Rheumatoid Arthritis

Back

Practice Essentials

Rheumatoid arthritis (RA) is a chronic systemic inflammatory disease of unknown cause. An external trigger (eg, cigarette smoking, infection, or trauma) that triggers an autoimmune reaction, leading to synovial hypertrophy and chronic joint inflammation along with the potential for extra-articular manifestations, is theorized to occur in genetically susceptible individuals. See the image below.



View Image

Rheumatoid changes in the hand. Photograph by David Effron MD, FACEP.

See Rheumatoid Arthritis: In and Out of the Joint, a Critical Images slideshow, to help identify the distinguishing features of RA as well as the signs of extra-articular manifestations of this disfiguring disease.

Signs and symptoms

In most patients with RA, onset is insidious, often beginning with fever, malaise, arthralgias, and weakness before progressing to joint inflammation and swelling.

Signs and symptoms of RA may include the following:

The physical examination should address the following:

During the physical examination, it is important to assess the following:

Guidelines for evaluation

See Presentation for more detail.

Diagnosis

No test results are pathognomonic; instead, the diagnosis is made by using a combination of clinical, laboratory, and imaging features. Potentially useful laboratory studies in suspected RA include the following:

Potentially useful imaging modalities include the following:

Joint aspiration and analysis of synovial fluid may be considered, including the following:

See Workup for more detail.

Management

Nonpharmacologic, nonsurgical therapies include the following:

Guidelines for pharmacologic therapy

Nonbiologic disease-modifying antirheumatic drugs (DMARDS) include the following:

Biologic tumor necrosis factor (TNF)–inhibiting DMARDs include the following:

Biologic non-TNF DMARDs include the following:

Other drugs used therapeutically include the following:

Surgical treatments include the following:

See Treatment and Medication for more detail.

Background

The hallmark feature of rheumatoid arthritis (RA) is persistent symmetric polyarthritis (synovitis) that affects the hands and feet, though any joint lined by a synovial membrane may be involved. Extra-articular involvement of organs such as the skin, heart, lungs, and eyes can be significant. (See Presentation.)

No laboratory test results are pathognomonic for RA, but the presence of anti-cyclic citrullinated protein antibody (ACPA) and rheumatoid factor (RF) is highly specific for this condition. (See Workup.)

Optimal care of patients with RA requires an integrated approach that includes nonpharmacologic therapies and pharmacologic agents such as nonbiologic and biologic disease-modifying antirheumatic drugs (DMARDs), nonsteroidal anti-inflammatory drugs (NSAIDs), analgesics, and corticosteroids. (See Treatment and Medication.)

Early therapy with DMARDs has become the standard of care; it not only can more efficiently retard disease progression than later treatment but also may induce more remissions. (See Treatment.) Many of the newer DMARD therapies, however, are immunosuppressive in nature, leading to a higher risk for infections. (See Complications.)

Macrophage activation syndrome is a life-threatening complication of juvenile idiopathic arthritis (JIA) that necessitates immediate treatment with high-dose steroids and cyclosporine. (See Complications.)

The following guidelines on treating RA to therapeutic target were issued in 2015 by an international task force of rheumatologists, patient representatives, and a rheumatology nurse[7, 8] :

Pathophysiology

The pathogenesis of RA is not completely understood. An external trigger (eg, cigarette smoking, infection, or trauma) that sets off an autoimmune reaction, leading to synovial hypertrophy and chronic joint inflammation along with the potential for extra-articular manifestations, is theorized to occur in genetically susceptible individuals.

Synovial cell hyperplasia and endothelial cell activation are early events in the pathologic process that progresses to uncontrolled inflammation and consequent cartilage and bone destruction. Genetic factors and immune system abnormalities contribute to disease propagation.

CD4 T cells, mononuclear phagocytes, fibroblasts, osteoclasts, and neutrophils play major cellular roles in the pathophysiology of RA, whereas B cells produce autoantibodies (ie, rheumatoid factors). Abnormal production of numerous cytokines, chemokines, and other inflammatory mediators has been demonstrated in patients with RA, including the following:

Ultimately, inflammation and exuberant proliferation of the synovium (ie, pannus) leads to destruction of various tissues, including cartilage (see the image below), bone, tendons, ligaments, and blood vessels. Although the articular structures are the primary sites involved by RA, other tissues are also affected.



View Image

This gross photo shows destruction of the cartilage and erosion of the underlying bone with pannus from a patient with rheumatoid arthritis.

Etiology

The cause of RA is unknown. Genetic, environmental, hormonal, immunologic, and infectious factors may play significant roles. Socioeconomic, psychological, and lifestyle factors (eg, tobacco use, the main environmental risk[9] ) may influence disease development and outcome.

Genetic factors

Genetic factors account for 50% of the risk for developing RA.[10] About 60% of RA patients in the United States carry a shared epitope of the human leukocyte antigen (HLA)-DR4 cluster, which constitutes one of the peptide-binding sites of certain HLA-DR molecules associated with RA (eg, HLA-DR beta *0401, 0404, or 0405). HLA-DR1 (HLA-DR beta *0101) also carries this shared epitope and confers risk, particularly in certain southern European areas. Other HLA-DR4 molecules (eg, HLA-DR beta *0402) lack this epitope and do not confer this risk.

Genes other than those of the major histocompatibility complex (MHC) are also involved. Results from sequencing genes of families with RA suggest the presence of several resistance and susceptibility genes, including PTPN22 and TRAF5.[11, 12]

Juvenile idiopathic arthritis (JIA), also known as juvenile rheumatoid arthritis (JRA), is a heterogeneous group of diseases that differs markedly from adult RA. JIA is known to have genetically complex traits in which multiple genes are important for disease onset and manifestations, and it is characterized by arthritis that begins before the age of 16 years, persists for more than 6 weeks, and is of unknown origin.[13] The IL2RA/CD25 gene has been implicated as a JIA susceptibility locus, as has the VTCN1 gene.[14]

Some investigators suggest that the future of treatment and understanding of RA may be based on imprinting and epigenetics. RA is significantly more prevalent in women than in men,[15, 16] which suggests that genomic imprinting from parents participates in its expression.[17, 18] Imprinting is characterized by differential methylation of chromosomes by the parent of origin, resulting in differential expression of maternal over paternal genes.[19]

Epigenetics is the change in DNA expression that is due to environmentally induced methylation and not to a change in DNA structure. Clearly, the research focus will be on environmental factors in combination with immune genetics.

Infectious agents

For many decades, numerous infectious agents have been suggested as potential causes of RA, including Mycoplasma organisms, Epstein-Barr virus (EBV), and rubella virus. This suggestion is indirectly supported by the following evidence:

Emerging evidence also points to an association between RA and periodontopathic bacteria. For example, the synovial fluid of RA patients has been found to contain high levels of antibodies to anaerobic bacteria that commonly cause periodontal infection, including Porphyromonas gingivalis.[20, 21]

Hormonal factors

Sex hormones may play a role in RA, as evidenced by the disproportionate number of females with this disease, its amelioration during pregnancy, its recurrence in the early postpartum period, and its reduced incidence in women using oral contraceptives. Hyperprolactinemia may be a risk factor for RA.[22]

Immunologic factors

All of the major immunologic elements play fundamental roles in initiating, propagating, and maintaining the autoimmune process of RA. The exact orchestration of the cellular and cytokine events that lead to pathologic consequences (eg, synovial proliferation and subsequent joint destruction) is complex, involving T and B cells, antigen-presenting cells (eg, B cells, macrophages, and dendritic cells), and various cytokines. Aberrant production and regulation of both proinflammatory and anti-inflammatory cytokines and cytokine pathways are found in RA.

T cells are assumed to play a pivotal role in the initiation of RA, and the key player in this respect is assumed to be the T helper 1 (Th1) CD4 cells. (Th1 cells produce IL-2 and interferon [IFN] gamma.) These cells may subsequently activate macrophages and other cell populations, including synovial fibroblasts. Macrophages and synovial fibroblasts are the main producers of TNF-a and IL-1. Experimental models suggest that synovial macrophages and fibroblasts may become autonomous and thus lose responsiveness to T-cell activities in the course of RA.

B cells are important in the pathologic process and may serve as antigen-presenting cells. B cells also produce numerous autoantibodies (eg, RF and ACPA) and secrete cytokines.

The hyperactive and hyperplastic synovial membrane is ultimately transformed into pannus tissue and invades cartilage and bone, with the latter being degraded by activated osteoclasts. The major difference between RA and other forms of inflammatory arthritis, such as psoriatic arthritis, lies not in their respective cytokine patterns but, rather, in the highly destructive potential of the RA synovial membrane and in the local and systemic autoimmunity.

Whether these 2 events are linked is unclear; however, the autoimmune response conceivably leads to the formation of immune complexes that activate the inflammatory process to a much higher degree than normal. This theory is supported by the much worse prognosis of RA among patients with positive RF results.

Epidemiology

Worldwide, the annual incidence of RA is approximately 3 cases per 10,000 population, and the prevalence rate is approximately 1%, increasing with age and peaking between the ages of 35 and 50 years. RA affects all populations, though it is much more prevalent in some groups (eg, 5-6% in some Native American groups) and much less prevalent in others (eg, black persons from the Caribbean region).

First-degree relatives of individuals with RA are at 2- to 3-fold higher risk for the disease. Disease concordance in monozygotic twins is approximately 15-20%, suggesting that nongenetic factors play an important role. Because the worldwide frequency of RA is relatively constant, a ubiquitous infectious agent has been postulated to play an etiologic role.

Women are affected by RA approximately 3 times more often than men are,[15, 16] but sex differences diminish in older age groups.[15] In investigating whether the higher rate of RA among women could be linked to certain reproductive risk factors, a study from Denmark found that the rate of RA was higher in women who had given birth to just 1 child than in women who had delivered 2 or 3 offspring.[23] However, the rate was not increased in women who were nulliparous or who had a history of lost pregnancies.

Time elapsed since pregnancy is also significant. In the 1- to 5-year postpartum period, a decreased risk for RA has been recognized, even in those with higher-risk HLA markers.[24]

The Danish study also found a higher risk of RA among women with a history of preeclampsia, hyperemesis during pregnancy, or gestational hypertension.[23] In the authors’ view, this portion of the data suggested that a reduced immune adaptability to pregnancy may exist in women who are predisposed to the development of RA or that there may be a link between fetal microchimerism (in which fetal cells are present in the maternal circulation) and RA.[23]

Prognosis

Outcome in RA is compromised when diagnosis and treatment are delayed. The clinical course of RA is generally one of exacerbations and remissions. Approximately 40% of patients with this disease become disabled after 10 years, but outcomes are highly variable.[25] Some patients experience a relatively self-limited disease, whereas others have a chronic progressive illness.

Prognostic factors

Intervention with DMARDs in very early RA (symptom duration < 12 weeks at the time of first treatment) provides the best opportunity for achieving disease remission.[26] Better detection of early joint injury has provided a previously unappreciated view of the ubiquity and importance of early joint damage. Nonetheless, predicting the long-term course of an individual case of RA at the outset remains difficult, though the following all correlate with an unfavorable prognosis in terms of joint damage and disability:

In a retrospective study that used logistic regression to analyze clinical and laboratory assessments in patients with RA who took only methotrexate, the authors found that measures of C-reactive protein (CRP) and swollen joint count after 12 weeks of methotrexate administration were most associated with radiographic progression at week 52.[27]

The prognosis of RA is generally much worse among patients with positive RF results. For example, the presence of RF in sera has been associated with severe erosive disease.[28, 29] However, the absence of RF does not necessarily portend a good prognosis.

Other laboratory markers of a poor prognosis include early radiologic evidence of bony injury, persistent anemia of chronic disease, elevated levels of the C1q component of complement, and the presence of ACPA (see Workup). In fact, the presence of ACPA and antikeratin antibodies (AKA) in sera has been linked with severe erosive disease,[28] and the combined detection of these autoantibodies can increase the ability to predict erosive disease in RA patients.[29]

RA that remains persistently active for longer than 1 year is likely to lead to joint deformities and disability.[30] Periods of activity lasting only weeks or a few months followed by spontaneous remission portend a better prognosis.

A study by Mollard et al of 8189 women in a US-wide observational cohort who developed RA before menopause found greater functional decline in postmenopausal women than in premenopausal ones; furthermore, the trajectory of functional decline worsened and accelerated after menopause.  However, ever-use of hormonal replacement therapy, ever having a pregnancy, and longer length of reproductive life were associated with less functional decline.[31]

Morbidity and mortality

Most data on RA disability rates derive from specialty units caring for referred patients with severe disease. Little information is available on patients cared for in primary care community settings. Estimates suggest that more than 50% of these patients remain fully employed, even after 10-15 years of disease, with one third having only intermittent low-grade disease and another one third experiencing spontaneous remission.

RA is associated with traditional and nontraditional cardiovascular risk factors. The leading cause of excess mortality in RA is cardiovascular disease, followed by infection, respiratory disease, and malignancies. The effects of concurrent therapy, which is often immunosuppressive, may contribute to mortality in RA. However, studies suggest that control of inflammation may improve mortality.

Nontraditional risk factors appear to play an important role in cardiovascular morbidity and mortality. Myocardial infarction, myocardial dysfunction, and asymptomatic pericardial effusions are common; symptomatic pericarditis and constrictive pericarditis are rare. Myocarditis, coronary vasculitis, valvular disease, and conduction defects are occasionally observed. A large Danish cohort study suggested the presence of an increased risk of atrial fibrillation and stroke in patients with RA.[32]

The overall mortality in patients with RA is reportedly 2.5 times higher than that of the general age-matched population. In the 1980s, mortality among those with severe articular and extra-articular disease approached that among patients with 3-vessel coronary disease or stage IV Hodgkin lymphoma. Much of the excess mortality derives from infection, vasculitis, and poor nutrition. With the exception of lymphoma, mortality from cancer is unchanged.

Patient Education

Patient education and counseling help to reduce pain, disability, and frequency of physician visits. These may represent the most cost-effective intervention for RA.[33, 34]

Informing patient of diagnosis

With a potentially disabling disease such as RA, the act of informing the patient of the diagnosis takes on major importance. The goal is to satisfy the patient’s informational needs regarding the diagnosis, prognosis, and treatment in appropriate detail. To understand the patient’s perspective, requests, and fears, the physician must employ careful questioning and empathic listening.

Telling patients more than they are intellectually or psychologically prepared to handle (a common practice) risks making the experience so intense as to trigger withdrawal. Conversely, failing to address issues of importance to the patient compromises the development of trust. The patient needs to know that the primary physician understands the situation and is available for support, advice, and therapy as the need arises. Encouraging the patient to ask questions helps to communicate interest and caring.

Discussing prognosis and treatment

Patients and families do best when they know what to expect and can view the illness realistically. Many patients fear crippling consequences and dependency. Accordingly, it is valuable to provide a clear description of the most common disease manifestations. Without encouraging false hopes, the physician can point out that spontaneous remissions can occur and that more than two thirds of patients live independently without major disability. In addition, emphasize that much can be done to minimize discomfort and to preserve function.

A review of available therapies and their efficacy helps patients to overcome feelings of depression stemming from an erroneous expectation of inevitable disability.[35] (See Treatment). Even in those with severe disease, guarded optimism is now appropriate, given the host of effective and well-tolerated disease-modifying treatments that are emerging.

Dealing with misconceptions

Several common misconceptions regarding RA deserve attention. Explaining that no known controllable precipitants exist helps to eliminate much unnecessary guilt and self-recrimination. Dealing in an informative, evidence-based fashion with a patient who expresses interest in alternative and complementary forms of therapy can help limit expenditures on ineffective treatments.

Another misconception is that a medication must be expensive to be helpful. Generic NSAIDs, low-dose prednisone,[25] and the first-line DMARDs are quite inexpensive yet remarkably effective for relieving symptoms, a point that bears emphasizing. The belief that one must be given the latest TNF inhibitor to be treated effectively can be addressed by a careful review of the overall treatment program and the proper role of such agents in the patient’s plan of care.

Active participation of the patient and family in the design and implementation of the therapeutic program helps boost morale and to ensure compliance, as does explaining the rationale for the therapies used.

The family also plays an important part in striking the proper balance between dependence and independence. Household members should avoid overprotecting the patient (eg, the spouse refraining from intercourse out of fear of hurting the patient) and should work to sustain the patient’s pride and ability to contribute to the family. Allowing the patient with RA to struggle with a task is sometimes constructive.

Supporting patient with debilitating disease

Abandonment is a major fear in these individuals. Patients are relieved to know that they will be closely observed by the primary physician and healthcare team, working in conjunction with a consulting rheumatologist and physical/occupational therapist, all of whom are committed to maximizing the patient’s comfort and independence and to preserving joint function. With occupational therapy, the treatment effort is geared toward helping the patient maintain a meaningful work role within the limitations of the illness.

Persons with long-standing severe disease who have already sustained much irreversible joint destruction benefit from an emphasis on comfort measures, supportive counseling, and attention to minimizing further debility. Such patients need help in grieving for their disfigurement and loss of function.

An accepting, unhurried, empathic manner allows the patient to express feelings. The seemingly insignificant act of touching does much to restore a sense of self-acceptance. Attending to pain with increased social support, medication, and a refocusing of attention to function is useful. A trusting and strong patient-doctor relationship can do much to sustain a patient through times of discomfort and disability.

For more information, see the Arthritis Center and Pain Management Center, as well as Rheumatoid Arthritis, Juvenile Rheumatoid Arthritis, Rheumatoid Arthritis Medications, Chronic Fatigue Syndrome, and Chronic Pain.

History

The hallmark feature of rheumatoid arthritis (RA) is persistent symmetric polyarthritis (synovitis) that affects the hands and feet, although any joint lined by a synovial membrane may be involved. The severity of RA may fluctuate over time, but chronic RA most commonly results in the progressive development of various degrees of joint destruction, deformity, and a significant decline in functional status. Extra-articular involvement of organs such as the skin, heart, lungs, and eyes can also be significant.

Juvenile idiopathic arthritis (JIA), sometimes referred to as juvenile rheumatoid arthritis (JRA), is the most common form of childhood arthritis. In most patients, the immunogenic associations, clinical pattern, and functional outcome of JIA are different from those of adult-onset RA.

Patients with RA may report difficulty performing activities of daily living (ADLs), such as dressing, standing, walking, personal hygiene, or use of their hands. In addition to articular deterioration, constitutional symptoms (eg, fatigue, malaise, morning stiffness, weight loss, and low-grade fever) may be present.

In most patients, RA has an insidious onset. It may begin with systemic features (eg, fever, malaise, arthralgias, and weakness) before the appearance of overt joint inflammation and swelling. A small percentage (approximately 10%) of patients with this disease have an abrupt onset with the acute development of synovitis and extra-articular manifestations. Spontaneous remission is uncommon, especially after the first 3-6 months.

Physical Examination

During the physical examination, it is important to assess the following:

Joint involvement is the characteristic feature of RA. In general, the small joints of the hands and feet are affected in a relatively symmetric distribution. In decreasing frequency, the metacarpophalangeal (MCP), wrist, proximal interphalangeal (PIP), knee, metatarsophalangeal (MTP), shoulder, ankle, cervical spine, hip, elbow, and temporomandibular joints are most commonly affected.

Affected joints show inflammation with swelling, tenderness, warmth, and decreased range of motion (ROM). Atrophy of the interosseous muscles of the hands is a typical early finding. Joint and tendon destruction may lead to deformities such as ulnar deviation, boutonniere and swan-neck deformities, hammer toes, and, occasionally, joint ankylosis.

Other commonly observed musculoskeletal manifestations include the following:

Most patients with RA have muscle atrophy from disuse, which is often secondary to joint inflammation.

Examination of upper extremities

Fingers

The boutonniere deformity (see the image below) describes nonreducible flexion at the PIP joint along with hyperextension of the distal interphalangeal (DIP) joint of the finger.



View Image

Boutonniere deformity.

This deformity occurs as a result of synovitis stretching or rupturing the PIP joint through the central extensor tendon, with concomitant volar displacement of the lateral bands. When the lateral bands have subluxed far enough to pass the transverse axis of the joint, they become flexors of the PIP joint. Hyperextension of the DIP joint occurs as the tendons shorten with time. A compensatory and reducible hyperextension may occur at the MCP joint. Consequences of boutonniere deformity are loss of thumb mobility and pincher grasp.

Swan-neck deformity of the finger describes hyperextension at the PIP joint with flexion of the DIP joint (see the image below).



View Image

Rheumatoid changes in the hand. Photograph by David Effron MD, FACEP.

This deformity may be initiated either (a) by disruption of the extensor tendon at the DIP joint, with secondary shortening of the central extensor tendon and hyperextension of the PIP joint, or (b) by volar herniation of the PIP joint capsule due to weakening from chronic synovitis, with subsequent tightening of the lateral bands and central extensor tendon. The lateral bands may become shortened over time and lie dorsally, limiting PIP flexion and ineffectively extending the DIP joint.

Tightness of intrinsic muscles (eg, the interossei and lumbricals) may cause major declines in finger mobility. This tightness is ascertained on examination when the PIP joint cannot be flexed while the MCP joint is fully extended but can be flexed while the MCP is in flexion (Bunnell test); primary PIP joint pathology is evident with the MCP joint in either position. For accurate assessment, the phalanx must be aligned with the metacarpal; when ulnar deviation at the MCP joint exists, the intrinsic muscles on the ulnar side are slack, allowing more motion.

Flexor tenosynovitis of the fingers is common and suggests a poor prognosis. “Triggering” of the finger occurs when thickening or nodule formation of the tendon interacts with the concomitant tenosynovial proliferation, trapping the tendon in a flexed position (stenosing tenosynovitis). Tendon rupture may occur as a consequence of infiltrative synovitis in the digit or bony erosion of the tendon at the wrist (especially the flexor pollicis longus).

Arthritis mutilans (sometimes called opera glass hands) results if destruction is severe and extensive, with dissolution of bone. In the small joints of the hands, the phalanges may shorten, and the joints may become grossly unstable. Pulling on the fingers during examination may lengthen the digit in a manner resembling the opening of opera glasses, or the joint may bend in unusual directions merely under the pull of gravity.

Metacarpophalangeal joints

Two typical deformities that alter the alignment of the palmar skeletal arches and the stability of the fingers may occur at the MCP joints: volar subluxation and ulnar deviation (see the image below).



View Image

Subluxation in the metacarpophalangeal joints, with ulnar deviation, in a patient with rheumatoid arthritis of the hands.

Most cases of ulnar deviation are accompanied by counterpoised radial deviation of the wrist, roughly proportional to the degree of ulnar deviation of the fingers. The volar plate is firmer and more substantial than other portions of the MCP joint capsule, thus effectively limiting extension and dorsal movement at the joint. The greater strength of the flexor muscles relative to the extensor muscles causes volar migration of the proximal phalanx after synovial-based inflammation has weakened ligament and tendon insertions about the MCP joint capsule.

Ulnar deviation occurs after synovitis has led to stretching and attenuation of the volar plate and collateral ligaments, allowing dislocation of the flexor tendon volarward and ulnarward. The supporting structures of the extensor tendons also may become attenuated or destroyed by synovial distention and invasion, loosening the tendons so that they no longer ride centrally and dorsally over the metacarpal head but move into the cleft between the MCP joints.

If the extensor tendon subluxation is beyond the transverse axis of the MCP joint, the tendon becomes a flexor at that joint, further limiting the active extension of the fingers.

Wrists

Multiple deformities may occur in the wrist. Disruption of the radioulnar joint with dorsal subluxation of the ulna (caput ulnae) as well as rotation of the carpus on the distal radius with an ulnarly translocated lunate are common. The combination of an ulnar drift of the fingers and carpal rotation is known as a zigzag deformity. Shortening of the carpal height (noted on radiographs), due in part to cartilage loss, is seen with rotational deformities.

Dorsal subluxation of the ulna often allows the ulnar styloid to be depressed volarly on examination, much as a piano key is depressed when played. It may lead to rupture of the extensor tendons of the little, ring, and long fingers because the end of the distal ulna is roughened secondary to erosion of bone and may abrade the tendons as they move during normal hand function, much as a rope is frayed when rubbed over a sharp rock (see the image below). This process is especially likely to lead to tendon rupture if tenosynovitis is present.



View Image

Coronal, T1-weighted magnetic resonance imaging scan shows characteristic pannus and erosive changes in the wrist in a patient with active rheumatoid ....

Entrapment neuropathy may result from synovitis about the flexor tendons. Entrapment of the median nerve as it passes through the carpal tunnel leads to decreased sensation on the palmar aspect of the thumb, index finger, and long finger and on the radial aspect of the ring finger; weakness and atrophy of the muscles in the thenar eminence also occurs. Entrapment of the ulnar nerve at the wrist, a less frequent event, causes decreased sensation over the little finger and the ulnar aspect of the ring finger and decreased interosseous muscle strength and mass.

Elbow

Elbow involvement is often detected by palpable synovial proliferation at the radiohumeral joint and is commonly accompanied by a flexion deformity, such as in contractures. Involvement of the olecranon bursa is common, as are rheumatoid nodules in the bursa and along the extensor surface of the ulna (see the image below).



View Image

Rheumatoid nodules at the elbow. Photograph by David Effron MD, FACEP.

Shoulders

RA commonly involves the shoulders and is manifested by tenderness, nocturnal pain, and limited motion. Initially, swelling occurs anteriorly, but it may be difficult to detect and is present on examination in a minority of patients at any point in time.

Rotator cuff degeneration secondary to synovitis may limit abduction and rotation. Superolateral migration of the humerus occurs with complete tears. Glenohumeral damage results in pain with motion and at rest and typically leads to severely restricted motion or “frozen shoulder syndrome.” Acromioclavicular arthritis is not as frequent or as disabling as the other manifestations of this disease.

Examination of lower extremities

Ankles and feet

The ankle joint itself is rarely involved without midfoot or MTP involvement. Because it is a mortise joint, it does not often deform. Major structural changes occur in the midfoot and foot as a result of the combination of chronic synovitis and weight bearing. Posterior tibialis tendon involvement or rupture may lead to subtalar subluxation, which results in eversion and migration of the talus laterally. Midfoot disease leads to loss of normal arch contour with flattening of the feet.

The MTP joints are inflamed in most patients and, because of the heavy loads they bear, often become deformed over time. The great toe typically develops hallux valgus (a bunion); subluxation of the phalanx at the MTP joint of the other toes predominantly occurs dorsally. Toes may exhibit compensatory flexion due to a fixed length of the flexor tendons—so-called hammer toes. The second and third metatarsal heads commonly protrude and may become the primary weight-bearing surface at the MTP joints. Calluses and pain upon weight bearing result.

Knees

Affected knees may develop large effusions and abundant accumulation of synovium. Knee effusions and synovial thickening are common in RA and are easily detected during the early course of the disease. Persistent effusions may lead to inhibition of quadriceps function by spinal reflexes, resulting in subsequent atrophy.

Instability may develop after progressive loss of cartilage and weakening of ligaments; deformity may include genu valgus or varus, as well as flexion deformities. The patient’s energy expenditure for standing or walking increases substantially if there are flexion deformities of the knees.

Hips

The hips are commonly involved in RA; however, because of their deep location, their involvement is not always readily apparent early in the course of the disease. Hips are difficult to examine by means of direct inspection or palpation.

Limited motion or pain on motion and weight bearing are the hallmarks of hip involvement. The Patrick maneuver (flexion, external rotation, and abduction) is abnormal in this situation. A flexion deformity may be demonstrable by conducting a Thomas test, which is performed by flexing one hip (with the patient supine) while restricting pelvic motion by keeping the other hip in the neutral position on the examination table. If the hip cannot be maintained in the neutral position, a contracture is present.

Cervical spine

Cervical spine involvement (see the following image) usually affects C1-C2 and has the potential to cause serious neurologic consequences. Patients who are to undergo intubation or procedures that may involve manipulation of the neck should undergo careful evaluation of the cervical spine.



View Image

Ankylosis in the cervical spine at several levels due to long-standing juvenile rheumatoid arthritis (also known as juvenile idiopathic arthritis).

Neck pain on motion and occipital headache are common manifestations of cervical spine involvement. Most patients with cervical spine involvement (see the image below) have had the disease for more than 10 years.



View Image

Lateral view of the cervical spine in a patient with rheumatoid arthritis shows erosion of the odontoid process.

Clinical manifestations of early cervical spine disease consist primarily of neck stiffness that is perceived throughout the entire arc of motion. The atlantoaxial joint is a synovial-lined joint and is susceptible to the same proliferative synovitis and subsequent instability seen in the peripheral joints. Patients with severe destruction in the hands (arthritis mutilans) are very likely to have symptomatic cervical spine abnormalities, as are those patients taking significant amounts of corticosteroids for control of RA.

Neurologic involvement in the cervical spine ranges from radicular pain to a variety of spinal cord lesions that may result in weakness (including quadriparesis), sphincter dysfunction, sensory deficits, and pathologic reflexes.

Transient ischemic attacks (TIAs) and cerebellar signs may reflect vertebral artery impingement from cervical subluxation or basilar artery impingement from upward migration of the dens. Tenosynovitis of the transverse ligament of C1 may lead to C1-C2 instability. Myelopathy secondary to rupture of the transverse ligament may lead to neurologic deficits. Radiculopathy is most common at the C2 root, though symptomatic subluxations may occur at any level.

Symptoms of cervical myelopathy are gradual in onset and are often unrelated to either the development or the accentuation of neck pain. When neck pain does occur, it frequently radiates over the occipital region in the distribution of the C1-3 nerve roots. The Lhermitte sign, in which tingling paresthesia that descends through the thoracolumbar spine occurs as the cervical spine is flexed, is typically observed.

For more information, see Rheumatoid Arthritis of the Cervical Spine.

Stiffness, Tenderness, and Pain on Motion

On physical examination, stiffness in patients with RA is determined by limitation of motion, which may vary with the time of day. However, stiffness that is due to articular surface derangement or soft tissue contractures about the joint does not vary with the time of day.

Severe stiffness in the hands may improve with heat, but it is most effectively relieved with active exercise. These modalities reduce stiffness immediately after application, but unfortunately, they do not prevent the return of stiffness.

Direct palpation can elicit joint tenderness, which can vary significantly among patients and with the method of application of force used. To minimize variation over time, the examiner should try to apply approximately the same pressure for each patient examined.

The enlarged synovial membrane, periarticular ligaments, and supporting structures are the major pain-sensitive structures. Muscles may also become tender, but rarely is this due to myositis. Muscle tenderness is not specific for RA. Severe muscle tenderness should suggest another differential diagnosis, including fibromyalgia or a regional pain disorder (see DDx). Bony prominences are generally tender; periarticular structures tend to be more vulnerable to palpation at these sites.

Pain on motion is often used as a surrogate for tenderness in joints that are not readily amenable to direct palpation because of overlying muscle and other tissues. Such areas include the cervical spine, shoulder, and hip.

Pain on motion of the joint may be due to noninflammatory processes that also interfere with the joint’s normal, almost frictionless motion, including damage of cartilage and bone. Additionally, joint instability or subluxation causes pain on motion because of musculotendinous imbalances across the joint. Documenting the positions of motion at which pain occurs can be useful.

Swelling, Deformity, and Limitation of Motion

In RA, enlargement of the synovial membrane is noted on physical examination as thickening of the synovium that may obscure joint margins. This thickening is most evident in the small joints of the hands and feet. In the MCP and MTP joints, the outline of the base of the proximal phalanx may become indistinct, and in the PIP joints of the fingers, a fusiform swelling is noted that is due to the anatomy of the synovial reflections (see the image below).



View Image

Soft-tissue swelling and early erosions in the proximal interphalangeal joints in a patient with rheumatoid arthritis of the hands.

If synovial proliferation is abundant, the resultant soft-tissue mass may have a doughy texture on palpation. Such synovial proliferation is commonly identified in the PIP, MCP, elbow, ankle, MTP, and knee joints, as well as in the flexor tendons of the fingers, the common extensor compartment of the dorsal wrist, and the extensor carpi ulnaris tendon sheath.

Joint effusions may also contribute to swelling by distending the joint. When the effusion is put under increased pressure with joint flexion, the synovium may be forced between articular structures, with the result that a portion becomes trapped and separated from the rest of the joint, forming a Baker cyst (see the image below). More fluid is forced into the structure with subsequent loading of the distended joint, and a 1-way valve effect may prevent the fluid from returning to the joint.



View Image

Anteroposterior radiograph of the knee shows uniform joint-space loss in the medial and lateral knee compartments without osteophytosis. A Baker cyst ....

Baker cysts may be seen in most peripheral joints and are most commonly recognized in the knee.[36] The larger the effusion, the more likely it is that a painful cyst will develop in the popliteal fossa. Rupture of a Baker cyst at the knee may resemble acute thrombophlebitis, with distal dissection of inflammatory joint contents along fascial planes as far as the ankle and dorsal foot.

Deformity of the joint may develop over time as articular and supporting structures are damaged by the inflammatory process. By the time deformity has developed, the diagnosis of RA is in little doubt; however, optimal management of RA requires that the inflammatory aspects of the arthritis be recognized before the development of deformity.

Loss of cartilage from proteolytic and mechanical degradation, combined with stretching and weakening of the periarticular ligaments and their attachments, allows forces acting across the joints to deform them. The small joints in the hands and feet are most commonly deformed in this manner; more than 10% of patients with RA develop deformity of the small joints of the hands within the first 2 years of the disease, and at least 33% develop such deformities over time. Joint instability is seen if disruption of supporting structures has occurred.

Limitation of motion occurs as a result of articular surface damage, joint and tendon sheath swelling, or alteration of joint supporting structures. Effusion may limit joint motion through pain or by causing sufficient tightness of the joint capsule to impede joint mobility. Fibrosis involving tendons and muscles may limit normal joint motion and result in flexion contractures. Joint deformities and subluxations invariably limit motion because of mechanical factors.

Extra-articular Manifestations

RA is a systemic disease, and most individuals with this condition experience extra-articular manifestations such as generalized malaise and fatigue. Rarely, a patient presents with extra-articular manifestations before the onset of arthritis. Some of these manifestations are more common in men (eg, pleural involvement, vasculitis, and pericarditis), but overall, the sex distribution of extra-articular manifestations of the disease is similar to that of RA.

Rheumatoid nodules

Rheumatoid nodules occur in approximately 25% of patients with RA, but they occur in fewer than 10% of patients during the first year of the disease. These lesions are most commonly found on extensor surfaces or sites of frequent mechanical irritation.

The olecranon process, the proximal ulna, the back of the heel, the occiput, and the ischial tuberosities are common periosteal sites for rheumatoid nodule development. Nodules may also form in subcutaneous tissues of the fingers, in toe and heel pads, in tendons, and in viscera. Rheumatoid factor (RF) is almost invariably present in patients with rheumatoid nodules; the absence of RF suggests other diagnoses.

Frequently, there is a discrepancy between the level of articular inflammation and the progression of nodule formation. Patients with rheumatoid nodulosis have a great number of nodules, usually subcutaneous, and may have little active synovitis. In a similar fashion, patients whose articular inflammation responds well to treatment with methotrexate (MTX) may have a seemingly paradoxical rapid increase in the number of nodules.

Effects on organ systems

RA affects several organ systems, as follows:

Subcutaneous nodules (rheumatoid nodules) develop in many RA patients whose RF value is abnormal, often over pressure points (eg, olecranon). Vasculitic lesions of the skin may manifest as palpable purpura or skin ulceration (eg, leg ulceration). Additionally, palmar erythema and pyoderma gangrenosum may be noted.

Cardiovascular morbidity and mortality are increased in patients with RA. Nontraditional risk factors appear to play an important role. Myocardial infarction, myocardial dysfunction, and asymptomatic pericardial effusions are common; symptomatic pericarditis and constrictive pericarditis are rare. Myocarditis, coronary vasculitis, valvular disease, and conduction defects are occasionally observed.

RA involvement of the lungs may take several forms, including pleural effusions, interstitial fibrosis, nodules, and bronchiolitis obliterans organizing pneumonia. Methotrexate therapy can induce interstitial fibrosis that may be difficult to distinguish from that which naturally occurs in patients with RA.

The kidneys usually are not directly affected by RA. Secondary involvement is common, including that due to medications (eg, nonsteroidal anti-inflammatory drugs [NSAIDs], gold, and cyclosporine), inflammation (eg, amyloidosis), and associated diseases (eg, Sjögren syndrome with renal tubular abnormalities).

GI involvement, like renal involvement, is often secondary to associated processes such as medication effects, inflammation, and other diseases. The liver may be affected in patients with Felty syndrome (ie, RA, splenomegaly, and neutropenia).

Vasculitic lesions can occur in any organ, but they are most commonly found in the skin. Lesions may present as palpable purpura, skin ulcers, or digital infarcts.

Some patients with active RA have an anemia of chronic disease. Several hematologic parameters parallel disease activity, including normochromic-normocytic anemia, thrombocytosis, and eosinophilia, though the last of these is uncommon. Leukopenia is a finding in patients with Felty syndrome.

Nerve entrapment is common, as with the median nerve in carpal tunnel syndrome. Vasculitic lesions, mononeuritis multiplex, and cervical myelopathy may cause serious neurologic consequences. Peripheral myopathy may be noted as well.

Keratoconjunctivitis sicca is common in individuals with RA, and this condition is often the initial manifestation of secondary Sjögren syndrome. The eye may also have episcleritis, uveitis, and nodular scleritis that may lead to scleromalacia.

2010 ACR/EULAR Diagnostic Criteria

Historically, the diagnosis of RA was based on the 1987 American College of Rheumatology (ACR) criteria. These criteria were based on the persistence of arthritic symptoms over time; however, this classification system failed to identify patients with early inflammatory arthritis.

It is now recognized that early therapeutic intervention significantly improves clinical outcomes and reduces irreversible joint damage and disability. With this focus, the ACR and the European League Against Rheumatism (EULAR) devised new classification criteria for early arthritis, which assess joint involvement, autoantibody status, acute-phase response, and symptom duration, as well as revised criteria for classifying RA in newly presenting patients, those with erosive disease typical of RA, and those with inactive disease with or without treatment.[37]

Classification of RA

The 2010 ACR/EULAR classification criteria for RA are designed to identify patients with unexplained inflammatory arthritis in at least 1 peripheral joint and a short duration of symptoms who would benefit from early therapeutic intervention. They represents a paradigm shift from the 1987 ACR criteria, which lacked the sensitivity to detect early RA.[1]

According to the ACR/EULAR criteria, patients who should be tested are those (1) who have at least 1 joint with definite clinical synovitis and (2) whose synovitis is not better explained by another disease (eg, lupus, psoriatic arthritis, or gout).

The ACR/EULAR classification system is a score-based algorithm for RA that incorporates the following 4 factors:

The maximum number of points possible is 10. A classification of definitive RA requires a score of 6/10 or higher. Patients with a score lower than 6/10 should be reassessed over time. If patients already have erosive changes characteristic of RA, they meet the definition of RA, and application of this diagnostic algorithm is unnecessary.

Joint involvement consists of swelling or tenderness upon examination. The presence of synovitis may be confirmed on imaging studies. Points are allocated as follows:

At least 1 serology test result is needed for RA classification. Points are allocated as follows:

Negative serology test results are defined as international unit (IU) values that do not exceed the upper limit of normal (ULN) for the reporting laboratory or assay. Low-positive results are defined as IU values that exceed the ULN but are no more than 3 times the ULN for the reporting laboratory or assay. High-positive results are IU values that are more than 3 times higher than the ULN for the reporting laboratory or assay. When RF is available only as a positive or negative finding, a positive result should be scored as low-positive RF.

At least 1 test acute-phase reactant test result is needed for classification. Local laboratory standards determine which results are normal and which are abnormal. Points are allocated as follows:

Points for the patient’s self-reporting of the duration of signs or symptoms of synovitis in clinically involved joints are allocated as follows:

A 2014 systematic literature review by Radner et al concluded that the 2010 ACR/EULAR criteria have a pooled sensitivity for RA of 0.82 (95% confidence index [CI], 0.79-0.84) and a specificity of 0.61 (95% CI, 0.59-0.64). These authors' review of research that directly compared the 2010 ACR/EULAR criteria with 1987 ACR criteria found that the ACR/EULAR criteria had higher overall sensitivity (+0.11 compared with 1987 criteria) at the cost of lower overall specificity (-0.04).[38]

 

 

2012 ACR Disease Activity Measures

In April 2012, the ACR published new recommendations for the measurement of disease activity for clinical use at the point of care. The scales used included the following 6 recommended measures[2] :

The 6 measures above result in a single continuous index with defined ranges for low, moderate, or high disease activity or clinical remission.[2] In addition, the data collected from these measures are not solely dependent on the patient’s report, the clinician’s assessment, or laboratory measurements.

For more detailed information on how to use these tools and how to calculate the measurement tools, see the 2012 Rheumatoid arthritis disease activity measures: American College of Rheumatology Recommendations for use in clinical practice.

Measurement of disease progression

To determine the progression of RA, patients are categorized by clinical and radiologic criteria into 4 stages, as follows:

In the 2012 ACR-recommended RA disease activity measures, the following indicate disease activity cutoffs for patient-driven composite tools (all 3 scales, 0-10)[2] :

For the CDAI patient and provider composite tool (scale, 0-76), low/minimal disease is in the range of greater than 2.8 to 10.00; moderate disease, greater than 10.0 to 22.0; and high/severe disease, greater than 22.0.[2]

The following disease activity cutoffs are for the ACR-recommended RA patient, provider, and laboratory composite tools[2] :

Measurement of disease remission

In 2011, the ACR and the EULAR published a Boolean-based definition of remission in RA, according to which a patient must satisfy all of the following to be considered in remission[3] :

In addition, the ACR/EULAR index-based definition considers RA patients to be in remission if they have an SDAI score of 3.3 or lower. The SDAI score is defined as the sum of the tender joint count (28 joints), swollen joint count (28 joints), patient global assessment (0-10 scale), and CRP measurement (mg/dL).[3]

Given the cost and potential adverse effects of newer drugs used to treat RA, individualized treatment requires knowledge of poor prognostic factors. A systematic review of prospective studies identified 16 independent clinical, laboratory, and genetic factors predictive of RA remission. Although the predictive value of these factors remains to be confirmed, early aggressive treatment has been shown to improve outcomes regardless of poor prognostic factors.[39]

In the 2012 ACR-recommended RA disease activity measures, the following indicate disease remission cutoffs for patient-driven composite tools (all 3 scales, 0-10)[2] :

For the CDAI patient and provider composite tool (scale, 0-76), remission is indicated by a measurement of 2.8 or less.[2]

The following disease remission cutoffs are for the ACR-recommended RA patient, provider, and laboratory composite tools[2] :

Measurement of functional status

Patients with RA are categorized into 4 functional classes:

Complications

RA itself is not fatal, but complications of the disease may shorten survival by years in some individuals. In general, RA is progressive and cannot be cured, but in some patients, the disease gradually becomes less aggressive, and symptoms may even improve. However, if bone and ligament destruction and any deformities have occurred, the effects are permanent.

Joint disability and pain with daily life are common. Affected joints can become deformed, and the performance of even ordinary tasks may be very difficult or impossible; these factors can severely affect patients’ quality of life. In addition, RA is a systemic disease that can affect other parts of the body in addition to joints. These effects include the following:

Approach Considerations

No test results are pathognomonic for rheumatoid arthritis (RA); instead, the diagnosis is made by using a combination of clinical, laboratory, and imaging features. For clinical and radiologic criteria used in determining disease progression, see 2012 ACR Disease Activity Measures. Bone scanning findings may help distinguish inflammatory from noninflammatory changes in patients with minimal swelling, and densitometry findings are useful for helping diagnose changes in bone mineral density that are indicative of osteoporosis.

Laboratory Studies

Routine viral screening by serologic testing does not significantly facilitate the diagnosis of RA in patients with early RA, nor is it helpful as a potential identifier of disease progression.[40] Potentially useful laboratory studies in suspected RA fall into 3 categories—markers of inflammation, hematologic parameters, and immunologic parameters—and include the following:

Markers of inflammation

The ESR and the CRP level are associated with disease activity. The CRP value over time correlates with radiographic progression.

Hematologic parameters

The CBC commonly demonstrates anemia of chronic disease and correlates with disease activity; it improves with successful therapy. Hypochromic anemia suggests blood loss, commonly from the gastrointestinal (GI) tract (associated with the use of nonsteroidal anti-inflammatory drugs [NSAIDs]). Anemia may also be related to disease-modifying antirheumatic drug (DMARD) therapy.

Thrombocytosis is common and is also associated with disease activity. Thrombocytopenia may be a rare adverse event of therapy and may occur in patients with Felty syndrome. Leukocytosis may occur but is usually mild. Leukopenia may be a consequence of therapy or a component of Felty syndrome, which may then respond to DMARD therapy.

Immunologic parameters

Immunologic parameters include autoantibodies (eg, RF, anti-CCP antibodies, and ANAs). RF is an immunoglobulin (Ig) M antibody directed against the Fc fragment of IgG that is present in approximately 60-80% of patients with RA over the course of their disease (but in fewer than 40% of patients with early RA). RF values fluctuate somewhat with disease activity, though titers of RF generally remain high even in patients with drug-induced remissions.

RF is not specific for RA but is also present in other connective tissue diseases, infections, and autoimmune disorders, as well as in 1-5% of healthy people. The presence of RF predicts radiographic progression of bone erosions, independent of disease activity.[41]

Although ANAs are present in approximately 40% of patients with RA, test results for antibodies to most nuclear antigen subsets are negative.

Assays for anti-citrullinated protein antibody (ACPA; often tested as anti-CCP) are now being used clinically for diagnosing RA. ACPA-positive and ACPA-negative RA may be 2 distinct disease subsets, with different underlying pathogeneses and risks for developing RA.[42, 43] ACPA-positive patients may have a more erosive RA disease course than ACPA-negative patients.[44] However, a 2011 study suggests that reassessment of ACPA or IgM RF during the first year after onset of arthritis does not provide significant additional information.[45]

The sensitivity of anti-MCV assays has been reported to be comparable to that of ACPA[46] ; however, other studies have found the specificity of anti-MCV to be slightly lower than that of ACPA.[47, 48] Anti-MCV and anti-CCP levels may correlate with disease activity.[46, 49]

Studies of anti-CCP antibodies suggest a sensitivity and specificity equal to or better than those of RF, with an increased frequency of positive results in early RA; the presence of both anti-CCP antibodies and RF is highly specific for RA. Additionally, the presence of anti-CCP antibodies, like that of RF, indicates a worse prognosis.

A trial that tested for 4 novel RA biomarkers improved the diagnosis of early RA in patients who tested negative on conventional tests.[50, 51] In the study comprising 293 RA patients, 97 healthy control subjects, and 87 rheumatic control patients with other arthritides, the panel of 4 biomarkers—UH-RA.1, UH-RA.9, UH-RA.14 and UH-RA.21had 83% specificity for RA.[51] These markers were found in 37% of patients with early RA and 26% of those who were seronegative for rheumatoid factor and anticitrullinated protein antibody. The investigators suggested that in addition to an improved diagnosis in early RA, use of these biomarkers may also have prognostic potential.[51]

Pregnancy

RA often goes into remission during pregnancy. The presence of RF neither helps predict nor correlates with the outcome of arthritis during pregnancy. The ESR cannot be used to assess RA disease activity during pregnancy, because pregnancy alters the normal values. The volume expansion that occurs during pregnancy can result in lower hematocrit values.

Radiography

Radiography remains the first choice for imaging in RA; it is inexpensive, readily available, and easily reproducible, and it allows easy serial comparison for assessment of disease progression.[52] Views of the hands, wrists, knees, feet, elbows, shoulders, hips, cervical spine, and other joints should be assessed with radiography when indicated (see the images below). Erosions may be present in the feet, even in the absence of pain and in the absence of erosions in the hands.



View Image

Widespread osteopenia, carpal crowding (due to cartilage loss), and several erosions affecting the carpal bones and metacarpal heads in particular in ....



View Image

Lateral view of the cervical spine in a patient with rheumatoid arthritis shows erosion of the odontoid process.



View Image

Anteroposterior radiograph of the knee shows uniform joint-space loss in the medial and lateral knee compartments without osteophytosis. A Baker cyst ....

Magnetic Resonance Imaging

Magnetic resonance imaging (MRI) provides a more accurate assessment and earlier detection of lesions than radiography does[53] ; however, the cost of the examination and the small size of the joints involved militate against its widespread use. MRI is used primarily in patients with abnormalities of the cervical spine (see the images below); early recognition of erosions on the basis of MRI images has been sufficiently validated.



View Image

T1-weighted sagittal magnetic resonance image of the cervical spine shows basilar invagination with cranial migration of an eroded odontoid peg. There....



View Image

Sagittal T2-weighted magnetic resonance image of cervical spine in same patient as in previous image. Compromised foramen magnum is easily appreciated....

Ultrasonography

Ultrasonography of joints (see the image below) is gaining increased widespread acceptance in clinical practice; however, its use in RA is not yet the standard of care.[54, 55, 56, 57] Ultrasonography allows recognition of effusions in joints that are not easily accessible (eg, the hip and, in obese patients, the shoulder) and of cysts (Baker cysts). In addition, high-resolution sonograms allow visualization of tendon sheaths, changes and degree of vascularization of the synovial membrane, and even erosions. Ultrasonography can often be done in the office.



View Image

Ultrasonography-guided synovial biopsy of the second metacarpophalangeal joint of the right hand in a patient with rheumatoid arthritis of the hands. ....

Imaging of Specific Structures

Hand

Hand imaging in RA can include radiography, MRI, ultrasonography, and computed tomography (CT), though the last of these plays only a minimal role. Radiography is the mainstay of imaging RA in the hands: It is inexpensive and easily reproducible, and it allows easy serial comparison for assessment of disease progression. Its main disadvantage is the absence of specific radiographic findings in early disease; erosions may only be visualized later.

MRI is sensitive than radiography to early changes in RA, and in the appropriate clinical setting, it is more accurate than plain radiography in the diagnosis of the disease. However, a systematic literature review concluded that widespread use of MRI for the diagnosis of early RA and for helping determine the prognosis of early RA is not currently recommended, though MRI bone edema may be predictive of progression in certain RA populations.[58]

Ultrasonography has been applied to the assessment of RA with the goal of improving on the current standard of conventional radiography. Like MRI, ultrasonography serves as an early diagnostic tool and can help in evaluating the cause of joint swelling in a patient with RA.[59, 60] However, the results of one study suggested that NSAID use may mask the ultrasonographic gray scale and power Doppler signal in the assessment of synovitis in RA, resulting in lower scoring despite continuing disease activity.[61]

See Rheumatoid Arthritis Hand Imaging for complete information on this topic.

Spine

Spinal imaging in RA involves radiography, MRI, and CT. As with hand imaging in RA, the mainstay of imaging remains plain radiography. Only about 50% of patients with radiographic evidence of atlantoaxial subluxation are actually symptomatic.

The role of plain radiography is to establish whether the patient has risk factors for cord compression. The major role for CT and MRI is in preoperative assessment of the 2 main indications for surgical intervention—namely, neurologic deficit and severe pain.[62, 63] Although CT scanning can document bone damage and alignment abnormalities, especially with more detailed multiplanar reconstruction, MRI has become the preferred modality for evaluation of the spinal cord and neural elements.[64]

See Rheumatoid Arthritis Spine Imaging for complete information on this topic.

Joint Aspiration

Consider joint aspiration when making the definitive diagnosis of RA or when ruling out coexistent infection or crystal arthritis in an acutely swollen joint. New-onset monoarticular arthritis or an unusual pattern of a joint flare in a patient with RA should encourage strong consideration of joint aspiration and synovial fluid analysis.

Analysis of synovial fluid includes Gram staining, cell count, culture, and assessment of overall appearance. In patients with RA, analysis typically reveals inflammation (white blood cell [WBC] count >2000/µL, generally in the range of 5000-50,000/µL). Usually, neutrophil predominance (60-80%) is observed in the synovial fluid (in contrast with mononuclear cell predominance in the synovium). Because of a transport defect, glucose levels of synovial fluids (as well as pleural and pericardial fluids) in patients with RA are often low compared with serum glucose levels.

Histologic Findings

Early in the course of the RA disease process, there is an influx of inflammatory cells into the synovial membrane, with subsequent angiogenesis, proliferation of chronic inflammatory (mononuclear) cells and resident synovial cells, and marked histologic changes—a 2-cell-layer lining membrane changes to a thickened membrane that often has villous projections into the joint space (see the images below).



View Image

The hallmark of rheumatoid arthritis is a perivascular mononuclear cell infiltrate in the synovium (pictured here). The early stages are noted to have....



View Image

The inflammation involved in rheumatoid arthritis can be intense. It is composed of mononuclear cells and can resemble a pseudosarcoma.

The lymphoplasmacytic infiltration of the synovium with neovascularization seen in RA is similar to that seen in other conditions characterized by inflammatory synovitis. Early rheumatoid nodules are characterized by small-vessel vasculitis and later by granulomatous inflammation.

Approach Considerations

Optimal care of patients with rheumatoid arthritis (RA) consists of an integrated approach that includes both pharmacologic and nonpharmacologic therapies. Many nonpharmacologic treatments are available for this disease, including exercise, diet, massage, counseling, stress reduction, physical therapy, and surgery. Active participation of the patient and family in the design and implementation of the therapeutic program helps boost morale and ensure compliance, as does explaining the rationale for the therapies used.

Medication-based therapies comprise several classes of agents, including nonsteroidal anti-inflammatory drugs (NSAIDs), nonbiologic and biologic disease-modifying antirheumatic drugs (DMARDs), immunosuppressants, and corticosteroids. Early therapy with DMARDs has become the standard of care, because it can both retard disease progression more efficiently than later treatment and, potentially, induce more remissions.[65, 66, 67]

In pregnant patients with RA, no special obstetric monitoring is indicated beyond what is performed for usual obstetric care. However, some of the medications used in treating RA can have adverse effects on the fetus and may have to be discontinued several months before conception is planned.

Surgical procedures used in the treatment of RA include the following:

Pharmacologic Therapy

In 2008, the American College of Rheumatology (ACR) developed recommendations and algorithms for the use of nonbiologic and biologic DMARDs for patients with RA[68] ; an updated version was published in April 2012.[4]

Once a diagnosis is made, the main treatment goals are to control disease activity and slow the rate of joint damage, in addition to minimizing pain, stiffness, inflammation, and complications. Pharmacologic therapies that are used include nonbiologic and biologic DMARDs and adjunctive agents such as corticosteroids, NSAIDs, and analgesics.

A study by Callhoff et al found that biologic agents were significantly more effective than nonbiologic treatments in improving physical function in RA. In the investigation, a meta-analysis of 35 studies that included 8733 treated patients with RA and 4664 controls. More than 50% of patients treated with biologics experienced clinically relevant improvement. Etanercept and rituximab were the most effective treatments, both in patients who had never before taken antirheumatic drugs and in those who had shown an inadequate response to them.[84]

The ACR recommends that before patients undergo pharmacologic treatment with either nonbiologic or biologic DMARDS, they should receive not only the pneumococcal, hepatitis, and influenza vaccinations (as indicated in the 2008 ACR recommendations)[68] but also vaccinations for human papillomavirus (HPV) and herpes zoster virus (HZV) (added in the 2012 updated ACR recommendations).[4]

AHRQ recommendations

A 2007 clinician’s guide from the Agency for Healthcare Research and Quality (AHRQ) advised the following, with a medium level of confidence[6] :

Early DMARD therapy

DMARDs can be classified into nonbiologic and biologic agents. The nonbiologic DMARDs include the following:

The recognition of TNF-α and interleukin (IL)-1 as central proinflammatory cytokines has led to the development of biologic agents that block these cytokines or their effects. In addition to improving signs and symptoms and quality of life, all biologic agents significantly retard radiographic progression of joint erosions. Biologic DMARDs include agents such as adalimumab, certolizumab, etanercept, golimumab and infliximab.

DMARDs represent the most important measure in the successful treatment of RA. These agents can retard or prevent disease progression and, thus, joint destruction and subsequent loss of function. Successful DMARD therapy may eliminate the need for other anti-inflammatory or analgesic medications; however, until the full action of DMARDs takes effect, anti-inflammatory or analgesic medications may be required as bridging therapy to reduce pain and swelling.

Many studies have revealed that early treatment of RA (ie, within months of onset) with DMARDs not only can retard disease progression more efficiently than later treatment but also may induce more remissions.[65, 66, 67] Thus, early DMARD therapy (< 6 months after the onset of symptoms) has become the standard of care.[68] Patients with early forms of arthritis should be evaluated by and, if necessary, referred to physicians who are experienced in the diagnosis and treatment of RA.

Nonbiologic DMARDs

The results of a retrospective cohort study found that the use of HCQ may decrease the risk of diabetes in patients with RA. Further studies are needed to determine its preventive role in other patients at high risk for diabetes.[85] The results of a retrospective cohort study involving 121,280 patients found a lower adjusted risk of diabetes mellitus among individuals with RA or psoriasis who started a TNF inhibitor or HCQ than among those taking other nonbiologic DMARDs.[86]

In terms of frequency of remissions and time to onset of action, MTX and SSZ are the most active compounds and provide the best risk-benefit ratios. MTX, either alone or in combination with other agents, has become the standard of care for moderate to severe RA. Injectable gold salts and penicillamine rarely induce sustained remission and thus have largely been supplanted by more effective agents.

In October 2013, the US Food and Drug Administration (FDA) approved the first single-dose, self-administered, disposable MTX subcutaneous autoinjector (Otrexup).[87] Otrexup is indicated for adults with severe, active RA who have either responded inadequately to or cannot tolerate first-line therapy, as well as for children with active polyarticular juvenile idiopathic arthritis (JIA) (also known as juvenile rheumatoid arthritis).[87]

In a meta-analysis of 158 trials, triple therapy (MTX, SSZ, and HCQ) was found to have a statistically significant benefit of inhibiting joint damage compared with oral MTX alone.  Also, there was no statistically significant difference in preventing joint damage between triple therapy and MTX in combination with a biologic (adalimumab, certolizumab, etancercept, or inflximab) or the combination of MTX and tofacitinib.[88]

The FDA also approved Otrexup to control symptoms in adults with severe, recalcitrant, disabling psoriasis if the disease has not adequately responded to other treatments.[87]

Minocycline may act as a DMARD through its action as a matrix metalloproteinase inhibitor (MMPI). Leflunomide is the most recent addition to the nonbiologic DMARDs and has activity similar to that of SSZ and MTX. Most of these drugs have been shown to improve signs and symptoms (as well as quality of life) and to significantly retard radiographic progression of RA.

Biologic DMARDs: TNF inhibitors

The TNF inhibitors that bind TNF and thus prevent its interaction with its receptors include the following:

Precautions and adverse effects

Biologic agents are expensive. Consensus statements do not recommend their use until at least one nonbiologic DMARD, usually MTX, has been administered without sufficient success. In clinical trials, as many as 70% of patients achieve significant responses, but remissions are not usually observed.[89]

Adverse effects associated with the biologic agents include the generation of antibodies against these compounds, emergence of antinuclear antibodies (ANAs), occasional drug-induced lupuslike syndromes, and infections (including tuberculosis). Rarely, demyelinating disorders and bone marrow suppression occur. Immunogenicity, such as the development of anti-drug antibodies, has been shown to occur in adalimumab and infliximab, potentially leading to decreased drug efficacy. Concomitant use of MTX may reduce the frequency of anti-drug antibodies.[90]

Acute and chronic infections, demyelinating disorders, class III or IV heart failure, and recent malignancies are contraindications to the use of TNF inhibitors. Thoroughly searching for latent tuberculosis using purified protein derivative (PPD) testing or an interferon (IFN) gamma release assay (IGRA), with or without chest radiography, is recommended before these agents are started.

Patients taking anti-TNF agents must avoid live-virus vaccines. Giving live vaccines to patients receiving immunosuppressive drugs leads to a higher risk for serious infection.

The results of one study noted that the use of anti-TNF therapy may double the risk of septic arthritis in patients with RA, with the risk being highest in the early months of therapy. Although not significantly influenced by anti-TNF therapy, previous joint replacement surgery was also noted as a risk factor for septic arthritis.[91]

Hepatitis B virus (HBV) reactivation can occur in both hepatitis B surface antigen (HBsAg)–positive and HBsAg-negative/hepatitis B core antibody (anti-HBc)-positive patients with detectable occult HBV infection during anti−TNF-α therapy. Antiviral prophylaxis may effectively reduce this reactivation.[92]

In three Swedish registries that analyzed the cancer risk in 6366 RA patients taking TNF inhibitors, no increased risk of cancer with these agents was observed.[93] These patients were compared with 61,160 biologics-naive RA patients, 5989 patients starting MTX, 1838 patients starting DMARD combination therapy, and the general Swedish population. Of patients taking TNF inhibitors, 240 developed first-time cancer, yielding a relative risk (RR) of 1.00 compared with the biologics-naive cohort. Similar RRs were shown with the other cohorts.

TNF inhibitors vs MTX

In one study, bone erosions showed a higher rate of repair in RA patients treated with TNF inhibitors than in patients treated with MTX.[94] After a 1-year follow-up, the group treated with TNF blockers showed a mean width of 2 mm and a mean depth of 2.3 mm; the MTX-treated group showed a mean width of 2.4 mm and a mean depth of 2.4 mm. Deeper lesions in the TNF-inhibitor group were also particularly prone to repair when compared with more shallow lesions.[94]

Van Vollenhoven et al reported that in patients with early RA who have MTX-treatment failure, the addition of a TNF antagonist was superior to the addition of conventional DMARDs.[95] In this study, 258 patients with early RA who did not achieve low disease activity after 3-4 months of MTX (up to 20 mg/wk) were randomized to receive additional treatment (in addition to MTX) either with SSZ and HCQ or with infliximab.

In the SSZ and HCQ group, 32 of 130 (25%) achieved the primary outcome, defined as a good response according to the European League Against Rheumatism (EULAR); in the infliximab group, 50 of 128 (39%) attained the primary outcome.[95]

A systematic review by Visser et al suggested that an initial oral MTX dose of 15 mg/wk, with escalation of 5 mg/mo to achieve target doses of 25-30 mg/wk or maximum tolerable doses, was the optimal evidence-based dosing strategy.[96] Starting at higher initial doses or escalating too rapidly may be limited by toxicity. Conversion from oral to subcutaneous (SC) administration of MTX is suggested for patients who have an inadequate response to oral therapy.

Adalimumab

A 5-year analysis of an open-label extension (OLE) study concluded that a delay of 52 weeks for adding adalimumab to concomitant MTX therapy contributed to inferior radiographic, functional, and clinical outcomes in patients with active RA.[97]

According to data from a study of 221 consecutive RA patients, adalimumab blood levels of 5 to 8 μg/mL have the greatest effect on disease activity. In the study, adalimumab trough levels greater than 8 μg/mL had no additional beneficial effect on disease activity.[98, 99] Study participants were treated with 40 mg adalimumab subcutaneously every other week for 28 weeks and stratified according to whether or not they were taking concomitant MTX. Patients treated with concomitant MTX reached recommended blood levels at lower adalimumab doses. At 28-week follow-up, mean adalimumab levels were 4.1 μg/mL in patients receiving monotherapy and 7.4 μg/mL in patients receiving concomitant MTX. MTX might contribute to increasing adalimumab blood levels by reducing inflammation and lowering the number of targets for adalimumab to bind to.[98]

Certolizumab

Fleischmann et al found that monotherapy with certolizumab effectively reduced the signs and symptoms of active RA in patients in whom DMARD therapy had failed.[100] In this study, 200 patients were randomized on a 1:1 basis to receive certolizumab 400 mg or placebo every 4 weeks for 24 weeks. At 24 weeks, 45.5% of the certolizumab group achieved a 20% improvement, according to ACR criteria, compared with 9.3% of the placebo group. Statistically significant differences were observed as early as week 1 through week 24.[100]

A study by Smolen et al found that certolizumab plus MTX was more efficacious than placebo plus MTX, rapidly and significantly improving signs and symptoms of RA and physical function and inhibiting radiographic progression.[101] In this study, 619 patients were randomized to receive certolizumab 400 mg at weeks 0, 2, and 4, followed by 200 mg or 400 mg plus MTX every 2 weeks or placebo plus MTX every 2 weeks.

Significantly more patients who received certolizumab 200 mg (57.3%) or 400 mg (57.6%) achieved 20% improvement by ACR criteria than did those who received placebo (8.7%).[101] Radiographic progression was significantly inhibited with certolizumab 200 mg or 400 mg, as compared with placebo. When compared with placebo plus MTX, certolizumab plus MTX significantly relieved signs and symptoms, improved physical function, and inhibited radiographic progression in patients with RA.[101]

Golimumab

Golimumab is a human anti−TNF-α monoclonal antibody that inhibits TNF-α bioactivity, thereby modulating immune activity in patients with RA. Using a modified intention-to-treat analysis, researchers demonstrated that golimumab plus MTX is more efficacious than MTX alone (and that golimumab alone is about as efficacious as MTX alone) in reducing disease signs and symptoms in MTX-naive patients.[102]

In this 52-week, randomized, double-blind, placebo-controlled study, which was was followed by an open-label extension through 5 years, 637 patients were randomized to receive placebo plus MTX (group 1), golimumab 100 mg SC plus placebo (group 2), golimumab 50 mg SC plus MTX (group 3), or golimumab 100 mg SC plus MTX (group 4).[102] Intent-to-treat analysis showed no significant differences in the primary endpoint between group 1 and groups 3 and 4 combined, indicating efficacy of subcutaneous golimumab. The incidence of serious adverse events was similar across all groups.

In July 2013, the FDA approved golimumab IV.[103, 104, 105] Approval was supported by a phase 3 study of 592 patients with moderately to severely active RA who had been receiving background MTX for at least 3 months.

In this study, 58.5% (n = 231/395) of patients receiving treatment with golimumab IV plus MTX experienced significant improvements in signs and symptoms at week 14 compared with 24.9% of patients receiving placebo plus MTX (n = 49/197).[105] Improvement was demonstrated by at least a 20% increase in ACR 20 score, the study’s primary endpoint. A higher proportion of patients receiving golimumab plus methotrexate achieved at least a 50% improvement in ACR criteria (ACR 50) at week 14 (30%) compared with patients receiving placebo plus MTX (9%).

The rate of adverse events and serious adverse events, respectively, at week 24 were 53% and 4% in the golimumab group and 49% and 2% in the placebo group.[103, 104] The most common adverse events were "infections and infestations," including upper respiratory tract infection (>5% of patients), urinary tract infection, and nasopharyngitis. Exacerbation of RA occurred in 5.6% of patients receiving placebo plus MTX.[103]

At week 52, the rate of adverse events and serious adverse events in the golimumab group were 65% and 9%, respectively.[103] No serious opportunistic infections occurred through week 52. However, in the golimumab group, a single case of tuberculosis was reported, and a patient died from a myocardial infarction secondary to community-acquired pneumonia.[103]

Biologic DMARDs: non-TNF agents

Rituximab

Rituximab is most often used in combination with MTX. It has been shown to be effective for reducing signs and symptoms in adult patients with moderately to severely active RA who have had an inadequate response to therapy with one or more TNF inhibitors.[106, 107, 108] The ORBIT study in 295 biological-treatment naive patients with RA found that initial treatment with rituximab is noninferior to initial TNF inhibitor treatment, and is cost saving over 12 months.[109]

Treatment with rituximab may deplete CD20+ B cells. A study by Bingham et al suggested that polysaccharide and primary immunizations should be administered before rituximab infusions in order to maximize treatment responses.[110] The study showed that responses to pneumococcal polysaccharide vaccine were less in RA patients receiving rituximab and MTX (57% showed a 2-fold rise in titer in response to 1 or more serotypes) than in those receiving MTX alone (82%).

Decreased response to keyhole limpet hemocyanin (KLH) (neoantigen) was also seen in the rituximab-treated patients (47% vs 93%).[110] However, the ability to maintain a positive delayed-type hypersensitivity to a Candida albicans skin test was comparable in both treatment groups, as was response to tetanus toxoid.

Anakinra

Anakinra is a recombinant nonglycosylated form of the human IL-1 receptor antagonist (IL-1ra). IL-1ra occupies the IL-1 receptor without triggering it and prevents receptor binding of IL-1. In clinical trials, a significant response was observed in approximately 40% of patients with RA.

Abatacept

Abatacept is a selective costimulation modulator that inhibits T-cell activation by binding to CD80 and CD86, thereby blocking their interaction with CD28. CD28 interaction provides a signal needed for the full T-cell activation that is implicated in RA pathogenesis.

Maintenance doses of abatacept may be administered as a monthly intravenous (IV) infusion or by the patient as a weekly SC injection.[111] In patients with RA who have previously had treatment failure with anti-TNF therapy, abatacept has been shown to demonstrate consistent safety and efficacy that are maintained from 6 months to 5 years of therapy.[112]

A head-to-head phase IIIb randomized noninferiority trial found that subcutaneous (SC) abatacept and SC adalimumab were equally effective in RA patients, with comparable safety (though adalimumab was associated with more injection-site reactions).[113] Adding either treatment to background methotrexate produced similar American College of Rheumatology 20% improvement response (ACR20) rates and similar rates of radiographic nonprogression. In view of these findings, clinicians may reasonably conclude that the 2 agents are substantially equivalent for treating RA.

Tocilizumab

Tocilizumab, an IL-6 receptor inhibitor, is available as either an IV infusion or SC injection. It is indicated for moderate-to-severe active RA in adults who have had an inadequate response to 1 or more TNF-antagonist therapies. It may be used either alone or in combination with MTX or other DMARDs. However, Dougados et al found that in patients with active RA, combination therapy with IV tocilizumab and MTX did not yield better clinical results than tocilizumab monotherapy and that was more often associated with transaminase increases.[114]

In patients with inadequate response to TNF inhibitors, tocilizumab treatment results in significant, clinically meaningful, rapid, and sustained improvements in a number of patient-reported outcomes.[115, 116] A 2012 consensus statement confirmed the efficacy and safety of IL-6 pathway blockade in adult rheumatoid arthritis.[117]

In October 2013, the FDA approved an SC injection of tocilizumab that can be self-administered after proper training. The SC formulation has been shown to be as effective as the IV infusion and has the same safety profile except for increased injection site reactions with SC administration.[116]

Sarilumab

Also an IL-6 inhibitor, sarilumab is a human monoclonal antibody that binds to both soluble and membrane-bound IL-6 receptors (sIL-6R and mIL-6R). It is approved for adults with RA who have had an inadequate response or intolerance to one or more DMARDs. Administration is by SC injection every 2 weeks. Sarilumab may be prescribed as monotherapy or with MTX or other conventional DMARDs.

Approval of sarilumab was based on the MOBILITY and TARGET trials. In the MOBILITY study, patients treated with sarilumab plus MTX had reduced signs and symptoms and improved physical function, and demonstrated significantly less radiographic progression of structural damage, compared with placebo plus MTX. At 24 weeks of treatment, the ACR20 score was 66% for sarilumab 200 mg, 58% for sarilumab 150 mg, and 33% for placebo.[118]

The TARGET study combined sarilumab with a DMARD. Results showed reduced signs and symptoms and improved physical function compared with placebo plus DMARD. Results were similar to the MOBILITY trial. At 24 weeks, ACR20 was 61% for sarilumab 200 mg, 56% for sarilumab 150 mg, and 34% for placebo.[119]

Tofacitinib

Janus kinases (JAKs) consist of a group of intracellular tyrosine kinases that transmit signals from cytokine or growth factor–receptor interactions on the cellular membrane to influence cellular processes of hematopoiesis and immune cell function. Within the signaling pathway, JAKs phosphorylate and activate signal transducers and activators of transcription (STATs), which modulate intracellular activity, including gene expression.

JAK inhibitors modulate the signaling pathway at the point of JAKs, preventing the phosphorylation and activation of STATs. These signals maintain the inflammatory condition in RA. Inhibition of JAKs reduces production of and modulates proinflammatory cytokines central to RA.

Tofacitinib is an oral JAK inhibitor[120] that was approved by the FDA in November 2012 as second-line treatment for moderate to severe active RA.[121] The indication is specific for patients who have had an inadequate response to or are intolerant of MTX. Tofacitinib may be given as monotherapy or in combination with MTX or other nonbiologic DMARDs. It should not be used in combination with biologic DMARDs or potent immunosuppressive agents (eg, azathioprine or cyclosporine).

Tofacitinib has been associated with reductions in signs and symptoms of RA and improvement in physical function.[122] Fleischmann et al demonstrated that ACR criteria for a 20% response were met in 59.8% of patients receiving monotherapy with tofacitinib 5 mg twice daily, compared with 26.7% of patients receiving placebo.[123] Health Assessment Questionnaire Disability Index (HAQ-DI) reduction was also greater in the tofacitinib group (−0.50 points) than in the placebo group (−0.19 points).

In another study, in which 717 patients who received stable MTX doses over 12 months were randomized to also receive 5 mg or 10 mg of tofacitinib orally twice daily, adalimumab 40 mg every 2 weeks, or placebo, ACR 20% response rates at 6 months were higher among patients receiving 5 mg or 10 mg of tofacitinib (51.5% and 52.6%, respectively) and among those receiving adalimumab (47.2%) than among those receiving placebo (28.3%).[124]

A once-daily 11-mg extended-release dosage form was approved by the FDA in February 2016 as an alternative to the 5-mg twice-daily regimen.

Baricitinib

The FDA approved a second JAK inhibitor, baricitinib (Olumiant), in June 2018 as a second-line treatment of moderately to severely active RA in adults who have had an inadequate response to one or more TNF antagonists. Baricitinib may be used as monotherapy or in combination with methotrexate or other nonbiologic DMARDs. It should not be used in combination with biologic DMARDs or potent immunosuppressive agents (eg, azathioprine or cyclosporine).

The dosage of baricitinib is one 2-mg tablet daily. The FDA declined to approve 4-mg tablets, citing safety concerns. The drug's prescribing information will contain a boxed warning about the risk for serious infections, malignancies, and thrombosis.[125]

Approval of baricitinib was supported by the RA-BUILD, RA-BEYOND, and RA-BEACON phase III clinical trials. The RA-BUILD study was a double-blind 24-week study that included 684 biologic DMARD–naïve patients with RA and inadequate response or intolerance to 1 or more conventional synthetic DMARDs. At week 12, 62% of patients taking baricitinib 4 mg achieved ACR20 response, compared with 39% of those taking placebo (P ≤0.001).[126]

In the RA-BEYOND study,  radiographic progression at 24 and 48 weeks was statistically significantly lower for either baricitinib 2 or 4 mg compared with placebo. However, only baricitinib 4 mg demonstrated statistically significant inhibition of progressive radiographic joint damage compared with patients initially randomized to placebo when observed at week 48.[127]

In the RA-BEACON study (n=527), patients with RA received baricitinib 2 mg, baricitinib 4 mg, or placebo in addition to conventional DMARDs they were currently taking. Patients in the study had demonstrated an inadequate response or intolerance to at least one TNF inhibitor therapy. Study participants could have undergone prior therapy with other biological DMARDs. At week 12, patients taking baricitinib had significantly higher ACR20 response rates (49%, versus 27% for placebo) and gains in all individual ACR20 component scores.[128]

Upadacitinib 

Another JAK inhibitor, upadacitinib (Xenleta), was approved in August 2019 for moderately to severely active RA in adults who have had an inadequate response or are intolerant to methotrexate. It may be used as monotherapy or in combination with methotrexate or other nonbiological DMARDs.

Approval was based on data from the SELECT phase 3 RA program that enrolled over 4000 patients with moderate to severe RA across 5 studies. In the SELECT-MONOTHERAPY study, 598 or 648 patients completed the study. At week 14, an ACR20 response was achieved by 89 (41%) of 216 patients in the continued methotrexate group, 147 (68%) of 217 patients receiving upadacitinib 15 mg, and 153 (71%) of 215 patients receiving upadacitinib 30 mg (p < 0.0001 for both doses vs continued methotrexate).[156]

The SELECT-COMPARE study evaluated safety and effectiveness of upadacitinib compared with adalimumab (each with methotrexate) over 48 weeks. Results showed low disease activity, clinical remission, and improvements in pain and function remained superior for upadacitinib compared with adalimumab at 26 weeks and also from weeks 26 to 48.[157]

Combination DMARD therapy

In clinical trials, 30-70% of patients using DMARDs, either as monotherapy or in combination therapy, achieve partial responses, as determined by the ACR disease activity score. Currently, it is not possible to predict which patients will not have a treatment response.

In clinical practice, three strategies are employed to reduce disease activity as much as possible in patients whose disease does not respond or in those with clinical responses that are regarded as insufficient:

Because patients may require 2-3 months to achieve a full response to DMARDs, decisions regarding changes in medication are often delayed until that time.

Combination therapy appears to be helpful in patients whose disease partly or completely fails to respond to DMARD monotherapy. One study found that in patients with early, active RA, combination DMARD therapy with downward titration or intensive triple-DMARD combination therapy is more cost-effective than DMARD monotherapy.[129] Several combinations have proved successful without posing unexpected added risks; most include MTX (eg, MTX plus SSZ plus HCQ, MTX plus leflunomide, or MTX plus biologic DMARDs).

MTX combined with infliximab[130] or rituximab[131] yields a better response than monotherapy does. MTX combined with etanercept provides a higher rate of meaningful clinical response. MTX combined with cyclosporine, though not a commonly used combination, results in greater clinical improvement than MTX alone. Triple therapy with MTX, SSZ, and HCQ may provide substantially greater clinical improvement than either MTX alone or SSZ plus HCQ.[132]

The toxicities of these drug combinations are rarely more significant than those occurring with any of the individual agents used alone, though liver and bone marrow toxicity may be increased if MTX and leflunomide are combined.

Adverse effects

When used with appropriate clinical and laboratory control monitoring, combination therapy with the above agents is usually well tolerated. Adverse events typically become rarer after the first 2-3 months of therapy. Most adverse events are reversible with cessation of the drugs or with reduction of the doses.

The most important and most common adverse events relate to liver and bone marrow toxicity (MTX, SSZ, leflunomide, azathioprine, gold compounds, and D-penicillamine), renal toxicity (cyclosporine, parenteral gold salts, and D-penicillamine), pneumonitis (MTX), allergic skin reactions (gold compounds and SSZ), autoimmunity (D-penicillamine, SSZ, and minocycline), and infections (azathioprine and cyclosporine).

Antimalarial agents may cause ocular toxicity. In addition to an assessment of renal and liver function, it is recommended that baseline ophthalmologic screening for patients using these agents should include an evaluation for visual impairment and a recording of near visual acuity.[133] In 2011, the American Academy of Ophthalmology issued Revised Recommendations on Screening for Chloroquine and Hydroxychloroquine Retinopathy.

Complications of DMARD treatment

Patients with an established diagnosis of RA who are being treated with DMARDs, particularly those treated with combination therapy, including biologic agents such as TNF antagonists, may present with serious infections, malignancies, or both.[134, 135, 136] Additionally, adverse events from RA medications may include liver toxicity, renal toxicity, bone marrow depression, lung inflammation, and skin manifestations.

TNF precautions and mortality

Patients taking anti-TNF agents must avoid live-virus vaccines (eg, measles-mumps-rubella [MMR], HZV, varicella-zoster virus [VZV], and bacillus Calmette-Guérin [BCG] vaccines) to avoid the potential for serious infection.

A large national prospective cohort study over a mean of 4 years demonstrated that anti-TNF therapy for RA was not associated with a significant increase or decrease in mortality when compared with standard nonbiologic DMARD therapy.[137] The results from another study confirmed that the risk of serious infection and malignancy is not increased in patients receiving anti-TNF therapy when the patients have early RA and have not been previously treated with MTX or other DMARDs.[138]

In a systematic review and meta-analysis reporting on the risk of malignancy in patients with RA treated with TNF inhibitors, the data reviewed showed that these agents did not increase the risk of malignancy, particularly lymphoma; however, they did appear to increase the risk of skin cancer, including melanoma.[139]

Corticosteroids

Corticosteroids are potent anti-inflammatory drugs that are commonly used in patients with RA to bridge the time until treatment with DMARDs is effective.[5, 140] These agents are effective adjuncts to DMARD or NSAID therapy. Timely dose reductions and cessation are important because of the adverse effects associated with long-term steroid use. Corticosteroids can be administered by oral, IV, or intra-articular routes.

When Buttgereit et al studied circadian rhythms in 288 patients with active RA, half of whom were randomly assigned to a modified-release (MR) prednisone tablet and the other half to an immediate-release (IR) prednisone tablet, there was a clinically relevant reduction of morning stiffness of the joints with the MR product as compared with the IR product.[141]

A 9-month extension of the same study showed that the MR prednisone taken at bedtime was well tolerated and provided a sustained improvement.[142] A third study that added low-dose MR prednisone to existing DMARD treatment also showed improvements in RA signs and symptoms, including a reduction in morning stiffness as compared with baseline (35% vs 55%).[143]

A comparison of high-dose IV steroids with infliximab in the 18-month randomized, double-blind IDEA study found similar rates of remission induction and sustained remission, as well as the time to sustained remission, with the two agents.[144, 145] The study, which included 112 patients with new-onset DMARD-naïve RA, compared the efficacy of infliximab and IV steroid therapy, both in combination with methotrexate, as remission induction in early RA, followed by a treat-to-target approach.

Adverse effects

One study found that the use of corticosteroids was associated with heart failure in patients with RA, independent of cardiovascular risk factors and coronary heart disease (CHD). Those patients who currently used MTX showed a lower risk of heart failure.[146]

Nonsteroidal anti-inflammatory drugs

NSAIDs interfere with prostaglandin synthesis through inhibition of the enzyme cyclooxygenase (COX), thus reducing swelling and pain. However, they do not retard joint destruction and thus are not sufficient to treat RA when used alone. Like corticosteroids, NSAIDs can be reduced in dose or discontinued with successful DMARD therapy.

The several dozen NSAIDs are available can be classified into several different groups of compounds. Commonly used NSAIDs include ibuprofen, naproxen, ketoprofen, piroxicam, and diclofenac.

In the early 1990s, a second isoform of COX was discovered (COX-2). COX-1 has a protective role, particularly in the stomach, whereas COX-2 is strongly upregulated during inflammation. Traditional NSAIDs are nonselective COX inhibitors, inhibiting both COX-1 and COX-2. Several coxibs (selective COX-2 inhibitors) were developed that had a significant preference for COX-2 over COX-1. Currently, however, only one COX-2 inhibitor remains on the US market—namely, celecoxib.

Adverse effects

Coxibs, with their selectivity for COX-2, have been shown to be clinically efficacious and are accompanied by reduced gastrointestinal (GI) toxicity, the major adverse event related to the use of nonselective COX inhibitors (ie, NSAIDs). Other adverse effects, such as water retention, hypertension, and abnormal transaminase levels, are observed with both nonselective COX inhibitors and selective COX-2 inhibitors.

Whether and to what degree nonaspirin NSAIDs, coxibs, or both have increased cardiovascular toxicity has not been definitively settled. An analysis of 6 placebo-controlled trials comparing celecoxib with placebo found that the risk of cardiovascular death, myocardial infarction, stroke, heart failure, or thromboembolic events increased after celecoxib treatment in a dose-dependent fashion.[147]

Analgesics

Acetaminophen, tramadol, codeine, opiates, and various other analgesic medications can also be used to reduce pain. These agents do not affect swelling or joint destruction.

Experimental therapies

Despite significant advances over the past decades, RA continues to be a chronic disease. It remains active in many patients whose conditions partially or completely fail to respond to DMARDs. Therefore, the vigorous search for new therapeutic agents continues.

Several new CD20 B-cell−targeted biologic agents are under investigation, including atacicept, AMG 623, B3-FCc, Br3-Fc, belimumab, epratuzumab, ofatumumab, ocrelizumab, and TRU-015. Small molecules directed at enzymes involved in signal transduction of TNF and other proinflammatory cytokines are effective in treating RA.

A phase II study reported that in comparison with placebo, fostamatinib, an inhibitor of spleen tyrosine kinase (Syk), reduced disease activity in RA patients who did not have a response to MTX therapy.[148] Further investigation is required to determine the safety and efficacy of this agent in RA patients.

Inhibition of matrix metalloproteinases (MMPs), though initially unsuccessful, could prove to be efficacious, as could inhibition of osteoclast activation. Apheresis procedures are also being investigated. High-dose immunosuppression combined with autologous stem cell transplantation has been used in study protocols for patients whose conditions are resistant to other therapies.

Recommendations for Use of DMARDS and Biologic Agents

2016 EULAR Recommendations

Guidelines from the European League Against Rheumatism (EULAR) are designed to prevent the overtreatment of 20% to 30% of patients with rheumatoid arthritis (RA). Based on 3 systematic reviews, the 2016 update of the EULAR RA management recommendations includes 12 recommendations. Highlights include the following[5] :

2015 ACR Recommendations

This section summarizes the 2015 ACR recommendations for the use of nonbiologic and biologic DMARDs in the treatment of RA (see Table below).[4, 68] The ACR recommends that low disease activity or remission be the target for all patients with early or established disease who are receiving a DMARD or a biologic agent.

 

Table 1. 2015 American College of Rheumatology Guideline for the Treatment of Rheumatoid Arthritis



View Table

See Table

Use of biologic agents in patients with hepatitis, malignancy, or CHF

Hepatitis

Patients with RA requiring treatment who are infected with hepatitis C should not be treated differently than RA patients not infected with hepatitis C.[4] In RA patients with untreated chronic hepatitis B virus (HBV) infection, as well as those with treated chronic HBV disease of Child-Pugh class B and higher, biologic agents are not recommended. In patients with HBV infection who are receiving or have received antiviral therapy, treatment recommendations are the same as in patients without HBV infection.[4]

Malignancies

Any biologic agent may be started or resumed in patients with RA who were treated more than 5 years ago for either solid malignancies or nonmelanoma skin cancer, if such patients would otherwise receive this RA treatment.[4]

Recommendations for RA patients with a previous history of treated or untreated malignancy are as follows[4] :

Chronic heart failure

Anti−tumor necrosis factor (TNF) biologic agents are not recommended in RA patients with New York Heart Association (NYHA) congestive heart failure (CHF) class III (cardiac disease causing marked limitation of physical activity; less than ordinary physical activity causing fatigue, palpitation, dyspnea, or anginal pain; asymptomatic at rest) or IV (cardiac disease causing inability to carry on any physical activity without discomfort; presence of symptoms of cardiac insufficiency or angina syndrome at rest, which increase with physical activity) who have an ejection fraction of 50% or less. ACR recommends combination DMARDs or non-TNF biologic agents or tofacinitib over anti-TNF agents.

Screening for tuberculosis

In patients with RA who are under consideration for beginning or receiving treatment with biologic agents, screening for latent tuberculosis (TB) infection (LTBI) is recommended, regardless of whether these individuals have risk factors for LTBI. Nevertheless, clinicians should review patients’ medical histories to identify the following LTBI risk factors, as defined by the Centers for Disease Control and Prevention (CDC)[4] :

TB screening tests

Regardless of the risk factors for LTBI, the initial screening test for TB in patients with RA who are starting therapy with biologic agents should be the tuberculin skin test (TST) or interferon (IFN) gamma release assays (IGRAs). IGRAs are the preferred screening test in patients with a history of a bacillus Calmette-Guérin (BCG) vaccination; TST results in these patients have a high rate of false-positives.

ACR recommendations for further evaluation on the basis of the patient’s initial or repeat screening results are summarized in Table 2 below.

Table 2. 2015 ACR Recommendations for Further Evaluation After Initial/Repeat TB Screening Results



View Table

See Table

Vaccinations in patients beginning or receiving therapy with DMARDs or biologic agents

Before RA patients start or while they are on nonbiologic DMARD monotherapy (hydroxychloroquine [HCQ], leflunomide, MTX, minocycline, or SSZ), DMARD combination therapy (double [mostly MTX-based] or triple [HCQ, MTX, and SSZ]), or anti-TNF (adalimumab, certolizumab, etanercept, golimumab, or infliximab) or non-TNF (abatacept, rituximab, or tocilizumab) biologic agents, administer all killed vaccines (pneumococcal, intramuscular influenza, and HBV) and human papillomavirus (HPV) recombinant vaccine.[4]

The herpes zoster virus (HZV) live attenuated vaccine should only be administered before RA patients start DMARD monotherapy, combination DMARDs, and anti-TNF or non-TNF biologic agents, as well as in those already receiving DMARD monotherapy or combination DMARDs. This vaccine is not recommended for RA patients already on anti-TNF or non-TNF biologic agents.[4] The ACR recommends consulting the vaccine instructions and the CDC dosing and timing recommendations.

Considerations for Specific Patient Presentations

Acute presentations in patients with RA are generally because of an exacerbation of known disease or manifestations in other organ systems or other disease sequelae. Patients presenting with an initial onset of previously undiagnosed possible RA require symptomatic treatment with NSAIDs and rapid referral for definitive diagnosis and institution of DMARD therapy. A delay of as little as 2-3 months in initiating joint-sparing therapy results in significant irreversible joint damage measured radiographically at 5 years.

In patients with known disease, increased pain, edema, and dysfunction are characteristics of rheumatoid flare (exacerbation). Flares may be local or systemic in nature. Laboratory evaluation may reveal elevation in acute-phase reactants. Treatment consists of rest, NSAIDs, DMARDs, short courses of steroids (2-4 weeks), and, possibly, intra-articular steroid injections. Pain relief is important and may necessitate short-term use of narcotic analgesics.

Felty syndrome

Felty syndrome is a triad of RA, neutropenia, and splenomegaly. Patients with Felty syndrome are prone to serious bacterial infections that result in higher morbidity and mortality than those reported for other patients with RA. Prompt diagnosis and initiation of antibiotic therapy are required.

Baker cysts

Ruptured Baker cysts are often confused with deep vein thrombosis (DVT). Baker cysts often occur fairly early in the course of the disease, with pain, edema, and inflammation in the posterior knee and calf. The diagnosis is best made with ultrasonography. Treatment includes rest, elevation, needle puncture of the calf, knee joint aspiration, and referral.

Carpal tunnel syndrome

Carpal tunnel syndrome (median nerve compression neuropathy) is evinced by pain or paresthesias in the median nerve distribution of the hand, a positive Phalen or positive Tinel test, or positive findings on electromyography (EMG). Therapy includes rest, temporary immobilization, NSAIDs, and surgery.

Pregnancy

Pregnancy alters the immune state, possibly contributing to a change in the course of RA.[69, 70] For decades, the ameliorating effects of pregnancy on disease activity in women with RA have been observed. No specific guidelines address obstetric monitoring in patients with RA.

Because limited available data suggest a significant risk of preterm birth, preeclampsia, or fetal growth restriction in pregnant patients with RA, no special obstetric monitoring is indicated beyond what is performed for usual obstetric care (eg, stabilizing the disease before conception and using drugs safe for pregnancy and lactation).[71]

Medications considered low-risk in pregnancy include immunomodulating drugs, low-dose corticosteroids, antimalarial agents, SSZ, and azathioprine. Fetal toxicity is rarely associated with HCQ at the dosage used for RA and connective tissue disease (6.5 mg/kg body weight).[72] Anakinra may be used until conception.

Before therapy is initiated, patients should be counseled about the teratogenicity and adverse effects of the medications used to treat RA. NSAIDs should be avoided in the third trimester. MTX is contraindicated in pregnancy, because it is an abortifacient and has teratogenic effects, including craniofacial abnormalities, limb defects, and central nervous system (CNS) defects such as anencephaly, hydrocephaly, and meningomyelopathy, especially with first-trimester exposure.[73] Leflunomide is also contraindicated. Both MTX and leflunomide should be discontinued at least 3 months before pregnancy, and blood levels of leflunomide should be assessed.

Use of anti-TNF drugs during pregnancy is not covered by current guidelines; the ACR guidelines recommend this topic as a target for future research.[4] Stopping TNF inhibitors at conception has been recommended.[71] However, TNF inhibitors do not cross the placenta until the end of the second trimester. Thus, concern over possible effects on the development of the fetal immune system has led to the suggestion, in the inflammatory bowel disease literature, that anti-TNF drugs can continue from conception through the first trimester but should be stopped during the second trimester.[74]

Analysis of data from the health registries in Denmark and Sweden determined that women who received anti-TNF agents during pregnancy had a slightly higher risk of having children with birth defects. However, the increased risk was not statistically significant.[75]

Patients may need a reminder about the importance of using contraception during DMARD therapy and about the necessity of discontinuing some of these medications several months before conception is planned. In addition to discontinuance, some patients who take DMARDs may require treatment with other medications to enhance their clearance.

Patients with RA must be monitored closely after delivery because of the potential for arthritis flareups to occur during the postpartum period.

For complete information on this topic, see Rheumatoid Arthritis and Pregnancy.

Exercise, Education, Assistive Devices, and Occupational/Physical Therapies

Goals of rehabilitation for RA patients include the following:

Nonpharmacologic therapeutic modalities that are available to the physiatrist to assist patients in achieving these goals includ the following[76, 77, 78] :

Heat and cold therapies

Application of heat, either superficial or deep, is an effective modality for the relief of joint pain and stiffness caused by RA. In addition, it is used to treat joints in preparation for ROM, stretching, and muscle-strengthening exercises. Superficial and deep heating methods have been shown to raise the intra-articular temperature in patients with RA. Heat may be delivered via the following:

Cold is preferable for treatment of an acutely inflamed joint. Application of cold results in decreased pain and decreased muscle spasm. Cold may be delivered via ice packs, ice sticks, topical sprays, or ice water.

Orthotics and splints

Orthotic devices play an important role in the rehabilitation management of patients with RA. These devices are used to decrease pain and inflammation, improve function, reduce deformity, and correct biomechanical malalignment.

Lower-extremity orthoses are prescribed to provide stability and proper alignment or to shift weight-bearing off the affected limb. The most common orthoses used for the lower extremity involve the foot and ankle joints.

Approximately 80% of patients affected by RA have significant foot involvement. These problems are easily accommodated by providing a deep, wide, soft leather shoe. A metatarsal pad or bar is typically used to remove weight from painful metatarsophalangeal (MTP) joints, and a rocker-bottom sole can be used to facilitate rolloff. Hindfoot pronation should be addressed with custom inserts.

Knee orthoses may be used to control the following problems:

Therapeutic exercise

Fatigue and decreased endurance are frequent symptoms in patients with RA. When these patients are compared with age-matched subjects who do not have RA, a reduction in aerobic capacity and muscle strength is noted, both because of the disease itself and because of the lack of physical activity in these patients. Thus, exercise is an important part of rehabilitation management of RA.

Aerobic conditioning in affected patients (if tolerated) improves maximum oxygen uptake and decreases perceived exertion at submaximal workloads, reportedly without causing adverse effects in the joints. In addition, patients undergoing long-term endurance training have been known to feel less isolated, to take less sick leave, and to develop improved function in activities of daily living (ADLs). Patients with well-controlled RA should be encouraged to engage in 30 minutes of daily aerobic exercise several times a week.

A 21-week study by Katz et al in 96 patients with RA found that providing a pedometer and step-monitoring diary, with or without a step target,  increased activity levels and decreased fatigue. A control cohort that received education only showed a trend toward decreased steps.[79]

Muscle atrophy often accompanies RA and is exacerbated by inactivity, bed rest, splints, and medications. Isometric exercises restore and maintain strength in affected patients without producing pain. Resistance exercises may be initiated when the isometric program has been well established and when the patient is free of pain.[80]

In a review addressing hand function in RA patients, O’Brien suggested that strengthening hand exercises could yield significant improvements in the overall functioning of patients with this disease.[81] If surgery is not an option, medical management should be pursued in conjunction with rather thorough physical therapy in order to maintain and preserve hand function.[81]

Occupational therapy

Occupational therapy can be very useful for patients with RA.[82] Occupational therapy is initiated to help patients achieve the following:

An occupational therapist may work in conjunction with the physical therapist to ensure that patients are able to meet their goals. The occupational therapist may also assist in the recommendation and use of splints and orthotics, especially when the upper extremity is affected.

Upper-extremity orthoses may be classified as either static or dynamic. Static splints are used to support a weak or unstable joint, to rest a joint for pain relief, or to maintain functional alignment. Dynamic splints traditionally have been used to manage the postoperative hand, but they may also be used to increase manual dexterity. The most commonly used splints for the hand are the finger-ring splint and the thumb-post splint. The functional wrist splint and the resting hand splint are commonly used for wrist splinting.

Adaptive equipment

Many assistive devices are available to patients with RA and are used to maximize function, maintain independence, reduce joint stress, conserve energy, and provide pain relief. Equipment is available to assist patients with transfers, dressing, feeding, toileting, cooking, and ambulation. Physical and occupational therapists can assist with training in the use of adaptive equipment.

Joint-protection education

Joint-protection education provides the patient with techniques and recommendations for preventing joint overuse and avoiding of biomechanical torques that excessively bend the joint.[76] The use of adaptive equipment is important. Other components of a good joint-protection program include the following:

Energy-conservation education

Fatigue is a major component of RA, and it is due to the systemic nature of the disease, as well as to the decreased cardiovascular endurance observed in patients with this inflammatory disorder.

The goal of energy conservation techniques is to save energy while maximizing function. Adaptive equipment is an essential part of this program. Other elements include maintaining joint ROM and strength, improving cardiovascular fitness, and taking short rest periods during the day. Every individual with RA should implement joint-protection and energy-conservation programs into his or her lifestyle.

Williams et al found that in older women with either RA or osteoarthritis of the lower limb, an individualized home program of balance-training exercise can improve balance and gait stability.[83] In this study, 39 women (mean age, 69.3 years) underwent a 4-month program of balance exercises conducted by a physical therapist. Before exercise training, 64% of patients reported having had fallen during the previous 12 months, and 42% of patients had a moderate fall-risk score.

After the 4-month program, the patients demonstrated improvement on most balance measurements, including their fall-risk score and measurements of activity level, fear of falling, functional reach, and step width.[83] Improvements were also seen in patients’ body mass index (BMI) and in their sit-to-stand rising index.

Surgical Intervention

Surgical intervention in patients with RA can achieve pain relief, deformity correction, and functional improvement.[76] A number of surgical procedures are available to obtain these goals, such as myofascial techniques, excisions, reconstructions, joint fusions, and joint replacements. The timing of surgery is a complex decision; the patient’s age, stage of disease, and level of disability, as well as the location of the involved joints, must be considered. Early surgical intervention may help maintain a patient’s functional level of independence.

Deformities of the hand or wrist lead to loss of the ability to grip, grasp, and pinch, often leaving the patient unable to perform the activities of daily living. The surgical treatments for RA of the hand and wrist include the following:

Cervical spine instability may be observed in patients with established RA who have degeneration of the ligaments and bone in the C-spine area. Degeneration of the transverse ligament can lead to instability at the C1-C2 level. Minor trauma can lead to neurologic sequelae due to inherent instability.

Exercise caution in evaluating RA patients after minor falls, motor vehicle accidents, or other injuries. Cervical spine injury may occur spontaneously. Patients with long-standing disease may need careful evaluation perioperatively to detect any cervical spine instability before neck manipulation or intubation during surgery. Patients with RA of the cervical spine who have refractory pain, clearly evident neurologic compromise, or intrinsic spinal cord signal changes on magnetic resonance imaging (MRI) are generally candidates for surgical intervention.

For more information on these topics, see Hand and Wrist Surgery in Rheumatoid Arthritis and Rheumatoid Arthritis of the Cervical Spine.

Guidelines Summary

Disease Classification

The 2010 American College of Rheumatology (ACR)/European League Against Rheumatism (EULAR) classification criteria for RA are designed to identify patients with unexplained inflammatory arthritis in at least 1 peripheral joint and a short duration of symptoms who would benefit from early therapeutic intervention. They represents a paradigm shift from the 1987 ACR criteria, which lacked the sensitivity to detect early RA.[1]

According to the ACR/EULAR criteria, patients who should be tested are those with at least 1 joint with definite clinical synovitis that is not better explained by another disease (eg, lupus, psoriatic arthritis, or gout).[1]

The ACR/EULAR classification system is a score-based algorithm for RA that incorporates the following 4 factors:

The maximum number of points possible is 10. A classification of definitive RA requires a score of 6/10 or higher. Patients with a score lower than 6/10 should be reassessed over time. If patients already have erosive changes characteristic of RA, they meet the definition of RA, and application of this diagnostic algorithm is unnecessary.[1]

Disease Activity Measures

In 2012, the ACR published revised recommendations for the measurement of disease activity for clinical use at the point of care. The scales used included the following recommended measures[2] :

The 6 measures above result in a single continuous index with defined ranges for low, moderate, or high disease activity or clinical remission.[2] See Table 3, below.

Table 3. Rheumatoid Arthritis Disease Activity Cutoffs



View Table

See Table

Measurement of disease remission

In the 2012 ACR-recommended RA disease activity measures, the following cutoffs indicate disease remission[2] :

In 2011, the ACR and the EULAR jointly released two definitions for evaluating remission of RA in clinical trials, one a Boolean-based definition and the other based on a composite index of RA activity, the SDAI. The Boolean-based definition requires that the patient satisfy all of the following to be considered in remission[3] :

To be considered in remission using the traditional index definition, the patient must have an SDAI score of less than 3.3.[3]

Treating to Therapeutic Target

The following revised guidelines on treating RA to therapeutic target were issued in 2015 by an international task force of rheumatologists, patient representatives, and a rheumatology nurse[7, 8] :

Management of Early RA

In 2016, the European League Against Rheumatism (EULAR) updated its 2007 guidelines for the management of early arthritis, which put renewed emphasis on early intervention, preventive lifestyle measures, and careful clinical examination rather than reliance on ultrasound or advanced imaging. Key recommendations, based on evidence and expert opinion, are as follows[149] :

In its 2015 revised guidelines for the management of RA, the American College of Rheumatology (ACR) included the following recommendations for treatment of early RA[4] :

A change from 2012 to 2015 guidelines is that the 2015 guidelines do not recommend initial combination DMARD therapy in early RA with moderate to high disease activity.

Management of Established RA

Guidelines for pharmacologic therapy for RA have been issued by the following organizations:

The 2016 update of the EULAR RA management guidelines includes the following key recommendations for pharmacologic therapy[5] :

In its 2015 revised guidelines, the ACR included the following recommendations for treatment of patients with established RA[4] :

A change from 2012 to 2015 guidelines is that instead of switching from one anti-TNF biologic to another anti-TNF biologic because of continued activity, it is recommended to change first to a non-TNF biologic.

Medication Summary

Optimal care of patients with rheumatoid arthritis (RA) requires an integrated approach that includes both nonpharmacologic therapies and pharmacologic agents such as nonbiologic and biologic disease-modifying antirheumatic drugs (DMARDs), nonsteroidal anti-inflammatory drugs (NSAIDs), analgesics, and corticosteroids.

Leflunomide (Arava)

Clinical Context:  Leflunomide is indicated for the treatment of active RA to reduce signs and symptoms, inhibit structural damage and improve physical function. Corticosteroids, aspirin, or other NSAIDs may be continued during leflunomide use. Leflunomide is contraindicated in women who are or may become pregnant.

Leflunomide is a pyrimidine synthesis inhibitor that blocks autoimmune antibodies and reduces inflammation. It also inhibits dihydroorotate dehydrogenase, an enzyme in the de novo pyrimidine synthesis pathway and has antiproliferative activity. 

Sulfasalazine (Azulfidine, Azulfidine EN-tabs)

Clinical Context:  Sulfasalazine (SSZ) is indicated for the treatment of patients with RA who have had an inadequate response to salicylates or other NSAIDs. It acts locally to decrease inflammatory response and systemically inhibits prostaglandin synthesis.

SSZ delayed-release tablets do not have an immediate response; therefore, concurrent treatment with NSAIDs or other analgesics is recommended at least until the effect of the delayed-release tablets is apparent. The initial dosage is 0.5-1 g/day. The dosage can be adjusted to a dose of 3 g/day after 12 weeks if an adequate clinical response is not seen.

Hydroxychloroquine (Plaquenil)

Clinical Context:  Hydroxychloroquine (HCQ) is approved for the treatment of acute or chronic RA. The initial dosage is 400-600 mg/day; dosages should be computed on the basis of patient body weight. If a good clinical response is seen over a period of 4 to 12 weeks, the dosage can be reduced by 50% and continued at a level of 200-400 mg/day. The risk of retinopathy is greater when this dosage is exceeded. It should be noted that each 200 mg tablet contains only 155 mg of active drug.

Patients must have a baseline eye examination (including color and vision testing, funduscopic examination, and visual-field testing) performed before starting HCQ therapy. Most rheumatologists recommend an HCQ eye examination every 6-12 months.

Rituximab (Rituxan)

Clinical Context:  Rituximab is most often used in combination with methotrexate (MTX). It has been shown to be effective at reducing signs and symptoms in adult patients with moderately to severely active RA who have had an inadequate response to therapy with 1 or more tumor necrosis factor (TNF) antagonists. Treatment with rituximab may deplete CD20+ B cells. The usual regimen consists of 2 intravenous (IV) infusions of 1000 mg, separated by 2 weeks, in combination with MTX.

Class Summary

DMARDs represent the most important measure in the successful treatment of rheumatoid arthritis. These agents can retard or prevent disease progression and, thus, joint destruction and subsequent loss of function. Successful DMARD therapy may eliminate the need for other anti-inflammatory or analgesic medications; however, until the full action of DMARDs takes effect, anti-inflammatory or analgesic medications may be required as bridging therapy to reduce pain and swelling.

Methotrexate (Rheumatrex, Trexall, Otrexup)

Clinical Context:  MTX is a folic acid antagonist that is approved for the management of severe active RA in patients who have had an insufficient therapeutic response to or are intolerant of an adequate trial of first-line therapy, including full-dose NSAIDs. It inhibits dihydrofolic acid reductase, which ultimately interferes with DNA synthesis, repair, and cellular replication.

Tocilizumab (Actemra)

Clinical Context:  Tocilizumab is an IL-6 receptor inhibitor. It is indicated for moderate to severe active RA in adults who have had an inadequate response to 1 or more TNF-antagonist therapies. It has been approved as an IV infusion or SC injection that may be used alone or in combination with MTX or other DMARDs. 

Sarilumab (Kevzara)

Clinical Context:  Human monoclonal antibody that binds to both soluble and membrane-bound IL-6 receptors (sIL-6R and mIL-6R). It is administered as a SC injection. May be prescribed as monotherapy or with methotrexate or other conventional DMARDs.

Azathioprine (Imuran)

Clinical Context:  Although azathioprine is not a first-line agent, it is sometimes used in the treatment of active RA to reduce signs and symptoms, particularly in patients who may have coinciding connective tissue diseases, such as systemic lupus erythematosus. Aspirin, NSAIDs, or low-dose glucocorticoids may be continued during treatment with azathioprine. The mechanism whereby azathioprine affects autoimmune diseases is unknown; however, it works primarily on T cells.

The initial dosage is 1 mg/kg/day (50-100 mg/day) given as a single dose or in divided doses twice daily. The dosage may be increased by 0.5 mg/kg/day at 6-8 weeks and thereafter at 4-week intervals, up to a maximum dosage of 2.5 mg/kg/day. Azathioprine is available in tablet form for oral administration or in 100-mg vials for IV injection.

Cyclosporine (Gengraf, Neoral)

Clinical Context:  Although cyclosporine is approved for the treatment of patients with severe active RA in which the disease has not adequately responded to MTX, it is not commonly used to treat RA, because of its nephrotoxicity. When cyclosporine is used, patients' renal function must be closely monitored.

Cyclosporine can be used in combination with MTX in RA patients who do not have an adequate response to MTX alone.

Anakinra (Kineret)

Clinical Context:  Anakinra is a recombinant, nonglycosylated form of the human interleukin (IL)-1 receptor antagonist (IL-1Ra) that is indicated for reducing signs and symptoms and slowing the progression of structural damage of moderately to severely active RA that has failed treatment with 1 or more DMARDs. Anakinra can be used alone or in combination with DMARDs other than TNF-blocking agents.

Abatacept (Orencia)

Clinical Context:  Abatacept is a selective costimulation modulator that inhibits T-cell activation by binding to CD80 and CD86, thereby blocking their interaction with CD28. CD28 interaction provides a signal needed for the full T-cell activation that is implicated in RA pathogenesis.

This agent is approved for reducing signs and symptoms, inducing major clinical response, inhibiting the progression of structural damage, and improving physical function in adult patients with moderately to severely active RA. Abatacept can be used as monotherapy or concomitantly with DMARDs other than TNF antagonists.

Class Summary

DMARDs represent the most important measure in the successful treatment of RA.

Infliximab (Remicade, Inflectra, infliximab-dyyb, Renflexis, infliximab-abda, Ixifi, infliximab-qbtx)

Clinical Context:  Infliximab, a chimeric monoclonal antibody against TNF-α, is approved for reducing signs and symptoms, inhibiting the progression of structural damage, and improving physical function in patients with moderately to severely active RA, in combination with MTX. This agent binds to cells that express membrane TNF. FDA approved infliximab-dyyb, infliximab-abda, and infliximab-qbtx as biosimilars and not as an interchangeable drug.

Etanercept (Enbrel, Erelzi, etanercept-szzs)

Clinical Context:  Etanercept, a bivalent p75–TNF receptor linked to the Fc portion of human immunoglobulin G (IgG), is approved for reducing signs and symptoms, inducing major clinical response, inhibiting the progression of structural damage, and improving physical function in patients with moderately to severely active RA. It can be given alone or in combination with MTX. This agent binds lymphotoxin (formerly termed TNF-β) in addition to soluble TNF-α. 

Golimumab (Simponi, Simponi Aria)

Clinical Context:  Golimumab, a human monoclonal antibody to TNF-α, inhibits TNF-α bioactivity, thereby modulating immune activity in patients with RA. It is approved for the treatment of adults with moderately to severely active RA, in combination with MTX. It may be administered as either a SC injection every month, or as an IV infusion every 2 months following 2 once monthly doses. Golimumab should be given in combination with MTX.

Certolizumab (Cimzia)

Clinical Context:  Certolizumab is a pegylated anti−TNF-α agent, which results in disruption of the inflammatory process in RA. It is indicated for the treatment of adults with moderately to severely active RA. 

Adalimumab (Amjevita, Cyltezo, Humira, Hadlima, Hyrimoz, Adalimumab-atto, Adalimumab-adbm, Adalimumab-bwwd, Adalimumab-adaz)

Clinical Context:  Adalimumab is a recombinant human IgG1 monoclonal antibody that is specific for human TNF. It binds specifically to TNF-α and blocks interaction with p55 and p75 cell-surface TNF receptors. It is indicated to reduce inflammation and inhibit progression of structural damage in moderate to severe RA, alone or in combination with MTX or other nonbiologic DMARDs. This agent is reserved for those who experience an inadequate response to 1 or more DMARDs. Adalimumab can be used alone or in combination with MTX or other DMARDs. FDA approved adalimumab-atto, adalimumab-adbm, adalimumab-adaz, adalimumab-bwwd, as biosimilars and not as interchangeable drugs.

Class Summary

The recognition of TNF-α and IL-1 as central proinflammatory cytokines has led to the development of agents that block either these cytokines or their effects. In addition to improving signs and symptoms and quality of life, all biologic agents significantly retard radiographic progression of joint erosions.

The TNF inhibitors, which bind TNF and thus prevent its interaction with its receptors, include etanercept, infliximab, golimumab, certolizumab, and adalimumab. Consensus statements do not recommend their use until at least one xenobiotic DMARD, usually MTX, has been administered without sufficient success.

Adverse effects associated with the biologic agents include the generation of antibodies against these compounds, the emergence of antinuclear antibodies (ANAs), occasional drug-induced lupuslike syndromes, and infections. Rarely, demyelinating disorders and bone marrow suppression occur.

Acute and chronic infections, demyelinating disorders, New York Heart Association (NYHA) class III or IV heart failure, and recent malignancies are contraindications for the use of TNF inhibitors. A thorough search for latent tuberculosis using chest radiography or purified protein derivative (PPD) testing is recommended before these agents are started. Patients taking anti-TNF agents must avoid live-virus vaccines to avoid the risk of serious infection.

Tofacitinib (Xeljanz, Xeljanz XR)

Clinical Context:  Tofacitinib is an oral JAK inhibitor. It is indicated as second-line treatment for moderate to severe active RA in patients with an inadequate response to or intolerance of MTX. Tofacitinib may be used as monotherapy or in combination with MTX or other nonbiologic DMARDs, but it should not be used in combination with biologic DMARDs or potent immunosuppressive agents (eg, azathioprine and cyclosporine). Tofacitinib is a JAK inhibitor that modulates the signaling pathway at the point of JAKs, preventing the phosphorylation and activation of STATs.

Baricitinib (Olumiant)

Clinical Context:  Oral JAK inhibitor indicated for moderately to severely active RA as second-line treatment in adults who have had an inadequate response to one or more TNF-antagonists. It may be used as monotherapy or in combination with methotrexate or other nonbiologic DMARDs. It should not be used in combination with biologic DMARDs or potent immunosuppressive agents (eg, azathioprine or cyclosporine).

Upadacitinib (Rinvoq)

Clinical Context:  Indicated for moderately to severely active RA in adults who have had an inadequate response or are intolerant to methotrexate. It may be used as monotherapy or in combination with methotrexate or other nonbiological DMARDs.

Class Summary

Janus kinase (JAK) pathway inhibitors consist of a group of intracellular tyrosine kinases that transmit signals from cytokine or growth factor-receptor interactions on the cellular membrane to influence cellular processes of hematopoieses and immune cell function. Within the signaling pathway, JAKs phosphorylate and activate Signal Transducers and Activators of Transcription (STATs) which modulate intracellular activity including gene expression.

Minocycline (Dynacin, Minocin)

Clinical Context:  The anti-inflammatory effects of minocycline may result from inhibition of inflammatory cell migration and transformation of lymphocytes.

Class Summary

Although tetracyclines are not typically used to treat RA, the ACR recommends minocycline monotherapy in patients with a disease duration shorter than 24 months who have low disease activity and no poor prognostic factors.[69, 4]

Ibuprofen (Advil, Motrin)

Clinical Context:  Ibuprofen is indicated for patients with mild to moderate pain. It inhibits inflammatory reactions and pain by decreasing prostaglandin synthesis.

Naproxen (Naprosyn, Aleve, Anaprox, Anaprox DS, Naprelan)

Clinical Context:  Naproxen is used to relieve mild to moderate pain. This agent inhibits inflammatory reactions and pain by decreasing the activity of cyclooxygenase, which is responsible for prostaglandin synthesis.

Diclofenac (Voltaren, Cambia)

Clinical Context:  Diclofenac is one of a series of phenylacetic acids that have demonstrated anti-inflammatory and analgesic properties in pharmacologic studies. It is believed to inhibit COX, which is essential in the biosynthesis of prostaglandins.

Ketoprofen

Clinical Context:  Ketoprofen is used to relieve mild to moderate pain and inflammation. It reversibly inhibits cyclooxygenase-1 and 2 (COX-1 and 2) enzymes, which results in decreased formation of prostaglandin precursors.

Celecoxib (Celebrex)

Clinical Context:  Celecoxib is approved for the relief of signs and symptoms of RA. It primarily inhibits COX-2, which is considered an inducible isoenzyme (induced during pain and inflammatory stimuli). Inhibition of COX-1 may contribute to NSAID gastrointestinal (GI) toxicity. At therapeutic concentrations, celecoxib does not inhibit COX-1, thus potentially resulting in decreased GI toxicity. Administer the lowest possible dose for each patient. Use of celecoxib has been associated with an increased risk of cardiovascular toxicity.

Class Summary

NSAIDs interfere with prostaglandin synthesis through inhibition of the enzyme cyclooxygenase (COX), thus reducing swelling and pain. However, they do not retard joint destruction and therefore are not sufficient to treat RA when used alone. Like corticosteroids, NSAIDs can be reduced in dose or discontinued with successful DMARD therapy. These agents decrease intraglomerular pressure and decrease proteinuria.

The several dozen NSAIDs available can be classified into several different groups of compounds. Commonly used NSAIDs include ibuprofen, naproxen, ketoprofen, celecoxib, and diclofenac.

Acetaminophen (Tylenol)

Clinical Context:  Acetaminophen is used for analgesia in patients with documented hypersensitivity to aspirin or NSAIDs, those with upper GI disease, and those who are taking oral anticoagulants. This agent does not have anti-inflammatory properties.

Class Summary

Analgesics such as acetaminophen may be used in patients with RA.

Tramadol (Ultram)

Clinical Context:  The immediate-release formulation of tramadol is approved for the management of moderate to moderately severe pain in adults. The extended-release formulation is used for the management of moderate to moderately severe chronic pain in adults who require around-the-clock treatment of pain for an extended period of time. Tramadol and its active metabolite (M1) binds to mu-opiate receptors in the central nervous system causing inhibition of ascending pain pathways, altering perception of and response to pain.

Class Summary

Tramadol has been used to reduce pain in patients with RA. However, this agent only provides analgesic effects and does not have anti-inflammatory properties.

Diclofenac topical (Voltaren)

Clinical Context:  Diclofenac topical gel is approved in patients with osteoarthritis at a dosage of 32 g/day applied over all affected joints. It has also been used to provide analgesic effects in patients with RA.

Class Summary

Topical agents such as diclofenac can provide analgesia for patients with RA. This agent is commonly used in patients who experience acute pain.

Prednisone (Prednisone Intensol, Rayos)

Clinical Context:  Prednisone is an immunosuppressant for the treatment of autoimmune disorders; it may decrease inflammation by reversing increased capillary permeability and suppressing polymorphonuclear leukocyte activity. Prednisone stabilizes lysosomal membranes and suppresses lymphocytes and antibody production.

Circadian (ie, bedtime) administration of modified-release prednisone (Rayos) has been shown shown to decrease morning stiffness with RA.

Methylprednisolone (Depo-Medrol, Medrol, Solu-Medrol)

Clinical Context:  Methylprednisolone decreases inflammation by suppressing the migration of polymorphonuclear leukocytes (PMNs) and reversing increased capillary permeability. It is as adjunctive therapy for short term administration (for an acute episode or exacerbation) in RA.

Prednisolone (Orapred ODT, Pediapred)

Clinical Context:  Prednisolone controls or prevents inflammation by controlling the rate of protein synthesis, suppressing the migration of PMNs and fibroblasts, reversing capillary permeability, and stabilizing lysosomes at the cellular level. It is as adjunctive therapy for short term administration (for an acute episode or exacerbation) in RA.

Class Summary

Corticosteroids are potent anti-inflammatory drugs commonly used in patients with RA to bridge the time until DMARDs become effective. Prednisone dosages as high as 10 mg/day are typically used, but some patients may require even higher dosages. Adverse events associated with long-term steroid use make dose reductions and cessation important in due course.

Penicillamine (Cuprimine, Depen)

Clinical Context:  Penicillamine depresses circulating IgM rheumatoid factor and T-cell activity (but not B-cell activity). It is indicated in patients with severe, active RA who have failed to respond to an adequate trial of conventional therapy.

Class Summary

Chelation therapy in RA may suppress inflammation and arthritis by depressing T-cell activity.

What is rheumatoid arthritis (RA)?What are the signs and symptoms of rheumatoid arthritis (RA)?Which assessments should be included in the physical exam for rheumatoid arthritis (RA)?Which lab studies are helpful in the diagnosis of rheumatoid arthritis (RA)?Which imaging modalities are helpful in the diagnosis of rheumatoid arthritis (RA)?Is joint aspiration useful in the diagnosis of rheumatoid arthritis (RA)?Other than medication and surgery, which therapies exist for rheumatoid arthritis (RA)?Which nonbiologic DMARDS are used to treat rheumatoid arthritis (RA)?Which biologic tumor necrosis factor (TNF)-inhibiting DMARDS are used to treat rheumatoid arthritis (RA)?Which biologic non–tumor necrosis factor (TNF) DMARDS are used to treat rheumatoid arthritis (RA)?What drugs other than DMARDS are used to treat rheumatoid arthritis (RA)?Which surgical procedures are used to treat rheumatoid arthritis (RA)?What are the main features of rheumatoid arthritis (RA)?What lab test results suggest rheumatoid arthritis (RA)?How is rheumatoid arthritis (RA) treated?What is macrophage activation syndrome as it relates to juvenile idiopathic arthritis (JIA)?What are the treatment guidelines for managing rheumatoid arthritis (RA) to therapeutic target?What is the pathogenesis of rheumatoid arthritis (RA)?Which cells have a major role in the pathophysiology of rheumatoid arthritis (RA)?What causes the tissue destruction in rheumatoid arthritis (RA)?Is rheumatoid arthritis (RA) genetic?What causes rheumatoid arthritis (RA)?What is juvenile idiopathic arthritis (JIA), and is it genetically similar to rheumatoid arthritis (RA)?What is the role of genomic imprinting and epigenetics in the etiology of rheumatoid arthritis (RA)?Which infectious agents may cause rheumatoid arthritis (RA)?Do sex hormones increase the risk of developing rheumatoid arthritis (RA)?What is the role of immunologic factors in the development of rheumatoid arthritis (RA)?What is the role of T cells in the pathogenesis of rheumatoid arthritis (RA)?What is the role of B cells in the pathology of rheumatoid arthritis (RA)?What is the role of synovial membrane in the pathology of rheumatoid arthritis (RA)?What is the global prevalence of rheumatoid arthritis (RA) among different age groups and ethnicities?Does rheumatoid arthritis (RA) in first-degree relatives increase risk?Is rheumatoid arthritis (RA) more common in women or men?What are the possible clinical outcomes of rheumatoid arthritis (RA)?How can early treatment improve the prognosis in rheumatoid arthritis (RA)?What factors correlate with an unfavorable prognosis in rheumatoid arthritis (RA)?Which findings are associated with radiographic progression in patients who received methotrexate (MTX) for rheumatoid arthritis (RA)?Which lab results suggest a poor prognosis in rheumatoid arthritis (RA)?How does duration of rheumatoid arthritis (RA) affect prognosis?What is the prevalence of full employment among patients with rheumatoid arthritis (RA)?What risk factors contribute to mortality in rheumatoid arthritis (RA)?What nontraditional cardiovascular risk factors contribute to mortality and morbidity in rheumatoid arthritis (RA)?What is the mortality rate in rheumatoid arthritis (RA)?What are the benefits of patient education and counseling in rheumatoid arthritis (RA)?What is the best way to inform patients about their diagnosis of rheumatoid arthritis (RA)?What is the best way to communicate prognosis and treatment options to patients and their families after a diagnosis of rheumatoid arthritis (RA)?Is it beneficial to tell a patient that there are no controllable causes for rheumatoid arthritis (RA)?What is the best response to a patient expressing interest in alternative and complementary forms of therapy for rheumatoid arthritis (RA)?What are the common misconceptions about the effectiveness of less costly rheumatoid arthritis (RA) treatments?What are the benefits of active participation of families in the management of patients with rheumatoid arthritis (RA)?What is a common fear in patients with rheumatoid arthritis (RA)?What is the best approach to supporting patients with severe rheumatoid arthritis (RA)?What are the main features of rheumatoid arthritis (RA)?What is juvenile idiopathic arthritis (JIA)?What is the impact of rheumatoid arthritis (RA) on a patient’s ability to perform activities of daily living (ADLs)?Is the onset of rheumatoid arthritis (RA) insidious or abrupt?What should be assessed in a physical exam for rheumatoid arthritis (RA)?How does rheumatoid arthritis (RA) affect the elbow?How does rheumatoid arthritis (RA) affect the cervical spine?Which joints are affected by rheumatoid arthritis (RA) and what are the symptoms?What are the musculoskeletal manifestations of rheumatoid arthritis (RA) other than joint involvement?What is a boutonniere deformity in rheumatoid arthritis (RA)?What is a swan-neck deformity of the finger in rheumatoid arthritis (RA)?How is tightness of intrinsic muscles due to rheumatoid arthritis (RA) assessed?What is the prognosis of flexor tenosynovitis/trigger finger due to rheumatoid arthritis (RA)?What is arthritis mutilans (opera glass hands) due to rheumatoid arthritis (RA)?How does rheumatoid arthritis (RA) affect metacarpophalangeal (MCP) joints?How does rheumatoid arthritis (RA) affect the wrist?How does rheumatoid arthritis (RA) affect the shoulders?How does rheumatoid arthritis (RA) affect the ankles and feet?How does rheumatoid arthritis (RA) affect the knees?How does rheumatoid arthritis (RA) affect the hips?What are clinical manifestations of early cervical spine disease due to rheumatoid arthritis (RA)?What are the manifestations of neurologic involvement in the cervical spine due to rheumatoid arthritis (RA)?What do transient ischemic attacks (TIAs) and cerebellar signs indicate in rheumatoid arthritis (RA)?What are symptoms of cervical myelopathy in rheumatoid arthritis (RA)?How can stiffness caused by rheumatoid arthritis (RA) be determined during physical exam?How is stiffness in rheumatoid arthritis (RA) relieved?How are pain and tenderness assessed during physical exam in rheumatoid arthritis (RA)?How is enlargement of the synovial membrane noted on physical exam in rheumatoid arthritis (RA)?How is synovial proliferation identified during a physical exam in rheumatoid arthritis (RA)?How does a Baker cyst form in rheumatoid arthritis (RA)?Where are Baker cysts most commonly found in rheumatoid arthritis (RA)?How does rheumatoid arthritis (RA) cause joint deformity?How does rheumatoid arthritis (RA) cause limitation of motion?What are the common extra-articular manifestations of rheumatoid arthritis (RA)?How do rheumatoid nodules affect patients with rheumatoid arthritis (RA)?Which organ systems are affected by rheumatoid arthritis (RA)?How does rheumatoid arthritis (RA) affect the heart/cardiovascular system?How does rheumatoid arthritis (RA) affect the lungs?How does rheumatoid arthritis (RA) affect the kidneys?How does rheumatoid arthritis (RA) affect the GI system?Where do vasculitis lesions develop in rheumatoid arthritis (RA)?Is anemia a presentation of rheumatoid arthritis (RA)?Is nerve entrapment a presentation of rheumatoid arthritis (RA)?How does rheumatoid arthritis (RA) affect the eye?What are the ACR and EULAR diagnostic criteria for rheumatoid arthritis (RA)?Who should be tested for rheumatoid arthritis (RA), according to 2010 ACR/EULAR criteria?What is the 2010 ACR/EULAR classification system for rheumatoid arthritis (RA)?What are the 2012 ACR recommendations for the measurement of disease activity in rheumatoid arthritis (RA)?What are the 4 stages of progression of rheumatoid arthritis (RA)?What are the ACR–recommended rheumatoid arthritis (RA) disease activity cutoffs?What is the 2011 ACR/EULAR definition of remission in rheumatoid arthritis (RA)?How is remission in rheumatoid arthritis (RA) predicted?What are the rheumatoid arthritis (RA) disease activity measure cutoffs that indicate disease remission for patient-driven composite tools?What are the rheumatoid arthritis (RA) disease remission cutoffs for RA patient, provider, and lab composite tools?What are the 4 functional classes of rheumatoid arthritis (RA)?Is rheumatoid arthritis (RA) fatal?How can rheumatoid arthritis (RA) affect daily life?Which parts of the body are affected by rheumatoid arthritis (RA) in addition to joints?How is rheumatoid arthritis (RA) differentiated from other diseases?How is rheumatic fever differentiated from rheumatoid arthritis (RA)?How is systemic lupus erythematosus (SLE) differentiated from rheumatoid arthritis (RA)?How is degenerative joint disease (DJD) differentiated from rheumatoid arthritis (RA)?How is gouty arthritis differentiated from rheumatoid arthritis (RA)?How is pyogenic arthritis differentiated from rheumatoid arthritis (RA)?How is chronic Lyme disease differentiated from rheumatoid arthritis (RA)?How is polymyalgia rheumatica differentiated from rheumatoid arthritis (RA)?How are carcinomas differentiated from rheumatoid arthritis (RA)?What are the differential diagnoses for Rheumatoid Arthritis?How is rheumatoid arthritis (RA) diagnosed?Which lab studies are useful in the diagnosis of rheumatoid arthritis (RA)?Which inflammation markers are associated with rheumatoid arthritis (RA)?What are the hematologic findings consistent with a diagnosis of rheumatoid arthritis (RA)?What are the immunologic findings consistent with a diagnosis of rheumatoid arthritis (RA)?Is rheumatoid factor (RF) specific to rheumatoid arthritis (RA)?What is the significance of antinuclear antibodies (ANAs) in rheumatoid arthritis (RA)?Are assays for anti-citrullinated protein antibodies (ACPA) used to diagnose rheumatoid arthritis (RA)?Can anti-cyclic citrullinated peptide (anti-CCP) antibodies testing be used to diagnose rheumatoid arthritis (RA)?Can rheumatoid arthritis (RA) biomarkers be used to diagnose early RA?How does pregnancy affect lab results in rheumatoid arthritis (RA)?How is radiography used to assess rheumatoid arthritis (RA)?How is MRI used to assess rheumatoid arthritis (RA)?Is ultrasonography useful in the assessment of patients with rheumatoid arthritis (RA)?What is the most effective method of hand imaging in rheumatoid arthritis (RA)?What is the most effective method of spinal imaging in rheumatoid arthritis (RA)?When should joint aspiration and synovial fluid analysis be considered in patients with rheumatoid arthritis (RA)?Which synovial fluid analysis findings suggest rheumatoid arthritis (RA)?Which histologic findings suggest rheumatoid arthritis (RA)?What is the most effective management approach for rheumatic arthritis (RA)?What are the nonpharmacological treatment options for rheumatoid arthritis (RA)?What can a doctor do to boost morale and ensure compliance with therapy in patients with rheumatoid arthritis (RA)?What are the pharmacologic treatment options for rheumatoid arthritis (RA)?What impact does pregnancy have on the treatment of rheumatoid arthritis (RA)?What are the surgical options in the treatment of rheumatoid arthritis (RA)?Should patients with rheumatoid arthritis (RA) taking anti-tumor necrosis factor (TNF) agents avoid live-virus vaccines?Are live-virus vaccines safe in patients with rheumatoid arthritis (RA) who are taking anti-tumor necrosis factor (TNF) agents?Which medications are used in the treatment of rheumatoid arthritis (RA)?Are biologic agents or nonbiologic DMARDs more effective in improving physical function in rheumatoid arthritis (RA)?Which vaccinations are recommended by the ACR prior to pharmacologic treatment for rheumatoid arthritis (RA)?What are the Agency for Healthcare Research and Quality (AHRQ) recommendations for the pharmacologic therapy of rheumatoid arthritis (RA)?Which nonbiologic and biologic DMARDs are used for rheumatoid arthritis (RA)?What is the role of DMARDs in the management of rheumatoid arthritis (RA)?What are the benefits of early treatment of rheumatoid arthritis (RA) with DMARDs?Which nonbiologic DMARDs decrease the risk of diabetes in rheumatoid arthritis (RA)?Which DMARDs are most effective in triggering remission of rheumatoid arthritis (RA)?What are the indications of methotrexate (MTX) subcutaneous autoinjector (Otrexup) in rheumatoid arthritis (RA)?What is leflunomide and is it effective to treat rheumatoid arthritis (RA)?Which tumor necrosis factor (TNF) inhibitors are used in the treatment of rheumatoid arthritis (RA)?When should biologic agents be considered for treatment of rheumatoid arthritis (RA)?What are the adverse effects of biologic agents used to treat rheumatoid arthritis (RA)?What are the contraindications to the use of tumor necrosis factor (TNF) inhibitors in the treatment of rheumatoid arthritis (RA)?What are the risks of anti-tumor necrosis factor (TNF) therapy in rheumatoid arthritis (RA)?How do tumor necrosis factor (TNF) inhibitors compare to methotrexate (MTX) in the treatment of rheumatoid arthritis (RA)?At what blood level is adalimumab (Humira) most effective on disease activity in rheumatoid arthritis (RA)?What are the effects of certolizumab in rheumatoid arthritis (RA)?What is golimumab and is it effective in treating rheumatoid arthritis (RA)?What is the role of rituximab in the treatment of rheumatoid arthritis (RA)?What is the role of anakinra in the treatment of rheumatoid arthritis (RA)?What is the role of abatacept in the treatment of rheumatoid arthritis (RA)?How is abatacept administered in patients with rheumatoid arthritis (RA)?How is tocilizumab administered in patients with rheumatoid arthritis (RA)?How is sarilumab administered in patients with rheumatoid arthritis (RA)?What is the role of janus kinases (JAKs) in the treatment of rheumatoid arthritis (RA)?When is tofacitinib indicated in the treatment of rheumatoid arthritis (RA)?How effective is tofacitinib in the treatment of rheumatoid arthritis (RA)?How effective are DMARDs in the treatment of rheumatoid arthritis (RA)?How is disease activity reduced in refractory rheumatoid arthritis (RA)?When should drug combination therapy be considered for the treatment of rheumatoid arthritis (RA)?How are the adverse effects of combination therapy for rheumatoid arthritis (RA) managed?What are the most common adverse effects of combination therapy for rheumatoid arthritis (RA)?What are the possible adverse effects of antimalarial agents used for rheumatoid arthritis (RA)?What are possible adverse effects of DMARDs in rheumatoid arthritis (RA)?Does anti-tumor necrosis factor (TNF) therapy for rheumatoid arthritis (RA) increase mortality rates in comparison to standard nonbiologic DMARD therapy?Do tumor necrosis factor (TNF) inhibitors increase the risk of malignancy in rheumatoid arthritis (RA)?What role do corticosteroids play in the treatment of rheumatoid arthritis (RA)?What are possible adverse effects of corticosteroid use in the treatment of rheumatoid arthritis (RA)?What is the role of NSAIDS in the treatment of rheumatoid arthritis (RA)?What are possible adverse effects of NSAIDS in the treatment of rheumatoid arthritis (RA)?What is the role of analgesics in the treatment of rheumatoid arthritis (RA)?What experimental therapies are under investigation for the management of rheumatoid arthritis (RA)?Are biologic agents recommended in rheumatoid arthritis (RA) with chronic heart failure?What are the EULAR guidelines to prevent overtreatment of rheumatoid arthritis (RA)?What are the ACR recommendations for the use of DMARDS in the treatment of rheumatoid arthritis (RA)?Is the use of biologic agents recommended in rheumatoid arthritis (RA) and hepatitis?Is the use of biologic agents safe in patients with rheumatoid arthritis (RA) who have had cancer?What are the treatment recommendations for rheumatoid arthritis (RA) in patients who have had cancer?Which patients should be screened for latent TB infection (LTBI) before initiating biologic agents for the treatment of rheumatoid arthritis (RA)?Which latent TB infection (LTBI) screening tests should be performed before initiating treatment of rheumatoid arthritis (RA) with biologic agents?Which vaccinations should be administered prior to treatment of rheumatoid arthritis (RA) with DMARDs or biologic agents?Are there guidelines for the use of anti-tumor necrosis factor (TNF) drugs to treat rheumatoid arthritis (RA) during pregnancy?Which treatments should be considered for acute presentations of rheumatoid arthritis (RA)?How is Felty syndrome treated in rheumatoid arthritis (RA)?How are Baker cysts treated in rheumatoid arthritis (RA)?How is carpal tunnel syndrome (CTS) treated in rheumatoid arthritis (RA)?How does pregnancy affect the course of rheumatoid arthritis (RA)?Which rheumatoid arthritis (RA) medications are considered low-risk in pregnant women?What should be considered before initiating therapy in pregnant patients with rheumatoid arthritis (RA)?Is contraception required for women treated with DMARD therapy for rheumatoid arthritis (RA)?Should women with rheumatoid arthritis (RA) be monitored during the postpartum period?What are the goals of rheumatoid arthritis (RA) rehabilitation?What treatments are available to a physiatrist working with patients with rheumatoid arthritis (RA)?Do heat and cold therapies relieve the symptoms of rheumatoid arthritis (RA)?Are orthotic devices and splints beneficial in the treatment of rheumatoid arthritis (RA)?What is the role of therapeutic exercise in the management of rheumatoid arthritis (RA)?What are the benefits of aerobic conditioning in rheumatoid arthritis (RA)?What interventions have been shown to increase activity levels in rheumatoid arthritis (RA)?Which exercises are beneficial in rheumatoid arthritis (RA) and muscle atrophy?Do strengthening hand exercises improve hand function in rheumatoid arthritis (RA)?What are the goals of occupational therapy in rheumatoid arthritis (RA)?What is the role of an occupational therapist in the management of rheumatoid arthritis (RA)?What is the role of splints used to treat upper-extremity orthoses in rheumatoid arthritis (RA)?Are there assistive devices for patients with rheumatoid arthritis (RA)?What are the components of a joint-protection program in rheumatoid arthritis (RA)?Is an energy conservation program beneficial in rheumatoid arthritis (RA)?What is the purpose of surgical intervention in rheumatoid arthritis (RA)?What are the surgical options for rheumatoid arthritis (RA) of the hand and wrist?What causes cervical spine instability in rheumatoid arthritis (RA)?Who should be tested for rheumatoid arthritis (RA), according to ACR/EULAR criteria?What is the ACR/EULAR classification system for rheumatoid arthritis (RA)?What are the ACR recommendations for the measurement of rheumatoid arthritis (RA) disease activity?What are the ACR guidelines on the measurement of disease remission for rheumatoid arthritis (RA)?What are the guidelines for treating rheumatoid arthritis (RA) to therapeutic target?What are the EULAR treatment guidelines for early rheumatoid arthritis (RA)?What are the ACR treatment guidelines for early rheumatoid arthritis (RA)?What are the EULAR guidelines for pharmacologic therapy in rheumatoid arthritis (RA)?What are the ACR treatment guidelines for established rheumatoid arthritis (RA)?Which medications are used in the treatment of rheumatoid arthritis (RA)?Which medications in the drug class DMARDs, Other are used in the treatment of Rheumatoid Arthritis?Which medications in the drug class DMARDs, Immunomodulators are used in the treatment of Rheumatoid Arthritis?Which medications in the drug class DMARDs, TNF Inhibitors are used in the treatment of Rheumatoid Arthritis?Which medications in the drug class DMARDs, JAK Inhibitors are used in the treatment of Rheumatoid Arthritis?Which medications in the drug class Tetracyclines are used in the treatment of Rheumatoid Arthritis?Which medications in the drug class Nonsteroidal Anti-inflammatory Drugs are used in the treatment of Rheumatoid Arthritis?Which medications in the drug class Analgesics, Other are used in the treatment of Rheumatoid Arthritis?Which medications in the drug class Opioid Analgesics are used in the treatment of Rheumatoid Arthritis?Which medications in the drug class Topical Skin Products are used in the treatment of Rheumatoid Arthritis?Which medications in the drug class Corticosteroids are used in the treatment of Rheumatoid Arthritis?Which medications in the drug class Chelators are used in the treatment of Rheumatoid Arthritis?

Author

Howard R Smith, MD, Director of the Lupus Clinic, Department of Rheumatic and Immunologic Diseases, Cleveland Clinic

Disclosure: Nothing to disclose.

Coauthor(s)

Adam Brown, MD, Fellow, Department of Rheumatology, Cleveland Clinic

Disclosure: Nothing to disclose.

Chief Editor

Herbert S Diamond, MD, Visiting Professor of Medicine, Division of Rheumatology, State University of New York Downstate Medical Center; Chairman Emeritus, Department of Internal Medicine, Western Pennsylvania Hospital

Disclosure: Nothing to disclose.

Additional Contributors

Katherine K Temprano, MD, Assistant Professor of Internal Medicine, Division of Rheumatology, St Louis University School of Medicine

Disclosure: Partner received honoraria from Baxter for speaking and teaching.

Acknowledgements

Edward Bessman, MD Chairman, Department of Emergency Medicine, John Hopkins Bayview Medical Center; Assistant Professor, Department of Emergency Medicine, Johns Hopkins University School of Medicine

Edward Bessman, MD is a member of the following medical societies: American Academy of Emergency Medicine, American College of Emergency Physicians, and Society for Academic Emergency Medicine

Disclosure: Nothing to disclose.

Sarjoo M Bhagia, MD Consulting Staff, OrthoCarolina; Voluntary Teaching Faculty, Carolinas Rehabilitation

Sarjoo M Bhagia, MD is a member of the following medical societies: American Academy of Physical Medicine and Rehabilitation, Association of Academic Physiatrists, North American Spine Society, and Physiatric Association of Spine, Sports and Occupational Rehabilitation

Disclosure: Nothing to disclose.

Heather Lyn Carone, MD Attending Physician, Department of Emergency Medicine, St Vincent Mercy Medical Center

Heather Lyn Carone, MD is a member of the following medical societies: American College of Emergency Physicians, American Medical Association, and Society for Academic Emergency Medicine

Disclosure: Nothing to disclose.

Gino A Farina, MD, FACEP, FAAEM Associate Professor of Clinical Emergency Medicine, Albert Einstein College of Medicine; Program Director, Department of Emergency Medicine, Long Island Jewish Medical Center

Gino A Farina, MD, FACEP, FAAEM is a member of the following medical societies: American Academy of Emergency Medicine, American College of Emergency Physicians, and Society for Academic Emergency Medicine

Disclosure: Nothing to disclose.

Patrick M Foye, MD Associate Professor of Physical Medicine and Rehabilitation, Co-Director of Musculoskeletal Fellowship, Co-Director of Back Pain Clinic, Director of Coccyx Pain Service (Tailbone Pain Service: www.TailboneDoctor.com), University of Medicine and Dentistry of New Jersey, New Jersey Medical School

Patrick M Foye, MD is a member of the following medical societies: American Academy of Physical Medicine and Rehabilitation, American Association of Neuromuscular and Electrodiagnostic Medicine, Association of Academic Physiatrists, and International Spine Intervention Society

Disclosure: Nothing to disclose.

Harris Gellman, MD Consulting Surgeon, Broward Hand Center; Voluntary Clinical Professor of Orthopedic Surgery and Plastic Surgery, Departments of Orthopedic Surgery and Surgery, University of Miami, Leonard M Miller School of Medicine

Harris Gellman, MD is a member of the following medical societies: American Academy of Medical Acupuncture, American Academy of Orthopaedic Surgeons, American Orthopaedic Association, American Society for Surgery of the Hand, and Arkansas Medical Society

Disclosure: Nothing to disclose.

Elliot Goldberg, MD Dean of the Western Pennsylvania Clinical Campus, Professor, Department of Medicine, Temple University School of Medicine

Elliot Goldberg, MD is a member of the following medical societies: Alpha Omega Alpha, American College of Physicians, and American College of Rheumatology

Disclosure: Nothing to disclose.

Kavita Gupta, DO, MEng Department of Orthopedics, Center of Physical Medicine and Rehabilitation, University of Dentistry and Medicine of New Jersey

Kavita Gupta, DO, MEng is a member of the following medical societies: American Academy of Physical Medicine and Rehabilitation, American Osteopathic Association, Association of Academic Physiatrists, and Pennsylvania Medical Society

Disclosure: Nothing to disclose.

John A Kare, MD Assistant Professor of Emergency Medicine, Charles R Drew University of Medicine and Science/UCLA, Director of Research, Department of Emergency Medicine, Martin Luther King Jr/Charles R Drew Medical Center

John A Kare, MD is a member of the following medical societies: American Academy of Emergency Medicine, American Medical Student Association/Foundation, and Emergency Medicine Residents Association

Disclosure: Nothing to disclose.

Randall W King, MD, FACEP Assistant Clinical Professor of Emergency Medicine, The University of Toledo College of Medicine; Director, Emergency Medicine Residency Program, Associate Chair, Department of Emergency Medicine, St Vincent Mercy Medical Center

Randall W King, MD, FACEP is a member of the following medical societies: American College of Emergency Physicians, American Medical Association, Council of Emergency Medicine Residency Directors, Ohio State Medical Association, and Society for Academic Emergency Medicine

Disclosure: Challenger corporation None Physician Advisory Board; Ohio ACEP Consulting fee Editor Rivers review text Emergency Medicine

Milton J Klein, DO, MBA Consulting Physiatrist, Heritage Valley Health System-Sewickley Hospital and Ohio Valley General Hospital

Milton J Klein, DO, MBA is a member of the following medical societies: American Academy of Disability Evaluating Physicians, American Academy of Medical Acupuncture, American Academy of Osteopathy, American Academy of Physical Medicine and Rehabilitation, American Medical Association, American Osteopathic Association, American Osteopathic College of Physical Medicine and Rehabilitation, American Pain Society, and Pennsylvania Medical Society

Disclosure: Nothing to disclose.

Kristine M Lohr, MD, MS Professor, Department of Internal Medicine, Center for the Advancement of Women's Health and Division of Rheumatology, Director, Rheumatology Training Program, University of Kentucky College of Medicine

Kristine M Lohr, MD, MS is a member of the following medical societies: American College of Physicians and American College of Rheumatology

Disclosure: Nothing to disclose.

Robert J Nowinski, DO Clinical Assistant Professor of Orthopaedic Surgery, Ohio State University College of Medicine and Public Health, Ohio University College of Osteopathic Medicine; Private Practice, Orthopedic and Neurological Consultants, Inc, Columbus, Ohio

Robert J Nowinski, DO is a member of the following medical societies: American Academy of Orthopaedic Surgeons, American College of Osteopathic Surgeons, American Medical Association, American Osteopathic Association, Ohio Osteopathic Association, and Ohio State Medical Association

Disclosure: Tornier Grant/research funds Other; Tornier Honoraria Speaking and teaching

Robert E O'Connor, MD, MPH Professor and Chair, Department of Emergency Medicine, University of Virginia Health System

Robert E O'Connor, MD, MPH is a member of the following medical societies: American Academy of Emergency Medicine, American College of Emergency Physicians, American College of Physician Executives, American Heart Association, American Medical Association, Medical Society of Delaware, National Association of EMS Physicians, Society for Academic Emergency Medicine, and Wilderness Medical Society

Disclosure: Nothing to disclose.

Elizabeth Salt, ARPN, PhD Assistant Professor, Division of Rheumatology, University of Kentucky College of Nursing; Rheumatology Nurse Practitioner, University of Kentucky Chandler Medical Center

Elizabeth Scarbrough, MSN is a member of the following medical societies: American College of Rheumatology, Council for the Advancement of Nursing Science, and Sigma Theta Tau International

Disclosure: Nothing to disclose.

Roberto Sandoval, MD Consulting Staff, Department of Emergency Medicine, Anaheim Memorial Medical Center, La Palma Intercommunity Hospital

Roberto Sandoval, MD is a member of the following medical societies: American College of Emergency Physicians and American Medical Association

Disclosure: Nothing to disclose.

Joseph E Sheppard, MD Professor of Clinical Orthopedic Surgery, Chief of Hand and Upper Extremity Service, Department of Orthopedic Surgery, University of Arizona Health Sciences Center, University Physicians Healthcare

Joseph E Sheppard, MD is a member of the following medical societies: American Academy of Orthopaedic Surgeons, American Society for Surgery of the Hand, and Orthopaedics Overseas

Disclosure: Nothing to disclose.

Francisco Talavera, PharmD, PhD Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Medscape Salary Employment

Roman V Voytsekhovskiy, MD Fellow in Hand Surgery, Department of Orthopedic Surgery, Rush University Medical Center

Disclosure: Nothing to disclose.

Eleby R Washington III, MD, FACS Associate Professor, Department of Surgery, Division of Orthopedics, Charles R Drew University of Medicine and Science

Eleby R Washington III, MD, FACS is a member of the following medical societies: American Academy of Orthopaedic Surgeons, American College of Surgeons, American Medical Association, International College of Surgeons, and National Medical Association

Disclosure: Nothing to disclose.

Richard Worthington MD, Department of Emergency Medicine, Wood County Hospital

Richard Worthington is a member of the following medical societies: American College of Emergency Physicians, Ohio State Medical Association, and Society for Academic Emergency Medicine

Disclosure: Nothing to disclose.

References

  1. [Guideline] Aletaha D, Neogi T, Silman AJ, et al. 2010 Rheumatoid arthritis classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Arthritis Rheum. 2010 Sep. 62(9):2569-81. [View Abstract]
  2. [Guideline] Anderson J, Caplan L, Yazdany J, et al, for the American College of Rheumatology. Rheumatoid arthritis disease activity measures: American College of Rheumatology Recommendations for use in clinical practice. Arthritis Care Res (Hoboken). 2012. 64:640-7. [View Abstract]
  3. [Guideline] Felson DT, Smolen JS, Wells G, Zhang B, van Tuyl LH, et al. American College of Rheumatology/European League Against Rheumatism provisional definition of remission in rheumatoid arthritis for clinical trials. Arthritis Rheum. 2011 Mar. 63 (3):573-86. [View Abstract]
  4. [Guideline] Singh JA, Saag KG, et al. 2015 American College of Rheumatology Guideline for the Treatment of Rheumatoid Arthritis. http://dx.doi.org/10.1002/art.39480 (2015). Arthritis Care and Research. 2015. [View Abstract]
  5. [Guideline] Smolen JS, Landewé R, Bijlsma J, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2016 update. Ann Rheum Dis. 2017 Mar 6. 73(3):492-509. [View Abstract]
  6. Donahue KE, Jonas DE, Hansen RA, Roubey R, Jonas B, Lux LJ, et al. Agency for Healthcare Research and Quality. Choosing Medications for Rheumatoid Arthritis. 2012 Apr. [View Abstract]
  7. Kelly JC. Rheumatoid arthritis: Updated recommendations released. Medscape Medical News. Available at http://www.medscape.com/viewarticle/845495. May 28, 2015; Accessed: June 30, 2015.
  8. [Guideline] Smolen JS, Breedveld FC, Burmester GR, Bykerk V, Dougados M, et al. Treating rheumatoid arthritis to target: 2014 update of the recommendations of an international task force. Ann Rheum Dis. 2015 May 12. [View Abstract]
  9. Carlens C, Hergens MP, Grunewald J, et al. Smoking, use of moist snuff, and risk of chronic inflammatory diseases. Am J Respir Crit Care Med. 2010 Jun 1. 181(11):1217-22. [View Abstract]
  10. Barton A, Worthington J. Genetic susceptibility to rheumatoid arthritis: an emerging picture. Arthritis Rheum. 2009 Oct 15. 61(10):1441-6. [View Abstract]
  11. Begovich AB, Carlton VE, Honigberg LA, et al. A missense single-nucleotide polymorphism in a gene encoding a protein tyrosine phosphatase (PTPN22) is associated with rheumatoid arthritis. Am J Hum Genet. 2004 Aug. 75(2):330-7. [View Abstract]
  12. Potter C, Eyre S, Cope A, Worthington J, Barton A. Investigation of association between the TRAF family genes and RA susceptibility. Ann Rheum Dis. 2007 Oct. 66(10):1322-6. [View Abstract]
  13. Prakken B, Albani S, Martini A. Juvenile idiopathic arthritis. Lancet. 2011 Jun. 377(9783):2138-49. [View Abstract]
  14. Hinks A, Ke X, Barton A, Eyre S, Bowes J, Worthington J. Association of the IL2RA/CD25 gene with juvenile idiopathic arthritis. Arthritis Rheum. 2009 Jan. 60(1):251-7. [View Abstract]
  15. Ahlmen M, Svensson B, Albertsson K, Forslind K, Hafstrom I. Influence of gender on assessments of disease activity and function in early rheumatoid arthritis in relation to radiographic joint damage. Ann Rheum Dis. 2010 Jan. 69(1):230-3. [View Abstract]
  16. Areskoug-Josefsson K, Oberg U. A literature review of the sexual health of women with rheumatoid arthritis. Musculoskeletal Care. 2009 Dec. 7(4):219-26. [View Abstract]
  17. Martin-Trujillo A, van Rietschoten JG, Timmer TC, et al. Loss of imprinting of IGF2 characterises high IGF2 mRNA-expressing type of fibroblast-like synoviocytes in rheumatoid arthritis. Ann Rheum Dis. 2010 Jun. 69(6):1239-42. [View Abstract]
  18. Zhou X, Chen W, Swartz MD, et al. Joint linkage and imprinting analyses of GAW15 rheumatoid arthritis and gene expression data. BMC Proc. 2007. 1 Suppl 1:S53. [View Abstract]
  19. Barlow DP. Genomic imprinting: a mammalian epigenetic discovery model. Annu Rev Genet. 2011. 45:379-403. [View Abstract]
  20. Hitchon CA, Chandad F, Ferucci ED, et al. Antibodies to porphyromonas gingivalis are associated with anticitrullinated protein antibodies in patients with rheumatoid arthritis and their relatives. J Rheumatol. 2010 Jun. 37(6):1105-12. [View Abstract]
  21. Routsias JG, Goules JD, Goules A, Charalampakis G, Pikazis D. Autopathogenic correlation of periodontitis and rheumatoid arthritis. Rheumatology (Oxford). 2011 Jul. 50(7):1189-93. [View Abstract]
  22. Barrett JH, Brennan P, Fiddler M, Silman AJ. Does rheumatoid arthritis remit during pregnancy and relapse postpartum? Results from a nationwide study in the United Kingdom performed prospectively from late pregnancy. Arthritis Rheum. 1999 Jun. 42(6):1219-27. [View Abstract]
  23. Jorgensen KT, Pedersen BV, Jacobsen S, Biggar RJ, Frisch M. National cohort study of reproductive risk factors for rheumatoid arthritis in Denmark: a role for hyperemesis, gestational hypertension and pre-eclampsia?. Ann Rheum Dis. 2010 Feb. 69(2):358-63. [View Abstract]
  24. Guthrie KA, Dugowson CE, Voigt LF, Koepsell TD, Nelson JL. Does pregnancy provide vaccine-like protection against rheumatoid arthritis?. Arthritis Rheum. 2010 Jul. 62(7):1842-8. [View Abstract]
  25. Shah A, St. Clair EW. Rheumatoid Arthritis. In: Kasper DL, Fauci AS, Hauser SL, Longo DL, Jameson JL, Loscalzo J, Eds. Harrison's Principles of Internal Medicine. 19th ed. New York, NY: McGraw-Hill Education; 2016.
  26. Gremese E, Salaffi F, Bosello SL, et al. Very early rheumatoid arthritis as a predictor of remission: a multicentre real life prospective study. Ann Rheum Dis. 2013 Jun. 72(6):858-62. [View Abstract]
  27. Weinblatt ME, Keystone EC, Cohen MD, et al. Factors associated with radiographic progression in patients with rheumatoid arthritis who were treated with methotrexate. J Rheumatol. 2011 Feb. 38(2):242-6. [View Abstract]
  28. Agrawal S, Misra R, Aggarwal A. Autoantibodies in rheumatoid arthritis: association with severity of disease in established RA. Clin Rheumatol. 2007 Feb. 26(2):201-4. [View Abstract]
  29. Vencovsky J, Machacek S, Sedova L, et al. Autoantibodies can be prognostic markers of an erosive disease in early rheumatoid arthritis. Ann Rheum Dis. 2003 May. 62(5):427-30. [View Abstract]
  30. Sokka T, Kautiainen H, Mottonen T, Hannonen P. Work disability in rheumatoid arthritis 10 years after the diagnosis. J Rheumatol. 1999 Aug. 26(8):1681-5. [View Abstract]
  31. Mollard E, Pedro S, Chakravarty E, Clowse M, Schumacher R, Michaud K. The impact of menopause on functional status in women with rheumatoid arthritis. Rheumatology. 29 January 2018.
  32. Lindhardsen J, Ahlehoff O, Gislason GH, et al. Risk of atrial fibrillation and stroke in rheumatoid arthritis: Danish nationwide cohort study. BMJ. 2012. 344:e1257. [View Abstract]
  33. Hawley DJ. Psycho-educational interventions in the treatment of arthritis. Baillieres Clin Rheumatol. 1995 Nov. 9(4):803-23. [View Abstract]
  34. Tucker M, Kirwan JR. Does patient education in rheumatoid arthritis have therapeutic potential?. Ann Rheum Dis. 1991 Jun. 50 suppl 3:422-8. [View Abstract]
  35. Thompson A. Practical aspects of therapeutic intervention in rheumatoid arthritis. J Rheumatol Suppl. 2009 Jun. 82:39-41. [View Abstract]
  36. Komano Y, Harigai M, Koike R, Sugiyama H, Ogawa J, Saito K. Pneumocystis jiroveci pneumonia in patients with rheumatoid arthritis treated with infliximab: a retrospective review and case-control study of 21 patients. Arthritis Rheum. 2009 Mar 15. 61(3):305-12. [View Abstract]
  37. Aletaha D, Neogi T, Silman AJ, Funovits J, Felson DT, Bingham CO 3rd. 2010 rheumatoid arthritis classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Ann Rheum Dis. 2010 Sep. 69(9):1580-8. [View Abstract]
  38. Radner H, Neogi T, Smolen JS, Aletaha D. Performance of the 2010 ACR/EULAR classification criteria for rheumatoid arthritis: a systematic literature review. Ann Rheum Dis. 2014 Jan. 73 (1):114-23. [View Abstract]
  39. Katchamart W, Johnson S, Lin HJ, Phumethum V, Salliot C, Bombardier C. Predictors for remission in rheumatoid arthritis patients: A systematic review. Arthritis Care Res (Hoboken). 2010 Aug. 62(8):1128-43. [View Abstract]
  40. Varache S, Narbonne V, Jousse-Joulin S, et al. Is routine viral screening useful in patients with recent-onset polyarthritis of a duration of at least 6 weeks? Results from a nationwide longitudinal prospective cohort study. Arthritis Care Res (Hoboken). 2011 Nov. 63(11):1565-70. [View Abstract]
  41. Aletaha D, Alasti F, Smolen JS. Rheumatoid factor determines structural progression of rheumatoid arthritis dependent and independent of disease activity. Ann Rheum Dis. Jul 13 2012. [View Abstract]
  42. Scott IC, Steer S, Lewis CM, Cope AP. Precipitating and perpetuating factors of rheumatoid arthritis immunopathology: linking the triad of genetic predisposition, environmental risk factors and autoimmunity to disease pathogenesis. Best Pract Res Clin Rheumatol. 2011 Aug. 25(4):447-68. [View Abstract]
  43. Daha NA, Toes RE. Rheumatoid arthritis: Are ACPA-positive and ACPA-negative RA the same disease?. Nat Rev Rheumatol. 2011 Apr. 7(4):202-3. [View Abstract]
  44. van Venrooij WJ, van Beers JJ, Pruijn GJ. Anti-CCP antibodies: the past, the present and the future. Nat Rev Rheumatol. 2011 Jun 7. 7(7):391-8. [View Abstract]
  45. Mjaavatten MD, van der Heijde DM, Uhlig T, et al. Should Anti-citrullinated Protein Antibody and Rheumatoid Factor Status Be Reassessed During the First Year of Followup in Recent-Onset Arthritis? A Longitudinal Study. J Rheumatol. 2011 Nov. 38(11):2336-41. [View Abstract]
  46. Bang H, Egerer K, Gauliard A, et al. Mutation and citrullination modifies vimentin to a novel autoantigen for rheumatoid arthritis. Arthritis Rheum. 2007. 56(8):2503–11. [View Abstract]
  47. Coenen D, Verschueren P, Westhovens R, Bossuyt X. Technical and diagnostic performance of 6 assays for the measurement of citrullinated protein/peptide antibodies in the diagnosis of rheumatoid arthritis. Clin Chem. 2007. 53(3):498–504. [View Abstract]
  48. Soos L, Szekanecz Z, Szabo Z, et al. Clinical evaluation of anti-mutated citrullinated vimentin by ELISA in rheumatoid arthritis. J Rheumatol. 2007. 34(8):1658–63. [View Abstract]
  49. Szekanecz Z, Soos L, Szabo Z, et al. Anti-citrullinated protein antibodies in rheumatoid arthritis: as good as it gets?. Clin Rev Allergy Immunol. 2008. 34(1):26–31. [View Abstract]
  50. Goodman A. Novel biomarkers improve diagnosis in early RA. Medscape Medical News. June 20, 2013.
  51. De Winter L, Hansen W, Geusens P, et al. New autoantibodies as biomarkers for early and seronegative rheumatoid arthritis [abstract OP0181]. Presented at: EULAR 2013, the Annual Congress of the European League Against Rheumatism; Madrid, Spain; June 14, 2013. Ann Rheum Dis. 2013. 72(suppl 3):114.
  52. van der Heijde DM. Radiographic imaging: the ‘gold standard’ for assessment of disease progression in rheumatoid arthritis. Rheumatology (Oxford). 2000 Jun. 39 suppl 1:9-16. [View Abstract]
  53. Tan YK, Conaghan PG. Imaging in rheumatoid arthritis. Best Pract Res Clin Rheumatol. 2011 Aug. 25(4):569-84. [View Abstract]
  54. Wells AF, Haddad RH. Emerging role of ultrasonography in rheumatoid arthritis: optimizing diagnosis, measuring disease activity and identifying prognostic factors. Ultrasound Med Biol. 2011 Aug. 37(8):1173-84. [View Abstract]
  55. Bruno MA, Wakefield RJ. Chapter 5: Ultrasound of Rheumatoid Arthritis. Bruno MA, Mosher TJ, Gold GE. Arthritis in Color: Advanced Imaging of Arthritis. Philadelphia, PA: Saunders-Elsevier; 2009. 96-122.
  56. Cheung PP, Dougados M, Gossec L. Reliability of ultrasonography to detect synovitis in rheumatoid arthritis: a systematic literature review of 35 studies (1,415 patients). Arthritis Care Res (Hoboken). 2010 Mar. 62(3):323-34. [View Abstract]
  57. Fiocco U, Ferro F, Vezzu M, et al. Rheumatoid and psoriatic knee synovitis: clinical, grey scale, and power Doppler ultrasound assessment of the response to etanercept. Ann Rheum Dis. 2005 Jun. 64(6):899-905. [View Abstract]
  58. Suter LG, Fraenkel L, Braithwaite RS. Role of magnetic resonance imaging in the diagnosis and prognosis of rheumatoid arthritis. Arthritis Care Res (Hoboken). 2011 May. 63(5):675-88. [View Abstract]
  59. Cyteval C. Doppler ultrasonography and dynamic magnetic resonance imaging for assessment of synovitis in the hand and wrist of patients with rheumatoid arthritis. Semin Musculoskelet Radiol. 2009 Mar. 13(1):66-73. [View Abstract]
  60. Fukae J, Kon Y, Henmi M, Sakamoto F, Narita A, Shimizu M. Change of synovial vascularity in a single finger joint assessed by power doppler sonography correlated with radiographic change in rheumatoid arthritis: comparative study of a novel quantitative score with a semiquantitative score. Arthritis Care Res (Hoboken). 2010 May. 62(5):657-63. [View Abstract]
  61. Zayat AS, Conaghan PG, Sharif M, et al. Do non-steroidal anti-inflammatory drugs have a significant effect on detection and grading of ultrasound-detected synovitis in patients with rheumatoid arthritis? Results from a randomised study. Ann Rheum Dis. 2011 Oct. 70(10):1746-51. [View Abstract]
  62. Kelleher MO, McEvoy L, Yang JP, Kamel MH, Bolger C. Lateral mass screw fixation of complex spine cases: a prospective clinical study. Br J Neurosurg. 2008 Oct. 22(5):663-8. [View Abstract]
  63. Cakir B, Kafer W, Reichel H, Schmidt R. [Surgery of the cervical spine in rheumatoid arthritis. Diagnostics and indication]. Orthopade. 2008 Nov. 37(11):1127-40; quiz 1141. [View Abstract]
  64. Narvaez JA, Narvaez J, Serrallonga M, et al. Cervical spine involvement in rheumatoid arthritis: correlation between neurological manifestations and magnetic resonance imaging findings. Rheumatology (Oxford). 2008 Dec. 47(12):1814-9. [View Abstract]
  65. Verstappen SM, Albada-Kuipers GA, Bijlsma JW, et al, for the Utrecht Rheumatoid Arthritis Cohort Study Group (SRU). A good response to early DMARD treatment of patients with rheumatoid arthritis in the first year predicts remission during follow up. Ann Rheum Dis. 2005. 64:38-43. [View Abstract]
  66. Aletaha D, Funovits J, Keystone EC, Smolen JS. Disease activity early in the course of treatment predicts response to therapy after one year in rheumatoid arthritis patients. Arthritis Rheum. 2007. 56:3226-35. [View Abstract]
  67. Verschueren P, Esselens G, Westhovens R. Predictors of remission, normalized physical function, and changes in the working situation during follow-up of patients with early rheumatoid arthritis: an observational study. Scand J Rheumatol. 2009. 38:166-72. [View Abstract]
  68. [Guideline] Singh JA, Furst DE, Bharat A, Curtis JR, Kavanaugh AF, et al. 2012 update of the 2008 American College of Rheumatology recommendations for the use of disease-modifying antirheumatic drugs and biologic agents in the treatment of rheumatoid arthritis. Arthritis Care Res (Hoboken). 2012 May. 64 (5):625-39. [View Abstract]
  69. Ince-Askan H, Dolhain RJ. Pregnancy and rheumatoid arthritis. Best Pract Res Clin Rheumatol. 2015 Aug-Dec. 29 (4-5):580-96. [View Abstract]
  70. Ostensen M, Forger F, Nelson JL, Schuhmacher A, Hebisch G, Villiger PM. Pregnancy in patients with rheumatic disease: anti-inflammatory cytokines increase in pregnancy and decrease post partum. Ann Rheum Dis. 2005 Jun. 64(6):839-44. [View Abstract]
  71. Makol A, Wright K, Amin S. Rheumatoid arthritis and pregnancy: safety considerations in pharmacological management. Drugs. 2011 Oct 22. 71(15):1973-87. [View Abstract]
  72. Parke A, West B. Hydroxychloroquine in pregnant patients with systemic lupus erythematosus. J Rheumatol. 1996 Oct. 23(10):1715-8. [View Abstract]
  73. Temprano KK, Bandlamudi R, Moore TL. Antirheumatic drugs in pregnancy and lactation. Semin Arthritis Rheum. 2005 Oct. 35(2):112-21. [View Abstract]
  74. Androulakis I, Zavos C, Christopoulos P, Mastorakos G, Gazouli M. Safety of anti-tumor necrosis factor therapy during pregnancy in patients with inflammatory bowel disease. World J Gastroenterol. 2015 Dec 21. 21 (47):13205-11. [View Abstract]
  75. Bröms G, Granath F, Ekbom A, Hellgren K, Pedersen L, Sørensen HT, et al. Low Risk of Birth Defects for Infants Whose Mothers Are Treated With Anti-Tumor Necrosis Factor Agents During Pregnancy. Clin Gastroenterol Hepatol. 2016 Feb. 14 (2):234-41.e1-5. [View Abstract]
  76. Luqmani R, Hennell S, Estrach C, et al. British Society for Rheumatology and British Health Professionals in Rheumatology guideline for the management of rheumatoid arthritis (after the first 2 years). Rheumatology (Oxford). 2009 Apr. 48(4):436-9. [View Abstract]
  77. Goksel Karatepe A, Gunaydin R, Turkmen G, Kaya T. Effects of home-based exercise program on the functional status and the quality of life in patients with rheumatoid arthritis: 1-year follow-up study. Rheumatol Int. 2011 Feb. 31(2):171-6. [View Abstract]
  78. Kamioka H, Tsutani K, Okuizumi H, Mutoh Y, Ohta M, Handa S. Effectiveness of aquatic exercise and balneotherapy: a summary of systematic reviews based on randomized controlled trials of water immersion therapies. J Epidemiol. 2010. 20(1):2-12. [View Abstract]
  79. Katz P, Margaretten M, Gregorich S, Trupin L. Physical Activity to Reduce Fatigue in Rheumatoid Arthritis: A Randomized, Controlled Trial. Arthritis Care Res (Hoboken). 2017 Apr 5. [View Abstract]
  80. Lemmey AB, Marcora SM, Chester K, Wilson S, Casanova F, Maddison PJ. Effects of high-intensity resistance training in patients with rheumatoid arthritis: a randomized controlled trial. Arthritis Rheum. 2009 Dec 15. 61(12):1726-34. [View Abstract]
  81. O'Brien ET. Surgical principles and planning for the rheumatoid hand and wrist. Clin Plast Surg. 1996 Jul. 23(3):407-20. [View Abstract]
  82. Macedo AM, Oakley SP, Panayi GS, Kirkham BW. Functional and work outcomes improve in patients with rheumatoid arthritis who receive targeted, comprehensive occupational therapy. Arthritis Rheum. 2009 Nov 15. 61(11):1522-30. [View Abstract]
  83. Williams SB, Brand CA, Hill KD, Hunt SB, Moran H. Feasibility and outcomes of a home-based exercise program on improving balance and gait stability in women with lower-limb osteoarthritis or rheumatoid arthritis: a pilot study. Arch Phys Med Rehabil. 2010 Jan. 91(1):106-14. [View Abstract]
  84. Callhoff J, Weiss A, Zink A, Listing J. Impact of biologic therapy on functional status in patients with rheumatoid arthritis--a meta-analysis. Rheumatology (Oxford). 2013 Dec. 52(12):2127-35. [View Abstract]
  85. Bili A, Sartorius JA, Kirchner HL, et al. Hydroxychloroquine use and decreased risk of diabetes in rheumatoid arthritis patients. J Clin Rheumatol. 2011 Apr. 17(3):115-20. [View Abstract]
  86. Solomon DH, Massarotti E, Garg R, et al. Association between disease-modifying antirheumatic drugs and diabetes risk in patients with rheumatoid arthritis and psoriasis. JAMA. 2011 Jun 22. 305(24):2525-31. [View Abstract]
  87. Brooks M. FDA OKs Methotrexate Autoinjector (Otrexup). Medscape Medical News. Oct 18 2013.
  88. Glen S Hazlewood, Cheryl Barnabe, George Tomlinson, Deborah Marshall, Dan Devoe, Claire Bombardier. 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:[View Abstract]
  89. Furst DE, Breedveld FC, Kalden JR, et al. Updated consensus statement on biological agents for the treatment of rheumatic diseases, 2007. Ann Rheum Dis. 2007 Nov. 66 Suppl 3:iii2-22. [View Abstract]
  90. Garces S, Demengeot J, Benito-Garcia E. The immunogenicity of anti-TNF therapy in immune-mediated inflammatory diseases: a systematic review of the literature with a meta-analysis. Ann Rheum Dis. 2013 Dec. 72(12):1947-55. [View Abstract]
  91. Galloway JB, Hyrich KL, Mercer LK, et al. Risk of septic arthritis in patients with rheumatoid arthritis and the effect of anti-TNF therapy: results from the British Society for Rheumatology Biologics Register. Ann Rheum Dis. 2011 Oct. 70(10):1810-1814. [View Abstract]
  92. Lan JL, Chen YM, Hsieh TY, et al. Kinetics of viral loads and risk of hepatitis B virus reactivation in hepatitis B core antibody-positive rheumatoid arthritis patients undergoing anti-tumour necrosis factor alpha therapy. Ann Rheum Dis. 2011 Oct. 70(10):1719-25. [View Abstract]
  93. Askling J, van Vollenhoven RF, Granath F, et al. Cancer risk in patients with rheumatoid arthritis treated with anti-tumor necrosis factor alpha therapies: does the risk change with the time since start of treatment?. Arthritis Rheum. 2009 Nov. 60(11):3180-9. [View Abstract]
  94. Finzel S, Rech J, Schmidt S, et al. Repair of bone erosions in rheumatoid arthritis treated with tumour necrosis factor inhibitors is based on bone apposition at the base of the erosion. Ann Rheum Dis. 2011 Sep. 70(9):1587-93. [View Abstract]
  95. van Vollenhoven RF, Ernestam S, Geborek P, Petersson IF, Coster L, Waltbrand E. Addition of infliximab compared with addition of sulfasalazine and hydroxychloroquine to methotrexate in patients with early rheumatoid arthritis (Swefot trial): 1-year results of a randomised trial. Lancet. 2009 Aug 8. 374(9688):459-66. [View Abstract]
  96. Visser K, van der Heijde D. Optimal dosage and route of administration of methotrexate in rheumatoid arthritis: a systematic review of the literature. Ann Rheum Dis. 2009 Jul. 68(7):1094-9. [View Abstract]
  97. Keystone EC, Kavanaugh A, Weinblatt ME, Patra K, Pangan AL. Clinical consequences of delayed addition of adalimumab to methotrexate therapy over 5 years in patients with rheumatoid arthritis. J Rheumatol. 2011 May. 38(5):855-62. [View Abstract]
  98. Kelly J. Rheumatoid Arthritis: Target Adalimumab Level Determined. Medscape Medical News. Available at http://www.medscape.com/viewarticle/818102. Accessed: December 23, 2013.
  99. Pouw MF, Krieckaert CL, Nurmohamed MT, et al. Key findings towards optimising adalimumab treatment: the concentration-effect curve. Ann Rheum Dis. 2015 Mar. 74(3):513-8. [View Abstract]
  100. Fleischmann R, Vencovsky J, van Vollenhoven RF, Borenstein D, Box J, Coteur G. Efficacy and safety of certolizumab pegol monotherapy every 4 weeks in patients with rheumatoid arthritis failing previous disease-modifying antirheumatic therapy: the FAST4WARD study. Ann Rheum Dis. 2009 Jun. 68(6):805-11. [View Abstract]
  101. Smolen J, Landewe RB, Mease P, Brzezicki J, Mason D, Luijtens K. Efficacy and safety of certolizumab pegol plus methotrexate in active rheumatoid arthritis: the RAPID 2 study. A randomised controlled trial. Ann Rheum Dis. 2009 Jun. 68(6):797-804. [View Abstract]
  102. Emery P, Fleischmann RM, Moreland LW, Hsia EC, Strusberg I, Durez P. Golimumab, a human anti-tumor necrosis factor alpha monoclonal antibody, injected subcutaneously every four weeks in methotrexate-naive patients with active rheumatoid arthritis: twenty-four-week results of a phase III, multicenter, randomized, double-blind, placebo-controlled study of golimumab before methotrexate as first-line therapy for early-onset rheumatoid arthritis. Arthritis Rheum. 2009 Aug. 60(8):2272-83. [View Abstract]
  103. Brown T. FDA approves intravenous golimumab (Simponi Aria) for rheumatoid arthritis. Medscape Medical News. July 18, 2013.
  104. Janssen Biotech, Inc. Simponi Aria (golimumab) for infusion receives FDA approval for treatment of moderately to severely active rheumatoid arthritis [press release]. July 18, 2013.
  105. Weinblatt ME, Bingham CO 3rd, Mendelsohn AM, et al. Intravenous golimumab is effective in patients with active rheumatoid arthritis despite methotrexate therapy with responses as early as week 2: results of the phase 3, randomised, multicentre, double-blind, placebo-controlled GO-FURTHER trial. Ann Rheum Dis. 2013 Mar. 72(3):381-9. [View Abstract]
  106. Edwards JC, Szczepanski L, Szechinski J, Filipowicz-Sosnowska A, Emery P, Close DR. Efficacy of B-cell-targeted therapy with rituximab in patients with rheumatoid arthritis. N Engl J Med. 2004 Jun 17. 350(25):2572-81. [View Abstract]
  107. Peterfy C, Emery P, Tak PP, Østergaard M, DiCarlo J, Otsa K, et al. MRI assessment of suppression of structural damage in patients with rheumatoid arthritis receiving rituximab: results from the randomised, placebo-controlled, double-blind RA-SCORE study. Ann Rheum Dis. 2016 Jan. 75 (1):170-7. [View Abstract]
  108. Emery P, Gottenberg JE, Rubbert-Roth A, et al. Rituximab versus an alternative TNF inhibitor in patients with rheumatoid arthritis who failed to respond to a single previous TNF inhibitor: SWITCH-RA, a global, observational, comparative effectiveness study. Ann Rheum Dis. 2015 Jun. 74(6):979-84. [View Abstract]
  109. Porter D, van Melckebeke J, Dale J, Messow CM, McConnachie A, Walker A, et al. Tumour necrosis factor inhibition versus rituximab for patients with rheumatoid arthritis who require biological treatment (ORBIT): an open-label, randomised controlled, non-inferiority, trial. Lancet. 2016 Jul 16. 388 (10041):239-47. [View Abstract]
  110. Bingham CO 3rd, Looney RJ, Deodhar A, Halsey N, Greenwald M, Codding C. Immunization responses in rheumatoid arthritis patients treated with rituximab: results from a controlled clinical trial. Arthritis Rheum. 2010 Jan. 62(1):64-74. [View Abstract]
  111. Orencia (abatacept) [package insert]. Princeton, NJ: Bristol-Myers Squibb. 2011. Available at
  112. Genovese MC, Schiff M, Luggen M, et al. Longterm safety and efficacy of abatacept through 5 years of treatment in patients with rheumatoid arthritis and an inadequate response to tumor necrosis factor inhibitor therapy. J Rheumatol. Aug 2012. 39(8):1546-54. [View Abstract]
  113. Weinblatt ME, Schiff M, Valente R, et al. Head-to-head comparison of subcutaneous abatacept versus adalimumab for rheumatoid arthritis: Findings of a phase IIIb, multinational, prospective, randomized study. Arthritis Rheum. 2013 Jan. 65(1):28-38. [View Abstract]
  114. Dougados M, Kissel K, Sheeran T, et al. Adding tocilizumab or switching to tocilizumab monotherapy in methotrexate inadequate responders: 24-week symptomatic and structural results of a 2-year randomised controlled strategy trial in rheumatoid arthritis (ACT-RAY). Ann Rheum Dis. Jul 7 2012. [View Abstract]
  115. Strand V, Burmester GR, Ogale S, Devenport J, John A, Emery P. Improvements in health-related quality of life after treatment with tocilizumab in patients with rheumatoid arthritis refractory to tumour necrosis factor inhibitors: results from the 24-week randomized controlled RADIATE study. Rheumatology (Oxford). Jun 28 2012. [View Abstract]
  116. Burmester GR, Rubbert-Roth A, Cantagrel A, et al. A randomised, double-blind, parallel-group study of the safety and efficacy of subcutaneous tocilizumab versus intravenous tocilizumab in combination with traditional disease-modifying antirheumatic drugs in patients with moderate to severe rheumatoid arthritis (SUMMACTA study). Ann Rheum Dis. 2014 Jan. 73(1):69-74. [View Abstract]
  117. Smolen JS, Schoels MM, Nishimoto N, et al. Consensus statement on blocking the effects of interleukin-6 and in particular by interleukin-6 receptor inhibition in rheumatoid arthritis and other inflammatory conditions. Ann Rheum Dis. 2013 Apr. 72(4):482-92. [View Abstract]
  118. Genovese MC, Fleischmann R, Kivitz AJ, Rell-Bakalarska M, Martincova R, Fiore S, et al. Sarilumab Plus Methotrexate in Patients With Active Rheumatoid Arthritis and Inadequate Response to Methotrexate: Results of a Phase III Study. Arthritis Rheumatol. 2015 Jun. 67 (6):1424-37. [View Abstract]
  119. Fleischmann R, van Adelsberg J, Lin Y, Castelar-Pinheiro GD, Brzezicki J, Hrycaj P, et al. Sarilumab and Nonbiologic Disease-Modifying Antirheumatic Drugs in Patients With Active Rheumatoid Arthritis and Inadequate Response or Intolerance to Tumor Necrosis Factor Inhibitors. Arthritis Rheumatol. 2017 Feb. 69 (2):277-290. [View Abstract]
  120. Fleischmann R. Novel small-molecular therapeutics for rheumatoid arthritis. Curr Opin Rheumatol. 2012 May. 24(3):335-41. [View Abstract]
  121. FDA approves Xeljanz for rheumatoid arthritis [press release]. November 6, 2012. Available at http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm327152.htm. Accessed: November 28, 2012.
  122. van der Heijde D, Tanaka Y, Fleischmann R, et al; ORAL Scan Investigators. Tofacitinib (CP-690,550) in patients with rheumatoid arthritis receiving methotrexate: twelve-month data from a twenty-four-month phase III randomized radiographic study. Arthritis Rheum. 2013 Mar. 65(3):559-70. [View Abstract]
  123. Fleischmann R, Kremer J, Cush J, et al. Placebo-controlled trial of tofacitinib monotherapy in rheumatoid arthritis. N Engl J Med. Aug 9 2012. 367(6):495-507. [View Abstract]
  124. van Vollenhoven RF, Fleischmann R, Cohen S, et al. Tofacitinib or adalimumab versus placebo in rheumatoid arthritis. N Engl J Med. Aug 9 2012. 367(6):508-19. [View Abstract]
  125. Brown T. FDA Approves Baricitinib for Rheumatoid Arthritis. Medscape Medical News. 2018 Jun 01. Available at https://www.medscape.com/viewarticle/897516
  126. Dougados M, van der Heijde D, Chen YC, Greenwald M, Drescher E, Liu J, et al. Baricitinib in patients with inadequate response or intolerance to conventional synthetic DMARDs: results from the RA-BUILD study. Ann Rheum Dis. 2017 Jan. 76 (1):88-95. [View Abstract]
  127. van der Heijde D, Dougados M, Chen YC, Greenwald M, Drescher E, Klar R, et al. Effects of baricitinib on radiographic progression of structural joint damage at 1 year in patients with rheumatoid arthritis and an inadequate response to conventional synthetic disease-modifying antirheumatic drugs. RMD Open. 2018. 4 (1):e000662. [View Abstract]
  128. Smolen JS, Kremer JM, Gaich CL, DeLozier AM, Schlichting DE, Xie L, et al. Patient-reported outcomes from a randomised phase III study of baricitinib in patients with rheumatoid arthritis and an inadequate response to biological agents (RA-BEACON). Ann Rheum Dis. 2017 Apr. 76 (4):694-700. [View Abstract]
  129. Tosh JC, Wailoo AJ, Scott DL, Deighton CM. Cost-effectiveness of combination nonbiologic disease-modifying antirheumatic drug strategies in patients with early rheumatoid arthritis. J Rheumatol. 2011 Aug. 38(8):1593-600. [View Abstract]
  130. Lipsky PE, van der Heijde DM, St Clair EW, et al. Infliximab and methotrexate in the treatment of rheumatoid arthritis. Anti-Tumor Necrosis Factor Trial in Rheumatoid Arthritis with Concomitant Therapy Study Group. N Engl J Med. 2000 Nov 30. 343(22):1594-602. [View Abstract]
  131. Rigby W, Ferraccioli G, Greenwald M, et al. Effect of rituximab on physical function and quality of life in patients with rheumatoid arthritis previously untreated with methotrexate. Arthritis Care Res (Hoboken). 2011 May. 63(5):711-20. [View Abstract]
  132. O'Dell JR, Haire CE, Erikson N, et al. Treatment of rheumatoid arthritis with methotrexate alone, sulfasalazine and hydroxychloroquine, or a combination of all three medications. N Engl J Med. 1996 May 16. 334(20):1287-91. [View Abstract]
  133. Jones SK. Ocular toxicity and hydroxychloroquine: guidelines for screening. Br J Dermatol. 1999 Jan. 140(1):3-7. [View Abstract]
  134. Bongartz T, Sutton AJ, Sweeting MJ, Buchan I, Matteson EL, Montori V. Anti-TNF antibody therapy in rheumatoid arthritis and the risk of serious infections and malignancies: systematic review and meta-analysis of rare harmful effects in randomized controlled trials. JAMA. 2006 May 17. 295(19):2275-85. [View Abstract]
  135. Sohl S, Renner R, Winter U, et al. [Drug-induced lupus erythematosus tumidus during treatment with adalimumab]. Hautarzt. 2009 Oct. 60(10):826-9. [View Abstract]
  136. Ramos-Casals M, Brito-Zeron P, Soto MJ, Cuadrado MJ, Khamashta MA. Autoimmune diseases induced by TNF-targeted therapies. Best Pract Res Clin Rheumatol. 2008 Oct. 22(5):847-61. [View Abstract]
  137. Lunt M, Watson KD, Dixon WG, Symmons DP, Hyrich KL. No evidence of association between anti-tumor necrosis factor treatment and mortality in patients with rheumatoid arthritis: results from the British Society for Rheumatology Biologics Register. Arthritis Rheum. 2010 Nov. 62(11):3145-53. [View Abstract]
  138. Thompson AE, Rieder SW, Pope JE. Tumor necrosis factor therapy and the risk of serious infection and malignancy in patients with early rheumatoid arthritis: A meta-analysis of randomized controlled trials. Arthritis Rheum. 2011 Jun. 63(6):1479-85. [View Abstract]
  139. Mariette X, Matucci-Cerinic M, Pavelka K, et al. Malignancies associated with tumour necrosis factor inhibitors in registries and prospective observational studies: a systematic review and meta-analysis. Ann Rheum Dis. 2011 Nov. 70(11):1895-904. [View Abstract]
  140. Hoes JN, Jacobs JW, Buttgereit F, Bijlsma JW. Current view of glucocorticoid co-therapy with DMARDs in rheumatoid arthritis. Nat Rev Rheumatol. 2010 Dec. 6(12):693-702. [View Abstract]
  141. Buttgereit F, Doering G, Schaeffler A, et al. Efficacy of modified-release versus standard prednisone to reduce duration of morning stiffness of the joints in rheumatoid arthritis (CAPRA-1): a double-blind, randomised controlled trial. Lancet. Jan 19 2008. 371(9608):205-14. [View Abstract]
  142. Buttgereit F, Doering G, Schaeffler A, et al. Targeting pathophysiological rhythms: prednisone chronotherapy shows sustained efficacy in rheumatoid arthritis. Ann Rheum Dis. Jul 2010. 69(7):1275-80. [View Abstract]
  143. Buttgereit F, Mehta D, Kirwan J, et al. Low-dose prednisone chronotherapy for rheumatoid arthritis: a randomised clinical trial (CAPRA-2). Ann Rheum Dis. 2013 Feb. 72(2):204-10. [View Abstract]
  144. Boggs W. Infliximab, steroids offer similar RA remission rates. Medscape Medical News. August 28, 2013.
  145. Nam JL, Villeneuve E, Hensor EM, et al. Remission induction comparing infliximab and high-dose intravenous steroid, followed by treat-to-target: a double-blind, randomised, controlled trial in new-onset, treatment-naive, rheumatoid arthritis (the IDEA study). Ann Rheum Dis. 2014 Jan. 73(1):75-85. [View Abstract]
  146. Myasoedova E, Crowson CS, Nicola PJ, et al. The influence of rheumatoid arthritis disease characteristics on heart failure. J Rheumatol. 2011 Aug. 38(8):1601-6. [View Abstract]
  147. Solomon SD, Wittes J, Finn PV, et al, for the Cross Trial Safety Assessment Group. Cardiovascular risk of celecoxib in 6 randomized placebo-controlled trials: the cross trial safety analysis. Circulation. 2008 Apr 22. 117(16):2104-13. [View Abstract]
  148. Weinblatt ME, Kavanaugh A, Genovese MC, Musser TK, Grossbard EB, Magilavy DB. An oral spleen tyrosine kinase (Syk) inhibitor for rheumatoid arthritis. N Engl J Med. 2010 Sep. 363(14):1303-12. [View Abstract]
  149. [Guideline] Combe B, Landewe R, Daien CI, et al. 2016 update of the EULAR recommendations for the management of early arthritis. Ann Rheum Dis. 2016 Dec 15. [View Abstract]
  150. Nordberg LB, Lillegraven S, Lie E, Aga AB, Olsen IC, Hammer HB, et al. Patients with seronegative RA have more inflammatory activity compared with patients with seropositive RA in an inception cohort of DMARD-naïve patients classified according to the 2010 ACR/EULAR criteria. Ann Rheum Dis. 2017 Feb. 76 (2):341-345. [View Abstract]
  151. Ahlmen M, Svensson B, Albertsson K, Forslind K, Hafstrom I, BARFOT Study Group. Influence of gender on assessments of disease activity and function in early rheumatoid arthritis in relation to radiographic joint damage. Ann Rheum Dis. 2010 Jan. 69(1):230-3. [View Abstract]
  152. Kineret [package insert] [package insert]. Amgen. Personal communication with Kijung Sung-Thay, PharmD. 2008.
  153. Axelsen MB, Eshed I, Horslev-Petersen K, et al; OPERA study group. A treat-to-target strategy with methotrexate and intra-articular triamcinolone with or without adalimumab effectively reduces MRI synovitis, osteitis and tenosynovitis and halts structural damage progression in early rheumatoid arthritis: results from the OPERA randomised controlled trial. Ann Rheum Dis. 2015 May. 74(5):867-75. [View Abstract]
  154. Callhoff J, Weiss A, Zink A, Listing J. Impact of biologic therapy on functional status in patients with rheumatoid arthritis--a meta-analysis. Rheumatology (Oxford). 2013 Dec. 52(12):2127-35. [View Abstract]
  155. Chambers CD, Johnson DL, Luo Y, Xu R, Jones KL. Pregnancy outcomes in women exposed to adalimumab: an update on the OTIS Autoimmune Diseases in Pregnancy project. American College of Rheumatology. Available at http://acrabstracts.org/abstract/pregnancy-outcome-in-women-treated-with-adalimumab-for-the-treatment-of-rheumatoid-arthritis-an-update/. 2014 ACR/ARHP Annual Meeting.Abstract Number: 821; Accessed: April 6, 2017.
  156. Smolen JS, Pangan AL, Emery P, Rigby W, Tanaka Y, Vargas JI, et al. Upadacitinib as monotherapy in patients with active rheumatoid arthritis and inadequate response to methotrexate (SELECT-MONOTHERAPY): a randomised, placebo-controlled, double-blind phase 3 study. Lancet. 2019 Jun 8. 393 (10188):2303-2311. [View Abstract]
  157. Fleischmann RM, Genovese MC, Enejosa JV, Mysler E, Bessette L, Peterfy C, et al. Safety and effectiveness of upadacitinib or adalimumab plus methotrexate in patients with rheumatoid arthritis over 48 weeks with switch to alternate therapy in patients with insufficient response. Ann Rheum Dis. 2019 Jul 30. [View Abstract]

Rheumatoid changes in the hand. Photograph by David Effron MD, FACEP.

This gross photo shows destruction of the cartilage and erosion of the underlying bone with pannus from a patient with rheumatoid arthritis.

Boutonniere deformity.

Rheumatoid changes in the hand. Photograph by David Effron MD, FACEP.

Subluxation in the metacarpophalangeal joints, with ulnar deviation, in a patient with rheumatoid arthritis of the hands.

Coronal, T1-weighted magnetic resonance imaging scan shows characteristic pannus and erosive changes in the wrist in a patient with active rheumatoid arthritis. Courtesy of J. Tehranzadeh, MD, University of California at Irvine.

Rheumatoid nodules at the elbow. Photograph by David Effron MD, FACEP.

Ankylosis in the cervical spine at several levels due to long-standing juvenile rheumatoid arthritis (also known as juvenile idiopathic arthritis).

Lateral view of the cervical spine in a patient with rheumatoid arthritis shows erosion of the odontoid process.

Soft-tissue swelling and early erosions in the proximal interphalangeal joints in a patient with rheumatoid arthritis of the hands.

Anteroposterior radiograph of the knee shows uniform joint-space loss in the medial and lateral knee compartments without osteophytosis. A Baker cyst is seen medially (arrowhead).

Widespread osteopenia, carpal crowding (due to cartilage loss), and several erosions affecting the carpal bones and metacarpal heads in particular in a child with advanced juvenile rheumatoid arthritis (also known as juvenile idiopathic arthritis).

Lateral view of the cervical spine in a patient with rheumatoid arthritis shows erosion of the odontoid process.

Anteroposterior radiograph of the knee shows uniform joint-space loss in the medial and lateral knee compartments without osteophytosis. A Baker cyst is seen medially (arrowhead).

T1-weighted sagittal magnetic resonance image of the cervical spine shows basilar invagination with cranial migration of an eroded odontoid peg. There is minimal pannus. The tip of the peg indents the medulla, and there is narrowing of the foramen magnum due to the presence of the peg. Inflammatory fusion of several cervical vertebral bodies is shown.

Sagittal T2-weighted magnetic resonance image of cervical spine in same patient as in previous image. Compromised foramen magnum is easily appreciated, and there is increased signal intensity within upper cord; this is consistent with compressive myelomalacia. Further narrowing of canal is seen at multiple levels.

Ultrasonography-guided synovial biopsy of the second metacarpophalangeal joint of the right hand in a patient with rheumatoid arthritis of the hands. The biopsy needle is seen as a straight echogenic line on the left side of the image in an oblique orientation.

The hallmark of rheumatoid arthritis is a perivascular mononuclear cell infiltrate in the synovium (pictured here). The early stages are noted to have plasma cells as well, and syphilis needs to be part of the differential diagnosis.

The inflammation involved in rheumatoid arthritis can be intense. It is composed of mononuclear cells and can resemble a pseudosarcoma.

Ankylosis in the cervical spine at several levels due to long-standing juvenile rheumatoid arthritis (also known as juvenile idiopathic arthritis).

Widespread osteopenia, carpal crowding (due to cartilage loss), and several erosions affecting the carpal bones and metacarpal heads in particular in a child with advanced juvenile rheumatoid arthritis (also known as juvenile idiopathic arthritis).

Rheumatoid changes in the hand. Photograph by David Effron MD, FACEP.

Rheumatoid nodules at the elbow. Photograph by David Effron MD, FACEP.

Soft-tissue swelling and early erosions in the proximal interphalangeal joints in a patient with rheumatoid arthritis of the hands.

Subluxation in the metacarpophalangeal joints, with ulnar deviation, in a patient with rheumatoid arthritis of the hands.

Coronal, T1-weighted magnetic resonance imaging scan shows characteristic pannus and erosive changes in the wrist in a patient with active rheumatoid arthritis. Courtesy of J. Tehranzadeh, MD, University of California at Irvine.

Lateral view of the cervical spine in a patient with rheumatoid arthritis shows erosion of the odontoid process.

Boutonniere deformity.

Anteroposterior radiograph of the knee shows uniform joint-space loss in the medial and lateral knee compartments without osteophytosis. A Baker cyst is seen medially (arrowhead).

Ultrasonography-guided synovial biopsy of the second metacarpophalangeal joint of the right hand in a patient with rheumatoid arthritis of the hands. The biopsy needle is seen as a straight echogenic line on the left side of the image in an oblique orientation.

Plain lateral radiograph of the normal cervical spine taken in extension shows measurement of anterior atlantodental interval (yellow line) and posterior atlantodental interval (red line).

Lateral flexion view of the cervical spine shows atlantoaxial subluxation.

Lateral view of the cervical spine in a patient with rheumatoid arthritis shows erosion of the odontoid process.

T1-weighted sagittal magnetic resonance image of the cervical spine shows basilar invagination with cranial migration of an eroded odontoid peg. There is minimal pannus. The tip of the peg indents the medulla, and there is narrowing of the foramen magnum due to the presence of the peg. Inflammatory fusion of several cervical vertebral bodies is shown.

Sagittal T2-weighted magnetic resonance image of cervical spine in same patient as in previous image. Compromised foramen magnum is easily appreciated, and there is increased signal intensity within upper cord; this is consistent with compressive myelomalacia. Further narrowing of canal is seen at multiple levels.

Lateral radiograph of the same patient as in Images 4-5. Midcervical vertebral-body fusions are shown. The eroded peg is difficult to visualize, but inferior subluxation of the anterior arch of C1 is shown.

Lateral radiograph of a normal cervical spine shows the McGregor line. The odontoid tip should not protrude more than 4.5 mm above the line, which is drawn from the posterior edge of the hard palate to the most caudal point of the occiput.

Normal lateral magnified radiograph of the cervical spine shows the Ranawat method of detection of cranial settling. This method is used to measure the distance from the center of the pedicles (sclerotic ring) of C2 to a line drawn connecting the midpoints of the anterior and posterior arches of C1. (Normal values are 15 mm or greater for males and 13 mm or greater for females.)

Lateral radiograph of the cervical spine shows how the cervical height index (CHI) is calculated. The distance from the center of the sclerotic ring of C2 to the tip of the spinous process of C2 (dotted line) is measured. This is then divided into the distance from the center of the sclerotic ring of C2 to the midpoint of the inferior border of the body of C7. A CHI of less than 2 mm is a sensitive predictor of neurologic deficit.

X-ray shows total hip replacement, with prosthesis, in patient with osteoarthritis.

This gross photo shows destruction of the cartilage and erosion of the underlying bone with pannus from a patient with rheumatoid arthritis.

The hallmark of rheumatoid arthritis is a perivascular mononuclear cell infiltrate in the synovium (pictured here). The early stages are noted to have plasma cells as well, and syphilis needs to be part of the differential diagnosis.

The inflammation involved in rheumatoid arthritis can be intense. It is composed of mononuclear cells and can resemble a pseudosarcoma.

A 72-year-old man with long-standing rheumatoid arthritis (RA) developed blue-grayish discoloration of his skin. He had been on hydroxychloroquine for approximately 15 years. The diagnosis was hydroxychloroquine-related hyperpigmentation. Image courtesy of Jason Kolfenbach, MD, and Kevin Deane, MD, Division of Rheumatology, University of Colorado Denver School of Medicine.

A 64-year-old woman with rheumatoid arthritis has developed nodules on the dorsal and volar aspect of her fingers, as well as the posterior aspect of her heels. The diagnosis is RA nodules with methotrexate-induced accelerated nodulosis. Image courtesy of Jason Kolfenbach, MD, and Kevin Deane, MD, Division of Rheumatology, University of Colorado Denver School of Medicine.

A 64-year-old woman with rheumatoid arthritis has developed nodules on the dorsal and volar aspect of her fingers, as well as the posterior aspect of her heels. The diagnosis is RA nodules with methotrexate-induced accelerated nodulosis. Image courtesy of Jason Kolfenbach, MD, and Kevin Deane, MD, Division of Rheumatology, University of Colorado Denver School of Medicine.

Disease Activity Recommendations
Early RA (< 6 months)
  • Administer DMARD monotherapy in patient with low-high disease activity
  • If disease activity remains moderate /high despite DMARD monotherapy, use combination DMARDs or a TNF inhibitor or a non-TNF biologic
(Notable change from 2012 to 2015 guidelines: The 2015 guidelines do not recommend initial combination DMARD therapy in early RA with moderate to high disease activity)
Established RA (=6 month or meets 1987 ACR RA classification criteria)If disease activity remains moderate or high despite DMARD monotherapy, ACR guidelines recommend one of the following:



  • Combination DMARDs
  • Add an anti-TNF biologic
  • Non-TNF biologic
  • Tofacitinib
  



If disease activity remains moderate or high despite use of a single anti-TNF biologic:
  • Switch to a non-TNF biologic  with or without MTX over another anti-TNFi or Tofacitinib
If disease activity remains moderate or high despite use of one anti-TNF biologic and one non-TNF biologic:
  • Use another non-TNF biologic with or without MTX over Tofacitinib
  • If still uncontrolled use Tofacitinib
(Notable change from 2012 to 2015 guidelines: Instead of switching from one anti-TNF biologic to another anti-TNF biologic because of continued activity, it is recommended to change first to a non-TNF biologic)
  
Result of Initial/Repeat TST or IGRA Recommendation
Positive
  • Obtain chest x-ray
  • If the chest x-ray is suspicious for active TB, obtain sputum examination for active disease
NegativeIn patients with RA but without risk factors or clinical suspicion for TB
  • No further workup is needed
In patients with RA and immunosuppression plus LTBI risk factors
  • LBTI is not excluded
  • Repeat the TST or IGRA 1-3 wk after an initial negative test result
Active/latent TB
  • Treat with appropriate antitubercular therapy
  • Refer to a specialist as necessary
  • Initiate or resume biologic agents after either 1 mo of treatment of LTBI with antitubercular regimen or completion of treatment for active TB
  • Screen annually in individuals with RA who (1) are continuing on biologic agents while living, traveling, or working in situations of likely TB exposure and (2) have a positive baseline for TST or IGRA; TST or IGRA may still be positive after successful TB therapy; monitor for clinical signs or symptoms of recurrent TB
ACR = American College of Rheumatology; IGRA = interferon gamma release assay; LBTI = latent tuberculosis infection; RA = rheumatoid arthritis; TB = tuberculosis; TST = tuberculin skin test.



Source: Singh JA, Saag KG, et al2015 American College of Rheumatology Guideline for the Treatment of Rheumatoid Arthritis. Arthritis Care and Research http://dx.doi.org/10.1002/art.39480 (2015).[4]



Tool Type Tool Low/Minimal Disease Moderate Disease High/Severe Disease
Patient-driven compositePAS, PAS0.26–3.703.71 to < 8.08.00–10.00
PASII0.26–3.703.71 to < 8.08.00–10.00
Patient and provider compositeRAPID-3>1.0–2.0>2.0–4.0>4.0–10
Patient, provider, and laboratory compositeCDAI2.8–10.0>10.0–22.0>22.0
DAS282.6 to < 3.23.2–5.1>5.1
SDAI>3.3–11.0>11.0 to 26>26
PAS= Patient Activity Scale; RAPID-3 = Routine Assessment of Patient Index Data with 3 measures; CDAI = Clinical Disease Activity Index; DAS28 = 28-joint Disease Activity Score (with either erythrocyte sedimentation rate or C-reactive protein level); SDAI = Simplified Disease Activity Index