• 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

1/182

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;

182 Cards in this Set

  • Front
  • Back
Erythrocyte: lineage and precursors
Myeloid lineage
Pluripotent hematopoietic stem cell
proerythroblast
reticulocyte
erythrocyte
Neutrophils, eosinophils, basophils: lineage and precursors
Myeloid lineage
Myeloblast
Promyelocyte
Myelocyte
Metamyelocyte
Stab (Band) cell
N, E, B cells
Platelets: lineage and precursors
Myeloid lineage
Megakaryoblast
Megakaryocyte
Platelet
Macrophages: lineage and precursors
Myeloid lineage
Monoblast
Monocyte
Macrophage
Plasma cell: lineage and precursors
Lymphoid stem cell lineage
Lymphoblast
B cell
Plasma cell
Active T cell: lineage and precursors
Lymphoid lineage
Lymphoid stem cell
Lymphoblast
T cell
Activated T cell
Natural killer cell: lineage and precursors
Lymphoid lineage
Lymphoid stem cell
Pro-NK
Natural killer cell
What is the main source of energy for RBC? Where is it derived from?
Glucose

90% from anaerobic respiration to lactate

10% from the HMP shunt
What are the components of the dense granules of platelets?
ADP and calcium
What are the components of the α-granules of platelets?
vWF and fibrinogen
What is the lifespan of a platelet?
8-10 days
Where can 1/3 of the platelet pool be found?
In the spleen
What is the normal count for leukocytes?
4,000 to 10,000 / mcL
What do the granules of basophils contain?
Heparin, histamine, leukotrienes (LTD4), and other vasoactive amines
What is released upon mast cell degranulation? Can bind what to its membrane? What prevents degranulation? Involved in what type of HSR?
Histamine, heparin, eosinophil chemotactic factors.

Can bind IgE to membrane.

Cromolyn sodium prevents degranulation.

Involved in type I HSR
What does eosinophils use to defend against protozoa and helminths? Produces what? Why?
Major basic protein

Produces histaminASE and arylsulfatase (mutated in metachromatic leukodystrophy) to help limit the reaction after mast cell degranulation.

NAACP

Neoplasm
Asthma
Allergic processes
Collagen vascular disease
Parasites (invasive)
What do the granules of neutrophils contain?
hydrolytic enzymes, lysozyme, myeloperoxidase, and lactoferrin.
What activates macrophages? Can present what type of MHC? secretes what?
IFN-γ

Can present MHC type II

Secretes:
TNF-α
IL-1
IL-6
IL-12 (activates NK-cells and Th1 cells)
Dendritic cells: present what on the surface?
MHC II and Fc receptor

Main inducers of primary antibody response
Dendritic cells express what 2 receptors on their surface?
MHC II and Fc receptor
Where do B cells mature? Where do B cells migrate to? What CDs are on surface?
Mature in the marrow and then migrate to the peripheral lymphoid tissues (follicles of LN, white pulp of the spleen, unencapsulated lymphoid tissue).

Express
CD19
CD20
CD40 (which binds to CD40L on T cells)
What CD do all T cells have?
CD3
Blood type A has what antigen and what antibody?
A antigen and B antibody
Blood type AB has what antigen and what antibody?
A and B antigens and NO antibodies (universal recipient)
Hemophilia A: deficiency of what?
of factor VIII --> intrinsic pathway defect --> inc PTT

Macrohemorrhage in hemophilia - hemarthroses (bleeding into joints), easy bruising, inc PT and PTT.
Hemophilia B: deficienty of what?
of factor IX

Macrohemorrhage in hemophilia - hemarthroses (bleeding into joints), easy bruising, inc PT or PTT
Vitamin K deficieny: deficiency of what? What drug inhibits these? What values do you have to follow with these?
II, VII, IX, X, proteins C and S

Warfarin inhibits these

Follow PT
What does antithrombin do? What does heparin do to antithrombin? What lab values should be followed?
Antithrombin inhibits II, IXa, Xa, XIa, and XIIa

Antithrombin is activated by heparin

Follow PTT
vonWillebrand factor connects what to what?
connects subendothelial collagen to platelets via GpIb
What is the function of clopidogrel and ticlopidine?
They irreversibly block ADP receptors on platelets and prevent the expression of the GpIIb/IIIa receptors on the surface.
Bernard-Soulier syndrome: defect? Platelet count? Bleeding time?
Deficiency of GpIb, expressed on platelets and connects platelet to the subendothelial collagen via von Willebrand factor.

Platelet count is down, bleeding time is up.
Glanzmann's thrombasthenia: defect? platelet count? bleeding time?
Deficiency of GpIIb/IIIa, which connects platelets and platelets via fibrinogen.

Labs: blood smear shows no platelet clumping

Platelet count is normal, bleeding time is increased
What do aggregated platelets express?
Thromboxane A2
Thrombomodulin: where can it be found? What does it do?
Thrombomodulin is expressed on the surface of (damaged?) vascular endothelial cells.

