• 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

1/133

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;

133 Cards in this Set

  • Front
  • Back
What is the mechanism of Heparin?
Cofactor for the activation of antithrombin, ↓ thrombin, and ↓ factor Xa

Short t1/2
What are the uses of Heparin?
- Immediate anticoagulation for Pulmonary Embolism, Acute Coronary Syndrome, MI, and Deep Venous Thrombosis
- Used during pregnancy (doesn't cross placenta)
What do you need to monitor in patients taking Heparin?
Follow PTT
What are the toxic side effects of Heparin?
- Bleeding
- Thrombocytopenia (HIT)
- Osteoporosis
- Drug-drug interactions
What is HIT?
Heparin Induced Thrombocytopenia (HIT)
- Development of IgG antibodies against heparin bound to platelet factor 4 (PF4)
- Antibody-heparin-PF4 complex activates platelets → thrombosis and thrombocytopenia
How do you rapidly reverse an overdose of Heparin? Mechanism?
Protamine Sulfate (positively charged molecule that binds negatively charged heparin)
What are the types of low molecular weight heparins (LMWH)? Mechanism? How are they different?
Enoxaparin and Dalteparin
- Act more on Factor Xa
- Better bioavailability and 2-4 times longer half-life
- Can be administered subcutaneously and without lab monitoring
- Not as easily reversible
What drugs can you use in a patient who requires anticoagulation but was taking heparin and got heparin-induced thrombocytopenia (HIT)? Source?
Argatroban or Bivalirudin
- Derivatives of hirudin, the anticoagulant used by leeches
What is the mechanism and use of Argatroban?
- Inhibits thrombin directly
- Used instead of heparin for anticoagulating patients with Heparin-induced thrombocytopenia (HIT)
What is the mechanism and use of Bivalirudin?
- Inhibits thrombin directly
- Used instead of heparin for anticoagulating patients with Heparin-induced thrombocytopenia (HIT)
What is the mechanism of Warfarin?
Interferes with normal synthesis and γ-carboxylation of vitamin K-dependent clotting factors II, VII, IX, and X and proteins C and S
What clotting factors and proteins are affected by Warfarin?
- Factor II
- Factor VII
- Factor IX
- Factor X
- Protein C
- Protein S
What is the effect of warfarin on blood tests?
Increases PT (extrinsic pathway) / INR
What are the clinical uses of Warfarin?
- Chronic anti-coagulation (after STEMI, venous thromboembolism prophylaxis, and prevention of stroke in atrial fibrillation)

