• 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

1/120

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;

120 Cards in this Set

  • Front
  • Back
  • 3rd side (hint)
Adjuvant therapy
therapy after surgery or radiation therapy

for Solid Tumors

given to people who do not have detectable cancer but are likely to relapse
biological therapy
therapeutic agents that are not cytotoxic
es) IL-2, retinoids, angiogenesis inhibitors
Carcinoma
epithelial tumors (eg breast, colon, lung)
most common types of human cancer
treated by organ of origin

there are no carcinomas of the spleen
concurrent chemotherapy
in conjunction with radiotherapy

localized but unresectable cancers
consolidation
chemotherapy given to those in remission to extend disease-free survival

refers to hematopoetic malignancies as opposed to adjuvant therapy for solid cancers
epigenetic
heritable modification of genome that does not change DNA sequence

often permanent silencing of gene or locus through modification of histones or hypermethylation of certain cytosines w/in promotor or regulatory regions

Contribute to malignant transformation by:
inducing genomic instability by inhibiting DNA repair
inactivating tumor suppressor genes in association w/ promoter DNA methylation
tumor grade
based on histological appearance
consideres: nuclear: cytoplasmic ratio, mitotic index, cellular heterogeneity, etc
higher grade usually means more aggressive & worse prognosis
In Situ cancer
malignant-looking cells that have not invaded the basement membrane
very high grade dysplasia
almost always merits therapy
Induction
high-dose initial chemotherapy used to induce remission
Leukemia
malignant cells circulating in blood & bone marrow

most leukemia occurs in adults

some cytogenetic lesions are favorable in leukemia
epigenetic
heritable modification of genome that does not change DNA sequence

often permanent silencing of gene or locus through modification of histones or hypermethylation of certain cytosines w/in promotor or regulatory regions
tumor grade
based on histological appearance
consideres: nuclear: cytoplasmic ratio, mitotic index, cellular heterogeneity, etc
higher grade usually means more aggressive & worse prognosis
In Situ cancer
malignant-looking cells that have not invaded the basement membrane
very high grade dysplasia
almost always merits therapy
Induction
high-dose initial chemotherapy used to induce remission
Leukemia
malignant cells circulating in blood & bone marrow
lymphoma
malignant lymphoid cells in lymph nodes
maintenance chemotherapy
prlonged (can be >18 months) chemotherapy to maintain remission

used for some leukemias, lymphomas & pediatric tumors (not usually in adult solid tumors)
Micrometastases
can't be detected clinically at time of tumor diagnosis (too small for CT scan)
often relapse w/ distant disease later on
target of adjuvant therapy
Neo-adjuvant therapy
chemo before surgery or radiation therapy
makes surgery simpler by shrinking tumor
can tell if tumor is sensitive to chemotherapy being given
Performance status
most universal predictor fo patient outcomes across wide variety of tumor types
measured by "Karnofsky score" or "ECOG score"

PS0: no symptoms, no limits from disease
PS1: restricted in strenuous activity but ambulatory 7 able to carry out light work
PS2: ambulatory and capable of self-care but unable to carry out work activities; up and about more than 50% of waking hours
PS3: limited self-care, confined to bed or chair more than 50% of waking hours
PS4: completely disabled
partial response
>50% decrease in tumor mass that last for more than 3 months
complete response
complete resolution of disease for more than 3 months
Response Rate
partial response + complete response
Sarcoma
Mesenchymal tumors of 2 flavors:
soft tissue sarcomas
osteosarcomas
rarer than carcinomas in adults
clinically very heterogeneous
histology more important than location (leiomyosarcoma treated similarly no matter the location)
staging
measure of degree of spread
should be done multiple times at different points
requires complete history, physical exam, lab, radiographic & pathologic assessment of primary tumor, regional lymph nodes & metastatic sites
tumor suppressor gene
leads to:
modulation of DNA repair
senescence
or apoptosis
estimate tumor burden by
extrapolate from size of a lesion using radiologic assessment

measuring a marker protein produced by the tumor (eg immunoglobulin in multiple myeloma)
refractory cancer
does not respond to therapy well

ex) pancreatic cancer, melanoma
most common sites of metastases
lungs, liver, bones, brain, adrenals, subcutaneous

