Integrity: Cancer Pathology Flashcards

1
Q

Definitiion of atrophy

A
  • Atrophy
  • Shrunken tissue with reduced cell size (± number)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Definition of hypertrophy

A

Hypertrophy
* Enlargement of a tissue with increased cell size

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

Definition of hyperplasia

A

Hyperplasia
* Increased number of otherwise normal cells in a tissue

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

Definition of transdifferentiation

A

Transdifferentiation
* A switch of differentiation direct from one mature lineage to
another which is normally present in that tissue

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

Definition of metaplasia

A

Metaplasia
* A switch of differentiation from one mature phenotype to
another which is not normally present in that tissue,
in response to an environmental change.

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

Definition of dysplasia

A

Dysplasia
* Disordered microscopic appearance and maturation of cells,
implying neoplasia.

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

Definition of a tumour

A

Tumour
* Abnormal lump of no specific cause (often presumptively a neoplasm)

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

Definition of a cyst

A

Cyst
* Abnormal fluid-filled lesion lined with epithelium

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

Definition of a hamartoma

A

Hamartoma
* Disorganised but mature normal tissue elements, lacking autonomous growth

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

Definition of neoplasm

A

Neoplasm
* Abnormal accumulation of cells derived from a mutated ancestor ‘seed’ cell
* growth is autonomous of environmental restraining signals

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

Deinition of cancer

A

Cancer = a malignant neoplasm

  • Invasion: crosses tissue boundaries
  • Metastasis: discontinuous spread to survive & grow at remote sites
  • carriage in: blood / lymph / serous cavity fluid / CSF (cerebrospinal fluid)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

How is the craniospinal venous system relevent in metastasis?

A
  • 2-way venous flow (no valves)
  • Links cranial + vertebral circulation
    with intercostal, abdominal & pelvic
    venous plexuses
  • Direct ’back-door’ route for metastasis
    to spine and brain
  • skips lungs, lymphatics
  • e.g. from prostate, breast, thyroid.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

6 appearances of surface neoplasms

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

Definition of carcinoma

A

Carcinoma is cancer that forms in epithelial tissue

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

What is a sarcoma?

A

Sarcoma is the general term for a broad group of cancers that begin in the bones and in the soft (also called connective) tissues (soft tissue sarcoma)

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

What is Cachexia

A

progressive muscle dysfunction + lean muscle wasting (+/- adipose loss), not reversible with nutritional support

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

What is paraneoplastic?

A

Paraneoplastic = a problem caused by a neoplasm but not attributable to cancer invasion or secretion of indigenous tissue hormone aka it’s directly, physically or anatomically affecting an organ

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

What is clonal expansion?

A

This is a normal process where you aquire cancer driving mutations (onces that encourage growth) but they don’t progress further, if these fill a neish well they will expand and outcompete the surrounding cells

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

What happens with clonal expansion and age?

A

Clonal expansion increases with age and to varying degrees in different sites

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

What is a cancerisation field?

A

Cancerisation field
A tissue with accumulated genetic & epigenetic changes that favour
cancer emergence

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

Field cancerisation:

Mutation count & clonal patch sizes increase with…

A

Mutation count & clonal patch sizes increase with

  • chronic inflammation (cycles of destruction and regeneration)
  • carcinogens: sun exposure, smoking, alcohol
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Consequence of non-cancerous clonal haematopoiesis

A

clonal haematopoiesis is pro-inflammatory
* Coronary heart disease & stroke (larger risk than hypertension)

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

What has the highest risk of tissue that is
Normal
Hyperplastic
Metaplastic
Dysplastic

A

Dysplastic

Usually means it has gotten much bigger

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

How does cancer arise from clonal expansion: clonal sweep

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

What are the hydrodynamics of a tumour and why is it relevent?

A

The core has a very high interstitial (between cells) pressure resulting in an area with poor diffusion, hypoxia, acidic and ripe for ischemia.

This hypoxia produces a strong selection bias for angiogenesis in growing cancers (through HIF - hypoxia driven gene expression and angiogenic factors)

The flow of fluid is also radially outward from the core and then towards the lymphatic drains.

This makes it hard for drugs to diffuse into the core and promotes cells migration towards the lymphatics

Key points:
* High core pressure
* Hypoxia in core which promotes angiogenesis selection bias
* Prevents drugs from reaching core and promotes spread outward towards lymphatics

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

WHat is angiogenesis?

A

The creation of new blood vessels

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

What’s happening here?

A

Competition for blood in cancer creates dissorganised angiogenesis

This results in non-hierarchical unstable haemodynamics

  • pooling, eddies, flow reversals
  • pO2 varies in “waves & tides”
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What is a cancer driver gene?

