• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/131

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

131 Cards in this Set

  • Front
  • Back
Benign vs Malignant tumors
BASED ON POTENTIAL
Benign: microscopic and gross characteristics are innocent. It is localized, doesn't spread and treatable with surgical intervention

Malignant = can invade and destroy adjacent structures. Can spread to distant sites. May cause death
Two components of tumors
Parenchyma (transformed cells)
Non-neoplastic Stroma (supporting cells that tumor needs for growth and survival such as connective tissue, blood vessels and inflammatory cells)
Naming of benign tumors
-oma
except for:
hepatoma
seminoma
melanoma
lymphoma
mesothelioma
myeloma
sarcoma
carcinoma
What is an adenoma?
benign tumor of the glands (from glands or in glandular pattern
What is a papilloma?
benign epithelial neoplasm, finger like projections
WARTS
What is a polyp?
mass that projects above the mucosal surface
What is a cystaderoma?
hollow cystic mass
Sarcomas
cancers of cells with mesenchymal origin (bone, cartilage, fat, connective tissue, and muscle)
Carcinoma
From epithelia, most common type of malignant tumor. Cancers can thus be in all three germ layers
Lymphoma
cancer arising from lymph nodes and tissues
Leukemias
cancer of immature blood cells that grown in the marrow and accumulate in the blood stream
Benign vs Malignant: 4 things that separate them
1) Degree of differentiation (malignant can be anywhere on differentiation scale but benign tumors are always well differentiated)
2) Rate of Tumor Growth (malignant is faster)
3) Presence of local invasion (malignant)
4) Ability to metastasize (only malignant)
Better differentiated cells retain?
functional capabilities or gain unintended functions
Name two unintended functions gained by well differentiated tumors
1) AFP ---> secrete fetal protein

ECTOPIC HORMONE SECRETION:
2) Small Cell Lung Carcinoma ---> ADH secretion causing hypernatremia
3) Squamous lung cancer ---> PTH causing hypercalcemia
What is Anaplasia?
malignant neoplasms of undifferentiated cells. It is a hallmark of malignancy
Tissue and Cellular characteristics of Anaplasia
Tissue: Loss of polarity, anarchic growth
Cellular: Pleumorphism (variation in size and shape), Hyperchromic and large nuclei (N:C ration increases); Chromatin clumping; Large nucleoli, and large # of mitoses with multiple spindles
Grading is used to measure?
differenciation
Scaling is used to measure
spread of primary tumor
What is dysplasia? How is it different from malignancy?
Disorderly but non-neoplastic proliferation normally in epithelium. Unlike malignancy it can be unchanged for long periods, regress spontaneously, progress to malignancy
Cervical dysplasia and carcinoma
Mild Dysplasia (CIN1) = failure of proper maturation lowest 1/3 of mucosa
Moderate Dysplasia (CIN II) = failure of maturation in cell in lower 2/3 of mucosa
Severe displasia (CIN III) = abnormality affects >2/3 of epithelia; mitoses at all levels
Carcinoma in situ = entire thickness replaced by atypical dysplastic cells, no orderly differentiation. Basement membrane NOT crossed
Top three causes of cancer death in men and women
Men: Lung/Bronchus, Prostate, Colon/Rectum
Women: Lung/Bronchus, Breast, Colon/Rectum
Has lung cancer begun to decrease in men or women
In men but in women it has only plateaued
Death vs incidence for following cancers: Lung, Prostate
Lung: new case=deaths
Prostate: new cases> deaths
Pancreatic: new cases = deaths
Breast: new cases> deaths
Cancers and stages
Early: Breast, colorectal, melanoma, prostate
Late: Lung, ovary pancrease
What cancer peaks at middle age?
Testicular
What cancer has a bimodal distribution?
Hodgekins
Oncogenes are?
Tumor Supressor Genes are?
Oncogenes are growth factors: prepare cells to divide, stimulate proliferation
Tumor Supressor Genes are Growth Inhibiting Factors: CDK inhibitors (CIP/INK 4a) which inhibit cyclin D/cdk4 and cyclin E/cdk2; Also RB which is a cell cycle regulator at G1-S transitions
What cyclin is over expressed in B Cell lymphomas, carcinomas?
