Cancer Flashcards

(663 cards)

1
Q

What is metaplasia?

A

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

Adaptive

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

What is 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

Increased mitotic figures (abundant, abnormal and in places where not usually found)
Loss of architectural orientation
Loss in uniformity in uniformity of individual cells
Hyperchromatic, enlarged nuclei

Potentially (but rarely) reversible

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

When is dysplasia common?

A
Cervix= HPV infection
Bronchus= smoking-> squamous dysplasia develops in lung
Colon= UC
Larynx= smoking
Stomach= pernicious anaemia
Oesophagus= acid reflux
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

When stained, what does it mean if dysplasia cells are dark?

A

Nuclei seen

High grade dysplasia

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

What is neoplasia?

A

Tumour, malignancy

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
6
Q

Outline benign tumour characteristics

A
Don't invade metastasise
Encapsulated (compresses tissue around it to be in a capsule)
Usually well differentiated
Slowly growing
Normal mitoses

NB. exceptions to this e.g. unencapsulated benign

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

When can benign tumours be fatal?

A

If in a dangerous place e.g. meninges (-> epilespy), pituitary (adenoma)

If it secretes something dangerous e.g. insulinoma

Get infected: bladder (block ureter)

Bleeds= stomach

Ruptures= liver adenoma

Torts (twisted)= ovarian cyst
- Twist on avascular pedestral-> lose blood supply-> die-> necrotic tissue

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

Outline malignant tumours characteristics

A
Invade surrounding tissues
Spread to distant sites
No capsule
Well to poorly differentiated (normally poorly)
Rapidly growing
Abnormal mitoses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What is a metastasis?

A

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

Depend on lymphatic and vascular drainage of the primary site

Lymph node involvement has a worse prognosis

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

Benign and malignant tumours are distinguished from each other by all of the following except:

Degree of differentiation
Speed of growth
Capsulation 
Invasiveness
Site
A

Site

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

Well differentiated tumours are characterised by all of the following, except:

A small numbers of mitoses.
Lack of nuclear pleomorphism
A high nuclear-cytoplasmic ratio
Relatively uniform nuclei
Close resemblance to the corresponding normal tissue
A

A high nuclear-cytoplasmic ratio

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

Define: papilloma

A

Benign epithelial tumour

Of surface epithelium e.g. skin, bladder

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

Define: adenoma

A

Benign epithelial tumour

Of glandular epithelium e.g. stomach, thyroid, colon, kidney, pituitary, pancreas

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

Define: carcinoma

A

Malignant tumour derived from epithelium

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

Define: squamous cell carcinoma

A

Skin cancer (malignant derived from epithelium)

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

Define: adenocarcinoma

A

From glandular structures (malignant derived from epithelium)

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

Define: transitional cell carcinoma

A

Mostly urinary tract (malignant derived from epithelium)

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

Define: basal cell carcinoma

A

From the skin (malignant derived from epithelium)

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

Define: osteoma

A

Benign soft tissue tumour

Of bone

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

Define: sarcoma

A

Malignant tumour derived from connective tissue (mesenchymal) cells

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

Define: liposarcoma

A

Fat sarcoma (malignant tumour from mesenchymal cells)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Define: osteosarcoma

A

Bone sarcoma (malignant tumour from mesenchymal cells)

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

Define: chrondrosarcoma

A

Cartilage sarcoma (malignant tumour from mesenchymal cells)

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

Define: rhabdomyosarcoma

A

Striate muscle sarcoma (malignant tumour from mesenchymal cells)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Define: leiomyosarcoma
Smooth muscle sarcoma (malignant tumour from mesenchymal cells)
26
Define: malignant peripheral nerve sheath tumour
Nerve sheath sarcoma (malignant tumour from mesenchymal cells)
27
Define: leukaemia
Malignant tumour of bone marrow derived cells which circulate in the blood ``` Acute= blastic Chronic= more mature ```
28
Define: lymphoma
Malignant tumour of lymphocytes (usually) in lymph nodes Found in every organ
29
Define: teratoma
Tumour derived from germ cells, which have the potential to develop into tumours of all 3 germ cell layers Ectoderm Mesoderm Endoderm
30
How do teratomas differ in men and women?
Commonly affect testes and ovaries (gonadal teratomas) ``` Ovaries= often benign Testes= almost always malignant ```
31
Define: 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
32
A benign tumour of glandular tissue is: ``` An adenoma A leiomyoma An adenocarcinoma A squamous papilloma A lymphoma ```
An adenoma
33
A malignant tumour derived from soft tissue is a: ``` Carcinoma Sarcoma Teratoma Lymphoma Melanoma ```
Sarcoma
34
What are the criteria for assessing differentiation of a malignant tumour?
Evidence of normal function still present production of: Keratin, mucin, bile, hormones Various grading systems- for Ca breast, prostate, colon No differentation= anaplastic carcinoma
35
What is TNM?
TUMOUR NODE METASTASIS Grade= degree of differentiation Stage= how far tumour has spread Tumours of higher grade (i.e. poorly differentiated) tend to be of higher stage (i.e. spread further) STAGE IS MORE IMPORTANT THAN GRADE in determining prognosis
36
How does cell division vary in specific cells?
Different cells divide at different rates Embryonic vs adult cells Complexity of system Necessity for renewal (e,g. intestinal epithelial cells much quicker than hepatocytes) State of differentiation (some never divide e.g. neurons and cardiac myoctyes) Tumour cells
37
What does premature, aberrant mitosis cause?
Premature, aberrant mitosis results in cell death
38
What does it mean if a tumour is aneuploid?
Abnormal chromosome number and content Mutations in oncogenes and tumour suppressor genes-> most solid tumours are aneuploid
39
What is chromosome instability?
Loose and gain whole chromosomes during cell division
40
What can cause abnormal mitosis in cell division?
Perturbation of protein levels of cell cycle regulators is found in different tumours- abnormal mitosis
41
True or false: contact inhibition of growth can affect cell division?
True
42
What can attacking the machinery that regulates chromosome segregation do?
One of the most successful anti-cancer strategies in clinical use
43
What is the cell cycle?
Orderly sequence of events in which a cell duplicates its contents and divides in two Duplication Division Co-ordination
44
What is regulated progression through the cell cycle?
M-phase= mitosis (division) - Nuclear division - Cell division (cytokinesis) Interphase (duplication= G0, G1, S, G2) - DNA - Organelles - Protein synthesis
45
Why is mitosis the most vulnerable period of the cell cycle?
Cells are more easily killed (irradiation, heat shock, chemicals) DNA damage can not be repaired Gene transcription silenced Maybe metabolism?
46
Outline the eukaryotic cell cycle
M phase= mitosis ``` Interphase: G0= cell cycle machinery dismantled G1 phase (Gap)- decision point S phase= synthesis of DNA/ protein G2 phase (Gap)- decision point ```
47
What happens in the S phase?
Replication for division DNA replication Protein synthesis= initiation of translation and elongation increased: capacity is also increased Replication of organelles (centrosomes, mitochondria, Golgi etc) in case of mitochondria, needs to coordinate with replication of mitochondrial DNA
48
What is the centrosome?
Consists of 2 centrioles (barrels of nine triplet microtubules) - mother and daughter centriole Functions= microtubule organizing centre (MTOS) and mitotic spindle
49
What happens to centrosomes during mitosis?
G1= pair of centrosomes Then split up and duplicate themselves-> elongated microtubules Then they continuously divide Microtubules growing from centrosomes at nucleating sites (from y-tubulin ring complexes)
50
What are the 6 phases of mitosis?
``` Prophase Prometaphase Metaphase (Metaphase to anaphase transition) Anaphase Telophase ``` Cytokinesis
51
What happens in prophase?
Condensation of chromatin (replicated chromosomes condense) Duplicated centrosomes migrate to opposite sides of the nucleus and organize the assembly of spindle microtubules Mitotic spindle forms outside nucleus between the 2 centrosomes
52
What does a condensed chromosome consist of?
Each consists of 2 sister chromatids, each with a kinetochore (around the centromere)
53
Outline spindle formation
Radial microtubule arrays (ASTERS) form around each centrosome (microtubule organising centres- MTOC) Radial arrays meet Polar microtubules form NB. Microtubules are in a dynamic state
54
Outline metaphase
Chromosomes aligned at equator of the spindle NB. Prometaphase early and late
55
What happens in early prometaphase?
Breakdown of nuclear membrane Spindle formation largely complete Attachment of chromosomes to spindle via kinetochores (centromere region of chromosome)
56
What happens in late prometaphase?
Microtubule from opposite pole is captured by sister kinetochore Chromosomes attached to each pole congress to the middle Chromosome slides rapidly towards centre along microtubules
57
What is CENP-E?
Centromere protein E (kinetochore tension sensing)
58
What happens in anaphase?
Paired chromatids separate to form 2 daughter chromosomes Cohesin holds sister chromatids together Anaphase A and B
59
What happens in anaphase A?
Breakdown cohesin Microtubules get shorter Daughter chromosomes pulled toward opposite spindle poles
60
What happens in anaphase B?
1- Daughter chromosomes migrate towards poles | 2- Spindle poles (centrosomes) migrate apart
61
What happens in telophase?
Daughter chromosomes arrive at spindle Nuclear envelope reassembles at each pole Assembly of contractile ring
62
What happens in cytokinesis?
New membrane inserted Acto-myosin ring contracts Midbody begins to form Chromatic decondenses Nuclear substructures reform Interphase microtubule array reassembles
63
What happens at the spindle assembly checkpoint?
During transition out of metaphase Senses completion of chromosome alignment and spindle assembly (monitors kinetochore activity) *Unattached kinetochores generate checkpoint signals* Requires CENP-E and BUB protein kinases (Need BUBs to dissociated to-> anaphase)
64
What involvement do BUBs have in spindle assembly?
BUBs dissociate from kinetochore when chromosomes are properly attached to the spindle When all dissociated, anaphase proceeds
65
What is an amphellic attachment?
When the sister kinetochores are correctly connected to microtubules from opposite poles -> bioriented chromosome
66
What is a monotelic attachment?
Only one of the sister chromatids is connected to a spindle pole, the chromosome is mono-orientated
67
What is a syntelic attachment?
Both sister kinetochores are attached to a single spindle pole and the chromosome is mono-orientated
68
What is a merotelic attachment?
Usually one (or rarely both) sister kinetochores are connected to both poles instead of one Chromosomes are bioriented in merotelic attachments
69
How does aneuploidy occur?
Mis-attachment of microtubules to kinetochores Aberrant centrosome/DNA duplicaiton
70
In aneuploidy: outline mis-attachment of microtubules to kinetochores
Cohesion defects/synthelic attachment-> both sister chromatatids at the same pole Merotelic attachment-> chromosome loss at cytokinesis
71
In aneuploidy: outline aberrant centrosome/DNA duplication
Aberrant cell cycle (DNA and centrosome duplication affected)-> multipolar spindle-> aberrant cytokines
72
How does anti-cancer therapy work by inducing gross chromosome mis-segregations?
Inhibition of attachment-error-correction mechanism Checkpoint kinase inhibitior
73
What are taxanes and vinca alkaloids?
Breast and ovarian cancers Alter microtubule dynamics Produces unattachment kinetochores Causes long-term mitotic arrest
74
What happens if something goes wrong during the cell cycle?
CELL CYCLE ARREST At check points (G1 and spindle check point) Can be temporary (i.e. following DNA repair) PROGRAMMED CELL DEATH (APOPTOSIS) DNA damage too great and cannot be repaired Chromosomal abnormalities Toxic agents Cell cycle progression aborted and cell destroyed
75
What are the cell cycle checkpoints preventing tumour progression?
G2 checkpoint checks for DNA damage (TUMOURS INHIBIT THIS) Metaphase checkpoint checks for sister chromatid alignment (TUMOURS INHIBIT THIS) G1 checkpoint for growth factors (TUMOURS CAUSE THIS)
76
What happens to the cell cycle during tumorigenesis?
De-regulation of cell cycle Exit cell cycle (G0)-> dismantle cell cycle apparatus (TUMOURS INHIBIT THIS)
77
What triggers a cell to enter the cell cycle and divide?
In the absence of stimulus, cells go into Go (quiescent phase) Most cells in the body which are differentiated to perform specific functions Cells are not dormant, but are non-dividing Exit from G0 highly regulated- requires growth factors and intracellular signalling cascades
78
What signally cascades are there through the cell?
Response to extracellular factors Signal amplification Signal integration Modulation by other pathways Regulation of divergent responses
79
How are peptide growth factors involved in signalling?
Ligand binds and activates the receptor - Epidermal growth factor (EGF): platelet-derived growth factor (PGDF) - Respective receptors found as monomeric, inactive state - Receptor Protein Tyrosine Kinase (RPTK) ---- In presence of ligand - Receptors form dimers - Are activated by phosphorylation (of AA residues in kinase domain)
80
What happens in protein phosphorylation?
Transfer of phosphate from ATP to hydroxyl groups The added phosphate group (negatively charged) can alter protein function by: - Causing a change in shape (conformation) leading to change in activity (+ve or –ve) - Creating a docking site for another protein
81
How does signalling by peptide growth factors affect phosphorylation?
Signalling by peptide growth factors - > receptors form dimers, are activated by phosphorylation - > receptor activation - > triggers different phosphorylation events (kinase cascades and binding of adapter proteins)
82
How do protein kinase cascades work?
Protein regulated by a kinase may be another kinase etc. Leads to signal amplification, diversification and opportunity for regulation NB. phosphorylation by kinases and reversed by phosphatases NB2. Protein kinase cascades driven by GFs
83
What phase do adult cells go into when they aren't receiving growth signals?
Most adult cells aren't constantly dividing In absence of growth signals they go into the G0 (or quiescent) phase E.g. liver hepatocytes
84
What is the restriction point?
Point in G1 (end towards S) | Where cell monitors its own size and external signals
85
What is the role of c-Myc?
c-Myc= transcription factor that stimulates the expression of cell cycle genes Key role in cell cycle entry
86
What are the key components of signalling pathways?
Regulation of enzymes activity by protein phosphorylation (kinases) Adapter proteins Regulation by GTP-binding proteins
87
Outline growth factor stimulation of signalling pathways
Mitogenic GF - > receptor protein tyrosine kinase - > small G (GTP-binding) protein (Ras) - > kinase cascade - > immediately early genes (c-Jun, c-Fos, c-Myc- TFs)- control the expression of other genes
88
Give examples of mitogenic GFs?
Growth signals from other cells e/g/ hepatocyte growth factor released after liver damage
89
What is Anti-HER2 ab used for?
Cancer treatment Prevents further activation Blocks all signals through the cell's interior
90
Where do adaptor proteins bind?
Tyrosine phosphorylation provides docking sites for adaptor proteins Protein-protein interactions: protein binding- bringing proteins together
91
Why are modular proteins useful?
Proteins are modular and contain domains Some domains are important in molecular recognition- have no enzymatic function of their own, simply bring other proteins together
92
Outline Src homology regions in Grb2
``` SH3= bind to proline-rich regions (constitutive) SH2= bind to phosphorylated tyrosines (inducible, specific sequence context) ``` SH3-SH2-SH3
93
Outline how GTP binding acts as a molecular switch in Ras?
Exchange factors e.g. Sos (exchange of GTP for GDP) - Signal in-> GTP binding activates-> Ras ON -> signaling by GTP-binding protein (not kinases) GAP (GTPase activating proteins) and GTP hydrolysis inactivates -> Ras OFF
94
How does Sos work?
Exchange factor | Constitutively (i.e. always bound to Grb2 via SH3 domains)
95
What do RPTKs signal to?
Ras
96
How is Ras oncogenically activated?
Mutations-> increase amount of active GTP-loaded Ras e. g. V12 Ras- constitutively active (glycine-> valine)= prevents GAP binding (i.e. prevents inactivation) e. g. L61 Ras- constitutively active (glutamine-> leucine)= prevents GTP hydrolysis
97
What Ras activates protein kinase cascades?
ERK (specifically)= EC signal-regulated kinase cascade MAPK (generically)= mitogen-activated protein kinase cascades
98
Outline the ERK casade
Raf MEK ERK (NB. generic= MAPKKK, MAPKK, MAPK)
99
How do protein kinases promote division?
Protein kinases stimulate changes in cell proteins and gene expression to promote division
100
What are Myc and Ras?
Oncogenes Activated by protein kinases
101
In relation to signalling from receptor protein tyrosine kinases (RPTKs), which of these statements is most accurate? RPTK ligands diffuse across the cell membrane and bind to the cytoplasmic domain of the receptor RPTKs use GTP as a phosphate donor Adaptor proteins phosphorylate target proteins Ras is activated by phosphorylation Ras bound to GTP activates the ERK cascade
Ras bound to GTP activates the ERK cascade
102
What do checkpoints in mitosis need?
Need correct timing and sequence
103
What are cyclin-dependent kinases (Cdks)?
Cell cycle based is on cyclically activated protein kinases Cdks are present in proliferating cells throughout cell cycle Activity is regulated by: - Interaction with cyclins - Phosphorylation
104
What do cyclins activate?
Cdks
105
What are cyclins?
Transiently expressed at specific points in the cell cycle Regulated at level of expression Synthesised, then degraded
106
Why are there different cyclin-Cdk complexes?
Different cyclin-Cdk complexes trigger different events in the cell cycle Different cyclins and different cdks required at different stages of cell cycle Cyclins activate Cdks but also alter substrate specificity Substrate accessibility changes through the cell cycle
107
What is MPF (mitosis promoting factor) comprised of?
Cdk1 and mitotic cyclin B
108
How are Cdks regulated by phosphorylation?
Cdks require activating phosphorylation And removal of inactivating phosphorylation
109
When is Cdk1 dephosphorylated?
At the end of interphase Dephosphorylation activates Cdk1 Positive feedback Reinforces activation of MPF and drives mitosis
110
How is cyclin B degraded?
Signal from fully attached kinetochores causes cyclin B to be degraded: Cdk1 inactivated Key substrates dephosphorylated Mitosis progresses
111
What happens when cdk1/cycB are active?
Mitosis 'on hold'- key substrates phosphorylated
112
What happens from G0 to G1?
Growth factor stimulates the pathway that promotes G0 to G1 transition Immediate early gene transcriptionfactors (e.g. c-jun, c-fos, c-myc) -> Stimulate transcription of other genes (e.g. cyclin D) (OVERALL- GFs bind to R-> Activated cell cycle control system)
113
How does cyclin D stimulate synthesis of cyclin E?
Cyclin D activates Cdk4 and Cdk6 to stimulate synthesis of cyclin E
114
How is the expression of cyclins and Cdks regulated?
Timing process Cdks become sequentially active and stimulate synthesis of genes required for next phase, e.g. cycD/Cdk4/6 stimulates expression of cycE – gives direction and timing to cycle Cyclins susceptible to degradation, hence cyclical activation
115
What do Cdks do?
Phosphorylate proteins (on serine or threonine) to drive cell cycle progression Cdk1 with mitotic cyclin (e.g. B) e.g. nuclear lamins (causes breakdown of nuclear envelope) Cd2 with G1 cyclin (e.g. E) e.g. retinoblastoma protein (pRb)
116
How is gene expression regulated by Rb?
pRb acts as a “brake” on the cell cycle (stops G0-> prolierating cell) Cdks phosphorylate (at multiple sites) and progressively inactivate pRb Rb is a “tumour suppressor”
117
What genes are regulated by the transcription factor E2F?
``` PROTO-ONCOGENES c-Myc N-Myc B-Myb IGF-1 ``` ``` CELL CYCLE E2F-1,2,3 pRb p107 cyclin A cyclin E CDK4 CDK2 ``` ``` DNA SYNTHESIS Thymidine kinase Thymidine synthetase Dihydrofolate reductase (DHFR) DNA Polymerase ```
118
How are Cdks regulated by CKIs (Cdk inhibitors)?
Cdk inhibitor binding-> inactivion of cyclin-cdk binding
119
What are the families of CKIs?
G1 PHASE CKIS= inhibit Cdk4/6 by displacing cycD ``` INK4 family: p15INK4b p16INK4a p18INK4c p19INK4d ``` -- S PHASE CKIS= inhibit Cdks by binding to the Cdk/cyc complex CIP/KIP family: p21CIP1/WAF1 p27KIP1 p57KIP2 Must be degraded to allow cell cycle progression
120
How do EFGR/HER2 oncogenes lead to cancer and how is it treated?
