The document discusses cell life cycles and cell growth kinetics which are important for understanding cancer chemotherapy. It describes the different phases of the cell cycle and explains how some chemotherapy agents target specific phases while others are non-specific or can target cells in all phases including resting cells. The concepts of cell kill, tumor doubling time, and drug resistance are also summarized. Details are provided about different classes of chemotherapy including alkylating agents and the specific drug cyclophosphamide, explaining their mechanisms of action and common adverse effects.
The document discusses cell life cycles and cell growth kinetics which are important for understanding cancer chemotherapy. It describes the different phases of the cell cycle and explains how some chemotherapy agents target specific phases while others are non-specific or can target cells in all phases including resting cells. The concepts of cell kill, tumor doubling time, and drug resistance are also summarized. Details are provided about different classes of chemotherapy including alkylating agents and the specific drug cyclophosphamide, explaining their mechanisms of action and common adverse effects.
Original Description:
Anticancer agents and their pharmacology. MBChB notes.
The document discusses cell life cycles and cell growth kinetics which are important for understanding cancer chemotherapy. It describes the different phases of the cell cycle and explains how some chemotherapy agents target specific phases while others are non-specific or can target cells in all phases including resting cells. The concepts of cell kill, tumor doubling time, and drug resistance are also summarized. Details are provided about different classes of chemotherapy including alkylating agents and the specific drug cyclophosphamide, explaining their mechanisms of action and common adverse effects.
The document discusses cell life cycles and cell growth kinetics which are important for understanding cancer chemotherapy. It describes the different phases of the cell cycle and explains how some chemotherapy agents target specific phases while others are non-specific or can target cells in all phases including resting cells. The concepts of cell kill, tumor doubling time, and drug resistance are also summarized. Details are provided about different classes of chemotherapy including alkylating agents and the specific drug cyclophosphamide, explaining their mechanisms of action and common adverse effects.
Download as PPTX, PDF, TXT or read online from Scribd
Download as pptx, pdf, or txt
You are on page 1of 120
ANTICANCER
Cell life cycle
• Knowledge of the cell life cycle and cell cycle kinetics is essential to the understanding of the activity of chemotherapy agents in the treatment of cancer Phases of the cell cycle 1. M phase, or mitosis, is the phase in which the cell divides into two daughter cells 2. G1 phase, or postmitotic gap, is when RNA and the proteins required for the specialized functions of the cell are synthesized in preparation for DNA synthesis 3. S phase is the phase in which DNA synthesis and replication occurs 4. G2 phase, or the premitotic or postsynthetic gap, is the phase in which RNA and the enzymes topoisomerase I and II are produced to prepare for duplication of the cell Phases of the cell cycle 5. G0 phase, or resting phase, is the phase in which the cell is not committed to division • Cells in this phase are generally not sensitive to chemotherapy • Some of these cells may reenter the actively dividing cell cycle • In a process called recruitment, some chemotherapy regimens are designed to enhance this reentry by killing a large number of actively dividing cells Cell growth kinetics • Several terms describe cell growth kinetics • Cell growth fraction is the proportion of cells in the tumor dividing or preparing to divide • As the tumor enlarges, the cell growth fraction decreases because a larger proportion of cells may not be able to obtain adequate nutrients and blood supply for replication • Cell cycle time is the average time for a cell that has just completed mitosis to grow and again divide and again pass through mitosis. Cell cycle time is specific for each individual tumor Cell growth kinetics • Tumor doubling time is the time for the tumor to double in size • As the tumor gets larger, its doubling time gets longer because it contains a smaller proportion of actively dividing cells owing to restrictions of space, nutrient availability, and blood supply. • The gompertzian growth curve illustrates these cell growth concepts Cell growth kinetics • Tumor cell burden is the number of tumor cells in the body • Because a large number of cells is required to produce symptoms and be clinically detectable (approximately 109 cells), the tumor may be in the plateau phase of the growth curve by the time it is discovered • The cell kill hypothesis states that a certain percentage of tumor cells will be killed with each course of cancer chemotherapy Cell growth kinetics • As tumor cells are killed, cells in G0 may be recruited into G1, resulting in tumor regrowth • Thus, repeated cycles of chemotherapy are required to achieve a complete response or remission • The percentage of cells killed depends on the chemotherapy dose • In theory, the tumor burden would never reach absolute zero because only a percentage of cells are killed with each cycle • Less than 104 cells may depend on elimination by the host’s immune system Cycle specificity of chemotherapy • Chemotherapeutic agents may be classified according to their reliance on cell cycle kinetics for their cytotoxic effect • Combinations of chemotherapy agents that are active in different phases of the cell cycle may result in a greater cell kill 1. Phase-specific agents are most active against cells that are in a specific phase of the cell cycle • These agents are most effective against tumors with a high growth fraction Phase-specific agents • Theoretically, administering phase-specific agents as continuous intravenous infusions or by multiple repeated doses may increase the likelihood of hitting the majority of cells in the specific phase at any one time • Therefore, these agents are also considered schedule-dependent agents • Examples are as follows: a) M phase: mitotic inhibitors (e.g., vinca alkaloids, taxanes) b) G1 phase: asparaginase, prednisone c) S phase: antimetabolites d) G2 phase: bleomycin, etoposide Phase-nonspecific agents 2. Phase-nonspecific agents are effective while cells are in the active cycle but do not require that the cell be in a particular phase • These agents generally show more activity against slow-growing tumors • They may be administered as single bolus doses because their activity is independent of the cell cycle • These drugs are also considered dose-dependent agents • Examples