• 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

1/553

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;

553 Cards in this Set

  • Front
  • Back
one word for each phase of hemostasis
primary - platelets
secondary - fibrin
tertiary - plasmin
primary screening test for platelet function

if abnormal...
bleeding time

everyone with thrombocytopenia will have long bleedings times

next step: platelet aggregation studies with ADP, epinephrine, collagen, ristocetin as agonists
dense granules in platelets contain
calcium, serotonin, other small molecule mediators of platelet function
alpha granules in platelets contain
protein mediators of platelet function

(fibrinectin, platelet factor 5, thrombospondin, vWF)
can have prolonged bleeding times (platelet function test) due to
congenital
drugs
alcohol
uremia
hyperglobulinemias
fibrin/fibrinogen split products
thrombocythemia
cardiac surgery
a congenital platelet adhesion disorder

inheritance
problem
test
Bernard Soulier disease

AR
abnormal GPIb-IX complex (receptor on platelets for vWF) so the monolayer of platelets doesn't form

test: ability to aggregate platelets in presence of ristocetin
delta-storage pool disease
normal platelet adhesion, but you don't release dense granules (ADP, serotonin, calcium) on activation to aggregate more platelets

--> MILD bleeding disorder
Gray platelet syndrome
alpha granules are leaky, so you don't get release of protein mediators of platelet aggregation (thrombospondin, vWF, fibronectin, etc)

--> MILD bleeding disorder
problems with platelet aggregation that lead to mild bleeding disorder
delta-storage pool disease
Gray platelet syndrome
Glanzmann's thrombasthenia

inheritance

consequence
lack of GP IIb/IIIa (fibrinogen receptor) on platelets...platelets cannot aggregate in response to usual stimuli

AR

SEVERE bleeding
Scott syndrome
loss of shufflase enzyme that flips PM inside out...so the acidic phosphilipid that form receptors for Factor 5a and 8a aren't produced...can't localize the clotting cascade to right place
drug induced platelet function defects cause by
alcohol
prostaglandin synthetase inhibitors (aspirin, NSAIDs, phenylbutzone)
ADP receptor inhibitors clopidogrel, ticlopidine, prasugrel)
causes of decreased platelet production

tx?
leukemia**
aplastic anemia
metastatic carcinoma
drugs
radiotherapy
primary marrow disorders

--> decreased megakaryotes, but life span of the platelets is normal

tx: good response to transfusion
causes of increased destruction of platelets
immune mediated:
ITP
lymphoma
lupus
drugs

note: platelet life span 12-24 hrs instead of 7-10 days; often see increased megakaryocytes and macroplatelets
*poor response to transfusion

consumption:
DIC
TTP
HUS

septicemia
ITP pathogenesis
IgG autoantibodies opsonize platelets for removal by macrophages

no good dx test...it's a dx of exclusion
ITP tx
steroids - first line
splenectomy - second line
HIV-associated thrombocytopenia:
pathogenesis
tx
early HIV: immune complexes sit on platelet and get destroyed in innocent bystander reaction
late HIV: marrow infiltration by fungus/TB/mycobacteria and pancytopenia

ART, esp. AZT
if more than one clotting factor is abnormal, get a
inhibitor screen
how are clotting tests prepared
blood collected in sodium citrate, a weak calcium chelator; amount designed to drop calcium to a level where spontaneous clot will not occur

RBCs and platelets centrifuged off, leaving plasma

PT: 50% patient plasma, 25% TF and phospholipid species, 25% calcium chloride (to activate clotting process) --> time to clot measured (10-14 s normal)

aPTT: 50% patient plasma, 25% phospholipid and surface active agent, 25% calcium chloride --> time to clot measured (normally 25-35)
clotting factor deficiency tests done only

how is it done
if aPTT or PT abnormal

50% patient plasma with 50% plasma deficient in a single clotting protein
PT abnormal implicates factors
2, 5, 7, or 10
aPTT abnormal implicates factors
12, 11, 9, 8 abnormal
von Willebrand disease

prevalence

inheritance
lack of vWF --> decreased factor VIII activity, defect in platelet adhesion;
most common congenital bleeding disorder; variable severity, even within one family; mild bleeding

0.8-2%

AD
vWF made in

stored in

secretion
endothelial cells and megakaryocytes

multimers stored in Weibel-Palade bodies in endothelial cells

constitutive and stimulated secretion
vWF will have a prolonged ___
aPTT (b/c of low Factor VIII activity)

also low ristocetin cofactor activity, increased bleeding time

but 20% of the time all of these will be normal in heterozygotes, so you have to test multiple times
types of vW disease
Type I: most common; quantitative defect; defect due to non-sense mutation; 50% normal levels

Type II: qualitative defect
IIa - no multimer formation (normal levels of factor VIII but adhesion defect)
IIb - decreased multimers, decreased platelets
IIc - hundred of defects in multimerization
IIn - defect in factor VIII binding - looks a lot like hemophilia

Type III: severe quantitative defect (double heterozygotes or homozygotes) - usually lack factor VIII and platelet adhesion properties
Hemophilia __ is more common

tx:
A (85%)

A = Factor 8
B = Factor 9
(clinically indistinguishable except by factor analysis)

tx: lethal without replacement therapy

incidence hasn't changed over time, so many mutations are new
strata of hemophilia severity
mild: >5% factor - bleeding with significant trauma

moderate: 1-5% factor - bleeding with mild trauma, occaional spontaneous hemarthroses

severe: <1% factor level - spontaneous hemarthroses and soft tissue bleeding

**within each kindred, similar severity in disease (unlike vWF)
**thousands of different mutations
vWF are __ in hemophiliacs
normal

if you add hemophiliac's blood to vWD patient, factor VIII levels go up b/c the vWF stabilizes what VIII they are making
in hemophilia A, bleeding time is __, aPTT is ___
normal (initially, but then plug breaks and they rebleed)

prolonged
in vW disease, bleeding time is ___ and aPTT is ___
prolonged

prolonged
Factor XI deficiency

tx:
Hemophilia C aka Jewish hemophilia

90% ashkenazi

mild bleeding (usually with procedures); #1 cause of lawsuits

AR

tx: fresh frozen plasma good for one week
causes of acquired clotting disorders
Vitamin K deficiency
liver disease
DIC
coumadin
heparin
Vitamin K deficiency bleeding almost always seen in __ because ___

lab abnormality

tx
hospital patients

need malnutrition and reduced gut flora

PT prolonged first due to factor VII's short half-life

tx: replace Vitamin K (response in 1-2 days)
liver disease and abnormal hemostasis

tx:
decr. synthesis of vitamin K dependent proteins

decr. clearance of activated proteins

incr. fibrinolysis secondary to decr. antiplasmin

dysfibrinogenemia secondary to synthesis of abnormal fibrinogen

increased fibrin split product

increased PT, aPTT
decreased platelets (hypersplenism)

Tx: replacement only if they are having a procedure or an active problem
Tissue Factor Pathway Inhibitor (TFPI) complexes with ___

function:
7a/10a/TF

inactivates 10a
antithrombin III is a classic serine protease inhibitor; it inactivates ___

this is normally a very slow reaction, but it's sped up by ___
9a, 10a, 11a, thrombin

heparin (which floats away and continues working after the reaction, as a true catalyst)
in addition to leading to fibrin formation/inactivating 5a and 8a/activating platelets, thrombin but it also binds to ___ on the surface of ___

this activates ___
thrombomodulin

endothelial cells

protein C
Protein S function
cofactor for Protein C...together they inactivate Va and VIIIa
though you usually need <10% of clotting factor to start having clinical problems, deficiencies of <__% of ATIII, Protein C, Protein S, thrombomodulin, plasminogen can

heterozygous protein deficiency -->

homozygous protein deficiency -->
50

increased venous thrombosis, occasional increased arterial thrombosis

neonatal purpura fulminans, fibrinogenolysis, chronic DIC (most don't survive first year of life; luckily it's rare)
dominant phenotype of anticoagulant protein deficiency
increased venous thrombosis
young age of thrombosis
no predisposing factors
increased thrombin generation in all patients
positive family history
recessive phenotype of anticoagulant protein deficiency
no family history
neonatal purpura fulminans in offspring
increased thrombin generation (similar to that seen in dominant)
why is it that the same mutation in anticoagulant protein can cause a dominant phenotype in one person and recessive in another
Activated Protein C resistance

failure of activated Protein C to prolong aPTT

98% of the time it's due to Factor V Leiden mutation
Factor VIIIa is inactivated by ...
APC
Protein S
Factor V ******
phospholipid

thus, Factor V Leiden deficiency leads to problems inactivating factor VIII as well
acquired hypercoagulable states
1. inflammatory diseases
2. nephrotic syndrome
3. anticardiolipin antibody syndrome
4. malignancy
5. immobilization
6. TTP
7. DIC
8. OCT
9. prosthetic valves
10. PNH
11. myeloproliferative diseases
12. atherosclerosis
13. surgery
14. diabetes mellitus
Factor V Leiden mutation
Arg 506 --> Gln

(this is at a cleavage site, so activated Protein C can't inactivate 5a properly)

found in >98% of patients with activated protein C resistance

5-20% of caucasians are carriers

increases risk of venous thromboembolism....4x in heterozygotes, 10x in homozygotes

in combination with other deficiencies (protein C, protein S, ATIII, plasminogen) has a synergistic effect

so in 50% of patients with dominant phenotype, they have both a mutation in the protein and a factor V Leiden mutation
Prothrombin G20210 --> A mutations leads to
increased prothrombin production, higher risk of thrombosis during pregnancy or venous thromboembolic disease

1-3% of Northern European population
one of the most potent prothrombotic disorders
anticardiolipin antibody syndrome (lupus anticoagulant)
why is inflammatory disease a hypercoagulable state?
increased C4b binding protein binds Protein S and removes it as a cofactor for Protein C

increased IL-1 and TNF downregulate thrombomodulin (via endocytosis and decreased transcription) so that thrombin becomes procoagulant
in nephrotic syndrome, ___ proteins are lost, but ___ is big enough to not get through
Protein C, protein S, ATIII

C4b (which binds any remaining protein S)

thus, nephrotic syndrome (and protein-losing enteropathy) are pro-thrombotic states!
someone with anticardiolipin antibody may have
lupus (only 30% of the time)

false + RPR (measures antiphospholipid antibodies)

spontaneous abortions

bleeding in vitro, thrombosis in vivo (try using different phospholipid source in vitro, e.g. platelets)
-true antigen is source of controversy
-no good assay to test...it's a clinical diagnosis
dx of anticardiolipid antibody syndrome
at least 2 abnormal tests more than 12 weeks apart
+ clinical syndrome of either:

3 1st trimester abortion, 1 2nd/3rd trimester abortion, or unexplained venous/arterial thrombotic event
causes of subacute/chronic DIC
acute leukemia
carcinomas (probably release of TF from tumor)
hemangiomata
aortic aneurysm
?liver disease
plasmin activation
12a --> kallikrein --> plasmin

urokinase --> plasmin

tPA --> plasmin
vitamin K dependent co-factors
2, 7, 9, 10
protein C and S
Fibrinogen split products (elevated in ___) include___

besides freeing up the lumen, they also
DIC

Fragment X (big core, largely insoluble)
Fragment D (<50% of end, soluble)
Fragment Y (>50% of end, soluble)
Fragment E (disulfide knot, soluble)

inhibit further fibrin polymerization
defibrination mechanisms
release of tissue procoagulants:
-tumor
-fetus/placenta
-prostatic cancer
-pancreatic cancer
-WBC
-RBC
-shock

damage to vascular tree:
-septicemia
-aortic aneurysm
-hemangioma
-tumor emboli

decreased clearance
-liver disease

brain tissue is very high in TF
lab values in acute DIC

1. PT
2. aPTT
3. fibrinogen
4. thrombin time
5. factor levels
6. platelet count
7. RBC fragmentation
8. fibrin split products
labs

