throbber
The Use of Methotrexate in Rheumatoid Arthritis
`Andrea T. Borchers, Carl L. Keen, Gurtej S. Cheema, and M. Eric Gershwin
`
`Objective: To address the long-term efficacy and toxicity issues related to
`methotrexate (MTX) and compare it with other disease-modifying antirheu-
`matic drugs (DMARDs).
`Methods: Review of the international literature on the clinical use of MTX in
`rheumatoid arthritis (RA) disease.
`Results: MTX has emerged as a relatively safe and effective treatment for RA
`that compares favorably with other therapies, particularly because of its
`considerably longer median drug survival. The toxicity profile of MTX is well
`established and includes serious and sometimes fatal liver disease, pneumo-
`nitis, and cytopenias. Hence, regular and careful monitoring of patients taking
`MTX is essential, particularly when MTX is combined with other DMARDs.
`Folate supplementation can reduce some of the most common side effects of
`MTX, but it has not yet been established whether this translates into a reduced
`risk of serious disease. Another potential approach to reducing the toxicity of
`MTX is therapeutic drug monitoring and dose individualization. However,
`correlations between pharmacokinetics and clinical response have been ad-
`dressed in only a few studies and with conflicting results.
`Conclusions: MTX is an effective DMARD with a relatively safe profile
`compared with other therapies. Folate supplementation can significantly re-
`duce the risk of MTX toxicity. Finally, it is essential that patients be monitored
`carefully to reduce the potential serious toxicities of MTX.
`Semin Arthritis Rheum 34:465-483. © 2004 Elsevier Inc. All rights reserved.
`
`INDEX WORDS: Bone marrow suppression; liver toxicity; methotrexate; rheu-
`matoid arthritis.
`
`RHEUMATOID ARTHRITIS (RA) is a sys-
`
`temic autoimmune disease characterized by
`chronic inflammation of synovial joints resulting in
`destruction of cartilage and, ultimately, bone.
`There is no cure for RA, and treatment aims at
`limiting joint damage, preventing loss of function,
`and decreasing pain. The armamentarium of drugs
`used for these purposes includes nonsteroidal an-
`tiinflammatory drugs, disease modifying antirheu-
`matic drugs (DMARDs), and corticosteroids. Un-
`fortunately, joint destruction can often progress
`despite treatment, leading to deformity and disabil-
`ity in a substantial number of patients. In recent
`years, several studies have shown that a greater
`impact on slowing disease progression can be
`achieved if patients with recent-onset RA are
`treated with DMARDs earlier than had previously
`been recommended (1). Hence, the current Amer-
`ican College of Rheumatology (ACR) guidelines
`recommend initiation of DMARD treatment within
`3 months of diagnosis (2). According to these
`guidelines, methotrexate (MTX) is the standard by
`
`which new DMARDs are evaluated, and MTX as
`monotherapy, or
`in combination with other
`DMARDs, should be instituted if a satisfactory
`
`From the Division of Rheumatology, Allergy and Clinical
`Immunology, and the Department of Nutrition, University of
`California at Davis, Davis, CA.
`Andrea T. Borchers, PhD: Visiting Postdoctoral Scholar,
`Division of Rheumatology, Allergy and Clinical Immunology,
`University of California at Davis, Davis, CA; Carl L. Keen,
`PhD: Professor and Chair, Department of Nutrition, University
`of California at Davis, Davis, CA; Gurtej S. Cheema, MD:
`Assistant Clinical Professor of Medicine, Division of Rheuma-
`tology, Allergy and Clinical Immunology, University of Cali-
`fornia at Davis, Davis, CA; M. Eric Gershwin, MD: Professor
`of Medicine, Division of Rheumatology, Allergy and Clinical
`Immunology, University of California at Davis, Davis, CA.
`Address reprint requests to M. Eric Gershwin, MD, Division
`of Rheumatology, Allergy and Clinical Immunology, University
`of California at Davis School of Medicine, TB 192, Davis, CA
`95616. E-mail: megershwin@ucdavis.edu
`© 2004 Elsevier Inc. All rights reserved.
`0049-0172/04/3401-0000$30.00/0
`doi:10.1016/j.semarthrit.2003.12.003
`
`Seminars in Arthritis and Rheumatism, Vol 34, No 1 (August), 2004: pp 465-483
`
`465
`
`Medac Exhibit 2013
`Koios Pharmaceuticals v. Medac
`IPR2016-01370
`Page 00001
`
`

`

`466
`
`BORCHERS ET AL
`
`response has not been achieved with hydroxychlo-
`roquine (HCQ) or sulfasalazine (SSZ). In patients
`with very active disease or a poor prognosis, it may
`even be advisable to use MTX as the initial
`DMARD. This has been the practice among rheu-
`matologists in the United States even before pub-
`lication of the updated guidelines (3). For patients
`with severe RA, MTX is the DMARD of first
`choice for rheumatologists in the United States and
`in Europe (England and The Netherlands), and in
`both regions it is the anchor drug in therapy based
`on the combination of 2 DMARDs (4,5).
`
`METHODS
`A Medline search of published studies with key
`words
`“methotrexate,”
`“rheumatoid arthritis,”
`“DMARDs,” and “amethopterin” from 1950 to
`2003 was conducted. From these studies and their
`accompanying references, a total of 1114 studies
`dating from 1952 to the present were reviewed and
`131 were determined to be pertinent to our discus-
`sion. These manuscripts included a variety of sub-
`jects, including pharmacology, therapy of connec-
`tive-tissue diseases, and use of DMARDs. The
`manuscripts were reviewed with particular refer-
`ence to the usage of MTX in RA, with emphasis on
`double-blinded studies and on mechanisms of ac-
`tion.
`
`Pharmacology
`MTX (amethopterin) is a methyl-derivative of
`aminopterin, a drug that was first used for the
`treatment of RA in 1951. Both aminopterin and
`MTX are folate analogues, but they are also folate
`antagonists in that they compete with folate for
`cellular uptake and intracellular polyglutamation
`and they reversibly inhibit dihydrofolate reductase
`(DHFR). DHFR participates in single carbon trans-
`fers necessary for methionine, purine, and thymi-
`dylate synthesis and, ultimately, DNA synthesis.
`The inhibition of other folate-dependent enzymes
`by polyglutamated MTX further contributes to this
`reduction of nucleotides available for cellular pro-
`liferation. Through partial substrate depletion and
`inhibition of thymidylate synthase, MTX polyglu-
`tamates block the de novo synthesis of thymidy-
`late. In addition, they interfere with de novo purine
`synthesis by directly blocking the activity of
`5-amino-imidazole-4-carboxamide ribonucleotide
`(AICAR) transformylase and by resulting in the
`accumulation of dihydrofolate polyglutamates,
`
`inhibitors of this
`
`which are even more potent
`enzyme (6).
`The inhibition of DNA synthesis rapidly affects
`proliferating cells, such as malignant cells, and
`also lymphocytes. This is likely the major mecha-
`nism of action of high-dose MTX in leukemia and
`other malignancies. “High-dose” refers to MTX
`therapy consisting of between 200 mg/m2 and 30
`g/m2 body surface administered intravenously over
`a period of 24 hours, first at weekly intervals and
`then at much longer intervals. However, patients
`with RA and psoriasis are treated with low-dose
`MTX (ie, doses ranging between 7.5 and 20 mg per
`week). However, we caution that “low-dose” refers
`to the comparative use of higher dosages in cancer
`and does not imply the doses used are nontoxic.
`The mechanism(s) of action of low-dose MTX
`have still not been fully elucidated. It has recently
`been reported that low concentrations of MTX
`either inhibit proliferation or induce apoptosis in
`activated, but not resting, peripheral blood lym-
`phocytes (7). These findings suggest that antipro-
`liferative and immunosuppressive effects contrib-
`ute to the effectiveness of
`low-dose MTX.
`However, somewhat conflicting results have been
`reported concerning the effect of MTX on prolif-
`erative responses, partly caused by the use of
`different methodologies for measuring prolifera-
`tion (8).
`MTX exerts a variety of antiinflammatory activ-
`ities (6,9). These are at least partly mediated via
`the increased release of adenosine as a conse-
`quence of AICAR transformylase inhibition by
`MTX polyglutamates. Inhibition of this enzyme
`results in the accumulation of AICAR, which in
`turn inhibits adenosine deaminase, thereby increas-
`ing the intracellular adenosine concentration and
`the subsequent release of adenosine into the extra-
`cellular fluid. Adenosine exhibits a variety of an-
`tiinflammatory activities, including the inhibition
`of leukocyte accumulation and neutrophil-medi-
`ated endothelial injury at sites of inflammation; the
`reduction in tumor necrosis factor (TNF)-␣synthe-
`sis; and inhibition of a variety of natural killer,
`monocyte/macrophage, and T-cell activities (6,9-
`11).
`In addition to its effects on de novo purine
`synthesis,
`low-dose MTX therapy was recently
`reported to be associated with decreased activity of
`several enzymes participating in the salvage path-
`way of purine synthesis (12). This effect was seen
`
`Page 00002
`
`

`

`CLINICAL USE FOR METHOTREXATE
`
`467
`
`after 48 weeks, but not after 6 weeks, of MTX
`treatment, was independent of folate supplementa-
`tion, and was not associated with measures of
`MTX efficacy or toxicity. An exception was 5⬘-
`nucleotidase, which exhibited decreased activity
`only in those patients who discontinued MTX
`because of hepatotoxicity.
`A further mechanism of action may involve an
`MTX-induced reduction in the synthesis of poly-
`amines via inhibiting the conversion of homocys-
`teine to methionine, the precursor for the methyl-
`donor S-adenosyl-methionine that is required for
`polyamine synthesis. Polyamines may play a role
`in the pathogenesis of RA and other inflammatory
`diseases (9).
`
`Efficacy in RA
`MTX is among the most commonly used
`DMARDs in the treatment of patients with RA. It
`is taken once a week in doses commonly ranging
`between 7.5 and 20 mg. Whereas in some clinical
`trials the weekly dose was divided into 3 equal
`aliquots taken in 12-hour intervals (13,14), patients
`with RA generally take the entire dose at once. The
`most common route of administration is oral, but
`parenteral (intramuscular, subcutaneous, and occa-
`sionally intravenous) routes are preferred for some
`patients. Although bioavailability of oral MTX is
`similar to that of parenteral MTX (15-17), there are
`reports of patients who respond to intramuscular
`injections but not to oral MTX. A possible expla-
`nation for this observation is that the bioavailabil-
`ity of oral MTX relative to intramuscular MTX
`was 13.5% lower at the usual maintenance dose
`(mean, 17 mg) than at the 7.5-mg dose commonly
`used at the initiation of MTX therapy (18).
`The efficacy of MTX in the treatment of RA has
`been shown in a number of randomized, controlled
`trials (RCTs) (19,20). A Cochrane systematic re-
`view concluded that, despite a fairly high (22%)
`withdrawal rate caused by adverse events, short-
`term MTX therapy provided substantial benefit to
`patients with RA and this benefit was both clini-
`cally and statistically significant (20). Compared
`with placebo, MTX therapy induces significant
`improvement in the number of tender and swollen
`joints, pain, physician and patient global assess-
`ment, and functional status. Approximately 60% to
`70% of patients have a significant response to
`MTX, although the estimates depend to some ex-
`tent on how “significant response” is defined (19).
`
`The onset of MTX-induced improvement is gener-
`ally within 3 months in the majority of patients
`who will eventually respond. After an initial period
`of rapid improvement, a plateau in the response is
`often reached after 6 to 12 months (21,22).
`In direct comparisons of traditional DMARDs in
`RCTs,
`their efficacy is generally quite similar,
`although MTX has been found to be superior to
`azathioprine and auranofin (19,23). The duration of
`most clinical trials is between 6 months and 2
`years, which is short compared with the natural
`history of RA in the majority of patients. In addi-
`tion, only select patients are included in RCTs and
`a variety of aspects of patient treatment, including
`dose adjustments and drug termination, are han-
`dled differently in RCTs than in common clinical
`practice. Long-term observational
`studies, al-
`though frequently not randomized and therefore
`liable to some bias, can offer a different perspec-
`tive on the efficacy and toxicity of a drug and on
`the reasons for its discontinuation.
`With few exceptions (24), long-term observa-
`tional studies indicate that toxicities–rather than
`lack of efficacy–are the most common cause of
`discontinuing MTX (22,25-32). As will be dis-
`cussed later, folic acid supplementation can reduce
`some of the toxicities of MTX. Many of the avail-
`able data stem from a time when patients treated
`with MTX did not routinely receive folate supple-
`mentation, and, in some of the more recent studies,
`information on the extent of supplementary folate
`intake was unavailable. This makes it difficult to
`assess whether the now-widespread use of folate
`has had an impact on the frequency of MTX
`discontinuations and the reasons for them.
`Studies comparing different second-line drugs
`show that discontinuations are less frequent
`in
`patients taking MTX than in those taking other
`DMARDs (24,26,27). Thus, MTX therapy has the
`highest median drug survival among DMARDs
`and the greatest cumulative probability of being
`continued for 5 years or even longer. The median
`time on MTX of 4.25 years was more than twice as
`long as that of other DMARDs (1.6 to 2 years)
`(26). Others have reported similar, occasionally
`even longer, median survival times (24,25,27). The
`percentage of patients still taking MTX after 5
`years ranges between 45% and 65% (25,27,31,33),
`but it has been reported to be as high as 73% in 1
`study (22) and only 25% in another (29). Even
`after 10 years, 30% of patients with RA continued
`
`Page 00003
`
`

`

`468
`
`BORCHERS ET AL
`
`to take MTX at a tertiary care center in Alabama
`(30). At last follow-up, 53% of patients with RA
`treated in 6 Australian community practices were
`still taking MTX after 12 years, but this percentage
`decreased to 38% if intermittent discontinuations
`of ⬎3 months were considered as terminations
`(32). It should be emphasized that careful moni-
`toring is required no matter how long a patient
`takes MTX.
`Notwithstanding its long median survival times,
`MTX is associated with some serious toxicities,
`discussed in more detail later. Thus, despite its
`proven effectiveness, it is important to examine
`whether newer DMARDs offer similar or even
`greater effectiveness with fewer toxicities. In re-
`cent years, several new agents for the treatment of
`RA have been developed, including leflunomide
`(Arava; Aventis, Bridgewater, NJ) and anticyto-
`kine agents, such as etanercept, infliximab, and
`anakinra.
`Leflunomide is a new DMARD approved for the
`treatment of RA by the Food and Drug Adminis-
`tration (FDA) in 1998. It is an isoxazolyl derivative
`that is converted by first-pass metabolism in the
`liver and gut to the active metabolite, A77 1726.
`This metabolite inhibits the rate-limiting enzyme
`in pyrimidine synthesis, dihydroorotate dehydro-
`genase, and thereby prevents cellular proliferation,
`particularly that of CD4⫹ T cells, which are known
`to proliferate rapidly during the initiation of RA.
`Recent comparisons between MTX and lefluno-
`mide indicate that they are similarly effective in
`treating RA (34,35), equally slowing radiographic
`progression (36) and both showing sustained clin-
`ical benefit over at least a 2-year period (35,37)
`(Tables 1 and 2). In 1 study, the clinical response
`rates to leflunomide were equivalent to those seen
`with MTX during the first year (34) but signifi-
`cantly higher after the second year (37). Con-
`versely, in another study, MTX resulted in greater
`improvements did than leflunomide in all outcome
`measures after 1 year, but the benefits from the 2
`drugs were similar at the end of the second year in
`the subgroup of patients who chose to continue the
`double-blind treatment (35). The frequency of ad-
`verse events, including serious ones, was similar
`with the 2 drugs, although the proportion of with-
`drawals because of toxicity tended to be higher in
`the leflunomide group in 1 trial (35). However, in
`the MTX group, there were 2 treatment-related
`deaths, whereas no deaths occurred in the lefluno-
`
`mide group. Asymptomatic transaminase eleva-
`tions resulting in withdrawal were more than twice
`as frequent in the leflunomide group than in the
`MTX group in 1 trial (7.1% vs 3.3%, respectively)
`(34), but half as frequent in another (1.6% vs 3.2
`%) (35).
`The effect of the addition of leflunomide to a
`stable, but clinically unsatisfactory, dose of MTX
`was assessed in an open-label trial (38) and an
`RCT (39). In the open-label trial, 20% of subjects
`met the ACR20 response criteria after 1 month of
`leflunomide treatment; this percentage increased to
`57% after 9 months, with a similar response rate
`maintained at 12 months. Similar results were
`reported in the RCT (39). Although generally well
`tolerated,
`the combination of
`leflunomide and
`MTX was associated with elevated liver enzymes
`in 63% of patients (38). Elevations of alanine
`aminotransferase (ALT) ⬎2 times the upper limit
`of normal were observed in 34% of the patients
`and elevations of aspartate aminotransferase (AST)
`were observed in 24%. For comparison, in a ran-
`domized study comparing MTX and leflunomide,
`serum transaminase elevations 2 times the upper
`limit of normal occurred in 9.3% of patients treated
`with MTX and 11% of patients treated with le-
`flunomide (34).
`The first report of serious liver disease (early
`cirrhosis) associated with the use of a combination
`of leflunomide and MTX was published in 2000
`(40). Since then, a number of serious hepatic ab-
`normalities have been described in postmarketing
`reports, resulting in considerable controversy over
`the safety of leflunomide (41). This issue was
`recently addressed by an FDA-appointed advisory
`panel. Based on the currently available evidence,
`this panel deemed the safety of leflunomide ade-
`quate and acceptable and its benefits greater than
`its potential safety risks.
`
`Anticytokine Agents
`Based on the recognition that TNF-␣ and inter-
`leukin (IL)-1 play important roles in the pathogen-
`esis of RA (42), several biologic agents that block
`the activity of these two proinflammatory cytokines
`have been developed. These include etanercept, a
`soluble TNF receptor (p75):Fc fusion protein; in-
`fliximab, a chimeric human-murine monoclonal
`anti–TNF-␣antibody; and anakinra, a recombinant
`human IL-1 receptor antagonist (IL-1ra).
`Etanercept (Enbrel; Immunex, Seattle, WA) has
`
`Page 00004
`
`

`

`CLINICAL USE FOR METHOTREXATE
`
`469
`
`Table 1: MTX Versus Leflunomide or Etanercept in 12-month RCTs
`
`MTX (34)
`
`Leflunomide
`
`MTX (35)
`
`Leflunomide
`
`MTX* (48)
`
`Etanercept
`
`7.5 mg/wk,
`increased
`to 15 mg/
`wk if active
`disease
`persisted
`
`Loading dose
`of 100 mg/
`d for 3 d;
`20 mg/d
`thereafter
`
`7.5 mg/wk in
`wks 1-4, 10
`mg/wk in
`wks 2-12,
`then 10-15
`mg/wk
`
`Loading dose
`of 100 mg/
`d for 3 d:
`20 mg/d
`thereafter
`
`46
`23
`9.5 (6.5) wk
`
`52
`34
`8.6 (7.4) wk
`
`64.8†
`—
`101 ⫾ 92.5 d†
`
`50.5
`—
`74 ⫾ 80 d
`
`25 mg sub-
`cutan-
`eously
`twice
`weekly
`
`7.5 mg/wk,
`increased
`to 15 mg/
`wk at wk 4,
`and to 20
`mg/wk at
`wk 8
`72
`65
`MTX ⬍ Etanercept†
`MTX ⬎ Etanercept†
`
`⫺6.6 ⫾ 7.6
`⫺5.4 ⫾ 5.5
`0.88 (3.3)†
`
`⫺7.7 ⫾ 7.8
`⫺5.7 ⫾ 6.5
`0.53 (4.5)
`
`⫺9.7 ⫾ 7.9†
`⫺9.0 ⫾ 7.3†
`—
`
`⫺8.3 ⫾ 7.9
`⫺6.8 ⫾ 7.3
`—
`
`—
`—
`1.59
`
`—
`—
`1.00
`
`—
`
`—
`
`0.03
`
`0.03
`
`—
`
`—
`
`0.48 (1.8)
`
`0.23 (2.2)
`
`—
`
`—
`
`1.03†
`
`0.47
`
`10‡
`
`24‡
`
`22
`
`17
`
`15‡
`
`3‡
`
`19
`
`7
`
`11†
`
`4
`
`5
`
`5
`
`Dose
`
`ACR20 (%)
`ACR50 (%)
`Time to reach
`ACR20
`Joint count
`Tender
`Swollen
`Change in
`Sharp
`score
`(units)
`Change in
`Larson
`score
`(units)
`Change in
`erosion
`score
`(units)
`Discontinuation
`Because of
`toxicity
`Because of
`lack of
`efficacy
`
`*Some data were presented as figures (ie, actual numbers were not provided).
`†Statistically significantly different from treatment comparison.
`‡No statistical evaluation was presented.
`
`been approved by the FDA for RA as monotherapy
`or in combination with MTX. The approval was
`based on the significant clinical benefit, as assessed
`by ACR criteria, that this agent exhibited either
`alone or in addition to MTX in multi-center, dou-
`ble-blind, placebo-controlled trials (43-45). Clini-
`cal trials of longer duration have further confirmed
`the efficacy of etanercept and have added the
`findings that the benefit is sustained over at least a
`2-year period and that etanercept can slow, or even
`halt, radiographic progression (46,47).
`A recent RCT directly compared MTX and et-
`
`anercept (48). The weekly MTX dose was in-
`creased from the initial 7.5-mg dose to 20 mg in all
`patients, but the dose had to be reduced in 15% of
`them because of adverse events or serum transam-
`inase elevations. Etanercept was administered sub-
`cutaneously twice a week at a dose of either 10 mg
`or 25 mg. Compared with MTX, the higher dose of
`etanercept was found to induce an ACR20 re-
`sponse faster and in a greater proportion of patients
`during the first 6 months of the 12-month trial.
`Thereafter, the percentages were similar in the 2
`groups. Several factors are likely to account for the
`
`Page 00005
`
`

`

`470
`
`BORCHERS ET AL
`
`Table 2: MTX Versus Leflunomide or Etanercept in 24-month RCTs
`
`MTX (37)
`
`Leflunomide
`
`MTX (35)
`
`Leflunomide MTX (47) Etanercept
`
`Dose
`
`7.5-15 mg/wk
`
`20 mg/d
`
`10-15 mg/wk
`
`20 mg/d
`
`ACR20 (%)
`ACR50 (%)
`Joint count
`Tender
`Swollen
`Change in Sharp score
`(units)
`Change in Larson score
`(units)
`Change in erosion score
`(units)
`Discontinuation because
`of adverse events
`Toxicity
`Lack of efficacy
`
`67*
`43†
`
`79
`56
`
`71.7
`—
`
`64.3
`—
`
`⫺8.8 ⫾ 9.0
`⫺7.7 ⫾ 6.7
`1.2 ⫾ 3.8
`
`⫺10.1 ⫾ 6.7 ⫺10.9 ⫾ 8.2
`⫺8.2 ⫾ 6.1 ⫺10.3 ⫾ 7.3*
`1.6 ⫾ 4.2
`—
`
`⫺10.5 ⫾ 8.1
`⫺9.1 ⫾ 7.7
`—
`
`—
`
`—
`
`MTX better than
`leflunomide‡
`
`0.6 ⫾ 1.7
`
`1.0 ⫾ 3.1
`
`—
`
`17
`—
`
`27
`—
`
`4
`6
`
`—
`
`6
`8
`
`20 mg/wk 25 mg twice
`weekly
`72
`49
`
`59*
`42
`
`—
`—
`3.2*
`
`—
`
`1.9*
`
`12*
`—
`
`—
`—
`1.3
`
`—
`
`0.7
`
`7
`—
`
`*Statistically significantly different from treatment comparison.
`†P ⫽ .053.
`‡In the second year, there was no further increase in joint damage in lefunomide and a small improvement in MTX, resulting in a
`small but statistically significant treatment difference.
`
`observation that etanercept gave more favorable
`results than MTX in the early phase of the trial.
`One is that MTX is started at a low dose and
`gradually increased, another is the previously men-
`tioned finding that it takes approximately 6 months
`for the response to MTX to plateau.
`The trial was continued with 512 of the original
`632 patients in an open-label manner for another
`12 months (47). During this time, patients contin-
`ued to receive the therapy to which they had been
`randomized. The benefits from both treatments
`were sustained, and etanercept continued to pro-
`vide a greater clinical response so that 72% of
`patients in the 25-mg etanercept group, after 24
`months, met ACR20 improvement compared with
`59% of the MTX group. Although the subjects
`participating in this trial had RA for ⱕ3 years, the
`eligibility criteria were designed to insure that only
`patients at high risk of radiographic progression
`were recruited. It is noteworthy that, at the end of
`the trial, significantly more patients in the 25 mg
`etanercept group than in the MTX group showed
`no radiographic progression from baseline (no in-
`crease in total Sharp score in 63% vs 51% and no
`increase in erosions in 70% versus 58% of pa-
`
`tients). In addition, the mean changes in total Sharp
`score and erosion score were significantly lower in
`the 25-mg etanercept group (1.3 and 0.7 units,
`respectively) than in the MTX group (3.2 and 1.9
`units, respectively).
`infliximab, particularly
`The effectiveness of
`when combined with MTX, has also been shown in
`multicenter RCTs (49,50) and infliximab has been
`approved by the FDA for use in RA. Infliximab has
`not been compared directly with MTX but it has
`been evaluated in combination with MTX. In an
`RCT involving patients with RA who were not, or
`only partially, responsive to MTX, addition of
`infliximab in 4 different doses to their stable dose
`of MTX was shown to result in a rapid improve-
`ment that was sustained over 30 weeks (50). Total
`ACR20 response rates were significantly higher in
`the MTX plus infliximab groups than in the MTX
`plus placebo group, and substantially more inflix-
`imab-treated patients achieved ACR50 and ACR70
`responses. The clinical benefit was sustained over
`24 weeks and, importantly, radiographic progres-
`sion was essentially halted in the patients receiving
`MTX plus infliximab, whereas a 10% increase in
`the total radiographic score was noted in the group
`
`Page 00006
`
`

`

`CLINICAL USE FOR METHOTREXATE
`
`471
`
`receiving MTX plus placebo (51). The frequency
`and types of adverse events,
`including serious
`ones, did not differ significantly between the MTX
`plus infliximab and the MTX plus placebo groups.
`Anakinra (recombinant IL-1ra) has been re-
`ported to significantly improve measures of clini-
`cal disease activity and slow the rate of radio-
`graphic progression in patients with RA (52-54),
`and it has been approved by the FDA for the
`treatment of RA. A direct comparison of this bio-
`logic agent with MTX has not yet been conducted,
`but the combination of the 2 in patients with only
`suboptimal responses to MTX alone induced an
`ACR20 response faster and in a greater proportion
`of patients than did MTX plus placebo (55). The
`response to anakinra was dose-dependent, and the
`changes seen with doses of 1.0 mg/kg and 2.0
`mg/kg reached statistical significance relative to
`placebo.
`The cost of biologic agents is considerably
`higher than that of most
`traditional DMARDs.
`According to the ACR, the estimated annual drug
`cost for usual maintenance doses in 2001 were
`$697 to $1859 for MTX, $509 to $763 for SSZ,
`$2938 for leflunomide, $15436 for Etanercept, and
`$13,940 to $36,694 for infliximab (not including
`physician’s office visit fees or laboratory costs
`associated with monitoring) (2). Cost effectiveness
`increasingly enters into physicians’ decisions of
`which drug to prescribe (56). Because of the lack
`of longitudinal data that allow for the calculation
`of the cost effectiveness of these newer treatment
`options, only some preliminary estimates are avail-
`able (2,57).
`
`Combination Therapy
`In addition to combinations of biologic agents
`and traditional DMARDs, regimens combining 2
`or 3 traditional DMARDs are being used with
`increasing frequency in clinical practice. This trend
`has developed despite the limited number of RCTs
`that have addressed the issue of whether combina-
`tions of DMARDs provide greater benefit than
`monotherapies without increasing toxicity. A sys-
`tematic review of 20 studies meeting the inclusion
`criteria covered 8 trials of MTX combined with
`other DMARDs (58). Whereas the double therapy
`consisting of MTX with azathioprine, auranofin, or
`SSZ did not provide greater benefit than MTX
`alone, the combination of MTX with cyclosporin
`was superior to MTX alone. Triple or quadruple
`
`therapy involving MTX plus HCQ plus SSZ with
`or without prednisone has been reported to result in
`higher response rates and less radiographic pro-
`gression than MTX alone or MTX combined with
`only 1 other DMARD (59-62). Direct comparisons
`of the results of such studies are often hampered by
`significant differences in study design, particularly
`with respect to the type of patients recruited, dos-
`ages and permitted dosage increases, primary out-
`come measures, and the criteria used to define
`them (Table 3).
`Whereas many of the published studies used a
`parallel or step-up approach, the step-down (from
`aggressive high dosages to less aggressive lower
`dosages) approach has also been investigated in
`only 1 study by comparing SSZ alone with a
`combination of SSZ, prednisolone, and MTX in
`patients with early but very active RA (63). The
`initial prednisolone dose of 60 mg/d was tapered
`rapidly to 7.5 mg/d by week 7, maintained at that
`level until week 28, then gradually tapered and
`stopped. MTX therapy consisted of 7.5 mg/wk
`until week 40, followed by gradual tapering and
`termination by the end of week 46. Patients in the
`combined-treatment group improved faster and
`achieved a higher response (ACR20) rate after 28
`weeks than the SSZ group. Once prednisone was
`discontinued, some of the initial benefit was lost,
`whereas the SSZ group continued to slightly im-
`prove. After 56 weeks, the differences in measures
`of clinical efficacy were no longer significant.
`However, joint destruction and erosion scores were
`significantly lower in the combined group at 28,
`56, and 80 weeks. This research study suggests that
`prednisolone can significantly slow radiographic
`progression. Others have reported similar findings
`(64,65), but follow-up in 1 of the study cohorts
`indicated that joint destruction resumed on with-
`drawal of steroids (66). This contrasts with the
`findings after 4 to 5 years of follow-up in the SSZ
`and combined SSZ/MTX/prednisolone groups de-
`scribed previously (63) that were recently pub-
`lished (67). Compared with the group treated with
`SSZ alone, the combined-therapy group continued
`to exhibit a significantly reduced rate of radio-
`graphic progression (3.7 points per year less, even
`after adjustment for differences in treatment and
`disease activity during follow-up).
`Although the results of some of these clinical trials
`with DMARD and/or prednisolone combinations are
`promising, it appears that the effects are additive
`
`Page 00007
`
`

`

`472
`
`BORCHERS ET AL
`
`Table 3: Endpoints Used in the Evaluation of DMARDs
`
`Description
`
`Comments
`
`To qualify as a responder, the patient
`must complete the trial and must
`meet ACR response criteria at the
`end of the trial; some researchers
`report the percentage of patients
`fulfilling the criteria at some point
`during the trial
`The actual number of joints should
`be 71 for tenderness and 68 for
`swelling, but this number varies
`greatly among studies
`A variety of different questionnaires
`have been used
`The possibility of tradeoffs between
`efficacy and toxicity does not enter
`into the ACR definition of response
`Not as widely used and accepted as
`the ACR response criteria
`
`Provide some measure of functional
`status, but are generally used only
`as secondary outcome measures
`This measure is particularly valuable
`for comparisons between drugs or
`between single drugs and drug
`combinations; unfortunately, the
`definitions used for what
`constitutes efficacy and toxicity
`necessitating withdrawal vary
`considerably among studies
`Radiologic assessment provides
`valuable information about the rate
`of disease progression
`
`Endpoint
`
`ACR 20, 50,
`70
`response
`
`ⱖ 20%, 50%, and 70% improvement
`in tender joint count and swollen
`joint count and in at least 3 of the
`following 5 criteria: patient global
`assessment, physician global
`assessment, pain intensity, HAQ,
`CRP, ESR
`
`Other
`composite
`measures
`Other
`individual
`measures
`
`Paulus criteria, DAS, DAS28, EULAR,
`WHO/ILAR criteria
`
`Duration of morning stiffness, 50-foot
`walking time, grip strength
`
`Withdrawal because of lack of
`efficacy or toxicity
`
`Radiographic
`progression
`
`Sharp
`Score
`Larson
`Score
`
`Assessment of joint erosion and joint
`space narrowing by using the
`Sharp or the Larsen scoring
`system or modifications thereof
`Counts the number of erosions from
`0 to 5 regardless of their size*
`Defines grades less precisely by
`using general terms and standard
`reference radiographs
`
`Abbreviations: CRP, C-reactive protein; DAS, Disease Activity Score; ESR, erythrocyte sedimentation rate; EULAR, European League
`Against Rheumatism; HAQ, health assessment questionnaire; WHO/ILAR, World Health Organization/International League Against
`Rheumatism.
`*The van der Heijde modification does take the size relative to the joint surface into account.
`
`rather than synergistic. No increase in toxicity was
`detected with combinations compared with single
`agents in a majority of the short term RCTs, but it
`remains to be established whether this will hold true
`
`for long-term combination treatment. Prednisolone
`therapy in RA, if used inappropriately, is fraught with
`potential toxicity. Further research is clearly needed
`to establish optimal combinations and dosages.
`
`Page 00008
`
`

`

`CLINICAL USE FOR METHOTREXATE
`
`473
`
`Radiographic Progression
`A meta-analysis of trials published until 1991
`indicated that, although MTX did not slow radio-
`graphic progression to a significantly greater extent
`than gold salts, it was superior to azathioprine (68).
`The inclusion in the meta-analysis of case-control
`and long-term observational studies along with
`RCTs may be questioned because treatment dura-
`tion in most of the RCTs was 6 to 12 months,
`whereas some of the observational studies fol-
`lowed up patients for up to 84 months. Data from
`long-term observational studies of patients treated
`with MTX show that radiographic progression oc-
`curs despite MTX therapy in a majority of patients
`(21,22,28,33). However, in all of these studies, a
`subset of patients did not experience radiographic
`worsening of lesions.
`A vast majority of patients included in these
`stud

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket