1 / 38

Alkylating Agents

Alkylating Agents. Largest class of antineoplastics General Properties/Mechanism: All are electrophilic molecules that covalently modify nucleophilic molecules in cells DNA most important adduct (N 7 and O 6 of Guanine) for anticancer properties General Types of Alkylating Agents :

bernad
Download Presentation

Alkylating Agents

An Image/Link below is provided (as is) to download presentation Download Policy: Content on the Website is provided to you AS IS for your information and personal use and may not be sold / licensed / shared on other websites without getting consent from its author. Content is provided to you AS IS for your information and personal use only. Download presentation by click this link. While downloading, if for some reason you are not able to download a presentation, the publisher may have deleted the file from their server. During download, if you can't get a presentation, the file might be deleted by the publisher.

E N D

Presentation Transcript


  1. Alkylating Agents Largest class of antineoplastics General Properties/Mechanism: • All are electrophilic molecules that covalently modify nucleophilic molecules in cells • DNA most important adduct (N7 and O6 of Guanine) for anticancer properties General Types of Alkylating Agents: • Monofunctional • Cause single strand DNA breaks • Bifunctional • Inhibit DNA replication and transcription by crosslinking DNA Subtypes: • Nitrogen mustards • Nitrosoureas • Platinum Compounds

  2. Nitrogen Mustards Compound • Mechlorethamine (Mustargen) • Hodgekin’s/other lymphomas • and chronic leukemias. • MOPP regimen MOPP (Mechlorethamine, Vincristine (Oncovin), Procarbazine, Prednisone) • Chlorambucil (Leukeran) • Chronic lymphocytic leukemia, • malignant lymphomas • Reactivity: >Alkeran, <Mustargen • Less alopecia and nausea • Cumulative bone marrow toxicity

  3. Mechlorethamine (Mustargan) • Mustargan serves as a prototype for the mustards (and other alkylating agents) because of its spectrum of activity, its toxicities and its mechanism of action. It is highly unstable and reactive – still in use in combination therapy, MOPP, 80% response, >50% cure • Toxicity: Nausea and vomiting (N&V). Dose limiting toxicity (DLT) is bone marrow suppression, white blood cells, esp. granulocytes. Max suppression at 10-12 days, recovery around 42 days. • ADME: t1/2 less than 30 min., admin in free flowing catheter • Analogs: Chlorambucil (Leukeran), Chronic Lymphocytic Leukemia; Melphalan (Alkeran), Multiple Myeloma (longer half-lives, p.o.); Busulfan (Busulfex), CML

  4. Most Common Nitrogen Mustards • Cyclophosphamide (Cytoxan) • Malingnant lymphomas, various carcinomas. • Activated by P450 enzymes • Most versatile nitrogen mustard • Better bone marrow recovery than Mustargen Ifosphamide (Isophosphamide, Ifex) Lung, breast, ovarian, lymphomas

  5. Cyclophosphamide (Cytoxan) • Use: Most widely used alkylating agent. Non-Hodgkin's lymphomas (CHOP), Breast (CMF increases 7 yr survival form 31% to 49 %) • Toxicity: N&V, anaphalactoid rxns. DLT, bone marrow suppression, max 10-12 days, recovery by 21 days (relatively platelet sparing); alopecia, hemorrhagic cystitis (minimized by diuresis), etc. • ADME: May be administered parenterally, in a large range of doses. Resistant cells may have aldehyde oxidase (like the liver) • Analog: Isofosfamide (Ifex)

  6. Nitrosoureas Compound Chloroethylcyclohexyl nitrosourea (Lomustine, CCNU) Bischloronitrosourea (Carmustine, BCNU) • BCNU and CCNUs:Brain tumors, Hodgkin’s disease, melanoma. • Lipid soluble: (CNS active) • O6 of Guanine preferred alkylation site • Prolonged cummulative myelosuppression • Teratogenic

