22.05.2022 Views

DƯỢC LÍ Goodman & Gilman's The Pharmacological Basis of Therapeutics 12th, 2010

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

elements formed by amplification of DHFR genes in response to

methotrexate treatment) or in stably integrated, homogeneously

staining chromosomal regions or amplicons. First identified as an

explanation for resistance to methotrexate (Schimke et al., 1978),

gene amplification of a target protein has since been implicated in the

resistance to many antitumor agents, including 5-FU and pentostatin

(2′-deoxycoformycin) and has been observed in patients with lung

cancer (Curt et al., 1985) and leukemia. Impaired transport and

polyglutamation have been implicated as mechanisms of resistance

in childhood leukemia.

To overcome resistance, high doses of methotrexate may permit

entry of the drug into transport-defective cells and may permit

the intracellular accumulation of methotrexate in concentrations that

inactivate high levels of DHFR.

The understanding of resistance to pemetrexed is incomplete.

In various cell lines, resistance to this agent seems to arise from loss

of influx transport, TS amplification, changes in purine biosynthetic

pathways, or loss of polyglutamation.

Absorption, Fate, and Excretion. Methotrexate is readily absorbed

from the GI tract at doses of <25 mg/m 2 , but larger doses are absorbed

incompletely and are routinely administered intravenously. Peak concentrations

of 1-10 μM in the plasma are obtained after doses of

25-100 mg/m 2 , and concentrations of 0.1-1 mM are achieved after

high-dose infusions of 1.5-20 g/m 2 . After intravenous administration,

the drug disappears from plasma in a triphasic fashion (Sonneveld

et al., 1986). The rapid distribution phase is followed by a second

phase, which reflects renal clearance (t 1/2

of ~2-3 hours). A third

phase has a t 1/2

of ~8-10 hours. This terminal phase of disappearance,

if unduly prolonged by renal failure, may be responsible for major

toxic effects of the drug on the marrow, GI epithelium, and skin.

Distribution of methotrexate into body spaces, such as the pleural or

peritoneal cavity, occurs slowly. However, if such spaces are

expanded (e.g., by ascites or pleural effusion), they may act as a site

of storage and slow release of the drug, resulting in prolonged elevation

of plasma concentrations and more severe bone marrow toxicity.

Approximately 50% of methotrexate binds to plasma proteins

and may be displaced from plasma albumin by a number of

drugs, including sulfonamides, salicylates, tetracycline, chloramphenicol,

and phenytoin; caution should be used if these drugs are

given concomitantly. Up to 90% of a given dose is excreted

unchanged in the urine within 48 hours, mostly within the first

8-12 hours. Metabolism of methotrexate in humans usually is minimal.

After high doses, however, metabolites are readily detectable;

these include 7-hydroxy-methotrexate, which is potentially nephrotoxic.

Renal excretion of methotrexate occurs through a combination

of glomerular filtration and active tubular secretion. Therefore,

the concurrent use of drugs that reduce renal blood flow (e.g., nonsteroidal

anti-inflammatory agents), that are nephrotoxic (e.g., cisplatin),

or that are weak organic acids (e.g., aspirin, piperacillin) can

delay drug excretion and lead to severe myelosuppression. Particular

caution must be exercised in treating patients with renal insufficiency.

In such patients, the dose should be adjusted in proportion to decreases

in renal function, and high-dose regimens should be avoided.

Methotrexate is retained in the form of polyglutamates for

long periods—for example, for weeks in the kidneys and for several

months in the liver.

It is important to emphasize that concentrations of methotrexate

in CSF are only 3% of those in the systemic circulation at steady

state; hence, neoplastic cells in the CNS probably are not killed by

standard dosage regimens. When high doses of methotrexate are

given (>1.5 g/m 2 ; see “Therapeutic Uses”), cytotoxic concentrations

of methotrexate reach the CNS.

Pharmacogenetics may influence the response to antifolates

and their toxicity. The C677T substitution in methylenetetrahydrofolate

reductase reduces the activity of the enzyme that generates

methylenetetrahydrofolate, the cofactor for TS, and thereby

increases methotrexate toxicity (Pullarkat et al., 2001). The presence

of this polymorphism in leukemic cells confers increased sensitivity

to methotrexate and might also modulate the toxicity and therapeutic

effect of pemetrexed, a predominant TS inhibitor. Likewise, polymorphisms

in the promoter region of TS govern the translation

efficiency of this message and, by governing the intracellular levels

of TS, modulate the response and toxicity of both antifolates (Pui et

al., 2004) and fluoropyrimidines.

Therapeutic Uses. Methotrexate (Amethopterin; RHEUMATREX, TREX-

ALL, others) has been used in the treatment of severe, disabling psoriasis

in doses of 2.5 mg orally for 5 days, followed by a rest period

of at least 2 days, or 10-25 mg intravenously weekly. It also is used

at low dosage to induce remission in refractory rheumatoid arthritis

(Hoffmeister, 1983). Awareness of the pharmacology, toxic potential,

and drug interactions associated with methotrexate is a prerequisite

for its use in these non-neoplastic disorders.

Methotrexate is a critical drug in the management of acute

lymphoblastic leukemia (ALL) in children. High-dose methotrexate

is of great value in remission induction and consolidation and in the

maintenance of remissions in this highly curable disease. A 6- to

24-hour infusion of relatively large doses of methotrexate may be

employed every 2-4 weeks (≥1-7.5 g/m 2 ) but only when leucovorin

rescue follows within 24 hours of the methotrexate infusion. Such

regimens produce cytotoxic concentrations of drug in the CSF and

protect against leukemic meningitis. For maintenance therapy, it is

administered weekly in doses of 20 mg/m 2 orally. Outcome of treatment

in children correlates inversely with the rate of drug clearance.

During methotrexate infusion, high steady-state levels are associated

with a lower leukemia relapse rate (Pui et al., 2004).

Methotrexate is of limited value in adults with AML, except for treatment

and prevention of leukemic meningitis. The intrathecal administration

of methotrexate has been employed for treatment or

prophylaxis of meningeal leukemia or lymphoma and for treatment

of meningeal carcinomatosis. This route of administration achieves

high concentrations of methotrexate in the CSF and also is effective

in patients whose systemic disease has become resistant to

methotrexate. The recommended intrathecal dose in all patients

>3 years of age is 12 mg (Bleyer, 1978). The dose is repeated every

4 days until malignant cells no longer are evident in the CSF.

Leucovorin may be administered to counteract the potential toxicity

of methotrexate that escapes into the systemic circulation, although

this generally is not necessary. Because methotrexate administered

into the lumbar space distributes poorly over the cerebral convexities,

the drug may be given via an intraventricular Ommaya reservoir in

the treatment of active intrathecal disease. Methotrexate is of established

value in choriocarcinoma and related trophoblastic tumors of

women; cure is achieved in ~75% of advanced cases treated sequentially

with methotrexate and dactinomycin and in >90% when early

diagnosis is made. In the treatment of choriocarcinoma, 1 mg/kg of

methotrexate is administered intramuscularly every other day for

1693

CHAPTER 61

CYTOTOXIC AGENTS

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!