• 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

1/54

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;

54 Cards in this Set

  • Front
  • Back
What are the treatment strategies for cancer?
Antineoplastics
Anti-angiogenic drugs
Vaccines
Immunomodulators
What are the rules of Skipper's model of cancer growth? How are they applied to therapy? What types does this model work for?
Doubling time of cancer cells is constant and number of cells killed during treatment is constant.
Repeated dosing with therapy agents can cure cancer. Works for leukemias
What are the rules of the Gompertzian growth model of cancer? How are they applied to therapy?
Solid tumors consist of proliferating and quiescent cells. The larger the tumor, the lower the percentage of proliferating cells.
A tumor is more likely to be cured when it is small. Increasing drug frequency is more effective than increasing dose.
What are the phases of the cell cycle?
M- phase: mitosis
G1- phase: Cell growth
S phase: DNA replication
G2- phase: Preparation for mitoss
What drugs block the G1 phase? S phase? G2 phase? M phase?
G1 blocked by cycline dependent kinases.
S phase blocked by DNA synthesis inhibitors (antimetabolites)
G2 phase blocked by microtubule and topoisomerase inhibitors
M phase blocked by microtubule inhibitors
What are the mechanisms of drug resistance (7)?
Decreased uptake
Increased efflux
Modification of targeted enzymes
Increased drug metabolism and detoxification
Modification of extracellular proteins
Increased DNA repair
Decreased apoptosis
What drugs gain resistance due to decreased uptake?
Antimetabolites
What proteins are involved in increased efflux as a mechanism of drug resistance?
P-glycoprotein
Multidrug resistance proteins
What drugs gain resistance due to increased efflux?
Hydrophobic compounds- anthracyclines, vinca alkaloids, taxanes
What molecule is increased to cause increased drug metabolism in drug resistance?
Glutathione
What drugs gain resistance by increased DNA repair mechanisms?
Alkylating agents
What molecule causes decreased apoptosis in drug resistance?
p53
What are the principles of the Goldi-Coldman model of combination chemotherapy?
Different working mechanisms
Non-overlapping toxicities
Non-cross-resistance mechanisms
What are some of the common side effects of antineoplastics?
Myelosuppression
Nausea/ vomiting
GI tract injury
Alopecia
Infertility
Cardiotoxicity
Neurotoxicity
Increased risk of secondary cancer
What are the results of myelosuppression associated with chemotherapy?
Higher infection rate
Increased bleeding
Anemia
Why does nausea and vomiting occur with chemotherapy? What drugs are used to treat it?
Chemoreceptor trigger zone and vomiting center express receptors that are sensitive to antineoplastics. Also due to GI toxicity.
Metoclopramide- dopamine receptor antagonist
Ondansetron and granisetron- Serotonin receptor antagonist
What is tumor lysis syndrome? What are symptoms?
In large or proliferating tumors, rapid cell death after therapy can cause massive release of cellular metabolites.
Hyperuricemia, hyperkalemia, hyperphospatemia, secondary hypocalcemia result.
Can result in organ failure and death.
Why is angiogenesis a target for cancer treatment?
Tumors induce angiogenesis by secreting VEGF, FGF, PDGF due to increased need for oxygen and nutrients.
Preventing angiogenesis can limit tumor growth.
In what ways to antiangiogenic drugs prevent angiogenesis?
Block matrix breakdown
Direct inhibition of endothelial cell activation
Blockade of angiogenesis activators
Inhibition of endothelial-specific integrin/ survival signal
What is active cellular immunotherapy? How is it done? Why is it not done often?
Approach to treat cancer that uses patient's antigen-presenting cells to re-engage the patient's T cells to attack specific cancer.
APCs are removed from the patient, presented to specific antigens and readministered to the patient.
ACI is very expensive and not shown to increase survival.
What are the classes of antineoplastic drugs?
Alyklating agents
Antibiotics
Antimetabolites
Plant alkaloids
Hormonal agents
Antibodies
What is the mechanism for alkylating cancer drugs? What are the side effects?
Form covalent alkyl bonds with DNA bases, causing cross linking. This inhibits DNA replication.
Myelosuppression, and future cancer can occur.
What are types of alkylating cancer drugs?
Nitrogen mustards- mechlorethamine, cyclophosphamide
Nitrosoureas- carmustine, streptozocin
Cisplatin
What was the first antineoplastic developed? What are side effects?
Mechlorethamine
Does not require bioactivation, so causes extravasation reaction and myelosuppression.
No longer used.
What is the mechanism of cyclophophamide? What is the toxicity? What drug is given to prevent side effects?
DNA cross-linking, requires bioactivation.
Bladder and renal toxicity, myocarditis, congestive heart failure. Treat renal toxicity with Mesna.
What is the mechanism of nitrosoureas? What are they used for?
Alkylation of DNA and RNA, does not require bioactivation.
Crosses the BBB- brain tumors.
What is the mechanism of Carmustine, Lomustine and Semustine? What is the indication?
DNA cross-linking (nitrosourea)
Primary brain tumors
What is the mechanism of Streptozocin? What are the side effects?
DNA alkylation, but no cross linking
Mild myelosuppression, renal toxicity, diabetes, extravasation injury.
What is the mechanism for Cisplatin and Carboplatin? What are the acute, subacute and chronic side effects?
DNA cross linking
Acute: hypersensitivity, nausea/ vomiting
Subacute: renal and gonadal toxicity, myelosuppression
Chronic: neurotoxicity, secondary tumors
What are the two major mechanisms for antibiotics against cancer?
DNA cross-linking
Inhibition of DNA topoisomerase
What is the mechanism of Actinomycin D? What are the side effects?
DNA cross linking; inhibition of topoisomerase II.
Myelosuppression, nausea/ vomiting, photosensitivity, radiosensitizer.
What is the mechanism for Mitomycin? What are the side effects?
DNA cross linking and free radical production.
Delayed myelosuppression, renal toxicity, lung fibrosis.
What is the mechanism for Bleomycin? What are the side effects?
DNA cross linking and free radical formation.
Fatal acute allergic reaction, skin and lung toxicity, pulmonary fibrosis, lack of myelosuppression.
What is the mechanism for Anthracycline? What are the side effects?
Inhibition of topoisomerase II and DNA intercalation.
Myelosuppression, severe extravasation injury, serious cardiac toxicity.
What is the mechanism of anti-metabolites? What phase of the cell cycle do they work?
Mimic folic acid, pyrimidine and purine to block enzymatic reactions.
Block the S phase of the cell cycle
What is the mechanism of methotrexate? What are the side effects? What drug is given to alleviate these?
Folic acid analogue that inhibits dihydrofolate reductase.
Myelosuppression, renal damage, liver toxicity.
Leucovorin (folinic acid) is used to bypass the folic acid cycle and reduce toxicity.
What is the mechanism of 5-fluorouracil? What are the side effects? What deficiency is associated with increased toxicity?
Pyrimidine analogue that inhibits thymidylate synthase.
Myelosuppression, long term CNS effects, hepatotoxicity, cardiotoxicity, photosesitivity.
Dehydropyrimidine dehydrogenase deficiency.
What is the mechanism of mercaptopurine? What are the side effects? What deficiency is associated with increased toxicity?
Purine analogue. Monophosphate inhibits purine synthesis enzymes. Triphosphate causes strand breakage.
Myelosuppression, hepatotoxicity.
Thiopurine methyltransferase
What drugs affect microtubules? What do they do?
Vinca alkaloids inhibit microtubule polymerization
Taxanes inhibit microtubule depolymerization
What are the side effects of vinblastine? Vincristine?
Vinblastine- myelosuppression
Vincristine- neurologic toxicity, peripheral neuropathy.
What are the side effects of paclitaxel and docetaxel?
Myelosuppression, type I hypersensitivity, peripheral neuropathy
What is the mechanism of epidodophyllotoxins (etoposide and teniposide)? What are the side effects?
Inhibition of topoisomerase II
Myelosuppression, peripheral neuropathy, secondary tumors
What is the mechanism of camptothecins (topotecan and irinotecan)? What are the side effects? What deficiency is associated with increased side effects? What is the indication?
Inhibition of topoisomerase I
Haemorrhagic cystitis, myelosuppression, severe diarrhea.
Gilbert's disease
Colorectal cancer
What is the mechanism of prednisone? What are the long term complications?
Prevents immune cell proliferation, induces cell death, side effect relief. Mostly synergistic.
Adrenal atrophy- Cushings; immunosuppression, glucose intolerance, osteoporosis, edema, hypertension.
What is the mechanism of monoclonal antibodies? what are the side effects?
Monoclonal IgG antibodies specific to a certain antigen.
Complement mediated cytotoxicity, radioactive molecules deliver drugs, block specific receptor, bind to growth factors.
Allergic reaction.
What is the antigen for Rituximab? What is the indication?
CD20 found on B cells
B cell non-Hodgkin's lymphoma
What is the antigen for Trastuzumab? What are the side effects? What is the indication?
Her2/neu
Cardiotoxicity, hypersensitivity.
Metastatic breast cancer
What is the antigen for Cetuximab? What are the side effects? What is the indication?
EGFR
Interstitial lung disease, skin reactions.
Used with irinotecan in colorectal cancer
What is the antigen for Bevacizumab? What are the side effects?
VEGF
Hypertension, bleeding
What is the mechanism of Imatinib? What are the indications? What are the side effects?
Inhibitor of tyrosine kinase for PDGF, BCF-ABL and c-kit.
Chronic myeloid leukemia, GIST
Neutropenia, thrombocytopenia, hepatotoxicity, renal toxicity, muscle cramps, edema.
What is the mechanism of Sunitinib? What are the indications? What are the side effects?
Inhibitor of tyrosine kinase on PDGF, VEGF, c-kit, RTK.
Renal cell carcinoma, GIST
Hypertension, skin discoloration
What is the mechanism of Gefitinib? What are the indications? What are the side effects?
EGFR tyrosine kinase blocker
Non small cell lung cancer
Fatal interstitial lung disease, bleeding
What is the mechanism of Bortezomib? What is the indication? What are the side effects?
Inhibits the 26S proteasome complex.
Multiple myeloma
Asthenia, peripheral neuropathy, myelosuppression, shingles
What is the mechanism of Thalidomide? What is the indication? What are the side effects?
Inhibits TNF alpha
Multiple myeloma
Blood clotting, TERATOGENIC