*NOT used in pregnant women (can cross placenta = teratogen)
What are the toxic side effects of Warfarin?
- Bleeding
- Teratogen
- Skin / tissue necrosis
- Drug-drug interactions
- Bleeding
- Teratogen
- Skin / tissue necrosis
- Drug-drug interactions
How can you reverse a warfarin overdose?
- Give Vitamin K
- For severe overdose, give fresh frozen plasma
What drugs are direct factor Xa inhibitors?
- Apixaban
- Rivaroxaban
What is the mechanism of Apixaban and Rivaroxaban?
Binds and directly inhibits activity of factor Xa
What are the clinical uses of Apixaban and Rivaroxaban?
- Treatment and prophylaxis of DVT and PE (rivaroxaban)
- Stroke prophylaxis in patients with atrial fibrillation
- Oral agents do not require coagulation monitoring!
What are the side effects of Apixaban and Rivaroxaban?
Bleeding (no specific reversal agent available)
What is the structure of heparin and warfarin?
- Heparin: large, anionic, acidic polymer
- Warfarin: small lipid-soluble molecule
What is the route of administration of heparin and warfarin?
- Heparin: parenteral (IV, SC)
- Warfarin: oral
What is the site of action of heparin and warfarin?
- Heparin: blood
- Warfarin: liver
What is the relative onset of action of heparin and warfarin?
- Heparin: rapid (seconds)
- Warfarin: slow, limited by half-lives of normal clotting factors
What is the mechanism of action of heparin and warfarin?
- Heparin: activates antithrombin, which ↓ the action of IIa (thrombin) and factor Xa
- Warfarin: impairs the synthesis of vitamin K-dependent clotting factors II, VII, IX, and X (vitamin K antagonist)
What is the duration of action of heparin and warfarin?
- Heparin: acute (hours)
- Warfarin: chronic (days)
Do heparin and warfarin inhibit coagulation in vitro?
- Heparin: yes
- Warfarin: no
How do you treat an acute overdose of heparin and warfarin?
- Heparin: protamine sulfate
- Warfarin: IV vitamin K and fresh frozen plasma
What should you monitor in patients taking heparin and warfarin?
- Heparin: PTT (intrinsic pathway)
- Warfarin: PT / INR (extrinsic pathway)
Do heparin and warfarin cross the placenta?
- Heparin: no
- Warfarin: yes (teratogenic)
What are the types of thrombolytics?
- Alteplase (tPA)
- Reteplase (rPA)
- Tenecteplase (TNK-tPA)
What is the mechanism of Alteplase (tPA), Reteplase (rPA), and Tenecteplase (TNK-tPA)?
Thrombolytics
- Directly or indirectly aids conversion of plasminogen to plasmin
- Plasmin cleaves thrombin and fibrin clots
- ↑ PT, ↑ PTT, and no change in platelet count
What are the clinical uses of Alteplase (tPA), Reteplase (rPA), and Tenecteplase (TNK-tPA)?
- Early MI
- Early ischemic stroke
- Direct thrombolysis of severe PE
What are the side effects and contraindications of Alteplase (tPA), Reteplase (rPA), and Tenecteplase (TNK-tPA)?
- Bleeding
- Contraindicated in patients with active bleeding, history of intracranial bleeding, recent surgery, known bleeding diatheses, or severe hypertension
How do you treat toxicity of Alteplase (tPA), Reteplase (rPA), and Tenecteplase (TNK-tPA)? Mechanism?
- Aminocaproic Acid - inhibits fibrinolysis

