• 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

1/462

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;

462 Cards in this Set

  • Front
  • Back
When B cells and T cells mature, what do they become, respectively?
B cells mature into plasma cells whereas T cells become activated T cells

2010-336
Monocytes develop from what immediate precursor blood cell type?
Monoblasts

2010-336
Promyelocytes, myelocytes, metamyelocytes, and stab cells are precursors for what three granulocytes?
Neutrophils, eosinophils, and basophils

2010-336
Neutrophils, eosinophils, and basophils differentiate from what common precursor blood cell type?
Myeloblasts

2010-336
Reticulocytes are precursors for what type of blood cell?
Erythrocytes

2010-336
Proerythroblasts, lymphoblasts, myeloblasts, monoblasts, and megakaryoblasts share what precursor cell type?
Pluripotent hematopoietic stem cells

2010-336
What precursors indicate that a stem cell has differentiated into the platelet lineage?
Megakaryoblasts, which become megakaryocytes that form platelets

2010-336
Lymphoblasts are the precursors for what two blood cell types?
B cells and T cells

2010-336
In a normal blood sample, name the differential of white blood cells from most to least numerous.
Neutrophils, Lymphocytes, Monocytes, Eosinophils, Basophils (remember: Neutrophils Like Making Everything Better)

2010-336
What molecule do erythrocytes depend upon exclusively for energy?
Glucose; remember, they cannot use ketones for energy

2010-336
What role does the erythrocyte play in acid/base physiology?
Erythrocytes carry carbon dioxide from the periphery to the lungs for elimination

2010-336
How do erythrocytes facilitate transport of carbon dioxide away from the peripheral tissues?
The chloride-bicarbonate antiport produces a physiologic chloride shift, resulting in cellular loss of chloride and gain of bicarbonate (and thus carbon dioxide)

2010-336
What is the average lifespan of an erythrocyte in a normal human host?
120 days

2010-336
What feature of the erythrocyte structure permits easy gas exchange of oxygen and carbon dioxide?
The biconcave disc shape permits a high surface area to volume ratio and facilitates gas exchange

2010-336
How would you describe a peripheral blood smear that shows an increased number of red blood cells and numerous immature red blood cells?
There is an erythrocytosis or polycythemia (increased number of cells) and a reticulocytosis (increased immature cells)

2010-336
Where does an erythrocyte obtain the adenosine triphosphate that it needs for energy?
Glucose: 90% is anaerobically degraded to lactate, whereas 10% goes to the hexose monophosphate shunt

2010-336
How would you describe a peripheral blood smear showing red blood cells of varying sizes and of varying shapes?
This smear exhibits anisocytosis (varying sizes) and poikilocytosis (varying shapes)

2010-336
Where in the body would you typically find the most platelets at any one time?
Approximately one third of the platelet pool is stored in the spleen

2010-336
What is the role of platelets immediately following an injury such as a laceration?
Primary hemostasis; for example, they help prevent leakage of red blood cells from damaged vessels

2010-336
What two types of granules do platelets contain?
Dense granules and alpha granules

2010-336
What are the contents of the dense granules of platelets?
Adenosine diphosphate, calcium

2010-336
You note multiple small red lesions on the skin of a patient that do not blanche with pressure; what are two possible etiologies of these lesions?
These are petechiae, which can be caused by a low platelet count (thrombocytopenia) or dysfunctional platelets

2010-336
What is the life span of a platelet?
8-10 days

2010-336
What happens to platelets when they are in the presence of damaged endothelium?
Platelet activation; "sticky" platelets aggregate and interact with fibrinogen to form a hemostatic plug

2010-336
How do mature platelets form from large megakaryocyte precursors?
Small portions of cytoplasm fragment off of megakaryocytes

2010-336
What are the contents of the α granules of platelets?
von Willebrand's factor, fibrinogen

2010-336
Name three types of granulocytes.
Basophils, eosinophils, and neutrophils

2010-336
What is the main function of leukocytes?
To defend against infection

2010-336
Granulocytes and mononuclear cells are what kind of blood cells?
Leukocytes (leuk = white; cyte = cell)

2010-336
How many leukocytes are normally found per microliter or blood?
4000 to 10,000

2010-336
Name two types of mononuclear cells.
Lymphocytes and monocytes

2010-336
What distinguishes the appearance of a basophil from other granulated cells?
The densely basophilic granules, which stain blue with basic stains

2010-337
What molecules, normally seen as part of the inflammatory cascade, are contained in basophilic granules?
Histamine (which causes vasodilation) and other vasoactive amines, as well as leukotrienes (LTD-4)

2010-337
You note many basophils on a peripheral smear of a patient undergoing an allergic reaction; do you also expect to see mast cells?
No; although both cells are involved in allergic reactions, basophils are found in the blood whereas mast cells are typically only found in tissues

2010-337
What stimulus causes mast cell degranulation?
Immunoglobulin E binding to receptors on the surface of the mast cell

2010-337
What medication can prevent the release of inflammatory compounds from mast cells in a patient with asthma?
Cromolyn sodium, which stabilizes mast cell membranes and prevents degranulation

2010-337
What substances are released when mast cells degranulate?
Histamine, heparin, eosinophilic chemotactic factors

2010-337
Compare and contrast the function and location of basophils and mast cells.
Both types of cells mediate allergic reactions and secrete histamine; however, mast cells bind immunoglobulin E and are found in tissue, whereas basophils are found in the blood

2010-337
Eosinophils are phagocytic cells, particularly in the presence of what?
Antigen-antibody complexes

2010-337
How do eosinophils and mast cells interact during an allergic reaction?
Eosinophils produce histaminase and arylsulfatase to limit the inflammatory reaction from mast cell degranulation

2010-337
Name five diagnoses to consider in a patient discovered to have eosinophilia.
Neoplasm, Asthma, Allergic reaction, Collagen vascular disease, Parasites (remember: NAACP)

2010-337
What distinguishes an eosinophil from other granulated cells?
They are full of large, uniform eosinophilic (pink) granules (eosin = a dye that stains pink; philic = loving)

2010-337
What is the role of major basic protein, found in eosinophils?
Major basic protein is released to fight helminth and protozoan infections

2010-337
A peripheral blood smear shows multiple neutrophils with nuclei that display 6, 7, and even 8 lobes; what lab tests are indicated?
B12 and folate tests; hypersegmented polys (neutrophils) are present in deficiencies of either vitamin

2010-337
What is the appearance of granules found in neutrophils?
Large, spherical, azurophilic granules

2010-337
What are the contents of the granules in neutrophils?
Hydrolytic enzymes, lysozyme, myeloperoxidase, and lactoferrin

2010-337
Under the microscope you note a neutrophil with several granules; what organelle are you looking at?
Lysosome

2010-337
Would you expect to find monocytes in a tissue biopsy?
No. Monocytes are found in the blood, not the tissue; when they enter tissue they differentiate into macrophages

2010-337
Describe the appearance of monocytes.
Large, kidney-shaped nucleus; frosted glass cytoplasm

2010-337
Which cytokine causes activation of macrophages?
γ-Interferon

2010-337
What role do macrophages play in the immunologic response to extracellular pathogens?
They are antigen-presenting cells using major histocompatibility complex class II molecules (present phagocytosed extracellular pathogens)

2010-337
What are dendritic cells in the skin called?
Langerhans cells

2010-338
What molecules do dendritic cells express on their surfaces that are integral to their role as antigen-presenting cells?
Major histocompatibility complex II and Fc receptor

2010-338
What is the major function of B lymphocytes?
To produce antibodies

2010-338
What is the function of T lymphocytes?
They control the cellular immune response and regulate the activity of B lymphocytes and macrophages

2010-338
What kind of antibody response occurs when the body encounters an antigen it has previously seen?
Rapidly secreted, previously formed antibodies because B cells have antigenic memory

2010-338
After maturation, where do B lymphocytes migrate to from the bone marrow?
Peripheral lymphoid tissue (lymph node follicles, white pulp of spleen, other uncapsulated lymphoid tissue)

2010-338
What major histocompatibility complex do B cells possess on the cell surface?
Major histocompatibility complex II, which allows B cells to act as antigen-presenting cells

2010-338
B lymphocytes are part of the _____ (cell-mediated/humoral) immune response.
Humoral

2010-338
What is the response of a B cell after encountering a recognized antigen?
B cells differentiate into plasma cells and secrete antibodies

2010-338
Where do B lymphocytes originate? Where do they mature?
They originate and mature in the bone marrow (remember: B = Bone marrow)

2010-338
You note that a patient's bone marrow shows a monoclonal proliferation of plasma cells; what is the diagnosis?
Multiple myeloma

2010-338
How are plasma cells formed?
When B cells encounter a recognized antigen, they differentiate into plasma cells (which have antigenic memory)

2010-338
What organelles create the appearance of the off-center nucleus and abundant cytoplasm in a plasma cell?
The rough endoplasmic reticulum and Golgi apparatus are plentiful (remember: the plasma cell is a protein (antibody) factory!)

2010-338
What is the site of origin for T lymphocytes? Where do they mature?
They originate in the bone marrow but mature in the thymus (remember: T cells mature in the Thymus)

2010-338
What is the predominate type of lymphocyte found in the circulation?
T cells, which make up approximately 80% of circulating lymphocytes

2010-338
T lymphocytes mediate the _____ (cellular/humoral) immune response.
Cellular

2010-338
Which major histocompatibility complex and CD molecules are expressed by cytotoxic T cells?
Major histocompatibility complex I and CD8 (MHC × CD = 8; MHC I × CD8 = 8)

2010-338
Which major histocompatibility complex and CD molecules are expressed by helper T cells?
Major histocompatibility complex II and CD4 (MHC × CD = 8; MHC II × CD4 = 8)

2010-338
Name the three types of mature cells into which T cells differentiate.
Cytotoxic T cells, helper T cells, and suppressor T cells

2010-338
What is the blood group of an individual with both A and B antigens on red blood cells?
Group AB (universal recipient)

2010-339
What is the blood group of an individual with the B antigen on red blood cells and A antibody in plasma?
Group B

2010-339
What is the blood type of an individual who is a universal recipient of blood cells?
Blood group AB; A and B antigen on red blood cells

2010-339
Which class of antibodies are produced against Rh factor?
Immunoglobulin G

2010-339
A patient receiving a blood transfusion suddenly develops symptoms of anaphylaxis, renal failure, and hypotension and has schistocytes on a smear; what is the likely cause?
Incompatible blood type transfusion

2010-339
If a mother is blood type O and her fetus is blood type B, will the fetus be at risk for hemolytic disease of the newborn?
No; antibodies against ABO blood groups are immunoglobulin M class and do not cross the placenta

2010-339
What happens when an Rh-negative mother with anti-Rh antibodies becomes pregnant with a child who is Rh positive?
Erythoblastosis fetalis; the anti-Rh immunoglobulin G crosses the placenta and causes hemolysis in the newborn

2010-339
What is the blood type of an individual who is a universal donor of blood products?
Blood group O; neither A nor B antigens on red blood cells

2010-339
What is the blood group of an individual with neither A nor B antigens on red blood cells and both A and B antibodies in plasma?
Group O (universal donor)

2010-339
Which class of antibodies are produced against the ABO blood groups?
Immunoglobulin M

2010-339
What is the blood group of an individual with the A antigen on red blood cells and B antibody in plasma?
Group A

2010-339
What happens when an Rh-negative mother is exposed to Rh-positive blood (for instance, during delivery)?
Exposure to Rh-positive blood (which has the Rho antigen) will result in the development of anti-Rh immunoglobulin G antibodies, which may complicate future pregnancies

2010-339
Why do susceptible individuals develop angioedema when taking an angiotensin-converting enzyme inhibitor?
Angiotensin-converting enzyme deactivates bradykinin; blocking this results in the unopposed activation of bradykinin by kallikrein

2010-339
What reaction accounts for the effects of heparin?
Heparin activates antithrombin, resulting in inhibition of thrombin and factors IXa, Xa, XIa, and XIIa

2010-339
What are two stimuli for the conversion of factor X to factor Xa?
Activated factor VIIa, or factor IXa with factor VIIIa as a cofactor; this is where the intrinsic and extrinsic pathways converge

2010-339
Kallikrein links the coagulation cascade to the kinin cascade through its activation of _____ and to the fibrinolytic system through its activation of _____.
Bradykinin; plasminogen (to plasmin)

2010-339
Which coagulation factors are specific to the extrinsic coagulation pathway?
Factor VII and tissue factor (thromboplastin)

2010-339
Once high-molecular-weight kininogen cleaves to form bradykinin, what are the effects of the kinin cascade?
Vasodilation, vascular permeability, and pain

2010-339
Which four reactions is thrombin involved in?
Thrombin (IIa) cleaves fibrinogen into fibrin monomers, activates factor XIII to help stabilize and cross-link the fibrin mesh, and activates both factor VIII and factor V

2010-339
What ion does factor XIII require to stabilize the fibrin mesh?
Calcium

2010-339
Which coagulation factors are specific to the intrinsic coagulation pathway?
Factor XII, factor XI, factor IX, and factor VIII

2010-339
Name two reactions initiated by high-molecular-weight kininogen.
High-molecular-weight kininogen is the initial reagent in the kinin cascade and stimulates the coagulation cascade by interacting with factor XII

2010-339
What factor converts prokallikrein to kallikrein and factor XI to XIa?
Factor XIIa; it is thus involved in both the kinin cascade and the coagulation cascade

2010-339
Which reactions in the coagulation cascade require calcium and a phospholipid surface as cofactors?
The conversion of factor IX to IXa, VII to VIIa, X to Xa, and II to IIa

2010-339
What is the end result of the coagulation cascade?
The conversion of fibrinogen into fibrin to form a fibrin mesh that will stabilize the platelet plug

2010-339
What disorder results from a deficiency of factor IX?
Hemophilia B

2010-339
What does plasmin create when it interacts with fibrin clots?
Fibrin split products (fibrin degradation)

2010-339
Which coagulation factors are common to both the intrinsic and extrinsic coagulation pathways?
Factor X, factor V, factor II (prothrombin), and factor XIII

2010-339
What part of the fibrinolytic system also activates the complement cascade by cleaving C3 to C3a?
Plasmin

2010-339
What disorder results from a deficiency of factor VIII?
Hemophilia A

2010-339
Name two initiators of the intrinsic coagulation pathway.
Factor XII is converted to XIIa in the presence of endothelial damage (collagen basement membrane and activated platelets) and by high-molecular-weight kininogen

2010-339
A patient is severely deficient in vitamin K; which coagulation factors will be affected?
This will result in decreased levels of factors II, VII, IX, X, and proteins C and S

2010-339
Via which molecule does von Willebrand's factor attach to platelets?
von Willebrand's factor binds to glycoprotein Ib, found on the platelet surface membrane

2010-340
Which contributors to thrombogenesis are found within platelets?
von Willebrand's factor, fibrinogen, and, upon cyclooxygenase activation of the arachidonic acid cascade, thromboxane A2

2010-340
What event causes expression of glycoprotein IIa/IIIb on the platelet membrane?
Upon adenosine diphosphate activation of the platelet, glycoprotein IIa/IIIb is inserted into the surface membrane

2010-340
Name four mediators of thrombus formation or degradation that are found within the vascular endothelial cell.
von Willebrand's factor, thromboplastin, tissue plasminogen activator, and prostaglandin I2

2010-340
Which molecule, found on the surface of vascular endothelial cells, is a cofactor in the activation of protein C, an anticoagulant?
Thrombomodulin, which is located on the surface of vascular endothelial cells

2010-340
How do platelets adhere to one another?
Glycoprotein IIb/IIIa binds fibrinogen, bridging platelets together

2010-340
Which molecule binds the subendothelial collagen that is exposed after endothelial damage?
von Willebrand's factor binds to the subendothelial collagen and attaches to activated platelets

2010-340
What is the difference between the function of glycoprotein Ib and glycoprotein IIb/IIIa on the platelet surface?
Glycoprotein Ib binds von Willebrand's factor on a damaged endothelial surface, whereas glycoprotein IIa/IIIb binds fibrinogen attached to other platelets

2010-340
What is the role of protein S in anticoagulation?
Protein S is a cofactor for protein C activation, which inactivates factors Va and VIIIa

2010-341
How does aspirin prevent clot formation?
Aspirin inhibits cyclooxygenase in the platelets, thereby preventing TxA2 formation and thus aggregation of platelets

2010-341
Which medication directly interferes with glycoprotein IIa/IIIb?
Abciximab

2010-341
What is the relationship between von Willebrand's factor and factor VIII?
von Willebrand's factor prolongs the half-life of factor VIII, which is why there is an increase in partial thromboplastin time in severe variants of von Willebrand's disease

2010-341
Which ion helps to strengthen the platelet plug once it has formed?
Calcium

2010-341
Which product of the arachidonic acid pathway favors aggregation of platelets? Which molecules inhibit aggregation?
Thromboxane, which is released by platelets, is proaggregation; prostacyclin and nitric oxide, released by endothelial cells, are antiaggregation

2010-341
Why do neonates receive an injection of vitamin K soon after birth?
Neonates lack enteric bacteria that produce vitamin K and can therefore be deficient in the vitamin K-dependent factors and suffer hemorrhagic disease

2010-341
Name three endogenous factors that initiate anticoagulation.
Antithrombin III, protein C, plasminogen

2010-341
What enzyme is involved in vitamin K activation?
Epoxide reductase

2010-341
How does plasminogen act as an anticoagulant?
Plasminogen is activated by tissue plasminogen activator to plasmin, which cleaves the fibrin clot

2010-341
What process occurs during the adhesion stage of platelet plug formation?
The platelet adheres to exposed subendothelial collagen via glycoprotein Ib and von Willebrand's factor

2010-341
What is the result of the factor V Leiden mutation?
The mutation results in a factor V that resists inactivation by activated protein C, leading to a hypercoagulable state

2010-341
What occurs during the swelling stage of platelet plug formation?
The platelet plug "swells" as many platelets bind together secondary to adenosine diphosphate activation of glycoprotein IIa/IIIb bridge formation

2010-341
How does warfarin act as an anticoagulant?
It inhibits epoxide reductase, thereby inhibiting the activation of vitamin K

2010-341
What is tissue plasminogen activator used for clinically?
As a thrombolytic in cases of acute stroke or myocardial infarction within hours of clot formation; tissue plasminogen activator activates plasminogen to plasmin

2010-341
Which stage of platelet plug formation is affected by ticlopidine and clopidogrel?
Swelling; these drugs inhibit the adenosine diphosphate-induced expression of glycoprotein IIa/IIIb

2010-341
What type of red blood cell morphology would you see in the peripheral blood that is associated with Heinz bodies?
Bite cells; the spleen "bites" off the precipitated hemoglobin in the cells

2010-343
Name two diseases associated with Heinz bodies.
α-Thalassemia and glucose-6-phosphate dehydrogenase deficiency

2010-343
What is the treatment for sideroblastic anemia due to a mutation in δ-ALA synthase?
Pyridoxine (vitamin B6)

2010-343
What symptoms and laboratory findings are consistent with β-thalassemia minor?
This condition is asymptomatic, and diagnosed by an hemoglobin A2 > 3.5% on electrophoresis

2010-343
What x-ray finding should raise your suspicion of β-thalassemia major?
Bone marrow expansion, which can be seen as increased marrow space, bone deformities ("chipmunk facies"), and a "crew-cut" appearance of the skull x-ray

2010-343
What is the role of the mean corpuscular volume in an anemia workup?
A mean corpuscular volume < 80 fL is a microcytic anemia; a volume > 100 fL is a macrocytic anemia; a volume between those values is a normocytic anemia

2010-343
What would you see on hemoglobin electrophoresis in a fetus with deletions of all four α globin genes?
Hemoglobin Barts (γ4); this is a lethal condition and will result in hydrops fetalis

2010-343
What biochemical pathway is interrupted in iron deficiency anemia?
The formation of heme (which requires iron)

2010-343
You see a patient with tongue inflammation and atrophy, dysphagia for solids, and a hemoglobin of 9 g/dL; what is the cause of his anemia?
This patient has Plummer-Vinson syndrome, the triad of esophageal webs, atrophic glossitis, and iron deficiency anemia

2010-343
What is the inheritance pattern associated with a mutation in the δ-aminolevulinic acid gene?
This mutation, which causes a hereditary sideroblastic anemia, is found on the X chromosome

2010-343
What disease do you expect to see in a child born to a father homozygous for hemoglobin S and a mother with β-thalassemia major?
This child will be a hemoglobin S/β-thalassemia heterozygote, which presents with mild or moderate sickle cell disease based on β chain production

