Neoplasia Flashcards

1
Q

What two components are all tumours composed of?

A

Neoplastic cells that constitute tumour parenchyma and reactive stroma made up of connective tissue, blood vessels and cells of the adaptive and innate immune system

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

Define hamartoma

A

Disorganised mass composed of cells indigenous to the involved tissue. (As in other neoplasms most have clonal chromosomal aberrations that are aquired through somatic mutation)

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

Define Choristoma

A

Is the term applied to at heterotopic (misplaced) rest of cells. Is not a neoplasm e.g. normally organised pancreatic tissue in the submucosa of the stomach.

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

What are six morphological changes that cancer cells can exhibit?
(Remembering there is a spectrum and not all show all)

A
  • Anaplasia
  • Pleomorphism (note tumour giant cells)
  • Abnormal nuclear morphology (e.g. disproportionately large)
  • Mitoses (most importantly atypical, bizarre mitotic figures)
  • Loss of polarity
  • Other changes (e.g. areas of ischemic necrosis)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Explain the differences between metaplasia, dysplasia and carcinoma in situ

A
  • Metaplasia is the replacement of one cell type to another. Usually in association with tissue damage, repair and regeneration. Prone to malignant transformation
  • Dysplasia is disordered growth with the affected area showing changes like those of cancer. It is a precursor to malignant transformation but does not always progress to cancer. With removal of inciting causes it may be completely reversible.
  • Carcinoma in situ is when dysplasia is severe and involves the full thickness of epithelium but does not penetrate the basement membrane. Unless treated have a high probability of progression to invasive cancers.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Next to the development of metastases, what is the most reliable discriminator of malignant and benign tumours?

A

Invasiveness

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

Why do most benign tumours develop a capsule?

A

Because they grow and expand slowly. Stromal cells and fibroblasts deposit ECM following activation by hypoxic damage due to the expanding tumour.

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

What three pathways can dissemination of cancers occur through?

A
  • Seeding of body cavities and surfaces
  • Lymphatic spread (common with carcinomas)
  • Hematogenous spread (common with sarcomas)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

List seven of the best established environmental factors affecting cancer risk

A
  • Infectious agents
  • Smoking
  • Alcohol consumption
  • Diet
  • Obesity
  • Reproductive history
  • Environmental carcinogens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What are three types of acquired predisposing conditions to developing cancer?

A
  • Chronic inflammation
  • Precursor lesions (hyperplasia, metaplasia, dysplasia and some benign neoplasms)
  • Immunodeficiency
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What four epidemiological factors are thought to contribute to the development of cancer?

A
  • Environmental factors
  • Age
  • Acquired predisposing conditions
  • Genetic predisposition
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What are the four classes of genes that are the principal targets for cancer causing mutations?

A
  • Proto-oncogenes
  • Tumour suppressor genes
  • Genes that regulate programmed cell death
  • Genes that are responsible for DNA repair
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What are the eight hallmarks of cancer?

A
  • Self-sufficiency in growth signals
  • Insensitivity to growth-inhibitory signals
  • Altered cellular metabolism (aerobic glycolysis)
  • Evasion of apoptosis
  • Limitless replicative potential
  • Sustained angiogenesis
  • Ability to invade and metastasise
  • Ability to evade host immune response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What are oncogenes?

A

Mutated genes that cause excessive cell growth, even in the absence of growth factors and other growth-promoting external cues

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

For the following three receptor tyrosine kinase genes, describe the effect of the mutation that tends to occur allowing constitutive activity in certain tumours:
1. ERBB1
2. ERBB2
3. ALK

A
  1. Point mutations in lung adenocarcinomas produce constitutive activation of EGF
  2. Gene Amplification leading to over-expression of HER2
  3. Constitutively active form created through gene rearrangement e.g. a deletion on ch 5 can lead to EML4-ALK fusion gene
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What kind of mutation involving what family of genes is the most common type of abnormality involving proto-oncogenes in human tumours?

A

Point mutations of RAS (HRAS, KRAS, NRAS)

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

In the receptor tyrosine kinase receptor signalling pathway, what two kinase families downstream of RAS can also frequently be involved in oncogenic gain of function mutations?

A
  • RAF e.g. BRAF in hairy cell leukemias
  • PI3K e.g. in 30% of breast carcinomas
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What is the classic example of an oncogenic mutation in nonreceptor tyrosine kinases (they activate same signalling pathways as RTKs)?

