Chapter 7: Neoplasia Flashcards

1
Q

What kind of tumor is derived from glands and applied to benign epithelial neoplasms?

A

Adenoma

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

What benign epithelial neoplasm has visible finger like or warty projections?

A

Papilloma

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

A polyp with glandular tissue is called what?

A

Adenomatous polyp

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

Malignant tumors arising from solid mesenchymal tissues are called what?

A

Sarcoma

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

Malignant neoplasms of epithelial cell origin, derived from any of the 3 germ layers, are called what?

A

Carcinomas

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

What is the most common mixed tumor?

Capable of producing what and is designated?

A
  • Salivary gland mixed tumor
  • Epithelial and myoepithelial cells = pleomorphic adenoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What is the term applied to a heterotopic rest of cells?

i.e., a small nodule of well-developed and normally organized pancreatic tissue found in the submucosa of the stomach, duodenum, or SI.

A

Choristoma

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

Lack of differentiation, or anaplasia, is considered a hallmark of?

A

Malignancy

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

What refers to variation of size and shape of cancer cells?

A

Pleomorphism

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

What best describes carcinoma in situ?

A
  • Dysplastic changes are marked and involve the full thickness of the epithelium
  • Lesions DO NOT penetrate the BM
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What malignant cancers invade early but rarely metastasize?

A
  • Gliomas
  • Basal cell carcinoma of the skin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Once tumor cells breach the basement membrane they are said to be?

A

Invasive

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

How is dysplasia related to malignant transformation?

Is all metastatic epithelium dysplastic?

A
  • May be a precursor to malignant transformation
  • Does NOT always progress to cancer
  • Not all metastatic epithelium is dysplastic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What are the 3 pathways of spread of cancer?

A

1) Direct seeding of body cavities or surfaces
2) Lymphatic spread
3) Hematogenous spread

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

In direct seeding of body cavities, sometimes appendiceal carcinomas or ovarian carcinomas fill the peritoneal cavity with what?

A

Pseudomyxoma peritonei

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

What is the most common pathway for the initial dissemination of carcinomas?

A

Lymphatic spread

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

Where do carcinomas of the breast in the upper outer quadrant disseminate 1st to?

Inner quadrants?

A
  • Axillary LNs
  • LNs along internal mammary arteries
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Where do carcinomas of the lung in the major respiratory passages metastasize first to?

A

Perihilar tracheobronchial and mediastinal nodes

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

Renal cell carcinoma invades what structure?

A

Renal vein —> IVC —-> right side of the heart (sometimes)

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

Hepatocellular carcinoma often penetrates what?

A

Portal and hepatic radicles

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

Breast carcinoma preferentially spreads to where?

A

Bone

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

Bronchogenic carcinoma preferentially spreads to where?

A

Adrenals and brain

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

Neuroblastoma preferentially spreads to where?

A

Liver and bones

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

To avoid the considerable surgical morbidity associated with full LN dissection, which technique is often used to assess the presence of metastatic lesions in the LNs?

A
  • Biopsy of sentinel nodes

*The first node in a regional lymphatic basin that receives lymph flow from the primary tumor

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

Which organs are most frequently involved in hematogenous dissemination of cancer?

A

Liver and lungs

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

Describe the following for benign cancers:

  • Differentiation
  • Rate of growth
  • Local invasion
  • Metastasis
A
  • Well differentiated; structure sometimes typical of tissue of origin
  • Progressive and slow growth
  • Cohesive, expansile, well-demarcated masses that do not invade
  • Absent metastasis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Describe the following for malignant cancers:

  • Differentiation
  • Rate of growth
  • Local invasion
  • Metastasis
A
  • Lack differentiation (anaplasia); structure often atypical
  • Erratic, slow or rapid growth
  • Locally invasive
  • Frequent metastasis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Cancers arising in close proximity to the vertebral column often embolize through where?

This pathway is involved in the frequent vertebral metastases of carcinomas of?

A
  • Paravertebral plexus
  • Thyroid and prostate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What are the most common tumors in Men?

Woman?

A

Men = prostate, lung, and colon

Woman = breast, lung and colon

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

What is responsible for a large majority of cervical carcinoma and increasing fraction of head and neck cancers?

A

HPV

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

What cancer is associated with benzene (i.e., light oil, paint, printing, dry cleaning, adhsives. etc.)?

