Anticancers Flashcards

1
Q

Cell cycle specific agents

A
  • antimetabolites
  • bleomycin
  • microtubule inhibitors
  • epipodophyllotoxins
  • camptothecins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

cell cycle nonspecific agents

A
  • alkylating agents
  • platinum coordination complexes
  • antitumour antibiotics
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

MULTIDRUG RESISTANCE

A
  • Mainly due to overexpression of membrane efflux pumps; P-glycoprotein is the most important efflux pump responsible for multidrug resistance.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

TOXICITY: COMMON ADVERSE EFFECTS

A
  • Doxorubicin causes cardiotoxicity.
  • Cyclophosphamide & ifosphamide cause hemorrhagic cystitis.
  • Bleomycin causes pulmonary fibrosis.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

MINIMIZING ADVERSE EFFECTS

A
  • Leucovorin rescues bone marrow from methotrexate.
  • Mesna reduces hemorrhagic cystitis caused by cyclophosphamide and ifosfamide.
  • Dexrazoxane reduces anthracycline-induced cardiotoxicity.
  • Filgrastim reverses neutropeniac aused by many anticancer agents.
  • Amifostine is a cytoprotective agent that reduces renal toxicity caused by cisplatin.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

ANTIMETABOLITES

A
  • FOLATE ANALOGS
  • PURINE ANALOGS
  • PYRIMIDINE ANALOGS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

MTX

A
  • folate analog
  • DHR inhibitor
  • Methotrexate undergoes conversion to a series of polyglutamates (MTX-PGs).
  • The process is catalyzed by the enzyme folylpolyglutamate synthase (FPGS).
  • MTX-PGs increase the inhibitory potency of MTX for additional sites, including thymidylate synthase and enzymes of the de novo purine nucleotide biosynthetic pathway.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

MTX - Adverse

A
  • Common: Stomatitis, mucositis, myelosuppression, alopecia, nausea, vomiting.
  • Renal Damage: Uncommon. Complication of high-dose methotrexate.
  • Hepatic fibrosis and cirrhosis.
  • Pneumonitis.
  • Neurologic Toxicities. With IT administration.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Leucovorin

A
  • Leucovorin is N5-formyl-THF.
  • Antidote to drugs that decrease levels of folic acid, such as methotrexate, to rescue the bone marrow.
  • Leucovorin provides the normal tissues with the reduced folate, thus circumventing the inhibition of DHFR.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Purine analogs

A
  • 6-MP
  • 6-Thioguanine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

6-MP

A
  • Thiol analog of hypoxanthine.
  • Converted to thio-IMP by th esalvage pathway enzyme, HGPRT.
  • Thio-IMP inhibits the first step of the de novo purine ring biosynthesis.
  • Thio-IMP also blocks formation of AMP and GMP from IMP.
  • Also,dysfunctional RNA and DNA result from incorporation of guanylate analogs.

PK

  • metabolized by Xanthine Oxidase to yield thiouric acid
  • also metabolized by TPMT
  • If allopurinol is given to reduce hyperuricemia, dose of 6-MP must be decreased to avoid accumulation of the drug.

Uses: ALL

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

6-Thioguanine

A
  • Converted to the nucleotide, which then inhibits purine synthesis and the phosphorylation of GMP to GDP.
  • It can be incorporated into RNA and DNA.
  • Used for acute non-lymphocytic leukemias.
  • Allopurinol does not potentiate 6-TG action because very little is metabolized to thiouric acid.
  • Toxicities: same as for 6-MP.

PK

  • 6-mercaptopurine and 6-thioguanine are also metabolized by the enzyme thiopurine methyltransferase (TPMT).
  • Patients who have weak activity TPMT are at increased risk for severe toxicities such as myelosuppression.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Pyrimidine analogs

A
  • 5-FU
  • Capecitabine
  • Cytarabine
  • Gecitabine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

5-FU

A
  • Carbecitabine is prodrug
  • Converted to the deoxyribonucleotide 5-FdUMP.
  • 5-FdUMP inhibits thymidylate synthase. DNA synthesis is inhibited.
  • ‘Thymineless death’ results.
  • 5-FU is also converted to 5-FUTP and incorporated into RNA, interfering with RNA processing and function.

**MOA: **Thymidylate synthase inhibition

  • Leucovorin potentiates 5-FU (but inhibits MTX)

Uses

  • 5-FU is used in the treatment of carcinomas of the breast and GI tract.
  • 5-FU can be used topically for keratoses and superficial basal-cell carcinoma.
  • 5-FU/leucovorin combination is used as chemotherapy for colorectal cancer.
  • 5-FU inhibits thymidylate synthase by forming a ternary complex involving the enzyme, the substrate (5-FdUMP), and the cofactor (N5, N10- methylene-THF).
  • **Increasing levels of N5,N10-methylene-THF potentiates the activity of 5-FU. **

Metabolism

  • 5-FU is mainly metabolized by the enzyme dihydropyrimidine dehydrogenase (DPD).
  • Deficiency of DPD is seen in up to 5% of cancer patients.
  • These patients may experience severe toxicity such as myelosuppression, neurotoxicity and life-threatening diarrhea.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

5-FU: Adverse

A
  • Nausea, vomiting, alopecia, bone marrow depression.
  • An erythematous desquamation of the palms and soles called the “hand-foot syndrome” is seen after extended infusions.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Capecitabine

A
  • Fluoropyrimidine carbamate.
  • Orally available prodrug of 5-FU.
  • Capecitabine’s cytotoxic activity is the same as that of 5-FU.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Cytarabine (ARA-C)

A
  • Analog of deoxycytidine.
  • Sequentially phosphorylated to the trisphosphate.
  • Incorporated into DNA.
  • The incorporated residue inhibits DNA polymerase.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Gemcitabine

A
  • Analog of deoxycytidine.
  • Anti-metabolite -> cell cycle non-specific

**MOA: **

  • Phosphorylatedbynucleosidekinasestothe nucleoside di- and triphosphate, which inhibit DNA synthesis.
  • Inhibitionresultsfromtwoactions:
  • Inhibition of ribonucleotide reductase.
  • Incorporation of gemcitabine triphosphate into DNA. This results in chain termination.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

ANTITUMOUR ANTIBIOTICS

A
  • Bind to DNA through intercalation between bases and block synthesis of new RNA or DNA (or both), cause DNA strand breakage, and interfere with cell replication.
  • includes anthracyclines & bleomycin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Anthracyclines

A
  • includes doxorubucin & daunorubicin
  • The anthracycline antibiotics are among the most important antitumor agents.
  • Doxorubicin is one of the most widely used anticancer drugs.

MOA: 4 major mechanisms:

  • Inhibition of topoisomerase II
  • Intercalation in DNA with consequent blockade of DNA & RNA synthesis and strand breakage.
  • Binding to cell membranes to alter fluidity and ion transport.
  • Generation of free radicals.
  • The free radicals are the cause of the cardiac toxicity.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Anthracyclines: Adverse

A
  • Myelosuppression: main toxicity.
  • Cardiotoxicity:
    • Dose-dependent, dilated cardiomyopathy associated with heart failure.
    • Due to free radicals.
    • The iron-chelating agent dexrazoxane can reduce the cardiotoxicity.
  • “Radiation recall reaction” with erythema at sites of prior radiation therapy.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Bleomycin

A
  • Mixture of glycopeptides.
  • Like the antracyclines, bleomycin causes breakage of DNA by oxidative processes.
  • Cell-cycle specific. Arrest cells in G2 phase.