It activates protein C
Platelet aggregation is a balance between pro-coagulant and anti-aggregation factors. What are they?
Pro-aggregation: thromboxane A2 (released by platelets)

anti-aggregation: PGI2 (inhibits platelets aggregation and promotes vascular permeability) and NO. Both are released by vascular endothelial cells
What is abciximab?
A mab that is a receptor antagonist of the GpIIb/IIIa receptor

Mainly used after procedures involving the coronary artery, like angioplasty
What effect does calcium have on the platelet plug?
It strengthens the plug.
What converts vitamin K to activated vitamin K? What inhibits this? What pts lack this?
epoxide reductase. Inhibited by warfarin. Neonates lack enteric bacteria which produce vitamin K.
von Willebrand factor carries/protects what factor?
factor VIII
What does activated protein C do?
It cleaves and inactivates Va and VIIIa
What is the pathophys of factor V Leiden mutation?
Factor V Leiden produces a factor V that is resistant to inactivation by protein C --> hypercoagulability
tPA converts what to what?
Plasminogen to plasmin
Acanthocyte (spur cell)
Liver disease, abetalipoproteinemia
Basophilic stippling
Baste the ox TAIL

Thalassemias
Anemia of chronic disease
Iron deficiency
Lead poisoning
Bite cell
G6PD deficiency
Elliptocyte
Hereditary elliptocytosis
Macro-ovalocyte
Megaloblastic anemia (also see hypersegmented PMNs), marrow failure
Ringed sideroblasts
Sideroblastic anemia
Schistocyte, helmet cell
DIC, TTP/HUS, traumatic hemolysis
Teardrop cell
Bone marrow infiltration (e.g. myelofibrosis)
Target cell
Hbc disease
Asplenia
Liver disease
Thalassemia

HALT said the hunter to his target

Note that lead poisoning presents with basophilic stippling, not target cells
Heinz bodies
Oxidation of iron from ferrous to ferric leads to denatured Hgb precipitation and damage to RBC membrane. Leads to formation of bite cells.

Assoc w α-thalassemia, G6PD deficiency.
Howell-Jolly bodies
Basophilic nuclear remnants found in RBCs.

Seen in pts with functional hyposplenia or asplenia
Pappenheimer bodies
Iron containing granules that are seen because the iron aggregates within mitos and ribosomes. Appear as faint violet or magenta speckles. Assoc w severe anemias and thalassemias
Cabot rings
Thin, red-staining threadlike strands in the shape of a loop or figure 8 that are found on rare occasions in RBCs.

Assoc w megaloblastic anemia, lead poisoning, disorders of erythropoiesis
What are the five main pathologies that lead to a microcytic, hypochromic (MCV <80) anemia?
1. Iron deficiency
2. α-thalassemia
3. β-thalassemia
4. Lead poisoning
5. Sideroblastic anemia
Iron deficiency anemia: what type of anemia will you see?
Microcytic (MCV < 80) hypochromic anemia
Plummer-Vinson syndrome
Iron deficiency anemia
Esophageal webs
Atrophic glossitis
α-thalassemia: Where is it prevalent? What is the defect? How many genes are involved? Consequences of more or less genes being involved?
Prevalent in Asia and Africa.

Defect: α-globin gene mutations that lead to dec α-globin synthesis

4 genes involved

Deletion of 4 genes is incompatible with life. This leads to Hb Barts (γ4), wich causes hydrops fetalis.

Deletion of 3 genes leads to HbH disease (β4)

Deletion of 1-2 genes is not assoc w anemia
Hb Barts
(γ4)

4 deletions in the α locus, not compatible with life --> hydrops fetalis

Note that the γ chains have a high affinity for O2 than normal, so it dec O2 delivery to tissues
HbH disease
β4

three deletions in the α locus --> HbH disease

HbH disease is comprised of Hb Barts γ4 and one Hb hemoglobin. Both of these have a higher affinity for hemoglobin, thus reducing oxygen delivery to tissues

See microcytic hypochromic anemia with

Target cells, Heinz bodies (precipitated HbH) as well as splenomegaly
α-thalassemia trait
Means that there are two deletions in the α locus. But two normal alleles are good enough for almost normal erythropoesis, but there is a mild microcytic hypochromic anemia

Can be cis (both deletions on one chr, seen in Asians) or can be trans (deletions are on different chr, seen in blacks)
β-thalassemia: prevalent in what pops? What is the defect?
Prevalent in Med pops

Defect is in point mutations in splicing sites and promoter sequences.
β-thalassemia minor: what is the defect? sx? diagnosis confirmed by?
Defect is that the β chain is underproduced.

Usually asymptomatic

Diagnosis is confirmed by increased HbA2 (> 3.5%)
See inc HbA2 and inc HbF, what do you think?
β-thalassemia minor
β-thalassemia major: what is the defect? what are the sx? what is the txt? Other sequelae?
β chain is absent --> severe anemia requireing blood transfusion (risk of secondary hemochromatosis)

See marrow expansion ("crew cut" on an X-ray) --> skeletal deformities and chipmunk facies

NOTE: also see the crew cut formation in sickle cell!
See inc HbF, what do you think?
Either β-thalassemia minor or major
"Crew cut" on skull x-ray: what do you think?
β-thalassemia major --> marrow expansion

Also see with sickle cell disease
HbS/β-thalassemia heterozygote
mild to moderate sickle cell disease depedning on the amount of β-globin production
What two enzymes does lead poisoning inhibit? What does this result in? Also leads to what and results in what?
Ferrochelatase and ALA dehydratase

Results in decreased heme synthesis

Also results in RNA degredation which leads to basophilic stippling
Sideroblastic anemia: what is the defect? Txt? Reversible etiologies? Effects on iron panel? Histo?
Defect in heme synthesis. Hereditary form is an X-linked defect in the δ-aminovulinic acid synthase gene (the rate-limiting step).