2010-343
What condition would you expect to see in a patient with a deletion of three of the four α globin genes?
Symptomatic microcytic anemia due to α-thalassemia; known as hemoglobin H disease (β4)

2010-343
Lead poisoning disrupts which two enzymes involved in the formation of hemoglobin?
Lead blocks ferrochelatase and ALA dehydratase, which decreases the synthesis of heme

2010-343
What are three etiologies of iron-deficiency anemia?
Loss (chronic bleeding), malnutrition or poor absorption, and increased demand (such as in pregnancy)

2010-343
What is the cause of hereditary sideroblastic anemia?
A mutation in the δ-aminolevulinic acid gene

2010-343
What type of mutation is present in a patient with β-thalassemia minor?
There is a mutation in one gene (heterozygote), causing underproduction of β globin; the mutation is often in a promoter or splice site

2010-343
What should you see on hemoglobin electrophoresis in a patient with β-thalassemia (major and minor) that, as in infants, increases the blood's affinity for oxygen?
Increased fetal hemoglobin (α2; γ2)

2010-343
A patient has an elevated serum iron and ferritin, and a normal total iron-binding capacity; what diagnosis should you suspect if a bone marrow aspirate shows blast forms of erythrocytes that have a dark ring around the nucleus?
Sideroblastic anemia; the aspirate describes the iron-laden mitochondria that create the appearance of a ringed sideroblast

2010-343
What is a long-term adverse effect of the treatment of β-thalassemia major?
Iron overload (secondary hemochromotosis) due to repeated blood transfusions

2010-343
What disease, seen in people that live in the Mediterranean region, is caused by point mutations in hemoglobin β chain genes?
β-Thalassemia; typically these mutations are at splicing sites and in the promoter regions

2010-343
A fetus undergoes amniocentesis and is found to have deletions of all four α globin genes; what is the likely outcome?
Hydrops fetalis; deletion of four genes is incompatible with life

2010-343
A mother is concerned that her child has deletions of two α globin genes; what symptoms will likely manifest?
Deletion of one or two α globin genes is not associated with symptomatic anemia

2010-343
What disease, common in Asia and Africa, is caused by deletions of the hemoglobin α chain gene?
α-thalassemia; gene deletions result in decreased α globin synthesis

2010-343
Name two reversible causes of sideroblastic anemia
Alcohol use and lead poisoning

2010-343
What is the cause of basophilic stippling associated with lead poisoning?
Lead disrupts the degradation of RNA, which builds up and stains with basic dyes

2010-343
What treatment is indicated in a patient with β-thalassemia major?
Because both genes are mutated, there is no β chain production and the patient requires many blood transfusions

2010-343
What kind of anemia is associated with congenital deficiencies in purine or pyrimidine synthesis?
Macrocytic anemia (eg, orotic aciduria) because of the relative delay in nucleus maturation compared to cytoplasm

2010-344
Name three ways that heavy alcohol use can cause macrocytic anemia.
As a result of vitamin B12 or folate deficiency (malnutrition), liver disease, or by directly affecting red blood cells and causing bone marrow suppression

2010-344
Name two conditions in which an increased demand for folate can result in a macrocytic anemia.
Pregnancy (folate is needed for fetal neural tube development) or hemolytic anemias (folate is needed to replenish hemolyzed blood)

2010-344
A patient has anemia with a mean corpuscular volume of 110 fL and glossitis and on the peripheral smear you note hypersegmented neutrophils; what are the two possible causes of this condition?
B12 or folate deficiency; both present with these features

2010-344
Why might a patient with Crohn's disease have a macrocytic anemia?
Crohn's disease often affects the terminal ileum, the site of vitamin B12 absorption; severe or extensive disease may cause malabsorption of folate as well

2010-344
What kind of anemia is seen in a patient with poor dietary vitamin intake?
Macrocytic anemia, due to B12 or folate deficiency (remember that alcoholics are also often malnourished)

2010-344
What change in mean corpuscular volume would you expect in an otherwise healthy patient who has had moderate to severe blood loss?
Increased mean corpuscular volume, because the patient will compensate with reticulocytosis, which have slightly higher mean corpuscular volumes than mature red blood cells

2010-344
Why is the mean corpuscular volume increased in megaloblastic macrocytic anemia?
There is a defect in DNA synthesis: the nucleus takes longer to mature than the cytosol and thus there is excess cytoplasm, creating a megaloblast; this is caused by B12 and folate deficiency

2010-344
What is the homocysteine level in patients with B12 and folate deficiency?
Both B12 and folate deficiencies have elevated homocysteine levels

2010-344
Name two drugs than can cause a folate deficiency and macrocytic anemia.
Methotrexate or trimethoprim, both of which interfere with folate metabolism

2010-344
What autoimmune disorder can result in a macrocytic anemia?
Pernicious anemia, which results in vitamin B12 deficiency and loss of intrinsic factor

2010-344
Name five medications that can cause macrocytic anemia.
Methotrexate, trimethoprim, fluorouracil, zidovudine, hydroxyurea

2010-344
Which parasite can cause vitamin B12 deficiency?
Diphyllobothrium latum, a tapeworm found in fish

2010-344
What laboratory test (other than a vitamin level) will distinguish between B12 and folate deficiency as a cause of megaloblastic anemia?
Methylmalonic acid is elevated in B12 deficiency but normal in folate deficiency; homocysteine is elevated in both deficiencies

2010-344
What symptoms can distinguish macrocytic anemia caused by vitamin B12 deficiency and macrocytic anemia caused by folate deficiency?
Neurologic symptoms (combined subacute degeneration) are seen in vitamin B12 deficiency but not in folate deficiency because of the role B12 plays in fatty acid pathways

2010-344
Name three causes of normocytic, normochromic anemia that are not due to hemolysis.
Anemia of chronic disease, aplastic anemia (bone marrow failure), renal disease (decreased erythropoietin)

2010-344
Generally, hemolysis that is intrinsic to the red blood cell occurs in the _____ (intravascular/extravascular) space, whereas hemolysis extrinsic to the red blood cell occurs in the _____ (intravascular/extravascular) space.
Extravascular; intravascular. Notable exceptions include paroxysmal nocturnal hemoglobinuria (intrinsic intravascular) and warm agglutinin autoimmune or microangiopathic hemolysis (extrinsic extravascular)

2010-344
Name six causes of normocytic, normochromic hemolytic anemia that are intrinsic to the red blood cell.
Hereditary spherocytosis, glucose-6-phosphate dehydrogenase deficiency, pyruvate kinase deficiency, sickle cell anemia, hemoglobin C defect, and paroxysmal nocturnal hemoglobinuria

2010-344
What is a cause of normocytic, normochromic hemolytic anemia that is intrinsic to the red blood cell and occurs in the intravascular space?
Paroxysmal nocturnal hemoglobinuria

2010-344
Name three causes of normocytic, normochromic hemolytic anemia that are extrinsic to the red blood cell.
Autoimmune hemolysis, microangiopathies like disseminated intravascular coagulation or thrombotic thrombocytopenic purpura–hemolytic uremic syndrome, or infections

2010-344
Name two causes of normocytic, normochromic hemolytic anemia that are extrinsic to the red blood cell and occur in the extravascular space.
Autoimmune hemolysis due to warm agglutinins (not cold) or microangiopathies like disseminated intravascular coagulation or thrombotic thrombocytopenic purpura–hemolytic uremic syndrome

2010-344
How are normocytic, normochromic hemolytic anemias further classified?
First by cause: intrinsic vs extrinsic to the red blood cell; and then by location: intravascular vs extravascular

2010-344
What are two possible explanations for aplastic anemia that is considered idiopathic?
There may be an autoimmune etiology or a primary defect in the stem cell itself

2010-345
You suspect that a patient has aplastic anemia due to an autoimmune process; what treatment can you pursue?
Immune suppression using antithymocyte globulin or cyclosporin

2010-345
What are four viral infections that can cause aplastic anemia?
Parvovirus B19 (especially in patients with sickle cell anemia), HIV, Epstein-Barr virus, and viral hepatitis

2010-345
Name three treatments for aplastic anemia that can be tried when other interventions have failed.
Red blood cell and platelet transfusion, dosing with granulocyte-macrophage colony-stimulating factor or granulocyte colony-stimulating factor, or allogeneic bone marrow transplantation

2010-345
How do chronic inflammatory processes cause anemia?
The inflammation increases hepcidin levels, which cause macrophages to retain iron, preventing heme from forming

2010-345
A patient presents with petechiae and bleeding from the gums, fatigue and pallor, and has had several recent infections; a bone marrow aspirate is "dry" making you suspect what process?
Aplastic anemia; this patient has symptoms suggestive of thrombocytopenia, anemia, and neutropenia, which are confirmed by the hypocellular aspirate

2010-345
Why do patients with end-stage renal disease have normocytic, normochromic anemia?
The kidneys normally produce erythropoietin; in renal failure the loss of erythropoietin results in anemia, which is amenable to treatment with exogenous erythropoietin

2010-345
A patient with pancytopenia (anemia, neutropenia, and thrombocytopenia) has fatty infiltrates on bone marrow biopsy; what is the diagnosis?
Aplastic anemia; the marrow is depleted, but the cell morphology will be normal

2010-345
Name four drugs that can result in aplastic anemia.
Benzene, chloramphenicol (eg, "gray baby syndrome"), antimetabolites, and alkylating agents (chemotherapy)

2010-345
What are four general etiologies of aplastic anemia?
Radiation or drugs, viral infections, Fanconi's anemia, or idiopathic aplastic anemia

2010-345
What are the iron, ferritin, and total iron-binding capacity levels in a patient with anemia of chronic disease?
Decreased iron and total iron-binding capacity, with increased ferritin

2010-345
What mechanism of aplastic anemia is seen in patients with Fanconi's anemia?
These patients have an inherited defect in DNA repair

2010-345
What is the inheritance pattern of pyruvate kinase deficiency?
Autosomal recessive

2010-346
What molecule is depleted in the red blood cells of patients that have glucose-6-phosphate dehydrogenase deficiency, leading to anemia?
Glutathione; without glucose-6-phosphate dehydrogenase, there is a depletion of glutathione, which results in red blood cell susceptibility to oxidative stress