A

Most human cancers have 2-8 mutant drivers

> 500 total but most early cancers have mutations in the same 9 genes

Typically they affect multiple cancer hallmarks, suppressing some and promoting others

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

How are do key drivers differ after treatment?

A

Key driver mutations are often ubiquitous in untreated cancer + its metastases

  • New mutations often have no selective impact: neutral passenger mutations in ‘subclones’

After treatment you are applying new pressures which unmask previously neutral or minority but resistant subclones

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

Key cancer mutation hallmarks

4 Early

4 Late

A

Early

  1. Promotes proliferation genes
  2. Surpresses anti-proliferation genes
  3. Unlimited copying - removes caps that normal cells have whereby they can only be copies so many times before the quality becomes worse
  4. Metabolic reprogramming for energy

Late

  1. Evasion of immune system
  2. Resisting aopoptitic mechanisms
  3. Invasion and metastasis
  4. Angiogenesis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What do we mean by plasticity when it comes to cancer cells

A

Many have an ability to change state, whether this is a speciality state or a stem state.

This means they can often adapt to differing pressures

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

What does epithelial-mesenchymal plasticity (EMP) mean?

A

Some cancer cells can switch between being epithelial and mesenchymal

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

How do cancer cells subvert immune activation
* release immunosuppressive factors
* TGFb, IL-10, PG
* repress antigen processing & presentation
* Small cell lung cancer
* reduce MHC I; display immune inhibitors (PDL1, CTLA4)

A

The first is by creating the space of metabolic stress this makes it had to immune cells to survive. Cancer cells can competitively steal glucose from T cells.

Aditionally:

  • release immunosuppressive factors TGFb, IL-10, PG
  • repress antigen processing & presentation
  • reduce MHC I; display immune inhibitors (PDL1, CTLA4)
34
Q

Why is relapse quite common?

A

Risidual cells are often left behind after treatment (these can be a handful of subclones)

These are then treatment resistant and spread making a second treatment less successful

35
Q

What is enhancer switching?

A

Enhancer switching is where cells can activate different enhancers to promote the same survival genes. Multiple different routes to the same end. This gives heritable and reversible resistance to targeted cancer therapy

36
Q

Three types of carcinogen

A

chemical - natural or synthetic
physical - UV or ionising radiation
biological - bacteria, viruses, parasites

37
Q

What is Replicative Senescence and what is used to overcome replicative senescence in germ and stem cells

A

Replicative senescence is the phenomenom where cells can only divide so many times.

The end of chromosomes are clipped with repetitive sequences called telomeres. These shorten with every division eventually resulting in apoptosis.

Telomerase is present in stem cells and germ cells in order to maintain telomere ends which results in unlimited replication.

Telomerase is abnormally upregulated in cancer cells

38
Q

What are the loops on the ends of chromosome designed to cap unlimited cell replication called?

A

Telomeres

39
Q

What substance can maintain telomeres during replication?

A

Telomerase

40
Q

What happens to telomerase in cancer cells?

A

Abnormally upregulated

41
Q

What is the cap on the number of times a cell can divide called?

A

Replicative senescence

42
Q

What is inherited predisposition to cancer in humans

A

In rare instances tumour mutations can be inherited in the germ line giving rise to an inherited predisposition to a particular cancer.

43
Q

What is Retinoblastoma?

A

Retinoblastoma is a rare childhood cancer of retinoblasts, with a peak
incidence at 3-4 years of age

44
Q

What is the difference between an inherited retinoblastoma and a sporadic one?

A

You need a loss of both RB1 genes.

In the inherited version one is lost in every cell already hence if the other spontaineously mutates in any cell it results in the retinoblastoma. Bilateral likely.

In spontaineous if one cell mutates loss of one RB1 gene then the mutation would have ot happen in the same cell therefore it would likely only be in one eye. Unilateral.

45
Q

What gene is involved in a retinoblastoma?

A

Rb

46
Q

What is an allele

A

One of two or more alternative forms of a gene that arise by mutation and found in the same part of the chromosome

47
Q

What is a zygote?

A

Fertilized egg

Pre-zygotic: before fertilization
Post-zygotic: after fertilization

48
Q

What does Heterozygous mean?

A

Posessing two different alleles of the same gene

An example of a heterozygous condition is inheriting different genes for eye colour from both biological parents. If there are two different versions, it is a heterozygous genotype for that particular gene.

49
Q

Gene involved in Familial adenomatous polyposis coli (FAP)

A

APC - regulates signal transduction

50
Q

What is Familial adenomatous polyposis coli

A

An inherited colon cancer, hundred of polyps through colon or rectum

51
Q

What is Li Fraumeni sydrome?