Mutationally activated or overexpressed in many breast cancers Treated= Herceptin antibody for the treatment of HER2-positive metastatic breast cancer
121
How do Ras oncogenes lead to cancer and how is it treated?
Mutationally activated in many cancers Treated= inhibitors of membrane attachment
122
How do Cyclin D1 oncogenes lead to cancer?
Over expressed in 50% of cancers
123
How do B-Raf oncogenes lead to cancer and how is it treated?
Mutationally activated in melanomas Treated= kinase inhibitors in trials
124
How do c-Myc oncogenes lead to cancer?
Overexpressed in many tumours
125
How do Rb tumour suppressors lead to cancer?
Inactivated in many cancers
126
How do p27KIP1 tumour suppressors lead to cancer?
Underexpression correlates with poor prognosis in many malignancies
127
In relation to cell cycle control, which of these statements is most accurate? Cyclin degradation activates Cdks Myc stimulates the cell cycle Cdk1 must be phosphorylated at 2 sites to be active Rb is activated by phosphorylation The E2F-Rb complex activates transcription of target genes
Cdk1 must be phosphorylated at 2 sites to be active
128
What are the main anti-cancer treatment modalities?
``` Surgery Radiotherapy Chemotherapy Immunotherapy Hormonal therapy ```
129
True or false: cancer is a disease of the genome?
True
130
What kinds of genetic mutations can cause cancer?
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
131
What kinds of systemic therapy are there?
Cytotoxic chemotherapy Target therapies
132
What kinds of cytotoxic chemotherapy are there?
``` Alkylating agents Antimetabolites Anthracyclines Vinca alkaloids and taxanes Topoisomerase inhibitors ```
133
What kinds of target therapies are there?
Small molecule inhibitors | Monoclonal antibodies
134
How do cytotoxic chemotherapies work biochemically?
'Select' rapidly dividing cells by targeting their structures (mostly the DNA)
135
How/when is cytotoxic chemotherapy given?
Given IV or by mouth Works systemically Given: - Post-op= adjuvant - Pre-op= neoadjuvant As a monotherapy or in combo Curative or palliative intent
136
Why are there so many side effects to cytotoxic chemo?
Non "targeted"- affects all rapidly dividing cells in the body
137
What are alkylating agents in cytotoxic therapies?
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-pairing - oncogenic
138
What are pseudo-alkylating agents in cytotoxic therapies?
Add platinum to guanine residues in DNA Same mechanism of cell death as akylating agents Examples: carboplatin, cisplatin, oxaliplatin
139
Give examples of alkylating agents
Chlorambucil, cyclophosphamide, dacarbazine, temozolomide
140
Give examples of pseudoalkylating agents
Carboplatin, cisplatin, oxaliplatin
141
What are the SEs of alkylating and psudoalkylating agents?
``` Hair loss (not carboplatin) Nephrotoxicity Neurotoxicity Ototoxicity (platinums) Nausea Vomiting Diarrhoea Immunosuppression Tiredness ```
142
What are anti-metabolites in cytotoxic therapies?
Masquerade as purine or pyrimidine residues leading to inhibition of DNA synthesis, DNA double strand breaks and apoptosis Block DNA replication and transcription Can be purine (A/G), pyrimidine (T/U/C) or folate antagonists
143
Give examples of anti-metabolites
Methotrexate (folate), 6-mercaptopurine, decarbazine and fludarabine (purine), 5-fluorouracil, capecitabine, gemcitabine (pyrimidine)
144
Why are folate-antagonists (anti-metabolites) used in cytotoxic chemo therapies?
Inhibit dihydrofolate reductase required to make folic acid, an important building block for all nucleic acids- especially thymine
145
What are the SEs of anti-metabolites (cytotoxic chemotherapies)
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) Fatigue
146
What do anthracyclines do (cytotoxic chemo)?
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
147
Give examples of anthracyclines (cytotoxic chemo)
Doxorubicin | Epirubicin
148
What are the SEs of anthracyclines (cytotoxic chemo)?
``` 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”) ```
149
What do vinca alkaloids and taxanes do (cytotoxic chemo)?
Originally derived from natural sources Work by inhibiting assembly (vinca alkaloids) or disassembly (taxanes) of mitotic microtubules causing dividing cells to undergo mitotic arrest
150
What are the SEs of microtubule targeting drugs (vinca alkaloids and taxanes- cytotoxic chemo)?
Nerve damage: peripheral neuropathy, autonomic neuropathy Hair loss Nausea Vomiting Bone marrow suppression (neutropenia, anaemia etc) Arthralgia Allergy
151
What are topoisomerase inhibitors (cytotoxic chemo)?
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 Effects through their action on DNA
152
Give examples of specific topoisomerase inhibitors (cytotoxic chemo)
Topotecan and irinotecan (topo I) Etoposide (topo II) Alter binding of the complex to DNA and allow permanent DNA breaks
153
What are the SEs of topoisomerase inhibitors?
(Irinotecan): Acute cholinergic type syndrome – diarrhoea, abdominal cramps and diaphoresis (sweating). Therefore given with atropine Hair loss Nausea, vomiting Fatigue Bone marrow suppression
154
If you're undergoing chemo, what temperature do you need to reach before you need to go to hospital?
>38.5%
155
What is 5-flurouracil (5-FU)?
Anti-metabolite (cytotoxic chemo) ``` SEs: PPE Nausea and vomiting Bone marrow suppression causing anaemia, neutropaenia and thrombocytopaenia Mucositis Diarrhoea ``` ``` Treatment: Pyridoxine (vitamin B6) Antiemetics Transfusions/platelets/GCSF/dose reduction Mouth washes Loperamide ```
156
What is epirubicin?
Anthracycline (cytotoxic chemo) ``` SEs: Cardiac toxicity Alopecia Neutropaenia Nausea and vomiting Fatigue Skin changes ``` ``` Treatment: Irreversible but can cap dosing Scalp cooling Antiemetics Transfusions/platelets/GCSF/dose reduction ```
157
What is cyclophosphamide?
Alkylating agent (cytotoxic chemo) ``` SEs: Alopecia Nausea Vomiting Diarrhoea Immunosuppression ``` Treatment: Scalp cooling Antiemetics Loperamide
158
Outline resistance mechanisms in cancer cytotoxic chemo treatment
DNA adducts replaced by Base Excision repair (using PARP) DNA repair mechanisms upregulated and DNA damage is repaired-> DNA double strand breaks Drug effluxed from the cell by ATP-binding cassette (ABC) transporters THE RESISTANT CELL SURVIVES
159
What is the difference in 'cancer wiring' in monogenic cancers and others?
You can “cut the wiring” in monogenic cancers but for others, parallel pathways or feedback cascades are activated
160
What is the use of 'dual kinase inhibitors'?
Prevent feedback loops but increase toxicities New therapeutic strategies required
161
What are the ten hallmarks of the cancer cell?
``` 6 FIRST KNOWN Self-sufficient Insensitive to anti-growth signals Anti-apoptotic Pro-invasive and metastatic Pro-angiogenic Non-senescent ``` ``` EMERGING HALLMARKS Dysregulated metabolism Evades the immune system Unstable DNA Inflammation ``` siappndeui= u pained pis
162
Why are cancer cells described as self-sufficient in growth signals?
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
163
Outline the GF R pathway?
GF Rs move closer to each other C terminal region connections Bind to SH2 Kinase cascade and signal amplification
164
Describe a GF receptor
Crosses membrane Ligand-binding site Kinase domain C-terminal region with lots of tyrosines (for autophosphorylation)
165
What percentage of RTKs are associated with human malignancies?
>50% associated with human malignancies
166
What happens in overexpression of receptors the GF pathway in cancer? (To HER2, EGFR, PDGFR, VEGF)
HER2= amplified and overexpressed in 25% breast cancer EGFR= overexpressed in breast (and colorectal) cancer PDGFR= glioma in brain cancer VEGF= prostate cancer, kidney cancer, breast cancer Overall there is increased kinase cascade and signal amplification
167
What happens in constitutive (ligand independent) receptor activation in cancer?
``` EGFR= lung cancer FGFR= head and neck cancers, myeloma ``` Increase kinase cascade and signal amplification
168
``` What kind of monoclonal antibodies end: - momab - ximab - zumab - mumab ? ```
- momab (derived from mouse antibodies) - ximab (chimeric) e.g cetuximab - zumab (humanised) e.g. bevacizumab trastuzumab - mumab (fully human) e.g. panitumumab
169
What happens to humanized monoclonal antibodies?
Murine regions interspersed within the light and heavy chains of the Fab portion
170
What happens to chimeric monoclonal antibodies?
Murine component of the variable region of the Fab section is maintained integrally
171
How do monoclonal antibodies target the EC component of the receptor?
They neutralise the ligand-> prevent receptor dimerisation Cause internalisation of receptor mAbs also activate Fcγ-receptor-dependent phagocytosis or cytolysis induces complement-dependent cytotoxicity (CDC) or antibody-dependent cellular cytotoxicity (ADCC)
172
What does bevacizumab do?
Monoclonal antibody Binds and neutralises VEGF Improves survival in colorectal cancer
173
What does cetuximab do?
Monoclonal antibody Targets EGFR
174
How do small molecule inhibitors affect the GF pathway?
Bind to the kinase domain of the tyrosine kinase within the cytoplasm and block autophosphorylation and downstream signalling (I.e. target internal TK)
175
What is glivec?
The first targeted therapy (cancer) A small molecule inhibitor and targets the ATP binding region within the kinase domain Doesn't work because all cancers aren't monogenic
176
Give examples of small molecule inhibitors that act on RTKs
Act on RTKs but also IC kinases= therefore can affect cell signalling pathways Inhibiting receptors include erlotinib (EGFR), gefitinib (EGFR), lapatinib (EGFR/HER2), sorafinib (VEGFR)
177
Why are small molecule inhibitors useful?
They block cancer hallmarks (e.g. VEGF inhibitors alter blood flow to a tumour) without the toxicity observed with cytotoxics
178
What are the advantages and disadvantages of monoclonal antibodies?
``` ADVANTAGES High target specificity Cause ADCC, complement mediated cytotoxicity and apoptosis induction Can be radiolabelled Cause target receptor internalisation Long half-life (lower dosing frequency) Good for haematological malignancies Liked by regualatory authorities (FDA) ``` DISADVANTAGES Resistance Large and complex structure (low tumour or BBB penetration), Less useful against bulky tumours Only useful against targets with extracellular domains Not useful for constitutively activated receptors Cause immunogenicity, allergy Parenteral (IV) administration Risky! (though humanisation reduces risk) Expensive
179
What are the advantages and disadvantages of small molecules in targeted therapy?
ADVANTAGES Can target TKs without an extracellular domain or which are constitutively activated (ligand independent) Pleiotropic targets (useful in heterogenic tumours/ cross talk) Oral administration Good tissue penetration Cheap DISADVANTAGES Resistance Shorter half-life, more frequent administration Pleiotropic targets (more unexpected toxicity)
180
What are the mechanisms of resistance to targeted therapies?
Mutations in ATP-binding domain (e.g BCR-Abl fusion gene and ALK gene, targeted by Glivec and crizotinib respectively) Intrinsic resistance (herceptin effective in 85% HER2+ breast cancers, suggesting other driving pathways) Intragenic mutations Upregulation of downstream or parallel pathways
181
What are anti-sense oligonucleotides used for?
Used in cancer for 'undruggable' targets 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
182
What are the types of targeted therapies (cancer)?
Small molecule inhibitors Monoclonal antibodies
183
What is RNA interference and what is it used for (cancer therapy)?
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)
184
What are the problems with tumour heterogeneity in the targeted approach to cancer?
Tracking heterogeneity and bottlenecks Tumor sampling bias Drivers of heterogeneity Actionable mutations
185
How does programmed cell death 1 (PD-1) lead to immune modulation?
Present on the surface of cancer cells Required to maintain T cell activation After binding the ligand PDL1, the body’s T cells can no longer recognise tumour cells as foreign If either is blocked, immune system is stimulated
186
What is nivolumab?
An anti-PD1 antibody Had an effect on improving melanoma
187
What would be useful to do to tumours in the future before starting therapy?
Sequence them Useful for treatment and prognostic info
188
What are the new therapeutic avenues for cancer treatments?
Nanotherapies- delivering cytotoxics more effectively Virtual screening technologies to identify “undruggable” targets Immunotherapies using antigen presenting cells to present “artificial antigens” Targeting cancer metabolism
189
What is cell behaviour?
The term used to describe the way cells interact with their external environment and their reactions to this Particularly proliferative and motile responses of cells
190
What external influences are detected by cells?
Chemical (and biochem)= hormones, GFs, ion concs, ECM, molecules on other cells, nutrients and dissolved gas (O2/CO2) concs Physical= mechanical stresses, temp, the topography or 'layout' of the ECM and other cells
191
Which external factors influencing cell proliferation relate to cancer cell behaviour?
Growth factors Cell-cell adhesion Cell-ECM adhesion
192
How do cells behave in culture?
Cell starts spherically Cell settling on culture surface Spreading (acquires polarity- front) Acquiring motility
193
How does cell spreading occur?
Requires energy to modulate cell adhesion and the cytoskeleton during spreading Not passive, gravity-dependent event Can be tidy or more random
194
When is a cell likely to 'bleb'?
When it has no contact with the ECM substratum E.g. directly above another cell
195
Why do you need an adhesive path to study cell-ECM adhesion?
Agar is non adhesive Cells require binding to ECM to be fully competent for responding to soluble growth factors
196
Why is cell spreading important?
If the cell can't spread it will die by apoptosis Cell spreads-> survives and grows
197
What happens in cell-ECM adhesion?
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
198
What do cell-ECM adhesion molecules look like?
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
199
What are integrins?
Most important ECM receptors Heterodimer complexes of alpha and beta subunits Associate extracellularly by their HEAD regions Each of their TAIL regions spans the plasma membrane
200
What are the subunits of integrins?
Alpha/beta heterodimers (both sub-units span the plasma membrane one) 10a and 8B known
201
How do integrins recognise peptide sequences?
Recognise short, specific peptide sequences (e.g. α5β1 fibronectin receptor binds arg-gly-asp (RGD) More than 20 combinations of α/β known Each combination specifically binds a particular peptide sequence Such peptide sequences found in more than one ECM molecule, e.g. RGD found in fibronectin, vitronectin, fibrinogen plus others
202
How do integrins link?
Linked, via actin-binding proteins, to the actin cytoskeleton (most integrins) Integrin complexes cluster to form focal adhesions (most) or hemidesmosomes= involved in signal transduction Integrins also bind to specific adhesion molecules on some cells
203
Where is the α6β4 intergin complex found?
The α6β4 intergin complex found in epithelial hemidesmosomes Linked to the cytokeratin (intermediate filament) network
204
How do peptide sequences bind to integrins?
More than 20 combos of a/B heterodimers (in integrins) known which bind specifically to short peptide sequence on ECM proteins
205
How are ECM receptors (e.g. integrins) involved in transducing signals?
"Outside-in" integrin signalling = ECM receptors act to transduce signals (receive info from its adhesion to ECM)-> can alter the phenotype of the cell E.g. ECM binding to an integrin complex can stimulate the complex to produce a signal inside the cell "Inside-out" integrin signalling = A signal generated inside the cell can act on an integrin complex to alter the affinity of an integrin (i.e. alter its affinity for ECM binding) E.g. in inflam or blood-clotting, switching on adhesion of circulating leukocytes
206
What does the amount of force generated at a focal adhesion on the ECM depend on?
Amount of force that is generated at a focal adhesion depends: - The force generated by the cytoskeleton (F cell) - The stiffness of the ECM
207
How does "inside-out" activation extend the flexed complex?
Ligand-binding opens the legs of the complex (conformational change)-> allows cytoplasmic signalling molecules to bind
208
What do integrins recruit?
Recruit cytoplasmic proteins which promote both signalling and actin assembly
209
The matrix type has profound effects on the phenotype of cells- how does this vary between type 1 collagen and laminin?
In interstitial matrix (type 1 collagen), mammary epithelium does not differentiate to secretory cells In basal lamina (basement membrane) matrix, mammary cells organise into “organoids” and produce milk proteins
210
True or false: flow of medium inhibits cell proliferation?
False Flow of medium stimulates cell proliferation
211
What happens when there is a confluent monolayer formed in culture?
Cells have cell-cell contact-> cease proliferating and slow down many other metabolic activities There is also competition for external growth factors Density-dependence of cell division
212
Outline the ERK MAP kinase cascade
(NB. required for cyclin D expression) Adapter on RTK - > Ras - > Raf (MAPKKK) - > MEK (MAPKK) - > ERK (MAPK) - > Gene expression (proliferation) in nucleus NB. There is cross-talk between ECM and GF signalling
213