A

Translocation of ABL gene (for ABL TK) from ch 9 to ch22 where it fuses with BCR, BCR drives self association, resulting in CML

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

What is oncogene addiction?

A

When tumour cells are highly dependent on the activity of one oncoprotein. e.g. in CML BCR-ABL inhibitors work very well (but do not cure as some CML “stem cells” persist).

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

MYC is a transcription factor regularly involved in cancers. It’s activation is common to most growth signalling pathways, it can also be directly affected by mutation as in Burkitt lymphoma where it is translocated from ch 8 to 14 where is fuses with an IG gene, leading to the enhancers instead enhancing MYC.
What are three activities of MYC that show how it contributes to multiple hallmarks of cancer?

A
  • Activated expression of many genes involved in cell growth e.g D cyclins, rRNA and those that lead to metabolic reprogramming
  • In some contexts upregulates expression of telomerase
  • Can act together with a handful of other transcription factors to reprogram somatic cells into pluripotent stem cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What are the four main groups of proto-oncogenes?

A

Growth factors, growth factor receptors and subsequent pathway protiens, transcription factors, and cyclins and cyclin-dependent kinases

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

In the cell cycle the G1/S checkpoint is more important in cancer because as well as leading to dysregulated growth it can impair DNA repair. What are the most frequently occurring oncogenes for this stage?

A

D cyclin and CDK4

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

Loss of normal cell cycle control is central to malignant transformation and at least one of the four key regulators of the cell cycle is dysregulated in the vast majority of human cancers (whether via direct mutation or upstream mutation).
What are the four key regulators?

A

p16/INK4a, cyclin D, CDK4, RB

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

RB is a key negative regulator of the G1/S cell cycle transition. In quiescent cells it exists in an active hypophosphorylated state in complex with E2F transcription factors, preventing them from activating transcription of S-phase genes. In what two ways can its function be compromised?

A
  • Loss of function mutations involving both RB alleles (or viral oncoproteins that bind and inhibit RB)
  • A shift to the inactive hyperphosphorylated state caused by gain of function mutations upregulating CDK/cyclin D activity or by loss of function mutations of CDK inhibitors (p16/INK4a)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What is the most frequently mutated gene in human cancers and what is its general role?

A

TP53 - regulates cell cycle progression, DNA repair, cellular senescence and apoptosis

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

MDM2 is an enzyme that ubiquitinates p53 in the normal cell, making it virtually non-existent. What are the two major mechanisms by which p53 is released from this inhibition?

A
  • Through phosphorylation of MDM2 and p53 stimulated by one of the protein kinases ATM or ATR in response to DNA damage and hypoxia
  • Binding of p14/ARF to MDM2 displacing p53 due to increased expression triggered by signalling from oncoproteins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q
  1. How does p53 induce transient cell cycle arrest and 2. assist in facilitating DNA repair?
A
  1. By inducing transcription of CDKN1A, producing p21 which inhibits CDK4/D cyclin complexes
  2. By inducing proteins such as GADD45 (growth arrest and dna damage) that enhance DNA repair
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Why does a cancer’s p53 status have important therapeutic implications?

A

Irradiation and conventional chemotherapy mediate their effects by inducing DNA damage and subsequent apoptosis. Therefore tumours with wild-type p53 alleles are more likely to be killed by such therapy than those with mutant p53 alleles.
Also, with mutant p53 means has a mutator phenotype, increasing the chances of resistant subclones to any therapy

29
Q

B-catenin is a proto-oncogene. What are two mechanisms by which it is normally allowed to translocate to the nucleus, stimulating genes like TCF that promote proliferation?

A
  • Through WNT signalling (an embryonic development pathway) which blocks the formation of a destruction complex initiated by the tumour-suppressor protein APC (adenomatous polyposis coli)
  • Through the disassociation from E-Cadherin that occurs when cell-cell contact is lost such as in epithelial injury.
30
Q

What triggers metabolic reprogramming to aerobic glycolysis (the Warburg effect)?

A

Signalling cascades downstream of growth factor receptors. e.g. RTK/PI3K/AKT signalling and MYC upregulation

31
Q

What is oncometabolism, and an example of an oncoprotein and its oncometabolite?

