocytes and placed around nucleus = protection

• Two types of melanin are formed:
Eumelanin – brown or black
Phaeomelanin – yellowish or reddish brown Melanin is formed from tryosine via a series of enzymes

• MCR1 gene
>20 gene polymorphisms
Variation in eumelanin : phaeomelanin produced
Explains different hair colour and skin types

Melanin dictates skin sensitivity to UV damage

138
Q

Describe tumours of melanocytes.

A

Malignant tumour of melanocytes
-Melanocytes become abnormal -Atypical cells and architecture

Caused by

  • UV exposure
  • Genetic factors

Risk of metastasis: Pagetoid spread, atypical melatocyes because located in epidermis normally in basement membrane (resembles Paget’s)

Lentigo maligna (melanoma in situ)

  • proliferation of malignant melanocytes within the epidermis
  • no risk of metastasis
  • irregular shape
  • light and dark brown colours
  • size usually > 2 cm

Lentigo maligna melanoma

  • local invasion
  • superficial spreading
  • lateral proliferation of malignant melanocytes
  • invade basement membrane
  • risk of metastasis
  • melanocytes in dermis and epidermis - radial growth phase - heading outwards and vertical growth phase - downwards
Diagnosis of superficial spreading malignant melanoma 
ABCD rule
Asymmetry
Border irregular
Colour variation (dark brown-black)
Diameter > 0.7 mm and increasing 
Erythema 

Nodular malignant melanoma
Vertical proliferation of malignant melanocytes (no previous horizontal growth)
Risk of metastasis

Nodular melanoma arising within a superficial spreading melanoma

  • downward proliferation of malignant melanocytes
  • following previous horizontal growth
  • nodule developing within irregular plaque
  • prognosis will become worse

Acral lentiginous melanoma
Pigmented plaques/lumps on sole

Amelanotic melanoma
Non-pigmented

Overall types of malignant melanoma:
• Superficial spreading 
• Nodular 
• Lentigo maligna melanoma 
• Acral lentiginous 
• Amelanotic

Melanoma recognition - ABCD
• Asymmetry • Border • Colour • Diameter

Prognosis of melanoma can be found by using Breslow thickness; see depth of invasion from granular epidermis to where it extends

139
Q

What are some risk factors for the development of melanoma?

A
  • family history of melanoma
  • ultraviolet irradiation
  • intermittent burning exposure in unacclimatised fair skin
  • personal history of melanoma
  • skin type I, II
140
Q

Distinguish between a keratoacanthoma and SCC.

A

Keratoacanthoma grows rapidly into a volcano like shape

SCC from keratinocytes, if well differentiated it will make keratin 
Malignant tumour of keratinocytes 
Caused by:
-UV exposure    
-HPV    
-Immunosuppression    
May occur in scars or scarring processes 
Risk of metastasis
141
Q

Describe basal cell carcinomas.

A
Malignant tumour arising from basal layer of epidermis 
Caused by:
-sun exposure    
-Genetics 
Slow growing 
Invades tissue, but does not metastasise Common on face
Glistening, telangiectasia 
Can be nodular, superficial
142
Q

Describe mycosis fungoides, Kaposi’s and epidermodysplasia veruciformis.

A

Mycosis fungoides
Cutaneous and cell lymphoma
Looks like psoriasis

Kaposi’s sarcoma HIV and HHV8 associated - rises from endothelial cells of lymphatics

Epidermodysplasia veruciformis
Rare autosomal recessive condition
Predisposition to HPV induced warts and SCCs