1. slightly to grossly prolonged
2. usually prolonged
3. usually low (consumption)
4. usually prolonged (b/c fibrinogen is low)
5. variable
6. usually low (platelets consumed in clots)
7. sometimes present (fibrin strands clothesline them)
8. usually present (D-dimer assay measures cross-linked fragment D)
DIC therapy
depends on primary manifestation
1. thrombosis --> anticoagulant therapy
2. bleed --> replacement therapy
--cryoprecipitate (give fibrinogen)
--fresh frozen plasma (gives other factors)
--platelets

TREAT UNDERLYING DISEASE WHEN POSSIBLE (DIC will almost always go away if you can...of course something like pancreatic cancer will be hard to treat)

heparin is rarely indicated, unless the underlying disease requires you to shut off the thrombotic process
lupus anticoagulant is a bad name for __ because ___
anticardiolipin antibody

only 30% have lupus, and it's not associated with bleeding except in rare cases...mostly associated with arterial or venous thrombosis (mechanism unknown)
cause of acute DIC
shock
sepsis (esp. Gram negative)
allergic reactions
mismatched transfusion
obstetrical problems (*always DIC until misproven*)
trauma
burns
extracorporeal circulation
acidosis
purpura fulminans
generally, subacute DIC --> __; tx with ___

acute DIC --> __ because ___;
clotting problems (DVT, PE, arterial)
anticoagulants

bleeding
DIC leads to consumptive coagulopathy...when you rev up the system to this extent by damage or exposure to large amounts of endotoxin, you cause thrombotic problems but you also consume your clotting factors in the process
in acute DIC you see decreases in both
coagulants and anticoagulants

(severity may relate to levels of anticoagulants)
the first mutation in most men with prostate cancer

others that may occur subsequently
GSTP1 (protects against oxidative damage) - normal part of aging for men, and a clear player in almost every case

RNASEL, MSR1 (germline mutations)
the mutation that marks the transition from normal prostate epithelium to prostatic intraepithelial neoplasia
decrease in NKX3.1 (tumor suppressor)

--> predisposes to DNA damage and increases cell survival
mutation present in 60% of prostate cancer, marking transition from PIN to localized prostate cancer (invasive cancer)

transition from localized to metastatic?
ETS translocation (AR-dependent), P27

PTEN, P53, RB, MYC
single clear driver of castration-resistant cancer
mutation in AR gene
between the ages of 50 and 75, risk of prostate cancer increases __ fold
7

(colon cancer only 4 fold)
who has most prostate cancer?
blacks (earlier and steeper slope for incidence and mortality)
risk factors for prostate cancer
clearly a disease of aging

also, western diet --> earlier onset, higher risk
incidence of prostate cancer has been going ___

mortality has been going __, and this is due to ___
up (and a bump is due to PSA screening)

down

something, but NOT screening
PSA is a

why is it an imperfect test?
serine protease that liquifies seminal coagulum

there is an age-dependent leakage of PSA, and can also see increases in BPH and prostatitis
PSA is a good indicator of
cancer recurrence post-tx
refinements in PSA test
PSA density (amt. in blood, size of prostate via ultrasound)

free vs bound PSA

PSA velocity (how fast it rises - this is completely useless in an undiagnosed male, but in a dx man, rapid rise is worse)
for prostate cancer, 10-yr survival with:
organ confined disease___
regional extension ___
distant metastases ___
69%
38.5%
15%
screening has picked up significant prostate cancer, but has not significantly improved cause-specific death at __
10 years out

(European study has shown benefit 13 years out for 50-59 and 65-69 y.o.)
most powerful prognostic index for newly diagnosed prostate cancer
Gleason score

a 1cm prostate cancer can be incredibly diverse in severity
Gleason grade of 1 =

5 =
well formed, well circumscribed glands with nice nuclei

almost no glandular architecture, palisading, pleiomorphic nuclei, cells that invade the stroma

not until you get to the higher grades that prostate cancer has <50% impact on mortality
whether we should even tx Gleason grade __ is still a major question
6
in prostate cancer (and in normal aging), you have multiple loci that are microscopically malignant, but
usually only one clone is the bad one
-there is therefore sampling error in the biopsy

if a 90-yr-old male dies of other causes, he has a 90% chance of having cancer in the prostate
prostate cancer staging:
T1a =
T1b =
T1c =

T2a =
T2b =
T2c =

T3, T4

N0, N1

M0
M1a
M1b
M1c
T1's are incidental histology findings
T1a = <5% of tissue
T1b = >5% of tissue
T1c = PSA detection (70-75% of diagnosis these days)

T2 is abnormal rectal exam
T2a = <half lobe
T2b = >half lobe
T2c = both lobes

T3, T4 = invasive local disease (firmness in seminal vesicles would indicate T3 disease)

N0, N1 = +/- regional nodes

M1a = distant mets
M1b = bone mets
if you feel a nodule on rectal exam, it has a __% chance of being malignant
50%
treatment of primary prostate cancer, locally invasive
Watchful waiting
Radical prostatectomy (highest rate in low-grade cancer)
External beam radiation (highest in high-grade cancer...becoming less popular)
Radiation seed implantation (Brachy...becoming more popular)
in prostate cancer, increased __ rates result from lead time bias
survival
a neurovascular bundle runs ___ to the prostate and supplies the ___
posterior

erectile mechanism
men undergoing radical prostatectomy have 30 year survival that matches the general population; is that because the surgery is really good or because it didn't make a difference
we don't know

one european study showed that watchful waiting was worse that prostatectomy, but that was in symptomatic patients, not screen-detected patients; the surgery was especially beneficial in those <65, but no difference for >65
confined prostate cancer > __ > __ > __
extracapsular extension > seminal vesicle invasion > LN+
estimate that __% of screen-detected cancers are unnecessarily found
47
after prostatectomy, get PSA every

cancer recurrence if

salvage radiotherapy...
6 months

2 consecutive rises

can be done but it markedly increases incontinence and GUARANTEES impotence
urinary incompetence post prostatectomy is more common in
1. older pts
2. comorbidity
3. less prolific centers (slightly)
all radiation for prostate cancer is given with a small margin, so you get some
bladder, rectum, proximal urethra, seminal vesicles
what is IMRT
intensity modulated radiation therapy

(a strategies to improve the risk benefit ratio...if MR can identify regions of more cancer, IMRT can tailor the dose accordingly)
advantage of brachytherapy over external beam radiation in prostate cancer

problems?
external beam is 6-8 wks, half hour every day

brachy is a 1 time implantation

proctatitis, obstruction, works in a small gland, but not done in a large gland

not shown to be any more effective than external beam
major complication of radical prostatectomy?

radiation therapy?
urinary strictures (17%)

radiation proctitis (18%)
in terms of physical composite score, there is a long-term cost to ___therapy
brachy
worst prostate cancer tx in terms of incontinence

all have a pretty bad effect on erectile function, but __ is the worst
RP (10% vs 4%)

RP (79.6% vs 61.5%)
tx of locally advanced prostate cancer
GnRH agonist (leuprolide, goserelin) + radiation
orchiectomy
finasteride (propecia...doesn't work)
adrenal blockade (ketoconazole)
antiandrogens (bicalutamide, flutamide)
when you give a GnRH agonist, there is a
brief spike in testosterone, then the pituitary production of LH shuts off, then you have chemical castration

these are injections that you have to keep getting
__% of circulating androgens come from the adrenal gland
10%
__ is responsible for male pattern baldness
5HT (a prostate-specific androgen)
finasteride works for __ but not __
BPH, baldness

prosate cancer
can you make more people candidates for prostatectomy by shrinking it with hormonal therapies?
no
greatest advance in tx of prostate cancer
addition of hormonal therapy to simultaneous radiation therapy for locally advanced disease

makes a huge difference! on any parameter
90% of the time, prostate cancer metastasizes to the __ first
bone
tx of metastatic prostate cancer

mean disease-free survival is
hormonal therapy (it's hormone sensitivity is retained for some period of time...study shows big benefit of early androgen ablation)

2-2.5 years
morbidity of androgen ablation
osteoporosis and fractures
fatigue
diabetes
cardiovascular risk
the hormone therapy used in metastatic disease

second-line hormonal therapy

management of mets
anti-androgen + GnRH agonist
(it's worth it)

-anti-androgen withdrawal (in case it's agonistic
-ketoconazole (adrenal blockade)
-adrenal blockade with steroids, aminoglutethimide

-prophylactic radiation in weight-bearing bones
-pain management (narcotics, NSAIDs, radioactive nuclides that are calcium analogs)
-second line hormonal therapy
-chemotherapy - last line...doesn't really help except maybe docetaxel)
androgen ablation is beneficial to __ prostate cancer but harmful for ___
poorly differentiated

moderately differentiated
in metastatic prostate cancer, androgen ablation should not be discontinued, even after
progression on a GnRH agonist

(there is a component of the tumor that clearly benefits from that therapy b/c prostate cancer will always, until death, depend on androgen receptor activity)
androgen receptor has these domains
DNA binding (Zn fingers)
steroid binding
transcription regulation

assembly of transcriptional complexes in front of genes that have androgen response elements
castration resistant prostate cancer is characterized by
1. AR gene amplification (ligand independence)
2. AR mutations (tunr anti-androgens into androgens)
3. AR phosphorylation (allows activation)
4. AR coactivator over expression
5. incr. expression fo androgen synthetic enzymes
6. alternate splicing to generate ligand independence

tells us that prostate cancer is always dependent on AR
what waste does the blood remove
CO2
nitrogenous waste
cellular toxins
normal RBC volume

diameter

thickness
80-100 femtoliters

8 microns (about the size of lymphocyte nucleus)

2 microns
automated counters measure __ using ___
size, number

impedance
*anisocytosis =
variable size of RBCs
term for variable sizes of RBCs
anisocytosis
*poikilocytosis =
variability in the shape of RBCs
variability in shape of RBCs
poikilocytosis
smaller RBCs are thinner and therefore look __ colorful
less
normal hemoglobin ranges
female: 12-16

male: 13.5-17.5

(androgens tend to stimulate hemoglobin production)
red cell counts and __ levels are often related to each other
hemoglobin
normal reticulocyte count
0.2-2.0%

(retics are the 1st day of a red cell's life)
normal platelet count
150,000-400,000
*definition of anemia
decrease in the number of circulating red blood cells

(most common hematologic disorder by far...more common than all of the others put together...for the most part it's a secondary disorder...usually not a problem with their blood cells)
*most common hematologic disorder
anemia

(by far)
*causes of anemia
1. blood loss
2. decreased RBC production (marrow failure)
3. increased RBC destruction (hemolysis)
how can you distinguish between hemolytic anemia and marrow failure
reticulocyte count
(decreased in marrow failure, increased in hemolysis)
*single most important marker of RBC production
retic count

(absolute value more accurate than percentage)
reticulocytes still have small amounts of __ present in them

tend to stain somewhat __er

slightly smaller/larger?

can be detected using
RNA (undergo removal of RNA on passing through spleen in 1st day of life)

blue

larger

supravital stain which stains the RNA directly
*how to calculate the absolute value of reticulocytes
retic % x RBC count

*more accurate way to assess body's response to anemia, rather than percentage
normal up to 100,000/ microleter
*if the anemia is a marrow failure due to cytoplasmic protein production problem, the anemia is usually __cytic

if a problem in nuclear division/maturation, it's usually __cytic
normo or micro

macro
anemia due to cytoplasmic protein production problems...name the types of disorders
disorders of globin synthesis
disorders of heme synthesis
causes of disorder in heme synthesis
1. decreased iron (if you don't have iron, in a utilizable form, you can't make RBCs)

2. iron not in utilizable form (anemia of chronic disease...iron can't get into the cells that produce RBCs)
most common cause of anemia

2nd most
anemia of chronic disease

iron deficiency
active form of iron

in __ conditions it's easy to maintain this state
Fe2+ (ferrous)