A

When oncometabolites are produced by mutated enzymes which alter the epigenome leading to oncogenic changes in gene expression.
Isocitrate dehydrogenase (IDH) of the krebs cycle can mutate to catalyse the production of the oncometabolite 2-hydroxyglutarate (2-HG)

32
Q

Tumour cells frequently contain mutations in genes that result in resistance to apoptotic cell death-particularly the intrinsic pathway. What are the two major mechanisms by which apoptosis is avoided by cancer cells?

A
  • Loss of TP53 function (as prevents upregulation of the pro-apoptotic protein PUMA in response to DNA damage and other stressors)
  • Overexpression of anti-apoptotic members of the BCL2 family such as in follicular B-cell lymphomas where BCL2 is overexpressed due to a 14;18 translocation
33
Q

What three interrelated factors appear to be critical to the immortality of cancer cells?

A

Evasion of senescence, evasion of mitotic crisis and self-renewal

34
Q

For a tumour to grow beyond 1-2mm it needs to be able to induce angiogenesis. What are some alterations that enhance the production of pro-angiogenic factors?

A
  • Relative lack of oxygen due to hypoxia. This stabilises HIF1a, a transcription factor that activates the transcription of VEGF and bFGF
  • Driver mutations in certain tumour suppressors and oncogenes favour angiogenesis. e.g. p54 induces the inhibitor thombospondin-1 and represses the likes of VEGF while RAS, MYC and MAPK upregulate VEGF
  • Proteases. Influence local balance of anti and pro angiogenesis factors e.g. by releasing them from the ECM
35
Q

Epithelial-mesenchymal transition (EMT) is controlled by the transcription factors SNAIL and TWIST. What is it believed that this process entails?

A

Downregulation of epithelial markers (e.g. E-cadherin) and upregulation of mesenchymal markers (e.g. vimentin and smooth muscle actin). This is believed to favour development of a promigratory phenotype. (for metastasis)

36
Q

What are the four steps of metastases invasion of the ECM (the first phase of metastasis)

A
  1. Dissociation of cancer cells from one another
  2. Degradation of the basement membrane and interstitial connective tissue (e.g. MMP-9)
  3. Attachment to “remodelled” ECM
  4. Locomotion
37
Q

What three types of factors may be involved in movement of metastatic cells through the ECM?

A
  • Tumour cell-derived cytokines (acting as autocrine motility factors)
  • Cleavage products of matrix components
  • Stromal cell-derived paracrine factors e.g. hepatocyte growth factor/scatter factor which binds to the RTK MET and stimulates motility
38
Q

What three factors appear to relate to where circulating tumour cells arrest and eventually form clinically significant metastatic deposits?

A
  • Location and vascular drainage of the primary tumour
  • Tropism of particular kinds of tumour cells for specific tissues
  • Escape from tumour dormancy
39
Q

What are three groups of protein antigens in tumours that can elicit CD8+ cytotoxic T-cell responses?

A
  • Neoantigens produced from genes bearing passenger and driver mutations
  • Overexpressed (e.g. tyrosinase in melanoma) or aberrantly expressed (e.g. cancer-testis antigens and MAGE family proteins) normal cellular proteins
  • Tumour antigens produced by oncogenic viruses
40
Q

How are APCs able to activate CD8+ T cells in lymphoid tissue, allowing migration to the affected tissue?

A

Through a mechanism called cross-presentation where APCs present the antigen in the context of MHC I molecules (as well as MHC II) with costimulatory molecules

41
Q

What are five mechanisms that can allow tumour cells to evade the immune system?

A
  • Selective outgrowth of antigen-negative variants
  • Loss or reduced expression of MHC molecules (this only stops T cells)
  • Engagement of pathways that inhibit T-cell activation
  • Secretion of immunosuppressive factors e.g. TGF-B
  • Induction of regulatory T cells
42
Q

What are two examples of pathways tumour cells can engage to inhibit T cell activation? (Immune checkpoints)

A
  • Block costimulatory CD28 B7 binding by promoting expression of CTLA-4 on tumour specific T cells (binds and blocks B7 from CD28)
  • Upregulation of PD-L1 and PD-L2 cell surface proteins (on the tumour cell). These activate PD-1 receptors on effector T cells, inhibiting activation.
43
Q

What is an example of a cancer syndrome caused by inheritance of an abnormal copy of a mismatch repair gene?
(side note - hallmark of mismatch-repair defects is microsatellite instability)

A

Hereditary nonpolyposis colon cancer (HNPCC) syndrome. Most commonly affected genes are MSH2 and MLH1

44
Q

1.What kind of DNA damage is repaired by the nucleotide excision repair system?
2. What syndrome can mutation in any of the genes in the above system cause?