MOA

  • A DNA-bleomycin-Fe2+ complex undergoes oxidation to bleomycin- Fe3+.
  • The liberated electrons react with O2 to form free radicals which cause strand breakage.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Bleomycin: Adverse

A
  • The most serious adverse reaction is pulmonary toxicity (pneumonitis, fibrosis).
  • Dose-limiting.
  • Very little myelosuppression.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Alkylating agents

A

includes

  • nitrogen mustards (mechlorethane, cyclophosphamide, Ifosfamide, melphalan)
  • alkyl sulfonates
  • nitrosoureas
  • triazenes
  • methylhydrazines
  • Exert cytotoxic effects via transfer of their alkyl groups to various cellular constituents.
  • Alkylation of DNA is probably what leads to cell death.
  • Alkylation of DNA can occur on a single DNA strand or on both strands through cross-linking, as most major alkylating agents are bi- functional.
  • Toxicities occur particularly in rapidly growing tissues like the bone marrow, GI tract and gonads.
  • Nausea and vomiting are common.
  • Emetic effects can be reduced with 5-HT3 receptor antagonists.
  • Alkylating agents are mutagenic and carcinogenic.
  • Cyclophosphamide is the most widely used alkylating agent.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Mechlorethamine including Adverse

A
  • Very unstable.
  • Solutions must be made up just prior to administration.
  • Powerful vesicant. Only given IV.

Adverse

  • Severe nausea and vomiting.
  • Severe bone marrow depression.
  • Alopecia.
  • Immunosuppression.
  • Largely replaced by cyclophosphamide, melphalan and other more stable alkylating agents.
26
Q

Cyclophosphamide

A
  • Can be given orally or IV.
  • Prodrug.
  • **Activated to 4-OH-cyclophosphamide by CYP2B. **
  • Broad clinical spectrum.
  • Essential component of many drug combinations.
  • Most widely used alkylating agent
27
Q

Cyclophosphamide - Adverse

A
  • Nausea and vomiting
  • Bone marrow depression
  • Hemorrhagic cystitis
  • Alopecia
  • Sterility
  • Acrolein, a metabolite of cyclophosphamide is responsible for the hemorrhagic cystitis caused by therapy with cyclophosphamide.
  • This can be prevented by parenteral administration of mesna, a sulfhydryl compound which reacts with acrolein in the bladder.
28
Q

IFOSFAMIDE including Adverse

A
  • Analog of cyclophosphamide.
  • Activated by hydroxylation in the liver by CYP3A4

Adverse

  • Similar toxicity profile to cyclophosphamide, but causes greater platelet suppression, neurotoxicity, and urinary tract toxicity.
  • Adequate hydration and administration of mesna permit its use.
29
Q

Melphalan

A

Primarily used to treat multiple myeloma.

30
Q

Alkyl sulfonate

A

BUSULFAN

  • Mainly used in chronic myelogenous leukemia.
  • Myelosuppression is the main toxicity.
  • May cause pulmonary fibrosis.
31
Q

Nitrosoureas

A

CARMUSTINE

LOMUSTINE

  • Very lipophilic.
  • Cross the blood-brain barrier.
  • Useful in treatment of brain tumours.
  • Nitrosoureas require biotransformation, which occurs non-enzymatically.
32
Q

TRIAZENES

A

DACARBAZINE

  • Acts as methylating agent after activation in the liver.
  • Given IV.
  • Indicated for malignant melanoma.
  • Also indicated for Hodgkin’s disease.
  • Toxicity includes nausea and vomiting.
  • Myelosuppression is usually mild to moderate.
33
Q

PROCARBAZINE

A
  • METHYLHYDRAZINES
  • Converted by liver P450 enzymes to alkylating metabolites.

USES

  • Used in combination with mechlorethamine, vincristine and prednisone (the MOPP regimen) for the treatment of Hodgkin’s disease.
  • Alternative regimens with less leukemogenic potential have replaced MOPP.
34
Q

PROCARBAZINE: ADVERSE EFFECTS

A
  • Leukopenia and thrombocytopenia.
  • MAO inhibitor
  • Disulfiram-like reactions.
  • Highly carcinogenic, mutagenic and teratogenic.
  • Potent immunosuppressive agent.
35
Q

PLATINUM COORDINATION COMPLEXES

A
  • CISPLATIN
  • CARBOPLATIN
  • Broad antineoplastic activity.
  • Have become the foundation for treatment of testicular cancer, ovarian cancer, and cancers of the **head and neck, bladder, esophagus, lung and colon. **
  • Inhibit DNA synthesis and binds DNA through formation of cross-links.
  • Given IV.

Uses

  • Approved for testicular, ovarian, and bladder cancers.
  • Its use with vinblastine and bleomycin has been major advance in development of curative therapy for testicular cancers.
36
Q

CISPLATIN: ADVERSE EFFECTS

A
  • Myelosuppression: mild-to-moderate.
  • Nausea and vomiting.
  • Ototoxicity.
  • Peripheral neuropathy.
  • Nephrotoxicity is abolished by hydration and diuresis.
  • Amifostine is a cytoprotective agent used to reduce the renal toxicity associated with cisplatin.
37
Q

Carboplatin - Adverse

A
  • Less nausea, neurotoxicity, ototoxicity and nephrotoxicity than cisplatin.
  • Dose-limiting toxicity is myelosuppression.
38
Q

MT inhibitors

A
  • Vinca alkaloids: vincristine & vinblastine
  • Taxanes: Paclitaxel & Docetaxel
39
Q

Vinca alkaloids including Adverse

A
  • includes vincristine and vinblastine

MOA

  • Vinca-tubulin and inhibit its ability to polymerize into microtubules.
  • This results in mitotic arrest in metaphase.
  • Cell division stops. Cells die by apoptosis.

Adverse

  • Vincristine: Peripheral neuropathy. Bone marrow depression is mild. Alopecia.
  • Vinblastine: Myelosuppression is the dose- limiting adverse effect. Other adverse effects include alopecia and peripheral neuropathy
40
Q

Taxanes

A
  • including Paclitaxel & Docetaxel

MOA

  • Taxanes bind to beta-tubulin subunit of microtubules and promote microtubule polymerization.
  • Stabilization of the microtubules in a polymerized state arrests cells in mitosis and leads to apoptosis.
41
Q

Taxanes - Adverse

A

PACLITAXEL

  • Hypersensitivity,myelosuppression,peripheral neuropathy, alopecia, etc
  • Hypersensitivity is reduced by premedication with dexamethasone, diphenhydramine and an H2 blocker
  • Abraxane is an albumin-bound form of paclitaxel which does not cause hypersensitivity, does not require premedication, and causes less myelosuppresion than the traditional paclitaxel.

DOCETAXEL

  • Myelosuppression, peripheral neuropathy, fluid retention, alopecia, mucositis.
  • Pretreatment with dexamethasone is required to prevent fluid retention.
  • Docetaxel does not cause neuropathy as frequently as paclitaxel.
  • Myelosuppression is dose-limiting.
42
Q

Epipododphyllotoxin

A
43
Q

Camptothecin

A
  • TOPOTECAN
  • IRINOTECAN
  • Natural products derived from the Camptotheca acuminata tree.
  • Inhibit topoisomerase I. Inhibition results in DNA damage.
44
Q

Relative myelosupression

A

High

  • Cytarabine
  • Alkylating agents
  • Doxorubicin
  • Daunorubicin
  • Vinblastine

Medium

  • Carboplatin
  • MTX
  • 5-FU

Low

  • Bleomycin
  • Vincristine
  • Asparaginase
45
Q

ESTROGEN INHIBITORS

A

Anti-estrogen approaches for therapy of hormone-positive breast cancer include:

  • SELECTIVE ESTROGEN-RECEPTOR MODULATORS (SERMs): Tamoxifen, Raloxifene
  • SELECTIVE ESTROGEN-RECEPTOR DOWNREGULATORS (SERDs): Fulvestrant
  • AROMATASE INHIBITORS (AIs): Anastrozole & Letrozole, Exemestane
46
Q

TAMOXIFEN

A
  • SERMS bind to estrogen receptors and act as agonists or antagonist depending on the tissue.
  • Tamoxifen is an antagonist on breast cancer.
  • Tamoxifen is an agonist in nonbreast tissues.
  • Used for receptor-positive breast cancer.
  • Chemopreventive agent in women at risk for breast cancer.
47
Q

RALOXIFENE

A
  • Raloxifene is an antiestrogen in the uterus and the breast, while promoting estrogenic effects in the bone to inhibit resorption.
  • Used for prevention of postmenopausal osteoporosis and prophylaxis of breast cancer in high risk postmenopausal women.
48
Q

FULVESTRANT

A
  • SERDs are devoid of estrogen agonist activity.
  • Fulvestrant binds to the estrogen receptor (ER) inhibits its dimerization and increases its degradation.
  • ER-mediated transcription is abolished.
49
Q

AROMATASE INHIBITORS

A
  • Aromatase converts androstenedione to estrone.
  • In postmenopausal women, this conversion is the primary source of circulating estrogens.
  • Aromatase inhibitors are the standard of care for adjuvant treatment of postmenopausal women with hormone receptor–positive breast cancer.