Fe3+ (ferric) cannot trasnport electrons or bind O2

anaerobic (but iron donates electrons readily to oxygen --> ROS)...green plants wiped out 95% of species and the ones that survive were the ones that could limit their exposure to iron
*iron compartments:

hemoglobin iron __%
storage iron (ferritin, hemosiderin) __%
myoglobin rion (critical for muscle metabolism) __%
labile pool and other tissue iron (important for electron transport and cytochrome sstem) __%
transport iron __%
Hb 67%
storage 27%
myoglobin 3.5%
labile/other tissue 2.5%
transport 0.08%
*senescent RBCs are eaten by macrophages in reticuloendothelial system, and the iron can either be:
1. bound to ferritin (IC storage)
2. bind transport molecule, go across membrane, bind transferrin in circulation (very tightly); transferrin transports iron back to bone marrow to RBC precursor where it binds to a specific transferrin receptor; complex is internalized, iron is released and incorporated into heme, heme into hemoglobin

the transferrin + receptor is taken back to membrane and transferrin released back into circulation
how is iron released from transferrin intracellularly
vacuole fuses with lysosome, acidified, which releases iron from lysosome into cytoplasm
most comon cause of anemia in men and post-menopausal women
GI bleed
*causes of iron deficiency anemia
GI bleed
menstrual blood loss

pregnancy
infancy (rapid growth rate)
adolescence (rapid growth rate)
polycythemia vera (increased production of red cells)

tropical sprue (malabsorption)
gastrectomy or bariatric surgery (absorption of iron requires an acid environment)
chronic atrophic gastritis (loss of parietal cells and acid production)

diertary inadequacy (rarely sole cause)
__ of iron in diet is soluble; __ of that actually gets taken up into intestinal cells; about __ of that is absorbed into system
1/2

1/2

1/4
how does iron get from lumen into GI cells? via __

how does iron get from intestinal cells to blood stream? via __
DMT-1

ferroportin
*__ measures the amount of iron present in the crypt cells and either stimulates production of more __ which leads to increased iron absorption or down-regulates production of those while up-regulating ___
HFE

DMT-1, ferroportin

ferritin (which leads to iron being bound in GI cell, which then gets sloughed into the lumen of the intestines)
iron production is regulated at the _NA level
RNA

(based on iron binding to an iron regulatory protein)

the IRP, in a low iron state with 3 irons attached, binds the stem loop of ferritin so that translation cannot occur or binds the stem loops of transferrin and DMT-1 such that it stabilizes those mRNAs

in a high iron state, 4 Fe's are attached to IRP and it can't bind the loops
can't be iron deficient until your __ is used up
storage pool
*progression of findings in iron deficiency anemia
1. stainable iron, bone marrow aspirate (loss of storage pool)
2. serum ferritin low
3. desaturation of transferrin
4. low serum iron
5. transferrin production increases
6. microcytosis/hypochromic/anisocytosis/poikilocytosis
7. anemia

reverse in same order
changes in iron compartments in anemia
hemoglobin iron is low, but now 87% of iron

storage iron is now 0%

rest is the same
*sx of iron deficiency
1. fatigue (can be out of proportion)
2. atrophic glossitis
3. pica
4. koilonychia (nail spooning)
5. esophageal web (Plummer Vinson syndrome, rare)
**tx iron deficiency anemia

side effects
oral iron replacement (100-900 mg)
+/- Vitamin C (Fe requires acid environment for absorption)

(takes a long time, at least 2-3 months, to replace since iron is poorly absorbed)

side effects: constipation, nausea, GI cramps

parenteral iron possible but problematic to allergic reactions...may have to do it if they had gastrectomy
response to iron replacement therapy
initial response takes 7-14 days (have to synthesize RBCs from earliest precursor)
modest reticulocytosis (7-10%) - modest b/c of difficulty in getting enough iron into the body

need 6 months of therapy beyond correction of anemia (which usually takes 2-3 months) b/c you need to replete stores, assuming no further loss of blood/iron in which case you need to be treated longer
__% of population has hemochromatosis

inheritance

defects in
2%

AD (varying degrees of penetrance)

HFE, sometime in DMT-1

(the HFE sensor is messed up, reads like you have constantly low levels...defect in HFE --> decreased iron uptake by crypt enterocytes --> decreased IC iron --> increased DMT-1 and increased absorption)
sequence of events in hemochromatosis
1. increased ferritin
2. increased transferrin saturation (normal 33%, >60% is a marker of disease, >95% can lead to free iron

albumin does buffer free iron a little bit
**IRREVERSIBLE diseases of iron deposition in tissues:
1. skin darkening
2. endocrinopathies (diabetes, hypothyroid, hypopituitarism)
3. liver damage (cirrhosis, HCC)
4. cardiac damage (cardiomyopathies --> CHF)
*when do you use iron chelation in iron overload?
generally for transfusion-induced iron overload (can't phlebotomize them)
**anemia of chronic disease is generally:

Hb:
Hct:

usually __cytic

retic count is

bilirubin is

EPO levels
mild

10
30%

normocytic (30% microcytic) with mild anisocytosis or poikilocytosis

normal (inappropriately low...also somewhat shortened RBC lifespan)

normal (b/c destruction is relatively normal)

increased but blunted for degree of anemia
*anemia of chronic diseass diseases:
thyroid disease

collage vascular disease (RA, SLE, polymyositis, polyarteritis nodosa)

IBD

malignancy

chronic infectious disease (osteomyelitis, tubercuosis)

Familial mediterranean fever
iron compartment profile in anemia of chronic disease
Hb is 40% (normal amt)
storage is 54%

can't get it to where it needs to go

decrease in transport iron b/c it's stuck in the storage pool
*what's the pathogenesis of anemia of chronic disease
TNF and IL-1 --> increased ferritin and decreased transferrin

also have decreased iron absorption in gut b/c hepcidin (upregulated in ACD) binds to and causes internalization of ferroportin leading to decreased iron uptake

iron stays trapped in the macrophage with ferritin, so it can't get back to bone marrow to red cell precursors
hepcidin
antimicrobial polypeptide produced in the liver

binds to and causes internalization of ferroportin --> decreased iron uptake from GI tract

upregulated in inflammation, down-regulated in iron deficiency

part of decreased iron absorption in anemia of chronic disease
*in iron deficiency:
serum iron is __
transferrin is __
ferritin is __

in anemia of chronic disease:
serum iron is __
transferrin is __
ferritin is __
low
increased (compensatory)
low

low
low
increased
in iron metabolism disorders, the problem is with RBCs and
no other cell types, really
tx for anemia of chronic disease
replacement therapy
correction of underlying cause
which is more dangerous, iron excess or iron deficiency?
iron excess

(in treating iron deficiency, you really can't overshoot unless you give it IV)
*most important cause of iron deficiency in men and post-menopausal women is
colon cancer! (in the US)

(b/c of blood loss)
white cell life span
12-24 hrs
platelet life span
7 days
__ is one of the most highly active organs in the body
bone marrow

any slowing of DNA production leads to bone marrow failure
any slowing of __ leads to bone marrow failure
DNA production
hallmark disease for marrow failure
megaloblastic anemia
in megaloblastic anemia,
hemoglobin production is __
defect is in __
affects __ cells
LDH is __
serum Fe is __
serum B12 or folate __
normal

nuclear replication and division(--> anemia)

all marrow derivatives (-->anemia, leukopenia, thrombocytopenia, reticulocytopenia) = pancytopenia

elevated (RBCs don't have mitochondria, make lactate, have LDH to detoxify)

normal or elevated (defect in utilization b/c of block in nuclear maturation and division)

low
trademark cells in megaloblastic anemia
oval macrocyte > 100 fl
hypersegmented neutrophil
structure of folic acid

bridge is
pteridine ring --> PABA --> glutamic acid tail

N-C-C-N b/w pteridine and PABA
all organisms need folic acid; bacteria in particular utilize folic acid for DNA production, and some abx work at this part
the N-C-C-N bridge between pteridine and PABA

prevent bacteria from growing, which kills them

mammals don't have the mechanisms to synthesize this
*folate's major role is to
transfer 1-carbon fragments to things
if you take methenyl THF and oxidize it, you get

if you take methenyl THF and reduce it, you get

if you then break a bond you get
formyl THF - one important for purine biosynthesis (not the most important one)

methylene THF - one important for thymine synthesis...major one for DNA synthesis (inhibited by cancer drugs)

methyl THF - the primary transport form of folic acid...what's in your blood
folate is readily absorbed from diet in methyl THF form...when that gets into cells its converted to THF by ...

then ...
an enzyme that uses B12

you can add the polyglutamate tail which traps it inside the cell; that then goes to methylene THF
***methotrexate (chemo agent) blocks
dihydrofolate reductase
(which converts DHF polyglutamate back to THF polyglutamate)

so does trimethoprim
*causes of folate deficiency
folate poor diet
--alcoholism
--severe poverty

increased folate requirement
--pregnancy
--severe hemolytic anemia
--severe psoriasis (increased skin cell production)

drug therapy (that's actually how chemo works, so you don't want to give folic acid b/c that could block the effect of the drug)

malabsorption
--tropical sprue
--celiac
manifestations of folate deficiency
megaloblastic anemia
glossitis/stomatitis (often lose their taste buds as a result)
GI malabsorption secondary to impaired GI epithelium (rare)...since turnover is rapid, it's susceptible
*B12 functions
1. cofactor for conversion of methyl-THF to THF
2. methylation of myelin
3. cofactor for conversion of methylmalonyl CoA to succinyl CoA (important in maximizing ox-phos)
*cobalamin structure

what are the different beta groups
4 pyrrole rings (like heme), except it has one straight C-C bond
-cobalamin is the coordinating metal instead of Fe

CN (inactive; can be converted)
OH (inactive; can be converted)
methyl (folate metabolism)
adenosyl (mutase activity - important for methyl malonyl CoA conversion to succinyl CoA)
when __ cobalamin converts methyl THF to THF, __ is converted to __
methyl

homocysteine

methionine
a more sensitive way to measure B12
homocysteine levels
methylmalonic acid levels

(caveat, everyone with renal insufficiency will have high levels of these b/c they're excreted by the kidney)
if you're deficient in B12 OR folate, you inhibit

if you're B12 deficient but not folate deficient, you'll have high levels of
conversion of homocysteine to methionine

methylmalonic acid
*high levels of methylmalonic acid can be seen in
B12 deficiency
renal insufficiency
maple syrup urine disease
*__ is only present in animal proteins, so vegans are prone to deficiency and should be supplemented
B12
*GI absorption of cobalamin
GI mucosa makes an R protein that binds B12 and gets it through stomach; as B12 goes into duodenum, R is stripped off, IF (made by parietal cells) binds; in terminal ileum, the IF-B12 complex goes into cell; complex splits, transcobalamin II binds B12; that complex circulates then goes into hematopoietic precursor cells where they split

there's another transport protein called transcobalamin I that isn't able to get into the hematopoietic precursors...we don't know what it does other than screw up our assays
*causes of cobalamin deficiency
gastric failure
--pernicious anemia (loss of parietal cells)
--total gastrectomy

ileal failure
--regional enteritis (Crohn's)
--ileal resection (due to Crohn's)
--tropical sprue (infection of ileum)

competing organisms
--bacterial overgrowth (blind loop...they love eating B12)
--diphyllobothrium latum (gefilte fish parasite)
*pernicious anemia is ___ destruction of ___

particularly associated with this disease
autoimmune

parietal cells

(antibodies against parietal cells, IF...achlorhydria is universal)

hashimoto's thyroiditis
increased incidence of pernicious anemia in
American blacks, northern europeans
*why does B12 deficiency lead to megaloblastic anemia
really it's the deficiency in intracellular folate...because you can't get to polyglutamate THF (folate) so the methyl THF leaves the cell freely
*manifestations of cobalamin deficiency
much like folate deficiency:
-megaloblastic anemia
-stomatitis/glossitis
-GI mucosa alterations

