• 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

1/86

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;

86 Cards in this Set

  • Front
  • Back
Decreases thrombin and Xa, short half-life -- used for immediate anticoagulation for pulmonary embolism, stokre, acute coronary syndrome, MI, DVT.
1. major mechanism
2. follow what lab?
Heparin
1. catalyzes the activation of antithrombin III
2. PTT
Protamine sulfate is used for the rapid reversal of the effects of what drug?
1. mechanism
Heparin
1. positively charged molecule that acts by binding negatively charged heparin
act more on Xa, and can be administered without lab monitoring of PTT
1. problem
low-molecular weight heparins (i.e. ENOXAPARIN)
1. not easily reversible
binds platelets causing autoantibody production that destroys platelets and overactivates the remaining ones, resulting in a thrombocytopenic, hypercoagulable state
Heparin causing Heparin-induced thrombocytopenia (HIT)
Pathogenesis of HIT
1. Heparin binds PLT factor 4 to form heparin:PF4 complex
2. Igs form against that complex -- autoantibody production leads to destruction of PLTs and activation of platelets --> thrombocytopenia AND hypercoagulable state
Hirudin derivatives; directly inhibit thrombin
1. used for
Lepirudin, bivalirudin
1. alternative to heparin for anticoagulating patients with HIT
Alternative to anticoagulating patients with HIT
1. mechanism
Lepirudin, bivalrudin
1. directly inhibits thrombin
Has a long half-life. Requires monitoring of PT.
1. mechanism
2. how metabolized?
Warfarin
1. interferes with normal synthesis of gamma-carboxylation of vitamin K-dependent clotting factors II, VII, IX, X, PC, and PS.
2. cytochrome P450
Affects the extrinsic pathway
1. clinical use
2. huge contraindication
Warfarin
1. Chronic anticoagulation
2. pregnancy -- because it crosses the placenta
Toxicities include bleeding, teratogenesis, skin/tissue necrosis, and drug-drug interactions.
Warfarin
Acetylates and irreversibly inhibits COX 1 and 2 preventing what process which promotes platelet aggregation:
1. has what effect on PT, PTT, bleeding time?
Aspirin (ASA)
inhibition of COX prevents conversion of arachidonic acid to TXA2 which promotes PLT aggregation
1. No effect on PT, PTT, but increases bleeding time
Toxicities include gastric ulceration, bleeding, hyperventilation, Reye's syndrome, and tinnitus (CN VIII)
1. clinical use
Aspirin (ASA)
1. antipyretic, analgesic, anti-inflammatory, antiplatelet
Inhibit PLT aggregation by irreversibly blocking ADP receptors.
1. inhibit PLT binding to fibrinogen how?
Clopidogrel, ticlopidine
1. prevent glycoprotein G2b/3a from being expressed
Monoclonal antibody that binds to glycoprotein receptor 2b/3a on activated platelets, preventing aggregation
1. toxicities
Abciximab
1. bleeding, thrombocytopenia
Used for acute coronary syndrome; coronary stenting. decreases the incidenece or recurrence of thrombotic stroke
1. toxicity
Clopidogrel, ticlopidine
1. Neutropenia (tocloopidine)
used for acute coronary syndromes, percutaneous transluminal coronary angioplasty, monoclonal antibody
1.mechanism
Abciximab
1. antibody to Gp2b/3a mimicking Glanzmann's disease
Pyrimidine analog bioactivated to a form that covalently complexes folic acid. This complex inhibits thymidylate synthease resulting in a decrease in dTMP
1. Toxicity
5-fluorouracil bioactivated to 5F-dUMP
1. myelosuppresion which is not reversible with leucovorin, photosensitivity
Thymidine is administered to "rescue" the patient from this treatment
1. mechanism
5-fluorouracil
1. bioactivated to 5F-dUMP which complexes with folic acid, this complex inhibits thymidylate synthase
Blocks de novo purine synthesis. activated by HGPRTase.
1. toxicity
2. metabolized by:
6-MP (mercaptopurine)
1. bone marrow, GI, liver.
2. xanthine oxidase
Toxicity increased with allopurinol, why?
6-MP
b/c allopurinol inhibits xanthine oxidase which metabolizes 6-MP
what is histiocytosis X?
proliferative disorder of dendritic langerhan cells from the monocyte lineage
What is the site of action for Warfarin & Heparin?
Warfarin acts in the liver

