• 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

1/223

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;

223 Cards in this Set

  • Front
  • Back
Reed-Sternberg (RS) cells are characteristic of what disease?
Hodgkin's disease
Note: ↓ RS cells and ↑ lymphocytes = good prognosis
Describe the appearance of Reed-Sternberg cells.
Owl's eyes: binucleate or bilobed with the 2 halves as mirror images
What are the general characteristics of Hodgkin's lymphoma?
Presence of Reed-Sternberg cells
Localized, single group of nodes (rarely extranodal)
Contiguous spread
B signs/sx (fever, night sweats, weight loss)
Mediastinal lymphadenopathy
Bimodal age distribution (young & old), mostly men
What are the general characteristics of Non-Hodgkin's lymphoma?
Multiple peripheral nodes, commonly extranodal
Noncontiguous spread
No hypergammaglobulinemia
Few B signs/sx (fever, night sweats, weight loss)
Peak incidence: 20-40 years of age
Majority are of B-cell origin
Which type of lymphoma is associated with HIV and immunosuppression?
Non-Hodgkin's
Which type of lymphoma is associated with EBV?
Hodgkin's (50% of mixed cellularity Hodgkin's cases)
What is the most common type of Hodgkin's lymphoma and who is it seen in?
Nodular sclerosing; seen in young adults, women more than men (excellent prognosis)
What type of Hodgkin's lymphoma is associated with the greatest number of RS cells?
Mixed cellularity
Note: seen in older men; strongly associated with EBV
Which type of Hodgkin's lymphoma has few RS cells, occurs in men <35 years old, and has an excellent prognosis?
Lymphocyte predominant
Which type of Hodgkin's lymphoma has a poor prognosis?
Lymphocyte depleted
Note: this is rare and is seen in older men with disseminated disease
This disease is characterized by a monoclonal immunoglobulin spike on serum protein electrophoresis and Ig light chains in urine.
Multiple myeloma
Monoclonal immunoglobulin spike = M protein
Ig light chains in urine = Bence Jones Protein
What are the clinical features related to multiple myeloma?
Lytic bone lesions (bone pain, pathologic fractures, hypercalcemia); Renal insufficiency (anemia); Immunosuppression (recurrent infections, death)
This is a monoclonal plasma cell cancer that arises in the marrow and produces large amounts of IgG or IgA.
Multiple myeloma
Name 3 types of B-cell Non-Hodgkin's lymphoma that are seen in adults.
Small lymphocytic lymphoma; Follicular lymphoma; Mantle cell lymphoma
This type of T-cell Non-Hodgkin's lymphoma is most often seen in children.
Lymphoblastic lymphoma
Note: commonly presents with ALL and mediastinal mass; very aggressive
Which type of Non-Hodgkin's lymphoma is decribed as having a Starry-sky appearance?
Burkitt's lymphoma (starry-sky represents sheets of lymphocytes with interspersed macrophages)
What is the difference between the endemic and sporadic forms of Burkitt's lymphoma?
Endemic (African) form presents as a jaw lesion while the sporadic form presents in the pelvis or abdomen
This is the most common form of adult non-Hodgkin's lymphoma.
Diffuse large cell lymphoma
Note: majority are B cell (80%); 20% occur in kids; aggressive, but 50% are curable
What disorder is associated with t(9;22)?
CML; this is the Philadelphia chromosome which produces a bcr-abl hybrid gene
What disorder is associated with t(8;14)?
Burkitt's lymphoma (c-myc becomes activated by moving nect to the heavy-chain Ig gene)
What disorder is associated with t(14;18)?
Follicular lymphoma (bcl-2 activation)
Note: bcl-2 inhibits apoptosis
What disorder is associated with t(15;17)?
M3 type of AML (acute promyelocytic leukemia)
Note: this disorder is responsive to all-trans retinoic acid
What disorder is associated with t(11;22)?
Ewing's sarcoma
What disorder is associated with t(11;14)?