2010-346
What organ is responsible for the anemia seen in hereditary spherocytosis?
The spleen; the small, inflexible membrane causes the spleen to remove affected red blood cells prematurely

2010-346
What happens to the hemoglobin in patients with sickle cell disease in the presence of low oxygen concentrations?
Without oxygen, the hemoglobin has an exposed charge that causes precipitation and formation of long polymers of hemoglobin that give the characteristic sickle shape to the red blood cell

2010-346
Name six complications seen in homozygotes for sickle cell disease.
Aplastic crisis (parvovirus infection), autosplenectomy, Salmonella osteomyelitis, painful vaso-occlusive crises, renal papillary necrosis, and splenic sequestration crisis

2010-346
What mutations are present in hereditary spherocytosis?
Ankyrin, band 3.1, or spectrin mutations; each normally interacts with the red blood cell skeleton and plasma membrane to preserve normal morphology

2010-346
Why does the hemolysis seen in paroxysmal nocturnal hemoglobinuria occur in the intravascular space?
There is complement-mediated hemolysis, an intravascular process due to decreased levels of decay accelerating factor; decay accelerating factor normally prevents the membrane attack complex from destroying red blood cells

2010-346
What is the significance of the hemoglobin C mutation?
Patients with hemoglobin S and C mutations have milder sickle cell disease than homozygotes for hemoglobin S

2010-346
You see an African-American patient who has crescent-shaped cells on the peripheral smear; what is the cell and what is the prevalence of this disease?
This is a sickle cell; the trait (heterozygotes) occurs in 8% of African-Americans, whereas 0.2% have sickle cell disease (homozygotes)

2010-346
What x-ray finding in sickle cell anemia may also be seen in thalassemia?
The "crew cut" skull radiograph, due to bone marrow expansion, is seen in both conditions

2010-346
What kind of anemia is seen in patients with hemoglobin C disease?
Intrinsic normocytic hemolytic anemia; this condition is due to a mutation in the β globin chain (different from hemoglobin S in sickle cell disease)

2010-346
What treatment options are available for patients with sickle cell disease?
Patients can be treated with hydroxyurea, which increases hemoglobin F concentration, or may undergo allogeneic bone marrow transplantation

2010-346
Why will a newborn with sickle cell disease initially be asymptomatic?
The newborn still has increased levels of hemoglobin F and will be asymptomatic until his hemoglobin F is replaced by hemoglobin S

2010-346
What is the inheritance pattern seen in glucose-6-phosphate dehydrogenase deficiency?
X-linked recessive; therefore, symptoms are usually seen in males

2010-346
What evolutionary advantage may explain the high prevalence of sickle cell trait in the population?
This mutation allows resistance to malaria, an intracellular parasite that infects erythrocytes

2010-346
What acute process should you suspect in a patient with red blood cells that have no central pallor, increased mean corpuscular hemoglobin concentration, and increased red blood cell distribution width if he contracts a parvovirus B19 infection?
Aplastic crisis; this patient likely has hereditary spherocytosis and is predisposed to aplastic anemia during parvovirus infection due to his increased reliance on erythropoiesis

2010-346
What lab test should you order to confirm the diagnosis in a patient who has spherocytes on the peripheral smear and splenomegaly on exam?
Osmotic fragility test; in hereditary spherocytosis the red blood cells will lyse more easily because of decreased membrane flexibility

2010-346
You see a patient with hereditary spherocytosis but no splenomegaly on exam; if he had Howell-Jolly bodies on peripheral smear what is the reason for your exam and lab findings?
This patient has likely had a splenectomy; this treatment can be used if the anemia in hereditary spherocytosis is severe and refractory to treatment

2010-346
A newborn is born with hemolytic anemia and you determine that there is no blood group or Rh antigen difference between the baby and mother; what congenital error in metabolism may be the cause?
Pyruvate kinase deficiency, which causes low adenosine triphosphate in red blood cells and a resultant rigidity of the membranes

2010-346
A patient develops anemia, indirect hyperbilirubinemia, and jaundice after eating fava beans; what condition should you suspect?
Glucose-6-phosphate dehydrogenase deficiency; fava beans, along with sulfa drugs and infections, are well-documented causes of oxidant stress

2010-346
What is the diagnosis in a patient who has Heinz bodies on the peripheral smear and multiple "bite cells"?
Glucose-6-phosphate dehydrogenase deficiency

2010-346
What is the mutation that causes sickle cell disease?
Glutamic acid to valine in the β globin chain

2010-346
What laboratory test would be abnormal in a patient with paroxysmal nocturnal hemoglobinuria?
Increased urine hemosiderin; the hemolysis occurs all day, but urine accumulates overnight and thus appears darker the next day

2010-346
What vaccinations should be given to patients with sickle cell disease?
Vaccinations to encapsulated organisms because of the increased risk of sepsis after autosplenectomy: Streptococcus pneumoniae, Haemophilus influenzae, and Neisseria meningitidis

2010-346
What type of antibody is involved in autoimmune hemolytic anemia due to warm agglutinins?
Immunoglobulin G (remember: Warm weather is GGGreat)

2010-347
What are two infections that can present with hemolytic anemia due to cold agglutinins?
Mycoplasma pneumoniae infection or infectious mononucleosis

2010-347
What type of hemolytic process causes erythroblastosis fetalis?
Warm agglutinin-mediated immune reaction; remember that this is caused by maternal immunoglobulin G antibodies against the fetal Rh group

2010-347
You note schistocytes or "helmet cells" on the peripheral smear of a patient; what mechanism causes this deformity?
This red blood cell morphology is seen in microangiopathic hemolytic anemias, thought to be due to damage as red blood cells pass through a narrow or obstructed vessel lumen

2010-347
Name two infections that cause hemolysis but are not associated with agglutinins.
Intracellular pathogens, like malaria and babesia, can lyse the erythrocyte and cause hemolytic anemia

2010-347
What does a positive direct Coombs' test represent?
A direct Coombs' test adds anti-immunoglobulin G antibodies to the patient's red blood cells; if agglutination occurs, it indicates that the patient's red blood cells are covered with immunoglobulin G antibodies

2010-347
What are two cardiac pathologies that can cause hemolytic anemia?
Severe aortic stenosis, or any mechanical valve, due to the shearing and mechanical destruction of red blood cells

2010-347
What are four conditions that may be the cause of "helmet cells" or schistocytes on a patient's peripheral blood smear?
Disseminated intravascular coagulation, thrombotic thrombocytopenic purpura–hemolytic uremic syndrome, systemic lupus erythematous, or malignant hypertension

2010-347
What does a positive indirect Coombs' test represent?
An indirect Coombs' test adds normal red blood cells to the patient's serum; if agglutination occurs, it indicates that the patient has anti-red blood cell antibodies in serum

2010-347
Name three conditions that are associated with immunoglobulin G-mediated autoimmune hemolysis.
Systemic lupus erythematous, chronic lymphocytic leukemia, and drug reactions (for instance, α-methyldopa)

2010-347
What type of antibody is involved in autoimmune hemolytic anemia due to cold agglutinins?
Immunoglobulin M (remember: Cold ice cream is MMM); cold agglutinins trigger anemia when exposed to cold temperatures

2010-347
What is the relationship between transferrin and total iron-binding capacity?
Total iron-binding capacity is an indirect measure of transferrin, which transports iron in the blood

2010-347
What are the relevant lab values for a patient with hemochromatosis?
The serum iron and ferritin are high and the total iron-binding capacity is low; the percent transferrin saturation is very high

2010-347
What are the relevant lab values in a patient with iron-deficiency anemia?
The serum iron and ferritin are low, whereas the total iron-binding capacity is elevated; the percent transferrin saturation is very low

2010-347
What changes are seen in serum iron studies for a patient who is either pregnant or using oral contraceptive pills?
The serum iron and ferritin are normal; the total iron-binding capacity is elevated due to estrogen exposure (increased binding proteins); the percent transferrin saturation is low

2010-347
What is the role of ferritin in the body?
It is the primary iron storage protein of the body

2010-347
What are the relevant lab values in a patient with anemia of chronic disease?
The serum iron and total iron-binding capacity are low, whereas the ferritin is elevated; the percent transferrin saturation is normal

2010-347
What is the evolutionary theory behind the etiology of anemia of chronic disease?
Pathogens need iron to thrive; sequestration of iron in macrophages may prevent pathogens from reproducing

2010-347
What cofactor is necessary for the rate-limiting step in heme synthesis?
Vitamin B6, which is needed to convert glycine and succinyl-Co-A to aminolevulinic acid

2010-348
What substrate of heme synthesis accumulates in the blood in lead poisoning?
Protoporphyrin

2010-348
What environmental exposures are common sources of lead poisoning in adults? In children?
Adults are exposed to lead in the workplace, such as a battery/ammunition/radiator factories, whereas children are exposed via lead paint

2010-348
What enzymatic defect is present in acute intermittent porphyria?
Porphobilinogen deaminase (also called uroporphyrinogen-I-synthase)

2010-348
Lead poisoning affects which two enzymes in the heme synthesis pathway?
Ferrochelatase and ALA dehydratase are inhibited by lead

2010-348
How does heme provide negative feedback for its own synthesis?
Heme inhibits ALA dehydratase, the second step in heme synthesis

2010-348
A child presents with a microcytic anemia, gastrointestinal distress, and renal disease; she has developmental regression and basophilic stippling on red blood cells; what is the diagnosis?
These are the symptoms of lead poisoning, commonly seen when children are exposed to lead paint

2010-348
What treatment is available for patients with acute intermittent porphyria?
Glucose or heme, both of which inhibit ALA synthase to decrease buildup of heme precursors

2010-348
What are the differences in neurologic outcomes between adults and children with lead poisoning?
Adults typically have headaches, memory loss, and demyelination, whereas children typically have developmental delays

2010-348
A patient who recently started phenobarbital presents after a day at the beach where she had multiple alcoholic drinks with abdominal pain, polyneuropathy, an altered mental status. Her urine turns pink on standing. What is the diagnosis?
Acute intermittent porphyria (remember the five P's: Painful abdomen, Pink urine, Polyneuropathy, Psychological disturbances, Precipitated by drugs [eg, phenobarbital or ethanol])

2010-348
A patient presents with blisters upon exposure to sunlight and you suspect a porphyria; what is the most likely cause?
Porphyria cutanea tarda, the most common porphyria