however, any solid tumor can spread to anywhere
effect of adjuvant therapy on long-term cure rates
increased in breast, colorectal, osteosarcoma

not increased in lung cancer
factors in likelihood of metastasis of a tumor
tumor motility
angiogenesis
patterns of gene expression
Principles of Chemotherapy
single drugs are rarely curative (b/c of resistance)
used for both curative and palliative treatment
Principles of Chemotherapy Combo Regimens
component drugs must be active as single agents
toxicities should be non-overlapping
drugs should be used in their optimal dose/schedule
combinations should be given at consistent intervals
mechanisms of Drug Resistance
- overexpression of drug metabolizing enzymes
- overexpression of drug targets
- drug efflux pumps
- increase in DNA repair mechanisms
consolidation/intensification
chemotherapy repeated after a remission is obtained to solidify the remission and increase cure or prolong remission

may be similar to previous regimens (consolidation) or intensified
maintenance chemotherapy
prolonged chemotherapy to maintain a remission

important in some leukemias & pediatric tumors but not in solid tumors
What cancers can be treated curatively even at advanced stage?
testicular, Hodgkin's, non-Hodgenkin's lymphoma
purpose of surgical oncology
- reduce or remove bulk
- resect metastatic disease (lung mets in sarcoma, liver mets in colon cancer)
- emergencies- spinal cord compression, bowl perforation
- palliation
- reconstruction
Phase I clinical trial
evidence of toxicity?
Phase II clinical trial
are drugs useful for treating the cancer?
Phase III clinical trials
how useful is the new drug compared to the gold standard treatment?
Phase IV clinical trials
get experience w/ new drug & with combinations of drugs/treatment
Synthetic Lethality
2 genes are in a synthetic lethal relationship if a mutation in either gene alone is not lethal but mutations in both lead to cell death

usefulness example)
BRCA1 & 2 defects lead to defective base-excision repair
when patients are treated w/ PARP (PolyADPRibose polymerase), it disables the homologous recombination repair
when both repair mechanisms are disable-->cell dies
PARP inhibitors
cause synthetic lethality in BRCA 1 & 2 related tumors
Oncogene examples of activation mechanisms
cyclin D1 - amplification
K-RAS - point mutation
c-MYC - translocation
akt activation by PTEN loss - loss of inhibitor
Tumor Suppressor Gene examples
p16INK4a (loss of senescence)
RB
p53
PTEN
BRCA (loss of DNA repair mechanism)
malignant cells of ALL
committed lymphoid progenitor cell (pre-T or -B cell)
malignant cells of CLL
mature B-cells
derived from pre or post-germinal center B cells
malignant cells of AML
committed myeloid progenitor
CML - how is it different from other leukemias?
- classified as myeloproliferative disorder (like Polycythemia vera, essential thrombocytosis & primary myelofibrosis)
- malignant cells MAINTAIN THE ABILITY TO DIFFERENTIATE
- transformation of hematopoietic progenitor cell (HPC)
- predilection for myeloid pathway (but does NOT cause Erythrocytosis or usually Anemia)
- MORPHOLOGICALLY HETEROGENEOUS - population of cells at all levels of myeloid differentiation
- can transform into Acute Myeloid Leukemia or Acute Lymphoblastic Anemia
Who gets CML?
1 to 2 ppl/100,000
median age at diagnosis: 66 yrs
male/female ratio: 1.4/4
risk factors: ionizing radiation exposure
Unique Clinical Presentation of CML
usually no adenopathy (like AML)
Leukocytosis - neutrophilia, basophilia, eosinophilia
Anemia or normal hemoglobin
Thrombocytosis
20-40% detected solely from labs

most common symptoms however, are fatigue, weight loss, abdominal fullness, & night sweats
splenomegaly (50%), sometimes hepatomegaly
Genetics of CML
Philadelphia Chromosome
t(9;22) required for diagnosis of CML
detected cytogenetically in 95% of cases
5% of cases detected by FISH
What does the Philadelphia chromosome do?
t(9;22)
- segment of abl gene coding for non-receptor tyrosine kinase is translocated to bcr gene on chromosome 22
- bcr-abl gene -->abnormal tyrosine kinase that's constitutively active
EXCEPTION TO MULTI-STEP MODEL OF NEOPLASIA
Philadelphia chromosome is the only mutation that is required for CML
How is CML diagnosed?
Peripheral smear review & bone marrow biopsy
- determine Chronic phase, accelerated phase or blast crisis

cytogenetics of blood and marrow
-detect most t(9;22) as well as other cytogenetic abnormalities

FISH
- BEST SCREENING TEST
- more sensitive than cytogenetics & can detect cryptic translocations
- does not test for other abnormalities

Quantitative PCR
- most quantitatively sensitive
- does not test for other abnormalities, can miss rare mutations
Which phase of CML is easiest to treat?
chronic phase is much easier to treat than accelerated phase or blast crisis
Prognosis of CML phases
Chronic phase (most people present at chronic phase)
--- median 3-5 years

Accelerated phase (increasingly unresponsive to therapy)
--median duration 6-9 months

Blast crisis (complications of cytopenias, extramedullary disease)

25-40% of patients skip accelerated phase and go directly to blast phase
--- median survival 3-6 months
Therapy for CML
2 options:
Abl tyrosine kinase inhibitors
-- Imatinib (Gleevec)
-- 2nd generation Abl inhibitors
Allgeneic stem cell transplant (only "cure)
Imatinib mesylate (Gleevec)
TYROSINE KINASE INHIBITOR FOR CML
small molecule inhibitor of BCR-ABL
avoid moving to Accelerated or Blast phases

What about resistance? -->side 3

-----preferentially binds inactive conformation of BCR-ABL
can take orally
well tolerated
excellent efficacy!
Primary Resistance (VERY UNUSUAL - NOT VERY IMPORTANT)
-- inadequte initial response
--- due to Low levels of imatinib (non-compliance, poor absorption, metabolism, reduced influx pump levels)

Secondary resistance
-- loss of previous response
--------overexpression of bcr-abl (10%)
-------point mutations within ABL kinase domain - can't bind (50-90%)

what to do? use 2nd generation inhibitors
Nilotinib (2nd generation ABL kinase inhibitor)
binds better & more specifically to BCR-ABL than imatinib
still active w/ most BCR-ABL point mutations
now FDA approved for initial treatment -- deeper responses than imatinib
Stem Cell Transplant in CML
high dose chemo +/- radiation-->infusion of donor HSCs

Graft vs Host disease is good!
-- decreased risk of relapse
-- depletion of donor HSC increases risk of relapse

can re-induce remission w/ donor lymphocyte infusions

Disease free survival after related donor SCT is 45-70%

only known cure but substantial morbidity
Who gets CLL?
most common adult leukemia
15,110 US cases in 2008
4,390 deaths

median age at diagnosis:72

Risk:
family members have 2-7x increased risk
Agent Orange

UNHEARD OF IN KIDS
"smudge cell"
what's it in?
characteristic of CLL on peripheral smear
Unique Presentation of CLL
frequent infections esp respiratory or encapsulated organisms
---- due to hypogammaglobulinemia

lymphadenopathy (like ALL)
Leukocytosis - SPECIFICALLY LYMPHOCYTOSIS
anemia
thrombocytopenia (unlike CML)
Autoimmunity in CLL
autoimmune hemolytic anemia in 10-25% of cases
Immune thrombocytopenia in 2%
pure red cell aplasia & autoimmune neutropenia (Evans Syndrome) is rare
Transformation of CLL
worse prognosis

evolution of more aggressive lymphoid neoplasim
---- clonally related to CLL in 60%

Richter's Transformation
---- CLL-->diffuse large cell lymphoma
----- occurs in 2 to 9% of CLL
--- worsening of normal symptoms

Prolymphocytic leukemia
-----10% of CLL
----- much bigger than CLL cells, much more cytoplasm
Dysregulated cell death in CLL
Bcl-2 is inhibitor of apoptosis
inhibits release of cytochrome c from mitochondria
overexpression -->malignant cells
Rai Classification used for CLL
Stage 0: lymphocytosis only -- survival >15 yrs
Stage 1: add lymphadenopathy -- survival 8 yrs
Stage 2: add splenomegaly --survival 6 yrs
Stage 3: add Anemia -- survival 3 yrs
Stage 4: add Thrombocytopenia -- survival 2 yrs
Treatment of CLL
NOT CURABLE
don't treat patients w/out symptoms -- no survival advantage -- early stage long term prognosis is still good

Chemotherapy - relieve symptoms
- weight loss, fevers, sweats, fatigue
- bulky lymphadenopathy, painful splenomegaly
- rapidly increases lymphocytosis
- autoimmune cytopenias like hemolytic anemia or thrombocytopenia
- increased frequency of bacterial infections (due to hypogammaglobulinemia!)

Drugs
- variety of conventional drugs, now some monoclonal antibodies
---- Rituximab - Anti-CD20 Ab
----Alemtuzumab - anti CD52 Ab
Who gets AML?
13000 cases in US in 2008
8,820 deaths (more fatal than CML & CLL)

Median age at diagnosis: 68

Risk factors:

Prior radiation

Prior chemotherapy
--Alkylating agents
--Topoisomerase II inhibitors

Benzene exposure

Prior history of MDS or a myeloproliferative disorder.

Familial syndromes
--Down syndrome
--Fanconi anemia
Unique Clinical Features
Faster onset than CML & CLL (eg 1-2 months)

Severe Anemia

Severe Neutropenia-->opportunistic infections (Staph, gram - rods, psuedomonas, invasive fungal infections like Aspergillosis)

Severe thrombocytopenia-->petechiae

Hyperleukocytosis
- stasis of blood flow w/ really high blast count
- mental status changes -> lethargy
- SOB
- diffuse bilateral pulmonary infiltrates
- Treatment: Leukophoresis before chemotherapy

Acute DIC
- most common in acute promyelocytic leukemia (APL) - often have acute DIC right from get go

Extramedullary Involvement
- CNS, gums
- skin - leukemia cutis - leukemic blasts in dermis of patients
Pathogenesis of Acute Leukemias
Class I mutations
- confer survival and/or proliferative advantage
ex) FLT-3

Class II mutations
- lead to block in hematopoietic cell differentiation
ex) PML-RARalpha
one of the 2 almost always found in ALL & AML
FLT-3
receptor tyrosine kinase
activating mutations seen in 30% of adult AML cases

can be either duplication or point mutation

result is deregulated proliferation
PML-RARalpha
retinoic acid receptor-alpha
bound by retinoids-->transcription of genes for differentiation

t(15;17) fusion gene PML-RARα central to the pathogenesis of acute promyelocytic leukemia (APL)

PML-RARα recruits the nuclear co-repressor (NCoR) complex and histone deacetylases to the RARα response element-->BLOCK TRANSCRIPTION OF GENES FOR DIFFERENTIATION

Give ATRA (all-trans retinoic acid) -->release inhibition of gene transcription and promote differentiation
Prognosis of AML
Age - older is worse

history of MDS or myeloproliferative disorder-->bad

prior chemo or radiation-->bad
-- associated w/ complex cytogenetic abnormalities, inc chromosome 5 & 7 deletion

PML-RARalpha - t(15;17) - good - KNOW THIS

t(8;21), inv(16) also good
deletions of chromosomes 5 or 7 - bad

Mixed lineage leukemia - 11q23 - bad
AML Treatment
30-40% cure rates in adults

Induction - reduce level of disease below level of detection -->complete remission & bone marrow works!

7+3 chemotherapy used for induction - less important
- 7 days continuous infusion of pyrimidine analog
- 3 days of anthracycline

Consolidation
- several cycles of high dose cytarabine (the pyrimidine analog)-->eradicate minimal residual disease

Stem Cell Transplant during 1st complete remission in patients w/ high risk disease
APL Treatment
cure rates 80% or more
induction7+3 chemotherapy + ATRA (all-trans retinoic acid)-->FORCE DIFFERENTIATION

then consolidation

stem cell transplant only for relapsed/refractory disease
ALL
less common than AML, fewer deaths

5,430 new cases in US in 2008
1460 deaths

median age at diagnosis: 11
Most common cancer in children

Peak incidence between ages 2 to 5.

Risk factors
Prior radiation
Prior chemotherapy
--Alkylating agents
--Topoisomerase II inhibitors
Familial syndromes
--Down syndrome
--Neurofibromatosis, etc
Unique Clinical Features of ALL
same symptoms as AML +
lymphadenopathy (like CLL)

mediastinal mass
- esp w/ precursor T-cell ALL

CNS
- have to prevent CNS relapse w/ intrathecal chemo or cranial radiation

Testicular
- strong predictor of CNS involvement/relapse in males

Tumor Lysis Syndrome
--from high cell turnover-->intracellular electrolyes released (urea, K+, phosphate)
--low Calcium
--high LDH
--acute renal failure due to uric acid in kidneys
--more common in ALL than AML
--Treatment: aggressive hydration, allopurinol (lower uric acid), rasburicase (break down uric acid)
Mutations in ALL
Type 1:
Bcr-Abl
t(9;22): The Philadelphia Chromosome.
33% of adult ALL cases.
4% of pediatric cases.
- worse prognosis (remember it was good prognosis in CML)

Characterized by the p190 Bcr-Abl product (as opposed to p210 of CML)

Constitutive activation of BCR-ABL→ ↑ cell proliferation.

Type II:
t(12;21): TEL-AML1
The most frequent cytogenetic abnormality in childhood B-cell ALL.
Associated with a good prognosis-overwhelming odds of cure
Fusion of TEL to AML1 -->abnormal recruitment of NCoR and HDACs → repression of AML1-mediated transcription-->no differentiation!
Prognosis for ALL
adults - 30-40% cure
kids - 80% cure

Age
<1, ≥10 → bad

Cytogenetics
Good
t(12;21)
Hyperdiploidy

Bad
t(9;22)
11q23 (MLL) translocations, t(4;11) in infant ALL.
Hypodiploidy

High white blood cell count
≥50x109/L→Bad
Treatment of ALL
very arduous & long
lots of cytotoxic drugs & corticosteroids

Induction:
prednisone
vincristine
daunorubicin
L-asparaginease

imatinib improves outcomes for those with Philadelphia Chromosome

Bone Marrow Transplant is treatment of choice for eligible high risk candidates (including Philadelphia Chromosome)
Distinguish Leukemias
Symptom onset over weeks to few months → acute leukemia; months to years → chronic.

Severe cytopenias → acute; less severe → chronic.

Blasts → AML or ALL; mature lymphs → CLL; spectrum of myelopoieisis → CML.

Remember more disease specific manifestations of the diseases
--DIC more likely in AML
--blasts and an anterior mediastinal mass in T-cell ALL
T-cell immunophenotype
CD 1, 2, 3, 4, 5, 7, 8
Granulocyte & Monocyte immunophenotypes
CD 13 - Granulocyte
CD 14 - Monocytic
CD 15 - Granulocyte

CD 33 is also myeloid
B cell immunophenotype
Nineteen and Early Twenties: B-cell phenotype
Be all you can “B” in the army at age 19-23
CD 19, 20, 21, 22, 23
Stem cell immunophenotype
34
Flow Cytometry
measure cellular properties as they move in stream past stationary detectors

identify phenotypes by using fluorescently labeled antibodies
WHO Classification of Leukemias
Builds on previous systems (i.e. FAB)
Recently revised (2008)
Diagnosed according the prominent cell type involved

Two major categories:
Acute: rapid onset, aggressive, usually poorly differentiated (blasts)

Chronic: insidious onset, usually less aggressive and more mature appearing

Acute Lymphoblastic Leukemia
Acute Myeloid Leukemia
Chronic Lymphoproliferative Disorder
Myeloproliferative Disorder
Myelodysplastic Syndrome
Chronic myeloid Leukemia Peripheral Blood & Bone Marrow Aspirate
bone marrow
-Hypercellular
-Increased M:E
-less variety of cell types (not as much trilineage hematopoiesis) but still much less homogenous than AML
Peripheral Blood
-Numerous immature
granulocytes (note they are still differentiated - not blast cells)
-Increased basophils, etc
CML vs Benign Neutrophilia
Left shift not as marked as in CML (may see bands but infrequently see
myelocytes and other immature forms)

Normal/Increased LAP

Neutrophils as opposed to variety
Leukocyte Alkaline Phosphatase in CML - increased or decreased?
decreased

benign process will have a normal or increased LAP
What is the CLL equivalent lymphoma?
small lymphocytic lymphoma
Other diseases associated with CLL
Autoimmune Hemolytic Anemia
Immune Thrombocytopenic Purpura
CLL Peripheral Smear and Bone Marrow Aspirate
Peripheral blood: CLL
-usually diagnostic
-lymphocytosis

Bone Marrow Aspirate: CLL
-variable involvement
-loss of heterogeneity
immature B cell marker
CD 10
CD 34
blast marker
combined with B cell marker-->B lymphocytic leukemia
Leukocyte Alkaline Phosphatase - what is it?
stain for Leukocyte Alkaline Phosphatase - quantitate stain

how do you determine between benign and leukemia?
leukemia - decreased LAP
reactive neutrophilia - increased
What is the best screening test for CML?
FISH
catch both cryptic and non-cryptic BCR-ABL mutations
When does BCR/ABL t(9;22) mutation occur?
in pluripotent stem cell
most cell lines still have BCR/ABL fusion gene

responsible for potential development of AML (80%) or AML (20%)
Flow Cytometry of CLL
CD 19
CD 5
CD 23
either kappa or lambda
CD 10
ALL
Follicular
Burkitt
CLL Cytogenetics; what to know
know some are better prognosis, others worse

deletion 11q23/mutation of MLL-->bad prognosis

deletion 17p13 (p53) -->bad prognosis
Deletion 11q23/mutation of MLL
bag prognosis in virtually all diseases/cancers
deletion of 17p13
includes p53
bad prognosis in CLL
AML morphology
>20% of blood or bone marrow is blasts

Auer rods - polymerized groups of myeloperoxidase granules
-- always AML for our purposes
Acute Myeloid Leukemia with Multilineage Dysplasia
follows MDS or MPD or MDS/MPD
genetics of Acute Promyelocytic Leukemia
t(15;17)
PML/RARalpha
5-8% of AML
Clinically ASSOCIATED WITH DIC up front
good prognosis after up front mortality

non15;17 translocations don't always respond to ATRA therapy

called promyelocytic b/c have technically moved out of blast phase "more mature"
t(15;17)
genetic mutation in ALL-->APL
PML/RARalpha
PML/RARalpha
t(15;17) translocation in ALL-->APL
Retinoic Acid Receptor alpha
Normally heterodimerizes with retinoid X receptor (RXR) and interacts with nuclear corepressor complex to transcriptionally block granulocyte differentiation

Retinoic acid releases the complex and allows progression of differentiation past the promyelocyte stage
has DIC
suspect/known ALL
has no CD34 or HLADR
what do they have now?
what should you do?
APL
immediate ATRA treatment
cytogenetics of AML
50% have no clonal cytogenetic abnormality

FLT3 - known mutation - poor prognosis
- constitutively active tyrosine kinase
- most frequent molecular abnormality in AML

NPM1 - also know
FLT3
most common molecular abnormality in AML

contitutively active tyrosine kinase

poor prognosis

internal tandem duplication (most common) or point mutation
is it possible to have "aleukemic" leukemia?
Yes,
ALL usually has leukocytosis but can be aleukemic
bone marrow is packed but they're not circulating cells

Usually present with pancytopenia
What's more common to see in T-cell ALL?
mediastinal or soft tissue mass

higher risk, worse prognosis

blasts that express T cell markers only
What if you suspect acute leukemia? what's the first step?
Immunohistochemistry
myeloid or lymphoid?
TDT & CD34 stain positive - immature cells
all cells on bottom left indicate early lymphoid precursor
*** check on this - thought granules stained -->myeloid
what type of lymphoma is more common with HIV?
B cell
favorable prognosis for adult ALL?
hyperdiploidy
t(12;21)
unfavorable prognosis for adult ALL?
t(9;22) - WORST CYTOGENETIC ABNORMALITY IN ALL
abn(11q23)
hypodiploidy