Lippincott Chapter 46: Anticancer Drugs Flashcards

1
Q

ANTIMETABOLITES

A

Azacitidine VIDAZA
Capecitabine XELODA
Cladribine LEUSTATIN
Cytarabine CYTOSINE ARABINOSIDE (ARA-C)
Fludarabine FLUDARA
5-Fluorouracil ADRUCIL
Gemcitabine GEMZAR
6-Mercaptopurine PURINETHOL
Methotrexate (MTX) TREXALL
Pemetrexed ALIMTA
Pralatrexate FOLOTYN

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

ANTIBIOTICS

A

Bleomycin BLENOXANE
Daunorubicin CERUBIDINE
Doxorubicin ADRIAMYCIN
Epirubicin ELLENCE
Idarubicin IDAMYCIN
Mitoxantrone

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

ALKYLATING AGENTS

A

Busulfan MYLERAN
Carmustine BICNU
Chlorambucil LEUKERAN
Cyclophosphamide CYTOXAN
Dacarbazine DTIC-DOME
Ifosfamide IFEX
Lomustine CEENU
Melphalan ALKERAN
Temozolomide TEMODAR

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

MICROTUBULE INHIBITORS

A

Docetaxel TAXOTERE
Paclitaxel TAXOL
Vinblastine
Vincristine VINCASAR PFS
Vinorelbine NAVELBINE

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

STEROID HORMONES AND THEIR
ANTAGONISTS

A

Anastrozole ARIMIDEX
Bicalutamide CASODEX
Estrogens VARIOUS
Exemestane AROMASIN
Flutamide
Goserelin ZOLADEX
Letrozole FEMARA
Leuprolide LUPRON
Megestrol acetate MEGACE
Nilutamide NILANDRON
Prednisone
Tamoxifen
Triptorelin TRELSTAR
Raloxifene
Fulvestrant FASLODEX

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

MONOCLONAL ANTIBODIES

A

Bevacizumab AVASTIN
Cetuximab ERBITUX
Rituximab RITUXAN
Trastuzumab HERCEPTIN

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

TYROSINE KINASE INHIBITORS

A

Dasatinib
TARCEVA
Imatinib GLEEVEC
Nilotinib TASIGNA
Sorafenib NEXAVAR
Erlotinib
Sunitinib

SPRYCEL
SUTENT

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

OTHERS

A

Abiraterone ZYTIGA
Carboplatin
Cisplatin PLATINOL
Enzalutamide XTANDI
Interferons PEG-INTRON
Irinotecan CAMPTOSAR
Oxaliplatin ELOXATIN
Procarbazine MATULANE
Topotecan HYCAMTIN
Asparaginase ERWINAZE
Etoposide TOPOSAR, VEPESID

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

ANTIMETABOLITES

Methotrexate, pemetrexed, and pralatrexate

A

Antimetabolites are structurally related to normal compounds that exist
within the cell (Figure 46.8). They generally interfere with the availability
of normal purine or pyrimidine nucleotide precursors, either by inhibit-
ing their synthesis or by competing with them in DNA or RNA synthesis.
Their maximal cytotoxic effects are in S-phase and are, therefore, cell
cycle specific.
A. Methotrexate, pemetrexed, and pralatrexate
The vitamin folic acid plays a central role in a variety of metabolic
reactions involving the transfer of one-carbon units and is essential
for cell replication. Folic acid is obtained mainly from dietary sources
and from that produced by intestinal flora. Methotrexate [meth-oh-
TREK-sate] (MTX), pemetrexed [pem-e-TREX-ed], and pralatrexate
[pral-a-TREX-ate] are antifolate agents.
1. Mechanism of action: MTX is structurally related to folic acid and
acts as an antagonist of the vitamin by inhibiting mammalian dihy-
drofolate reductase (DHFR), the enzyme that converts folic acid to
its active, coenzyme form, tetrahydrofolic acid (FH4
) (Figure 46.9).
The inhibition of DHFR can only be reversed by a 1000-fold excess
of the natural substrate, dihydrofolate (FH2
), or by administration of
leucovorin, which bypasses the blocked enzyme and replenishes
the folate pool (Figure 46.9). [Note: Leucovorin, or folinic acid, is
the N5
-formyl group–carrying form of FH4
.] MTX is specific for the
S-phase of the cell cycle. Pemetrexed is an antimetabolite similar
in mechanism to methotrexate. However, in addition to inhibiting
DHFR, it also inhibits thymidylate synthase and other enzymes
involved in folate metabolism and DNA synthesis. Pralatrexate is a
newer antimetabolite that also inhibits DHFR.

  1. Therapeutic uses: MTX, usually in combination with other drugs,
    is effective against acute lymphocytic leukemia, Burkitt lymphoma
    in children, breast cancer, bladder cancer, and head and neck car-
    cinomas. In addition, low-dose MTX is effective as a single agent
    against certain inflammatory diseases, such as severe psoriasis
    and rheumatoid arthritis, as well as Crohn disease. All patients
    receiving MTX require close monitoring for possible toxic effects.
    Pemetrexed is primarily used in non–small cell lung cancer.
    Pralatrexate is used in relapsed or refractory T-cell lymphoma.
  2. Resistance: Nonproliferating cells are resistant to MTX, probably
    because of a relative lack of DHFR, thymidylate synthase, and/
    or the glutamylating enzyme. Decreased levels of the MTX
    polyglutamate have been reported in resistant cells and may be due
    to its decreased formation or increased breakdown. Resistance in
    neoplastic cells can be due to amplification (production of additional
    copies) of the gene that codes for DHFR, resulting in increased
    levels of this enzyme. The enzyme affinity for MTX may also be
    diminished. Resistance can also occur from a reduced influx of MTX, apparently caused by a change in the carrier-mediated
    transport responsible for pumping the drug into the cell.
  3. Pharmacokinetics: MTX is variably absorbed at low doses from
    the GI tract, but it can also be administered by intramuscular, intra-
    venous (IV), and intrathecal routes (Figure 46.10). Because MTX
    does not easily penetrate the blood–brain barrier, it can be adminis-
    tered intrathecally to destroy neoplastic cells that are thriving in the
    sanctuary of the CNS. High concentrations of the drug are found
    in the intestinal epithelium, liver, and kidney, as well as in asci-
    tes and pleural effusions. MTX is also distributed to the skin. High
    doses of MTX undergo hydroxylation at the 7 position and become
    7-hydroxymethotrexate. This derivative is much less active as an
    antimetabolite. It is less water soluble than MTX and may lead to
    crystalluria. Therefore, it is important to keep the urine alkaline and
    the patient well hydrated to avoid renal toxicity. Excretion of the
    parent drug and the 7-OH metabolite occurs primarily via urine,
    although some of the drug and its metabolite appear in feces due
    to enterohepatic excretion.
  4. Adverse effects: Adverse effects of MTX are outlined in Figure
    46.8. Pemetrexed should be given with folic acid and vitamin B12
    supplements to reduce hematologic and GI toxicities. It is also rec-
    ommended to pretreat with corticosteroids to prevent cutaneous
    reactions. One of the more common side effects of pralatrexate is
    mucositis. Doses must be adjusted or withheld based on the sever-
    ity of mucositis. Pralatrexate also requires supplementation with
    folic acid and vitamin B12.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