ANASTROZOLE & LETROZOLE

  • Nonsteroidal.
  • Reversiblecompetitiveinhibitorsofaromatase.

EXEMESTANE

  • Steroidal.
  • Irreversibleinhibitorofaromatase.
50
Q

ANDROGEN INHIBITORS

A
  • The preferred approach for the therapy of prostate cancer is the use of GnRH agonists, alone or in combination with an androgen receptor blocker.
  • GONADOTROPIN-RELEASING HORMONE AGONISTS
  • ANDROGEN RECEPTOR BLOCKERS
51
Q

GONADOTROPIN-RELEASING HORMONE AGONISTS

A
  • GOSERELIN
  • LEUPROLIDE
  • When given continuously or as a depot, GnRH agonists cause an initial surge in LH and FSH levels, followed by inhibition of gonadotropin release.
  • This results in reduction of testicular production of testosterone to castrate levels.
  • Testosterone levels fall to 10% of their initial values after a month.
  • However,they increase significantly in the beginning, causing a transient flare of tumor activity and an increase in symptoms.
  • The flare phenomenon can be counteracted by concurrent administration of flutamide for 2-4 weeks.
52
Q

FLUTAMIDE

A
  • ANDROGEN RECEPTOR BLOCKERS
  • Synthetic, nonsteroidal antiandrogen.
  • Metabolized to an active metabolite that acts as a competitive antagonist at the androgen receptor, preventing its translocation to the nucleus.
53
Q

Inhibitors of EGF Receptors

A
  • Gefitinib:
  • Erlotinib:
  • Lapatinib: Inhibitor of EGFR and ErbB2 tyrosine kinases.
  • Cetuximab: mAb against EGFR
  • Trastuzumab: mAb against ErbB2
54
Q

Imatinib

A

Inhibits the tyrosine kinase of Bcr-Abl. Inhibits c-kit (a receptor tyrosine kinase.)

55
Q

Sorafenib

A
  • Inhibits the serine/threonine kinase RAF, which is immediately downstream of RAS
  • also inhibits VEFR-2, PDGFR and other RTK so it also inhibits angiogenesis
56
Q

Bortezomib

A
  • Inhibits the proteasome.
  • Induces growth inhibition and apoptosis of tumor cells with relatively few toxic effects on normal cells.
57
Q

Sunitinib

A
  • Inhibits VEGFR-1, VEGFR-2, and PDGFR
58
Q

ASPARAGINASE

A
  • Most normal tissues synthesize L-asparagine in amounts sufficient for protein synthesis.
  • Certain neoplastic tissues, however, require exogenous source.
  • Asparaginase hydrolyzes serum asparagine, depriving these cells of the asparagine necessary for protein synthesis, leading to cell death.
59
Q

ASPARAGINASE - ADVERSE EFFECTS

A
  • Hypersensitivity.
  • Decrease in clotting factors.
  • Liver abnormalities.
  • Pancreatitis, seizures, coma due to ammonia toxicity.
60
Q

HYDROXYUREA

A
  • Inhibits ribonucleotide reductase.
  • This leads to depletion of deoxynucleoside trisphosphate pools.
  • DNA synthesisis thereby inhibited.
  • Kills cells in S phase.
  • Given orally.
  • **Uses: **Melanoma, chronic myelocytic leukemia, carcinoma of the ovary, head and neck.
61
Q

​Interferons alpha

A

Approved for hairy cell leukemia, CML, malignant melanoma and Kaposi’s sarcoma.