therapy for rheumatoid arthritis: combinations of disease-modifying drugs and new paradigms of...

14
Therapy for Rheumatoid Arthritis: Combinations of Disease- Modifying Drugs and New Paradigms of Treatment By William S. Wilke and John D. Clough The last 10 years have witnessed a change in the way rheumatologists view rheumatoid arthritis (RA). It is no longer considered a slowly progres- sive disease limited to the joints, but rather an aggressive systemic disease that results in clini- cally significant morbidity early in its course and can contribute to excess mortality. Heightened awareness of the health impact of RA has spurred a search for effective therapy to be applied early in the course of disease for patients with moderate to severe RA. Combinations of disease-modifying antirheumatic drugs (DMARD) have become an increasingly popular alternative to sequential monotherapy. In this report, we review published series of patients with RA who have been treated with combinations of DMARDs, sometimes in- cluding chemotherapeutic agents, with some critical comment. Published paradigms of treat- “The basic program of rest, salicylates, and physical therapy is the most sensible beginning of therapy. The combination of more than two antiinflammatory drugs is not recommended. John A, Goldman, Evelyn B. Hess, 1970.’ “You know something’s happening But you don’t know what it is For the times they are A ‘changin’ ” Bob Dylan, circa 1965. C ONSERVATIVE THERAPY for rheuma- toid arthritis (RA) was the recognized and popular approach advocated by rheumatologists in the 1960s and early 1970s. This position was eminently defensible. Treatment with medica- tions in common usage, especially parenteral gold and D-penicillamine (DPN), was by no means universally effective, and resulted in significant adverse effects in at least one third of treated pa- tients.2*3 Trials of chemotherapeutic agents, such as the controlled study of methotrexate (MTX) by Black et aJ4 while demonstrating the efficacy of this drug class, proved that in high doses these drugs were very dangerous. Gradually, over the last 10 years, chemotherapeutic drugs have gained acceptance, first as third-line agents and more recently, in the case of MTX, as a possible second- line drug of choice for the treatment of RA.5,6 In addition, rheumatologists have adopted a more aggressive approach to the treatment of this dis- ment are also reviewed and a new strategy is pre- sented. The “step-down bridge” strategy allows early treatment with at least four DMARDs, but may place some patients with mild disease at an inappropriately high risk of adverse effects. The “sawtooth” strategy gives little guidance as to which DMARD(s) should be chosen for initial treatment. We describe a “graduated-step” strategy that provides numerical grading to match disease severity and disease activity with ap- propriate initial therapy and that facilitates ther- apeutic decisions throughout the course of treat- ment. Copyright 0 1991 by W.B. Saunders Company INDEX WORDS: Rheumatoid arthritis; combina- tion therapy; strategies of treatment; “graduated- step” strategy. eaSe and are willing to initiate disease-modifying antirheumatic drugs (DMARDs) earlier in the course of disease than in the past.’ This evolution in therapeutic thought has been driven by some recent changes in our understanding of RA and the therapeutic agents used to treat it. RA is no longer considered a benign disease that only has an impact on function. Severe rheumatoid disease has been shown to confer both long-term morbidity and early mortality.*,’ In addition, the high frequency of severe adverse effects expected with chemotherapeutic agents used early in the course of disease has not been realized. lo Although these agents are effective when they are used in low-dose monotherapy, disease may From the Department ofRheumatic and Immunologic Dis- ease, The Cleveiand Clinic Foundation, Cleveland, OH. William S. Wilke, MD: Staff; Department of Rheumatic and Immunologic Disease, Head, Section on Subspecialty Clinics, The Cleveland Clinic Foundation; John D. Clough, MD: Sta& Department of Rheumatic and Immunologic Dis- ease, Director of Health Affairs, The Cleveland Clinic Foun- dation. Address reprint requests to William S. Wilke, MD, De- partment ofRheumatic and Immunologic Diseases (Desk AjO), The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195-5028. Copyright 0 1991 by W.B. Saunders Company 0049-0172/91/2102-1004%5.00/0 Seminars in AIthriri.. and Rheumetism, Vol2 1, No 2, Suppl 1 (October), 199 1: pp 2 l-34 21

Upload: william-s-wilke

Post on 28-Aug-2016

214 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Therapy for rheumatoid arthritis: Combinations of disease-modifying drugs and new paradigms of treatment

Therapy for Rheumatoid Arthritis: Combinations of Disease- Modifying Drugs and New Paradigms of Treatment

By William S. Wilke and John D. Clough

The last 10 years have witnessed a change in the way rheumatologists view rheumatoid arthritis (RA). It is no longer considered a slowly progres- sive disease limited to the joints, but rather an aggressive systemic disease that results in clini- cally significant morbidity early in its course and can contribute to excess mortality. Heightened awareness of the health impact of RA has spurred a search for effective therapy to be applied early in the course of disease for patients with moderate to severe RA. Combinations of disease-modifying antirheumatic drugs (DMARD) have become an increasingly popular alternative to sequential monotherapy. In this report, we review published series of patients with RA who have been treated with combinations of DMARDs, sometimes in- cluding chemotherapeutic agents, with some critical comment. Published paradigms of treat-

“The basic program of rest, salicylates, and physical therapy is the most sensible beginning of therapy.

The combination of more than two antiinflammatory drugs is not recommended. ”

John A, Goldman, Evelyn B. Hess, 1970.’

“You know something’s happening But you don’t know what it is For the times they are A ‘changin’ ”

Bob Dylan, circa 1965.

C ONSERVATIVE THERAPY for rheuma- toid arthritis (RA) was the recognized and

popular approach advocated by rheumatologists in the 1960s and early 1970s. This position was eminently defensible. Treatment with medica- tions in common usage, especially parenteral gold and D-penicillamine (DPN), was by no means universally effective, and resulted in significant adverse effects in at least one third of treated pa- tients.2*3 Trials of chemotherapeutic agents, such as the controlled study of methotrexate (MTX) by Black et aJ4 while demonstrating the efficacy of this drug class, proved that in high doses these drugs were very dangerous. Gradually, over the last 10 years, chemotherapeutic drugs have gained acceptance, first as third-line agents and more recently, in the case of MTX, as a possible second- line drug of choice for the treatment of RA.5,6 In addition, rheumatologists have adopted a more aggressive approach to the treatment of this dis-

ment are also reviewed and a new strategy is pre- sented. The “step-down bridge” strategy allows early treatment with at least four DMARDs, but may place some patients with mild disease at an inappropriately high risk of adverse effects. The “sawtooth” strategy gives little guidance as to which DMARD(s) should be chosen for initial treatment. We describe a “graduated-step” strategy that provides numerical grading to match disease severity and disease activity with ap- propriate initial therapy and that facilitates ther- apeutic decisions throughout the course of treat- ment. Copyright 0 1991 by W.B. Saunders Company

INDEX WORDS: Rheumatoid arthritis; combina- tion therapy; strategies of treatment; “graduated- step” strategy.

eaSe and are willing to initiate disease-modifying antirheumatic drugs (DMARDs) earlier in the course of disease than in the past.’ This evolution in therapeutic thought has been driven by some recent changes in our understanding of RA and the therapeutic agents used to treat it.

RA is no longer considered a benign disease that only has an impact on function. Severe rheumatoid disease has been shown to confer both long-term morbidity and early mortality.*,’ In addition, the high frequency of severe adverse effects expected with chemotherapeutic agents used early in the course of disease has not been realized. lo

Although these agents are effective when they are used in low-dose monotherapy, disease may

From the Department ofRheumatic and Immunologic Dis- ease, The Cleveiand Clinic Foundation, Cleveland, OH.

William S. Wilke, MD: Staff; Department of Rheumatic and Immunologic Disease, Head, Section on Subspecialty Clinics, The Cleveland Clinic Foundation; John D. Clough, MD: Sta& Department of Rheumatic and Immunologic Dis- ease, Director of Health Affairs, The Cleveland Clinic Foun- dation.

Address reprint requests to William S. Wilke, MD, De-

partment ofRheumatic and Immunologic Diseases (Desk AjO), The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195-5028.

Copyright 0 1991 by W.B. Saunders Company 0049-0172/91/2102-1004%5.00/0

Seminars in AIthriri.. and Rheumetism, Vol2 1, No 2, Suppl 1 (October), 199 1: pp 2 l-34 21

Page 2: Therapy for rheumatoid arthritis: Combinations of disease-modifying drugs and new paradigms of treatment

22 WILKE AND CLOUGH

not be well controlled in every patient.“,” Higher doses of these agents not only increase their ef- fectiveness, but also result in at least a trend to- ward a higher frequency of adverse effects.‘3-‘5 To minimize the incidence and severity of ad- verse effects from high doses of individual agents, relatively low doses of two or more drugs are given simultaneously.

WHICH DRUGS SHOULD BE COMBINED?

An informed decision about which DMARDs to combine requires detailed information about the pharmacology and mechanisms of action of each separate drug. For instance, a renal-toxic drug such as cyclosporine A probably should not be combined with another drug that depends al- most entirely on renal excretion for its elimina- tion, such as MTX. Drugs that are combined ought to have different mechanisms of action that might lead to potentiation, as one drug acts more “proximally” in the inflammatory cascade, thereby allowing lower doses of other drugs that act more “distally.” The ideal combination would include drugs with actions that allow intracellular potentiation.

Combined DMARDs also should have sepa- rate toxicity profiles, which ought to reduce the likelihood of potentiated toxicity. Distinctly sep- arate toxicities also allow blame for adverse re- actions to be correctly assigned so that the of- fending agent may be reduced in dose or discontinued. The toxicity profile of the combi- nation should be acceptable.

Differing durations of onset might be exploited. Because the onset of action of MTX generally occurs within 3 to 6 weeks, it might be combined with oral gold, which has a delayed onset of ac- tion.16 Patients treated with this combination should experience early improvement of the dis- ease process with MTX. Later, as oral gold takes effect, the dose of MTX may be lowered or the drug may be discontinued.

Unfortunately, knowledge about drug inter- actions and mechanism(s) of action is incom- plete. The combination of DPN, which inhib- its T-cell function,‘7 and hydroxychloroquine (HCQ), which inhibits macrophage function,‘* would seem appropriate. In fact, as we will see, this combination shows little promise.

COMBINATION THERAPY IN THE LITERATURE

Combinations of DMARDs Excluding Chemotherapeutic Drugs

Six series have reported experience with “con- servative” DMARDs in combination (Table 1). Four separate series have reported the combi- nation of antimalarials with other DMARDs.

HCQ combined with DPN was compared with each drug alone in a 2-year double-blind con- trolled study of 56 patients.lg At 6 months, 43% of patients taking DPN alone were markedly im- proved, as were 17% of patients taking HCQ alone, whereas only 7% of patients who received combination therapy were markedly improved. This pattern of response continued throughout 24 months of therapy. Moderately severe adverse effects that resulted in lowering the dose of one or the other agent or in discontinuing an agent occurred in 29% of patients receiving combina- tion therapy, 43% of patients receiving DPN alone, and 22% of patients receiving HCQ alone.

In another study, 72 patients were given chlo- roquine phosphate 250 mg/d, DPN, or the com- bination of these two drugs, and monitored for 1 year2’ Chloroquine alone was the least effective treatment in this study. At 12 months, radio- graphs of the hands were compared. Forty-one percent of patients taking DPN and 45% of pa- tients taking the combination were judged to have x-rays that were unchanged or improved. Only 12% of the patients taking chloroquine showed x-ray changes that were unchanged or improved. Adverse effects occurred in 57% of patients taking the combination, 42% taking DPN alone, and 15% of patients taking chloroquine alone.

Neither of these series recommends the com- bination of antimalarials and DPN. In the first study, patients actually fared worse with com- bination therapy. In the second study, the addi- tion of chloroquine to DPN may have resulted in mildly additive improved efficacy, but also contributed to a higher incidence of adverse ef- fects.

Other series have combined HCQ with par- enteral gold. In a placebo-controlled 12-month prospective study that compared 52 patients re- ceiving parenteral gold and HCQ with 49 patients receiving parenteral gold and placebo, the group that received the combination showed a trend toward greater improvement in all measures.2’

Page 3: Therapy for rheumatoid arthritis: Combinations of disease-modifying drugs and new paradigms of treatment

COMBINATION THERAPY FOR RHEUMATOID ARTHRITIS 23

Table 1: Series in Which Combinations of DMARDs Without Chemotherapeutic Agents Were Used

Reference No. of

Patients DMARDs Efficacy Toxicity

BunchJg 56 HCQ 2.2 mg/kg/d

DPN 7 mg/kg/d

Gibson*’ 72 Chloroquine 250 mg/d

DPN maximum of 750 mg/d

Scot? 101 HCQ 400 mg/d

P. gold 50 mg/wk

Singletot? 10

Taggart23 30

HCQ 200 mg/d P. gold

SSZ 2 g/d

DPN 500 mg/d

Farr24 38 SSZ 1.5-3.0 g/d + either

DPN 500 mg/d or

P. gold 50 mg/wk

DPN alone more effective

DPN and combination

“equally effective”

Trend favoring

combination

Response in 80%

Combination significantly

better than SSZ alone

9 (24%) “good”

17 (46%) partial

remission in 2 patients

Led to D/C in:

Combination-2g%

DPN-43%

Combination-57%

DPN-42%

Chloroquine-15%

Withdrawals:

Combination-25

Gold alone- 17 .> safer than gold

alone”

Withdrawals:

Combination-7/l 5

ssz-3115

At 12 months-withdrawal:

DPN-3 1%

P. gold-33% 1 Abbreviations: HCQ, hydroxychloroquine; DPN, o-penicillamine; P. gold, parenteral gold; SSZ, sulfasalazine

Only improvement in C-reactive protein (CRP) showed significance at the 1% level. Unfortu- nately, there was a higher incidence of adverse effects in patients receiving combination therapy, and although they were generally nuisance tox- icities, such as rash or gastrointestinal upset, the investigators felt that adverse effects limited the usefulness of this combination.

A lower incidence of adverse effects was seen in an earlier series presented only as an abstract.** In this study, parenteral gold was added after 13 months of therapy with HCQ 200 mg/d. After a mean total dose of 750 mg of parenteral gold, eight of 10 patients improved or remitted. All patients experienced at least a one-tube dilution decrease of rheumatoid factor titer. Therapy was continued for 3 years, and no patient was with- drawn due to adverse effects. The investigators speculated that HCQ somehow protected patients from the toxicity of parenteral gold therapy.

These reports suggest that the combination of HCQ and parenteral gold may benefit patients. Divergent opinions about the safety of this com- bination might be answered by further long-term prospective trials.

Sulfasalazine (SSZ) also has been used in com- bination with other DMARDs.23 A 6-month open trial of SSZ 2 g alone in 15 patients or in com-

bination with DPN 500 mg/d in 15 patients showed moderately increased efficacy with the combination. With SSZ alone, only the eryth- rocy-te sedimentation rate (ESR) and grip strength showed statistical improvement. With the com- bination, the ESR, grip strength, and articular index all showed statistically significant im- provement. Unfortunately, adverse effects in- cluding rash, dysgeusia, and thrombocytopenia were much more common in patients treated with the combination and led to seven withdraw- als from combination therapy versus three with- drawals with SSZ alone.

In another study, 38 patients established on a constant dose of SSZ were given either DPN or parenteral gold.24 During the first year, approx- imately one third of the patients in each group withdrew due to toxicity. However, compared with baseline, the entire group experienced an overall clinical response of 70%, which indicated to the investigators that both combinations were more effective than SSZ alone.

Both of these studies suggest that the combi- nation of parenteral gold or DPN with SSZ is relatively safe, and that this combination is more effective than SSZ alone. Whether these combi- nations are more effective than parenteral gold or DPN alone is unknown. If the combination is

Page 4: Therapy for rheumatoid arthritis: Combinations of disease-modifying drugs and new paradigms of treatment

24

more effective, it lends itself to further study be- cause the typical adverse effects of SSZ differ from those of either DPN or parenteral gold.

Combinations Using One Chemotherapeutic Agent

All of the chemotherapeutic agents commonly used to treat RA have been combined with at least one other DMARD (Table 2). In addition, prednisone has been combined with a cytotoxic drug in a controlled study.25 High-dose cyclo- phosphamide (CTX) (1.7 mg/kg/d) in 19 patients was compared with low-dose (CTX ( 1.1 mg/kg/ d) combined with low-dose prednisone (7.7 mg/ d) in 13 patients. Improvement in the number of affected joints and grip strength was similar in both groups. Walking time and ring size showed greater improvement in high-dose CTX patients. On the other hand, 19 patients in the high-dose

WILKE AND CLOUGH

CTX group experienced 25 occurrences of ad- verse effects, including serious infection in two. Only one of 13 patients in the low-dose group experienced an adverse effect. In this series, pred- nisone was CTX-sparing.

CTX was one of the chemotherapeutic agents used in combination in an open trial of 21 pa- tients.26 Eleven RA patients were treated with parenteral antilymphocyte globulin 750 mg/d administered on 15 consecutive days and simul- taneous prednisolone 20 mg/d for 12 months. At 12 months, CTX 50 mg/d was added in one pa- tient, chlorambucil 4 mg/d in two patients, and azathioprine (AZA) 2.5 mg/kg/d in eight patients, At 6 and 12 weeks’ follow-up, the duration of morning stiffness, grip strength, and active joint count showed significant improvement for the group when compared with previous predniso- lone alone. Unfortunately, this improvement was

Table 2: Series in Which One Chemotherapeutic Agent Was Combined With Another DMARD

No. of Chemotherapeutic Reference Patients Agent DMARDs or Other EffiCaCy Toxicity

Smyth=k 13 CTX 1.1 mg/kg/d Prednisone 7.7 mg/d Equal to 1.7 mg/kg/d CTX -8% Binda?’ 11 AZA 2.5 mg/kg/d (8) Initial antilymphocyte globulin Overall significant at 3 mo 91%

Prednisolone 20 mg/d CHL 4 mg/d (2) CTX 50 mg/d (1 j’

Bittef’ 30 CHL 5 mg/d l Gold sodium thiomalate or Gold 30%

+ DPN 100 mg/d or + Levamisole 45 mg/wk

17 CHL 5 mg/d l DPN 100 mg/d or + 20%

+ Levamisole 450 mg/wk DPN best Let.+? 70 AZA 2.1 mg/kg/d Gold sodium thiomalate “Well controlled” (47.1%) No worse than gold

50 mg/mo alone

Shiroky” 4 MTX 11.25 mg/wk SSZ 2.3 g/d -88% improvement in None active joints

Lee” 18 MTX 10 mg/wk DPN 750 mg/d 8/l 2 achieved complete “. no dropouts”

remission Pred 10 -, 2.5 mg

Brawe?’ 7 MTX 11.5 mg/wk P. gold 75 mg/wk Recapture of good “No toxicity was response or remission observed”

in all

FtauJ2 48 MTX 7.5-15 mg/wk P. gold 50 mg/wk Overall reduction in No worse than MTX

disease activity alone

William+ 335 MTX 7.5 mg/wk AUR 8 mg/d Similar response for More withdrawals in

combination v. MTX combination for ADR

alone. Lack of response withdrawals = AUR 13%. MTX 7%. combination 2%

Kantor’O 287 MTX “standard dose” AUR “standard dose” 49% of pts taking AUR - 12% withdrawals alone after combination due to worsened clinically ADR with

combination

l Numbers in parentheses indicate number of patients treated.

Abbreviations: CTX, cyclophosphamide; AZA, azathioprine; CHL, chlorambucil; DPN, b-penicillamine; MTX, methotrexate; SSZ, sul-

fasalazine; P. gold, parenteral gold; AUR, auranofin; ADR, adverse drug reaction.

Page 5: Therapy for rheumatoid arthritis: Combinations of disease-modifying drugs and new paradigms of treatment

COMBINATION THERAPY FOR RHEUMATOID ARTHRITIS 25

not seen at 6 and 12 months. The lack of con- tinued statistically significant improvement may be due to the fact that prednisolone was reduced from 20 mg/d at the start of combination therapy to a mean of 13 mg/d at 6 months, and to 11 mg/d at 12 months.

In another open trial, chlorambucil was added to previous DPN 1 g/d or to previously initiated parenteral gold. 27 Other combinations included levamisole with DPN, and parenteral gold with DPN. Remission was most often seen in the par- enteral gold plus DPN and levamisole with DPN patients, during a follow-up of 38 to 113 weeks. Unfortunately, the number of patients in each group was small (five to 12), which makes claims of efficacy less certain. Approximately 25% of pa- tients treated with these combinations experi- enced adverse effects, including herpes zoster in- fection in one patient.

The combination of AZA 2.1 mg/kg/d and parenteral gold 50 mg/mo administered to 70 patients was reported in abstract form.28 Forty- seven percent of patients were “well controlled.” Adverse effects with this combination were re- ported to be no worse than with parenteral gold alone.

MTX has been combined with three different DMARDs. A short report of four patients treated for a mean period of 24 months with MTX mean dose 11.3 mg/wk and SSZ mean dose 2.3 g/d in patients who had not responded to SSZ in three cases and to MTX in one case, suggested that the combination was more effective than either drug alone.29 The authors reported no adverse effects. This combination of two folic acid inhibitors should be monitored carefully, because of a report of potentiated MTX toxicity in a patient receiving simultaneous trimethoprim-sulfamethoxazole.30

The combination of DPN and MTX was re- ported in abstract form.31 Sixteen patients who failed to respond optimally to DPN were given weekly oral MTX, mean dose 10 mg/wk. Dis- continuation due to drug intolerance did not oc- cur. Three patients were lost to follow-up and one patient died of unknown causes. Of the re- maining 12 patients, eight achieved complete re- mission, which was sustained for 7 months in seven of eight patients. The other four patients achieved 50% or greater improvement in impor- tant efficacy measures. The combination of MTX

and DPN was also shown to be corticosteroid- sparing compared with DPN alone.

MTX was added to background parenteral gold therapy. 32 Seven patients who received parenteral gold for a mean of 4 years in a dose of 75 mg/mo were given MTX, mean dose I 1.5 mg/wk, for an average of 19 months. This ab- stract reports either good response or remission in all treated patients. Toxicity was not en- countered.

In the European literature, Rau and Karger treated 46% of 10 1 RA patients with simulta- neous parenteral gold and MTX 7.5 to 15 mg/ wk.33 This combination resulted in a reduction of active joint count and daily corticosteroid re- quirement. The toxicity encountered was no worse than with MTX alone.

Two recent abstracts report divergent results observed with the combination of auranofin (AUR) and MTX. In the first double-blind, ran- domized study, 335 patients initially were given AUR 6 mg/d or MTX 7.5 mg/wk alone.34 A third group was given the combination of AUR 6 mg/ d and MTX 7.5 mg/wk. Two hundred eleven patients completed the trial. The investigators found no statistically significant differences in re- sponse among the groups. Withdrawals for lack of response were less frequent in the group re- ceiving both active agents; however, there were more withdrawals due to adverse effects in that same group.

In the second abstract, 267 patients with active RA initially received a combination of AUR and MTX for 6 months. I6 At 6 months, 89% of eval- uable patients showed clinical benefit defined as greater than 50% reduction in joint count and physician’s global assessment. In a second blinded phase of this trial, placebo was randomly substi- tuted for MTX in half the patients. Previous ben- efits were maintained in 74% of patients who re- mained on combination therapy and 5 1% of patients taking AUR alone. At 6 months’ follow- up, only 14% of patients who received both active agents withdrew because of adverse effects. The investigators believe that the risks of combination therapy were not greater than the risks of either agent alone.

The combination of MTX and HCQ has also been reported. We found that this combination was no more toxic than MTX alone in a retro- spective review of 87 RA patients monitored for

Page 6: Therapy for rheumatoid arthritis: Combinations of disease-modifying drugs and new paradigms of treatment

26

a mean of 42 months.35 More recently, Fries et al reported an unexpected benefit when MTX was combined with HCQ.36 In a computer-gen- erated review of nearly 2,600 RA patients, the percentage of patients with elevated serum trans- aminase levels was statistically significantly lower in those receiving MTX 8.8 mg/wk combined with HCQ (0%) than in patients receiving MTX 9.6 mg/wk alone (2.7%). Figure 1 contrasts serum transaminase levels in a subset of patients from that study.

Among these combinations, those that include CTX run the risk of long-term oncogenesis3’ The combination of MTX and low-dose oral gold is well tolerated. Our own unpublished anecdotal experience suggests that the addition of oral gold to background MTX and HCQ is more effective than MTX and HCQ alone. As with the com- bination of MTX and HCQ, further studies are needed.

The experience with prednisone and CTX suggests an interesting concept. Both increased efficacy and increased toxicity usually occur as the doses of chemotherapeutic agents are in- creased.‘2-‘4 The addition of lowdose prednisone (<O. 1 mg/kg) as a chemotherapeutic-sparing agent might be safer38,39 than increasing the dose of the chemotherapeutic agent.

Combinations Using Two Chemotherapeutic Agents

Whether or not the concept of RA as a benign clonal expansion of poorly regulated B cells has merit 40,41 this analogy has led to the use of com- binations of chemotherapeutic agents for treat- ment (Table 3). 42-50 Scherbel and Harrison were among the first clinicians in the United States to

WILKE AND CLOUGH

combine sequential parenteral nitrogen mustard followed by low-dose pulsed MTX, usually in combination with HCQ. Unfortunately, only the details of the nitrogen mustard therapy were published.51,S2 We continue to use these agents in sequence.53

When chemotherapeutic agents are combined to reduce the frequency and severity of adverse effects, they usually are prescribed in lower doses than when used as monotherapy. McCarty and Carrera were the first to publish results of two chemotherapeutic agents combined in low-dose with HCQ.42 They treated 17 patients with resis- tant RA using roughly */z dose equivalents of CTX and AZA combined with 200 to 400 mg/d of HCQ introduced sequentially. The dose of each chemotherapeutic agent was alternately increased over weeks to months until mild leukopenia oc- curred. Five patients achieved remission, two near-remission, seven partial response, and three showed no response. In a follow-up report, only four of the 16 patients who achieved remission showed worsening of erosions at 3.5 years’ follow- up.43 This response was remarkable, because all these patients had failed previous DMARD ther- apy, which often included parenteral gold and/ or AZA. Unfortunately, after 3.5 years of follow- up, four patients developed malignant neoplasms, which were fatal in three; pneumonia developed in five other patients, which necessitated with- drawal of at least one of the chemotherapeutic agents in two patients. 43 These investigators sug- gested the substitution of another chemothera- peutic agent for CTX. In a recent editorial, the senior author reported that he had successfully treated at least 33 additional patients with the combination of MTX, AZA, and HCQ.S3

Serum Glutamic Oxaloacetic

Traneaminase (SGOT) in units/ml

Methotrexate Alone Methotrexate + Hydroxychloroquine

Fig 1: Twenty-six pa- tients taking either MTX alone or MTX combined with HCQ. (Data from Fries et al.36)

Page 7: Therapy for rheumatoid arthritis: Combinations of disease-modifying drugs and new paradigms of treatment

COMBINATION THERAPY FOR RHEUMATOID ARTHRITIS 27

Table 3: Series in Which Two Chemotherapeutic Agents Were Used Simultaneously

No. of Chemotherapeutic

Reference Patients Agents DMARDs or Other Efficacy Toxicity

McCa@ 3 1 AZA 74 mg/d HCQ 2 11 mg/d 16 complete remission 4 malignancies

Csuka43 CTX 30 mg/d 14 excellent to good 2 pulmonary

1 no response infections

1 thrombocytopenia

ButleP 16 CTX 25 mg/d HCQ 400 mg/d Radiologic recortication 1 stomatitis

Tiliakos45.4B MTX 5 mg/wk 7/l 2 and 50-90%

improvement in

efficacy measures

in all

Fosdick4’ 16 MTX

I

doses not None 6 complete remission No serious toxicity

CTX specified g partial remission

Biro- 37 MTX g mg/wk HCQ 200-400 mg/d 4 joint count 6 1% 3 leukopenia

Wilke49 AZA 67 mg/d 4 WSR47% 1 herpes roster

1 bacterial pneumonia

LangevitP 12 MTX 5-12.5 mg/wk HCQ 200-400 mg/d J- joint count 59% 3 nausea

AZA 25-50 mg/d J WSR38%

Abbreviations: AZA, azathioprine; CTX, cyclophosphamide; HCQ, hydroxychloroquine; MTX, methotrexate; WSR, Westergren sedi- mentation rate.

Butler and Tiliakos reported a controlled trial in which two groups of patients, who received either MTX 5 mg/wk, CTX 25 mg/d, and HCQ 400 mg/d or MTX alone 10 to 15 mg/wk, were compared with a third group that continued pre- vious treatment without chemotherapeutic agents.44*45 During a follow-up ranging from 8 to 23 months, patients who received the two che- motherapeutic agents showed the most improve- ment. This experience was updated and 16 pa- tients were monitored for 2 years4’j These patients experienced 50% to 90% improvement in duration of morning stiffness, grip strength, number of painful joints, and Westergren sedi- mentation rate (WSR). X-ray evaluation showed recortication in seven patients. Adverse effects were rare; treatment was interrupted in only one patient due to stomatitis.

Fosdick reported 16 patients treated with MTX and CTX. MTX was started after CTX alone failed to produce the desired improvement.47 Few details are given in this abstract. Six patients achieved complete remission and nine, partial remission. Side effects included “self-limited sto- matitis” in all patients and elevation of a liver function test in one patient. No irreversible bone marrow depression, thrombocytopenia, or infec- tion were reported.

We reported our positive experience with combination therapy using AZA, MTX, and HCQ in preliminary form in 1987.48 The original 20 patients had failed previous single-agent che- motherapeutic treatment, including MTX in 17 patients and AZA in three. Either AZA or MTX was then added to the other stable chemothera- peutic agent. The majority of patients were also receiving HCQ.

Since then, we have retrieved data on all pa- tients in the department treated with this com- bination and lengthened the duration of follow- up. We now have information on 37 patients who received AZA (mean dose, 67 mg/d) and MTX (mean dose, 9 mg/wk) for a mean of 30 months. Thirty-one patients simultaneously were taking HCQ 200 to 400 mg/d. Active joint count was reduced from 14 at time zero to 9 at 12 months and 5 at 18 months. The WSR decreased from 48 mm/h at time zero to 36 mm/h at 12 months and 26 mm/h at 18 months. The frequency of serious toxicity was low. Three patients experi- enced reversible leukopenia, one patient herpes zoster, and one patient bacterial pneumonia. Other adverse effects included gastrointestinal toxicity in seven patients, elevation of serum transaminase level in two, alopecia in one, and hepatic fibrosis in a patient who had previously

Page 8: Therapy for rheumatoid arthritis: Combinations of disease-modifying drugs and new paradigms of treatment

28 WILKE AND CLOLJGH

Table 4: Disease Features of Rheumatoid Arthritis and Other Factors That Predict a Poor Outcome

in Selected Prospective Series

Follow- No. of Reference Year Up (vr) Patients IFC t APR AJC t RF Other

DuthieBS 1964 9 200 x X X X

McKonkey@ 1972 0.5-6 987 X

Jacoby6’ 1973 a-14 100 X X

Fleminge’ 1976 4.5 102 X X X X 4 Grip strength

Fcigenbaums* 1979 >5 50 X X 4 Grip strength, psychological

stress

Sjoblomao i 984 2 103 X X 4 Hgb concentration

Raskars2 1984 15 100 X X X 4 Hgb concentration presence

of rheumatoid nodules

Kaarelas3 I 985 a 200 X X Worsening radiographic grade,

symmetric swelling, EMS

> T HR

Sherrers9 1986 11.9 681 X Worsening radiographic grade,

t age, female sex

Scott” 1987 20 112 X X t Age, late presentation

Mottonen” 1988 2 58 X X X “Disease activity”

Erhardtsg i 989 a 64 Extra-articular involvement,

t cryoglobulins, (+) PASCA

Reilly8 1990 25 100 X X f Hgb concentration

Abbreviations: IFC, impaired initial functional capacity or worsening functional capacity; + APR, chronic elevation of acute-phase reactants; AJC, active joint count inversely related to prognosis; t RF, increasing or high titer rheumatoid factor; 4 Hgb, decreased hemoglobin concentration; EMS, duration of early morning stiffness; PASCA, positive test for precipitating antibodies to soluble cellular antigens.

taken MTX for more than 10 years. This low incidence may be due to the trial design, which excluded patients who were intolerant to che- motherapeutic agents. Patients who failed monotherapy had done so after taking MTX for a mean of 58 months or AZA for 15 months, proving they could tolerate chemotherapy. Our treatment also may have been made safer due to concomitant therapy; the majority of patients were receiving HCQ36 and 2 1 of 37 patients were receiving weekly folic acid.s5,56

This same combination was reported in the British literature.50 Of 12 patients observed for 14 to 18 months, the mean joint count before treatment was 11 .O and after treatment was 4.5. The WSR decreased from 45 mm/h to 28 mm/ h. Four patients were withdrawn from treatment, due to nausea in one and inefficacy in three. These results are similar to the Cleveland Clinic experience.

The combination of AZA and MTX seems surprisingly safe in the small number of patients reported. There is theoretical rationale for ex- pecting synergy between these agents at the in-

flammatory cell leve157 and with regard to drug metabolism.58 The de novo pathway of purine synthesis is blocked by MTX. Further purine synthesis is dependent on salvage pathways, which are in turn dependent on the enzyme hy- poxanthine-guanine phosphoribosyltransferase. This enzyme is also necessary for purine activa- tion. As this enzyme becomes more active, it converts a higher percentage of intracellular AZA to its effective form. Above the intracellular level, MTX inhibits xanthine oxidase, the enzyme that

Total - IO-21 months

Fig 2: Time course of therapy based on the

“treatment pyramid.”

Page 9: Therapy for rheumatoid arthritis: Combinations of disease-modifying drugs and new paradigms of treatment

COMBINATION THERAPY FOR RHEUMATOID ARTHRITIS 29

catabolizes 6-mercaptopurine, resulting in higher blood levels of active AZA.

If this drug combination is shown to be more effective than monotherapy with either agent in controlled studies, it may be a useful alternative to higher doses of MTX or AZA.

GOALS OF THERAPY

Eficacy Measures

The primary goal of therapy in RA is to im- prove or at least maintain function. Many ret- rospective and prospective series have attempted to discover which features or other factors present in early RA and/or during the disease process

predict poor functional outcome8,59-70 (Table 4). Older age, high titer of rheumatoid factor, early functional decline, abnormal psychological sta- tus, extra-articular involvement, low hemoglobin level, and poor initial radiographic grade predict later impaired function, morbidity, and some- times mortality. Clearly, one of the measures that affects function is radiographic change. Unfor- tunately, radiographic studies are expensive and subject to interobserver error.

Two predictive measures common to most long-term studies are the initial active joint count and sustained elevation of acute-phase reactants. That the number of active joints might predict

--A Other Combinations

or CSA or Biologicals or Apheresis or Tidal Lavage

______________

I AU (P) or DPN ! orMTXorAZA I k

I

z 8 w -

I I ! HCQ I + I + +

NSAlDs I + I 1 + (Analgesic?) , + (Analgesic?)

OTiPTlPt. Ed. ; + I + I + I I i I I

Mild (O-4) Moderate (5-7) Severe (8-l 3)

Disease Severity Classification

Reserved

Fig 3: The initial level of therapy is based on the sum of the disease activity index (Al; Table 5) and the

prognostic factor score (Table 6). Subsequent therapy decisions depend only on the disease Al (see text).

Therapies enclosed in the “consider” block may be used, but are not mandatory. This allows individual

flexibility while maintaining the spirit of the paradigm. The “reserved” step should not be used as initial

therapy and is only used if patients fail step 3, “severe” disease. Al Level 1: joint aspiration, corticosteroid

joint injectkrn, intramuscular administration of corticosteroids and/or corticotropin, and oral corticosteroid

(0.1 mg/kg prednisone equivalent). Al Level 2: includes “induction therapies” with intramuscular or joint

injection of chemotherapeutic agents, intravenous chemotherapeutic agents such as nitrogen mustard,

and intravenous high-dose “pulsed” corticosteroids. HCQ, hydroxychloroquine; NSAIDs, nonsteroidal

antilnflammatory drugs; OT, occupational therapy; PT. physical therapy; ED, patient education; AU (0).

oral gold; SSZ, sulfasalarine; MTX, mathotrexate; AU (P), parenteral gold; DPN, o-penicillamine; AZA,

azathioprfne; CSA, cyclosporine A.

Page 10: Therapy for rheumatoid arthritis: Combinations of disease-modifying drugs and new paradigms of treatment

30 WILKE AND CLOUGH

Table 5: Disease Activity

Disease

Activity

Pain

EMS (min)

S/T joints

WSR (min/h)

or

CRP (mg/dL)

Total ooints

Inactive Subacute Active

Marked

Response or Partial Poor

Remission Score Response Score Response Score

None 0 Intermittent 1 Constant 2

<15 0 15-30 1 >30 2

o-2 0 3-5 1 >5 2

<30 30-45 >45

0 1 2

<2.0 2.0-2.6 >2.6

0 4 8

NOTE. Disease activity index has been adapted from a drug response index conceived and tested by Scott et al.*‘,87 One numerical

value is assigned to each efficacy measure. Grip strength was not included because of diurnal variation and the inherent problems of

interpretation due to simultaneous median neuropathy in some patients, and fixed deformities and degenerative changes at the carpal/

metacarpal joint of the thumb in others.

Abbreviations: EMS, duration of early morning stiffness; S/T joints, total count of swollen and/or tender joints, ie, a joint that is swollen

and/or tender is given a value of 1; WSR, Westergren sedimentation rate; CRP, C-reactive protein.

declining functional capacity and worsening of the radiographic index is not surprising. In a sur- vey of 57 experienced rheumatologists, the joint tenderness count was the most popular measure of drug effectiveness. 5g Of interest, the WSR was among the least-preferred measures, despite the ability of “disease-modifying” agents to reduce acute-phase reactants levels5’

The magnitude of elevation of acute-phase reactants correlates well with radiographic grade5%72>73 and with active joint count.8,5g-62,- 65,66,70 Three series have found more severe in- volvement of peripheral joints72,73 and a higher rate of new bone erosions73 in patients with RA who have persistently elevated acute-phase reac- tants. In another prospective study of 64 patients who received second-line drugs for an adequate time period, patients who maintained a persis- tently low WSR showed less radiographic pro- gression than did patients with higher values.74 In a trial of 67 patients with RA, only those who received drugs that lowered the WSR showed a trend toward reduced rate of articular erosions.75

The prospective series by Dawes et al deserves special attention. 76 One hundred fifty patients with RA who received DMARDs were assessed during 1 year of therapy. Radiographs were ob- tained at the start, at 6 and 12 months. The WSR and CRP were measured regularly. At 12 months, patients were divided into groups based on the values for the WSR and CRP. At 6 months, all

patients showed significant worsening of radio- graphic scores compared with baseline. However, at 12 months, patients in whom the acute-phase reactants were normalized showed no progres- sion. In contrast, patients in whom these mea- sures were intermittently elevated or always el- evated showed radiographic deterioration. This study suggests that in a large population of pa- tients with RA, radiographic deterioration is minimized when acute-phase reactants are nor- malized.

If the degree of eventual physical dysfunction is directly related to the number of a&&d joints and the height of the elevation of acute-phase reactants, the objective goal of therapy must be to reduce the number of affected joints and to normalize acute-phase reactants. This should be accomplished quickly, soon after the diagnosis of RA is certain and before cartilage is damagecL40 Effective early treatment is necessary, because radiographic damage develops in the fnst 2 years in the majority of patients with moderate to se- vere disease.77~78 In addition, effective treatment may better control irreversible joint damage, which results in radiographic change.”

NEWER PARADIGMS OF TREATMENT

Strict adherence to a pyramidal approach to therapy may delay the initiation of effective mea- sures for many months to years in patients with moderate to severe RA (Fig 2). Because no rev-

Page 11: Therapy for rheumatoid arthritis: Combinations of disease-modifying drugs and new paradigms of treatment

COMBINATION THERAPY FOR RHEUMATOID ARTHRITIS 31

Table 6: Prognostic Factors

Factor Score

Deteriorating functional capacity* 1

Sustained (~6 mo) elevated WSR 1

RF titer 2 1:640 Latex 1 Extra-articular diseaset 1

Radiologic joint damage 1

Total points 5

* Includes worsening functional class, and/or change in employ-

ment capacity.

t Includes rheumatoid nodules, inflammatory eye disease, sicca

complex, vasculitis, and pleural or pulmonary involvement.

Abbreviations: WSR, Westergren sedimentation rate; RF, rheu-

matoid factor.

olutionary new modalities are available, new philosophies of therapy are necessary. Until safer and/or more effective treatments are available, we must use those we have innovatively. Wilske and Healey have suggested a “step-down bridge” paradigm in which an initial dose of prednisone 10 mg/d is administered for 1 month.” Patients who fail to respond adequately are then given the combination of antimalarials, oral gold, paren- teral gold, and MTX. Other DMARDs may be substituted. After 3 months of multiple drug therapy, tapering is attempted with the hope that antimalarials will maintain disease control.

Another strategy, the “sawtooth” paradigm, depends on setting a ceiling for disability.‘l Dis- ease-modifying drug therapy is serially changed whenever this ceiling is reached, in an attempt to maintain early disease control.

The step-down bridge paradigm certainly al- lows vigorous early therapy for RA. Unfortu- nately, it may place some patients with only moderate disease at unnecessary risk of adverse effects from exposure to multiple DMARDs. In addition, long-term maintenance of moderate to severe RA with HCQ alone has not been dem- onstrated.

Table 7: Disease Severity Classification

Score Classification

o-4 Mild

5-7 Moderate

B-13 Severe

NOTE. Disease activity index + prognostic factor score = disease

severity classification.

Table 8: Other Factors That Influence

Treatment Decisions I

A.

8.

C.

D.

E.

F.

G.

Drug allergy or intolerance

Age

Patient expectations

Coexisting disease

1 Peptic ulcer disease

2. Diabetes mellitus

3. Malignancy

4. Macular degeneration

5. Active infection

6. impaired renal function

7. Liver disease

Drug interaction

1. Anticoagulants

2. Anticonvulsants

3. Alcohol

4. Allopurinol

5. Antibiotics

6. Addiction

Noncompliance

Drug failure and need for surgery

The sawtooth paradigm assumes that when monotherapy or combinations of drugs result in disease control, this control will disappear over time. Although that phenomenon certainly oc- curs with gold therapy, it is usually because over time the dose and frequency of administration are changed. 82 That phenomenon does not ap pear to occur with some DMARDs, including MTX.83 In addition, initial disease control may be difficult to realize. Monotherapy with conser- vative DMARDs is only effective in a small per- centage of patients. 2,84,85 Even with chemother- apeutic agents, tight control is rarely achieved. In a long-term prospective study, 16 patients who continued to receive MTX for 36 months showed a mean WSR of 53 mm/h and an active joint count of 8.’ ’ This apparent limited response to monotherapy in some patients dictates that com- binations of DMARDs will likely be used early in the sawtooth paradigm. But this paradigm does not provide rationale for specific therapeutic choices.

To expose only those patients with sufficiently active disease to risk the potential adverse reac- tions of combination therapy, the Cleveland Clinic group has designed a graduated paradigm of treatment: “graduated-step therapy” (Fig 3). In this paradigm, at or near the time of diagnosis,

Page 12: Therapy for rheumatoid arthritis: Combinations of disease-modifying drugs and new paradigms of treatment

32

disease activity is computed by summing vari- ables derived from a response index developed and tested by Scott et al86*87 (Table 5). The nu- merical value for disease activity is then added to another value obtained by summing arbitrarily chosen prognostic indicators (Table 6). This sum is the numerical value for disease severity (Table 7). Patients are then entered into the paradigm and treated with the modalities corresponding to this value (Fig 3).

At 3 to 6 months, patients are reevaluated, but only the disease activity index is computed. Pa- tients with values of 0 to 3 are designated as hav- ing inactive disease and to have responded to therapy. They may be continued at the present treatment level, or it may be reduced. Patients with values of 6 to 8 have active disease that has not responded to therapy and treatment should be escalated. Patients with values of 4 to 5 have subacute disease. Decisions regarding change of therapy for these patients will depend on changes of efficacy measures that have not been included in the disease activity criteria, including time of

WILKE AND CLOUGH

onset of afternoon fatigue, change in grip strength, change in hemoglobin concentration, or the onset or worsening of extra-articular dis- ease. Other factors that might influence treatment decisions are also considered (Table 8).

This paradigm is, in fact, a formalized version of the past and current practice of our group. As we enter and prospectively monitor our patients using this paradigm, we will evaluate efficacy and toxicity on a regular basis. We anticipate that changes in the specific recommendations at each step will be necessary as this concept evolves.

New philosophies of therapy represent the best chance for contemporary “breakthroughs” in the treatment of RA. The early use of compatible combinations of drugs and modalities that are carefully chosen and appropriate for the individ- ual level of disease activity appears to be a rational paradigm to test in well-designed prospective trials.

ACKNOWLEDGMENT

The authors would like to thank Ann W. Fuentes for her technical assistance on the manuscript.

REFERENCES

1. Goldman JA, Hess EV: Treatment of rheumatoid ar- thritis- 1970. Bull Rbeum Dis 2 1:609-6 12, 1970

2. Grindulis KA, McKonkey B: Outcome of attempts to treat rheumatoid arthritis with gold, penicillamine, sulfasal- azine, or dapsone. Ann Rheum Dis 43:398-401, 1984

3. Thomas MH, Rothermich NO, Philips VK, et al: Gold vs. Dpenicillamine double blind study and follow-up. J Rheumatol 11:764-767, 1984

4. Black RL, O’Brien WM, VanScot El, et al: Methotrexate therapy in psoriatic arthritis: Double blind study on 21 pa- tients. JAMA 1891743-747, 1964

5. Willkens RF: Resolve: Methotmxate is the drug of choice after NSAIDs in rheumatoid arthritis. Semin Arthritis Rheum 20:76-80, 1990

6. Furst DE: Proposition: Methotrexate should not be the first second-line agent to be used in rheumatoid arthritis if NSAIDs fail. Semin Arthritis Rheum 20:69-75, 1990

7. Katz WA, Gottlieb NL, Jaffe I, et al: Criteria for initiat- ing therapy with disease-modifying anti-rheumatic drugs (DMARDs) in rheumatoid arthritis (RA). Arthritis Rheum 3OS61, 1987 (suppl)

8. Reilly PA, Cosh JA, Maddison PJ, et al: Mortality and survival in rheumatoid arthritis. A 25year prospective study of 100 patients. Ann Rheum Dis 49:363-369, 1990

9. Pincus T: Is mortality increased in rheumatoid arthritis? J Musculoskel Med 5:27-46, 1988

10. Zatarain E, Williams CA, Fries JF: Comparison of ad- verse reactions of methotrexate and other disease-modifying drugs. Arthritis Rheum 3lSll6, 1988 (suppl)

11. Weinblatt ME, Trentham DE, Fraser PA, et al: Long-

term prospective trial of lowdose metbotrexate in rheumatoid arthritis. Arthritis Rheum 31:167-175, 1988

12. Urowitz MB, Gordon DA, Smythe HA, et al: Azathio- prine in rheumatoid arthritis. A double-blind, crossover study. Arthritis Rheum 16:41 l-418, 1973

13. Furst DE, Koehnke R, Burmeister LF, et al: Increasing methotrexate effect with increasing dose in the treatment of resistant rheumatoid arthritis. J Rheumatoll6:3 13-320, 1989

14. Thompson RN, Watts C, Edelman J, et al: A controlled two-center trial of parenteral methotmxate therapy for re- fractory rheumatoid arthritis. J Rheumatol 11:760-763, 1984

15. Woodland J, Chaput de Saintonge DM, Evans SJ, et al: Azathioprine in rheumatoid arthritis: Double-blind study of full vs. half doses vs. placebo. Ann Rheum Dis 40:355-359, 1981

16. Kantor SM, Wallin BA, Grier CJ, et al: Combination of auranofin and methotrexate as initial DMARD in RA. Ar- thritis Rheum 33:S60, 1990 (suppl)

17. Lipsky PE, Ziff M: Inhibition of human helper T-cell function in vitro by bpenicillamine and CuSO+ J Clin Invest 65:1069-1076, 1980

18. Jones CJP, Jayson MIV: Chloroquine: Its effect on leu- kocyte auto and heterophagocytosis. Ann Rheum Dis 43:205- 212, 1984

19. Bunch TW, G’Dulfy JD, Tompkins RB, et al: Con- trolled trial of hydroxychloroquine and Dpenicillamine singly and in combination in the treatment of rheumatoid arthritis. Arthritis Rheum 27~267-276, 1984

20. Gibson T, Emery P, Armstrong RD, et al: Combined openicillamine and chloroquine treatment of rheumatoid

Page 13: Therapy for rheumatoid arthritis: Combinations of disease-modifying drugs and new paradigms of treatment

COMBINATION THERAPY FOR RHEUMATOID ARTHRITIS 33

arthritis-A comparative study. Br J Rheumatol26:279-284, 1987

2 I. Scott DL, Dawes PT, Tunn E, et al: Combination ther- apy with gold and hydroxychloroquine in rheumatoid arthritis: A prospective, randomized, placebo-controlled study. Br J Rheumatol 28:128-133, 1989

22. Singleton PT, Cervantes AG: Chrysotherapy and con- comitant use of antimalarial drug therapy in rheumatoid ar- thritis. Arthritis Rheum 25:Sl15, 1982 (suppl)

23. Taggart AJ, Hill J, Astbury C, et al: Sulfasalazine alone or in combination with rr-penicillamine in rheumatoid ar- thritis. Br J Rheumatol 26:32-36, 1987

24. Farr M, Kitas G, Bacon PA: Sulfasalazine in rheu- matoid arthritis: Combination therapy with Dpenicillamine or sodium aurothiomalate. Clin Rheumatol 7:242-248, 1988

25. Smyth CJ, Bartholomew BA, Mills DM, et al: Cyclo- phosphamide therapy for rheumatoid arthritis. Arch Intern Med 135:789-793, 1975

26. Binder AI, Ansell BM, Denman AM: Intensive im- munosuppression in intractable rheumatoid arthritis. Br J Rheumatol 25:380-383, 1986

27. Bitter T: Combined disease-modifying chemotherapy for intractable rheumatoid arthritis. Clin Rheum Dis 10:4 17- 428, 1984

28. Lewis P, Hazleman B, Bulgen D, et al: Clinical and immunological study of high dose azathioprine combined with gold therapy, in Gordon JL, Hazleman BL (eds): Rheumatoid Arthritis: Cellular Pathology and Pharmacology. Amsterdam, Netherlands, Elsevier North Holland, 1977, p 280

29. Shiroky JB, Watts CS, Neville C: Combination meth- otrexate and sulfasalazine in the management of rheumatoid arthritis: Case observations. Arthritis Rheum 32:1160-l 164, 1989

30. Thomas MH, Gutterman LA: Methotrexate toxicity in a patient receiving trimethoprim-sulfamethoxazole. J Rheumatol 13:440-44 1, 1986

3 1. Lee S, Solomon G: Treatment of rheumatoid arthritis (RA) with combination of Dpenicillamine (DPCN) and methotrexate (MTX). Arthritis Rheum 33:R39, 1990 (suppl)

32. Brawer AE: The combined use of oral methotrexate and intramuscular gold in rheumatoid arthritis. Arthritis Rheum 3l:RlO, 1988 (suppl)

33. Rau R, Karger T: The European experience, in Wilke WS (ed): Methotrexate Therapy in Rheumatic Disease. New York, NY, Marcel Dekker, 1989, pp 57-90

34. Wiiams HJ, Ward JR, Reading JC, for the Cooperative Systematic Studies of the Rheumatic Diseases: Comparison of auranofin, methotrexate, and the combination of both in the treatment of rheumatoid arthritis. Arthritis Rheum 33: s 10, 1990 (suppl)

35. Wilke WS, Calabrese LH, Km11 PL, et al: Incidence of toxicity in patients with rheumatoid arthritis treated with methotrexate, in Rau R (ed): Low-Dose Methotrexate Therapy in Rheumatic Disease, ~019. Base], Switzerland, Karger, 1986, pp 134-144

36. Fries JF, Singh G, Lenert L, et al: Aspirin, hydroxy- chloroquine, and hepatic enzyme abnormalities with meth- otrexate in rheumatoid arthritis. Arthritis Rheum 33:16 1 l- 1619, 1990

37. Baker GL, Kahl LE, Zee BC, et al: Malignancy follow- ing treatment of rheumatoid arthritis with cyclophosphamide.

Long-term case-control follow-up study. Am J Med 83: l-9, 1987

38. Harris ED Jr, Emkey RD, Nichols J, et al: Low-dose prednisone therapy in rheumatoid arthritis: A double blind study. J Rheumatol IO:7 13-72 1, 1983

39. Byron MA, Kirwan JR: Corticosteroids in rheumatoid arthritis: Is a trial of their ‘disease modifying” potential fea- sible? Ann Rheum Dis 46:171-173, 1986

40. Bitter T: Combined disease-modifying chemotherapy for intractable rheumatoid arthritis. Clin Rheum Dis 10:4 13- 428, 1984

41. Harris ED Jr: Rheumatoid arthritis. Pathophysiology and implications for therapy. N Engl J Med 322: 1277-1289, 1990

42. McCarty DJ, Carrera GF: Intractable rheumatoid ar- thritis: Treatment with combined cyclophosphamide, aza- thioprine and hydroxychloroquine. JAMA 248: 17 18- 1723, 1982

43. Csuka ME, Carrera GF, McCarty DJ: Treatment of intractable rheumatoid arthritis with combined cyclophos- phamide, azathioprine and hydroxychloroquine: A follow-up study. JAMA 25512315-2319, 1986

44. Tiliakos NA: Single-agent versus combination cytotoxic therapy: The case for combination therapy, in Willkens RF, Dahl SL (eds): Therapeutic Controversies in Rheumatic Dis- ease. Philadelphia, PA, Grune & Stratton, 1987, pp 295-305

45. Tiliakos NA: Low-dose cytotoxic drug combination therapy in intractable rheumatoid arthritis: Two years later. Arthritis Rheum 29:S79, 1986 (suppl)

46. Butler DE, Tiliakos NA: Low-dose cytotoxic drug combination therapy in rheumatoid arthritis. Arthritis Rheum 28:S15, 1985 (suppl)

47. Fosdick WM: Combined cytotoxic therapy in rheu- matoid arthritis (RA). Arthritis Rheum 14:161-162, 1971

(SUPPl) 48. Biro JA, Segal AM, Mackenzie AH, et al: The com-

bination of methotrexate (MTX) and azathiprine (AZA) for resistant rheumatoid arthritis (RA). Arthritis Rheum 30:s 18, 1987 (suppl)

49. Wilke WS, Sherrer YS, Clough JD: Combination che- motherapy for severe rheumatoid arthritis. Intern Med Spe- cialist 10:59-76, 1989

50. Langevitz P, Kaplinski N, Ehrentield M, et al: Intrac- table U-Treatment with combined methotrexate, azathio- prine, and hydroxychlorcquine. Br J Rheumatol28:27 l-272, 1989

5 1. Scherbel AL: Intravenous administration of nitrogen mustard alone and in combination with corticotropin for rheumatoid arthritis. Cleve Clin Q 24:71-76, 1957

52. Scherbel AL, Harrison JW: Chemotherapy in rheu- matoid arthritis. Postgrad Med 26:857-864, 1957

53. Wilke WS, Sexton C, Steck W: Parenteral nitrogen mustard for inflammatory arthritis. Cleve Clin J Med 57:643- 646, 1990

54. McCarty DJ: Suppress rheumatoid inflammation early and leave the pyramid to the Egyptians. J Rheumatol 17: 1115-1118, 1990

55. Morgan SL, Baggott JE, Vaughn WH, et al: The effect of folic acid supplementation on toxicity of low-dose meth- otrexate in patients with rheumatoid arthritis. Arthritis Rheum 33:9-18, 1990

Page 14: Therapy for rheumatoid arthritis: Combinations of disease-modifying drugs and new paradigms of treatment

34 WILKE AND CLOUGH

56. Stewart KA, Mackenzie AH, Clough JD, et al: Folate- supplementation in methotrexate-treated rheumatoid arthritis patients. Semin Arthritis Rheum 20:332-338, 1991

57. Jolivet J, Chabner BA: Methotrexate drug interactions. Intern Med Specialist 5:45-5 1, 1984

58. Balis FM, Holcenberg JS, Zimm S, et al: The effect of methotrexate on the hioavailability of oral 6-mercaptopurine. Clin Pharmacol Ther 41:384-387, 1987

59. Sherrer YS, Bloch DA, Mitchell DM, et al: The de- velopment of disability in rheumatoid arthritis. Arthritis Rheum 29:494-500, 1986

60. Sjoblom KG, Saxne T, Petterson H, et al: Factors re- lated to the progression of joint destruction in rheumatoid arthritis. Stand J Rheumatol 13:2 l-27, 1984

6 1. Fleming A, Crown JM, Corbett M: Prognostic value of early features in rheumatoid disease.. Br Med J 1: 1243- 1245, 1976

62. Rasker JJ, Cosh JA: The natural history of rheumatoid arthritis: A 15 year follow-up study. The prognostic significance of features noted in the first year. Clin Rheumatol 3:l l-20, 1984

63. Kaarela K: Prognostic factors and diagnostic criteria in early rheumatoid arthritis. Stand J Rheumatol 57:1-54, 1985 (suppl)

64. Scott DL, Coulton BL, Symmons DPM, et al: Long- term outcome of treating rheumatoid arthritis: Results after 20years. Lancet 1:1108-1111, 1987

65. Duthie J Jr, Brown PE, Truelove LH, et al: Course and prognosis in rheumatoid arthritis. A further report. Ann Rheum Dis 23:193-204, 1964

66. McKonkey B, Crockson RA, Crockson AP: The as- sessment of rheumatoid arthritis: A study based on measure- ments of the serum acute phase reactants. Q J Med 192: 115- 125, 1972

67. Jacoby RK, Jayson MIV, Cosh JA: Onset, early stages and prognosis of rheumatoid arthritis. A clinical study of 100 patients with 1 l-year follow-up. Br Med J 2:96-100, 1973

68. Feigenbaum SL, Masi AT, Kaplan SB: Prognosis in rheumatoid arthritis. A longitudinal study of newly diagnosed younger adult patients. Am J Med 66:377-384, 1979

69. Erhardt CC, Mumford PA, Venables PJW, et al: Factors predicting a poor life prognosis in rheumatoid arthritis. An 8 year prospective study. Ann Rheum Dis 48:7- 13, 1989

70. Mottonen TT: Prediction of erosiveness and rate of development of new erosions in early rheumatoid arthritis. Ann Rheum Dis 47:648-653, 1988

7 1. Bombardier C, Tugwell P, Sinclair A, et ah Preference for endpoint measures in clinical trials: Results of structured workshops. J Rheumatol9:798-801, 1982

72. Thompson PW, Silman AJ, Kitwan JR, et al: Articular indices of joint inflammation in rheumatoid arthritis. Cor- relation with the acute phase response. Arthritis Rheum 30: 6 18-623, 1987

73. Amos R, Constable T, Crockson R, et al: Rheumatoid arthritis: Relation of serum C-reactive protein and erythrocyte sedimentation rates to radiographic changes. Br Med J 1:195- 197, 1977

74. Scott DL, Grindulis KA, Struthers GR, et al: Progres- sion of radiological changes in rheumatoid arthritis. Ann Rheum Dis 43:8-17, 1984

75. Pullar T, Hunter JA, Cape11 HA: Does second-line therapy affect the radiological progression of rheumatoid ar- thritis? Ann Rheum Dis 43: 18-23, 1984

76. Dawes PT, Fowler PD, Clarke S, et al: Rheumatoid arthritis: Treatment which controls the c-reactive protein and erythrocyte sedimentation rate reduces radiological progres- sion. Br J Rheumatol 25:44-49, 1986

77. Larsen A, Thoen J: Hand radiographs of 200 patients with rheumatoid arthritis repeated after an interval of one year. Stand J Rheumatol 16:395-401, 1987

78. Brook A, Corbett M: Radiographic changes in early rheumatoid disease. Ann Rheum Dis 36:71-73, 1977

79. Lukkainen R, Kajander A, Isomaki H: Effect of gold on progression of erosions in rheumatoid arthritis. Better re- sults with early treatment. Stand J Rheumatol6: 189-192,1977

80. Wilske KR, Healey LA: Remodeling the pyramid-A concept whose time has come. J Rheumatol 16565-567, 1989

81. Fries JF: Re-evaluating the therapeutic approach to rheumatoid arthritis: The “sawtooth” strategy. J Rheumatol 17:12-15, 1990 (suppl)

82. Thomas MH, Rothermich NO, Philips VK, et al: Gold vs. Dpenicillamine double blind study and follow-up. J Rheumatol 11:764-767, 1984

83. Weinstein A, Marlowe S, Kom J, et al: Low-dose methotrexate treatment of rheumatoid arthritis. Long-term observations. Am J Med 79:331-337, 1985

84. Ferraccioli GF, Salaffi F, Nervetti A, et al: Long-term outcome with gold thiosulfate and tiopronin in 200 rheu- matoid patients. Clin Exp Rheumatol 7:577-581, 1989

85. Cape11 HA, Lewis D, Carey J: A three-year follow-up of patients allocated to placebo or oral or injectable gold ther- apy for rheumatoid arthritis. Ann Rheum Dis 45:705-711, 1986

86. Scott DL, Spector TD, McKonkey BL: What should we hope to achieve when treating rheumatoid arthritis? Ann Rheum Dis 48:256-26 1, 1989

87. Scott DL, Dacre JE, Greenwood A, et al: Can we de- velop simple response criteria for slow acting anti-rheumatic drugs? Ann Rheum Dis 49:196-198, 1990

88. Wright V, Dixson JS, Bird HA: Therapeutic significance of laboratory results in rheumatic disease. Semin Arthritis Rheum 15:8-13, 1985

89. Carvalho A, Graudal H: Radiographic progression of rheumatoid arthritis related to some clinical and laboratory parameters. Acta Radio1 Diagn 21:551-555, 1980

90. Fuchs HA, Caye JJ, Callahan LF, et al: Evidence of sign&cant radiographic damage in rheumatoid arthritis within the first two years of disease. J Rheumatol 16:585-591, 1989