Txt: pyridoxine (B6) therapy

Reversible etiologies: alcohol, lead

See
inc iron
normal TIBC
inc ferritin

Histo: ringed sideroblasts (with iron-laden mitos)
What is the composition of HbF and HbA? When does the switch from HbF to HbA occur?
HbF (α2γ2)
HbA (α2β2)

The switch occurs around 6 months old. Children with β-thalassemia become anemic around this time.
What are the three conditions that cause a macrocytic (MCV > 100) anemia?
1. Megaloblastic anemia caused by folate deficiency

2. Megaloblastic anemia caused by B12 deficiency

3. Nonmegaloblastic macrocytic anemias
Macrocytic anemia: what is the main problem?
Impaired DNA synthesis --> maturation of nucleus of RBC delayed relative to maturation of cytoplasm
What two enzymes does lead poisoning inhibit? What does this result in? Also leads to what and results in what?
Ferrochelatase and ALA dehydratase

Results in decreased heme synthesis

Also results in RNA degredation which leads to basophilic stippling
Sideroblastic anemia: what is the defect? What is the defect assoc w the hereditary form? Txt? Reversible etiologies? Effects on iron panel? Histo?
Defect in heme synthesis. Hereditary form is an X-linked defect in the δ-aminovulinic acid synthase gene (the rate-limiting step).

Txt: pyridoxine (B6) therapy

Reversible etiologies: alcohol, lead

See
inc iron
normal TIBC
inc ferritin

Histo: ringed sideroblasts (with iron-laden mitos)
What is the composition of HbF and HbA? When does the switch from HbF to HbA occur?
HbF (α2γ2)
HbA (α2β2)

The switch occurs around 6 months old. Children with β-thalassemia become anemic around this time.
What are the three conditions that cause a macrocytic (MCV > 100) anemia?
1. Megaloblastic anemia caused by folate deficiency

2. Megaloblastic anemia caused by B12 deficiency

3. Nonmegaloblastic macrocytic anemias
Macrocytic anemia: what is the main problem?
Impaired DNA synthesis --> maturation of nucleus of RBC delayed relative to maturation of cytoplasm
Megaloblastic anemia caused by folate deficiency: Findings, etiologies
Findings:
Hypersegmented neutrophils, glossitis, dec folate, inc homocysteine, but NORMAL methylmalonic acid

Etiologies: malnutrition (alcoholism), malabsorption, impaired matabolism (e.g. methotrexate, trimethoprim), inc requirement (e.g. hemolytic anemia, pregnancy)
Megaloblastic anemia caused by B12 deficiency
Findings:
Hypersegmented neutrophils, glossitis, dec B12, inc homocysteine, but INC methylmalonic acid

Etiologies: malnutrition (alcoholism), malabsorption (Crohn's), pernicious anemia, Diphyllobothrium latum (fish tapeworm)

Neurologic symptoms: subacute combined degeneration (due to involvement of B12 in fatty acid pathways)
What is a way to distinguish folate def from B12 def?
Folate: normal methylmalonic acid

B12 def: inc methylmalonic acid
Nonmegaloblastic macrocytic anemias
1. Liver disease
2. Alcoholism: macrocytosis and bone marrow suppression can occur in the absence of folate/B12 deficiency

3. Reticulocytosis: reticulocytes are bigger than mature RBCs, leading to an increased MCV

4. Metabolic disorder (e.g. orotic aciduria): congenital deficiencies of purine or pyrimidine synthesis

5. Drugs: 5-FU, AZT, hydroxyurea.
Anemia of chronic disease, aplastic anemia, kidney disease: what type of anemia do these cause?
Nonhemolytic, normocytic, normochromic anemia
Hereditary spherocytosis, G6PD def, pyruvate kinase def, sickle cell, HbC defect, paroxysmal nocturnal hemoglobinuria: what type of anemia do these cause?
Intrinsic hemolytic, normocytic, normochromic anemia
autoimmune, microangiopathic (DIC, TTP/HUS), infectious agents: what type of anemia do these cause?
Extrinsic hemolytic, normocytic, normochromic anemia.
PNH, autoimmune (cold agglutinins), mechanical destruction (e.g. aortic stenosis, mechanical valve), microangiopathies (DIC, TTP/HUS): what type of anemia do these cause?
Intravascular hemolysis, normocytic, normochromic anemia
Herediatry spherocytosis, G6PD deficiency, pyruvate kinase def, sickle cells anemia, HbC defect, autoimmune (warm agglutinins): what type of anemia do these cause?
Extravascular hemolysis, normocytic, normochromic anemia.
Anemia of chronic disease: Iron panel, what is up and what is down? What can happen in long standing disease?
Inflammation --> inc hepcidin (sequesters iron) --> less release of iron from macrophages --> dec iron --> dec TIBC --> inc ferritin

Can become microcytic, hypochromic in long-standing disease
Aplastic anemia: pathologic features? causes (4)? Sx? Treatment?
Pathologic features: pancytopenia charcterized by severe anemia, neutropenia, and thrombocytopenia. Normal cell morphology, but hypocellular bone marrow with fatty infiltration.