What do growth factors depend on in signals controlling proliferation of tissue cells?
Density dependence
214
What does the ECM depend on in signals controlling proliferation of tissue cells?
Anchorage dependence GF Rs and integrin signalling complexes can each activate identical signalling pathways (e.g. MAPK) Individually weak and/or transient but together= strong and sustained Separate signalling pathways act synergistically
215
What contact interactions are there between cells?
Short term= transient interactions b/w cells which don't form stable cell-cell junctions Long term= stable interactions resulting in formation of cell-cell junctions
216
What is contact inhibition of locomotion?
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 OVERALL EFFECT= Reponsible for preventing multi-layering of cells in culture and in vivo
217
What kind of cells have long-term cell-cell contacts?
Some cell types strongly adhere and form specific cell-cell junctions ``` Epithelial cells (form layers) Endothelial cells (form layers) Neurones (form synapses) ```
218
What is contact-induced spreading of epithelial cells?
Contact between epithelial cells leads to the mutual induction of spreading So 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
219
How does cell-cell adhesion affect cell proliferation?
When there are no cell-cell junctions, activated MAPK, decreased p27KIP1-> high proliferation When there are cell-cell junctions form, inactive MAPK, increased p27KIP1-> low proliferation NB. Can have adhesion blocking antibodies-> stimulates proliferation
220
Is B-catenin the link between cell-cell adhesion and proliferation?
Seems that cadherin binds to B-catenin which binds to a-catenin which binds to actinfilament
221
What is adenomatous polyposis coli (APC)?
Inherited colon cancer (number of familial forms) APC gene-product is a protein involved in the degradation of the junction-associated molecule (B catenin)
222
Outline B-catenin dynamics in cells
APC-> Degradation (cytoplasm) Cytoplasm-> APC Sequestered (bound to cadherin at plasma membrane) -> LEF-1-> gene transcription (nucleus) SEE DIAGRAM
223
What is the mechanism for contact inhibition of proliferation?
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 -> influences gene expression, leading to proliferation
224
What other adhesion-associated signalling pathways influence contact-induced inhibition of proliferation (not just B-catenin)?
Clustering of cadherins after cell-cell contact is known to alter the activation of small GTPases (Rac is activated, Rho is inhibited-> can influence proliferation) Some growth factor receptors are associated with cell-cell junctions -> reduced their capacity to promote proliferation
225
What does it mean that 'cells can lose their social skills'?
Cells can lose their behavioural constraints As a result, they will: - 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
226
What happens to contact inhibition in cancer cells?
Lost Forms a multilayer of uninhibited cancer cells (instead of contact-inhibited monolayer of normal cells)
227
Outline the mechanism of short-circuiting leading to uncontrolled proliferation?
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 proto-oncogenes This is the mechanism of short-circuiting -> uncontrolled proliferation as a result of loss of GF dependence etc.
228
Define: oncogene
Mutant gene which promotes uncontrolled cell proliferation
229
Define: proto-oncogene
Normal cellular gene corresponding to the oncogene
230
Give examples of proto-oncogenes
``` EGF receptor Ras (signalling intermediate) c-Raf (signalling intermediate) c-Jun (transcription Factor) ```
231
Give examples of oncogenes
V12Ras (Gly12Val mutation) L61Ras (Gln61Leu mutation) v-Raf (deletion of regulatory domain) v-Jun (deletion of regulatory domain)
232
What percentage of cancers have mutated Ras?
30% of all cancers
233
What happens during uncontrolled proliferation of tissue cells?
GF (density dependence lost) ECM (anchorage dependence lost) Signals short circuites Mutant gene products (constitutively active so upstream signals aren't required for the pathway to be 'on') -> nucleus-> proliferation
234
How do cancers spread?
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
235
How does a 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 (indicates its invasiveness and the prognosis)
236
What are the molecular mechanisms that regulate cell motility?
Microfilaments Regulation of actin dynamics Cytoskeletal proteins Signalling proteins
237
What are the overall steps of tumour progression from benign to malignant?
Homeostasis Genetic alterations Hyper-proliferation De-differentiation (disassembly, cell-cell contacts) Invasion (increased motility, cleavage of ECM proteins)
238
Outline the processes of metastasis
Epithelial cells in primary tumours are tightly bound together Metastatic tumour cells become mobile mesenchyme-type cells and enter the bloodstream Metastatic cells then travel through the bloodstream to a new location in the body Metastatic cells enter the circulation and invade a new organ Cancer cells lose their mesenchymal characteristics and form a new tumour
239
What are the types of migration strategy tumour cells use?
Ameoboid Mesenchmal (single cells or chains) Cluster/cohorts Multicellular strands/sheets
240
What kind of tumours have ameoboid migration?
Lymphoma Leukaemia SCLC
241
What kind of tumours have mesenchymal (single cell) migration?
Fibrosarcoma Glioblastoma Anaplastic tumours
242
What kind of tumours have cluster/cohorts migration?
Epithelial cancer | Melanoma
243
What kind of tumours have multicellular strands/sheets migration?
Epithelial cancer | Vascular tumours
244
What happens to the morphogenesis of cells during tumour cell metastasis?
``` 2D sheet Branching morphogenesis (mammary gland) Vascular sprouting Multi-cellular 3D invasion strands Border cells Detached cluster ```
245
What is the difference between the migration of primary glial cells and a glial tumour cell line?
Migration of primary glial cells= move as a coherent front until they touch eachother (contact inhibition of migration) Migration of a glial tumour cell line= move much fast, hyperactive motility in all directions with no regard for surrounding
246
What happens to genes in cytoskeleton regulation and motility machinery in primary tumours?
Up-regulated
247
What stimuli cause cells to move?
Organogenesis and morphogenesis Wounding GFs/chemoattractants Dedifferentiation (tumours)
248
What determines where cells move to and stop?
Where to go= directionality (polarity) When to stop= contact-inhibition motility (Moves with specialized structures e.g. focal adhesion, lamellae, filopodium)
249
What is filamentous action important for?
Important cellular functions as the mobility and contraction of cells during cell division
250
What is focal adhesion (in attachment to substratum, ECM proteins)?
Focal adhesion is a type of adhesive contact between the cell and extracellular matrix through the interaction of the transmembrane proteins integrins with their extracellular ligands, and intracellular multiprotein assemblies connected to the actin cytoskeleton
251
What are filopodia?
Finger-like protusions rich in actin filaments
252
What are lamellipodia?
Sheet-like protrusions rich in actin filaments
253
What control is needed in cell movement?
Within a cell to coordinate what is happening in different parts Regulate adhesion/release of cell-extracellular matrix receptors From outside to respond to external influence
254
What is haptotaxis?
Directional motility or outgrowth of cells
255
What is chemotaxis?
The movement of an organism in response to a chemical stimulus
256
What are the stages of cell motility?
Extension Adhesion Translocation De-adhesion
257
What is the difference between G and F actin?
``` G= small soluble units F= large filamentous polymer ```
258
How do actin filaments lead to polarity?
Signal (e.g. nutrient source) - > disassembly of filaments and rapid diffusion of subunits - > reassembly of filaments at a new site (F and G= different roles depending how they grow and shrink)
259
Outline filament organization and structure
Stress fibres= antiparallel contractile structures near nucleus of cells (with focal adhesions) Lamellipodium= branches and cross-linked filaments around outside of cell Filopodium= bundle of parallel filaments
260
How are F actin filaments remodelled?
``` Bundling Motor proteins Side-binding Cappnig Cross linking Severing -> G actin ```
261
How are G actin filaments remodelled?
Nucleating Sequestering -> F actin
262
Outline nucleation in actin filament remodelling
Limiting step in actin dynamics Formation of trimers to initiate polymerization Adding of actin, Arp2 and Arp3 at plus end Attaches to actin monomers-> nucleated actin filament
263
Outline elongation in actin filament remodelling
Reverse of nucleation Profilin (monomer binding) competes with thymosin for binding to actin monomers and promotes assembly Thymosin inhibits free actin monomers forming actin filaments
264
What is sequestering?
Biological processes in which an organism accumulates a compound or tissue
265
Outline capping in actin filament remodelling
Proteins added to end of actin chains + end= CapZ, Gelsolin, Fragmin/severin - end= tropomodulin, Arp complex
266
Outline severing in actin filament remodelling
In unsevered population, actin filaments grow and shrink relatively slowly In severed population, actin filaments grow and shrink more rapidly Severing proteins= gelsolin, ADF/cofilin, fragmin/severin
267
How does B4-thymosin lead to sequestering of actin?
Inhibits polymerization (so stops free actin monomers forming actin filaments)
268
How do actin functions cooperate to generate filaments?
All processes work together ``` Prolifilin-actin barbed end elongation Growth pre-existing end Annealing Barbed-end capping Filament severing ``` Needs ATP and ADP and proteins
269
Outline cross-linking and bundling in actin filament remodelling
Way of forming effective interactions between cytoskeletal filaments ``` Proteins involved: alpha-actinin fimbrin filamin spectrin villin vinculin ```
270
How do actin functions cooperate to organize filaments?
Severing Barbed-end capping Bundling Buckling ATP, ADP and proteins involved
271
Outline branching in actin filament remodelling
Branching-> more connections Branching protein= Arp complex
272
Outline Gel-sol transition by actin filament severing
Gel-> rigid | Sol-> can flow
273
Which one of these diseases is not caused by deregulation of actin cytoskeleton? High blood pressure Wiskott-Aldrich Syndrome – WAS (immunodeficiency, eczma, autoimmunity) Epidermolysis Bullosa (hereditary blistering diseases) Bullous Pemphigoid (autoimmune disease) Alzheimer (neurodegenerative
Alzheimer (neurodegenerative)
274
Where do actin activities participate during cell movement? E.g. polymerization, gel/sol transition, attachment ECM, contraction and detachment, disassembly/ nucleation/ branching/ severing/ capping/ bundling
Disassembly/ nucleation/ branching/ severing/ capping/ bundling-> EXTENSION (actin activities) Polymerization-> EXTENSION Gel/sol transition-> ADHESION Attachment ECM-> ADHESION Contraction-> TRANSLOCATION Detachment-> DE-ADHESION
275
Outline lamellae protrusion
Polymerization, disassembly, branching, capping Net filament assembly at leading edge Net filament disassembly behind leading edge
276
What are filopodia involved in?
Actin polymerization Bundling Cross linking
277
What signalling mechanisms regulate the actin cytoskeleton?
Ion flux changes (i.e. intracellular calcium) Phosphoinositide signalling (phospholipid binding) Kinases/phosphatases (phosphorylation cytoskeletal proteins) Signalling cascades via small GTPases
278
How is the actin cytoskeleton controlled by small g protein?
Rho subfamily proteins are activated by RTK, adhesion Rs and signal transduction pathways Expression levels up-regulated in different human tumours
279
What are the Rho subfamily?
Rho (subfamily of small GTPases belongs to the Ras super-family) - Family members: Rac, Rho, Cdc42 best known Participate in a variety of cytoskeletal processes Including control of actin cytoskeleton
280
What do the Rho subfamily activate?
Cdc42-> filopodia Rac-> lamellipodia Rho-> stress fibres
281
How does signalling from small GTPases regulate actin cytoskeleton and motility?
Actin binding proteins regulated by Rac/Cdc42 SEE DIAGRAM Rac.GTPCdc42.GTP Leads to actin polymerization/organization
282
How do small GTPases participate on cell migration?
Rac-> EXTENSION (involved in actin polymerization, branching) Rac and Rho-> ADHESION (focal adhesion, assembly) Rho-> TRANSLOCATION (stress fibres, tension, contraction) Rho-> DE-ADHESION Cdc42= filopodia, polarized motility, actin polymerization
283
What do we need programmed cell death for?
Harmful cells (e.g. cells with viral infection, DNA damage) Developmentally defective cells (e.g. B lymphocytes expressing antibodies against self antigens) Excess/unnecessary cells (e.g. embryonic development) Obsolete cells (e.g. mammary epithelium at the end of lactation) Exploitation (chemotherapeutic killing of cells)
284
Define: necrosis
Unregulated cell death associated with trauma, cellular disruption and an inflammatory response
285
Define: apoptosis
Programmed cell death Regulated cell death, controlled disassembly of cellular contents without disruption (no inflammatory response)
286
What happens in necrosis?
Plasma membrane becomes permeable Cells and organelles swell, chromatin condenses and rupture of cellular membranes Release of proteases leading to autodigestion and dissolution of the cell Cell lysis with invasion of phagocytic cells and localised inflammation
287
What are the phases of apoptosis?
Latent phase | Execution phase
288
What happens in the latent phase of apoptosis?
Death pathways are activated, but cells appear morphologically the same
289
What happens in the execution phase of apoptosis?
Loss of microvilli and intercellular junctions (chromatin begins to condense) Cell shrinkage (epithelium closes around) Loss of plasma membrane asymmetry (phosphatidylserine lipid appears in outer leaflet) Chromatin and nuclear condensation DNA fragmentation Formation of membrane blebs (violent blebs) Fragmentation of these blebs into membrane-enclosed apoptotic bodies Apoptotic bodies phagocytosed by neighbouring cells and moving macrophages (PLASMA MEMBRANE REMAINS INTACT- NO INFLAMMATION)
290
What DNA modification happens in apoptosis?
DNA fragmentation leads to more “ends” | can be labelled by adding an extra fluorescently-tagged base-> yellow nuclei on cells that are dying in TUNEL assay
291
What can apoptotic cells look like?
Dying becomes rough Loss of microvilli, cell shrinkage and cell blebbing
292
What are the kinds of cell death?
Necrosis Apoptosis Apoptosis-like PCD Necrosis-like PCD
293
What is apoptosis-like PCD?
Some, but not all, features of apoptosis Display of phagocytic recognition molecules before plasma membrane lysis
294
What is necrosis-like PCD?
Variable features of apoptosis before cell lysis | 'Aborted apoptosis'
295
Why can cell death be described as a graded response?
Features of both apoptosis and necrosis
296
What are the mechanisms of apoptotic cell death?
The executioners- Caspases Initiating the death programme - Death receptors - Mitochondria The Bcl-2 family (controllers of this) Stopping the death programme
297
What are caspases and what do they do?
Cysteine-dependent aspartate-directed proteases Executioners of apoptosis Activated by proteolysis Cascade of activation
298
What kind of caspases are there?
Initiator (2, 9) Initiator with DED (death effector domain) (10, 8) Effector (3, 6, 7)
299
What are homotypic protein-protein interactions of caspases?
Card domains bind to card domains Provide protein interactions at particular site they are needed
300
How is a procaspase converted to an active enzyme?
Cleavage of the inactive procaspase precursor is followed by folding of 2 large and 2 small chains to form an active L2S2 heterotetramer Tetramer is the activated species that you should worry about
301
What do initiator caspases do?
Trigger apoptosis by cleaving and activating
302
What do effector caspases do?
Carry out the apoptotic programme Cleave and inactivate proteins or complexes (e.g. nuclear lamins leading to nuclear breakdown) Activate enzymes (incl. protein kinases; nucleases, e.g. Caspase-Activated DNase, CAD) by direct cleavage, or cleavage of inhibitory molecules
303
What is the caspase cascade?
Amplification Divergent responses Regulation Initiator caspases-> effector caspases SEE DIAGRAM
304
What are the mechanisms of caspase activation?
Death by design= Receptor-mediated (extrinsic) pathways - External to cell - Cell senses signal and dies Death by default= Mitochondrial (intrinsic) death pathway - Signal from inside the cell usually involving mitochondria
305
What are death receptors?
Secreted or transmembrane ligands (trimeric) ``` Have cysteine rich domains= EC Transmembrane domain Death domain (DD)= IC ```
306
What are FADD and FLIP?
FADD= activates programme of cell death (DED and DD) FLIP= inhibits (DED and DED)
307
What is DD (in FADD)?
Death effector domain
308
What is DED (in FLIP and FADD)?
Death domain
309
How does signalling through death receptors e.g. Fas/Fas-ligand occur?
1. Receptor (Fas) trimerisation by ligand (Fas-L on lymphocyte) 2. Recruitment of adapter protein (FADD) through its DD to DD of Fas 3. Recruitment and oligomerisation of procaspase 8 through its DED to FADD DED –> Death-Inducing Signalling Complex (DISC)
310
How does initiator procaspase 8 oligomerisation result in cleavage and activation?
Trimerised R tails/FADD and initiator procaspase 2 - > cleavage of initiator procaspase 2 - > active initiator caspase 8 tetramer released from receptor Some initiator procaspases have intrinsic low catalytic activity (oligomerisation allows transcleavage) Others are activated by conformational change on oligomerisation Need at least 2 procaspases to form an active tetramer
311
Outline death receptor activation of caspase 8
Binds to caspase 9 and will block its ability to cleave itself Inhibited by FLIP FLIP - caspase homology in DED domain, but no proteolytic activity therefore competes with procaspase
312
How does FLIP inhibit procaspase 8 activation?
Trimerised receptor tials/FADD and initiator procaspase 8-> no cleavage Competes for binding to receptor tails/FADD via DED domains Incorporates into R-procaspase complexes and interferes with transcleavage
313
How does caspase 8 activate downstream effector caspases?
Activates caspase 3 and caspase 7 Caspase 3 activates caspase 6, 2 and 1 Caspase 6 activates caspase 10 (which activates caspase 8 and the loop)
314
Outline the mitochondrial regulation of apoptosis
Cellular stresses e.g. lack of or overstimulation by GFs, DNA damage (p53) ``` Loss of mitochondrial membrane potential (ΔΨ) ``` Release of cytochrome c and other apoptosis- inducing factors ``` Formation of the apoptosome complex (Apaf1, caspase 9) ```
315
What is the apoptosome?
'Wheel of death' Apaf-1 heptamer (Apaf-1 scaffold) ``` CONTAINS: Apaf-1 (can potentially bind to procaspase 9) Cytochrome c (bound to WD4repeats) ATP Procaspase 9 (binds to CARD domain) ```
316
How is the caspase cascade activated by the apoptosome/
Oligomerisation brings multiple procaspase 9s close together ->cleavage, activation and release as active caspase 9 tetramer Active caspase 9 tetramer initiates a caspase cascade leading to apoptosis
317
What are the principal mechanisms of apoptosis?
Bid links receptor | Mitochondrial death pathways
318
How do bid links receptor and mitochondrial death pathways contribute to apoptosis?
Caspase 8 cleaves Bid which enhances release of mitochondrial proteins, thus engaging the intrinsic pathway
319
Where do apoptosome's get energy from?
The apoptosome requires ATP Energy levels in the cell may determine whether death is by necrosis or apoptosis (+ATP= apoptosis, -ATP= necrosis)
320
What are the main modulators of apoptosis>
Bcl-2 family proteins Group I, II and III Group I= BH4, BH3. BH1. BH2, TM (Bcl-2) Group II= BH3, BH1, BH2, TM (Bax) Group III (BH3 only)= BH3 (maybe TM) (Bid, Bak, Bad)
321
What Bcl-2 family proteins are anti-apoptotic?
Bcl-2 Bcl-xL (Mitochondrial)
322
What Bcl-2 family proteins are pro-apoptotic?
Bid Bad Bax Bak (Move between cytosol and mitochondria)
323
Outline the PI3K signalling pathway in cell cycle and apoptosis regulation
GF e.g. ECF, insulin binds to GFR e.g. EGFR, insulin R - > PI3K (inhibited by PTEN) - > PDK-1 - > PKB/Akt - > cell survival, proliferation, protein synthesis
324
What is PI3K?
Phosphatidylinositol 3’-kinase (PI3’-K) A lipid kinase (not a protein kinase) involved in growth control and cell survival Activates PKB/Akt which is anti-apoptotic
325
What does PKB/Akt do?
Phosphorylates and inactivates Bad and caspase 9 Inactivates FOXO transcription factors (FOXOs promote expression of apoptosis-promoting genes) Other, e.g. stimulates ribosome production and protein synthesis
326
How do Bcl-2 family proteins regulate apoptosis via BH3 heterodimerisation (GF absent)?
GF absent Bad dephosphorylated and released Displaces Bcl-2/xL from -> Bax/Bak Pore-> apoptosis
327
How do Bcl-2 family proteins regulate apoptosis via BH3 heterodimerisation (GF present)?
GF present PI3K and PKB/Akt Heterodimers are inactive Phospho-Bad inactive Cell survival
328
What inhibits the converstion of Phosphatidylinositol 4,5-bisphosphate (PIP2) to Phosphatidylinositol 3,4,5-trisphosphate (PIP3)?
PTEN | Dephosphorylates
329
What do IAPs do in apoptosis?
Inhibitor of Apoptosis Proteins (IAPs) regulate Programed Cell Death Bind to procaspases and prevent activation Bind to active caspases and inhibit their activity
330
What are the cytoprotective/anti-apoptotic pathways?
Bcl-2, Bcl-xL: intrinsic pathway FLIP, IAPs: extrinsic pathway Growth factor pathways via PI3’-K and PKB/Akt
331
What proto-oncogenes/tumour suppressors are associated with apoptosis?
Bcl-2 PKB/Akt PTEN
332
What are the therapeutic uses of programmed cell death?
Harmful (oncogenic) cells (e.g. cells with viral infection, DNA damage) Chemotherapeutic killing of tumour cells, e.g. Dexamethasone stimulates DNA cleavage
333
Why is the cell cycle important in life, death and cancer?
Cycle checkpoints (growth arrest ensures genetic fidelity) 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
334
What do proto-oncogenes code for?
Essential proteins involved in maintenance of cell growth, division and differentiation
335
How is a proto-oncogene converted to an oncogene?
Mutation Oncogene protein product no longer responds to control influences
336
How can oncogenes be expressed?
Aberrantly expressed Over-expressed Aberrantly active
337
How is an oncogene activated?
Mutation in the coding sequence (point mutation of deletion) Gene amplification (multiple gene copies overproduced) Strong enhancer increases normal protein levels (e.g. Brukitt's lymphoma) Chromosomal translocation (chimeric genes) OR Fusion to actively transcribed gene overproduces protein or fusion protein is hyperactive (e.g. Philadelphia chromosome) Insertional mutagenesis (e.g. viral infection)
338
What happens to normal activity when proto-oncogenes are activated to oncogenes?
Normal activity disrupted Affects TK signalling, nuclear/cytosolic signalling and GPCR signalling Affects phosphorylation, proliferation, transcription and translation
339
What happens to mutant Ras?
Upon binding GTP, RAS becomes active Dephosphorylation of the GTP to GDP switches RAS off Mutant RAS fails to dephosphorylate GTP and remains active
340
``` SRC gene (oncogenes): Function Mechanism of activation Location Associated human cancers ```
Function= tyrosine kinase Mechanism of activation= overexpression/C-terminal deletion Location= cytoplasmic Associated human cancers= breast, colon, lung
341
``` MYC gene (oncogenes): Function Mechanism of activation Location Associated human cancers ```
Function= transcription factor Mechanism of activation= translocation Location= nuclear Associated human cancers= Burkitt's lymphoma
342
``` JUN gene (oncogenes): Function Mechanism of activation Location Associated human cancers ```
Function= transcription Mechanism of activation= overexpression/deletion Location= nuclear Associated human cancers= lung
343
``` Ha-RAS gene (oncogenes): Function Mechanism of activation Location Associated human cancers ```
Function= G protein Mechanism of activation= point mutation Location= cytoplasmic Associated human cancers= bladder
344
``` Ki-RAS gene (oncogenes): Function Mechanism of activation Location Associated human cancers ```
Function= G protein Mechanism of activation= point mutation Location= cytoplasmic Associated human cancers= colon, lung
345
Which one of the following statements is incorrect? a. Mutation can convert a protooncogene to an oncogene b. Gene amplification of a protooncogene can be oncogenic c. Chromosome translocation can lead to inappropriate expression of a protooncogene and to an oncogenic effect. d. A protooncogene can be activated to an oncogene by insertional mutagenesis e. Protooncogenes are not expressed in normal cells.
e. Protooncogenes are not expressed in normal cells. | What
346
What are tumour suppressor genes? How can they cause cancer?
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 lost of both copies means loss of control
347
What are the features of 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
348
What is a retinoblastoma?
Malignant cancer of developing retinal cells Sporadic disease usually involves one eye Hereditary cases can be unilateral or bilateral and multifocal
349
What causes retinoblastoma?
Sporadic or hereditary Hereditary: 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
350
What do tumour suppressor genes do?
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
351
p53 gene (TSG): Function Location Associated human cancer
Function= cell cycle regulator Location= nuclear Associated human cancer= many (colon, breast, bladder, lung etc)
352
BRCA1 gene (TSG): Function Location Associated human cancer
Function= cell cycle regulator Location= nuclear Associated human cancer= breast, ovarian, prostate
353
PTEN gene (TSG): Function Location Associated human cancer
Function= tyrosine and lipid phosphatase Location= cytoplasmic Associated human cancer= prostate, glioblastoma
354
APC gene (TSG): Function Location Associated human cancer
Function= cell signalling Location= cytoplasmic Associated human cancer= colon
355
p16-INK4A gene (TSG): Function Location Associated human cancer
Function= cell cycle regulator Location= nuclear Associated human cancer= colon and others
356
MLH1 gene (TSG): Function Location Associated human cancer
Function= mismatch repair Location= nuclear Associated human cancer= colon, gastric
357
Why is p53 important in cancer?
p53 is a 'guardian of the genome'= key player in decision making during the cell cycle Although p53 is a tumour supressor gene, mutants of p53 act in a dominant manner and mutation of a single copy is sufficient to get dysregulation of activity
358
How do mutations of the APC tumour suppressor gene lead to colorectal cancer?
FAMILIAL ADENOMATOUS POLYPOSIS COLI Due to a deletion in 5q21 resulting in loss of APC gene (tumour suppressor gene) Involved in cell adhesion and signalling Sufferers develop multiple benign adenomatous polyps of the colon There is a 90% risk of developing colorectal carcinoma
359
What pathway does the tumour suppressor gene APC participate in?
WNT signalling pathway
360
What does the APC protein do in the WNT signalling pathway?
APC protein helps control the activity of B-catenin and thereby preventing uncontrolled growth Mutation of APC is a frequent event in colon cancer
361
The protein products of tumour suppressor genes are not NOT involved with: a. Regulation of cellular proliferation b. Metabolism of drugs c. Regulation of cell cycle d. Repair of DNA damage e. Control of transcription
b. Metabolism of drugs
362
How do oncogenes, TSGs and proto-oncogenes cause cancer?
Oncogene Tumour suppressor gene Proto-oncogene Defective tumour suppressor gene -> Cell growth and proliferation-> cancer
363
How does normal epithelium develop into metastasis in colorectal cancer?
Normal epithelium (APC->) Hyperproliferative epithelium (K-ras and DNA hypomethylation->) Adenoma (p53->) Carcinoma Metastasis
364
What are the features of an oncogene?
``` Gene active in tumour Specific translocations/point mutations Mutations rarely hereditary Dominant at cell level Broad tissue specificity Leukaemia and lymphoma ```
365
What are the features of tumour suppressor genes?
``` Gene inactive in tumour Deletions or mutations Mutations can be inherited Recessive at cell level Considerable tumour specificity Solid tumours ```
366
Human cancer involves damage to DNA, or inheritance of aberrant sequences at.... These...
Critical gene targets These targets, proto-oncogenes and tumour suppressor genes, regulate cell cycle decisions (mitosis, arrest, differentiation, apoptosis)
367
When is angiogenesis useful physiologically?
Wound healing Menstrual cycle Embryonic development
368
What are the signs of insufficient angiogenesis?
Baldness MI- ischaemia Limb fractures Thrombosis
369
What are the signs of vascular malformations?
Angiodysplasia (HHT & VWD) Cerebral malformations (AVM/CCM)
370
What are the signs of excessive angiogenesis?
Retinal disease Cancers Atherosclerosis Obesity
371
How are blood vessels made?
VASCULOGENESIS Bone marrow progenitor cell (recruitment or mobilization, capillary plexus, mature network) ANIOGENESIS Sprouting (SMC recruitment) ``` ARTERIOGENESIS Collateral growth (shear stress, cytokines-> matrix remodelling/SMC growth) ```
372
Outline the model of sprouting angiogenesis (detailed)
Selection of sprouting ECs - Modulation of EC-EC contacts and PC contacts - Promoted by ECM degradation and GFs - Inhibited by lateral inhibition, growth inhibitors and pro-quiescent signals (local and systemic) Sprout outgrowth and guidance) - EC proliferation - Maintenance of junctions - Deposition of new ECM - Promoted by GFs, ECM and invasive behaviour - Inhibited by growth inhibition Sprout fusion and lumen formation - Stalk-cell proliferation - Vacuole formation and fusion - Promoted by tip cells encountering adhesion - Inhibited by tip cells encountering repulsion Perfusion and maturation - Stabilisation of EC-EC adhesion - Stabilization of PC contacts - Decreased EC proliferation - Inhibited by increased pro-quiescent signals
373
What are the main stages of sprouting angiogenesis?
Initiation Selection of sprouting ECs Tip-cell navigation Sprout outgrowth and guidance (stalk elongation) Sprout fusion and lumen formation Perfusion and oxygenation) Maturation and stabilization Quiescence
374
What effect does hypoxia have on angiogenesis?
Hypoxia-> activates GFs-> stimulates sprouting
375
What are the inhibitors of angiogenesis?
Thrombospondin-1 | Statins= angiostatin, endostatin, canstatin turnstatin
376
What are the activators of angiogenesis?
``` VEGFs FGFs PDGFB EGF LPA ```
377
Outline how hypoxia triggers angiogenesis
ABSENCE OF OXYGEN HIF-a doesn't bind to pVHL (there isn't any hydroxyproline) HIF-a binds to HIF-B Activates hypoxia-inducible genes (VEGF, PDGF-B, TGF-a and EPO)
378
What happens to HIF in angiogenesis in the presence of oxygen?
PRESENCE OF OXYGEN HIF-alpha has ubiquitin attachement, hydroproline head and is bound to pVHL Loses pVHL and hydroxyproline HIF-a with ubiquitin binds to proteasome HIF-a destroyed
379
List hypoxia-inducible genes
VEGF PDGF-B TGF-a EPO
380
What is VEGF? What are the members of this family?
Vascular endothelial growth factor ``` VEGF-A VEGF-B VEGF-C VEGF-D PIGF (placental growth factor) ```
381
What does VEGFR-2 do?
Major mediator of VEGF-dependent angiogenesis | Activates signalling pathways that regulate endothelial cell migration, survival and proliferation
382
What are the VEGF receptors?
Tyrosine kinase receptors VEGFR-1 VEGFR-2 VEGFR-3 ``` Co-receptors Neuropilin 1 (Nrp1) Neuropilin 2 (Nrp2) ```
383
What is tip cell selection based on?
Notch signalling between adjacent endothelial cells at the angiogenic front Determined by VEGFR02, NRP1, integrins, HIF-1a, PGC01a, MT1-MMP, JAGGED1, DLL4 Specialised endothelial tip cells lead the outgrowth of blood-vessel sprouts towards gradients of VEGF
384
What leads the outgrowth of blood-vessel sprouts?
Specialised endothelial tip cells lead the outgrowth of blood-vessel sprouts towards gradients of VEGF
385
What is canonical notch signalling?
Notch Rs and ligands are membrane-bound proteins that associate through their EC domains The IC domain of Notch (NICD) translocates to the nucleus and binds to the TF RBP-J Important for tip cells to bind to stalk cells
386
Outline VEGF/Notch signalling?
TIP CELL SELECTION In stable blood vessels, DII4 and Notch signalling maintain quiescence VEGF activation increases expression of DII4 INDUCTION OF SPROUTING DII4 drives Notch signalling which inhibits expression of VEGFR2 in the adjacent cell DII4-expressing tip cells acquire a motile, invasive and sprouting phenotype Adjacent cells (stalk cells) form the base of the emerging sprout, proliferate to support sprout elongation
387
What happens in stalk elongation and tip guidance? What do the stages involve?
Lumen formation (VE-caherin, CD34, sialomucins, VEGF) Pericyte recruitment (PDGF-B, ANG-1, NOTCH, ephrin-B2, FGF) Tip cell guidance and adhesion (semaphorins, ephrins, integrins) Liberation of angiogenic factors from ECM (VEGF, FGFs) Stalk elongation (VEGFR-1, NOTCH, WNT, NRARP, PIGF, FGFs, EGFL7) Myeloid cell recruitment (ANG-2, SDF-1a, PIGF)
388
What is the role of macrophages in vessel anastomosis?
Physiological and pathological angiogenesis Macrophages have been shown to carve out tunnels in the ECM-> provides avenues for subsequent capillary infiltration Tissue-resident macrophages associated with angiogenic tip cells during anastomosis
389
What happens in stabilisation and quiescence (angiogenesis)? What is involved in this?
Quiescent phalanx resolution: Transendothelial lipid transport (VEGF-B) Vascular maintenance (VEGF, ANG-1, FGFs, NOTCH) Phalanx cells (PHD2, HIF2a, VE-cadherin, TIE-2) Barrier formation (VE-cadherin, ANG-1) Basement membrane deposition (TIMPs, PAI-1) Pericyte maturation (PDGF-B, PDGFR-B, ephrin-B2, ANG-1, NOTCH, TGF-B1)
390
Where is VE-cadherin often found in endothelial cells?
In adherens junctions Constitutively expressed Homophilic interaction mediates adhesion b/w endothelial cells and IC signalling Controls contact inhibitIon of cell growth and promotes survival of EC
391
What is found in tight junctions of endothelial cells?
Caludins, occludin, JAMs, ESAM, nectin
392
What is VE-cadherin essential for?
Vessel stabilisation and quiescence Controls contact inhibitIon of cell growth and promotes survival of EC
393
What kind of cells help stabilise the neovessels in angiogenesis?
Mural cells (progenitor cells of pericytes) Work with pericytes to stabilise vessels
394
How do mural cells and pericytes help to stabilise neovessels in angiogenesis?
Stalk cells attached to pericytes which are recruited Pericytes-> TIMP3 and ANG1 act on stalk cells (Angiopoietin/Tie-2 system) DLL4 ligand binds to notch R
