• 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

1/75

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;

75 Cards in this Set

  • Front
  • Back
What is Gompertzian kinetics?
Tumors may outgrow their blood supply causing the cells to proliferate less, making them harder to kill
What is a one log kill?
90% of cells in the population are killed
What is a two log kill?
99% of the cells in a population are killed
If there are 10E12 cells in a population and a drug is used that kills 99% of the cells, how many will be left?
10E10 cells will remain because this would be a two log kill
The killing of cancer cells generally follows what kinetic pattern?
First-order kinetic pattern
What are some mechanisms that may allow a cell to become resistant to a particular chemotherapy?
gene amplification
increased production of P-glycoprotein-like efflux pumps
What are the basic principles of combination chemotherapy?
1) each drug must be individually active against the tumor type
2) the mechanisms of interaction must produce an optimum effect
3) drugs have different toxic effects
What is adjuvant chemotherapy?
Chemotherapy after surgery or radiation to improve the outcome by eliminating clinically unapparent tumor cells
What is neoadjuvant therapy?
Chemotherapy or radiation before surgery to shrink the tumor to make it more operable
What are the major divisions of alkylating agents?
1) bischloroethylamines
2) nitrosoureas
3) alkyl sulfonate
4) ethyleneimines
Name the bischloroethylamines. Of what broad class do they belong?
Bischolorethylamines are alkylating agents
1) mechlorethamine
2) melphalan
3) cyclophosphamide
4) chlorambucil
Name the nitrosoureas. Of what broad class do they belong?
They are alkylating agents
1) carmustine
2) lomustine
3) streptozocin
What is the alkyl sulfonate and to what broad class does it belong?
Busulfan is an alkyating agent
What is the ethyleneimine and to what broad class does it belong?
Thio-TEPA is an alkylating agent
What is the mechanism of action of alkylating agents?
They are activated to chemically reactive metabolites (spontaneously or by enzymes) that bind to bases in DNA
What are some common adverse affects to alkylating agents?
Myelosuppression- can be delayed
GI upset - nausea and vomiting
Reproductive effects - amenorrhea, decreased sperm production
Carcinogenicity
What are some possible mechanisms of resistance against alkylating agents?
1) repair of damaged DNA
2) decreased tumor uptake of drug
3) increased inactivation of drug
What drugs commonly causes hemorrhagic cystitis and how can this be overcome? To what class do these drugs belong?
Cyclophosphamide and Ifosfamide are bischloroethylamines (alkylating agents) that can cause hemorrhagic cystitis

This can be overcome by increased hydration or with the administration of the drug mesna
What bischoloroethylamine is commonly used as an effective immunosuppressant?
cyclophosphamide
What alkylating agents are used in the treatment of brain tumors because they cross the BBB?
The nitrosureas carmustine and lomustine
What are the non-classical alkylating agents?
1) procarbazine
2) dacarbazine
What are the toxicities associated with procarbazine?
1) MOA inhibition - tyramine induced hypertension
2) disulfaram-like effects
3) myelosuppression
What is the use of Dacarbazine?
Malignant melanoma
What is the mechanism of action of cisplatin?
It binds to DNA and inhibits DNA synthesis and function
What are the adverse side effects of cisplatin?
1) Nephrotoxicity - diminished by hydration and diuresis
2) HIGHLY EMETOGENIC
3) ototoxicity
4) myelosuppression
When administering cisplatin, how would you help reduce the chance of nephrotoxicity?
Make sure the patient was well hydrated and give diuretics
What are the drugs that work as antimetabolites?
1) methotrexate
2) 6-mercaptopurine
3) 6-thioguanine
4) 5-FU
5) cytarabine
6) gemcitabine
What is the mechanism of action of methotrexate?
It inhibits dihydrofolate reductase, preventing the formation of FH4 which is needed as a cofactor in the synthesis of thymidylate

Overall decrease in DNA, RNA , and protein synthesis

Polyglutamates are formed which also contributes to toxicity
What are the possible mechanisms of resistance against methotrexate?
increased synthesis of DHFR
mutated DHFR
decreased cellular uptake
increased efflux
What are some of the toxicities associated with methotrexate use?
1) myelosuppression
2) GI mucositis
3) hepatic with long term use (macrovesicular fatty change)
What are some of the non-cancer uses of methotrexate?
Rheumatoid arthritis
Psoriasis
What is leucovorin and for what is it used?
This is a form of folic acid that is administered as an antidote for excessive doses of methotrexate
What is leucovorin rescue?
This is the use of leucovorin as part of a high-dose methotrexate therapy to "rescue" from some of the adverse effects of methotrexate
What is the mechanism of action of pemtrexed?
It inhibits thymidylate synthetase and is converted into polyglutamates
What is given to supplement patients taking pemetrexed?
B12 and folic acid; they reduce toxicity but do not interfere with efficacy
What drugs are purine and pyrimadine analogs and how do they work?
1) 6-mercaptopurine
2) 6-thioguanine
3) 5-FU
4) cytarabine
5) gemcitabine
6) fludarabine phosphate
7) cladribine
They must be converted into active nucleotides and interfere with DNA synthesis/function
How is 6-mercaptopurine metabolized and how can this be affected?
It is metabolized to 6-thiouric acid by xanthine oxidase and can also be S-methlyated by thiopurine-S-methyltransferase

Some patients have low TPMT activity and these patients have increased risk of toxicity