Causes:
1. Radiation and drugs (benzene, chloramphenicol, alkylating agents, antimetabolites)
2. Viral agents (parvovirus B19, EBV, HIV)
3. Fanconi's anemia (inherited defect in DNA repair)
4. Idiopathic (immune mediated, primary stem cell defect) - may follow acute hepatitis

Sx: fatigue, malaise, pallor, purpura, mucosal bleeding, petechiae, infection.

Treatment: withdrawal of offending agent, immunosuppressive regimens (antithymocyte globulin, cyclosporin), allogenic BM transplant, RBC and platelet transfusion, G-CSF, or GM-CSF

Note that antithymocyte globulin is an infusion of horse or rabbit derived abs agains human T cells.
Hereditary spherocytosis: what type of anemia? defect? pathogenesis? findings? labs?
Intrinsic hemolytic normocytic anemia

Defect: defect in proteins interacting with RBC membrane skeleton and plasma membrane (e.g. ankyrin, band 3.1, or spectrin)

Path: less mem causes small and round RBCs with no central pallor (Inc MCHC, inc RDW (red cell distribution width)) --> premature removal by spleen

Findings: splenomegaly, aplastic crisis (B19 infection). Howell-Jolly bodies present after splenectomy. Can also see bilirubin gallstones secondary to hemolysis.

Labs: positive osmotic fragility test.
Pyruvate kinase deficiency: what type of anemia? Path? Presenation?
Intrinsic hemolytic normocytic anemia

Path: defect in pyruvate kinase --> dec ATP --> rigid RBCs

Presentation: hemolytic anemia in the newborn
Sickle cell disease: what % of AAs carry the trait? What % have the disease? Why are newborns usually asymptomatic? What is the mutation in SC? What promotes sickling?
Trait = 8%, disease = 0.2%

Newborns are usually asymptomatic due to inc HbF and dec HbS

The mutation is a singel aa replacement in the β chain (glutamic acid --> valine)

Low O2 or dehydration precipitates sickling.
What are some complications that sickle cell homozygotes face?
1. Aplastic crisis (due to parvovirus B19 infection)

2. Autosplenectomy --> inc risk of infection with encapsulated organisms

3. Salmonella osteomyelitis

4. Painful crisis (vaso-occlusive)

5. Renal papillary necrosis (due to low O2 in papilla, esp vasa recta)

6. Splenic sequestration crisis (spleen lacks collateral blood supply)
What is the treatment for Sickle cell anemia?
Hydroxyurea (inc HbF) and BM transplantation
HbC defect: path? presentation? histo?
This mutated form reduces the normal plasticity of host erythrocytes causing a hemoglobinopathy

In homozygotes, nearly all Hb is in the HbC form, resulting in mild hemolytic anemia

See target cells,
Paroxysmal nocturnal hemoglobinuria
Path: intravascular hemolysis due to inc complement-mediated RBC lysis (impaired sysnthesis of GPI anchor/decay-accelerating factor [CD55, CD59] in RBC membrane).

Especially susceptible to lysis by complement in an ACIDIC environment. Stress exacerbates lysis (infection, menstruation, surgical procedures, exposure to cold, vaccines, etc.).

Especially prone to VENOUS THROMBOSIS --> abdominal pain --> death (via Budd-Chiari)

Also see pigment gallstones and granulocytopenia leading to frequent infections.

Also aplastic anemia/leukemias

Labs: see increased URINE HEMOSIDERIN. Can also do the "sugar-water" test or the Ham test
Warm agglutinin: seen with what disease states?
Seen in CHRONIC anemia with SLE, in CLL, or with certain drugs (e.g. α-methyldopa)

Warm weather is GGGreat
Cold agglutinin: triggered by what? assoc w what diseases?
ACUTE anemia triggered by cold

Seen with Mycoplasma pneumoniae infections or infectious mononucleosis

Cold ice cream - MMM
Autoimmune hemolytic anemias are usually positive for what test?
For the Coombs test
Explain the direct Coombs test
Anti-Ig antibody added to pt's RBCs, agglutinate if RBcs are coated with Ig.
Explain the indirect Coombs test
Normal RBCs are added to pt's serum, agglutinate if serum has anti-RBC surface Ig.
In iron deficiency anemia, what is the:

Serum iron?
Transferrin and TIBC?
Ferritin?
% transferrin saturation (serum Fe/TIBC)?
Serum iron = low

Transferrin and TIBC = HIGH (the liver produces more tranferrin, presumably to maximize use of the little iron that is available)

Ferritin = Low

% transferrin saturation (serum Fe/TIBC) = Low
Anemia of chronic disease:

Serum iron?
Transferrin and TIBC?
Ferritin?
% transferrin saturation (serum Fe/TIBC)?
Serum iron = low (b/c the body "holds" iron intracellularly as ferritin)

Transferrin and TIBC = LOW (The body produces less transferrin to keep the Fe away from pathogens)

Ferritin = HIGH

% transferrin saturation (serum Fe/TIBC) = unchanged
Hemochromatosis:

Serum iron?
Transferrin and TIBC?
Ferritin?
% transferrin saturation (serum Fe/TIBC)?
Serum iron = INC

Transferrin and TIBC = low

Ferritin = high

% transferrin saturation (serum Fe/TIBC) = very high
Pregnancy/OCP use:
Serum iron?
Transferrin and TIBC?
Ferritin?
% transferrin saturation (serum Fe/TIBC)?
Serum iron = unchanged

Transferrin and TIBC = HIGH (just because the liver is working overtime???)