Mantle cell lymphoma
Note: this lymphoma is CD5+ and has a poor prognosis
What is the leukemoid reaction?
Very high leukocyte count with a left shift (80% bands) and ↑ leukocyte alkaline phosphatase
Name 3 consequences of leukemia due to bone marrow failure.
Anemia (↓ RBCs), infections (↓ mature WBCs), hemorrhage (↓ platelets)
This type of leukemia is TdT+, has many lymphoblasts, and may spread to the CNS and testes.
ALL
What is a characteristic histological finding of AML?
Auer rods (peroxidase-positive cytoplasmic inclusions in granulocytes and myeloblasts)
Note: Treatment of AML can release auer rods → DIC
This type of leukemia presents in older adults with lymphadenopathy and hepatosplenomegaly, but has few symptoms and an indolent course.
CLL
Note: CLL may also present with warm antibody autoimmune hemolytic anemia
What specialized type of cells are seen in the peripheral blood smear of a patient with CLL?
Smudge cells (small lymphocytes that are disrupted)
This disorder is defined by a myeloid stem cell proliferation and the Philadelphia chromosome.
CML
Describe the appearance of the cells in Hairy cell leukemia, and name the characteristic stain used.
The cells have filamentous, hair-like projections and stain positive for TRAP (tartrate-resistant acid phosphatase)
List the age profile for ALL, AML, CML, and CLL.
ALL: <15
AML: 15-40
CML: 30-60
CLL: >60
What is Histiocytosis X?
An infiltrate in the lung of Langerhans cells from the monocyte lineage. Primarily occurs in young adults who smoke. Birbeck granules are seen on EM.
What is the mechanism of heparin?
Catalyzes the activation of antithrombin III, ↓ thrombin (IIa) and Xa.
Name the drug used to reverse the actions of heparin and describe its mechanism.
Protamine sulfate is used. It is a positively charged molecule that acts by binding negatively charged heparin.
What is heparin used for?
Immediate anticoagulation for pulmonay embolism, stroke, angina, MI, DVT. Can be used during pregnancy (doesn't cross placenta)
What are the side effects/toxicity of heparin?
Bleeding, thrombocytopenia (HIT), osteoporosis, drug-drug interactions
How do the low-molecular-weight heparins (enoxaparin) differ from heparin?
LMW heparins act more on Xa, have better bioavailability and longer half-life, and can be given subcutaneously
What is the mechanism of heparin-induced thrombocytopenia (HIT)?
Heparin binds to platelets, causing autoantibody production that destroys platelets and overactivates the remaining ones, resulting in a thrombocytopenic, hypercoagulable state.
What test is used to monitor Heparin?
Partial thromboplastin time (PTT); intrinsic pathway
What are the mechanism and use of Lepirudin and Bivalirudin?
These drugs directly inhibit thrombin. They are used as an alternative to heparin for patients with HIT.
What is the mechanism of Warfarin?
Interferes with normal synthesis and γ-carboxylation of vit. K-dependent factors (2, 7, 9, 10, protein C and S)
What is Warfarin used for?
Chronic anticoagulation. Not used in pregnancy (crosses placenta).
What test is used to monitor Warfarin?
Prothrombin time (PT) or INR; extrinsic pathway
What are the side effects/toxicity of Warfarin?
Bleeding, teratogenic, skin/tissue necrosis, drug-drug interactions (metabolized by CYP450 system)
How do you treat an acute overdose of Warfarin?
Give IV vitamin K and fresh frozen plasma
Compare heparin and warfarin in terms of route of administration.
Heparin is given parenterally (IV, SC)
Warfarin is given orally
Compare heparin and warfarin in terms of site and duration of action.
Heparin works in the blood acutely (hours), warfarin works in the liver chronically (days)
Compare heparin and warfarin in terms of on-set of action.
Heparin is rapid (seconds), warfarin is slow (limited by the half-lives of normal clotting factors)
What are the thrombolytic drugs?