  7. BCNU (Carmustin) • Use: Primary glioblastoma (with surgery and radiation can increase live span from 20 weeks to 50 weeks) • Toxicity: N&V. DLT bone marrow suppression, max 28 days; recovery by 42 days. • ADME: Administered by injection. Unique among antineoplastics for its high oil-to-water partition coefficient (very lipophillic) and low ionization at physiological pH – risk of hematological toxicity persist due to storage in the liver & adipose tissue • Analog: CCNU (lomustine), may be administered orally. Temozolomide (Temodar)is a new non-nitrosourea used frequently in glioblastoma

  8. Platinum compounds • Cisplatin (Platinol) • testicular, ovarian, head and neck, lung, • bladder cancer • Curative in combination for metastatic testicular cancer • Carboplatin (Paraplatin) • ovarian, non-small cell and small cell lung cancers • less toxic (~45×) than cisplatin but is generally less active • Oxaliplatin (Eloxatin) • broad anticancer activity: colorectal, ovarian, pancreatic, non-Hodgkins lym, breast, lung, prostate, etc • Lacks cross-resistance with other platinum compounds • Generally less toxic than other platinum compounds

  9. Cisplatin (Platinol) Use: Most used in combination therapy - testicular tumors (PEB ~ 85% curative) Toxicity: DLT – nephrotoxicity, can be treated with Amifostine (a thiophosphate) Major N&V, hearing loss at high end. ADME : Administered i.v. after 1-2 liters saline and mannitol (chloride diuresis)  Analog: Carboplatin (Paraplatin), Oxaliplatin (Eloxatin) is now a first line drug for combination therapy of colon cancer

  10. Other Alkylators • Streptozocin (Zanosar), a nitrosourea • Use: Malignant pancreatic insulinoma, pancreatic carcinoid, doubles 1 yr survival rates (Nausea and vomiting notable, renal toxicity in 2/3 cases – bone marrow suppression in ~ 20%) • Procarbazine (Mutalane) • Use: Hodgkin’s Disease (MOPP protocol) • Few reports of cross resistance, minor toxicities (bone marrow suppression), psychic disturbances, nausea, vomiting, Weak MAO inhibitor - available orally • Dacarbazine (DTIC) • Temozolomide & DTIC are metabolized to same active compound: 5-amino-imidizole-4 carboxamide - thought to be the active alkylating species • DTIC Use: Melanoma, Hodgkin’s disease (ABVD), dose i.v. • Temozolomide Use: malignant gliomas

  11. Alkylating Agents - Summary • Largest class of antineoplastic drugs • Nitrogen Mustards • Nitrosoureas • Platinum Compounds • All are electrophilic compounds that react with DNA • Bifunctional: Nitrogen mustards & platinum compounds • Monofuctional: Nitrosoureas • Toxicities vary depending on the particular compound • Many have severe bone marrow suppression • Platinum compounds tend to show more renal toxicities

  12. Metabolites and their Antimetabolites General Properties/Mechanism: • Antimetabolites resemble cellular metabolites and act to interfere with DNA synthesis or the synthesis of DNA precursors. • These are classical cell cycle specific anti-cancer agents • Most are prodrugs that must be activated through incorporation into the normal biosynthetic pathways • Toxicities: only partially selective to tumor cell – toxic to all rapidly dividing normal cells • Bone marrow and intestinal epithelium are particularly sensitive • General Drug classes: • Antifolates • Antinucleotide analogs

  13. Folate Antimetabolites • Methotrexate, Amethopterin • Trophoblastic choriocarcinoma, lymphoblastic leukemia, Burkitt’s • lymphoma • Class: Folate antagonist • Mechanism: Dihydrofolate Reductase inhibitor: inhibits dTMP synthesis. Kills in S-phase (G1 and S RNA and protein synthesis) • Toxicity: bone marrow, GI, renal, alopecia, teratogen • Resistance: increased/altered DHFR, decreased uptake