Cyclin D
Cyclins and their CDK
D - 4
E - 2
A - 2
B - 1
CDK inhibitors regulating G1, G2
p21 and p27 (CIP and INK) -->induced by p53 in response to DNA damage, slowing cell cycle
p16 (INK4a): inhibits cyclin D/cdk4, inactive in cancers
How many alleles need to be altered in protooncogenes?
only 1 confers gain of function mutation
Oncogene point mutations
K-Ras (colon, lung, pancreas), H-Ras (bladder, kidney), N-Ras (melanoma)
What is the most frequently activated oncogene?
RAS
What is RAS
a G protein stimulated by growth factors that activates the MAP kinase pathways, activating transcription and helping cell cycle progression
RAS mutation
Gain of function mutation at codon 12 or 61. Alters GTPase activity, can't hydrolyze GTP and RAS is always turned on
What does MYC Proto-Oncogene do?
It is a transcription factor. Its over expression has the same functional consequence as RAS
Translocations causing oncogenes
ABL proto-oncogene translocation resulting in chronic myelogenous leukemia (slow progressing, enlarged spleen, fatigue);
What is the diagnostic marker for chronic myelogenous leukemia?
Philadelphia Chromosome
What is used to cure CML?
Drugs that target BCR-ABL fusion kinase (which insert into the catalytic cleft)
How to diagnose CML?
Lots of leukocytes seen in blood smear; Diagnostic Hallmark of CML is the Philadelphia Chromosome
What is the philadelphia chromosome?
a translocation between a portion of chromosome 9 and chromosome 22, forming a BCR-ABL Oncogene with unregulated tyrosine kinase activity. The fusion gene has self sufficiency of growth signals, insensitivity to growth-inhibitory signals, evasion of apoptosis.
Example of Gene Amplificaiton
L-MYC oncogene is amplified with some types of tobacco smoking;
N-MYC amplification in neuroblastoma
How can gene amplification be present?
Double minutes (pieces of genes)
OR
Homogenous-staining regions (HSR) ---> when copies integrated into chromosome
Overexpression of growth factor receptors, two examples
1) EGF-R in some lung cancers
2) HER2/NEU oncogene in 30% of patients with invasive breast cancer.
A subset of non-smoking women develop adenocarcinomas of the lung as a consequence of?
Overexpression of EGF-R. Can be treated with drugs that inhibit EGF-R signaling such as Cetuximab (binds EGF receptor), Trastuzumab (binds EGF) Erlotininb and Gefitinin (blocks activity of EGF receptor)
What is the gene that is a prognostic marker in breast cancer?
HER2/NEU...overexpressed in cancers...can see using FISH or Immunohistochemistry
What is a therapeutic target in breast cancer?
HER2/NEU...Antibodies (Herceptin) block extracellular domain of HER2/NEU and prolong survival of this subset of women with breast cancer.
Transformation of what causes cancer?
A single progenitor cell that incurred genetic damage and undergoes clonal expansion ---> monoclonal nature of tumors
When tumor is clinically detectable, what portion of its life cycle has occurred?
30 doublings before it's detectable and 10 doublings afterward.
What is tumor heterogeneity?
A tumor becomes heterogenous over time although it begins monoclonal in origin ----> result of multiple mutations that accumulate independently of different cells. Subclones that beat the odds become cancer genes.
Four classes of normal regulatory genes
1) growth promoting proto-oncogenes
2) growth inhibiting tumor suppressor genes
3) genes that regulate apoptosis
4) Genes involved in DNA repair
How do cancer cells develop limitless replication potential?
Telomerase activation in cancer cells void the telomere time clock which usually limits the proliferative capability of normal somatic cells. You first have shortened telomeres, which, in the absence of p53, form dicentric chromosomes which replicate to form new double stranded breaks. These cells can survive and be cancerous if telomerase activity is present.
What is tumor heterogeneity?