Streptokinase, urokinase, tPA (alteplase), APSAC (antistreplase)
What is the mechanism of the thrombolytic drugs?
aid in conversion of plasminogen to plasmin which cleaves thrombin and fibrin clots
What is the effect of the thrombolytics on PT, PTT, & platelet count?
Increased PT, Increased PTT, No change in platelet count
How do you treat toxicity with thrombolytics?
aminocaproic acid (an inhibitor of fibrinolysis)
Which platelet receptor binds to exposed collagen on an injured vascular endothelium?
GP Ia
Which platelet receptor forms fibrinogen bridges between platelets?
GP IIb/IIIa
Which platelet receptor binds vWF bound to collagen?
GP Ib
What is the mechanism of Aspirin?
irreversibly acetylates COX1 and COX2 preventing conversion of arachidonic acid to TXA2
What is the effect of aspirin on bleeding time, PT, & PTT?
Aspirin increases bleeding time with no effect on PT or PTT
What are toxicities associated with aspirin?
gastric ulceration, bleeding, hyperventilation, Reye's syndrome, tinnitus (CNVIII)
What is the mechanism of Clopidogrel & Ticlopidine?
inhibits platelet aggregation by irreversibly blocking ADP receptors. Inhibits fibrinogen binding by preventing glycoprotein IIb/IIIa expression
What toxicity is associated with Clopidogrel & Ticlopidine?
neutropenia
What is the mechanism of Abciximab?
monoclonal antibody that binds GP IIb/IIIa on activated platelets preventing aggregation
What phase of the cell cycle do vinca alkaloids inhibit?
M phase
What phase of the cell cycle do taxols inhibit?
M phase
What phase of the cell cycle do the antimetabolites inhibit?
S phase
What phases of the cell cycle does Etoposide inhibit?
S phase and G2 phase
What phase of the cell cycle does Bleomycin inhibit?
G2 phase
What is the mechanism of methotrexate?
S phase antimetabolite. Folic acid analog that inhibits DHFR causing a decrease in dTMP and therefore decreased DNA & protein synthesis
What are the clinical indications of methotrexate?
leukemias, lymphomas, choriocarcinoma, sarcomas, abortion, ectopic pregnancy, RA, psoriasis
What is a toxicity of methotrexate?
myelosuppression
How can you reverse myelosuppression due to methotrexate?
leucovorin (folinic acid)
What is the mechanism of 5-fluorouracil?
S phase antimetabolite. Pyrimidine analog bioactivated to 5F-dUMP which covalently complexes folic acid & thus inhibits thymidylate synthase. This causes a decrease in dTMP and therefore a decrease in DNA and protein synthesis.
What are the clinical indications of 5-fluorouracil?
colon cancer & other solid tumors & basal cell carcinoma
What is a toxicity of 5-fluorouracil?
myelosuppression
How can you reverse myelosuppression due to 5-fluorouracil?
thymidine (NOT leucovorin)
What is the mechanism & clinical indication of Cytarabine (ara-C)?
Inhibits DNA polymerase used to treat AML
What is the mechanism of Cyclophosphamide, ifosfamide?
alkylating agents which covalently cross link DNA at guanine N-7
What is the clinical use of Cyclophosphamide, ifosfamide?
Non-Hodgkin's lymphoma, breast & ovarian carcinomas, immunosuppressant
What is the mechanism of the nitrosureas (carmustine, lomustine, semustine, streptozocin)?
alkylate DNA (can cross BBB)
What is the clinical use of the nitrosureas?
brain tumors including glioblastoma multiforme
What is the mechanism of cisplatin & carboplatin?
alkylating agents which covalently cross link DNA at guanine N-7
What is the clinical use of cisplatin & carboplatin?
testicular, bladder, ovary, & lung carcinomas
What are the toxicities associated with cisplatin & carboplatin?
nephrotoxicity and acoustic nerve damage
What is the mechanism of Busulfan?
alkylates DNA
What is the clinical indication of Busulfan?