are alkylating agents and antitumor antibiotics Cell cycle–nonspecific agents 3. Cell cycle–nonspecific agents are effective in all phases, including G0 • Examples are carmustine and lomustine • Radiation therapy is also considered cell cycle nonspecific Drug resistance • A fundamental issue in cancer chemotherapy is the development of cellular drug resistance. Some tumor types, e.g. malignant melanoma, renal cell cancer, and brain cancer, exhibit primary resistance, i.e. absence of response on the first exposure, to currently available agents • The presence of inherent drug resistance is thought to be tightly associated with the genomic instability associated with the development of most cancers • For example, mutations in the p53 tumor suppressor gene occur in at least 50% of all human tumors • Preclinical and clinical studies have shown that loss of p53 function leads to resistance to radiation therapy as well as to resistance to a wide range of anticancer agents • Defects in the mismatch repair enzyme family, which are tightly linked to the development of familial and sporadic colorectal cancer, gives rise to resistance to unrelated anticancer agents, including the fluoropyrimidines, the thiopurines, and cisplatin/carboplatin Drug resistance • In contrast to primary resistance, acquired resistance develops in response to exposure to a given anticancer agent • Experimentally, drug resistance can be highly specific to a single drug and is usually based on a specific change in the genetic machinery of a given tumor cell with amplification or increased expression of one or more genes • In other instances, a multidrug-resistant phenotype occurs, associated with increased expression of the MDR1 gene, which encodes a cell surface transporter glycoprotein (P-glycoprotein) • This form of drug resistance leads to enhanced drug efflux and reduced intracellular accumulation of a broad range of structurally unrelated anticancer agents, including the anthracyclines, vinca alkaloids, taxanes, camptothecins, epipodophyllotoxins, and even small molecule inhibitors, such as imatinib Alkylating agents Alkylating agents • Examples • Containing bis(chloroethyl)amines functional group - cyclophosphamide, mechlorethamine, melphalan, and chlorambucil • Nitrosoureas - carmustine (BCNU), lomustine (CCNU), and semustine (methyl-CCNU) • Others – Ifosfamide, thiotepa and busulfan Mechanism of action • Alkylating agents exert their cytotoxic effects via transfer of their alkyl groups to various cellular constituents particularly in the nucleus • The general mechanism of action of these drugs involves intramolecular cyclization to form an ethyleneimonium ion that may directly or through formation of a carbonium ion transfer an alkyl group to a cellular constituent • In addition to alkylation, a secondary mechanism that occurs with nitrosoureas involves carbamoylation of lysine residues of proteins through formation of isocyanates Mechanism of action • The major site of alkylation within DNA is the N7 position of guanine • These interactions can occur on a single strand or on both strands of DNA through cross-linking, as most major alkylating agents are bifunctional, with two reactive groups • Alkylation of guanine can result in miscoding through abnormal base pairing with thymine or in depurination by excision of guanine residues • The latter effect leads to DNA strand breakage through scission of the sugar- phosphate backbone of DNA • Thus, although alkylating agents are not cell cycle specific, cells are most susceptible to alkylation in late G1 and S phases of the cell cycle and express blockage in G2 Resistance • The mechanism of acquired resistance to alkylating agents may involve: 1. Increased capability to repair DNA lesions 2. Decreased transport of the alkylating drug into the cell 3. Increased production of glutathione and glutathione-associated proteins, which are needed to conjugate the alkylating agent 4. Increased glutathione S-transferase activity, which catalyzes the conjugation Adverse effects • The adverse effects usually associated with alkylating agents are generally dose-related and occur primarily in rapidly growing tissues such as bone marrow, the gastrointestinal tract, and the reproductive system • Nausea and vomiting can be a serious issue with a number of these agents • In addition, they have direct vesicant effects and can damage tissues at the site of injection as well as produce systemic toxicity • As a class, alkylating agents are carcinogenic in nature, and there is an increased risk of secondary malignancies, especially acute myelogenous leukemia Cyclophosphamide • Cyclophosphamide is a widely used alkylating agent, • It has high oral bioavailability – it can be administered via the oral and intravenous routes with equal clinical efficacy • It is inactive in its parent form, and must be activated to cytotoxic forms by liver cytochrome P450 mixed-function oxidase system – converts cyclophosphamide to 4-hydroxycyclophosphamide, which is in equilibrium with aldophosphamide • These active metabolites are delivered to both tumor and normal tissue, where nonenzymatic cleavage of aldophosphamide to the cytotoxic forms—phosphoramide mustard and acrolein Cyclophosphamide • Hemorrhagic cystitis is a bladder toxicity that is seen most commonly after administration of cyclophosphamide and Ifosfamide • Acrolein, a metabolite of these agents, is thought to cause a chemical irritation of the bladder mucosa, resulting in bleeding • Prevention – aggressive hydration with subsequent frequent urination, and the administration of the uroprotectant mesna • Mesna acts by binding to acrolein and preventing it from contacting the bladder mucosa Nitrosoureas • These drugs appear to be non-cross-resistant with other alkylating agents; all require biotransformation, which occurs by nonenzymatic decomposition, to metabolites with both alkylating and carbamoylating activities • The nitrosoureas are highly lipid-soluble and are able to cross the blood- brain barrier, making them effective in the treatment of brain tumors • Mechanism of action • Although the majority of alkylations by the nitrosoureas are on the N7 position of guanine in DNA, the critical alkylation responsible for cytotoxicity is on the O6 of guanine, which leads to G-C crosslinks in DNA Nitrosoureas • Pharmacokinetics • After oral administration of lomustine, peak plasma levels of metabolites appear within 1–4 hours; CNS concentrations reach 30– 40% of the activity present in the plasma • Urinary excretion appears to be the major route of elimination from the body • One naturally occurring sugar-containing nitrosourea, streptozocin, is interesting because it has minimal bone marrow toxicity • This agent has activity in the treatment of insulin-secreting islet cell carcinoma of the pancreas Nonclassic alkylating agents Procarbazine • Mechanisms—Procarbazine is a reactive agent that forms hydrogen peroxide, which generates free radicals that cause DNA strand scission Pharmacokinetics • Procarbazine is orally active and penetrates into most tissues, including the cerebrospinal fluid • It is eliminated via hepatic metabolism. Clinical use—The primary use of the drug is as a component of regimens for Hodgkin’s and non-Hodgkin’s lymphoma, and brain tumors Procarbazine Toxicity—Procarbazine is a myelosuppressant and causes gastrointestinal irritation, CNS dysfunction, peripheral neuropathy, and skin reactions • Procarbazine inhibits many enzymes, including monoamine oxidase and those involved in hepatic drug metabolism • Adverse events can occur when procarbazine is given with other MAO inhibitors as well as with sympathomimetic agents and tyramine containing foods • Disulfiram-like reactions have occurred with ethanol • The drug is leukemogenic Nonclassic alkylating agents • Other nonclassic alkylating agents include: 1. Dacarbazine – administered parenterally • Used for malignant melanoma, Hodgkin's lymphoma, soft tissue sarcomas, and neuroblastoma • Main dose-limiting toxicity is myelosuppression, but nausea and vomiting can be severe and in some cases also a potent vesicant 2. Bendamustine Platinum analogs • Mechanism of action • They are thought to exert their cytotoxic effects in the same manner as alkylating agents • As such, they kill tumor cells in all stages of the cell cycle and bind DNA through the formation of intrastrand and interstrand cross-links, thereby leading to inhibition of DNA synthesis and function • The primary binding site is the N7 position of guanine Platinum analogs • Cisplatin • Indications – small cell and small cell lung cancer, esophageal and gastric cancer, head and neck cancer, and genitourinary cancers, particularly testicular, ovarian, and bladder cancer • Extensively cleared by the kidneys and excreted in the urine – dose modification is required in the setting of renal dysfunction • Toxicity – severe nausea and vomiting; nephrotoxicity, peripheral sensory neuropathy, ototoxicity, nerve dysfunction Platinum analogs • Carboplatin (2nd generation platinum analog) • It exhibits significantly less renal toxicity and gastrointestinal toxicity as compared to cicplatin • Its main dose-limiting toxicity is myelosuppression • It has therefore been widely used in transplant regimens to treat refractory hematologicmalignancies • Administration –vigorous intravenous hydration is not required for carboplatin therapy, therefore carboplatin is viewed as an easier agent to administer to patients Platinum analogs • Oxaliplatin (third-generation platinum analog) • Mechanism of action • Identical to those of cisplatin and carboplatin • However, tumors that are resistant to cisplatin or carboplatin on the basis of mismatch repair defects are not cross-resistant to oxaliplatin, and this finding may explain the activity of this platinum compound in colorectal cancer • Indications : colorectal cancer as part of the FOLFOX regimen (5- fluorouracil/leucovorin/oxaliplatin), colon cancer, and other gastrointestinal cancers, such as pancreatic, gastroesophageal, and hepatocellular cancers Platinum analogs - oxaliplatin • Toxicity • Neurotoxicity is the main dose-limiting toxicity and is manifested by a peripheral sensory neuropathy of two types: • Acute form that is often triggered and worsened by exposure to cold, and a chronic form that is dose-dependent • Although this chronic form is cumulative in nature, it tends to be reversible, in sharp contrast to cisplatin-induced neurotoxicity Antimetabolites Antimetabolites • They include: 1. Antifolates 2. Purine antagonists 3. Fluoropyrimidines 4. Deoxycytidine analogs Antifolates • Examples – methotrexate (mtx); pemetrexed • Methotrexate • Mechanism of action • MTX is a folic acid analog that binds with high affinity to the active catalytic site of dihydrofolate reductase (DHFR), interfering with the synthesis of tetrahydrofolate (THF), which serves as the key one-carbon carrier for enzymatic processes involved in de novo synthesis of thymidylate, purine nucleotides, and the amino acids serine and methionine • Inhibition of these various metabolic processes thereby interferes with the formation of DNA, RNA, and key cellular proteins Methotrexate • MTX mechanism continued… • Intracellular formation of polyglutamate metabolites, with the addition of up to 5–7 glutamate residues, is critically important for the therapeutic action of MTX, and this process is catalyzed by the enzyme folylpolyglutamate synthase (FPGS) • MTX polyglutamates are selectively retained within cancer cells, and they display increased inhibitory effects on enzymes involved in de novo purine nucleotide and thymidylate biosynthesis, making them important determinants of MTX's cytotoxic action Resistance to MTX 1. Decreased drug transport via the reduced folate carrier or folate receptor protein 2. Decreased formation of cytotoxic MTX polyglutamates 3. Increased levels of the target enzyme DHFR through gene amplification and other genetic mechanisms 4. Altered DHFR protein with reduced affinity for MTX 5. Recent studies have suggested that decreased accumulation of drug through activation of the multidrug resistance transporter P170 glycoprotein may also result in drug resistance Methotrexate • Administration and pharmacokinetics • MTX is administered by the intravenous, intrathecal, or oral route • However, oral bioavailability is saturable and erratic at doses greater than 25 mg/m2 • Renal excretion is the main route of elimination – dose modification is required in the setting of renal dysfunction • Care must also be taken when MTX is used in the presence of drugs such as aspirin, penicillin, cephalosporins, and nonsteroidal anti- inflammatory agents, as they inhibit the renal excretion of MTX Methotrexate • The biologic effects of MTX can be reversed by administration of the reduced folate leucovorin (5-formyltetrahydrofolate) or by L- leucovorin, which is the active enantiomer • Leucovorin rescue is used in conjunction with high-dose MTX therapy to rescue normal cells from undue toxicity, and it has also been used in cases of accidental drug overdose • Toxicity - Mucositis, diarrhea, myelosuppression with neutropenia and thrombocytopenia Fluoropyrimidines • 5-Fluorouracil • Mechanism of action • 5-Fluorouracil (5-FU) is inactive in its parent form and requires activation via a complex series of enzymatic reactions to ribosyl and deoxyribosyl nucleotide metabolites. One of these metabolites, 5- fluoro-2'-deoxyuridine-5'-monophosphate (FdUMP), forms a covalently ternary complex with the enzyme thymidylate synthase and the reduced folate 5,10-methylenetetrahydrofolate, a reaction critical for the de novo synthesis of thymidylate • This results in inhibition of DNA synthesis through "thymineless death.