- Fresh frozen plasma and cryoprecipitate can also be used to correct factor deficiencies
What is the mechanism of aspirin (ASA)?
- Irreversibly inhibits cyclooxygenase (both COX-1 and COX-2) enzyme by covalent acetylation
- Platelets cannot synthesize new enzyme so effect lasts until new platelets are produced
What lab values change in patients taking aspirin (ASA)?
- ↑ Bleeding time, ↓ TXA2 and prostaglandins
- No effect on PT or PTT
What are the clinical uses of aspirin (ASA)?
- Antipyretic
- Analgesic
- Anti-inflammatory
- Antiplatelet (↓ aggregation)
What are the toxic side effects of aspirin (ASA)?
- Gastric ulceration
- Tinnitus (CN VIII)
- Chronic use can lead to acute renal failure, interstitial nephritis, and upper GI bleeding
- Reye syndrome in children with viral infection
What can an overdose of aspirin (ASA) cause?
Respiratory alkalosis initially, which is then superimposed by metabolic acidosis
What is the cause of Reye syndrome?
Children with a viral infection who take aspirin
What are the types of ADP receptor inhibitors?
- Clopidogrel
- Ticlopidine
- Prasugrel
- Ticagrelor
What is the mechanism of Clopidogrel, Ticlopidine, Prasugrel, and Ticagrelor?
- Inhibit platelet aggregation by irreversibly blocking ADP receptors
- Inhibits fibrinogen binding by preventing glycoprotein IIb/IIIa from binding to fibrinogen
What are the clinical uses of Clopidogrel, Ticlopidine, Prasugrel, and Ticagrelor?
- Acute coronary syndrome
- Coronary stenting
- ↓ Incidence or recurrence of thrombotic stroke
What are the side effects of Clopidogrel, Ticlopidine, Prasugrel, and Ticagrelor?
- Neutropenia (ticlopidine)
- TTP / HUS may be seen
What drugs inhibit platelet aggregation via inhibition of phosphodiesterase III?
Cilostazol and Dipyridamole
What is the mechanism of Cilostazol and Dipyridamole?
- Phosphodiesterase III inhibitor
- ↑ cAMP in platelets, thus inhibiting platelet aggregation
- Vasodilates
What are the uses of Cilostazol and Dipyridamole?
- Intermittent claudication
- Coronary vasodilation
- Prevention of stroke or TIAs (combined with aspirin)
- Angina pectoris
What are the side effects of Cilostazol and Dipyridamole?
- Nausea
- Headache
- Facial flushing
- Hypotension
- Abdominal pain
What are the types of GpIIb/IIIa inhibitors?
- Abciximab
- Eptifibatide
- Tirofiban
What is the mechanism of Abciximab, Eptifibatide, and Tirofiban?
- Binds to the glycoprotein receptor IIb/IIIa on activated platelets, preventing aggregation
- Abciximab is made from monoclonal antibody Fab fragments
What are the uses of Abciximab, Eptifibatide, and Tirofiban (GpIIb/IIIa inhibitors)?
- Unstable angina
- Percutaneous transluminal coronary angioplasty
What are the side effects of Abciximab, Eptifibatide, and Tirofiban (GpIIb/IIIa inhibitors)?
- Bleeding
- Thrombocytopenia
What are the drugs that interact with the cell cycle? Which part?
- Antimetabolites (affect S)
- Etoposide (affects S and G2)
- Bleomycin (affects G2)
- Vinca alkaloids and taxols (affects M)
- Antimetabolites (affect S)
- Etoposide (affects S and G2)
- Bleomycin (affects G2)
- Vinca alkaloids and taxols (affects M)
What anti-neoplastic drugs affect nucleotide synthesis? How?
- Methotrexate and 5-Fluorouracil: ↓ thymidine synthesis
- 6-Mercaptopurine: ↓ purine synthesis
- Methotrexate and 5-Fluorouracil: ↓ thymidine synthesis
- 6-Mercaptopurine: ↓ purine synthesis
What anti-neoplastic drugs inhibit DNA synthesis? How?
- Alkylating agents, Cisplatin: cross-link DNA
- Dactinomycin, Doxorubicin: intercalates into DNA
- Etoposide: inhibits topoisomerase II
- Alkylating agents, Cisplatin: cross-link DNA
- Dactinomycin, Doxorubicin: intercalates into DNA
- Etoposide: inhibits topoisomerase II
What anti-neoplastic drugs inhibit cellular division? How?
- Vinca alkaloids: inhibit microtubule formation
- Paclitaxel: inhibits microtubule disassembly
- Vinca alkaloids: inhibit microtubule formation
- Paclitaxel: inhibits microtubule disassembly
What are the anti-metabolite drugs?
- Methotrexate (MTX)
- 5-Fluorouracil (5-FU)
- Cytarabine (Arabinofuranosyl Cytidine)
- Azathioprine, 6-Mercaptopurine (6-MP), 6-Thioguanine (6-TG)
What is the mechanism of Methotrexate (MTX)?
Folic acid analog that inhibits Dihydrofolate Reductase → ↓ dTMP → ↓ DNA and ↓ protein synthesis
What are the clinical uses of Methotrexate (MTX)?
- Cancers: leukemias, lymphomas, choriocarcinoma, sarcomas
- Non-neoplastic: abortion, ectopic pregnancy, rheumatoid arthritis, psoriasis, IBD
What are the toxic side effects of Methotrexate (MTX)?
- Myelosuppression
- Macrovesicular fatty change in liver
- Mucositis
- Teratogenic
How can you reverse the myelosuppression induced by Methotrexate (MTX)?
Reversible with leucovorin (folinic acid) "rescue"
What is the mechanism of 5-Flurouracil (5-FU)?
- Pyrimidine analog bioactivated to 5F-dUMP, which covalently complexes folic acid
- This complex inhibits thymidylate synthase → ↓ dTMP → ↓ DNA and ↓ protein synthesis
What is 5-fluorouracil (5-FU) used for?
- Colon cancer
- Pancreatic cancer
- Basal cell carcinoma (topical)
What are the toxic side effects of 5-fluorouracil (5-FU)?
- Myelosuppression
- Photosensitivity
How can you reverse the myelosuppression induced by 5-Fluorouracil (5-FU)?
Reversible with uridine "rescue"
What is the mechanism of Cytarabine (Arabinofuranosyl Cytidine)?
Pyrimidine analog → inhibition of DNA polymerase
What is Cytarabine (Arabinofuranosyl Cytidine) used for?
- Leukemias
- Lymphomas
What are the toxic side effects of Cytarabine (Arabinofuranosyl Cytidine)?
- Leukopenia
- Thrombocytopenia
- Megaloblastic anemia
- CYTarabine causes panCYTopenia
What is the mechanism of Azathioprine, 6-Mercaptopurine (6-MP), and 6-Thioguanine (6-TG)?
- Purine (thiol) analogs → ↓ de novo purine synthesis
- Activated by HGPRT
What are the clinical uses of Azathioprine, 6-Mercaptopurine (6-MP), and 6-Thioguanine (6-TG)?
- Preventing organ rejection
- Rheumatoid Arthritis