Heparin acts in the blood
Describe the onset of action of Heparin vs. Warfarin
heparin is raid in seconds

Warfarin is slow, limiedt by half lives of normal clotting factors
what is the limit of Warfarin's onset of effectiveness?
the half lives of normal clotting factors
What is the duration of action for Heparin & warfarin?
Hep has acute hours
Warfar has chronic (days)
Which inhibits coagulation in vitro Heparin or warfarin?
Heparin
What is the treatment of acute overdose of warfarin?
IV vitamin K & Fresh frozen plasma
Remember that Vit K will give you protein C & S faster so dont give Vit K first if their INR is real high
Which crosses the palcenta Warfarin or heparain?
Warfarin is teratogenic
how do thrombolytics affect PT & PTT?
they increase both because they are acting on the process which converts plasminogen to plasmin which breaks down the clot formed by either the intrinsic or extrinsic pathway
What is the clinical use for thrombolytics?

when are the contraindicated?
1. Early Mi
2. ischemic stroke

they are contraindicated in people with bleeding issues or sever HTN
What is urokinase and what endogenous compound is it similar to in its action?
urokinase is a thrombolytic and does the same thing as tPA
What is aminocaproic acid?
This is an antithrombolytic that inhibits the conversion of plasminogen to plasmin
How does streptokinase affect clotting?
Streptokinase combines with a proactivator and converts plasminogen to plasmin which breaks down the clot
How does anistreplase (APSAC) affect clotting?
Anestreplase converts plasminogen to plasmin
Synergy with MTX, why?
5-FU -- b/c it inhibits thymidylate synthase, which also prevents the synthesis of dTMP

MTX works by inhibiting dihydrofolate reductase so that it can't regenerate the THF that is needed to make dTMP from dUMP
converted to a nucleotide analog that inhibits DNA polymerase
1. toxicities
Cytarabine (ara-C)
converted to cytosine arabimoside triphosphate
1. megaloblastic anemia, luekopenia, thrombocytopenia
toxicities include leukopenia, thrombocytopenia, megaloblastic anemia
Cytarabine (C-ara)
1. inhibits DNA polymerase -- converted to cytosine arabinoside triphosphate
Alkylating agents that covalently cross-link DNA at guanine N-7
1. require bioactivation by:
Cyclophosphamide, ifosfamide
1. liver
Methotrexate is an antimetabolite and inhibits what phase of the cell cycle?
s phase
What drug inhibits dyhydrofolate reductase?
Methotrexate (it's a folate analog)
what drug is a folic acid analogue?
methotrexate
What is leucovorin (folinic acid)?
this is used with methotrexate to reverse its myelosuppressive effects.

It works by replacing folic acid. It is a metabolite that does not need to be acted on by dihydrofolate reductase so it can still be used and some DNA synth can occur?
What thrombolytic comes from beta-hemolytic strep?
streptokinase
What is the toxicity of methotrexate?
myelosuppression
What is the mechanism of action of Busulfan?
1. toxicity
it alkylates DNA
1. pulmonary fibrosis and hyperpigmentation
What 2 drugs can cause pulmonary fibrosis?
1. Busulfan
2. Bleomycin
What drug causes Hyper pigmentation?
Busulfan
what drugs have a red color?
Doxorubicin (adriamycin, daunorubicin

think RUBY red
Mechanism of Doxorubicin (adriamycin)?
generates free radicals & noncovalently intercalates in to DNA (this creates breaks in DNA strand to decrease replication)
What drug non-covalently intercalates into DNA?
Doxyrubicin/adriamycin
(also generates free radicals)
What drug is cardiotoxic and causes toxic extravasation?

what other toxicities?
Doxorubicin/adriamycin

alopeicia + myelosupression
What is the mechanism for Dactinomycin (actinomycin)D?

toxicitiy? (its pretty standard)
intercaltes into DNA

myelosuppression
What is the mechanism for bleomycin?
induces formation of free radicals, which cause breaks in DNA strands
What is the mechanism for hydroxyurea? & what phase of cell cylce is affected?
inhibts ribonucleotide reductase t/f DNA synthesis decreases

affects S phase
What is the mechanism for Etoposide (VP-16)? what phase of cell cylce?
G2 phase and S phase specific agent that inhibits topoisomerase 2 & increases DNA degredatoin
What is the mechanism for Prednisone?
may trigger apoptosis (but may work on undividing cells)
What is the mechanism for Tamoxifen / raloxifene? how does it help women (2 different ways)?
this is a SERM which is an estrogen receptor modulator