CML
What are the toxicities associated with Busulfan?
pulmonary fibrosis & hyperpigmentation
What is the mechanism of Doxorubicin (adriamycin) & daunorubicin?
generates free radicals & noncovalently intercalates in DNA causing strand breaks
What are the toxicities of Doxorubicin (adriamycin) & daunorubicin?
cardiotoxicity, myelosuppression, marked alopecia, toxic extravasation
What is the mechanism of Dactinomycin (actinomycin D)?
intercalated in DNA
What is the clinical use of Dactinomycin?
Wilm's Tumor, Ewing's Sarcoma, Rhabdomyosarcoma
What is the mechanism of bleomycin?
induces formation of free radicals which cause DNA strand breaks
What is the mechanism of hydroxyurea?
Inhibits ribonucleotide reductase which decrease DNA synthesis (S-phase specific)
What are the clinical uses of hydroxyurea?
Melanoma, CML, sickle cell disease
What is the mechanism of Etoposide (VP-16)?
G2 phase specific agent that inhibits topoisomerase II and increase DNA degradation
What are the clinical uses of Etoposide (VP-16)?
small cell carcinoma of the lung and prostate & testicular carcinoma
What is the mechanism of Prednisone?
may trigger apoptosis (may even work on nondividing cells)
What are the side effects of Prednisone?
cushing-like symptoms, immunosuppression, cataracts, acne, osteoporosis, hypertension, peptic ulcers, hyperglycemia, psychosis
What is the mechanism of Tamoxifen & Raloxifene?
Receptor antagonists in the breast and receptor agonists in bone; block the binding of estrogen to ER positive cells
What are the clinical uses of Tamoxifen & Raloxifene?
Breast Cancer & Osteoporosis Prevention
What is the mechanism of Trastuzumab (Herceptin)?
monoclonal antibody against HER-2 (erb-B2) which helps kill breast cancer cells that overexpress HER-2 (possibly through antibody mediated cytotoxicity)
What is the use of Trastuzumab (Herceptin)?
metastatic breast cancer
What is the major toxicity of Trastuzumab (Herceptin)?
cardiotoxicity
What is the mechanism of Imatinib (Gleevec)?
Philadelphia chromosome bcr-abl tyrosine kinase inhibitor
What are the clinical indications for Imatinib (Gleevec)?
CML & GI stromal tumors
What is the side effect associated with Imatinib (Gleevec)?
fluid retention
What is the mechanism of vincristine & vinblastine?
M-phase specific alkaloids that bind to tubulin & block the polymerization of microtubules so that the mitotic spindle cannot form
What are the clinical indications for vincristine & vinblastine?
lymphoma, Wilm's tumor, & choriocarcinoma
What are the side effects of vincristine?
neurotoxicity (areflexia, peripheral neuritis) & paralytic ileus
What is the major side effect of vinblastine?
bone marrow suppression
What is the mechanism of the taxols including Paclitaxel?
M phase specific agents that bind to tubulin & hyperstabilize polymerized microtubules so that the mitotic spindle cannot break down (anaphase cannot occur)
What are the clinical indications of the taxols?
ovarian and breast carcinomas
What are the toxicities associated with the taxols?
