Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Aug 1.
Published in final edited form as: Rheum Dis Clin North Am. 2018 Jun 12;44(3):371–391. doi: 10.1016/j.rdc.2018.03.010

Pharmacotherapy Pearls in Rheumatology for the Care of Older Adult Patients: Focus on Oral Disease-Modifying Antirheumatic Drugs and the Newest Small Molecule inhibitors

Blas Y Betancourt 1, Ann Biehl 2, James D Katz 1, Ananta Subedi 1
PMCID: PMC6047536  NIHMSID: NIHMS957460  PMID: 30001781

Abstract

Providing safe and effective pharmacotherapy to geriatric patients with rheumatological disorders is challenging. Multidisciplinary care involving rheumatologists, primary care physicians and other specialties can optimize benefit and reduce adverse outcomes. Oral disease-modifying antirheumatic drugs, including methotrexate, hydroxychloroquine, sulfasalazine, and leflunomide, and the small molecule inhibitors tofacitinib and apremilast have distinctive monitoring requirements and specific adverse reaction profiles. This article provides clinically relevant pearls for use of these interventions in older patients.

Keywords: Geriatrics, DMARDs, Rheumatology, Rheumatoid Arthritis, Tofacitinib, Apremilast

Introduction

Providing safe and effective pharmacotherapy to the geriatric patient population is an ongoing struggle for health care providers. The incidence of rheumatological disorders increases with advancing age. Recent National Health Interview Survey data (2013 – 2015) observed a prevalence of physician-diagnosed arthritis among adults aged 65 years and older approaches 50% with 44% of these patients having related activity limitation.1 It is estimated that rheumatoid arthritis (RA) affects 0.5% to 1% of the adult population in developed countries. This translates to approximately 1.3 million Americans, with an increasing prevalence with advancing age.2

Oral disease-modifying antirheumatic drugs (DMARDs) are not only effective in reducing morbidity and improving quality of life but can also have a positive impact on mortality.3 However, DMARDs alter the host immune system and could create a risk of significant adverse events including infection and malignancy. A rheumatologist involved in the care of the elderly should be aware of specific adverse reactions and drug interactions associated with the use of oral DMARDs.

Older patients are at increased risk for adverse drug reactions. Budnitz and colleagues found individuals over the age of 65 were more likely than younger persons to have adverse drug reactions requiring emergency room visits and hospitalization.4 Such therapeutic misadventures in geriatric patients can be due to age-related changes in pharmacokinetics and pharmacodynamics, polypharmacy contributing to increased risk of clinically significant drug-drug interactions, and alterations in cognitive faculties that impair health literacy and therapeutic adherence.511 These problems are likely compounded by age bias, manifesting as a reluctance to aggressively treat older patients, as well as economic barriers.12,13 Management of rheumatologic conditions also carries special risk due to rapidly evolving use of novel therapeutic agents with limited data guiding their use in geriatric patients.

This review provides an update regarding commonly used oral DMARDs for the treatment of inflammatory arthritis, including methotrexate (MTX), hydroxychloroquine (HCQ), sulfasalazine (SSZ), and leflunomide, as well as the newer oral antirheumatic agents tofacitinib and apremilast. Although nonsteroidal anti-inflammatories, prednisone, and injectable biologic agents are commonly used in the management of inflammatory arthritis, these are outside the scope of this article.

Age-related changes in pharmacokinetics and pharmacodynamics

Pharmacokinetics is the study of drug absorption, distribution, metabolism, and excretion in the body. Geriatric patients experience physiologic changes at every step of the pharmacokinetic process.7,9 However, understanding of the age-related changes on pharmacokinetic properties of particular medications has been hampered by the general lack of inclusion of older adults in clinical trials and drug-specific pharmacokinetic studies. The most clinically significant pharmacokinetic alteration in the geriatric population is a decline in renal function that decreases metabolite excretion. Several commonly used anti-rheumatic medications, such as MTX, require monitoring of renal function.

Pharmacodynamics is the study of the biochemical and physiologic effects of drugs in the body. The pharmacodynamic changes with aging are more difficult to study and less characterized than pharmacokinetic alterations. In general, the response in elderly is less predictable and subject to more interindividual variability.11 Aging patients experience changes at multiple levels including receptor, signal transduction or homeostatic mechanisms. This can not only affect the effectiveness but also the risk of adverse reactions. For example, a decrease in cell density and cell proliferation in the bone marrow in elderly individuals,7 can make these patients especially sensitive to the hematological side effects of MTX.

Polypharmacy

Polypharmacy has been defined in many ways. Some definitions focus on the number of medications whereas others consider clinical appropriateness and indication.10,1416 Consequences to polypharmacy include the risk for clinically significant drug-drug interactions, adverse drug reactions, and nonadherence.10

Age is an important risk factor for polypharmacy.14 Geriatric patients receiving multiple medications are at increased risk for cognitive impairment, falls, incontinence, and poor nutritional status.10 The complex medical regimens of RA place older patients at such risks. Treharne and colleagues17 found that the total number of RA medications was predicted by advancing age and longer disease duration. In addition, the total number of comorbidities contributed to this relationship. A 2001 study of hospitalized subjects with rheumatic diseases also found similar results, with older subjects having a higher likelihood of meeting the study’s definition of polypharmacy compared with younger subjects.18

Health literacy

Health literacy, defined as an individual’s overall capacity to obtain, process, and understand basic health information and services needed to make appropriate health decisions, is another area in which age-related changes may have an impact.19 Wong and colleagues noted that a third of patients prescribed common rheumatology medications followed the dosing instructions incorrectly.20 Several studies have found a relationship between older age and reduced health literacy. This relationship may be influenced by the educational level and age-related changes to functional status, such as visual impairment.5,2123 One study identified age older than 55 years as a risk factor for poor knowledge of MTX use in a diverse urban rheumatology clinic population in California.24

Age bias

Older patients may face yet another challenge in receiving safe and effective treatment of rheumatological disorders in the form of age bias, or disparities in the prescription of treatment by doctors based on patient age.13,25,26 In a 2010 choice-based conjoint analysis, Kievit and colleagues showed that among 135 rheumatologists, patient’s age was an important factor in the decision to escalate RA treatment.12 Tutuncu and colleagues found that patients with older-onset RA were less frequently treated with biologic drugs and combination DMARDs than those with younger-onset RA, even though they had comparable disease severity and activity.27 The reluctance to escalate therapy may result in older patients not receiving appropriate interventions, despite evidence of similar responsiveness to standard therapies when compared with younger patients.28

Due to these complexities, rheumatologists, working in partnership with other members of the allied health care team, can most effectively minimize the risk for therapeutic misadventure.

DMARD and the Risk of Infection

Autoimmune disease itself is a risk factor for infection. Patients with RA were found to have a higher risk of infection, when compared to controls with no RA.29 Addition of immunosuppressive and immunomodulatory agents for the treatment of autoimmune disease further increase the risk of infection. In a systemic literature review of observational studies and RA registries, the risk of serious infections was found to be higher among patients on biologic DMARDs (bDMARs) compared to the conventional synthetic DMARDs (csDMARDs), with a hazard ratio (HR) ranging from 1.1 to 2.4. The risk of tuberculosis was also significantly higher among patients on bDMARDs compared to the general population (HR: 34.9 [8.9, 137.2]) and when compared with patients on csDMARDs (HR: 12.5 [3.5, 44.7]).30 As with other DMARDs, risk of infection is a concern with the targeted synthetic DMARD (tsDMARD), tofacitinib. Pooled data from multiple phase 2, phase 3 and long-term extension studies of tofacitinib for RA found that the overall incidence rate of serious infections was 3 events per 100 patient-years. Pneumonia, herpes zoster, urinary tract infection and cellulitis were the most common. The incidence rates were found comparable to other bDMARDs (3.0 to 5.5 per patient-years) and TNF inhibitors (3.2 to 4.6 per patient-years). The incidence of infection did not increase with longer duration of tofacitinib use and was stable over time. The risk factors independently associated with increased risk of serious infection were: age more than 65, corticosteroid use (>7.5mg), diabetes and tofacitinib dose.31 Adequate vaccination of patients with rheumatological disease is the key strategy in preventing infectious complications. Hence the Advisory Committee on Immunization Practices (ACIP) has recommended vaccination of patients with chronic inflammatory disease on immunosuppression with Pneumococcal Vaccines (PCV-13 followed by PCV-23) and annual influenza vaccine.32

Methotrexate

MTX is the most common DMARD used to treat RA and remains the cornerstone of treatment for this condition. The therapeutic effect of MTX in inflammatory arthritis was first reported in the 1950s,33,34 but did not receive Food and Drug Administration (FDA) approval for the treatment of RA until 1988. It is also used to treat several other rheumatic conditions such as juvenile idiopathic arthritis,35,36 psoriatic arthritis,37 reactive arthritis,38 systemic lupus erythematosus,39 granulomatosis with polyangiitis,40 and polymyositis/dermatomyositis.41

The exact mechanism by which MTX exerts its therapeutic effects in RA is not fully understood. MTX is a folate analogue that enters the cells primary mediated by folate transporter 1 (FOLT), also known as reduced folate carrier 1 (RFC1). Once inside the cells, MTX undergoes polyglutamation catalyzed by folylpolyglutamate synthetase in the same manner as naturally occurring folate. MTX polyglutamates (MTX-glu) increase both the retention of MTX in the cells as well as its inhibitory actions in several intracellular enzymes. First, MTX inhibits dihydrofolate reductase (DHFR), the enzyme required for reduction of dihydrofolate (FH2) to tetrahydrolate (FH4), resulting in suppression of purines, thymidylate, serine, and methionine synthesis, and ultimately, DNA production. Second, MTX inhibits thymidylate synthetase (TYMS), an enzyme that converts deoxyuridine monophosphate (dUMP) to deoxythymidine monophosphate (dTMP), ultimately leading to decreased levels of pyrimidine that would be required for DNA biosynthesis. Finally, inhibition of 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) transformylase (ATIC) initiates a series of events that results in increased adenosine levels, which has multiple anti-inflammatory effects. For example, adenosine is associated with a decreased production of proinflammatory cytokines, such as tumor necrosis factor alpha (TNFα) and interleukin (IL)-6, as well as increased production of anti-inflammatory cytokines such as IL-10. It appears that adenosine, acting on adenosine receptors, is itself a key mediator of the anti-inflammatory effect of MTX whereas the inhibition of DHFR and TYMS seems to contribute more to antiproliferative effects.4244