- Azathioprine: Systemic Lupus Erythematosus (SLE)

- 6-MP and 6-TG: Leukemia and Inflammatory Bowel Disease
What are the toxic side effects of Azathioprine, 6-Mercaptopurine (6-MP), and 6-Thioguanine (6-TG)?
- Bone marrow, GI, and liver toxicity

- Azathioprine and 6-MP are metabolized by xanthine oxidase; thus both have ↑ toxicity with allopurinol, which inhibits their metabolism
What are the anti-tumor antibiotics?
- Dactinomycin (Actinomycin D)
- Doxorubicin (Adriamycin), Danorubicin
- Bleomycin
What is the mechanism of Dactinomycin (Actinomycin D)?
Intercalates in DNA
What are the clinical uses of Dactinomycin (Actinomycin D)?
- Wilms tumor
- Ewing sarcoma
- Rhabdomyosarcoma
- Used for childhood tumors ("children act out")
What are the toxic side effects of Dactinomycin (Actinomycin D)?
Myelosuppression
What is the mechanism of Doxorubicin (Adriamycin) and Daunorubicin?
- Generates free radicals
- Intercalates in DNA → breaks in DNA → ↓ replication
What are the clinical uses of Doxorubicin (Adriamycin) and Daunorubicin?
- Solid tumors
- Leukemias
- Lymphomas
What are the toxic side effects of Doxorubicin (Adriamycin) and Daunorubicin?
- Cardiotoxicity (dilated cardiomyopathy)
- Myelosuppression
- Alopecia
- Toxic to tissues following extravasation

- Dexrazoxane (iron chelating agent), used to prevent cardiotoxicity
What is the mechanism of Bleomycin?
Induces free radical formation, which causes breaks in DNA strands
What are the clinical uses of Bleomycin?
- Testicular cancer
- Hodgkin lymphoma
What are the toxic side effects of Bleomycin?
- Pulmonary fibrosis
- Skin changes
- Mucositis
- Minimal myelosuppression
What are the alkylating agent drugs?
- Cyclophosphamide, Ifosfamide
- Nitrosoureas (Carmustine, Lomustine, Semustine, Streptozocin)
- Busulfan
What is the mechanism of Cyclophosphamide and Ifosfamide?
- Covalently X-link (interstrand)
- DNA at guanine N-7
- Requires bioactivation by liver
What are the clinical uses of Cyclophosphamide and Ifosfamide?
- Solid tumors
- Leukemia
- Lymphomas
- Some brain cancers
What are the toxic side effects of Cyclophosphamide and Ifosfamide?
- Myelosuppression
- Hemorrhagic cystitis
How can you limit the hemorrhagic cystitis caused by Cyclophosphamide and Ifosfamide? Mechanism?
Mesna - thiol group of mesna binds toxic metabolites
What is the mechanism of Nitrosoureas (Carmustine, Lomustine, Semustine, and Streptozocin)?
- Requires bioactivation
- Crosses blood-brain-barrier → CNS
- Cross-links DNA
What are the clinical uses of Nitrosoureas (Carmustine, Lomustine, Semustine, and Streptozocin)?
Brain tumors (including glioblastoma multiforme)
What are the toxic side effects of Nitrosoureas (Carmustine, Lomustine, Semustine, and Streptozocin)?
CNS toxicity (convulsions, dizziness, ataxia)
What is the mechanism of Busulfan?
Cross-links DNA
What are the clinical uses of Busulfan?
- CML
- Also used to ablate patient's bone marrow before bone marrow transplantation
What are the toxic side effects of Nitrosoureas (Carmustine, Lomustine, Semustine, and Streptozocin)?
- Severe myelosuppression (in almost all cases)
- Pulmonary fibrosis
- Hyperpigmentation
What are the types of microtubule inhibitors?
- Vincristine and Vinblastine
- Paclitaxel and other Taxols
What is the mechanism of Vincristine and Vinblastine?
- Vinca alkaloids that bind β-tubulin
- Inhibits its polymerization into microtubules
- Prevents mitotic spindle formation (M-phase arrest)
What are the clinical uses of Vincristine and Vinblastine?
- Solid tumors
- Leukemias
- Lymphomas
What are the toxic side effects of Vincristine and Vinblastine?
- Vincristine: neurotoxicity (areflexia, peripheral neuritis), paralytic ileus

- Vinblastine: blasts bone marrow (myelosuppression)
What is the mechanism of Paclitaxel and other Taxols?
- Hyperstabilize polymerized microtubules in M phase so that mitotic spindle cannot break down (anaphase cannot occur)
- It is taxing to stay polymerized
What are the clinical uses of Paclitaxel and other Taxols?
Ovarian and breast carcinomas
What are the toxic side effects of Paclitaxel and other Taxols?
- Myelosuppression
- Alopecia
- Hypersensitivity
What is the mechanism and clinical uses of Cisplatin and Carboplatin?
- Cross-links DNA
- Used for testicular, bladder, ovary, and lung carcinomas
What are the toxic side effects of Cisplatin and Carboplatin?
- Nephrotoxicity
- Acoustic nerve damage
- Prevents nephrotoxicity with amifostine (free radical scavenger) and chloride diuresis
What is the mechanism and clinical uses of Etoposide and Teniposide?
- Inhibits topoisomerase II → ↑ DNA degradation
- Used for solid tumors (particularly testicular and small cell lung cancer), leukemias, and lymphomas
What are the toxic side effects of Etoposide and Teniposide?
- Myelosuppression
- GI irritation
- Alopecia
What is the mechanism and clinical uses of Irinotecan and Topotecan?
- Inhibits topoisomerase I and prevents DNA unwinding and replication
- Used for colon cancer (irinotecan)
- Used for ovarian and small cell lung cancers (topotecan)
What are the toxic side effects of Irinotecan and Topotecan?
- Severe myelosuppression
- Diarrhea
What is the mechanism and clinical uses of Hydroxyurea?
- Inhibits ribonucleotide reductase → ↓ DNA synthesis (S-phase specific)
- Used for melanoma, CML, sickle cell disease (↑ HbF)
What are the toxic side effects of Hydroxyurea?
- Bone marrow suppression
- GI upset
What is the mechanism of Prednisone and Prednisolone?
- May trigger apoptosis
- May even work on non-dividing cells
What are the clinical uses of Prednisone and Prednisolone?
- Most commonly used glucocorticoids in cancer chemotherapy
- Used in CLL, non-Hodgkin lymphomas (part of combination chemotherapy regimen)
- Also used as immunosuppressants (eg, autoimmune disease)
What are the toxic side effects of Prednisone and Prednisolone?
Cushing like symptoms:
- Weight gain
- Central obesity
- Muscle breakdown
- Cataracts
- Acne
- Osteoporosis
- Hypertension
- Peptic ulcers
- Hyperglycemia
- Psychosis
What is the mechanism of Tamoxifen and Raloxifene?
- Selective estrogen receptor modulator (SERMs)
- Receptor antagonists in breast and agonists in bone
- Blocks the binding of estrogen to ER (+) cells
What are the clinical uses of Tamoxifen and Raloxifene?
- Breast cancer treatment (tamoxifen only) and prevention
- Raloxifene also useful to prevents osteoporosis
What are the toxic side effects of Tamoxifen and Raloxifene?
- Tamoxifen: partial agonist in endometrium, which ↑ the risk of endometrial cancer, "hot flashes"

- Raloxifene: no ↑ in endometrial carcinoma because it is an endometrial antagonist
What is the mechanism of Trastuzumab (Herceptin)?
- Monoclonal antibody against HER-2 (c-erbB2), a tyrosine kinase receptor
- Helps kill breast cancer cells that overexpress HER-2, through inhibition of HER2-initiated cellular signaling and antibody-dependent cytotoxicity
What are the clinical uses of Trastuzumab (Herceptin)?
HER-2 positive breast cancer and gastric cancer (Tras2zumab)
What are the toxic side effects of Trastuzumab (Herceptin)?
- Cardiotoxicity ("HEART"ceptin damages the heart)
What is the mechanism of Imatinib (Gleevec)?
Tyrosine kinase inhibitor of bcr-abl (Philadelphia chromosome fusion gene in CML) and c-Kit (common in GI stromal tumors)
What are the clinical uses of Imatinib (Gleevec)?
- CML
- GI stromal tumors
What are the toxic side effects of Imatinib (Gleevec)?
Fluid retention
What is the mechanism of Rituximab?
Monoclonal Ab against CD20, which is found on most B-cell neoplasms
What are the clinical uses of Rituximab?
- Non-Hodgkin Lymphoma (CD20 positive cells)
- Rheumatoid arthritis (with MTX)
- Idiopathic Thrombocytopenic Purpura (ITP)
What are the toxic side effects of Rituximab?
Increased risk of progressive multifocal leukoencephalopathy
What is the mechanism and use of Vemurafenib?
- Small molecule inhibitor of forms of the B-Raf kinase with the V600E mutation
- Used for metastatic melanoma
What is the mechanism and clinical uses of Bevacizumab?
- Monoclonal antibody against VEGF, inhibits angiogenesis
- Used for solid tumors (colorectal cancer, renal cell carcinoma)
What are the toxic side effects of Bevacizumab?
Hemorrhage and impaired wound healing
Which chemo drug(s) cause(s) acoustic nerve damage?
Cisplatin / Carboplatin

(and nephrotoxicity)
Cisplatin / Carboplatin

(and nephrotoxicity)
Which chemo drug(s) cause(s) peripheral neuropathy?
Vincristine
Vincristine
Which chemo drug(s) cause(s) pulmonary fibrosis?
- Bleomycin
- Busulfan
- Bleomycin
- Busulfan
Which chemo drug(s) cause(s) cardiotoxicity?
- Doxorubicin
- Trastuzumab
- Doxorubicin
- Trastuzumab
Which chemo drug(s) cause(s) nephrotoxicity?
Cisplatin and Carbolatin (and acoustic nerve damage)
Cisplatin and Carbolatin (and acoustic nerve damage)
Which chemo drug(s) cause(s) hemorrhagic cystitis?
Cyclophosphamide
Cyclophosphamide
Which chemo drug(s) cause(s) myelosuppression?
- 5-FU
- 6-MP
- Methotrexate
- 5-FU
- 6-MP
- Methotrexate