A

Acute myeloid leukemia

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

What cancer is associated with beryllium (i.e., missle fuel, space vehicles)?

A

Lung carcinoma

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

What cancer is associated with cadmium (i.e., yellow pigments and phosphors; used in batteries, etc.)?

A

Prostate carcinoma

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

What cancer is associated with vinyl chloride (i.e., refrigerant, vinyl polymers, plastic adhesives, etc.)?

A

Hepatic angiosarcoma

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

What cancer is associated with chromium (i.e., metal alloys, paints, pigments, and preservatives)?

A

Lung carcinoma

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

What cancers are associated with nickel?

A

Lung and oropharyngeal carcinoma

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

What cancers are associated with Arsenic?

A

Lung carcinoma

Skin carcinoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What is the etiologic agent of osteomyelitis?

What neoplasm?

A
  • Bacterial infection
  • Carcinoma in draining sinuses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

What is the etiologic agent of Chronic Cystitis?

What neoplasm?

A
  • Schistosomiasis
  • Bladder carcinoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What is the associated neoplasm of Sjorgen syndrome and Hashimoto thyroiditis?

A

MALT lymphoma

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

What is the classic example of a benign, neoplastic, precursor lesion?

A

Colonic villous adenoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What are 2 examples of epigenetic modifications that contribute to the malignant properties of cancer cells?

A

1) DNA methylation: silencing of tumor suppressor genes
2) Histone modification

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

What are the 4 classes of regulatory genes that are principal targets of cancer-causing mutations?

A

1) Growth-promotong proto-oncogenes
2) Growth-inhibiting tumor suppressor genes
3) Genes that regulate apoptosis
4) DNA-repair genes

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

What are the 8 fundamental changes in cell physiology, which are considered the hallmarks of cancer?

A

1) Self-sufficiency in growth signals
2) Insensitivity to growth-inhibitory signals
3) Altered cellular metabolism
4) Evasion of apoptosis
5) Limitless replicative potential (immortality?)
6) Sustained angiogenesis
7) Ability to invade and metastasize
8) Ability to evade host immune response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

Define Oncoprotein

A

A protein encoded by an oncogene that drives increased cell proliferation through one of several mechanisms

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

What is MYC known as and what are its important functions?

A
  • The master transcription factor that regulates genes needed for rapid cell growth by deregulation through chromosomal translocations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

90% of what type of cancer contains RAS mutations?

A

Pancreatic adenocarcinomas

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

What is the major function of CDK4; D cyclins?

A

Form a complex that phosphorylates RB, allowing the cell to progress through the G1 restriction point

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

What is the major function of the cell cycle component, RB?

A
  • Tumor suppressive “pocket” protein that binds E2F transcription factors in its hypophosophorylated state, prevents G1/S transition
  • Interacts with several transcription factors that regulate differentiation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What induces the cell cycle inhibitor p21?

A

p21 is induced by tumor suppressor p53

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

What responds to growth suppressors such as TGF-B?

A

p27

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

What chromosome is p53 located on?

Acts mainly through what?

What negatively regulates p53?

A
  • Chromosome 17
  • Acts mainly through p21
  • Negatively regulated by MDM2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What inhibits MDM2 activity?

What does this do?

A
  • p14
  • Increases p53 activity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

What specifically binds to cyclinD-CDK4, blocking CDK4/cyclin D-mediated phosphorylation of RB; thereby reinforcing the RB checkpoint?

A

p16

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

What occurs in the familial form of retinoblastoma?

Inherited how?

A
  • All somatic cells inherit one mutated copy of RB gene from a carrier parent (first hit)
  • Only one additional RB mutation in a retinal cell is required for complete loss of RB function (second hit)
  • Autosomal dominant
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

What occurs in sporadic cases of Retinoblastoma?

A
  • Both normal RB alleles must undergo somatic mutation in the same retinoblast (two hits)
  • Probability of this event is low (explaining why this tumor is uncommon in th general population)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

What is the “two-hit” hypothesis of oncogenesis as it relates to retinoblastoma?

Which chromosome involved?

A
  • Two mutations (hits) involving both alleles of RB at chromosome locus 13q14 are required to produce retinoblastoma
  • In familial cases, children inherit one defective copy of the RB gene in the germline (first hit), and the other copy is normal. Retinoblastoma develops when the normal RB allele is mutated in retinoblasts as a result of spontaneous somatic mutation (second hit)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

What are the key initiators of p53 activity following DNA damage or cellular exposure to hypoxia?