myelosuppression & hypersensitivity
A antigens on RBCs, anti-B antibodies in plasma
Blood Groups
B antigens on RBCs, anti-A antibodies in plasma
Blood Groups
A and B antigens on RBCs, no anitbodies
Blood Groups
No antigens, A and B antibodies in plasma
Blood Groups
Type O
Blood Groups
Type AB
Blood Groups
Rh+ blood given to an Rh- recipient (massive IgG response)
Blood Groups
Biconcave
RBC Forms
Hereditary spherocytosis, autoimmune hemolysis
RBC Forms
herediatry elliptocytosis
RBC Forms
megaloblastic anemia, bone marrow failure
RBC Forms
DIC, traumatic hemolysis
RBC Forms
G-6-PD deficiency
RBC Forms
Spiny RBC seen in abetalipoproteinemia
RBC Forms
HbC disease, Asplenia, Liver disease, Thalassemias
RBC Forms
TTP/HUS, microvascular damage, DIC
RBC Forms
Thalassemias, Anemia of chronic disease, iron deficiency, lead poisoning
RBC Forms
Megaloblastic anemia
RBC Forms
Iron deficiency, Thalessemias, Lead poisoning
Anemia
Megaloblastic anemias (Vit B12/folate deficiency), DNA-synth inhibiting drugs,
Anemia
acute hemorrhage, enzyme defects (G6PD), RBC membrane defects, bon marrow disorders, autoimmune hemolysis, Anemia of chronic disease
Anemia
RBC hemolysis
Lab Values in Anemia
decr, incr, decr, very decreased
Lab Values in Anemia
decr, decr, incr, normal
Lab Values in Anemia
normal, incr, normal, decr
Lab Values in Anemia
incr, decr, incr, very increased
Lab Values in Anemia
pancytopenia resulting from failure/destruction of myeloid multipotent stem cells
Aplastic anemia
radiation, drugs/chemicals (benzene, immunosuppressants), viral (B19, EBV, HIV, acute hepatitis), Fanconi's anemia, idiopathic
Aplastic anemia
fatigue/malaise, infections, abnormal bleeding/petechiae/purpura
Aplastic anemia
pancytopenia (but normal cell morphologies), Bone marrow: hypocellular, increase fat infiltration
Aplastic anemia
withdraw offending agent, BM transplant, transfusion, G-CSF or GM-CSF
Aplastic anemia
glutamic acid --> valine in Beta chain of HgB gene
Blood Dyscrasias
aplastic crisis (B19 infection), autosplenectomy (incr risk of encapsulated bacterial infections, Salmonella osteomyelitis), vaso-occlusive pain crises, renal papillary necrosis, splenic sequestration syndrome
Blood Dyscrasias
hydroxyurea (increase HbF); BM transplant
Blood Dyscrasias
african americans
Blood Dyscrasias
malaria
Blood Dyscrasias
Underproduction of one or more of the alpha HgB chains (there are 4)
Blood Dyscrasias
Absent Beta chain production (homozygote), severe anemia
Blood Dyscrasias
Bone marrow expansion/abnormatlity; seen in Sickle Cell and Beta-Thal major
Blood Dyscrasias
Incr serum bilirubin (jaundice, pigmented gallstones), incr reticulocytes (marrow compensation)
Hemolytic Anemias
Paroxysmal nocturnal hemoglobinuria, microangiopathic anemia
Hemolytic Anemias
Autoimmune (most), hereditary spherocytosis
Hemolytic Anemias
Coombs Test (anti-Ig Abs added to patients RBCs, positive if agglutination occurs)
Hemolytic Anemias
IgG
Hemolytic Anemias
SLE, CLL, drugs (methyldopa)
Hemolytic Anemias
IgM
Hemolytic Anemias
infections (mycoplasma pneumoniae, infectious mononucleosis), triggered by cold
Hemolytic Anemias
Spectrin or ankyrin
Hemolytic Anemias
Incr MCHC and RDW
Hemolytic Anemias
Osmotic fragility test (increased in HS)
Hemolytic Anemias
Impaired GP I anchor synthesis (membrane defect --> increased sensitivity to complement lysis)
Hemolytic Anemias
DIC, TTP/HUS, SLE, malignant hypertension
Hemolytic Anemias
Schistocytes (helmet cells)
Hemolytic Anemias
coaguation cascade activation, microthrombi production, consumption of platelets, coagulation factors, and fibrin
DIC
Gm- sepsis, Trauma, OB/GYN complications, acute Pancreatis, malignancy, nephrotic syndrome, transfusion
DIC
Incr PT, PTT, fibrin split products (d-dimers), decr platelet count. Schistocytes on blood smear
DIC
frequent nose bleeds (epistaxis), bleeding gums, petechiae/purpura; incr bleeding time
Bleeding Disorders
platelyet defects --> mucous membrane defects
Bleeding Disorders
ITP, TTP, DIC, aplastic anemia, drugs
Bleeding Disorders
hemoarthrosis (joint bleeding), easy brusing, incr PT and/or PTT
Bleeding Disorders
Coagulation factor defects
Bleeding Disorders
Hemophilia A/B, von Willenbrand disease
Bleeding Disorders
decr, incr, normal, normal
Hemorrhagic disorders
normal, normal, normal, incr
Hemorrhagic disorders
normal, incr, normal, incr
Hemorrhagic disorders
decr, incr, incr, incr
Hemorrhagic disorders
normal, normal, incr, incr
Hemorrhagic disorders
decr, incr, normal, normal
Hemorrhagic disorders
normal, incr, normal, normal
Hemorrhagic disorders
platelet adhesion (GP Ib deficiency)
Hemorrhagic disorders
platelet aggregation (GP Iib-IIIa deficiency)
Hemorrhagic disorders
Extrinsic factors (II, V, VII, X)
Hemorrhagic disorders
Intrinsic factors all except VII
Hemorrhagic disorders
Which types of lymphocytes are of lymphoid stem cell origin
B Cells (including plasma cells) and T cells
What is the WBC differential from highest to lowest?