395
Outline the angiopoietin-Tie2 ligand-receptor system
Ang-1 and Ang-2 are antagonistic ligands of the Tie2 receptor Ang-1 binding to Tie2: Promotes- vessel stability Inhibits- inflammatory gene expression Ang-2 antagonises Ang-1 signalling Promotes- vascular instability and VEGF-dependent angiogenesis
396
What does Ang-1 binding promote and inhibit in the Ang/Tie2 system?
Ang-1 binding to Tie2 Promotes- vessel stability Inhibits- inflammatory gene expression
397
What does Ang-2 binding promote in the Ang/Tie2 system?
Ang-2 antagonises Ang-1 signalling | Promotes- vascular instability and VEGF-dependent angiogenesis
398
What diseases are likely to have increased Ang-2 plasma levels?
Congestive heart failure Sepsis Chronic kidney disease
399
Outline tumour angiogenesis and neovasculature
Tumours <1mm receive oxygen and nutrients by diffusion Larger tumours need vessel network (so tumour secretes angiogenic factors that stimulate migration, proliferation and neovessel formation by endothelial cells in adjacent established vessels) Once a tumour is newly vascularized, doesn't rely on diffusion solely so can grow progressively
400
What is the angiogenic switch?
Discrete step in tumour development that can occur at different stages in the tumour-progression pathway Depends on the nature of the tumour and its microenvironment
401
Describe tumour blood vessels
Irregularly shaped, dilated, tortuous Not organised into definitive venules, arterioles and capillaries Leaky and haemorrhagic, partly due to overproduction Perivascular cells often become loosely associated (Some tumours may recruit endothelial progenitor cells from the BM)
402
What agents target the VEGF pathway?
Anti-VEGF antibodies Soluble VEGF Rs (e.g. VEGF-Trap) Anti-VEGFR antibodies (e.g. IMC-1121b) Small molecule VEGFR inhibitors (e.g. vatalanib, sunitinib, ZD6474, AZO2171)
403
What happens to tumor growth with VEGF inhibition by soluble VEGFR1 (Flt-1)?
Reduces tumor growth VEGFR1 binds to VEGF and 'mops it up' preventing it from stimulating angiogenesis Flt-1 expression reduces tumor growth in vivo, without affecting tumor cell growth in vitro SO IT MUST BE ON VASCULATURE
404
What are the side effects of avastin?
Anti-VEGF humanised MAb ``` GI perforation Hypertension Proteinuria Venous thrombosis Haemorrhage Wound healing complications ``` NB. No overall survival advantage over chemo alone No QoL or survival advantage
405
Does anti-angiogenic therapy in cancer work?
In some cases= benefits are transitory Followed by a restoration of tumour growth and progression In other cases there's no objective benefit
406
What happens in anti-angiogenic therapy which normalises vasculature?
Reduces hypoxia Increase efficacy of conventional therapies
407
What happens in 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
408
What are the potential mechanisms of resistance to anti-VEGF therapy in cancer?
VEGF INHIBITION AGGRAVATES HYPOXIA -> increased tumour production of other angiogenic factors or increases tumour invasiveness TUMOUR VESSELS MAY BE LESS SENSITIVE TO VEGF INHIBITION Due to vessel lining by tumour cells or endothelial cells derived from tumours TUMOUR CELLS THAT RECRUIT PERICYTES May be less responsive to VEGF therapy
409
What is vasculogenic mimicry?
Tumour cell vasculogenic mimicry (VM) Plasticity of aggressive cancer cells forming de novo vascular networks (i.e. tumour cells organise themselves to form vessel-like channels which hook up to channels within the tumour mass) Associated with malignant phenotype and poor clinical outcome
410
How can anti-angiogenic cancer therapy be developed in the future?
In combo with other anti-cancer therapies Need to consider resistance (combinatorial strategies to inhibit angiogenesis and target drug resistance mechanisms) Novel non-VEGF targets
411
What is age-related macula degeneration?
AMD Abnormal growth of choroidal blood vessels: Leaky vessels cause oedema Visual impairment
412
What is the main anti-VEGF therapy for AMD?
Lucentis (ranibizumab) High efficacy of treating and improving vision Very expensive but more effective than Avastatin (bevacizumab)
413
What are the challenges for therapeutic strategies to inhibit angiogenesis in cancer?
Tumours are complex 3D structures with microenvironemnts Lack good in vitro models Studies generally performed on cell lines growing as 2D monolayers which don't mimic interplay b/w tumour cells and their EC environment Tumours receive nutrients and therapeutics through vasculature (can't study in vitro)
414
Outline the 'tumour-on-a-chip" platform
Microphysiological system that incorporates human cells in a 3D ECM Supported by perfused human microvessels Useful in drug screening (considers vasculature)
415
What can damage DNA?
``` CHEMICALS (carcinogens) Dietary Lifestyle Environmental Occupational Medical Endogenous ``` RADIATION Ionizing Solar Cosmic Damaged DNA can -> cancer
416
What kind of damage can be done to DNA by carcinogens?
DNA adducts and alkylation Base dimers and chemical cross-links Base hydroxylations and abasic sites formed Double and single strand breaks
417
What happens in phase 1 of mammalian metabolism?
Addition of functional groups e.g. oxidations, reductions, hydrolysis Mainly cytochrome p450-mediated
418
What happens in phase 2 of mammalian metabolism?
Conjugation of phase 1 functional groups E.g. sulphation, glucuronidation, acetylation, methylation, amino acid and glutathione conjugation Generates polar (water soluble) metabolites
419
What causes polycyclic aromatic hydrocarbons?
Common environmental pollutants Formed from combustion of fossil fuels Formed from combustion of tobacco
420
Outline the two step epoxidation of B[a]P
Benzo[a]pyrene - > (p450) - > TOXIC benzo[a]pyrene-7,8-oxide - > (EH) - > less toxic benzo]a]pyrene-7,8-dihydrodiol - > (p450) - > Benzo[a]pyrene-7,8 dihydrodiol-9,10- oxide Spontaneously breaks off Charge around C Incredibly reactive (with DNA and RNA) Electron rich molecule in body -> DNA adducts
421
What is aflatoxin B1?
Formed by Aspergillus flavus mould Common on poorly stored grains and peanuts Aflatoxin B1 is a potent human liver carcinogen, especially in Africa and Far-East - America have money to test for it and avoid it - Poorer countries can’t so eat it
422
Outline the epoxidation of aflatoxin B1
C=C substrate for p450 on aflatoxin B1 - > generate epoxide (p450) Aflatoxin B1. 2,3-epoxide - Adducts directly to guanine - Can lead to mutation in genetic sequence
423
What happens in the metabolism of 2-naphthylamine?
Aromatic 2-naphthylamine has amine group -> (CYP1A2) -> N-hydroxy-2-naphthylamine -> (glycuronyl transferase) -> Adds glucuronide -> (urine pH means nitrenium ion added) -> DNA-reactive electrophile causes bladder tumours
424
Why does metabolism of 2-naphthylamine cause bladder not liver cancer?
Metabolised by cytochrome p450 but bladder cancer (not liver) because of the way it is metabolically activated CYP1A2 forms hydroxyl amine which is very toxic -> acidic pH of urine leads to breakage of glucoronide-> release of nitrenium positive ion which reacts with DNA
425
Where is 2-naphthylamine found?
Past components of dye-stuffs Include 2-naphthylamine and benzidine German dye industry epidemiology
426
How does solar (UV) radiation lead to cancer?
Pyrimidine (thymine) dimers Skin cancer
427
How does ionising radiation lead to cancer?
Generates free radicals in cells Includes oxygen free radicals - Super oxide radical: O2* - Hydroxyl radical: HO* Possess unpaired electrons - Electrophilic and therefore seek out electron-rich DNA
428
How do oxygen free radicals attack DNA?
Double and single strand breaks Apurinic and apyrimidinic sites Base modifications: - Ring-opened guanine and adenine - Thymine and cytosine glycols - 8-hydroxyadenine and 8-hydroxyguanine (mutagenic)
429
What enzyme system is most frequently involved in the activation of chemicals to metabolites that can damage DNA? a. Glucuronyl transferase b. Haem oxygenase c. Cytochrome P450 d. Xanthine oxidase e. Glutathione transferase
c. Cytochrome P450
430
What is the role of p53 in dealing with cellular stress?
PRODUCTION OF P53 (stims MDM2, inhibited by MDM2) - Ribonucleotide depletion - Nitric oxide - Hypoxia - Oxidative stress - Mitotic apparatus dysfunction - Oncogene activation - DNA replication stress - Double-strand breaks - Telomere erosion ``` MILD/PHYSIOLOGICAL STRESS Regulates p53 target genes -> metabolic homeostasis -> antioxidant defence -> DNA repair -> growth arrest -> senescene -> apoptosis ``` SEVERE STRESS Protein-protein interactions -> apoptosis
431
What are the types of DNA repair?
Direct reversal of DNA damage Base excision repair (mainly for apurinic/apyrimidinic damage) Nucleotide excision repair (mainly for bulky DNA adducts) During- or post-replication repair
432
What happens in DNA repair: direct reversal of DNA damage?
Photolyase splits cyclobutane pyrimidine-dimers Methyltransferases and alkyltransferases remove alkyl groups from bases
433
What happens in DNA repair: base excision repair?
DNA glycosylases and apurinic/apyrimidinic endonucleases and other enzyme partners A repair polymerase (e.g. Polβ) fills the gap and DNA ligase completes the repair - Mutagen exposure - DNA-glycosylase - AP-endonuclease - DNA polymerase (replaces nucleotide base based on complementary base pairing) - DNA ligase
434
What happens in DNA repair: nucleotide excision repair?
Xeroderma pigmentosum proteins (XP proteins) assemble at the damage A stretch of nucleotides either side of the damage are excised Repair polymerases (e.g. Polδ/β) fill the gap and DNA ligase completes the repair - Mutagen exposure - Endonuclease - Helicase - DNA polymerase (replaces 100s of bases based on complementary base pairing) - DNA ligase
435
What happens in DNA repair: during- or post-replication repair?
Mismatch repair | Recombinational repair
436
What kind of DNA damage is most likely to cause a mutagenic event?
Endogenous damage: - Depurination - Depyrimidination - Single-strand breaks - Alkylation - Free radical base oxidations The greater the persistence of damage then the greater the chance of a mutagenic event Repair rate per cell vs damage per hour per cell
437
What is the
Carcinogen damage leading to altered DNA -> A, B or C A. Efficient repair-> normal cell B. Apoptosis -> cell death C. 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
438
How can you test for DNA damage?
Structural alerts/SAR e.g. amino group, aromatic group that could form an epoxide - > In vitro BACTERIAL gene mutation assay e. g. Ames test with S. typhimurium - > In vitro MAMMALIAN CELL assay e. g. chromosome aberration, TK mutation in mouse lymphoma cell, Micronucleus assay - > In vivo MAMMALIAN assay e. g. Bone marrow micronucleus test transgenic rodent mutation assay -> Investigative in vivo MAMMALIAN assays
439
What happens in the bacterial (Ames) test for muttagenicity of chemicals?
Chemical to be tested Has rat liver enzyme preparation (S9) added - Bacteria that don't synthesis histidine (e.g. salmonella strain) Conversion of chemical to reactive metabolite On histidine-free media: if mutations occur in bacterial genome then bacteria acquire ability to synthesise histidine = colonies
440
How do detect DNA damage (chromosomal abberrations) in mammalian cells?
Treat mammalian cells with chemical in presence of liver S9 Look for chromosomal damage e.g. chromosome interchanges, acentric ring etc.
441
How does an in vitro micronucleus assay work?
Cells treated with chemical and allowed to divide Cytokinesis blocked using cytochalasin-B 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)
442
Which of the following is involved in the repair of damaged DNA? ``` Mutation Epoxidation DNA adduction Base excision repair Sister chromatid exchange ```
Base excision repair
443
What happens if damaged DNA is incorrectly repaired?
Can lead to mutation and possibly neoplasia
444
True or false: Colorectal cancer is the 5th most common cancer in developed countries?
False 4th most common in cancer overall 2nd leading cause of cancer death
445
What does the colon do?
Extracts water from faeces (electrolyte balance) Faecal reservoir (evolutionary advantage) Bacterial digestion e.g. for vitamins B and K
446
Outline the microanatomy of the colon
Crypts of Lieberkuhn Differentiation UP Proliferation UP and DOWN Contains columnar, goblet, endocrine and mesenchymal cells
447
What is a polyp?
Projection from a mucosal surface into a hollow viscus, and may be hyperplastic, neoplastic, inflammatory, hamartomatous, etc.
448
What is an adenoma?
Benign neoplasm of mucosal epithelial cells
449
What does the APC mutation cause?
Prevents cell loss-> mutation Normally have protective mechanisms to eliminate genetically defective cells
450
What effect does proliferation have on cels?
Makes them vulnerable
451
What are the types of polyps?
``` Hyperplastic / metaplastic Adenomas Juvenile Peutz Jeghers Lipomas Others (essentially any circumscribed intramucosal lesions) ```
452
What is a hyperplastic polyp?
``` Very common <0.5cm 90% of LI polyps Often multiple Benign (no malignant potential) Commonly from K-ras mutation (15%_ ```
453
What are the types of colonic adenoma?
Type of cell: can be tubular or villous (or tubulovillous if mixed) Protrusion: can be pedunculated (tree) or sessile (rug) Sessile ones tend to be more villous, but both can be either
454
Describe a tubular adenoma
Columnar cells with nuclear enlargement, elongation, multilayering and loss of polarity Increased proliferative activity Reduced differentiation Complexity/disorganisation of architecture
455
Describe a villous adenoma
Mucinous 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
456
What is dysplasia?
Bad growth (literal meaning) Abnormal growth of cells with some features of cancer C.f. atypia Subjective analysis Indefinite, low grade and high grade (indefinite not used now)
457
How does UC lead to higher risk of colorectal cancer?
Ulcerative colitis causes increased proliferation in an attempt to repair damage, and the inflammation also damages the basement membrane, making invasion easier
458
Why is removing polyps a good idea to reduce cancer risk?
Most colorectal cancer comes for polyps
459
What is the adenoma-carcinoma sequence?
NORMAL Germline or somatic mutations of cancer suppressor genes (!st hit) MUCOSA AT RISK Methylation abnormalities Inactivation of normal alleles (2nd hit) ADENOMAS Protoncogene mutation Homozygous loss of additional cancer suppressor genes (Grow into lumen, submucosa extends) CARCINOMA Additional mutation Gross chromosomal alterations (Invasion through muscularis propria)
460
What is adenomatous polyposis coli (APC/FAP)?
5q21 gene mutation FAP (familial adenomatous polyposis)= inactivation of APC tumour suppressor genes
461
What percentage of adults have colonic adenomas at age 50? What percentage of these become cancers if left?
25% at 50y have colonic adenomas 5% become cancers if left
462
How long do adenomas usually take to develop into cancers?
10-15 years | Stay at curable stage 2 years approx
463
What size polyps have higher risks to develop into cancer?
Large (>1cm)
464
What do most colorectal carcinomas arise from?
Adenomas NB. Residual adenomas may remain (in 10-30% of CRCs)
465
What genetic pathway is involved in adenoma carcinoma sequence?
``` APC Kras Smads p53 Telomerase activation ```
466
What genetic pathway is involved in microsatellite instability?
HNPCC (hereditary non-polyposis colorectal cancer): due to microsattelite instability. Microsatellites are repeat sequences prone to misalignment, often mismatch repair genes
467
What are the main pathways for a genetic predisposition to colorectal cancer?
FAP | HNPCC
468
Where is colonic carcinoma particularly rare/common?
``` Rare= Japan, Mexico and Africa Common= US, Eastern Europe, Australia ``` (Generally 50-80y olds)
469
What dietary factors increase the risk of colonic carcinoma?
``` High fat Low fibre High red meat Refined carbohydrates Chemicals in food ```
470
Why do chemicals in food increase risk of colorectal cancer?
Food through GI system Contains carcinogens (also some anticancer agents) Head modifies chemicals further HCAs= heterocyclic amines (cooked e.g. in meat -> mutagenesis)
471
What anticancer food elements are useful in colorectal carcinoma?
Vit C= ROS scavenger Vit E= ROS scavenger Isothiocyanates )cruciferous veg) Polyphenoles (green tea, fruit juice)-> activate MAPK-> regulates Phase 2 detox enzymes and other genes that reduce DNA oxidation Also garlic associated apoptosis (Ajoene, allicin) Green tea (EGCG-induced telomerase activity)
472
What dietary deficiency is important in colorectal cancer?
Folates Co-enzyme for nucleotide synthesis and DNA methylation MTHFR Deficiency-> DNA synth disruption-> DNA instability-> genomic hypomethylation and focal hypermethylation-> gene activation and silencing
473
How does colorectal carcinoma present clinically?
``` The big ones: Change in bowel habit (more/less constipated, going more frequently, anything) Bleeding PR (peri rectum) Iron deficient anaemia (otherwise unexplained) ``` ``` Also potentially: Mucus PR Bloating Cramps (‘colic’) Constitutional changes (weight loss, fatigue, etc.) ```
474
Where do most colorectal cancers occur?
Most colorectal cancers occur in the sigmoid colon or rectum, the rest distributed fairly evenly 22% caecum/ascending colon 11% transverse 6% descending colon 55% rectosigmoid
475
How is colorectal carcinoma differentiated?
70% of colorectal cancers are moderately differentiated (10% well and 20% poorly)