BUT there are also the demyelination effects (dementia, psychological disturbance, loss of posterior and lateral columns (combined system disease...lose vibration and position sense first)), so you can't just treat it with folate, which would correct the others...treat with B12

neurological disease is stabilized with treatment but usually not reversed
in subacute combined degeneration they lose
position sense and vibration sense
*sequence of events in cobalamin deficiency

is this process fast or slow
1. rise in homocysteine and methylmalonic acid
2. cobalamin decrease
3. MCV rises, neutrophil hypersegmentation
4. MCV is macrocytic
5. anemia
6. symptoms

slow process...have about 10 years worth in storage pool if you're not a vegan...so if you destroy my parietal cells now, it will take 10 years to get anemia
*where is folic acid absorbed
duodenum/jejunum
*how long will your storage supply of folate last?
4-5 months
*dietary deficiency of B12 is
unheard of

(folate is possible, though not as common since fortification)

can supplement either o fthese as much as you want...no side effects
*folic acid is found in __ foods, cobalamin found in __

which is water soluble?
all

animal proteins

both
*in megaloblastic anemia, you have to draw a level before...
they eat (b/c one meal can replace folate levels)

after you draw blood, can feed them both; when you get the test result, only have to supplement the deficient one

don't transfuse!!! just give the vitamin and all the stuff stuck in the bone marrow can divide
*correction of megaloblastic anemia takes about
2 months

retic count peaks at about a week and a half
*pts with megaloblastic anemia also have a defect in __ metabolism; __ in particular leads to ~ absorption problems
iron

B12 deficiency (so when you start treating with B12, improvement will stop after a point, and you've unmasked the iron deficiency, so you have to work that up now)
*macrocytic anemia develops __ly, responds to tx __ly
slowly

quickly
oxygen binds to the __ in hemoglobin
the ferrous atom
the 4 globin chains normally have a __ association
loose
*at __ oxygen tensions, there's significant unloading into tissues
low

sigmoid shaped oxygen dissociation curve b/c of cooperative binding
*Epo function
1. stimulate proliferation of committed RBC precursors
2. stimulate maturation of those
nucleus is extruded from RBC __ it leaves the bone marrow
before
what causes Epo to be released from kidneys
oxygen sensing apparatus in the kidneys
*alpha like globin genes

on chromosome

promoter
zeta (fetal, though alpha turned on way early then stays constant)
2 identical alphas

16

HS-40
*beta like globin genes

chromosome

promoter
epsilon (embryonic)
2 gammas (each slightly different)
delta (dysfunctional beta - 1% of adult)
beta

11

beta-LCR (essential for developmental expression...through looping, it's intimately associated with gamma during fetal life and with beta and delta during adult life)
*gamma globin levels fall
pretty much at birth, though you don't get more beta than gamma until about 6 mo.

1% of adult is still gamma though
*important region for beta-LCR switiching from fetal to adult globins
PYR complex (pyrimidine rich region) in intergenic region b/w A-gamma and delta loci
***what is BCL11A
transcription regulator protein that is necessary for Sox 6 binding; without it, fetal gamma globin remains active

(trying to make drugs that are Ab to BCL11A)
globin genes contain __ exons and __ introns (spliced out)
3

2
hemoglobinopathy is a ___itative change

thalassemia is a ___ change
qualitative

quantitive (decreased or absent production of an otherwise normal globin chain)

heterozygotes of either have increased survival advantage over malaria (homozygotes do not)
*second most common Hb disorder

seen in

severity
HbE

SE asia

generally mild unless you have a sickle gene or thal gene (synergistic)
*Hb C seen in
West africa
thalassemias seen in
mediterranean, S. asia, SE asia
*Hb D seen in

important because
India

when inherited with a sickle gene --> sickling disorder
*beta thalassemia is due to __ gene defects
a number of (several hundreds)

usually involve splice sites (ineffective splicing) or defect in 5' regulation
*beta+ thal is a __zygous condition

difference b/w that and beta0 thal
homo

beta+ makes 10-30% beta chains
beta0 makes none

(both make 100% alpha)
*why are you anemic in beta thal
excess alpha globin aggregates and precipitates on RBC membrane, causing those developing RBCs to be hemolyzed IN THE BONE MARROW...they don't make it to circulation

gamma globin is insufficiently increased to compensate
***profile of beta thal trait
mild anemia (Hct 10.8) +
low MCV (60 fl)

in iron deficiency, if MCV were this low, the Hct would be <10
*in beta+ thal you see __ Hb F

in beta0?
slightly increased

slightly increased

(Hb A2 is variable in both)
*in beta+ thal, the anemia is __

in beta0 thal?
severe

severe
*δβ thal, HPFH and Corfu are deletion abnormalities at
the intergenic region b/w gamma and delta

impairs the binding of BCL11A, so fetal Hb is expressed
*δβ thal is a __ anemia with __ HbA and __ HbF
mild

absent

modestly increased
*HPFH is __ anemia with __ HbA and __ HbF
absence of

0%

100%
*corfu is a __ anemia with __ HbA and __ HbF
mild

10%

90%
*in __ bone marrow expands, and there's an attempt to make blood in extramedullary sites like spleen (site of blood production in fetal life) and liver/spine
beta thal major

but it's not sufficient --> poor growth and development
*tx of beta thal
chronic transfusions - will extend life significantly, but problems accompany this, including iron overload (complications e.g. cardiomyopathy = most common cause of death, usually happens in 30s)

chelation has problems of compliance, local skin reaction

(improvements like guidelines, leukodepletion filters, testing of blood, venous access, chelation improvements)
*alpha thal silent carrier

alpha thal trait

HbH disease

hydrops fetalis
deletion of one alpha locus (black)

deletion of two loci (on same or different chromosomes...two blacks or one oriental)

deletion of three alpha genes (oriental and one black)

deletion of all alpha genes (two oriental)
*HbH disease
tetramer of 4 beta chains

deletion of 3 alpha globin genes (one black, two asian)

moderate/manageable hemolytic anemia (unstable Hb, tends to precipitate and hemolyze)...most live through adult life
*hydrops fetalis
deletion of all four alpha globin genes
*single alpha globin deletion seen in

double deletions in
20% of blacks (--> silent carrier)

orientals (alpha thal trait)

two "black mutations" would also be alpha thal trait
*profile of alpha thal trait
like beta thal in that MCV is low, anemia is mild, but you don't see elevated Hb A2
*acquired gene defect in alpha thalassemia myelodysplasia
ATRX chromatin remodeling protein --> clumps of Hb H
*sickle mutation
codon 6 on beta globin (glu-->val)
*if you inherit one HbC and one HbS, you get

homozygous HbC -->
a sickling disease

mild anemia, target cells
*mutations that cause polycythemias

why?
Yakima, Chesapeake

Hb doesn't release oxygen well, so more Epo is produced thanks to oxygen sensor
*mutations that cause oxidation of hemoglobin --> cyanosis
M Boston, M milwaukee
*Hb mutations that causes instability --> hemolysis (alone or with drug stimulation)
Zurich,
Koln,
Hammersmith
*sickling shape does not occur when the __ form is present
oxy

(when deoxy happens, it starts to sickle; but if you're a sickle trait, you have one beta S and one beta A, so the reaction is too slow/dilute to sickle)
*Hb __ is favorable with a sickle trait
A or F

(C is synergistic --> sickling)
*sickle cell patients have an ONGOING ___
hemolytic anemia

(vaso-occlusive crisis is episodic, and what brings them to the hospital)
*why is sickle crisis episodic
there's only sometimes slowing down of flow or constriction in post-capillary venule --> vaso-occlusive crisis
folks with SS trait can get
hematuria (renal papillae can slough if they're dehydrated or high altitude)
*SC sickling is __ in severity; prediliction for __ damage
moderate

bone, eye
the worst sickle cell syndromes
SS and Sbeta0

Sbeta+ is less trouble

SF is mild

SC is moderate
*dx of sickle cell disease
hemoglobin electrophoresis
+
sickling test
**in sickle cell, hemolysis --> consumption of NO, reduced NO -->
leg ulceration
childhood stroke
priapism
PE

so the complications of SS are due to both physical plugging up and endothelial problems
*tx for SS?
1. hydroxyurea allows a little more HbF production (by shifting to stem cells that divide less) --> some improvement

also reduces risk of stroke by decreasing flow velocity in cranial vessels (increased flow is a warning sign of stroke)...stroke happens b/c of accumulation of debris, inflammation, interaction of RBC/WBC/damaged endothelial cells

2. exchange transfusion - but you have to keep it constant, and there is risk of iron overload; has been shown to help avoid stroke

3. BM Transplant - works in the right setting; more successful the younger you are
can you diagnose SS disease antenatally?
yes
can counsel, abort
*features of hemolytic anemia:
__cytic
__RBC survival
__retic count
normo
shortened
increased
*bone marrow has the capacity to increase production of retics by __fold; so you don't become anemic from hemolytic anemia until your RBC survival is < __ days
6

20
*findings CONSISTENT with hemolysis:
bilirubin __
LDH __
haptoglobin __
urine Hb __
urine hemosiderin __
urine urobilinogen __
up (unconjugated)
up
decreased/absent
present
present in chronic hemolysis

but these are not sensitive or specifc
**what is haptoglobin?
protein that binds and removes Hb from circulation, b/c free Hb is nephrotoxic
*most common blood smear finding in hemolytic anemia is
normal blood smear!
*tests to define the CAUSE of hemolysis (but do not demonstrate the presence of hemolysis)
Hb electrophoresis
RBC enzymes (G6PD, PK, etc)
antiglobulin tests (immune-mediated hemolysis)
cold agglutinins (some immune hemolysis)
osmotic fragility (spherocytosis)
CD55/CD59 (PNH)
clotting profile (DIC)
*categories of causes of hemolytic anemia
INTRAcorpuscular
--membrane abnormalities (e.g. hereditary spherocytosis)
--metabolic abnormalities
--hemoglobinopathies

EXTRAcorpuscular
--immune
--nonimmune
*when red cells develop and increased sensitivity to complement, you have __
paroxysmal nocturnal hemoglobinuria
possible membrane defects in hemolytic anemia
microskeletal defects (spherocytosis, elliptocytosis)
membrane permeability (stomatocytosis)
increased sensitivity to complement (PNH)
*what gives RBC its nice shape?

in hereditary spherocytosis, __ is defective or absent

where is the hemolysis
spectrin lattice, with actin wound between it, all anchored by glycophroin C

spectrin

spleen (extravascular)
*describe splenic hemolysis in hereditary spherocytosis
low oxygen tension in spleen b/c cells sit around longer; lower tension --> build-up of metabolites, swelling, of cells, which they don't tolerate well b/c they have less membrane --> rupture
in spherocytosis, you start to get hemolysis at __% normal saline; in normal person, it's at __%
0.6

0.55
*in PNH, the hemolysis is
intravascular and extravascular
PNH is an acquired deficit of __ protein; which does what?
GPI-associated, including Decay Activating factor (CD 59)...or could be lack of GPI anchor (located on X chromosome)

if complement binds to RBC, which it generally shouldn't, DAF is there to degrade it. no DAF --> assembly or membrane attack complex --> poking holes in membrane

more active when pH is lower, which happens at night when CO2 is high; but it occurs through the day and night
*G6PD leads to ___ because ___
hemolytic anemia

it regenerates NADPH, which is necessary to make glutathione from its oxidized form; glutathione protects against oxidative stress in RBCs

without it, oxidative Hb forms and precipitates on RBC membrane as Heinz bodies, which get picked off in spleen --> loss of membrane --> eventual lysis during oxidative stress

(causes of oxidative stress = infection, medications, fava beans)
*causes of oxidative stress

oxidative stress in G6PD deficiency leasd to __ formation and __vascular hemolysis
Infection

Meds
--antimalarials
--aspirin
--nitroglycerin

Fava beans

Heinz body

extra
*normal G6PD falls off over the life span of the cell, but not to a dangerous level; the __ variant is worse and the __variant is worst
African (unstable enzyme) - poops out at 50 days, so older cells are susceptible