Ferritin = unchanged

% transferrin saturation (serum Fe/TIBC) = low
Lead poisoning: What are the two affected enzymes? What is the accumulated substrate? What are the sx in general? In children? In adults?
Ferrochetalase and ALA dehydratase

Protoporphyrin (blood)

General sx: Microcytic anemia, GI, and kidney disease. Lead lines on gingivae (Burton's lines) and on the epiphyses of long bones on x-ray. See encephalopathy and Erythrocyte basophilic stippling. Abdominal colic and sideroblastic anemia. DROPS - wrist and foot drop.

sx/txt in children: exposure to lead paint --> MR. Succimer for kids (It "sucks" to be a kid who eats lead)

sx/txt in adults: environmental exposure (battery/ammunition factory) --> HA, memory loss, demyelination. Dimercaprol and EDTA are first line.
Acute intermittent porphyria: affected enzyme? Accumulated substrate? Sx? Txt?
Affected enzyme: Porphobilinogen deaminase (aka uroporphyrinogen-I-synthase)

Accumulated substrate: porphobilinogen, δ-ALA, uroporphyrin (in urine)

Sx: 5 P's

Painful abdomen
Pink urine
Polyneuropathy
Psychological disturbances
Precipitated by drugs (esp inducers of CYP 450 like barbs/alcohol)

Note that pts are normal except when affected by env, drugs.

Seen more commonly in women
Txt: glucose and heme, which inhibit ALA synthase (and thus the accumulation of toxic precursors)
Porphyria cutanea tarda: affected enzyme? Accumulated substrate? sx?
Affected enzyme: Uroporphyrinogen decarboxylase

Accumulated substrate: Uroporphyrin (tea-colored urine)

Presentation: blistering cutaneous photosensitivity. Most common porphyria
Idiopathic thrombocytopenic purpura (ITP): defect? Platelet count? Bleeding time? Nature of this illness? Histo? See inc what type of cell?
Anti GpIIb/IIIa antibodies --> peripheral platelet destruction

Systemic illness is not present in ITP!

No schistocytes!

Platelet count down, bleeding time up.

Labs: increased megakaryocytes
Thrombotic thrombocytopenic purpura (TTP): defect? causing what? Labs? PC/BT? sx?
Deficiency of ADAMTS13 (vWF metalloprotease) --> dec degredation of vWF multimers.

Pathogenesis: inc amounts of large vWF multimers --> inc platelet aggregation and thrombosis

Labs: schistocytes, inc LDH

PC down
BT up

Sx: pentad of:
neurologic
renal symptoms
fever
thrombocytopenia
microangiopathic hemolytic anemia

NOTE that TTP is similar to HUS, but HUS has no neuro sx.
von Willebrans's disease: path?


PC:
BT:
PT:
PTT:
vWF carries/protects FVIII, so dece vWF leads to an intrinsic pathway defect and normal or inc PTT

dec vWF --> defect in platelet to collagen adhesion

Mild but most common bleeding disorder

PC: normal
BT: inc
PT: normal
PTT: normal or inc
DIC: path? labs? PC/BT?PT/PTT?
Widespread activation of clotting leads to a deficiency in clotting factors, which creates a bleeding state.

Labs:
Schistocytes
Inc fibrin split products (D-dimers)
DEC fibrinogen
Dec factors V and VIII


PC: dec
BT: inc
PT: inc
PTT: inc
Prothrombin gene mutation: associated with what?
mutation in the 3' untranslated region, assoc w venous clots.
ATIII deficiency: see what after administration of heparin?
Inherited deficiency of antithrombin III; reduced inc in PTT upon administration of heparin (because heparin works through antithrombin)
Protein C or S deficiency: has what consequences? associated with danger upon giving what drug?
Dec ability to inactivate factors V and VIII.

Inc risk of hemorrhagic skin necrosis following administration of warfarin.

WISN is a potentially catastrophic complication of warfarin therapy that arises as a consequence of the different half-lives of the vitamin K-dependent proteins. One day after initiation of usual doses of warfarin, protein C activity is reduced by approximately 50%. Owing to their longer half-lives, the levels of the vitamin K-dependent clotting factors II, IX, and X decline more slowly (factor VII activity declines at approximately the same rate as protein C). The reduced level of protein C activity relative to these other procoagulant molecules creates a transient hypercoagulable state. This effect is more pronounced when large loading doses of warfarin are administered. Indeed, WISN typically occurs during the first few days of warfarin therapy, often when daily doses in excess of 10 mg are administered
Hodgkin's lymphoma:
Distinctive cell type?
Typical sx?
See what type of lymphadenopathy?
50% of cases assoc w what?
Demographics?
Good prognosis = what?
Hodgkin's lymphoma:
Distinctive cell type: Reed-Sternberg cell

Typical sx: Constitutional "B" signs/sx: low-grade fever, night sweats, weight loss

See what type of lymphadenopathy: nontender mediastinal lymphadenopathy

50% of cases assoc w what: EBV

Demographics: bimodal: young and old; more common in men except for nodular sclerosing type.