Neutrophils > Lymphocytes > Monocytes > Eosinophils > Basophils
Main energy source for RBCs
Glucose
Definition of anisocytosis
Varying sizes of RBCs
Definition of poikilocytosis
Varying shapes of RBCs
WBCs (2) that mediate allergic reaction
Basophil, mast cell
Substances (4) found in basophil's granules
Heparin, histamine, leukotrienes, other vasoactive amines
Which type of WBC does a mast cell resemble structurally and functionally?
Basophil
What type of hypersensitivity reactions are mast cells involved in?
Type I
Which type of WBC defends against helminthic and protozoan infections?
Eosinophil
What are 5 causes of eosinophilia?
Neoplastic, Asthma, Allergic processes, Collagen vascular diseases, Parasites
Which type of WBC has a multilobed nucleus?
Neutrophil
In what disease states does one see hypersegmented neutrophils?
Vitamin B12/folate deficiency
Which WBC has a kidney shaped nucleus and a frosted glass cytoplasm
Monocyte
What is the main function of macrophages?
Phagocytose bacteria, cell debris, and senescent RBCs and scavenges damaged cells and tissues
What is a substance that activates macropages?
IFN-gamma
Macrophages use which MHC class to present antigens?
Class II
What cells are professional APCs?
Dendritic cells
Which WBCs have round, densely staining nucleus?
Lymphocytes
Where do B cells arise from?
Stem cells in bone marrow
When a B cell encounters antigen, what does a B cell differentiate to, and produce?
Plasma cells, and they produce antibodies
Which type ofWBC has an off-center nucleus and clock-face chromatin?
Plasma cells, and they produce antibodies
Multiple myeloma is a neoplasm of what cells?
Multiple myeloma
Where do T cells originate from?
Stem cells in bone marrow, and mature in thymus
The majority of circulating lymphocytes are what kind of cells?
T cells
Cytotoxic T cells are CD_____ and receive antigen presentation from MHC Class ______
CD8, MHCI
Helper T cells are CD ______ and receive antigen prenstation from MHC _____
CD4, MHC II
What factors are involved in the intrinsic clotting pathway (before convergence)
XII, XI, IX, VIII, X
What factors are involved in the extrinsic clotting pathway (before convergence)
VII, tissue factor
Factor Va converts what to what?
Prothrombin to thromin
Thrombin converts what to what?
Fibrinogen to fibrin
What are the vitamin K-dependent coagulation factors?
II, VII, IX, X, protein C, protein S
What does activated protein C do?
Inactivates Factors Va and VIIIa
What does antithrombin III do?
Inactivates thrombin, IX a, X a, XI a
What does tPA do?
Genetrates plasmin from plasminogen, which cleaves fibrin clots
Factor V Leiden mutation causes resistance to what?
Activated Protein C
Activation of the kinin cascade causes what three effects?
Increased vasodilation, increased permeability, increased pain