Sunteți pe pagina 1din 3

CHAPTER 62 IMMUNOSUPPRESSIVE DRUGS 993

Many drugs such as erythromycin, azole antifungal drugs, Dosage


and some calcium channel antagonists that inhibit CYP3A4
(inhibiting the metabolism of cyclosporine) also inhibit Tacrolimus is not routinely used for rheumatologic condi-
Pgp. Drug interactions mediated by these dual mechanisms tions. Different dosages have been used in clinical studies
may result in a twofold to fivefold increase in cyclosporine (see the next paragraph).
concentrations. Azithromycin, in contrast to erythromycin
and clarithromycin, seems unlikely to alter cyclosporine Clinical Indications
levels. The plasma concentrations and clinical toxicity of
several statin lipid-lowering agents are increased substan- Tacrolimus has been studied in a number of rheumatic con-
tially by cyclosporine, but the pharmacokinetics of fluvas- ditions with variable success. In RA, tacrolimus showed
tatin and pravastatin, because they are not metabolized only modest efficacy, and with the availability of a plethora
primarily by CYP3A4, are altered less by cyclosporine.98 of effective conventional and biologic disease-modifying
Nevertheless, the pravastatin AUC curve, a measure of drug anti-rheumatic drugs, the need for a novel immunosuppres-
exposure, was five times higher in patients also receiving sant such as tacrolimus has waned.104,105 Topical 1% tacro-
cyclosporine.99 Of the calcium channel antagonists, dil- limus has been used with moderate success in patients with
tiazem, nicardipine, and verapamil increase cyclosporine resistant skin disease resulting from SLE, subacute cutane-
concentrations; nifedipine and amlodipine have variable ous lupus erythematosus, and discoid lupus erythemato-
effects; and isradipine and nitrendipine do not generally sus.106 A large open-label, randomized, controlled trial in
affect concentrations.100 It is controversial whether nonste- 150 patients with biopsy-confirmed active lupus nephritis
roidal anti-inflammatory drugs (NSAIDs) increase cyclo- showed that remission induction with tacrolimus (0.6 mg/
sporine nephrotoxicity. In many clinical studies, cyclosporine kg/day) was not inferior to MMF (2-3 g/day) when given
and NSAIDs have been safely co-administered79,101; how- with prednisolone (0.6 mg/kg/day) for 6 weeks and followed
ever, increased cyclosporine-associated nephrotoxicity with by azathioprine maintenance, with major infectious epi-
NSAIDs has been reported. Currently, many patients start- sodes occurring in 5.4% versus 9.2% of patients, respec-
ing cyclosporine also take an NSAID. If the creatinine tively.107 In a retrospective study in patients who had
increases, in addition to decreasing the dose of cyclosporine, interstitial lung disease associated with polymyositis or der-
discontinuation of the NSAID may be tried. Grapefruit matomyositis, patients treated with tacrolimus in addition
juice increases plasma concentrations of cyclosporine, so to conventional therapy had a significantly improved sur-
patients should be warned to avoid consuming grapefruit. vival compared with patients who were treated with con-
ventional therapy alone.108

TACROLIMUS (FK506)
Toxicity
Structure
The adverse effects of tacrolimus are dose related and
Tacrolimus, previously known as FK506, is a macrolide include nephrotoxicity, hypertension, hyperkalemia, hyper-
derived from an actinomycete and is widely used in organ uricemia, tremor, hyperglycemia, and gastrointestinal intol-
transplantation as an alternative to cyclosporine. Studies in erance.102 Among patients with RA who took 3 mg of
rheumatic autoimmune disease are less advanced. tacrolimus daily for more than 1 year, 59% experienced an
adverse effect, probably or possibly due to the drug, includ-
ing diarrhea (15%); nausea (10%); tremor (9%); headache
Mechanisms of Action
(9%); abdominal pain (8%); increased creatinine (7%);
Tacrolimus is about 100 times more potent than cyclospo- hypertension (5%); and pneumonia, pancreatitis, hypergly-
rine and, although structurally different, is also a calcineurin cemia, and diabetes mellitus (all <1%). A creatinine
inhibitor. Tacrolimus binds to an intra-cellular binding increase of greater than 30% from baseline to the end of the
protein (FK binding protein), and this drug-immunophilin study was found in 40%.109
complex, in association with calcineurin, suppresses tran-
scription of cytokines such as IL-2, inhibiting the early steps
of T lymphocyte activation (see Figure 62-3).74 MYCOPHENOLATE MOFETIL
Structure
Pharmacology
MMF, a prodrug, is the inactive 2-morpholinoester of myco-
Absorption of tacrolimus after oral administration is phenolic acid, which is hydrolyzed to the active mycophe-
poor and highly variable (range, 4% to 93%; average, nolic acid (MPA), an antibiotic with immunosuppressive
25%).102 Tacrolimus is lipophilic, is widely distributed in effects.110
tissues, and is almost completely metabolized, with an elimi-
nation half-life of 5 to 16 hours.103 As with cyclosporine, Mechanism of Action
Pgp and CYP3A4 in the liver and gut are important deter-
minants of the metabolism and disposition of tacrolimus. Two pathways exist for the synthesis of guanine nucleotides:
Drugs that inhibit CYP3A4 or Pgp can increase tacrolimus the de novo pathway and the salvage pathway. MPA revers-
concentrations (see Table 62-4).102 Impaired hepatic func- ibly inhibits inosine monophosphate dehydrogenase, a
tion, but not impaired renal function, increases tacrolimus crucial enzyme for the de novo synthesis of guanosine
concentrations.103 purines.110,111 Lymphocytes, in contrast to many other cells,