How do they act; what is p53 released from?

A
  • ATM/ATR family
  • These kinases phosphorylate p53, liberating it from inhibitors such as MDM2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

What affect does the active hypophosphorylated RB in complex with E2F transcription factors have?

A
  • Binds DNA, recruits chromatin-remodeling factors (histone deacetylases and histone methyltransferases)
  • Inhibits transcription of genes who products are required for the S phase of the cell cycle
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

What phosphorylates RB (inactivating it)?

What occurs when RB is phosphorylated?

A
  • Phosphorylated by cyclin D-CDK4 and CDK6; also cyclin E-CDK2
  • Causes RB to release E2F
  • E2F is then able to activate transcription of S-phase genes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

Virtually all cancer cells show dysregulation of the G1-S checkpoint as a result of mutation in one of four genes that regulate the phosphorylation of RB; what are these genes?

A
  • RB (loss of function mutations)
  • CDK4 (gene amplifications)
  • -* Genes encoding cyclin D proteins (gene amplifications)
  • CDKN2A (p16): loss of cyclin-dependent kinase inhibitors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

Which GFs activate Cyclins D/CDK4,6 and Cyclin E/CDK2 leading to hyperphosphorylated RB (inactive)?

A

EGF and PDGF

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

Explain the interaction between the E7 protein of HPV and RB, where does it bind?

Which form of HPV is higher risk and why?

A
  • Binds to hypophosphorylated form of RB in the same pocket which normally sequesters E2F transcription factors
  • HPV16 confers a high risk for development of cervical carcinoma and expresses E7 protein variants with higher affinity for RB than do lower risk viral types
  • RB protein is unable to bind E2F transcription factors and is functionally inactivated by these viral oncoproteins, and the E2F factors are free to cause cell cycle progression
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

Normal GF signaling leads to RB _______ and __________, thus promoting cell cycle progression

A

Normal GF signaling leads to RB hyperphosphorylation and inactivation, thus promoting cell cycle progression

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

Tumors with wild type or mutated TP53 alleles are more susceptible to chemotherapy?

What kind of cancers?

A
  • Wildtype
  • Testicular Teratocarcinomas and Childhood Acute Lymphoblastic Leukemia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

Activation of normal p53 by DNA-damaging agents or by hypoxia leads to cell cycle arrest in which phase?

Causes induction of DNA repair by transcriptional upregulation of what?

A
  • Arrest in G1
  • Upregulation of cyclin-dependent kinase inhibitor, CDKN1A (encoding p21): inhibitor of CDK4/D cyclin
  • Upregulation of GADD45 genes (DNA repair)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

If DNA repair fails, what does p53 trigger?

A
  • Apoptosis (through BAX and PUMA)
  • Senescence
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

Mutated TP53 alleles are mostly resistant to chemotherapy and irradiation and include what cancers?

A

Lung and colorectal cancers

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What do cells with defective p53 acquire and what does this result in?

A
  • Mutator phenotype
  • Tendency to acquire additional mutations at a higher rate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

Rare patients with Li-Fraumeni syndrome inherit how many defective copies of TP53?

A

Inherit one defective copy of TP53 and have a very high incidence of a wide variety of cancers

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

The majority of human cancers demonstrate ________ mutations in TP53

A

The majority of human cancers demonstrate biallelic loss-of-function mutations in TP53

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

Like RB, p53 can also be inactivated by?

A

Viral oncoproteins, such as the E6 protein of HPV

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

What is the “gatekeeper of colonic neoplasia?”

Functions by?

A
  • Adenomatous polyposis coli (APC)
  • Downregulates growth-promoting signaling pathways
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

Germline loss-of-function mutations involving APC (5q21) locus are associated with what disorder?

Inheritance pattern?

A
  • Familial Adenomatous Polyposis = Autosomal Dominant
  • Individuals born w/ one mutant allele develop thousands of adenomatous polyps in the colon during their teens or 20s
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

What is the role of APC in regulating the stability and function of β-catenin in both resting and WNT activated colonic epithelial cells?

Forms what?

What does β-catenin do?

A
  • In resting colonic epithelial cells (not exposed to WNT), β-catenin forms macromolecular complex containing the APC protein. This complex leads to destruction of β-catenin, and intracellular levles are low
  • When stimulated by WNT, the destruction complex is deactivated, β-catenin degradation does not occur, and cytoplasmic levels increase
  • β-catenin translocates to the nucleus, where it binds to TCF, a TF that activates genes involved in cell cycle progression and proliferation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

Germline loss-of-function mutations of the von Hippel-Lindau (VHL) gene on chromosome 3p are associated with what?

A
  • Hereditary renal cell cancers
  • Pheochromocytomas
  • Hemangioblastomas of the CNS
  • Retinal angiomas
  • Renal cysts
77
Q

When APC is mutated or absent, as frequently occurs in colonic polyps and cancers, what happens to β-catenin?

How do they cells behave?

A
  • The destruction of β-catenin cannot occur.
  • β-catenin translocates to nucleus and coactivates genes that promote entry into the cell cycle and cells behave as if they are under constant stimulation by the WNT pathway
78
Q

VHL encodes what, which is reponsible for?

How does this function in oxygen rich and hypoxic enviornments?

A
  • Encodes a component of a ubiquitin ligase that is responsible for the degradation of hypoxia-induced factors (HIFs)
  • In presence of oxygen, HIF1α is hydroxylated and binds to VHL, leading to its ubiquitination and degradation
  • In hypoxia the hydroxylation rxn does NOT occur and HIF1α escapes recognition by VHL and accumulates in nuclei of hypoxic cells turning on genes encoding VEGF, PDGF, and GLUT1
79
Q

In chronic myelogenous leukemia (CML) and some acute lymphoblastic leukemias (ALL), which translocation occurs?

What does this fusion create?

A
  • ABL gene is translocated from chromosome 9 to chromosome 22 where it fuses with the BCR gene
  • Encodes a constituitively active, oncogenic BCR-ABL tyrosine kinase
80
Q

Which portion of the BCR-ABL fusion has the tyrosine kinase activity?

What is the function of the other gene?

A
  • ABL has tyrosine kinase activity
  • BCR drives self association of BCR-ABL and unleashes activity of ABL
81
Q

What 6 ways are tumor cells able to evade cell death by apoptosis?

A

1) Loss of p53 –> reducedfunction of pro-apoptotic factors such asBAX
2) Reduced egress of cytochrome c as a result of upregulation of anti-apoptotic factors such as BCL2, BCL-XL, and MCL-1
3) Loss of apoptotic peptidase activating factor 1 (APAF1)
4) Upregulation of inhibitors of apoptosis (IAP)
5) Reduced CD95 level
6) Inactivation of death-induced signaling complex FADD, needed for Fas-FasL

82
Q

Which pathway of apoptosis is most commonly affected in cancer cells?

A

Intrinsic (mitochondrial)

83
Q

In >85% of follicular B-cell lymphomas, what gene is overexpressed?

Due to what translocation?

A
  • Anti-apoptotic gene BCL2
  • (14;18) translocation
84
Q

What are the most common general category of tumor in adults?

Children?

A

Adults = Carcinomas

Children = Acute Leukemia and distinctive neoplasms of CNS

85
Q

What is an abundant collagenous stroma called?

How can this also be described?

A
  • Desmoplasia
  • Scirrhous
86
Q

What chromosome is the RB gene located?

How is it inherited?

A
  • Chromosome 13
  • Autosomal dominant
87
Q

What cancer is specific to a mutation in BRCA2?

A

Chronic lymphocytic leukemia

88
Q

What is the function of GADD45?

What induces it?

A
  • DNA repair
  • p53
89
Q

What does CDKN2A activate?

A

p16 and p14

90
Q

What is a novel gene expression assay that is being used to predict prognosis and treatment response in cancer patients?

A
  • Expression of CEN/KT (centromere and kinetochore) genes
  • Centromere and kinetochore gene expression score (CES) signature
91
Q

How does the Warburg effect work (3 signaling mechanisms)?

A

1) PI3K/AKT signaling
2) RTK activity
3) MYC

92
Q

What opposes the actions of the Warburg effect via blockage of PI3K/AKT signaling?

A

PTEN

93
Q

Mutation of PTEN results in what?

Located on what chromosome?

A
  • Cowden syndrome
  • Chromosome 10
94
Q

Hypoxia triggers angiogenesis through the actions of?

A

HIF1α on the transcription of the proangiogenic factor VEGF

95
Q

How does p53 regulate angiogenesis?

RAS, MYC, and MAPK?

A
  • p53 Induces synthesis of the angiogenesis inhibitor thrombospondin-1
  • RAS, MYC, MAPK all upregulate VEGF expression and stimulate angiogenesis
96
Q

Invasion of the ECM, a hallmark of malignancy, occurs in what 4 steps?

A

1) Loosening of cell-cell contacts
2) Degradation of ECM
3) Attachment to novel ECM components
4) Migration and invasion of tumor cells

97
Q

Via which enzymes and factors are tumor cells able to degrade the basement membrane and interstitial CT during the second step of invasion?

A
  • Tumor cells may secrete proteolytic enzymes themselves
  • May induce stromal cells (i.e., fibroblasts and inflammatory cells) to elaborate proteases, such as MMP, Cathepsin D, and Urokinase plasminogen activator
98
Q

What is loss of adhesion to the BM called?

Triggers what?

A
  • Anoikis
  • Apoptosis
99
Q

Solid tumors and normal T lymphocytes is use what to spread to LNs and other metastatic sites?

A
  • Binding of CD44 to hyaluronate on high endothelial venules
  • Solid tumors express CD44, which appears to enhance their spread to LNs and other metastatic sites
100
Q

What mechanisms may account for the organ tropism exhibited by tumor cells and why metastasis sometimes occurs in locations unrelated to the initial site of the tumor?

A
  • Tumor cells may have adhesion molecules whose ligands are expressed preferentially on the endothelial cells of the target organ
  • Chemokines may play an important role. Some breast cancer cells express the chemokine receptors CXCR4 and CCR7
  • In some cases, the target tissue may be a nonpermissive enviornment - “unfavorable soil”
101
Q

The prolonged survival of micrometastases without progression (dormancy) like in breast cancer metastases to bone is accomplished how?

A
  • Breast cancer cells secrete PTHRP, which stimulates osteoblasts to make RANKL
  • RANKL activates osteoclasts, which degrade the bone matrix and release GFs embedded within, like IGF and TGF-B
  • GFs help make the metastatic site habitable for the cancer cells.
102
Q

What protein is often found overexpressed in drug-resistant cancers?

A

MCL-1

103
Q

What areas of the body rarely have metastasis?

Why?

A
  • Spleen and Skeletal muscle
  • Represent “unfavorable soil”
104
Q

What are the main pro angiogenic factors?

GOF mutations in what may upregulate them?

A
  • VEGF, bFGF
  • GOF mutations in RAS or MYC
105
Q

What are the oncofetal antigens whose expression at high levels serve as markers that aid in tumor diagnosis and clinical management?

A
  • CEA (carcinoembryonic antigen)
  • AFP (α-fetoprotein)
106
Q

The first step in invasion of the ECM by tumor cels involves loss of?

A

E-cadherin function

107
Q

Where do most metastasis occur?

What organs?

A
  • First capillary bed available
  • Lung and liver
108
Q

Which oncogene products and tumor suppressor gene products may enter MHC class I and class II for detection by the immune system?

A

Mutated RAS, p53, and BCR-ABL

109
Q

Which overexpressed and aberrantly expressed cellular proteins may lead to detection of tumor cells by the immune system?

A

Overexpressed: tyrosinase, gp100, MART in melanomas

Aberrantly expressed: cancer-testis Ags (MAGE, BAGE)

110
Q

Which T lymphocytes have a clear protective role against virus-associated neoplasms and increased number of these cells correlates with a better prognosis in a variety of cancers?

A

CD8+ CTLs

111
Q

Which immune cells may collaborate in anti-tumor reactivity?

A

T cells, NK cells, and Macrophages

112
Q

Which cytokines activate NK cells and what is their effect on tumor cells?

A
  • IL-2 and IL-15
  • NK cells can lyse a wide range of human tumors, including many that seem to be nonimmunogenic for T cells
113
Q

What mode of inheritance is hereditary nonpolyposis colon cancer (HNPCC)?

Defect in what?

Genome shows what?

A
  • Autosomal dominant
  • Mismatch repair system

- Microsatellite instability (hallmark) = changes in length of short repeats throughout the genome

114
Q

Antibodies against which transmembrane protein expressed on the surface of all normal B-cells have been used in the treatment of B-cell lymphomas and leukemias?

What are these antigens called?

A
  • CD20
  • Differentiation antigens
115
Q

Which mutations leading to genomic instability are important causes of lymphoid neoplasms?

A

Mutations in lymphoid cells due to expression of gene products that induce genomic instability (RAG1, RAG2, AID)

116
Q

What leads to anemia in patients with advanced cancers?

A

Inflammation-induced sequestration of iron and downregulation of EPO production

117
Q

What has been shown to decrease the incidence of colonic adenomas and are now approved for tx of patients with familial adenomatous polyposis?

A

COX2 inhibitors

118
Q

What mucins are expressed on ovarian carcinomas?

A

CA-125

CA19-9

119
Q

Which gene responsible for nucleosome positioning/chromatin remodeling is mutated in 100% of Malignant rhabdoid tumors?

A

SNF5

120
Q

Which genes responsible for histone methylation are seen in 90% of Acute leukemia in infants and Follicular Lymphomas?

A
  • MLL1 =* Acute leukemia in infants
  • MLL2* = Follicular lymphoma
121
Q

In the molecular model for the evolution of colorectal cancers through the adenoma-carcinoma sequence what are the steps?

What are the “first hit” and “second hit”?

A

“First hit” = loss of APC tumor suppressor genes at 5q21

“Second hit” = methylation abnormalities; APC – β-catenin

  • Activation of RAS
  • Loss of TP53 at chromosome 18
122
Q

Where is MUC-1 expressed?

A

Ovarian and breast carcinomas

123
Q

What is expressed on T cell lymphomas and Hodgkin lymphomas and antibodies to it may be used as treatment?

A

CD30

124
Q

People with xeroderma pigmentosum are at increased risk for what?

Defect in what?

A
  • Cancer of skin due to UV radiation –> pyrimidine dimers
  • Nucleotide excision repair (NER)
125
Q

Mutations in what genes account for 30% of HNPCC?

A

MSH2 and MLH1

126
Q

Defects in the homologous recombination DNA repair system results in what 3 disorders?

A

1) Bloom syndrome
2) Ataxia-Telangiectasia
3) Fanconi anemia

127
Q

Infiltrating cancers invoke chronic inflammatory states with what classic symptoms?

A
  • Anemia
  • Fatigue
  • Cachexia
128
Q

What miR is believed to be important in invasiveness and metastasis?

A

MiR-200

129
Q

What miR is overexpressed in B cell lymphomas and upregulates MYC?

A

miR-155

130
Q

Deletions of what miRs lead to CLL?

Their deletion upregulates what?

A
  • miR-15 and miR-16
  • BCL-2
131
Q

Defects in DICER lead to what cancers?

A

Rare Ovarian and Testicular tumors

132
Q

Which genotype seen in 10% of the white population increases the risk of developing lung cancer in light smokers nearly 7-fold?

A

CYP1A1

133
Q

What carcinogen does Aspergillus make?

Causes what cancer?

A
  • Aflatoxin B1
  • Hepatocellular carcinoma in Africa and the Far East
134
Q

Aflatoxin B1-associated hepatocellular carcinomas have which particular mutation in TP53?

A

G:C –> T:A transversion in codon 249 producing an arginine –> serine substitution in the p53 protein

135
Q

What is responsible for the induction of cutaneous cancers?

Due to the formation of?

What wavelength?

A
  • UVB
  • Formation of pyrimidine dimers in DNA (NER); particularly adjacent thymidine residues in the same strand of DNA
  • 280-320 nm
136
Q

What are the most frequent radiation-induced tumors?

A
  • Myeloid leukemias, Thyroid cancer (only in the young)
  • Intermediate category: breast, lungs, and salivary gland cancers
137
Q

What sites are relatively resistant to radiation-induced neoplasia?

A

Skin, bone, and GI tract

138
Q

What is the main oncogenic RNA virus?

What does it cause?

Endemic where?

A

HTLV-1

Adult T-cell leukemia/lymphoma (ATLL)

Japan, Caribbean, South America, Africa

139
Q

What protein is responsible for the transforming activity and essential for viral replication in HTLV-1?

A

Tax

140
Q

How does the protein Tax produced by the HTLV-1 virus contribute to increased pro-growth signaling and cell survival?