In the mid-1980’s, four clinical trials in patients with active RA showed that MTX was superior to placebo in decreasing disease activity in the short-term. 4548 Since then, MTX has been clearly established as a mainstay in the therapy of RA. A systematic review of 7 randomized, placebo-controlled trials (n=732 patients) in RA showed that MTX (doses between 5 mg and 25 mg weekly) was associated with a clinically important and statistically significant improvement in the American College of Rheumatology (ACR) 50 response rate at 52 weeks when compared with placebo (relative risk [RR] 3.0, 95% confidence interval [CI] 1.5 to 6.0). Radiographic progression rates were significantly lower in the MTX-treated group compared with the placebo group (RR 0.31, 95% CI 0.11 to 0.86) but no significant differences were observed in radiographic scores.49 A 2016 network meta-analysis (158 trials, more than 37,000 patients) showed that the combination of MTX, SSZ, and HCQ (i.e., triple therapy) was superior to MTX for ACR50 response and was not statistically different from MTX plus any biologic or tofacitinib in patients with no previous MTX use or with inadequate response to this medication.50 The 2015 ACR guideline for the treatment of RA has recommended the use of MTX as a preferred initial DMARD for most RA patients and to serve as the anchor drug in combination with biological therapy.51

In rheumatic conditions, MTX is prescribed as a low-dose regimen (typically dosed ≤ 25 mg weekly). It can be administered once weekly via oral, subcutaneous, or intramuscular route. There is no difference in the bioavailability of intramuscular versus subcutaneous route of administration, 52 but the former is not frequently used. Oral bioavailability varies broadly among patients and decreases with increasing dose. A phase 2 study that evaluated the relative bioavailability of oral vs. subcutaneous MTX, showed that systemic exposure of oral MTX plateaued at doses ≥15 mg weekly, whereas subcutaneous administration at the same dose resulted in linear increases in systemic exposure. Hence, subcutaneous injection at higher doses is an alternative approach that can improve bioavailability.53 Another strategy to improve the bioavailability of oral administration includes splitting of the dose of MTX, such as giving one-half dose repeated 12 hours apart on the same day, once weekly. The efficacy of MTX dose splitting has only been addressed in limited studies.54

MTX is well tolerated in most RA patients. In a 2014 systematic review of 7 placebo-controlled trials, the adverse event rate at 12 weeks in the MTX monotherapy group was 45% (vs. 15% in the placebo group; RR 3.0, 95% CI 1.4 to 6.4) and MTX was associated with a higher rate of discontinuation due to adverse events compared to placebo (16% vs. 8%; RR 2.1, 95% CI 1.3 to 3.3).49 Another review of 21 prospective studies (2009) found that the long-term therapy (>2 years of treatment; n=3463 patients) with MTX was associated with frequent adverse reactions (73%) but they were generally mild and caused discontinuation in around 11% of the patients.55

The most common adverse reactions of MTX are gastrointestinal (nausea, vomiting, abdominal pain, diarrhea and stomatitis), elevated hepatic enzymes, dermatological (skin rash, pruritus, and alopecia), neurological (headache, vertigo, lethargy, and cognitive dysfunction), hematological (cytopenia), and pulmonary (such as pneumonitis). The rate of toxicity increases in patients with renal impairment.56 MTX is eliminated primarily via renal excretion and may accumulate in the setting of reduced renal function.57,58 Elevated liver enzymes are frequent adverse reactions and an important reason for treatment withdrawal. It occurs in up to 20% of patients and leads to discontinuation in 4% of patients during long-term use.55 Although, these events are usually self-limited, they can potentially lead to liver cirrhosis.59 Risk factors for liver injury during MTX therapy are alcohol use, history of liver disease (e.g., hepatitis B or C), other comorbidities (diabetes, obesity, hyperlipidemia), exposure to hepatotoxic medications, lack of folate supplementation, persistent abnormal liver panel, and family history of heritable liver disease.60

Several measures can be used to decrease adverse reactions associated with MTX. Laboratory monitoring for MTX toxicity should start with determining baseline complete blood cell counts, liver enzymes, and serum creatinine level; and thereafter followed by monitoring at regular intervals.51 Folic acid or folinic acid supplementation may ameliorate some of the folate-pathway-dependent adverse effects, including hematologic, gastrointestinal and hepatic side effects.61 Switching from oral to parenteral route therapy or splitting the dose of MTX are other strategies that have been recommended albeit with limited scientific support.62

Few elderly patients were included in the initial trials with MTX but accumulating data over time has helped to better understand MTX in this population. The efficacy response of geriatric patients with active RA to MTX treatment is comparable to that of younger patients.28 Given the safety profile of this medication, extra caution must be exercised when treating geriatric patients. A decline in renal function may be associated with impaired clearance and increased toxicity of MTX,5658 which is particularly relevant in the elderly. Close monitoring for signs of hepatic and hematologic toxicity should be emphasized as well.

Elderly patients are vulnerable to clinically significant drug interactions with MTX. Trimethoprim-sulfamethoxazole (TMP-SMX), a commonly prescribed antibiotic with folate antagonist effect, can increase the occurrence of hematologic manifestations and life-threatening pancytopenia. In a 2010 systematic review of 67 articles addressing MTX-drug interactions,63 cytopenia and elevation of liver enzymes were the main reported toxicities. In this review, clinically significant interactions with TMP-SMX therapy were noted in one observational study and in 17 case reports. TMP-SMX was mostly indicated for urinary tract infections. There were no reported cases of toxicity with use of prophylactic doses of 3 times per week. High-dose aspirin was also found to exacerbate the toxicity of MTX.63 Other drugs known to cause hepatotoxicity such as SSZ, leflunomide, and azathioprine may increase the incidence of liver toxicity when used in combination with MTX.64 Because MTX can accumulate in the setting of reduced clearance, medications that affect renal function should be avoided or used with caution (Box 1).

Box 1. Clinical pearls: MTX.

  • Monitor renal function; dose adjust accordingly.

  • Folic acid or folinic acid supplementation can improve tolerability of MTX.

  • Avoid concomitant use of TMP-SMX when dosed daily.

Hydroxychloroquine

HCQ is another commonly used DMARD for the management of rheumatological disorders. The precise mechanism of action of HCQ is unknown but is thought to have immunomodulatory and anti-inflammatory activity through stabilization of the lysosomal membrane, down-regulation of antigen presentation and inhibition of cell-mediated cytotoxicity.65 It also interferes with the innate immune response by inhibiting the Toll-like receptors (TLR)66.

HCQ carries FDA approval for the treatment of RA and systemic lupus erythematosus (SLE). In RA, it is best used in mild, early disease or as a component of combination therapy. Treatment with HCQ was found to be an independent determinant of remission in RA in a multicenter cross-sectional study. HCQ has exhibited synergistic effects in improving disease activity when used in combination with other DMARDs, including MTX and SSZ (triple therapy). 67,68

The use of HCQ is well established in cutaneous forms of SLE. In a comparative study of HCQ and acitretin, there was similar clinical efficacy, with 50% of the patient having a complete resolution of discoid lupus.69 HCQ is beneficial in the management of non-organ threatening disease manifestations including arthralgia, fatigue, fever, and rash. In SLE patients, it has been demonstrated to decrease the risk of flare70, decrease the risk of thromboembolism in patients with antiphospholipid antibodies,71 lower total cholesterol in patients taking steroids,72 and lower fasting blood glucose concentration.73 HCQ was also shown to have a beneficial effect on the survival of lupus patients74 and a protective effect on the risk of organ damage.75

HCQ is generally regarded as well tolerated, with gastrointestinal distress being the most common adverse effect. Skin hyperpigmentation is also a known side effect of long-term HCQ therapy. HCQ-induced blue-black dyschromia has been clinically misinterpreted as elder abuse. These cases were resolved after a thorough history or, in some cases, a skin biopsy.76,77

Post-marketing cases of cardiomyopathy and QT interval prolongation have been reported with the use of HCQ. Other rare adverse events reported include hypoglycemia, proximal myopathy and neuropathy. For this reason, caution is recommended when co-administering with hypoglycemic agents or medications with arrhythmic potential.78

HCQ has the potential to cause irreversible retinal toxicity. A 2014 retrospective case-control study reviewed more than 2300 patient records of HCQ users with at least 5 years of treatment duration in a large integrated health organization of 3.4 million overall members. The overall prevalence of HCQ-related retinopathy was reported to be 7.5%, with risk dependent on dosage and duration of use. Patient on more than 5 mg/kg had 10% risk within 10 years of use and a 40% risk after 20 years of use. In patients using less than 5 mg/kg, the risk was reduced to 2% within the first 10 years and 20% after 20 years of use. Other risk factors for retinopathy include renal disease (GFR< 60ml/minute) or concurrent use of tamoxifen. Notably, age was not identified as a risk factor.79 Based on these data, the American Academy of Ophthalmology has recommended a maximum daily dosing of less than 5 mg/kg real body weight and a baseline fundus examination to rule out preexisting maculopathy. In patients with no major risk factors, the annual screening should be done after 5 years of HCQ use. The recommended modalities of screening are visual fields and spectral-domain optical coherence tomography (SD-OCT).80 (Box 2)

Box 2. Clinical pearls: HCQ.

  • HCQ is a mainstay in the management of skin manifestations caused by SLE.

  • Judicious dosing to avoid toxicity should be practiced using a 5.0 mg/kg actual body weight cutoff.

  • Routine ophthalmology examinations are recommended to screen for the development of retinal toxicity.

