Onc Flashcards

1
Q

What are the characteristics of neoplasia?

A

abnormal collection of new cells with autonomous/unregulated proliferation
clonal population-same initial genetic changes

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

What is the definition of a tumor?

A

swelling

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

How does a malignant tumor differ from a benign tumor?

A

malignant-can invade, destroy adjacent structures and/or metastasize
benign-bland appearance, localized

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

What is a pseudotumor?

A

non-neoplastic tumor (swelling from inflammation)

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

What is scirrhous?

A

dense desmoplasia making a tumor rock hard

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

What are violations of the -oma/benign naming scheme?

A
leukemia-malignant
plasmacytoma/multiple myeloma-malignant
melanoma-malignant
glioma/glioblastoma-malignant (meningioma is benign)
mesothelioma
hepatoma
seminoma/dysgerminoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What is characteristic for a true papillary structure?

A

has a central blood vessel

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

What is a mixed tumor?

A

single clone with different lines of differentiation
mixed tumor of salivary gland-pleomorphic adenoma
uterine tumor-3MT carcinosarcoma

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

What is the difference between desmoplasia and neoplasia?

A

normal cells reacting to a neoplasm (do not contain the mutations that the neoplasm does)

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

What is the difference between a mature and immature teratoma?

A

teratoma-cells of more than 1 germ cell layer (totipotential cells)-usually found in ovary or testicle
mature-mature elements
immature-immature elements, particularly immature neural cells, aggressive

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

What is a dermoid cyst?

A

tumor of ovary

lined with skin and skin appendages; may have hair, teeth

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

What is a hamartoma?

A

right cells for the right place but in wrong arrangement

benign, clonal proliferation

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

What is a choristoma?

A

wrong cells in the wrong place but in the right arrangement

ex-gastric heteropia of the esophagus

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

What is differentiation?

A

determines the grade
well-1 (trouble calling it cancer)
moderate-2 (no trouble calling it a specific kind)
poorly differentiated-3 (trouble figuring out kind of cancer)

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

What is anaplasia? What are the hallmarks of anaplasia?

A
lack of differentiation
pleomorphism
hyperchromasia-darker nuclei
incrased nuclear:cytoplasmic ratio
large nucleoli 
atypical mitoses 
loss of polarity-nucleus away from lumen in colon
tumor giant cells
necrosis-outgrow blood supply
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What does differentiation reveal about cell function?

A

well differentiated may maintain normal function
endocrine-secrete hormones
squamous-make carcinoma
less differentiated can also secrete hormones but they are frequently hormones not found in normal tissue ex AFP from hepatocellular carcinoma

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

What is metaplasia?

A

replacement of one normal cell type with a different normal cell type
ex. smokers keratin for protection, Barretts Esophagus

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

What is dysplasia?

A

neoplastic replacement of normal cells by abnormal cells-starting march to cancer
pleomorphism
loss of polarity
mitotic figures

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

What is carcinoma in situ?

A

severe dysplasia with marked pleomorphism
full thickness changes in epithelium but no invasion of basement membrane
when invasion of basement membrane-invasive

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

What is the tumor stage?

A
more pathologic/prognostic than grade
TNM system
T-size and extent of invasion
N-number and location of involved lymph nodes
M-metastasis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What limits the rate of growth?

A

doubling time

growth fraction-what fraction of cells is replicative

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

Why does rate of growth correlate to differentiation?

A

well differentiated cells take more time to grow

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

What influences rate of growth?

A

hormone-leiomyoma grows with high estrogen stimulation

blood supply

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

How do cancer stem cells differ from normal stem cells?

A

In normal-divide asymmetrically (daughter cell with limited proliferative capacity)
cancer-has immortality, arise from normal tissue stem cells

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

Why are cancer stem cells resistant to conventional therapy?

A

low rate of cell division
multiple drug resistance
tumor initiating cells may remain-cause more cancer

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

What is the Warburg effect?

A

tumors lack oxygen dependent ATP generation
“glucose hunger”
build up of Kreb cycle intermediates-catabolic building for new daughter cells
PET measures 18F-flurodeoxyglucose uptake

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

How can a cancer cell avoid apoptosis?

A
decrease CD95 (Fas)-->extrinsic pathway
FLIP expression to inhibit death complex
decrease p53
loss of APAF-1 (normally decreases caspase activation)
increased Bcl-2
increased IAPs (block caspase 9)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What are tumor suppressor genes?