Patients given Allopurinol to block xanthine oxidase must have their dose of 6-mercaptopurine decreased to avoid toxicity
Describe the metabolism of 6-thioguanine
It is methylated by TMPT and deaminated to 6-thioxanthine which happens before the xanthine oxidase step, so these patients don't require a dose change when taking allopurinol
What is the mechanism of action of fludarabine phosphate?
It is a nucleotide that inhibits DNA synthesis and repair as well as inhibition of ribonucleotide reductase; it induces apoptosis
What is the mechanism of action of 5-FU?
It inhibits thymidylate synthase which decreases the production of TMP and DNA synthesis; it can also be incorporated into DNA and RNA
What are the platinum containing drugs?
cisplatin
carboplatin
oxaliplatin
What is the mechanism of gemcitabine?
Ribonucleotide reductase inhibition
incorporation into DNA
inhibition of DNA synthesis and repair
What drug causes mucositis and damage to the oral mucosa and mucosa of the GI tract?
Methotrexate
What is the form of tetrahydrofolate given in patients treated with excess methotrexate?
leucovorin
What drugs inhibit thymidylate synthetase?
5-FU and pemetrexed
What are the antibiotic anticancer drugs?
1) Dactinomycin (Actinomycin D)
2) Doxorubacin
3) Daunorubicin
4) Bleomycin
5) Mitomycin C
What is the mechanism of action of dactinomycin?
It intercalates between base pairs of DNA and interferes with RNA synthesis
What are the mechanisms of action of doxorubicin and duanorubicin?
They intercalate between DNA bases and inhibit DNA and RNA synthesis; they also inhibit topoisomerase II and cause DNA strand breaks
What are the adverse reactions associated with doxorubicin?
1) Cardiotoxicity - cardiomyopathy and heart failure, also arrhythmia and myocarditis/peridcarditis
2) myelosuppression
3) nausea, stomatitis
4) hepatic dysfunction
What is the mechanism of action of bleomycin?
It binds to DNA and causes strand breaks; it also generates free radicals
What are the major toxicities associated with bleomycin?
1) pumlonary fibrosis
2) skin manifestations such as erythema and tenderness
3) allergic reactions
What drug can cause hemolytic uremic syndrome?
mitomycin C
Name the vinca alkaloids and their mechanism of action
The vinca alkaloids include vincristine and vinblastine

They are mitotic inhibitors by binding to the tubulin and inhibiting the polymerization to microtubules
Which vinca alkaloid causes less myelosuppression? Which causes less neurotoxicity?
Vincristine causes less myelosuppression and vinblastine causes less neurotoxicity
What is the mechanism of action of paclitaxel?
It binds to microtubules and promotes the formation of microtubules, which leads to an inhibition of mitosis (opposite of vinca alkaloids); the mitotic spindle cannot break down during anaphase
What are the toxicities associated with paclitaxel?
hypersensitivity reactions
myelosuppression
peripheral neuropathy
arrhythmia
What is the mechanism of action of etoposide?
It inhibits topoisomerase II and leads to DNA strand breaks
What is the mechanism of action of topotecan and irinotecan?
They inhibit topoisomerase I causing DNA damage
What enzyme is used to treat cancer and how does it work?
L-Asparaginase depletes asparagine in certain leukemic cells which cannot synthesize the amino acid, leading to decreased protein synthesis
What are the toxicities associated with L-asparaginase?
Allergic reactions, hepatotoxicity, clotting factor changes, neurotoxicity, pancreatitis, and hyperglycemia
What is the mechanism of action of imatinib?
It inhibits the Bcr-Abl tyrosine kinase expressed by CML cells by preventing the binding of ATP
What drug is used to treat CML?
imatinib
What is the mechanism of action of dasaitinib and nilotinib and when are they used?
They are used to treat imatinib resistant CML; they also inhibit the Bcr-Abl tyrosine kinase
Name the growth factor receptor inhibitors
1) cetuximab
2) panitumumab
3) Gefitinib
4) erlotinib
5) bevicizumab
6) sorafenib
7) sunitinib
What is the mechanism of action of cetuximab and panitumumab?
They are monoclonal antibodies that bind to epidermal growth factor receptor and inhibit downstream receptor signaling
What are the adverse effects of cetuximab and panitumumab?
infusion reactions
acneifrom rash
interstitial lung disease
What is the mechanism of action of gefitinib and erlotinib?
They inhibit the tyrosine kinase domain of EDGF, leading to the inhibition of receptor signaling
What are the adverse effects of gefitinib and erlotinib?
diarrhea
acneiform rash
interstitial lung disease
What is the mechanism of action of bevicizumab?
It is a monoclonal antibody that binds to vascular endolethial growth factor
What is the mechanism of action of sorafenib and sunitinib?
They inhibit many tyrosine kinases including VEGF receptors and PDGF beta receptors
What are the adverse effects associated with sorafenib and sunitinib
hypertension
hand-foot syndrome
possible heart failure (sunitinib)
rash
How does all-trans retinoic acid work and what cancer is it used to treat?
It induces the differentiation of cancer cells in acute promyelocytic leukemia
What is the mechanism of arsenic trioxide and what toxicities are associated with it?
It induces differentiation but can cause QT prolongation, arrhythmias, and retinoic acid syndrome
What is the mechanism of action of bortezomib?
It is a proteasome inhibitor
What is the mechanism of action of hydroxyurea and what are the side effects?
It inhibits ribonucleotide reductase but causes bone marrow suppression, N and V, skin reactions