Sulfasalazine

SSZ is a well-established DMARD that is most commonly used as a second-line agent in RA combination therapy but is also indicated to treat other inflammatory arthritides and inflammatory bowel disease. SSZ is composed of sulfapyridine and 5-aminosalicylic acid and it is thought that the antiarthritic activity of this compound is mostly conferred by the sulfapyridine moiety. The precise mechanism of action of SSZ is not elucidated but may involve several anti-inflammatory and immunomodulatory effects such as inhibition of proinflammatory cytokines (e.g., TNF-alpha and IL-8) and increase of adenosine release at inflamed sites (similar to MTX).8183

Genetic polymorphisms may play a role in the efficacy of the drug as well as the propensity for adverse effects. A prolonged half-life and accumulation of the sulfapyridine metabolite of SSZ with a subsequent increase in toxicity may be seen in slow acetylators.84 Patients with glucose-6 phosphate dehydrogenase (G6PD) deficiency are at increased risk of hemolytic anemia after initiation SSZ treatment.85

Although it may be used alone, SSZ is typically prescribed with MTX and HCQ as part of so-called RA ‘triple therapy’. SSZ is the most commonly discontinued drug in this regimen secondary to adverse effects.86 It is associated with gastrointestinal, central nervous system (headache, dizziness), cutaneous, and hematologic adverse reactions. The gastrointestinal complaints are usually mild in nature. They resolve with discontinuation or dose reduction and are better tolerated with the use of an enteric-coated formulation. 87,88

A syndrome of fever, rash, and abnormal liver tests can occur in the setting of SSZ therapy. In the presence of eosinophilia, this reaction is termed drug rash with eosinophilia and systemic symptoms (DRESS).89 Finally, a rare adverse effect of SSA is crystalluria with intra-tubular precipitation of sulfasalazine metabolites, and subsequent acute kidney injury.90,91 Therefore, vigilance toward maintaining adequate hydration and monitoring renal function is prudent in older individuals treated with SSZ (Box 3).

Box 3. Clinical pearls: SSZ.

  • Initiation therapy with a gradual dose increase and interval laboratory monitoring can minimize the risk of adverse reactions.

  • Slow acetylators and patients with G6PD are at increased risk of toxicity.

  • Crystalluria is a rare adverse effect. Patients should maintain adequate hydration.

Leflunomide

Leflunomide is an isoxazole derivative that inhibits dihydroorotate dehydrogenase in the pyrimidine pathway. T lymphocytes are dependent on the de novo synthesis of pyrimidine. Decreased pyrimidine leads to decreased T cell proliferation. Leflunomide is also known to modulate T-cell immunology by shifting the T-helper (Th)1/Th2 balance.92 Consequently, it has successfully been used in the treatment of various inflammatory arthritides. It received FDA approval for use in RA in 1998. There are no large head to head randomized clinical trials comparing leflunomide with MTX, but the available evidence suggests a comparable clinical efficacy93. The use of leflunomide is limited by the lack of enough evidence for its use in combination with other biological DMARDs.

Leflunomide has a similar safety profile as MTX. The most common reactions include gastrointestinal upset, hepatotoxicity, alopecia, and risk of infection.93 Adverse events are similar regardless of patient age.94 Safe prescribing of this agent relies on monitoring not only the blood count and liver function but also blood pressure. In a long-term study in patients with RA treated with leflunomide, clinically relevant increase in blood pressure was found in 5% of the patients but in most cases normalized during ongoing treatment.95 Leflunomide is also associated with significant but modest weight loss compared to other RA medications.96 In a small study of patients with RA treated with leflunomide, significant weight loss was found in 5 out of 70 patients (7%) and the weight loss ranged from 19 to 53 pounds (14%–26% decrease from baseline). Most patients had weight loss early (within 6 months) of initiation of the medication and on further continuation of medication the weight stabilized.97 Hepatotoxicity is an important adverse reaction of leflunomide and cases of severe liver injury have been reported; therefore, it is not recommended to use in patients with preexisting liver disease.98

Due to low hepatic clearance and enterohepatic recycling, the pharmacokinetic profile of leflunomide is notable for a long elimination half-life of approximately 2 weeks.99 In the setting of severe toxicity or other condition necessitating rapid withdrawal, cholestyramine can be used to bind the active metabolite of leflunomide in the intestine and interrupt enterohepatic or entero-entero recycling, thus reducing the half-life to 1 to 2 days.99102 (Box 4)

Box 4. Clinical pearls: leflunomide.

  • Hypertension and unintentional weight loss are two adverse effects related to leflunomide therapy that may be of particular concern in older patients.

  • Regular monitoring for hepatotoxicity is recommended.

  • In the clinical scenarios requiring rapid withdrawal of leflunomide, a cholestyramine washout can be implemented to decrease the half-life of the active metabolite.

Janus Kinase Inhibitors

Janus kinase inhibitors are synthetic DMARDs that target the janus kinase (JAK) and signal transducer and activator of transcription (STAT) intracellular signaling pathway. The JAK/STAT pathway mediates intracellular signaling in a variety of ways. The JAK1-JAK3 complex is involved in lymphocyte proliferation induced by interleukins such as IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21. The JAK 2 homodimer is essential in intracellular signaling by erythropoietin and granulocyte-macrophage colony stimulating factor, which is required for erythropoiesis, myelopoiesis, and thrombopoiesis.103 The JAK-STAT pathway is also involved in the host immune response against viral infection and mycobacterial infection.

Tofacitinib is a JAK inhibitor, which is FDA approved for the treatment of RA. It inhibits JAK1 and JAK3, but also inhibits JAK 2 to a lesser degree.104 It is indicated for the treatment of adult patients with moderate to severe RA with inadequate response or intolerance to MTX.105108 It may be used as monotherapy or in combination with MTX or other DMARDs. Tofacitinib is also approved for active psoriatic arthritis. Trials are currently underway studying the agent’s effects in the management of other disease states, including juvenile idiopathic arthritis, SLE, psoriatic arthritis, and ankylosing spondylitis.

Tofacitinib is FDA-approved for daily dosing at 5 mg twice daily or with the extended-release formulation at 11 mg once daily. Tofacitinib is metabolized via cytochrome P450 (CYP) 3A4 and a lesser degree via CYP2C19. Strong inhibitors of CYP 3A4 increase the effect of tofacitinib, increasing the toxicity whereas inducers of CYP3A4 decrease the effect of tofacitinib (Table 1).

Table 1.

Clinically Relevant Medication Interactions with Tofacitinib. Data from refs109112

CYP subset Inhibitors Inducers

CYP3A4 Clarithromycin, Rifampin
Erythromycin Phenytoin
Ketoconazole Carbamazepine
Itraconazole
Diltiazem
Verapamil
Nelfinavir
Ritonavir

CYP2C19 Fluoxetine Rifampicin
Fluvoxamine Phenytoin
Isoniazd Carbamazepine
Ritonavir

Herpes zoster infection is a concern in geriatric RA patients on tofacitinib. Older age is one of the most important risk factors for the development of herpes zoster infection.113 In addition, RA is an independent risk factor for herpes zoster (adjusted hazard ratio = 1.9 [95% CI = 1.8–2.0]).114 In phase II, phase III and long-term extension studies, the incidence rate of herpes zoster was found higher among the tofacitinib group compared to the placebo (crude incidence rate of 4.4 per 100 patient-years [95% CI 3.8– 4.9]) and was more common among Japanese and Korean patients. Not surprisingly, older age was associated with higher risk (odds ratio 1.9, 95% CI 1.5–2.6). Among the patients with herpes zoster, 7% of them had serious disease requiring hospitalization or use of intravenous antiviral treatment.115 The risk of herpes zoster infection was found to be higher in patients taking tofacitinib in combination with glucocorticoids compared to those on monotherapy with only tofacitinib.116 These studies underscore the importance of adequate vaccination before initiation of tofacitinib. The 2015 ACR guideline for the treatment of RA has recommended the administration of herpes zoster vaccine before starting biologic or tofacitinib in RA patients ages ≥50 years. After vaccination, they recommended a 2-week waiting period before starting a biologic.51 The Infectious Disease Society of America (IDSA) has recommended administration of live vaccine greater than or equal to 4 weeks before the use of immunosuppressive agents.117 Despite the safety and benefit of this approach, vaccination against herpes zoster is woefully underutilized in patients with RA.118 A new recombinant zoster vaccine (brand name, Shingrex) was recommended by ACIP in 2017 for immunocompetent patients above the age of 50 years. The vaccine is recommended at day 0 with a repeat dose after 2 months.119 This vaccine is found to be more efficacious and reduces the risk of herpes zoster and post-herpetic neuralgia by more than 90%.120 Due to the inactivated nature of the vaccine, it could be administered 2 weeks prior to the initiation of tofacitinib and this strategy could potentially mitigate the underutilization of such vaccination.

Winthrop and colleagues illustrated a diminished responsiveness to the 23-valent pneumococcal polysaccharide vaccine (PPSV-23) in subjects commencing tofacitinib therapy at a dose of 10 mg twice daily. The response was further diminished in subjects with concomitant MTX. However, the response to annual influenza vaccination was unaltered in subjects receiving tofacitinib, with or without MTX. For current users, a 2-week holiday from tofacitinib around the time of vaccination did not appreciably improve immunogenicity for the PPSV-23 vaccine. Although most users developed sufficient responses to both vaccines, administration of the pneumococcal vaccine before initiation of tofacitinib may improve overall response and should be considered (BOX 5).121

Clinical pearls: tofacitinib.

  • Tofacitinib is a new and effective treatment option for RA, which could be used with or without MTX.

  • Patients should receive screening for tuberculosis before therapy initiation.

  • Risk of infections including herpes zoster may be higher in patients receiving tofacitinib, specifically Asian patients.

  • Tofacitinib therapy may blunt the immune response to specific vaccines; therefore, patients should be assessed for appropriate immunizations against herpes zoster, influenza and pneumococcus before commencing therapy.