  14. Nucleotide Analogs • 5-Fluorouracil; 5-FU (Efudex) • Colorectal, breast, gastric, pancreatic colon cancers (35% decrease in recurrence). Used topically for premalignant skin lesions. • Class: Pyrimidine analog • Mechanism: irreversible inhibition of thymidylate synthase (TS). Incorporation into DNA/RNA. Kills in G1 and S-phases. Penetrates CNS. • Toxicity: N&V, GI toxicity. DLT bone marrow suppression • Resistance: increased/altered TS • F-UMP: incorporated into RNA affecting processing/function • F-dUMP: Inhibits Thymidylate Synthase • ADME: Administered i.v., half-life in the blood of ~10-20 min • Analog: Floxuridine (FUDR), often infused into the hepatic artery for liver cancer

  15. Modulation of 5-FU & MTX with Leucovorin Rescue • Leucovorin (reduced form of folic acid) is commonly used to rescue cells exposed to folate antagonists (i.e., methotrexate) - does not function as a cytotoxic chemotherapy agent • High-dose methotrexate with leucovorin rescue (HDM-L) is a component of the standard regimen for adjuvant therapy of osteosarcoma, produces a high complete response rate in CNS lymphomas, and is a part of standard curative therapy for childhood ALL • Leucovorin promotes 5-FU inhibition of Thymidylate Synthase – used in colon cancer

  16. More nucleotide analogs… • Cytosine arabinoside; araC; Cytarabine(Cytosar) • Acute myelogenous leukemia • Class: Nucleoside sugar analog • Mechanism: activated to ara-CTP (DNA polymerase inhibitor) by dCMP kinase. Kills in S-phase • Toxicity: severe bone marrow hypoplasia, GI • Resistance: polymerase alteration, decreased activation, increased deactivation (dCMP deaminase) • Seen as a 2'-deoxynucleoside • Phosphorylated and competes with dCTP for incorporation into DNA • 2'-OH in  position: Steric hindrance with base affects base stacking interactions in DNA helix • 1 AraC per 2000 bases reduces growth rate by 50%

  17. More nucleotide analogs… • 6-Mercaptopurine; 6-MP (Purinethol) • Acute leukemias (40% remission in children) • Class: Purine analog • Mechanism: HPRTase: Hypoxanthine-guanine phosphoribosyltransferase makes the nucleoside phosphate from the free base - inhibition of purine synthesis; incorporation into RNA/DNA unclear. Kills in S-phase. • Toxicity: bone marrow, nausea, vomiting • Resistance: decreased HPRTase activity (needed to incorporate 6-MP into RNA/DNA) 6-Thioguanine (Thioguanine) Same as 6-MP Used against granulocytic Leukemia with cytarabine

  18. Antimetabolites - Summary • Classical cell-cycle dependent agents • Require bioactivation • Classes • Antifolates • Antinucleotide analogs • Inhibit enzymes required for DNA synthesis • Toxicities extend to tissues with higher rates of cellular turnover

  19. Antibiotics General classes of carcinolytic antibiotics: • Cyclic pentapeptides • Anthracyclines • Complex glycopeptides General Properties/Mechanism: • All bind to DNA or DNA associated proteins. • All inhibit RNA synthesis (transcription) • Most are Isolated from Steptomyces broths • Some induce DNA strand breaks by free radical production and/or inhibition of Topoisomerase II (Topo-II) Pure Topoisomerase Inhibitors: • Two Classes: Topo I and Topo II • Inhibit topoisomerase enzymes resulting in DNA breaks

  20. Actinomycin D (Cosmogen) • Wilm’s tumor (kidney, in children), Ewing’s and Kaposi’s sarcomas, MTX-resistant choriocarcinoma. • Class: Cyclic pentapeptide • Mechanism: DNA intercalation; inhibits transcription. Kills in the G1 and S phases. • Toxicity: Myelosuppression, GI, skin. • RNA polymerase much more sensitive than DNA polymerase. • Causes single-strand DNA breaks. • Free radical formation • Topo II inhibition One of the most potent anticancer drugs per mole

  21. Anthracyclines • Doxorubicin (Adriamycin, ADM) • Breast, head, neck, thyroid, bladder, testes, prostate, ovary, Wilm’s and Ewing’s sarcomas, leukemias and lymphomas • Class: Anthracycline • Mechanism: DNA intercalator, DNA breaks by free radical damage and Topo-II interference. Kills in S phase. • Toxicity: DLT - cardiotoxicity (total dose, often irreversible), myelosuppression • Resistance: decreased/altered Topo-II, P-glycoprotein activity (decreased accumulation) Doxorubicin-polymer conjugates have reduces toxicity, increases targeting

  22. Doxorubicin (Adriamycin) Mech: Intercalation and blockage of topoisomerase II, oxygen radical production because of quinone structure. One of the most useful cancer drugs – new formulations extending uses. Toxicity: Myelosuppression max in week 2; recovery in week 4. Radiation recall. Toxicity limiting total dose is a cardiomyopathy, a unique toxicity in 1/3 of patients if the total dose exceeds 550 mg/m2. Dexrazoxane (Zinecard) an antioxidant metal chelator can reduce anthracycline cardiotoxicity. ADME: Administered i.v., cleared by the liver. Analogs: Daunomycin (Daunorubicin), doxorubicin, about ½ as potent, Mitoxantrone (Novantrone), a synthetic doxorubicin with reduced cardiotoxicity; Idarubacin (Idamycin).

  23. Bleomycin A2 and B2 Complex Glycopeptides • Bleomycin (Blenoxane, BLM; mixture of A2 and B2) • Head, neck, testes (Curative with vinblastine and cisplatin), uterine, cervix, Hodgkin’s and non-Hodgkin’s lymphomas. • Class: Complex glycopeptide • Mechanism: DNA binding region. DNA breaks by free radical damage. Kills in G2 phase. • Toxicity: Cardiotoxicity, edema, lung and skin fibrosis - Minimal myelo- or immunosuppression • Resistance: Increased DNA repair

  24. Bleomycin (Blenoxane) • Mech.: Has DNA binding region & metal binding region (Cu2+ or Fe2+)Transfers e- from M2+ to O2 to form activated oxygen species. Causes double strand breaks in DNA - Cytotoxicity correlates with DNA fragmentation • Use: Testicular tumors - curative in combination with cisplatin and etoposide (PEB) • Toxicity: N&V, skin toxicity. DLT - pulmonary fibrosis at total dose of 400 U. • ADME: Bleomycin is excreted primarily by the kidneys Pulmonary toxicity is probably enhanced when dosage is not adjusted for renal dysfunction. • Resistance: Bleomycin appears to undergo extensive metabolism by a specific cysteine proteinase, bleomycin hydrolase – low levels in the lungs and skin.

  25. Pure Topo-II Inhibitor • Etoposide (VePesid, VP-16) • Testicular, small cell lung • Mechanism: Stabilizes Topo-II-DNA complex. Causes double strand break. Kills in G2 and S phases. • Toxicity: DLT - bone marrow, nausea, diarrhea (GI), UNCERTAIN effect on secondary neoplasia • ADME: Administered p.o. and i.v, largely cleared by kidney • Resistance: P-glycoprotein activity (decreased accumulation) Source: mandrake plant (podophyllum peltatum) • Podophyllotoxin binds microtubules and inhibits assembly. • Etoposide does not inhibit assembly of microtubules

  26. CPT 11, Irinotecan (Camptosar) Mech: “Pure” Topoisomerase I poison - source: derivatives of camptothecin (CPT), a cytotoxic plant alkaloid isolated from the Chinese tree Camptotheca acuminata Use: Colorectal cancer, and small cell and non-small cell lung cancer - has also shown positive responses in pancreatic cancer, refractory lymphoma, malignant gliomas, and gynecologic cancers Toxicity: GI toxicity, severe diarrhea. DLT - bone marrow suppression ADME: Administered i.v., cleared by kidney Analogs: topotecan (Hycamtin) Pure Topo-I Inhibitor

  27. Taxanes • Paclitaxel (Taxol) • Breast, ovary, lung, head and neck • Mechanism: Stabilizes microtubule assembly “mitotic spindle poison” G2/M arrest. • Toxicity: DLT - Myelosuppression, peripheral neuropathy, allergic reactions to injection are a problem • ADME: Administered i.v., cleared by the liver, susceptible to MDR • Resistance: Altered tubulin, P-glycoprotein activity (decreased accumulation) • Analog: docetaxel – semisynthetic, similar to paclitaxel • Taxol source: Taxus brevifolia • Limited supply • Now made semisynthetically • From needles of the Himalayan Yew (Taxus bacatta)

  28. Vinca alkaloids • Vincristine (Oncovin) • Hodgkin’s disease, childhood leukemia • Mechanism: Destabilizes microtubule assembly “mitotic spindle poison” G2/M arrest. • Toxicity: DLT - peripheral neuropathy • Resistance: Altered tubulin, P-glycoprotein activity (decreased accumulation) • Vinblastin (Velban) • Testicular (cisplatin, bleomycin) • Mechanism: Destabilizes microtubule assembly “mitotic spindle poison” G2/M arrest. • Toxicity: DLT – bone marrow suppression • Resistance: Altered tubulin, P-glycoprotein activity (decreased accumulation) Source: periwinkle plant (Vinca rosea) Despite similar structures, incomplete cross-resistance Administered i.v., susceptible to MDR, cleared largely by the liver

  29. Misc. agents • Hydroxyurea • Use: AML or the blastic phase of CML • Because of its radiosensitizing effects, it is used with radiation therapy to treat head and neck carcinoma • Mechanism: Hydroxyurea blocks ribonucleotide reductase, the rate-limiting enzyme of DNA synthesis. • Hydroxyurea inhibits ribonucleotide reductase by binding to the M2 subunit and disrupting the iron complex. • This enzyme is responsible for converting ribonucleotides to deoxyribonucleotide triphosphates (dNTPs), which are critical to DNA synthesis and repair. • Toxicity: Hematopoietic depression is the major toxic effect; UNCERTAIN risk of secondary leukemia; teratogen • Resistance: alterations in sensitivity of ribonucleotide reductase

  30. Misc. Agents cont. – Hormonal • Tamoxifen (Nolvadex) • Estrogen Receptor (ER) positive breast cancer • Class: Antiestrogen SERMS (synthetic estrogen receptor modulators) • Mechanism: Block estrogen response by competitive inhibition of ER. Inhibits G1 to S transition. • Toxicity: rarely severe adverse reactions to tamoxifen include vasomotor symptoms (hot flushes), N&V & vaginal bleeding . Tamoxifen also increases the incidence of endometrial cancer by two- to threefold • Anastrozole (Arimidex) • Fourth generation of aromatase inhibitor - effective against ER positive breast cancer • Class: Aromatase inhibitor • Mechanism: Anastrozole significantly suppresses serum estradiol levels Anastrozole inhibits aromatase (CYP19), the enzyme that catalyzes the final step in estrogen production. • Toxicity: rarely severe adverse reactions – some musculoskeletal toxicity

  31. Other Hormone/hormone-related • Prednisone • Hodgkin's disease, acute lymphocytic leukemia (ALL), lymphomas • Class: Corticosteroid • Mechanism: Antianabolic effects • Toxicity: Cushingoid symptoms • Diethylstibestrol, DES • Prostate cancer • Class: Estrogenic (non-steroid) • Mechanism: Block androgen dependence • Toxicity: developmental mutagen/teratogen

  32. Leuprolide (Lupron), GnRH peptide, used with Flutamide Mech.: Acts on pituitary to inhibit FSH and LH release Constant GnRH Causes Testosterone or Estrogen Levels to Fall to Castration Levels Use : Prostate cancer, and breast cancer prevention Toxicity: Less than DES, N&V, hot flashes Analog: Goserelin (Zoladex) Hormones-related therapy cont.