6-Mercaptopurine

A

6-Mercaptopurine [mer-kap-toe-PYOOR-een] (6-MP) is the thiol ana-
log of hypoxanthine. 6-MP and 6-thioguanine were the first purine
analogs to prove beneficial for treating neoplastic disease. [Note:
Azathioprine, an immunosuppressant, exerts its cytotoxic effects after
conversion to 6-MP.] 6-MP is used principally in the maintenance of
remission in acute lymphoblastic leukemia. 6-MP and its analog, aza-
thioprine, are also beneficial in the treatment of Crohn disease.
1. Mechanism of action:
a. Nucleotide formation: To exert its antileukemic effect, 6-MP
must penetrate target cells and be converted to the nucleotide
analog, 6-MP-ribose phosphate (better known as 6-thioinosinic
acid or TIMP; Figure 46.11). The addition of the ribose phosphate
is catalyzed by the salvage pathway enzyme, hypoxanthine–
guanine phosphoribosyltransferase (HGPRT).
b. Inhibition of purine synthesis: A number of metabolic pro-
cesses involving purine biosynthesis and interconversions are
affected by the nucleotide analog, TIMP. Similar to nucleotide
monophosphates, TIMP can inhibit the first step of de novo
purine ring biosynthesis (catalyzed by glutamine phosphoribo-
syl pyrophosphate amidotransferase). TIMP also blocks the for-
mation of adenosine monophosphate and xanthinuric acid from
inosinic acid.

c. Incorporation into nucleic acids: TIMP is converted to thio-
guanine monophosphate, which after phosphorylation to di- and
triphosphates can be incorporated into RNA. The deoxyribo-
nucleotide analogs that are also formed are incorporated into
DNA. This results in nonfunctional RNA and DNA.
2. Resistance: Resistance is associated with 1) an inability to
biotransform 6-MP to the corresponding nucleotide because of
decreased levels of HGPRT, 2) increased dephosphorylation,
or 3) increased metabolism of the drug to thiouric acid or other
metabolites.
3. Pharmacokinetics: Oral absorption is erratic and incomplete.
Once it enters the blood circulation, the drug is widely distributed
throughout the body, except for the cerebrospinal fluid (CSF). The
bioavailability of 6-MP can be reduced by first-pass metabolism in
the liver. 6-MP is converted in the liver to the 6-methylmercapto-
purine derivative or to thiouric acid (an inactive metabolite). [Note:
The latter reaction is catalyzed by xanthine oxidase.] The parent
drug and its metabolites are excreted by the kidney.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Fludarabine

A

Fludarabine [floo-DARE-a-been] is the 5′-phosphate of 2-fluoro-
adenine arabinoside, a purine nucleotide analog. It is useful in the
treatment of chronic lymphocytic leukemia, hairy cell leukemia, and
indolent non-Hodgkin lymphoma. Fludarabine is a prodrug, the phos-
phate being removed in the plasma to form 2-F-araA, which is taken up
into cells and again phosphorylated (initially by deoxycytidine kinase).
Although the exact cytotoxic mechanism is uncertain, the triphosphate
is incorporated into both DNA and RNA. This decreases their synthe-
sis in the S-phase and affects their function. Resistance is associ-
ated with reduced uptake into cells, lack of deoxycytidine kinase, and
decreased affinity for DNA polymerase, as well as other mechanisms.
Fludarabine is administered IV rather than orally, because intestinal
bacteria split off the sugar to yield the very toxic metabolite, fluoroad-
enine. Urinary excretion accounts for partial elimination.

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

Cladribine

A

Another purine analog, 2-chlorodeoxyadenosine or cladribine [KLA-
dri-been], undergoes reactions similar to those of fludarabine, and it
must be phosphorylated to a nucleotide to be cytotoxic. It becomes
incorporated at the 3′-terminus of DNA and, thus, hinders elonga-
tion. It also affects DNA repair and is a potent inhibitor of ribonucleo-
tide reductase. Resistance may be due to mechanisms analogous
to those that affect fludarabine, although cross-resistance is not
observed. Cladribine is effective against hairy cell leukemia, chronic
lymphocytic leukemia, and non-Hodgkin lymphoma. The drug is given
as a single, continuous infusion. Cladribine distributes throughout the
body, including into the CSF.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

5-Fluorouracil

A

5-Fluorouracil
5-Fluorouracil [flure-oh-YOOR-ah-sil] (5-FU ), a pyrimidine analog, has
a stable fluorine atom in place of a hydrogen atom at position 5 of the uracil ring. The fluorine interferes with the conversion of deoxyuridylic
acid to thymidylic acid, thus depriving the cell of thymidine, one of the
essential precursors for DNA synthesis. 5-FU is employed primarily in
the treatment of slowly growing solid tumors (for example, colorectal,
breast, ovarian, pancreatic, and gastric carcinomas). When applied
topically, 5-FU is also effective for the treatment of superficial basal
cell carcinomas.
1. Mechanism of action: 5-FU itself is devoid of antineoplastic
activity. It enters the cell through a carrier-mediated transport
system and is converted to the corresponding deoxynucleotide
(5-fluorodeoxyuridine monophosphate [5-FdUMP]; Figure 46.12),
which competes with deoxyuridine monophosphate for thymidylate
synthase, thus inhibiting its action. DNA synthesis decreases
due to lack of thymidine, leading to imbalanced cell growth and
“thymidine-less death” of rapidly dividing cells. [Note: Leucovorin
is administered with 5-FU, because the reduced folate coenzyme
is required in the thymidylate synthase inhibition. For example,
a standard regimen for advanced colorectal cancer is irinotecan
plus 5-FU/leucovorin.] 5-FU is also incorporated into RNA, and
low levels have been detected in DNA. In the latter case, a glyco-
sylase excises the 5-FU, damaging the DNA. 5-FU produces the
anticancer effect in the S-phase of the cell cycle.
2. Resistance: Resistance is encountered when the cells have
lost their ability to convert 5-FU into its active form (5-FdUMP)
or when they have altered or increased thymidylate synthase
levels.
3. Pharmacokinetics: Because of its severe toxicity to the GI tract,
5-FU is given IV or, in the case of skin cancer, topically. The drug
penetrates well into all tissues, including the CNS. 5-FU is rapidly
metabolized in the liver, lung, and kidney. It is eventually con-
verted to fluoro-β-alanine, which is removed in the urine. The dose
of 5-FU must be adjusted in impaired hepatic function. Elevated
levels of dihydropyrimidine dehydrogenase (DPD) can increase
the rate of 5-FU catabolism and decrease its bioavailability. The
DPD level varies from individual to individual and may differ by
as much as sixfold in the general population. Patients with DPD
deficiency may experience severe toxicity manifested by pancy-
topenia, mucositis, and life-threatening diarrhea. Knowledge of
an individual’s DPD activity should allow more appropriate dosing
of 5-FU.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Capecitabine

A

Capecitabine [cape-SITE-a-been] is a novel, oral fluoropyrimidine car-
bamate. It is used in the treatment of colorectal and metastatic breast
cancer. After being absorbed, capecitabine, which is itself nontoxic,
undergoes a series of enzymatic reactions, the last of which is hydro-
lysis to 5-FU. This step is catalyzed by thymidine phosphorylase, an
enzyme that is concentrated primarily in tumors (Figure 46.13). Thus,
the cytotoxic activity of capecitabine is the same as that of 5-FU and
is tumor specific. The most important enzyme inhibited by 5-FU (and,
thus, capecitabine) is thymidylate synthase. Capecitabine is well
absorbed following oral administration. It is extensively metabolized to 5-FU and is eventually biotransformed into fluoro-β-alanine.
Metabolites are primarily eliminated in the urine.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Cytarabine

A

Cytarabine
Cytarabine [sye-TARE-ah-been] (cytosine arabinoside or ara-C) is
an analog of 2′-deoxycytidine in which the natural ribose residue is
replaced by d-arabinose. Cytarabine acts as a pyrimidine antago-
nist. The major clinical use of cytarabine is in acute nonlymphocytic
(myelogenous) leukemia (AML). Cytarabine enters the cell by a
carrier-mediated process and, like the other purine and pyrimidine
antagonists, must be sequentially phosphorylated by deoxycytidine
kinase and other nucleotide kinases to the nucleotide form (cyto-
sine arabinoside triphosphate or ara-CTP) to be cytotoxic. Ara-CTP
is an effective inhibitor of DNA polymerase. The nucleotide is also
incorporated into nuclear DNA and can terminate chain elongation.
It is, therefore, S-phase (and, hence, cell cycle) specific.
1. Resistance: Resistance to cytarabine may result from a defect
in the transport process, a change in activity of phosphorylating
enzymes (especially deoxycytidine kinase), or an increased pool
of the natural dCTP nucleotide. Increased deamination of the drug
to uracil arabinoside (ara-U) can also cause resistance.
2. Pharmacokinetics: Cytarabine is not effective when given orally,
because of its deamination to the noncytotoxic ara-U by cytidine
deaminase in the intestinal mucosa and liver. Given IV, it distrib-
utes throughout the body but does not penetrate the CNS in suf-
ficient amounts. Therefore, it may also be injected intrathecally.
A liposomal preparation that provides slow release into the CSF is
also available. Cytarabine undergoes extensive oxidative deamina-
tion in the body to ara-U, a pharmacologically inactive metabolite.
Both cytarabine and ara-U are excreted in urine.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Azacitidine

A

Azacitidine [A-zuh-SITE-i-dine] is a pyrimidine nucleoside analog of
cytidine. It is used for the treatment of myelodysplastic syndromes
and AML. Azacitidine undergoes activation to the nucleotide metabo-
lite azacitidine triphosphate and gets incorporated into RNA to inhibit
RNA processing and function. It is S-phase cell cycle specific. The
mechanism of resistance is not well described.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Gemcitabine

A

Gemcitabine [jem-SITE-ah-been] is an analog of the nucleoside deoxy-
cytidine. It is used most commonly for pancreatic cancer and non–
small cell lung cancer. Gemcitabine is a substrate for deoxycytidine
kinase, which phosphorylates the drug to 2′,2′-difluorodeoxycytidine
triphosphate (Figure 46.14). Resistance to the drug is probably due
to its inability to be converted to a nucleotide, caused by an altera-
tion in deoxycytidine kinase. In addition, the tumor cell can produce
increased levels of endogenous deoxycytidine that compete for the
kinase, thus overcoming the inhibition. Gemcitabine is infused IV.
It is deaminated to difluorodeoxyuridine, which is not cytotoxic, and
is excreted in urine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Anthracyclines: Doxorubicin, daunorubicin, idarubicin,
epirubicin, and mitoxantrone

A

Doxorubicin [dox-oh-ROO-bi-sin] and daunorubicin [daw-noe-ROO-
bi-sin] are classified as anthracycline antibiotics. Doxorubicin is the
hydroxylated analog of daunorubicin. Idarubicin [eye-da-ROO-bi-sin],
the 4-demethoxy analog of daunorubicin, epirubicin [eh-pee-ROO-
bih-sin], and mitoxantrone [mye-toe-ZAN-trone] are also available.
Applications for these agents differ despite their structural similarity
and their apparently similar mechanisms of action. Doxorubicin is one
of the most important and widely used anticancer drugs. It is used in
combination with other agents for treatment of sarcomas and a vari-
ety of carcinomas, including breast and lung, as well as for treatment
of acute lymphocytic leukemia and lymphomas. Daunorubicin and
idarubicin are used in the treatment of acute leukemias, and mitoxan-
trone is used in prostate cancer.
1. Mechanism of action: Doxorubicin and other anthracyclines
induce cytotoxicity through several different mechanisms. For
example, doxorubicin-derived free radicals can induce membrane lipid peroxidation, DNA strand scission, and direct oxidation of
purine or pyrimidine bases, thiols, and amines (Figure 46.16).
2. Pharmacokinetics: All these drugs must be administered IV,
because they are inactivated in the GI tract. Extravasation is a seri-
ous problem that can lead to tissue necrosis. The anthracycline
antibiotics bind to plasma proteins as well as to other tissue com-
ponents, where they are widely distributed. They do not penetrate
the blood–brain barrier or the testes. These agents undergo exten-
sive hepatic metabolism, and dosage adjustments are needed
in patients with impaired hepatic function. Biliary excretion is the
major route of elimination. Some renal excretion also occurs, but
dosage adjustments are generally not needed in renal dysfunc-
tion. Because of the dark red color of the anthracycline drugs, the
veins may become visible surrounding the site of infusion, and red
discoloration of urine may occur.
3. Adverse effects: Irreversible, dose-dependent cardiotoxicity, appar -
ently a result of the generation of free radicals and lipid peroxida-
tion, is the most serious adverse reaction and is more common with
daunorubicin and doxorubicin than with idarubicin and epirubicin.
Addition of trastuzumab to protocols with doxorubicin or epirubicin
increases congestive heart failure. There has been some success
with the iron chelator dexrazoxane in protecting against the cardio-
toxicity of doxorubicin. The liposomal-encapsulated doxorubicin is
reported to be less cardiotoxic than the usual formulation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Bleomycin

A

Bleomycin [blee-oh-MYE-sin] is a mixture of different copper-chelating
glycopeptides that, like the anthracycline antibiotics, cause scission
of DNA by an oxidative process. Bleomycin is cell cycle specific and
causes cells to accumulate in the G2
phase. It is primarily used in the
treatment of testicular cancers and Hodgkin lymphoma.
1. Mechanism of action: A DNA–bleomycin–Fe2+ complex appears
to undergo oxidation to bleomycin–Fe3+. The liberated electrons
react with oxygen to form superoxide or hydroxyl radicals, which, in
turn, attack the phosphodiester bonds of DNA, resulting in strand
breakage and chromosomal aberrations (Figure 46.17).
2. Resistance: Although the mechanisms of resistance have not
been elucidated, increased levels of bleomycin hydrolase (or
deaminase), glutathione S-transferase, and possibly, increased
efflux of the drug have been implicated. DNA repair also may
contribute.
3. Pharmacokinetics: Bleomycin is administered by a number of
routes. The bleomycin-inactivating enzyme (a hydrolase) is high in
a number of tissues (for example, liver and spleen) but is low in the
lung and is absent in skin (accounting for the drug’s toxicity in those
tissues). Most of the parent drug is excreted unchanged in the
urine, necessitating dose adjustment in patients with renal failure.
4. Adverse effects: Mucocutaneous reactions and alopecia are
common. Hypertrophic skin changes and hyperpigmentation of the hands are prevalent. There is a high incidence of fever and
chills and a low incidence of serious anaphylactoid reactions.
Pulmonary toxicity is the most serious adverse effect, progressing
from rales, cough, and infiltrate to potentially fatal fibrosis. The
pulmonary fibrosis that is caused by bleomycin is often referred as
“bleomycin lung.” Bleomycin is unusual in that myelosuppression
is rare.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