A

A heriditary condition that predisposes people to several forms of cancer

52
Q

Gene involved in Li Fraumeni

A

p53 or TP53 - Cell cycle control/DNA damage

53
Q

What is Lynch / Hereditary
non polyposis colon cancer

A

Another heriditary colon cancer

54
Q

Gene involved in Lynch syndrome?

A

MLH1, MSH2 - DNA Mismatch repair

55
Q

Gene involved in Familial breast and ovarian
cancer

A

BRCA-1, BRCA-2
- DNA repair (d/s break repair)

56
Q

What is Basal cell naevus

A

A condition with multiple complications but a big one is numerous basal cell carcinomas

Basal cells are the cells at the bottom of the epidermis that make new skin cells

57
Q

Gene involved in basal cell naevus?

A

Ptch - Signal transduction

58
Q

Syndromes in which the heterozygotes express the tumour phenotype

(one mutation inherited form one parent however there will be one unmutated allele)

A
59
Q

Syndromes in which the homozygotes
have an increased risk of cancer
(you need to inherit two of the same in order to have an increased risk of cancer?)

A
60
Q

Common carcinogen for gastric carcinoma

A

Helicobacter pylori

61
Q

Parasitic cause of infection and carcinoma of the bladder

A

Schistosoma haematobium

62
Q

Virus linked to carcinoma of the cervix

A

HPV
human papillomavirus

63
Q

Viruses linked to hepatocellular carcinoma

A

hepatitis B virus, hepatitis C virus

64
Q

Virus linked to Nasopharyngeal carcinoma

A

EBV
Epstein Barr virus

65
Q

Virus linked to Adult T cell leukaemia

A

HTLV 1
Human T cell lymphotropic virus
type 1

66
Q

What is naphthylamine

A

Involved in the dye industry

Bladder cancer in workers exposed to naphthylamine

67
Q

What is a proto-oncogene?

A

This is a normal gene involved in regulating cell function that has to potential to become mutated and become an oncogene

68
Q

In terms of oncogene activation, what is a point mutation?

A

This is where the mutation occurs by one part of a codon switching

69
Q

In terms of oncogene activation, what is amplification

A

This is where one gene get copied over and over thus amplifying it’s effect

70
Q

In terms of oncogene activation, what is innapropriate regulation

A

This is where a promotor is present where it shouldn’t be thus activating a gene where it shouldn’t be activated

71
Q

Are oncogenes considered dominant or recessive?

A

Oncogenes activate tumour promoting factors therefor if one is switched on it has an effect hence they are considered dominant

72
Q

Are tumour suppressor genes considered dominant or recessive?

A

With a tumour suppressor genes these are genes whic work to surpress tumour function. You only need one to function in order to suppress the tumour hence both need to mutate in order for suppression to fail so they are considered recessive

73
Q

What is p53 protein?

A

This is a protein which is released in response to DNA damage which arrests the cell and stops it proliferating.

The p53 gene is therefore a tumour suppressing gene

74
Q

How do mutations in mismatch repair genes (MLH1, MSH2) encourage cancer?

A

By inhibiting missmatch repair the chance of DNA mutation will be greatly increased thus increasing the chance of cancer

75
Q

How does damage to exision repair encourage cancer?

A

DNA repair by exision of mutation is key to maining DNA. If this is inhibited mutation are more likely to stick

76
Q

How does damage to DNA strand break repair cause cancer?

A

BRCA1 & BRCA2 involved in d/s break repair. If the genes producing BRCA1 & BRCA2 are mutated then DNA is less likely to able to repair itself

77
Q

Three things invasion requires

A

Change and/or loss in cell-cell and
cell-matrix adhesion
Changes in adhesion are essential for motility
Focal proteolysis of the matrix

Movement to occupy the space

78
Q
A
78
Q

What are integrin receptors?

A

These are responsible for cell-matrix adhesion & signaling for cell survival
& proliferation.

There are often lost and rearranged in cancer cells to allow for invasion

79
Q

Things that determine where metastatic spread goes

A

Anatomy

nature of lymphatic and venous drainage
eg breast cancer -> local axilla nodes,
colorectal cancer -> local mesenteric nodes & liver

Organ specificity

adhesion molecules on the cancer that permit preferential attachment to different capillary beds

Different organs have different chemokine receptors

Common metastatic sites: liver, lung, bone, brain

80
Q

Blood markers of cancer

A
  • PSA- Prostate Ca
  • CA 125- Ovarian Ca
  • CEA- Colorectal Ca
  • AFP- Hepatocellular Ca
  • CA-15-3- Breast Ca
  • CA19-9- Pancreatic Ca
    Potential: cell-free DNA in blood – tumour mutations