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.

2007, Chappuis et al., 2005; Priotto et al., 2006, 2008; Robays et al.,

2008a). In contrast, the drug is largely ineffective for East African

trypanosomiasis. Eflornithine’s difficult treatment regimen is the

primary limitation to its use in the field. However, the negotiation of

the World Health Organization (WHO) of a stable supply of eflornithine

through 2011 has led to the widespread availability of the drug and to

its use in control and research programs run by world health agencies.

Eflornithine is both safer and more efficacious than melarsoprol

for late-stage Gambiense sleeping sickness, and it is now the

recommended first-line treatment for this disease when adequate

care can be provided for its administration. Eflornithine is no longer

available for systemic use in the U.S. but is available for treatment

of Gambian trypanosomiasis by special request from the CDC.

Antitrypanosomal Effects. The effects of eflornithine have been evaluated

both on drug-susceptible and drug-resistant T. brucei in vitro

and in infections with these parasites in rodent models (reviewed in:

Barrett et al., 2007). Eflornithine is a cytostatic agent that has multiple

biochemical effects on trypanosomes, all of which are a consequence

of polyamine depletion. The polyamine putrescine is depleted

to undetectable levels, intracellular levels of spermidine and trypanothine

are reduced by 25-50%, and methionine metabolism is altered.

Depletion of the polyamines, or of trypanothione, would be expected

to be lethal to the cells based on genetic studies that have disrupted the

biosynthetic genes in the pathway. As a consequence, macromolecular

biosynthesis is depressed, and the parasites transform from the

long, slender dividing forms into the short, stumpy nonreplicating

forms. These latter parasites are unable to synthesize variable cell surface

glycoprotein and eventually are cleared by the immune system.

The molecular mechanism of eflornithine action clearly is

inhibition of ornithine decarboxylase. Eflornithine irreversibly

inhibits both mammalian and trypanosomal ornithine decarboxylases,

thereby preventing the synthesis of putrescine, a precursor of

polyamines needed for cell division. Eflornithine inactivates the

enzyme through covalent labeling of an active-site cysteine residue,

and an X-ray structure of the T. brucei enzyme bound to the drug has

been reported (Grishin et al., 1999). A number of studies demonstrate

conclusively that ornithine decarboxylase is the target of eflornithine

action that leads to cell death. Mutant bloodstream trypanosomes

lacking ornithine decarboxylase or wild-type trypanosomes treated

with eflornithine cannot replicate, and mice inoculated with these null

parasites become resistant to infection by wild-type parasites

(Mutomba et al., 1999). The product of the ornithine decarboxylase

reaction, putrescine, rescues the growth deficit in both the mutant parasites

and eflornithine-treated cells. The null mutant parasites grown

with putrescine are not affected by eflornithine, demonstrating the

selectivity of the drug for the target enzyme.

The mechanisms of selective toxicity between the host and parasite,

or between the different species of T. brucei, are less clear

(Barrett et al., 2007). The parasite and human enzymes are equally

susceptible to inhibition by eflornithine; however, the mammalian

enzyme is turned over rapidly, whereas the parasite enzyme is stable,

and this difference likely plays a role in the selective toxicity. In addition,

mammalian cells may be able to replenish polyamine pools

through uptake of extracellular polyamines, whereas the slender

bloodstream forms of human trypanosomes divide within human

blood, which contains only very low levels of these essential compounds.

Further, the parasites lack efficient transport mechanisms.

T. brucei rhodesiense cells are less sensitive to eflornithine

inhibition than T. brucei gambiense cells, and studies in vitro suggest

that the effective doses are increased by 10-20 times in the refractory

cells. The molecular basis for the higher dose requirement in T. brucei

rhodesiense is still poorly understood; however, it has been postulated

to involve differences both in enzyme stability and in the

metabolism of S-adenosylmethionine compared with the sensitive

T. brucei gambiense cell lines.

Absorption, Fate, and Excretion. Eflornithine is given by intravenous

infusion. The drug does not bind to plasma proteins but is

well distributed and penetrates into the CSF, where it is estimated

that concentrations of at least 50 μM must be reached to clear parasites

(Burri and Brun, 2003). Despite the ability of the drug to penetrate

the blood-brain barrier, studies in mice suggest that

eflornithine crosses the healthy blood-CNS interface poorly, and that

it is the breakdown of this barrier caused by T. brucei infection that

allows greater penetration to occur (Sanderson et al., 2008). In

these studies, suramin enhanced eflornithine uptake into the CNS,

suggesting that this combination might lead to lower dose requirements

for eflornithine. Renal clearance after intravenous administration

is rapid (2 mL/minute per kilogram), with >80% of the drug

cleared by the kidney largely in unchanged form (Burri and Brun,

2003). There is some evidence that eflornithine displays dosedependent

pharmacokinetics at the highest doses used clinically.

Therapeutic Uses. Eflornithine is used for the treatment of late-stage

West African trypanosomiasis caused by T. brucei gambiense

(Balasegaram et al., 2006a, 2008; Chappuis, 2007; Chappuis et al.,

2005; Priotto et al., 2006, 2008; Robays et al., 2008a). Most patients

in the reported studies had advanced disease with CNS complications.

The preferred regimen for adult patients was found to be

100 mg/kg given intravenously every 6 hours as a 2-hour infusion for

14 days. Virtually all patients improved on this regimen unless they

were extremely ill, and the WHO and Médecins Sans Frontières

report improved rates >90%. The probability of disease-free survival

2 years after treatment was calculated to be 0.88 in one study (Priotto

et al., 2008) and the case-fatality rate of 1% for eflornithine was the

lowest reported for second-stage sleeping sickness in any study

(Chappuis et al., 2005). Children (<12 years of age) received higher

doses of eflornithine (150 mg/kg given intravenously every 6 hours

for 14 days) based on prior findings that eflornithine trough concentrations

in both the CSF and blood were significantly lower among

children than in adults (Milord et al., 1993). Patients who failed therapy

in this study tended to have trough CSF concentrations <50 μM.

Equal doses of eflornithine were less effective when given by the

oral route probably because of limited bioavailability. The problem

cannot be overcome simply by increasing the oral dose because of

ensuing osmotic diarrhea.

Eflornithine has proven to be less successful for treating

AIDS patients with West African trypanosomiasis, presumably

because host defenses play a critical role in clearing drug-treated

T. brucei gambiense from the bloodstream.

The standard course eflornithine treatment regime is very challenging

to administer in rural settings in Africa. A randomized phase

III trial testing nifurtimox-eflornithine combination therapy for second-stage

T. brucel gambiense suggested that the treatment course for

eflornithine could be reduced to 7 days in combination with nifurtimox.

A recently concluded clinical trial confirms the efficacy of

1425

CHAPTER 50

CHEMOTHERAPY OF PROTOZOAL INFECTIONS

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

Saved successfully!

Ooh no, something went wrong!