A
  1. Cross-linking of pyrimidine residues preventing normal DNA replication, caused by UV radiation.
  2. Xeroderma pigmentosum
45
Q

What are five factors that if affected by mutation could cause genomic instability?

A
  • DNA mismatch repair factors
  • Nucleotide excision repair factors
  • Homologous recombination repair factors
  • DNA polymerase- specifically the ‘proofreading’ function (most heavily mutated of all human cancers)
  • Regulated genomic instability in lymphoid cells (receptors gene recombination and class switching)
46
Q

Chromosomal translocations are the most common mechanism for activating oncogenes. What are the two ways in which they can do this, and an example of each?

A
  • Promotor or enhancer substitution (proto-oncogene one swapped with another, typically highly expressed gene’s one) e.g. Burkitt lymphoma MYC
  • Fusion gene formation leading to expression of a novel chimeric protein with oncogenic properties e.g. CML BCR-ABL
47
Q

What is the first example of differentiation therapy? (In which immortal tumour cells are induced to differentiate into their mature, limited lifespan, progeny)

A

All-trans retinoic acid for acute promyelocytic leukemia. (Binds to chimeric oncoprotein PML-RARA causing it and recruited repressor complexes to disassociate from DNA, allowing differentiation to neutrophils).

48
Q

What are two tumour suppressor genes commonly affected by chromosomal deletions in malignancy and their associated malignancies?

A
  • RB - retinoblastoma
  • VHL - renal cell carcinomas
49
Q

Clinically, what are the two most important gene amplifications and their associated malignancies?

A
  • NMYC - Neuroblastoma
  • ERBB2 - Breast cancer
50
Q

What three categories can epigenetic changes in malignancy fall under?

A
  • Silencing of tumour suppressor genes by local hypermethylation of DNA
  • Global changes in DNA methylation
  • Changes in histones
51
Q

What is the best characterised non-coding RNA that plays a role in carcinogenesis and how?

A

microRNAs. onco-miRs by suppressing tumour suppressor genes if overexpressed, or tumour suppressor function lost by loss of the miRNA allowing over-expression of a protooncogene

52
Q

Define 1. initiation and 2. promotion in the context of carcinogenesis

A
  1. Results from exposure of cells to a sufficient dose of a carcinogenic agent causing permanent DNA damage
  2. Induction of tumours to arise from initiated cells, not tumourgenic in isolation,
53
Q

Of the following direct acting carcinogens, which are alkylating agents and which are acylating agents?
- I-Acetyl-imidazole
- Diepoxybutane
- Anticancer drugs (cyclophosphamide, chlorambucil, nirtosoureas etc)
- Dimethyl sulphate
- Dimethylcarbamyl chloride
- B-Propiolactone

A
  • Alkylating agents: Diepoxybutane, Anticancer drugs (cyclophosphamide, chlorambucil, nirtosoureas etc), Dimethyl sulphate, B-Propiolactone
  • Acylating agents: I-Acetyl-imidazole, Dimethylcarbamyl chloride
54
Q

Two of some of the most potent indirect chemical carcinogens are metabolised by CYP1A1, of which 10% of the European population carry a highly inducible form significantly increasing the risk of cancer development on exposure to these carcinogens. What are they and their main sources?

A
  • polycyclic hydrocarbons, fossil fuels, tobacco smoking
  • benzo(a)pyrene soot, tobacco smoking
55
Q

Chemical carcinogens usually damage DNA (due to having highly reactive electophile groups) in a “hotspot” fashion depending on chemical make up (so kinda random but not). What is an example of a mutation almost exclusively related to exposure to a particular carcinogen?

A

G:C -> T:A (arginine->serine) in codon 249 of TP53 in hepatocellular carcinomas associated with aflatoxin-B1 (from aspergillus contaminated food)

56
Q

What is the name of the only human retrovirus implicated in the pathogenesis of human cancer (only 5% of infected 40-60 years later), and what are two of it’s genes suspected to be important for this?

