Dermatologic Clinics
Volume 23 • Number 2 • April 2005
Copyright © 2005 W. B. Saunders Company
The Use of Systemic Immune Moderators in Dermatology: An Update
Dana Kazlow Stern, MD a
Jackie M. Tripp, MD b
Vincent C. Ho, MD b
Mark Lebwohl, MD c, *
--------------------------------------------------------------------------------
a Department of Dermatology, Mount Sinai Medical Center, Mount Sinai School of
Medicine, 1 Gustave L. Levy Place, Box 1048, New York, NY 10029-6574, USA
b Division of Dermatology, Department of Medicine, Vancouver Hospital and Health
Sciences Center and University of British Columbia, 835 West 10th Avenue,
Vancouver, BC V5Z 4E8, Canada
c Department of Dermatology, The Mount Sinai School of Medicine, Mount Sinai
Medical Center, Clinical Trials Center, 5 East 98th Street, 5th floor, New York,
NY 10029-6574, USA
--------------------------------------------------------------------------------
* Corresponding author
--------------------------------------------------------------------------------
E-mail address: mark.lebwohl@mssm.edu
Dr. Stern has been an investigator for Biogen Idec, Inc., Genentech, Inc.,
Amgen, Inc., and Centocor Inc. Dr. Lebwohl has been an investigator, speaker,
and consultant for Centocor, Inc., Biogen Idec, Inc., Genentech, Inc., and Amgen
Inc., and he has been a speaker and investigator for Abbott, Inc.
PII S0733-8635(04)00092-0
--------------------------------------------------------------------------------
In addition to corticosteroids, dermatologists have access to an array of
immunomodulatory therapies. Azathioprine, cyclophosphamide, methotrexate,
cyclosporine, and mycophenolate mofetil are the systemic immunosuppressive
agents most commonly used by dermatologists. In addition, new developments in
biotechnology have spurred the development of immunobiologic agents that are
able to target the immunologic process of many inflammatory disorders at
specific points along the inflammatory cascade. Alefacept, efalizumab,
etanercept, and infliximab are the immunobiologic agents that are currently the
most well known and most commonly used by dermatologists. This article reviews
the pharmacology, mechanism of action, side effects, and clinical applications
of these therapies.
--------------------------------------------------------------------------------
When inflammatory dermatoses are severe or recalcitrant to topical therapies,
dermatologists are occasionally required to prescribe immunomodulatory drugs.
The first-line systemic anti-inflammatory therapy is the oral administration of
corticosteroids, but frequently other immunomodulatory drugs are needed to
control disease activity, while at the same time limiting or even eliminating
the need for corticosteroids. These immunomodulatory therapies can be divided
into two broad categories: the more traditional and well-known nonsteroidal
immune-modifying drugs, also commonly referred to as the systemic therapies, and
the newer immunobiologic agents.
The most common nonsteroidal immune-modifying drugs include the following
agents: azathioprine, cyclophosphamide, cyclosporine, methotrexate, and
mycophenolate mofetil (MMF). These agents are predominantly cytotoxic, but data
seem to suggest that methotrexate has significant anti-inflammatory activity as
well. The other category encompasses the plethora of newer immunobiologic agents
that are being developed, the most well known of which include alefacept,
efalizumab, etanercept, and infliximab. The current authors and others have
previously reviewed the pharmacology, mode of action, and adverse effects of
some of these agents [1], [2], [3], [4], [5], [6]. The current article reviews
new data concerning the pharmacology, mechanism of action, adverse effects, and
dermatologic applications of these two categories of therapeutic agent.
Familiarity with the disease-specific clinical efficacy of each of these
medications and their dosages and side-effect profiles allows for the proper and
efficacious use of these drugs to treat dermatologic disease.
AZATHIOPRINE
Inititially used as an immunosuppressant for renal transplantation, azathioprine
has been available for more than 40 years, and because of its relatively
favorable therapeutic index, it is one of the more commonly used
immunomodulatory drugs in dermatology. Structurally, it is a synthetic purine
analog formed by attaching an imidazole ring to 6-mercaptopurine (6-MP). This
analog is a prodrug and is quickly converted to 6-MP by a nonenzymatic
nucleophilic attack by sulfhydryl compounds present in erythrocytes and body
tissues.
PHARMACOLOGY
A comparison of the main pharmacologic attributes of the immunosuppressants
discussed in this article is presented in Table 1. Azathioprine is rapidly
absorbed after oral administration. The drug has a plasma half-life of only 3
hours because of its conversion to 6-MP. Active metabolites have a long
half-life, however, allowing once-daily dosing. The 6-MP is metabolized by means
of three competing pathways: thiopurine methyltransferase (TPMT) catalyzes
S-methylation to 6-methyl mercaptopurine, an inactive compound. Xanthine oxidase
catalyzes oxidation to the inactive 6-thiouric acid. Hypoxanthine-guanine
phosphoribosyl transferase (HGPRT) catalyzes the conversion of 6-MP to the
active 6-thioguanine metabolites. Erythrocyte levels of 6-thioguanine
nucleotides correlate with absolute neutrophil counts [7], and steady-state
levels may take days to years to achieve. A genetic polymorphism controls TPMT
activity. In a recent review of more than 3000 patients, approximately 80% of
the patients had normal TPMT activity, 9% had enzymatic activity that was above
normal, and 10% displayed low activity [8]. It was also found that 0.45% (1:220)
of the study population had no detectable enzyme activity. This ratio is higher
than the previously reported 1 in 300 occurrence of undetectable TPMT levels in
the population reported in other smaller studies. In renal transplant patients,
low TPMT activity has been found to correlate with a higher incidence of
leukopenia caused by azathioprine, and those with higher levels were found to be
less immunosuppressed [9]. Patients with intermediate TPMT activity may be more
likely to develop late-onset neutropenia [10].
Table 1 . Pharmacology of the common immunosuppressants used in dermatology
Features Azathioprine Cyclophosphamide Methotrexate Cyclosporine MMF
Mechanism of action Cell cycle–specific antimetabolite, inhibition of neutrophil
trafficking, inhibition of cellular cytotoxicity Non–cell cycle–specific
antimetabolite, selective macrophage inhibition, selective B-cell suppression
Cell cycle–specific antimetabolite, inhibition of neutrophil chemotaxis,
inhibition of IL-1, IL-6, and IL-8 release Inhibition of signal transduction and
IL-2 production in lymphocytes Cell cycle–specific antimetabolite, inhibition of
Tc and B-cell proliferation
Rate of onset of clinical immunosuppression Slow (4–8 wk) Moderately rapid (2–4
wk) Moderately rapid (2–4 wk) Rapid (1–2 wk) Slow (4–8 wk)
Route of administration Per os Per os/IV Per os/SC/intramuscular/IV Per os Per
os
Route of elimination Hepatic metabolism and renal excretion Hepatic metabolism
and renal excretion Renal excretion by active tubular secretion Hepatic
metabolism Hepatic metabolism and renal excretion
Dose modification in presence of:
Renal insufficiency Moderate decrease in dosage may be required Moderate
decrease in dosage may be required Relatively contraindicated, significant dose
reduction necessary No change in dosage necessary Moderate decrease in dosage
may be required
Hepatic insufficiency No change in dose necessary Moderate decrease in dosage
may be required Moderate decrease in dosage may be required Significant dose
alteration needed No change in dose necessary
Major potential drug interactions Allopurinol, warfarin Allopurinol Probenecid,
NSAIDs, cotrimazole Multiple (see text) Acyclovir, iron, antacids,
cholestyramine
Major toxicity Bone marrow depression, carcinogenesis Bone marrow depression,
hemorrhagic cystitis, carcinogenesis Hepatotoxicity, bone marrow depression,
carcinogenesis Hypertension, nephrotoxicity, carcinogenesis Bone marrow
depression, carcinogenesis
The determination of pretreatment red blood cell TPMT activity has been
advocated to detect those patients at risk for early-onset neutropenia [11].
This test can be accomplished using either a radiochemical assay of enzyme
activity or by high-pressure liquid chromotography. Recently, the isolation and
characterization of mutant alleles causing TPMT deficiency have enabled
identification of more than 80% of all TPMT mutant alleles in white persons and
have allowed the diagnosis of TPMT deficiency and heterozygosity based on
genotype [12]. If the drug is used without pretreatment measurement of TPMT
activity, it is now recommended that patients be counseled about the associated
risks and the inadequacy of blood count monitoring as a method of preventing
early toxicity [13]. TPMT measurement is becoming more widely available, and one
recent cost-effectiveness study has shown that polymerase chain reaction testing
to identify TPMT polymorphisms before treatment represents good economic value
in certain health care settings [14].
Other factors affect TPMT activity. Sulfasalazine and its metabolite
5-aminosalicylic acid inhibit TPMT activity, whereas 6-MP itself and diuretics
can induce its activity [15]. Further, very low lymphocyte activities of
5-nucleotidase have been documented in transplant recipients with normal TPMT
activity who developed neutropenia when given azathioprine. All patients require
tailoring of this medication, guided by leukocyte and platelet counts.
The 6-thioguanine metabolites prevent the interconversion of purine bases and
inhibit de novo biosynthesis of purine bases in an S-phase–specific fashion.
This antiproliferative effect may not be solely responsible for the
immunosuppressive action of azathioprine, because effects on natural killer cell
(NKC) function, T-cell signaling, T-cell cytolytic activity, prostaglandin
production, and neutrophil activity have been noted [1]. No significant changes
in interleukin (IL) levels have been noted.
ADVERSE EFFECTS
HEMATOLOGIC
Myelosuppression, manifesting as leukopenia, thrombocytopenia, or pancytopenia,
is the most significant adverse effect of azathioprine. As stated previously,
early toxicity, which is of rapid onset and severe, may be predicted by a
knowledge of TPMT activity. Continued monitoring is required, because late-onset
myelosuppression occurs more slowly and may be delayed for years. Significant
leukopenia was seen in 10% of patients (3 of 29) who had pemphigus vulgaris
treated at a mean dosage of 2 mg/kg per day for 4 to 12 years [16], and, in a
survey of azathioprine use by dermatologists in the United Kingdom, 45% of
respondents had observed significant myelosuppression [17].
GASTROINTESTINAL
Nausea and vomiting are the most frequent side effects, occurring 12.4 times and
5.1 times per 100 patient-years, respectively, in patients receiving standard
dosages for rheumatoid arthritis (RA), an average of 104 mg per day [18]. A
recent study has shown that azathioprine-related gastrointestinal side effects
are independent of TPMT polymorphism [19]. Hepatotoxicity was noted in 8% of
patients receiving azathioprine and prednisolone for bullous pemphigoid [20].
OPPORTUNISTIC INFECTIONS
An increased susceptibility to opportunistic infections has been reported in
patients receiving both azathioprine and corticosteroids, even in the absence of
leukopenia. Eruptions of herpes simplex, herpes zoster, and verrucae may be more
common [1].
ONCOGENIC POTENTIAL
Azathioprine has been associated with an increased incidence of malignancy in
renal transplant patients. Malignancies have included lymphomas, Kaposi's
sarcoma, renal carcinomas, carcinomas of the cervix and vulva, and skin cancers
[21]. An analysis of the risk of skin cancer in renal transplant recipients,
with a multivariate analysis of subgroups on either long-term cyclosporine or
azathioprine with or without corticosteroids, showed no differences between the
groups, suggesting that the increased risk is associated with the drug-induced
immunosuppression and is independent of the agents used to achieve this [22]. An
analysis of 1191 patients who have multiple sclerosis (MS) identified patients
who have cancer, matched them to patients who do not have cancer, and compared
them in terms of exposure to azathioprine therapy [23]. MS patients given
azathioprine were found to have a relative increase in cancer risk of 1.3 when
treated less than 5 years, of 2.0 when treated 5 to 10 years, and of 4.4 when
treated more than 10 years. Wide confidence intervals undermined these results,
but overall, the long-term risk for these patients was low. A more recent
retrospective study of 626 patients taking azathioprine who were observed for a
mean duration of 6.9 years showed no increased risk of cancer diagnosis [24].
HYPERSENSITIVITY REACTIONS
Several reports call attention to azathioprine hypersensitivity syndrome [25],
[26], [27], [28]. This syndrome has been reviewed comprehensively by Saway et al
[29]. The time of onset is from 3 hours to 42 days after onset of medication
use, with a mean of 14 days. Hypersensitivity syndrome is rare, with some 30
case reports in the literature. Manifestations may include the following:
hypotension; shock; a maculopapular, urticarial, or vasculitic eruption; fever;
acute hepatotoxicity; pancreatitis; rhabdomyolysis; acute renal failure; and
pneumonitis. Recently, a higher incidence of acute febrile toxic reaction has
been reported in 4 of 43 patients (9.3%) with RA who were on long-term
methotrexate and in whom azathioprine in usual dosages was added, suggesting a
possible adverse drug interaction [30].
MISCELLANEOUS
Azathioprine, used in conjunction with isotretinoin, has been reported to induce
curling of hair [31]. Azathioprine and isotretinoin also have been associated
independently with the development of curly hair. In addition, azathioprine
administered following corneal transplantation has been reported to cause
bilateral macular hemorrhage secondary to aplastic anemia [32].
CLINICAL USE IN DERMATOLOGY
A 1997 survey of current practice in the use of azathioprine by dermatologists
in the United Kingdom revealed that the most common indications for its use were
the immunobullous disorders (pemphigoid and pemphigus, where it was used in
conjunction with corticosteroids), atopic dermatitis and chronic actinic
dermatitis (both more often in the absence of corticosteroids), and
dermatomyositis (again as a steroid-sparing agent) [17]. Other common uses
included pyoderma gangrenosum, systemic lupus erythematosus, psoriasis, lichen
planus, and cutaneous lupus erythematosus. There remain very few controlled
trials to support the drug's use in dermatology. The data supporting the
adjuvant use of azathioprine in pemphigus were reviewed; these data consisted of
seven studies totalling 105 patients with no significant difference in outcome
between patients treated with cyclophosphamide or azathioprine [33]. Likewise,
the use of azathioprine in pemphigoid has been variably shown to have a
steroid-sparing effect [1], and in a study in which dosages of both
corticosteroid and azathioprine were fixed [20], to result in more adverse
effects. Azathioprine may be effective as a steroid-sparing agent in moderately
severe systemic lupus, but there is conflicting evidence as to its efficacy in
severe disease [1]. The efficacy of azathioprine in dermatomyositis-polymyositis
also has been questioned [34].
A retrospective review of 35 patients who had severe, longstanding atopic
dermatitis treated with azathioprine showed that 3 of 35 patients had severe
nausea, which resulted in treatment discontinuation. The median difference in
disability index before and after treatment was highly significant in the 32
patients who tolerated the drug [35]. More recently, a double-blind, randomized,
placebo-controlled trial of azathioprine was performed in 37 adult patients who
had severe atopic dermatitis (AD), and it similarly showed significant
improvement in symptom scores. There were, however, gastrointestinal side
effects in 14 patients [36]. A favorable short-term response to azathioprine in
four patients with intractable prurigo nodularis also has been reported [37].
The efficacy of azathioprine in controlling the ocular and extraocular
manifestations of Behçet's disease was shown in a double-blind controlled trial
[38]. The follow-up of this study showed that the beneficial effect of treatment
favorably affected the long-term prognosis of these patients [39].
Azathioprine has been shown to be an effective steroid-sparing agent for
generalized lichen planus [40] and also has been used successfully as
monotherapy in two patients [41].
USAGE GUIDELINES
Azathioprine is contraindicated in patients with known hypersensitivity to the
drug. Dosage adjustment, as guided by serial blood counts, may be required in
patients with hepatic or renal insufficiency. The drug should not be used during
pregnancy unless the benefits outweigh the risks. Both the parent drug and its
metabolites can cross the placenta and act as potential teratogens. The
magnitude of the teratogenic risk has been evaluated as minimal to small [42],
[43]. In contrast to the other cytotoxic agents, there is no evidence that
azathioprine produces gonadotoxicity or infertility.
DRUG INTERACTIONS
Because allopurinol inhibits the metabolism of azathioprine by xanthine oxidase,
the two drugs should not be used concurrently if possible. During concurrent
therapy, the dosage of azathioprine should be decreased by at least two thirds,
although this dosage reduces, but does not abolish, the risk of myelotoxicity
[44]. In this clinical situation, the monitoring of 6-thioguanine levels would
seem warranted [45].
Angiotensin-converting enzyme (ACE) inhibitors have been shown to potentiate
anemia in renal transplant patients concurrently on azathioprine [46]. This
finding seems to be caused by the erythropoietin-lowering effect of the ACE
inhibitors rather than from a pharmacokinetic interaction between the two drugs
[47]. To the current authors' knowledge, a clinically significant interaction
has not been documented in a nonrenal transplant setting, although the current
American College of Rheumatology guidelines advise against this combination
[48]. Both azathioprine and trimethoprim-sulfamethoxazole are antimetabolites
that have a synergistic effect in inhibiting bone marrow proliferation [49].
Again, however, no significant interaction was noted in renal transplant
patients treated with concomitant therapy over 4 months [50]. Because of
inhibition of TPMT activity, sulfasalazine may potentiate azathioprine toxicity.
Finally, warfarin resistance has been noted in patients treated with
azathioprine [51], [52].
DOSAGE AND MONITORING
Azathioprine is supplied for oral use as scored 50-mg tablets. The usual
starting dose is 1 to 2 mg/kg per day in single or two divided dosages. One
recent study of 48 children receiving azathioprine for atopic dermatitis
initiated therapy at dose levels of 2.5 to 3.5 mg/kg in those with a normal TPMT
level [53]. Therapeutic response for azathioprine occurs in 6 to 8 weeks. After
6 to 8 weeks, the dose may be increased by 0.5 mg/kg at 4-week intervals
according to white blood cell counts and clinical response, and generally should
not exceed 3 mg/kg per day. If there is no beneficial response in 12 to 16
weeks, treatment using azathioprine should be discontinued. There are no formal
consensus guidelines for monitoring azathioprine when used for dermatologic
indications. The current authors' recommendations have been adapted from
published guidelines in the dermatologic literature [54] and are presented in
Table 2.
Table 2 . Monitoring guidelines for the use of immunosuppressants Drug [Ref.]
Monitoring tests Frequency Guidelines for dosage modification
Azathioprine [54] CBC with differential WBC and platelets Weekly ×4, every 2 wk
×2, then monthly Decrease dose if WBC < 4 × 109/L, platelets < 1011/L,
discontinue if WBC < 2.5 × 109/L
Cyclophosphamide [54] CBC with differential WBC and platelets Weekly ×8, then
monthly Decrease dose if WBC < 4 × 109/L, platelets < 1011/L, discontinue if WBC
< 2.5 × 109/L
Renal function tests: BUN, creatinine Monthly
urinalysis weekly ×8, then every 2 wk, urine cytology when > 50 g total dose
monthly Discontinue if hematuria
Liver function tests Monthly
Methotrexate [105] CBC with differential WBC and platelets Weekly ×2, every 2 wk
×2, then monthly Discontinue for 2–3 wk if WBC < 3.5 × 109/L, discontinue if WBC
< 2.5 × 109/L or platelets < 1011/L, increased MCV necessitates folate
administration
Renal function tests: BUN, creatinine Every 3–4 mo
Liver function tests Every 1–2 mo, ensure it is at least 1 week after most
recent dose If persistently elevated, withhold for 1–2 wk and repeat; if
elevation persists for 2–3 mo perform liver biopsy
Liver biopsy If no risk factors, at cumulative doses of 1.5 g, 3 g, and 4 g. If
risk factors, then first at 2–4 mo of therapy, then at cumulative doses of 1–1.5
g, 3 g, 4 g. After a cumulative dose of 4 g, biopsy every 1–1.5 g.
Cyclosporine [189] CBC with differential WBC and platelets Every month, then
every 2–3 mo
Renal function tests: BUN and creatinine (×3) Biweekly ×6, then monthly Decrease
dose if creatinine increase is >30% over baseline; if persistent over 2
evaluations, discontinue
Blood pressure Biweekly ×6, then monthly
Urinalysis Biweekly ×2, then monthly
Liver function tests Biweekly ×2, then monthly
Potassium, uric acid, magnesium, cholesterol, triglycerides Biweekly ×2, then
monthly
Creatinine clearance Not done routinely
MMF [207] CBC with differential WBC and platelets Weekly ×4, every 2 wk ×4, then
monthly Decrease dose or discontinue if neutropenic
For all immunosuppressants, lymph node examination, complete physical
examination, stool guaiac, skin cancer examination, and PAP smear every 6 mo are
recommended.
Abbreviations: BUN, blood urea nitrogen; CBC, complete blood (cell) count; MCV,
molluscum contagiosum virus; WBC, white blood cell (count).
CYCLOPHOSPHAMIDE
Cyclophosphamide is another cytotoxic agent used on occasion by dermatologists
as an immune modulator. Because of the risk of bladder toxicity, myelotoxicity,
gonadotoxicity, and malignancy, its use is reserved for serious and potentially
life-threatening disorders in which the benefit outweighs the risks.
PHARMACOLOGY
Cyclophosphamide belongs to the nitrogen mustard family of alkylating agents. It
was initially synthesized as an orally active form of chlorethamine (nitrogen
mustard). It is well absorbed after oral administration, with peak plasma levels
within 3 hours. The plasma half-life is 5 to 6 hours, with 12% to 14% of the
drug being plasma bound [55]. The drug is metabolized by hepatic cytochrome P450
microsomal enzymes and oxidized to form the active metabolites phosphoramide
mustard and acrolein. No consistent association between renal or hepatic
insufficiency and cyclophosphamide toxicity has been found, and the drug can be
metabolized independently of hepatic metabolism.
Phosphoramide mustard is the metabolite that alkylates DNA and inhibits DNA
replication. Unlike azathioprine metabolites, the action of phosphoramide
mustard is not S-phase specific and it can affect noncycling cells. There is a
differential cytotoxicity to various lymphoid cell populations, with selective
suppression of B cells [56]. Hence, this drug has its greatest effect in
suppressing humoral immunity. Cyclophosphamide has a more variable response on
cell-mediated immunity, and low-dose cyclophosphamide has been shown to
potentiate immune responses in experimental systems. Immunostimulation has been
related to increased IL-12 gene expression [57]. Variable effects on macrophages
also have been noted [58].
ADVERSE EFFECTS
HEMATOLOGIC
Myelosuppression, consisting primarily of leukopenia, is the most common adverse
reaction and can be dose-limiting. After intravenous (IV) administration, the
nadir occurs at 8 to 12 days and recovery takes 18 to 25 days. A similar lag is
noted with oral therapy. During the leukopenia, the patient is at increased risk
of infection by pathogenic and opportunistic organisms. Thrombocytopenia and
anemia occur less frequently.
GASTROINTESTINAL
Anorexia, nausea, and vomiting occur commonly with IV administration. These
symptoms are dose-related and have been associated with the phosphoramide
mustard metabolite. Oral ondansetron and dexamethasone have been shown to
control nausea resistant to standard antiemetics in patients treated with IV
cyclophosphamide for lupus nephritis [59]; however, a recent double-blind study
of 258 patients undergoing emetic chemotherapy showed that a regimen of oral
granisetron and oral dexamethasone did not achieve significantly better results
than a course of metoclopramide, and that the former regimen was thus not
cost-effective [60].
UROLOGIC
Urologic adverse effects are one of the major toxicities of this drug and can
include dysuria, urgency, hematuria, bladder fibrosis, and necrosis [61]. Death
from hemorrhagic cystitis has occurred. Hemorrhagic cystitis can develop at
significantly lower dosages and with shorter durations of therapy in patients
treated intravenously rather than orally. At the dose commonly used in
dermatology, the average risk is 5% to 10% [62]. A review of 145 patients with
Wegener's granulomatosis treated with oral cyclophosphamide at a dosage of 2
mg/kg, however, identified nonglomerular hematuria in 50% of patients (73 of
145) [63]. Nonglomerular hematuria occurred earlier in smokers and was chronic
and recurrent in 38% of patients, even after discontinuation of the drug.
Acrolein is the metabolite believed to be responsible for urotoxicity.
Dehydration is an undisputed risk factor. High fluid intake and concurrent
administration of mesna (sodium 2-mercaptoethane sulfonate) reduce the risk of
this complication during IV administration. Mesna binds to acrolein in the
bladder, producing an inactive compound that is eliminated in the urine. The
relative efficacy of these two maneuvers is still a matter of debate [64].
ONCOGENIC POTENTIAL
Patients taking long-term oral cyclophosphamide have up to a 45-fold increase in
the incidence of bladder cancer [65]. The estimated incidence of transitional
cell carcinoma of the urinary tract, after first exposure to cyclophosphamide in
patients who have Wegener's granulomatosis, was 5% at 10 years and 16% at 15
years [63]. Nonglomerular hematuria identified a subgroup at increased risk but
was not invariably present. In patients who had RA and who were receiving oral
cyclophosphamide, a 4-fold increase in solid tumors and a 16-fold increase in
lymphoreticular malignancies have been observed [66], [67]. In a case-control
study in which patients were followed up for 2 decades after treatment, the
relative risk of cancer for those treated with cyclophosphamide was 1.5 and
bladder cancer was noted as late as 17 years after discontinuation of the drug
[68]. There was a recent report of a bladder carcinoma arising as late as 20
years after oral therapy with cyclophosphamide [69].
REPRODUCTIVE TOXICITY
Azoospermia and amenorrhea have been noted after both oral and IV pulse therapy.
The incidence is as high as 50%, and risk factors include age, longer period of
treatment, and degree of marrow suppression. Male patients may consider a sperm
bank before treatment. A study of men given testosterone before and during an
8-month cycle of cyclophosphamide for nephrotic syndrome suggests that this
method may help preserve fertility [70]. Suppressing ovarian function with oral
contraceptives may protect the ovary, and some advocate the use of
coadministering gonadotropins [71].