Mediterranean (really unstable, almost all cells susceptible)
*non immune types of extracorpuscular hemolysis
mechanical
infectious
chemicla
thermal
osmotic
*causes of microangiopathic hemolytic anemia
vascular abnormalities (no coag abnormalities)
--TTP
--renal lesions
--vasculitis
--AV fistula

intravascular coagulation predominant (activation of clotting system)
--abruptio placentae
--DIC
**clinical diagnosis of TTP
Fever
Anemia (microangiopathic hemolytic)
Thrombocytopenia
Renal dysfunction (mild)
Neuro (variable severity)

need to have the A and T
causes of TTP
HIV, SLE, SS, drugs (plavix, cyclosporine, tacrolimus)
*pathophys of TTP

test?
autoantibody to vWF cleaving factor (ADAMTS13); usually preceded by viral infection

there is also a rare congenital form (relapsing)

no good test; clinical dx; could check level of ADAMTS13, but could be too late
*TTP tx
95% fatal without tx

plasmapharesis
steroids
retuximab, vincristine, splenectomy (resistant disease)

could be like a month that they're hooked up to this every day before they get better; ADH decreases, Bili, decreases, platelets go up
*immune hemolytic anemia --> __vascular hemolysis
intra or extra
(destroyed in vascular tree if complement is activated vs tagged for destruction by macrophages via opsonization with Fc or C3b)

the antigen-Ab rxn depends on class of antibody, number and spacing of antigenic sites, availability of complement, environment tmemperature, functional status of RE system
*Direct Coombs test
test for immune hemolytic anemia

looks for Ig and/or complement on surface of RBC (red cells don't have Fc receptors, so they'd be bound via antigen combining site)

Coombs reagent = anti-human Ig and anti-human complement + patient's cells ... if Ig or complement is on surface, coombs reagent will link cells together and cause agglutination of RBCs

if negative, the cells stay in suspension

any positive test is abnormal
*serum is
plasma that has been clotted (no clotting factors)
*Indirect coombs test
add patients SERUM to a panel of RBCs with known antigens; add coombs reagent; if there were Abs in the patient serum, agglutination occurs
*types of immune hemolytic anemia
drug-related hemolysis
--immune complex mechanism
--haptenic mechanism
--true autoimmune

alloimmune hemolysis
--transfusion reaction
--hemolytic disease of newborn

autoimune hemolysis
--warm
--cold
drug-related immune hemolysis:

immune complex mechanim drugs?
haptenic mechanism?
true autoimmune mechanism?

tx?
1. quinidine, quinine, isoniazid - drug and Ab bind in plasma and either activate complement in plasma or sit on RBC...RBC gets hemolyzed in innocent bystander reaction

2. penicillins, cephalosporins - these alter the RBC membrane, creating a neoantigen

3. L-DOPA, procaineamide, ibuprofen - alters RBC membrane that leads to Ab formation...cause Abs that react with normal antigens on RBC surface; you don't need the drug to be in the system!!! for the other two you do

stop the drug!
*type of alluimmune hemolysis from hemolytic transfusion reaction
immediate intravascular hemolysis - within 5-10 minutes, life-threatening due to nephrotoxicity of Hb - preformed Ab's that can fix complement

slow extravacular hemolysis - days - usually due to repeat exposure to a foreign antigen - more commonly seen - reactivation of memory B cells pretty quickly

delayed sensitization - weeks - usually due to 1st exposure to foreign antigen - asx - normal rate of hemolysis until you get enough Ab to form a complex, then it drops off
*testing of blood before transfusion
ABO and Rh type of both people

antibody screen, indirect coombs of both people

major cross-match (recipient serum and donor red cells) - looks for minor Ab's
*scenario where you get hemolytic disease of newborn

tx?
mom is Rh-, first baby is Rh+...some baby blood spills into mom at birth; subsequent baby, she revs up antibodies against Rh, which go to baby; if baby is Rh+ --> hemolysis

can cause severe anemia, hyperbilirubinemia (kernicterus)

Tx with RhoGam after EACH pregnancy...can totally prevent this reaction
*autoimmune hemolysis often associated with either
lymphoproliferative disease
-CLL

collagen vascular disease
-SLE
*types of autoimmune hemolysis
Warm type
-IgG
-Ab's bind at all temps
-don't fix much complement
-Fc receptors/C3b recognized by macrophages, therefore...
-EXTRAvascular hemolysis
-70% associated with other illnesses

Cold type
-IgM
-antibodies bind best at 30 degrees or lower (so you typically see only complement on cells)
-fix entire complement cascade
-membrane attack complex
-INTRAvascular hemolysis
-90% associated with other illnesses
***tx warm type autoimmune hemolysis

cold type?
steroids, splenectomy - responds well

plasmapheresis (not steroids or splenectomy) b/c IgM is very large and only in plasma
*in hemolytic anemia, marrow function is usually

often require extra __ to maintain hematopoiesis
normal

folic acid

(they depend on bone marrow being hyperfuncitonal, so anything that decreases marrow function will be life-threatening
pediatric cancers are 1/__ the rate of breast cancer or prostate cancer
10

1/330 children gets one
1/750 20-year olds is a survivor
overall survival for childhood cancer is
80%
*most curable child cancers

~50% curable

least curable
RB
hodgkin's
wilm's tumor

brain tumors, neuroblastoma

AML
*most common pediatric cancers
Leukemia > brain tumors > lymphoma > neuroblastoma
*cancers that are primarily in 1-2 y.o. then drop off dramatically
RB
Neuroblastoma
Wilm's tumor
*pediatric cancer that's seen increasingly with adolescents than with younger kids
lymphoma
*___ (cancer) has a peak at 3-4 y.o. but has pretty high rates at other ages
ALL
*retinoblastoma (primitive neuroectodermal tumor) exists in three forms:

mutation is the __ gene
1. familial - 10% - almost complete penetrance; increased risk of other cancers
2. sporadic hereditable - 30% - new germline mutation - increased risk of other cancers
3. sporadic non-herditable - 60% - two hits occur after conception - always unilateral

tumor suppressor RB1
*bilateral RB appears ___er; __ hit
earlier

one (familial form leads to bilateral in 90% of cases - second hit occurs after conception)
*non-heritable RB is always
unilateral
variation occurs in the incidence of __ retinoblastoma
unilateral (more in poor living conditions)
*most common presenting sx of RB
leukocoria
(often visible only with electric light or camera flash) - often not visible until tumor is large enough to have affected vision

usually painless

more advanced disease - proptosis, erythema, pain
*RB 5 year survival

tx
97%

enucleation --> chemo/radiation

local therapy (laser and cryo) could treat less affected eye in bilateral disease
*secondary malignancies often see after retinoblastoma (36% in those with germline mutations)
bone (esp. orbits), nasal cavity, soft tissue, pineal

later increased risk of melanoma, breast, lung, bladder cancer

limit radiation exposure in these kids!
*Wilm's tumor makes up 95% of
child renal tumors

(others are rhabdoid, clear cell, RCC)
*Wilms tumor is associated with weird __ in 15% of patients
congenital anomalies like:

aniridia
hemihypertrophy
cryptorchidism
hypospadias

can be divided into syndromes with or without overgrowth features
*WT1 gene (a __) is mutated in __% of wilms tumor
zinc finger transcription factor

5-20%
*what is WAGR?
Wilms tumor with Aniridia, GU anomalies, and Retardation

without overgrowth features
*aniridia alone is a mutation in

with new onset, look to see if
PAX6

WT1 is also mutated (nearby), they're at risk for developing wilm's tumor
*syndrome of Wilm's tumor with overgrowth features

what is the mutations
Beckwith-Wiedemann (but Wilms tumor occurs in only 5-7% of B-W) - this is an imprinting disorder

visceromegaly, macroglossia, omphalocele, microcephaly, gigantism; increased risk of adrenal carcinoma, hepatoblastoma, gonadoblastoma

WT2 - region of IGF2 and H19...oppositely imprinted - IGF-2 is doubly expressed and H19 (which normally balances IGF-2) is turned off...so too much insulin-like growth factor...don't know why it only occurs in half of body
Wilm's tumor has a lot of histological diversity, but has varying proportions of 3 cell types
stromal, epithelial, blastemal...trying to recreate a kidney, but doesn't do it well
*Favorable histology (FH) of wilms tumor

in non-favorable histology there is a high incidence of __ mutation
no anaplasia

(anaplasia more likely in patients >5 y.o.)

p53 (86% vs 0.8 in FH)
*most common presentation of Wilms tumor
palpable mass in abdomen - slow growing so may not be noticed by parent

also hypertension and hematuria sometimes

**usually asx
Wilms tumor staging
1 - limited to kidney and excised
2 - not limited to kidney but all excised
3 - residual non-hematogenous tumor in abdomen
4 - hematogenous metastases
5 - bilateral renal involvement at dx
***tx Wilms tumor

survival rate
radical nephrectomy
+ radiation
+ chemo (dactinomycin)

90%
most common pediatric abdominal tumor
neuroblastoma
*neuroblastoma arises from __ cells
neural crest (which give rise to adrenal gland and symp. ganglia)
*this pediatric cancer (___) can have widely varying outcomes, including __
neuroblastoma

spontaneously regress
differentiate into benign
metastasize with high mortality rate (2/3)

among the metastatics, one group is young infants who do well, another is children 2-3 who do not
*most common cancer of infancy
NB
*why have we seen in increase in NB in infants, but not 1 year olds?
prenatal diagnosis of cases that spontaneously regress
genetic defect in NB
deletion at 1p often seen, LOH

sometimes see MYCN amplification (more often in advanced disease, rapid progression, poor outcome)
benign NB
ganglioneuroma
*NBs can originate from

at presentation __ have metastatic disease, often in __-
any site in SNS (cervical and thoracic common in infants, adrenal common in older)

1/2-2/3
bone marrow 70%
bone 55%
lymph nodes 30%
liver 29%
cranial 18%
lung, skin
*opsoclonus-myoclonus

tx?
paraneoplastic syndrome associated with NB (or EBV or coxsackie B)
dancing eye and dancing feet - acute cerebellar encephalopaty, truncal ataxia, rapid and random eye movements - can lead to permanent neuro deficits, decreased IQ, behavioral problems

kids can be nervous, irritable, lethargic

tx: ACTH, IVIG, chemo
*work-up for NB
urinary catecholamine metabolites HVA and VMA

CT/MRI

bone scan, bone marrow aspirate to assess for mets
NB staging
1 - localized and excised
2A - localized, incomplete resection, ipsilateral LN-
2B - ipsilateral LN+
3 - unresectable or contralateral LN+
4 - eissemination

*4S - localized primary tumor, dissemination limited to skin, liver (big mets), and/or bone marrow; children <1 year - DO NOTHING, usually regress spontaneously
*tx of low grade NB
just take out tumor (no chemo) --> 90% survival

high risk is another story
***tx of 4S neuroblastoma
supportive care in 55% (good prognosis)

symptomatic 45% - cyclophosphamide +/- hepatic RT (babies <2 mo who don't have enough space in abdomen for everything) - some of these die from organ failure, hepatomegaly
*genetic/histo features of 4S NB
96% FH
no MYC amplification
*NB survival much better in children __1 year old and without ___
less than

MYC amplification

even if you're non-MYC amplified, outcome is poor if you're 3 y.o.
***best therapy for high grade NB
BMT+ retinoic acid > BMT > chemo + retinoic acid > chemo
*pathophys of ALL (most common pediatric cancer)
single lymphocyte loses ability to apoptose --> accumulation of abnormal lymphocytes in marrow, crowding out normal elements

--> varying degrees of anemia, granulocytopenia, thrombocytopenia
*epidemiology of ALL
boys > girls
more whites
*common symptoms of ALL
1. fatigue (90%) - often secondary to
2. anemia (80%)
3. thrombocytopenia (50%) - petechiae
4. splenomegaly (65%)
5. bleeding (50%)

fever, bone pain, chest mass
6. LAD (50%)
*dx of ALL
CBC
smear
bone marrow aspirate
flow cytometry
cytogenetics
CXR (look for chest mass)
chemistries for hemolysis
LP to see if it went to CSF
D-dimer
infectious disease profile
*many leukemias have a "signature" which can be appreciated by __ but not __
flow cytometry

morphology alone

(useful for monitoring disease, stratifying patient, targeting therapy in future)
***the clonal cell of ALL: ___

has flow cytometry positive for
precursor B cells

HLA-DR
CD19
CD10
***cytogenetics in ALL: hyperdiploidy is ___; hypodiploidy is __


philadelphia chromosome (___) is ___

TEL/AML (t12,21) is
good

bad
(near haploid is very bad)

t(9,22)

bad

good
*leukemia standard risk:
age ___
initial WBC count __
no __ disease
disease of __ cells

high risk:
age ___
initial WBC count __
presence of __ disease
disease of __ cells
1-10
<50,000
CNS
precursor B

<1 year or >10 years
>50,000
CNS
T cells
*typical ALL tx is __ years of chemo

if Philadelphia chromosome, tx is

if MLL

if hypodiploidy
3-3.5

immediate BMT after initial induction

additional consolidation therapy

additional consolidation therapy
*minimal residual disease in __ can be detected by __
ALL

flow cytometry
PCR

(can detect 1K - 100K cells)

MRD > 0.001 after 30 days of tx suggests poor prognosis; give more intensive therapy, but don't know if that works
***general scheme of ALL tx
1. steroid prophase
2. remission induction (4 wk inpatient)
3. consolidation (inpatient)
4. CNS (cranial radiation or intrathecal; outpatient)
5. consolidation II (30 wks outpatient)
6. continuation/maintenance (70 wks outpatient)
*what fraction of people get cancer before they die
1/3-1/4
*lung cancer is going __ in men, __ in women
down

up
*colon cancer is gradually ___ing
decreasing

(probably because of awareness)
men have more cancer b/c of
prostate cancer
all cancer incidence is ___ing

all cancer mortality is __ing
increasing

decreasing

(so something good is happening...screening and tx)
virtually all colon cancer is due to
diet
non-hodgkin lympoma is #__ cancer; it is kind of __ing
6

rising
prostate/breast > lung > __
colorectal
by far the most common cause of cancer
smoking
*cigarettes cause these cancers
lung (90% of them)
pancreas (very high correlation)
esophageal
bladder (very strong dose-response correlation...stronger than for lung...the more you smoke, the more likely you are to get bladder cancer)
most cancer deaths
lung > prostate/breast > colon > pancreas
increased cup size and breast cancer in Japan attributed to
Western diet

combination of fat and hormones
*cancers that can be caused by diet
prostate (meat and fat intake)
breast (fat intake)
colorectal (grilled meat has carcinogens)
*direct link between benzene and
leukemia
HPV is a _NA virus

makes proteins that can bind __ proteins and immortalize cells
DNA

tumor suppressors (p53, RB...act as ubiquitin e3 ligase)
*why does EBV make more Burkitt's lymphoma (non-hodgkin lymphoma) in Africa
children are infected younger, when B cells are at a different stage of development (more Ig gene rearrangement and higher chance for genetic errors)

<50% of Burkitt's lymphoma in west is due to EBV
*HTLV is indigenous to ___ and causes this cancer
Japan, Caribbean

T-cell leukemia (a lot in Brooklyn where there are caribbean immigrants)
*cancers caused by infectious agents
HCC (hepatitis)
cervical
non-Hodgkin lymphoma (EBV)
leukemias (HTLV)
gastric (H. pylori)
3-legged stool of cancer causation
epidemiologic evidence - observation

mutation potential

animal model (recapitulates with no cofounders)
how does asbestos cause mutations
crocidolite fibers disrupt mitotic spindel (b/c they are so small)

inject a mouse intraperitoneally with crocidolite and it will get peritoneal mesothelioma
mutagenesis in cancer:

__% are sporadic mutations
__% are inherited
__% are exposure to mutagens and hereditary predisposition
15% - e.g. childhood leukemia
5% - high penetrance, low incidence
80% - why only 10% of smokers get lung cancer

MZ twins only have 10-20% concordance...something stochastic is going on
only __% of colon cancers are APC or MSH/MLH

__% of breast is BRCA
1-2%

1-%
*ATM mutation increases RR of __ cancer by 1.5-5x
breast
one cancer can provide and produce hundreds to thousands of cells with metastatic potential (independent, can intravasate, attach to endothelium, extravasate, set up an new colony) but
very few ever result in macroscopic metastases
***progression of mutations in colon cancer
1. APC
2. KRAS
3. p53
most cancers have ___ mutations, but not all are important
60-100
*tumor suppressor genes are __

oncogenes are __

cancer can also be caused by gene __
recessive

dominant (good targets so far)

methylation (changes levels of expression)...cancers as a group have different patterns of gene methylation than adjacent normal tissue
*cancer genes fall into 4 categories
signal transduction
cell cycle/checkpoints
DNA damage or repair
apoptosis
***some cell cycle/checkpoint cancer genes are recessive (e.g. ___), some are dominant (e.g. ___)

they are mutated in most cancers
p53, RB

CDKs
***DNA damage or repair genes
BRCA
MSH/MLH (mismatch repair; HNPCC)
ATM (DNA damage recognition)
XP (DNA repair)
*apoptosis mutations (least prevalent class)
BAX - colon cancer
BCL2 - leukemias
*high salt diet --> __ cancer
gastric
*single most carcinogenic thing we eat
aphyllotoxin - aspergillus fungus, grows in grain silos

solution? fresh food
*aphyllotoxin --> __ cancer
HCC
the offending hormone in breast cancer is
progesterone

(estrogen + progesterone replacement in post-menopausal women --> cancer)

earlier periods, fewer children increase breast cancer
*baby aspirin and COX-2 inhibitors can help prevent __ cancer
colon

(unfortunately COX-2 has toxicity, so only give if APC mutation)
which are specific, kinases or phosphatases?
kinases...many more of those are cancer genes
first proof of cancer kinase
Raus sarcoma virus, under a certain temp, would phosphorylate and cause malignancy...raise temp and things are hypophosphorylated and benign
*receptor tyrosine kinases -->
Ras or PI3K

Ras can --> PI3K

both can activate mTOR
cancer will never mutate 2 steps in the same pathway; what is hierarchical activation?

reactivation after resistance arises via
have to activate some pathways before they activate others

reactivation of the same pathway you shot with your arrow - the cancer has to have that pathway!!!
cancer kinases are often tyrosine kinases, but they could be
serine/threonine kinases
*human genome has __ tyrosine kinases and __ total kinases
90

518

(you can tell b/c they have common sequences)
*Ret receptor mutated in
thyroid cancer
IGFR is very important in cancer, but drugs have a side effect of hypoglycemia
t
*Her2 is in the __ family
EGFR

it is ErbB2
*each tyrosine in the EGFR tail
when phosphorylated --> downstream effectors

different between cells and at stages in development
*the 4 members of EGFR family can make lots of dimer combos, and each combo has preferred ligand; but the rules are that
Her2 has no ligand binding domain

Her 3 has no active kinase domain

(so don't put two of those together)
***the kinase domain of the EGFR is mutated in 10% of __ cancers

the drug that blocks this effect is

how does resistance develop
lung

erlotinib - blocks ATP binding - causes regression of the lung cancer in weeks


RESISTANCE:
1. Thr790 then becomes mutated, which is in the binding pocket for erlotinib, and it decreases affinity; knowing this, can make drugs that target that

2. could also develop increased phosphoHer3 --> increased Akt --> activated EGFR

3. Met amplification; Met transphosphorylates Her3, reactivates that pathway
***cetuximab is an antibody to
EGFR, but it's not as effective as erlotinib at regressing lung cancer
***a cancer where you have an EGFR variant III, deletion in first 5 exons makes it constitutively active

tx?
glioma

also responds to erlotinib
***Her2 is amplified in 25% of breast cancers, and is essential for growth in those (when breast cancer is diagnosed, you do IHC or FISH for Her2 amplification)

in these patients, there is indication for higher dose ___

the drug that targets Her2 is ___
doxorubicin

trastuzumab (which also sensitizes to taxanes) - not great as a single drug, but VERY dramatic improvement as adjuvant therapy
***trastuzumab MOA

toxicity
targets Her2 (in breast cancers where Her2 is amplified...which is 25%)

CHF, anaphylaxis, rash
Ras is bound to
inner plasma membrane

mutated in 90% of pancreas, 80% of lung, 45% of colon cancers
***DBL
a GEF

oncogene deleted in B-cell lymphoma
*neurofibromin
a GAP

oncogene of NF
*N-RAS (non-essential) mutated in

H-RAS (non-essential) mutated in
rare melanomas

salivary, Spitz nevi (innocent melanoma)
***the cancer mutations that arise in Ras are
12,13, 61 - such that GAP can't bind, so Ras is constitutively active
*lovostatin was shown to inhibit
the FA addition to RAS, which anchors it to PM...however that hasn't been an effective drug for pancreatic cancer yet...too much toxicity, not enough efficacy
***a big intracellular tyrosine kinase

mutated form does what

drug that targets it
ABL

has an mRNA with BCR and ABL sequences that makes an abnormal tyrosine kinase, constitutively active in CML (it lacks the autoinhibitory cap) --> cancer in neutrophils

imatinib
***imatinib blocks:

corresponding cancers:

resistance?
ABL - CML
KIT - GIST
PDGFR-A -hypereosinophilic syndrome
PDGFR-B -myeloproliferative syndrome

all inhibited by imatinib

1. most commonly, see mutation in imatinib binding pocket (can be targeted if we know the sequence)
2. can also see gene amplification, which stops if you stop imatinib (b/c it can)
GIST is in young adults, generally doesn't metastasize, but kills by
occupying abdomen
*CML in natural history transforms into a more aggressive leukemia that kills in a matter of months (blast crisis), even then, imatinib extends survival to a year in __oid types
myeloid
*1/__ chance of getting a spontaneous mutation in any cell in the body at any time
10 million

use 2 drugs at same time and chances of both mutations is 1/10^14
*lessons for therapeutic strategies
1. target dominant oncogenes (they drive the malignant phenotype)

2. pathway blockade DOWNstream of activation

3. resistance mechanisms reactive the SAME pathway; you know where to look, use multiple drugs on that pathway
*Shh pathway activated in __ cancer

tx?
BCC

mutation either disrupts Patched inhibition or causes Smoothened activation

can use derivatives of the rocky mountain thing that blocked Shh and --> cycloptic sheep (cyclopamine)

showed response in 85% of metastatic BCC
*what is the phenotype in each:
acute leukemia
chronic leukemia
lymphoma
myeloma
precursor cells
differentiated cells
differentiated cells
differentiated cells
*myeloma is a disease of __ cells
terminally differentiated B cells
*WBC profile for each:

1. leukemoid reaction:
2. acute leukemia:
3. CML
4. CLL
1. 82% neutrophils, 13% bands
2. 82% blasts (or promyelocytes in APL)
3. whole spectrum of differentiation
4. 98% lymphocytes
*what is ANLL
acute non-lymphocytic leukemia

= acute myeloid leukemia
(includes granulocytic, erythroid and megakaryocyte lineages)
*acute leukemia is basically and imbalance between __ and __

the majority of cells are not __
proliferation and differentiation

dividing (presents a therapeutic challenge)
***majority of leukemias have visible chromosomal abnormalities; some tumor-specific translocations:

acute promyelocytic leukemia ___
CML ___
Burkitt's lymphoma/leukemia
t(15,17) -- almost always
t(9,22)
t(8,14)
deletions of tumor suppressor genes are typically more __
aggressive, difficult to treat
***in Burkitt's lymphoma, ___ is overactive
myc (a TF)
*causes of leukemia
radiation
chemo
combination of two (like in treating hodgkin's lymphoma)
benzene
hereditary
HTLV-1
lymphohematopoietic stem cell -->
1. hematopoietic stem cell
2. lymphoid stem cell
*hematopoietic stem cell -->
1. erythroid progenitor
2. megakaryocyte progenitor
3. granulocyte monocyte progenitor --> all white cells other than B and T lymphocytes

if a chromosomal abnormality arises in a progenitor, everything downstream will have that
*in CML you often see the philadelphia chromosome as far back as
granulocyte monocyte progenitor, hematopoietic stem cell, or lymphohematopoietic stem cel
*myelodysplastic syndrome, seen in ___, is a precursor to ___

can commonly see __cytic anemia

other morphologic abnormalities
elderly (often)