A

Human T-Cell Leukaemia Virus Type 1 (HTLV-1) . Tax (stimulates transcription from 5’) and HBZ (a transcription factor)

57
Q

What are the five identified oncogenic DNA viruses?

A

HPV, EBV, HBV, Merkel cell polyomavirus and HHV8

58
Q

The high risk for carcinogenesis HPV types are 16, 18 and 31. What makes these variants higher risk than others?

A
  • Integrate with host genome
  • Increased expression of HPV E6 and E7 genes due to loss of E2 in integration
  • E6 higher affinity for p53 (promotes degradation) note also stimulates TERT
  • E7 higher affinity for RB (displacing E2F promoting cell cycle) and binds and presumably activated cyclins A and E. note also inactivates CDK inhibitors p21 and 27.
59
Q

What virus is associated with a number of cancer forms, enters B cells by binding the CD21 receptor, makes infected B cells immortal, however is easily found and destroyed by T cells in most cases, and is thought to “set the stage” for a translocation up-regulating MYC and other mutations ultimately producing a full blown cancer?

A

Epstein-Barr Virus

60
Q

HBV and HCV worldwide are associated with 70-85% of hepatocellular carcinomas. What is thought to be the dominant oncogenic effect?

A

Immunologically mediated chronic inflammation and hepatocyte death leading to proliferation and over time, genomic damage

61
Q

What two malignancies is Helicobacter pylori implicated in the development of?

A

Gastric adenocarcinomas and lymphomas

62
Q

Define grading and staging of tumours

A
  • Grading - Based on the degree of differentiation of the tumour cells and in some cancers the number of mitoses or architectural features.
  • Staging - Based on the size of the primary lesion (T), whether it has spread to regional lymph nodes (N) and the presence or absence of blood-borne metastases (M)
63
Q

What are the limits of histology and exfoliative cytology in cancer diagnosis?

A

Can be difficult to determine the nature of the tissue if origin in a poorly differentiated tumour and specific tumour types are difficult to distinguish based on morphologic appearance alone (e.g. various acute leukemias and lymphomas)

64
Q

What are five types of specimens commonly examined in cytologic smears for cancer cells?

A

Cervical, urine, cerebrospinal fluid, pleural effusions and bronchial washes

65
Q

In immunohistochemistry use of specific antibodies assists in categorising undifferentiated malignancies. For example, the presence of what intermediate filaments indicate that the malignancy is of 1. epithelial origin, 2. muscle cell origin or 3. haematologic origin

A
  1. Cytokeratins
  2. Desmin
  3. Nil
66
Q

What are three examples of the utility of immunohistochemistry in the the diagnosis or management of malignant neoplasms?

A
  • Categorisation of undifferentiated malignant tumours
  • Determination of site of origin of metastatic tumours (e.g. PSA for prostate)
  • Detection of molecules that have prognostic or therapeutic significance (e.g. oestrogen receptors in breast cancer)
67
Q

Flow cytometry requires viable cells in suspension. It is mainly used to identify cellular antigens expressed by “liquid” tumours (arise from blood forming tissues). What is an advantage of flow cytometry over immunohistochemistry?

A

Multiple antigens are assessed simultaneously on individual cells using combinations of specific antibodies linked to different fluorescent dyes.

68
Q

What are six uses of molecular diagnostics and cytogenetics (PCR, FISH, DNA microarrays etc) in malignancy?

A
  • Diagnosis of malignant neoplasms (e.g. PCR for reactive (polyclonal) vs neoplastic (monoclonal) lymphoid proliferations)
  • Prognosis of malignant neoplasms
  • Detection of minimal residual disease (currently only used in acute leukaemia)
  • Diagnosis of hereditary predisposition to cancer
  • Guiding therapy with oncoprotein-directed drugs
  • Identifying mechanisms of drug resistance: liquid biopsies
69
Q

List six tumour makers with their associated tumour types

A
  • Prostate specific antigen (PSA)- prostate adenocarcinoma
  • Carcinoembryonic antigen (CEA) - carcinomas of colon, pancreas, stomach, lung, heart and breast
  • Alpha-Fetoprotien (AFP) - Hepatocellular carcinomas, yolk sac tumours
  • Human chorionic gonadotropin (HCG) - testicular tumours
  • CA-125 - Ovarian tumours
  • Monoclonal immunoglobulins - multiple myeloma and other gammopathies