Apremilast

Apremilast is an oral small molecule that belongs to a class of new drugs known as phosphodiesterase-4 (PDE4) inhibitors. It was approved by the FDA for the treatment of psoriasis and psoriatic arthritis. PDE-4 is a superfamily of enzymes that catalyze the hydrolysis of cyclic adenosine monophosphate (cAMP).122 Inhibition of PDE4 increases the levels of cAMP, a well-known intracellular second messenger that leads to activation of cAMP-dependent protein kinase A (PKA). This causes modifications of several transcription factors such as the activation of cAMP-response element binding protein (CREB) and inhibition of nuclear factor kappa B (NF-kB). 123,124 The specific mechanism of action by which apremilast exerts its therapeutic effects remains incompletely understood but it is known to modify the production of several cytokines, presumably enhancing cAMP actions at the transcriptional level. PDE4 inhibition reduces secretion of pro-inflammatory cytokines such as TNFα, IFNγ, and IL-2; and increases production of anti-inflammatory modulators such as IL-10.125,126

The efficacy and safety of apremilast for the treatment of psoriatic arthritis has been evaluated in four randomized, double-blinded, placebo-controlled phase 3 trials in the Psoriatic Arthritis Long-term Assessment of Clinical Efficacy programme (PALACE 1–4 trials).127131 Patients were randomized to receive apremilast 20 or 30 mg twice daily or placebo. PALACE 1–3 enrolled a total of 1493 patients with active psoriasis arthritis despite prior traditional DMARD or biologic treatment. PALACE 4 included 527 patients with no prior DMARD or biological therapy. The primary endpoint was the ACR20 response rate at week 16 and the key secondary endpoint was the change from baseline in the Health Assessment Questionnaire-Disability Index (HAQ-DI) score at week 16. Apremilast improved signs and symptoms in patients with active psoriatic arthritis when compared to placebo. At week 16, the ACR20 response rate was significantly higher with apremilast 30 mg twice daily than with placebo across all trials (40 vs. 19% in PALACE 1, 32 vs. 19% in PALACE 2, 41 vs. 18% in PALACE 3, and 31 vs. 16 in PALACE 4). Improvements in HAQ-DI were also seen with apremilast in PALACE trials. For example, in PALACE 1 the mean changes from baseline at week 16 were −0.09 (standard error 0.04) in the placebo group and −0.25 (0.04) in apremilast 30 mg BID (p=0.0015 vs. placebo) in the per protocol population. Compared to placebo, a significantly greater proportion of patients receiving apremilast 30 mg BID achieved minimal clinically important differences (MCID) of ≥0.13 (39% placebo vs. 50% apremilast 30 mg BID; p=0.0334) and ≥0.30 (27% placebo vs. 40% apremilast 30 mg BID; p=0.0149) as measured by the HAQ-DI at week 16. This efficacy was sustained and the benefit of apremilast in psoriatic arthritis has been reported in extensions up to 4 years of treatment.132

The efficacy and safety of apremilast in active psoriatic arthritis was also evaluated in a phase 3B, randomized, double-blind, placebo-controlled trial (ACTIVE trial).133 Patients (n=219) who may have had one prior conventional therapy and were not previously treated with a biological agent were randomized to apremilast 30 mg twice daily (n=110) or placebo (n=109) for 24 weeks. A significantly greater ACR20 response at week 16 (primary outcome) was observed with apremilast versus placebo (38% vs. 20%; P=0.004). This trial also assessed the onset of apremilast efficacy at earlier time points (beginning at week 2) than in previous studies. At week 2 (first assessment), response rates were 16% in apremilast group versus 6% (P=0.025) in the placebo group. Improvements in other manifestations, including swollen and tender joints, enthesitis, physical impairment and morning stiffness severity, were also observed with apremilast at week 2. The benefit was maintained with continued treatment through week 52.

Apremilast has an acceptable safety profile and, in general, is well tolerated. The most frequent adverse reactions are diarrhea, nausea, upper respiratory tract infection, and headache. The gastrointestinal manifestations are the most common reactions and typically are mild or moderate in severity, occur early in therapy, and resolve with continued treatment.127129 In the PALACE 1, the frequency of the most common adverse events reported with apremilast 30 mg BID (n=168) in the placebo-controlled phase (weeks 0–24) were diarrhea (19.0%), nausea (18.5%), headache (10.7%), and upper respiratory tract infection (4.2%).127 Weight loss has also been reported (5% to 10% of body weight loss in 10% to 12% of subjects; ≥10% of body weight loss in 2%). Therefore, it is recommended to monitor weight regularly in patients treated with apremilast, and consider discontinuation if unexplained or clinically significant weight loss occurs. Apremilast is also associated with an increase in reports of depression. Hence the risks and benefits of this medication should be weighed in patients with a history of depression or mood changes.134 Treatment with apremilast is not associated with clinically meaningful laboratory abnormalities and routine laboratory monitoring is not required. However, baseline serum creatinine is important because the dose should be reduced to 30 mg once daily in patients with an estimated creatinine clearance less than 30 mL/min. In a pooled safety analysis of three trials (PALACE 1, 2, and 3), no new safety concerns were identified with long-term exposure over 52 weeks.135

The acceptable safety profile of apremilast makes this drug an appealing therapeutic option for geriatric patients. Of the total number of patients with psoriatic arthritis who enrolled in PALACE 1–3, around 10% (146 out of 1493) were ≥65 years of age. No overall differences were obtained in the safety profile of this age group compared with the younger group in the clinical studies. Similar results were obtained in the clinical trials of apremilast in psoriasis. In the Efficacy and Safety Trial Evaluating the Effects of Apremilast in Psoriasis (ESTEEM 1 and 2) phase III clinical trial program,136,137 about 9% (108 out of 1257) of the subjects who enrolled were ≥65 years of age. No overall differences were observed in the efficacy and safety in patients ≥65 years of age and younger patients.134

The clinical use of apremilast has also been explored in other rheumatic and dermatological conditions including RA, Behçet’s disease, and different forms of dermatitis.138 In RA, a phase 2, randomized, placebo-controlled trial in patients with active arthritis who had an inadequate response to MTX failed to meet the primary efficacy endpoint.139 Further studies are needed to clarify the radiographic effect in psoriatic arthritis and to determine the actual therapeutic role compared to conventional DMARDs and biologics (Box 6).

Clinical pearls: apremilast.

  • Apremilast has a favorable safety profile and is likely a viable therapeutic option for elderly patients with psoriatic arthritis.

  • Evaluate risks of depression and monitor weight during therapy.

  • Routine laboratory monitoring is not required during apremilast therapy.

Summary

An effective treatment strategy targeting rheumatological disorders in the elderly should be directed at maximizing the quality of life. In patients with RA, a treat-to-target approach with the goal of remission or low disease activity has improved outcomes.140 However, treatment in geriatric patients is challenging because they are particularly vulnerable to adverse reactions. Factors known to increase this risk for adverse reactions include age-related changes in pharmacokinetics and pharmacodynamics, comorbidities, polypharmacy, and drug compliance issues. Moreover, pharmacotherapy in rheumatology is evolving rapidly while guidance on how to apply the new advances to older individuals is frequently missing. Regular monitoring of clinical and laboratory parameters that is tailored to the specific medication and the comorbidities is crucial to minimize negative outcomes. Finally, educating the patient or caregiver and involving them in making decisions and setting the treatment goals is paramount to ensure an optimal outcome.

Key points.

  • Older patients with rheumatic disorders are at increased risk for therapeutic misadventure due to age-related pharmacokinetic and pharmacodynamic changes, polypharmacy, comorbidities, impaired health literacy secondary to decreased cognition, and provider age bias.

  • Rheumatologists along with other members of the allied health care team, can most effectively minimize the risk for medication-related adverse reactions in older patients.

  • Familiarity with dosing, monitoring, adverse reactions, medication interactions, and amelioration strategies can improve the safety of disease-modifying antirheumatic drugs in the older rheumatology patient.

Acknowledgments

Disclosure Statement: This work was supported by the Intramural Program of the National Institute of Arthritis and Musculoskeletal and Skin Diseases of the National Institutes of Health. This article reflects the views of the authors and should not be construed to represent FDA’s views or policies.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

The authors do not have any conflicts of interest related to this work.

References

  • 1.Barbour KE, Helmick CG, Boring M, Brady TJ. Vital Signs: Prevalence of Doctor-Diagnosed Arthritis and Arthritis-Attributable Activity Limitation - United States, 2013–2015. MMWR Morb Mortal Wkly Rep. 2017;66(9):246–253. doi: 10.15585/mmwr.mm6609e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Helmick CG, Felson DT, Lawrence RC, et al. Estimates of the prevalence of arthritis and other rheumatic conditions in the United States. Part I. Arthritis Rheum. 2008;58(1):15–25. doi: 10.1002/art.23177. [DOI] [PubMed] [Google Scholar]
  • 3.Choi HK, Hernan MA, Seeger JD, Robins JM, Wolfe F. Methotrexate and mortality in patients with rheumatoid arthritis: a prospective study. Lancet. 2002;359(9313):1173–1177. doi: 10.1016/S0140-6736(02)08213-2. [DOI] [PubMed] [Google Scholar]
  • 4.Budnitz DS, Pollock DA, Weidenbach KN, Mendelsohn AB, Schroeder TJ, Annest JL. National surveillance of emergency department visits for outpatient adverse drug events. JAMA. 2006;296(15):1858–1866. doi: 10.1001/jama.296.15.1858. [DOI] [PubMed] [Google Scholar]
  • 5.Baker DW, Gazmararian JA, Sudano J, Patterson M. The association between age and health literacy among elderly persons. J Gerontol B Psychol Sci Soc Sci. 2000;55(6):S368–374. doi: 10.1093/geronb/55.6.s368. [DOI] [PubMed] [Google Scholar]
  • 6.Joplin S, van der Zwan R, Joshua F, Wong PK. Medication adherence in patients with rheumatoid arthritis: the effect of patient education, health literacy, and musculoskeletal ultrasound. Biomed Res Int. 2015;2015:150658. doi: 10.1155/2015/150658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Turnheim K. When drug therapy gets old: pharmacokinetics and pharmacodynamics in the elderly. Exp Gerontol. 2003;38(8):843–853. doi: 10.1016/s0531-5565(03)00133-5. [DOI] [PubMed] [Google Scholar]
  • 8.Ranganath VK, Furst DE. Disease-modifying antirheumatic drug use in the elderly rheumatoid arthritis patient. Rheum Dis Clin North Am. 2007;33(1):197–217. doi: 10.1016/j.rdc.2006.12.011. [DOI] [PubMed] [Google Scholar]
  • 9.Shi S, Klotz U. Age-related changes in pharmacokinetics. Curr Drug Metab. 2011;12(7):601–610. doi: 10.2174/138920011796504527. [DOI] [PubMed] [Google Scholar]
  • 10.Shah BM, Hajjar ER. Polypharmacy, adverse drug reactions, and geriatric syndromes. Clin Geriatr Med. 2012;28(2):173–186. doi: 10.1016/j.cger.2012.01.002. [DOI] [PubMed] [Google Scholar]
  • 11.Trifiro G, Spina E. Age-related changes in pharmacodynamics: focus on drugs acting on central nervous and cardiovascular systems. Curr Drug Metab. 2011;12(7):611–620. doi: 10.2174/138920011796504473. [DOI] [PubMed] [Google Scholar]
  • 12.Kievit W, van Hulst L, van Riel P, Fraenkel L. Factors that influence rheumatologists’ decisions to escalate care in rheumatoid arthritis: results from a choice-based conjoint analysis. Arthritis Care Res (Hoboken) 2010;62(6):842–847. doi: 10.1002/acr.20123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Juby A, Davis P. An evaluation of the impact of seniors on a rheumatology referral clinic: demographics and pharmacotherapy. Clin Rheumatol. 2011;30(11):1507–1509. doi: 10.1007/s10067-011-1845-8. [DOI] [PubMed] [Google Scholar]
  • 14.Hovstadius B, Petersson G. Factors leading to excessive polypharmacy. Clin Geriatr Med. 2012;28(2):159–172. doi: 10.1016/j.cger.2012.01.001. [DOI] [PubMed] [Google Scholar]
  • 15.Hajjar ER, Cafiero AC, Hanlon JT. Polypharmacy in elderly patients. Am J Geriatr Pharmacother. 2007;5(4):345–351. doi: 10.1016/j.amjopharm.2007.12.002. [DOI] [PubMed] [Google Scholar]
  • 16.Haider SI, Johnell K, Weitoft GR, Thorslund M, Fastbom J. The influence of educational level on polypharmacy and inappropriate drug use: a register-based study of more than 600,000 older people. J Am Geriatr Soc. 2009;57(1):62–69. doi: 10.1111/j.1532-5415.2008.02040.x. [DOI] [PubMed] [Google Scholar]
  • 17.Treharne GJ, Douglas KM, Iwaszko J, et al. Polypharmacy among people with rheumatoid arthritis: the role of age, disease duration and comorbidity. Musculoskeletal Care. 2007;5(4):175–190. doi: 10.1002/msc.112. [DOI] [PubMed] [Google Scholar]
  • 18.Viktil KK, Enstad M, Kutschera J, Smedstad LM, Schjott J. Polypharmacy among patients admitted to hospital with rheumatic diseases. Pharm World Sci. 2001;23(4):153–158. doi: 10.1023/a:1011909827909. [DOI] [PubMed] [Google Scholar]
  • 19.Simonds SK. Health education as social policy. Health Educ Q. 1974;2:1–10. [Google Scholar]
  • 20.Wong PK, Christie L, Johnston J, et al. How well do patients understand written instructions?: health literacy assessment in rural and urban rheumatology outpatients. Medicine (Baltimore) 2014;93(25):e129. doi: 10.1097/MD.0000000000000129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Baker DW, Gazmararian JA, Williams MV, et al. Functional health literacy and the risk of hospital admission among Medicare managed care enrollees. Am J Public Health. 2002;92(8):1278–1283. doi: 10.2105/ajph.92.8.1278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Buchbinder R, Hall S, Youd JM. Functional health literacy of patients with rheumatoid arthritis attending a community-based rheumatology practice. J Rheumatol. 2006;33(5):879–886. [PubMed] [Google Scholar]
  • 23.Caplan L, Wolfe F, Michaud K, Quinzanos I, Hirsh JM. Strong association of health literacy with functional status among rheumatoid arthritis patients: a cross-sectional study. Arthritis Care Res (Hoboken) 2014;66(4):508–514. doi: 10.1002/acr.22165. [DOI] [PubMed] [Google Scholar]
  • 24.Barton JL, Schmajuk G, Trupin L, et al. Poor knowledge of methotrexate associated with older age and limited English-language proficiency in a diverse rheumatoid arthritis cohort. Arthritis Res Ther. 2013;15(5):R157. doi: 10.1186/ar4340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Fraenkel L, Rabidou N, Dhar R. Are rheumatologists’ treatment decisions influenced by patients’ age? Rheumatology (Oxford) 2006;45(12):1555–1557. doi: 10.1093/rheumatology/kel144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Radovits BJ, Fransen J, Eijsbouts A, van Riel PL, Laan RF. Missed opportunities in the treatment of elderly patients with rheumatoid arthritis. Rheumatology (Oxford) 2009;48(8):906–910. doi: 10.1093/rheumatology/kep129. [DOI] [PubMed] [Google Scholar]
  • 27.Tutuncu Z, Reed G, Kremer J, Kavanaugh A. Do patients with older-onset rheumatoid arthritis receive less aggressive treatment? Ann Rheum Dis. 2006;65(9):1226–1229. doi: 10.1136/ard.2005.051144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Koller MD, Aletaha D, Funovits J, Pangan A, Baker D, Smolen JS. Response of elderly patients with rheumatoid arthritis to methotrexate or TNF inhibitors compared with younger patients. Rheumatology (Oxford) 2009;48(12):1575–1580. doi: 10.1093/rheumatology/kep291. [DOI] [PubMed] [Google Scholar]
  • 29.Doran MF, Crowson CS, Pond GR, O’Fallon WM, Gabriel SE. Frequency of infection in patients with rheumatoid arthritis compared with controls: a population-based study. Arthritis Rheum. 2002;46(9):2287–2293. doi: 10.1002/art.10524. [DOI] [PubMed] [Google Scholar]
  • 30.Ramiro S, Gaujoux-Viala C, Nam JL, et al. Safety of synthetic and biological DMARDs: a systematic literature review informing the 2013 update of the EULAR recommendations for management of rheumatoid arthritis. Ann Rheum Dis. 2014;73(3):529–535. doi: 10.1136/annrheumdis-2013-204575. [DOI] [PubMed] [Google Scholar]
  • 31.Cohen S, Radominski SC, Gomez-Reino JJ, et al. Analysis of infections and all-cause mortality in phase II, phase III, and long-term extension studies of tofacitinib in patients with rheumatoid arthritis. Arthritis Rheumatol. 2014;66(11):2924–2937. doi: 10.1002/art.38779. [DOI] [PubMed] [Google Scholar]
  • 32. [Accessed on January 24, 2018]; https://www.cdc.gov/vaccines/hcp/acip-recs/general-recs/immunocompetence.html.
  • 33.Gubner R. Therapeutic suppression of tissue reactivity. I. Comparison of the effects of cortisone and aminopterin. Am J Med Sci. 1951;221(2):169–175. doi: 10.1097/00000441-195102000-00008. [DOI] [PubMed] [Google Scholar]
  • 34.Gubner R, August S, Ginsberg V. Therapeutic suppression of tissue reactivity. II. Effect of aminopterin in rheumatoid arthritis and psoriasis. Am J Med Sci. 1951;221(2):176–182. [PubMed] [Google Scholar]
  • 35.Silverman E, Mouy R, Spiegel L, et al. Leflunomide or methotrexate for juvenile rheumatoid arthritis. N Engl J Med. 2005;352(16):1655–1666. doi: 10.1056/NEJMoa041810. [DOI] [PubMed] [Google Scholar]
  • 36.Ruperto N, Murray KJ, Gerloni V, et al. A randomized trial of parenteral methotrexate comparing an intermediate dose with a higher dose in children with juvenile idiopathic arthritis who failed to respond to standard doses of methotrexate. Arthritis Rheum. 2004;50(7):2191–2201. doi: 10.1002/art.20288. [DOI] [PubMed] [Google Scholar]
  • 37.Lie E, van der Heijde D, Uhlig T, et al. Effectiveness and retention rates of methotrexate in psoriatic arthritis in comparison with methotrexate-treated patients with rheumatoid arthritis. Ann Rheum Dis. 2010;69(4):671–676. doi: 10.1136/ard.2009.113308. [DOI] [PubMed] [Google Scholar]
  • 38.Schmitt SK. Reactive Arthritis. Infect Dis Clin North Am. 2017;31(2):265–277. doi: 10.1016/j.idc.2017.01.002. [DOI] [PubMed] [Google Scholar]
  • 39.Fortin PR, Abrahamowicz M, Ferland D, Lacaille D, Smith CD, Zummer M. Steroid-sparing effects of methotrexate in systemic lupus erythematosus: a double-blind, randomized, placebo-controlled trial. Arthritis Rheum. 2008;59(12):1796–1804. doi: 10.1002/art.24068. [DOI] [PubMed] [Google Scholar]
  • 40.De Groot K, Rasmussen N, Bacon PA, et al. Randomized trial of cyclophosphamide versus methotrexate for induction of remission in early systemic antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis Rheum. 2005;52(8):2461–2469. doi: 10.1002/art.21142. [DOI] [PubMed] [Google Scholar]
  • 41.Fendler C, Braun J. Use of methotrexate in inflammatory myopathies. Clin Exp Rheumatol. 2010;28(5 Suppl 61):S164–167. [PubMed] [Google Scholar]
  • 42.Chabner BA, Allegra CJ, Curt GA, et al. Polyglutamation of methotrexate. Is methotrexate a prodrug? J Clin Invest. 1985;76(3):907–912. doi: 10.1172/JCI112088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Chan ES, Cronstein BN. Molecular action of methotrexate in inflammatory diseases. Arthritis research. 2002;4(4):266–273. doi: 10.1186/ar419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Brown PM, Pratt AG, Isaacs JD. Mechanism of action of methotrexate in rheumatoid arthritis, and the search for biomarkers. Nat Rev Rheumatol. 2016;12(12):731–742. doi: 10.1038/nrrheum.2016.175. [DOI] [PubMed] [Google Scholar]
  • 45.Thompson RN, Watts C, Edelman J, Esdaile J, Russell AS. A controlled two-centre trial of parenteral methotrexate therapy for refractory rheumatoid arthritis. J Rheumatol. 1984;11(6):760–763. [PubMed] [Google Scholar]
  • 46.Weinblatt ME, Coblyn JS, Fox DA, et al. Efficacy of low-dose methotrexate in rheumatoid arthritis. N Engl J Med. 1985;312(13):818–822. doi: 10.1056/NEJM198503283121303. [DOI] [PubMed] [Google Scholar]
  • 47.Williams HJ, Willkens RF, Samuelson CO, Jr, et al. Comparison of low-dose oral pulse methotrexate and placebo in the treatment of rheumatoid arthritis. A controlled clinical trial. Arthritis Rheum. 1985;28(7):721–730. doi: 10.1002/art.1780280702. [DOI] [PubMed] [Google Scholar]
  • 48.Andersen PA, West SG, O’Dell JR, Via CS, Claypool RG, Kotzin BL. Weekly pulse methotrexate in rheumatoid arthritis. Clinical and immunologic effects in a randomized, double-blind study. Ann Intern Med. 1985;103(4):489–496. doi: 10.7326/0003-4819-103-4-489. [DOI] [PubMed] [Google Scholar]
  • 49.Lopez-Olivo MA, Siddhanamatha HR, Shea B, Tugwell P, Wells GA, Suarez-Almazor ME. Methotrexate for treating rheumatoid arthritis. Cochrane Database Syst Rev. 2014;(6) doi: 10.1002/14651858.CD000957. CD000957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Hazlewood GS, Barnabe C, Tomlinson G, Marshall D, Devoe D, Bombardier C. Methotrexate monotherapy and methotrexate combination therapy with traditional and biologic disease modifying antirheumatic drugs for rheumatoid arthritis: abridged Cochrane systematic review and network meta-analysis. BMJ. 2016;353:i1777. doi: 10.1136/bmj.i1777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Singh JA, Saag KG, Bridges SL, Jr, et al. 2015 American College of Rheumatology Guideline for the Treatment of Rheumatoid Arthritis. Arthritis Rheumatol. 2016;68(1):1–26. doi: 10.1002/art.39480. [DOI] [PubMed] [Google Scholar]
  • 52.Jundt JW, Browne BA, Fiocco GP, Steele AD, Mock D. A comparison of low dose methotrexate bioavailability: oral solution, oral tablet, subcutaneous and intramuscular dosing. J Rheumatol. 1993;20(11):1845–1849. [PubMed] [Google Scholar]
  • 53.Schiff MH, Jaffe JS, Freundlich B. Head-to-head, randomised, crossover study of oral versus subcutaneous methotrexate in patients with rheumatoid arthritis: drug-exposure limitations of oral methotrexate at doses >/=15 mg may be overcome with subcutaneous administration. Ann Rheum Dis. 2014;73(8):1549–1551. doi: 10.1136/annrheumdis-2014-205228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Dhaon P, Das SK, Srivastava R, Agarwal G, Asthana A. Oral Methotrexate in split dose weekly versus oral or parenteral Methotrexate once weekly in Rheumatoid Arthritis: a short-term study. Int J Rheum Dis. 2016 doi: 10.1111/1756-185X.12910. [DOI] [PubMed] [Google Scholar]
  • 55.Salliot C, van der Heijde D. Long-term safety of methotrexate monotherapy in patients with rheumatoid arthritis: a systematic literature research. Ann Rheum Dis. 2009;68(7):1100–1104. doi: 10.1136/ard.2008.093690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.The effect of age and renal function on the efficacy and toxicity of methotrexate in rheumatoid arthritis. Rheumatoid Arthritis Clinical Trial Archive Group. J Rheumatol. 1995;22(2):218–223. [PubMed] [Google Scholar]
  • 57.Bressolle F, Bologna C, Kinowski JM, Arcos B, Sany J, Combe B. Total and free methotrexate pharmacokinetics in elderly patients with rheumatoid arthritis. A comparison with young patients. J Rheumatol. 1997;24(10):1903–1909. [PubMed] [Google Scholar]
  • 58.Bressolle F, Bologna C, Kinowski JM, Sany J, Combe B. Effects of moderate renal insufficiency on pharmacokinetics of methotrexate in rheumatoid arthritis patients. Ann Rheum Dis. 1998;57(2):110–113. doi: 10.1136/ard.57.2.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wood PR, Caplan L. Drug-Induced Gastrointestinal and Hepatic Disease Associated with Biologics and Nonbiologic Disease-Modifying Antirheumatic Drugs. Rheum Dis Clin North Am. 2018;44(1):29–43. doi: 10.1016/j.rdc.2017.09.003. [DOI] [PubMed] [Google Scholar]
  • 60.Kalb RE, Strober B, Weinstein G, Lebwohl M. Methotrexate and psoriasis: 2009 National Psoriasis Foundation Consensus Conference. J Am Acad Dermatol. 2009;60(5):824–837. doi: 10.1016/j.jaad.2008.11.906. [DOI] [PubMed] [Google Scholar]
  • 61.Shea B, Swinden MV, Tanjong Ghogomu E, et al. Folic acid and folinic acid for reducing side effects in patients receiving methotrexate for rheumatoid arthritis. Cochrane Database Syst Rev. 2013;(5) doi: 10.1002/14651858.CD000951. CD000951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Katchamart W, Bourre-Tessier J, Donka T, et al. Canadian recommendations for use of methotrexate in patients with rheumatoid arthritis. J Rheumatol. 2010;37(7):1422–1430. doi: 10.3899/jrheum.090978. [DOI] [PubMed] [Google Scholar]
  • 63.Bourre-Tessier J, Haraoui B. Methotrexate drug interactions in the treatment of rheumatoid arthritis: a systematic review. J Rheumatol. 2010;37(7):1416–1421. doi: 10.3899/jrheum.090153. [DOI] [PubMed] [Google Scholar]
  • 64.Anelli MG, Scioscia C, Grattagliano I, Lapadula G. Old and new antirheumatic drugs and the risk of hepatotoxicity. Ther Drug Monit. 2012;34(6):622–628. doi: 10.1097/FTD.0b013e31826a6306. [DOI] [PubMed] [Google Scholar]
  • 65.Fox R. Anti-malarial drugs: possible mechanisms of action in autoimmune disease and prospects for drug development. Lupus. 1996;5(Suppl 1):S4–10. [PubMed] [Google Scholar]
  • 66.Kuznik A, Bencina M, Svajger U, Jeras M, Rozman B, Jerala R. Mechanism of endosomal TLR inhibition by antimalarial drugs and imidazoquinolines. J Immunol. 2011;186(8):4794–4804. doi: 10.4049/jimmunol.1000702. [DOI] [PubMed] [Google Scholar]
  • 67.Moreland LW, O’Dell JR, Paulus HE, et al. A randomized comparative effectiveness study of oral triple therapy versus etanercept plus methotrexate in early aggressive rheumatoid arthritis: the treatment of Early Aggressive Rheumatoid Arthritis Trial. Arthritis Rheum. 2012;64(9):2824–2835. doi: 10.1002/art.34498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Wang GY, Zhang SL, Wang XR, et al. Remission of rheumatoid arthritis and potential determinants: a national multi-center cross-sectional survey. Clin Rheumatol. 2015;34(2):221–230. doi: 10.1007/s10067-014-2828-3. [DOI] [PubMed] [Google Scholar]
  • 69.Jessop S, Whitelaw DA, Grainge MJ, Jayasekera P. Drugs for discoid lupus erythematosus. Cochrane Database Syst Rev. 2017;5 doi: 10.1002/14651858.CD002954.pub3. CD002954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Mok CC, Penn HJ, Chan KL, Tse SM, Langman LJ, Jannetto PJ. Hydroxychloroquine Serum Concentrations and Flares of Systemic Lupus Erythematosus: A Longitudinal Cohort Analysis. Arthritis Care Res (Hoboken) 2016;68(9):1295–1302. doi: 10.1002/acr.22837. [DOI] [PubMed] [Google Scholar]
  • 71.Petri M. Use of hydroxychloroquine to prevent thrombosis in systemic lupus erythematosus and in antiphospholipid antibody-positive patients. Curr Rheumatol Rep. 2011;13(1):77–80. doi: 10.1007/s11926-010-0141-y. [DOI] [PubMed] [Google Scholar]