476
How is colorectal cancer graded?
TNM classification is used in colorectal cancer management today Duke’s classification was the first staging system which proved that staging a cancer improved its management
477
Outline Duke's classification
Duke’s A - Growth limited to wall (muscularis propria) - Nodes negative Duke’s B - Growth beyond muscularis propria - Nodes negative Duke’s C1 - Nodes positive - Apical lymph node negative Duke’s C2 - Apical lymph node positive (There is a D)
478
How do clinical features of colorectal carcinomas have an effect on prognosis?
Diagnosis in asymptomatic patients- unknown (probably improved) Rectal bleeding-> improved prognosis Bowel obstruction-> diminished prognosis Tumour location-> colon better than rectum, left better than right Age <30-> diminished prognosis Preop serum CEA-> diminished prognosis with high CEA level Distant metastases-> markedly diminished prognosis
479
How do pathological features of colorectal carcinomas have an effect on prognosis?
Depth of bowel wall penetration-> increased penetration, diminish prognosis No. of regional lymph nodes-> 1-4 better than >4 Degree of differentiation-> well is better than poorly Mucinous (colloid) or signet ring cell-> diminished prognosis Venous invasion-> diminished prognosis Lymphatic invasion-> diminished prognosis Perineural invasion-> diminished prognosis Local inflamm and immuno reaction-> improved prognosis
480
How does staging affect treatment for colorectal cancer?
``` I= surgery II= surgery, 5FU III= surgery, 5FU/leucovorin IV= surgery, metastatectomy, chemo, palliative rT ```
481
What patient's are high risk for colorectal cancer and are therefore screened?
Patients who have.... Had a previous adenoma A close relative affected by colorectal cancer <45yo 2 close relatives affected by colorectal cancer at all Evidence of a dominant familial cancer trait Ulcerative colitis or Crohn’s disease
482
Define: population screening
The practice of investigating apparently healthy individuals with the object of detecting unrecognised disease or a high risk of developing disease, and of intervening in ways that will prevent the occurrence of disease or improve the prognosis when it develops
483
When should a screening programme be implemented?
Condition should be important in respect in respect to seriousness and/or frequency Natural history of disease must be known (to ID where screening can take place and enable effects of any intervention to be assessed) Consider test characteristics - Simple and acceptable to patient - Sensitive and selective Screening population should have equal access to screening procedure Cost effective
484
What NHS screening is available for colon cancer?
FOB (fecal occult blood) Positives referred for: - 60-75y colonoscopy - 55-60y sigmoidoscopy
485
If a 76 year old man presents with new onset rectal bleeding... A. Haemorrhoids must be excluded in first instance B. Factor 5 leiden abnormalities are the likely cause C. GP should reassure and send patient home D. Colorectal malignancy must be excluded in the first instance E. Crohn's disease must be excluded in the first instance
D. Colorectal malignancy must be excluded in the first instance
486
With respect to the aetiology of colorectal adenocarcinoma: A. Many carcinomas are derived from adenomas B. Adenomas are invasive tumours C. UC is the underlying cause in many cases D. Many carcinomas are derived from hyperplastic polyps E. Angiodysplasia is the underlying cause in many cases
A. Many carcinomas are derived from adenomas
487
What is leukaemia?
Cancer of the bone marrow and other blood-forming organs (5% all cancers) Most common cancer in M&F 15-24
488
What mutations lead to leukaemia?
Series of mutations in pluripotential haematopoietic stem cells, single lymphoid cell (stem cell, pre B lymphocyte or pre T lymphocyte) or myeloid stem cell Cells show abnormalities in proliferation, differentiation or cell survival-> expansion of cell clone
489
Why is leukaemia different from other cancers?
MATTER STATES Most cancers exist as solid tumours- leukaemic cells replace normal bone marrow cells and circulating freely in the blood stream BEHAVIOUR Haemopoietic and lymphoid cells behave differently from other body cels Recirculate METASTASIS Leukaemia doesn't have concepts of invasion and metastasis Instead 'benign'= chronic (goes on) 'Malignant'= acute (very aggressive if untreated)
490
How can leukaemia be classified?
``` 'Benign'= chronic (goes on) 'Malignant'= acute (very aggressive if untreated) ``` Lymphoid or myeloid - Lymphoid can be B or T lineage - Myeloid can be any combo of granulocytic, monocytic, erthroid or megakaryocytic
491
What are the main types of leukaemia?
Acute lymphoblastic leukaemia (ALL) Acute myeloid leukaemia (AML) Chronic lymphocytic leukaemia (CLL) Chronic myeloid leukaemia (CML)
492
What are the important leukaemogenic mutations that have been recognized?
Mutation in a known proto-oncogene Creation of a novel gene, e.g. a chimeric 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 result from deletion or mutation If there is tendency to increased chromosomal breaks-> increased risk
493
What 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
494
What are the identifiable causes of leukaemogenic mutations?
Irradiation Anti-cancer drugs Cigarette smoking Chemicals e.g. benzene
495
What rare inherited abnormalities can lead to leukaemia?
Immune deficiency | Bone marrow failure syndrome
496
What is AML?
Generally due to mutations affecting TFs (so transcription of multiple genes is affected) Often oncogene product prevents normal protein function Dominant negative effect Cells continue to proliferate but they no longer mature - > build up of the most immature cells (myeloblasts/blast cells) in BM (spread into blood) - > failure of production of normal functioning end cells e.g. neutrophils, monocytes, erythrocytes, platelets
497
What is CML?
Generally due to mutations affecting a gene encoding a protein in the signalling pathway between a cell surface R and nucleus Protein may be membrane receptor or a cytoplasmic protein Cell kinetics and function not as seriously affected as in AML Cell becomes independent of external signals (altered stromal interactions, reduced apoptosis-> cells survive longer)
498
What is the overall difference between AML and CML?
``` AML= failure of production of end cells (increased lymphoblasts) CML= increased production of end cells (abnormal mature cells) ```
499
What happens to lymphoblasts in AML?
Increase in very immature cells | Failure to develop into mature T and B cells
500
What kind of cells are affected in CML?
Leukaemic cells are mature T cells or B cells (although abnormal)
501
How does leukaemia cause the disease characteristics?
Accumulation of abnormal cells Metabolic effects Crowding out of normal cells
502
What are the metabolic effects of leukaemia cell proliferation?
Hyperuricaemia and renal failure Weight loss Low grade fever Sweating
503
What does accumulation of abnormal cells lead to in leukaemia?
ACCUMULATION OF ABNORMAL CELLS - > Leucocytosis - > Bone pain (if leukaemia is acute) - > Hepatomegaly - > Splenomegaly - > Lymphadenopathy (if lymphoid) - > Thymic enlargement (if T lymphoid) - > Skin infiltration
504
What does crowding out of normal cells lead to (in leukaemia)?
Anaemia Neutropenia Thrombocytopenia
505
What can be seen on the hands of a patient with leukaemia?
Palor Haemorrhages (Signs of anaemia and thrombocytopenia)
506
What can be seen in gums of leukaemia patients?
Monocytic differentiation Haemorrhage into gums (thrombocytopenia) Thickened gums due to infiltration via leukaemia cells
507
Who does acute lymphoblastic leukaemia usually effect?
Children
508
What is suggested to cause B-lineage in acute lymphoblastic leukaemia (ALL)?
Delayed exposure to a common pathogen OR CONVERSELY Early exposure to pathogens protect Theorised because of studies of family size, new towns, socio-economic class, early social interactions, variations between countries More affluence-> more like to get leukaemia
509
What can increase leukaemias in infants and young children?
Irradiation in utero In utero exposure to certain chemicals (e.g. Baygon, Dipyrone) Epstein-Batt virus infection Rarely due to mutagenic drug
510
What causes the clinical features of ALL?
Accumulation of abnormal cells Crowding out of normal cells
511
What clinical features of ALL result from accumulation of abnormal cells?
``` Bone pain Hepatomegaly Splenomegaly Lymphadenopathy Thymic enlargement (mass) Testicular enlargement (bilateral) ```
512
What clinical features of ALL result 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)
513
What are the haematological features of ALL?
Leucocytosis with lymphoblasts in the blood Anaemia (normocytic, normochromic) Neutropenia Thrombocytopenia Replacement of normal bone marrow cells by lymphoblasts
514
How can you investigate ALL?
Blood count and film - ALL-> increased numbers, more immature cells Check of liver and renal function and uric acid Bone marrow aspirate Cytogenetic/molecular analysis - Immunophenotyping - Fluorescent antibodies are applied to cells recognise antigens on cell surface Chest X-ray
515
Why is cytogentic/molecule analysis of ALL useful?
How to manage patient (info about prognosis) Advances knowledge of leukaemia
516
What do hyperdiploidy and t(4;11) in cytogenetic analysis of ALL show?
Hyperdiploidy= good prognosis t(4;11)= poor prognosis
517
What are the leukaemogenic mechanisms of ALL?
Formation of a fusion gene (due to translocation) Dysregulation of a prot-oncogene by juxtaposition of it to the promoter of another gene e.g. T-cell receptor gene Point mutation in a proto-oncogene
518
How can you detect fusion gene formation (by translocation)?
FISH= fluorescence in situ hybridization Detected by 2 fluorescent probes (different for different regions) (e.g. red and green) When a fusion gene is formed the two colours fuse to give a combined signal (e.g. yellow)
519
How is ALL treated?
Supportive= red cells, platelets, antibiotics Systemic chemotherapy Intrathecal chemotherapy
520
Why are migrant studies informative about cancer epidemiology? What do rapid and slow changes in risk following migration show?
Rapid change implies lifestyle/environment factors act late in carcinogenesis Slow change implies exposures early in life are the most relevant Persistence of rates between generations suggests genetic susceptibility is important in determining risk
521
Give an overview of cancer epidemiology
Incidence is increasing for common cancer sites in both high-income (now with plateauing and even decreases) and low-income countries (e.g. breast, colorectum, prostate)- effects of earlier diagnosis, screening, changes in risk factors? Mortality is decreasing in most high-income countries but not in low income countries Total burden increasing becuase of demographic changes (ageing populations and increasing size) and Westernization of lifestyles
522
What type of cancer does hereditary retinoblastoma lead to?
Retinoblastoma
523
What type of cancer does xeroderma pigmentosum lead to?
Skin cancer
524
What type of cancer does Wilms' tumor lead to?
Kidney cancer
525
What type of cancer does Li-Fraumeni syndrome lead to?
Sarcomas, brain, breast, leukaemia
526
What type of cancer does FAP lead to?
Colon, rectum cancer
527
What type of cancer does Paget's disease lead to?
Bone cancer
528
What type of cancer does Fanconi's aplastic anemia lead to?
Leukemia, liver, skin
529
What are the main risk factors for cancer?
``` Smoking Diet Alcohol Infection Occupation Reproductive hormone ```
530
What percentage of cancers in the UK does Parkin estimate are preventable?
45% of cancers in men and 40% in women These could have been prevented had risk factors been reduced to the optimal levels or eliminated (like tobacco)
531
What percentage of cancer deaths are caused by smoking?
30% | 90% of lung cancer deaths in men and 80% in women
532
What kinds of cancers can alcohol be involved in?
Oral cavity, pharynx, larynx, oesophagus, liver Mechanisms poorly understood Synergism with tobacco Balance with preventive effect for CHD
533
What are the dietary guidelines for preventing cancer?
World Cancer Research Fund Be as lean as possible without becoming underweight Be physically active for at least 30 mins every day Avoid sugary drinks and limit consumption of energy-dense foods Eat more of a variety of vegetables, fruits, wholegrains and pulses Limit consumption of red meats and avoid processed meats Limit alcoholic drinks (2 for men and 1 for women a day) Limit consumption of salty foods and foods processed with sodium Don't use supplements to protect against cancer just eat well
534
What types of hormonal influences cause breast cancer?
Exogenous and endogenous hormones | E.g. pill
535
What Western lifestyle factors contribute to cancer?
Energy dense diet, rich in fat, refined carbs and animal protein Low physical activity Smoking and drinking
536
What does 'Westernization' of lifestyle lead to?
Greater adult body height Early menarche Obesity Diabetes Cardiovascular disease Hypertension Cancer
537
What percentage of cancer is likely caused by infectious agents?
16% worldwide
538
What kind of infectious agents can cause cancer?
HPV-> cervix, head and neck EBV-> Hodkin's lymphoma, Burkitts HCV/HBV-> liver H. pylori-> stomach
539
What do we know from epidemiology of cancer?
Cancer incidence is related to: Age Common environmental causes Geographical variation and secular trends
540
Are there inherited/familial cancers?
Yes but rare | Provide valuable info in understanding the process of carcinogenesis
541
What is the leading female cancer?
Breast (1 in 8 will develop in UK and USA, 1 in 5 cancer deaths) Liver more common overall
542
What countries have higher rates of breast cancer?
Developed countries
543
What is the current trend in breast cancer incidence and mortality?
Incidence rising | Mortality falling
544
Why is breast cancer mortality falling?
Early diagnosis Chemo and radiotherapies Hormonal therapies
545
Why is the breast an unusual organ?
Only organ that develops post-natally Dramatic changes in size, shape and function through infantile growth, puberty, pregnancy, lactation, weaning and postmenopausal regression
546
What kind of cancers are most breast cancers?
Carcinomas (tumour of epithelial cells)
547
What kinds of cancers/disease can affect the breast? Where do these affect?
Lobular cancer (lobule) Ductal cancer (duct) Duct ecstasia (duct) Paget's disease (lactiferous sinus) Phyllodes (fat and connective tissue stroma= very rare, sarcomal)
548
When is the main spurt of breast growth?
Puberty | Dependent on high levels of estrogen and progesterone produced by ovary
549
What happens to the breast in post-pubertal development?
Cyclical increases in ductal branching -> Resulting in extensive branching in the fat pad
550
What stages of breast development require oxygen?
Estrogen not necessary for the prenatal development of the mammary gland Estrogen required for prepubertal and pubertal gland development
551
What happens to the breast shape in pregnancy?
Characterised by large increases in side branching and development of secretory acini from the terminal ductal alveoli
552
How does the mammary gland return to near pre-pregnancy state after weaning?
Involved extensive apoptosis
553
What is unusual about the epithelial cells in the mammary glands?
2 layers of epithelial cells Inner- luminal (make contact with lumen) Outer- myoepithelial (make contact with basement membrane)
554
What acts on luminal and myoepithelial cells?
Inner= luminal epithelial cells (steroid hormones act on these) Outer= myoepithelial cells (able to contract in response to hormonal cues e.g. oxytocin) During lactation this is important
555
What are the most common types of breast cancer?
Invasive ductal carcinoma (80%) | Invasive lobular carcinoma (5-15%)
556
Where do most common breast cancers originate? How do they progress?
In the terminal duct lobular unit Progress from an initial hyperproliferative stage to a pre-cancerous in situ carcinoma stage Then progresses to invasive breast cancer Gives rise to range of carcinomas e.g. medullary carcinoma, carcinoma and lobular carcinoma
557
Why is immunohistochemical staining using antibodies against the Human Estrogen Receptor (ER) informative?
Any cell that expresses ER will be senstivie to oestrogen (oestrogen drives growth of cell) Different labs have different cutoff points for the calling the cancer either ER-positive or ER-negative Any positive result at all suggests hormonal therapy could be used
558
What risk factors are important in breast cancer growth?
Age of onset of menarche Age to first full-term pregnancy (having children earlier-> lower incidence) Some contraceptive pills Some hormone-replacement therapies Late age to menopause Diet, physical activity, height, medication (aspirin) Obesity
559
How does estrogen act on cells?
Gets across cell membranes easily | Can only have an action if cell has estrogen R (nuclear receptor)
560
What happens when an estrogen receptor is activated?
Binding estrogen activates estrogen receptor Gene expression induced by binding to specific DNA sequences called estrogen response elements Estrogen-induced gene products increase cell proliferation-> breast cancer
561
List some important estrogen regulated genes
Progesterone receptor (estrogen makes cells with ERs also receptive to progesterone) Cyclin D1 c-myc TGF-alpha
562
What percent of premenospausal women with advanced breast cancer will respond to oophorectomy?