  33. Antiandrogens • Flutamide (Eulixen) • Oral nonsteroidal antiandrogen - metastatic prostatic carcinoma • Mechanism: Inhibits the uptake and/or nuclear binding of testosterone and dihydrotestosterone by prostatic tissue - most effective when combined with LHRH agonists • Toxicity: rarely severe adverse • Bicalutamide (Casodex) • oral nonsteroidal antiandrogen structurally related to flutamide but has a long plasma half-life (once daily dosing) • Mechanism: Inhibits the uptake and/or nuclear binding of testosterone and dihydrotestosterone by prostatic tissue - most effective when combined with LHRH agonists - bicalutamide is more selective for the peripheral androgen receptor and has less activity at the central androgen receptor • Toxicity: rarely severe adverse

  34. Hormone-related - Summary • Breast and Prostate cancers frequently demonstrate hormone-dependent growth (at least in early stage disease) • Classes: • GnRH agonists • Antiestrogens (includine for prevention) • Aromatase inhibitors • Antiandrogens • Corticosteroids • Toxicities tend to be more mild

  35. Newer Molecularly Targeted Agents (1) • Tyrosine Kinase Inhibitors • imatinib (Gleevec) – Bcr-Abl/c-kit kinase inhibitor (CML, GIST) • Toxicity: generally mild – N&V, edema & muscle cramps, Neutropenia and thrombocytopenia • gefitinib (Iressa) - EGFR tyrosine kinase inhibitor (non-small cell lung cancer) Antibodies • Trastuzumab (Herceptin) – anti-HER2/neu monoclonal (breast cancer) • Bevacizumab (Avastin) - anti-VEGF monoclonal (colon cancer) • Cetuximab (Erbitux) - anti-EGFR monoclonal antibody (colon cancer) • Others – including antibody conjugates to drug/toxin

  36. Newer Molecularly Targeted Agents (2) • Differentiation Inducers • Retinoic Acid derivatives • Tretinoin, ATRA (Avita) • Uses: acute promyelocytic leukemia (APL) and undergoing phase III investigation in the treatment of Kaposi's sarcoma. Frequently given in combination with an anthracycline • Toxicities: Generally mild - cause dry skin, cheilitis, reversible hepatic enzyme abnormalities, bone tenderness, and hyperlipidemia • Others - proteasome inhibitor • Bortezomib (Velcade) – • Uses: Multiple Myeloma, in trials for other leukemias • dose-limiting toxicities in the following organ systems: gastrointestinal, hematopoietic, lymphatic, and renal

  37. Important Cytokines and Biological Response Modifiers • Biologic Agents Used to bolster immune response: • IL-2 (interleukin-2) T cell stimulation, highly toxic, used in metastatic melanoma • Interferons (Inf 2a,b) used in hairy cell leukemia • Filgrastim (Neupogen) G-CSF • Sargramostim (Leukine) GM-CSF • BCG (Bacillus Calmette-Guerin Live- Mycobacterium bovis ) used in bladder cancer

  38. Combination Chemotherapy Considerations • Most cancers are/become refractory to treatment by a single agent, combinations of anticancer drugs are often used. The following rules apply to combining drugs in cancer chemotherapy: • The drugs each must have some activity against the cancer. • The drugs should act via different mechanisms. • The drugs should have minimal overlapping toxicity. • Cellular resistance to each drug should occur by different mechanisms. •  The first drug combination, made famous by Dr. V. DeVita and others at NIH, is called MOPP (Mechlorethamine, oncovonin, procarbazine, and prednisone). MOPP is a curative treatment for Hodgkin's disease and its development was a major step forward. 

More Related