  • 72.Stojan G, Petri M. Atherosclerosis in systemic lupus erythematosus. J Cardiovasc Pharmacol. 2013;62(3):255–262. doi: 10.1097/FJC.0b013e31829dd857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Chen YM, Lin CH, Lan TH, et al. Hydroxychloroquine reduces risk of incident diabetes mellitus in lupus patients in a dose-dependent manner: a population-based cohort study. Rheumatology (Oxford) 2015;54(7):1244–1249. doi: 10.1093/rheumatology/keu451. [DOI] [PubMed] [Google Scholar]
  • 74.Alarcon GS, McGwin G, Bertoli AM, et al. LUMINA Study Group. Effect of hydroxychloroquine on the survival of patients with systemic lupus erythematosus: data from LUMINA, a multiethnic US cohort (LUMINA L) Ann Rheum Dis. 2007;66(9):1168–72. doi: 10.1136/ard.2006.068676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Akhavan PS, Su J, Lou W, Gladman DD, Urowitz MB, Fortin PR. The early protective effect of hydroxychloroquine on the risk of cumulative damage in patients with systemic lupus erythematosus. J Rheumatol. 2013;40(6):831–841. doi: 10.3899/jrheum.120572. [DOI] [PubMed] [Google Scholar]
  • 76.Cohen PR. Hydroxychloroquine-associated hyperpigmentation mimicking elder abuse. Dermatol Ther (Heidelb) 2013;3(2):203–210. doi: 10.1007/s13555-013-0032-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.True DG, Bryant LR, Harris MD, Bernert RA. Clinical images: Hydroxychloroquine-associated mucocutaneous hyperpigmentation. Arthritis Rheum. 2002;46(6):1698. doi: 10.1002/art.10278. [DOI] [PubMed] [Google Scholar]
  • 78. [Accessed on January 24, 2018]; https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/009768s037s045s047lbl.pdf.
  • 79.Melles RB, Marmor MF. The risk of toxic retinopathy in patients on long-term hydroxychloroquine therapy. JAMA Ophthalmol. 2014;132(12):1453–1460. doi: 10.1001/jamaophthalmol.2014.3459. [DOI] [PubMed] [Google Scholar]
  • 80.Marmor MF, Kellner U, Lai TY, Melles RB, Mieler WF. Recommendations on Screening for Chloroquine and Hydroxychloroquine Retinopathy (2016 Revision) Ophthalmology. 2016;123(6):1386–1394. doi: 10.1016/j.ophtha.2016.01.058. [DOI] [PubMed] [Google Scholar]
  • 81.Rodenburg RJ, Ganga A, van Lent PL, van de Putte LB, van Venrooij WJ. The antiinflammatory drug sulfasalazine inhibits tumor necrosis factor alpha expression in macrophages by inducing apoptosis. Arthritis Rheum. 2000;43(9):1941–1950. doi: 10.1002/1529-0131(200009)43:9<1941::AID-ANR4>3.0.CO;2-O. [DOI] [PubMed] [Google Scholar]
  • 82.Gadangi P, Longaker M, Naime D, et al. The anti-inflammatory mechanism of sulfasalazine is related to adenosine release at inflamed sites. J Immunol. 1996;156(5):1937–1941. [PubMed] [Google Scholar]
  • 83.Volin MV, Campbell PL, Connors MA, Woodruff DC, Koch AE. The effect of sulfasalazine on rheumatoid arthritic synovial tissue chemokine production. Exp Mol Pathol. 2002;73(2):84–92. doi: 10.1006/exmp.2002.2460. [DOI] [PubMed] [Google Scholar]
  • 84.Tarnowski M, Paradowska-Gorycka A, Dabrowska-Zamojcin E, Czerewaty M, Sluczanowska-Glabowska S, Pawlik A. The effect of gene polymorphisms on patient responses to rheumatoid arthritis therapy. Expert Opin Drug Metab Toxicol. 2016;12(1):41–55. doi: 10.1517/17425255.2016.1121233. [DOI] [PubMed] [Google Scholar]
  • 85.Mechanick JI. Coombs’ positive hemolytic anemia following sulfasalazine therapy in ulcerative colitis: case reports, review, and discussion of pathogenesis. Mt Sinai J Med. 1985;52(8):667–670. [PubMed] [Google Scholar]
  • 86.Cummins L, Katikireddi VS, Shankaranarayana S, et al. Safety and retention of combination triple disease-modifying anti-rheumatic drugs in new-onset rheumatoid arthritis. Intern Med J. 2015;45(12):1266–1273. doi: 10.1111/imj.12896. [DOI] [PubMed] [Google Scholar]
  • 87.Okubo S, Nakatani K, Nishiya K. Gastrointestinal symptoms associated with enteric-coated sulfasalazine (Azulfidine EN tablets) Mod Rheumatol. 2002;12(3):226–229. doi: 10.3109/s101650200039. [DOI] [PubMed] [Google Scholar]
  • 88.Weaver A, Chatwell R, Churchill M, et al. Improved gastrointestinal tolerance and patient preference of enteric-coated sulfasalazine versus uncoated sulfasalazine tablets in patients with rheumatoid arthritis. J Clin Rheumatol. 1999;5(4):193–200. doi: 10.1097/00124743-199908000-00003. [DOI] [PubMed] [Google Scholar]
  • 89.Raithatha N, Mehrtens S, Mouyis M, Manson J. Rash and fever after sulfasalazine use. BMJ. 2014;349:g5655. doi: 10.1136/bmj.g5655. [DOI] [PubMed] [Google Scholar]
  • 90.DeMichele J, Rezaizadeh H, Goldstein JI. Sulfasalazine crystalluria-induced anuric renal failure. Clin Gastroenterol Hepatol. 2012;10(2):A32. doi: 10.1016/j.cgh.2011.09.027. [DOI] [PubMed] [Google Scholar]
  • 91.Durando M, Tiu H, Kim JS. Sulfasalazine-Induced Crystalluria Causing Severe Acute Kidney Injury. Am J Kidney Dis. 2017;70(6):869–873. doi: 10.1053/j.ajkd.2017.05.013. [DOI] [PubMed] [Google Scholar]
  • 92.Fragoso YD, Brooks JB. Leflunomide and teriflunomide: altering the metabolism of pyrimidines for the treatment of autoimmune diseases. Expert Rev Clin Pharmacol. 2015;8(3):315–320. doi: 10.1586/17512433.2015.1019343. [DOI] [PubMed] [Google Scholar]
  • 93.Singer O, Gibofsky A. Methotrexate versus leflunomide in rheumatoid arthritis: what is new in 2011? Curr Opin Rheumatol. 2011;23(3):288–292. doi: 10.1097/BOR.0b013e328344f2e4. [DOI] [PubMed] [Google Scholar]
  • 94.Alivernini S, Mazzotta D, Zoli A, Ferraccioli G. Leflunomide treatment in elderly patients with rheumatoid or psoriatic arthritis: retrospective analysis of safety and adherence to treatment. Drugs Aging. 2009;26(5):395–402. doi: 10.2165/00002512-200926050-00003. [DOI] [PubMed] [Google Scholar]
  • 95.Kalden JR, Schattenkirchner M, Sorensen H, et al. The efficacy and safety of leflunomide in patients with active rheumatoid arthritis: a five-year followup study. Arthritis Rheum. 2003;48(6):1513–1520. doi: 10.1002/art.11015. [DOI] [PubMed] [Google Scholar]
  • 96.Baker JF, Sauer BC, Cannon GW, et al. Changes in Body Mass Related to the Initiation of Disease-Modifying Therapies in Rheumatoid Arthritis. Arthritis Rheumatol. 2016;68(8):1818–1827. doi: 10.1002/art.39647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Coblyn JS, Shadick N, Helfgott S. Leflunomide-associated weight loss in rheumatoid arthritis. Arthritis Rheum. 2001;44(5):1048–1051. doi: 10.1002/1529-0131(200105)44:5<1048::AID-ANR184>3.0.CO;2-V. [DOI] [PubMed] [Google Scholar]
  • 98. [Accessed on January 24, 2018]; https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/020905s031lbl.pdf.
  • 99.Rozman B. Clinical pharmacokinetics of leflunomide. Clin Pharmacokinet. 2002;41(6):421–430. doi: 10.2165/00003088-200241060-00003. [DOI] [PubMed] [Google Scholar]
  • 100.Wong SP, Chu CM, Kan CH, Tsui HS, Ng WL. Successful treatment of leflunomide-induced acute pneumonitis with cholestyramine wash-out therapy. J Clin Rheumatol. 2009;15(8):389–392. doi: 10.1097/RHU.0b013e3181c3f87e. [DOI] [PubMed] [Google Scholar]
  • 101.Laub M, Fraser R, Kurche J, Lara A, Kiser TH, Reynolds PM. Use of a Cholestyramine Washout in a Patient With Septic Shock on Leflunomide Therapy: A Case Report and Review of the Literature. J Intensive Care Med. 2016;31(6):412–414. doi: 10.1177/0885066615610108. [DOI] [PubMed] [Google Scholar]
  • 102.Hajdyla-Banas I, Banas T, Rydz-Stryszowska I, et al. Pregnancy course and neonatal outcome after exposure to leflunomide--2 cases report and review of literature. Przegl Lek. 2009;66(12):1069–1071. [PubMed] [Google Scholar]
  • 103.Winthrop KL. The emerging safety profile of JAK inhibitors in rheumatic disease. Nat Rev Rheumatol. 2017;13(4):234–243. doi: 10.1038/nrrheum.2017.23. [DOI] [PubMed] [Google Scholar]
  • 104.Clark JD, Flanagan ME, Telliez JB. Discovery and development of Janus kinase (JAK) inhibitors for inflammatory diseases. J Med Chem. 2014;57(12):5023–5038. doi: 10.1021/jm401490p. [DOI] [PubMed] [Google Scholar]
  • 105.Tanaka Y, Suzuki M, Nakamura H, Toyoizumi S, Zwillich SH. Phase II study of tofacitinib (CP-690,550) combined with methotrexate in patients with rheumatoid arthritis and an inadequate response to methotrexate. Arthritis Care Res (Hoboken) 2011;63(8):1150–1158. doi: 10.1002/acr.20494. [DOI] [PubMed] [Google Scholar]
  • 106.Fleischmann R, Kremer J, Cush J, et al. Placebo-controlled trial of tofacitinib monotherapy in rheumatoid arthritis. N Engl J Med. 2012;367(6):495–507. doi: 10.1056/NEJMoa1109071. [DOI] [PubMed] [Google Scholar]
  • 107.Fleischmann R, Cutolo M, Genovese MC, et al. Phase IIb dose-ranging study of the oral JAK inhibitor tofacitinib (CP-690,550) or adalimumab monotherapy versus placebo in patients with active rheumatoid arthritis with an inadequate response to disease-modifying antirheumatic drugs. Arthritis Rheum. 2012;64(3):617–629. doi: 10.1002/art.33383. [DOI] [PubMed] [Google Scholar]
  • 108.Kremer JM, Cohen S, Wilkinson BE, et al. A phase IIb dose-ranging study of the oral JAK inhibitor tofacitinib (CP-690,550) versus placebo in combination with background methotrexate in patients with active rheumatoid arthritis and an inadequate response to methotrexate alone. Arthritis Rheum. 2012;64(4):970–981. doi: 10.1002/art.33419. [DOI] [PubMed] [Google Scholar]