OPPORTUNISTIC INFECTIONS
Pathogenic and opportunistic infections can occur during treatment. They may
occur in the absence of leukopenia. In a series of 100 patients with systemic
lupus erythematosus, infections occurred with equal prevalence in those who
received IV (39%) or oral (40%) medication. Risk factors for severe infections
included a white blood cell count of less than 3000 and sequential IV and oral
therapy [72].
MISCELLANEOUS
Anagen effluvium, mucositis, and hyperpigmentation have been reported. When used
in higher dosages, cardiomyopathy, pneumonitis, pulmonary fibrosis,
hepatotoxicity, and syndrome of inappropriate antidiuretic hormone also have
been reported [61].
CLINICAL USE IN DERMATOLOGY
Cyclophosphamide continues to be used for potentially life-threatening
dermatoses that are resistant to other forms of treatment. These may include
systemic necrotizing vasculitis, severe forms of systemic lupus, severe
blistering disorders, multicentric reticulohistiocytosis, relapsing
polychondritis, and pyoderma gangrenosum [2]. Cyclophosphamide is believed to be
a more effective steroid-sparing agent than azathioprine in the treatment of
resistant pemphigus, although there is no incontrovertible evidence in the
literature to support this view [33]. There are also reports describing
successful cyclosphosphamide therapy in recalcitrant pemphigus foliaceous [73]
and pemphigus vulgaris [74]. IV pulse cyclophosphamide is being used more
frequently by dermatologists based on its effectiveness in lupus erythematosus
nephritis [75]. With adequate hydration and use of mesna, the incidence of
hemorrhagic cystitis is lower than with oral therapy, and it is hoped that the
risk of cancer is lowered as well. Using a combination of dexamethasone pulse,
cyclophosphamide pulse, and oral cyclophosphamide, clinical remissions have been
claimed in 61 of 79 patients who have pemphigus [76]. The combined use of IV
pulse and oral cyclophosphamide was recently reported in ocular pemphigoid [77]
and in resistant bullous pemphigoid [78] to decrease total cyclophosphamide
dose. A patient who had recalcitrant herpes gestationis also has responded to
pulse IV cyclophosphamide after delivery of the infant [79]. A report of a
patient with toxic epidermal necrolysis responding to IV cyclophosphamide [80]
adds to the four cases in the literature [81]. The feasibility of outpatient
monthly oral bolus cyclophosphamide therapy has been documented in patients with
active lupus erythematosus [82]. Patients were instructed to maintain a fluid
intake of 2 to 3 L per day and were given concurrent oral mesna. The safety and
patient acceptance remain to be documented. The comparative efficacy and
toxicities of pulse versus continuous oral administration in dermatologic
conditions also remain to be determined. Reports of the successful use of oral
cyclophosphamide in scleromyxedema [83] and in multicentric histiocytosis (in
combination with methotrexate and corticosteroids) [84] add to the anecdotal
literature of the use of this drug in rare, recalcitrant dermatoses.
USAGE GUIDELINES
Cyclophosphamide is contraindicated in patients who have demonstrated
hypersensitivity, in pregnancy, and during breast feeding.
DRUG INTERACTIONS
Allopurinol increases the toxicity of cyclophosphamide through an unknown
mechanism. There is a potential risk of increasing the incidence of lung
toxicity in those taking amiodarone [85]. Drugs that alter the P450 system may
affect cyclophosphamide pharmacokinetics.
DOSAGE AND MONITORING
Cyclophosphamide is supplied for oral use as 25- and 50-mg tablets. The
recommended oral dosage is 1 to 3 mg/kg per day (50–200 mg/d). This drug should
be taken in the morning followed by adequate hydration throughout the day (3 L
of fluid/d). In monthly IV pulse therapy, 0.5 to 1.0 g/m2 is infused over 1 hour
followed by vigorous IV hydration, and optional mesna usually at 160% of the
cyclophosphamide dose separated into four doses at 0, 3, 6, and 9 hours. Some
clinicians follow this treatment with daily oral cyclophosphamide at a dose of
50 mg/day. Further pulses are adjusted to obtain a white blood cell count nadir
of 2 to 3 × 109 /L after 10 to 14 days. Monitoring guidelines are outlined in
Table 2. It is recommended that all patients have a urinalysis every 3 to 6
months, even after the drug is discontinued, and that microscopic hematuria be
evaluated by cystoscopy. Urine cytology should be done every 6 to 12 months, and
routine cystoscopy should be considered every 1 to 2 years for all patients who
have a history of microscopic hematuria [63].
METHOTREXATE
Methotrexate is a synthetic analog of folic acid that has been used in
dermatologic therapy since 1951. Like azathioprine and cyclophosphamide, it has
cytotoxic properties. There is now increasing evidence for separate mechanisms
of anti-inflammatory activity.
PHARMACOLOGY
Methotrexate is usually given orally for the treatment of cutaneous disease but
can be administered subcutaneously with similar kinetics [86]. Occasionally,
this agent is given intramuscularly in the presence of gastrointestinal
intolerance. There is marked variability in the bioavailability of oral
methotrexate [87]. Approximately 35% to 50% of the drug is bound to albumin.
Maximum blood levels occur 1 to 2 hours after oral and intramuscular
administration [88], [89]. Polyglutamate derivatives are formed intracellularly
and are the principal bioactive metabolites that are long lasting. Hepatic
oxidation forms 7-hydroxy-methotrexate, a minor metabolite. The serum half-life
is 6 to 7 hours for the methotrexate but much longer for the polyglutamate
derivatives. The drug is excreted by the kidney, with 80% of a dose eliminated
unchanged within 24 hours. Methotrexate is filtered by the glomeruli and then
undergoes bidirectional transport within the renal tubule. Enterohepatic
recirculation occurs to a minor degree.
Methotrexate binds intracellularly to dihydrofolate reductase, thereby
preventing the reduction of folate cofactors (the conversion of dihydrofolate to
tetrahydrofolate) and inhibiting the thymidylate synthesis. Methotrexate
polyglutamates further inhibit thymidylate synthase and other folate-dependent
enzymes, such as aminoimidazole-carboxamide ribonucleoside (AICAR)
transformylase. Purine synthesis, required for DNA, RNA, and protein synthesis,
is thereby inhibited. The methylation of homocysteine to methionine is
inhibited, affecting the synthesis of polyamines such as spermidine and spermine
[90]. Methotrexate may directly inhibit epidermal cell proliferation by these
mechanisms, although lymphoid and macrophage cell lines have been shown to be
much more susceptible to the growth inhibitory and cytotoxic effects of
methotrexate than epidermal cell lines [91]. Recently, it has been shown that
the mechanism of action of methotrexate is highly dependent on the production of
reactive oxygen species [92]. In addition, the antimetabolite effect of
methotrexate is primarily S-phase specific. The inhibition of AICAR
transformylase is believed to lead to intracellular accumulation of AICAR, which
leads to release of adenosine into the extracellular space [90], [93]. This
process results, by way of interaction with specific cell surface adenosine
receptors, in potent inhibition of polymorphonuclear chemotaxis and adherence
and the inhibition of secretion of tumor necrosis factor α (TNF-α), IL-6, and
IL-8 by monocyte/macrophages. This process may thus be responsible for the
drug's anti-inflammatory effects. A recent study, however, downplays the role of
adenosine release in the mechanism of action of methotrexate [94]. Inhibitory
effects on IL-1, IL-2r, and the 5-lipoxygenase pathway also have been shown
[95].
ADVERSE EFFECTS
HEMATOLOGIC
Cytopenia occurs in 10% to 20% of patients on long-term methotrexate for
psoriasis manifesting as leukopenia, thrombocytopenia, or pancytopenia [96].
Potential risk factors include renal insufficiency, increased mean corpuscular
volume, older age, and concomitant use of trimethoprim-sulfamethoxazole or
nonsteroidal anti-inflammatory drugs (NSAIDs) [97]. The use of either folic acid
(1–5 mg/d) or folinic acid (leucovorin, 2.5–5 mg, 24 h after the methotrexate)
has been advocated to reduce hematotoxicity and is indicated in patients who
develop macrocytosis [98]. In a survey of British dermatologists, 75% use folate
supplementation in patients on methotrexate for psoriasis, one fourth of this
group use it for all patients on therapy, and three fourths use it only in
certain circumstances, namely in patients with macrocytosis [99]. The survey
also revealed the wide variety of opinion that exists regarding the indications
and dosing regimens for folate supplementation during methotrexate therapy and
pointed to the need for further studies to establish better guidelines.
GASTROINTESTINAL
Nausea and vomiting are the most frequent adverse effects and are dose-related.
Diarrhea and stomatitis may also occur. Folic acid administration, dose
splitting, and parenteral administration may control symptoms [100].
Hepatotoxicity is a major concern, with a 7% overall risk of severe
fibrosis/cirrhosis in patients with psoriasis but an apparent lower incidence in
patients with RA [101], [102]. Risk factors include age, total dose, obesity,
heavy alcohol intake, diabetes, and daily dosage [103]. Reports of severe
fibrosis continue to be published and fibrosis may occur despite serial normal
liver function tests [104]. Pretreatment complete blood count, liver function
tests, and hepatitis A, B, and C serology should be performed [105], [106].
Baseline and serial liver biopsies are also sometimes performed, but in the
absence of certain risk factors, such as history of liver disease or alcoholism
and elevated liver function tests, the usefulness of a baseline biopsy is not
recommended [105], [107], [108]. It has been suggested that serial biopsies may
not be required in the absence of abnormal liver function tests if the initial
biopsy sample is normal [109] or if the weekly dose of the drug is 15 mg or less
[108]. One recent study, which emphasized the use of methotrexate in developing
countries, claimed that for certain short-term methotrexate regimens in the
treatment of psoriasis, the need for liver biopsies can be safely minimized
[110]. Serial liver function tests continue to be recommended [105], however,
and several authors promote the serial use of serum type III procollagen
aminopeptide (PIIINP) as a test for fibrinogenesis [104], [111]. A recent
10-year retrospective study of 70 patients with psoriasis who were taking
methotrexate showed that the presence of repeatedly normal levels of PIIINP on
serial blood examinations correlated to a minimal risk of developing liver
toxicity [112]; however, this screening technique has not yet found common usage
or availability.
ONCOGENIC POTENTIAL
Methotrexate has been shown to be a significant independent risk factor
(relative risk, 2.1) for developing squamous cell carcinoma (SCC) in patients
with severe psoriasis [113], and this risk seems to be increased in patients
with psoriasis who have undergone treatment with psoralen plus ultraviolet A
(PUVA) [114]. Two patients with mixed-cellularity Hodgkin's disease have been
reported in association with methotrexate use for psoriasis [115]. There also
have been reports of an Epstein-Barr virus–associated lymphoproliferative
disorder in patients on low-dose methotrexate [116], [117], [118]. Contrary to
cyclophosphamide and azathioprine, methotrexate generally was not associated
with an overall increased risk of lymphoma in a large-scale retrospective study
of 16,263 patients with RA [119]. In addition, smaller retrospective studies
found no increased risk of lymphoproliferative disease in patients with
psoriasis on methotrexate [116].
REPRODUCTIVE TOXICITY
Methotrexate is a known abortifacient and teratogen. Teratologists have
recommended a delay of at least 12 weeks between drug discontinuation and
conception in women [120]. Conception should be avoided during therapy and 3
months afterward by couples in which the man is taking the drug [105].
OPPORTUNISTIC INFECTIONS
Opportunistic infections are reported in patients on low-dose methotrexate; in
patients with RA on methotrexate, these infections seem to be more common than
in those treated with azathioprine, cyclophosphamide, or cyclosporine [121].
Live vaccines should not be administered to patients on methotrexate.
MISCELLANEOUS
Idiosyncratic pneumonitis is rare in patients with psoriasis who are taking
methotrexate, but it can be life threatening. A chest radiograph is warranted if
pulmonary symptoms develop. Anaphylactoid reactions have been reported after
low-dose methotrexate and can occur during initial exposure [122]. Painful
erosion of psoriatic plaques is a cutaneous sign of methotrexate toxicity and
occurred in two patients who had an alteration in methotrexate dosage and
concomitant use of NSAIDs [123]. One report, however, described cutaneous
ulceration that was attributed to methotrexate therapy in a patient without
psoriasis [124]. Low-dose methotrexate may contribute to the development of
stress fractures of long bone. One study recently reported on three patients on
low-dose methotrexate with a triad of osseous pain, radiologic osteoporosis, and
distal tibial stress fractures [125]. Alopecia, hyperpigmentation, UV burn
recall, and toxic epidermal necrolysis also have been reported [89]. Central
nervous system (CNS) toxicities, including headaches, dizziness, fatigue, and
mood alterations, may occur.
CLINICAL USE IN DERMATOLOGY
The most widely accepted use for methotrexate in cutaneous disease is for the
treatment of severe or refractory psoriasis. It is also used for pityriasis
rubra pilaris, sarcoidosis, chronic urticaria, dermatomyositis, and
lymphoproliferative diseases, such as pityriasis lichenoides et varioliformis
acuta, lymphomatoid papulosis, and cutaneous T-cell lymphoma [89]. A recent
review of clinical experience with low-dose methotrexate to treat 113 patients
with severe psoriasis at a Dutch center revealed that prolonged improvement was
achieved in 81% of patients [96]. The drug has been found to be of value in
psoriatic polyarthritis as well [126]. In a case series of patients with adult
pityriasis rubra pilaris, the combined use of retinoids and methotrexate has
been advocated for patients whose disease is resistant to retinoids. Of 11
patients, 9 experienced significant clinical improvement at 16 weeks [127].
Others have warned against this combination over concern for the potential of
increased hepatotoxicity, however [128]. A review of the use of methotrexate in
45 patients with lymphomatoid papulosis and Ki-1 lymphoma revealed satisfactory
long-term control in 87% of patients with doses of 25 mg or less given at 1- to
4-week intervals for up to several years [129].
The use of methotrexate as corticosteroid-sparing agent in inflammatory
dermatoses continues to receive attention. Kasteler and Callen [130] reported on
the successful use of methotrexate (2.5–30 mg/wk) as a corticosteroid-sparing
agent in 13 patients with dermatomyositis and therapy-resistant cutaneous
features. Although older reports suggest methotrexate should be avoided in the
treatment of pemphigus vulgaris [131], it has been used at a dose of 5 to 10 mg
per week as an efficacious steroid-sparing agent in an elderly population with
bullous pemphigoid [132]. There is a single case report claiming a
steroid-sparing effect of methotrexate in the treatment of pyoderma gangrenosum
[133]. There also has been some benefit shown in the treatment of widespread
morphea [134]. Finally, a case report of multicentric reticulohistiocystosis
responding to low weekly dosages of methotrexate in addition to prednisone adds
to the two cases in the literature [135].
USAGE GUIDELINES
Patients with a history of significant liver or kidney disease should be
excluded. The reader is referred to the published guidelines for use in patients
with psoriasis [105] and, for comparison, to the guidelines for use in RA [101].
Pregnancy is an absolute contraindication, and active infection, excessive
alcohol consumption, patient unreliability, anemia, leukopenia, and
thrombocytopenia are all relative contraindications.
DRUG INTERACTIONS
Methotrexate toxicity has been precipitated by probenecid,
trimethoprim-sulfamethoxazole, omeprazole, and penicillins, possibly by
interference with tubular secretion, and possibly by interference with plasma
protein binding of the methotrexate. Nephrotoxic drugs, such as NSAIDs, can
reduce the renal clearance of methotrexate. Trimethoprim-sulfamethoxazole and
possibly sulfasalazine may increase bone marrow toxicity. Concurrent use of
nonabsorbed oral antibiotics, such as vancomycin, may decrease gastrointestinal
absorption of methotrexate [87].
DOSAGE AND MONITORING
Methotrexate is commonly given either as a single weekly dose or in three
divided doses over 36 hours. Usual dosages are from 5 to 10 mg/m2 per week
(7.5–30 mg/wk). The drug is available orally as a 2.5-mg pill or can be given as
a liquid (intended for injection). Monitoring guidelines are shown in Table 2.
CYCLOSPORINE
Cyclosporine is a lipophilic cyclic undecapeptide isolated from the fungus
Tolypocladium inflatum. It has been used for the treatment of severe psoriasis
since the early 1980s and has more recently found application in other
inflammatory dermatoses. Unlike the three previous immunomodulatory drugs, this
drug's main effect is on the intracellular signal transduction pathways of cells
that are primarily of the lymphoid lineage.
PHARMACOLOGY
Cyclosporine is insoluble in water. In a traditional formulation, given orally
as a soft gelatin capsule, the absorption is erratic, with a bioavailability of
approximately 30%, with high interpatient and intrapatient variability [136].
Peak blood levels occur after 2 to 4 hours, with an average serum half-life of
18 hours. Tissue concentrations are highest in adipose tissue and 80% of the
drug is bound to lipoproteins. There is poor CNS penetration. The drug is
metabolized by the cytochrome P450 3A system with bile excretion and
enterohepatic recirculation. Metabolites are inactive and only 6% of the drug is
excreted in the urine in the form of metabolites.
The microemulsion formulation of cyclosporine is now the standard formulation in
Canada and Europe. It is absorbed more rapidly, with a peak serum concentration
at 1 hour earlier than the original formulation, and has an increase in
bioavailability, resulting in higher maximum concentrations and lower
intrapatient and interpatient variability [137]. This effect is associated with
an increase in clinical efficacy [138] and in a comparable safety profile. A
decreased tolerability with a 1:1 dose conversion has been noted with an
increase in reported gastrointestinal adverse effects [137]. A 1:1 dose
conversion is still recommended when converting patients from the original
formulation to the microemulsion formulation, however [139]. Use of the new
formulation seems to result in an improved outcome at a lower dosage
[140].Cyclosporine acts primarily on T cells. The drug forms a complex with
cyclophilin intracellularly, which then binds to and inhibits calcineurin. This
process prevents the activation of nuclear factors involved in the transcription
of genes encoding several cytokines, including IL-2 and interferon γ (IFN-γ)
[141], [142]. This process then results in the inhibition of T-cell activation
and of T-cell–mediated potentiation of the immune response.
ADVERSE EFFECTS
NEPHROTOXICITY AND HYPERTENSION
Cyclosporine can cause structural and functional changes in the kidney.
Functional changes are caused by dose-related vasoconstriction in the renal
microcirculation. Structural changes include an obliterative microangiopathy
with tubular atrophy and interstitial fibrosis [143]. A recently proposed
mechanism of nephrotoxicity is that cyclosporine, through the inhibition of the
adaptive response to hypertonicity, interferes with the urinary concentration
mechanism [144]. Risk factors include a daily dose of more than 5 mg/kg,
persistent elevations of creatinine to more than 30% of baseline, and older age
[145]. Structural changes may also correlate with drug-induced hypertension
[146] and can even occur with low-dose cyclosporine [147], [148]. Hypertension
is seen in 10% to 15% of patients. This condition is reversible with drug
discontinuation and can be controlled with calcium channel blockers [149],
[150]. Studies in animal models have shown that such supplements as magnesium
[151], glycine [152], and L-arginine [153] may reduce the risk of
nephrotoxicity.
ONCOGENIC POTENTIAL
Cyclosporine is not mutagenic, but immunosuppression results in an increased
risk of skin cancers and possibly lymphomas. There have been reports of
lymphomas occurring while patients were taking cyclosporine [154], although some
authors believe that these cases of lymphoma were not directly related to the
cyclosporine, and that cyclosporine use in psoriasis does not necessarily convey
an increased risk of lymphoma [155]. The increased risk in skin cancers has been
estimated from 2.6-fold in patients with RA to 7.5-fold in patients with
psoriasis [139]. A recent prospective study of 1252 patients with severe
psoriasis on cyclosporine therapy showed a sixfold increase in cutaneous
malignancy attributable to cyclosporine [156]. This study also noted that the
increased risk for cutaneous malignancy was apparent after 2 years of therapy.
Previous PUVA therapy and previous immunosuppressive therapy may be predisposing
factors in the development of skin cancer in patients with psoriasis [156],
[157].
MISCELLANEOUS
Hyperlipidemia, hyperkalemia, hypomagnesemia, and hyperuricemia can occur but
are usually mild. Neurologic side effects can include hand tremors,
paresthesias, dysesthesias, and headache. Mucocutaneous side effects include
gingival hyperplasia, hypertrichosis, pilomatrix dysplasia, facial dysmorphism,
acne, eruptive angiomas, and folliculitis. Transient nausea, vomiting, diarrhea,
abdominal discomfort, and severe fatigue also have been noted.
CLINICAL USE IN DERMATOLOGY
Cyclosporine has become an accepted treatment for severe psoriasis and atopic
dermatitis. The efficacy of its use as maintenance therapy in severe psoriasis
has been shown [158], and the safety of cyclosporine in the treatment of
psoriasis for up to 2 years has been documented [159]. It is effective for
psoriatic arthritis as well, although symptomatic improvement can be slow [160].
Strategies to minimize risk and optimize treatment include intermittent
short-course therapy, rotational therapy, and short-term use for crisis
management [139], [161]. Cyclosporine is safe and effective for the short-term
treatment of severe atopic dermatitis [162]. In an open trial, 65 of 100
patients completed 48 weeks of therapy with significant improvement at an
average dose of 3 mg/kg per day [163]. A more recent study has shown that
cyclosporine microemulsion is effective for the short-term treatment of severe
atopic dermatitis using a bodyweight-independent dosing scheme [164]. The study
results also showed that a starting dose of 150 mg per day was preferable to a
starting dose of 300 mg per day, mainly because the former was observed to be
much less nephrotoxic. As with psoriasis, relapses are the rule, but a fraction
of patients have prolonged remissions [165]. The efficacy of cyclosporine at 3
mg/kg per day in recalcitrant hand dermatitis was similar to topical
betamethasone dipropionate in a randomized double-blind study [166].
The use of cyclosporine in other dermatoses is based mostly on case reports and
case series of successful outcomes. These have been summarized in review
articles on the subject [167], [168], [169]. Cyclosporine is considered by some
to be first-line treatment for pyoderma gangrenosum [168]. It is also excellent
treatment for extensive cutaneous lichen planus and may result in prolonged
remission even at very low dosages (1–5 mg/kg/d) [170]. Oral and genital lichen
planus may be somewhat less responsive [1]. Reports of the efficacy of
cyclosporine in the bullous disorders are mixed. In one report, long-lasting
remissions were obtained in six patients with pemphigus vulgaris refractory to
corticosteroids and other immunosuppressive agents [171]; however, a study
comparing prednisone alone with prednisone plus cyclosporine and prednisone plus
cyclophosphamide for the treatment of oral pemphigus found no significant
difference between the groups [172]. A more recent study showed that combination
treatment with corticosteroids and cyclosporine (at 5 mg/kg) offers no advantage
over treatment with corticosteroids alone in patients with pemphigus [173];
however, some maintain that there is value in using cyclosporine as maintenance
therapy for pemphigus [174]. Several case reports document the efficacy of
cyclosporine as a steroid-sparing agent in bullous and cicatricial pemphigoid,
and in epidermolysis bullosa acquisita [1]. Acute and juvenile dermatomyositis
respond to cyclosporine, and the drug should be considered in severe or
refractory disease [175]. Dramatic improvement also has been seen in the
mucocutaneous manifestations of Behçet's disease [168]. Cyclosporine in addition
to low-dose prednisone yielded cosmetically acceptable results in only two of
eight patients treated for alopecia areata in a recent report without induction
of durable remissions [176], and therefore does not seem warranted for this
indication. Recent reports have suggested impressive responses in the treatment
of severe cytophagic histiocytic panniculitis [177], toxic epidermal necrolysis
[178], hidradenitis suppurativa [179], and prurigo nodularis [180]. Further
study is warranted in these diseases. Also, there are reports of the successful
use of cyclosporine in the following conditions: actinic reticuloid,
Hailey-Hailey disease [181], granuloma annulare [182], [183], chronic actinic
dermatitis [184], recurrent Reiter's syndrome [185], eosinophilic cellulites
[186], and Sweet's syndrome [187].
USAGE GUIDELINES
Guidelines for the use of cyclosporine in psoriasis have been published [139],
[188], [189] and can be adopted for the use of cyclosporine in other dermatoses.
Absolute contraindications are severe concurrent infection, uncontrolled
hypertension, or renal insufficiency. Relative contraindications include the
following: current or past malignancy, immunodeficiency, high risk of
noncompliance, use of concomitant nephrotoxic drugs, gout, liver disease, and
pregnancy [189].
DRUG INTERACTIONS
Cyclosporine, in either oral formulation, interacts with many drugs [139]. Most
of the clinically relevant drug interactions with cyclosporine relate to
competitive inhibition or induction of the cytochrome P450 3A microsomal
enzymes. Anticonvulsant agents (phenytoin, phenobarbital, carbamazepine),
antibiotics (rifampin, trimethoprim-sulfamethoxazole, nafcillin), and
phenylbutazone can decrease cyclosporine levels; other antibiotics
(erythromycin, doxycycline), antifungal agents (ketoconazole, itraconazole,
fluconazole), calcium channel blockers (verapamil, diltiazem, nicardipine),
steroid hormones, and diuretics (furosemide, thiazides) can increase
cyclosporine levels. Several drugs may also synergistically increase
cyclosporine nephrotoxicity (diuretics, NSAIDs, aminoglycosides,
trimethoprim-sulfamethoxazole, amphotericin B, and melphalan).
DOSAGE AND MONITORING GUIDELINES
The microemulsion formulation is available in 25-mg, 50-mg, and 100-mg capsules.
The recommended starting dose [189] for patients with stable, generalized
psoriasis or for patients whose disease severity lies between moderate and
severe is 2.5 mg/kg per day in one or two divided doses. If improvement is not
noted within 2 to 4 weeks, the cyclosporine dose can then be increased in
increments of 0.5 to 1.0 mg/kg per day every 2 weeks to a maximum of 5 mg/kg per
day. For patients in whom a rapid improvement is critical (ie, those with
severe, inflammatory flares of psoriasis or the presence of other diseases that
have failed previous therapies), the starting dose should be 5 mg/kg per day.
(Note: the US Food and Drug Administration [FDA] has decreased the upper-limit
dose for cyclosporine treatment of psoriasis to 4 mg/kg/d.) In this scenario, as
soon as the patient has a significant response, the dose can be decreased by 0.5
to 1 mg/kg per day every week until the minimum effective maintenance dose is
attained. Monitoring guidelines are presented in Table 2. In either the high- or
low-dose scheme, dosage adjustments should be made in response to adverse
effects. If, for instance, the serum creatinine level increases by more than 30%
above baseline, and a repeat creatinine level 2 weeks later still shows the
increase, the dose of cyclosporine should be decreased by at least 1 mg/kg per
day, with the creatinine level to be rechecked in 1 month. If, at that point,
serum creatinine decreases to less than 30% above the patient's baseline,
treatment can be continued, and if not, then therapy should be discontinued and
not resumed until the creatinine levels return to within 10% of the patient's
baseline. Another adverse effect is an increase in blood pressure. When this
occurs, the physician can either attempt to control this through
antihypertensive therapy or by decreasing the cyclosporine dose by 25% to 50%
[189].
MYCOPHENOLATE MOFETIL
Mycophenolate mofetil (MMF) is a morpholinoethyl ester of mycophenolic acid
(MPA) [190], which, on biotransformation after absorption, converts to MPA, the
active metabolite. As an immune suppressant, MMF has been widely applied in
organ transplantation medicine since the early 1990s. More recently, it has
found other clinical applications in dermatology, rheumatology, and
gastroenterology.
PHARMACOLOGY
MMF, or 2-morpholinoethyl
(E)-6-(1,3-dihydro-4-hydroxy-6-methoxy-7-methyl-3-oxo-5-isobenzofuranyl)-4-methyl-4-hexenoate,
is the 2-morpholinoethyl ester of MPA. It is rapidly absorbed following oral
administration and then undergoes ester hydrolysis in the liver to form
mycophenolic acid, the active metabolite [191], [192]. Its oral bioavailability
is high at 94.1% in healthy volunteers [193]. The active metabolite MPA is a
noncompetitive inhibitor of the cellular inosine monophosphate dehydrogenase
required for de novo purine biosynthesis [190], [194], [195]. As a result, it
blocks RNA and DNA synthesis, which is critical for the immune response. Because
the lymphocytes rely predominantly on de novo purine biosynthesis rather than
the salvage pathway for purine synthesis (HGPRT), the T- and B-cell
proliferative response is inhibited [196], [197]. Therefore, MMF inhibits
antibody production and the formation of cytotoxic T lymphocytes [190], [195].
The selectivity for lymphocytes has another mechanism. MPA is 5 times more
active against the type 2 isoform of inosine monophosphatase dehydrogenase
(IMPDH), which is activated in activated lymphocytes, than against the type 1
IMPDH, which is active in most cell types [198]. Once ingested, MPA has a
half-life of 17.9 hours in healthy volunteers, and as part of its metabolism, it
undergoes glucuronidation to form phenolic glucuronide of MPA, which is not
pharmacologically active. Most of the metabolite (93%) is excreted in the urine,
with 6% excreted in the feces.
ADVERSE EFFECTS
The reported side effects of MMF are mainly related to gastrointestinal and
hematologic systems, with little effect on hepatic and renal function [199],
[200]. The side-effect profile makes MMF highly complementary to many other
immune suppressants used for dermatologic diseases, such as cyclosporine,
methotrexate, cyclophosphamide, and azathioprine, which have renal or hepatic
toxicities [1].
GASTROINTESTINAL
The most common side effects of MMF include nausea, vomiting, diarrhea, and
abdominal cramps, with an incidence of 12% to 36% [201]. In a recent study of
MMF in 54 patients with systemic lupus, 39% of patients had gastrointestinal
adverse events [202]. Recently, it has been suggested that MMF caused erosive
enterocolitis in renal transplant patients receiving the drug; however, a
direct, causal link has not been established [203]. If gastrointestinal symptoms
develop, the daily dose can often be spread over more than two daily doses. If
adverse effects persist for a longer period of time and seem more serious, MMF
may have to be withdrawn while a comprehensive invasive diagnostic process is
performed to rule out any opportunistic infections. Severe gastrointestinal
complications with MMF are rare, but when they do occur they may require
extensive diagnosis and treatment [204].
HEMATOLOGIC
For 11% to 34% of patients, the most serious hematologic side effect is
leukopenia. Severe leukopenia is only present in 0.5%–2.0% of patients. These
effects are generally reversible on drug discontinuation [201], [205]. In a
study of 57 patients who underwent liver transplants, approximately half
received MMF, and the rest received azathioprine [206]. Of the MMF-treated
patients, 21% had thrombocytopenia, whereas leukopenia was seen in 7% of
MMF-treated patients. Both of these adverse effects occurred less frequently
with MMF than with azathioprine.
ONCOGENIC POTENTIAL
In a clinical trial involving 1483 patients enrolled for the prevention of renal
transplant rejection, there was a slight increase of lymphoproliferative
malignancies in the MMF group after more than 1 year (incidence, 0.6%–1.0%) on
therapy compared with placebo or azathioprine (incidence, 0.3%) [201]. The
3-year safety data showed no unexpected changes in malignancy incidences
compared with the 1-year data [207]. In a recent retrospective examination of
106 renal transplant patients receiving azathioprine and 106 receiving MMF, four
patients using azathioprine were diagnosed with a malignancy (three
post-transplant lymphoproliferative disorders, one SCC) compared with two MMF
patients (prostate cancer, basal skin cell carcinoma) [208]. In a more recent
3-year safety study of 128 renal transplant patients, there were slightly more
malignancies (predominantly cutaneous) in patients given MMF (14.2%) versus
those patients on azathioprine (10.2%) [209]. In the previously described
studies involving transplant patients, however, the different patient groups in
these studies were all taking cyclophosphamide as well, so the above numbers are
relevant only in that they convey the risk of neoplasm from MMF relative to
azathioprine.
INFECTIONS
Because MMF is an immune suppressant, it has been associated with increased
infection rates in patients, primarily herpetic viral infections, with a
relative risk of 2 to 3 times normal rates [205]. The studies that showed an
increased risk of infection primarily reported on transplant patients. A recent
retrospective study of 23 transplant patients on 2 g of MMF per day of MMF
showed 10 patients developing 22 opportunistic or serious viral or bacterial
infections [210]. The link of MMF specifically to cytomegalovirus (CMV)
infection is more controversial. One study on transplant patients found that the
addition of MMF to the cyclosporine-prednisone combination did not result in an
increase in primary CMV infections, but CMV infections led more often to CMV
disease in patients treated with MMF than in those not receiving MMF [211]. In a
similar study, MMF did not increase the overall incidence of CMV infection in
renal transplant patients but increased the severity of CMV infection in terms
of the frequency of organ involvement and number of organs involved [212]. More
recently, however, a study of 48 patients showed that MMF puts transplant
patients at a significant increased risk (with a calculated relative risk of 19)
of a CMV infection [213].
CLINICAL USE IN DERMATOLOGY
The primary indication for MMF is for the prophylaxis of organ transplant
rejection, in combination with cyclosporine and corticosteroids [201]. Since the
report of the successful treatment of bullous pemphigoid and phemphigus vulgaris
with MMF in 1997 [214], [215], however, dermatologic applications of this new
immune suppressant have been expanded to many other bullous and inflammatory
conditions. Also since this initial report, other reports have confirmed the
usefulness of MMF for treating bullous pemphigoid [216], [217], [218], [219].
Similarly, other bullous disorders have been reported to be responsive to MMF,
including pemphigus vulgaris [214], [217], [220], [221], pemphigus foliaceous
[222], bullous and hypertrophic lichen planus [223], linear IgA bullous
dermatosis [224], cicatricial pemphigoid [225], and paraneoplastic pemphigus
[226], [227]. Epidermolysis bullosa acquisita failed to respond to a combination
treatment of MMF and prednisone in one report [227],but succeeded in another
[228]. A recent case series of 17 patents with pemphigus (12 with pemphigus
vulgaris, 4 with pemphigus foliaceous, and 1 with paraneoplastic pemphigus)
treated with MMF showed improvement in 12 patients [229]. In general, most of
the reports are on combinations of MMF and glucocorticoids. One exception is the
report of successful treatment with MMF monotherapy of a patient who had
pemphigus vulgaris [220]. The dosages in these reports ranged from 2 to 3 g per
day, and patients generally tolerated the treatment well. In addition, MPA was
used in the 1970s for the successful treatment of psoriasis [230].
Since MMF became available clinically in the early 1990s, Haufs et al [231]
reported that one patient responded to MMF at a dosage of 1 to 1.5 g twice daily
in 6 weeks, with a 50% reduction in Psoriasis Area and Severity Index (PASI)
score. Subsequently, Nousari et al [218] reported on another patient with plaque
psoriasis who responded to MMF plus topical steroid and calcipotriol. In a
series of five patients with plaque-type psoriasis taking 1 g of MMF twice daily
for 10 weeks, two patients with mild to moderate disease showed improvement,
whereas those with severe psoriasis did not respond [232]. In a study of 11
patients with severe, stable plaque-type psoriasis, it was shown that MMF at
dosages of 1 g twice daily for 3 weeks was an effective and seemingly safe
treatment option [233]. In a more recent study, 23 patients with moderate to
severe psoriasis were treated with MMF at a dosage of 2 to 3 g per day for 12
weeks in an open-label uncontrolled trial; approximately three fourths of the
patients were observed to have a significant reduction in symptom scores, with
only few adverse events [234]. MMF (2–3 g/d) has been used in combination with
cyclosporine for the treatment of recalcitrant pyoderma gangrenosum [235],
[236], [237], [238]. Grundmann–Kollmann et al [239] reported on two patients
with severe atopic dermatitis who dramatically responded to MMF monotherapy
after 2 to 3 weeks, with no side effects. The effectiveness of MMF for atopic
dermatitis was supported in two separate pilot studies, each involving 10
patients with moderate-to-severe or severe atopic dermatitis [240], [241]. In
another report, however, none of the five patients with atopic dermatitis
responded to MMF [242]. Pickenacker et al [243] reported on a patient with
severe dyshidrotic eczema who responded to MMF at a dosage of 3 g per day, but
in another report, a patient apparently developed dyshidrotic eczema while on
MMF therapy [244]. Clinical experiences have been reported for successful
treatment of systemic [245], [246], [247], [248], subacute cutaneous [249], and
discoid [250] lupus erythematosus, and other dermatologic diseases, including
vasculitis [251], [252], [253], [254], [255], sarcoidosis [256], ulcerated
necrobiosis lipoidica [257], and dermatomyositis [258]. Almost all of the
reports of the effectiveness of MMF in patients with dermatologic diseases are
case reports, case series, or small-scale, uncontrolled clinical trials.
USAGE GUIDELINES
MMF is contraindicated in patients with hypersensitivity to MMF, MPA, or any
component of the drug product [201], [207]. It is also contraindicated in
pregnancy. Caution should be exercised in patients with gastrointestinal
disorders because rare gastrointestinal bleeding has been reported. Patients
with severe renal function impairment (glomerular filtration rate of < 25
mL/min/1.73 m3) should not receive more than 2 g of MMF per day [201].
DRUG INTERACTIONS
No major drug interactions have been reported. Although there is some evidence
to suggest that glucocorticoids interfere with the bioavailability of MMF [259],
it is unclear as to the clinical significance of this phenomenon. The
coadministration of MMF with antacids, iron, and cholestyramine causes a
reduction of MMF absorption [201], [260]. One case report attempted to ascribe
the cause of a patient's neutropenia to an interaction between MMF and
valacyclovir [261]. Also, because of its similar effects on purine metabolism
and bone marrow suppression, MMF should not be used concomitantly with
azathioprine.
DOSAGE AND MONITORING
MMF is available as 250- and 500-mg tablets. Typical daily dosages are 2 to 3 g
per day in twice-daily dosing. Although MMF has been used in dermatologic
conditions, official approval for its use in the treatment of such diseases is
still not available. There are therefore no formal consensus guidelines for
monitoring MMF when used for dermatologic indications. The current authors'
recommendations have been adapted from previous guidelines [207] and are
presented in Table 2. Recent literature has suggested a possible benefit in
monitoring serum levels of MMF in transplant patients, but the relationship
between pharmacokinetic parameters and adverse events remains unclear [262].
THE USE OF SYSTEMIC IMMUNOBIOLOGIC AGENTS
Alefacept, efalizumab, etanercept, and infliximab are currently the
immunobiologic agents most commonly used in dermatology. At present, within the
dermatologic therapeutic armamentarium, these therapies are used mainly for the
treatment of psoriasis and psoriatic arthritis; however, their utility has been
shown in several other cutaneous disorders as well. A focus on the use of these
agents for the treatment of psoriasis, with anecdotal commentary on other
disease-specific clinical applications of these medications, and the
pharmacology, mechanism of action, dosages, and side-effect profiles of these
drugs will allow for a more thorough understanding of these agents for the
treatment of cutaneous disease.
A greater understanding of the role that T lymphocytes play in the pathogenesis
of cutaneous inflammatory disease has transformed physicians' understanding of
the mechanism of these disorders and resulted in a therapeutic revolution. It is
now understood that psoriasis, for example, is an autoimmune disease, with an
unknown antigenic stimulus. The epidermal hyperplasia of psoriasis is a CD8+ and
CD4+ T-lymphocyte–mediated reaction that occurs within focal skin regions [263].
Much of the current understanding of this process has come from experiments
using the severe combined immunodeficiency, or SCID, mouse model of psoriasis in
which clinically normal skin from a patient with psoriasis is transplanted onto
an immunodeficient mouse. When syngeneic T cells are activated in vitro and
injected into the transplanted skin, the skin develops a phenotypic expression
nearly identical to psoriatic skin [264], [265]. These experiments have
confirmed the paramount role of activated T cells in the induction of the
psoriasis plaque. Furthermore, studies using monoclonal antibodies have
elucidated the specific subsets of T cells involved in the formation and
maintenance of the psoriasis plaque and their roles at precise sites within
psoriatic skin. For example, CD8+ T cells (mainly cytotoxic or “killer
lymphocytes”) are found in high concentrations in the epidermis, whereas CD4+ T
cells predominate in the dermis [266], [267]. Psoriasis is considered to be a
Th1 immune disease because in psoriasis both CD4+ and CD8+ T cells produce
mainly type 1 cytokines. The understanding of the Th1 phenotype has facilitated
the creation of therapies that target specific steps within the Th1 pathway.
Many now recognize psoriasis to be one of the most prevalent T-cell–mediated
inflammatory diseases in humans, and it is considered to be the model Th1 immune
disorder by many researchers [263], [268].
The understanding of the T-cell–mediated pathogenesis of psoriasis has resulted
in an explosive research effort toward the development of biologic therapies
designed to target the immunologic process at specific points along the
inflammatory cascade. The biologics can be divided into three classes of agents:
(1) recombinant human cytokines or growth factors, (2) monoclonal antibodies,
and (3) fusion proteins. The classes and mechanism of action of the four most
commonly used immunobiologic agents used in dermatology are outlined in Table 3.
Alefacept, efalizumbab, etanercept, and infliximab, examples of the latter two
kinds of biologic agents, target highly specific steps in the aforementioned
T-cell–mediated inflammatory cascade. These therapies therefore are able to
control psoriatic disease with minimal impairment of immune function and
significantly fewer side effects than traditional immunosuppressive agents.
Table 3 . Summary of the common immunobiologic agents used in dermatology
Features Alefacept (Amevive) Efalizumab (Raptiva) Etanercept (Enbrel) Infliximab
(Remicade)
Type of agent Fusion protein Humanized monoclonal antibody Fusion protein
Chimeric monoclonal antibody
Mechanism of action Blocks T-cell activation. Selective reduction of memory
effector T cells Anti CD11a blocks T-cell activation and reduces trafficking of
T cells to inflamed skin TNF-α inhibitor TNF-α inhibitor
Route of administration IM SC SC IV
Approval status Psoriasis: FDA approved for chronic moderate to severe
plaque-type psoriasis, January 2003 Psoriasis: FDA approved for chronic moderate
to severe plaque-type psoriasis, October 2003 Psoriasis: FDA approved for
chronic moderate to severe plaque-type psoriasis, April 2004 Psoriasis: Phase 3
Abbreviation: IM, intramuscular.
ALEFACEPT
PHARMACOLOGY/MECHANISM OF ACTION
Alefacept is given as an intramuscular injection in a dose containing 15 mg of
alefacept per 0.5 mL of reconstituted solution. In patients who received
alefacept as a 7.5-mg IV administration, the mean volume of distribution was 94
mL/kg, the mean clearance was 0.25 mL/kg per hour, and the mean elimination
half-life was approximately 270 hours. Bioavailability was found to be 63%
post-intramuscular injection [269]. The pharmacokinetics of alefacept in
pediatric patients has not been studied. The effects of renal or hepatic
impairment on the pharmacokinetics of alefacept also have not been studied.
Alefacept is a fusion protein consisting of the first extracellular domain of
human leukocyte function antigen-3 (LFA-3) fused to the hinge Ch2 and Ch3
sequences of human IgG1 [268]. The drug is produced by recombinant DNA
technology in a Chinese hamster ovary mammalian cell expression system [269].
Alefacept acts as an immunosuppressive agent by inhibiting T-cell activation and
proliferation by binding to CD2 on T cells and blocking the LFA-3–CD2
interaction [270], [271], [272]. Alefacept also engages FcγRIII IgG receptors
(on NKCs and macrophages), resulting in the apoptosis of T cells that express
high levels of CD2 [264]. Because CD2 expression is higher on activated memory
(CD4+ CD45RO+ and CD8+ CD45R0+) than on naive (CD45RA+) T cells, alefacept
produces a selective reduction in memory T cells [268], [273], [274], [275],
[276].
Because the targeting of memory T cells is selective, naive (CD45RA+)
populations are minimally affected. This situation allows patients to maintain
the ability to mount appropriate immune responses to infectious agents [277].
This result was shown when alefacept-treated patients and controls were able to
mount comparable antibody responses to immunization with a novel
T-cell–dependent antigen (bacteriophage phiX174) and a recall antigen (tetanus
toxoid) [278].
The T-cell reductive effect observed in the circulation has been shown in the
skin. Patients with psoriasis who were treated with alefacept showed decreased
activation of T cells and memory T cells and a reduction in IFN-γ production in
lesional skin [279], [280]. Because the skin takes time to repopulate, by
selectively targeting the epidermal T cells, alefacept is able to induce longer
lasting remissions than many other biologic agents.
ADVERSE EFFECTS
ONCOGENIC POTENTIAL
The package insert for alefacept warns that the drug may increase the risk of
acquiring a malignancy. No statistically significant increased incidence of
malignancy has been observed in any studies to date, however. In a study that
looked at repeat courses of alefacept, five cases of malignancies (two systemic,
three cutaneous) were reported [281]. In each case, the patient had additional
potential risk factors for developing a malignancy, and the overall incidence of
cancers in the study was similar to what would be statistically expected in
untreated individuals of the same age. As is true of all of the biologic agents,
alefacept is a novel therapy, and the adverse effect profile may not be
established until after years of clinical experience. Patients with a history of
systemic malignancy should not be treated with alefacept.
INFECTIONS
One or two 12-week courses of intramuscular alefacept were shown in phase 3
studies to have safety and tolerability profiles similar to placebo [281]. No
increase in the incidence of opportunistic infections was noted [282]. No
association between infections and CD4+ T-cell counts was observed. In the
largest phase 3 trial, the only adverse event that occurred with an incidence of
5% or more in the alefacept group versus the placebo group in course 1 was
chills (10% versus 1%) [283]. The incidence of chills was temporally related to
dosing. Chills tended to occur soon after dosing, and the frequency of chills
was lower in the second course. Of note, chills were not a side effect in the
other large-scale phase 3 study [282] that examined the efficacy and
tolerability of intramuscular alefacept, and this side effect can therefore most
likely be attributed to the IV route of administration, which is no longer
available. In course 2 of the aforementioned phase 3 trial, accidental injury
and pharyngitis were the only adverse events that occurred with an incidence of
5% or more in the alefacept group versus the placebo group (20% versus 15%, and
16% versus 11%, respectively) [283].
Alefacept is an immunosuppressive agent and therefore should not be administered
to patients with clinically significant infections. To avoid clinically
significant immunosuppression, other immunosuppressive agents should be avoided
or used with caution in patients receiving alefacept. If a patient develops a
serious infection, alefacept should be discontinued and the patient should be
closely monitored. Therapy can be reinitiated on resolution of infection at the
physician's clinical discretion.
EFFECTS ON THE IMMUNE SYSTEM
The biologic agents, as with any exogenous protein molecule administered as
therapy, have the potential for immunogenicity. Antibody titers were low (<
1:40) in both of the largest phase 3 trials, and no immune hypersensitivity
reactions occurred [282], [283].
INJECTION SITE REACTIONS
In the largest phase 3 clinical trial that evaluated the intramuscular route of
administration, injection site pain and injection site inflammation were adverse
events that occurred at least 5% more frequently in alefacept-treated patients
than in those taking a placebo. Injection site reactions were typically
classified as mild, were usually single episodes, and did not result in any
patient discontinuing the study [282].
SUMMARY OF EFFICACY DATA FROM IMPORTANT CLINICAL TRIALS
Two important randomized, double-blind, placebo-controlled phase 3 trials were
conducted to evaluate the efficacy and tolerability of alefacept. In the first
trial, two 12-week courses of once-weekly 7.5-mg IV alefacept versus placebo
were given in a randomized double-blind study, and patients were followed up for
12 weeks after each treatment course [283]. Clinical outcomes were measured as
follows: a 75% or more reduction in the PASI, a 50% or more PASI reduction, or a
Physician Global Assessment (PGA) of “clear” or “almost clear” [283]. Because
many patients show continued clinical improvement after cessation of therapy as
shown by phase 2 data [276], the overall response rate was evaluated during both
the 12-week treatment period and during the 12-week follow-up period.
Of patients treated during course 1, 28% achieved a 75% or greater reduction in
PASI as compared with 8% of placebo-treated patients (P < 0.001); of those who
achieved a 75% or greater reduction in PASI, 23% achieved a PGA of clear/almost
clear as compared with 6% of placebo-treated patients (P < 0.001). Of patients
treated during course 1, 56% achieved a 50% or greater reduction in PASI as
compared with 24% of placebo-treated patients (P < 0.001).
A second course of alefacept provided additional benefit. Of patients who
received two 12-week courses of alefacept, 40% achieved a 75% or greater
reduction in PASI, 71% achieved a 50% or greater reduction in PASI, and 32%
achieved a PGA of clear/almost clear. Patients who were treated with two courses
of alefacept had a maximum mean reduction of 54% from baseline PASI at 6 weeks
post treatment [283].
The data showing that alefacept produced durable clinical improvements among
patients who responded were significant. Those patients, who received a single
course of alefacept and achieved 75% or greater overall PASI reduction, were
able to maintain a 50% or greater reduction in PASI for a median duration of
more than 7 months. Patients who achieved a PGA of clear or almost clear
maintained a 50% or greater reduction in PASI for a median duration of
approximately 8 months. Two courses of alefacept resulted in a greater duration
of response than a single course. This durable response makes alefacept the only
psoriasis therapy besides PUVA [284], the Goeckerman regimen [285], [286], and
possibly UVB [287] that can be considered a remittive treatment. Psoriasis
therapies can be either suppressive, where symptoms of disease recur shortly
after withdrawal of therapy, or remittive. Remittive therapies, such as
alefacept, provide long-lasting remission by mechanisms that reduce T cells in
the skin.
In the other pivotal phase 3 trial, the first randomized controlled trial of
weekly intramuscular doses of alefacept, 507 patients were randomized to one of
three treatment groups (10 mg, 15 mg, or placebo) of intramuscular alefacept
administered once weekly for 12 weeks with a 12-week follow-up [282]. To assess
overall response rate, the clinical outcome was measured throughout both the
treatment and follow-up periods as follows: a 75% or greater PASI reduction, a
50% or greater PASI reduction, or a PGA of clear/almost clear. Response rates
were compared among dosage groups.
The percentage of patients who achieved a 75% or greater reduction in PASI was
significantly higher (P < 0.001) in patients who received 15 mg of alefacept
(33%) or 10 mg of alefacept (28%) as compared with the placebo group (13%). Of
patients who achieved a 50% or greater PASI reduction throughout the study
period, 57% were in the 15-mg alefacept group (P < 0.001 versus placebo), 53%
were in the 10-mg alefacept group (P = 0.002 versus placebo), and 35% were in
the placebo group (P < 0.001). Overall response rates for a PGA of clear or
almost clear were 24%, 22%, and 8% of patients in the 15-mg, 10-mg, and placebo
groups, respectively (P < 0.001 for comparisons of both doses of alefacept
versus placebo). Again, the clinical response was durable and attributed to the
drug's mechanism of action. Of patients in the 15-mg group who had achieved at
least a 75% reduction from baseline 2 weeks after the last dose, 74% maintained
at least a 50% reduction in PASI during the 12-week follow-up period. Of
patients in the 15-mg group who achieved between a 50% and 75% PASI reduction,
79% maintained at least a 25% reduction in PASI from baseline during the 12-week
follow-up period [282].
CLINICAL USE IN DERMATOLOGY
Alefacept is indicated for the treatment of adult patients with moderate to
severe plaque-type psoriasis (∼1.5 million in the United States) [288] who are
candidates for systemic therapy or phototherapy. Patients who have
comorbidities, such as liver and renal disease, for whom systemic
immunosuppressive therapies are not options, represent an additional potential
patient population that can benefit from biologic therapies like alefacept. As
mentioned previously, alefacept is considered to be a remittive therapy that
allows patients to enjoy long periods of treatment-free, disease-free living. It
is therefore advantageous for the patient who needs a reprieve from chronic
therapy.
Unlike most biologic agents that require the patient to self-inject
subcutaneously, alefacept is a physician-administered intramuscular injection.
This mode of administration represents a clear advantage for patients who, for
either psychologic or physical reasons, are not able to self-inject.
USAGE GUIDELINES
Alefacept is contraindicated in patients with a known hypersensitivity to the
drug or any of its components [269]. Alefacept has not been formally studied in
women who are pregnant or nursing. It is not known whether alefacept is excreted
in human milk. The drug has been labeled pregnancy category B and should
therefore only be used during pregnancy when clinically necessary. Reproductive
toxicology studies were performed in cynomolgus monkeys with alefacept in doses
of up to 5 mg/kg per week (∼62 times the human dose based on body weight) and
revealed no evidence of impaired fertility or harm to the fetus. Weekly IV bolus
injections of alefacept administered to cynomolgus monkeys from organogenesis to
gestation did not cause abortifacient or teratogenic effects [269]. Caution
should be used when treating elderly individuals because of the higher incidence
of infections and malignancies in this population. Alefacept is not indicated
for the treatment of pediatric patients.
DRUG INTERACTIONS
No formal interaction studies have been performed [269].
DOSAGE AND MONITORING
Alefacept is approved by the FDA as a single 15-mg intramuscular dose given
weekly for 12 weeks. The IV formulation is no longer available. After a 12-week
rest period off the drug, patients can be treated with a second 12-week course.
The drug is supplied as a sterile, preservative-free, lyophilized powder to be
reconstituted with 0.6 mL of sterile water before injection. Monitoring
guidelines are presented in Table 4.
Table 4 . Monitoring guidelines for use of immunobiologic agents Drug Monitoring
tests Frequency Guidelines for dosage modification
Alefacept CBC and platelets, including total lymphocytes and CD4+ T-cell counts
(both must be within normal limits to begin therapy) Baseline Hold therapy if
CD4+ T-lymphocyte counts are below < 250 cells/μL
CD4+ T-cell counts Weekly Discontinue drug if CD4+ counts remain below 250
cells/μL for 1 mo
Efalizumab Platelet counts Baseline and once per month for the initial 3-mo
treatment period; periodic testing should be done throughout the treatment
period Discontinue if thrombocytopenia develops
Etanercept None required — —
Infliximab Tuberculin skin test Baseline —
EFALIZUMAB
PHARMACOLOGY/MECHANISM OF ACTION
Efalizumab is administered as a weekly subcutaneous (SC) injection. Patients (n
= 26) with moderate to severe plaque psoriasis who received a starting (SC) dose
of 0.7 mg/kg followed by 11 weekly SC doses of 1 mg/kg per week reached
steady-state serum concentrations at 4 weeks, with a mean trough concentration
of approximately 9 μg/mL. The mean steady-state clearance was 24 mL/kg per day
(range, 5–76 mL/kg/d; n = 25). After the last steady-state dose, the mean
elimination time of efalizumab was 25 days (range, 13–35 d; n = 17). Mean
efalizumab SC bioavailability was estimated to be 50%. Body weight was found to
be the most significant covariate affecting clearance of the drug. Patients who
received weekly SC doses of 1 mg/kg of efalizumab had similar exposure across
body weight quartiles [289].
Efalizumab is a humanized monoclonal antibody directed against CDlla, the α
subunit of LFA-1. This binding reversibly blocks the interaction between LFA-1
and intracellular adhesion molecule 1 (ICAM-1). LFA-1 expression is increased on
memory T cells, and ICAM-1 is expressed on vascular endothelial cells at sites
of inflammation and on keratinoctyes in various T-cell–mediated disorders [290],
[291], [292], [293]. Thus, the mechanism of efalizumab prevents the binding of T
cells to endothelial cells and blocks T-cell movement into the skin without
depleting memory effector T lymphocytes.
ADVERSE EFFECTS
ONCOGENIC POTENTIAL
Of 2762 patients who received efalizumab at various doses for a median duration
of 8 months [289], 31 patients were diagnosed with 37 malignancies.
Efalizumab-treated patients had a 1.8 per 100 patient-years incidence of
malignancies compared with 1.6 per 100 patient-years in placebo-treated
patients. Most malignancies were nonmelanoma skin cancers. The incidence of
noncutaneous solid tumors and malignant melanoma were within the range that
would be expected in the general, non–efalizumab-treated population. Efalizumab,
like alefacept, is a novel immunosuppressive agent without long-term safety and
efficacy data. Therefore, the role of efalizumab in the long-term development of
malignancies will require longer follow-up before any role in the development of
malignancy can be excluded.
INFECTIONS
In a pivotal phase 3 multicenter, randomized, placebo-controlled, double-blind
study [294] that evaluated the efficacy and safety of efalizumab in dosages of 1
or 2 mg/kg per week, subjects (N = 597) were treated with efalizumab for a
maximum of 24 weeks with no increased rate of infection observed among
efalizumab-treated patients [294]. The package insert for efalizumab states that
the overall incidence of hospitalization for infections was 1.6 per 100
patient-years for efalizumab-treated patients compared with 1.2 per 100
patient-years for placebo-treated patients [289].
EFFECTS ON THE IMMUNE SYSTEM
In the previously described large-scale phase 3 study of 597 patients, 5% of the
subjects who were treated with efalizumab developed antiefalizumab antibodies
[294]. These effects were not believed to be clinically significant.
THROMBOCYTOPENIA
During clinical trials, thrombocytopenia, as defined as platelet counts at or
below 52,000 cells per μL, was observed in eight (0.3%) of efalizumab-treated
patients and no placebo-treated patients [289]. Five of the eight patients were
treated with systemic steroids. The thrombocytopenia resolved in seven of the
eight patients (the eighth patient was lost to follow-up).
MISCELLANEOUS
First-dose reactions are sometimes associated with the administration of
monoclonal antibodies. An analysis of the pooled adverse events from the
efficacy and safety data for 1095 patients who were treated with efalizumab
showed a higher incidence of acute adverse events (defined as headache, chills,
fever, nausea, vomiting, or myalgia that occurred on the day of injection or 2
days post injection) than placebo controls (43% versus 27%) [295]. These events
were generally mild to moderate in severity and tended to occur after the
initial injection, and decreased with each subsequent injection. By the third
dose, the incidence of acute adverse events was similar to the placebo-treated
group.
SUMMARY OF EFFICACY DATA FROM IMPORTANT CLINICAL TRIALS
Data on efalizumab pooled from two phase 3 clinical trials from 1095 patients
with moderate to severe psoriasis showed an improvement of 75% or greater in
PASI relative to baseline on day 84 in 29.2% of patients who received efalizumab
in dosages of 1.0 mg/kg per week and in 27.6% of patients who received dosages
of 2.0 mg/kg per week compared with 3.4% of placebo-treated patients [296]. Of
patients who achieved an improvement of 50% or more in PASI, 55.6% had been
treated with efalizumab in dosages of 1.0 mg/kg per week and 54.5% had been
treated with efalizumab in dosages of 2.0 mg/kg per week compared with 15.1% of
patients who were treated with placebo. An extended treatment trial (ACD2059g)
was conducted in responders (≥75% PASI improvement from baseline) and partial
responders (≥50% PASI improvement from baseline) to examine the effects of
continued treatment with the same weekly dosing schedule versus a potentially
more convenient every-other-week dosing schedule versus discontinuation of
therapy to assess time to relapse. Results showed that every-other-week dosing
was sufficient to maintain an improvement of 75% or more in PASI in responders,
but weekly dosing resulted in a better clinical response rate among partial
responders. After cessation of therapy, psoriasis relapsed at a loss of 50% of
the original PASI improvement, with a median time to relapse of 60 to 80 days
[295]. Of note, the patients who “rebounded” were discontinued from efalizumab
therapy abruptly, without taper or transition to other therapeutic modalities.
Currently, there are several ongoing phase 3 trials designed to establish more
optimal strategies for cessation of efalizumab therapy, to determine optimal
dosing schedules, and to further support efalizumab as a potential long-term
therapy.
CLINICAL USE IN DERMATOLOGY
Efalizumab was FDA-approved in October 2003 for the treatment of chronic
moderate to severe plaque psoriasis. Efalizumab can be compared favorably with
the current systemic antipsoriasis agents in that it is associated with rapid
onset and continued clinical improvement in patients with moderate to severe
plaque-type psoriasis. It is also similar in that on cessation of therapy, some
patients in clinical trials have shown relapse or exacerbation of disease. Of
2589 clinical trial patients treated with efalizumab, 19 (0.7%) experienced
worsening of their psoriasis (n = 5) or worsening past baseline after
discontinuation of efalizumab (n = 14) [289]. Some of these patients developed
pustular or erythrodermic psoriasis. In comparison to systemic immunosuppressive
agents, however, efalizumab's mechanism of action is highly specific, and
clinical trial data have shown its potential role as a safe and effective
long-term therapy without the dose-limiting side effects of the systemic agents.
To date, although the long-term safety and efficacy data supporting efalizumab
as a chronic therapy have not been established, the favorable side-effect
profile and the convenience of a weekly self-administered SC injection make the
long-term administration of efalizumab feasible. Trials are currently being
conducted to evaluate efalizumab as a potential therapeutic agent for chronic
psoriasis.
USAGE GUIDELINES
Efalizumab is contraindicated in patients with a known hypersensitivity to the
drug or any of its components. No reproductive toxicity studies have been
conducted in pregnant women. Efalizumab has been labeled pregnancy category C.
It is not known whether efalizumab is excreted in human milk. The drug should
not be used in pregnancy unless the clinical benefits clearly outweigh the
risks.
In a developmental toxicity study that used antimouse CD11a antibody at up to 30
times the recommended clinical dose of efalizumab, there was no evidence of
maternal toxicity, embryotoxicity, or teratogenicity observed during
organogenesis [289].
Caution should be used when treating elderly individuals because of the higher
incidence of infections and malignancies in this population. Efalizumab has not
been studied in pediatric patients.
DRUG INTERACTIONS
Drug interaction studies have not been performed. Efalizumab should not be used
concomitantly with other immunosuppressive agents [289].
DOSAGE AND MONITORING
Efalizumab is administered as a single 0.7-mg/kg, SC conditioning dose followed
by a weekly SC dosage of 1 mg/kg. The maximum dosage should not exceed 200 mg
weekly [289]. The drug is supplied as a lyophilized, sterile powder to be
reconstituted with 1.3 mL of sterile water before injection. Monitoring
guidelines are presented in Table 2.
ETANERCEPT
PHARMACOLOGY/MECHANISM OF ACTION
Etanercept is a dimeric fusion protein that competitively inhibits the action of
TNF-α, rendering it biologically inactive. Etanercept is created with two
identical p75 TNF-α receptor peptides fused to the Fc region of IgG1 [297]. It
is this dimeric property of the drug that enables it to bind to two receptors on
TNF-α with greater affinity than natural monomeric receptors [298].
TNF-α is produced by type 1 helper T cells within psoriatic plaques. TNF-α has
an integral role in the amplification and maintenance of the inflammatory
cascade within psoriatic plaques by stimulating the production of chemokines and
the expression of adhesion molecules by keratinocytes and vascular endothelial
cells [299]. The importance of TNF-α in the inflammatory process has been
demonstrated by showing that levels of TNF-α in the serum [300] and blister
fluids [301] of involved psoriatic skin are higher than in those of controlled
nonpsoriatic skin. In addition, TNF-α levels have been significantly correlated
with PASI scores, and decreased levels are associated with clinical improvement
after treatment [300].
Pharmacokinetic studies of 25 patients with RA showed that administration of 25
mg of etanercept by a single SC injection resulted in a mean (± standard
deviation) half-life of 102 (±30) hours with a clearance of 160 (±80) mL per
hour [302]. Pharmacokinetic studies have not been conducted to study the effects
of renal or hepatic impairment in patients taking etanercept.
ADVERSE EFFECTS
ONCOGENIC POTENTIAL
The incidence of malignancies in the etanercept-treated population is best
evaluated using data from RA studies because these studies represent the longest
experience available and published phase 3 data evaluating etanercept in
psoriasis clinical trials are currently limited. Moreland et al [303] examined
the safety of etanercept therapy in a population of 1272 patients with RA 5
years after they had been treated and found the incidence of malignancies to be
35 in the treatment group. This number was what would be expected according to
the National Cancer Institute, Surveillance, Epidemiology, and End Results
database.
INFECTIONS
Similar to data on oncogenic potential, data concerning the incidence of
infections are also best gleaned from RA studies that have longer clinical
experience with the drug. Safety data from 5 years of clinical experience with
etanercept therapy in a population of 1272 patients with RA showed the frequency
of infections requiring hospitalization or IV antibiotics to be 0.04 per
patient-year in the total population [303]. This rate was the same as the rate
in the control group.
More recent short-term data from placebo-controlled trials investigating
etanercept for the treatment of psoriasis also have examined rates of infection.
In a double-blind, placebo-controlled 24-week study that examined the safety and
efficacy of etanercept as monotherapy in 672 patients with moderate to severe
psoriasis, rates of infection were similar across all treatment groups [304].
The study reported that 11% of placebo-treated patients, 10% of low-dose (25-mg
1×/wk) etanercept-treated patients, 9% of medium-dose (25-mg 2×/wk)
etanercept-treated patients, and 5% of high-dose (50-mg 2×/wk)
etanercept-treated patients developed an upper respiratory tract infection
during the first 12 weeks of therapy. There were no cases of opportunistic
infections or tuberculosis (TB) reported during the study. Similarly, in a
randomized, double-blind, placebo-controlled 12-week study [305] that looked at
the efficacy and safety of etanercept in dosages of 25 mg twice weekly, versus
placebo in 60 patients with psoriasis and psoriatic arthritis, the safety
profile of etanercept-treated patients was similar to the profiles previously
reported in the RA population [306], [307]. Upper respiratory tract infections
were one of the most common adverse events, but no adverse event occurred in a
significantly greater proportion in the treatment group relative to the placebo
group.
Postmarketing reports have reported serious infections and sepsis, including
fatalities, associated with the use of etanercept. Many of these serious
infections occurred in patients using concomitant immunosuppressive therapy
[302]. Usage guidelines concerning infections are discussed in the “Usage
guidelines” section.
The reactivation of latent TB has been observed in patients treated with
etanercept and infliximab, the other biologic agent that targets TNF. TB also
has been reported in much lower numbers in etanercept-treated patients. Thirteen
patients (eight in the United States and five in Europe) of approximately
102,000 patients with RA developed TB [299]. The etanercept-related cases
occurred sporadically and later during the course of therapy than the infliximab
cases. Currently, TB screening is required by the FDA before starting infliximab
but not before the initiation of therapy with etanercept. Some experts believe
that testing should be done before the initiation of etanercept as well.
EFFECTS ON THE IMMUNE SYSTEM
Data from clinical trials showed that 11% of etanercept-treated patients
developed a new positive antinuclear antibody (ANA) titer of 1:40 or greater
compared with 5% of placebo-treated patients [299]. Anti-dsDNA antibodies
occurred in 15% of etanercept-treated patients compared with 4% of
placebo-treated patients [299]. In addition, cases of drug-induced systemic
lupus and subacute cutaneous lupus have been reported [308], [309]. These
conditions resolved after therapy with etanercept was discontinued, and in one
case after corticosteroid treatment was initiated without withdrawal of
etanercept therapy [308]. Full cases of systemic lupus erythematosus associated
with etanercept therapy are rare [310].
DEMYELINATING DISEASE
In a review of the Adverse Events Reporting System of the FDA, Mohan et al [311]
identified 19 patients who developed neurologic symptoms, 17 following
etanercept therapy and 2 following infliximab therapy. The neurologic symptoms
were in most cases associated with demyelinating lesions of the CNS and were all
temporally related to anti–TNF-α therapy. Symptoms improved in all patients
after discontinuation of anti–TNF- α therapy. One patient's symptoms returned on
rechallenge with etanercept. It is important to note that 4 of the 19 patients
had a prior history of MS or MS-like symptoms. The authors suggested avoiding
anti-TNF-α therapy in patients with MS or in patients with a family history of
MS. An outline of the clinical approach to patients receiving anti–TNF-α therapy
who develop new neurologic signs suggestive of demyelination is also provided in
this study [311].
INJECTION SITE REACTIONS
In controlled clinical trials, injection site reactions were the only adverse
event that occurred significantly more often in etanercept-treated patients than
in placebo-treated patients. Approximately between 42% and 49% of
etanercept-treated patients in controlled clinical trials experienced injection
site reactions [306], [307]. In a study that retrospectively reviewed medical
records of patients who had received etanercept injections and who were still
being actively treated with etanercept, 21 (20%) of 103 patients reported
injection site reactions [312]. The reactions occurred within the first months
of therapy, within 1 to 2 days of injecting, and resolved within a few days. The
lower incidence of injection site reactions found in this study compared with
the 42% to 49% incidence in controlled clinical trials was attributed to the
retrospective nature of the study. The authors of this study performed a
histologic and immunophenotypic analysis of skin biopsy specimens and concluded
that the reactions may be an example of a T-lymphocyte–mediated delayed-type
hypersensitivity reaction. Injection site reactions can be treated with cool
compresses and 1% hydrocortisone ointment. Rotating sites of injection is also
recommended.
SUMMARY OF EFFICACY DATA FROM IMPORTANT CLINICAL TRIALS
Several clinical trials have investigated etanercept as monotherapy for the
treatment of psoriasis. A 24-week phase 2, multicenter, double-blind,
placebo-controlled trial of 112 patients with chronic moderate to severe
plaque-type psoriasis who were treated with etanercept in SC dosages of 25 mg
twice weekly found that, at week 12, 30% of etanercept-treated patients achieved
at least a 75% improvement in PASI, compared with 2% of placebo-treated patients
(P < 0.001) [313]. By week 24, 56% of etanercept-treated patients and 5% of
placebo-treated patients achieved an improvement of 75% or greater in PASI (P <
0.001).
A phase 3, placebo-controlled, double-blind, parallel-group study assessed the
safety and efficacy of a 24-week course of etanercept in SC dosages of 25 mg
once weekly (low dose), 25 mg twice weekly (medium dose), 50 mg twice weekly
(high dose), or placebo in 672 patients with moderate to severe plaque-type
psoriasis [304]. At week 12, 14%, 34%, 49%, and 4% of the patients in the low,
medium, high, and placebo groups, respectively, achieved a PASI improvement of
at least 75% (P < 0.001 for all three comparisons with the placebo group). As
early as week 2 of the study, the mean percentages of PASI improvements were
statistically significant for all three etanercept-treatment groups compared
with baseline. At week 12, mean levels of improvement were 40.9%, 52.6%, 64.2%,
and 14% in the low-dose, medium-dose, high-dose, and placebo groups,
respectively. Results at week 24 showed an improvement of 75% or greater in 25%,
44%, and 59% of the patients in the low-, medium-, and high-dose groups,
respectively. There was no placebo comparison group at this point in the study.
CLINICAL USE IN DERMATOLOGY
Etanercept has more than 130,000 patient-years of treatment experience (mostly
in the RA population) in both controlled clinical trials and postmarketing
experience [299]. Etanercept was FDA-approved in April 2004 for the treatment of
adult patients with chronic moderate to severe plaque-type psoriasis. Etanercept
was previously granted FDA approval for the treatment of psoriatic arthritis. It
is also indicated for reducing signs and symptoms of moderate to severe active
polyarticular-course juvenile RA. Because between 5% and 42% of patients with
psoriasis also have concomitant psoriatic arthritis [314], etanercept represents
an ideal biologic agent in physicians' current armamentarium for the treatment
of both of these disorders. In addition, similar to efalizumab, etanercept can
be administered with methotrexate, a benefit that has numerous favorable
clinical implications when treating patients with psoriasis or psoriatic
arthritis. Like other SC self-administered biologics, etanercept can be
administered by the patient at home, a feature that offers tremendous patient
independence. Unlike many biologics, etanercept requires no laboratory
monitoring during treatment. Data extrapolated from RA studies suggest that
etanercept therapy may (similar to efalizumab) have a role as a therapeutic
agent in chronic conditions administered on a long-term continuous basis.
Etanercept has been reported to be clinically effective in several other
cutaneous disorders, including the following: scleroderma [315]; cicatricial
pemphigoid [316]; in a patient with common variable immunodeficiency who
presented with scarring alopecia, arthritis, diarrhea, and recurrent infections
[317]; and for recurrent apthous stomatitis [318].
USAGE GUIDELINES
Etanercept is contraindicated in patients with a known hypersensitivity to the
drug or any of its components and should not be administered to patients with
sepsis. Treatment with etanercept should not be initiated in patients with
active or chronic localized infections. The manufacturer of etanercept cautions
that the drug should not be administered to patients who develop serious
infections or sepsis and caution should be taken when considering treating
patients with conditions that may predispose to infections, such as poorly
controlled or advanced diabetes. Caution should also be exercised when
considering using etanercept in patients with pre-existing or recent-onset CNS
demyelinating disorders [302].
Developmental toxicity studies of etanercept performed in rats and rabbits in
doses between 60- to 100-fold higher than the human dose have not resulted in
harm to the fetus [303]. Whether etanercept can impair fertility in humans is
unknown. The drug should not be used during pregnancy unless the clinical
benefits clearly outweigh the risks. Etanercept has been labeled as pregnancy
category B. It is not known whether etanercept is excreted in human milk.
Caution should be used when treating elderly individuals because of the higher
incidence of infections and malignancies in this population. Etanercept is
indicated for the treatment of polyarticular-course juvenile RA, but it has not
been studied in children younger than 4 years [302].
DRUG INTERACTIONS
No formal interaction studies have been performed. In a study of patients with
RA who were treated with etanercept and anakinra therapy for up to 24 weeks,
however, the rate of serious infections was 7% in the dual-treatment group
compared with 0% in those with etanercept alone [302].
DOSAGE AND MONITORING
Etanercept is indicated for adult patients with moderate to severe plaque-type
psoriasis as a step-down dosing regimen of 50 mg administered twice weekly for 3
months, followed by a 50-mg weekly maintenance dose. Etanercept is also
indicated for adults with psoriatic arthritis or RA as a 25-mg twice-weekly SC
injection administered 72 to 96 hours apart [302]. The drug is supplied as a
sterile, preservative-free, lyophilized powder to be reconstituted with 1 mL of
bacteriostatic water before injection. Monitoring guidelines are presented in
Table 4.
INFLIXIMAB
PHARMACOLOGY/MECHANISM OF ACTION
Pharmacokinetic study data from 17 patients with moderate to severe psoriasis
treated with either 5 mg/kg or 10 mg/kg doses of infliximab showed maximum
infliximab concentrations at day 14 [319]. For the 5- and 10-mg/kg–dose groups,
the concentrations were 158.14 mg/mL and 298.89 mg/mL,
respectively—concentrations that were directly proportional to the administered
doses. The median elimination half-life for infliximab was 7.62 (interquartile
range, 6.62–10.15) and 9.97(interquartile range, 6.17–10.1432) days for the 5
mg/kg and 10 mg/kg doses, respectively. The half-life and maximum infliximab
concentrations in the 10-mg/kg psoriasis group were similar to those seen in
patients with Crohn's disease who were treated with 5 mg/kg of infliximab.
Infliximab is a monoclonal antibody that targets TNF-α. It is composed of a
human constant region and a murine variable region, factors that make infliximab
chimeric in nature. By binding to both soluble and transmembrane forms of TNF-α,
infliximab can trigger complement-mediated lysis of cells expressing TNF- α
[320]. As previously described, TNF-α is an important inflammatory cytokine to
target because it is central to the pathogenesis of psoriasis. TNF-α fuels the
synthesis of many key cytokines involved in the inflammatory cascade, such as
IL-1, IL-6, and IL-8, and stimulates Langerhans' cell maturation and migration
from the skin to lymph nodes [321]. TNF-α activates nuclear factor κB, a
transcription factor that is central to the inflammatory process leading to
psoriasis [322]. TNF-α also has been shown to increase keratinocyte
proliferation in vitro [319]. In addition, both psoriatic lesional skin and the
plasma of patients with psoriasis have shown increased levels of TNF-α [323].
ADVERSE EFFECTS
ONCOGENIC POTENTIAL
The long-term risks of development of infliximab-associated malignancies remain
unknown. Nahar et al's [324] review of the use of infliximab for the treatment
of Crohn's disease and RA reported that 18 of 1372 infliximab-treated patients
developed 19 new or recurrent malignancies over 1430 patient-years of follow-up
[324]. The authors concluded that it could not be determined whether these
malignancies were induced by infliximab treatment. There are several factors
that complicate the ability to determine whether the reported occurrences of
lymphoma and other malignancies are related to infliximab treatment. Although
controversial, rates of lymphoproliferative disease have been shown to be higher
among RA, Crohn's disease, and psoriasis populations [325], [326], [327], [328],
[329], [330], [331], [332], [333]. In addition, lymphoproliferative disorders
tend to occur at an increased rate in patients who are immunosuppressed [332],
[333]. Many patients who have chronic inflammatory diseases, such as RA, Crohn's
disease, or psoriasis, have long exposure histories to immunosuppressive agents,
complicating the ability to analyze the potential causal relationship even
further. Last, because Epstein-Barr virus is a well-known cause of lymphoma in
organ transplant patients [334], [335], the virus may also have an integral role
in the development of malignancies in patient populations where
immunosuppressive agents are commonly used.
INFECTIONS
Infections, and especially reactivation of TB, have been a major issue of
concern associated with the use of infliximab. Keane et al [336] performed an
analysis of all reports as of May 29, 2001, of TB cases that occurred after
infliximab therapy through the FDA's MedWatch system. Details of this system are
available at http://www.fda.gov/cder/aers/. At the time of the analysis,
approximately 147,000 patients worldwide had received infliximab. The analysis
showed 70 reported cases of TB that occurred after treatment with infliximab for
a median of 12 weeks. TB developed in 48 of the patients after three or fewer
infusions of infliximab. Of the 70 cases, 40 were extrapulmonary disease. Most
cases (64) occurred in countries with a low incidence of TB. The reported
frequency of TB associated with infliximab therapy was much higher than the
reported frequency of other opportunistic infections. Several cases of
histoplasmosis also have been reported, however. Although the authors were not
able to completely assess the TB status of the patients pretreated with
infliximab, they concluded that the TB was likely reactivation TB.
As mentioned previously, histoplasmosis is another potentially life-threatening
infection that has been reported to the FDA as a possible complication
associated with infliximab therapy. In a review of the FDA's passive
surveillance database for monitoring postlicensure adverse events, Lee et al
[337] identified all cases of histoplasmosis received through July 2001 in
patients who had been treated with either infliximab or etanercept, the two
licensed anti–TNF-α biologic agents. Ten cases of Histoplasma capsulatum (HC)
infection were reported. Nine of these cases were associated with infliximab
therapy, and one case was associated with etanercept therapy. Clinical
manifestations of histoplasmosis, including fever, malaise, cough, dyspnea, and
interstitial pneumonitis, occurred within 1 week to 6 months of the first dose.
It is important to note that all of the patients had been treated with
concomitant immunosuppressive agents during infliximab therapy, and all of the
patients resided in HC-endemic areas. A detailed discussion of the clinical
implications of the association of infliximab with reactivation TB and
histoplasmosis will be addressed in the “Usage guidelines” section.
In a review of the published literature and presentations at scientific meetings
from a search of Medline and pre-Medline English-language articles published
from 1966 to June 2002 of infliximab for the treatment of RA and Crohn's
disease, Nahar et al [324] found that treated infections were reported in 36% of
patients receiving infliximab compared with 26% of placebo-treated patients.
Respiratory and urinary tract infections were the most frequently reported type
of infection. In addition, several serious infections were reported, including
pneumonia, abscess, cellulites, skin ulceration, pyelonephritis, sepsis, two
cases of TB, and one case of disseminated coccidioidomycosis. The authors
pointed out that most of the patients were on concomitant immunosuppressive
therapies in two of the large clinical trials in the review, A Crohn's Disease
Clinical Trial Evaluating Infliximab in a New Long-Term Treatment Regimen
(ACCENT) [338] and Anti-TNF Trial in Rheumatoid Arthritis with Concomitant
Therapy (ATTRACT) [339].
Chaudhari et al [340] assessed the clinical benefit and safety of infliximab for
the treatment of moderate to severe plaque psoriasis in a double-blind
randomized trial of 33 patients. In contrast to the aforementioned data from the
literature, the investigators did not observe any serious adverse events during
the 10-week study period where patients were randomized to receive 5 mg/kg, 10
mg/kg, or placebo in a 1:1:1 ratio. Patients enrolled in the study did not use
any other treatments for their psoriasis during the study and had stopped taking
systemic therapy (including cyclosporine, methotrexate, acitretin, and UVB and
PUVA) at least 4 weeks before the first dose of study medication. Therefore,
unlike many of the patients in previous clinical trials who were receiving
concomitant immunosuppressive agents, the patients in this trial were not, a
factor that may explain the lack of serious adverse event occurrences. In
addition, all patients had a clear chest radiograph within 1 month of receiving
the first dose of the study medication.
EFFECTS ON THE IMMUNE SYSTEM
In the review of the literature of patients with RA and Crohn's disease treated
with infliximab, Nahar et al [324] found that approximately 10% of all patients
in clinical trials developed antibodies to infliximab. The development of
antibodies to the drug was associated with an increased likelihood of developing
infusion reactions. Approximately 44% of patients with Crohn's disease treated
with infliximab developed ANAs between screening and the last clinical
evaluation [324].
In the ATTRACT study [339], a multicenter, randomized, double-blind,
placebo-controlled phase 3 trial, 428 patients with active RA were treated with
continuous methotrexate and randomized to one of four infliximab treatment
regimens. Of 342 exposed patients, 16% (54) developed anti-dsDNA antibodies
using the Farr assay of 10 units/mL compared with 0% of placebo-treated
patients. Of the 54 patients, 15 (4%) had titers greater than 25 units/mL.
INFUSION REACTIONS
Infusion reactions, defined as any adverse event occurring during an infusion or
within 1 to 2 hours after an infusion, occurred in approximately 20% of
infliximab-treated patients compared with 10% of placebo-treated patients in
clinical trials [341]. Less than 1% were considered serious reactions
characterized by clinical symptoms of anaphylaxis, convulsions, erythematous
rash, or hypotension. Of the patients who experienced infusion reactions,
approximately 3% discontinued infliximab therapy because of the reactions.
Infusion reactions can be avoided by slowing down the infusion, premedicating
with antihistamines, and, in some cases, administering systemic corticosteroids
[310]. It is therefore recommended that medications such as antihistamines,
corticosteroids, and epinephrine be available at the infusion site.
There have been reports of patients experiencing delayed infusion reactions in
instances where infliximab was restarted [324]. In a study in which 37 of 41
patients with Crohn's disease were retreated after a 2- to 4-year lapse between
treatments, 10 of the patients experienced adverse events that occurred 3 to 12
days following the infusion. Clinical signs and symptoms included myalgia or
arthralgia, with fever or rash, pruritus, edema of the face, hand, or lip,
dysphagia, urticaria, sore throat, and headache. None of the patients had
experienced an infusion-related adverse event previously. Approximately 39% (9
of 23) of the patients had received a liquid formulation, which is no longer
available, and 7% (1 of 14) had received the lyophilized formulation. The makers
of the drug do not know if occurrences of these reactions were caused by
differences in formulation [341].
CONGESTIVE HEART FAILURE
Infliximab has been associated with adverse outcomes in patients with congestive
heart failure. In a randomized study of 150 patients with stable New York Heart
Association class 3 or 4 heart failure and left ventricular ejection fraction of
35% or less, patients were randomized to receive either placebo (n = 49),
infliximab doses of 5 mg/kg (n = 50), or infliximab doses of 10 mg/kg (n = 51)
[342]. The results suggested that infliximab therapy over a 6-week period was
associated with an increased risk of worsening heart failure in patients in the
high-dose (10 mg/kg) group. The authors concluded that although a similar risk
was not observed in the low-dose (5 mg/kg) group, longer term treatment with
infliximab at this dose could result in more severe adverse events.
Postmarketing reports of worsening heart failure in patients being treated with
infliximab have been reported. These reports have occurred in patients with and
without precipitating factors. In addition, there have been rare postmarketing
reports of new-onset heart failure in patients without diagnosed cardiac
disease. There is a detailed discussion of the clinical implications of treating
patients who have heart failure with infliximab in the “Usage guidelines”
section.
DEMYELINATING DISEASE
As mentioned previously, neurologic symptoms associated with demyelination of
the CNS have been reported to be associated with the anti–TNF-α therapies.
SUMMARY OF EFFICACY DATA FROM IMPORTANT CLINICAL TRIALS
The first case documenting the efficacy of infliximab for the treatment of
psoriasis was described in a case report of a patient with Crohn's disease and
severe psoriasis who was treated with a single 5-mg/kg infusion of infliximab
and achieved an improvement in PASI of 22 points within 4 weeks [343].
Subsequent to that case, several reports have attested to the efficacy of
infliximab for the treatment of moderate to severe psoriasis [344], [345].
A small-scale study was conducted to garner information about the efficacy of
infliximab for the treatment of psoriasis in patients with concomitant psoriatic
arthritis [346]. Six patients with psoriasis and psoriatic arthritis
unresponsive to methotrexate therapy were treated with 5-mg/kg doses of
infliximab at weeks 0, 2, and 6, and PASI evaluations were performed at baseline
and 10 weeks after therapy started. At week 10, all six patients achieved
improvement in psoriasis as measured by PASI score.
In a randomized, double-blind, placebo-controlled clinical trial designed to
assess the clinical benefit and safety of infliximab as monotherapy for patients
with moderate to severe psoriasis, 33 patients were assigned to receive placebo
(n = 11), infliximab doses of 5 mg/kg (n = 11), or infliximab doses of 10 mg/kg
(n = 11) at weeks 0, 2, and 6 [340]. Assessments were performed at week 10 using
the PGA as the primary endpoint and the PASI score as the secondary endpoint.
Nine of 11 (82%) of patients in the 5-mg/kg treatment group and 10 of 11 (91%)
of patients in the 10-mg/kg group achieved the primary endpoint on the PGA at
week 10 of good, excellent, or clear. This finding was in comparison to 11 (18%)
of patients in the placebo group. The difference between infliximab doses of 5
mg/kg and placebo was 64% (95% confidence interval [CI], 20–89; P = 0.0089). The
difference between infliximab doses of 10 mg/kg and placebo was 73% (CI, 30–94;
P = 0.0019). Data regarding the secondary endpoint of the study, change in PASI
score, were also significant. Nine of 11 (82%) of patients in the 5-mg/kg group
and 8 of 11 (73%) of patients in the 10-mg/kg group showed an improvement of 75%
or more in PASI score compared with 11 (18%) patients in the placebo group. The
difference between infliximab doses of 5 mg/kg and placebo was 64% (CI, 20–90; P
= 0.0089); and the difference between infliximab doses of 10 mg/kg and placebo
was 55% (CI, 9–85; P = 0.03). The mean percentage improvements in PASI scores
were significantly higher among the infliximab-treated patients as early as week
2 of therapy (P < 0.0003). The median time to response in both treatment arms
was 4 weeks.
CLINICAL USE IN DERMATOLOGY
Infliximab is currently approved for treatment of RA and Crohn's disease and has
been reported to have clinical efficacy in several cutaneous disorders,
including the following: psoriasis [340], [344], [345], [346]; psoriatic
arthritis [346]; pustular psoriasis [347]; hidradenitis suppurativa [348];
pyoderma gangrenosum [344], [349], [350], [351], [352]; Behçet's syndrome [353],
[354], [355]; synovitis, acne, pustulosis, hyperostosis, and osteitis, or SAPHO,
syndrome [356]; subcorneal pustular dermatosis [357]; and toxic epidermal
necrolysis [358]. Similar to the clinical response to cyclosporine, high
percentages of patients respond to infliximab, and response rates tend to be
rapid. Unlike cyclosporine therapy, however, infliximab is not associated with
the concerning cumulative dose-related side effects of decreased renal function
and hypertension. Another advantage of infliximab is that, similar to efalizumab
and etanercept, infliximab has shown clinical efficacy for both psoriasis and
psoriatic arthritis, although it is not yet FDA-approved for either indication.
In addition, psoriasis is more prevalent in patients who have inflammatory bowel
disease [359]. Infliximab therapy could be beneficial therefore for patients
with concomitant inflammatory bowel disease and psoriasis. Although both
etanercept and infliximab inhibit TNF-α, infliximab has shown superior clinical
efficacy for the treatment of psoriasis. Chaudhari et al [340] offered some
explanations for this discrepancy. They asserted that differences may be from
the route of administration (IV for infliximab versus SC for etanercept),
differences in the patient populations in clinical trials, or differences in the
mechanism by which the drugs work. The different mechanisms have been shown by
in vitro data, which have shown that, unlike etanercept, infliximab is able to
trigger complement-mediated lysis of TNF-α–expressing cells [320]. If this
mechanism is accurate, then the implications are that inflximab is able to
abolish activated T cells and antigen-presenting cells, a potential explanation
for the rapidity and thoroughness of the clinical responses observed in trials.
USAGE GUIDELINES
A black box warning has been issued by the manufacturer of infliximab cautioning
that the drug should not be given to patients with clinically important active
infections. Caution should also be taken when considering the use of infliximab
for patients with chronic or recurrent infections. If a patient develops a
serious infection while being treated with infliximab, the treatment should be
discontinued. TB (frequently disseminated or extrapulmonary at clinical
presentation), invasive fungal infections, and other opportunistic infections
have been observed in patients being treated with infliximab [341]. Some
infections have been fatal. Patients should be evaluated for latent TB infection
with a tuberculin skin test, and treatment of latent TB infections should be
started before infliximab therapy. For patients who have resided in regions
where histoplasmosis or coccidioidomycosis is endemic, the benefits and risks of
infliximab therapy should be carefully considered before initiation of therapy.
Infliximab should only be used in patients with heart failure after other
treatment options have been considered. If a decision is made to administer
infliximab to these patients, the patients should be closely monitored during
therapy and treatment should be discontinued if new or worsening symptoms of
heart failure appear [341].
Physicians should exercise caution when considering the use of infliximab for
the treatment of patients with pre-existing or recent-onset CNS demyelinating or
seizure disorders [341].
Infliximab should not be administered to patients with known hypersensitivity to
any murine proteins or other component of the drug. To date, no animal
reproduction studies have been performed. It is therefore advised that
infliximab only be used in pregnancy when clearly clinically necessary [341].
Infliximab only cross-reacts with TNF-α in humans and chimpanzees. In a
developmental toxicity study using cV1q antimouse TNF-α, an analogous antibody
that inhibits mouse TNF-α, no evidence of carcinogenesis, mutagenesis, or
impairment of fertility was observed [341]. Doses of up to 40 mg/kg did not
produce any adverse effects in animal reproduction studies. It is not known
whether infliximab can cause harm to the fetus, if it can affect reproductive
capacity, or if it is excreted in human milk or absorbed systemically post
ingestion. Infliximab has been labeled pregnancy category B.
Because of the higher incidence of infections in the elderly population, caution
should be used when treating elderly patients. Infliximab is currently not
indicated for pediatric use.
DRUG INTERACTIONS
Drug interaction studies with infliximab have not been conducted. In RA and
Crohn's disease clinical trials, most patients were treated with one or more
concomitant medications. Patients with RA were treated with concomitant
methotrexate, NSAIDs, folic acid, corticosteroids, or narcotics. Patients with
Crohn's disease in clinical trials were treated with concomitant antibiotics,
antiviral medications, corticosteroids, 6-MP/azathioprine, and aminosalicylates.
Patients with Crohn's disease who received immunosuppresants tended to have
fewer infusion reactions. As previously mentioned, Nahar et al [324] reported
that concomitant treatment with immunosuppresants may have predisposed patients
in the ACCENT [338] and ATTRACT [339] clinical trials to increased rates of
serious infections.
DOSAGE AND MONITORING
Infliximab is indicated for the treatment of RA and Crohn's disease. Infliximab
is administered as an IV drip over the course of several hours. The mode of
administration of infliximab is a potential drawback for both patients and
physicians because of the time commitment involved, increased need for skilled
health care workers in the office at the time of administration, and potential
physical burden for arthritic patients who may have difficulty sitting in one
position for the length of the infusion. For the treatment of RA, infliximab is
given as a 3-mg/kg IV infusion followed by infusions at weeks 2 and 6, and then
every 8 weeks thereafter. Infliximab should be administered in combination with
methotrexate for the treatment of RA. Doses of up to 10 mg/kg may be used for
patients with RA who have incomplete clinical responses to lower doses. For
treatment of patients with Crohn's disease, the recommended dose of infliximab
is 5 mg/kg as an induction dose at baseline and weeks 2 and 6. Maintenance doses
of 5 mg/kg every 8 weeks after induction can be used to treat moderate to severe
active Crohn's disease or fistulizing disease. Doses of up to 10 mg/kg may be
used for the treatment of Crohn's disease. For treatment of psoriasis and
Crohn's disease, the lower 5-mg/kg dose of infliximab has shown greater efficacy
than the 10-mg/kg dose [340], [360].
The drug is supplied as a sterile, white, lyophilized powder for IV infusion.
Infliximab vials do not contain preservatives and must therefore be used
immediately after reconstitution. Monitoring guidelines are presented in Table
4.
SUMMARY
Azathioprine, cyclophosphamide, methotrexate, cyclosporine, and MMF are the
nonsteroidal immunosuppressive agents most commonly used by dermatologists.
Azathioprine has a relatively good safety profile and is therefore often
preferred for the treatment of chronic eczematous dermatitides and bullous
disorders. Awareness of the role of genetic polymorphisms in its metabolism can
increase the efficacy and safety of this drug. Cyclophosphamide is an
antimetabolite that has a more rapid onset of immunosuppressive effect than
azathioprine but has significant short- and long-term toxicity. It is of use in
fulminant, life-threatening cutaneous disease. Methotrexate is an antimetabolite
that has significant anti-inflammatory activity. Despite its hepatotoxicity, its
role in inflammatory dermatoses is broadening. Cyclosporine has potent T-cell
inhibitory effects secondary to interference with intracellular signal
transduction, and its use in dermatology is rapidly expanding. Given the
evidence for cumulative renal toxicity, it currently has a role in the
short-term treatment of refractory psoriasis and atopic dermatitis, and in
select inflammatory dermatoses. MMF is an immune suppressant with wide potential
dermatologic applications, especially for bullous disorders, pyoderma
gangrenosum, and psoriasis. It has a mechanism of action similar to
azathioprine, and it is generally well tolerated. Although some studies found a
slightly higher incidence of lymphoproliferative malignancies, MMF has a better
overall safety profile than azathioprine. It is more expensive, however. Both
mycophenolate and azathioprine should be used in conjunction with oral
corticosteroids because they are “steroid sparing” but not “steroid replacing.”
They are usually required when it is impossible to reduce steroids to an
acceptably low level. When they are added, as they become effective, the
corticosteroid taper is resumed toward the target maintenance dose.
Alefacept, efalizumab, etanercept, and infliximab are the immunobiologic agents
currently most commonly used by dermatologists. Alefacept, the first
immunobiologic agent to be approved for the treatment of adult patients with
moderate to severe plaque-type psoriasis, is a fusion protein that blocks T-cell
activation and selectively reduces memory T cells. Because of its unique
mechanism of action, alefacept is considered to be the only remittive biologic
agent. Therefore, patients who respond effectively to alefacept can enjoy
long-lasting therapy-free reprieves for more than 7 months. Alefacept is also
safe; safety and tolerability profiles were shown to be similar to placebo in
phase 3 clinical trials. Similar to all of the immunobiologic agents, data on
the potential for the development of malignancies require longer follow-up
periods before a possible role of these agents in the development of malignancy
can be excluded. The intramuscular route of administration, and the need for
weekly CD4+ T-cell counts, also distinguishes alefacept, because it requires the
patient to make weekly visits to a physician's office. Efalizumab, a humanized
monoclonal antibody directed against CD11a, is indicated for the treatment of
chronic moderate to severe plaque-type psoriasis. Unlike alefacept, efalizumab
is administered as a weekly SC injection. Thrombocytopenia was reported in a
small percentage of clinical trial patients; therefore, platelet counts must be
monitored throughout treatment. Efalizumab is generally well tolerated but,
similar to many of the systemic immunomodulators previously discussed, on
cessation of therapy, some patients have experienced clinically significant
relapse or exacerbation of psoriasis. Although long-term safety data have not
been established for any of the immunobiologic agents, efalizumab has been shown
to have a favorable side-effect profile and potential as a long-term therapeutic
agent for chronic psoriasis. Etanercept is a dimeric fusion protein that
inhibits TNF-α. Etanercept is FDA-approved for the treatment of psoriasis,
psoriatic arthritis, and RA, and for reducing signs and symptoms of moderate to
severe active polyarticular-course juvenile RA. Etanercept has a favorable
safety profile. It is the only immunobiologic agent of the four discussed that
does not require laboratory monitoring. Infliximab is a chimeric monoclonal
antibody that targets TNF-α. It is currently indicated for the treatment of RA
and Crohn's disease but has shown efficacy for a wide array of dermatologic
conditions, especially psoriasis and psoriatic arthritis, pyoderma gangrenosum,
and the cutaneous manifestations of Behçet's syndrome. High percentages of
patients with psoriasis respond to infliximab, and response rates tend to be
rapid and long-lasting. Infliximab has several potential drawbacks, however,
including its route of administration, which is by means of an IV drip over the
course of several hours. Infusion reactions also have been a concern, although
less than 1% of infusion reactions in clinical trials were considered serious.
Other issues have been postmarketing reports of worsening heart failure,
reactivation TB, and other opportunistic infections. In addition, some patients
have developed neutralizing antibodies. Unlike methotrexate and cyclosporine,
for example, infliximab is neither hepatotoxic nor nephrotoxic; it therefore
represents a useful therapeutic agent for patients with severe psoriasis who
have contraindications to other immunosuppressive agents or who are recalcitrant
to other therapies.
--------------------------------------------------------------------------------
REFERENCES:
[1] Dutz J., Ho V., Immunosuppressive agents in skin disorders. Clin Immunother
(1996) 5 : pp 268-293.
[2] Ho V.C., Zloty D.M., Immunosuppressive agents in dermatology. Dermatol Clin
(1993) 11 : pp 73-85. Abstract
[3] Dutz J.P., Ho V.C., Immunosuppressive agents in dermatology. An update.
Dermatol Clin (1998) 16 : pp 235-251. Full Text
[4] Zhou Y., Dutz J.P., Ho V.C., Mycophenolate mofetil (CellCept (R)) for
dermatological diseases. J Cutan Med Surg (2001) 5 : pp 26-29.
[5] Lebwohl M., Future psoriasis therapy. Dermatol Clin (1985) 13 : pp 415-423.
[6] Lebwohl M., Advances in psoriasis therapy. Dermatol Clin (2000) 18 : pp
13-21. Full Text
[7] Lennard L., Rees C., Lilleyman J., Maddocks J.L., Childhood leukemia: a
relationship between intracellular 6-mercaptopurine metabolism and neutropenia.
Br J Clin Pharmacol (1983) 16 : pp 359-363. Abstract
[8] Holme S.A., Duley J.A., Sanderson J., Routledge P.A., Anstey A.V.,
Erythrocyte thiopurine methyl transferase assessment prior to azathioprine use
in the UK. QJM (2002) 95 : pp 439-444. Abstract
[9] Chocair P., Duley J., Simmonds H., Cameron J.S., The importance of
thiopurine methyltransferase activity for the use of azathioprine in transplant
patients. Transplantation (1992) 53 : pp 1051-1056. Abstract
[10] Lennard L., Harrington C., Wood M., Maddocks J.L., Metabolism of
azathioprine to 6-thioguanine nucleotides in patients with pemphigus vulgaris.
Br J Clin Pharmacol (1987) 23 : pp 229-233. Abstract
[11] Snow J., Gibson L., A pharmacogenetic basis for the safe and effective use
of azathioprine and other thiopurine drugs in dermatology patients. J Am Acad
Dermatol (1995) 32 : pp 114-116. Full Text
[12] Yates C., Krynetski E., Loennechen T., Molecular diagnosis of thiopurine
S-methyltransferase deficiency: genetic basis for azathioprine and
mercaptopurine intolerance. Ann Intern Med (1997) 126 : pp 608-614. Abstract
[13] Anstey A., Management of immunobullous disorders: the clinical significance
of interindividual variation in azathioprine metabolism. Clin Exp Dermatol
(1997) 21 : pp 244-251.
[14] Marra C.A., Esdaile J.M., Anis A.H., Practical pharmacogenetics: the cost
effectiveness of screening for thiopurine s-methyltransferase polymorphisms in
patients with rheumatological conditions treated with azathioprine. J Rheumatol
(2002) 29 : pp 2507-2512. Abstract
[15] Vesell E., Therapeutic lessons from pharmacogenetics [editorial]. Ann
Intern Med (1997) 126 : pp 653-655. Citation
[16] Aberer W., Wolff-Schreiner E.C., Stingl G., Wolff K., Azathioprine in the
treatment of pemphigus vulgaris. J Am Acad Dermatol (1987) 16 : pp 527-533.
Abstract
[17] Tan B.B., Lear J.T., Gawkrodger D.J., English J.S., Azathioprine in
dermatology: a survey of current practice in the UK. Br J Dermatol (1997) 136 :
pp 351-355. Abstract
[18] Singh G., Fries J.F., Spitz P., Williams C.A., Toxic effects of
azathioprine in rheumatoid arthritis: a national post-marketing perspective.
Arthritis Rheum (1989) 32 : pp 837-843. Abstract
[19] Schwab M., Schaffeler E., Marx C., Azathioprine therapy and adverse drug
reactions in patients with inflammatory bowel disease: impact of thiopurine
S-methyltransferase polymorphism. Pharmacogenetics (2002) 12 : pp 429-436.
Abstract
[20] Guillaume J.C., Vaillant L., Bernard P., Controlled trial of azathioprine
and plasma exchange in addition to prednisolone in the treatment of bullous
pemphigoid. Arch Dermatol (1993) 129 : pp 49-53. Abstract
[21] Penn I., Cancers in cyclosporine-treated versus azathioprine-treated
patients. Transplant Proc (1996) 28 : pp 876-878. Citation
[22] Bouwes Bavinck J.N., Hardie D.R., Green A., The risk of skin cancer in
renal transplant recipients in Queensland, Australia. A follow-up study.
Transplantation (1996) 61 : pp 715-721. Abstract
[23] Confavreux C., Saddier P., Grimaud J., Moreau T., Adeleine P., Aimard G.,
Risk of cancer from azathioprine therapy in multiple sclerosis: a case-control
study. Neurology (1996) 46 : pp 1607-1612. Full Text
[24] Fraser A.G., Orchard T.R., Robinson E.M., Jewell D.P., Long-term risk of
malignancy after treatment of inflammatory bowel disease with azathioprine.
Aliment Pharmacol Ther (2002) 16 : pp 1225-1232. Abstract
[25] Beckett C.G., Hill P., Hine K.R., Leucocytoclastic vasculitis in a patient
with azathioprine hypersensitivity [review]. Postgrad Med J (1996) 72 : pp
437-438. Abstract
[26] Compton M.R., Crosby D.L., Rhabdomyolysis associated with azathioprine
hypersensitivity syndrome [letter]. Arch Dermatol (1996) 132 : pp 1254-1255.
Citation
[27] Knowles S., Gupta A., Shear N., Sauder D., Azathioprine
hypersensitivity-like reactions—a case report and review of the literature. Clin
Exp Dermatol (1995) 20 : pp 353-356. Abstract
[28] Corbett M., Schlup M., Azathioprine hypersensitivity mimicking underlying
inflammatory bowel disease. Intern Med J (2001) 31 : pp 366-367. Citation
[29] Saway P.A., Heck L.W., Bonner J.R., Kirklin J.K., Azathioprine
hypersensitivity. Am J Med (1988) 84 : pp 960-964. Abstract
[30] Blanco R., Martinez-Taboada V.M., Gonzalez-Gay M.A., Acute febrile toxic
reaction in patients with refractory rheumatoid arthritis who are receiving
combined therapy with methotrexate and azathioprine [review]. Arthritis Rheum
(1996) 39 : pp 1016-1020. Abstract
[31] van der Pijl J.W., Bavinck J.N., de Fijter J.W., Isotretinoin and
azathioprine: a synergy that makes hair curl [letter]?. Lancet (1996) 348 : pp
622-623. Citation
[32] Sudhir R.R., Rao S.K., Shanmugam M.P., Padmanabhan P., Bilateral macular
hemorrhage caused by azathioprine-induced aplastic anemia in a corneal graft
recipient. Cornea (2002) 21 : pp 712-714. Abstract
[33] Bystryn J.C., Steinman N.M., The adjuvant therapy of pemphigus. An update
[review]. Arch Dermatol (1996) 132 : pp 203-212. Abstract
[34] Villalba L., Adams E.M., Update on therapy for refractory dermatomyositis
and polymyositis [review]. Curr Opin Rheumatol (1996) 8 : pp 544-551. Abstract
[35] Lear J.T., English J.S., Jones P., Smith A.G., Retrospective review of the
use of azathioprine in severe atopic dermatitis. J Am Acad Dermatol (1996) 35 :
pp 642-643. Full Text
[36] Berth-Jones J., Takwale A., Tan E., Azathioprine in severe adult atopic
dermatitis: a double-blind, placebo-controlled, crossover trial. Br J Dermatol
(2002) 147 : pp 324-330. Abstract
[37] Lear J.T., English J.S., Smith A.G., Nodular prurigo responsive to
azathioprine [letter]. Br J Dermatol (1996) 134 : pp 1151-. Citation
[38] Yazici H., Pazarli H., Barnes C.G., A controlled trial of azathioprine in
Behcet's syndrome. N Engl J Med (1990) 322 : pp 281-285. Abstract
[39] Hamuryudan V., Ozyazgan Y., Hizli N., Azathioprine in Behcet's syndrome:
effects on long-term prognosis. Arthritis Rheum (1997) 40 : pp 769-774. Abstract
[40] Klein L.R., Callen J.P., Azathioprine: effective steroid-sparing therapy
for generalized lichen planus. South Med J (1992) 85 : pp 198-201. Citation
[41] Lear J.T., English J.S., Erosive and generalized lichen planus responsive
to azathioprine. Clin Exp Dermatol (1996) 21 : pp 56-57. Abstract
[42] Briggs G.G. Freeman R.K. Yaffe S.J. Drugs in pregnancy and lactation, a
reference guide to fetal and neonatal risk 4th edition. 1994Baltimore, MD:
Williams & Wilkins : pp 79a-82a.
[43] Friedman J.M., Polifka J.E., Teratogenic effects of drugs, a resource for
clinicians 1994Baltimore, MD: John Hopkins University Press : pp 53-55.
[44] Cummins D., Sekar M., Halil O., Banner N., Myelosuppression associated with
azathioprine-allopurinol interaction after heart and lung transplantation.
Transplantation (1996) 61 : pp 1661-1662. Abstract
[45] Schutz E., Gummert J., Mohr F., Armstrong V.H., Oellerich M., Should
6-thioguanine nucleotides be monitored in heart transplant recipients given
azathioprine?. Ther Drug Monit (1996) 18 : pp 228-233. Abstract
[46] Gossmann J., Kachel H.G., Schoeppe W., Scheuermann E.H., Anemia in renal
transplant recipients caused by concomitant therapy with azathioprine and
angiotensin-converting enzyme inhibitors. Transplantation (1993) 56 : pp
585-589. Abstract
[47] Gossmann J., Thurmann P., Bachmann T., Mechanism of angiotensin converting
enzyme inhibitor-related anemia in renal transplant recipients. Kidney Int
(1996) 50 : pp 973-978. Abstract
[48] American College of Rheumatology Ad Hoc Committee on Clinical Guidelines
Guidelines for monitoring drug therapy in rheumatoid arthritis. Arthritis Rheum
(1996) 39 : pp 723-731.
[49] Bradley P.P., Warden G.D., Maxwell J.G., Rothstein G., Neutropenia and
thrombocytopenia in renal allograft recipients treated with
trimethoprim-sulfamethoxazole. Ann Intern Med (1980) 93 : pp 560-562. Abstract
[50] Olsen S.L., Renlund D.G., O'Connell J.B., Prevention of Pneumocystis
carinii pneumonia in cardiac transplant recipients by trimethoprim
sulfamethoxazole. Transplantation (1993) 56 : pp 359-362. Abstract
[51] Rivier G., Khamashta M.A., Hughes G.R., Warfarin and azathioprine: a drug
interaction does exist [letter]. Am J Med (1993) 95 : pp 342-. Citation
[52] Havrda D.E., Rathbun S., Scheid D., A case report of warfarin resistance
due to azathioprine and review of the literature. Pharmacotherapy (2001) 21 : pp
355-357. Abstract
[53] Murphy L.A., Atherton D., A retrospective evaluation of azathioprine in
severe childhood atopic eczema, using thiopurine methyltransferase levels to
exclude patients at high risk of myelosuppression. Br J Dermatol (2002) 147 : pp
308-315. Abstract
[54] McDonald C.J., Cytotoxic agents for use in dermatology I. J Am Acad
Dermatol (1985) 12 : pp 753-775. Abstract
[55] Moore M.J., Clinical pharmacokinetics of cyclophosphamide. Clin
Pharmacokinet (1991) 20 : pp 194-208. Abstract
[56] Hemendinger R.A., Bloom S.E., Selective mitomycin C and cyclophosphamide
induction of apoptosis in differentiating B lymphocytes compared to T
lymphocytes in vivo. Immunopharmacology (1996) 35 : pp 71-82. Abstract
[57] Rothe H., Burkart V., Faust A., Kolb H., Interleukin-12 gene expression
mediates the accelerating effect of cyclophosphamide in autoimmune disease. Ann
N Y Acad Sci (1996) 795 : pp 397-399. Citation
[58] Marcinkiewicz J., Bryniarski K., Ptak W., Cyclophosphamide uncovers two
separate macrophage subpopulations with opposite immunogenic potential and
different patterns of monokine production. Cytokine (1994) 6 : pp 472-477.
Abstract
[59] Yarboro C.H., Wesley R., Amantea M.A., Klippel J.H., Pucino F., Modified
oral ondansetron regimen for cyclophosphamide-induced emesis in lupus nephritis.
Ann Pharmacother (1996) 30 : pp 752-755. Abstract
[60] Aapro M.S., Thuerlimann B., Sessa C., De Pree C., Bernhard J., Maibach R.,
A randomized double-blind trial to compare the clinical efficacy of granisetron
with metoclopramide, both combined with dexamethasone in the prophylaxis of
chemotherapy-induced delayed emesis. Ann Oncol (2003) 14 : pp 291-297. Abstract
[61] Fraiser L., Kanekal S., Kehrer J., Cyclophosphamide toxicity.
Characterising and avoiding the problem. Drugs (1991) 42 : pp 781-791. Abstract
[62] Stillwell T.J., Benson R.C., Jr, Cyclophosphamide-induced hemorrhagic
cystitis—a review of 100 patients. Cancer (1988) 61 : pp 451-457. Abstract
[63] Talar-Williams C., Hijazi Y.M., Walther M.M., Cyclophosphamide-induced
cystitis and bladder cancer in patients with Wegener granulomatosis. Ann Intern
Med (1996) 124 : pp 477-484. Abstract
[64] Haselberger M., Schwinghammer T., Efficacy of mesna for the prevention of
hemorrhagic cystitis after high-dose cyclophosphamide therapy. Ann Pharmacother
(1995) 29 : pp 918-920. Abstract
[65] Fairchild W.V., Spence C.R., Soloman H.D., The incidence of bladder cancer
after cyclophosphamide therapy. J Urol (1979) 122 : pp 163-164. Abstract
[66] Baker G.L., Kahl L.E., Zee B.C., Malignancy following treatment of
rheumatoid arthritis with cyclophosphamide; long-term case-control follow-up
study. Am J Med (1987) 83 : pp 1-9. Abstract
[67] Baltus J.A., The occurrence of malignancies in patients with rheumatoid
arthritis treated with cyclophosphamide: a controlled retrospective follow up.
Ann Rheum Dis (1983) 42 : pp 368-373. Abstract
[68] Radis C.D., Kahl L.E., Baker G.L., Effects of cyclophosphamide on the
development of malignancy and on long-term survival of patients with rheumatoid
arthritis. A 20-year follow-up study. Arthritis Rheum (1995) 38 : pp 1120-1127.
Abstract
[69] Volm T., Pfaff P., Gnann R., Kreienberg R., Bladder carcinoma associated
with cyclophosphamide therapy for ovarian cancer occurring with a latency of 20
years. Gynecol Oncol (2001) 82 : pp 197-199. Abstract
[70] Masala A., Faedda R., Alagna S., Use of testosterone to prevent
cyclophosphamide-induced azoospermia. Ann Intern Med (1997) 126 : pp 292-295.
Abstract
[71] Blumenfeld Z., Haim N., Prevention of gonadal damage during cytotoxic
therapy. Ann Med (1997) 29 : pp 199-206. Abstract
[72] Pryor B.D., Bologna S.G., Kahl L.E., Risk factors for serious infection
during treatment with cyclophosphamide and high-dose corticosteroids for
systemic lupus erythematosus. Arthritis Rheum (1996) 39 : pp 1475-1482. Abstract
[73] Cohen M.A., Cohen J.J., Kerdel F.A., Immunoablative high-dose
cyclophosphamide without stem cell rescue in pemphigus foliaceus. Int J Dermatol
(2002) 41 : pp 340-344. Abstract
[74] Hayag M.V., Cohen J.A., Kerdel F.A., Immunoablative high-dose
cyclophosphamide without stem cell rescue in a patient with pemphigus vulgaris.
J Am Acad Dermatol (2000) 43 : pp 1065-1069. Full Text
[75] Werth V.P., Pulse intravenous cyclophosphamide for treatment of autoimmune
blistering disease. Is there an advantage over oral routes [editorial]?. Arch
Dermatol (1997) 133 : pp 229-230. Citation
[76] Pasricha J.S., Khaitan B.K., Curative treatment for pemphigus [letter].
Arch Dermatol (1996) 132 : pp 1518-1519. Citation
[77] Pandya A.G., Warren K.J., Bergstresser P.R., Cicatricial pemphigoid
successfully treated with pulse intravenous cyclophosphamide [letter]. Arch
Dermatol (1997) 133 : pp 245-247. Citation
[78] Itoh T., Hosokawa H., Shirai Y., Horio T., Successful treatment of bullous
pemphigoid with pulsed intravenous cyclophosphamide. Br J Dermatol (1996) 134 :
pp 931-933. Abstract
[79] Castle S.P., Mather-Mondrey M., Bennion S., David-Bajar K., Huff C.,
Chronic herpes gestationis and antiphospholipid antibody syndrome successfully
treated with cyclophosphamide. J Am Acad Dermatol (1996) 34 : pp 333-336. Full
Text
[80] Frangogiannis N.G., Boridy I., Mazhar M., Mathews R., Gangopadhyay S., Cate
T., Cyclophosphamide in the treatment of toxic epidermal necrolysis. South Med J
(1996) 89 : pp 1001-1003. Abstract
[81] Heng M.C.Y., Allen S.G., Efficacy of cyclophosphamide in toxic epidermal
necrolysis. J Am Acad Dermatol (1991) 25 : pp 778-786. Abstract
[82] Dawisha S.M., Yarboro C.H., Vaughan E.M., Austin H.A., III, Balow J.E.,
Klippel J.H., Out-patient monthly oral bolus cyclophosphamide therapy in
systemic lupus erythematosus. J Rheumatol (1996) 23 : pp 273-278. Abstract
[83] Craig N.M., Putterman A.M., Roenigk R.K., Wang T.D., Roenigk H.H., Multiple
periorbital cutaneous myxomas progressing to scleromyxedema. J Am Acad Dermatol
(1996) 34 : pp 928-930. Full Text
[84] Liang G.C., Granston A.S., Complete remission of multicentric
reticulohistiocytosis with combination therapy of steroid, cyclophosphamide, and
low-dose pulse methotrexate. Case report, review of the literature, and proposal
for treatment [review]. Arthritis Rheum (1996) 39 : pp 171-174. Abstract
[85] Bhagat R., Sporn T.A., Long G.D., Folz R.J., Amiodarone and
cyclophosphamide: potential for enhanced lung toxicity. Bone Marrow Transplant
(2001) 27 : pp 1109-1111. Abstract
[86] Balis F.M., Mirro J., Reaman G.H., Pharmacokinetics of subcutaneous
methotrexate. J Clin Oncol (1988) 6 : pp 1882-1886. Abstract
[87] Bannwarth B., Pehourcq F., Schaeverbeke T., Dehais J., Clinical
pharmacokinetics of low-dose pulse methotrexate in rheumatoid arthritis
[review]. Clin Pharmacokinet (1996) 30 : pp 194-210. Abstract
[88] Bannwarth B., Labat L., Moride Y., Schaeverbeke T., Methotrexate in
rheumatoid arthritis, an update. Drugs (1994) 47 : pp 25-50. Abstract
[89] Olsen E.A., The pharmacology of methotrexate. J Am Acad Dermatol (1991) 25
: pp 306-318. Abstract
[90] Cronstein B.N., Molecular therapeutics. Methotrexate and its mechanism of
action [review]. Arthritis Rheum (1996) 39 : pp 1951-1960. Citation
[91] Jeffes E.W.B., III, McCullough J.L., Pittelkow M.R., Methotrexate therapy
of psorisas: differential sensitivity of proliferating lymphoid and epithelial
cells to the cytotoxic and growth-inhibitory effects of methotrexate. J Invest
Dermatol (1995) 104 : pp 183-188. Abstract
[92] Phillips D.C., Woollard K.J., Griffiths H.R., The anti-inflammatory actions
of methotrexate are critically dependent upon the production of reactive oxygen
species. Br J Pharmacol (2003) 138 : pp 501-511. Abstract
[93] Cronstein B.N., Molecular therapeutics. Methotrexate and its mechanism of
action [review]. Arthritis Rheum (1996) 39 : pp 1951-1960. Citation
[94] Andersson S.E., Johansson L.H., Lexmuller K., Ekstrom G.M., Anti-arthritic
effect of methotrexate: is it really mediated by adenosine?. Eur J Pharm Sci
(2000) 9 : pp 333-343. Abstract
[95] Cronstein B.N., Naime D., Ostad E., The antiinflammatory mechanism of
methotrexate. J Clin Invest (1993) 92 : pp 2675-2682. Abstract
[96] Van Dooren-Greebe R.J., Kuijpers A.L., Mulder J., De Boo T., Van de Kerkhof
P.C., Methotrexate revisited: effects of long-term treatment in psoriasis. Br J
Dermatol (1994) 130 : pp 204-210. Abstract
[97] Al-Awadhi A., Dale P., McKendry R.J., Pancytopenia associated with low dose
methotrexate therapy. A regional survey. J Rheumatol (1993) 20 : pp 1121-1125.
Abstract
[98] Guzzo C., Folic acid supplementation with methotrexate therapy: what
benefit if any?. J Am Acad Dermatol (1994) 31 : pp 689-. Citation
[99] Kirby B., Lyon C.C., Griffiths C.E., Chalmers R.J., The use of folic acid
supplementation in psoriasis patients receiving methotrexate: a survey in the
United Kingdom. Clin Exp Dermatol (2000) 25 : pp 265-268. Abstract
[100] Duhra P., Treatment of gastrointestinal symptoms associated with
methotrexate therapy for psoriasis. J Am Acad Dermatol (1993) 28 : pp 466-469.
Abstract
[101] Kremer J.M., Alarcon G.S., Lightfoot R.W., Jr, Methotrexate for rheumatoid
arthritis: suggested guidelines for monitoring liver toxicity. Arthritis Rheum
(1994) 37 : pp 316-328. Abstract
[102] Whiting-O'Keefe Q., Fye K.H., Sack K.D., Methotrexate and histologic
hepatic abnormalities: a meta-analysis. Am J Med (1991) 90 : pp 711-716.
Abstract
[103] Auerbach R., Psoriasis symposium. Methotrexate. Semin Dermatol (1992) 11 :
pp 23-29. Citation
[104] Zachariae H., Sogaard H., Heickendorff L., Methotrexate-induced liver
cirrhosis. Clinical, histological and serological studies—a further 10-year
followup. Dermatology (1996) 192 : pp 343-346. Abstract
[105] Roenigk H.H., Jr, Auerbach R., Maibach H.I., Weinstein G., Lebwohl M.,
Methotrexate in psoriasis: consensus conference. J Am Acad Dermatol (1998) 38 :
pp 478-485. Full Text
[106] Rothe M.J., Grin-Jorgensen C., Ramsey W.H., Grant-Kels J.M., Monitoring
patients taking methotrexate. J Am Acad Dermatol (1995) 32 : pp 680-. Full Text
[107] Petrazzuoli M., Rothe M.J., Grin-Jorgensen C., Ramsey W.H., Grant-Kels
J.M., Monitoring patients taking methotrexate for hepatotoxicity. J Am Acad
Dermatol (1994) 31 : pp 969-977. Abstract
[108] Rosenthal D., Guenther L., Kelly J., Current thoughts on the use of
methotrexate in patients with psoriasis. J Cutan Med Surg (1997) 2 : pp 41-46.
[109] Boffa M., Chalmers R., Haboubi N., Shomaf M., Mitchell D.M., Sequential
liver biopsies during long-term methotrexate treatment for psoriasis: a
reappraisal. Br J Dermatol (1995) 133 : pp 774-778. Abstract
[110] Kumar B., Saraswat A., Kaur I., Short-term methotrexate therapy in
psoriasis: a study of 197 patients. Int J Dermatol (2002) 41 : pp 444-448.
Abstract
[111] Boffa M.J., Smith A., Chalmers R.J., Serum type III procollagen
aminopeptide for assessing liver damage in methotrexate-treated psoriatic
patients. Br J Dermatol (1996) 135 : pp 538-544. Abstract
[112] Zachariae H., Heickendorff L., Sogaard H., The value of amino-terminal
propeptide of type III procollagen in routine screening for methotrexate-induced
liver fibrosis: a 10-year follow-up. Br J Dermatol (2001) 144 : pp 100-103.
Abstract
[113] Stern R.S., Laird N., The carcinogenic risk of treatments for severe
psoriasis. Cancer (1994) 73 : pp 2759-2764. Abstract
[114] Zumtobel U., Schwarze H.P., Favre M., Taieb A., Delaunay M., Widespread
cutaneous carcinomas associated with human papillomaviruses 5, 14 and 20 after
introduction of methotrexate in two long-term PUVA-treated patients. Dermatology
(2001) 202 : pp 127-130. Abstract
[115] Kamel O.W., Weiss L.M., van de Rijn M., Colby T.V., Kingma D.W., Jaffe
E.S., Hodgkin's disease and lymphoproliferations resembling Hodgkin's disease in
patients receiving long-term low-dose methotrexate therapy. Am J Surg Pathol
(1996) 20 : pp 1279-1287. Abstract
[116] Paul C., LeTourneau A., Cayuela J.M., Epstein-Barr virus-associated
lymphoproliferative disease during methotrexate therapy for psoriasis. Arch
Dermatol (1997) 133 : pp 867-871. Abstract
[117] Ebeo C.T., Girish M.R., Byrd R.P., Roy T.M., Mehta J.B.,
Methotrexate-induced pulmonary lymphoma. Chest (2003) 123 : pp 2150-2153. Full
Text
[118] Hirose Y., Masaki Y., Okada J., Epstein-Barr virus-associated B-cell type
non-Hodgkin's lymphoma with concurrent p53 protein expression in a rheumatoid
arthritis patient treated with methotrexate. Int J Hematol (2002) 75 : pp
412-415. Abstract
[119] Moder K.G., Tefferi A., Cohen M.D., Menke D.M., Luthra H.S., Hematologic
malignancies and the use of methotrexate in rheumatoid arthritis: a
retrospective study. Am J Med (1995) 99 : pp 276-281. Abstract
[120] Donnenfeld A.E., Pastuszak A., Noah J.S., Schick B., Rose N.C., Koren G.,
Methotrexate exposure prior and during pregnancy [letter]. Teratology (1994) 49
: pp 79-81. Citation
[121] Boerbooms A.M.T., Kerstens P.J., van Loenhout J.W., Mulder J., van de
Putte L.B., Infections during low-dose methotrexate treatment in rheumatoid
arthritis. Semin Arthritis Rheum (1995) 24 : pp 411-421. Abstract
[122] Alkins S.A., Byrd J.C., Morgan S.K., Ward F.T., Weiss R.B., Anaphylactoid
reactions to methotrexate [review]. Cancer (1996) 77 : pp 2123-2126. Abstract
[123] Pearce H.P., Wilson B.B., Erosion of psoriatic plaques: an early sign of
methotrexate toxicity [review]. J Am Acad Dermatol (1996) 35 : pp 835-838. Full
Text
[124] Ben-Amitai D., Hodak E., David M., Cutaneous ulceration: an unusual sign
of methotrexate toxicity—first report in a patient without psoriasis. Ann
Pharmacother (1998) 32 : pp 651-653. Abstract
[125] Zonneveld I.M., Bakker W.K., Dijkstra P.F., Bos J.D., van Soesbergen R.M.,
Dinant H.J., Methotrexate osteopathy in long-term, low-dose methotrexate
treatment for psoriasis and rheumatoid arthritis. Arch Dermatol (1996) 132 : pp
184-187. Abstract
[126] Pigatto P.D., Gibelli E., Ranza R., Rossetti A., Methotrexate in psoriatic
polyarthritis. Acta Derm Venereol (1994) 74 : pp 114-115.
[127] Clayton B.D., Jorizzo J.L., Hitchcock M.G., Adult pityriasis rubra
pilaris: a 10 year case series. J Am Acad Dermatol (1997) 36 : pp 959-964. Full
Text
[128] Andersen W.K., Feingold D.S., Adverse drug interactions clinically
important for the dermatologist. Arch Dermatol (1995) 131 : pp 468-473. Abstract
[129] Vonderheid E.C., Sajjadian A., Kadin M.E., Methotrexate is effective
therapy for lymphomatoid papulosis and other primary cutaneous CD30-positive
lymphoproliferative disorders. J Am Acad Dermatol (1996) 34 : pp 470-481. Full
Text
[130] Kasteler J.S., Callen J.P., Low-dose methotrexate administered weekly is
an effective corticosteroid-sparing agent for the treatment of the cutaneous
manifestations of dermatomyositis. J Am Acad Dermatol (1997) 36 : pp 67-71. Full
Text
[131] Carson P.J., Hameed A., Ahmed A.R., Influence of treatment on the clinical
course of pemphigus vulgaris. J Am Acad Dermatol (1996) 34 : pp 645-652. Full
Text
[132] Paul M.A., Jorizzo J.L., Fleischer A.J., White W.L., Low-dose methotrexate
treatment in elderly patients with bullous pemphigoid. J Am Acad Dermatol (1994)
31 : pp 620-625. Abstract
[133] Teitel A.D., Treatment of pyoderma gangrenosum with methotrexate. Cutis
(1996) 57 : pp 326-328. Abstract
[134] Seyger M.M., van den Hoogen F.H., de Boo T., de Jong E.M., Low-dose
methotrexate in the treatment of widespread morphea. J Am Acad Dermatol (1998)
39 : pp 220-225. Full Text
[135] Franck N., Amor B., Ayral X., Multicentric reticulohistiocytosis and
methotrexate. J Am Acad Dermatol (1995) 33 : pp 524-525. Full Text
[136] Faulds D., Goa K.L., Benfield P., Cyclosporin: a review of its
pharmacodynamic and pharmacokinetic properties, and therapeutic use in
immunoregulatory disorders. Drugs (1993) 45 : pp 953-1040. Abstract
[137] Erkko P., Granlund H., Nuutinen M., Reitamo S., Comparison of cyclosporin
A pharmacokinetics of a new microemulsion formulation and standard oral
preparation in patients with psoriasis. Br J Dermatol (1997) 136 : pp 82-88.
Abstract
[138] Bourke J.F., Berth-Jones J., Holder J., Graham-Brown R.A., A new
microemulsion formulation of cyclosporin (Neoral) is effective in the treatment
of cyclosporin-resistant dermatoses. Br J Dermatol (1996) 134 : pp 777-779.
Abstract
[139] Ho V., Cloutier M.A.R., Gratton D., Neoral in the treatment of psoriasis:
consensus treatment guidelines. J Cutan Med Surg (1997) 1 : pp 209-215.
[140] Gulliver W.P., Murphy G.F., Hannaford V.A., Primmett D.R., Increased
bioavailability and improved efficacy, in severe psoriasis, of a new
microemulsion formulation of cyclosporin. Br J Dermatol (1996) 48 : pp 35-39.
Abstract
[141] Liu J., FK506 and cyclosporin, molecular probes for studying intracellular
signal transduction. Immunol Today (1993) 14 : pp 290-295. Abstract
[142] Wong R.L., Winslow C.M., Cooper K.D., The mechanisms of action of
cyclosporin A in the treatment of psoriasis [review]. Immunol Today (1993) 14 :
pp 69-74. Abstract
[143] Pei Y., Chronic cyclosporine nephrotoxicity in rheumatoid arthritis. J
Rheumatol (1996) 23 : pp 4-5. Citation
[144] Sheikh-Hamad D., Nadkarni V., Choi Y.J., Cyclosporine A inhibits the
adaptive responses to hypertonicity: a potential mechanism of nephrotoxicity. J
Am Soc Nephrol (2001) 12 : pp 2732-2741. Full Text
[145] Feutren G., Mihatsch M., Risk factors for cyclosporine-induced nephropathy
in patients with autoimmune diseases. N Engl J Med (1992) 326 : pp 1654-1660.
Abstract
[146] Lowe N.J., Wieder J.M., Rosenbach A., Long-term low-dose cyclosporine
therapy for psoriasis: effects on renal function and structure. J Am Acad
Dermatol (1996) 35 : pp 710-719. Full Text
[147] Pei Y., Scholey J.W., Katz A., Schachter R., Murphy G.F., Cattran D.,
Chronic nephrotoxicity in psoriatic patients treated with low-dose cyclosporine.
Am J Kidney Dis (1994) 23 : pp 528-536. Abstract
[148] Powles A.V., Cook T., Hulme B., Renal function and biopsy findings after 5
years' treatment with low-dose cyclosporin for psoriasis. Br J Dermatol (1993)
128 : pp 159-165. Abstract
[149] Feutren G., Abeywickrama K., Friend D., von Graffenried B., Renal function
and blood pressure in psoriatic patients treated with cyclosporin A. Br J
Dermatol (1990) 122 : pp 57-69. Abstract
[150] Rodicio J.L., Calcium antagonists and renal protection from cyclosporine
nephrotoxicity: long-term trial in renal transplantation patients. J Cardiovasc
Pharmacol (2000) 35 : pp S7-S11. Abstract
[151] Asai T., Nakatani T., Yamanaka S., Magnesium supplementation prevents
experimental chronic cyclosporine a nephrotoxicity via renin-angiotensin system
independent mechanism. Transplantation (2002) 74 : pp 784-791. Abstract
[152] Thurman R.G., Zhong Z., von Frankenberg M., Stachlewitz R.F., Bunzendahl
H., Prevention of cyclosporine-induced nephrotoxicity with dietary glycine.
Transplantation (1997) 63 : pp 1661-1667. Abstract
[153] Andoh T.F., Gardner M.P., Bennett W.M., Protective effects of dietary
L-arginine supplementation on chronic cyclosporine nephrotoxicity.
Transplantation (1997) 64 : pp 1236-1240. Abstract
[154] Zackheim H.S., Cyclosporine-associated lymphoma [letter]. J Am Acad
Dermatol (1999) 40 : pp 1015-1016. Full Text
[155] Koo J., Lebwohl M., Reply. J Am Acad Dermatol (1999) 40 : pp 1016-.
[156] Paul C.F., Ho V.C., McGeown C., Risk of malignancies in psoriasis patients
treated with cyclosporine: a 5 y cohort study. J Invest Dermatol (2003) 120 : pp
211-216. Abstract
[157] van de Kerhof P.C.M., de Rooij M.J.M., Multiple squamous cell carcinomas
in a psoriatic patient following high-dose photochemotherapy and cyclosporin
treatment: response to long-term acitretin maintenance. Br J Dermatol (1997) 136
: pp 275-278. Abstract
[158] Shupack J., Abel E., Bauer E., Cyclosporine as maintenance therapy in
patients with severe psoriasis. J Am Acad Dermatol (1997) 36 : pp 423-432. Full
Text
[159] Laburte C., Grossman R., Abi-Rached J., Abeywickrama K.H., Dubertret L.,
Efficacy and safety of oral cyclosporin A (CyA; Sandimmun) for long-term
treatment of chronic severe plaque psoriasis. Br J Dermatol (1994) 130 : pp
366-375. Abstract
[160] Mahrle G., Schulze H.J., Brautigam M., Anti-inflammatory efficacy of
low-dose cyclosporin A in psoriatic arthritis. A prospective multicentre study.
Br J Dermatol (1996) 135 : pp 752-757. Abstract
[161] Ho V.C., Griffiths C.E., Berth-Jones J., Intermittent short courses of
cyclosporine microemulsion for the long-term management of psoriasis: a 2-year
cohort study. J Am Acad Dermatol (2001) 44 : pp 643-651. Full Text
[162] Camp R.D., Reitamo S., Friedmann P.S., Ho V., Heule F., Cyclosporin A in
severe, therapy-resistant atopic dermatitis: report of an international
workshop. Br J Dermatol (1993) 129 : pp 127-220.
[163] Berth-Jones J., Graham-Brown R.A., Marks R., Long-term efficacy and safety
of cyclosporin in severe adult atopic dermatitis. Br J Dermatol (1997) 136 : pp
76-81. Abstract
[164] Czech W., Brautigam M., Weidinger G., Schopf E., A body-weight-independent
dosing regimen of cyclosporine microemulsion is effective in severe atopic
dermatitis and improves the quality of life. J Am Acad Dermatol (2000) 42 : pp
653-659. Full Text
[165] Granlund H., Erkko P., Sinisalo M., Reitamo S., Cyclosporin in atopic
dermatitis: time to relapse and effect of intermittent therapy. Br J Dermatol
(1995) 132 : pp 106-112. Abstract
[166] Granlund H., Erkko P., Eriksson E., Reitamo S., Comparison of cyclosporine
and topical betamethasone-17, 21-diproprionate in the treatment of severe
chronic hand eczema. Acta Derm Venereol (1996) 76 : pp 371-376. Abstract
[167] Lim K.K., Su W.P., Schroeter A.L., Sabers C.J., Abraham R.T., Pittelkow
M.R., Cyclosporine in the treatment of dermatologic disease: an update. Mayo
Clin Proc (1996) 71 : pp 1182-1191. Abstract
[168] Ruzicka T., Cyclosporin in less common immune-mediated skin diseases
[review]. Br J Dermatol (1996) 48 : pp 40-42. Abstract
[169] Capella G.L., Casa-Alberighi O.D., Finzi A.F., Therapeutic concepts in
clinical dermatology: cyclosporine A in immunomediated and other dermatoses. Int
J Dermatol (2001) 40 : pp 551-561. Citation
[170] Levell N.J., Munro C.S., Marks J.M., Severe lichen planus clears with very
low-dose cyclosporin [letter]. Br J Dermatol (1992) 127 : pp 66-67. Citation
[171] Mobini N., Padilla T., Jr, Ahmed A.R., Long-term remission in selected
patients with pemphigus vulgaris treated with cyclosporine. J Am Acad Dermatol
(1997) 36 : pp 264-266. Full Text
[172] Chrysomallis F., Ioannides D., Teknetzis A., Panagiotidou D., Minas A.,
Treatment of oral pemphigus vulgaris. Int J Dermatol (1994) 33 : pp 803-807.
Abstract
[173] Ioannides D., Chrysomallis F., Bystryn J.C., Ineffectiveness of
cyclosporine as an adjuvant to corticosteroids in the treatment of pemphigus.
Arch Dermatol (2000) 136 : pp 868-872. Abstract
[174] Vardy D.A., Cohen A.D., Cyclosporine therapy should be considered for
maintenance of remission in patients with pemphigus [letter]. Arch Dermatol
(2001) 137 : pp 505-506. Citation
[175] Zeller V., Cohen P., Prieur A.M., Guillevin L., Cyclosporin a therapy in
refractory juvenile dermatomyositis. Experience and longterm follow-up of 6
cases. J Rheumatol (1996) 23 : pp 1424-1427. Abstract
[176] Shapiro J., Lui H., Tron V., Ho V., Systemic cyclosporine and low-dose
prednisone in the treatment of chronic severe alopecia areata: a clinical and
immunopathologic evaluation. J Am Acad Dermatol (1997) 36 : pp 114-117. Full
Text
[177] Ostrov B.E., Athreya B.H., Eichenfield A.H., Goldsmith D.P., Successful
treatment of severe cytophagic histiocytic panniculitis with cyclosporine A.
Semin Arthritis Rheum (1996) 25 : pp 404-413. Abstract
[178] Zaki I., Patel S., Reed R., Dalziel K.L., Toxic epidermal necrolysis
associated with severe hypocalcemia, and treated with cyclosporin [letter]. Br J
Dermatol (1995) 133 : pp 337-338. Citation
[179] Buckley D.A., Rogers S., Cyclosporin-responsive hidradenitis suppurativa.
J R Soc Med (1995) 88 : pp 289P-290P. Abstract
[180] Berth-Jones J., Smith S.G., Graham-Brown R.A.C., Nodular prurigo responds
to cyclosporin. Br J Dermatol (1995) 132 : pp 795-799. Abstract
[181] Berth-Jones J., Smith S.G., Graham-Brown R.A.C., Benign familial chronic
pemphigus (Hailey-Hailey disease) responds to cyclosporin. Clin Exp Dermatol
(1995) 20 : pp 70-72. Abstract
[182] Filotico R., Vena G.A., Coviello C., Angelini G., Cyclosporine in the
treatment of generalized granuloma annulare. J Am Acad Dermatol (1994) 30 : pp
487-488. Citation
[183] Ho V., Cyclosporine in the treatment of generalized granuloma annulare
[letter]. J Am Acad Dermatol (1995) 33 : pp 298-. Full Text
[184] Stinco G., Codutti R., Frattasio A., De Francesco V., Patrone P., Chronic
actinic dermatitis treated with cyclosporine-A. Eur J Dermatol (2002) 12 : pp
455-457. Abstract
[185] Kiyohara A., Takamori K., Niizuma N., Ogawa H., Successful treatment of
severe recurrent Reiter's syndrome with cyclosporine. J Am Acad Dermatol (1997)
36 : pp 482-483. Full Text
[186] Herr H., Koh J.K., Eosinophilic cellulitis (Wells' syndrome) successfully
treated with low-dose cyclosporine. J Korean Med Sci (2001) 16 : pp 664-668.
Abstract
[187] von den Driesch P., Steffan C., Zobe A., Hornstein O.P., Sweet's
syndrome—therapy with cyclosporin. Clin Exp Dermatol (1994) 19 : pp 274-277.
Abstract
[188] Berth-Jones J., Voorhees J.J., Consensus conference on cyclosporin A
microemulsion for psoriasis. Br J Dermatol (1996) 135 : pp 775-777. Citation
[189] Lebwohl M., Ellis C., Gottlieb A., Cyclosporine consensus conference: with
emphasis on the treatment of psoriasis. J Am Acad Dermatol (1998) 39 : pp
464-475. Full Text
[190] Ransom J.T., Mechanism of action of mycophenolate mofetil. Ther Drug Monit
(1995) 17 : pp 681-684. Abstract
[191] Parker G., Bullingham R., Kamm B., Hale M., Pharmacokinetics of oral
mycophenolate mofetil in volunteer subjects with varying degrees of hepatic
oxidative impairment. J Clin Pharmacol (1996) 36 : pp 332-344. Abstract
[192] Langman L.J., Shapiro A.M., Lakey J.R., LeGatt D.F., Kneteman N.M.,
Yatscoff R.W., Pharmacodynamic assessment of mycophenolic acid-induced
immunosuppression by measurement of inosine monophosphate dehydrogenase activity
in a canine model. Transplantation (1996) 61 : pp 87-92. Abstract
[193] Bullingham R., Monroe S., Nicholls A., Hale M., Pharmacokinetics and
bioavailability of mycophenolate mofetil in healthy subjects after single-dose
oral and intravenous administration. J Clin Pharmacol (1996) 36 : pp 315-324.
Abstract
[194] Kitchin J.E., Pomeranz M.K., Pak G., Washenik K., Shupack J.L.,
Rediscovering mycophenolic acid: a review of its mechanism, side effects, and
potential uses. J Am Acad Dermatol (1997) 37 : pp 445-449. Full Text
[195] Morris R.E., Mechanisms of action of new immunosuppressive drugs. Ther
Drug Monit (1995) 17 : pp 564-569. Abstract
[196] Allison A.C., Eugui E.M., Immunosuppressive and other effects of
mycophenolic acid and an ester prodrug, mycophenolate mofetil. Immunol Rev
(1993) 136 : pp 5-28. Citation
[197] Eugui E.M., Allison A.C., Immunosuppressive activity of mycophenolate
mofetil. Ann N Y Acad Sci (1993) 685 : pp 309-329. Citation
[198] Allison A.C., Eugui E.M., Mycophenolate mofetil and its mechanisms of
action. Immunopharmacology (2000) 47 : pp 85-118. Abstract
[199] Lipsky J.J., Mycophenolate mofetil. Lancet (1996) 348 : pp 1357-1359.
Citation
[200] Yamani M.H., Starling R.C., Goormastic M., The impact of routine
mycophenolate mofetil drug monitoring on the treatment of cardiac allograft
rejection. Transplantation (2000) 69 : pp 2326-2330. Abstract
[201] CellCept (mycophenolate mofetil) [package insert] 1997. Mississauga
(Canada): Hoffmann–La Roche Ltd.
[202] Riskalla M.M., Somers E.C., Fatica R.A., McCune W.J., Tolerability of
mycophenolate mofetil in patients with systemic lupus erythematosus. J Rheumatol
(2003) 30 : pp 1508-1512. Abstract
[203] Maes B.D., Dalle I., Geboes K., Erosive enterocolitis in mycophenolate
mofetil-treated renal-transplant recipients with persistent afebrile diarrhea.
Transplantation (2003) 75 : pp 665-672. Abstract
[204] Behrend M., Adverse gastrointestinal effects of mycophenolate mofetil:
aetiology, incidence and management. Drug Saf (2001) 24 : pp 645-663. Abstract
[205] Simmons W.D., Preliminary risk–benefit assessment of mycophenolate mofetil
in transplant rejection. Drug Saf (1997) 17 : pp 75-92. Abstract
[206] Sterneck M., Fischer L., Gahlemann C., Gundlach M., Rogiers X., Broelsch
C., Mycophenolate mofetil for prevention of liver allograft rejection: initial
results of a controlled clinical trial. Ann Transplant (2000) 5 : pp 43-46.
Abstract
[207] CellCept (mycophenolate mofetil) 2003. Hoffmann–La Roche Ltd.
[208] Triemer H.L., Pearson T.C., Odom K.L., Larsen C.P., Analysis of a
single-center experience with mycophenolate mofetil based immunosuppression in
renal transplantation. Clin Transplant (2000) 14 : pp 413-420. Abstract
[209] Mycophenolate Mofetil Acute Renal Rejection Study Group Mycophenolate
mofetil for the treatment of a first acute renal allograft rejection: three-year
follow-up. Transplantation (2001) 71 : pp 1091-1097.
[210] Baudard M., Vincent A., Moreau P., Kergueris M.F., Harousseau J.L.,
Milpied N., Mycophenolate mofetil for the treatment of acute and chronic GVHD is
effective and well tolerated but induces a high risk of infectious
complications: a series of 21 BM or PBSC transplant patients. Bone Marrow
Transplant (2002) 30 : pp 287-295. Abstract
[211] ter Meulen C.G., Wetzels J.F., Hilbrands L.B., The influence of
mycophenolate mofetil on the incidence and severity of primary cytomegalovirus
infections and disease after renal transplantation. Nephrol Dial Transplant
(2000) 15 : pp 711-714. Abstract
[212] Sarmiento J.M., Dockrell D.H., Schwab T.R., Munn S.R., Paya C.V.,
Mycophenolate mofetil increases cytomegalovirus invasive organ disease in renal
transplant patients. Clin Transplant (2000) 14 : pp 136-138. Abstract
[213] Hambach L., Stadler M., Dammann E., Ganser A., Hertenstein B., Increased
risk of complicated CMV infection with the use of mycophenolate mofetil in
allogeneic stem cell transplantation. Bone Marrow Transplant (2002) 29 : pp
903-906. Abstract
[214] Enk A.H., Knop J., Treatment of pemphigus vulgaris with mycophenolate
mofetil [letter]. Lancet (1997) 350 : pp 494-. Citation
[215] Bohm M., Beissert S., Schwarz T., Metze D., Luger T., Bullous pemphigoid
treated with mycophenolate mofetil [letter]. Lancet (1997) 349 : pp 541-.
Citation
[216] Popovsky J.L., Camisa C., New and emerging therapies for diseases of the
oral cavity. Dermatol Clin (2000) 18 : pp 113-125. Full Text
[217] Grundmann-Kollmann M., Kaskel P., Leiter U., Treatment of pemphigus
vulgaris and bullous pemphigoid with mycophenolate mofetil monotherapy [letter].
Arch Dermatol (1999) 135 : pp 724-725. Citation
[218] Nousari H.C., Sragovich A., Kimyai-Asadi A., Orlinsky D., Anhalt G.J.,
Mycophenolate mofetil in autoimmune and inflammatory skin disorders. J Am Acad
Dermatol (1999) 40 : pp 265-268. Full Text
[219] Nousari H.C., Griffin W.A., Anhalt G.J., Successful therapy for bullous
pemphigoid with mycophenolate mofetil. J Am Acad Dermatol (1998) 39 : pp
497-498. Full Text
[220] Bredlich R.O., Grundmann-Kollmann M., Behrens S., Kerscher M., Peter R.U.,
Mycophenolate mofetil monotherapy for pemphigus vulgaris [letter]. Br J Dermatol
(1999) 141 : pp 934-. Citation
[221] Enk A.H., Knop J., Mycophenolate is effective in the treatment of
pemphigus vulgaris. Arch Dermatol (1999) 135 : pp 54-56. Abstract
[222] Katz K.H., Marks J.G., Jr, Helm K.F., Pemphigus foliaceus successfully
treated with mycophenolate mofetil as a steroid-sparing agent. J Am Acad
Dermatol (2000) 42 : pp 514-515. Full Text
[223] Nousari H.C., Goyal S., Anhalt G.J., Successful treatment of resistant
hypertrophic and bullous lichen planus with mycophenolate mofetil [letter,
comment]. Arch Dermatol (1999) 135 : pp 1420-1421. Citation
[224] Glaser R., Sticherlin M., Successful treatment of linear IgA bullous
dermatosis with mycophenolate mofetil [letter]. Acta Derm Venereol (2002) 82 :
pp 308-309. Citation
[225] Megahed M., Schmiedeberg S., Becker J., Ruzicka T., Treatment of
cicatricial pemphigoid with mycophenolate mofetil as a steroid-sparing agent. J
Am Acad Dermatol (2001) 45 : pp 256-259. Full Text
[226] Williams J.V., Marks J.G., Jr, Billingsley E.M., Use of mycophenolate
mofetil in the treatment of paraneoplastic pemphigus. Br J Dermatol (2000) 142 :
pp 506-508. Abstract
[227] Schattenkirchner S., Eming S., Hunzelmann N., Krieg T., Smola H.,
Treatment of epidermolysis bullosa acquisita with mycophenolate mofetil and
autologous keratinocyte grafting [letter]. Br J Dermatol (1999) 141 : pp
932-933. Citation
[228] Trebing D., Ziemer A., [Acquired epidermolysis bullosa with a highly
varied clinical picture and successful treatment with mycophenolate mofetil].
Hautarzt (2001) 52 : pp 717-721. Abstract
[229] Powell A.M., Albert S., Al Fares S., An evaluation of the usefulness of
mycophenolate mofetil in pemphigus. Br J Dermatol (2003) 149 : pp 138-145.
Abstract
[230] Lynch W., Roenigk H.H.J., Mycophenolic acid in psoriasis. Arch Dermatol
(1977) 113 : pp 1203-1208. Abstract
[231] Haufs M.G., Beissert S., Grabbe S., Shutte B., Luger T.A., Psoriasis
vulgaris treated successfully with mycophenolate mofetil. Br J Dermatol (1998)
138 : pp 179-181. Abstract
[232] Grundmann-Kollmann M., Mooser G., Schraeder P., Treatment of chronic
plaque-stage psoriasis and psoriatic arthritis with mycophenolate mofetil. J Am
Acad Dermatol (2000) 42 : pp 835-837. Full Text
[233] Geilen C.C., Arnold M., Orfanos C.E., Mycophenolate mofetil as a systemic
antipsoriatic agent: positive experience in 11 patients. Br J Dermatol (2001)
144 : pp 583-586. Abstract
[234] Zhou Y., Rosenthal D., Dutz J., Ho V., Mycophenolate mofetil (CellCept(R))
for psoriasis: a two-center, prospective, open-label clinical trial. J Cutan Med
Surg (2003) 7 : pp 193-197. Abstract
[235] Michel S., Hohenleutner U., Mohr V., Landthaler M., [Therapy-resistant
pyoderma gangrenosum—treatment with mycophenolate mofetil and cyclosporine A].
[in German] Hautarzt (1999) 50 : pp 428-431. Abstract
[236] Hohenleutner U., Mohr V.D., Michel S., Landthaler M., Mycophenolate
mofetil and cyclosporin treatment for recalcitrant pyoderma gangrenosum
[letter]. Lancet (1997) 350 : pp 1748-. Citation
[237] Nousari H.C., Lynch W., Anhalt G.J., Petri M., The effectiveness of
mycophenolate mofetil in refractory pyoderma gangrenosum. Arch Dermatol (1998)
134 : pp 1509-1511. Citation
[238] Gilmour E., Stewart D.G., Severe recalcitrant pyoderma gangrenosum
responding to a combination of mycophenolate mofetil with cyclosporin and
complicated by a mononeuritis. Br J Dermatol (2001) 144 : pp 397-400. Abstract
[239] Grundmann-Kollmann M., Korting H.C., Behrens S., Successful treatment of
severe refractory atopic dermatitis with mycophenolate mofetil [letter]. Br J
Dermatol (1999) 141 : pp 175-176. Citation
[240] Neuber K., Schwartz I., Itschert G., Dieck A.T., Treatment of atopic
eczema with oral mycophenolate mofetil. Br J Dermatol (2000) 143 : pp 385-391.
Abstract
[241] Grundmann-Kollmann M., Podda M., Ochsendorf F., Boehncke W.H., Kaufmann
R., Zollner T.M., Mycophenolate mofetil is effective in the treatment of atopic
dermatitis. Arch Dermatol (2001) 137 : pp 870-873. Abstract
[242] Hansen E.R., Buus S., Deleuran M., Andersen K.E., Treatment of atopic
dermatitis with mycophenolate mofetil. Br J Dermatol (2000) 143 : pp 1324-1326.
Citation
[243] Pickenacker A., Luger T.A., Schwarz T., Dyshidrotic eczema treated with
mycophenolate mofetil [letter]. Arch Dermatol (1998) 134 : pp 378-379. Citation
[244] Semhoun-Ducloux S., Ducloux D., Miguet J.P., Mycophenolate mofetil-induced
dyshidrotic eczema [letter]. Ann Intern Med (2000) 132 : pp 417-. Citation
[245] Ruiz-Irastorza G., Khamashta M.A., Hughes G.R., Therapy of systemic lupus
erythematosus: new agents and new evidence. Expert Opin Invest Drugs (2000) 9 :
pp 1581-1593.
[246] Chan T.M., Li F.K., Tang C.S., Efficacy of mycophenolate mofetil in
patients with diffuse proliferative lupus nephritis. Hong Kong–Guangzhou
Nephrology Study Group. N Engl J Med (2000) 343 : pp 1156-1162. Abstract
[247] Austin H.A., Balow J.E., Treatment of lupus nephritis. Semin Nephrol
(2000) 20 : pp 265-276. Abstract
[248] Gaubitz M., Schorat A., Schotte H., Kern P., Domschke W., Mycophenolate
mofetil for the treatment of systemic lupus erythematosus: an open pilot trial.
Lupus (1999) 8 : pp 731-736. Abstract
[249] Schanz S., Ulmer A., Rassner G., Fierlbeck G., Successful treatment of
subacute cutaneous lupus erythematosus with mycophenolate mofetil. Br J Dermatol
(2002) 147 : pp 174-178. Abstract
[250] Goyal S., Nousari H.C., Treatment of resistant discoid lupus erythematosus
of the palms and soles with mycophenolate mofetil. J Am Acad Dermatol (2001) 45
: pp 142-144. Full Text
[251] Worm M., Sterry W., Kolde G., Mycophenolate mofetil is effective for
maintenance therapy of hypocomplementaemic urticarial vasculitis. Br J Dermatol
(2000) 143 : pp 1324-. Citation
[252] Nowack R., Gobel U., Klooker P., Hergesell O., Andrassy K., van der Woude
F.J., Mycophenolate mofetil for maintenance therapy of Wegener's granulomatosis
and microscopic polyangiitis: a pilot study in 11 patients with renal
involvement. J Am Soc Nephrol (1999) 10 : pp 1965-1971. Full Text
[253] Waiser J., Budde K., Braasch E., Neumayer H.H., Treatment of acute
c-ANCA-positive vasculitis with mycophenolate mofetil. Am J Kidney Dis (1999) 34
: pp e9-. Abstract
[254] Gross W.L., New concepts in treatment protocols for severe systemic
vasculitis. Curr Opin Rheumatol (1999) 11 : pp 41-46. Abstract
[255] Nowack R., Birck R., van der Woude F.J., Mycophenolate mofetil for
systemic vasculitis and IgA nephropathy [letter]. Lancet (1997) 349 : pp 774-.
Citation
[256] Kouba D.J., Mimouni D., Rencic A., Nousari H.C., Mycophenolate mofetil may
serve as a steroid-sparing agent for sarcoidosis. Br J Dermatol (2003) 148 : pp
147-148. Abstract
[257] Reinhard G., Lohmann F., Uerlich M., Bauer R., Bieber T., Successful
treatment of ulcerated necrobiosis lipoidica with mycophenolate mofetil
[letter]. Acta Derm Venereol (2000) 80 : pp 312-313. Citation
[258] Gelber A.C., Nousari H.C., Wigley F.M., Mycophenolate mofetil in the
treatment of severe skin manifestations of dermatomyositis: a series of 4 cases.
J Rheumatol (2000) 27 : pp 1542-1545. Abstract
[259] Cattaneo D., Perico N., Gaspari F., Gotti E., Remuzzi G., Glucocorticoids
interfere with mycophenolate mofetil bioavailability in kidney transplantation.
Kidney Int (2002) 62 : pp 1060-1067. Abstract
[260] Morii M., Ueno K., Ogawa A., Impairment of mycophenolate mofetil
absorption by iron ion. Clin Pharmacol Ther (2000) 68 : pp 613-616. Abstract
[261] Royer B., Zanetta G., Berard M., A neutropenia suggesting an interaction
between valacyclovir and mycophenolate mofetil. Clin Transplant (2003) 17 : pp
158-161. Abstract
[262] Cox V.C., Ensom M.H., Mycophenolate mofetil for solid organ
transplantation: does the evidence support the need for clinical pharmacokinetic
monitoring?. Ther Drug Monit (2003) 25 : pp 137-157. Abstract
[263] Krueger J.G., The immunologic basis for the treatment of psoriasis with
new biologic agents. J Am Acad Dermatol (2002) 46 : pp 1-23. Full Text
[264] Nickoloff B.J., Characterization of lymphocyte-dependent angiogenesis
using a SCID mouse: human skin model of psoriasis. J Investig Dermatol Symp Proc
(2000) 5 : pp 67-73. Abstract
[265] Nickoloff B.J., Kunkel S.L., Burdick M., Strieter R.M., Severe combined
immunodeficiency mouse and human psoriatic skin chimeras. Validation of a new
animal model. Am J Pathol (1995) 146 : pp 580-588. Abstract
[266] Bos J.D., Hagenaars C., Das P.K., Krieg S.R., Voorn W.J., Kapsenberg M.L.,
Predominance of “memory” T cells (CD4+ CDw29+) over “naïve” T cells (CD4+,
CD45R+) in both normal and diseased skin. Arch Dermatol Res (1989) 281 : pp
24-30. Abstract
[267] Austin L.M., Coven T.R., Bhardwaj N., Steinman R., Krueger J.G.,
Intraepidermal lymphocytes in psoriatic lesions are activated GMP-17(TIA-1)+
CD8+ CD3+ CTLs as determined by phenotypic analysis. J Cutan Pathol (1998) 25 :
pp 79-88. Abstract
[268] Gottlieb A.B., Psoriasis. Immunopathology and immunomodulation. Dermatol
Clin (2001) 19 : pp 1-13. Full Text
[269] Available at:Accessed November 2003 Alefacept [package insert] 2003.
Cambridge (MA): Biogen Idec.
http://www.fda.gov/cder/foi/label/2003/alefbio013003LB.htm.
[270] Chisholm P.L., Williams C.A., Jones W.E., The effects of an
immunomodulatory LFA3-IgG1 fusion protein on nonhuman primates. Ther Immunol
(1994) 1 : pp 205-216. Abstract
[271] Miller G.T., Hochman P.S., Meier W., Specific interaction of lymphocyte
function-associated antigen 3 with CD2 can inhibit T-cell responses. J Exp Med
(1993) 178 : pp 211-222. Abstract
[272] Meier W., Gill A., Rogge M., Immunomodulation by LFA3TIP, and LFA-3/IgG1
fusion protein: cell line dependent glycosylation effects on pharmacokinetics
and pharmacodynamic markers. Ther Immunol (1995) 2 : pp 159-171. Abstract
[273] Majeau G.R., Meier W., Jimmo B., Kioussis D., Hochman P.S., Mechanism of
lymphocyte function-associated molecule 3-Ig fusion proteins inhibition of T
cell responses. Structure/function analysis in vitro and in human CD2 transgenic
mice. J Immunol (1994) 152 : pp 2753-2767. Abstract
[274] Sanders M.W., Makgoba M.W., Sharrow S.O., Human memory T lymphocytes
express increased levels of three cell adhesion molecules (LFA-3, CD2, and
LFA-1) and three other molecules (UCHL1, CDw29, and Pgp-1) and have enhanced
IFN-γ production. J Immunol (1988) 140 : pp 1401-1407. Abstract
[275] Majeau G.R., Whitty A., Yim K., Meier W., Hochman P.S., Low affinity
binding of an LFA-3/IgG1 fusion protein to CD2+ T cells is independent of cell
activation. Cell Adhes Commun (1999) 7 : pp 267-269. Abstract
[276] Ellis C.N., Krueger G.G., Treatment of chronic plaque psoriasis by
selective targeting of memory effector T lymphocytes. N Engl J Med (2001) 345 :
pp 248-255. Abstract
[277] Gottlieb A.B., Bos J.D., Recombinantly engineered human proteins:
transforming the treatment of psoriasis. Clin Immunol (2002) 105 : pp 105-116.
Abstract
[278] Gottlieb A.B., Casale T., Frankel E., CD4+ T-cell-directed antibody
responses are maintained in patients with psoriasis receiving alefacept: results
of a randomized study. J Am Acad Dermatol (2003) 49 : pp 816-825. Full Text
[279] Krueger J.G., Gilleaudeau P., Kikuchi T., Lee E., Psoriasis-related
subpopulations of memory CD4+ and CD8+ T cells are selectively reduced by
alefacept. J Invest Dermatol (2002) 119 : pp 823-.
[280] Lowe N., Gonzalez J., Bagel J., Caro I., Ellis C., Menter A., Repeat
courses of intravenous alefacept in patients with chronic plaque psoriasis
provide consistent safety and efficacy. Int J Dermatol (2003) 42 : pp 224-230.
Abstract
[281] Akhavan A., Lebwohl M., Systemic psoriasis treatment with alefacept.
Psoriasis Forum (2003) 9 : pp 4-5.
[282] Lebwohl M., Enno C., Langley R., An international, randomized,
double-blind, placebo-controlled phase 3 trial of intramuscular alefacept in
patients with chronic plaque psoriasis. Arch Dermatol (2003) 139 : pp 719-727.
Abstract
[283] Krueger G.G., Papp K.A., Stough D.B., Loven K.H., Gulliver W.P., Ellis
C.N., A randomized, double-blind, placebo-controlled phase III study evaluating
efficacy and tolerability of 2 courses of alefacept in patients with chronic
plaque psoriasis. J Am Acad Dermatol (2002) 47 : pp 821-833. Full Text
[284] Spuls P.I., Witkamp L., Bossuyt P.M., Bos J.D., A systematic review of
five systemic treatments for severe psoriasis. Br J Dermatol (1997) 137 : pp
943-949. Abstract
[285] Koo J., Lebwohl M., Duration of remission of psoriasis therapies. J Am
Acad Dermatol (1999) 41 : pp 51-59. Full Text
[286] Menter A., Cram D.L., The Goeckerman regimen in two psoriasis day care
centers. J Am Acad Dermatol (1983) 9 : pp 59-65. Abstract
[287] Krueger G.G., Ellis C.N., Alefacept therapy produces remission for
patients with chronic plaque psoriasis. Br J Dermatol (2003) 148 : pp 784-788.
Abstract
[288] Krueger G.G., Callis K.P., Development and use of alefacept to treat
psoriasis. J Am Acad Dermatol (2003) 49 : pp S87-S97. Full Text
[289] Available at:Accessed October 2003 Efalizumab [package insert] 2003. South
San Francisco (CA): Genentech, Inc.
http://www.google.com/search?q=cache:fCZgcnAKE6IJ:www.bbriefings.com/pdf/790/genentech.pdf+efalizumab+package+insert&hl=en&ie=UTF-8.
[290] Grakoui A., Bromley S.K., Sumen C., The immunological synapse: a molecular
machine controlling T cell activation. Science (1999) 285 : pp 221-227. Abstract
[291] Sanders M.E., Makgoba M.W., Sharrow S.O., Human memory T lymphocytes
express increased levels of three cell adhesion molecules (LFA-3, CD2, and
LFA-1) and three other molecules (UCHL1, CDw29, and Pgp-1) and have enhanced
IFN-γ production. J Immunol (1988) 140 : pp 1401-1407. Abstract
[292] Van Seventer G.A., Shimizu Y., Horgan K.J., Ginter Luce G.E., Webb D.,
Shaw S., Remote T cell costimulation via LFA-1/ICAM-1 and CD2/LFA-3:
demonstration with immobilized ligand/mAb and in monocyte-mediated
costimulation. Eur J Immunol (1991) 21 : pp 1711-. Abstract
[293] Nickoloff B.J., Mitra R.S., Green J., Accessory cell function of
keratinocytes for superantigens. J Immunol (1993) 150 : pp 2148-2159. Abstract
[294] Lebwohl M., Tyring S.K., Hamilton T.K., A novel targeted T-cell modulator,
efalizumab, for plaque psoriasis. N Engl J Med (2003) 349 : pp 2004-2013.
Abstract
[295] Leonardi C.L., Efalizumab: an overview. J Am Acad Dermatol (2003) 49 : pp
S98-S104. Full Text
[296] Lebwohl M., Papp K.A., Tyring S., et al. Continued treatment with
subcutaneous efalizumab is safe: pooled results from two phase III trials.
Presented at the American Academy of Dermatology. New York, July 31–August 4,
2002
[297] Nussbaum R., Krueger J.G., Treatment of inflammatory dermatoses with novel
biologic agents: a primer. Adv Dermatol (2002) 18 : pp 45-89. Citation
[298] Weinberg J.M., Saini R., Biologic therapy for psoriasis: the tumor
necrosis factor inhibitors—infliximab and etanercept. Cutis (2003) 71 : pp
25-29. Abstract
[299] Goffe B., Cather J.C., Etanercept: an overview. J Am Acad Dermatol (2003)
49 : pp S105-S111. Full Text
[300] Mussi A., Bonifati C., Carducci M., Serum TNF-alpha levels correlate with
disease severity and are reduced by effective therapy in plaque-type psoriasis.
J Biol Regul Homeost Agents (1997) 11 : pp 115-118. Abstract
[301] Bonifati C., Carducci M., Cordiali F.P., Correlated increases of tumour
necrosis factor-alpha, interleukin-6 and granulocyte monocyte-colony stimulating
factor levels in suction blister fluids and sera of psoriatic
patients—relationships with disease severity. Clin Exp Dermatol (1994) 19 : pp
383-387. Abstract
[302] Available at:Accessed December 2003 Etanercept [package insert] 2003.
Thousand Oaks (CA): Amgen.
http://www.google.com/search?q=cache:6CgATD0-sNEJ:www.fda.gov/cder/foi/label/2003/etanimm060503LB.pdf+Etanercept+package+insert&hl=en&ie=UTF-8.
[303] Moreland L., Cohen S., Fleischmann R.M., et al. Safety and efficacy of up
to five years etanercept (Enbrel) therapy in rheumatoid arthritis. Presented at
European League Against Rheumatism. June 12–15, 2002, Stockholm, Sweden
[304] Leonardi C.L., Powers J.L., Matheson R.T., Etanercept as monotherapy in
patients with psoriasis. N Engl J Med (2003) 349 : pp 2014-2022. Abstract
[305] Mease P.J., Goffe B.S., Metz J., VanderStoep A., Finck B., Burge D.J.,
Etanercept in the treatment of psoriatic arthritis and psoriasis: a randomized
trial. Lancet (2000) 356 : pp 385-390. Abstract
[306] Weinblatt M.E., Kremer J.M., Bankhurst A.D., Trial of etanercept, a
recombinant tumor necrosis factor receptor: Fc fusion protein, in patients with
rheumatoid arthritis receiving methotrexate. N Engl J Med (1999) 340 : pp
253-259. Abstract
[307] Moreland L.W., Schiff M.H., Baumgartner S.W., Tindall E.A., Fleischmann
R.M., Bulpitt K.J., Etanercept theapy in rheumatoid arthritis: a randomized,
controlled trial. Ann Intern Med (1999) 130 : pp 478-486. Abstract
[308] Shakoor N., Michalska M., Harris C.A., Block J.A., Drug-induced systemic
lupus erythematosus associated with etanercept therapy. Lancet (2002) 359 : pp
579-580. Abstract
[309] Bleumink G.S., ter Borg E.J., Ramselaar C.G., Stricker B.H., Etanercept
induced subacute cuatneous lupus erythematosus. Rheumatology [Oxford] (2001) 40
: pp 1317-1319. Citation
[310] Lebwohl M., Psoriasis. Lancet (2003) 361 : pp 1197-1204. Abstract
[311] Mohan N., Edwards E.T., Cupps T.R., Demyelination occurring during
anti-tumor necrosis factor α therapy for inflammatory arthritides. Arthritis
Rheum (2001) 44 : pp 2862-2869. Abstract
[312] Zeltser R., Valle L., Tanck C., Holyst M.M., Ritchlin C., Gaspari A.A.,
Clinical, histological, and immunophenotypic characteristics of injection site
reactions associated with etanercept. Arch Dermatol (2001) 137 : pp 893-899.
Abstract
[313] Gottlieb A.B., Matheson R.T., Lowe N., A randomized trial of etanercept as
monotherapy for psoriasis. Arch Dermatol (2003) 139 : pp 1627-1632. Abstract
[314] Brockbank J., Gladman D.D., Psoriatic arthritis. Expert Opin Investig
Drugs (2000) 9 : pp 1511-1522. Abstract
[315] Ellman M.I.I., MacDonald P.A., Hayes F.A., Etanercept as treatment for
diffuse scleroderma: a pilot study. Arthritis Rhem (2000) 43 : pp S392-.
[316] Sacher C., Rubbert A., König C., Scharffetter-Kochanek K., Krieg T.,
Hunzelmann N., Treatment of recalcitrant cicatricial pemphigoid with tumor
necrosis factor α antagonist etanercept. J Am Acad Dermatol (2002) 46 : pp
113-115. Full Text
[317] Smith K.J., Skelton H., Common variable immunodeficiency treated with a
recombinant human IgG, tumour necrosis factor-α receptor fusion protein. Br J
Dermatol (2001) 144 : pp 597-600. Abstract
[318] Robinson N.D., Guitart J., Recalcitrant, recurrent aphthous stomatitis
treated with etanercept. Arch Dermatol (2003) 139 : pp 1259-1261. Citation
[319] Gottlieb A.B., Masud S., Ramamurthi R., Pharmacodynamic and
pharmacokinetic response to anti-tumor necrosis factor-α monoclonal antibody
(infliximab) treatment of moderate to severe psoriasis vulgaris. J Am Acad
Dermatol (2003) 48 : pp 1-11.
[320] Scallon B.J., Arevalo Moore M., Trinh H., Knight D.M., Ghrayeb J.,
Chimeric anti-TNF-α monoclonal antibody cA2 binds recombinant transmembrane TNF-
α and activates immune effector functions. Cytokine (1995) 7 : pp 251-259.
Abstract
[321] Gottlieb A.B., Infliximab for psoriasis. J Am Acad Dermatol (2003) 49 : pp
S112-S117. Full Text
[322] Barnes P.J., Karin M., Nuclear factor-kappaB: a pivotal transcription
factor in chronic inflammatory disease. N Engl J Med (1997) 336 : pp 1066-1072.
Citation
[323] Ettehadi P., Greaves M.W., Wallach D., Aderka D., Camp R.D.R., Elevated
tumor necrosis factor-alpha (TNF-alpha) biological activity in psoriatic skin
lesions. Clin Exp Immunol (1994) 96 : pp 146-151. Abstract
[324] Nahar I.K., Shojania K., Marra C.A., Alamgir A.H., Anis A.H., Infliximab
treatment of rheumatoid arthritis and Crohn's disease. Ann Pharmacother (2003)
37 : pp 1256-1265. Abstract
[325] Anis C.N., Blanchard J.F., Kliewer E., Wajda A., Cancer risk in patients
with inflammatory bowel disease: a population-based study. Cancer (2001) 91 : pp
854-862. Abstract
[326] Greenstein A.J., Gennuso R., Sachar D.B., Heimann T., Janowitz H.D.,
Aufses A.H., Jr, Extraintestinal cancers in inflammatory bowel disease. Cancer
(1985) 56 : pp 2914-2921. Abstract
[327] Baecklund E., Sundstrom C., Ekbom A., Lymphoma subtypes in patients with
rheumatoid arthritis: increased proportion of diffuse large B cell lymphoma.
Arthritis Rheum (2003) 48 : pp 1543-1550. Abstract
[328] Kamel O.W., Holly E.A., van de Rijn M., Lele C., Sah A., A population
based, case-control study of non-Hodgkin's lymphoma in patients with rheumatoid
arthritis. J Rheumatol (1999) 26 : pp 1676-1680. Abstract
[329] Gridley G., McLaughlin J.K., Ekbom A., Incidence of cancer among patients
with rheumatoid arthritis. J Natl Cancer Inst (1993) 85 : pp 307-311. Abstract
[330] Thomas E., Brewster D.H., Black R.J., Macfarlane G.J., Risk of malignancy
among patients with rheumatic conditions. Int J Cancer (2000) 88 : pp 497-502.
Abstract
[331] Hannuksela-Svahn A., Pukkala E., Läärä E., Poikolainen K., Karonen J.,
Psoriasis, its treatment, and cancer in a cohort of Finnish patients. J Invest
Dermatol (2000) 114 : pp 587-590. Abstract
[332] Margolis D., Bilker W., Hennessy S., Vittorio C., Santanna J., Strom B.L.,
The risk of malignancy associated with psoriasis. Arch Dermatol (2001) 137 : pp
778-783. Abstract
[333] Gelfand J.M., Berlin J., Voorhees A.V., Margolis D.J., Lymphoma rates are
low but increased in patients with psoriasis. Arch Dermatol (2003) 139 : pp
1425-1429. Abstract
[334] Gross T.G., Steinbuch M., DeFor T., B cell lymphoproliferative disorders
following hematopoietic stem cell transplantation: risk factors, treatment and
outcome. Bone Marrow Transplant (1999) 23 : pp 251-258. Abstract
[335] Okano M., Gross T.G., A review of Epstein-Barr virus infection in patients
with immunodeficiency disorders. Am J Med Sci (2000) 319 : pp 392-396. Abstract
[336] Keane J., Gershon S., Wise R.P., Tuberculosis associated with infliximab,
a tumor necrosis factor α-neutralizing agent. N Engl J Med (2001) 345 : pp
1098-1104. Abstract
[337] Lee J.-H., Slifman N.R., Gershon S.K., Life-threatening histoplasmosis
complicating immunotherapy with tumor necrosis factor α antagonists infliximab
and etanercept. Arthritis Rheum (2002) 46 : pp 2565-2570. Abstract
[338] Hanauer S.B., Feagan B.G., Lichtenstein G.R., Maintenance infliximab for
Crohn's disease: the ACCENT I randomized trial. Lancet (2002) 359 : pp
1541-1549. Abstract
[339] Maini R., St. Clair E.W., Breedveld F., for the ATTRACT Study Group
Infliximab (chimeric anti-tumor necrosis factor alpha monoclonal antibody)
versus placebo in rheumatoid arthritis patients receiving concomitant
methotrexate: a randomized phase III trial. Lancet (1999) 354 : pp 1932-1939.
Abstract
[340] Chaudhari U., Romano P., Mulcahy L.D., Dooley L.T., Baker D.G., Gottlieb
A.B., Efficacy and safety of infliximab monotherapy for plaque-type psoriasis: a
randomised trial. Lancet (2001) 357 : pp 1842-1847. Abstract
[341] Available at:Accessed 2003 Infliximab [package insert] 2003. Malvern (PA):
Centocor, Inc. http://www.remicade.com/PI/interactive_PI.jsp.
[342] Chung E.S., Packer M., Lo K.H., Fasanmade A.A., Willerson J.T.,
Randomized, double-blind, placebo-controlled, pilot trial of infliximab, a
chimeric monoclonal antibody to tumor necrosis factor-α, in patients with
moderate-to severe heart failure. Circulation (2003) 107 : pp 3133-3140.
Abstract
[343] Oh C.J., Das K.M., Gottlieb A.B., Treatment with anti-tumor necrosis
factor α (TNF-α) monoclonal antibody dramatically decreases the clinical
activity of psoriasis lesions. J Am Acad Dermatol (2000) 42 : pp 829-830. Full
Text
[344] Tan M.-H., Gordon M., Lebwohl O., George J., Lebwohl M., Improvement of
pyoderma gangrenosum and psoriasis associated with Crohn disease with anti-tumor
necrosis factor α monoclonal antibody. Arch Dermatol (2001) 137 : pp 930-933.
Abstract
[345] O'Quinn R.P., Miller J.L., The effectiveness of tumor necrosis factor α
antibody (infliximab) in treating recalcitrant psoriasis. Arch Dermatol (2002)
138 : pp 644-648. Abstract
[346] Ogilvie A.L.J., Antoni C., Dechant C., Treatment of psoriatic arthritis
with antitumour necrosis factor-α antibody clears skin lesions of psoriasis
resistant to treatment with methotrexate. Br J Dermatol (2001) 144 : pp 587-589.
Abstract
[347] Newland M.R., Weinstein A., Kerdel F., Rapid response to infliximab in
severe pustular psoriasis, von Zumbusch type. Int J Dermatol (2002) 41 : pp
449-452. Abstract
[348] Sullivan T.P., Welsh E., Kerdel F.A., Burdick A.E., Kirsner R.S.,
Infliximab for hidradenitis suppurativa. Br J Dermatol (2003) 149 : pp
1046-1049. Abstract
[349] Zaccagna A., Bertone A., Puiatti P., Anti-tumor necrosis factor alpha
monoclonal antibody (infliximab) for the treatment of pyoderma gangrenosum
associated with Crohn's disease. Eur J Dermatol (2003) 13 : pp 258-260. Abstract
[350] Kugathasan S., Miranda A., Nocton J., Drolet B.A., Raasch C., Binion D.G.,
Dermatologic manifestations of Crohn disease in children: response to
infliximab. J Pediatr Gastroenterol Nutr (2003) 37 : pp 150-154. Abstract
[351] Romero-Gomez M., Sanchez-Munoz D., Infliximab induces remission of
pyoderma gangrenosum. Eur J Gastroenterol Hepatol (2002) 14 : pp 907-. Citation
[352] Triantafillidis J.K., Cheracakis P., Sklavaina M., Apostolopoulou K.,
Favorable response to infliximab treatment in a patient with active Crohn
disease and pyoderma gangrenosum. Scand J Gastroenterol (2002) 37 : pp 863-865.
Abstract
[353] Estrach C., Mpofu S., Moots R.J., Behcet's syndrome: response to
infliximab after failure of etanercept. Rheumatology [Oxford] (2002) 41 : pp
1213-1214. Citation
[354] Saulsbury F.T., Mann J.A., Treatment with infliximab for a child with
Behcet's disease. Arthritis Rheum (2003) 49 : pp 599-600. Citation
[355] Katsiari C.G., Theodossiadis P.G., Kaklamanis P.G., Markomicheiakis N.N.,
Successful long-term treatment of refractory Adamantiades-Behcet's disease (ABD)
with infliximab: report of two patients. Adv Exp Med Biol (2003) 528 : pp
551-555. Citation
[356] Olivieri I., Padula A., Ciancio G., Salvarani C., Niccoli L., Cantini F.,
Successful treatment of SAPHO syndrome with infliximab: report of two cases. Ann
Rheum Dis (2002) 61 : pp 375-376. Citation
[357] Voigtlander C., Luftl M., Schuler G., Hertl M., Infliximab (anti-tumor
necrosis factor alpha antibody): a novel, highly effective treatment of
recalcitrant subcorneal pustular dermatosis (Sneddon-Wilkinson disease). Arch
Dermatol (2001) 137 : pp 1571-1574. Citation
[358] Fischer M., Fiedler E., Marsch W.C., Wolhlrab J., Antitumour necrosis
factor-α antibodies (infliximab) in the treatment of a patient with toxic
epidermal necrolysis. Br J Dermatol (2002) 146 : pp 707-708. Citation
[359] Yates V.M., Watkinson G., Kelman A., Further evidence for an association
between psoriasis, Crohn's disease, and ulcerative colitis. Br J Dermatol (1982)
106 : pp 323-. Abstract
[360] Targan S.R., Hanauer S.B., van Deventer S.J., A short-term study of
chimeric monoclonal antibody cA2 to tumor necrosis factor-alpha for Crohn's
disease. N Engl J Med (1997) 337 : pp 1029-1035. Abstract