A tumor becomes heterogenous over time although it begins monoclonal in origin ----> result of multiple mutations that accumulate independently of different cells. Subclones that beat the odds become cancer genes.
Four classes of normal regulatory genes
1) growth promoting proto-oncogenes
2) growth inhibiting tumor suppressor genes
3) genes that regulate apoptosis
4) Genes involved in DNA repair
How do cancer cells develop limitless replication potential?
Telomerase activation in cancer cells void the telomere time clock which usually limits the proliferative capability of normal somatic cells. You first have shortened telomeres, which, in the absence of p53, form dicentric chromosomes which replicate to form new double stranded breaks. These cells can survive and be cancerous if telomerase activity is present.
How do tumors sustain angiogenesis?
Induced by hypoxia; Normally VHL (Von Hipple Lindan Factor) binds to HIF-1alpha and is destroyed. However in hypoxia, HIF-1alpha cannot bind (a process that requires oxygen), translocates to the nucleus and activates VEGF.
In addition, FGF is released by stroma and tumor cells in ECF
What type of cancer invades but rarely metastasizes?
Basel Cell Carcinoma
In general, the more anapestic the more likely to metastasize. There is one exception, what is it?
Small cell lung carcinoma
What are the steps in tumor invasion?
1) Detachment of tumor cells from each other via loss of E-cadherin function. B Catenin are part of this complex, and allow for the transmition of antigrowth signals.
2) Local degradation of basement membrane and interstitial connective tissue via secretion of proteolytic enzymes (MMPS, Cathesin, Urokinase Plasminogen Activator)
3) Activation of tumor cells to ECM proteins via fibronectin and lamin receptor
4) Migration that involves many receptors and signal transmit ion pathways that converge on actin skeleto; Tumor derived cytokines cleave products of matrix and growth factors; Stromal cells produce growth factors like HGF, SCF
What are the three pathways of metastasis and what types of cancers take them
1) direct seeding (neoplasm invades body cavity) in ovarian cancer
2) Hematogenous spread (lung and liver) favored for sarcomas, usually venous
3) Lymphatic system, favored for carcinomas
What is the Knudson Hypothesis?
Retinoblastoma results from 2 mutations in the following senarios:
1) Hereditary: 1st mutation is germinal, present in all cells. Second mutation is only in retinoblast. Tumor is in both eyes.
Non-hereditary: Two somatic mutations, usually only one eye
Is retinoblastomas a autosomal dominant or recessive condition
dominant; these individuals are also susceptible to bone tumors, soft tissue sarcomas.
How is RB lost?
Deletion, point mutation, functional inactivation by viral oncoprotein, epigenetic silencing via methylation
How do microRNAs play a role in cancer?
Function is to control cell growth, differentiation, and cell survival.
MicroRNAs can increase expression of oncogenes or reduce expression of tumor suppressor genes.
What is the normal function of RB
Rb is normally hypophosphorylated in a complex with E2F in GI. It prevents cell cycle progression by sequestering E2F and by recruiting histone deacetylases and methyltransferases which bind to promoters of E2F-resposive genes on Cyclin E--->
However, mitogenic signaling leads to cyclin D expression and activation of Cyclin D Cdk4/6 complexes. Cyclins complexes phosphorylate E2F/RB resulting in release of EF2 which activates transcription of S phase genes
What controls cyclins?
Growth inhibitors such as TGF-B and p53 activate CDK inhibitors (p16, INK4a)----> inactivate cyclins
Growth factors EGF and PDGF----> activate cyclin
An epigenetic point mutation in CDK inhibitors results in what?
lack of CDK inactivation and thus growth (melanoma, pancreatic, esophageal, breast
What would mimic RB loss?
CDK4 overexpression
Cyclin D overexpression, amplification, translocation
CDK21 (p16) inactivation
where is p53 located
On chromosome 17, it is a tumor suppressor gene
There is homozygous loss of ___ in almost all cancers
p53
mutations of p53 are usually inherited or somatic?
somatic
What is an example of an inherited p53 mutation?
Li-Frauremi Syndrome (younger age, multiple sites)
What is normal p53 function?
- Arrests cell cycle in G1 to repair damaged DNA via p21 (CDKI)
- Repairs DNA via GADD45
- Transcribes BAX leading to apoptosis
p16 inactivates?
p21 activates
cyclins (tumor supressor)
CDKIs (leading to G1 arrest)
What senses DNA damage
ATM/ATR "sensors" which are protein kinases ----> phosporylate various targets (incl. p53 and DNA-repair proteins) and pause cell cycle and stimulation of DNA repair problems.
what's p53's relationship with microRNA?
p53 activates transcription of mir34 microRNA, which binds to cognate 3' untranslated regions. This 1) inhibits translation of growth promoting genes like MYC and CDK4 and inhibits translation of anti-apoptotic genes like BCL-2
What is the role of MDM2?
In non-stressed cells, p53 has a short half life, associating with MDM2 makes it inactive and undergoes degradation.
Under conditions of stress, p53 undergoes post-transcriptional modifications that release it from MDM2, prolonging its half life
Binding of MDM2 to ARF does what?
MDM2 can no longer interact with p53, activating it
If you lose ARF, you lose or gain p53 function
Lose it
What does TNM stand for?
----system of staging-----
T = size of Tumor
N = Nodal involvement
M - Metastases
Functional loss of p53 can be due to what?
1) mutation of p53
2) binding to viral oncoproteins (HPV viral oncoprotein E6)
3) Increase in MDM2
4) Deletion of p14ARF indirectly inactivates tumor suppressor pathway
What is APC? What is it's function?
Its a member of another class of growth suppressor genes and is part of the WNT signaling pathway. It's function is to down regulate B-Catenin. In resting cells, B-Catenin is bound to APC and destroyed. When cells are stimulated by WNT molecules, the destruction complex is deactivated, and degradation does not occur and cytoplasmic levels increase. B-Catenin translocated to nucleus and activates genes involved in Cell cycle progression. When APC is mutated, destruction of B catenin cannot occur---->cells under constant stimulation.
What happens if there is a germ-line mutation of APC
You can get the disorder "Familial Adenomatous Polyposis" in which individuals who inherit a single mutant APC allele develop thousands of adenomatous colon polyps in teens or early twenties....colon cancer is inevitable by 50.
Relationship between APC and Colon cancer
Normal APC but activating mutations in B-Catenin so that it is not destroyed.
What is the progression of mutations for colon cancer?
1) loss of one normal APC gene occurs early followed by inactivation of second APC gene later in life.
2) K-Ras mutation
3) p53 mutation, LOH, SMAD 2 and 4 (TGF-B)
4) Telomerase

Order is irrelevant. It's their accumulation that matters
Mechanisms for Self sufficiency in growth signals?
RAS mutation, MYC amplification, BCR-ABL translocation, EGF-R activation
Mechanisms for Insensitivity to growth inhibitory signals
BCR-ABL oncogene, pRb inactivation, p16 or INK4a, inactivation of APC
Evasion of Apoptosis - mechanisms
p53 mutations, BCR-ABL
Limitless replication potential mechanisms
activation of telomerase, inactivation of p53
mechanisms for development of sustained angiogenesis
overexpression of angiogenic factors, activation of HIF-1alpha
mechanisms for ability to invade and metastasize
inactivation of E-cadherin and cell-cell contact, activation of proteases, enhanced motility
What is Cowden Syndrome?
PTEN mutations, which is a tumor suppressor gene
Presents: Big head, autism
Facial and bucal lesions
What is Li Fraumeni Syndrome?
Inherited p53 mutation
Early onset sarcome, leukemia, Brain cancer,
Adrenocortical cancer
Breast Cancer
What is Peutz Jeghers syndrome?
Autosomal dominant mutation in tumor supressor gene.
Presentation:
Childhood GI heratomas
Pigmentation of lips, freckles on lips and fingertips
Prophylactic mastectomy reduces risk by how much? What about oophorectomy?
Decrease BC by 90%.
Decrease BC by 50% and Ovarian by 90%
Evolutionary theory of aging
aging confers a evolutionary advantage
selection pressure based on older individual being less useful
Mutation accumulation theory
mutation that kills late in life will not be selected against.
Aging is NON-Adaptive
Disposable Soma Theory
longevity, reproduction, and antagonistic pleiotrophy
1) Sexual reproduction may be at the expense of longevity (Bamboo)
2) gene or gene product has two effect: beneficial and detrimental (Antagonistic pleiotrophy (e.g., high testosterone)
R selection
density independent
selects for high reproductive rate
hazardous environment
rapid development, small body size, short life span (mice)
K Selection
Density dependent
selects for survival at max density.
Non hazardous environment
Delayed development, larger body size, longer lifespan
mitochondrial DNA damage in comparison to nuclear?
Mt DNA is more susceptible to ROS and it has worse repair mechanisms but the impact is less because it is redundant
What is cellular senescence?
loss of cell's ability to replicate.
senescent cells are larger than proliferating cells;
most common cause is telomeres shortening
Hutchinson-Gilford Progeria syndrome is the result of what?
Point mutation in LMNA gene (truncated form called progerin) which results in problems with Lamin A (nuclear envelope) making it hard for cells to divide
What are some examples of changes in gene expression as we age
decreased transcription of growth and stress related genes.
Increased transcription of inflammatory cytokines and matrix mellaloproteinases
longevity is related to what?
insulin and IGF-1
Decreased daf-2 expression in worms
size and height
caloric intake
How does one tell you got the whole tumor when excising?
You ink the margins and see if it has positive or negative margins. H and E most often used.
What is the most common dye used in surgical pathology?
H and E (Hematoxylin --basic, blue; Eocin - Acidic, red)
what is the gross appearance of colonic adenoma
strawberry
Tumor staging is done how?
Tumor (primary) --- 1 (mucosa) and 2 (musularis propria)
N (Node) - 3 (lymph node)
M (Metastasis) -4 (distant site)
what the benefit of freezing pathological sections
rapid, but lower quality, labor intensive
Prussian blue dye stains?
Iron
Trichrome dye stains?
Collagen (Fibrosis)
Verhoeff dye stains?
Elastin
Ziehl Nielsen dye stains?
Mycobacteria
Silver Methanamine stains
Fungi
Why to use immunohistochemistry
- identify origin of poorly differentiated or undifferentiated cells
- subclassify lymphomas, identify metastases, prognostic information
What receptors can be identified using immunohistochemistry
estrogen (TOMAXIPHAN)
progesterone
Her2/NEU (HERCEPTIN)
Colorectal cancers over express?
EGF-R
EGF-R therapy can be given to which patients?
Patients without K-Ras mutation.
Lynch Syndrome
MLH-1 mutations leading to bad DNA repair.
What is used to stain astrocytomas?
GFAP stains intermediate filaments of astrocytes
What are the specialized cells of the CNS that line the ventricular system?
Ependymal cells
What produces CSF
Choroid Plexus
tumor of the neurons
ganglioctyoma
how do you stage brain tumors?
You can't:
1) no CNS lymphatic drainage
2) No concept of margins (bc infiltration is the rule)
3) Rarely metastasize to lower origins
What is the only proven environmental factor for brain tumors?
Radiation:
given for TInia Capitis, brian tumors, acute lymphoma leukemia. Most come type of cancer is Meningioma 10 years post exposure
Grading Brain Tumors
1+ AMEN factor (1=Pilocytotic Astrocytoma, 2=Astrocytoma, 3=Anaplastic Astrocytoma, 4=Glioblastoma)
Atypia (of nucleus)
Mitoses
Endothelial proliferation of capillaries
Necrosis
Genetic characteristics of primary and secondary glioblastomas
Primary is usually EGF-R over expression
Secondary is usually Tp53 gene mutation
When do you have a duty to warn?
Special relationship
identifiable victim
harm in the forceable future and its serious
When do you test children
- with risk for genetic disease that has preventative measures (YES)
- If there are no available treatments but onset is in childhood, parent has discretion
- If onset is in adulthood and there are no preventable measures, then recommended NO
- discouraged for benefit of other family members
GINA Act
Can't discriminate based on genetic conditions.