booksmedicos.org
994 PART 8 PHARMACOLOGY OF ANTI-RHEUMATIC DRUGS

are critically dependent on the de novo purine synthesis patients.120 Another observational study also pointed to the
pathway and are a relatively selective target for MPA, potential benefits of therapeutic monitoring in 34 patients
accounting for the ability of the drug to inhibit reversibly with lupus nephritis when dosing was adjusted to achieve
B cell and T cell proliferation without myelotoxicity.112 an AUC of 30 to 60 mg*h/L.121 In this study MPA exposure
MPA results in decreased guanine synthesis and decreased was not associated with adverse events, and patients
DNA synthesis, decreased lymphocyte proliferation, altered with an AUC of 30 mg*h/L or greater had a greater renal
microRNA expression, and decreased antibody produc- response at 1 year. The major glucuronide metabolite of
tion.111-114 MPA also inhibits proliferation of fibroblasts, MPA accumulates in patients with impaired renal function
endothelial cells, and arterial smooth muscle cells and pre- and may cause increased gastrointestinal adverse effects.
vents deposition and contraction of collagen, extra-cellular Because MPA is highly protein bound, it is not cleared by
matrix proteins, and smooth muscle actin.115 hemodialysis.122

Pharmacology Toxicity
MMF is rapidly and completely absorbed and de-esterified MMF is generally well tolerated. The most common adverse
to the active MPA, which is highly (98%) protein bound. effects are gastrointestinal, such as diarrhea, nausea, abdom-
Most MPA (>99%) is found in plasma, with little in cells; inal pain, and vomiting. Occasional infections, leukopenia,
most is glucuronidated to the poorly active, stable phenolic lymphocytopenia, and elevated liver enzymes can occur. Of
glucuronide, which is eliminated in the urine (Figure 54 patients with SLE treated with MMF over a 3-year
62-4).116 Minor metabolites, some of which may be active, period, 16% withdrew because of adverse events, with
have also been described. Peak levels of MPA occur 1 to 2 73% continuing treatment at 12 months.123 In patients with
hours after administration, and secondary peaks, thought to lupus nephritis, diarrhea was more common and serious
be due to enterohepatic circulation, can be seen. The half- infections were less common with MMF than with cyclo-
life of MPA is 16 hours.116 MPA concentrations may vary phosphamide.41 Enteric-coated mycophenolate sodium and
fivefold to tenfold in persons receiving the same dose.117 A MMF have similar rates of adverse effects.124 Opportunistic
small amount of this variability may be due to genetic varia- infections, including one that was fatal, occurred in 3 of 10
tion in uridine-glucuronosyltransferase enzymes.118 Renal patients with idiopathic dermatomyositis who were treated
disease and liver disease have relatively minor effects on the with glucocorticoids and MMF.125
disposition of the active drug, MPA. Generally, dosage
adjustments are not required,116 but because free MPA con- Dosage
centrations are approximately doubled in patients with
severe renal impairment (creatinine clearance <20 to Effective daily dosages of MMF range from 0.5 to 1.5 g
30 mL/min),119 they may be necessary sometimes. A retro- twice a day. In 71 patients with lupus nephritis, the initial
spective study in 16 patients with lupus nephritis showed dose was 1 g/day with a target of 3 g/day. The mean maxi-
that concentration-controlled dose adjustments with a mal dose was 2680 mg/day, and 63% of patients tolerated
target MPA-AUC (from intake until 12 hours after inges- 3 g/day.39
tion) of 60 to 90 mg*h/L was associated with optimized
MPA exposure and excellent renal outcome at 12 months,
Clinical Indications
although adverse effects were reported in one third of
patients, resulting in a switch to azathioprine in two MMF is used as a safer alternative to cytostatic agents in
the treatment of several rheumatic diseases, notably SLE,126
systemic sclerosis,127 vasculitis,128 and inflammatory muscle
disease.125,129 In a 24-week study in persons with lupus
nephritis, mycophenolate was more effective than monthly
Mycophenolate mofetil pulse cyclophosphamide, with a failure rate (without com-
plete or partial remission at 24 weeks, plus those who
stopped treatment for any reason) of 34 of 71 (47.9%)
MPA (active) compared with 48 of 69 in the cyclophosphamide group
(69.6%; P = 0.01).39 In a systematic review of four trials
involving 618 patients, MMF was not superior to cyclophos-
Enterohepatic UGT1A9 UGT2B7
circulation phamide for renal remission, and there was no significant
difference for adverse events (infections, leukopenia, gastro-
MPAG (inactive) AcMPAG (active) intestinal symptoms, herpes zoster, end-stage renal disease,
and death) except for a lower incidence of alopecia and
amenorrhea with the use of MMF compared with cyclo-
Renal excretion
phosphamide.130 Notwithstanding its appeal as a safer alter-
native to cyclophosphamide for remission induction therapy
Figure 62-4 Mycophenolate mofetil is converted to mycophenolic of lupus nephritis, MMF is most commonly used for main-
acid (MPA) and is subsequently metabolized to its glucuronide (Glu) tenance therapy of lupus nephritis, although systematic
conjugates, MPAG and AcMPAG, by different isoforms of UDP glucuroni-
dyl transferases (UGT) in the liver. Pathways of enterohepatic circulation
reviews comparing its efficacy and safety with azathioprine
of MPA via the glucuronide conjugate metabolites are shown. The major- have not yielded consistent results.131,132 A study on repeat
ity of MPAG is excreted in urine. kidney biopsies in 30 patients with lupus nephritis failed

booksmedicos.org
CHAPTER 62 IMMUNOSUPPRESSIVE DRUGS 995

to detect differences between azathioprine and MMF as CONCLUSION


maintenance therapy after a short course of IV cyclophos-
phamide.133 MMF is an alternative remission induction Immunosuppressive drugs are key therapeutic tools in the
agent for patients with anti-neutrophil cytoplasmic management of many rheumatic diseases. They include
antibody–associated vasculitis and evidence of low disease alkylating agents such as cyclophosphamide and purine ana-
activity and who are not at risk of experiencing organ logue cytotoxic drugs such as azathioprine with a long
damage.134 MMF was slightly less effective than azathioprine history of clinical use in rheumatology, as well as noncyto-
in maintaining remission, however.134a In seven patients toxic immunosuppressants such as MMF. In contrast to
with myositis, six had a good clinical and biochemical extra-cellular, exquisitely targeted therapeutics represented
response to mycophenolate,129 an observation that was con- by biologics, our understanding of the in vivo mechanism
firmed in another study in six patients who had refractory of action of immunosuppressive drugs is still limited. In
myositis.135 In addition, in three studies in patients who had contrast, their potential efficacy and safety profiles are gen-
interstitial lung disease associated with dermatomyositis (n erally well known, and serious toxicities can usually be
= 4) or other connective tissue diseases, including RA (n = prevented by careful monitoring of laboratory tests for white
10) and systemic sclerosis (n = 13), MMF was effective in blood cell counts, liver and renal function, and electrolytes.
improving signs and symptoms of lung disease.136-138 MMF Nevertheless, vigilance is required, because the risk of
may be useful as an alternative immunosuppressant to aza- (opportunistic) infection is increased.144,145 As a general
thioprine, particularly in patients with gout who require rule, combination therapy of the different immunosuppres-
therapy with allopurinol because, in contrast to azathio- sants discussed earlier should be avoided. The individual
prine, it does not seem to interact significantly with allopu- response to immunosuppressive therapy can be highly vari-
rinol.72,112 Mycophenolate, 1 g twice daily, was not more able, and decisions to continue a chosen immunosuppres-
effective than placebo in two clinical trials involving 443 sant should be revisited on a regular basis, weighing the
patients with refractory RA139 as assessed with ACR20 benefits and adverse effects.
responses, and a larger mycophenolate-cyclosporine trial
was stopped prematurely. Treatment-related adverse events
were experienced by 51.6%, 73.1%, and 36.1% of patients Full references for this chapter can be found on
receiving MMF, cyclosporine, and placebo, respectively. ExpertConsult.com.
Hypertension, increased serum creatinine, muscle cramps,
hirsutism, and hypertrichosis were more than twice as
common with cyclosporine as with MMF. In all three trials SELECTED REFERENCES
the incidence of serious adverse events with MMF was 1. de Jonge ME, Huitema AD, Rodenhuis S, et al: Clinical pharmaco-
12.1% (compared with 11.3% and 7.5% for cyclosporine kinetics of cyclophosphamide. Clin Pharmacokinet 44:1135–1164,
2005.
and placebo, respectively). Although mycophenolate is 2. Fauci AS, Wolff SM, Johnson JS: Effect of cyclophosphamide upon
used in psoriasis as an effective alternative to methotrexate, the immune response in Wegener’s granulomatosis. N Engl J Med
there are only anecdotal reports of its utility in persons with 285:1493–1496, 1971.
psoriatic arthritis. 3. Struck RF, Alberts DS, Horne K, et al: Plasma pharmacokinetics of
cyclophosphamide and its cytotoxic metabolites after intravenous
versus oral administration in a randomized, crossover trial. Cancer
Pregnancy and Lactation Res 47:2723–2726, 1987.
4. Takada K, Arefayene M, Desta Z, et al: Cytochrome P450 pharma-
MMF is an FDA Pregnancy Category C drug. Mycophenolic cogenetics as a predictor of toxicity and clinical response to pulse
acid is associated with miscarriage and congenital malfor- cyclophosphamide in lupus nephritis. Arthritis Rheum 50:2202–2210,
2004.
mations when used during pregnancy, and therefore it 5. Haubitz M, Bohnenstengel F, Brunkhorst R, et al: Cyclophosphamide
should be avoided whenever possible by women trying to pharmacokinetics and dose requirements in patients with renal insuf-
conceive. Switching from MMF to azathioprine in 19 ficiency. Kidney Int 61:1495–1501, 2002.
patients with SLE did not result in deterioration of global 6. Moroni G, Raffiotta F, Trezzi B, et al: Rituximab vs mycophenolate
and vs cyclophosphamide pulses for induction therapy of active lupus
disease activity, and another study in 54 patients with SLE nephritis: a clinical observational study. Rheumatology (Oxford) 53:
who had quiescent lupus nephritis showed that replacing 1570–1577, 2014.
MMF with azathioprine rarely leads to renal flares, while 7. Illei GG, Austin HA, Crane M, et al: Combination therapy with
pregnancy outcomes were favorable.140,141 MMF is trans- pulse cyclophosphamide plus pulse methylprednisolone improves
ferred into the mother’s milk, and extreme caution should long-term renal outcome without adding toxicity in patients with
lupus nephritis. Ann Intern Med 135:248–257, 2001.
be used in women with childbearing potential and lactating 8. Houssiau FA, Vasconcelos C, D’Cruz D, et al: The 10-year follow-up
mothers. data of the Euro-Lupus Nephritis Trial comparing low-dose and
high-dose intravenous cyclophosphamide. Ann Rheum Dis 69:61–64,
2010.
Drug Interactions 9. Trevisani VF, Castro AA, Neves Neto JF, et al: Cyclophosphamide
versus methylprednisolone for treating neuropsychiatric involvement
Because MPA is glucuronidated and is not metabolized by in systemic lupus erythematosus. Cochrane Database Syst Rev 2:
CYP oxidation, there are few clinically significant drug CD002265, 2006.
interactions. Antacids reduce bioavailability by approxi- 10. Tashkin DP, Elashoff R, Clements PJ, et al: Cyclophosphamide versus
mately 15%, and cholestyramine reduces bioavailability by placebo in scleroderma lung disease. N Engl J Med 354:2655–2666,
2006.
approximately 40%.142 Rifampin treatment reduced MPA 11. Hoyles RK, Ellis RW, Wellsbury J, et al: A multicenter, prospective,
concentrations twofold to threefold.143 Co-administration randomized, double-blind, placebo-controlled trial of corticosteroids
with azathioprine is not recommended. and intravenous cyclophosphamide followed by oral azathioprine for

booksmedicos.org

S-ar putea să vă placă și