ALKYLATING AGENTS

Cyclophosphamide and ifosfamide

A

Cyclophosphamide and ifosfamide
These drugs are very closely related mustard agents that share most
of the same primary mechanisms and toxicities. They are cytotoxic
only after generation of their alkylating species, which are produced
through hydroxylation by cytochrome P450 (CYP450). These agents
have a broad clinical spectrum, being used either singly or as part of
a regimen in the treatment of a wide variety of neoplastic diseases,
such as non-Hodgkin lymphoma, sarcoma, and breast cancer.
1. Mechanism of action: Cyclophosphamide [sye-kloe-FOSS-fah-
mide] is the most commonly used alkylating agent. Both cyclophos-
phamide and ifosfamide [eye-FOSS-fah-mide] are first biotrans-
formed to hydroxylated intermediates primarily in the liver by the
CYP450 system (Figure 46.19).The hydroxylated intermediates
then undergo breakdown to form the active compounds, phos-
phoramide mustard and acrolein. Reaction of the phosphoramide
mustard with DNA is considered to be the cytotoxic step. The par-
ent drug and its metabolites are primarily excreted in urine.
2. Pharmacokinetics: Cyclophosphamide is available in oral or IV
preparations, whereas ifosfamide is IV only. Cyclophosphamide
is metabolized in the liver to active and inactive metabolites, and
minimal amounts are excreted in the urine as unchanged drug.
Ifosfamide is metabolized primarily by CYP450 3A4 and 2B6 iso-
enzymes. It is mainly renally excreted.
3. Resistance: Resistance results from increased DNA repair,
decreased drug permeability, and reaction of the drug with thiols
(for example, glutathione). Cross-resistance does not always occur.
4. Adverse effects: A unique toxicity of both drugs is hemorrhagic
cystitis, which can lead to fibrosis of the bladder. Bladder
toxicity has been attributed to acrolein in the urine in the case of
cyclophosphamide and to toxic metabolites of ifosfamide. Adequate
hydration as well as IV injection of mesna (sodium 2-mercaptoethane
sulfonate), which neutralizes the toxic metabolites, can minimize
this problem. A fairly high incidence of neurotoxicity has been
reported in patients on high-dose ifosfamide, probably due to the
metabolite, chloroacetaldehyde.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Nitrosoureas

Carmustine

A

Carmustine [KAR-mus-teen, BCNU] and lomustine [LOE-mus-teen,
CCNU] are closely related nitrosoureas. Because of their ability to
penetrate the CNS, the nitrosoureas are primarily employed in the
treatment of brain tumors.
1. Mechanism of action: The nitrosoureas exert cytotoxic effects
by an alkylation that inhibits replication and, eventually, RNA
and protein synthesis. Although they alkylate DNA in resting
cells, cytotoxicity is expressed primarily on cells that are actively
dividing. Therefore, nondividing cells can escape death if DNA
repair occurs. Nitrosoureas also inhibit several key enzymatic
processes by carbamoylation of amino acids in proteins in the
targeted cells.

  1. Pharmacokinetics: In spite of the similarities in their struc-
    tures, carmustine is administered IV and as chemotherapy wafer
    implants, whereas lomustine is given orally. Because of their lipo-
    philicity, they distribute widely in the body, but their most striking
    property is their ability to readily penetrate the CNS. The drugs
    undergo extensive metabolism. Lomustine is metabolized to active
    products. The kidney is the major excretory route for the nitro-
    soureas (Figure 46.20).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Dacarbazine

A

Dacarbazine [dah-KAR-bah-zeen] is an alkylating agent that must
undergo biotransformation to an active metabolite, methyltriazeno-
imidazole carboxamide (MTIC). This metabolite is responsible for the
drug’s activity as an alkylating agent by forming methylcarbonium
ions that can attack the nucleophilic groups in the DNA molecule.
Thus, similar to other alkylating agents, the cytotoxic action of dacar-
bazine has been attributed to the ability of its metabolite to methylate
DNA on the O6
position of guanine. Dacarbazine has found use in the
treatment of melanoma and Hodgkin lymphoma.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Temozolomide

A

The treatment of tumors in the brain is particularly difficult.
Temozolomide [te-moe-ZOE-loe-mide], a triazene agent, has been
approved for use against glioblastomas and anaplastic astrocytomas.
It is also used in metastatic melanoma. Temozolomide is related
to dacarbazine, because both must undergo biotransformation to
an active metabolite, MTIC, which probably is responsible for the
methylation of DNA on the 6 position of guanine. Unlike dacarbazine,
temozolomide does not require the CYP450 system for metabolic
transformation, and it undergoes chemical transformation at normal
physiological pH. Temozolomide also has the property of inhibiting
the repair enzyme, O6
-guanine-DNA alkyltransferase. Temozolomide
differs from dacarbazine in that it crosses the blood–brain barrier.
Temozolomide is administered intravenously or orally and has
excellent bioavailability after oral administration. The parent drug and
metabolites are excreted in urine (Figure 46.21).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Other alkylating agents

Mechlorethamine
Mephalan
Chlorambucil
Busulfan

A

Mechlorethamine [mek-lor-ETH-ah-meen] was developed as a
vesicant (nitrogen mustard) during World War I. Its ability to cause
lymphocytopenia led to its use in lymphatic cancers. Melphalan [MEL-
fah-lan], a phenylalanine derivative of nitrogen mustard, is used in the
treatment of multiple myeloma. This is a bifunctional alkylating agent
that can be given orally. Although melphalan can be given orally, the
plasma concentration differs from patient to patient due to variation in
intestinal absorption and metabolism. The dose of melphalan is care-
fully adjusted by monitoring the platelet and white blood cell counts.
Chlorambucil [clor-AM-byoo-sil] is another bifunctional alkylating
agent that is used in the treatment of chronic lymphocytic leukemia.
Both melphalan and chlorambucil have moderate hematologic toxici-
ties and upset the GI tract. Busulfan [byoo-SUL-fan] is another oral
agent that is effective against chronic granulocytic leukemia. In aged patients, busulfan can cause pulmonary fibrosis (“busulfan lung”).
Like other alkylating agents, all of these agents are leukemogenic.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Cyclophosphamide

A

Route: IV/PO
AE: Myelosuppression, hemorrhagic
cystitis, N/V/D, alopecia,
amenorrhea, secondary
malignancies
DI: Phenobarbital, phenytoin (P450);
digoxin, anticoagulants
Monitoring: Urinalysis; CBC; renal,
hepatic function
Good hydration to prevent bladder
toxicity (mesna with high doses)

26
Q

Ifosfamide

A

Route: IV
AE:Myelosuppression, hemorrhagic
cystitis, N/V, neurotoxicity, alopecia,
amenorrhea
DI:Phenobarbital, phenytoin (P450);
cimetidine, allopurinol, warfarin
Monitoring: Urinalysis; neurotoxicity
Notes: Use mesna and hydration to
prevent bladder toxicity

27
Q

Carmustine (BCNU)

A

Route: IV
AE:Myelosuppression, N/V, facial
ushing, hepatotoxicity, pulmonary
toxicity, impotence, infertility
DI: Cimetidine, amphotericin B,
digoxin, phenytoin
Monitoring:CBC; PFTs; renal,
hepatic function
Notes: Also available as an implantable
wafer (brain)

28
Q

Lomustine (CCNU)

A

Route: PO
AE:Myelosuppression, N/V, pulmonary
toxicity, impotence, infertility,
neurotoxicity
DI:Cimetidine, alcohol
Monitoring:CBC; PFTs; renal
function
Notes: Administer on an empty stomach

29
Q

Dacarbazine

A

Route:IV
AE:Myelosuppression, N/V, u-like
syndrome, CNS toxicity,
hepatotoxicity, photosensitivity
DI:Phenytoin, phenobarbital (P450)
Monitoring: CBC; renal, hepatic
function
Notes: Vesicant

30
Q

Temozolomide

A

Route: PO
AE: N/V, myelosuppression, headache,
fatigue, photosensitivity
Monitoring:CBC; renal, hepatic
function
Notes: Requires PCP prophylaxis

31
Q

Melphalan

A

Route: IV/PO
AE: Myelosuppression, N/V/D,
mucositis, hypersensitivity (IV)
DI: Cimetidine, steroids,
cyclosporine
Monitoring:CBC; renal, hepatic
function; Adjust in
renal dysfunction
Notes: Take on an empty stomach

32
Q

Chlorambucil

A

Route: PO
AE: Myelosuppression, skin rash,
pulmonary brosis (rare),
hyperuricemia, seizures
DI: Phenobarbital, phenytoin (P450)
Note: Take with food

33
Q

Busulfan

A

Route: IV
AE:Myelosuppression, N/V/D,
mucositis, skin rash, pulmonary
brosis, hepatotoxicity
DI:Acetaminophen, itraconazole,
phenytoin
Monitoring:CBC; pulmonary
symptoms; renal,
hepatic function
Notes: Busulfan lung

34
Q

MICROTUBULE INHIBITORS

Vincristine and vinblastine

A

Vincristine [vin-KRIS-teen] (VX) and vinblastine [vin-BLAS-teen]
(VBL) are structurally related compounds derived from the periwinkle
plant, Vinca rosea. They are, therefore, referred to as the Vinca alka-
loids. A less neurotoxic agent is vinorelbine [vye-NOR-el-been] (VRB).
Although the Vinca alkaloids are structurally similar to one another,
their therapeutic indications are different. They are generally adminis-
tered in combination with other drugs. VX is used in the treatment of
acute lymphoblastic leukemia in children, Wilms tumor, Ewing soft tis-
sue sarcoma, and Hodgkin and non-Hodgkin lymphomas, as well as
some other rapidly proliferating neoplasms. [Note: VX (former trade
name, Oncovin) is the “O” in the R-CHOP regimen for lymphoma. Due
to relatively mild myelosuppressive activity, VX is used in a number of
other protocols.] VBL is administered with bleomycin and cisplatin for
the treatment of metastatic testicular carcinoma. It is also used in the
treatment of systemic Hodgkin and non-Hodgkin lymphomas. VRB is
beneficial in the treatment of advanced non–small cell lung cancer,
either as a single agent or with cisplatin.

  1. Mechanism of action: VX, VRB, and VBL are all cell cycle spe-
    cific and phase specific, because they block mitosis in metaphase
    (M-phase). Their binding to the microtubular protein, tubulin, blocks
    the ability of tubulin to polymerize to form microtubules. Instead,
    paracrystalline aggregates consisting of tubulin dimers and the
    alkaloid drug are formed. The resulting dysfunctional spindle appa-
    ratus, frozen in metaphase, prevents chromosomal segregation
    and cell proliferation (Figure 46.23).
  2. Pharmacokinetics: IV injection of these agents leads to rapid
    cytotoxic effects and cell destruction. This, in turn, can cause
    hyperuricemia due to the oxidation of purines that are released
    from fragmenting DNA molecules. The Vinca alkaloids are concen-
    trated and metabolized in the liver by the CYP450 pathway and
    eliminated in bile and feces. Doses must be modified in patients
    with impaired hepatic function or biliary obstruction.
  3. Adverse effects: VX and VBL have certain toxicities in common.
    These include phlebitis or cellulitis, if the drugs extravasate dur-
    ing injection, as well as nausea, vomiting, diarrhea, and alopecia.
    However, the adverse effects of VX and VBL are not identical. VBL
    is a more potent myelosuppressant than VX, whereas peripheral
    neuropathy (paresthesias, loss of reflexes, foot drop, and ataxia) is
    associated with VX. Constipation is more frequently encountered
    with VX. These agents should not be administered intrathecally.
    This potential drug error can result in death, and special precau-
    tions should be in place for administration.
35
Q

Paclitaxel and docetaxel

A

Paclitaxel [PAK-li-tax-el] was the first member of the taxane family to
be used in cancer chemotherapy. A semisynthetic paclitaxel is now
available through chemical modification of a precursor found in the
needles of Pacific yew species. An albumin-bound form is also avail-
able. Substitution of a side chain has resulted in docetaxel [doe-see-
TAX-el], which is the more potent of the two drugs. Paclitaxel has
shown good activity against advanced ovarian cancer and metastatic
breast cancer. Favorable results have been obtained in non–small cell
lung cancer when administered with cisplatin. Docetaxel is commonly
used in prostate, breast, GI, and non–small cell lung cancers.
1. Mechanism of action: Both drugs are active in the G2
/M-phase of
the cell cycle, but unlike the Vinca alkaloids, they promote polym-
erization and stabilization of the polymer rather than disassembly,
leading to the accumulation of microtubules (Figure 46.24). The
overly stable microtubules formed are nonfunctional, and chromo-
some desegregation does not occur. This results in death of the
cell.
2. Pharmacokinetics: These agents undergo hepatic metabolism
by the CYP450 system and are excreted via the biliary system.
Dose modification is not required in patients with renal impairment,
but doses should be reduced in patients with hepatic dysfunction.
3. Adverse effects: The dose-limiting toxicities of paclitaxel and
docetaxel are neutropenia and leukopenia. Alopecia occurs, but vomiting and diarrhea are uncommon. [Note: Because of serious
hypersensitivity reactions (including dyspnea, urticaria, and hypo-
tension), patients who are treated with paclitaxel should be pre-
medicated with dexamethasone and diphenhydramine, as well as
with an H2
blocker.]

36
Q

Prednisone
Prednisone [PRED-ni-sone] is a potent, synthetic, anti-inflammatory
corticosteroid with less mineralocorticoid activity than cortisol (see
Chapter 27). [Note: At high doses, cortisol is lymphocytolytic and
leads to hyperuricemia due to the breakdown of lymphocytes.]
Prednisone is primarily employed to induce remission in patients with
acute lymphocytic leukemia and in the treatment of both Hodgkin and
non-Hodgkin lymphomas. Prednisone is readily absorbed orally. Like
other glucocorticoids, it is bound to plasma albumin and transcortin.
Prednisone itself is inactive and must first undergo 11-β-hydroxylation
to prednisolone in the liver. Prednisolone is the active drug. This ste-
roid then binds to a receptor that triggers the production of specific
proteins (Figure 46.26A). The latter is glucuronidated and excreted in
urine along with the parent compound.

A

Prednisone [PRED-ni-sone] is a potent, synthetic, anti-inflammatory
corticosteroid with less mineralocorticoid activity than cortisol (see
Chapter 27). [Note: At high doses, cortisol is lymphocytolytic and
leads to hyperuricemia due to the breakdown of lymphocytes.]
Prednisone is primarily employed to induce remission in patients with
acute lymphocytic leukemia and in the treatment of both Hodgkin and
non-Hodgkin lymphomas. Prednisone is readily absorbed orally. Like
other glucocorticoids, it is bound to plasma albumin and transcortin.
Prednisone itself is inactive and must first undergo 11-β-hydroxylation
to prednisolone in the liver. Prednisolone is the active drug. This ste-
roid then binds to a receptor that triggers the production of specific
proteins (Figure 46.26A). The latter is glucuronidated and excreted in
urine along with the parent compound.

37
Q

Tamoxifen

A

Tamoxifen [tah-MOX-ih-fen] is an estrogen antagonist with some
estrogenic activity, and it is classified as a selective estrogen receptor
modulator (SERM). It is used for first-line therapy in the treatment of
estrogen receptor–positive breast cancer. It also finds use prophy-
lactically in reducing breast cancer occurrence in women who are at
high risk. However, because of possible stimulation of premalignant
lesions due to its estrogenic properties, patients should be closely
monitored during therapy.
1. Mechanism of action: Tamoxifen binds to estrogen receptors in
the breast tissue, but the complex is unable to translocate into the
nucleus for its action of initiating transcriptions. That is, the com-
plex fails to induce estrogen-responsive genes, and RNA syn-
thesis does not ensue (Figure 46.26B). The result is a depletion
(down-regulation) of estrogen receptors, and the growth-promoting effects of the natural hormone and other growth factors are sup-
pressed. [Note: Estrogen competes with tamoxifen. Therefore, in
premenopausal women, the drug is used with a gonadotropin-
releasing hormone (GnRH) analog such as leuprolide, which
lowers estrogen levels.]
2. Pharmacokinetics: Tamoxifen is effective after oral adminis-
tration. It is partially metabolized by the liver. Some metabolites
possess antagonist activity, whereas others have agonist activ-
ity. Unchanged drug and metabolites are excreted predominantly
through the bile into the feces. Tamoxifen is an inhibitor of CYP3A4
and P-glycoprotein.
3. Adverse effects: Side effects caused by tamoxifen include hot
flashes, nausea, vomiting, skin rash, and vaginal bleeding and
discharge (due to estrogenic activity of the drug and some of its
metabolites). Hypercalcemia may occur, requiring cessation of
the drug. Tamoxifen can also lead to increased pain if the tumor
has metastasized to bone. Tamoxifen has the potential to cause
endometrial cancer. Other toxicities include thromboembolism and
effects on vision. [Note: Because of a more favorable adverse effect
profile, aromatase inhibitors are making an impact in the treatment
of breast cancer.]

38
Q

Fulvestrant and raloxifene

A

Fulvestrant [fool-VES-trant] and raloxifene [ral-OKS-i-feen] are two
agents that interact with the estrogen receptor to prevent some of the
downstream effects. Fulvestrant is an estrogen receptor antagonist that
is given via intramuscular injection to patients with hormone receptor–
positive metastatic breast cancer. This agent binds to and causes estro-
gen receptor down-regulation on tumors and other targets. Raloxifene
is a SERM given orally that acts to block estrogen effects in the uterine and breast tissues, while promoting effects in the bone to inhibit resorp-
tion. This agent has been shown to reduce the risk of estrogen receptor–
positive invasive breast cancer in postmenopausal women. These
agents are known to cause hot flashes, arthralgias, and myalgias.

39
Q

Aromatase inhibitors

Anastrozole and letrozole

A

The imidazole aromatase inhibitors,
such as anastrozole [an-AS-troe-zole] and letrozole [LE-troe-zole],
are nonsteroidal aromatase inhibitors. They do not predispose
patients to endometrial cancer and are devoid of the androgenic
side effects that occur with the steroidal aromatase inhibitors
such as aminoglutethimide. Although anastrozole and letrozole
are considered second-line therapy after tamoxifen for hormone-
dependent breast cancer in the United States, they have become
first-line drugs in other countries for the treatment of breast can-
cer in postmenopausal women. They are orally active and cause
almost a total suppression of estrogen synthesis. Both drugs are
extensively metabolized in the liver, and

40
Q

Exemestane

A

A steroidal, irreversible inhibitor of aromatase, exemes-
tane [ex-uh-MES-tane], is orally well absorbed and widely distributed.
Hepatic metabolism is by the CYP3A4 isoenzyme. Because the
metabolites are excreted in urine, doses of the drug must be adjusted
in patients with renal failure. Its major toxicities are nausea, fatigue,
and hot flashes. Alopecia and dermatitis have also been noted.

41
Q

Progestins

Megestrol acetate

A

Megestrol [me-JESS-trole] acetate is a progestin that was widely used
in treating metastatic hormone-responsive breast and endometrial neo-
plasms. It is orally effective. Other agents are usually compared to it in
clinical trials; however, the aromatase inhibitors are replacing it in therapy

42
Q

Leuprolide, goserelin, and triptorelin

A

GnRH is normally secreted by the hypothalamus and stimulates the
anterior pituitary to secrete the gonadotropic hormones: 1) luteinizing
hormone (LH), the primary stimulus for the secretion of testosterone
by the testes, and 2) follicle-stimulating hormone (FSH), which stimu-
lates the secretion of estrogen. Leuprolide [loo-PROE-lide], goserelin
[GOE-se-rel-in], and triptorelin [TRIP-to-rel-in] are synthetic analogs of
GnRH. As GnRH analogs, they occupy the GnRH receptor in the pitu-
itary, which leads to its desensitization and, consequently, inhibition of
release of FSH and LH. Thus, both androgen and estrogen syntheses
are reduced (Figure 46.27). Response to leuprolide in prostatic can-
cer is equivalent to that of orchiectomy with regression of tumor and
relief of bone pain. These drugs have some benefit in premenopausal women with advanced breast cancer and have largely replaced estro-
gens in therapy for prostate cancer. Leuprolide is available as 1) a
sustained-release intradermal implant, 2) a subcutaneous depot injec-
tion, or 3) an intramuscular depot injection to treat metastatic carci-
noma of the prostate. Goserelin acetate is a subcutaneous implant,
and triptorelin pamoate is injected intramuscularly. Levels of androgen
may initially rise but then fall to castration levels. The adverse effects
of these drugs, including impotence, hot flashes, and tumor flare, are
minimal compared to those experienced with estrogen treatment.

43
Q

Estrogens

A

Estrogens, such as ethinyl estradiol, had been used in the treatment
of prostatic cancer. However, they have been largely replaced by the
GnRH analogs because of fewer adverse effects. Estrogens inhibit the
growth of prostatic tissue by blocking the production of LH, thereby
decreasing the synthesis of androgens in the testis. Thus, tumors that
are dependent on androgens are affected. Estrogen treatment can
cause serious complications, such as thromboemboli, myocardial
infarction, strokes, and hypercalcemia. Men who are taking estrogens
may experience gynecomastia and impotence.

44
Q

Flutamide, nilutamide, and bicalutamide

A

Flutamide [FLOO-tah-mide], nilutamide [nye-LOO-ta-mide], and
bicalutamide [bye-ka-LOO-ta-mide] are synthetic, nonsteroidal
antiandrogens used in the treatment of prostate cancer. They compete
with the natural hormone for binding to the androgen receptor and prevent
its translocation into the nucleus (Figure 46.27). These antiandrogens
are taken orally and are cleared through the kidney. [Note: Flutamide
requires dosing three times a day and the others once a day.] Side
effects include gynecomastia and GI distress. Rarely, liver failure has
occurred with flutamide. Nilutamide can cause visual problems

45
Q

MONOCLONAL ANTIBODIES

Trastuzumab

A

In patients with metastatic breast cancer, overexpression of trans-
membrane human epidermal growth factor receptor protein 2 (HER2)
is seen in 25% to 30% of patients. HER2 overexpression is also noted
in gastric and gastroesophageal cancers. Trastuzumab [tra-STEW-
zoo-mab], a humanized monoclonal antibody, specifically targets the
extracellular domain of the HER2 growth receptor that has intrinsic
tyrosine kinase activity. [Note: At least 50 tyrosine kinases mediate
cell growth or division by phosphorylating signaling proteins. They
have been implicated in the development of many neoplasms by an
unknown mechanism.]
1. Mechanism of action: Trastuzumab binds to HER2 sites in
breast cancer, gastric cancer, and gastroesophageal tissues
and inhibits the proliferation of cells that overexpress the HER2
protein, thereby decreasing the number of cells in the S-phase.
By binding to HER2, it blocks downstream signaling pathways,
induces antibody-dependent cytotoxicity, and prevents the
release of HER2.
2. Adverse effects: The most serious toxicity associated with the
use of trastuzumab is congestive heart failure. The toxicity is wors-
ened if given in combination with anthracyclines. Extreme caution
should be exercised when giving the drug to patients with preexist-
ing cardiac dysfunction.