It acts as an estrogen antagonist in breast & agonist in the bone
what drug inhibits topoisomerase 2?
Etoposide (VP-16)
What is the toxicity of tamoxifen? how is it different from raloxifene?
increase risk of endometrial carcinoma due to its agonist effects

also causes hot flashes

Raloxifene does not increase risk for endometrial carcinoma because its an endometrial agonist
What drug can cause Cushing's like symptoms, acen, immunosuppression, cataracts, acne?
prednisone
what drug can cause hyperglycemia?
prednisone
what drug can cause GI ulcers & psychosis?
prednisone
Hydroxyurea is used in CML and what other non-cancer disease?
sickle cell b/c it increases fetal Hb
Does bleomycin cause myelosuppresion?
No daddy, it doesnt
Bleomycin inhibits what phase of the cell cycle?
G2 phase
cause hemorrhagic cystitis
1. which can partially be prevented with:
Cyclophosphamide, ifosfamide
1. mesna (sulfhydryl groups of mesna bind the intermediate metabolite acrolein which is toxic to the bladder)
Cause myelosuppresion, which is why they are sometimes used as immunosuppressants
Cyclophophamide, ifosfamide
Alkylate DNA, but require bioactivation. cross the blood-brain barrier which is why they are used for brain tumors (including glioblastoma multiforme)
Nitrosoureas
Carmustine, lomustine, semustine, streptozocin
cause CNS toxicity (dizziness, ataxia) used for brain tumors
1. mechanism
Nitrosoureas -- Carmustine, lomustine, semustine, streptozocin
1. alkylating agents that require bioactivation, cross blood-brain barrier
Carmustine, lomusstine, semustine, streptozocin
1. mechanism
Nitrosoureas
1. alkylate DNA, cross blood-brain barrier
Cross-link DNA -- cause nephrotoxicity and acoustic nerve damage
Cisplatin, carboplatin
used for testicular, bladder, ovary, and lung carcinomas
1. cross-link DNA
Cisplatin, carboplatin
1. mechanism
Monoclonal antibody against HER-2 (erb-B2)
1. helps kill breast cancer cells that:
Trastuzumab
1. overexpress HER-2, possibly through antibody-dependent cytotoxicity
causes cardiotoxicity, used for metastatic breast cancer
1. mechanism
Trastuzumab (Herceptin)
1. monoclonal antibody against HER-2 (erb-B2) which is expressed on breast cancer cells
Philadelphia chromosome bcr-abl tyrosine kinase inhibitor
1. toxicity
Imatinib (Gleevec)
1. fluid retention
used for CML, GI stromal tumors
1. mechanism
Imatinib (Gleevec)
1. Philadelphia chrosome bcr-abl tyrosine kinase inhibitor
M-phase specific alkaloids that bind tubulin and block polymerization of microtubules so that mitotic spindle cannot form
1. toxicities
Vincristine, vinblastine
1. vincristine --> neurotoxicity (areflexia, peripheral neuritis, paralytic ileus
2. vinblastine --> bone marrow suppression (Blast = Bone marrow
causes neurotoxicity (areflexia, peripheral neuritis), paralytic ileus
1. mechanism
Vincristine
1. M-phase specific alkaloid that binds tubulin and blocks polymerization of microtubules so that mitotic spindle cannot form
bone marrow suppression, M-phase-specific alkaloid
vinBLASTine BLASTS the bone marrow
M-phase-specific agents that bind to tubulin and hyperstabilize polymerized microtubules so that mitotic spindle cannot break down (anaphase cannot occur)
1. toxicities
Paclitaxel, other taxols
1. myelosuppresion and hypersensitivity
causes myelosuppression and hypersensitivity
Paclitaxel, other taxols
prevents anaphase from occuring
Paclitaxel, other taxols
Cladribine is used for:
1. Mechanism
Hairy cell leukemia
1. cytotoxic purine resistant to degradation by Adenosine deaminase
What drug limits the nephrotoxicity caused by Cisplatin?
What is the mechanism?
Amifostine
Scavenges free radicals
What limits the cardiotoxicity of Doxo/Daunorubicin?
What is the mechanism?
Dexrazoxane
Iron-chelating agent
Leucovorin can be used to potentiate the toxicity of which drug?
Leucovorin strengthens the association of 5-FU with thymidylate synthase