ANLL

macro (without B12 or folate deficiency)

pancytopenia (can happen) with aniso~, poikilo~, neutrophil hypogranularity/hyposegmentation, macroplatelets, hypolobated/smallmegakaryocytes, increased blast count
*tx of MDS
Epo, GCSF, hypomethylating agents (doesn't respond well to tx...just have to stabilize)
*Auer rods =
APL

distorted primary granules
***DIC is common with __ leukemia
APL
*tx of APL
arsenic, retinoic acid + chemo

(cells mature to PMNs)

80% cure
*t(15,17) is __ genes
RA receptor (with RA promotes differentiation) and PML (apoptosis gene)

with the translocation you get no apoptosis or differentation
***features of ALL vs ANLL wrt
age
myeloperoxidase stain
auer rods
terminal transferase
cell surface Ag
Ig or T cel receptor gene rearrangement
ALL:
-kids
-no (neutrophils contain)
-no (derived from myeloid precursor granules)
-yes TdT
-B or T
-yes (Ig if B, T cell if T)

ANLL
-adults
-yes
-yes
-no TdT
-myeloid
-no
*manifestations of acute leukemia
marrow failure (neutrophenia, anemia, thrombocytopenia) --> infections, fatigue, bleeding

hyperuricemia (high turnover of purines) --> acute renal failure

DIC (especially in APL; decreased platelets, abnormal clotting) - purpura, intracranial hemorrhage
***in acute leukemia, you can get bone pain (b/c of expansion of medullary cavity, esp. in children)

enlarged liver, spleen, nodes (more in __)

hypertrophied gums (esp. in ___)

meningeal infiltratino (esp. ___) --> cranial nerve palsies, headache
ALL

AML

ALL
*blast leukocytosis is an emergency because
rising number of blasts can --> leukostasis in small vessels of brain/lungs/kidneys --> tachypnea, tinnitus, lethargy, stupor, death
***tx of acute leukemia
aggressive (except APL)
CNS prophylaxis
BMT if relapse
allopurinol (uric acid builds up from purine breakdown)
transfusions
3-4 broad spectrum abx if infection
*median survival of child ALL?
adult ALL or adult ANLL?

5 year survival for child ALL?
adult ALL?
adult ANLL?
6+ years
1-2 years

70%
30%
15% (except APL) - elderly have more resistant form, usually in background of MDS
***in addition to CML, many patients with ___ have the philly chromosome
adult ALL
*two phases of CML
chronic phase (stable)
blastic phase (terminal phase)

transition is accelerated phase
***symptoms of CML chronic phase

median duration if untreated
weakness, weight loss
purpura (related to platelets)
thrombocytosis (2-4 million)
leukocytosis
normocytic anemia
splenomegaly 80%)
priapism
can see eos and basophils too
but low alk phos

3-4 years
*CML test in non-developed countries

if you can't see Ph chromosome on cytogenetics,
alk phos

can see in FISH or PCR
*ABL is on chromosome

BCR is on

both end up on

BCR ABL activates __
9
22

little 22

Ras
*if CML is going to affect lymphoid cells, they will more often be
B cells
***tx of CML chronic phase
-hydroxyurea stabilizes blood counts, but disease progresses

-interferon leads to cure in 10-15% (cure = Ph-negative)

imatinib (60% become Ph negative)

allogeneic BMT for those who don't respond to TK inhibitors
*myeloablative conditioning used in patients getting
autologous stem cells

(also in allogeneic, but not as important b/c of graft v malignancy effect)
*transplant related mortality is __ in autologous tx, __ in allogeneic
2-4%

10-30% (mostly GVHD)
***autologous stem cell tx has highest curative potential for __
hodgkin's lymphoma (40-50%) or diffuse large B-cell lymphoma when it relapses
*how can you get stem cells from peripheral blood for transplant
give GCSF and they will proliferate into blood, then you can do leukopharesis
*GVHD is associated with __ damage
skin, liver, gut
*increasing incidence of GVHD
unrelated donor who is HLA identical (considerable risk) > HLA-identical sibling (ideal) > T cell depleted sample from HLA sibling > identical twin (no GVHD)

but there is an inverse risk of relapse (GVM effect)
* in pts whose CML relapses after alloSCT, trasfusion of __ from donor WITHOUT additional chemoradiotherapy can induce complete remission
lymphocytes only

(but you do risk GVHD again)
*blastic phase of CML presents like
acute leukemia almost...

weight loss, fever, sweats, bone pain
worsening splenomegaly, anemia, platelet counts low or high

death in weeks/month from leukostasis
*adult ALL has __ prognosis

__% are Ph positive
poor

30-40%
(BCR-ABL or an even stronger mutation)
***all chronic myeloproliferative disorders will respond to ___, but only one is curable (CML)
hydroxyurea
***chronic myeloproliferative disorders

least aggressive is __

hematocrit high in __

anemia in __

primarily platelets elevated in __

tendency to bleed in all, but also to clot it ___
1. CML
2. Myelofibrosis with myeloid metaplasia
3. polycythemia vera
4. essential thrombocythemia

ET is least aggressive
Hct high in PV (everything elevated)
anemia in MMM
platelets elevated in ET
can clot in PV

splenomegaly seen in all 4
all can convert to acute leukemia
last 3 are Ph negative
*JAK2 mutation common in
myeloproliferative d/o's, esp. P. vera
*polyclonal B cell disorder will have surface ___

monoclonal B cell disorder will have
kappa and lambda light chains, innumberable kappa and lambda gene rearrangement bands

either kappa or lambda surface light chains, and a single kappa or lambda gene rearrangement band
*Hodgkin's age distribution

nodes seen

cause?
20-30 yrs. and >50 yrs

cervical >mediastinal>paraaortic
(tends to be asymmetric, starting in neck)

familial
HLA association
geographic clustering ("epidemics") sometimes
weak assoc with EBV
***tx of Hodgkin's lymphoma
radiation +/- multiagent chemo with different MOAs and non-overlapping toxicities
---prototype MOPP


if this is unsuccessful, intensify chemotherapy

then go to autologous SCT with leukapharesis and myeloablation-rescue...recovers in 2 weeks

(single agent is good for nothing)
*incidence of non-Hodgkin's is ___

mortality is ___
increasing

increasing

(both are decreasing for hodgkin's)
***Hodgkin's tends to be in __ patients

cell of origin is

usual stage at presentation

extranodal sites are __

Reed-Sternberg cells are __

architecture is ___
young or old (NHL is middle-aged)

B cell (also most of NHL)

I or II (vs III or IV in NHL)

follicular (diffuse in NHL)

uncommon (common in NHL)

present in HL, absent in NHL
***follicular lymphoma is most common indolent NHL; age usually __

stage usually __

translocation __ -->

33% transform to

prognosis
50-60 years

III-IV

t(14,18)

overexpression of BCL-2 (an anti-apoptosis protein) with IgH juxtaposition

aggressive large cell lymphoma

incurable but treatable (median survival 7-8 years)
***tx indolent lymphoma
asx - observe

radiation for localized cancer

alkylators and purine analogs are very helpful

rituximab (anti-CD20) +/- chemo: high response rate

rituximab with radioactive label

alloSCT sometimes in younger people (generally not helpful)
***rituximab MOA

treats

toxicity
anti-CD20 monoclonal Ab

treat indolent and aggressive NHL and CLL;
1. binds B cell then promotes apoptosis
2. binds complement to promote lysis
3. leads to antibody-dependent cytotoxicity

fatal infusion rxn, HepB reactivation w/ fulminant hepatitis
tumor lysis syndrome w/ severe renal toxicity
cardiac dysrhythmias
*difference b/w indolent and aggressive NHL
aggressive commonly has CNS involvement, survival is months (not years) but it's potentially curable (30-60%)
***tx of diffuse large B-cell lymphoma
Stage I or II:
RCHOP and radiation
(rituximab, cclophosphamide, etc, prednisone)

Stage III, IV
more RCHOP +/- radiation
30-60% cure rate

relapse
salve chemo and rituximab followed by SCT
***Burkitt's lymphoma tx
cyclophosphamide in CHOP like regimen
*cutaneous T cell lymphoma is a malignancy of __ T cells
Helper CD4+
***difference b/w adult T cell leukemia and Sezary syndrome (cutaneous T cell lymphoma)

geography:
skin:
lytic bone:
nuclei:
pathogenesis:
which is more malignant?
leukemic phase
TCR clonality
ATL has clusters in Japan, Caribbean (Sezary, none)

Sezary ALWAYS has skin changes (ATL often does)

ATL has lytic bone lesions (Sezary does not)

ATL has flower cells, Sezary has cerebriform

ATL from HTLV-1; Sezary unknown

ATL is v. malignant; high IL-2 expression

both have leukemic phase

both have TCR clonality
*myeloma is a disease of

produces
plasma cells (terminally differentiated B cells)

monoclonal Ig or light chain
*pre-B-cell malignancy:
B-cell malignancy:
transformed B cell ~:
B-ALL
CLL (decreased gamma globulins)
macroglobulinemia (monoclonal IgM)
***most common leukemia
CLL (B cell origin)

(also least dramatic)

age >50 yrs
*signs of CLL

labs

prognosis
LAD
splenomegaly
hepatomegaly
asx or vague complaints
recurrent pneumococcus infection

kappa or lambda light chain
single Ig gene rearrangement
hypoimmunoglobulinemia
lymphocytosis

few months to >20 years...mean is 5 years
***CLL tx
Stage 0 or 1 - no evidence for tx
Asx - watch and wait
sx - radiation good for local
chemo
rituximab - best of drugs

SCT is the only curative tx
*multiple myeloma more common in __s
blacks

any adult, but mostly older people

increasing incidence, unknown cause
***diagnostic features of multiple myeloma
1. almost always plasmacytosis (much >10% where there should be <5%)

2. monoclonal Ig and/or light chain in serum or urine (spike on electrophoresis) - other causes include CLL, lymphoma, benign monoclonal gammopathy in elderly

3. lytic disease of bone

other causes of plasmacytosis - inflamm, cirrhosis, AIDS, infection
*biggest problem in dx myeloma
benign monoclonal gammopathy is more common, only 10% progress to myeloma; most remain stable and have no bone disease or kidney disease

but most patients have sx at diagnosis...bone pain, pneumococcal infections, anemia sx
*hyperviscosity syndrome in ___ is due to ___
myeloma

aggregating paraprotein; circulatory insufficency, abnormal hemostasis; brain/lung/kidney manifestions like bleeding, dyspnea, encephalopathy and visual disturbances
*bacterial infections in myeloma

why?
early: s. pneumoniae
late: staph aureus or Gram negative rods

normal Ig levels are reduced; TGF beta and other causes
*amyloidosis in myeloma
due to light chain deposition in tissue

lambda > kappa

skin, tongue, heart, peripheral nerves, kidneys, soft tissues

no effective therapy
***tx for multiple myeloma
radiotherapy - most sensitive malignancy

thalidomide - anti angiogenesis
lenalidomide - anti angiogenesis
bortezomib - proteosome inhibitor
+corticosteroids

SCT for control, not cure
***thalidomide MOA

treats

toxicity
antiangiogenesis

tx multiple myeloma

teratogen, peripheral neuropathy
***lenalidomide MOA

treats

toxicity
anti angiogenesis

tx multiple myeloma

increased DVT risk, myelosuppression
***bortezomib MOA

treats
proteasome inhibitor - very active

tx multiple myeloma

GI, cardiac, all the ~penias
*sarcomas arise from __ cells (<1% of all cancers)
mesenchymal - bone, cartilage, muscle, fat

fleshy tumors

chondrosarcomas
liposarcomas
leiomyosarcomas
rhabdomyosarcomas
fibrosarcomas
osteosarcomas
classes of sarcomas
pediatric vs adult

translocation-related vs complex karyotype (most are complex)

bone vs soft tissue

origin known vs origin unknown
*pediatric sarcomas - 10% of childhood cancers: ___
rhabdomyosarcoma (most common)
--embryonal (better prognosis)
--alveolar (translocation related)
osteosarcoma
Ewing's sarcoma
--small round blue cell tumor (neuorectoderm?); translocation related
*osteosarcoma
pleiomorphic high grade tumor composed of fibroblasts, myofibrobalsts, histiocytes

in long bones usually

painless mass of several months duration

lesions are destructive

cslerosis is either from tumor or reactive

Codman's triangle of bone
sunburst patterns
adult sarcoma presentation age
40-50s
*difference in tx of pediatric vs adult sarcomas
peds: LOT of chemo +/- surgery
adult: LOT of surgery +/- chemo
***genetic syndromes associated with sarcomas
NF - von Recklinghausen's disease
Li-Fraumeni syndrom - p53 - osteosarcomas
RB - sarcomas
Gardner's (FAP) - desmoids
***translocation associated sarcomas
1. alveolar RMS (PAX)
2. Ewing's (EWS-Fli)
3. synovial
4. clear cell

rest are complex
*chondrosarcoma occurs in this age group
80s
*chemo efficacy for following:
Ewing's
osteosarcoma
chondrosarcoma
ewings = good
osteosarcoma = moderate (need surgery too)
chondro = bad (chemo really doesn't work)
*sarcomas of unknown origin
Ewing's sarcoma
clear cell sarcoma (looks likemelanoma of soft tissue parts)
synovial sarcoma

we think MFH comes from mesenchymal cells
***side effect of imatinib (oral)
puffy eyes
some swelling
can't drink grapefruit juice (increases metabolism)
*about __ of cancer pts are cured with local surgery and/or local radiation
1/3

only 10% of cancers curable by chemo (70% of childhood cancers)
*neoadjuvant therapy
for inoperable cancer; tx of locally advanced disease

maybe reduce from stage 3 or 2b to stage 2a (operable)
***curable cancers
ALL, AML
Hodgkin's and non-Hodgkin's
germ cell cancer
small cell lung cancer
chriocarcinoma
GIST
CML
cell kill in follows __ kinetics
first order (constant % every time)

our natural immune system can only handle 100 million cells, but cancer may have 10^12...so that's why you need so much chemo
***___ are the only drugs that kill non-growing cells
lipophilic alkylators

(temazolamide, BCNU, CCNU)
growth fraction of tumor (% of cells actively progressing through cell cycle) is maximal when it is about __ maximum size
37%

this is when it's most responsive to chemo

another reason why advanced cancer is hard to tx

some drugs are cell-cycle non-specific, so cells don't have to be in active cell cycle
***what phase does each attack:
antimetabolites
taxanes
vinca alkaloids (vincristine)
antitumor abx (e.g. bleomycin)
S
M
M
G2/M
***all nature-derived chemo drugs are ___ metabolized except ___

all synthetics (antimetabolites, platinum agents, alkylators) are __ metabolized

except
hepatically

BET - bleomycin, etoposide, topotecam

renally excreted

cyclophosphamide which must be hepatically metabolized first
***cell cycle non-specific agents
1. alkylating agents (looks for electron dense parts of DNA to add alkyl and inhibit)

2. anthracyclines (e.g. doxorubicin; topoisomerase II inhibitors, doesn't allow reannealment --> apoptosis)

3. camptothecins (e.g. irinotecan; topoisomerase 1 inhibitor)

4. platinum analogs - atypical alkylators; all have chlorines which come off at physiological pH, loosk for electron dense region to alkylate
chemotherapy doesn't kill cell, it makes cell kill itself
t
about __% of drug cures occur in __% of cancer types
90

10
drugs known to be partially active against tumor when used alone should be
used in combination
tumor resistance can be
intrinsic (the more genetically unstable the tumor is, the more mutations will occur before you even expose it to chemo) - p53, mismatch repair defect, Bcl-2, NF-kappaB

or

acquired (pumps that kick stuff out, phosphorylases that inactivate drug, decreased conversion of drug like cyclophosphamide to active form, reduced affinity of target enzyme for drug like in methotrexate/DHFR, enhanced DNA repair, increased expression of target enzyme like DHFR)
normal tissues never become resistant to drugs...why?
b/c of wild type p53...many times resistance requires gene amplification, which you need mutatnt p53 for
probably could cure cancer with high doses, but toxicity is
just too high
in general liquid/solid tumors are more amenable to high dose chemo
liquid
why has chemo + small molecules not worked
small molecules are cytostatic, chemo works at level of DNA to be cytocidal and requires active cells
***ways to target VEGF pathway
1. Ab to VEGF
2. decoy receptor to bind VEGF
3. Ab to VEGFR
4. ribozymes that degrade VEGFR
5. TKI of VEGFR
***ER_ breast cancer is eligible for hormonal therapy
+
1/__ compounds screened make it to market
10K

20 year patent protection

takes about 16 years to get through pipeline, 1 billion dollars

(and antineoplastic agents have lowest success rate)

2x as many developing than abx or cv drugs
clinical trial phases
I:
safety and tolerability
pharmacokinetics
normal and targeted pops (in cancer, mostly just cancer pts)

II:
single arm, single institution
proof of concept - efficacy in different tumor types - look for 1 in 12-15 (1 in 5 has a good chance of approval)
dose ranging
safety and pharmacokinetics refined in special population

III:
large, multi-center
placebo-controlled
often replicated

IV:
after approval
adverse event reporting
new indications and uses
average 4-5 years
endpoints for FDA
time to progression
survival
relief of symptoms
delay of event
***phase I metabolism most often by
CYP3A4/5/7

UGTs
***5FU metabolism can be affected by

irinotecan?
DPD polymorphism

UGT polymorphism
***sorafenib MOA

treats

toxicity
inhibits b-RAF and c-RAF (kinases)

RCC

bleeding, rash, hypertensions
**monitor BP
***sunitinib MOA

treats
TKI - antiangiogenesis

RCC, GIST
***methotrexate class

MOA


toxcitiy
anti-metabolite (for cancer)

folic acid analog that blocks S phase

teratogen; myelosuppression, reversible with leucovorin
***5-FU class

MOA

toxicity
anti-metabolite (for cancer)

act on S phase; pyrimidine analog that complexes folic acid, inhibits thymidylate

cerebellar toxicity!!!
myelosuppression reversible with thymidine
photosensitivity
***cyclophosphamide class

MOA

toxicity
alkylating agent

alkylates at guanines

requires bioactivation by liver

hemorrhagic cystritis
***paclitaxel MOA

toxicity
taxane

acts on M phase; inhibits microtubule disassembly

allergy to castor oil in formulation
infertility
rash, flushing, chest pain
***docetaxotere MOA

toxicity
taxane

acts on M phase; inhibits microtubule disassembly

allergy to castor oil in formulation
infertility
rash, flushing, chest pain
***vincristine class

MOA

toxicity
vinca alkaloids

acts on M phase; inhibits microtubule formation by binding MTs

neurotoxicity
paralytic ileus
***irinotecan class

MOA

toxicity
camptothecins

topoisomerase I inhibitor

alopecia!!!
GI
***doxorubicin class

MOA

toxicity
anthracyclines

topoisomerase II inhibitor (DNA intercalation)

cardiotoxicity
***bleomycin class

MOA

skin changes
antibiotic

acts on G2; induces free radical damage

pulmonary fibrosis, skin changes
***carboplatin MOA

toxicity
cross links DNA --> apoptosis

**nephrotoxicity, acoustic nerve damage
***cisplatin MOA

toxicity
cross links DNA --> apoptosis

**nephrotoxicity, acoustic nerve damage
***oxaliplatin MOA

toxicity
alkylating agent --> evenutal apoptosis

does not cause alopecia;
irreversible neurotoxicity (cold intolerance, paresthesias)
***bevacizumab MOA

toxicity
Ab against VEGF

bleeding, thrombosis, wound healing problems...wait 6 weeks until surgery!
***cetuximab MOA

toxicity
Ab against EGFReceptor; prevents dimerization

full body rash...but this is a good sign! good efficacy, better prognosis

efficacy blocked by downstream Ras mutations
***tamoxifen MOA

treats

toxicity
SERM - blocks binding of estrogen to receptor

ER+ breast ca

may increase endometrial Ca b/c of partial agonist effects
hot flashes
***leuprolide MOA

treats

toxicities
GnRH agonist - shuts down estrogen/androgen production

ER+ breast cancer, prostate cancer

hot flashes, vaginal bleeding, amenorrhea, AVOID preganancy

gynecomastia, impotence, transient rise in testosterone, alopecia, weight gain
***anastrazole MOA

treats

toxicity
aromatase inhibitor; interferes with estrogen production in peripheral tissues

ER+ breast cancer

hot flashes, osteoporosis
***bicalutamide MOA

treats

toxicity
non-steroidal anti-androgen that binds to cytosoal ARs

prostate cancer

hot flashes, gynecomastia
***arsenic trioxide treats
APL - induces morphological changes and DNA fragmentation in NB4
*benign adult renal tumors

malignant
renal papillary adenoma
angiomyolipoma
oncocytoma

RCC
urothelial carcinoma (pelvis)
*__% of renal cancers are RCC
85% (others are urothelial Ca)

(usually older people, 2:1 M:F)

3% of all new cancers

it's an adenocarcinoma
*RCC risk factors
smoking
obesity (esp in women)
hypertension
unopposed estrogen
metal exposure
CRF
tuberous sclerosis
*most RCCs are ___
sporadic

familial 4%, occur in younger age group

vonHippel Lindau
hereditary clear cell
hereditary papillary carcinoma
*types of RCC
clear cell Ca (most common - proximal tuule)
papillary Ca
chromophobe RCC
collecting duct Ca (1% but most aggressive...associated with medullary carcinoma, SS disease...can become sarcamoid)
urothelial carcinoma stratification
80% in situ
20% invasive (all malignant)

in situ could be:
papillary (benign papilloma, low malignant potential, malignant)
or flat urothelial CIS (malignant)
presentation of bladder tumors
painless hematuria

tend to recur and progress

60% are single
70% are localized
urothelial carcinoma variants
squamous cell
adeno
mixed
small cell

all worse prognosis
mesenchymal tumors of bladder
leiomyoma (benign)

rhabdomyosarcoma (child, malignant)
leiomyosarcoma (adult, malignant)
familial melanoma caused by
CDK inhibitor mutation (p16 mutation)
when RB is active, it is a ___
brake on the cell cycle
either CDKs gain function or __ lose function...not both
CDKi's or RB
***overall, p53 mutated in __% of cancers
50
can inactivated p53 by activating __ (degrades it)
MDM2

maybe you can block this with Nutlin?
Cyclin D1 is overexpressed in a lot of breast cancers; if the cancer is ER-, the prognosis is __; if ER+, __

if D1+ and Her2+,
worse

better

worse prognosis

thus limited predictive value...complex issue
***p27 loss leads to __ cancer
prostate, breast

(enhanced proteolysis, mislocalization)
***RB inhibits proliferation through formation of __ that inhibit __ phase

RB is inhibited by ___

that complex is inhibited by __
transcriptional repressor complexes

S

CyclinD1-CDK4 complex (which phosphorylates RB)

p16 (so p16 allows RB to put on its brake)
p27 is degraded by
Skp2

increased Skp2 is bad
MDM2 ubiquitination of p53...
targets it for degradation