A

genes that reduce genomic instability

slow cell growth (cancer wants to inactivate these)

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

What are oncogenes?

A

genes that increase genomic instability

promote cell growth (cancer wants to activate these)

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

How can you test for tumor suppressor mutations?

A

PCR for microsatellite instability-mutations hint at genomic instability

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

What is the two hit hypothesis?

A

need to mutations for tumor suppressors to be inactivated

mutations that activate tumor suppressors do not help cancer

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

How many mutations are necessary for oncogenes?

A

only one mutation needs to be activated for cancer cell to benefit
mutations that inactivate oncogenes do not help cancer

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

Why is there an increase in p53 in cancer?

A

mutant p53 when cell is under stress makes more inactive p53

creates a functional but not quantitative deficiency

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

How does HPV interact with tumor suppressors?

A

E6 binds to p53 and promotes degradation

E7 binds and inactivates Rb

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

What is Li-Fraumeni syndrome?

A

inherited p53 mutation
another somatic mutation
leads to multiple tumors at a young age
predisposed to various sarcomas and carcinomas

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

What is the APC/beta catenin pathway?

A

APC-tumor suppressor

down regulates growth promoting signal of beta-catenin

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

How many mutations are necessary within the APC/beta catenin pathway for cancer progression?

A

APC-2
Beta catenin-1
E-cadherin (functional APC can still handle this unless WNT is activated and blocks APC)
WNT-1 (works as an oncogene)

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

What is INK4a/ARF associated with?

A

familial melanoma

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

What is TGF-beta associated with?

A

pancreas

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What is PTEN associated with?

A

Cowden syndrome (breast carcinomas)

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

What is NF1/NF2 associated with?

A

neurofibromatosis

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

What is VHL associated with?

A

von Hippel-Lindau syndrome

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

What is WT1/WT2 associated with?

A

Wilms tumor

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

What is PTCH associated with?

A

nevoid basal cell carcinoma syndrome

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

What does the RET gene cause?

A

cell survival in neuroendocrine cells

MEN2A and MEN2B

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

How can MEN2A and MEN2B be differentiated?

A

MEN2A-extracellular, thyroid, parathyroid, adrenal

MEN2B-intracellular, thyroid and adrenal

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

What mutation is responsible for gastrointestinal stromal tumor?

A

C-kit

treat with Imatinib

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

What translocation is seen in chronic myelomonocytic leukemia?

A

t (5;12)

PDGFR fused with TEL transcription factor (active without stimulus)

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

What translocation is seen in the Philadelphia chromosome? What gene product is seen and what does it cause?

A
t (9;22)
BCR-ABL fusion gene
M-CML and ALL
m-ALL
can be treated with imatinib mesylate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What happens to growth receptors in cancer cells? What is an example?

A

upregulated receptors
Her2/neu overexpressed in breast cancers
treat with Trastuzumab

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

Where is EGFR activated?

A
activated in carcinomas
lung and colon
upregulated in some glioblastomas 
treat with erlotinib, cetuximab (glio)
only works if activating mutations further down the chain are not present
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

Where are KRAS, NRAS, and HRAS mutations seen?

A

KRAS-common in GI and pancreatic tumors (codon 12, 13)

NRAS, HRAS-more common in thyroid neoplasms (codon 61)

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

Where are BRAF mutations seen?

A

V600E mutation
common in thyroid, melanoma, colon
can be targeted in metastatic melanoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

Where are PI3KCA mutations seen?

A

associated with breast, lung, colon, gynecologic cancers

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

How do tumors kill?

A
Cachexia/wasting
hormones or paraneoplastic syndromes
infarction
bleeding
infection
bad location
invasion and metastasis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

What causes cachexia?

A

increased BMR

probably due to cytokines

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

What is the difference between hormone secretion and paraneoplastic syndromes?

A

hormone-release hormones produced by tissue type

paraneoplastic-molecules not normally made by the tumor tissue creating symptoms

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

What cancers are associated with Cushing syndrome?

A

lung small cell
pancreatic
neural tumors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

What cancers are associated with SIADH?

A

lung small cell

intracranial neoplasms

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

What cancers are associated with hypercalcemia?

A

lung squamous cell
breast
renal
adult T cell leuk/lymph

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

What is carcinoid syndrome?

A

elevated serotonin and bradykinin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

What cancers are associated with polycythemia?

A

gastric
renal
cerebellar hemangioma
hepatocellular ca

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

What is hypertrophic osteroarthropathy?

A

seen in bronchogenic carcinoma

clubbing of digits (also seen in liver disease, diffuse lung disease, congenital heart disease)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

What is associated with migratory thrombophlebitis?

A

lung and pancreas cancer

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

What is associated with DIC?

A

acute promyelocytic leukemia

prostate cancer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

What is nonbacterial thrombotic endocarditis?

A

sterile fibrin vegetations on valve leaflets

seen in non-neoplastic causes like rheumatic fever and lupus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

What cancer is the most thrombogenic?

A

mucinous adenocarcinoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

What are examples of bleeding seen in cancer?

A

melena-coln cancer eroded by fecal flow

hematuria in bladder cancer

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

What is an example of a metastasis that seeds the body cavities?

A

pseudomyxoma peritonei-from mucous secreting appendix carcinoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

What is the sentinel lymph node?

A

first node in a regional lymphatic basin receiving lymph flow from tumor

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

What is a skip metastasis?

A

lymph node metastasis beyond first line lymph nodes due to anastomoses of lymphatics

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

What is hematogenous spread? What are examples?

A

veins are more easily penetrated than arteries
tend to rest in first capillary bed encountered
portal vein-liver metastases in colon cancer
veins-lung metastasis
paravertebral venous plexus-prostate and thyroid vertebral metastasis

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

What is tumor emboli homing? What are examples?

A

Organ tropism for metastaiss
breast-bone
lung-adrenal gland and brain
neuroblastoma-liver and bones

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

What cancers are most known for dormancy?

A

breast and melanoma

prolonged survival of micrometastases without progression

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

What does a tumor secrete to make metastatic site habitable?

A

cytokines
growth factors
ECM molecules

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

What are the universal properties of cancer?

A

uncontrolled proliferation-autocrine growth stimulation and do not exhibit contact inhibition
immortalization
protection from antiproliferative signaling
protection from apoptosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

What are the properties of solid tumors?

A

angiogenesis

invasion and metastasis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

What are the emerging properties of tumors?

A

deregulation of cellular energetics

evasion of immune system

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

What are enabling properties of tumors?

A

genome instability and mutation

tumor-promoting inflammation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

How can clonal relationships be established in cancer?

A

same G6PD

mixture of different A/B isotypes are not 1:1

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

What are the results of gain of function mutations in oncogenes?

A
sustained cellular proliferation
advancement of cell cycle progression
decreased requirement of growth factors
promotion of metastasis
protection from apoptosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

How are proto-oncogenes converted?

A

gain of function-increased expression or loss of regulation
viral mechanisms or mutational mechanisms
-deletion or point mutation
-gene amplification
-overexpression (novel promoter)
-novel product (new coding region)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

What disorders are associated with defects in Rb?

A

retinoblastoma

sporadic-small cell lung carcinoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

What disorders are associated with p53 mutations?

A

Li-Fraumeni

sporadic-lung and breast cancer

85
Q

What disorders are associated with BRCA1?

A

familial breast cancer due to mutated ds break repair

sporadic-breast and ovarian cancer

86
Q

What disorders are associated with NF1?

A

neurofibromatosis due to mutated GTPase activator

87
Q

What are the differences between familial and sporadic retinoblastoma?

A

familal-multiple tumors, early onset, bilateral

sporadic-single unilateral tumor, late onset

88
Q

What is LOH? How can it occur

A
loss of heterozygosity is loss of one allele
discoverd by RFLP
-local events
-somatic recombination
-loss and duplication
-chromosome loss
89
Q

What is FAP?

A

familial adenomatous polyposis due to mutation in APC
presents with rectal bleeding
attenuated (100 polyps) or fulminant (100-1000 polyps)

90
Q

What genes are associated with hereditary nonpolyposis colorectal cancer syndrome?

A

MLH1, MSH2, MSH6, or PMS2

91
Q

What is chromosomal instability?

A

increased mutability to a cancer

translocations can be defining or diagnostic markers

92
Q

What causes chromosomal instability?

A

mutations in nucleotide metabolism, DNA replication, chromosome segregation
telomere attrition
aneuploidy
hypomethylation
ds breaks
cellular stress-hypoxia, pH, high osmolarity
VDJ recombination therapies

93
Q

How do epigenetics influence cancer?

A

gene silencing of tumor suppressors by hypermethylation
activation of oncogenes by hypomethylation
genomic instability
reversible changes

94
Q

What epigenetic factors can contribute to cancer progression?

A

reactivation of retrosposons and elimination of methylation borders secondary to hypomethylation
Alu elements harbor 33% of CpG
strongest risk of hypomethylation is age

95
Q

How do epigenetics influence therapy?

A

epigenetic alterations may be driving drug resistance
decitabine
HDAC inhibitors have been developed

96
Q

What is the relationship between IDH1 and glioblastoma?

A

90% of IDH1 mutations are R132H in glioblastoma
epigenetically driven responds poorly to surgery
(most of secondary glioblastomas are IDH1 mutant)

97
Q

What is the difference between a carcinogen and mutagen?

A

carcinogen-agent that induces cancer (most are mutagens)

mutagen-raises the frequency of mutation above the spontaneous rate

98
Q

What is remission?

A

cancer not detectable by the most sensitive measures

99
Q

What is cure?

A

zero cancer cells present

100
Q

What is first line therapy?

A

first treatment for a disease

101
Q

What is used for Hodgkin’s disease?

A

MOPP, ABVD

102
Q

What is used for non-Hodgkin’s disease?

A

CHOP

103
Q

What is used for breast cancer?

A

CMF, CAF

104
Q

What is used for small cell lung cancer?

A

PACE

105
Q

What is used for germ line cancer?

A

VIP

106
Q

What is used for cervical cancer?

A

BIP

107
Q

What is used for lymphomas?

A

M-BACOD

108
Q

What is for ovarian cancer?

A

BEP

109
Q

What is used for pheochromocytoma?

A

CVD

110
Q

What is used for testicular cancer?

A

PEB

111
Q

What drugs are M phase specific?

A

vincristine, vinblastine, paclitaxel

112
Q

What drugs are S phase specific?

A

cytosine arabinoside

hydroxyurea

113
Q

What are the problems with cancer chemotherapy?

A

dose-limiting toxicities
resistance
multidrug glycoprotein pumps
second cancers-leukemia and lymphoma

114
Q

What are the characteristics of alyklating agents?

A

cross linking DNA-blocks DNA replication and transcription
cytotoxic-proliferative dependent, mainly affect G1 and S
resistance-decreased permeability or uptake, increased nucleophiles or repiar
toxicity-decreased bone marrow, nausea, fetal death

115
Q

Which drug resembles phenylamine?

A

melphalon

116
Q

What is the MOA of alkylating agents?

A

reaction with N7 of guanine in DNA

needs both strands for cross-linking to occur

117
Q

What are the unique characteristics of cylcosphamide?

A

must be activated by p450, causes hemorrhagic cystitis and SIADH

118
Q

What is the toxicity caused by chlorambucil?

A

hepatotoxicity

119
Q

What can be given to decrease the hemorrhagic cystitis?

A

MESNA (byproduct acrolein is responsible for the adverse reaction)
MESNA reacts and detoxes

120
Q

What is a unique toxicity of busulfan?

A

pulmonary fibrosis and hyperpigmented skin

121
Q

What are carmustine, lomustine, semustine, streptozocin used to treat?

A

first line of therapy for brain tumors

122
Q

What should be avoided when giving procarbazine?

A

MAOI and alcohol

123
Q

What is the MOA of dacarbazine and temozolamide?

A

methylates DNA and RNA

124
Q

What is the MOA for platinum containing drugs?

A

binds to guanine and forms intrastrand crosslinks

also binds to protein

125
Q

What are the toxicities associated with cisplatin?

A

nephrotoxicity-give forced diuresis
avoid if renal clearance is below 60
ototoxicity
peripheral neuropathy

126
Q

What is the MOA for anthracyclines?

A

tight bindign between base pairs in DNA
blocks activity of topo II
inhibition of DNA repair
cause histone eviction from open chromatin

127
Q

What is a unique toxicity of antracyclines, particularly doxorubicin?

A

cardiotoxicity (can be prevented by dexrazoxane)

128
Q

What is the MOA for mitoxantrone?

A

produce double stranded breaks through complexes but does not produce free radicals
lowers liklihood of cardiotoxicty

129
Q

What drugs are a derivative of podophyllotoxin? What is the MOA?

A

epipodophyllotoxins (etoposide and tenopside)
form a complex with DNA topo II
kills in S and G2

130
Q

What is the MOA for camptothecin analogs?

A

captothecin, topetecan, irinotecan inhibit topo I

act in S phase

131
Q

What is unique about irinotecan?

A

prodrug activated in tissue by carboxyesterases (high in carcinoma)

132
Q

What are bleomycins and when are they active?

A

combination of structurally related antibiotics

active in G2 phase

133
Q

What are the unique toxicities associated with bleomycins?

A

pulmonary fibrosis and pneumonitis
cutaneous reactions
low grade fever
minimal BM suppression

134
Q

What is the mechanism of action for dactinomycin?

A

interaction of peptide loops of dactinomycin at purine-pyrimidine (dG-dC) base pairs and intercalate to prevent DNA transcription

135
Q

What are the toxicities associated with dactinomycin?

A

oral and GI ulceration

stomatitis

136
Q

What is the MOA of folic acid analogs?

A

MTX inhibits dihydrofolate reductase to block biosyntehsis of thymidylate and purines-decreases DNA synthesis
pemetrexed also directly inhibits thymidylate synthase
S phase specific

137
Q

What does pemetrexed inhibit?

A

tymidylate synthase, GAR formyltransferase, AICAR formyltransferase

138
Q

What are the unique toxicities associated with methotrexate and pemetrexed?

A

oral and GI ulceration; hepatotoxicity; pulmonary toxicity

139
Q

What is leucovorin?

A

minimizes the toxic effects of folate depletion

given 24-36 hours after MTX (bypass block without minimizing anti-tumor activity)

140
Q

What is the MOA of 5-flurouracil?

A

decrease DNA synthesis by inhibition of thymidylate synthase, incorporation into DNA
G1 and S phase

141
Q

What are the toxicities associated with 5FU?

A

oral and GI ulceration

142
Q

What is the MOA for cytarbine?

A

S phase

inhibits polymerase alpha

143
Q

What is the MOA for mercaptopurine?

A

S phase

inhibits synthesis of adenine and guanine by blocking conversion of inosinate to purine precursors

144
Q

What are adenosine deaminase inhibitors?

A

pentostatin, cladribine, fludarabine

inhibits ADA leading to buildup of adenosine and deoxyadenosine nucleotides

145
Q

What are the vinca alkaloids?

A

vincristine, vinblastine, vinorelbine

146
Q

What are the yew alkaloids?

A

paclitaxel, docetaxel

147
Q

What is the MOA for vincristine?

A

M phase

bind to soluble tubulin and blocks polymerization

148
Q

What is the MOA for paclitaxel?

A

M phase

prevents depolymerization

149
Q

What are the toxicities associated with vincristine?

A

alopecia, peripheral neuropathy, SIADH

150
Q

What are the toxicities associated with paclitaxel?

A

peripheral neuropathy

151
Q

What is the MOA of asparaginase?

A

depletion of L asparagine leads to inhibition of RNA and DNA synthesis

152
Q

What is mitotane?

A

similar to DDT
used for adrenocortical cancers
blocks hormone production of adrenal
steroid hormone pills must be given to minimize side effects

153
Q

What are interferons?

A

cytokines

INF-alpha slows G1 to S and S to G2

154
Q

What is prednisone?

A

anti-inflammatory used to cause apoptosis in leukemic cells

155
Q

What are progestins used for?

A

used in hormone responsive cancers expressing progesterone receptor

156
Q

What are antiestrogens (tamoxifen, toremefine) used for?

A

used in estrogen receptor positive breast cancer

157
Q

What is tamoxifen?

A

not a steroid, has activity against estrogen

158
Q

What are the anti-androgens (bicalutamide, flutamide, and nilutamide) used for?

A

blocks androgen growth

159
Q

What is the purpose of leuprolide?

A

luteinizing hormone releasing hormone agonist

combination leads to total androgen ablation

160
Q

What is bevaczimab used for?

A

blocks VEGF and prevents angiogenesis

161
Q

What is denusomab used for?

A

blocks RANK-RANKL in bone

162
Q

What is trastuzumab used for?

A

blocks Her2/neu

163
Q

What is gemtuzumab used for?

A

blocks CD33 on leukemia

164
Q

What is rituximab used for?

A

blocks CD20 on B cells

165
Q

What is alemtuzumab used for?

A

blocks CD52 on lymphocytes

166
Q

What drugs are CLTA4 antibodies?

A

ipilimumab and tremelimumab

cause colitis

167
Q

What drugs are PD1 antibodies?

A

nivolumab and pembrolizumab

cause colitis and skin rash

168
Q

What is the MOA for imatinib?

A

blocks Bcr-Abl kinase

169
Q

What is the MOA for erlotinib, gefitinib?

A

blocks EGFR

170
Q

What is the MOA for crizotinib?

A

blocks ALK-1 kinase

171
Q

What is the MOA for bortezomib?

A

blocks proteosomal degradation

172
Q

What is the MOA for sorafenib?

A

blocks B-raf kinase

173
Q

What drug binds to Bcl-2?

A

venetoclax

174
Q

What are some HDAC inhibitors?

A

vorinostat

romidepsin

175
Q

What is a PARP-1 inhibitor?

A

olaparib

176
Q

What is amirubicin?

A

anthracycline with reduced cardiotoxicity

177
Q

What is the MOA of fulvestrant?

A

ER blocker, partial estrogen activity

178
Q

What is abraxane?

A

nanoparticle for formulation of paclitaxel, chemophor free (decrease in hypersensitivity)

179
Q

What is picoplatin?

A

does not cause platinum resistance like other platinum containing drugs

180
Q

What is cytology used from?

A

fine needle aspirate
pap smear
body fluid

181
Q

What is immunochemistry useful for?

A

stain specific antigens in a cell

prognostic or predictive-Her2/neu presence

182
Q

Why is electron microscopy frequently not used?

A

special handling and fixation

outrageously expensive

183
Q

What is flow cytometry used for?

A

count individual cells and characterize cell antigens/DNA content
used for lymphomas and leukemias

184
Q

How can residual disease be detected?

A

FISH, PCR, RT-PCR

problem-low sensitivity and specificity when testing for tumor markers

185
Q

What is PSA?

A

prostate adenocarcinoma marker

186
Q

What is alpha FP?

A

hepatoma and seminoma marker

187
Q

What is HCG?

A

choriocarcinoma marker

188
Q

What is CEA?

A

colon cancer marker

189
Q

What is CA19.9?

A

marker for pancreatic cancer

190
Q

What is CA125?

A

marker for ovarian cancer

191
Q

What are some GWA genetic markers?

A

prostate-8q24

lung-15q25

192
Q

When is ALK inhibitor crizotinib most beneficial?

A

neuroblastoma with ALK mutations R1275Q or F11742

193
Q

What did ch14.18 target?

A

GD2 on surface of neuroblastoma

194
Q

What was ATRA added to treat?

A

APL, increased survival

195
Q

What is CART T?

A

T cells programmed against CD19 cells

196
Q

What are the most common cancers by incidence?

A

prostate/breast
lung
colorectal

197
Q

What are the most common cancers by death?

A

lung
bresast/prostate
colorectal

198
Q

Why have death rates declined?

A

improved detection and treatment

decreased smoking

199
Q

What does chronic inflammation do?

A

cellular damage created that can lead to cancer

non-hereditary predisposing condition

200
Q

What cancers are the immunodeficient susceptible to?

A

Lymphomas-diffuse large B cell lymphomas

Kaposi sarcoma

201
Q

What does UV exposure increase risk of?

A

cumulative-nonmelanoma skin cancer

intermittent but intense-melanoma

202
Q

What tissues are most sensitive to ionizing radiation?

A

directly fractures double helix
hematopoietic-leukemia
thyroid in young
breast, lung, salivary gland

203
Q

What is the difference between direct and indirect acting agents?

A

direct-no metabolic conversion needed, weak carcinogens (alkylating agents)
indirect-require metabolic conversion, enzymes matter (CYP1A1 increases lung cancer in smokers) (PCV, amides, nitrites)

204
Q

What are the major infection associated malignancies?

A

stomach-H pylori
cervical-HPV
liver-Hep B and C
Burkitts lymphoma and nasopharyngeal-EBV
Kaposi sarcoma and non-Hodgkin lymphoma-HIV/HHV8
bladder and colon-schistosomiasis
adult T cell leukemia/lymphoma-human T cell lymphotrophic virus type 1

205
Q

How do infections induce malignancies?

A
immunosuppression
chronic inflammation
oxidative stress
DNA damage and mutation
cell injury 
cell division
206
Q

What is aflatoxin B associated with?

A

Hepatocellular carcinoma (when given a high protein diet)

207
Q

What are examples of primary prevention?

A
decrease alcohol and tobacco use
decreased exposure to carcinogens
decreased exposure to radiation and excessive sunlight
access to clean water
safe sex
immunization and treatment of infections
208
Q

What are examples of secondary prevention?

A

mass population and selective cancer screening through IARC and SEER