1/3 Sensitive to effects of estrogen
563
What is paradoxical about breast cancer in postmenopausal women?
Responds to high dose therapy with synthetic estrogens i.e.g causes breast tumour regression
564
What percentage of breast cancers have over expressed estrogen receptors?
70% | Presence is indicative of better prognosis
565
What i the role of estrogen in an ER-positive case?
Estrogen regulates the expression of genes involved in cellular proliferation leading to breast cancer
566
What are the major kinds of primary therapy to treat breast cancer?
SURGERY Mastectomy= removal of breast Lumpectomy= surgery to remove tumour and a small amount of normal tissue around it (breast conserving- almost always includes radiation therapy)
567
What are the major treatment approaches for breast cancer?
Surgery Radiation therapy Chemotherapy Endocrine therapy
568
What is an adjuvant therapy for breast cancer?
Any kind of treatment given after primary therapy to increase long-term disease-free survival Useful to kill any cancer cells that may have spread Increase chance of long-term survival
569
What are the main aims of endocrine therapy in breast cancer treatment?
Ovarian suppression Blocking estrogen production by enzymatic inhibition Inhibiting estrogen responses
570
When are estrogen levels highest in the menstrual cycle?
End of follicular phase just before ovulation
571
Outline hormonal control of target tissues in the LHRH system
PREMENOPAUSAL Hypothalamus releases LHRH-> pituitary gland releases gonadotrophins (FSH + LH) -> ovary (aromatization) releases esterogens and progesterone-> acts on body PRE/POST MENOPAUSAL Hypothalamus releases ACTH-> adrenal glands release corticosteroids, progesterone and androgens Androgens undergo peripheral conversion to estrogens -> acts on body
572
True or false; fat is good for peripheral conversion
True
573
What is ovarian ablation carried out by?
Surgical oophorectomy | Ovarian irradiation
574
What are the major problems associated with ovarian ablation?
Morbidity and irreversibility Now can have reversible/ reliable medical ovarian ablation
575
Where does most estrogen biosynthesis happen in pre-menopausal women?
Ovary
576
How can reversible ovarian ablation be achieved?
Using LHRH agonists Act on pituitary gland = LHRH agonists bind to LHRH Rs in the pituitary-> R down-regulation and suppression of LH release and inhibition of ovarian function (including estrogen production) Prevent production of FSH and LH
577
Give examples of LHRH agonists
Goserelin Buserelin Leuprolide Triptorelin
578
What are the targets for breast cancer treatment?
LHRH agonists (pit gland LHRH Rs) Aromatase inhibitors (aromatization in ovary and peripheral conversion) Antiestrogens (inhibit estrogen activity)
579
What is trans-activation by estrogen receptors?
Upon binding ligand, the receptor dissociates from hsp90 dimerises and binds to response elements in regulated genes, enabling transcriptional activation These inhibit the effects triggered by activation of the response element (prevent TATA-> gene expression)
580
What is the developed anti-estrogen?
Tamoxifen
581
What is tamoxifen?
Anti-estrogen (selective estrogen receptor modulator= SERM) Competitive inhibitor of estradiol binding to the ER Administered as tamoxifen citrate and undergoes extensive metabolism in GI tract and liver (particularly by hydroxylation)
582
Who is tamoxifen effective on?
For metastatic disease in postmenopausal patients (approx 1/3 respond) As effective in pre-menopausal as in post-menopausal women
583
How do anti-estrogens work?
Negate stimulatory effects of estrogen by blocking ER-> holds cell at G1 phase of cell cycle
584
Why does tamoxifen has estrogenic effects in bone and cardiovascular system?
BONE Long-term administration of an anti-estrogen has the potential to precipitate premature osteoporosis (Because estrogen important to maintain bone in premenopausal women) CV SYSTEM Following menopause, women at same risk for CHD as men Long-term administration of an antiestrogen could produce a population at risk for premature coronary heart disease (Because estrogen lowers LDL levels and raises HDL levels)
585
What are the undesirable effects of tamoxifen?
SEs= 29% have hot flushes LIVER= Possible subsequent thromboembolic episodes UTERUS= Endometrial thickening (-> cancer), hyperplasia, fibroids, polyps, vaginal discharge HYPOTHALAMUS= Increased vasomotor symptoms EYE= Increased cataracts DVT
586
What are the desirable effects of tamoxifen?
BREAST= reduces breast cancer LIVER & HEART= lowers cholesterol, reduces atherosclerosis and heart attacks BONE= maintains density to help prevent bone loss
587
What are the desirable effects of estrogen?
BRAIN= improves cognitive function BREAST= programs glands to produce milk LIVER & HEART= lowers cholesterol, reduces atherosclerosis and heart attacks UTERUS= programs uterus to nourish a fetus BONE= maintains density to help prevent bone loss
588
What are the undesirable effects of estrogen?
BREAST= promotes breast cancer LIVER= increases thromboembolism UTERUS= promotes endometrial cancer
589
What compounds are currently being considered to treat all stages of breast cancer?
Toremifene ICI 182, 780 Raloxifene (faslodex SERM)
590
What have tamoxifen trials focused on?
High risk patients - Previous benign breast pathology (5 years) - Previous family history
591
What have tamoxifen vs placebo trials shown?
38% reduction in overall breast cancer incidence No effect on ER negative breast cancer incidence No association between prevention and patient age
592
Why are aromatase inhibitors used often in postemnopausal women?
Major source of estrogen in post-menopausal women derives from conversion of adrenal hormone (androstenedione, A and sometimes testosterone to estroen, E2)
593
Where is androstenedione (and sometimes testosterone) converted to estroen? Catalyst?
Extra-adrenal or peripheral sites e.g. fat, liver and muscle Catalysed by aromatase enzyme complex (3 separate steroid hydroxylations) -> production of estrone sulphate (which is circulated in the plasma)
594
What does aromatase consist of?
A complex containing a cytochrome P450 heme containing protein as well as the flavoprotein NADPH cytochrome P450 reductase
595
What are the classes of aromatase inhibitors?
Type 1= irreversible, mechanism-based/ suicide inhibitors (initially compete with natural substrate then irreversibly inactive enzyme) e.g. exemestane drug Type 2= competitive inhibitors (reversible binding) e.g. anastrozole drug
596
Why are progestins important in breast cancer?
Progesterone= dominant naturally occurring progestin Influence proliferation and differentiated functions in human breast
597
Why are progestins used in endocrine treatment of uterine and breast cancer?
Clinically proven antineoplastic properties Used for metastatic breast cancer as 2nd or 3rd line therapy following selective estrogen
598
What is the principal progestin used for metastatic breast cancer?
Megestrol acetate
599
What happens when patients with metastatic breast cancer become resistant to endocrine therapies?
Happens in significant proportion of patients Most cases continue to demonstrate estrogen responses and contain estrogen receptor
600
What is the clinical problem with targeting estrogen receptors in breast cancer?
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
601
How can you prevent relapse due to resistance during prolonged endocrine therapy (ER in breast cancers)?
Continue endocrine therapies | But with additional therapeutic agents/strategies for endocrine resistant, metastatic disease
602
How can ERs be used in treatment of breast cancer?
>60% of ERα-positive tumours respond to endocrine therapy Anti-estrogens e.g. tamoxifen Inhibitors of estrogen synthesis e.g. exomestane
603
How is ER-positive primary breast cancer treated in pre-menopausal women?
``` Ovarian ablation (and then if relapse with metastatic disease= tamoxifen, aromatase inhibitors, progestins, ?faslodex) ``` Tamoxifen (and then if relapse with metastatic disease= ovarian ablation, aromatase inhibitors, progestins, ?faslodex)
604
How is ER-positive primary breast cancer treated in post-menopausal women?
Tamoxigen | and then if relapse with metastatic disease= aromatase inhibitors, progestins, ?faslodex
605
What is the screening programme for breast cancer?
Mammography to screen all women between 50-64 (being extended to age 70) Once every 3 years More than 70% women attend 6/100 asked to go back for more tests
606
What percentage of breast tumours are first spotted by women themselves?
90%
607
What are the molecular sub-types of breast cancer?
ERa -ve - Basal-like(metaplastic/ medullary/ mucinous/ others) - ErbB2 +ve (herceptin) - Normal-like ERa +ve - Luminal B/C (hormone therapy) - Luminal A (hormone therapy)
608
What is the patient history of breast cancer?
Lump detected (self exam/GP) Referred to hospital Examined by surgical team (mammogram, FNA) Surgery performed (lumpectomy/ mastectomy) Tumour examined pathologically (ER/ PR) See physician for first time ER+ (tamoxifen 5 years) ER- (chemo 6 months) Disease free interval If patient returns with secondary tumour (no cure)
609
What are the malignant skin cancer types?
``` Basal cell carcinoma Squamous cell carcinoma Malignant melanoma Kaposi’s sarcoma Mycosis fungoides ```
610
What are the layers of the skin?
Epidermis (1st line of defence) Dermis Hypodermis NB. Have blood vessels, hair follicles and sebaceous glands
611
Outline the structure of the epidermis?
``` OLD at top, YOUNG at bottom Dead keratinocytes (at surface- flake off) Stratum corneum Stratum lucidum Stratum granulosum Stratum spinosum (includes dendritic cells and living keratinocytes) Stratum basale (includes melanocytes) Dermis (includes sensory nerve ending) ```
612
What types of skin cancer are keratinocyte derived?
Basal cell carcinoma | Squamous cell carcinoma (aka non- melanoma skin cancer, NMSC)
613
What type of skin cancer is melanocyte derived?
Malignant melanoma
614
What types of skin cancer are vasculature derived?
Kaposi's sarcoma, angiosarcoma
615
What type of skin cancer is lymphoocyte derived?
Mycosis fungoides (not actually a fungus just incorrectly named)
616
How does accumulation of genetic mutations lead to skin cancer?
Uncontrolled cell proliferation
617
What genetic syndromes can cause skin cancer?
Gorlin’s syndrome= defect in patch gene-> basal cell Xeroderma pigmentosum= genetic condition (DNA repair damaged- BCCs, MCCs, and melanoma)
618
What viral infections cause skin cancer?
HHV8 in Kaposi’s sarcoma HPV in SCC
619
What kinds of cancer result from UV light?
BCC, SCC and malignant melanoma
620
How does immunosuppression cause skin cancer?
Drugs, HIV, old age, leukaemia
621
What are the distinguishing features of a malignant melanoma?
Irregular margin and colour | Often on trunk
622
What are the distinguishing features of a basal cell carcinoma?
Most common kind of skin cancer Glistening surface Dilated vessels on surface Often on head
623
What are the wavelengths of UVC, B and A?
``` UVC= 100-280 UVB= 280-310 UVA= 310-400 ```
624
Where can UVC, UVB and UVA reach on the earth?
``` C= only through ozone layer B= to sea level (main cause of skin cancer) A= to Dead Sea level ```
625
Why is sunlight essential for life?
Essential for photosynthesis (plants) Infrared spectra provide warmth Effect on human mood Stimulates the production of vitamin D in the skin
626
Is UVA or B more penetrative?
A= 100x more UVA penetrates to Earth's surface
627
What does UVA cause?
Skin ageing | Contribution to skin carcinogenesis (B most important)
628
How does UVB contribute to skin cancer?
UVB directly induces abnormalities in DNA e.g. mutations UVB induces photoproducts (mutations): 1) Affects pyrimidines - i.e. Cytosine and Thymine bases cyclobutane pyrimidine dimers - 6-4 pyrimidine pyrimidone photoproducts 2) Usually repaired quickly by nucleotide excision repair
629
How does UVA contribute to skin cancer?
DNA forming cyclobutane butane pyrimidine dimers Less efficiently than UVB Free radicals which damage DNA and cell membrane
630
What genes are commonly affected by UV damage to DNA?
Cell division DNA repair Cell cycle arrest
631
How are photoproducts removed? What happens when this stops working?
Removed by Nucleotide Excision Repair Defective-> Xeroderma pigmentosum
632
What is Xeroderma pigmentosum?
Genetic condition with defective Nucleotide Excision Repair Any skin type Hundreds/thousands of skin cancers Die at early age If diagnosed early may have a relatively normal life
633
What types of mutations cause cancer?
Mutations that stimulate uncontrolled cell proliferation Mutations that alter responses to growth stimulating / repressing factors Mutations that inhibit apoptosis
634
What happens in sun burn?
Very rapid accumulations of mutations in DNA Skin reacts by causing apoptosis (to remove UV damaged cells which might become cancer cells) - Keratinocyte cell apoptosis Sun burn cells= apoptotic cells in UV overexposed skin
635
What happens in photocarcinogenesis?
Normally UV-> DNA damage-> repair of DNA-> normal cell Cancer UV-> p53 mutation inactivated wildtype -> skin cancer Apoptosis UV-> DNA damage-> damage too severe so can't repair-> apoptosis
636
Outline the immunomodulatory effects of UV light?
UVA and UVB effect the expression of genes involved in skin immunity (depletes Langerhans cells in the epidermis) Reduced skin immunocompetence and immunosurveillance (Basis for UV phototherapy for e.g. psoriasis) Further increases the cancer causing potential of sun exposure
637
What happens if Langerhans cells are damaged by UV?
Immune response-> cell death (malignant transformation) Skin cancer develops
638
What are the Fitzpatrick Phototypes?
I - 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 - Afrocaribbean
639
What is melanin?
Pigment responsible for skin colour Produced by melanocytes within the basal layer of the epidermis Colour depends on melanin type/amount not density of melanocytes (which is fairly constant) 2 types (eumelanin and phaeomelanin) Dictates skin sensitivity to UV damage
640
Where are melanocytes and melanin found?
Melanocytes normally only on BM | Melanin is equally distributed along basal layer
641
What are the two types of melanin formed?
Eumelanin= brown or black Phaeomelanin= yellowish or reddish brown Melanin is formed from tryosine via a series of enzymes
642
What is the MCR1 gene?
Codes for melanin >20 gene polymorphisms Variation in eumelanin: phaeomelanin produced (explains different hair colour and skin types)
643
How is melanin produced?
Produced by melanocytes: Tyrosine-> Dopa-> Dopaquinone -> Eumelanin and pheomelanin -> melanin Transfered into keratinocytes
644
What is a malignant melanoma?
Malignant tumour of melanocytes (become abnormal, atypical cells and architecture) Cause by UV exposure and genetic factors Risk of metastasis
645
What is a lentigo maligna?
Melanoma in situ Flat large tumours, irregular shape, light/dark brown colours, usually >2cm, often on face Proliferation of malignant melanocytes within the epidermis No risk of metastasis (hasn't invaded deeply through BM)
646
What is a superficial spreading malignant melanoma?
Malignant melanocytes proliferate laterally Invade basement membrane Risk of metastasis
647
How can you diagnose superficial spread malignant melanoma?
ABCD rule ``` Asymmetry Border irregular Colour variation (dark brown-black) Diameter >0.7mm and increasing Erythema (redness) ```
648
What is a nodular malignant melanoma?
Vertical proliferation of malignant melanocytes (no previous horizontal growth) Risk of metastasis
649
What is a nodular melanoma arising within a superficial spreading melanoma?
Downward proliferation of malignant melanocytes Following previous horizontal growth Nodule develops within irregular plaque Prognosis worse than superficial spreading melanoma
650
What is an acral lentiginous melanoma?
``` Acral= hands and feet Lengtiginous= flat ``` Occur in darker skin people
651
What is an amelanotic melanoma?
Tumour that doesn't make melanin
652
What are the types of malignant melanoma?
``` Superficial spreading Nodular Lentigo maligna melanoma Acral lentiginous Amelanotic ```
653
How can you determine melanoma prognosis?
Breslow thickness Measurement from granular layer to bottom of tumour Ticker= worse
654
What are the main risk factors for the development of melanoma?
Family history of dysplastic nevi or melanoma UV irradiation Sunburns during childhood Intermittent burning exposure in unacclimatized fair skin Atypical/dysplastic nervus syndome Skin type I or II Personal history of melanoma
655
What is a keratoaceanthoma?
Sudden growth no risk of metastasis
656
Does a squamous cell carcinoma have a risk of metastasis?
Yes
657
What is a squamous cell carcinoma and what is it caused by?
Malignant tumour of keratinocytes Caused by: UV exposure, HPV, immunosuppression, smoking, scarring process Risk of metastasis
658
What causes a cutaneous horn?
Well differentiated squamous cell carcinoma
659
What is a basal cell carcinoma and what is it caused by?
Malignant tumour arising from basal layer of epidermis Cause by sun exposure and genetics Slow growing Invades tissue but doesn't metastasis Common on phase
660
What are the little vessels called in BCC?
Arborizing vessels | Telangiectasia
661
How long does mycosis fungoides take to progress?
Takes years or decades to progress
662
What is Kaposi's sarcoma associated with?
HIV and HHV8 associated
663
What is epidermodysplasia veruciformis?
Rare autosomal recessive condition | Predisposition to HPV induced warts and SCCs