2010-348
Which step in heme synthesis requires iron?
The final step, where protoporphyrin is converted to heme by ferrochelatase

2010-348
In porphyria cutanea tarda, there is a defect in the enzyme _____, resulting in an accumulation of _____.
Uroporphyrinogen decarboxylase; uroporphyrin, causing "tea-colored" urine

2010-348
What enzyme is affected in patients with X-linked sideroblastic anemia?
δ-ALA synthase, which is responsible for the rate-limiting step of heme synthesis; conversion of glycine and succinyl-Co-A to aminolevulinic acid

2010-348
What pathologic process occurs if there is a defect in one of the steps of heme synthesis?
Porphyria, in which heme precursors accumulate and enter various bodily fluids

2010-348
What substances accumulate in acute intermittent porphyria?
Porphobilinogen, δ-ALA, and uroporphyrin (in the urine)

2010-348
What condition should be suspected in a patient who has bluish lines on his gums and thick white lines on long bone epiphyses on x-ray?
Lead poisoning; the blue lines around teeth are Burton's lines, and the white lines near the epiphysis are lead lines

2010-349
Name two hematologic manifestations of lead poisoning.
Basophilic stippling on erythrocytes and sideroblastic anemia

2010-349
What treatment is indicated in a child who has lead poisoning?
Succimer (remember: it sucks to be a kid who eats lead)

2010-349
Name three neurologic manifestations of lead poisoning.
Encephalopathy, abdominal pain, and wrist and foot drop

2010-349
What are the main treatments for lead poisoning in adults?
Chelation therapy; dimercaprol or EDTA are first-line treatments

2010-349
What is the role that von Willebrand's multimers play in the development of thrombotic thrombocytopenic purpura?
The large von Willebrand's multimers cause increased platelet aggregation (bind glycoprotein Ib) and thrombosis

2010-349
Glanzmann's thrombasthenia causes a defect in what platelet interaction?
Poor platelet-to-platelet aggregation in the platelet plug due to decreased glycoprotein IIb/IIIa

2010-349
Which platelet disorder has an increased bleeding time but normal platelet count?
Glanzmann's thrombasthenia; all have an increased bleeding time, whereas Bernard-Soulier disease, immune thrombocytopenic purpura, and thrombotic thrombocytopenic purpura have decreased platelet counts

2010-349
Bernard-Soulier disease causes a defect in the platelet's interaction with what molecule?
Endothelial collagen; there is a decrease in the molecule glycoprotein Ib for platelet-to-collagen adhesion

2010-349
A patient has bleeding from the gums, epistaxis, petechiae, and purpura; what is the likely defect in coagulation?
These describe the microhemorrhages associated with a platelet disorder; there may or may not be a decreased number of platelets

2010-349
A patient develops renal failure, confusion, thrombocytopenia, fever, and schistocytes on peripheral smear; what is the diagnosis?
This is the classic pentad of thrombotic thrombocytopenic purpura

2010-349
What enzyme is deficient in patients with thrombotic thrombocytopenic purpura?
ADAMTS-13, a metalloproteinase that degrades von Willebrand's multimers

2010-349
What is the antibody target in immune thrombocytopenic purpura?
Immune thrombocytopenic purpura is caused by antibodies to glycoprotein IIb/IIIa causing platelet destruction

2010-349
What coagulation test will indicate there is a problem with platelet function?
Bleeding time will be increased; there will be no change in prothrombin time or partial thromboplastin time

2010-349
A patient presents to the emergency room with a large swollen joint tapped and found to be full of blood. He has a history of easy bruising. What condition should you consider in your diagnosis?
Spontaneous hemarthrosis, which is a macrohemorrhage seen in hemophilia (compared to petechiae or purpura in platelet disorders)

2010-349
What are the prothrombin time and partial thromboplastin time results in a patient with hemophilia (A or B)?
The prothrombin time will be normal and the partial thromboplastin time will be elevated because hemophilia affects the intrinsic pathway

2010-349
What coagulation factors are evaluated when you order a prothrombin time test?
Factors I, II, V, VII, and X; the extrinsic pathway

2010-349
What coagulation factors are evaluated when you order a partial thromboplastin time test?
All coagulation factors except VII and XIII; the intrinsic pathway

2010-349
What are the prothrombin time and partial thromboplastin time changes in a patient with vitamin K deficiency?
Vitamin K deficiency causes both prothrombin time and partial thromboplastin time elevations

2010-349
Hemophilia A is caused by a deficiency of _____, whereas hemophilia B (Christmas disease) is caused by a deficiency of _____.
Factor VIII; factor IX

2010-349
What is the most common inherited bleeding disorder?
von Willebrand's disease, an autosomal dominant disease that can have varying severity (typically mild)

2010-350
Name seven causes of disseminated intravascular coagulation.
Gram-negative Sepsis, Trauma, Obstetric complications, acute Pancreatitis, Malignancy, Nephrotic syndrome (loss of ATIII), or Transfusion (remember: STOP Making New Thrombi)

2010-350
Why can you see an elevated partial thromboplastin time in a patient with von Willebrand's disease?
von Willebrand's factor carries and protects factor VIII; deficiencies in von Willebrand's factor can cause intrinsic pathway coagulopathies related to factor VIII deficiency

2010-350
What are the platelet count, bleeding time, prothrombin time, and partial thromboplastin time results of a patient in disseminated intravascular coagulation?
Widespread activation of all coagulation pathways results in global depletion: a low platelet count and an increased bleeding time, prothrombin time, and partial thromboplastin time

2010-350
What are the platelet count, bleeding time, prothrombin time, and partial thromboplastin time in a patient with von Willebrand's disease?
There is an elevated bleeding time due to a defect in platelet adhesion; in severe von Willebrand's deficiency, there is also an increase in partial thromboplastin time due to decreased half-life of factor VIII

2010-350
A patient presents with petechiae and gastrointestinal bleeding; initial labs show schistocytes, elevated fibrin split products, and decreased fibrinogen, factor V, and factor VIII. What condition should you be concerned about?
Disseminated intravascular coagulation

2010-350
Why does protein C or S deficiency lead to a hypercoaguable state?
There is an inability to inactivate factors V and VIII

2010-350
What is the risk associated with administering warfarin in the setting of protein C or S deficiency?
Hemorrhagic skin necrosis

2010-350
What is the pathogenesis of factor V Leiden?
There is a mutation in factor V, rendering it unable to be degraded by protein C, leading to a hypercoaguable state

2010-350
How would a patient with antithrombin III deficiency react to administration of heparin?
The patient would not have the expected increase in partial thromboplastin time after heparin administration because heparin's mechanism of action is dependent on AT III

2010-350
What is the difference in tumor location between leukemia and lymphoma?
In leukemia, tumor cells are typically found in the blood and bone marrow, whereas in lymphoma the tumor cells are lymphoid masses (large lymph nodes)

2010-350
What demographic group is most affected by Hodgkin's lymphoma?
Hodgkin's lymphoma is more common among men and is bimodal, seen in the young and elderly

2010-351
Which virus is implicated in Hodgkin's lymphoma? Non-Hodgkin's lymphoma?
Epstein-Barr virus is associated with 50% of cases of Hodgkin's lymphoma; HIV and other immunosuppressed states are risk factors for non-Hodgkin's lymphoma

2010-351
Typically, non-Hodgkin's lymphoma is a tumor derived from what type of cell?
B lymphocytes, except for the few that have lymphoblastic T-cell origin

2010-351
What findings on biopsy portend a good prognosis in a patient with Hodgkin's lymphoma?
A low number of Reed-Sternberg cells and an increased number of lymphocytes

2010-351
What malignancy is associated with Reed-Sternberg cells (cells with a distinctive bilobed nucleus that looks like "owl's eyes")?
Hodgkin's lymphoma; these cells are necessary but not sufficient for the diagnosis

2010-351
A patient has night sweats, fevers, weight loss, and new palpable lymph nodes. Which type of lymphoma is more likely: Hodgkin's or non-Hodgkin's?
Hodgkin's lymphoma; these are "B" symptoms, more common in Hodgkin's

2010-351
What pattern of lymph node involvement is seen in Hodgkin's lymphoma? How does this differ from non-Hodgkin's lymphoma?
Hodgkin's lymphoma tends to be localized (especially mediastinal) and spread to contiguous nodes, whereas non-Hodgkin's is more likely to be peripheral, with extranodal involvement and noncontiguous spread

2010-351
Lacunar cells are variants of Reed-Sternberg cells that are found in what variant of Hodgkin's disease?
Nodular sclerosing (65% to 75% of all Hodgkin's cases)

2010-351
What is the CD profile of Reed-Sternberg cells?
CD30+ and CD15+

2010-351
Which type of Hodgkin's lymphoma has a poor prognosis and is most often found in older men with disseminated disease?
Lymphocyte depleted (rare)

2010-351
Which type of Hodgkin's lymphoma has an intermediate prognosis and is marked by numerous Reed-Sternberg cells and lymphocytes?
Mixed cellularity (25% of all Hodgkin's cases)

2010-351
Which two types of Hodgkin's lymphoma have the most favorable prognosis?
Nodular sclerosing (65% to 75% of all Hodgkin's cases) and lymphocyte predominant (6% of all Hodgkin's cases)

2010-351
How does the ratio of Reed-Sternberg cells to lymphocytes correlate with prognosis of Hodgkin's lymphoma?
The higher the lymphocyte to Reed-Sternberg cell ratio, the better the prognosis

2010-351
Histopathology in a 30-year-old female with Hodgkin's lymphoma shows few Reed-Sternberg cells, many lymphocytes, and collagen banding with lacunar cells; what is the lymphoma type?
Nodular sclerosing, the most common type of Hodgkin's lymphoma (65% to 75% of all Hodgkin's cases), seen in young adults, in women more than men

2010-351
What is the histological appearance of Burkitt's lymphoma?
Sheets of lymphocytes interspersed with macrophages ("starry-sky" appearance)

2010-352
What is the gene product of the translocation associated with Burkitt's lymphoma?
c-myc oncogene moves next to heavy-chain immunoglobulin gene promoter and is thus produced in large quantities

2010-352
What infectious etiology is associated with Burkitt's lymphoma?
Epstein-Barr virus

2010-352
Which variant of non-Hodgkin's lymphoma is associated with t(14;18)/bcl-2 expression?
Follicular lymphoma (small cleaved cell); bcl-2 inhibits apoptosis in this B-cell lymphoma

