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.

capsules and a 100-mg/mL oral solution. Because the original and

microemulsion formulations are not bioequivalent, they cannot be

used interchangeably without supervision by a physician and monitoring

of drug concentrations in plasma. Generic preparations of

both NEORAL and SANDIMMUNE, now widely available, are bioequivalent

by FDA criteria. When switching between formulations,

increased surveillance is recommended to ensure that drug levels

remain in the therapeutic range. This need for increased monitoring

is based on anecdotal experience rather than validated differences.

Because SANDIMMUNE and NEORAL differ in terms of their pharmacokinetics

and definitely are not bioequivalent, their generic versions

cannot be used interchangeably. This has been a source of

confusion to pharmacists and patients. Transplant units need to educate

patients that SANDIMMUNE and its generics are not the same as

NEORAL and its generics, such that one preparation cannot be substituted

for another without risk of inadequate immunosuppression or

increased toxicity.

Blood is most conveniently sampled before the next dose

(a C 0

or trough level). Although convenient to obtain, C 0

concentrations

do not reflect the area under the drug concentration curve (AUC)

as a measure of cyclosporine exposure in individual patients. As

a practical solution to this problem and to better measure the overall

exposure of a patient to the drug, it has been proposed that levels

be taken 2 hours after a dose administration (so-called C 2

levels)

(Cole et al., 2003). Some studies have shown a better correlation of

C 2

with the AUC, but no single time point can simulate the exposure

as measured by more frequent drug sampling. In complex patients

with delayed absorption, such as diabetics, the C 2

level may underestimate

the peak cyclosporine level obtained, and in others who are

rapid absorbers, the C 2

level may have peaked before the blood sample

is drawn. In practice, if a patient has clinical signs or symptoms

of toxicity, or if there is unexplained rejection or renal dysfunction,

a pharmacokinetic profile can be used to estimate that person’s exposure

to the drug. Many clinicians, particularly those caring for transplant

patients some time after the transplant, monitor cyclosporine

blood levels only when a clinical event (e.g., renal dysfunction or

rejection) occurs. In that setting, either a C 0

or C 2

level helps to ascertain

whether inadequate immunosuppression or drug toxicity is present.

As described above, cyclosporine absorption is incomplete

following oral administration and varies with the individual patient

and the formulation used. The elimination of cyclosporine from the

blood generally is biphasic, with a terminal t 1/2

of 5-18 hours (Noble

and Markham, 1995). After intravenous infusion, clearance is ~5-7

mL/min/kg in adult recipients of renal transplants, but results differ

by age and patient populations. For example, clearance is slower in

cardiac transplant patients and more rapid in children. Thus, the intersubject

variability is so large that individual monitoring is required.

After oral administration of cyclosporine (as NEORAL), the

time to peak blood concentrations is 1.5-2 hours (Noble and

Markham, 1995). Administration with food delays and decreases

absorption. High- and low-fat meals consumed within 30 minutes

of administration decrease the AUC by ~13% and the maximum concentration

by 33%. This makes it imperative to individualize dosage

regimens for outpatients.

Cyclosporine is distributed extensively outside the vascular

compartment. After intravenous dosing, the steady-state volume of

distribution reportedly is as high as 3-5 L/kg in solid-organ transplant

recipients.

Only 0.1% of cyclosporine is excreted unchanged in urine.

Cyclosporine is extensively metabolized in the liver by CYP3A and to

a lesser degree by the GI tract and kidneys. At least 25 metabolites

have been identified in human bile, feces, blood, and urine. All of the

metabolites have reduced biological activity and toxicity compared to

the parent drug. Cyclosporine and its metabolites are excreted principally

through the bile into the feces, with ~6% being excreted in the

urine. Cyclosporine also is excreted in human milk. In the presence of

hepatic dysfunction, dosage adjustments are required. No adjustments

generally are necessary for dialysis or renal failure patients.

Therapeutic Uses. Clinical indications for cyclosporine

are kidney, liver, heart, and other organ transplantation;

rheumatoid arthritis; and psoriasis. Its use in dermatology

is discussed in Chapter 65. Cyclosporine generally

is recognized as the agent that ushered in the modern

era of organ transplantation, increasing the rates of

early engraftment, extending kidney graft survival, and

making cardiac and liver transplantation possible.

Cyclosporine usually is combined with other agents,

especially glucocorticoids and either azathioprine or

mycophenolate and, most recently, sirolimus.

The dose of cyclosporine varies, depending on the organ

transplanted and the other drugs used in the specific treatment

protocol(s). The initial dose generally is not given before the transplant

because of the concern about nephrotoxicity. Especially for

renal transplant patients, therapeutic algorithms have been developed

to delay cyclosporine or tacrolimus introduction until a threshold

renal function has been attained. The amount of the initial dose

and reduction to maintenance dosing is sufficiently variable that no

specific recommendation is provided here. Dosing is guided by signs

of rejection (too low a dose), renal or other toxicity (too high a dose),

and close monitoring of blood levels. Great care must be taken to

differentiate renal toxicity from rejection in kidney transplant

patients. Ultrasound-guided allograft biopsy is the best way to assess

the reason for renal dysfunction. Because adverse reactions have

been ascribed more frequently to the intravenous formulation, this

route of administration is discontinued as soon as the patient can

take the drug orally.

In rheumatoid arthritis, cyclosporine is used in severe cases

that have not responded to methotrexate. Cyclosporine can be combined

with methotrexate, but the levels of both drugs must be monitored

closely. In psoriasis, cyclosporine is indicated for treatment of

adult immunocompetent patients with severe and disabling disease

for whom other systemic therapies have failed. Because of its mechanism

of action, there is a theoretical basis for the use of cyclosporine

in a variety of other T cell–mediated diseases. Cyclosporine reportedly

is effective in Behçet’s acute ocular syndrome, endogenous

uveitis, atopic dermatitis, inflammatory bowel disease, and nephrotic

syndrome, even when standard therapies have failed.

Toxicity. The principal adverse reactions to cyclosporine therapy are

renal dysfunction and hypertension; tremor, hirsutism, hyperlipidemia,

and gum hyperplasia also are frequently encountered.

Hypertension occurs in ~50% of renal transplant and almost all cardiac

transplant patients. Hyperuricemia may lead to worsening of

1011

CHAPTER 35

IMMUNOSUPPRESSANTS, TOLEROGENS, AND IMMUNOSTIMULANTS

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

Saved successfully!

Ooh no, something went wrong!