“ 5-Fluorouracil • Mechanism continued • 5-FU is converted to 5-fluorouridine-5'-triphosphate (FUTP), which is then incorporated into RNA, where it interferes with RNA processing and mRNA translation • 5-FU is also converted to 5-fluorodeoxyuridine-5'-triphosphate (FdUTP), which can be incorporated into cellular DNA, resulting in inhibition of DNA synthesis and function • Thus, the cytotoxicity of 5-FU is thought to be the result of combined effects on both DNA- and RNA-mediated events 5-Fluorouracil • Pharmacokinetics and administration • 5-FU is normally administered intravenously with a half-life of 10–15 minutes • The clinical activity of this drug is highly schedule-dependent and because of its extremely short half-life, standard schedules of administration should be followed • Up to 80–85% of an administered dose of 5-FU is catabolized by the enzyme dihydropyrimidine dehydrogenase • A pharmacogenetic syndrome that involves partial or complete deficiency of the DPD enzyme, and in this setting, which is seen in up to 5% of all cancer patients, severe toxicity in the form of myelosuppression, diarrhea, nausea and vomiting, and neurotoxicity, has been observed 5-Fluorouracil • Indications • Colorectal cancer • Cancers of the breast, stomach, pancreas, esophagus, liver, head and neck, and anus • Toxicity • Major toxicities include myelosuppression, gastrointestinal toxicity in the form of mucositis and diarrhea, skin toxicity manifested by the hand-foot syndrome, and neurotoxicity Leucovorin • Mechanism of leucovorin in FOLFOX regimen • Enhances the effects of 5-fluorouracil’s metabolite (5-flurouridylic acid acid) by stabilizing its bond with thymidilate synthase to inhibit DNA and RNA synthesis Capecitabine • Capecitabine is a fluoropyrimidine carbamate prodrug with 70–80% oral bioavailability • It is metabolized first in the liver then at the tumour site to 5-FU • Indications: metastatic breast cancer, colon cancer, colorectal cancer (XELOX regimen) • Toxicity • The main toxicities of capecitabine include diarrhea and the hand-foot syndrome • While myelosuppression, nausea and vomiting, and mucositis are also observed with this agent, the incidence is significantly less than that seen with intravenous 5-FU Deoxycytidine analogs • Examples – Cytarabine, gemcitabine • Cytarabine • Cytarabine (ara-C) is an S phase-specific antimetabolite that is converted by deoxycytidine kinase to the 5'-mononucleotide (ara-CMP) • Ara-CMP is further metabolized to the diphosphate and triphosphate metabolites, and the ara-CTP triphosphate is felt to be the main cytotoxic metabolite • Ara-CTP competitively inhibits DNA polymerase alpha- and DNA polymerase beta-, thereby resulting in blockade of DNA synthesis and DNA repair, respectively Cytarabine • Pharmacokinetics and administration • After intravenous administration, the drug is cleared rapidly, with most of an administered dose being deaminated to inactive forms • The clinical activity of this drug is highly schedule-dependent and because of its rapid degradation, it must be given by continuous infusion over a 5–7 day period Cytarabine • Indications • Its activity is limited exclusively to hematologic malignancies, including acute myelogenous leukemia and non-Hodgkin's lymphoma. This agent has absolutely no activity in solid tumors • Toxicities include myelosuppression, mucositis, nausea and vomiting, and neurotoxicity when high-dose therapy is administered • Cytarabine induced kerato-conjunctivitis has been known to occur in patients receiving high dose (>1000 mg/m2) or multiday regimens; it can be prevented by prophylactic administration of corticosteroid eye drops (every 4-6 hours up to 48 hours after last cytarabine administration) Purine antagonists • 6-Thiopurines • Examples - 6-Mercaptopurine (6-MP), 6-Thioguanine • 6-Mercaptopurine (6-MP) • Indications - This agent is used primarily in the treatment of childhood acute leukemia, and a closely related analog, azathioprine, is used as an immunosuppressive agent 6-Mercaptopurine (6-MP) • Mechanism of action • 6-MP is inactive in its parent form and must be metabolized by hypoxanthine-guanine phosphoribosyl transferase (HGPRT) to form the monophosphate nucleotide 6-thioinosinic acid, which in turn inhibits several enzymes of de novo purine nucleotide synthesis • The monophosphate form is eventually metabolized to the triphosphate form, which can then get incorporated into both RNA and DNA 6-Mercaptopurine (6-MP) • Metabolism - 6-MP is converted to an inactive metabolite (6-thiouric acid) by an oxidation reaction catalyzed by xanthine oxidase • The purine analog allopurinol, a potent xanthine oxidase inhibitor, is frequently used as a supportive care measure in the treatment of acute leukemias to prevent the development of hyperuricemia that often occurs with tumor cell Lysis • Because allopurinol inhibits xanthine oxidase, simultaneous therapy with allopurinol and 6-MP would result in increased levels of 6-MP, thereby leading to excessive toxicity • In this setting, the dose of mercaptopurine must be reduced by 50–75% • In contrast, such an interaction does not occur with 6-TG, which can be used in full doses with allopurinol. 6-Mercaptopurine (6-MP) • The thiopurines are also metabolized by the enzyme thiopurine methyltransferase (TPMT), in which a methyl group is attached to the thiopurine ring • There is a pharmacogenetic syndrome in which there is partial or complete deficiency of this enzyme • Patients with this genotype are at increased risk for developing severe toxicities in the form of myelosuppression and gastrointestinal toxicity with mucositis and diarrhea Fludarabine • Mechanism of action • Fludarabine phosphate is rapidly dephosphorylated to 2-fluoro- arabinofuranosyladenosine and then phosphorylated intracellularly by deoxycytidine kinase to the triphosphate form • The triphosphate metabolite interferes with the processes of DNA synthesis and DNA repair through inhibition of DNA polymerase alpha and DNA polymerase beta • The triphosphate form can also be directly incorporated into DNA, resulting in inhibition of DNA synthesis and function • The diphosphate metabolite of fludarabine inhibits ribonucleotide reductase, leading to inhibition of essential deoxyribonucleotide triphosphates Fludarabine • Indications – Low-grade non-Hodgkin's lymphoma and chronic lymphocytic leukemia (CLL), acute myeloid leukemia • PK and administration - It is given parenterally and up to 25–30% of parent drug is excreted in the urine Fludarabine • Toxicity • The main dose-limiting toxicity is myelosuppression • This agent is a potent immunosuppressant with inhibitory effects on CD4 and CD8 T cells. Patients are at increased risk for opportunistic infections, including fungi, herpes, and Pneumocystis jiroveci pneumonia (PCP) • Patients should receive PCP prophylaxis with trimethoprim-sulfamethoxazole (double strength) at least three times a week, and this should continue for up to 1 year after stopping fludarabine therapy • The agent cladribine has similar properties to fludarabine including prolonged immunosuppression characterized by low levels of CD4 and CD8 T cells Natural product cancer chemotherapy drugs Vinca alkaloids • These include vinblastine, vincristine and vinorelbine • Vinblastine • Vinblastine is an alkaloid derived from the periwinkle plant Vinca rosea • Indications - Hodgkin's and non-Hodgkin's lymphomas, breast cancer, and germ cell cancer • Mechanism of action - involves inhibition of tubulin polymerization, which disrupts assembly of microtubules, an important part of the cytoskeleton and the mitotic spindle • This inhibitory effect results in mitotic arrest in metaphase, bringing cell division to a halt, which then leads to cell death Vinblastine • Pharmacokinetics • Vinblastine and other vinca alkaloids are metabolized by the liver P450 system, and the majority of the drug is excreted in feces via the biliary system • As such, dose modification is required in the setting of liver dysfunction Vinblastine • Toxicity • Adverse effects include nausea and vomiting, bone marrow suppression, and alopecia • This agent is also a potent vesicant, and care must be taken in its administration Vincristine • Vincristine is an alkaloid derivative of Vinca rosea and has similar pharmacologic properties as vinblastine but its toxicity profile is different • Indications – ALL, the lymphomas, pediatric tumors including rhabdomyosarcoma, neuroblastoma, Ewing's sarcoma, and Wilms' tumor Vincristine • Toxicity • The main dose-limiting toxicity is neurotoxicity, usually expressed as a peripheral sensory neuropathy, although autonomic nervous system dysfunction with orthostatic hypotension, urinary retention, paralytic ileus, or constipation, cranial nerve palsies, ataxia, seizures, and coma have been observed • While myelosuppression occurs, it is generally milder and much less significant than with vinblastine • The other potential adverse effect that can develop is the syndrome of inappropriate secretion of antidiuretic hormone (SIADH) Taxanes & related drugs • Paclitaxel is an alkaloid ester derived from the Pacific yew (Taxus brevifolia) and the European yew (Taxus baccata) • Indications - broad range of solid tumors, including ovarian, advanced breast, non-small cell and small cell lung, head and neck, esophageal, prostate, and bladder cancers and AIDS-related Kaposi's sarcoma • Mechanism of action • The drug functions as a mitotic spindle poison through high-affinity binding to microtubules with enhancement of tubulin polymerization • This promotion of microtubule assembly by paclitaxel occurs in the absence of microtubule-associated proteins and guanosine triphosphate and results in inhibition of mitosis and cell division Paclitaxel • Pharmacokinetics • It is metabolized extensively by the liver P450 system, and nearly 80% of the drug is excreted in feces via the hepatobiliary route • Dose reduction is required in the setting of liver dysfunction • Toxicity • Hypersensitivity reactions may be observed in up to 5% of patients, but the incidence is significantly reduced by premedication with dexamethasone, diphenhydramine, and an H2 blocker • Other adverse effects include myelosuppression, peripheral sensory neuropathy, nausea, vomiting, hypotension, arrhythmias Paclitaxel • A novel albumin-bound paclitaxel formulation (Abraxane) is approved for use in metastatic breast cancer • In contrast to paclitaxel, this formulation is not associated with hypersensitivity reactions, and premedication to prevent such reactions is not required • Moreover, this agent has significantly reduced myelosuppressive effects compared with paclitaxel, and the neurotoxicity that results appears to be more readily reversible than is typically observed with paclitaxel Docetaxel • Docetaxel is a semisynthetic taxane derived from the European yew tree • Its mechanism of action, metabolism, and elimination are identical to those of paclitaxel • Indications - advanced breast cancer, non-small cell lung cancer, head and neck cancer, small cell lung cancer, gastric cancer, advanced platinum-refractory ovarian cancer, and bladder cancer • Toxicity - hypersensitivity neurotoxicity, fluid retention, myelosuppression with neutropenia Ixabepilone • Although not a taxane, ixabepilone is a novel microtubule inhibitor indicated for metastatic breast cancer • Of note, this agent continues to have activity in drug-resistant tumors that overexpress P-glycoprotein or tubulin mutations • The main adverse effects include myelosuppression, hypersensitivity reactions, and neurotoxicity in the form of peripheral sensory neuropathy Epipodophyllotoxins • Examples – etoposide and teniposide • Etoposide is a semisynthetic derivative of podophyllotoxin, which is extracted from the mayapple root (Podophyllum peltatum) • Indications - Etoposide has clinical activity in germ cell cancer, small cell and non-small cell lung cancer, Hodgkin's and non-Hodgkin's lymphomas, and gastric cancer • Mechanism of action - involves inhibition of topoisomerase II, which results in DNA damage through strand breakage induced by the formation of a ternary complex of drug, DNA, and enzyme Etoposide • Administration and pharmacokinetics • The oral bioavailability is about 50%, requiring the oral dose to be twice that of an intravenous dose • Up to 30–50% of drug is excreted in the urine, and dose reduction is required in the setting of renal dysfunction Camptothecins • The camptothecins are natural products derived from the Camptotheca acuminata tree • Mechanism of action - they inhibit the activity of topoisomerase I, the key enzyme responsible for cutting and religating single DNA strands • Inhibition of this enzyme results in DNA damage • Topotecan • Indications - advanced ovarian cancer, small cell lung cancer • PK - The main route of elimination is renal excretion, and dosage must be adjusted in patients with renal impairment Irinotecan • Irinotecan is a prodrug that is converted mainly in the liver by the carboxylesterase enzyme to the SN-38 metabolite, which is 1000-fold more potent as an inhibitor of topoisomerase I than the parent compound • Indications - metastatic colorectal cancer • PK - In contrast to topotecan, irinotecan and SN-38 are mainly eliminated in bile and feces, and dose reduction is required in the setting of liver dysfunction Irinotecan • Toxicity • Myelosuppression and diarrhea are the two most common adverse events • There are two forms of diarrhea: an early form that occurs within 24 hours after administration and is thought to be a cholinergic event effectively treated with atropine, and a late form that usually occurs 2–10 days after treatment • The late diarrhea can be severe, leading to significant electrolyte imbalance and dehydration in some cases Antitumor antibiotics Anthracyclines • The anthracycline antibiotics, isolated from Streptomyces peucetius var caesius • Examples – doxorubicin, Daunorubicin, idarubicin, epirubicin • Mechanism of action – anthracyclines intercalate between DNA base pairs, inhibit topoisomerase II, and generate free radicals; They block the synthesis of RNA and DNA and cause DNA strand scission; Membrane disruption also occurs to alter fluidity and ion transport • The free radical mechanism is the cause of the cardiotoxicity associated with the anthracyclines Anthracyclines • Administration and pharmacokinetics • Administered via the intravenous route • The anthracyclines are metabolized extensively in the liver; up to 50% of drug is eliminated in the feces via biliary excretion, and dose reduction is required in the setting of liver dysfunction • Although anthracyclines are usually administered on an every-3-week schedule, alternative schedules such as low-dose weekly or 72–96 hour continuous infusions have been shown to yield equivalent clinical efficacy with reduced toxicity Anthracyclines • Clinical use • Doxorubicin – is very widely used in combination with other anticancers in many adult and pediatric solid tumors and lymphomas • Daunorubicin and idarubicin are used in acute myeloid leukemia Anthracyclines • Toxicity • The main dose-limiting toxicity of all anthracyclines is myelosuppression, with neutropenia more commonly observed than thrombocytopenia • In some cases, mucositis is dose-limiting • Cardiotoxicity • Two forms are observed: 1. The acute form occurs within the first 2–3 days and presents as arrhythmias and conduction abnormalities, other electrocardiographic changes, pericarditis, and myocarditis. This form is usually transient and in most cases is asymptomatic Anthracycline toxicity 2. The chronic form results in a dose-dependent, dilated cardiomyopathy associated with heart failure • The chronic cardiac toxicity appears to result from increased production of free radicals within the myocardium • This effect is rarely seen at total doxorubicin dosages below 500–550 mg/m2 (the cumulative maximum lifetime dose for epirubicin is 900 mg/m2) • Use of lower weekly doses or continuous infusions of doxorubicin appear to reduce the incidence of cardiac toxicity Anthracycline toxicity • Chronic cardiotoxicity cont… • In addition, treatment with the iron-chelating agent dexrazoxane is currently approved to prevent or reduce anthracycline-induced cardiotoxicity in women with metastatic breast cancer who have received a total cumulative dose of doxorubicin of 300 mg/m2 • The anthracyclines can also produce a "radiation recall reaction," with erythema and desquamation of the skin observed at sites of prior radiation therapy Mitoxantrone • Mitoxantrone is an anthracene compound whose structure resembles the anthracycline ring • Mechanisms - It binds to DNA to produce strand breakage and inhibits both DNA and RNA synthesis • Clinical use - It is indicated for prostate cancer, non-Hodgkin lymphoma, adult and pediatric acute myeloid leukemia Mitoxantrone • Toxicity • Myelosuppression with leukopenia is the dose-limiting toxicity, and mild nausea and vomiting, mucositis, and alopecia also occur • Although the drug is thought to be less cardiotoxic than doxorubicin, both acute and chronic cardiac toxicity are reported • A blue discoloration of the fingernails, sclera, and urine is observed 1–2 days after drug administration Mitomycin • Mitomycin (mitomycin C) is an antibiotic isolated from Streptomyces caespitosus • Mechanism - It undergoes metabolic activation through liver enzyme- mediated reduction to generate an alkylating agent that cross-links DNA • Hypoxic tumor stem cells of solid tumors exist in an environment conducive to reductive reactions and are more sensitive to the cytotoxic actions of mitomycin than normal cells and oxygenated tumor cells • It is active in all phases of the cell cycle, and is the best available drug for use in combination with radiation therapy to attack hypoxic tumor cells. • Clinical uses – squamous cell carcinomas of the anus, cervix and for breast, gastric, and pancreatic cancer Mitomycin • Administration and pharmacokinetics • Mitomycin is given intravenously and is rapidly cleared via hepatic metabolism • Intravesically administered mitomycin for superficial bladder cancer is not absorbed systemically hence no systemic toxicity • Toxicity - Myelosuppression, mucositis, anorexia and fatigue, nausea/vomiting, hemolytic-uremic syndrome manifested as microangiopathic hemolytic anemia, thrombocytopenia, and renal failure Bleomycin • Mechanism of action • Bleomycin is a small peptide that contains a DNA-binding region and an iron-binding domain at opposite ends of the molecule. It acts by binding to DNA, which results in single-strand and double-strand breaks following free radical formation, and inhibition of DNA biosynthesis • The fragmentation of DNA is due to oxidation of a DNA-bleomycin- Fe(II) complex and leads to chromosomal aberrations • Bleomycin is a cell cycle-specific drug that causes accumulation of cells in the G2 phase of the cell cycle Bleomycin • Clinical uses • Hodgkin's and non-Hodgkin's lymphomas, germ cell tumor, head and neck cancer, and squamous cell cancer of the skin, cervix, and vulva • Administration and pharmacokinetics • One advantage of this agent is that it can be given subcutaneously, intramuscularly, or intravenously • Elimination of bleomycin is mainly via renal excretion; dose modification is recommended in patients with renal dysfunction Bleomycin • Toxicity • Pulmonary toxicity is dose-limiting for bleomycin and usually presents as pneumonitis with cough, dyspnea, dry inspiratory crackles on physical examination, and infiltrates on chest x-ray • The incidence of pulmonary toxicity is increased in patients older than 70 years of age, in those who receive cumulative doses greater than 400 units, in those with underlying pulmonary disease, and in those who have received prior mediastinal or chest irradiation • Other toxicities include skin hyperpigmentation, keratosis and stomatitis Miscellaneous anticancer drugs Miscellaneous anticancer drugs • A large number of newer anticancer drugs that do not fit traditional categories will be covered here including targeted therapies • Targeted cancer therapies are drugs or other substances that block the growth and spread of cancer by interfering with specific molecules ("molecular targets") that are involved in the growth, progression, and spread of cancer Tyrosine kinase inhibitors (TKIs) • Imatinib • Mechanism – it is an inhibitor of the tyrosine kinase domain of the Bcr-Abl oncoprotein and prevents phosphorylation of the kinase substrate by ATP • It is indicated for the treatment of chronic myelogenous leukemia (CML), a pluripotent hematopoietic stem cell disorder characterized by the t(9:22) Philadelphia chromosomal translocation • This translocation results in the Bcr-Abl fusion protein, the causative agent in CML, and is present in up to 95% of patients with this disease • This agent also inhibits other receptor tyrosine kinases for platelet-derived growth factor receptor (PDGFR), stem cell factor, and c-kit Imatinib • Clinical use – Used in CML (chronic phase and blast crisis); Imatinib is also effective also for treatment of gastrointestinal stromal tumors expressing the c-kit (CD117) tyrosine kinase • Administration and pharmacokinetics • Imatinib is well absorbed orally, and it is metabolized in the liver, with elimination of metabolites occurring mainly in feces via biliary excretion • Toxicity - diarrhea, myalgia, fluid retention, and congestive heart failure Other TKIs • Dasatinib and nilotinib are TKIs with similar mechanisms and uses as Imatinib; both are active against Imatinib resistant CML • Pharmacokinetics of TKIs • Imatinib, dasatinib, and nilotinib are all metabolized in the liver, mainly by the CYP3A4 liver microsomal enzyme. A large fraction of each drug is eliminated in feces via the hepatobiliary route • It is important to review the patient's current list of prescription and nonprescription drugs because these agents have potential drug-drug interactions, especially with those that are also metabolized by the CYP3A4 system • In addition, patients should avoid grapefruit products (CYP3A4 inhibitor) and the use of St. John's wort (CYP3A4 inducer), as they may alter the clinical activity of these small molecule inhibitors Growth factor receptor inhibitors • Trastuzumab, a monoclonal antibody, recognizes a surface protein in breast cancer cells that overexpress the (human epidermal growth factor receptor 2) HER-2/neu receptor for epidermal growth factor (25-30%) • By binding to the HER-2/neu protein, trastuzumab inhibits growth of tumour cells and mediates antibody dependent cellular cytotoxicity (ADCC) in cancer cells overexpressing HER-2/neu • Acute toxicity of this antibody includes nausea and vomiting, chills, fevers, and headache • Trastuzumab may cause cardiac dysfunction, including heart failure Growth factor receptor inhibitors • Several other drugs inhibit an epidermal growth factor receptor (EGFR) that is distinct from the HER-2/neu receptor for the epidermal growth factor that is targeted by trastuzumab • The EGFR regulates signaling pathways involved in cellular proliferation, invasion and metastasis, and angiogenesis • It is also implicated in inhibiting the cytotoxic activity of some anticancer drugs and radiation therapy Growth factor receptor inhibitors • Cetuximab is a chimeric monoclonal antibody directed against the extracellular domain of the EGFR, and it is presently approved for use in combination with irinotecan for metastatic colon cancer in the refractory setting or as monotherapy in patients who are deemed to be irinotecan-refractory • Cetuximab is of the G1 isotype, its antitumor activity may be mediated in part by immunologic-mediated mechanisms • This agent is also approved for use in combination with radiation therapy in patients with locally advanced head and neck cancer Cetuximab • Administration/pharmacokinetics • Although this antibody was initially approved to be administered on a weekly schedule, pharmacokinetic studies have shown that an every- 2-week schedule provides the same level of clinical activity as the weekly schedule • Toxicity • Cetuximab is well tolerated, with the main adverse effects being an acneiform skin rash, hypersensitivity infusion reaction, and hypomagnesemia Panitumumab • Panitumumab is a fully human monoclonal antibody directed against the EGFR and works through inhibition of the EGFR signaling pathway • In contrast to cetuximab, this antibody is of the G2 isotype, and as such, it would not be expected to exert any immunologic-mediated effects • Presently, panitumumab is approved for patients with refractory metastatic colorectal cancer who have been treated with all other active agents • As this is a fully human antibody, infusion-related reactions are seen only rarely, and acneiform skin rash and hypomagnesemia are the two main adverse effects associated with this agent. Gefitinib & Erlotinib • Gefitinib and erlotinib are small molecule inhibitors of the tyrosine kinase domain associated with the EGFR, and both are used in the treatment of non-small cell lung cancer that is refractory to at least one prior chemotherapy regimen • Patients who are nonsmokers and who have a bronchoalveolar histologic subtype appear to be more responsive to these agents • In addition, erlotinib has been approved for use in combination with gemcitabine for the treatment of advanced pancreatic cancer Gefitinib & Erlotinib • Pharmacokinetics • Both agents are metabolized in the liver by the CYP3A4 enzyme system, and elimination is mainly hepatic with excretion in feces • Caution must be taken when using these agents with drugs that are also metabolized by the liver CYP3A4 system, such as phenytoin and warfarin, and the use of grapefruit products should be avoided • Toxicity • An acneiform skin rash, diarrhea, and anorexia and fatigue are the most common adverse effects observed with these small molecules Agents acting on VEGF signalling • The vascular endothelial growth factor (VEGF) is one of the most important angiogenic growth factors • The growth of both primary and metastatic tumors requires an intact vasculature • As a result, the VEGF-signaling pathway represents an attractive target for cancer treatment Agents acting on VEGF signalling • Bevacizumab is a recombinant humanized monoclonal antibody that targets all forms of VEGF-A; this antibody binds to and prevents VEGF-A from interacting with the target VEGF receptors • Clinical uses – metastatic colorectal cancer, non-small lung cancer; breast cancer • Use for age-related macular degeneration (ARMD) • VEGF has been demonstrated in human specimens of choroidal neovascularization (CNV) and animal models have confirmed that this protein is capable of inducing CN • Therefore, targeted inhibition of VEGF is a reasonable approach for the treatment of CNV in ARMD Bevacizumab • Toxicity • One potential advantage of this antibody is that it does not appear to exacerbate the toxicities typically observed with cytotoxic chemotherapy • The main safety concerns associated with bevacizumab include hypertension, an increased incidence of arterial thromboembolic events (transient ischemic attack, stroke, angina, and myocardial infarction), wound healing complications and gastrointestinal perforations, and proteinuria Sorafenib & Sunitinib • Sorafenib is a small molecule that inhibits multiple receptor tyrosine kinases (RTKs), especially VEGF-R2 and VEGF-R3, platelet-derived growth factor- (PDGFR-), and raf kinase • It was initially approved for advanced renal cell cancer, and recently was approved for advanced hepatocellular cancer • Sunitinib is similar to sorafenib in that it inhibits multiple RTKs, although the specific types are somewhat different; they include PDGFR- and PDGFR-, VEGFR-R1, VEGF-R2, VEGF-R3, and c-kit • It is approved for the treatment of advanced renal cell cancer and for the treatment of gastrointestinal stromal tumors (GIST) after disease progression on or with intolerance to imatinib Sorafenib & Sunitinib • Pharmacokinetics • Both sorafenib and sunitinib are metabolized in the liver by the CYP3A4 system, and elimination is primarily hepatic with excretion in feces • Each of these agents has potential interactions with drugs that are also metabolized by the CYP3A4 system, especially warfarin • In addition, patients should avoid grapefruit products and the use of St. John's wort, as they may alter the clinical activity of these agents Sorafenib, & Sunitinib • Toxicity • Hypertension, bleeding complications, and fatigue are the most common adverse effects seen with both agents • With respect to sorafenib, skin rash and the hand-foot syndrome are observed in up to 30–50% of patients • For sunitinib, there is also an increased risk of cardiac dysfunction, which, in some cases, can lead to congestive heart failure Asparaginase • Asparaginase (L -asparagine amidohydrolase) is an enzyme used to treat childhood ALL. • Mechanism - It hydrolyzes circulating L-asparagine to aspartic acid and ammonia; because tumor cells in ALL lack asparagine synthetase, they require an exogenous source of L-asparagine • Thus, depletion of L-asparagine results in effective inhibition of protein synthesis • In contrast, normal cells can synthesize L-asparagine and thus are less susceptible to the cytotoxic action of asparaginase Asparaginase • Toxicity • The main adverse effect of this agent is a hypersensitivity reaction manifested by fever, chills, nausea and vomiting, skin rash, and urticarial • Severe cases can present with bronchospasm, respiratory failure, and hypotension • Other adverse effects include an increased risk of both clotting and bleeding as a result of alterations in various clotting factors, pancreatitis, and neurologic toxicity with lethargy, confusion, hallucinations, and in severe cases, coma Retinoic acid derivatives • Tretinoin (all-trans-retinoic acid) is active in patients with acute promyelocytic leukemia (APL) through the induction of terminal differentiation, in which the leukemic promyelocytes lose their ability to proliferate • Clinical use - this agent is approved for use in APL following progression or relapse with anthracycline-based chemotherapy and for patients in whom anthracycline-based chemotherapy is contraindicated Retinoic acid derivatives • Toxicity - vitamin A toxicity manifesting as headache, fever, dry skin and mucous membranes, skin rash, pruritus, and conjunctivitis; retinoic acid syndrome (fever, leukocytosis, dyspnea, weight gain, diffuse pulmonary infiltrates, and pleural or pericardial effusions); increased serum cholesterol and triglyceride levels; central nervous system toxicity in the form of dizziness, anxiety, depression, confusion, and agitation; abdominal pain and diarrhea; and transient elevations in liver function tests Arsenic trioxide (As2O3) • Mechanism • It functions by inducing differentiation through degradation of the chimeric PML/RAR- protein • In addition, it induces apoptosis through a mitochondrion-dependent process, resulting in subsequent release of cytochrome C with caspase activation • Clinical use - Arsenic trioxide is used for induction of remission in patients with APL with the t(15:17) chromosomal translocation refractory to or relapsed following first-line therapy with all-trans- retinoic acid- and anthracycline-based chemotherapy Arsenic trioxide (As2O3) • Pharmacokinetics/administration • This drug is administered via the intravenous route, and it is widely distributed in the body • Toxicity • The main toxicities are fatigue, electrocardiographic changes with QT prolongation, arrhythmias, and a syndrome characterized by fever, dyspnea, skin rash, fluid retention, and weight gain Rituximab • Rituximab is a monoclonal antibody that binds to a surface protein in non-Hodgkin’s lymphoma cells and induces complement-mediated lysis, direct cytotoxicity, and induction of apoptosis • It is currently used with conventional anticancer drugs (eg, cyclophosphamide plus vincristine plus prednisone) in low-grade lymphomas • Rituximab is associated with hypersensitivity reactions and myelosuppression (with profound neutropenia) Proteasome Inhibitors • Bortezomib and carfilzomib are inhibitors of the chymotrypsin like activity of the 26S proteasome in mammalian cells • The 26S proteasome is a large protein complex that degrades ubiquitinated proteins, such as cyclin-dependent kinases • Inhibition results in down-regulation of the nuclear factor kappa B (NF-κB) signaling pathway • Clinical use – multiple myeloma Proteasome Inhibitors • Pharmacokinetics – eliminated by the liver; it is advantageous for use in patients with renal impairment as a result of multiple myeloma • Toxicity • Adverse effects include peripheral neuropathy, thrombocytopenia, heart failure, and hypotension Hormonal anticancer agents Gonadal hormone antagonists • Tamoxifen, a selective estrogen receptor modulator blocks the binding of estrogen to receptors of estrogen-sensitive cancer cells in breast tissue; other examples include torimefene, raloxifene • Clinical use - The drug is used in receptor-positive breast carcinoma and has been shown to have a preventive effect in women at high risk for breast cancer • Toxicity • Because it has agonist activity in the endometrium, tamoxifen increases the risk of endometrial hyperplasia and neoplasia • Other adverse effects include nausea and vomiting, hot flushes, vaginal bleeding, and venous thrombosis Gonadal hormone antagonists • Flutamide is an androgen receptor antagonist used in prostatic carcinoma • Adverse effects include gynecomastia, hot flushes, and hepatic dysfunction Gonadotropin-Releasing Hormone (GnRH) Analogs • Leuprolide, goserelin, and nafarelin are GnRH agonists, effective in prostatic carcinoma • When administered in constant doses so as to maintain stable blood levels, they inhibit release of pituitary luteinizing hormone (LH) and follicle-stimulating hormone (FSH) • Physiologic secretion of GnRH is pulsatile; constant serum levels cause down-regulation of GnRH recpetors and a drop in gonadotropin levels leading to reduced sex hormone levels to castrate levels Gonadotropin-Releasing Hormone (GnRH) Analogs • Toxicity • Leuprolide may cause bone pain, gynecomastia, hematuria, impotence, and testicular atrophy