2010-352
A Japanese patient presents to your office with multiple skin lesions and lab tests reveal HTLV-1 infection; what malignancy should you consider?
Adult T-cell lymphoma, an aggressive neoplasm seen in regions endemic for HTLV-1: Japan, West Africa, and the Caribbean

2010-352
What lesion is associated with the endemic form of Burkitt's and where is this lymphoma endemic?
Jaw lesions, Africa; in the United States, pelvic or abdominal lesions are often the presenting complaint

2010-352
What is the prognosis in a patient who has follicular lymphoma?
This has a slow, indolent course, but is difficult to cure so it becomes a chronic disease

2010-352
Lymphoma immunohistochemistry reveals a t(11:14) translocation and CD5 positivity; what is the prognosis?
This is mantle cell lymphoma, a type of non-Hodgkin's lymphoma seen in older males that has poor prognosis

2010-352
Which variant of non-Hodgkin's lymphoma is associated with t(8;14)/c-myc expression?
Burkitt's lymphoma, a B-cell lymphoma

2010-352
What translocation is associated with mantle cell lymphoma?
t(11;14)

2010-352
A patient presents with multiple cutaneous lesions, and a biopsy is consistent with mycosis fungoides; what is the prognosis?
Mycosis fungoides, or Sézary syndrome, is a T-cell lymphoma seen in adults that has an indolent course

2010-352
Which form of non-Hodgkin's lymphoma can be derived from either B or T cells?
Diffuse large-cell lymphoma (80% B-cell, 20% T-cell origin), the most common adult non-Hodgkin's lymphoma

2010-352
A patient has a monoclonal spike on protein electrophoresis; what symptoms and imaging would suggest this is due to Waldenstrom's macroglobulinemia rather than multiple myeloma?
Waldenstrom's macroglobulinemia is more likely to have symptoms of hyperviscosity due to immunoglobulin M pentamers and does not have bone lesions, unlike myeloma

2010-353
What findings on radiograph should elevate your suspicion for multiple myeloma?
Round "punched-out" lytic bone lesions

2010-353
A patient with multiple myeloma presents with a fever; what dangerous complication of his disease is he at risk for?
Infection. Multiple myeloma is an immunosuppressed state: the immunoglobulins produced are nonfunctional and neoplastic plasma cells crowd out functional leukocytes

2010-353
A 65-year-old man has a monoclonal immunoglobulin spike on serum protein electrophoresis. He is asymptomatic. What is the likely diagnosis?
Monoclonal gammopathy of undetermined significance; the condition may or may not progress to multiple myeloma

2010-353
Name a characteristic finding of multiple myeloma that can be seen on peripheral blood smear.
Rouleaux formation, in which red blood cells appear to be stacked like poker chips

2010-353
What are two minimally invasive tests that can be used to further corroborate the diagnosis in a patient you suspect has multiple myeloma?
Bence Jones proteins (immunoglobulin light chain) in the urine, and monoclonal immunoglobulin spike on protein electrophoresis; bone marrow is required for definitive diagnosis

2010-353
Compared to Waldenstrom's macroglobulinemia, multiple myeloma predominantly produces which immunoglobulin?
Myeloma produces immunoglobulin G (55%) or immunoglobulin A (25%), compared to immunoglobulin M produced in Waldenstrom's macroglobulinemia

2010-353
A 55-year-old male undergoes bone marrow biopsy, which reveals a proliferation of plasma cells that have a "fried egg" appearance; what is the diagnosis?
This is likely multiple myeloma, a plasma cell neoplasm that is the most common primary bone tumor in patients older than 40-50 years

2010-353
What is the etiology of renal failure in multiple myeloma?
Immunoglobulin light chains excreted in the urine (Bence Jones proteins) are toxic to renal tubules; the elevated proteins also cause primary (AL) amyloidosis

2010-353
A patient presents with lower back pain, and in your work-up you note he has anemia, elevated creatinine, and elevated calcium; what diagnosis should you consider?
Multiple myeloma (think CRAB: hyperCalcemia, Renal insufficiency, Anemia, and Bone/Back pain)

2010-353
A child presents with a mediastinal mass, and immunohistochemistry reveals a population of cells that are positive for terminal deoxynucleotidyl transferase (TdT) and common acute lymphoblastic leukemia antigen (CALLA); what is the diagnosis?
Acute lymphocytic leukemia/lymphoma; TdT is a marker of prelymphocytes

2010-354
The peripheral smear of an adult patient has multiple granulated blast forms and you note small, stick-like crystals in the cytoplasm; what is the diagnosis?
This patient has myeloblasts (granulated blasts), and Auer rods ("bundles of sticks") consistent with acute myelogenous leukemia

2010-354
In the course of chronic myelogenous leukemia, what event causes the beginning of the terminal phase of the illness?
Transformation to either acute myelogenous leukemia or acute lymphoblastic leukemia (ALL), termed a "blast crisis"; until this phase, chronic myelogenous leukemia typically follows a more indolent course

2010-354
What is the treatment for the M3 type of acute myelogenous leukemia?
All-trans retinoic acid, which causes the myeloblasts to differentiate into mature forms

2010-354
Bone marrow biopsy of a child with a mediastinal mass and immature white blood cell forms in the peripheral blood reveals an extreme elevation in lymphoblasts; what cell characteristic would portend a better prognosis?
Translocation (12;21) conveys a better prognosis; although acute lymphoblastic leukemia (ALL) is generally responsive to chemotherapy

2010-354
What diagnosis is confirmed by the presence of the Philadelphia chromosome (t[9;22], bcr-abl)?
Chronic myelogenous leukemia

2010-354
What lab test can help distinguish between chronic myelogenous leukemia and a leukemoid reaction in a patient with an elevated white blood cell count?
Leukocyte alkaline phosphatase; it will be low in chronic myelogenous leukemia and elevated in a leukemoid reaction (exuberant leukocyte response to infection)

2010-354
Patients with chronic myelogenous leukemia are most likely to be in what age range?
Patients with chronic myelogenous leukemia are typically between the ages of 30 and 60 years

2010-354
Patients with acute lymphoblastic leukemia are most likely to be in what age range?
Patients with acute lymphoblastic leukemia (ALL) are typically younger than than 15 years of age

2010-354
Patients with chronic lymphocytic leukemia are most likely to be in what age range?
Patients with chronic lymphocytic leukemia are typically over the age of 60 years

2010-354
What is the treatment of choice of chronic myelogenous leukemia?
Imatinib, a compound that inhibits the bcr-abl tyrosine kinase

2010-354
An elderly male presents with fatigue and you note anemia with a positive Coombs test and smudge cells on the peripheral smear; what is the likely diagnosis?
Chronic lymphocytic leukemia (indistinguishable from small lymphocytic lymphoma)

2010-354
A patient with massive splenomegaly has a leukocyte count of 100,000 with a differential including elevated neutrophils, metamyelocytes, basophils, and occasional immature myeloid cells; what is the likely diagnosis?
Chronic myelogenous leukemia; all myeloid cell lines are increased in this condition

2010-354
An elderly patient presents with a mature B-cell tumor that has cells on the peripheral smear with filamentous, hairlike projections; what lab study should you obtain to confirm the diagnosis?
Tartrate-resistant acid phosphatase test; the factors suggest hairy cell leukemia, which will stain tartrate-resistant acid phosphatase positive

2010-354
In which tissues, outside of the blood and lymphatic system, might neoplastic leukocytes be found in a patient with acute lymphoblastic leukemia (ALL)?
ALL often metastasizes to the central nervous system and testes

2010-354
A patient has an unregulated proliferation of clonal leukocytes in the bone marrow; what is the effect on circulating red blood cells, white blood cells, and platelets?
The patient has leukemia, which crowds the bone marrow causing decreased red blood cells (anemia), decreased mature white blood cells (and variable numbers of circulating white blood cells), and decreased platelets (bleeding risk)

2010-354
What are two laboratory tests that would indicate a patient more likely has a leukemoid reaction than leukemia?
Increased white blood cell count with a left shift (approximately 80% bands) and increased levels of leukocyte alkaline phosphatase

2010-355
What is a complication of treatment of acute promyelocytic leukemia (M3)?
Treatment of acute myelogenous leukemia M3 with all-trans retinoic acidretinoic acid can lead to release of Auer rods, which can lead to disseminated intravascular coagulation

2010-355
What are Auer rods?
Peroxidase-positive cytoplasmic inclusions found in granulocytes and myeloblasts in patients with acute promyelocytic leukemia

2010-355
Name the chromosomal translocation associated with promyelocytic leukemia (M3).
t(15;17)

2010-355
Name the abnormal protein and the chromosomal translocation that are associated with follicular lymphomas.
bcl-2; t(14;18)

2010-355
Name the chromosomal translocation that is associated with Ewing's sarcoma.
t(11;22)

2010-355
Name the abnormal protein and the chromosomal translocation that are associated with Burkitt's lymphoma.
c-myc; t(8;14)

2010-355
Name the abnormal protein and the chromosomal translocation that are associated with chronic myelogenous leukemia.
bcr-abl; t(9;22), the "Philadelphia chromosome" (remember: Philadelphia CreaML cheese)

2010-355
Name the chromosomal translocation that is associated with mantle cell lymphoma.
t(11;14)

2010-355
Histiocytosis X can commonly present in three disease patterns known by what names?
Letterer-Siwe disease (hepatosplenomegaly and seborrhea), Hand-Schüller-Christian disease (exophthalmos, lytic skull lesions, and diabetes insipidus), or eosinophilic granulomas

2010-355
Immunohistochemistry reveals cells that express S-100 and CD1a surface proteins, and you note "tennis racket"-shaped objects in cells on electron microscopy; what is the diagnosis?
Histiocytosis X; the "tennis racket" inclusions are Birbeck granules

2010-355
From what lineage do the cells that cause histiocytosis X originate?
Histiocytosis X is a proliferation of dendritic cells (Langerhans cells) that are from the monocyte lineage

2010-355
What myeloproliferative disorders are associated with the JAK2 mutation (involved in hematopoietic signaling)?
Polycythemia vera, essential thrombocytosis, myelofibrosis

2010-356
What is the basic pathophysiology of polycythemia vera?
An abnormal stem cell clone acquires a JAK2 mutation, which allows it to proliferate independent of growth factors

2010-356
What are the relative levels of red blood cells, white blood cells, and platelets in myelofibrosis?
Decreased in red blood cells with variable levels of white blood cells and platelets

2010-356
What are the relative levels of red blood cells, white blood cells, and platelets in polycythemia vera?
Increase in all three lines

2010-356
What are the relative levels of red blood cells, white blood cells, and platelets in essential thrombocytosis?
Increase in platelets, no change in red blood cell and white blood cell levels

2010-356
What is the expected erythropoietin level in a patient with polycythemia vera?
Low; because of the JAK2 mutation, erythrogenic precursor cells divide independently of erythropoietin, and increased red blood cells provide negative feedback to the kidneys

2010-356
What are the relative levels of red blood cells, white blood cells, and platelets in chronic myelogenous leukemia?
Decrease in red blood cells, increase in white blood cells and platelets

2010-356
Name five clinical uses for heparin.
Immediate anticoagulation for pulmonary embolism, stroke, acute coronary syndrome, myocardial infarction, deep vein thrombosis

2010-356
What are the benefits of enoxaparin that allow it to be used in the outpatient setting, as opposed to heparin?
Enoxaparin can be administered subcutaneously, has a longer half-life, and does not need monitoring

2010-356
What difference between heparin and enoxaparin should be considered when there is a concern of excessive anticoagulation?
Enoxaparin is not easily reversible, whereas heparin can be reversed by the administration of protamine sulfate; heparin also has a shorter half-life

2010-356
How can you rapidly reverse the anticoagulation induced by heparin administration?
Give protamine sulfate, a positively charged molecule that binds negatively charged heparin

2010-356
What laboratory test can be used to determine if the heparin dose is in the therapeutic range?
The partial thromboplastin time, because heparin activates antithrombin III, which is involved in the intrinsic coagulation pathway

2010-356
During pregnancy, is heparin or warfarin the preferred method of anticoagulation?
Heparin; unlike warfarin, which is a teratogen, heparin does not cross the placenta and thus can be used during pregnancy

2010-356
What is the mechanism by which heparin anticoagulates?
It catalyzes the activation of antithrombin III and reduces levels of thrombin and factor Xa

2010-356
Name four toxicities of heparin.
Bleeding, thrombocytopenia, osteoporosis, and drug-drug interactions

2010-356
A 70-year-old woman is given prophylactic heparin while hospitalized for pneumonia. Five days later she develops deep vein thromboses and has a low platelet count. What disease is the likely cause of her sudden thrombocytopenic, hypercoagulable state?
Heparin-induced thrombocytopenia; this is an antibody-mediated process that destroys some platelets and overactivates those that remain

2010-356
Compare enoxaparin and heparin.
Enoxaparin is more specific for factor Xa, has higher bioavailability, and 2-4 times longer half-life

2010-356
A patient who was started on heparin therapy develops heparin-induced thrombocytopenia; what change should be made to his treatment regimen?
Discontinue the heparin and continue anticoagulation using lepirudin or bivalirudin (direct thrombin inhibitors)

2010-356
What is the mechanism of action of lepirudin or bivalirudin?
These are hirudin derivatives (an anticoagulant produced by leeches) that directly inhibit thrombin

2010-356
What is the mechanism by which warfarin anticoagulates?
It interferes with normal synthesis and the gamma-carboxylation of the vitamin K-dependent clotting factors II, VII, IX, and X and proteins C and S

2010-357
Name four toxicities of warfarin.
Bleeding, teratogenic effects (contraindicated in pregnancy), drug-drug interactions, and skin/tissue necrosis

2010-357
What is the therapy of choice for patients who require chronic anticoagulation: heparin or warfarin?
Warfarin; this is preferred to heparin because it can be administered orally

2010-357
How is warfarin metabolized?
By the cytochrome P450 pathway in the liver

2010-357
What laboratory test is used to determine if a patient taking warfarin is in the therapeutic range?
The prothrombin time will be increased because warfarin affects the EXtrinsic pathway (factor VII) (remember: the EX-PresidenT went to WAR(farin))

2010-357
What is the difference in the onset of action between heparin and warfarin?
Heparin acts within seconds, whereas warfarin has a slow onset dependent on the half-lives of the normal clotting factors

2010-357
Describe the treatment of acute warfarin overdose.
Intravenous vitamin K and fresh frozen plasma

2010-357
What is the difference in administration between heparin and warfarin?
Heparin is administered parenterally (intravenously or subcutaneously) whereas warfarin is given orally

2010-357
What major complication can be seen from the administration of thrombolytics?
Bleeding

2010-358
What is the pharmacologic treatment for thrombolytic toxicity?
Aminocaproic acid, an inhibitor of fibrinolysis; this medication can also be given to hemophiliacs for dental procedures

2010-358
How do thrombolytics affect the prothrombin time, partial thromboplastin time, and platelet count?
There is an increase in both the prothrombin time and partial thromboplastin time due to deactivation of thrombin, and no effect on the platelet count

2010-358
What is the primary enzymatic reaction that thrombolytics affect?
They either directly or indirectly aid the conversion of plasminogen to plasmin, which initiates fibrinolysis

2010-358
Name four drugs that can be used to lyse an existing thrombus.
Thrombolytics include streptokinase, urokinase, tissue plasminogen activator (alteplase), and anisoylated plasminogen streptokinase activator complex (anistreplase)

2010-358
What treatment option can potentially reverse the pathology of an early myocardial infarction or ischemic stroke?
Prompt administration of thrombolytics

2010-358
Name five contraindications for thrombolytics that are related to increased risk of bleeding.
Patients with active bleeding, a history of intracranial bleeding, recent surgery, known bleeding disorders, or severe hypertension

2010-358
Which terminal pathway in the coagulation cascade is enhanced by the administration of thrombolytics?
Degradation of fibrin and fibrinogen to fibrin-split-products and deactivation of thrombin

2010-358
How does abciximab downregulate platelet aggregation?
Abciximab is a monoclonal antibody that binds to the glycoprotein receptor IIb/IIIa on activated platelets, thereby preventing aggregation

2010-358
Name four clinical effects of aspirin administration.
Antipyretic, analgesic, antiinflammatory, and antiplatelet

2010-359
Name five common toxicities of aspirin.
Gastric ulceration, bleeding, hyperventilation, Reye's syndrome, and tinnitus (cranial nerve VIII)

2010-359
What is the effect of aspirin on bleeding time, prothrombin time, and partial thromboplastin time?
Aspirin increases bleeding time but has no effect on prothrombin time or partial thromboplastin time (it only affects platelets)

2010-359
What is the mechanism of action of aspirin as an anticoagulant?
Aspirin acetylates and irreversibly inhibits cyclooxygenase-1 and cyclooxygenase-2 to prevent the conversion of arachidonic acid to thromboxane A2

2010-359
What are three common clinical uses of clopidogrel and ticlopidine?
Acute coronary syndrome, during coronary stenting, and reducing the incidence or recurrence of thrombotic stroke

2010-359
What is the mechanism of action of clopidogrel and ticlopidine?
They inhibit platelet aggregation by irreversibly blocking adenosine diphosphate receptors; without adenosine diphosphate activation there is no glycoprotein IIb/IIIa expressed to bind fibrinogen

2010-359
Name a potentially life-threatening toxicity of the antiplatelet drug ticlopidine.
Neutropenia

2010-359
Name the two most common toxicities of abciximab.
Bleeding and thrombocytopenia

2010-359
Name a monoclonal antibody that binds the glycoprotein IIb/IIIa receptor on activated platelets and that prevents aggregation.
Abciximab

2010-359
What are clinical uses of abciximab?
Coronary artery disease: acute coronary syndromes or during percutaneous transluminal coronary angioplasty

2010-359
What occurs during stage G1 of the cell cycle?
The synthesis of components that are needed for DNA synthesis

2010-359
During which stage of the cell cycle do antimetabolite drugs work?
S

2010-359
During which stage of the cell cycle do vinca alkaloids and taxols work?
M

2010-359
During which stages of the cell cycle does etoposide work?
S and G2

2010-359
What occurs during stage G2 of the cell cycle?
The synthesis of components that are needed for mitosis

2010-359
What occurs during stage S of the cell cycle?
DNA synthesis

2010-359
What occurs during stage M of the cell cycle?
Mitosis

2010-359
During which stage of the cell cycle does bleomycin work?
G2

2010-359
How do vinca alkaloids exert their antineoplastic effect?
They inhibit microtubule formation, which results in an inability of the cell to perform mitosis

2010-359
What reaction is inhibited by the administration of 6-mercaptopurine?
The synthesis of purines

2010-359
What reaction is inhibited by the administration of either methotrexate or fluorouracil?
The synthesis of thymidine, a pyrimidine

2010-359
How do alkylating agents and cisplatin exert an antineoplastic effect?
These agents introduce cross-links in the DNA structure

2010-359
Name five antineoplastic drugs that function by damaging DNA.
Alkylating agents, cisplatin, dactinomycin, doxorubicin, etoposide

2010-359
How does paclitaxel exert its antineoplastic effect?
Paclitaxel inhibits microtubule disassembly, which interrupts the dynamic process required for cell division

2010-359
Which antineoplastic drugs interfere with DNA by intercalating into the double helix?
Doxorubicin, dactinomycin

2010-359
Name three antineoplastic drugs that function by interfering with nucleotide synthesis.
Methotrexate, fluorouracil, and 6-mercaptopurine

2010-359
How does etoposide exert its antineoplastic effect?
Etoposide inhibits topoisomerase II, which is needed to relieve DNA supercoils

2010-359
Name two antineoplastic drugs that function by interfering with cellular division.
Vinca alkaloids, paclitaxel

2010-359
What precaution should patients on fluorouracil take, especially in the summer?
Sun protection; fluorouracil can cause photosensitivity

2010-360
How does cytarabine exert its antineoplastic effect?
It is a pyrimidine antagonist, which inhibits DNA polymerase (needed in S phase)

2010-360
What adverse effects are seen in patients treated using 6-mercaptopurine or 6-thioguanine?
Bone marrow suppression and liver disease; patients on 6-mercaptopurine also often have gastrointestinal complaints

2010-360
What drug, used as an antineoplastic agent, is a folic acid analog that works by inhibiting dihydrofolate reductase?
Methotrexate; inhibiting dihydrofolate reductase results in less deoxythymidine monophosphate, which is needed for DNA and protein synthesis

2010-360
What agent can be administered to reverse the bone marrow suppression seen with methotrexate use?
Leucovorin (folinic acid) can be used to "rescue" the bone marrow by competing with methotrexate for binding sites; however, it does not reverse the effects of fluorouracil

2010-360
Often given in conjunction with methotrexate, fluorouracil is administered in the treatment of which tumors?
Colon cancer and solid tumors (it is a part of the FOLFOX regimen); it is applied topically for basal cell carcinoma

2010-360
A patient is given an excessive dose of fluorouracil; what can be administered to the patient to reduce toxicity?
Thymidine (remember: folinic acid will not reverse fluorouracil)

2010-360
6-mercaptopurine is used in the treatment of what types of cancers?
Leukemias and lymphomas (although not used in chronic lymphocytic leukemia or Hodgkin's lymphoma)

2010-360
Name four cancers that methotrexate can be used to treat.
Leukemias, lymphomas, choriocarcinomas, and sarcomas (all have many dividing tumor cells)

2010-360
What neoplasms are commonly treated using cytarabine?
Cytarabine is used to treat leukemias and lymphomas (acute myelogenous leukemia, acute lymphoblastic leukemia, and high-grade non-Hodgkin's lymphoma)

2010-360
Other than myelosuppression, what are three toxicities of treatment with methotrexate?
Patients can develop a painful mucositis and microvesicular fatty changes in the liver, and it is a teratogen (remember: used for abortion in ectopic pregnancy)

2010-360
Name three nonneoplastic conditions in which methotrexate can be used.
Abortion in ectopic pregnancy (blocks cell proliferation), rheumatoid arthritis, and psoriasis (decreased immune proliferation and response)

2010-360
What are three adverse effects seen in the use of cytarabine?
Leukopenia, thrombocytopenia, and megaloblastic anemia

2010-360
Which disease is treated using 6-thioguanine?
Acute lymphoblastic leukemia

2010-360
You are treating a patient with leukemia and are considering using either 6-mercaptopurine or 6-thioguanine. Which should be used if you are also going to treat with allopurinol to prevent tumor lysis syndrome?
6-thioguanine; 6-mercaptopurine is metabolized by xanthine oxidase, the enzyme blocked by allopurinol and may thus reach toxic levels if given together

2010-360
What is the mechanism by which 6-mercaptopurine and 6-thioguanine exert an antineoplastic effect?
These are purine analogs, which inhibit de novo purine synthesis; they are activated by hypoxanthine-guanine phosphoribosyltransferase

2010-360
What enzyme is inhibited by the 5F-dUMP/folic acid complex?
This complex inhibits thymidylate synthase, which is needed to form deoxythymidine monophosphate for DNA and protein synthesis

2010-360
How does fluorouracil, a pyrimidine, interact with folic acid?
Fluorouracil is converted to 5F-dUMP, which covalently binds folic acid rendering it ineffective (noncompetitive inhibition)

2010-360
Which chemotherapeutic drugs are most likely to cause alopecia as an adverse effect?
Doxorubicin, daunorubicin, and etoposide

2010-361
After several rounds of chemotherapy, a patient complains of severe dyspnea and your exam fails to find symptoms of heart failure; what antitumor antibiotic is likely a part of his chemotherapy regimen?
Bleomycin, an antineoplastic agent that has the well-documented adverse effect of pulmonary fibrosis; you may also see skin changes as an adverse effect

2010-361
Which of the antitumor antibiotics has the least myelosuppression as an adverse effect?
Bleomycin; myelosuppression is a common adverse effect of all other antitumor antibiotics

2010-361
What are three neoplasms in which dactinomycin is commonly used?
Ewing sarcoma, Wilms' tumor, and rhabdomyosarcoma (think of childhood tumors: ACTinomycin D because children ACT out)

2010-361
A patient presents late in the course of chemotherapy treatments with new symptoms of heart failure; what antineoplastic drug was likely a part of his regimen?
Doxorubicin and daunorubicin are notorious for dose-dependent cardiotoxicity; patients may also develop alopecia or other tissue toxicity

2010-361
Name three neoplasms commonly treated using etoposide.
Small-cell carcinoma of the lung, small-cell carcinoma of the prostate, and testicular cancer

2010-361
Which malignancies are commonly treated with doxorubicin or daunorubicin?
Hodgkin's lymphoma (the A in the ABVD regimen), myeloma, sarcoma, and solid tumors like breast, lung, or ovarian cancer

2010-361
Name two neoplasms commonly treated with bleomycin.
Testicular cancer and Hodgkin's lymphoma (the B in the ABVD regimen)

2010-361
When does etoposide interfere with the cell cycle?
Between G2 and S phase; etoposide inhibits topoisomerase II, which is needed to uncoil DNA strands and synthesize a sister chromatid

2010-361
Name three antineoplastic drugs that function by intercalating into DNA strands.
Dactinomycin (actinomycin D), daunorubicin, and doxorubicin work by intercalating in DNA strands, causing structural and functional distortions

2010-361
What common mechanism is seen in the antineoplastic drugs bleomycin, daunorubicin, and doxorubicin?
These drugs generate free radicals, which can cause DNA strand breaks and halt cell replication

2010-361
Which alkylating agents are effective at penetrating the blood-brain barrier to access the central nervous system?
Nitrosoureas, including carmustine, lomustine, semustine, and streptozotocin

2010-361
Which alkylating agents are used especially in treating central nervous system neoplasms?
Nitrosoureas; since they penetrate the blood-brain barrier they are used to treat brain tumors like glioblastoma multiforme, but also have central nervous system toxicities like dizziness and ataxia

2010-361
Which alkylating agent requires activation by the liver (and thus might not be effective in a patient with liver failure)?
Cyclophosphamide or ifosfamide; these cause DNA crosslinks at the 7 position of guanine

2010-361
What toxicity is common to both busulfan and bleomycin?
Pulmonary fibrosis; both busulfan and bleomycin can result in lung disease

2010-361
A patient who has been receiving chemotherapy for breast cancer presents with dark urine and is found to have many red blood cells in urine; this could have been avoided using what medication?
Mesna; this patient has hemorrhagic cystitis from cyclophosphamide use

2010-361
Which alkylating agent would you choose in a patient about to undergo hematopoietic stem cell transplantation?
Busulfan, which will ablate the host bone marrow before transplantation; it is also used in the treatment of chronic myelogenous leukemia

2010-361
Cyclophosphamide and ifosfamide are commonly used to treat which conditions?
These are used in breast cancer, ovarian cancer, and non-Hodgkin's lymphoma; they are also used at lower doses as immunosuppressants

2010-361
What toxicities are associated with vincristine treatment?
Neurotoxicity, including areflexia and peripheral neuritis, as well as paralytic ileus

2010-362
Which neoplasms are commonly treated using taxols?
Breast and ovarian carcinomas

2010-362
What antineoplastic drug functions by hyperstabilizing the mitotic spindle so that tumor cells are unable to complete anaphase?
PacliTAXel or other TAXols (remember: it is TAXing to stay polymerized [the microtubule stays polymerized])

2010-362
Which antineoplastic drugs bind to tubulin during the M phase, inhibiting the formation of microtubules?
Vincristine or vinblastine; microtubules are the vines of your cells, and without them the mitotic spindle cannot form

2010-362
What toxicity is associated with vinblastine treatment?
Bone marrow suppression (remember: vinBLASTine BLASTs Bone marrow)

2010-362
Name three neoplasms commonly treated using a vinca alkaloid (vincristine or vinblastine).
Hodgkin's lymphoma (the MOPP regimen contains vincristine), Wilms' tumor, and choriocarcinoma

2010-362
What are the clinical uses of cisplatin and carboplatin?
Testicular, bladder, ovary, and lung carcinomas

2010-362
Name two major toxicities of cisplatin and carboplatin.
Nephrotoxicity and acoustic nerve damage

2010-362
Which drug inhibits ribonucleotide reductase, acting during the S phase to decrease DNA synthesis?
Hydroxyurea

2010-362
What are the three clinical uses for hydroxyurea?
Melanoma, chronic myelogenous leukemia, and sickle cell disease (hydroxyurea increases concentration of hemoglobin F)

2010-362
Which cancers have prednisone as part of their treatment regimen?
Chronic lymphocytic leukemia, Hodgkin's lymphoma (MOPP regimen)

2010-362
How is prednisone thought to exert an antineoplastic effect?
Prednisone is thought to induce apoptosis, and may even affect nondividing cells

2010-362
What is the most commonly used glucocorticoid in cancer chemotherapy?
Prednisone

2010-362
A postmenopausal woman who recently started on a selective estrogen receptor modulator for breast cancer complains of vaginal spotting; what condition should you be concerned about?
Endometrial carcinoma; because of its partial agonist effects, tamoxifen increases the risk of endometrial carcinoma; raloxifene does not increase the risk of endometrial carcinoma

2010-362
What is the primary mechanism of action of tamoxifen and raloxifene?
They are selective estrogen receptor modulators that block binding of estrogen to estrogen-receptor-positive cells

2010-362
Name two selective estrogen receptor modulators that are used to treat breast cancer.
Tamoxifen and raloxifene

2010-362
Why is raloxifene not associated with an increased risk of endometrial carcinoma?
Raloxifene acts as an antagonist at the level of the endometrium; tamoxifen acts as an agonist (increasing endometrial cancer risk)

2010-362
The presence of what tumor marker would prompt you to use tamoxifen in the treatment of breast cancer?
Selective estrogen receptor modulators, like tamoxifen, act on the estrogen receptor, so the tumor should be estrogen receptor positive

2010-362
What is the proposed mechanism of action of trastuzumab?
Helps kills breast cancer cells that overexpress HER-2, possibly through antibody-dependent cytotoxicity

2010-363
What treatment option is available in a patient who has metastatic breast cancer that is HER-2/erb-B2 positive?
Trastuzumab, a monoclonal antibody against HER-2

2010-363
What organ should be monitored closely for adverse effects in a patient taking trastuzumab?
The heart; an adverse effect of trastuzumab therapy is cardiotoxicity

2010-363
What is the mechanism of action of imatinib?
This molecule binds to and inhibits the tyrosine kinase formed by the bcr-abl translocation found in chronic myelogenous leukemia

2010-363
What are the clinical uses for imatinib?
Chronic myelogenous leukemia and gastrointestinal stromal tumors (both express the bcr-abl tyrosine kinase)

2010-363