46
Q

Rituximab

A

Rituximab [ri-TUCKS-ih-mab] was the first monoclonal antibody to be
approved for the treatment of cancer. It is a genetically engineered,
chimeric monoclonal antibody directed against the CD20 antigen that
is found on the surfaces of normal and malignant B lymphocytes.
CD20 plays a role in the activation process for cell cycle initiation
and differentiation. The CD20 antigen is expressed on nearly all
B-cell non-Hodgkin lymphomas but not in other bone marrow cells. Rituximab is effective in the treatment of lymphomas, chronic lympho-
cytic leukemia, and rheumatoid arthritis.
1. Mechanism of action: The Fab domain of rituximab binds to the
CD20 antigen on the B lymphocytes, and its Fc domain recruits
immune effector functions, inducing complement and antibody-
dependent, cell-mediated cytotoxicity of the B cells. The antibody
is commonly used with other combinations of anticancer agents,
such as cyclophosphamide, doxorubicin, vincristine (Oncovin),
and prednisone (CHOP).
2. Adverse effects: Severe adverse reactions have been fatal. It is
important to infuse rituximab slowly. Hypotension, bronchospasm,
and angioedema may occur. Chills and fever commonly accom-
pany the first infusion (especially in patients with high circulating
levels of neoplastic cells), because of rapid activation of comple-
ment which results in the release of tumor necrosis factor-α and
interleukins. Pretreatment with diphenhydramine, acetaminophen,
and corticosteroids can ameliorate these problems. Tumor lysis
syndrome has been reported within 24 hours of the first dose of
rituximab. This syndrome consists of hyperkalemia, hypocalcemia,
hyperuricemia, hyperphosphatasemia (an abnormally high content
of alkaline phosphatase in the blood), and acute renal failure that
may require dialysis

47
Q

Bevacizumab

A

The monoclonal antibody bevacizumab [be-vah-SEE-zoo-mab] is an
IV antiangiogenesis agent. Bevacizumab is approved for use as a
first-line drug against metastatic colorectal cancer and is given with
5-FU–based chemotherapy. It attaches to and stops vascular endo-
thelial growth factor from stimulating the formation of new blood ves-
sels (neovascularization). Without new blood vessels, tumors do not
receive the oxygen and essential nutrients necessary for growth and
proliferation

48
Q

Cetuximab and panitumumab

A

Cetuximab [see-TUX-i-mab] is another chimeric monoclonal antibody
infused intravenously and approved to treat KRAS wild-type metastatic
colorectal cancer and head and neck cancers. [Note: KRAS is a form
of RAS proteins, which are mediators of proliferation and differentia-
tion.] It exerts its antineoplastic effect by targeting the epidermal growth
factor receptor (EGFR) on the surface of cancer cells and interfering
with their growth. Cetuximab, panitumumab [pan-i-TUE-moo-mab], and
other agents that target this receptor cause a distinct acneiform-type
rash. The appearance of this rash has been associated with a positive
response to therapy.

49
Q

PLATINUM COORDINATION COMPLEXES

Cisplatin, carboplatin, and oxaliplatin

A

Cisplatin [SIS-pla-tin] was the first member of the platinum coordi-
nation complex class of anticancer drugs, but because of its severe toxicity, carboplatin [KAR-boe-pla-tin] was developed. The mecha-
nisms of action of the two drugs are similar, but their potency, phar-
macokinetics, patterns of distribution, and dose-limiting toxicities
differ significantly (Figure 46.29). Cisplatin has synergistic cytotoxicity
with radiation and other chemotherapeutic agents. It has found wide
application in the treatment of solid tumors, such as metastatic testic-
ular carcinoma in combination with VBL and bleomycin, ovarian car-
cinoma in combination with cyclophosphamide, or alone for bladder
carcinoma. Carboplatin is used when patients cannot be vigorously
hydrated, as is required for cisplatin treatment, or if they suffer from
kidney dysfunction or are prone to neuro- or ototoxicity. Oxaliplatin
[ox-AL-ih-pla-tin] is a closely related analog of carboplatin used in the
setting of colorectal cancer.
1. Mechanism of action: The mechanism of action for this class of
drugs is similar to that of the alkylating agents. In the high-chloride
milieu of the plasma, cisplatin persists as the neutral species, which
enters the cell and loses its chlorides in the low-chloride milieu. It
then binds to guanine in DNA, forming inter- and intrastrand cross-
links. The resulting cytotoxic lesion inhibits both polymerases for
DNA replication and RNA synthesis. Cytotoxicity can occur at any
stage of the cell cycle, but cells are most vulnerable to the actions
of these drugs in the G1
and S-phases.
2. Pharmacokinetics: These agents are administered via IV infu-
sion. Cisplatin and carboplatin can also be given intraperitoneally
for ovarian cancer and intra-arterially to perfuse other organs. The
highest concentrations of the drugs are found in the liver, kidney,
and intestinal, testicular, and ovarian cells, but little penetrates into
the CSF. The renal route is the main avenue for excretion.
3. Adverse effects: Severe, persistent vomiting occurs for at least
1 hour after administration of cisplatin and may continue for as
long as 5 days. Premedication with antiemetic agents is required.
The major limiting toxicity is dose-related nephrotoxicity, involving
the distal convoluted tubule and collecting ducts. This can be pre-
vented by aggressive hydration. Other toxicities include ototoxic-
ity with high-frequency hearing loss and tinnitus. Unlike cisplatin,
carboplatin causes only mild nausea and vomiting, and it is rarely nephro-, neuro-, or ototoxic. Its dose-limiting toxicity is myelosup-
pression. Oxaliplatin has a distinct side effect of cold-induced
peripheral neuropathy that usually resolves within 72 hours of
administration. It also causes myelosuppression and cumulative
peripheral neuropathy. Hepatotoxicity has also been reported.
These agents may cause hypersensitivity reactions ranging from
skin rashes to anaphylaxis

50
Q

TOPOISOMERASE INHIBITORS

Camptothecins

A

Camptothecins are plant alkaloids originally isolated from the Chinese
tree Camptotheca. Irinotecan [eye-rin-oh-TEE-kan] and topotecan
[toe-poe-TEE-kan] are semisynthetic derivatives of camptothecin
[camp-toe-THEE-sin]. Topotecan is used in metastatic ovarian cancer
when primary therapy has failed and also in the treatment of small
cell lung cancer. Irinotecan is used with 5-FU and leucovorin for the
treatment of colorectal carcinoma.
1. Mechanism of action: These drugs are S-phase specific and
inhibit topoisomerase I, which is essential for the replication of
DNA in human cells (Figure 46.31). SN-38 (the active metabolite
of irinotecan) is approximately 1000 times as potent as irinotecan
as an inhibitor of topoisomerase I. The topoisomerases relieve
torsional strain in DNA by causing reversible, single-strand breaks.
2. Adverse effects: Bone marrow suppression, particularly neutrope-
nia, is the dose-limiting toxicity for topotecan. Frequent blood counts
should be performed on patients taking this drug. Myelosuppression
is also seen with irinotecan. Acute and delayed diarrhea may be
severe and require treatment with atropine during the infusion or
high doses of loperamide in the days following the infusion.

51
Q

Etoposide

A

Etoposide [e-toe-POE-side] is a semisynthetic derivative of the plant
alkaloid, podophyllotoxin. It blocks cells in the late S- to G2
phase of the cell cycle. Its major target is topoisomerase II. Binding of the
drug to the enzyme–DNA complex results in persistence of the tran-
sient, cleavable form of the complex and, thus, renders it susceptible
to irreversible double-strand breaks (Figure 46.32). Etoposide finds its
major clinical use in the treatment of lung cancer and in combination
with bleomycin and cisplatin for testicular carcinoma. Etoposide may
be administered either IV or orally. Dose-limiting myelosuppression
(primarily leukopenia) is the major toxicity.

52
Q

Summary of topoisomerase inhibitors.

A

Irinotecan
Route:IV
AE:Diarrhea, myelosuppression, N, V
DI:CYP3A4 substrates
Monitoring CBC, electrolytes

Acute and delayed (life-
threatening) diarrhea

53
Q

Topotecan

A

Route: IV, PO
AE: Myelosuppression, N, V
DI:P-glycoprotein inhibitors (PO)
Monitoring CBC
Notes: Diarrhea common with PO

54
Q

Etoposide

A

Route: IV,PO
AE: Myelosuppression, hypotension,
alopecia, N, V
Monitoring CBC

Notes:May cause secondary malignancies
(leukemias)

55
Q

TYROSINE KINASE INHIBITORS

Imatinib, dasatinib, and nilotinib

A

Imatinib [i-MAT-in-ib] mesylate is used for the treatment of chronic
myelogenous leukemia (CML) as well as GI stromal tumors. It acts
as a signal transduction inhibitor, used specifically to inhibit tumor
tyrosine kinase activity. A deregulated BCR-ABL kinase is present
in the leukemia cells of almost every patient with CML. In the case
of GI stromal tumors, an unregulated expression of tyrosine kinase
is associated with a growth factor. The ability of imatinib to occupy
the “kinase pocket” prevents the phosphorylation of tyrosine on the
substrate molecule and, hence, inhibits subsequent steps that lead
to cell proliferation. Nilotinib [ni-LOT-in-ib] and dasatinib [da-SAT-in-ib]
are also first-line options for CML. These agents are all available in
oral formulations, and they are associated with notable toxicities, such
as fluid retention and QT prolongation (Figure 46.33).

56
Q

Erlotinib

A

Erlotinib [er-LOT-tih-nib] is an inhibitor of the epidermal growth factor
receptor tyrosine kinase. It is an oral agent approved for the treat-
ment of non–small cell lung cancer and pancreatic cancer. Erlotinib
is absorbed after oral administration and undergoes extensive
metabolism in the liver by the CYP3A4 isoenzyme. The most com-
mon adverse effects are diarrhea, nausea, acne-like skin rashes, and
ocular disorders. A rare but potentially fatal adverse effect is interstitial
lung disease, which presents as acute dyspnea with cough.

57
Q

Sorafenib and sunitinib

A

Sorafenib [SOR-af-i-nib] and sunitinib [su-NIT-ti-nib] are oral serine/
threonine and tyrosine kinase inhibitors used mainly in renal cell car-
cinoma. Sorafenib is also part of the treatment strategy for hepato-
cellular carcinoma, and sunitinib is used in GI stromal tumors and
pancreatic neuroendocrine tumors. These agents target cell surface
kinases that are involved in tumor signaling, angiogenesis, and apop-
tosis, thus slowing tumor growth. Adverse effects include diarrhea,
fatigue, hand and foot syndrome, and hypertension.

58
Q

MISCELLANEOUS AGENTS

Procarbazine

A

Procarbazine [proe-KAR-ba-zeen] is used in the treatment of Hodgkin
disease and other cancers. Procarbazine rapidly equilibrates between
the plasma and the CSF after oral administration. It must undergo a
series of oxidative reactions to exert its cytotoxic action that causes
inhibition of DNA, RNA, and protein synthesis. Metabolites and the
parent drug are excreted via the kidney. Bone marrow depression is
the major toxicity, and nausea, vomiting, and diarrhea are common.
The drug is also neurotoxic, causing symptoms ranging from drowsi-
ness to hallucinations to paresthesias. Because it inhibits monoamine
oxidase, patients should be warned against ingesting foods that con-
tain high levels of tyramine (for example, aged cheeses, beer, and
wine) as this could cause a hypertensive crisis. Ingestion of alcohol
leads to a disulfiram-like reaction. Procarbazine is both mutagenic
and teratogenic. Nonlymphocytic leukemia has developed in patients
treated with the drug.

59
Q

Asparaginase and pegaspargase

A

Some neoplastic cells require an external source of asparagine
because of limited capacity to synthesize sufficient amounts of the
amino acid to support growth and function. l-Asparaginase [ah-
SPAR-a-gi-nase] and the pegylated formulation pegaspargase [peg-
ah-SPAR-jase] catalyze the deamination of asparagine to aspartic
acid and ammonia, thus depriving the tumor cells of this amino acid,
which is needed for protein synthesis. The form of the enzyme used
chemotherapeutically is derived from bacteria. l-Asparaginase is
used to treat childhood acute lymphocytic leukemia in combination
with VX and prednisone. The enzyme must be administered either
IV or intramuscularly, because it is destroyed by gastric enzymes

Toxicities include a range of hypersensitivity reactions (because it is
a foreign protein), a decrease in clotting factors, liver abnormalities,
pancreatitis, seizures, and coma due to ammonia toxicity.

60
Q

Interferons

A

Interferons
Human interferons are biological response modifiers and have
been classified into the three types α, β, and γ on the basis of their
antigenicity. The α interferons are primarily leukocytic, whereas the
β and γ interferons are produced by connective tissue fibroblasts
and T lymphocytes, respectively. Recombinant DNA techniques in
bacteria have made it possible to produce large quantities of pure
interferons, including two species designated interferon-α-2a and 2b
that are employed in treating neoplastic diseases. Interferon-α-2a is
currently approved for the management of hairy cell leukemia, CML,
and acquired immunodeficiency syndrome (AIDS)-related Kaposi
sarcoma. Interferon-α-2b is approved for the treatment of hairy cell
leukemia, melanoma, AIDS-related Kaposi sarcoma, and follicular
lymphoma. Interferons interact with surface receptors on other
cells, at which site they exert their effects. Bound interferons are
neither internalized nor degraded. As a consequence of the binding
of interferon, a series of complex intracellular reactions take place.
These include synthesis of enzymes, suppression of cell proliferation,
activation of macrophages, and increased cytotoxicity of lymphocytes.
However, the exact mechanism by which the interferons are cytotoxic
is unknown. Interferons are well absorbed after intramuscular or
subcutaneous injections. An IV form of interferon-α-2b is also available.
Interferons undergo glomerular filtration and are degraded during
reabsorption, but liver metabolism is minimal. Flu-like symptoms
and GI upset are common with these agents. Suicidal ideation and
seizures have been reported.

61
Q

Abiraterone

A

Abiraterone [ab-er-AT-er-own] acetate is an oral agent used in the treatment
of metastatic castration–resistant prostate cancer (Figure 46.34).
Abiraterone acetate is used in conjunction with prednisone to inhibit
the CYP17 enzyme (an enzyme required for androgen synthesis),
resulting in reduced testosterone production. Coadministration with
prednisone is required to help lessen the effects of mineralocorticoid
excess resulting from CYP17 inhibition. Hepatotoxicity may occur, and
patients should be closely monitored for hypertension, hypokalemia,and fluid retention. Joint and muscle discomfort, hot flushes, and
diarrhea are common side effects with this agent.