A
  • Interacts with PI3K and thereby stimulates AKT
  • Directly upregulates expression of cyclin D2 and represses function of CDK inhibitors
  • Activates the TF NF-kB
141
Q

What are the high-risk DNA viruses associated with squamous cell carcinomas of the cervix, anogenital region, and head/neck?

A

HPV 16 and 18

142
Q

What binds to and mediates the degradation of p53?

What does it stimulate?

Polymorphism of Arg72 in p53 leads to what?

A
  • E6 protein
  • Stimulates expression of TERT, catalytic subunit of telomerase
  • Cervical carcinomas
143
Q

What binds RB protein and displaces E2F?

What can it inactivate?

What can it activate?

A
  • E7 protein
  • Inactivates CDK inhibitors p21 and p27
  • Activates cyclins E and A
144
Q

HPV itself is not sufficient for Carcinogenesis, what else does it need for full malignant transformation?

A

Co-transfection with mutated RAS

145
Q

What are the most common EBV associated tumors?

A
  • Those derived from B cells (i.e., African Burkitt Lymphoma and B-cell lymphomas)
  • Nasopharyngeal carcinoma
146
Q

What does EBV use to infect B cells?

A

Complement receptor 2 (CR2, CD21)

147
Q

What does EBV use as an oncogene?

How does it act?

A
  • LMP-1
  • Behaves like a constituitively active CD40 receptor = stimulates B cell growth
  • Activates NF-kB and JAK/STAT signaling pathways and promotes B cell survival and proliferation
  • Prevents apoptosis by activating BCL2
148
Q

What is the most common childhood tumor in central Africa and New Guinea?

A

Burkitt lymphoma (EBV related)

149
Q

In the case of Burkitt lymphoma it seems that EBV is not directly oncogenic, but instead acts how?

A

Acts as a polyclonal B-cell mitogen setting the stage for acquisition of the (8;14) translocation and other mutations that ultimately produce full-blown cancer

150
Q

Which tumor is endemic in Southern China, some parts of Africa, and in the Inuit population?

Association with what virus?

A
  • Nasopharyngeal carcinoma
  • EBV
151
Q

While the oncogenic effects of HBV and HCV are multifactorial, what is the dominant effect in the development of hepatocellular carcinomas?

A

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

152
Q

Which key molecular step within hepatocytes blocks apoptosis, allowing the dividing hepatocytes to incur genotoxic stress and to accumulate mutations characteristc of HBV induced hepatocellular carcinoma?

A

Activation of NF-kB pathway

153
Q

Which protein of HBV and the HCV core protein can activate signal transduction pathways that may contribute to carcinogenesis?

Where is HBV endemic?

A
  • HBx protein
  • Endemic in: Far East and Africa (highest incidences of hepatocellular carcinoma)
154
Q

H.pylori causes what cancers?

What associated gene is present?

A
  • Gastric adenocarcinoma and MALT (gastric) lymphomas
  • CagA, stimulates growth factor pathways
155
Q

Chronic H.pylori infection leads to polyclonal B-cell proliferations that may give rise to a?

A

Monoclonal B-cell tumor (MALT lymphoma) of the stomach

156
Q

What is characteristic of neoplasms in the gut and urinary tract?

A

Melena (blood in stool) and Hematuria

157
Q

Cancer cachexia is associated with what 3 events?

A

1) Equal loss of both fat and lean muscle
2) Elevated BMR
3) Evidence of systemic inflammation (i.e., increase in acute phase reactants)

158
Q

Grading of tumors is based on what?

A

Cytologic appearance; based on idea that behavior and differentiation are related

159
Q

What are the 3 things staging is graded on?

A

T - size of primary lesion

N - extent of spread to regional LNs

M - presence or absence of blood-borne metastases

160
Q

T0 means what?

A

Indicative of In situ lesion

161
Q

What are the major forms of underlying cancer associated with Cushing Syndrome?

A

Small-cell carcinoma of lung

Pancreatic adenoma

Neural tumors

162
Q

Lung cancer patients with Cushing syndrome have elevated serum levels of?

A
  • POMC
  • Corticotropin
163
Q

What are the 2 general processes involved in cancer-associated hypercalcemia?

Which of the 2 is considered to be paraneoplastic?

A

1) Osteolysis induced by cancer
2) Production of calcemic humoral substances by extraosseous neoplasms (only one considered paraneoplastic)

164
Q

Which factors have been causally implicated in the hypercalcemia of malignancy?

Which is the most important?

A
  • PTHRP = most important
  • IL-1
  • TGF-α
  • TNF
165
Q

Which tumors are most often associated with paraneoplastic hypercalcemia?

A
  • Squamous cell carcinoma of lung
  • Breast carcinoma
  • Renal carcinoma
  • Adult T-cell leukemia/lymphoma
166
Q

What are the 2 major forms of underlying cancer seen in Myasthenia?

A

1) Bronchogenic carcinoma
2) Thymic neoplasms

167
Q

What is the major form of underlying cancer seen in disorders of the CNS and PNS?

A

Breast carcinoma

168
Q

What are the 3 major forms of underlying cancer seen in patients with Acanthosis nigricans?

Due to what?

A
  • Gastric carcinoma
  • Lung carcinoma
  • Uterine carcinoma
  • Due to EGF secretions
169
Q

What are the 2 major forms of underlying cancer seen in patients with Hypertrophic osteoarthropathy and clubbing of the fingers?

A

1) Bronchogenic carcinoma
2) Thymic neoplasms

*Also seen in Myasthenia syndromes

Big Ten Hype

170
Q

What are the 2 major forms of underlying cancer seen in patients with Migratory thrombophlebitis (Trousseau phenomenon)?

A

1) Pancreatic carcinoma
2) Bronchogenic carcinoma

171
Q

Disseminated intravascular coagulation is most commonly associated with what 2 cancers?

A

1) Acute promyelocytic leukemia
2) Prostatic adenocarcinoma

DICAP-itated

172
Q

What screening technique is used for carcinoma of the cervix, endometrial carcinoma, lung carcinoma, bladder and prostatic tumors, etc

A

Cytologic smears

173
Q

What technique is used for assessment of breast, thyroid, and LN cancer?

A

Fine needle aspiration (FNA)

174
Q

Presence of cytokeratins, detected by immunohistochemistry points to?

A

An epithelial origin (carcinoma)

175
Q

What does presence of ALK in a immunohistochemical stain indicate?

A

Lung cancer and lymphomas

176
Q

Flow cytometry is mainly used to identify cellular antigens expressed by what kind of tumors?

A
  • “Liquid” tumors, those that arise from blood-forming tissues
  • B- and T-cell lymphomas and leukemias, as well as myeloid neoplasms
177
Q

HCG is a marker for what tumors?

A
  • Trophoblastic tumors
  • Testicular tumors
178
Q

CA-125 is a marker for what?

A
  • Ovarian tumors
  • Immunoglobulin in multiple myeloma
179
Q

Alpha-fetoprotein (AFP) is a marker for what?

A
  • Hepatocellular carcinomas
  • Embryonal cell carcinomas (yolk sac tumors)
180
Q

Neuron specific enolase is a marker for what 2 tumors?

A

1) Small-cell cancer of lung
2) Neuroblastoma

181
Q

TP53, APC, RAS mutants in stool and serum used as marker for what cancer?

A

Colon cancer

182
Q

Chronic Inflammatory Bowel disease increases the risk for what cancer?

A

Colorectal carcinoma

183
Q

What is one major differenc between Burkitt Lymphomas and Nasopharyngeal carcinomas associated with EBV?

A
  • Burkitt lymphomas associated with EBV do NOT express LMP-1
  • Nasopharynheal carcinomas associated with EBV DO express LMP-1
184
Q

What are the 2 major forms of underlying cancers associated with Dermatomyositis?

A

1) Bronchogenic carcinoma
2) Breast carcinoma

185
Q

What is the major form of underlying cancer associated with Red cell aplasia?

A

Thymic neoplasms

186
Q

Hypertrophic osteoarthropathy is characterized by what 3 abnormalities?

A

1) Periosteal new bone formation, primarily at distal ends of long bones, metatarsals, metacarpals, and prox. phalanges
2) Arthritis of the adjacent joints
3) Clubbing of the digits

187
Q

Advances mucin-secreting adenocarcinomas often show lesions of the heart where?

Known as?

A
  • Often left-sided heart valves
  • Nonbacterial thrombotic endocarditis
188
Q

CA-15-3 is used as a marker for what cancer?

A

Breast cancer