Search This Blog

July 23, 2014

Symptoms persist

I remain somewhat frustrated with the sporadic and persistent nature of my symptoms. One minute I forget I'm flaring and then...WHAM!...I'm reminded with a reality dose of stiffness and pain. The fatigue and difficulty concentrating are practically the default mode, but I have been able to manage despite it. I have a three day motorcycle trip planned this weekend so I am hoping not to be in too much pain during the ride, or that will make for a crappy weekend. Trying to stay positive.

July 19, 2014

A few weeks of symptoms

I have to admit that it's been a rough few weeks of being symptomatic with heel pain, left eye pressure/ache, tight traps, and incredible fatigue. So far no night sweats. It's been difficult to concentrate at work because the fatigue and heel pain have been quite overwhelming even for me. I rest on the thread that this will hopefully pass. Until then, I'm returning to another nap. 

June 28, 2014

Feeling stiff

I've been feeling quite stiff today. It's been difficult to rise from a chair and it hurts my lower back when I sneeze. My left Achilles tendon has also been markedly tender for five days. I have taken an Advil Extra Strength...now it's a wait and see game hoping for relief. 

June 12, 2014

Fatigue

It's been a challenging week. I've been feeling extremely fatigued and have been experiencing night sweats each evening for the past week. I came home from work this afternoon and slept for five solid hours, forgoing my tennis aerobics from 6-7pm and my tennis league from 9:30-11pm. I could not mentally function in the manner required for a strategic game given my exhaustion. Fortunately, I have not noticed an increase in the intensity of my baseline ever-present back and neck pain. It's been a while since I've felt so run down. 

June 03, 2014

Clinical manifestations of ankylosing spondylitis in adults

Ankylosing spondylitis (AS), a form of spondyloarthritis (SpA), is a chronic inflammatory disease of the spine (axial skeleton) manifested by back pain and progressive stiffness of the spine; it can also involve the hips, shoulders, and peripheral joints. Extraarticular manifestations, including uveitis, may also be seen in patients with AS and other forms of SpA. AS typically develops in young adults, with a peak age of onset between 20 and 30 years.

The name “ankylosing spondylitis” is derived from the Greek root “ankylosis,” meaning stiffening of a joint, and “spondylos,” which refers to a vertebra. “Ankylosing spondylitis” thus refers to the inflammatory disorder associated with fibrous or bony bridging of joints in the spine, including the bridging of intervertebral discs.

NOMENCLATURE AND CLASSIFICATION CRITERIA

Ankylosing spondylitis (AS) is the prototypic form of spondyloarthritis (SpA), a family of disorders characterized by inflammation around the entheses (the sites of ligament insertion into bone) and an association with the human leukocyte antigen (HLA)-B27.

Classically, the diseases which comprise the SpA are AS, undifferentiated SpA, reactive arthritis, psoriatic arthritis, juvenile SpA, and arthritis/spondylitis associated with inflammatory bowel diseases (IBD).
SpA with predominantly axial involvement is also designated as “axial SpA,” and SpA with predominantly peripheral involvement is also designated as “peripheral SpA.” Axial SpA in the absence of plain radiographic changes of sacroiliitis is termed non-radiographic axial SpA, abbreviated as nr-axSpA.

Several different sets of classification criteria have been used to define the clinical spectrum of AS and axial SpA more generally; and the specific criteria set used for a particular study will influence the epidemiologic, clinical, and radiographic findings found among the study population. Since the mid-1980s, the modified New York classification criteria for AS has been used most often to define the classic form of axial SpA, historically termed AS. These criteria require the presence of specific abnormalities both clinically and upon plain radiography to meet classification criteria for AS. However, radiographic abnormalities may take several years to develop, and their recognition is subject to interobserver variation. Thus, it is likely that figures reported in the literature are an underestimation of true incidence and prevalence.
The spectrum of patients considered to have axial SpA, including AS, has also been defined by the 2009 Assessment of SpondyloArthritis International Society (ASAS) classification criteria, which do not necessarily require evidence of disease on plain radiography. Other classification criteria for SpA include the Amor criteria and the European Spondyloarthritis Study Group (ESSG) criteria sets.

EPIDEMIOLOGY

General population

The incidence and prevalence of ankylosing spondylitis (AS) in a population depend upon the ethnic group studied, the selection of subjects for evaluation, and the criteria for diagnosis. Estimates of the prevalence of AS in different studies range from zero to 1.4 percent [1-5]. In general, there is a clear correlation between the prevalence of AS in a given population and the prevalence of human leukocyte antigen (HLA)-B27 in that group, with the prevalence of AS being approximately 5 to 6 percent among people who are HLA-B27-positive. 

The prevalence of AS in the US was surveyed in the 1971 to 1975 National Health and Nutrition Examination Survey (NHANES) I study consisting of 6913 subjects of 25 to 74 years of age. The prevalence of AS was estimated to be 0.2 to 0.5 percent [7]. In the US, the prevalence of HLA-B27 varies among different ethnic communities; in a 2009 national survey, the prevalence rates of HLA-B27 in several groups were [8]:

  • Mexican-Americans – 4.6 percent
  • Whites who are non-Hispanic – 7.5 percent
  • Blacks who are non-Hispanic – 1.1 percent
Familial risk depends partly upon the presence of HLA-B27. An individual's risk of developing AS is increased 5.6- to 16-fold if there is a first-degree relative with AS, and 10 to 20 percent of HLA-B27-positive individuals with affected first-degree relatives develop AS [9].

The actual male-to-female ratio is probably about 2:1 to 3:1 in AS. In a large Dutch cohort, more radiographic damage was found in the spine in men [10]. Men with non-radiographic axial spondyloarthritis (SpA) in a German cohort were at increased risk for developing radiographic sacroiliitis compared with women, and therefore at increased risk for evolution to AS [11]. Some cross-sectional studies have suggested that cervical involvement is more frequent in women, and lumbar involvement more frequent in men [12,13]; however, other studies, including longitudinal studies, have not found such a difference [10,14-16].

People with chronic back pain — The prevalence of AS among working adults with chronic (greater than three months) low back pain was 4.6 percent in one study [17]. In another group with chronic back pain, the prevalence of either AS or undifferentiated SpA with axial involvement was 5 percent [18].

MUSCULOSKELETAL SYMPTOMS AND FINDINGS

Ankylosing spondylitis (AS) is characterized by involvement of the spine and sacroiliac (SI) joints, peripheral joints, and entheses, although extraskeletal organs may also be affected. Spinal and SI involvement typically results in inflammatory back pain. Skeletal involvement may also result in complications including fracture and neurologic compromise in some patients. The major musculoskeletal features include:

  • Spinal and SI involvement
  • Hip and shoulder ("root" joint) involvement
  • Costovertebral, manubriosternal, sternoclavicular, and costochondral inflammation
  • Inflammation of extraspinal entheses
  • Peripheral arthritis
Low back pain — Almost all patients with AS report back pain, which frequently, but not invariably, has characteristics suggesting an inflammatory etiology. Inflammatory back pain typically exhibits at least four of the following five features [19]:

  • Age of onset <40 li="" years="">
  • Insidious onset
  • Improvement with exercise
  • No improvement with rest
  • Pain at night (with improvement upon arising)

Inflammatory back pain, by this definition, is present in 70 to 80 percent of patients with AS, but also in 20 to 25 percent of the patients with mechanical causes of back pain [20]. Despite its prevalence among patients with AS, in a population study only a minority of individuals with symptoms characteristic of inflammatory back pain will have SpA [21].

Limited spinal mobility and chest expansion — Postural abnormalities, especially hyperkyphosis, may already become apparent within the first 10 years of disease in some patients. Increasing flexion deformity of the neck, increased thoracic kyphosis, loss of normal lumbar lordosis, and flexion deformities of the hips generate a characteristic stooped posture. Lateral lumbar flexion and the Schober test are the measurements for spinal mobility which are affected earliest.

Buttock pain — Buttock pain, especially alternating between the two sides, may be indicative of SI involvement. In a study of 101 patients with AS and 112 with mechanical low back pain, alternating buttock pain was present in 37 percent of patients with AS and only 12 percent of patients with mechanical low back pain [23].

Hip pain — Hip involvement, resulting in hip joint pain, is present in 25 to 35 percent of patients with AS and is associated with higher degrees of disability and a worse prognosis. The typical symptom is groin pain, but as with other disorders of the hip joint, pain may be referred to the medial thigh or even the knee. Flexion deformities of the hips may develop. Hip involvement may be more severe in patients with early onset of AS, axial, and entheseal disease; and such patients may be more likely to eventually require hip replacement surgery [24].

Peripheral arthritis — Patients with AS may experience pain, stiffness, and swelling of joints outside the spine. Peripheral arthritis occurs in approximately 35 to 50 percent of patients with AS over the course of disease, and the prevalence of peripheral arthritis within a cohort of patients with AS at a single time point in one study was about 20 percent [25,26].
In one cohort, 36 percent of patients available for follow-up 33 years after enrollment in the cohort had experienced arthritis outside the spine [22]; the most commonly affected joints, in order of frequency, were the shoulders, hips, and knees. In another study, which involved 147 patients, the most frequently involved peripheral joints were ankles (40 percent), hips (36 percent), knees (29 percent), shoulders (19 percent), and the sternoclavicular joints (14 percent) [27]. Patients may also report temporomandibular joint (TMJ) pain. TMJ involvement is very uncommon in our experience, but one report has suggested that it might be more common than usually reported [28].

Enthesitis — The enthesis is the region of attachment of tendons and ligaments to bone; enthesitis (inflammation of the enthesi is a classic feature of AS and other spondyloarthritides. Enthesitis manifests as pain, stiffness and tenderness of insertions, usually without much swelling, although swelling may be a prominent feature at large insertions of the lower limbs. Enthesitis may occur in approximately 40 to 70 percent of patients with AS at some time during the disease course [11,26]. Extraspinal areas that may be involved by enthesitis in patients with AS include the calcaneal attachments of the Achilles tendon and the plantar fascia, the shoulders, the costochondral junctions, the manubriosternal and sternoclavicular joints, and along the superior iliac crest. Tenderness at these and other entheseal sites is suggestive of enthesitis.

The prevalence of enthesitis across one cohort at a single point in time was about 20 percent [11,26]; the frequency is probably dependent upon the duration of the disease. The heel is the most frequently involved site, with enthesitis present in 22 percent of patients at this site in one cross-sectional study [27]. Ultrasound techniques are frequently used in studies of enthesitis, but interpretation of these data has been limited by the great variation in acquisition methods, and there is no consensus regarding the threshold to be used in clinical practice [29]. Magnetic resonance imaging (MRI) is also used for the detection of osteitis at the entheseal insertion to the bone and soft tissue edema at the enthesis and adjacent areas.
Inflammation at the insertion of the supraspinatus tendon into the greater tuberosity of the humerus may be associated with shoulder pain and limited mobility of the affected shoulder or shoulders.

Dactylitis — Dactylitis (sausage digits) is characterized by diffuse swelling of toes or fingers. In one report of 1072 AS patients, dactylitis was present in 8 percent [30]. In two other AS studies, dactylitis at any time during disease was reported in 6 percent of patients [11,26], while prevalent dactylitis was seen in only 2 percent at a single time point [11].

EXTRAARTICULAR COMORBIDITIES

Patients with ankylosing spondylitis (AS) can exhibit a range of disease manifestations in extraskeletal organ systems. For some of these, a causal relationship with AS is more likely, such as pulmonary or cardiac involvement, while for others the relationship is less clear and may be more complex. Examples of the latter include uveitis, inflammatory bowel disease (IBD), and psoriasis, for which prevalence is disproportionally increased in patients with AS compared with the general population; these conditions may affect quality of life and function. Additionally, other problems, such as some neurologic manifestations, may be complications of changes due to the disease process. Renal disease may rarely occur, either as a comorbidity or a complication of the disease or its treatment.

Patients with AS also sometimes complain of constitutional symptoms, including considerable fatigue. Poor sleep is common, and is thought to be related in part to pain caused by active disease. Fever is an infrequent manifestation of AS in adults.

Acute anterior uveitis — Unilateral uveitis is the most common extraarticular complication of AS, occurring in 25 to 40 percent of patients [31-33]. Uveitis typically presents as acute unilateral pain, photophobia, and blurring of vision. Uveitis may be the first problem to require medical evaluation and should alert clinicians to the possibility of AS. About 50 percent of patients with acute recurrent unilateral anterior uveitis have a form of spondyloarthritis (SpA) [34]. The activity and severity of the eye disease are not correlated with the activity and severity of the articular disease. However, the occurrence of uveitis is associated with longer AS disease duration and the presence of human leukocyte antigen (HLA)-B27 [33].

Recurrence is common despite the effectiveness of local therapy, but seldom leads to permanent impairment of vision. In one study of 148 patients with HLA-B27-associated uveitis performed before the availability of tumor necrosis factor (TNF) inhibitors, the median duration of an attack was six weeks, and there were a median number of three recurrences for patients with more than 12 months of follow-up [35]. Cataracts and glaucoma can also occur. They are associated with posterior synechiae, increased intraocular pressure, and cystoid macular edema

Inflammatory bowel disease — Ileal and colonic mucosal ulcerations, which are almost always asymptomatic, can be detected by histological examination in 50 to 60 percent of patients with AS [36,37]. A minority of those with histologic inflammation develop clinically apparent IBD, whether Crohn’s disease or ulcerative colitis. Overt IBD occurs in approximately 5 to 10 percent of patients with AS, and about 4 to 10 percent of patients with IBD have concomitant findings of AS [33,38,39]. A considerable proportion of IBD patients have asymptomatic radiographic sacroiliitis [40]; the association between HLA-B27 and AS is weaker in the presence of established IBD, compared with AS alone [41]. However, the clinical presentation is very similar.

Psoriasis — Psoriasis is present in up to approximately 10 percent of patients with AS [33,38]. Patients with concomitant psoriasis have more frequent peripheral joint involvement and possibly a more severe AS disease course compared with AS patients without psoriasis [42]. Some experts have suggested that all psoriatic arthritis should be classified is part of the spondyloarthritis family, but this remains a matter of debate [43].

Psychosocial status — AS can adversely affect psychological health and socioeconomic status in addition to causing pain and disability. Taken together, these factors influence the patient’s overall quality of life, and they are collectively responsible for the intensity of pain, fatigue, and sleep disturbance [44,45].
In small studies, symptoms of pain and fatigue in patients with AS were associated with structural differences in certain regions in white and grey matter of the brain compared with controls, as shown on MRI, along with measures of emotional strength and spinal mobility [46]. A sense of helplessness (a feeling of an inability to actively cope with the disease), was an important contextual factor identified in both longitudinal and cross-sectional studies that affected health-related quality of life and functional measures [47-49].

Cardiovascular disease — AS is associated with an increased risk of both aortic regurgitation and cardiovascular disease overall:

  • The estimated frequencies of aortic regurgitation and conduction disturbances are both increased but vary considerably, ranging from 6 to 10 percent for aortic regurgitation, and from 3 to 33 percent for conduction disturbances [50-53].
  • There is also an increased risk of other cardiovascular diseases in AS. In a cross-sectional study during 2001 and 2002, involving 1843 patients with AS and 7372 controls (matched for age, gender, and geography), there were statistically significant increases among patients with AS in the prevalence of cardiovascular disease, heart failure, and peripheral vascular disease (prevalence ratios of 1.7, 95% CI 1.3-2.3; 1.6, 95% CI 1.2-2.2; and 1.8, 95% CI 1.2-2.6, respectively) [54]. There were nonsignificant trends suggesting a possible increase in ischemic heart disease and atherosclerosis. The prevalence of hypertension and hyperlipidemia was also significantly greater among the patients with AS. A 2011 metaanalysis of longitudinal studies found a statistically nonsignificant trend suggesting a possible increase in the risk of myocardial infarction in patients with AS and increased risk of the metabolic syndrome [55].
  • Pulmonary disease — Pulmonary compromise can occur in patients with AS due to restrictive changes caused by the musculoskeletal disease and to changes in the lungs themselves, including interstitial, nodular, and parenchymal abnormalities.
  • Restrictive pulmonary disease is related largely to diminished chest wall and spinal mobility. In one study where this was seen, pulmonary function tests (PFTs) were compared between patients with AS and matched controls; 18 percent of patients with AS, but none of the controls, showed a restrictive pattern [56]. Vital capacity is decreased and functional residual capacity is increased.
  • A small percentage of patients, with estimates ranging from 1.3 to 15 percent, have evidence of other pulmonary changes such as apical pulmonary fibrosis by plain radiography [57-60]. The apical fibrosis is usually asymptomatic and associated with a long duration of disease.
  • Abnormalities detected on a high-resolution computed tomography (CT) are much more common than findings on plain radiography. These include a mosaic pattern, subpleural nodules, and parenchymal bands, and may be present even early in the disease [59,60].
  • Fibrotic disease and cigarette smoking may be risk factors for spontaneous pneumothorax, a complication that is otherwise rare in AS [61].
The causes of these intrinsic pulmonary abnormalities are unknown. There is a lack of evidence for alveolitis, as bronchoalveolar lavage is generally unremarkable [58]. However, in one report transbronchial biopsies revealed interstitial fibrosis of variable degree in 5 out of 12 patients, indicating that the radiologic appearance of apical fibrosis may actually reflect a more generalized process [58].

COMPLICATIONS

Several types of complications can occur in patients with ankylosing spondylitis (AS), most frequently as a consequence of longstanding disease. These include low bone mass, fractures, and neurologic compromise due to changes in the spine. Renal complications are rare.

Osteopenia

Low bone mineral density (BMD) and bone loss have been documented in patients with AS, and are evident within the first 10 years of disease [62-64]. In more than half of patients with longstanding and persistently active disease, BMD is decreased at both the spine and the hip [65,66]. However, with advanced disease, dual photon absorptiometry (DEXA) measurement of bone mineral density of the spine may provide falsely high values because of the superimposed effect of syndesmophytes and periosteal bone formation [67]. However, estimation of bone density in the presence of syndesmophytes may be facilitated by dual-energy quantitative computed tomography (CT) scanning of the spine; and in using this technique, it has been shown that patients with syndesmophytes have lower bone mass than those without this finding [68]. Osteopenia or frank osteoporosis may predispose to fracture, especially in the spine.

Fractures in axial SpA

Patients with ankylosed spines can develop vertebral fractures with minimal (and sometimes no recollected) trauma [69]. Additionally, hyperkyphosis of the spine is common in patients with longstanding AS, and many of these patients have anterior wedging of the thoracic vertebrae [70].
In one study, the risk of a clinical vertebral fracture was significantly increased in patients with AS, compared with matched controls (odds ratio 3.3, 95% CI 1.5-7.0), although the risks of forearm or hip fractures were not [71]. Estimates of the lifetime incidence of clinical vertebral fracture in patients with AS range from 4 to 18 percent [72]. Cervical fractures that can be visualized by magnetic resonance imaging (MRI) or CT may often not be visualized on plain radiographs.

Neurologic manifestations

Neurologic symptoms can occur by several mechanisms, including cord or spinal nerve compression due to fracture of the ankylosed spine or atlantoaxial subluxation. Additionally, subclinical neuropathy, myelopathy, and myopathy are common but rarely symptomatic [73].

  • Spinal cord injury Spinal cord injury is 11 times more common in AS than in the general population and affects the cervical spine, including the lower cervical spine, more often than the thoracic and lumbar spine [74]. About 65 percent of spinal fractures are associated with neurological complications, although the initial complaints are often subtle and might be missed without a high index of suspicion [75]. The morbidity of spinal injury is very high, causing paraparesis or tetraparesis in some patients [72].
  • Atlantoaxial subluxation Clinically significant spontaneous subluxation of the atlantoaxial joint (C1-C2) occurs in patients with AS and can lead to spinal cord compression if it is not recognized and stabilized. In one study of 103 patients, for example, anterior or posterior atlantoaxial subluxation was observed in 21 and 2 percent of individuals, respectively [76]. In a two-year follow-up, progression of anterior subluxation was observed in about 50 percent. In other reports, clinically significant spontaneous subluxation occurred in approximately 2 percent of individuals. This problem is managed in a fashion similar to that in rheumatoid arthritis.
  • Cauda equina syndrome The cauda equina syndrome is a rare complication of AS, typically reported in patients with longstanding disease who have marked ankylosis of the spine [72,77,78]. The symptoms are those of damage to the lumbosacral nerve roots, probably caused by arachnoiditis. Patients may complain of abnormalities in cutaneous sensation and motor function, problems with bladder and bowel control, and impotence.

Renal disease

Renal disease is very uncommon in AS, but non-specific glomerulopathy, immunoglobulin A (IgA) nephropathy, and renal amyloidosis may be seen [79-81]. In a cohort of 681 patients with AS abnormal findings on urinalysis were seen in 8 percent, including proteinuria and/or hematuria. Patients with proteinuria exceeding 1000 mg per day underwent renal biopsy. The biopsies showed nonspecific glomerulopathy (3 patients), IgA nephropathy (2 patients), and amyloidosis (1 patient) [79]. This spectrum of renal diseases is in agreement with an earlier review of literature [80]. Renal amyloidosis is rare [79,82,83]. 

PREGNANCY

Ankylosing spondylitis (AS) does not appear to adversely affect pregnancy. The relationship between the activity of AS and pregnancy has been evaluated by questionnaire in patients from the United States, Canada, and 10 European countries, in whom no adverse effects upon fertility, pregnancy outcome, or the neonate were identified [84]. Disease activity was improved, remained unchanged, or had worsened in equal numbers of patients. A postpartum flare was experienced in about 60 percent of patients, particularly in the peripheral joints.

LABORATORY FINDINGS

Laboratory findings in ankylosing spondylitis (AS) are generally nonspecific. An elevated acute phase response may be present, including an elevated erythrocyte sedimentation rate (ESR) and elevated C-reactive protein (CRP), in about 50 to 70 percent of patients with active disease; thus, a normal ESR and CRP do not exclude AS [11,26,85]. The ESR and CRP do not correlate well with axial disease activity, but might correlate better with active peripheral arthritis. A normochromic normocytic anemia is occasionally seen, most typically in patients with very active disease [85,86].

The level of serum bone-specific alkaline phosphatase may be elevated in severe disease, and serum levels of immunoglobulin A (IgA) are commonly elevated above the normal range [87]. Synovial fluid findings are typical of inflammatory arthritis, with an increased white blood cell count that is predominated by polymorphonuclear leukocytes. Human leukocyte antigen (HLA)-B27 is present, but not invariably so, in most patients with AS (90 to 95 percent of patients of European ancestry), and commonly present in other forms of spondyloarthritis compared with the prevalence in the general population.

MUSCULOSKELETAL IMAGING

Radiographic changes from joint and spine involvement with ankylosing spondylitis (AS) usually become evident over time. Evidence of AS can sometimes be demonstrated by magnetic resonance imaging (MRI) before damage is evident on plain radiography.

Sacroiliac joints

An abnormal appearance of the sacroiliac (SI) joint using an appropriate imaging technique is a hallmark of AS. Radiographic findings progress from subtle joint space narrowing and sclerosis to erosive change, and may result eventually in bony ankylosis (fusion). Plain radiography and MRI are the principal techniques used to assess the SI joints; additional methods that have been used to detect abnormalities include computed tomography (CT), scintigraphy, ultrasonography, and conventional tomography. Scintigraphy lacks specificity for SI, and conventional tomography involves substantial radiation exposure; thus, neither are generally used clinically.

Plain radiographs of the pelvis/SI joints — The initial radiographic changes of AS occur in the SI joints in about 95 percent of patients. In general, radiographic damage is slow to develop. Plain radiographs may be normal or may only reveal slight or suspicious changes during the first few years of disease, but usually will become abnormal within the first decade [14]. In a study of 228 patients with newly diagnosed axial spondyloarthritis (SpA), radiographic changes of the SI joints typical of AS were found in 33 percent of patients with symptoms for less than one year, about 47 percent of patients with symptoms for one to six years, and in 68 percent of those with symptoms for between six and nine years [88].

Changes characteristic of sacroiliitis can be well visualized with anteroposterior (AP) views of the pelvis. However, the SI joints are not as clearly visualized on plain radiography as are other joints, such as the knees or digits, and more subtle findings can be missed. This is because the direction of the SI joint is oblique relative to the coronal plane, being nearer to the central axis posteriorly than anteriorly. In addition, the direction is also oblique relative to the sagittal plane, being nearer the central axis inferiorly than superiorly. The surfaces on both the sacral and the iliac sides are also irregular in shape. X-ray films should be of good quality, as under-penetrated films can be misread.

With a high-quality film of the AP view of the pelvis, the joint spaces of both the right and the left SI joints can be well seen. The right and left sides should be read separately. The most obvious abnormalities as disease becomes more advanced are fusion and large erosions. Erosions are seen as obvious craters, usually associated with widening of the joint space. In advanced disease of the joint, when an SI joint is fused, the joint space disappears, so that the bone is continuous between the sacrum and the ilium. On the x-ray film, the joint space becomes a thin, narrow line. Presence of either fusion or obvious erosion even on one of the two SI joints is consistent with AS. The presence of sclerosis of the SI joint is a sign indicating possible AS, but is not diagnostic by itself.
Sacroiliitis has been divided into five grades, primarily for use in epidemiologic and clinical research:
  • Grade 0: normal (image 2)
  • Grade 1: suspicious changes (image 3)
  • Grade 2: minimal abnormality – small localized areas with erosions or sclerosis, without alteration in the joint width (image 3)
  • Grade 3: unequivocal abnormality – moderate or advanced sacroiliitis with one or more of the following: erosions, evidence of sclerosis, widening, narrowing, or partial ankylosis (image 4A-B)
  • Grade 4: total ankylosis (image 5A-B)
By expert consensus it has been deemed that definite sacroiliitis is present in patients with bilateral changes of at least grade 2 in severity or at least unilateral changes of at least grade 3 severity. [89]. In practice, clinicians can regard the x-ray changes to be diagnostic of sacroiliitis if there is ankylosis or if there are obvious erosions. Other examples of the various grades are available in the slide library of the Assessment of Spondyloarthritis International Society.

Additional changes typical but not diagnostic of AS that may be seen on plain radiography of the pelvis include erosions and osteitis at the ischial tuberosities, iliac crests, symphysis pubis, femoral trochanters, and hip. Radiographs from these sites typically show destructive changes without much new bone formation. The severity of radiographic hip involvement correlates with more severe spinal disease [14].

MRI of the SI joints — The most characteristic change of active sacroiliitis on MRI is termed "bone marrow edema" (BME), which is a hyperintense signal in the bones adjacent to the affected joints, as seen in the STIR (short tau inversion recovery) images or the T2-weighted images with fat absorption. MRI views that are most helpful in AS, according to expert consensus, are the semi-coronal views parallel to the axes of the SI joints. Administration of contrast medium is normally not necessary to visualize these changes. BME can also be present in other causes of chronic low back pain, such as degenerative spine disease, where it may be present in up to 20 percent of such patients [90].
MRI changes other than BME, including structural changes such as erosions, fatty degeneration, and sclerosis visualized by T1-weighted images, can also be present; these findings can be useful in clinical assessment.

Spine

A series of radiographic changes characteristic of AS can develop progressively over the course of disease [14]:

  • Squaring of the vertebral bodies due to anterior and posterior spondylitis is an early radiographic sign of inflammatory and destructive spinal involvement due to axial SpA.
  • Changes that are more easily distinguished and found at later stages of AS include syndesmophytes, ankylosis of the facet joints, and calcification of the anterior longitudinal ligament.
  • The abnormality that is most easily recognizable is the bamboo spine in late ankylosing spondylitis.
Anterior atlantoaxial (C1-C2) subluxation is also be visualized by use of plain radiographs taken in flexion and extension of the cervical spine. Other findings that may also be apparent on radiographs of the spine are spondylodiscitis and fractures [91].

MRI of the lumbar spine may show changes in the absence of abnormalities on plain radiographs, and infrequently even in the absence of SI joint inflammation [92]. Spinal changes in the absence of significant SI joint inflammation were more frequent in a study population of patients with clinically active, longstanding, non-radiographic axial SpA (nr-axSpA) [93]. Abnormalities seen in AS and other SpA that experts consider consistent with inflammatory change in the spine include anterior/posterior spondylitis in at least three sites on a STIR sequence [94]. The MRI views should be sagittal and should include the STIR or the T2-weighted images with fat absorption sequences. Another characteristic lesion on MRI of the spine is fatty change, which may be present in several corners of the vertebral bodies, especially in younger adults and in the presence of a ‘healthy’ disc, seen on T1-weighted sequences.

Entheses

Ultrasonography is increasingly being used for the detection and assessment of enthesitis, such as Achilles tendonitis. Findings in 2011 of an expert task force, based upon a systematic review of published data, were that the utility of ultrasound in assessing enthesitis required further validation, despite a growing body of literature [29]. Major problems included a lack of standardization of techniques, the diversity of equipment used in different studies, and a lack of consensus for the definitions of enthesitis and treatment response.

SUMMARY AND RECOMMENDATIONS


  • Estimates of the prevalence of ankylosing spondylitis (AS) vary from 0 to 1.4 percent, depending upon the ethnic group, the prevalence of human leukocyte antigen (HLA)-B27, the selection of subjects for evaluation, and the criteria for diagnosis. The prevalence may be underestimated because of the difficulty in establishing the diagnosis clinically and radiographically; it is increased to approximately 5 to 6 percent in subjects who are HLA-B27 positive. The prevalence of AS or axial spondyloarthritis (SpA) among adults with chronic back pain is about 5 percent. 
  • The symptoms of AS include inflammatory back pain, buttock pain, limited spinal mobility, limited chest expansion, hip and shoulder pain, and enthesitis. Patients may have peripheral arthritis, dactylitis, and constitutional features. Inflammatory low back pain is characterized by age of onset <40 .="" li="">
  • Extraarticular involvement may include anterior uveitis, psoriasis, and symptomatic as well as asymptomatic inflammatory bowel diseases (IBD). Additional extraarticular features include cardiovascular disease, including aortic regurgitation and conduction abnormalities; pulmonary disease, especially restricted chest expansion and apical pulmonary fibrosis; and manifestations of psychosocial distress. Complications of AS may include osteopenia, neurologic manifestations of cord or spinal nerve compression due to spinal fracture or atlantoaxial subluxation, and other rare neurologic events.
  • Laboratory findings are generally nonspecific, and may include the presence of HLA-B27 and occasionally a normochromic normocytic anemia. An elevated acute phase response may be present, including an elevated erythrocyte sedimentation rate (ESR) and elevated C-reactive protein (CRP), but the ESR and CRP do not correlate well with axial disease activity, and a normal ESR and CRP do not exclude AS or active disease. 
  • Imaging abnormalities characteristic of AS include sacroiliitis and spondylitis, which can be seen on plain radiographs in more advanced disease. However, they are only detected during early disease, when plain radiographs may be normal or may have only minimal changes, by the finding of bone marrow edema (BME) and other changes on magnetic resonance imaging (MRI). Ultrasonography may be helpful in the detection of enthesitis but requires further standardization and validation. 

Authors: David T Yu, MD, Astrid van Tubergen, MD, PhD
Section Editor: Joachim Sieper, MD
Deputy Editor: Paul L Romain, MD

All topics are updated as new evidence becomes available and our peer review process for UptoDate is complete. Literature review current through: Apr 2014. | This topic last updated: Mar 13, 2014.

REFERENCES

  1. Lawrence RC, Helmick CG, Arnett FC, et al. Estimates of the prevalence of arthritis and selected musculoskeletal disorders in the United States. Arthritis Rheum 1998; 41:778.
  2. Khan MA. HLA-B27 and its subtypes in world populations. Curr Opin Rheumatol 1995; 7:263.
  3. Braun J, Bollow M, Remlinger G, et al. Prevalence of spondylarthropathies in HLA-B27 positive and negative blood donors. Arthritis Rheum 1998; 41:58.
  4. Ng SC, Liao Z, Yu DT, et al. Epidemiology of spondyloarthritis in the People's Republic of China: review of the literature and commentary. Semin Arthritis Rheum 2007; 37:39.
  5. Helmick CG, Felson DT, Lawrence RC, et al. Estimates of the prevalence of arthritis and other rheumatic conditions in the United States. Part I. Arthritis Rheum 2008; 58:15.
  6. Zochling J, Smith EU. Seronegative spondyloarthritis. Best Pract Res Clin Rheumatol 2010; 24:747.
  7. Reveille JD. Epidemiology of spondyloarthritis in North America. Am J Med Sci 2011; 341:284.
  8. Reveille JD, Hirsch R, Dillon CF, et al. The prevalence of HLA-B27 in the US: data from the US National Health and Nutrition Examination Survey, 2009. Arthritis Rheum 2012; 64:1407.
  9. Baron M, Zendel I. HLA-B27 testing in ankylosing spondylitis: an analysis of the pretesting assumptions. J Rheumatol 1989; 16:631.
  10. van Tubergen A, van der Heijde D, Dougados M, et al. Are syndesmophytes most prevalent in the lumbar or in the cervical spine in patients with ankylosing spondylitis and do they develop in a specific direction? Rheumatology (Oxford) 2012; 51:1432.
  11. Rudwaleit M, Haibel H, Baraliakos X, et al. The early disease stage in axial spondylarthritis: results from the German Spondyloarthritis Inception Cohort. Arthritis Rheum 2009; 60:717.
  12. Gran JT, Husby G, Hordvik M, et al. Radiological changes in men and women with ankylosing spondylitis. Ann Rheum Dis 1984; 43:570.
  13. Atagunduz P, Aydin SZ, Bahadir C, et al. Determinants of early radiographic progression in ankylosing spondylitis. J Rheumatol 2010; 37:2356.
  14. Jang JH, Ward MM, Rucker AN, et al. Ankylosing spondylitis: patterns of radiographic involvement--a re-examination of accepted principles in a cohort of 769 patients. Radiology 2011; 258:192.
  15. Brophy S, Mackay K, Al-Saidi A, et al. The natural history of ankylosing spondylitis as defined by radiological progression. J Rheumatol 2002; 29:1236.
  16. El Maghraoui A, Bensabbah R, Bahiri R, et al. Cervical spine involvement in ankylosing spondylitis. Clin Rheumatol 2003; 22:94.
  17. Calin A, Garrett S, Whitelock H, et al. A new approach to defining functional ability in ankylosing spondylitis: the development of the Bath Ankylosing Spondylitis Functional Index. J Rheumatol 1994; 21:2281.
  18. Underwood MR, Dawes P. Inflammatory back pain in primary care. Br J Rheumatol 1995; 34:1074.
  19. Sieper J, van der Heijde D, Landewé R, et al. New criteria for inflammatory back pain in patients with chronic back pain: a real patient exercise by experts from the Assessment of SpondyloArthritis international Society (ASAS). Ann Rheum Dis 2009; 68:784.
  20. Rudwaleit M, van der Heijde D, Khan MA, et al. How to diagnose axial spondyloarthritis early. Ann Rheum Dis 2004; 63:535.
  21. Weisman MH. Inflammatory back pain: the United States perspective. Rheum Dis Clin North Am 2012; 38:501.
  22. Carette S, Graham D, Little H, et al. The natural disease course of ankylosing spondylitis. Arthritis Rheum 1983; 26:186.
  23. Rudwaleit M, Metter A, Listing J, et al. Inflammatory back pain in ankylosing spondylitis: a reassessment of the clinical history for application as classification and diagnostic criteria. Arthritis Rheum 2006; 54:569.
  24. Vander Cruyssen B, Muñoz-Gomariz E, Font P, et al. Hip involvement in ankylosing spondylitis: epidemiology and risk factors associated with hip replacement surgery. Rheumatology (Oxford) 2010; 49:73.
  25. Vander Cruyssen B, Ribbens C, Boonen A, et al. The epidemiology of ankylosing spondylitis and the commencement of anti-TNF therapy in daily rheumatology practice. Ann Rheum Dis 2007; 66:1072.
  26. Ciurea A, Scherer A, Exer P, et al. Tumor necrosis factor α inhibition in radiographic and nonradiographic axial spondyloarthritis: results from a large observational cohort. Arthritis Rheum 2013; 65:3096.
  27. Sampaio-Barros PD, Bertolo MB, Kraemer MH, et al. Primary ankylosing spondylitis: patterns of disease in a Brazilian population of 147 patients. J Rheumatol 2001; 28:560.
  28. Ramos-Remus C, Major P, Gomez-Vargas A, et al. Temporomandibular joint osseous morphology in a consecutive sample of ankylosing spondylitis patients. Ann Rheum Dis 1997; 56:103.
  29. Gandjbakhch F, Terslev L, Joshua F, et al. Ultrasound in the evaluation of enthesitis: status and perspectives. Arthritis Res Ther 2011; 13:R188.
  30. Pérez Alamino R, Maldonado Cocco JA, Citera G, et al. Differential features between primary ankylosing spondylitis and spondylitis associated with psoriasis and inflammatory bowel disease. J Rheumatol 2011; 38:1656.
  31. Maksymowych WP, Chou CT, Russell AS. Matching prevalence of peripheral arthritis and acute anterior uveitis in individuals with ankylosing spondylitis. Ann Rheum Dis 1995; 54:128.
  32. Zeboulon N, Dougados M, Gossec L. Prevalence and characteristics of uveitis in the spondyloarthropathies: a systematic literature review. Ann Rheum Dis 2008; 67:955.
  33. Stolwijk C, van Tubergen A, Castillo-Ortiz JD, Boonen A. Prevalence of extra-articular manifestations in patients with ankylosing spondylitis: a systematic review and meta-analysis. Ann Rheum Dis 2013.
  34. Muñoz-Fernández S, Martín-Mola E. Uveitis. Best Pract Res Clin Rheumatol 2006; 20:487.
  35. Tay-Kearney ML, Schwam BL, Lowder C, et al. Clinical features and associated systemic diseases of HLA-B27 uveitis. Am J Ophthalmol 1996; 121:47.
  36. Leirisalo-Repo M, Turunen U, Stenman S, et al. High frequency of silent inflammatory bowel disease in spondylarthropathy. Arthritis Rheum 1994; 37:23.
  37. De Keyser F, Mielants H. The gut in ankylosing spondylitis and other spondyloarthropathies: inflammation beneath the surface. J Rheumatol 2003; 30:2306.
  38. El Maghraoui A. Extra-articular manifestations of ankylosing spondylitis: prevalence, characteristics and therapeutic implications. Eur J Intern Med 2011; 22:554.
  39. Palm O, Moum B, Ongre A, Gran JT. Prevalence of ankylosing spondylitis and other spondyloarthropathies among patients with inflammatory bowel disease: a population study (the IBSEN study). J Rheumatol 2002; 29:511.
  40. WRIGHT V, WATKINSON G. SACRO-ILIITIS AND ULCERATIVE COLITIS. Br Med J 1965; 2:675.
  41. Rudwaleit M, Baeten D. Ankylosing spondylitis and bowel disease. Best Pract Res Clin Rheumatol 2006; 20:451.
  42. Edmunds L, Elswood J, Kennedy LG, Calin A. Primary ankylosing spondylitis, psoriatic and enteropathic spondyloarthropathy: a controlled analysis. J Rheumatol 1991; 18:696.
  43. Eder L, Gladman DD. Psoriatic arthritis: phenotypic variance and nosology. Curr Rheumatol Rep 2013; 15:316.
  44. Li Y, Zhang S, Zhu J, et al. Sleep disturbances are associated with increased pain, disease activity, depression, and anxiety in ankylosing spondylitis: a case-control study. Arthritis Res Ther 2012; 14:R215.
  45. Ward MM, Reveille JD, Learch TJ, et al. Impact of ankylosing spondylitis on work and family life: comparisons with the US population. Arthritis Rheum 2008; 59:497.
  46. Wu Q, Inman RD, Davis KD. Fatigue in ankylosing spondylitis is associated with the brain networks of sensory salience and attention. Arthritis Rheumatol 2014; 66:295.
  47. Jang JH, Green CE, Assassi S, et al. The contribution of disease activity on functional limitations over time through psychological mediators: a 12-month longitudinal study in patients with ankylosing spondylitis. Rheumatology (Oxford) 2011; 50:2087.
  48. Baysal O, Durmuş B, Ersoy Y, et al. Relationship between psychological status and disease activity and quality of life in ankylosing spondylitis. Rheumatol Int 2011; 31:795.
  49. Gordeev VS, Maksymowych WP, Evers SM, et al. Role of contextual factors in health-related quality of life in ankylosing spondylitis. Ann Rheum Dis 2010; 69:108.
  50. Eder L, Sadek M, McDonald-Blumer H, Gladman DD. Aortitis and spondyloarthritis--an unusual presentation: case report and review of the literature. Semin Arthritis Rheum 2010; 39:510.
  51. Slobodin G, Naschitz JE, Zuckerman E, et al. Aortic involvement in rheumatic diseases. Clin Exp Rheumatol 2006; 24:S41.
  52. Vinsonneau U, Brondex A, Mansourati J, et al. Cardiovascular disease in patients with spondyloarthropathies. Joint Bone Spine 2008; 75:18.
  53. Brunner F, Kunz A, Weber U, Kissling R. Ankylosing spondylitis and heart abnormalities: do cardiac conduction disorders, valve regurgitation and diastolic dysfunction occur more often in male patients with diagnosed ankylosing spondylitis for over 15 years than in the normal population? Clin Rheumatol 2006; 25:24.
  54. Han C, Robinson DW Jr, Hackett MV, et al. Cardiovascular disease and risk factors in patients with rheumatoid arthritis, psoriatic arthritis, and ankylosing spondylitis. J Rheumatol 2006; 33:2167.
  55. Mathieu S, Gossec L, Dougados M, Soubrier M. Cardiovascular profile in ankylosing spondylitis: a systematic review and meta-analysis. Arthritis Care Res (Hoboken) 2011; 63:557.
  56. Berdal G, Halvorsen S, van der Heijde D, et al. Restrictive pulmonary function is more prevalent in patients with ankylosing spondylitis than in matched population controls and is associated with impaired spinal mobility: a comparative study. Arthritis Res Ther 2012; 14:R19.
  57. Boushea DK, Sundstrom WR. The pleuropulmonary manifestations of ankylosing spondylitis. Semin Arthritis Rheum 1989; 18:277.
  58. Kchir MM, Mtimet S, Kochbati S, et al. Bronchoalveolar lavage and transbronchial biopsy in spondyloarthropathies. J Rheumatol 1992; 19:913.
  59. Quismorio FP Jr. Pulmonary involvement in ankylosing spondylitis. Curr Opin Pulm Med 2006; 12:342.
  60. Kiris A, Ozgocmen S, Kocakoc E, et al. Lung findings on high resolution CT in early ankylosing spondylitis. Eur J Radiol 2003; 47:71.
  61. Lee CC, Lee SH, Chang IJ, et al. Spontaneous pneumothorax associated with ankylosing spondylitis. Rheumatology (Oxford) 2005; 44:1538.
  62. van der Weijden MA, Claushuis TA, Nazari T, et al. High prevalence of low bone mineral density in patients within 10 years of onset of ankylosing spondylitis: a systematic review. Clin Rheumatol 2012; 31:1529.
  63. Geusens P, Lems WF. Osteoimmunology and osteoporosis. Arthritis Res Ther 2011; 13:242.
  64. Roux C. Osteoporosis in inflammatory joint diseases. Osteoporos Int 2011; 22:421.
  65. Gratacós J, Collado A, Pons F, et al. Significant loss of bone mass in patients with early, active ankylosing spondylitis: a followup study. Arthritis Rheum 1999; 42:2319.
  66. Dos Santos FP, Constantin A, Laroche M, et al. Whole body and regional bone mineral density in ankylosing spondylitis. J Rheumatol 2001; 28:547.
  67. El Maghraoui A, Borderie D, Cherruau B, et al. Osteoporosis, body composition, and bone turnover in ankylosing spondylitis. J Rheumatol 1999; 26:2205.
  68. Karberg K, Zochling J, Sieper J, et al. Bone loss is detected more frequently in patients with ankylosing spondylitis with syndesmophytes. J Rheumatol 2005; 32:1290.
  69. Vosse D, Feldtkeller E, Erlendsson J, et al. Clinical vertebral fractures in patients with ankylosing spondylitis. J Rheumatol 2004; 31:1981.
  70. Vosse D, van der Heijde D, Landewé R, et al. Determinants of hyperkyphosis in patients with ankylosing spondylitis. Ann Rheum Dis 2006; 65:770.
  71. Vosse D, Landewé R, van der Heijde D, et al. Ankylosing spondylitis and the risk of fracture: results from a large primary care-based nested case-control study. Ann Rheum Dis 2009; 68:1839.
  72. Mundwiler ML, Siddique K, Dym JM, et al. Complications of the spine in ankylosing spondylitis with a focus on deformity correction. Neurosurg Focus 2008; 24:E6.
  73. Khedr EM, Rashad SM, Hamed SA, et al. Neurological complications of ankylosing spondylitis: neurophysiological assessment. Rheumatol Int 2009; 29:1031.
  74. Chaudhary SB, Hullinger H, Vives MJ. Management of acute spinal fractures in ankylosing spondylitis. ISRN Rheumatol 2011; 2011:150484.
  75. Thumbikat P, Hariharan RP, Ravichandran G, et al. Spinal cord injury in patients with ankylosing spondylitis: a 10-year review. Spine (Phila Pa 1976) 2007; 32:2989.
  76. Ramos-Remus C, Gomez-Vargas A, Guzman-Guzman JL, et al. Frequency of atlantoaxial subluxation and neurologic involvement in patients with ankylosing spondylitis. J Rheumatol 1995; 22:2120.
  77. Bartleson JD, Cohen MD, Harrington TM, et al. Cauda equina syndrome secondary to long-standing ankylosing spondylitis. Ann Neurol 1983; 14:662.
  78. Sant SM, O'Connell D. Cauda equina syndrome in ankylosing spondylitis: a case report and review of the literature. Clin Rheumatol 1995; 14:224.
  79. Lee SH, Lee EJ, Chung SW, et al. Renal involvement in ankylosing spondylitis: prevalence, pathology, response to TNF-a blocker. Rheumatol Int 2013; 33:1689.
  80. Strobel ES, Fritschka E. Renal diseases in ankylosing spondylitis: review of the literature illustrated by case reports. Clin Rheumatol 1998; 17:524.
  81. Levy AR, Szabo SM, Rao SR, et al. Estimating the occurrence of renal complications among persons with ankylosing spondylitis. Arthritis Care Res (Hoboken) 2014; 66:440.
  82. Gratacós J, Collado A, Sanmartí R, et al. Coincidental amyloid nephropathy and IgA glomerulonephritis in a patient with ankylosing spondylitis. J Rheumatol 1993; 20:1613.
  83. Lance NJ, Curran JJ. Amyloidosis in a case of ankylosing spondylitis with a review of the literature. J Rheumatol 1991; 18:100.
  84. Ostensen M, Ostensen H. Ankylosing spondylitis--the female aspect. J Rheumatol 1998; 25:120.
  85. Sieper J, Braun J, Rudwaleit M, et al. Ankylosing spondylitis: an overview. Ann Rheum Dis 2002; 61 Suppl 3:iii8.
  86. Niccoli L, Nannini C, Cassarà E, et al. Frequency of anemia of inflammation in patients with ankylosing spondylitis requiring anti-TNFα drugs and therapy-induced changes. Int J Rheum Dis 2012; 15:56.
  87. Maksymowych WP. What do biomarkers tell us about the pathogenesis of ankylosing spondylitis? Arthritis Res Ther 2009; 11:101.
  88. Poddubnyy D, Brandt H, Vahldiek J, et al. The frequency of non-radiographic axial spondyloarthritis in relation to symptom duration in patients referred because of chronic back pain: results from the Berlin early spondyloarthritis clinic. Ann Rheum Dis 2012; 71:1998.
  89. van Tubergen A, Heuft-Dorenbosch L, Schulpen G, et al. Radiographic assessment of sacroiliitis by radiologists and rheumatologists: does training improve quality? Ann Rheum Dis 2003; 62:519.
  90. Marzo-Ortega H, McGonagle D, O'Connor P, et al. Baseline and 1-year magnetic resonance imaging of the sacroiliac joint and lumbar spine in very early inflammatory back pain. Relationship between symptoms, HLA-B27 and disease extent and persistence. Ann Rheum Dis 2009; 68:1721.
  91. van der Heijde D, Landewé R. Imaging in spondylitis. Curr Opin Rheumatol 2005; 17:413.
  92. Rudwaleit M, van der Heijde D, Landewé R, et al. The development of Assessment of SpondyloArthritis international Society classification criteria for axial spondyloarthritis (part II): validation and final selection. Ann Rheum Dis 2009; 68:777.
  93. van der Heijde D, Sieper J, Maksymowych WP, et al. Spinal inflammation in the absence of sacroiliac joint inflammation on magnetic resonance imaging in patients with active nonradiographic axial spondyloarthritis. Arthritis Rheumatol 2014; 66:667.
  94. Hermann KG, Baraliakos X, van der Heijde DM, et al. Descriptions of spinal MRI lesions and definition of a positive MRI of the spine in axial spondyloarthritis: a consensual approach by the ASAS/OMERACT MRI study group. Ann Rheum Dis 2012; 71:1278.

Chest wall complications from AS

Ankylosing spondylitis is a chronic inflammatory disease affecting joints of the axial skeleton with resultant fibrosis and ossification of the ligamentous structures of the spine, sacroiliac joints, and rib cage [1]. Patients present most frequently with chronic low back pain, and clinical findings often include limited spinal range of motion, bilateral sacroiliitis on plain radiographs, and positive serology for HLA-B27 in 90 to 95 percent of patients. Individuals may rarely complain of exercise intolerance. Extraarticular manifestations of the illness include ocular disease (anterior uveitis, 25 percent) and cardiovascular disease (including aortitis, aortic insufficiency, aortic root dilation, thoracic aortic aneurysms, conduction abnormalities), and peripheral arthritis occurs in 10 to 20 percent of patients [2].

Ankylosing spondylitis causes pleuropulmonary disease in less than 2% of patients, most commonly in the form of chest wall restriction and upper lobe fibrocystic parenchymal disease. Less commonly, ankylosing spondylitis affects the cricoarytenoid joint, and patients can present with hoarseness, sore throat, upper airway obstruction, or respiratory failure. In addition, these patients are at increased risk for spinal cord injury when undergoing endotracheal intubation.

Pathophysiology

Ankylosing spondylitis causes fixation of the chest wall through fusion of the costovertebral joints from inflammation [4-6]. Similarly, the anterior chest wall can be affected by enthesitis (inflammation of the muscular or tendinous attachments to bone) of the manubriosternal symphysis and sternoclavicular joints. Clinical evidence of chest wall disease includes limitation of chest wall expansion to 2.5 centimeters when measured at the level of the fourth intercostal space [7].

Pulmonary function

Despite chest wall restriction, patients will often manifest only a mild restrictive ventilatory defect, with a mild decrease in the total lung capacity (TLC) and vital capacity (VC) [4]. Relative preservation of lung volumes has been attributed to compensation by the diaphragm with increased abdominal excursion, fixation of the thorax at greater lung volumes, and preservation of chest wall symmetry and rib excursion [4-6,8]. The functional residual capacity (FRC) and residual volume (RV) are normal or increased due to fixation of the rib cage in an inspiratory position. Gas exchange, airflow, and lung compliance are usually normal. Maximal inspiratory pressure (MIP) and maximal expiratory pressure (MEP) may be mildly reduced [8,9].

Exercise tolerance

Patients may rarely complain of exercise intolerance. Formal testing may reveal exercise limitation with decreased maximal oxygen consumption (VO2max), but the extent to which this correlates with limited chest wall expansion is debated [10,11]. Some authors report that exercise limitation does not occur until the vital capacity (VC) is less than 75 percent predicted [12], while others have invoked additional factors such as deconditioning and inspiratory muscle fatigue to account for the exercise limitation [13]. Non-specific body fatigue can confound interpretation of self-reported exercise capacity capability [14].

Treatment

Treatment is primarily preventive and supportive. There is debate about whether disease activity, as assessed by the erythrocyte sedimentation rate (ESR), correlates with the decline in VC [5,15]. Anti-inflammatory therapy has not been shown to influence the pulmonary disease or the underlying illness. Spinal extension exercises may be beneficial in maintaining mobility [6]. A number of preliminary reports on the use of TNF blockers in ankylosing spondylitis suggest short-term improvement in disease activity ratings and quality of life measures [16-18]. Changes in pulmonary function tests and respiratory symptoms have not been assessed consistently.

Parenchymal disease

Apical fibrobullous disease develops in approximately 1 percent of patients, with a male to female predominance of 50:1 [2,19]. Affected patients have usually suffered from ankylosing spondylitis for at least 15 years. Possible etiologies of this complication include:
  • Inflammatory lung disease
  • Repeated aspiration from associated esophageal muscle dysfunction (with diminished clearance of secretions due to chest wall disease)
  • Decreased ventilation of, or altered apical stresses to, the upper lung fields as a result of chest wall disease.
The course of the fibrosis may be progressive, and no therapy of uncomplicated apical fibrosis has been shown to alter the direction of the illness [4,19]. This condition is usually clinically silent until superinfection occurs, most commonly with Aspergillus or mycobacterial species [2]. Because of the location of pulmonary disease and its radiographic appearance, tuberculosis must be excluded. These patients are also at increased risk for spontaneous pneumothorax. 

Assessment and treatment of ankylosing spondylitis in adults

Ankylosing spondylitis (AS) is a chronic inflammatory disease manifested by back pain and progressive spinal stiffness. AS characteristically affects young adults with a peak age of onset between 20 and 30 years. Although classically thought of as a spinal disease, transient acute arthritis of peripheral joints occurs in up to 50 percent of patients. In addition, other organs such as the eyes, lungs, heart, and kidneys can be affected. The assessment, monitoring, prognosis, and treatment of ankylosing spondylitis in adults are presented here. The clinical manifestations and diagnosis of ankylosing spondylitis in adults, and a detailed discussion of the clinical manifestations, approach to diagnosis, pathogenesis, and treatment of SpA in children, are presented separately.

GOALS OF MANAGEMENT

The primary goal of management for patients with ankylosing spondylitis (AS) is to maximize long-term health-related quality of life through the following:

  • Relief of symptoms – To eliminate symptoms such as pain, stiffness, and fatigue or to reduce them to the minimal possible level
  • Maintenance of function – To maintain the best possible functional capacity
  • Prevention of complications of spinal disease – To prevent flexion contractures, especially dorsal kyphosis (hunching over from the spine)
  • Minimization of extraspinal and extraarticular manifestations and comorbidities – To reduce the impact of AS-associated disorders such as uveitis and aortic valve insufficiency
The authors' recommendations are generally consistent with those jointly developed by the Assessment of SpondyloArthritis international Society (ASAS) and the European League Against Rheumatism (EULAR), and also with those proposed by an international task force [1-3]. This task force advocates a “treat to target” approach to spondyloarthritis management, with an ideal goal of disease remission, and an alternative goal, appropriate particularly for some patients with more longstanding disease, of minimal disease activity [3]. Management of AS is lifelong for each patient, and shared decision-making by patients and their clinicians is important in management.

The symptoms of AS and the rate of progression fluctuate with time and can vary substantially between patients. Functional limitations can result directly from disease activity but also indirectly from the feelings of helplessness and depression, which may be associated with disease activity and which also require recognition and appropriate treatment intervention [4].

ASSESSMENT AND MONITORING

Treatment decisions depend upon an accurate assessment of disease impact. In general, four approaches are required:

  1. Information is elicited in the patient medical history to assess the degree of disease activity, functional disability, and health-related quality of life.
  2. Physical examination is used to detect inflammation, contractures, and limitations in range of motion.
  3. Blood tests are used to assist in assessing disease activity.
  4. Imaging is used to assess skeletal damage and, in some patients, disease activity.

1. Patient medical history 


— The medical history is used to assess disease activity, functional disability, and health-related status. The following parameters are recommended:

  • The patient global assessment on a scale of 0 to 10 or (1 to 10).
  • The physician global assessment on a scale of 1 (mild) to 4 (severe).
  • The Bath Ankylosing Spondylitis Disease Activity Index (BASDAI) disease activity questionnaire 
A) Functional disability and psychological status – In clinical practice, the particular daily activities and quality-of-life parameters to be assessed depend upon the judgement of clinician. In clinical research on ankylosing spondylitis (AS), functional disability and health-related quality of life are numerically scored using the Bath Ankylosing Spondylitis Functional Index (BASFI) and the AS Quality of Life instrument (ASQoL), respectively [5-8]. The BASFI focuses on functional ability, while the ASQoL also addresses social and psychological domains.

Disease activity scores:
A.1) BASDAI — The BASDAI disease activity questionnaire consists of six questions concerning overall symptoms during the week immediately prior to answering the questions, including [9]:

  1. How would you describe the overall level of fatigue/tiredness you have experienced?
  2. How would you describe the overall level of AS neck, back, or hip pain you have had?
  3. How would you describe the overall level of pain/swelling in joints other than neck, back, hips you have had?
  4. How would you describe the level of discomfort you have had from an area tender to touch or pressure?
  5. How would you describe the level of morning stiffness you have had from the time you wake up?
  6. How long does your morning stiffness last from the time you wake up?
Each question is scored on a scale of 0 to 10. Except for the last question, 0 indicates none and 10 indicate very severe. For the last question, 0 is 0 hours, 5 is one hour, and 10 is two or more hours.

To calculate the BASDAI score, the sum of the scores for questions one through four is added to the mean of the sum of the scores for questions five and six; the total is divided by five to calculate the disease activity score. A BASDAI score of at least 4.0 (out of a maximum of 10) indicates severe disease activity. An interactive webpage is available at http://basdai.com/BASDAI.php.

A.2) ASDAS — The Ankylosing Spondylitis Disease Activity Score (ASDAS) is a composite measure of disease activity. It incorporates information from the level of acute phase reactants and various clinical parameters (a patient global score and several questions also used for the BASDAI) [10]. An ASDAS calculator is available from the Assessment of SpondyloArthritis international Society (ASAS), together with information regarding its use and interpretation [11,12]. The ASDAS has the advantage of categorizing the disease activity as inactive, moderate, high, or very high. It also defines the degree of change in the score that would classify the response to treatment as a clinically important improvement or major improvement.

2. Physical examination 

In addition to the general physical examination, the physical examination in AS focuses on three areas:

  • axial joints, including the hips; 
  • peripheral joints; and 
  • entheses  (inflammation of the muscular or tendinous attachments to bone). 
Details of the assessment used in research are available (http://www.asas-group.org/ and [13-15]).The parameters below are those used in clinical practice. There is considerable variation among healthy individuals in the ranges of motion of the cervical, thoracic and lower spine. Thus, the threshold values given should be considered as estimates, rather than precise cut-off points between normal and abnormal [16].

  • Cervical spine – AS may cause forward stooping of the thoracic and cervical spine. The degree of flexion deformity is measured by asking the patient to stand erect with heels and buttocks against a wall and to extend his or her neck, while keeping the chin (mandible) horizontal in an attempt to touch the wall. Almost all normal individuals can touch the wall with the occiput (bony prominence at the back of the head). The distance between the occiput and the wall in a patient with AS reflects the degree of cervical deformity. In addition, the range of flexion, extension, lateral flexion, and rotation are also recorded.
  • Thoracic spine – The range of motion of the costovertebral joints is measured by the degree of chest expansion. Chest expansion is measured at the level of the fourth intercostal space or just below the breasts in females. The patients are instructed to raise their arms above their heads and to exert a maximal forced expiration followed by a maximal inspiration. Normal expansion is usually 2.5 cm.
  • Lower spine – Range of motion of the lower spine is tested in the sagittal plane by the Schober test, and in the coronal plane by the extent of lateral spinal flexion.
                               •Schober test – The Schober test measures the forward flexion of the lumbar spine. The patient stands erect; a mark is placed at the midpoint of a line joining the posterior superior iliac spines (dimples of Venus); another mark is made 10 cm above it in the midline. The patient then bends forward maximally without bending the knees and the distance is measured again. In normal individuals, the difference between the two measurements should exceed 4 cm.
                                •Lateral spinal flexion – With the patient standing erect with heel and back against a wall and knees and hands extended, the distance between the tip of the middle finger and the floor is measured. The patient is then instructed to bend sideways without bending the knees or lifting the heels. A second measurement is made and the difference between the two is recorded. The final result is the averaged measurements of right and left flexion. Normal is greater than 10 cm.

  • Hip joint – Hip involvement should be suspected when a patient shows an abnormal gait. It is verified by testing whether there is limitation of flexion, internal and external rotation, or pain at the extremes of these joint motions. Destructive disease at the hip joints in AS can lead to flexion deformities. However, a unilateral flexion deformity is frequently masked by compensatory motion in the spine. To eliminate this problem during physical examination, the patient is asked to lie supine and maximally flex one hip. If there is a flexion deformity of the contralateral hip, the knee of the contralateral limb will be raised and the degree of flexion deformity can be measured by the angle of the contralateral thigh.
  • Peripheral joint count – This involves a 44-joint count of the number of tender and/or swollen joints, including those of the ankles, feet and the sternoclavicular joints (page II40 in handbook in http://www.asas-group.org/) [17].
  • Sausage digit (dactylitis) – The number of digits that have sausage appearance should be determined.
  • Enthesitis count (inflammation of the muscular or tendinous attachments to bone) – At least 18 entheseal sites are assessed during clinical trials (http://www.asas-group.org/). In practice, the minimum should be at the heel, at the sites where the Achilles tendon and the plantar fascia are each attached to the calcaneus.
Extraarticular comorbidities — The methods of monitoring extraarticular involvement, such as uveitis, are based upon standards used for the organs being involved. AS is associated with increased risk of both osteoporosis and cardiovascular disease.

Recommendations for office practice — The following are a suggested set of indicators for minimum evaluation in initial visits and for monitoring disease progression:
  • Patient self-assessment of pain (1 to 10)
  • BASDAI and/or ASDAS, depending upon the availability of information on acute phase reactants, as the CRP or ESR value is required for calculating the ASDAS
  • BASFI or those daily activities that are limited by disease
  • Physician's global assessment on a scale of 1 (mild) to 4 (severe)
  • ESR or CRP
The frequency of monitoring depends on the course of symptoms, severity of disease and the therapies being used.

3. Acute phase reactants in the blood

The erythrocyte sedimentation rate (ESR) and/or C-reactive protein (CRP) are useful for monitoring disease activity, if they are elevated at baseline of treatment.

4. Imaging

In research, radiographic damage of the spine is assessed using lateral views of the cervical and lumbar spine, graded vertebra by vertebra, and scored, for example, using the Modified Stokes Ankylosing Spondylitis Spine Score (mSASSS) [18]. In clinical practice, it is useful to examine the anterosuperior and anteroinferior margins of the lumbar vertebrae on a lateral spine radiograph, and to record those with syndesmophytes. In advanced disease, plain radiographs of the spine will reveal "bamboo spine" with virtually complete fusion of the vertebral column. At that stage there is usually fusion of the sacroiliac joints. Because disease progression is slow, unless changes are suspected, it is not necessary to repeat radiographs of the vertebrae more than once every two years.

NONPHARMACOLOGICAL TREATMENT 

Patient education, exercise, and medications are all very important in the management of ankylosing spondylitis (AS) [3]. Patient education and support, in addition to that provided by the patient’s clinician, is frequently available from support groups.
  • Smoking cessation is recommended for everyone who smokes. It is a modifiable risk factor for poor functional outcome in AS [19]. 
  • Home exercises are effective, but supervised exercise programs or formal physical therapy can be of greater benefit [20]. Optimally, an initial evaluation and training by a physical therapist should be part of the therapeutic regimen. Exercises include postural training, range of motion stretching, recreational activities, and perhaps hydrotherapy. In addition, pain relief measures such as local heat or cold can be given a trial. At a minimum, patients with AS should participate in an unsupervised home exercise program [21]. Inpatient rehabilitation is rarely needed [22]. Many patients, however, do not exercise on a frequent basis, despite widespread appreciation of the benefits of exercise [23]. Video demonstrations of exercises tailored for ankylosing spondylitis are available for viewing through the National Ankylosing Spondylitis Society in the UK [24].Guide books and audio and video aids on compact disc and digital video disc are available from the Spondylitis Association of America. Even those patients who are doing well clinically with pharmacologic treatment will also benefit from education and exercise [25-28]. 

PHARMACOLOGIC THERAPY

Pharmacotherapy includes one or more of the following:
  • nonsteroidal antiinflammatory drugs (NSAIDs), 
  • analgesics, 
  • sulfasalazine (SSZ), and 
  • anti-tumor necrosis factor (TNF) agents. 
  • Systemic glucocorticoids have a limited role, but intraarticular injections may be helpful to some patients.

Nonsteroidal antiinflammatory drugs 

NSAIDs should be the first line of treatment for all symptomatic ankylosing spondylitis (AS) patients, unless contraindicated. In many patients, NSAIDs are the only medications required. About 70 to 80 percent of AS patients report substantial relief of their symptoms, including back pain and stiffness, with NSAIDs [29]. This is significantly more than the 15 percent observed in mechanical low back pain. NSAID use also results in decreased pain in peripheral joints [30]. There are two issues related to NSAID therapy in AS:
  • The choice of NSAID, including the possible role of selective cyclooxygenase (COX)-2 inhibitors – Initial anecdotal evidence suggested that indomethacin is the most effective NSAID [31]. However, multiple studies support the view that other NSAIDs are also effective [32]. Regardless of the NSAID used, the maximum dose is usually required. To assess the usefulness of a particular NSAID, it should be given at a sustained dose on a regular basis for at least four weeks. A table of recommended doses is available [29]. Short-term studies indicate that selective COX-2 inhibitors, such as celecoxib and etoricoxib, are as effective as nonselective NSAIDs [33-36].
  • Continuous versus on-demand therapy – AS patients who have persistent, active, symptomatic disease should use continuous NSAIDs to control symptoms. This approach is in agreement with the 2010 Assessment of SpondyloArthritis international Society (ASAS)/European League Against Rheumatism (EULAR) recommendations [1,2]. In addition, a randomized trial and several additional studies have shown that patients who use either continuous daily NSAIDs or high-dose NSAIDs regardless of symptoms may have a lower rate of radiographic progression in the spine [37-40].
As an example, in one trial involving 215 patients, which suggested that continuous daily use of NSAIDs may suppress radiographic progression, patients were randomly assigned to receive either continuous or on-demand NSAID therapy [37]. At two years, the degree of radiographic progression was statistically significantly less (by 73%) in the patients who were treated continuously with NSAIDs. Patients with more active inflammation seem to particularly benefit from continuous NSAID therapy. In one study, continuous NSAID therapy was especially effective in patients with an elevated erythrocyte sedimentation rate (ESR) [39]. In another study, continuous NSAIDs were most effective in patients with high C-reactive protein (CRP) and with the presence of syndesmophytes at baseline [40]. However, the exact clinical meaning of these findings remains to be determined.

Analgesics and Opiods 

When used alone, analgesics and opioids are seldom effective during active AS. However, they can be considered for residual pain, or when NSAIDs are contraindicated.

Tumor necrosis factor alpha antagonists

The anti-TNF-alpha agents available in Canada, the United States, and Europe for use in AS are infliximab, etanercept, adalimumab, and golimumab. Concomitant use of an immunomodulatory drug, such as methotrexate (MTX), is not required, as this provides no additional benefit but may increase the cost and the risk of adverse effects [41].

Overall efficacy — A 2007 meta-analysis indicated that all three of the anti-TNF alpha agents then available (adalimumab, etanercept, and infliximab) were similar in efficacy in patients with AS [42]. These medications have not been compared directly with each other, but indirect comparisons were unable to distinguish significant differences. At week 12 of trials, patients treated with the anti-TNF agents were 3.6-fold more likely, compared with those treated with placebo, to achieve 50 percent improvement (using a composite measure, the ASAS50). Approximately 80 percent of patients with AS respond to treatment with one of these agents and approximately one half get at least 50 percent improvement in a composite index, modified from the one adopted by the Ankylosing Spondylitis Assessment Group that includes [43]:
  • Patient global assessment
  • Patient assessment of pain
  • A functional assessment, such as ability to carry out certain activities with which the patient had difficulty prior to treatment
  • Degree of inflammation as assessed by morning stiffness
The clinical responses are typically rapid. Eighty percent of patients who experienced a 50 percent response according to BASDAI by 12 weeks did so within the first six weeks of treatment. The long-term effects of anti-TNF therapy appear to be durable, with continued efficacy for at least two years in two observational studies, and for five years in a third study [44-46]. Patients who do not respond to or do not tolerate one anti-TNF agent may respond to an alternate anti-TNF agent [47]. Why some patients but not others respond to these interventions is unknown.

Use of anti-TNF therapy may also decrease the frequency of recurrences of uveitis in patients with AS [48]. Despite their efficacy, the indiscriminate use of anti-TNF-alpha drugs is discouraged because of cost concerns and a lack of long-term safety data [49]. A guideline for selecting patients for whom anti-TNF therapy is indicated, dosing (amount and frequency), and monitoring for efficacy and adverse effects has been developed and updated by an international panel of clinicians with expertise in the treatment of AS [50-52]. A summary of guidelines is presented separately.

Effect on radiographic progression —  Whether long-term use of anti-TNF agents can halt the radiographic progression of the disease has been uncertain; such effects may not be evident until patients have been followed for several years. In a prospectively followed cohort of 334 patients treated with standard therapies for AS, in whom TNF inhibitors were administered to 201 patients, treatment with a TNF inhibitor was associated with a 50 percent reduction in the odds of radiographic progression (odds ratio 0.52, 95% CI 0.30-0.88) [53]. Such benefit was more evident in patients in whom these agents were begun earlier in the disease course and in whom the follow-up was at least four years. This finding was confirmed in a separate study in which patients with AS who were treated with the TNF inhibitor infliximab showed a similar rate of radiographic progression, during the first four years of follow-up, compared with a historical control group that did not receive a TNF inhibitor, but significantly less progression, compared with the historical controls, from years four to eight [54].

In contrast, analyses of imaging data in patients treated for only two years, comparing patients who received these drugs in several clinical trials with a historical cohort of patients who did not receive TNF inhibitors, failed to demonstrate that treatment with infliximab, adalimumab, etanercept, or golimumab [55] could halt radiologic progression of AS, despite evidence of clinical improvement and reduced spinal inflammation [56-58].

Use of TNF antagonists in early and advanced axial spondyloarthritis — Most studies of anti-tumor necrosis factor (TNF) therapy have recruited patients whose states of disease are neither very early nor very advanced. However, in trials that have evaluated these populations, the highest remission rates are observed with patients with very early disease [59], and patients with advanced disease have also benefited from anti-TNF therapy [60].

Predictors of response — The following parameters are possible predictors of a good response to TNF antagonists [61,62]:
  • Shorter disease duration, which is the best predictor
  • Elevated CRP
  • Young age
Improvement of greater than 50 percent may be seen in up to 80 percent of such patients [63].

Switching to a second TNF antagonist — A large open-label study addressed the effectiveness of switching to adalimumab when patients failed etanercept or infliximab because of either an inadequate response or adverse effect. Such patients were compared with those begun on adalimumab who had never received etanercept or infliximab. After 12 weeks, a Bath Ankylosing Spondylitis Disease Activity Index (BASDAI) 50 response was observed in 63 percent of patients who had never received TNF antagonists, and 40.8 percent of patients who had been treated with prior TNF antagonists [64]. In general, switching to another TNF antagonist is more useful for patients who have loss of response to the first one.

Stopping TNF antagonists when patients have obtained a major clinical response — The majority of patients will relapse soon after discontinuing the TNF antagonists [63]. It is possible that the dose or interval of treatment can be modified, but this has not been systematically studied.

Improvement in bone density — Some data suggest that use of anti-TNF biologic agents may be associated with improved bone density. Three studies utilizing anti-TNF biologic agents in the treatment of 75 patients with various forms of spondyloarthritis were reviewed [65]; when the changes in bone density from baseline to the end of observation were compared, patients receiving anti-TNF agents had increases in bone mineral density of 3.2 to 3.6 percent at the lumbar spine and 1.8 to 2.0 percent at the hip.

General side effects — Potential adverse effects such as an increased risk of reactivation of latent tuberculosis and the exacerbation or development of demyelinating disease must be considered when using these agents. Infliximab, etanercept, and adalimumab, when used in patients with AS, have been temporally associated with the development of psoriasis, including the palmoplantar pustulosis type [66,67]. Three cases of retropharyngeal abscess were also noted among 107 patients with a variety of spondyloarthritis (incidence of 1.6 per 100 patient years).

Infliximab 
Significant benefit has been noted in patients with axial as well as peripheral spondyloarthritis using the chimeric mouse-human monoclonal anti-TNF alpha antibody infliximab [66,68-72]. Infliximab use also resulted in improvement in the synovial histopathology [73]. Decreases in vascularity, thickness of the lining layer, and the degree of infiltration of synovium by neutrophils, macrophages, and T cells were seen; these findings have been confirmed in a subsequent study in two additional cohorts [74].

The largest study of this anti-TNF agent randomly assigned 357 North American and European patients to receive infliximab (5 mg/kg at week 0, 2, 6, 12, and 16) or placebo (The ASSERT trial) [75]. Patients with complete spinal ankylosis were excluded from this study, as were those with a recent infection, active or latent tuberculosis, or immunosuppression due to HIV/AIDS or solid organ transplantation. Patients with other diseases that could be exacerbated by infliximab, including demyelinating disorders and heart failure, were also excluded. MTX and other disease-modifying antirheumatic drugs (DMARDs) were prohibited. The primary outcome assessed at 24 weeks was a change that met ASAS criteria for a 20 percent improvement (ASAS20).

This and other controlled trials of infliximab utilized a 5 mg/kg dose [76,77], which is higher than the usual dose of 3 mg/kg for patients with rheumatoid arthritis. The 5 mg/kg dose appears to remain effective when used for periods of at least three years [46,78,79]. Whether a lower dose would be equally effective is uncertain. When 21 patients were given the 3 mg/kg dose in one of the observational studies cited above [71], infliximab appeared to be effective; however, three patients were given the 5 mg/kg dose after three months of therapy because of inadequate control of symptoms on the lower dose. Some patients who are not controlled with infusions every eight weeks may benefit from a reduced interval between treatments (eg, six weeks) [80].

The time to maximal response may be as long as six weeks, though some patients experience pain relief shortly after the first infusion. Our experience suggests that patients with longstanding disease are less likely to respond to infliximab therapy.

Most initial studies with infliximab in AS were carried out with patients who had definite sacroiliitis by plain radiography. A placebo-controlled randomized trial was carried out in a group of patients in whom the majority did not have plain radiographic sacroiliitis. These were inflammatory back pain patients with positive magnetic resonance imaging (MRI) changes of sacroiliitis and positive testing for HLA-B27. Infliximab was also effective in these patients [81]. In one case report, infliximab was effective in reversing symptoms of cauda equina syndrome [82]. No other interventions have previously been of benefit, but the utility of this approach in other patients has not yet been demonstrated.

MTX, although routinely used in combination with infliximab to treat rheumatoid arthritis, has not been found to be necessary or provide additional beneficial when infliximab is being used to treat AS [83]. The importance of coadministration of MTX on efficacy has not been studied with other TNF-alpha antagonists in AS.

Biosimilar for infliximab — The immunoglobulin (Ig)G1 chimeric human–murine monoclonal antibody, CT-P13 [84,85], was developed as an anti-TNF alpha monoclonal antibody biosimilar to the original (termed “innovator”) infliximab, which had first became commercially available in 1999. A “biosimilar” is defined by the World Health Organization (WHO) as a “biotherapeutic product which is similar in terms of quality, safety and efficacy to an already licensed reference biotherapeutic product” [86]; “similarity” is defined as the “absence of a relevant difference in the parameter of interest.” CT-P13 has an identical amino acid sequence to infliximab and is produced in the same type of cell line; it exhibits highly similar in vitro and in vivo pharmacodynamics, binding specificities and affinities, and other biologic and pharmacologic characteristics [84,85].

CT-P13 has shown equivalent clinical efficacy to the innovator infliximab in a small number of clinical trials, including the PLANETAS (Programme evaluating the autoimmune disease investigational drug CT-P13 in AS patients) trial, a randomized phase 1 trial in patients with AS, which also showed equivalent pharmacokinetic profiles of CT-P13 and infliximab [85]. In this trial, which involved 250 patients with active AS, treatment with CT-P13 demonstrated equivalent efficacy to infliximab up to week 30 (ASAS20 of 71 and 72 percent, odds ratio [OR] 0.91, 95% CI 0.51-1.62; ASAS40 of 52 and 47 percent, OR 1.19, 95% CI 0.70-2.00). Multiple other response measures were also comparable, and there were no differences detected in immunogenicity or safety. The initial trials of CT-P13 were the first clinical trials comparing a biosimilar to an innovator biologic agent for a rheumatic disease [87]. The worldwide availability of this agent is limited and will depend upon regulatory requirements in different regions and local patent laws and patent expiration dates [87].

Etanercept 
Several open studies and randomized trials have demonstrated the efficacy of etanercept in AS [88-93]. Limited data suggest that the symptomatic benefits of etanercept are quickly lost after cessation of therapy [94]. As with infliximab, etanercept use also resulted in improvement in synovial pathology in the peripheral joints [95].

The largest trial confirmed the efficacy of etanercept; it was a well-designed, international trial that randomly assigned 277 patients to receive etanercept (25 mg sc twice weekly) or placebo [91]. After 12 weeks of treatment, the proportion of patients achieving improvements of 20, 50, or 70 percent were about 60, 45, and 25 percent, respectively. Seventeen percent of patients achieved partial remission. These were significantly higher than the corresponding placebo values. In a subset of 40 subjects who had serial MRI examinations of the spine, there was a mean 54 percent decrease in T2-weighted signal in those treated with etanercept, suggesting decreased spinal inflammation, while the placebo group had a slight worsening (increase of 13 percent) [96]. Significant improvement was also noted in spinal and chest wall mobility. Hence, these are also useful follow-up parameters for patients receiving anti-TNF therapies. Only injection site reactions were more frequent in the etanercept group. In an unblinded extension of this study, beneficial effects were sustained during nearly two years of continued etanercept treatment [97].

Further support for the efficacy of etanercept in AS was provided by a multicenter randomized trial carried out at 14 European sites; 84 ankylosing spondylitis patients were randomly assigned to etanercept (25 mg SC twice a week) or placebo for 12 weeks [92]. Significant improvement was observed in the etanercept group by week 2, and significantly more patients achieved an ASAS50 at week 12 (48.9 versus 10.3 percent).
The results of the multiple randomized trials summarized above show convincingly that etanercept is effective for patients with ankylosing spondylitis. Weekly dosing with 50 mg appears to result in similar efficacy to 25 mg twice weekly [98,99].


Adalimumab
Adalimumab, a humanized anti-TNF monoclonal antibody, is also far more efficacious than placebo [59,100,101]. This was illustrated in a study that randomly assigned 315 patients in a 2:1 ratio to adalimumab (40 mg every other week by subcutaneous injection) or placebo injections [100]. The study population was predominantly Caucasian, male, and HLA-B27 positive with a mean duration of disease of 10 years. A significantly greater proportion of adalimumab-treated patients achieved the primary efficacy endpoint, an ASAS20 response at the end of 12 weeks (58 versus 21 percent). Patients who did not achieve an ASAS20 after 12 weeks were eligible to receive 40 mg weekly. At week 24 all patients were switched to or continued adalimumab 40 mg every other week in an open-label extension of the trial [45]. Of 61 patients treated weekly, ASAS20 responses were achieved after six weeks and after one year by 23 and 34 percent, respectively. At week 24 patients were switched to or continued adalimumab 40 mg every other week in an open-label extension of the trial [45]. At two years, for patients who were still receiving adalimumab, an ASAS20 was achieved by 64.5 percent. The proportion of these patients who experienced over 50 percent improvement in the BASDAI was 71 percent [45].

Thus, the advantage of adalimumab treatment was maintained for up to 24 weeks in the randomized phase of the study, and subsequently maintained for up to two years of follow-up in the open-label phase of the study.
Adalimumab has also been tested in randomized trials using patients with axial SpA who do not have radiographic signs of sacroiliitis (nr-axSpA). There is a significantly higher ASAS40 response at week 12 using adalimumab compared with placebo [102]. At two years, the frequency of patients with serious adverse events that were possibly drug-related was 5.5 percent. No cases of tuberculosis, heart failure, lupus-like symptoms, or demyelinating disease were reported.

Golimumab
Golimumab is a human anti-TNF alpha monoclonal antibody that is administered by subcutaneous injections once every four weeks. Efficacy for patients with AS appears similar to that of other anti-TNF agents. This was illustrated in a study that randomly assigned 356 patients to golimumab 50 mg, golimumab 100 mg, or placebo injections every four weeks [103]. After 14 weeks ASAS20 responses occurred in 59, 60, and 22 percent of patients in the three groups, respectively. These responses appear to be comparable to those of other TNF antagonists. The recommended dose is 50 mg subcutaneously every four weeks. The precautions and possible side effects are the same as those for the other FDA-approved TNF antagonists.

Certolizumab
An additional TNF inhibitor that is available for use in patients with active axial spondyloarthritis (SpA), including AS, is certolizumab pegol (CZP), a pegylated Fab fragment of a humanized monoclonal antibody directed against TNF-alpha that is administered by subcutaneous injection. In a randomized trial involving 325 patients with active axial SpA, including patients with AS and non-radiographic axial SpA, the patients who received CZP (200 mg every two weeks or 400 mg every four weeks) were significantly more likely to achieve improvement in disease activity compared with patients receiving placebo (ASAS20 58 and 64 versus 38 percent, ASAS40 43 and 49 versus 18 percent) [104]. Benefit was sustained at week 24. The response was similar to that seen with other TNF inhibitors, and no additional safety concerns were identified.

Disease modifying agents (DMARDs)
The only traditional DMARD regarded as potentially useful in AS is sulfasalazine.

Sulfasalazine (SSZ)
We typically use sulfasalazine in patients with peripheral arthritis, such as those with oligoarthritis of the knees and/or ankles, for whom a drug other than an anti-TNF agent is desirable, and prefer it to MTX in this setting. We discontinue sulfasalazine if there is no improvement after a four- to six-month trial at maximum dose. We also consider stopping the agent after a remission has been obtained, with its reinstitution in those who relapse. We do not recommend using sulfasalazine in those with only symptoms or signs of axial disease.

A 2006 meta-analysis that included data from 11 randomized clinical trials of SSZ concluded that it was significantly more effective than placebo in reducing spinal stiffness and in lowering the ESR [105]. However, most of the clinical trials of SSZ have been relatively small. Two of the larger trials involved 89 and 264 patients with AS, respectively [106,107]. In both of these trials, SSZ was more effective for the treatment of peripheral arthritis than for axial disease.

The use of SSZ has declined very considerably since the introduction of the TNF antagonists, which are more effective for both axial and peripheral disease [108,109]. Most experts in the care of patients with spondyloarthropathy recommend SSZ only for AS patients with peripheral arthritis, and many experts do not recommend its use if TNF antagonists are available [52]. In one study, SSZ was less effective in reducing disease activity compared with etanercept, whether or not peripheral joint involvement was present [110].

A typical dose of SSZ used in clinical trials was one 500 mg tablet daily for the first week, increasing by one tablet per day each subsequent week to a total of four to six tablets daily, divided and given twice a day. For those on the higher daily dose, the dose was reduced to four tablets if the higher dose was not tolerated. Approximately 60 percent of the SSZ-treated patients reported side effects, all reversible on withdrawal of the drug. The most common side effects were nausea, dizziness, headache, and rash. There is no guideline as to how patients on SSZ should be monitored for side effects. Leukopenia and neutropenia can develop quite suddenly, with an incidence varying from 1 to 5 percent. We recommend that blood counts be monitored at least every three months, and more frequently in the initial period. 

Methotrexate
Some studies have suggested that MTX may be effective in some patients with AS [111,112]. However, a 2006 meta-analysis of the efficacy of MTX in AS found no evidence of benefit in this disease [113]. Additionally, the combination of MTX and infliximab did not increase the efficacy or decrease the risk of adverse effects compared with infliximab alone [83,114,115]. Thus, there is currently a lack of evidence to support the use of MTX in the treatment of AS. Nevertheless, some rheumatologists do prescribe methotrexate for AS patients with peripheral arthritis.

Leflunomide
Limited data suggest that leflunomide is of little or no benefit for patients with AS [116,117].

Glucocorticoids
We suggest not using systemic glucocorticoids on a long-term basis for patients with AS, although very limited data suggest that relatively high doses of prednisolone may have some benefit for very short-term therapy. There is only one small randomized trial of oral glucocorticoid therapy in AS, involving a total of 39 patients [118]. This two-week trial compared the responses of NSAID-resistant patients who were randomly allocated to receive prednisolone (either 50 or 20 mg daily) or placebo. The proportion of patients achieving a 50 percent reduction in the BASDAI did not differ significantly between the groups, although there was a statistically significant greater improvement in the BASDAI score in the patients receiving prednisolone 50 mg but not 20 mg, compared with placebo. Similarly, pain scores in the patients receiving prednisolone 50 mg daily differed significantly from the controls, but not in the patients receiving 20 mg daily. Additional trials would be required to establish whether glucocorticoid therapy has a role in the treatment of AS, especially given the high dose of glucocorticoids required to achieve a significant effect, the small number of patients studied, and the short duration of therapy. In addition, patients with AS generally already have significant loss of bone density, which can be exacerbated by glucocorticoid therapy. 

Intraarticular injections, although never evaluated in a well-designed clinical trial to our knowledge, and local injections into painful plantar fasciae can be helpful for peripheral arthritis and plantar fasciitis, respectively. Local injections into the area of Achilles tendon are not recommended as rupture of the tendon may follow glucocorticoid infiltration at this site. Injection of long-acting glucocorticoids into the sacroiliac joints may be beneficial in patients who complain of marked pain at the sacroiliac joints that is unresponsive to systemic medications. Benefit of injection of long-acting glucocorticoid has been found in some but not all studies [119,120]. A double-blind study reported more than 70 percent relief in over 80 percent of injected sacroiliac joints [120]. Relief persisted for as long as six months or more after the injection and no complications were seen.

Other therapies possibly beneficial 
Several agents have shown evidence of potential benefit for AS but require further study. These include:
  • Biologics blocking the IL23-IL17 axis – The IL-23/IL17 axis is being studied as a major axis in the pathogenesis of SpA [121]. The results of two small proof-of-concept investigations suggest that blocking these mediators may be of benefit:
                         •Secukinumab – Secukinumab is an anti-interleukin (IL)-17A monoclonal antibody that has shown benefit in a randomized proof-of-concept trial in which 24 patients with moderate to severe AS (mean disease duration of 10 years) were assigned to receive secukinumab (10 mg/kg administered intravenously twice, three weeks apart) and 6 patients were assigned to receive placebo [122]. ASAS20 response rates at week six were significantly greater in patients allocated to receive secukinumab (59 versus 24 percent). Much larger trials of a substantially greater duration are required to assess the potential benefit of this approach. Secukinumab and other IL-17 inhibitors are of particular interest in AS because of the potential importance of IL-17 in the pathogenesis of the disease and possible benefit of IL-17 inhibition in patients with psoriatic arthritis.
                       •Ustekinumab - The other biologic against tested in AS that is directed against the IL23/IL17 axis is ustekinumab. It is an antibody directed against the p40 protein subunit shared by IL12 and IL23. In a 24-week open-label study the drug was administered to 20 patients with active AS, with substantial improvement in disease activity measures at 24 weeks; an ASAS40 was achieved in 65 percent of the patients, and greater than 50 percent improvement in the BASDAI was reported in 55 percent of the patients. Randomized trials are needed [123].
                       •Pamidronate – Pamidronate, a bisphosphonate, has been evaluated in the treatment of AS, because of some evidence of antiinflammatory activity and its effect on bone turnover [124-127]. Dose-dependent benefit of pamidronate treatment was demonstrated in a trial involving 84 patients with AS with a suboptimal response to NSAID therapy who were randomly assigned to one of two doses of pamidronate (60 mg/month or 10 mg/month), administered intravenously for six months [125]. Other observational studies have reported, at the most, only modest clinical benefit with pamidronate (60 mg/month), and several patients have experienced serious adverse effects with this regimen [126-128]. Additional data from randomized trials would be required before pamidronate could be considered for use in routine clinical practice for the treatment of AS.
                     •Thalidomide – The potential benefit of thalidomide has been examined in AS because of its immunomodulatory properties, including its effects on TNF [129-131]. A 2002 review of observational studies reported that among more than 50 patients treated with thalidomide, 68 percent improved and 19 percent withdrew from treatment due to lack of efficacy or adverse effects [131]. Randomized trials are needed to better define the role of thalidomide in the treatment of AS, and the seriousness of the potential side effects will need to be considered in determining whether it has a role in therapy.
                       •Rituximab – Rituximab, a monoclonal antibody that depletes B cells, has undergone preliminary study in 20 patients with active AS, based in part upon previous findings of dense B cell infiltration in subchondral bone of inflamed sacroiliac joints of patients with AS [132]. Significant efficacy at week 24 following initial intravenous administration of rituximab was observed in patients who were naive to TNF inhibitor therapy, but not in those who had already failed to respond to a TNF inhibitor (ASAS20 response 50 versus 30 percent, BASDAI50 response 50 versus 0 percent). Further study, including randomized trials, will be required to determine whether there is a role for rituximab in the treatment of AS.
                        •Apremilast – Apremilast, an oral phosphodiesterase 4 (PDE4) inhibitor with antiinflammatory and immunomodulatory effects, has shown some evidence of benefit in a randomized trial involving 38 patients with symptomatic AS and active disease on MRI [133]. There were trends suggesting greater improvement with apremilast, compared with placebo, in several clinical measures at 12 weeks, but these differences did not achieve statistical significance. The improved measures returned to their baseline values by four weeks following discontinuation of the trial medications. Larger trials of greater duration would be required to determine if this medication will be of benefit to patients with AS.
  • Other biologic agents probably not beneficial – Inhibitors of interleukin IL-12/23 and other agents have been the subject of investigation, but there is presently a lack of sufficient evidence for the use in AS of any biologics other than the TNF antagonists [1,134,135].  Several agents that have been shown to be effective in patients with rheumatoid arthritis or other forms of inflammatory arthritis have not been found effective in patients with ankylosing spondylitis. These include:
                        •Abatacept – Abatacept, a biologic agent which blocks T-cell costimulation, has not been found to be useful in AS in a 24-week open-label study involving 30 patients [136].
                        •Tocilizumab – Tocilizumab, a monoclonal antibody directed against IL-6 receptors, showed no benefit in composite clinical outcomes compared with placebo in a 12-week randomized trial involving 102 patients with AS, despite a decrease in C-reactive protein in the patients receiving tocilizumab [137].

SURGERY

Hip and spine surgery may be beneficial in selected patients with ankylosing spondylitis (AS). Clinicians and anesthetists should be cautioned that such patients may have reduced chest expansion and more rigid cervical spines.

Total hip arthroplasty

Total hip arthroplasty (THA, total hip replacement) is indicated in AS whenever there is severe, persistent pain or severe limitation in mobility and quality of life due to hip involvement. THA is more common in patients with early onset of disease, axial, and entheseal involvement [138]. Although there has been concern that patients with AS undergoing THA at a young, active age will experience a higher incidence of replacement failure compared with older, less active patients, excellent long-term results were found in a group of 340 patients with AS who had undergone initial THA at a mean age of 40 years [139]. The proportions of such hips that did not require revision at 10, 15, and 20 years following operation were 90, 78, and 64 percent, respectively. In another analysis of 181 hips, the survival of the prosthetic joint was 71 percent at 27 years [140]. Currently, the Assessment of SpondyloArthritis international Society (ASAS)/European League Against Rheumatism (EULAR) recommendations are that age not be a factor in decisions regarding THA.

Patients with AS may be at higher risk of developing heterotopic ossification following joint replacement, but this remains an unusual complication [141]. If, however, this complication has occurred following a prior joint arthroplasty, prophylactic therapy has been recommended, such as a nonsteroidal antiinflammatory drug (NSAID) beginning on the day of surgery or radiation therapy (pre- or postoperative).

Spinal surgery

Cervical fusion is indicated for the very small number of patients who develop atlantoaxial subluxation with impairment in neurologic function. This problem is managed in a fashion similar to that in rheumatoid arthritis. Wedge osteotomy is indicated in those patients who develop flexion deformities severe enough to impair the ability to look in a forward direction. Spinal surgeons should be consulted in the event of acute vertebral fractures, which should be suspected in patients who develop unusual neck or back pain, even in the absence of trauma. 

PROGNOSIS

Most ankylosing spondylitis (AS) patients with mild disease that is restricted to a small area of involvement are able to maintain almost full functional and employment capacity. However, a minority of patients develop severe skeletal restrictions or life-threatening extra-musculoskeletal complications. A questionnaire survey found that employment is affected in approximately 30 percent of the males with AS. However, the majority of patients can work well into their fifties [142].

Disease activity usually fluctuates in the individual patient, with symptoms usually persisting over decades. Approximately 1 percent of patients develop a stage of "burn-out" of disease activity and enter long-term remission [143].

Prognostic indicators

A number of prognostic indicators were identified before the use of biologics in patients with AS. One study, for example, evaluated 328 patients with spondyloarthritis; seven variables at entry correlated with increased disease severity [144]:
  • Hip arthritis – odds ratio (OR) 23
  • Sausage-like finger or toe – OR 8
  • Poor efficacy of NSAIDs – OR 8
  • High erythrocyte sedimentation rate (>30 mm/h) – OR 7
  • Limitation in range of motion of the lumbar spine – OR 7
  • Oligoarthritis – OR 4
  • Onset less than 16 years of age – OR 3
A mild outcome was likely if none of these factors was present at entry (sensitivity 93 percent, specificity 78 percent). However, a severe outcome was predictable if the hip were involved or if three factors were present (sensitivity 50 percent), and mild disease could be virtually excluded (specificity 98 percent). Patients with these features should therefore be followed at more frequent intervals.

A number of other factors have been associated with a poor outcome in patients with AS [19,145,146]. These include cigarette smoking, increasing severity of radiographic changes, active disease as assessed by a disease activity index, functional impairment as assessed by a self report, lower educational attainment, presence of other diseases related to spondyloarthritis (eg, psoriasis, inflammatory bowel disease), male sex, a history of uveitis, and occupational activities involving either dynamic flexibility (ability to quickly and repeatedly bend, twist, and stretch) or exposure to whole-body vibration (such as driving a truck or operating heavy equipment). Other predictors of poor outcome are HLA-B27, age, the presence of enthesitis, poor functional ability, and elevated C-reactive protein (CRP) [147].

Elevated CRP levels are associated with increased risk of radiographic progression both in the sacroiliac joints and in the spine [148,149]. Other factors associated with an increased degree of radiographic progression include the baseline severity of radiographic change and cigarette smoking [53,150].

Tumor necrosis factor inhibitors might be more effective in controlling inflammation when used early in the disease course, prior to the development of changes on plain radiographs [63]. Some evidence also suggests that such therapy may prevent or reduce radiographic progression.

Spinal cord injury
Patients with AS suffer an increased rate of spinal fractures. In those with AS, a majority of spinal cord injuries resulted from slips and falls, an event that was rarely the cause of cord damage in those without AS (53 versus 7 percent, respectively).

Cardiovascular risk and mortality
It is uncertain whether patients with AS have significantly higher cardiovascular risk compared with the general population. One longitudinal, population-based study found a significant increase in the risk of ischemic heart disease among young patients (ages 18 to 45) with newly diagnosed AS, compared with controls without AS, during three years of follow-up.[152].

Mortality among patients with AS admitted to the hospital for medical care is about 1.5 times higher than that of the general population [153]. Overall, mortality may be modestly increased. This was illustrated in a review of eight studies on mortality in AS that concluded that there was an increase in mortality rates compared with the general population (standardized mortality ratios ranging from 1.32 to 2.62) [154]. The major causes of death in patients with AS were infections, cancer, and diseases of the respiratory and cardiovascular systems [155]. In contrast to some other chronic rheumatic diseases (eg, rheumatoid arthritis and Sjögren's syndrome) the risk of developing lymphoma does not appear to be significantly increased in patients with AS [156].

SUMMARY AND RECOMMENDATIONS

  • Treatment of patients with ankylosing spondylitis (AS) must be individualized. The level of disease activity, presence of poor prognostic factors or concomitant illnesses, degree of functional impairment, patient's tolerance of the risk of adverse effects, expectations of treatment, and other psychosocial factors must be considered. The level of disease activity and the extent of improvement can be scored by using the Bath Ankylosing Spondylitis Disease Activity Index (BASDAI) questionnaire and/or the Ankylosing Spondylitis Disease Activity Score (ASDAS). 
  • We recommend use of a nonsteroidal antiinflammatory drug (NSAID) as initial therapy (Grade 1A). Any NSAID may be effective. Regular use for a period of four weeks allows the maximal effect of a given NSAID to be assessed. If some but not satisfactory improvement is noted, the NSAID may be continued indefinitely while other interventions are initiated
  • The NSAID can be supplemented by use of an analgesic; occasionally, low-potency opioids may be used. 
  • We recommend an exercise program for all patients (Grade 1B). Individual physical therapy may be needed by some. 
  • We suggest NOT using systemic glucocorticoids (Grade 2C). We suggest intraarticular glucocorticoids for persistent peripheral joint involvement, enthesitis at sites other than the Achilles tendon, and for pain of sacroiliitis (Grade 2B).
  • Traditional nonbiologic disease-modifying antirheumatic drugs (DMARDs) (eg, sulfasalazine, methotrexate, leflunomide, or penicillamine) are ineffective for those with axial disease. 
  • For patients with axial disease who do not respond to NSAIDs we recommend an anti-tumor necrosis factor (TNF) agent (Grade 1A). If there is failure to one anti-TNF agent, it is possible that an alternate anti-TNF agent can be effective. Prior to initiating anti-TNF therapy, active infection and latent tuberculosis should be excluded.
  • In patients with predominantly peripheral arthritis who do not respond adequately to NSAIDs and for whom a TNF inhibitor is unavailable or is contraindicated, we recommend sulfasalazine, unless there are contraindications (eg, allergy to sulfonamide antibiotics) to this drug (Grade 1A). Methotrexate is being used by some clinicians as an alternative.
  • There is a paucity of data to guide therapy if the treatments recommended above are ineffective or contraindicated.
  • Surgical interventions are used selectively. Total joint replacement may be necessary for those whose joints are already destroyed. Wedge osteotomy of the spine is reserved for those patients with severe spinal deformities, as in patients with neck flexion so pronounced that useful forward vision is difficult or impossible. Fusion of the atlantoaxial joint of the cervical spine is needed if there is significant neck or occipital pain or evidence of neurologic dysfunction due to C1-C2 (atlantoaxial) subluxation, as occurs in patients with rheumatoid arthritis.

Author: David T Yu, MD
Section Editor: Joachim Sieper, MD
Deputy Editor: Paul L Romain, MD
    
Source: UptoDate
All topics are updated as new evidence becomes available and the peer review process for UptoDate is complete. Literature review current through: Apr 2014. | This topic last updated: Feb 10, 2014. 

REFERENCES

  1. Braun J, van den Berg R, Baraliakos X, et al. 2010 update of the ASAS/EULAR recommendations for the management of ankylosing spondylitis. Ann Rheum Dis 2011; 70:896.
  2. Zochling J, van der Heijde D, Burgos-Vargas R, et al. ASAS/EULAR recommendations for the management of ankylosing spondylitis. Ann Rheum Dis 2006; 65:442.
  3. Smolen JS, Braun J, Dougados M, et al. Treating spondyloarthritis, including ankylosing spondylitis and psoriatic arthritis, to target: recommendations of an international task force. Ann Rheum Dis 2014; 73:6.
  4. Jang JH, Green CE, Assassi S, et al. The contribution of disease activity on functional limitations over time through psychological mediators: a 12-month longitudinal study in patients with ankylosing spondylitis. Rheumatology (Oxford) 2011; 50:2087.
  5. Calin A, Garrett S, Whitelock H, et al. A new approach to defining functional ability in ankylosing spondylitis: the development of the Bath Ankylosing Spondylitis Functional Index. J Rheumatol 1994; 21:2281.
  6. Bath Ankylosing Spondylitis Functional Index (BASFI) http://basdai.com/BASFI.php (Accessed on February 10, 2014).
  7. Doward LC, Spoorenberg A, Cook SA, et al. Development of the ASQoL: a quality of life instrument specific to ankylosing spondylitis. Ann Rheum Dis 2003; 62:20.
  8. Ankylosing spondylitis quality of life instrument (ASQol) http://ard.bmj.com/content/suppl/2002/12/20/62.1.20.DC1/62120Appendices.pdf (Accessed on February 10, 2014).
  9. Garrett S, Jenkinson T, Kennedy LG, et al. A new approach to defining disease status in ankylosing spondylitis: the Bath Ankylosing Spondylitis Disease Activity Index. J Rheumatol 1994; 21:2286.
  10. van der Heijde D, Lie E, Kvien TK, et al. ASDAS, a highly discriminatory ASAS-endorsed disease activity score in patients with ankylosing spondylitis. Ann Rheum Dis 2009; 68:1811.
  11. Assessment of SpondyloArthritis International Society. Ankylosing Spondylitis Disease Activity Score. http://www.asas-group.org/research.php?id=01#null (Accessed on October 15, 2013).
  12. Machado P, van der Heijde D. How to measure disease activity in axial spondyloarthritis? Curr Opin Rheumatol 2011; 23:339.
  13. Spondyloarthritis research and treatment network. www.spartangroup.org (Accessed on August 11, 2006).
  14. Gladman DD, Inman RD, Cook RJ, et al. International spondyloarthritis interobserver reliability exercise--the INSPIRE study: I. Assessment of spinal measures. J Rheumatol 2007; 34:1733.
  15. Gladman DD, Inman RD, Cook RJ, et al. International spondyloarthritis interobserver reliability exercise--the INSPIRE study: II. Assessment of peripheral joints, enthesitis, and dactylitis. J Rheumatol 2007; 34:1740.
  16. Chilton-Mitchell L, Martindale J, Hart A, Goodacre L. Normative values for the Bath Ankylosing Spondylitis Metrology Index in a UK population. Rheumatology (Oxford) 2013; 52:2086.
  17. Sieper J, Rudwaleit M, Baraliakos X, et al. The Assessment of SpondyloArthritis international Society (ASAS) handbook: a guide to assess spondyloarthritis. Ann Rheum Dis 2009; 68 Suppl 2:ii1.
  18. Wanders A, Landewé R, Spoorenberg A, et al. Scoring of radiographic progression in randomised clinical trials in ankylosing spondylitis: a preference for paired reading order. Ann Rheum Dis 2004; 63:1601.
  19. Doran MF, Brophy S, MacKay K, et al. Predictors of longterm outcome in ankylosing spondylitis. J Rheumatol 2003; 30:316.
  20. Dagfinrud H, Kvien TK, Hagen KB. Physiotherapy interventions for ankylosing spondylitis. Cochrane Database Syst Rev 2008; :CD002822.
  21. Elyan M, Khan MA. Does physical therapy still have a place in the treatment of ankylosing spondylitis? Curr Opin Rheumatol 2008; 20:282.
  22. Lubrano E, D'Angelo S, Parsons WJ, et al. Effectiveness of rehabilitation in active ankylosing spondylitis assessed by the ASAS response criteria. Rheumatology (Oxford) 2007; 46:1672.
  23. Passalent LA, Soever LJ, O'Shea FD, Inman RD. Exercise in ankylosing spondylitis: discrepancies between recommendations and reality. J Rheumatol 2010; 37:835.
  24. National Ankylosing Spondylitis Society. Back to Action online exercise videos. http://www.nass.co.uk/exercise/exercise-for-your-as/watch-exercise-videos/ (Accessed on October 15, 2013).
  25. Masiero S, Bonaldo L, Pigatto M, et al. Rehabilitation treatment in patients with ankylosing spondylitis stabilized with tumor necrosis factor inhibitor therapy: a randomized controlled trial. J Rheumatol 2011; 38:1335.
  26. Spadaro A, De Luca T, Massimiani MP, et al. Occupational therapy in ankylosing spondylitis: Short-term prospective study in patients treated with anti-TNF-alpha drugs. Joint Bone Spine 2008; 75:29.
  27. Dubey SG, Leeder J, Gaffney K. Physical therapy in anti-TNF treated patients with ankylosing spondylitis. Rheumatology (Oxford) 2008; 47:1100.
  28. Lubrano E, D'Angelo S, Spadaro A, et al. Rehabilitation for ankylosing spondylitis in the era of biologics: any room left for this treatment? J Rheumatol 2011; 38:1228.
  29. Song IH, Poddubnyy DA, Rudwaleit M, Sieper J. Benefits and risks of ankylosing spondylitis treatment with nonsteroidal antiinflammatory drugs. Arthritis Rheum 2008; 58:929.
  30. Zochling J, van der Heijde D, Dougados M, Braun J. Current evidence for the management of ankylosing spondylitis: a systematic literature review for the ASAS/EULAR management recommendations in ankylosing spondylitis. Ann Rheum Dis 2006; 65:423.
  31. Calin A, Elswood J. A prospective nationwide cross-sectional study of NSAID usage in 1331 patients with ankylosing spondylitis. J Rheumatol 1990; 17:801.
  32. Sidiropoulos PI, Hatemi G, Song IH, et al. Evidence-based recommendations for the management of ankylosing spondylitis: systematic literature search of the 3E Initiative in Rheumatology involving a broad panel of experts and practising rheumatologists. Rheumatology (Oxford) 2008; 47:355.
  33. van der Heijde D, Baraf HS, Ramos-Remus C, et al. Evaluation of the efficacy of etoricoxib in ankylosing spondylitis: results of a fifty-two-week, randomized, controlled study. Arthritis Rheum 2005; 52:1205.
  34. Barkhuizen A, Steinfeld S, Robbins J, et al. Celecoxib is efficacious and well tolerated in treating signs and symptoms of ankylosing spondylitis. J Rheumatol 2006; 33:1805.
  35. Dougados M, Béhier JM, Jolchine I, et al. Efficacy of celecoxib, a cyclooxygenase 2-specific inhibitor, in the treatment of ankylosing spondylitis: a six-week controlled study with comparison against placebo and against a conventional nonsteroidal antiinflammatory drug. Arthritis Rheum 2001; 44:180.
  36. Sieper J, Klopsch T, Richter M, et al. Comparison of two different dosages of celecoxib with diclofenac for the treatment of active ankylosing spondylitis: results of a 12-week randomised, double-blind, controlled study. Ann Rheum Dis 2008; 67:323.
  37. Wanders A, Heijde Dv, Landewé R, et al. Nonsteroidal antiinflammatory drugs reduce radiographic progression in patients with ankylosing spondylitis: a randomized clinical trial. Arthritis Rheum 2005; 52:1756.
  38. Haroon N, Kim TH, Inman RD. NSAIDs and radiographic progression in ankylosing spondylitis Bagging big game with small arms? Ann Rheum Dis 2012; 71:1593.
  39. Kroon F, Landewé R, Dougados M, van der Heijde D. Continuous NSAID use reverts the effects of inflammation on radiographic progression in patients with ankylosing spondylitis. Ann Rheum Dis 2012; 71:1623.
  40. Poddubnyy D, Rudwaleit M, Haibel H, et al. Effect of non-steroidal anti-inflammatory drugs on radiographic spinal progression in patients with axial spondyloarthritis: results from the German Spondyloarthritis Inception Cohort. Ann Rheum Dis 2012; 71:1616.
  41. Sieper J. Developments in the scientific and clinical understanding of the spondyloarthritides. Arthritis Res Ther 2009; 11:208.
  42. McLeod C, Bagust A, Boland A, et al. Adalimumab, etanercept and infliximab for the treatment of ankylosing spondylitis: a systematic review and economic evaluation. Health Technol Assess 2007; 11:1.
  43. Anderson JJ, Baron G, van der Heijde D, et al. Ankylosing spondylitis assessment group preliminary definition of short-term improvement in ankylosing spondylitis. Arthritis Rheum 2001; 44:1876.
  44. Baraliakos X, Brandt J, Listing J, et al. Outcome of patients with active ankylosing spondylitis after two years of therapy with etanercept: clinical and magnetic resonance imaging data. Arthritis Rheum 2005; 53:856.
  45. van der Heijde D, Schiff MH, Sieper J, et al. Adalimumab effectiveness for the treatment of ankylosing spondylitis is maintained for up to 2 years: long-term results from the ATLAS trial. Ann Rheum Dis 2009; 68:922.
  46. Braun J, Baraliakos X, Listing J, et al. Persistent clinical efficacy and safety of anti-tumour necrosis factor alpha therapy with infliximab in patients with ankylosing spondylitis over 5 years: evidence for different types of response. Ann Rheum Dis 2008; 67:340.
  47. Cantini F, Niccoli L, Benucci M, et al. Switching from infliximab to once-weekly administration of 50 mg etanercept in resistant or intolerant patients with ankylosing spondylitis: results of a fifty-four-week study. Arthritis Rheum 2006; 55:812.
  48. Braun J, Baraliakos X, Listing J, Sieper J. Decreased incidence of anterior uveitis in patients with ankylosing spondylitis treated with the anti-tumor necrosis factor agents infliximab and etanercept. Arthritis Rheum 2005; 52:2447.
  49. Jois RN, Gaffney K, Keat A. Anti-tumour necrosis factor therapy for ankylosing spondylitis--unresolved issues. Rheumatology (Oxford) 2007; 46:899.
  50. Braun J, Pham T, Sieper J, et al. International ASAS consensus statement for the use of anti-tumour necrosis factor agents in patients with ankylosing spondylitis. Ann Rheum Dis 2003; 62:817.
  51. Braun J, Davis J, Dougados M, et al. First update of the international ASAS consensus statement for the use of anti-TNF agents in patients with ankylosing spondylitis. Ann Rheum Dis 2006; 65:316.
  52. van der Heijde D, Sieper J, Maksymowych WP, et al. 2010 Update of the international ASAS recommendations for the use of anti-TNF agents in patients with axial spondyloarthritis. Ann Rheum Dis 2011; 70:905.
  53. Haroon N, Inman RD, Learch TJ, et al. The impact of tumor necrosis factor α inhibitors on radiographic progression in ankylosing spondylitis. Arthritis Rheum 2013; 65:2645.
  54. Baraliakos X, Haibel H, Listing J, et al. Continuous long-term anti-TNF therapy does not lead to an increase in the rate of new bone formation over 8 years in patients with ankylosing spondylitis. Ann Rheum Dis 2014; 73:710.
  55. Braun J, Baraliakos X, Hermann KG, et al. The effect of two golimumab doses on radiographic progression in ankylosing spondylitis: results through 4 years of the GO-RAISE trial. Ann Rheum Dis 2013.
  56. van der Heijde D, Landewé R, Baraliakos X, et al. Radiographic findings following two years of infliximab therapy in patients with ankylosing spondylitis. Arthritis Rheum 2008; 58:3063.
  57. van der Heijde D, Salonen D, Weissman BN, et al. Assessment of radiographic progression in the spines of patients with ankylosing spondylitis treated with adalimumab for up to 2 years. Arthritis Res Ther 2009; 11:R127.
  58. van der Heijde D, Landewé R, Einstein S, et al. Radiographic progression of ankylosing spondylitis after up to two years of treatment with etanercept. Arthritis Rheum 2008; 58:1324.
  59. Haibel H, Rudwaleit M, Listing J, et al. Efficacy of adalimumab in the treatment of axial spondylarthritis without radiographically defined sacroiliitis: results of a twelve-week randomized, double-blind, placebo-controlled trial followed by an open-label extension up to week fifty-two. Arthritis Rheum 2008; 58:1981.
  60. Dougados M, Braun J, Szanto S, et al. Efficacy of etanercept on rheumatic signs and pulmonary function tests in advanced ankylosing spondylitis: results of a randomised double-blind placebo-controlled study (SPINE). Ann Rheum Dis 2011; 70:799.
  61. Rudwaleit M, Listing J, Brandt J, et al. Prediction of a major clinical response (BASDAI 50) to tumour necrosis factor alpha blockers in ankylosing spondylitis. Ann Rheum Dis 2004; 63:665.
  62. Rudwaleit M, Claudepierre P, Wordsworth P, et al. Effectiveness, safety, and predictors of good clinical response in 1250 patients treated with adalimumab for active ankylosing spondylitis. J Rheumatol 2009; 36:801.
  63. Haibel H, Sieper J. Editorial review: how early should ankylosing spondylitis be treated with a tumor necrosis factor-blocker? Curr Opin Rheumatol 2010; 22:388.
  64. Rudwaleit M, Van den Bosch F, Kron M, et al. Effectiveness and safety of adalimumab in patients with ankylosing spondylitis or psoriatic arthritis and history of anti-tumor necrosis factor therapy. Arthritis Res Ther 2010; 12:R117.
  65. Barnabe C, Hanley DA. Effect of tumor necrosis factor alpha inhibition on bone density and turnover markers in patients with rheumatoid arthritis and spondyloarthropathy. Semin Arthritis Rheum 2009; 39:116.
  66. Baeten D, Kruithof E, Van den Bosch F, et al. Systematic safety follow up in a cohort of 107 patients with spondyloarthropathy treated with infliximab: a new perspective on the role of host defence in the pathogenesis of the disease? Ann Rheum Dis 2003; 62:829.
  67. Ko JM, Gottlieb AB, Kerbleski JF. Induction and exacerbation of psoriasis with TNF-blockade therapy: a review and analysis of 127 cases. J Dermatolog Treat 2009; 20:100.
  68. Brandt J, Haibel H, Cornely D, et al. Successful treatment of active ankylosing spondylitis with the anti-tumor necrosis factor alpha monoclonal antibody infliximab. Arthritis Rheum 2000; 43:1346.
  69. Van den Bosch F, Kruithof E, Baeten D, et al. Effects of a loading dose regimen of three infusions of chimeric monoclonal antibody to tumour necrosis factor alpha (infliximab) in spondyloarthropathy: an open pilot study. Ann Rheum Dis 2000; 59:428.
  70. Stone M, Salonen D, Lax M, et al. Clinical and imaging correlates of response to treatment with infliximab in patients with ankylosing spondylitis. J Rheumatol 2001; 28:1605.
  71. Maksymowych WP, Jhangri GS, Lambert RG, et al. Infliximab in ankylosing spondylitis: a prospective observational inception cohort analysis of efficacy and safety. J Rheumatol 2002; 29:959.
  72. Temekonidis TI, Alamanos Y, Nikas SN, et al. Infliximab therapy in patients with ankylosing spondylitis: an open label 12 month study. Ann Rheum Dis 2003; 62:1218.
  73. Baeten D, Kruithof E, Van den Bosch F, et al. Immunomodulatory effects of anti-tumor necrosis factor alpha therapy on synovium in spondylarthropathy: histologic findings in eight patients from an open-label pilot study. Arthritis Rheum 2001; 44:186.
  74. Kruithof E, Baeten D, Van den Bosch F, et al. Histological evidence that infliximab treatment leads to downregulation of inflammation and tissue remodelling of the synovial membrane in spondyloarthropathy. Ann Rheum Dis 2005; 64:529.
  75. van der Heijde D, Dijkmans B, Geusens P, et al. Efficacy and safety of infliximab in patients with ankylosing spondylitis: results of a randomized, placebo-controlled trial (ASSERT). Arthritis Rheum 2005; 52:582.
  76. Braun J, Brandt J, Listing J, et al. Treatment of active ankylosing spondylitis with infliximab: a randomised controlled multicentre trial. Lancet 2002; 359:1187.
  77. Van Den Bosch F, Kruithof E, Baeten D, et al. Randomized double-blind comparison of chimeric monoclonal antibody to tumor necrosis factor alpha (infliximab) versus placebo in active spondylarthropathy. Arthritis Rheum 2002; 46:755.
  78. Braun J, Brandt J, Listing J, et al. Two year maintenance of efficacy and safety of infliximab in the treatment of ankylosing spondylitis. Ann Rheum Dis 2005; 64:229.
  79. Sieper J, Baraliakos X, Listing J, et al. Persistent reduction of spinal inflammation as assessed by magnetic resonance imaging in patients with ankylosing spondylitis after 2 yrs of treatment with the anti-tumour necrosis factor agent infliximab. Rheumatology (Oxford) 2005; 44:1525.
  80. Collantes-Estevez E, Muñoz-Villanueva MC, Zarco P, et al. Effectiveness of reducing infliximab dose interval in non-responder patients with refractory spondyloarthropathies. An open extension of a multicentre study. Rheumatology (Oxford) 2005; 44:1555.
  81. Barkham N, Keen HI, Coates LC, et al. Clinical and imaging efficacy of infliximab in HLA-B27-Positive patients with magnetic resonance imaging-determined early sacroiliitis. Arthritis Rheum 2009; 60:946.
  82. Cornec D, Devauchelle Pensec V, Joulin SJ, Saraux A. Dramatic efficacy of infliximab in cauda equina syndrome complicating ankylosing spondylitis. Arthritis Rheum 2009; 60:1657.
  83. Breban M, Ravaud P, Claudepierre P, et al. Maintenance of infliximab treatment in ankylosing spondylitis: results of a one-year randomized controlled trial comparing systematic versus on-demand treatment. Arthritis Rheum 2008; 58:88.
  84. Yoo DH, Hrycaj P, Miranda P, et al. A randomised, double-blind, parallel-group study to demonstrate equivalence in efficacy and safety of CT-P13 compared with innovator infliximab when coadministered with methotrexate in patients with active rheumatoid arthritis: the PLANETRA study. Ann Rheum Dis 2013; 72:1613.
  85. Park W, Hrycaj P, Jeka S, et al. A randomised, double-blind, multicentre, parallel-group, prospective study comparing the pharmacokinetics, safety, and efficacy of CT-P13 and innovator infliximab in patients with ankylosing spondylitis: the PLANETAS study. Ann Rheum Dis 2013; 72:1605.
  86. World Health Organization. Expert Committee on Biological Standardization. Guidelines on evaluation of similar biotherapeutic products. Geneva, 2009. http://www.who.int/biologicals/areas/biological_ (Accessed on September 28, 2013).
  87. Kay J, Smolen JS. Biosimilars to treat inflammatory arthritis: the challenge of proving identity. Ann Rheum Dis 2013; 72:1589.
  88. Barthel HR. Rapid remission of treatment-resistant ankylosing spondylitis with etanercept--a drug for refractory ankylosing spondylitis? Arthritis Rheum 2001; 45:404.
  89. Marzo-Ortega H, McGonagle D, O'Connor P, Emery P. Efficacy of etanercept in the treatment of the entheseal pathology in resistant spondylarthropathy: a clinical and magnetic resonance imaging study. Arthritis Rheum 2001; 44:2112.
  90. Gorman JD, Sack KE, Davis JC Jr. Treatment of ankylosing spondylitis by inhibition of tumor necrosis factor alpha. N Engl J Med 2002; 346:1349.
  91. Davis JC Jr, Van Der Heijde D, Braun J, et al. Recombinant human tumor necrosis factor receptor (etanercept) for treating ankylosing spondylitis: a randomized, controlled trial. Arthritis Rheum 2003; 48:3230.
  92. Calin A, Dijkmans BA, Emery P, et al. Outcomes of a multicentre randomised clinical trial of etanercept to treat ankylosing spondylitis. Ann Rheum Dis 2004; 63:1594.
  93. Song IH, Hermann K, Haibel H, et al. Effects of etanercept versus sulfasalazine in early axial spondyloarthritis on active inflammatory lesions as detected by whole-body MRI (ESTHER): a 48-week randomised controlled trial. Ann Rheum Dis 2011; 70:590.
  94. Brandt J, Khariouzov A, Listing J, et al. Six-month results of a double-blind, placebo-controlled trial of etanercept treatment in patients with active ankylosing spondylitis. Arthritis Rheum 2003; 48:1667.
  95. Kruithof E, De Rycke L, Roth J, et al. Immunomodulatory effects of etanercept on peripheral joint synovitis in the spondylarthropathies. Arthritis Rheum 2005; 52:3898.
  96. Baraliakos X, Davis J, Tsuji W, Braun J. Magnetic resonance imaging examinations of the spine in patients with ankylosing spondylitis before and after therapy with the tumor necrosis factor alpha receptor fusion protein etanercept. Arthritis Rheum 2005; 52:1216.
  97. Davis JC Jr, van der Heijde DM, Braun J, et al. Efficacy and safety of up to 192 weeks of etanercept therapy in patients with ankylosing spondylitis. Ann Rheum Dis 2008; 67:346.
  98. van der Heijde D, Da Silva JC, Dougados M, et al. Etanercept 50 mg once weekly is as effective as 25 mg twice weekly in patients with ankylosing spondylitis. Ann Rheum Dis 2006; 65:1572.
  99. Braun J, McHugh N, Singh A, et al. Improvement in patient-reported outcomes for patients with ankylosing spondylitis treated with etanercept 50 mg once-weekly and 25 mg twice-weekly. Rheumatology (Oxford) 2007; 46:999.
  100. van der Heijde D, Kivitz A, Schiff MH, et al. Efficacy and safety of adalimumab in patients with ankylosing spondylitis: results of a multicenter, randomized, double-blind, placebo-controlled trial. Arthritis Rheum 2006; 54:2136.
  101. Lambert RG, Salonen D, Rahman P, et al. Adalimumab significantly reduces both spinal and sacroiliac joint inflammation in patients with ankylosing spondylitis: a multicenter, randomized, double-blind, placebo-controlled study. Arthritis Rheum 2007; 56:4005.
  102. Sieper J, van der Heijde D, Dougados M, et al. Efficacy and safety of adalimumab in patients with non-radiographic axial spondyloarthritis: results of a randomised placebo-controlled trial (ABILITY-1). Ann Rheum Dis 2013; 72:815.
  103. Inman RD, Davis JC Jr, Heijde Dv, et al. Efficacy and safety of golimumab in patients with ankylosing spondylitis: results of a randomized, double-blind, placebo-controlled, phase III trial. Arthritis Rheum 2008; 58:3402.
  104. Landewé R, Braun J, Deodhar A, et al. Efficacy of certolizumab pegol on signs and symptoms of axial spondyloarthritis including ankylosing spondylitis: 24-week results of a double-blind randomised placebo-controlled Phase 3 study. Ann Rheum Dis 2014; 73:39.
  105. Chen J, Liu C. Is sulfasalazine effective in ankylosing spondylitis? A systematic review of randomized controlled trials. J Rheumatol 2006; 33:722.
  106. Dougados M, vam der Linden S, Leirisalo-Repo M, et al. Sulfasalazine in the treatment of spondylarthropathy. A randomized, multicenter, double-blind, placebo-controlled study. Arthritis Rheum 1995; 38:618.
  107. Clegg DO, Reda DJ, Abdellatif M. Comparison of sulfasalazine and placebo for the treatment of axial and peripheral articular manifestations of the seronegative spondylarthropathies: a Department of Veterans Affairs cooperative study. Arthritis Rheum 1999; 42:2325.
  108. Ward MM. Sulfasalazine for the treatment of ankylosing spondylitis: relic or niche medication? Arthritis Rheum 2011; 63:1472.
  109. Braun J, van der Horst-Bruinsma IE, Huang F, et al. Clinical efficacy and safety of etanercept versus sulfasalazine in patients with ankylosing spondylitis: a randomized, double-blind trial. Arthritis Rheum 2011; 63:1543.
  110. Braun J, Pavelka K, Ramos-Remus C, et al. Clinical efficacy of etanercept versus sulfasalazine in ankylosing spondylitis subjects with peripheral joint involvement. J Rheumatol 2012; 39:836.
  111. Sampaio-Barros PD, Costallat LT, Bertolo MB, et al. Methotrexate in the treatment of ankylosing spondylitis. Scand J Rheumatol 2000; 29:160.
  112. Gonzalez-Lopez L, Garcia-Gonzalez A, Vazquez-Del-Mercado M, et al. Efficacy of methotrexate in ankylosing spondylitis: a randomized, double blind, placebo controlled trial. J Rheumatol 2004; 31:1568.
  113. Chen J, Liu C, Lin J. Methotrexate for ankylosing spondylitis. Cochrane Database Syst Rev 2006; :CD004524.
  114. Marzo-Ortega H, McGonagle D, Jarrett S, et al. Infliximab in combination with methotrexate in active ankylosing spondylitis: a clinical and imaging study. Ann Rheum Dis 2005; 64:1568.
  115. Li EK, Griffith JF, Lee VW, et al. Short-term efficacy of combination methotrexate and infliximab in patients with ankylosing spondylitis: a clinical and magnetic resonance imaging correlation. Rheumatology (Oxford) 2008; 47:1358.
  116. van Denderen JC, van der Paardt M, Nurmohamed MT, et al. Double blind, randomised, placebo controlled study of leflunomide in the treatment of active ankylosing spondylitis. Ann Rheum Dis 2005; 64:1761.
  117. Haibel H, Rudwaleit M, Braun J, Sieper J. Six months open label trial of leflunomide in active ankylosing spondylitis. Ann Rheum Dis 2005; 64:124.
  118. Haibel H, Fendler C, Listing J, et al. Efficacy of oral prednisolone in active ankylosing spondylitis: results of a double-blind, randomised, placebo-controlled short-term trial. Ann Rheum Dis 2014; 73:243.
  119. Hanly JG, Mitchell M, MacMillan L, et al. Efficacy of sacroiliac corticosteroid injections in patients with inflammatory spondyloarthropathy: results of a 6 month controlled study. J Rheumatol 2000; 27:719.
  120. Maugars Y, Mathis C, Berthelot JM, et al. Assessment of the efficacy of sacroiliac corticosteroid injections in spondylarthropathies: a double-blind study. Br J Rheumatol 1996; 35:767.
  121. Smith JA, Colbert RA. The IL-23/IL-17 axis in spondyloarthritis pathogenesis: Th17 and beyond. Arthritis Rheum 2013.
  122. Baeten D, Baraliakos X, Braun J, et al. Anti-interleukin-17A monoclonal antibody secukinumab in treatment of ankylosing spondylitis: a randomised, double-blind, placebo-controlled trial. Lancet 2013; 382:1705.
  123. Poddubnyy D, Hermann KG, Callhoff J, et al. Ustekinumab for the treatment of patients with active ankylosing spondylitis: results of a 28-week, prospective, open-label, proof-of-concept study (TOPAS). Ann Rheum Dis 2014; 73:817.
  124. Maksymowych WP, Lambert R, Jhangri GS, et al. Clinical and radiological amelioration of refractory peripheral spondyloarthritis by pulse intravenous pamidronate therapy. J Rheumatol 2001; 28:144.
  125. Maksymowych WP, Jhangri GS, Fitzgerald AA, et al. A six-month randomized, controlled, double-blind, dose-response comparison of intravenous pamidronate (60 mg versus 10 mg) in the treatment of nonsteroidal antiinflammatory drug-refractory ankylosing spondylitis. Arthritis Rheum 2002; 46:766.
  126. Cairns AP, Wright SA, Taggart AJ, et al. An open study of pulse pamidronate treatment in severe ankylosing spondylitis, and its effect on biochemical markers of bone turnover. Ann Rheum Dis 2005; 64:338.
  127. Grover R, Shankar S, Aneja R, et al. Treatment of ankylosing spondylitis with pamidronate: an open label study. Ann Rheum Dis 2006; 65:688.
  128. Toussirot E, Wendling D. Antiinflammatory treatment with bisphosphonates in ankylosing spondylitis. Curr Opin Rheumatol 2007; 19:340.
  129. Breban M, Gombert B, Amor B, Dougados M. Efficacy of thalidomide in the treatment of refractory ankylosing spondylitis. Arthritis Rheum 1999; 42:580.
  130. Huang F, Gu J, Zhao W, et al. One-year open-label trial of thalidomide in ankylosing spondylitis. Arthritis Rheum 2002; 47:249.
  131. Huang F, Wei JC, Breban M. Thalidomide in ankylosing spondylitis. Clin Exp Rheumatol 2002; 20:S158.
  132. Song IH, Heldmann F, Rudwaleit M, et al. Different response to rituximab in tumor necrosis factor blocker-naive patients with active ankylosing spondylitis and in patients in whom tumor necrosis factor blockers have failed: a twenty-four-week clinical trial. Arthritis Rheum 2010; 62:1290.
  133. Pathan E, Abraham S, Van Rossen E, et al. Efficacy and safety of apremilast, an oral phosphodiesterase 4 inhibitor, in ankylosing spondylitis. Ann Rheum Dis 2013; 72:1475.
  134. Song IH, Poddubnyy D. New treatment targets in ankylosing spondylitis and other spondyloarthritides. Curr Opin Rheumatol 2011; 23:346.
  135. Kiltz U, Heldmann F, Baraliakos X, Braun J. Treatment of ankylosing spondylitis in patients refractory to TNF-inhibition: are there alternatives? Curr Opin Rheumatol 2012; 24:252.
  136. Song IH, Heldmann F, Rudwaleit M, et al. Treatment of active ankylosing spondylitis with abatacept: an open-label, 24-week pilot study. Ann Rheum Dis 2011; 70:1108.
  137. Sieper J, Porter-Brown B, Thompson L, et al. Assessment of short-term symptomatic efficacy of tocilizumab in ankylosing spondylitis: results of randomised, placebo-controlled trials. Ann Rheum Dis 2014; 73:95.
  138. Vander Cruyssen B, Muñoz-Gomariz E, Font P, et al. Hip involvement in ankylosing spondylitis: epidemiology and risk factors associated with hip replacement surgery. Rheumatology (Oxford) 2010; 49:73.
  139. Sweeney S, Gupta R, Taylor G, Calin A. Total hip arthroplasty in ankylosing spondylitis: outcome in 340 patients. J Rheumatol 2001; 28:1862.
  140. Joshi AB, Markovic L, Hardinge K, Murphy JC. Total hip arthroplasty in ankylosing spondylitis: an analysis of 181 hips. J Arthroplasty 2002; 17:427.
  141. Nilsson OS, Persson PE. Heterotopic bone formation after joint replacement. Curr Opin Rheumatol 1999; 11:127.
  142. Koh WH, Jones SD, Garrett Sl, et al. Risks and determinants of unemployment in ankylosing spondylitis (abstract). Arthritis Rheum 1994; 37:263.
  143. Kennedy LG, Edmunds L, Calin A. The natural history of ankylosing spondylitis. Does it burn out? J Rheumatol 1993; 20:688.
  144. Amor B, Santos RS, Nahal R, et al. Predictive factors for the longterm outcome of spondyloarthropathies. J Rheumatol 1994; 21:1883.
  145. Pradeep DJ, Keat A, Gaffney K. Predicting outcome in ankylosing spondylitis. Rheumatology (Oxford) 2008; 47:942.
  146. Ward MM, Reveille JD, Learch TJ, et al. Occupational physical activities and long-term functional and radiographic outcomes in patients with ankylosing spondylitis. Arthritis Rheum 2008; 59:822.
  147. Vastesaeger N, van der Heijde D, Inman RD, et al. Predicting the outcome of ankylosing spondylitis therapy. Ann Rheum Dis 2011; 70:973.
  148. Poddubnyy D, Rudwaleit M, Haibel H, et al. Rates and predictors of radiographic sacroiliitis progression over 2 years in patients with axial spondyloarthritis. Ann Rheum Dis 2011; 70:1369.
  149. Poddubnyy D, Haibel H, Listing J, et al. Baseline radiographic damage, elevated acute-phase reactant levels, and cigarette smoking status predict spinal radiographic progression in early axial spondylarthritis. Arthritis Rheum 2012; 64:1388.
  150. Poddubnyy D, Haibel H, Listing J, et al. Cigarette smoking has a dose-dependent impact on progression of structural damage in the spine in patients with axial spondyloarthritis: results from the GErman SPondyloarthritis Inception Cohort (GESPIC). Ann Rheum Dis 2013; 72:1430.
  151. Alaranta H, Luoto S, Konttinen YT. Traumatic spinal cord injury as a complication to ankylosing spondylitis. An extended report. Clin Exp Rheumatol 2002; 20:66.
  152. Huang YP, Wang YH, Pan SL. Increased risk of ischemic heart disease in young patients with newly diagnosed ankylosing spondylitis--a population-based longitudinal follow-up study. PLoS One 2013; 8:e64155.
  153. Lehtinen K. Mortality and causes of death in 398 patients admitted to hospital with ankylosing spondylitis. Ann Rheum Dis 1993; 52:174.
  154. Zochling J, Braun J. Mortality in ankylosing spondylitis. Clin Exp Rheumatol 2008; 26:S80.
  155. Mok CC, Kwok CL, Ho LY, et al. Life expectancy, standardized mortality ratios, and causes of death in six rheumatic diseases in Hong Kong, China. Arthritis Rheum 2011; 63:1182.
  156. Askling J, Klareskog L, Blomqvist P, et al. Risk for malignant lymphoma in ankylosing spondylitis: a nationwide Swedish case-control study. Ann Rheum Dis 2006; 65:1184.

Racing Accomplishments

  • 2002 Nov--Run For Life 10K
  • 2002 Oct--Canadian International Marathon 5K
  • 2002 Sept--Community Power Challenge 5K
  • 2003 Oct--Canadian International Marathon 21.1K
  • 2003 Sept--Longboat Toronto Island 10K
  • 2004 May--Ottawa National Capital Race 21.1K
  • 2004 Oct--Toronto International Marathon 21.1K
  • 2004 Sept--Scotiabank Waterfront Marathon 21.1K
  • 2006 April--London Spring Run Off 10K
  • 2006 Aug--Midsummer's Night Run 15K
  • 2006 Dec--Honolulu Marathon 42.2K
  • 2006 July--5 Peaks Durham Regional Forest 5K
  • 2006 June--Race the Lake 10K
  • 2006 Oct--Vulture Bait Ultra Trail 10K
  • 2006 Sept--Scotiabank Waterfront Marathon 21.1K
  • 2007 March--Around the Bay 30K
  • 2007 April--Harry's Spring Run Off 8K
  • 2007 May--SportingLife 10K
  • 2007 Oct--Toronto International Marathon 21.1K
  • 2008 May--Ottawa National Capital Race 21.1K
  • 2008 Aug--Iroquois Trail Test 32K
  • 2008 Sept--Scotiabank Waterfront Marathon 21.1K
  • 2008 Oct--Run for the Toad 50K
  • 2008 Oct--Toronto Zoo Run 10K