This site is 100% ad supported. Please add an exception to adblock for this site.

Chemotherapy II

Terms

undefined, object
copy deck
What are cell cycle specific drugs that work at S phase?
Purine antimetabolites Antifolates Pyrimidine antimetabolites Ribonucleotide reductase inhibitors DNA polymerase inhibitors
Is a topoisomerase II inhibitor a cell cycle specific drug?
yes
how does Epipodophyllotoxins work?
topoisomerase inhibition
which drugs work at m phase? how do they work?
Vinca alkaloids Taxanes block mitotic spindle
is a topoisomerase I inhibitor a cell cycle specific drug?
no
what does Camptothecins do to inhibit cell cycle?
topoisomerase I inhibition
is a dna crosslinker cell cycle specific chemotherapy?
no
how do Alkylating Agents Platinums work?
dna crosslinker
what type of chemotherapy is nitrogen mustards? name some examples of nitrogen mustards
Alkylating Agents Nitrogen mustards Cyclophosphamide (Cytoxan) Mechlorethamine (Mustargen) Mephalan (Alkeran) Chlorambucil (Leukeran)
what type of chemo is a Nitrosureas? name some examples
alkylating agent Carmustine (BiCNU) Lomustine (CeeNU) Streptozocin (Zanosar)
What type of chemotherapy agent is an ethylinimne? give an example of one type of drug
Thiotepa (Thioplex)
What type of chemotherapy is an alkyl sulfonate? give an example
Alkyl sulfonates Busulfan (Myleran)
What type of chemotherapy is triazine? give an example
Triazines Dacarbazine (DTIC-Dome) Temozolamide (Temodal)
What is the mechanism of action of alkylating agents?
Transfers alkyl groups to a nucleophile (sulfhydryl, amino, hydroxyl, carboxyl, and phosphate groups on proteins or DNA) Bifunctional crosslinks to DNA that interfere with base pairing DNA strand breaks G2 cell cycle arrest
What is the dose limiting toxicity of an alkylating agent?
Myelosuppression
What type of chemo is Cisplatin? and what it its dose limiting toxicity? mechanism of action?
Platinum, nephrotoxicicty broad action agains solid tumors, esp squamous cell carcinoma
What type of chemo is Carboplatin (Paraplatin)? dose lmiting toxicity and mechanism of action?
Platinum myelosuppression dose limiting mechanism of action is a broad activity agains solid tumors
what is the dose limiting toxicity of Oxaliplatin and what is its mechanism of action?
2nd string drug for colorectal cancer and it has neurotoxicity
what is the general mechanism of action for platinums?
Crosslinks N7 guanine on DNA - similar to alkylating agents
What type of chemotherapy is folic acid? and give some examples
Folic acid is an antimetabolite antagonists Methotrexate (Amethopterin) Ralitrexed (Tomudex) Premetrexed (Alimta)
What type of drug is a purine analogue and give some examples
Purine analogues are antimetabolites Mercaptopurine (Purinethol) 6-thioguanine (Thioguanine) Fludarabine (Fludara) Cladribine (Leustatin)
what type of chemotherapy is a pyrimidine analogue?
Pyrimidine analogues are antimetabolites Fluorouracil (Adrucil) Capecitibine (Xeloda) Cytarabine (Cytosar-U) Gemcitabine (Gemzar)
what is the dose limiting toxicity for a folic acid chemotherapy?
myelosuppression, gi and oral ulceration
what is folic acid's mechanism of action
Mechanisms of Action Folic acid binds the catalytic domain on dihydrofolate reductase (DHFR) to generate an essential cofactor required for DNA, RNA and protein synthesis. Methotrexate competes with folic acid for the active site of DHFR Premetrexed also inhibits TS.
what is the clinical indication for some folic acid treatments?
Methotrexate:Burkitt’s lymphoma, acute lymphoblastic leukemia, trophoblastic choriocarcinoma and metastatic breast, head and neck, ovarian, and cervical carcinomas Ralitrexed: Colon cancer Premetrexed (multi-targeted anti-folate): Mesothelioma, lung cancer in combo with cisplatin
What are the indications for Pyrimidine antagonists
Fluorouracil: colon cancer and other solid tumors Capecitabine: breast cancer in combination with taxanes Cytarabine: acute myelogenous leukemia Gemcitabine: pancreatic cancer Clorfarabine: pediatric AML
What is the dose limiting toxicity for pyrimidine antagonist?
Myelosuppression, oral and GI ulceration
what is the mechanism of action for pyrimidine antagonist?
Inhibition of DNA synthesis by interfering with enzymes involved in pyrimidine synthesis (5-FU: thymidylate synthetase, GEM: ribonucleotide reductase, Cytarabine: DNA polymerase) Cell cycle arrest in G1 phase
folic acid blocks what in the dna synthesis pathway
dhfr
pyrimidine blocks what in dna synthesis
Thymidylate synthetase
Purine antagonists are what type of chemotherapy?
antimetabolites
what are the purine antagonist indications?
Mercaptopurine: pediatric acute leukemia Thioguanine: adult acute leukemia in combination with cytarabine Fludarabine: Non-Hodgkin’s lymphoma and chronic lymphocytic leukemia Cladribine: Hairy cell leukemia, CLL, and low grade NHL
what is the dose limiting toxicity for purine antagonists?
Myelosuppression
what is the mechanism of action for purine antagonist?
Inhibits enzymes involve in the synthesis of nucleotides from purine bases conversion to inhibit DNA and RNA synthesis Fludarabine inhibits DNA polymerase and ribonucleotide reductase
what are some types of plant alkaloids?
microtubule disrupters, microtubule stabilizers, Topoisomerase inhibitors, Type 2 inhibitors
what are some examples of type two inhibitors
Epipodophyllotoxins Etoposide (VePesid) Teniposide (Vumon)
what are some examples of topoisomerase inhibitors?
Topoisomerase inhibitors Type 1 inhibitors Camptothecins Irinotecan (Camptosar) Topotecan (Hycamptin) Type 2 inhibitors
what are some examples of microtubule stabilizers
Microtubule stabilizers Taxanes Paclitaxel (Taxol) Docetacel (Taxotere)
what are some examples of microtubule disrupters
Microtubule disrupters Vinca Alkaloids Vinblastine (Velban) Vincristine (Vincasar) Vinorelbine (Navelbine)
what are some of the dose limiting toxcicities of vinca alkaloids?
Vinblastine: myelosuppression Vincristine: neurotoxicity (bone marrow sparing) Vinorelbine: myelosuppression
what are some of the indications for vinca alkaloids
Vincristine - Same activity as vinblastine, less myelotoxicity Vinorelbine – Additional activity against non-small cell lung cancer
what is the mecanism of action for vinca alkaloids?
Inhibition of microtubule polymerization, disruption of mitotic spindle and chromosome separation, M phase cell cycle arrest
Taxanes indications?
Indications – broad range of solid tumors Paclitaxel – breast, ovarian. Lung, bladder, prostate, gastric, esophageal cancers Docetaxel – semisynthetic derivative with similar activity as paclitaxel
what is the toxicity of taxanes?
Toxicity Myelosuppression
what is the mechanism of action for taxanes?
Mechanism of action Stabilizes microtubule assembly, inhibition of mitosis and cell division (M phase block)
what are the indications of campothecins
Indications (2nd line therapy – used for drug resistant cancers) Irinotecan (CPT11) – Colorectal cancer (5-FU - 1st line therapy; Oxaliplatin - currently 2nd line) Topotecan – Lung and ovarian cancer (taxol or platininum-based therapy - 1st line therapy)
what is the dose limiting of campothecin?
Myelosuppression and diarrhea (CPT11)
what is the mech of action of campothecin?
Inhibition of topoisomerase I – enzyme that cuts one strand of the DNA to reanneal the cut strand – necessary for unwinding of DNA during replication Campthothecins keep DNA wound into chromosomes so the cell cannot make proteins needed for replication. Cell cycle independent inhibitor SN38 – active metabolite of CPT11
what are the indications for Epipodophyllotoxins
Etoposide – testicular, lung, gastric and hematological malignancies Teniposide – acute lymphoplastic leukemia in children
dose limiting toxicity of Epipodophyllotoxins
Dose limiting toxicity Nausea, vomiting, myelosuppression, alopecia
what are the mechanisms of action of Epipodophyllotoxins
Topoisomerase II inhibition – enzyme that cuts both strands of DNA, passes an unbroken strand of DNA through to reanneal the cut strands.
what are some Antitumor antibiotics
Anthracyclines Daunorubicin (Daunamycin and Mitoxantrone (Mutamycin) and Dactinomycin (Actinomycin D) Mitomycin (Mutamycin) Bleomycin (Blenoxane) Mithramycin (Mithracin)
Anthracyclines indications?
Doxorubicin and epirubicin – solid tumors Daunamycin and idarubicin – acute leukemias Dose limiting toxicity
anthracyclines mech of action
Inhibition of topoisomerase II, DNA intercalation, alteration in membrane fluidity and ion transport, and generation of free radicals Mechanisms of resistance Increased drug efflux by P-gp and MRPs, increased glutathione conjugation, topoisomerase 2 mutations, DNA repair
mitoxantrone mech of action, clinical indication and does limiting toxicity
Indications Hormone refractory prostate cancer, non-Hodgkin’s lymphoma, breast cancer, acute myeloid leukemia Dose limiting toxicity Myelosuppression – less cardiotoxic compared to doxorubicin Mechanism of action DNA strand breakage and inhibition of DNA or RNA synthesis
Bleomycin indications? this drug was in first aid ...
Squamous cell carcinoma of the cervix, head and neck, skin and vulva Hodkin’s and non-Hodkin’s lymphoma Ovarian and testicular tumors
bleomycin mech of action
Mechanism of action Small peptide that binds DNA to cause DNA strand breaks following free radical attack; induces G2 cell cycle arrest
bleomycin dose limiting toxicity
Dose limiting toxicity Pulmonary toxicity
Cytokines: what is an example?
Interferon-alpha (Intron A)
what is intro-a's indication
Indications Hairy cell leukemia, Kaposi sarcoma, squamous cell carcinomas
what is intron a's mech of action?
Mechanism of action Stimulate tumoricidal macrophages Direct cytotoxicity to cancer cells
cytokine: what is another example?
Interleuken-2 (Proleuken)
what is interleuken-2 / proleuken's mech of action? and indications?
Indications Malignant melanoma, squamous cell carcinomas Mechanism of action Increase activity of natural killer cells

Deck Info

63

swimaline

permalink