Good prognosis = high lymphocytes, low RS cells
Non-hodgkin's lymphoma:
Assoc w what diseases?
Spread?
Sx?
Demographics?
Assoc w what diseases: HIV and immunosuppression

Spread: multiple, peripheral nodes; extranodal involvement common; noncontiguous spread

Sx: fewer constitutional signs/sx

Demographics: peak incidence for certain subtypes at 20-40 years of age
Reed-sternberg cells: description? CDs? RS cells are undergoing what?
binucleate or bilobed with the 2 halves as mirror images ("owl's eyes")

Necessary but not sufficient for dx

CD30+ and CD15+

Cells in RS are undergoing "hypersomatic hypermutation"
Name the four types of Hodgkin's lymphoma in order of decending incidence
Nodular sclerosing (65-75%)
Mixed cellularity (25%)
Lymphocyte predominant (6%)
Lymphocyte depleted (rare)
Nodular sclerosing type of Hodgkin's disease:

RS
Lymphocytes
Prognosis
Comments: path? demo?
RS: low
Lymphocytes: high
Prognosis: excellent
Comments: Most common; see collagen banding, lacunar cells; women > men; primarily young adults
Mixed cellularity type Hodgkin's disease:


RS
Lymphocytes
Prognosis
Comments
RS: ++++
Lymphocytes: +++
Prognosis: intermediate
Comments: numerous RS cells
Lymphocyte predominant Hodgkin's lymphoma:


RS
Lymphocytes
Prognosis
Comments
RS: +
Lymphocytes: ++++
Prognosis: excellent
Comments: < 35 year old males
Lymphocyte depleted type Hodgkin's lymphoma:


RS
Lymphocytes
Prognosis
Comments
RS: high relative to lymphocytes

Comments: older males with disseminated disease
Burkitt's lymphoma:

Ages?
Translocation? what genes are involved?
Variations?
Histopath?
Associated with what? What other malignancies can this cause?
2 common presentations
Adolescents and young adults

t(8;14) c-myc gene moves next to the heavy-chain Ig gene (14)

t(8;22) myc/lambda
t(2;8) kappa/myc

"Starry sky" appearance

Assoc w EBV (which is assoc with nasopharyngeal cancer)

Endemic form in Africa: jaw lesion
Sporadic form in abdomen or pelvis
Diffuse large cell lymphoma:

Occurs in
Comments:
Non-Hodgkin's lymphoma

Occurs in usually older adults, but 20% in children.

Most common adult NHL
May be mature T cell in origin
Mantle cell lymphoma

demo?
translocation?
prognosis?
CD?
NHL

Older adults

t(11;14)

Poor prognosis

CD5+
Follicular lymphoma:

Occurs in?
Translocation?
Expression of what?
Course?
NHL

Occurs in adults

t(14;18) --> bcl-2 expression

Difficult to cure, indolent course; bcl-2 inhibits apoptosis
Adult T-cell lymphoma:

Caused by?
Presentation?
See in what pops?
Course?
Caused by HTLV-1

Adults present with cutaneous lesions; esp affects pops in Japan, West Africa, Caribbean

Aggressive
Mycosis fungoides/Sezary syndrome: presentation? Course?
Cutaneous T cell lymphoma

Adults present with cutaneous patches/nodules

Indolent
Malignant histiocytosis: description
A common subtype of NHL. Large cells w/ vesicular nucleus and prominent nucleolus. Nontender LNs or extranodal involvement
Multiple myeloma:

Produces large amounts of what?
Hyper what?
General sx?
Assoc w what amyloidosis?
lesions on x-ray?
Characterized by what on serum protein electrophoresis?
See what other distintive finding in urine?
RBC effects?
Produces large amounts of IgG (55%) or IgA (25%)

Hypercalcemia

Renal insufficiency, inc susceptibility to infection, anemia

AL

Punched out lesions on x-ray

Characterized by monoclonal immunoglobin spike (M protein) on serum protein electrophoresis.

See Ig light chains in urine (Bence Jones protein [most are VkCk])

Blood smear shows RBCs stacked like poker chips (Rouleaux formation).
Waldenstrom's macrogobulinemia: excess of what? leading to what sx? Lack of what lesions?
Excess IgM --> hyperviscosity symptoms

Lack of lytic bone lesions
Plasmacytoma
Solitary myeloma

Involves soft tissues (lungs, nasopharynx, nasal sinuses). Has plasma cell proliferation with M-spike of IgG, resembling m.myeloma but without metastatic potential. Some develop into mm in 10-20 years
Heavy chain disease
Excessive amounts of heavy chain gamma (IgG), alpha (IgA), or mu (IgM) are made
MGUS: basic description
asymptomatic monoclonal gammopathy
Acute lymphoblastic leukemia/lymphoma (ALL):

CP in children? adolescent males?
Bone marrow replaced by what?
Postive for what?
Responsive to therapy?
May spread to what two places?
What translocation indicates a better prognosis?
May present with BM involvement in childhood or mediastinal mass in adolescent males.

Bone replacement replaced by INC lymphoblasts

TdT+ (marker of pre-T and pre-B cells), CALLA+

Most responsive to therapy

May spread to CNS and testes

t(12;21) --> better prognosis
Small lymphocytic lymphoma (SLL)/Chronic lymphocytic leukemia (CLL):

Affects who?
sx?
see what on peripheral smear?
abs?
What is the difference bet SLL and CLL?
Affects older adults > 60

Often asymptomatic

Smudge cells on peripheral smear

Warm antibody autoimmune hemolytic anemia

SLL is the same as CLL, except CLL has inc peripheral blood lymphocytosis
Hairy cell leukemia

Affects who?
Tumor of what type of cell?
sx?
typical marker?
Affects the elderly

Mature B-cell tumor, cells have filamentous, hairlike projections

Often assoc w massive splenomegaly

Stains with TRAP (tartrate-resistant acid phosphatase)
AML

Characteristic histo?
See inc what on peripheral smear?
responds to what?
Auer rods

Inc circulating myeloblast on peripheral smear

M3 responds to all-trans retinoic acid (vitamin A), inducing differentiation of myeloblasts.
Chronic myelogenous leukemia?

translocation? products?
proliferation of?
May accelerate or transformation into what?
characteristic lab level?
responds to what?
Defined by the Phildelphia chromosome (t[9;22]) makes bcr-abl

Myeloid stem cell proliferation, presents with inc neutrophils, metamyelocytes, basophils

See splenomegaly

May accelerate and transform to AML or ALL ("blast crisis")

Very low LEUKOCYTE ALKALINE PHOSPHATASE (vs. leukemoid reaction)

Responds to imatinib
M0
undifferentiated (MPO negative)
M1/M2
Myeloblastic leukemia without maturation (has MPO)
M4
Myelomonocytic leukemia, has both myeloblasts and monoblasts (nonspecific esterase)
M5
Monocytic leukemia (nonspecific esterase)

High incidence of infiltration, esp of gums
M6
Erythroleukemia, see abnormal erythroid precursors
M7
Acute megakaryocytic leukemia; assoc w myelofibrosis due to PDGF

platelets
Approximate ages:
ALL
AML
CML
CLL
ALL: < 15
AML: median onset ~ 60
CML: 30-60
CLL: > 60
See what inc lab value with the "leukemoid reaction?"
inc leukocyte alkaline phosphatase
Langerhans cell histiocytoses (histiocytosis X):

lineage?
markers?
characteristic histo?
older terms?
Proliferative disorders of dendritic (Langerhans) cells from the monocyte lineage. Defective cells expres S-100 and CD1a. Birbeck granules ("tennis rackets" on EM) are characteristic.

Older terms include:
Letterer-Siwe disease
Hand-Schuller-Christian disease
Eosinophilic granulomas
Polycythemia vera:

RBCs
WBCs
Platelets
Philadelphia chr
JAK2 mutations
Abnormal clone of hematopoietic stem cells are increasingly sensitive to growth factors. Assoc w low levels of EPO

RBCs: high
WBCs: high
Platelets: high
Philadelphia chr: -
JAK2 mutations: +
Essential thrombocytosis

RBCs
WBCs
Platelets
Philadelphia chr
JAK2 mutations
Similar to p. vera, but specific for megakaryocytes

RBCs: normal
WBCs: normal
Platelets: high
Philadelphia chr: -
JAK2 mutations: + (30-50%)
(myeloid metaplasia) with myelofibrosis

RBCs
WBCs
Platelets
Philadelphia chr
JAK2 mutations
Fibrotic obliteration of bone marrow

RBCs: low
WBCs: variable
Platelets: variable
Philadelphia chr: -
JAK2 mutations: + (30-50%)
CML:

RBCs
WBCs
Platelets
Philadelphia chr
JAK2 mutations
bcl-abl transformation leads to inc cell division and inhibition of apoptosis

RBCs: low
WBCs: high
Platelets: high
Philadelphia chr: +
JAK2 mutations: -
Myelodysplastic syndrome (MDS)
refers to myeloid disorders featuring maturation defects and ineffective hematopoesis. MDS produces cytopenia and ACUTE leukemias, whereas myeloproliferative disorders produce polycythemia and CHRONIC leukemia.
Heparin:

What does it do?
Half life?
Onset of action/indications?
Can use during pregnancy because?
Use what for reversal?
Cofactor for the activation of antithrombin, decreases thrombin and Xa

short half life

Immediate anticoagulation for PE, stroke, acute coronary syndrome, MI, DVT.

Doesn't cross placenta, so can be used in pregnancy

For rapid reversal, use protamine sulfate (positively charged molecule that can act by binding negatively charged heparin
Pros and cons of low-molecular weight heparins
Pros:
act more on Xa
have better bioavailability
2-4x longer half life
can be admin subQ and w/o monitoring labs

Cons: not easily reversible
Heparin-induced thrombocytopenia
Heparin binds to platelets causing autoantibody production that destroys platelets and overactivates the remainging ones, resulting in a thrombocytopenic, hypercoagulable state
What drugs are used as alternatives to heparin in pts with HIT?
Lepirudin, bivalrudin
What do you give in the case of a warfarin overdose?
IV vitamin K and FFP
Thrombolytics: name four. MOA? Effect on labs? Treat toxicity with what?
Streptokinase, urokinase, tPA (alteplase), APSAC (antistreplase)

Directly or indirectly aid conversion of plasminogen to plasmin, which cleaves thrombin and fibrin clots.

PT up, PTT up, no effect on PC

Treat tox with: aminocapric acid (an inhibitor of fibrinolysis)
Aspirin toxicity
Aspirin is a weak acid --> OD caused hyperventilation --> pCO2 down --> resp alkalosis --> anaerobic glycolysis --> lactic acid buildup --> elevated anion gap --> metabolic acidosis
Toxicity of ticlopidine
Neutropenia
Vinca alkaloids vs. paclitaxel: MOA
Vinca: inhibit microtubule formaion

Paclitaxel: inhibit microtubule disassembly
MTX causes myelosuppression which is reversible with what drug? What other SE are assoc w MTX?
Reversible with leucovorin (folinic acid)

Macrovascular fatty change in liver; mucositis; teratogenic
5-fluoruracil (5-FU): notable about SE.
Myelosuppresion is not reversible with leucovorin

Rescue with thymidine
6-MP: what does it do? inc tox with what? why?
dec de novo purine synthesis.

Metabolized by XO, thus inc tox with allopurinol
6-thioguanine: has what advantage over 6-MP?
can be given with allopurinol
Cytarabine: what is it? Tox?
Pyrimidine antagonist --> inhibition of DNA polymerase

Leukopenia, thrombocytopenia, megaloblastic anemia
Dactinomycin: used for what? tox?
Used for childhood tumors (children ACT out)

Wilm's tumor, Ewing's sarcoma, rhabdomyosarcoma.

Tox: myelosuppression
Doxorubicin (Adriamycin), daunorubicin: MOA? Part of what drug regimen? Main toxs?
Generate free radicals, noncovalently intercalate in DNA --> breaks in DNA --> dec replication

Part of the ABVD drug regimen for Hodgkin's lymphoma

Cardiotoxicity
Myelosuppression
Marked alopecia
Toxic to tissues with extravasation
Bleomycin: specific to what stage? MOA? Part of what regimen? Tox?
Specific to G2

Induces formation of free radicals, whcih cause breaks in DNA strands

Part of the ABVD regimen for Hodgkin's

Pulmonary fibrosis
Skin changes
minimal myelosuppression
Etoposide (VP-16), teniposide: specific for what stage? MOA? Toxs?
Late S to G2 phase specific.

Inhibits topoisomerase II --> inc DNA degredation

Myelosuppression
GI irritation
alopecia
Cyclophosphamide, ifosfamide: MOA? tox?
Covalently cross link DNA at guanine N-7

Requires bioactivation by the liver

Myelosuppression
Hemorrhagic cystitis - can be partially prevented with mesna
Nitrosureas (carmustine, lomustine, semustine, streptozocin): requires what? goes where? used for what? tox?
Require bioactivation. Cross the BBB.

Used to txt brain tumors, including glioblastoma multiforme

CNS tox (dizziness, ataxia)
Busulfan: MOA? Used for? tox?
Alkylates DNA

Used for CML, also for ablating BM in hematopoietic stem cell transplants

Pulmonary fibrosis, hyperpigmentation
Vincristine, vinblastine: specific for what phase? Part of what drug regimen? tox of vincristine? tox of vinblastine?
Alkaloids that bind to tubulin in the M phase and block polymerization of microtubules so that the mitotis spindle cannot form.

Part of the MOPP (vincristine = Oncovin) regimen for Hodgkin's lymphoma, Wilm's tumor, choriocarcinoma

Vincristine:
Neurotox (areflexia, peripheral neuritis), paralytic ileus

VinBLASTine: BLASTs bone marrow (supression)
Paclitaxel, other taxols: MOA? used for? tox?
Hyperstabilize polymerized microtubules in M phase so that mitotic spindle cannot break down (anaphase acannot occur)

Ovarian and breast carcinomas

Myleosuppression and hypersensitivity
Hydroxyurea: MOA? specific for what phase? used for? tox?
Inhibits Ribonucleotide reductase --> dec DNA synthesis (S-phase specific)

Melanoma, CML, sickle cell disease (inc HbF)

BM suppression, GI upset
Tox assoc w tamoxifen? How is this different from raloxifene?
Tamoxifen may inc the risk of endometrial cancer via partial agonist effects. May also cause hot flashes.

Raloxifene does not cause endometrial carcinoma because it is an endometrial antagonist
Trastuzumab (Herceptin): MOA? Tox?
Monoclonal ab against Her-2 (erb-2). Helps kill cancer cells that overexpress Her-2

Cardioxicity
Imatinib: MOA? use? tox?
Philadelphia chromosome bcr-abl tyrosine kinase inhibitor

CML, GI stromal tumors

Fluid retention