• 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

1/69

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;

69 Cards in this Set

  • Front
  • Back
What is the mechanism of action of Cetuximab and what are some of its toxicities?
MOA: binds to EGFR (ErbB-1/HER1), and in effect competitively inhibits EGF/TGF-α binding, thus blocking signaling for tumor cell growth, angiogenesis, and metastasis; Toxicities: Acneform rash (like Erlotinib), infusion reactions
What is the difference between Filgrastim and Pegfilgrastim and what drug interactions are associated with them?
Pegfilgrastim is pegylated, thus substantially increasing its half-life (since it cannot be filtered out at the glomerulus); Both drugs should not be used concomitantly with cycle-specific chemotherapy (e.g. with 5-FU) since it can worsen myelosuppression
What is the mechanism of action of Sargramostim and what are its toxicities?
MOA: since it is a GM-CSF, it has the same MOA as Filgrastim; Toxicities: also similar to Filgrastim, but includes myalgias
What is Oprelvekin and what is its mechanism of action?
IL-11; MOA: stimulates multiple stages of megakaryopoiesis and thrombopoiesis
What two drugs are interleukins and how do they differ?
Aldesleukin (IL-2): stimulates the immune system, severe toxicities associated with it (e.g. hypotension), only used as a last resort; Denileukin (fusion protein of diphtheria toxin fragments and IL-2): targets delivery of toxin to cells expressing an IL-2 receptor, used to destroy cells in some leukemias/lymphomas
What is Alpha interferon used for and what toxicities are associated with it?
Leukemias, lymphomas, melanomas, myelomas – toxicities: flu-like symptoms, n/v, weight loss
What are the biologic effects of interferons?
Antiproliferative; Immunomodulatory; Differentiation (monocytes --> macrophages); Antigen expression (increased Ig expression on lymphocytes); Oncogene expression (reduced)
What is the primary site of metabolism for recombinant drugs and why?
Kidney – since they are small, the drugs can leak through the glomerulus; however, when they are reabsorbed in the nephron, they are degraded by local proteases
How does the effectiveness of treatment compare when looking at biologic response modifiers, between continuous SC infusion and bolus IV?
Continuous SC is far more effective than bolus IV
What are the nonspecific immunostimulants?
BCG; Levamisole
What problems are associated with MoAb use?
Cross-reactivity with normal tissues; Antigenic modulation (receptor internalization); Development of anti-mouse antibodies by patient (can only give one dose); Tumor heterogeneity in antigen expression; Tumor lacks adequate blood supply; Lack of direct toxicity; Lack of host accessory effectors
What is the nomenclature used for MoAb?
Common suffix is “mab”; Antibody source: u = human, o = murine; xi = chimeric
What are compounds called that modify the host’s biologic response to tumor cells?
Biologic Response Modifiers
What is particular about the half-life of monoclonal antibodies (MoAb) and what is the most important organ for clearance?
Half-life is long; Liver and reticuloendothelial system are most important organs for clearance
What two drugs are interleukins and how do they differ?
Aldesleukin (IL-2): stimulates the immune system, severe toxicities associated with it (e.g. hypotension), only used as a last resort; Denileukin (fusion protein of diphtheria toxin fragments and IL-2): targets delivery of toxin to cells expressing an IL-2 receptor, used to destroy cells in some leukemias/lymphomas
What is Alpha interferon used for and what toxicities are associated with it?
Leukemias, lymphomas, melanomas, myelomas – toxicities: flu-like symptoms, n/v, weight loss
What are the biologic effects of interferons?
Antiproliferative; Immunomodulatory; Differentiation (monocytes --> macrophages); Antigen expression (increased Ig expression on lymphocytes); Oncogene expression (reduced)
What is the primary site of metabolism for recombinant drugs and why?
Kidney – since they are small, the drugs can leak through the glomerulus; however, when they are reabsorbed in the nephron, they are degraded by local proteases
How does the effectiveness of treatment compare when looking at biologic response modifiers, between continuous SC infusion and bolus IV?
Continuous SC is far more effective than bolus IV
What are the nonspecific immunostimulants?
BCG; Levamisole
What is the mechanism of action of Trastuzumab and what are some of its toxicities?
MOA: binds to HER2 receptor leading to cell cycle arrest, and acts as a mediator of antibody-dependent cell-mediated cytotoxicity via natural killer cells and monocytes; Toxicity: hypersensitivity, CHF (especially in combination with Doxorubicin – signs may be delayed)
What is the mechanism of action of Filgrastim and Pegfilgrastim and what are some of their toxicities?
MOA: they are G-CSF and thus stimulate multiple stages of neutrophil production as well as increase the activity of individual neutrophils; Toxicity: bone pain, hyperuricemia
What are the established roles of neutrophil-related growth factors?
Prevention (not treatment) of neutropenic fever following “standard-dose” chemotherapy; Acceleration of hematopoietic recovery following “high-dose” chemotherapy; Generation of autologous peripheral blood progenitor cells; Treatment of chronic neutropenic states
What are the indications for EPO?
Anemia secondary to renal failure, malignancy, or chemotherapy
Besides causing hematopoiesis, what negative side-effects are associated with EPO?
Cancer-related angiogenesis (via VEGF), promotion of cancer cell migration; also hypertension, thrombotic events, and seizures
What is the mechanism of action of Tositumomab?
MOA: it is murine IgG conjugated with Iodine131, which binds to CD20 on B-cells, leading to complement-dependent toxicity, antibody-dependent cellular toxicity, and direct toxicity of radioactivity (to the bound cell and adjacent cells)
What is the mechanism of action of Gemtuzumab and what are some of its toxicities?
MOA: it is humanized IgG conjugated with the toxin calicheamicin, which becomes internalized after CD33 binding on leukemic cells (causes dsDNA breaks); Toxicities: myelosuppression
What is the mechanism of action of Bevacizumab and what are some of its toxicities?
MOA: binds to VEGF, thus inhibiting tumor-related angiogenesis; Toxicities: hypertension, proteinuria, problematic wound healing (since it inhibits angiogenesis)
What is the mechanism of action of Rituximab and what are some of its toxicities?
MOA: binds to CD20 on B-cells (CD20 regulates cell cycle), and causes B cell lysis; Toxicities: Infusion reactions, risk of acute death – Rituximab is used for B-cell non-Hodgkin’s lymphoma
How are Paclitaxel and Docetaxel metabolized?
By CYP3A4 and 28C (Paclitaxel only) – they are easily cleared from the CNS thus reducing the effects there; this can lead to increased relapse rates in the brain
What is the mechanism of action of Paclitaxel and Docetaxel?
Promotes microtubule assembly and then inhibits depolymerization, thus stabilizing the microtubules in their non-functional state
What important toxicities and drug interactions are associated with Irinotecan?
Myelosuppression; n/v; Diarrhea: early (treated with atropine), late (treated with loperimide) – Drug interactions: CYP3A4 inducers/inhibitors
What is the mechanism of action of Vincristine and what is its half-life?
Binds tubulin, thus inhibiting microtubule formation (i.e. M phase specific = mitotic arrest) – it’s half-life is long (>20 hrs)
What are the mitotic inhibitors used in chemotherapy?
Vincristine, Vinblastine, Vinorelbine, Paclitaxel, Docetaxel, Ixabepilone
What is the mechanism of action of Temsirolimus?
It binds to FKBP-12 and inhibits mTOR (like Sirolimus) --> this leads to growth arrest
What is the mechanism of action of Tamoxifen and what are its important toxicities?
Competitive estrogen antagonist (mostly G2 specific) – Toxicities: menopausal symptoms, thromboembolism, flare reaction, ocular/endometrial/uterine cancer
What are the estrogen receptor modulators used in chemotherapy?
Fulvestrant, Tamoxifen, Toremifene
What are the two subclassifications of anti-estrogen therapies and what characterizes each?
Receptor modulators: used pre- or post-menopausal, toxicities include blood clots / uterine cancer;
Aromatase inhibitors: used post-menopausal, toxicities include osteoporosis / fractures
What pharmacological class is Vorinostat and what is its mechanism of action?
Histone deacetylase inhibitor – MOA: inhibits HDAC1,2,3,6 which catalyze removal of acetyl groups from histones and transcription factors --> cell cycle arrest and/or apoptosis
What pharmacological class is Bortezomib and what is its mechanism of action?
It is a proteasome inhibitor – MOA: reversible inhibitor of ubiquitin proteasome pathway
What is the pharmacologic class of Arsenic Trioxide and Tretinoin
They are differentiating agents
What two signal transduction inhibitors are competitive inhibitors of ATP on the tyrosine kinase Bcr-Abl and how do they differ?
Imatinib and Dasatinib – Dasatinib has activity in Imatinib-resistant cell lines
Which signal transduction inhibitors are used for renal cell cancer and what is characteristic of them?
Sorafenib and Sunitinib – both are multi-kinase inhibitors causing inhibition of proliferation and antiangiogenesis; since they affect multiple tyrosine kinase inhibitors, they have more extensive toxicities associated with them
Which signal transduction inhibitors are associated with the EGFR/ErbB-1/ HER1 tyrosine kinases and what differentiates between them?
Erlotinib and Lapatinib – Erlotinib: causes cell cycle arrest and inhibition of angiogenesis, and is associated with a rash (no rash = no efficacy); Lapatinib has a dual action on ErbB-1 and ErbB-2, making it useful in HER-2 positive breast cancer
What is the mechanism of action of Leuprolide?
It is a GnRH agonist --> causes an initial surge of LH/FSH from the pituitary, temporarily increasing the levels of testosterone (~ 1 week) --> ultimately secretion of LH/FSH is decreased by continuous receptor stimulation – used to treat prostate cancer (also breast/ovarian cancer)
What important toxicities are associated with Leuprolide?
Hot flashes, reduced libido, impotence, gynecomastia
What is particular about the pharmacokinetics of Tamoxifen?
It is metabolized by several CYP450 enzymes (3A, 2C, 2D6); however, in order to achieve maximal efficacy, Tamoxifen must be metabolized by CYP2D6 to highly active metabolites; since ~10% of Caucasians do not have CYP2D6, this can cause problems; Inhibitors of CYP2D6 should also be avoided
What are the androgen receptor antagonists, used in chemotherapy and for what specific cancer are they important treatment options?
Bicalutamide, Flutamide – used in prostate cancer
What is the mechanism of action of Anastrozole and what toxicities are associated with it?
MOA: blocks aromatase conversion of androgens to estrogens in peripheral tissues; Toxicities: menopausal symptoms, vaginal dryness/bleeding, GI effects, osteoporosis
What are the aromatase inhibitors used in chemotherapy?
Aminoglutethimide, Anastrozole, Letrozole, Exemestane
What problems are associated with MoAb use?
Cross-reactivity with normal tissues; Antigenic modulation (receptor internalization); Development of anti-mouse antibodies by patient (can only give one dose); Tumor heterogeneity in antigen expression; Tumor lacks adequate blood supply; Lack of direct toxicity; Lack of host accessory effectors
What is the nomenclature used for MoAb?
Common suffix is “mab”; Antibody source: u = human, o = murine; xi = chimeric
What are compounds called that modify the host’s biologic response to tumor cells?
Biologic Response Modifiers
What is particular about the half-life of monoclonal antibodies (MoAb) and what is the most important organ for clearance?
Half-life is long; Liver and reticuloendothelial system are most important organs for clearance
What important pharmacokinetics are associated with Doxorubicin?
It has a large volume of distribution; Elimination is via bile, so bilirubin levels must be checked
What is the most common antitumor antibiotic and what is its mechanism of action?
Doxorubicin; MOA: free radical formation, topoisomerase II interaction (!), DNA intercalation
What are the mechanisms of action of 5-Fluorouracil (5-FU)?
Inhibition of thymidylate synthase (= decreased thymidine); Incorporation into RNA and DNA leading to apoptosis – some S phase specificity
What are important toxicities associated with 5-FU and what drug is used in association with 5-FU?
Toxicities (continuous infusion): mucositis, diarrhea, hand-and-foot syndrome (paresthesias, redness --> pain --> peeling --> resolution); Toxicities (daily bolus): myelosuppression; Drug interactions: Leucovorin (folinic acid) stabilizes/prolongs binding of 5-FU to thymidylate synthase
What compound causes deactivation of 5-FU?
Dihydropyrimidine dehydrogenase (DPD); 5-FU has a short half-life (min.) – DPD deficiency often results in lethal toxicity
What are the antitumor antibiotics?
Doxorubicin, Epirubicin, Idarubicin, Mitoxantrone, Actinomycin D, Bleomycin
What is the active metabolite of Irinotecan and how is it excreted?
Irinotecan is converted by carboxyesterases to SN-38 (100-1000x more active); SN-38 is glucoronidated and excreted in the bile; CYP3A4 also metabolizes Irinotecan, but forms inactive metabolites
What is the mechanism of action of Irinotecan?
Stabilization of a ternary complex between topoisomerase I and dsDNA, inducing single strand breaks, thus preventing cells from entering mitosis (G2 arrest)
What are the topoisomerase I inhibitors?
Irinotecan, Topotecan
What toxicities are found with Doxorubicin?
Cardiac problems: acute arrhythmias and chronic CHF (keep lifetime dose <550 mg/m2); Vesicant; Radiosensitizer; May make patient’s fluids/tissues red
What important toxicities are associated with Vincristine?
Neuropathies: decreased DTR --> paresthesias --> autonomic dysfunction; SIADH; Photosensitivity; NOT myelosuppression (sets it apart) – If accidentally given intrathecal, it is almost always lethal
What is characteristic of the metabolism of signal transduction inhibitors?
They are all metabolized by CYP3A4
What are the signal transduction inhibitors used in chemotherapy?
Dasatinib, Erlotinib, Imatinib, Lapatinib, Sorafenib, Sunitinib, Temsirolimus
What sets Ixabepilone apart from Paclitaxel and Docetaxel?
Although it has a similar MOA as Paclitaxel/Docetaxel, it is less susceptible to MDR/P-gp and may therefore work on vinca/taxane/anthracycline resistant cells
What important toxicities are associated with Paclitaxel and Docetaxel, respectively?
Paclitaxel: acute anaphylaxis, myelosuppression, peripheral neuropathy; Docetaxel: myelosuppression, peripheral neuropathy, fluid retention