  • 109.Dresser GK, Spence JD, Bailey DG. Pharmacokinetic-pharmacodynamic consequences and clinical relevance of cytochrome P450 3A4 inhibition. Clin Pharmacokinet. 2000;38(1):41–57. doi: 10.2165/00003088-200038010-00003. [DOI] [PubMed] [Google Scholar]
  • 110.Sager JE, Lutz JD, Foti RS, Davis C, Kunze KL, Isoherranen N. Fluoxetine- and norfluoxetine-mediated complex drug-drug interactions: in vitro to in vivo correlation of effects on CYP2D6, CYP2C19, and CYP3A4. Clin Pharmacol Ther. 2014;95(6):653–662. doi: 10.1038/clpt.2014.50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Westphal JF. Macrolide - induced clinically relevant drug interactions with cytochrome P-450A (CYP) 3A4: an update focused on clarithromycin, azithromycin and dirithromycin. Br J Clin Pharmacol. 2000;50(4):285–295. doi: 10.1046/j.1365-2125.2000.00261.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Pelkonen O, Turpeinen M, Hakkola J, Honkakoski P, Hukkanen J, Raunio H. Inhibition and induction of human cytochrome P450 enzymes: current status. Arch Toxicol. 2008;82(10):667–715. doi: 10.1007/s00204-008-0332-8. [DOI] [PubMed] [Google Scholar]
  • 113.Insinga RP, Itzler RF, Pellissier JM, et al. The incidence of herpes zoster in a United States administrative database. J Gen Intern Med. 2005;20(8):748–53. doi: 10.1111/j.1525-1497.2005.0150.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Smitten AL, Choi HK, Hochberg MC, et al. The risk of herpes zoster in patients with rheumatoid arthritis in the United States and the United Kingdom. Arthritis Rheum. 2007;57(8):1431–1438. doi: 10.1002/art.23112. [DOI] [PubMed] [Google Scholar]
  • 115.Winthrop KL, Yamanaka H, Valdez H, et al. Herpes zoster and tofacitinib therapy in patients with rheumatoid arthritis. Arthritis Rheumatol. 2014;66(10):2675–2684. doi: 10.1002/art.38745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Winthrop KL, Curtis JR, Lindsey S, et al. Herpes zoster and tofacitinib: Clinical outcomes and risk of concomitant therapy. Arthritis Rheumatol. 2017;69(10):1960–1968. doi: 10.1002/art.40189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Rubin LG, Levin MJ, Ljungman P, et al. 2013 IDSA Clinical practice guideline for vaccination of the immunocompromised host. Clin Infect Dis. 2014;58(3):309–18. doi: 10.1093/cid/cit816. [DOI] [PubMed] [Google Scholar]
  • 118.Zhang J, Delzell E, Xie F, et al. The use, safety, and effectiveness of herpes zoster vaccination in individuals with inflammatory and autoimmune diseases: a longitudinal observational study. Arthritis Res Ther. 2011;13(5):R174. doi: 10.1186/ar3497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. [Accessed on January 24, 2018]; https://www.fda.gov/downloads/…/Vaccines/ApprovedProducts/UCM581605.pdf.
  • 120.Cunningham AL, Lal H, Kovac M, et al. Efficacy of the Herpes Zoster Subunit Vaccine in Adults 70 Years of Age or Older. N Engl J Med. 2016;375(11):1019–1032. doi: 10.1056/NEJMoa1603800. [DOI] [PubMed] [Google Scholar]
  • 121.Winthrop KL, Silverfield J, Racewicz A, et al. The effect of tofacitinib on pneumococcal and influenza vaccine responses in rheumatoid arthritis. Ann Rheum Dis. 2016;75(4):687–695. doi: 10.1136/annrheumdis-2014-207191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Man HW, Schafer P, Wong LM, et al. Discovery of (S)-N-[2-[1-(3-ethoxy-4-methoxyphenyl)-2-methanesulfonylethyl]-1,3-dioxo-2,3-dihy dro-1H–isoindol-4-yl] acetamide (apremilast), a potent and orally active phosphodiesterase 4 and tumor necrosis factor-alpha inhibitor. J Med Chem. 2009;52(6):1522–1524. doi: 10.1021/jm900210d. [DOI] [PubMed] [Google Scholar]
  • 123.Mayr B, Montminy M. Transcriptional regulation by the phosphorylation-dependent factor CREB. Nat Rev Mol Cell Biol. 2001;2(8):599–609. doi: 10.1038/35085068. [DOI] [PubMed] [Google Scholar]
  • 124.Ollivier V, Parry GC, Cobb RR, de Prost D, Mackman N. Elevated cyclic AMP inhibits NF-kappaB-mediated transcription in human monocytic cells and endothelial cells. J Biol Chem. 1996;271(34):20828–20835. doi: 10.1074/jbc.271.34.20828. [DOI] [PubMed] [Google Scholar]
  • 125.Claveau D, Chen SL, O’Keefe S, et al. Preferential inhibition of T helper 1, but not T helper 2, cytokines in vitro by L-826,141 [4-[2-(3,4-Bisdifluromethoxyphenyl)-2-[4-(1,1,1,3,3,3-hexafluoro-2-hydroxypropan- 2-yl)-phenyl]-ethyl]3-methylpyridine-1-oxide], a potent and selective phosphodiesterase 4 inhibitor. J Pharmacol Exp Ther. 2004;310(2):752–760. doi: 10.1124/jpet.103.064691. [DOI] [PubMed] [Google Scholar]
  • 126.Eigler A, Siegmund B, Emmerich U, Baumann KH, Hartmann G, Endres S. Anti-inflammatory activities of cAMP-elevating agents: enhancement of IL-10 synthesis and concurrent suppression of TNF production. J Leukoc Biol. 1998;63(1):101–107. doi: 10.1002/jlb.63.1.101. [DOI] [PubMed] [Google Scholar]
  • 127.Kavanaugh A, Mease PJ, Gomez-Reino JJ, et al. Treatment of psoriatic arthritis in a phase 3 randomised, placebo-controlled trial with apremilast, an oral phosphodiesterase 4 inhibitor. Ann Rheum Dis. 2014;73(6):1020–1026. doi: 10.1136/annrheumdis-2013-205056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Cutolo M, Myerson GE, Fleischmann RM, et al. A Phase III, Randomized, Controlled Trial of Apremilast in Patients with Psoriatic Arthritis: Results of the PALACE 2 Trial. J Rheumatol. 2016;43(9):1724–1734. doi: 10.3899/jrheum.151376. [DOI] [PubMed] [Google Scholar]
  • 129.Edwards CJ, Blanco FJ, Crowley J, et al. Apremilast, an oral phosphodiesterase 4 inhibitor, in patients with psoriatic arthritis and current skin involvement: a phase III, randomised, controlled trial (PALACE 3) Ann Rheum Dis. 2016;75(6):1065–1073. doi: 10.1136/annrheumdis-2015-207963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Wells A, Adebajo AO, Aelion JA, et al. Apremilast, an oral phosphodiesterase 4 inhibitor, is associated with long-term (52-week) improvement in the signs and symptoms of psoriatic arthritis in DMARD-naive patients: results from a phase 3, randomized, controlled trial [abstract no. 1543] Arthritis Rheumatol. 2014;66(Suppl 10):S680. [Google Scholar]
  • 131.Efficacy and Safety Study of Apremilast to Treat Active Psoriatic Arthritis (PsA) (PALACE4) [Accessed January 28, 2018];ClinicalTrials.gov website. https://clinicaltrials.gov/ct2/show/results/NCT01307423. Identifier: NCT01307423. Updated August 21, 2017.
  • 132.Edwards CJ, Blanco FJ, Crowley JJ, et al. Apremilast Is Associated with Long-Term DAS-28 (CRP) Remission and Improvements in Skin Disease: Results from a Phase III Study in DMARD/Biologic-Experienced Active Psoriatic Arthritis Patients [abstract no. 1734] Arthritis Rheumatol. 2016;68(Suppl 10) [Google Scholar]
  • 133.Nash P, Ohson K, Walsh J, et al. Early and sustained efficacy with apremilast monotherapy in biological-naive patients with psoriatic arthritis: a phase IIIB, randomised controlled trial (ACTIVE) Ann Rheum Dis. 2018 doi: 10.1136/annrheumdis-2017-211568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. [Accessed on January 14, 2018]; https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/206088s000lbl.pdf.
  • 135.Mease PJ, Kavanaugh A, Gladman D, et al. Long-term safety and tolerability of apremilast, an oral phosphodiesterase 4 inhibitor, in patients with psoriatic arthritis: pooled safety analysis of three phase 3, randomized, controlled trials [abstract no. 310] Arthritis Rheumatol. 2013;65(Suppl 10):S131. [Google Scholar]
  • 136.Papp K, Reich K, Leonardi CL, et al. Apremilast, an oral phosphodiesterase 4 (PDE4) inhibitor, in patients with moderate to severe plaque psoriasis: Results of a phase III, randomized, controlled trial (Efficacy and Safety Trial Evaluating the Effects of Apremilast in Psoriasis [ESTEEM] 1) J Am Acad Dermatol. 2015;73(1):37–49. doi: 10.1016/j.jaad.2015.03.049. [DOI] [PubMed] [Google Scholar]
  • 137.Paul C, Cather J, Gooderham M, et al. Efficacy and safety of apremilast, an oral phosphodiesterase 4 inhibitor, in patients with moderate-to-severe plaque psoriasis over 52 weeks: a phase III, randomized controlled trial (ESTEEM 2) Br J Dermatol. 2015;173(6):1387–1399. doi: 10.1111/bjd.14164. [DOI] [PubMed] [Google Scholar]
  • 138.Sakkas LI, Mavropoulos A, Bogdanos DP. Phosphodiesterase 4 Inhibitors in Immune-mediated Diseases: Mode of Action, Clinical Applications, Current and Future Perspectives. Curr Med Chem. 2017;24(28):3054–3067. doi: 10.2174/0929867324666170530093902. [DOI] [PubMed] [Google Scholar]
  • 139.Genovese MC, Jarosova K, Cieslak D, et al. Apremilast in Patients With Active Rheumatoid Arthritis: A Phase II, Multicenter, Randomized, Double-Blind, Placebo-Controlled, Parallel-Group Study. Arthritis Rheumatol. 2015;67(7):1703–1710. doi: 10.1002/art.39120. [DOI] [PubMed] [Google Scholar]
  • 140.Smolen JS, Breedveld FC, Burmester GR, et al. Treating rheumatoid arthritis to target: 2014 update of the recommendations of an international task force. Ann Rheum Dis. 2016;75(1):3–15. doi: 10.1136/annrheumdis-2015-207524. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES