Open access peer-reviewed chapter

Perspective Chapter: Pulmonary System and Sjogren’s Syndrome

Written By

Moiz Ehtesham, Anupama Tiwari, Rose Sneha George and Ruben A. Peredo

Submitted: 25 October 2021 Reviewed: 21 January 2022 Published: 26 August 2022

DOI: 10.5772/intechopen.102803

From the Edited Volume

Idiopathic Pulmonary Fibrosis

Edited by Salim Surani and Venkat Rajasurya

Chapter metrics overview

120 Chapter Downloads

View Full Metrics

Abstract

Sjogren’s syndrome (SS) is a connective tissue disease targeting the exocrine glands with subsequent sicca symptoms mainly in eyes and mouth. Respiratory symptoms may be the most frequent extraglandular manifestation following fatigue and pain. Mucosal dysfunction may affect the upper and lower airways, being the small airways more frequently involved. Parenchymal disease carries most of the morbidity and mortality. Nonspecific interstitial pneumonia (NSIP) is the most common radiographic feature, whereas the fibrotic NSIP type is the most reported finding in biopsies. Pulmonary lymphoma may arise from bronchial-associated lymphoid tissue lesions, and although rare, it is prevalent in SS. Chronic hypertrophic bronchial wall changes may ascribe to the various cystic lesions. Under their presence, possible lymphocytic interstitial pneumonia, amyloidosis, and lymphoma should be explored. Pulmonary arterial hypertension may present as frequently as in lupus, especially in Asian populations. Advanced knowledge in the pathogenesis has helped in understanding the various presentations within the respiratory system, contrasting with the scarce therapeutic options to treat both the airway and parenchymal disease. Anti-fibrotic parenchymal lung therapy offers promising outcomes. The pulmonary involvement in SS may associate with a decline in quality of life and reduced life expectancy. Subsequently, clinicians should know these facts for a timely intervention.

Keywords

  • Sjogren’s syndrome
  • interstitial lung disease
  • airway disease
  • lymphoma
  • cystic lung disease

1. Introduction

Sjogren’s syndrome (SS) is a chronic, progressive, and systemic autoinflammatory disease, with exocrine (mainly salivary and lacrimal) glands as the main target organs, leading to the development of sicca symptoms [1, 2]. They are the main clinical feature of the disease. Fatigue, diffuse pain, cognitive dysfunction, and arthralgias follow, constituting common findings [3, 4]. A subset of patients may express disease in extraglandular organs/systems, reflecting the systemic nature of the disease [5, 6, 7]. This pattern is more prevalent in the pediatric population [8]. Articular, peripheral neurological and pulmonary manifestations are described often [5], followed by other disease features: hematologic, gastrointestinal, renal, cutaneous, and endocrine [5, 9]. A subset of patients may progress to develop lymphoproliferative diseases, mainly stemming from mucosal-associated lymphoid tissue but also presenting as other types of non-Hodgkin (mainly B-cell) lymphomas [10]. Pulmonary manifestations are reported with various frequencies, depending on the criteria and methodology used to define them averaging an estimate of 9–24% in most of the studies [9, 11, 12]. Sjogren’s syndrome when presenting alone is labeled as primary SS (pSS), while if it associates with another autoimmune (and mainly connective tissue) disease [13].

Advertisement

2. Epidemiology of Sjogren’s syndrome

The disease affects more women than men in a proportion of 9:1 [14, 15] and peaks in the fourth to sixth decade of life [16]. The pooled incidence ratio for primary SS (pSS) is 6.92 per 100,000 person-years, and has a prevalence ranging from 0.05% to 0.23% [15, 17, 18, 19]. It is considered the second (if not first) most common connective tissue disorder, with an estimate of 1–3% of the population being affected [20].

Advertisement

3. Pathogenesis

3.1 Histopathology

The cellular [21] and humoral [22] components participate actively in the gland dysfunction and eventual destruction. Invasion of mononuclear cellular infiltrate, mainly composed of lymphocytes, tends to localize around the salivary ducts, vessels, and adjacent to the intact mucous acini [23]. This infiltrate tends to aggregate forming clusters, and if they count ≥50 cells, they are named as focal lymphocytic sialadenitis (FLS) (Figure 1). Quantifying the number of FLS within 4 mm2 and dividing it by the area of normal glandular tissue will give an outcome. If this result is ≥1, it is reported as a focus score [24]. Focus scoring constitutes the main histopathological definition of SS [23, 25] and may range from 1 to 12. Higher values are obviated as they will be difficult to interpret due to the confluency of lymphocytic aggregates. Presence of a focus score helps the expert to define SS and to differentiate it from other inflammatory sialadenitides, including nonspecific chronic sialadenitis, sclerosing chronic sialadenitis, and others (Figure 1) [23].

Figure 1.

a (10×), b (20×): H&E section of this excisional biopsy of minor salivary gland tissue reveals periductular lymphoplasmacytic infiltrate. Multiple foci of periductular nodular lymphoplasmacytic infiltrates are identified. The number of mononuclear cell infiltrate per nodular focus is estimated to be greater than 50 lymphocytes per 4 mm2 of tissue examined, and each lobule examined contains at least one focus of inflammation. Thus, the focus score for the above biopsy was estimated to be ≥1. Therefore, this picture of chronic sialadenitis with a focus score ≥ 1 in the right clinical context is consistent with Sjögren’s syndrome.

3.2 Serology

Specific serology in SS associates with characteristic disease phenotypes and helps defining the disease [26]. The most characteristic and specific antibody is anti-SSA/Ro [27], and it is included in the 2016 classification criteria of SS [28]. Other serology, although less specific or not so prevalent, associates with SS as well, but is excluded from the currently accepted European-American classification criteria for Sjogren’s syndrome. For instance, in a large multicenter cohort study of 10,500 patients, serology at the time of diagnosis of SS showed the following frequencies in decreasing order: antinuclear antibody (ANA) (79.3%), anti-SSA/Ro (73.2%), rheumatoid factor (48.6%), and anti-SSB/La antibodies (45.1%) [26]. Despite this distribution, anti-SSA/Ro and anti-SSB/La antibodies have been specifically identified to participate in SS’s pathogenesis. They are also present in other connective tissue diseases (CTDs), lowering their specificity [29]. Cryoglobulins and low complements, mainly low C4, may reveal disease activity and define prognosis [27, 30, 31]. Their presence is additionally predictive of lymphoma [32]. Presence of circulating autoantibodies in patients with SS prior to the diagnosis suggests that the immune activation has been previously triggered by an unknown antigen, and it may take months or years to progress onto a phenotypical expression [33]. A two-hit hypothesis, in this scenario, may possibly be the most likely explanation for this phenomenon. Sensitization and priming of the immune system by a prior insult (first hit) may define and determine the fate of the upcoming a programmed immune response following the exposure of a second stressor (second hit). During this second insult the immune tolerance seems to be breached, with which the sequence of autoimmune events activate the disease [34, 35]. Presence of other circulating antibodies reported in SS may be the result of the polygenic nature of the disease and possibly due to different antigens activating the immune system, and hitting specific targets [26, 36, 37, 38, 39]. Novel autoantibodies linked with sicca eyes reveal our still limited knowledge in SS’s pathogenesis [40, 41].

Advertisement

4. Symptoms and disease definition

Symptoms associated with dry mucosae in the eyes and mouth (sicca) are complains the clinicians should explore to consider the disease [42, 43]. To define the dry eyes and mouth, several techniques objectively measure their quantity [2, 44]. In addition, recommendations on to elaborate questions regarding sicca symptoms, fatigue, arthralgia, Raynaud’s, and other remarkable features common in SS, are detailed in the new consensus guidelines for the evaluation and management of pulmonary disease in Sjogren’s syndrome [45]. Sicca symptoms constitute the core finding in SS and have always been included in any classification criteria. The composite of sicca eyes, mouth, positive serology (anti-SSA/Ro antibodies) and abnormal findings in the histopathology (a focus score of ≥1), constitute the current four pivotal components to fulfill the SS classification criteria [28]. Along the last five or more decades, the classification criteria have been modified more than 15 times [46, 47, 48, 49]. These changes reveal the difficulties met on agreements to define the disease, and mainly due to variability in the cohorts used, the protean disease manifestations, variability in the serology on different populations, and the need for more than one expert (specialist) to define each one of the criteria components. In addition, the continuous changes in the classification criteria mirror the difficulties to understand the intricate and still poorly understood immunopathogenesis [12, 50].

The initial descriptions of pulmonary manifestations are detailed in accurate observations almost a century ago, and are described ahead.

Advertisement

5. History

5.1 Sjogren’s syndrome as a systemic illness

The initial description of Sjogren’s syndrome (SS) included the dry eye, and Leber [51] described filamentary keratitis (FK) [51], a finding that years later was linked with the lacrimal gland dysfunction (described by Stock in 1925). Around the same time, descriptions of a combination of dry eyes and mouth, detailed by Hadden [52], was followed by further clinical associations of deforming arthritis, and detailed by several authors in case series [53, 54, 55]. In his doctoral thesis in 1933, Henrik Sjogren, a Swedish ophthalmologist, accurately detailed what we know as the syndrome that carries his name. In his treaty, he accurately depicted the disease as we currently know, “…ocular changes because of dry eyes and hypofunction of salivary secretion, along with the arthritis and other systemic symptoms deals with a generalized disease and is not purely a coincidence” [56]. Ever since we know the concept of SS and its extent. Dr. Sjogren’s pristine and sharp description made it possible to link all the clinical manifestations within a syndrome. This concept of a systemic illness manifesting in various organs was already familiar facilitating him to launch it as a unique disease. Decades prior, Dr. William Osler described systemic lupus on several patients (in 1895 and 1903) who presented with multiorgan involvement, other than the skin [57]. During the postwar era, in the 1950s, several scientists uncovered thyroid antibodies, and following Dr. Hashimoto’s hypothesis from 1912, in which he sustained the thyroid gland to be the target of specific autoantibodies. Dr. Jones applied this knowledge in SS. In this case, the salivary/lacrimal glands were the main target rather than the thyroid gland [58]. And indeed, the collaborative group of scientists from the National Institutes of Health in Maryland, USA, were able to identify them [53]. Two other concepts evolved as well: primary SS (pSS), when the disease presented alone; and secondary SS (sSS), when it was associated with other CTDs [13, 59].

5.2 Sjogren’s syndrome and the respiratory system in history

The pulmonary involvement was documented in the 1950s. In a registry of pSS, pulmonary infiltrates were reported in 7/40 cases [60]. In the original case series, Dr. Sjogren early on (in the 1930s and 1940s) described diverse respiratory findings including rhinitis sicca, pharyngitis sicca, and laryngitis sicca, and considered them to be components of the whole dryness spectrum added to keratoconjunctivitis sicca and xerostomia. In the mid-1940s, Dr. Weber extended this concept to other tissues, proposing that the exocrine gland’s dysfunction and destruction might precede sicca manifestations with an inflammatory continuum in the nasal, pharyngeal, and laryngeal mucosa, and other distant organs: the skin, vagina, gastric mucosa (this later with subsequent achlorydia) [61]. Management was mostly symptomatic. Further progression of several discoveries made it possible to elaborate definitions of lung compromise within SS’s disease spectrum. Baruch and coauthors launched the concept of SS to be classified in two types: (1) those related to major CTD, and (2) sicca complex in the lungs that encompassed the following: chronic bronchitis, subsegmental atelectasis, bronchiectasis, pneumonia, lymphoproliferative pulmonary infiltrates, and chronic interstitial pneumonia, later leading to fibrosis [62]. Bloch published a series of 62 SS cases 37 (60%) who complained of nasal dryness and adherent crusts. One of them had sudden hearing loss linked to otitis, and chronic sinusitis was present in 4 (6%), throat dryness in 28 (45%), hoarseness in 20 (32%), and chronic dry cough in 5 (24%) [49]. In the lower respiratory tract, Bloch and coauthors reported: pleurisy, pleural adhesions, focal and lipoid pneumonia, pulmonary atelectasis, and fibrosis [49].

5.3 Pathology in history

Histopathological findings of most cases revealed submucous gland atrophy and lymphocytic infiltration intermixed with plasma cells at all levels of the respiratory tract [49, 63, 64]. The descriptions of pulmonary disease detailed different scenarios, from asymptomatic to severely ill patients. In this latter group, authors described two cases of acute parenchymal infiltrates in the setting of recurrent bronchitis and pneumonia. These cases presented with pneumonia composed of different cellular types with lymphocytic predominance and nodular lesions without evidence of an underlying infection or malignancy [49]. Brown attributed the cellular clustering to the diminished secretion of mucus, poor bronchial drainage, and secondary infection. Poor cellular immune response was considered [65], but it was also linked secondarily to a phenomenon known as pseudolymphoma [66, 67]. The latter consisted of marked cervical lymphadenopathy, pulmonary infiltrates of lymphocytes without enough atypia or monoclonality to label it as lymphoma [49]. Years later (1972), a full description further reinforced the diversity on pulmonary presentations, ranging from asymptomatic cases to overwhelming lymphoproliferation. Examples within this process were considered, such as pseudolymphoma, Waldenstrom’s macroglobulinemia, reticulum-cell sarcoma within the lymph nodes and other types [68]. Similar findings were described in lymphoproliferative processes arisen in other autoimmune diseases (e.g., lupus), certain immune deficiency states, and hydantoin and use of other anticonvulsant drugs [68]. Finally, knowledge of SS in the respiratory system was expanded. Cases of amyloid in the lungs in patients with SS were reported in the 1970s [69], and other lower airway manifestations such as bronchiolitis, asthma, bronchiectasis, bronchiolitis obliterans with organizing pneumonia and also parenchymal disease, such as the interstitial pneumonia with potential to lead to diffuse interstitial pulmonary fibrosis, were linked with SS [62, 70, 71].

Advertisement

6. Prevalence and patterns of pulmonary disease in Sjogren’s syndrome

As described in the history, respiratory symptoms exhibit a plethora of manifestations with variable ranges of severity of different areas within the respiratory system. The airways and the lung parenchyma, or an admixture of both, may present alone and combined. Rarely the pleura may show inflammatory changes [72, 73], and pulmonary hypertension, although rare, has been more frequently recognized in East Asian populations [74]. Each one of the compartments may present with a range of different pathologies expanding the disease variety. For instance, in the lung parenchyma, NSIP may prevail [75, 76], but other manifestations have been reported [77].

The prevalence of lung and respiratory manifestations fluctuates from 9 to 24% [9, 11, 12] that include symptoms and abnormal pulmonary function tests or abnormal radiographic findings. Prevalence can go up to [78] 43%-75% [79] if patients are followed prospectively and analyzed based on a composite of multiple studies [80, 81, 82, 83]. Symptoms may represent an estimate of an average of 40–66% [84, 85, 86], with an increase in sensitivity if radiographic images are included. The involvement of lower airways seem to be the most common respiratory presentation in SS [84, 87, 88]. The cumulative incidence of interstitial lung disease (ILD) at 1 year of pSS diagnosis was found to be of 10%, and went up to 20% after 5 years and 47% at 15 years, a fact that becomes relevant as SS patients age, making an impact on the prevalence [89]. The high prevalence of SS and the respiratory system involvement are a concern for the clinician, alerting her/him to have a full evaluation consisting of obtaining a detailed medical history, at the onset and during the follow-up appointments [45].

Advertisement

7. Morbidity, mortality, and prognostic factors in Sjogren’s syndrome and pulmonary compromise

Many of patients with SS and respiratory manifestations, and mainly interstitial lung disease associated with SS (ILD-SS), experience a decline in their quality of life [90, 91]. This seems to be tightly associated with increased morbidity that ultimately will decrease their life expectancy [89]. Mortality risk increases fourfold in a 10-year timeframe [90], making lung involvement one of the most common causes of death [92] and a predictor for mortality [89]. In a meta-analysis of large cohorts of patients with pSS, the overall mortality risk was 1.46-fold higher than that of the general population, and patient profiles with this higher risk revealed to be in the European group, older age, males, presence of ILD, cryoglobulinemia, positive serology (anti-SSB/La), and low complement) [93]. This coincided with another meta-analysis, in which results of mortality risk showed the same, plus additional factors, such as the parotid enlargement, abnormal parotid scintigraphy, and extraglandular involvement [94]. The hazard ratio (HR) for death in pSS and ILD is between 2.1 and 3.2 [89, 95]. Among patients with pSS and already established ILD, respiratory failure accounted for the most common cause of death, and risk factors for mortality were older patients, with smoking habit, and carriers of severe ILD [96], either based on the number of reticulations on the chest HRCT and lymphoblastic foci in the biopsy [97].

Advertisement

8. Radiographic features in Sjogren’s syndrome and respiratory involvement

The chest X-ray may disclose features of both the airway, parenchymal and pleural involvement, but has a lower sensitivity than the chest HRCT [98]. The high-resolution chest computed tomography (HRCT) represents the most sensitive technique to uncover pulmonary features even in asymptomatic patients, followed by the pulmonary function tests (including plethysmography) [82]. Computed tomography (CT) changes have been reported in 34–50% [98]. In a cohort of 527 patients with pSS, prevalence of ILD was identified in 39.1% (206/527) based on abnormal chest HRCT. In this large cohort, the most common characteristics in the HRCT reported were associated with parenchymal disease in decreasing order: reticular pattern in 92.7%, ground-glass attenuation in 87.4%, and bronchovascular bundle thickening in 82% [99].

Advertisement

9. Pulmonary function testing in SS and respiratory involvement

The pulmonary function tests are of relevant utility since they will describe patterns of either obstructive, restrictive or a combination of both diseases. Prevailing features in pSS are seen in a pattern of obstruction in most patients and is mainly observed in the maximal expiratory flows (MEFs) 25–50% that test the small airway disease [84, 100]. Decreases in DLco have been reported in several studies [99, 101, 102, 103, 104], but the significance of such findings is still unclear. Usually, they precede the FVC decline. Correlations of DLco and higher Schurawitzki score on the chest HRCT have been made, representing a prognostic factor for mortality [104]. Disproportionally low DLco in equivalence to the FVC might represent alveolitis in ILD cases, or pulmonary arterial hypertension (PAH), seen in pSS more than thought in the past. The decline in FVC is more prominent once ILD is established [105, 106].

Advertisement

10. Bronchoalveolar lavage in Sjogren’s syndrome and respiratory involvement

Bronchioalveolar lavage in patients with SS may represent an extraordinary tool to define on whether the respiratory system is involved, especially in patients who would present with symptoms and negative changes on PFTs or chest HRCT [100, 107]. Results may disclose prevailing specific CD4(+) T lymphocytes in the cellular differential [108]. Thus, it may improve the sensitivity to detect disturbances in the respiratory system [84], especially for patients with unexplained respiratory symptoms with normal HRCT and PFTs. Furthermore, BAL will reveal an inflammatory pattern on cases with alveolitis, showing the T lymphocyte predominance [108].

11. Biopsy utility in Sjogren’s syndrome and respiratory involvement

Biopsy is currently limited to specific scenarios to (1) determine a clear etiology of the disease, (2) define fibrotic non-interstitial pneumonia (NSIP) vs. usual interstitial pneumonia (UIP), or admixed patterns (3), and establish underlying malignancy, especially in cases with lymphocytic interstitial pneumonia (LIP). In the biopsy, most of results may disclose a fibrotic NSIP pattern (Figures 2 and 3) [97]. Furthermore, many of the biopsies will reveal presence of small airway inflammatory changes in association with ILD. Amyloidosis will be identified in association with SS and nodular lung disease, and UIP will increase a yield to up to 33% with the biopsy [97, 103, 109, 110].

Figure 2.

H&E sections from a right upper lobe of lung wedge resection reveal focal areas of subpleural scar and fibrosis admixed with cystic airway distention (image a). These areas are associated with an adjacent prominent lymphoplasmacytic inflammation with lymphoid follicular hyperplasia (images b and c). The lymphoid expansion is associated with frequent airspace cholesterol clefts, histiocytes, eosinophilic debris, and sparse neutrophils (image A), mostly resembling changes secondary to localized airspace obstruction possibly secondary to the degree of hyperplastic lymphoplasmacytic reaction. Subpleural cysts and lymphoid hyperplasia are seen in Sjogren’s syndrome.

Figure 3.

H&E sections from this right upper lobe lung wedge biopsy (image a) reveal a cellular interstitial inflammatory infiltrate predominantly composed of lymphocytes admixed with interstitial fibrosis, which is mostly centrilobular. These constellations of findings are consistent with a cellular interstitial pneumonitis with fibrosis. Sections from the right lower lobe wedge biopsy (images b and c) reveal a more markedly altered lung parenchyma compared with the right upper lobe. There is extensive panlobular collagen fibrosis with multifocal areas demonstrating cystic spaces lined by bronchiolar epithelium and fibrotic walls (honeycomb changes). The findings identified in the lower lobe biopsy are not specific to an interstitial lung process but points toward the differential diagnosis of lesions that can have a nonspecific interstitial pneumonia (NSIP) pattern with temporal heterogeneity and extensive honeycombing, which would include connective tissue disorders such as Sjögren’s syndrome.

The significance of SS exhibiting extraglandular manifestations represents a higher inflammatory state. This is remarkably noticed when the respiratory system is affected. Hypergammaglobulinemia reflects this notion [81, 107]. Additionally, positive anti-SSA/Ro and anti-SSA/La antibodies may associate with the respiratory system [78, 111], so were other acknowledged makers: ANA and the rheumatoid factor (RF) [81, 83]; in addition, some authors consider the evidence of serology as a predictor for lung disease in SS conflicting [80].

12. European league against rheumatism and measurers of disease activity in Sjogren’s syndrome

The European League Against Rheumatism (EULAR) task force on SS has created the EULAR-SS Disease Activity Index (ESSDAI) to determine specific organ expression [112, 113]. It is used now as an index to quantify the disease activity in pSS, and applied in randomized control trials [114]. This index includes 12 domains, and representing the target organs and systems affected by the disease (organ systems that are explored are: cutaneous, respiratory, renal, articular, muscular, peripheral nervous system (PNS), central nervous system (CNS), hematological, glandular, constitutional, lymphatic, and immunological). For each domain, the scoring assigns the disease activity in 3–4 levels depending on the severity. Low activity is the ESSDAI of < 5 points; moderate-activity falls between 5 ≤ ESSDAI ≤ 13; and high activity scores the ESSDAI ≥ 14 [115]. A minimal clinically important improvement (MCII) is established as a decrease of at least three points in follow-up visits, when the prior scoring showed moderate activity [115]. The pulmonary domain is divided into four categories, based on the activity level, and range from no activity equivalent to 0, or to symptoms unrelated to pSS, to high activity level or scored as 4 (Table 1). The range between low and high activity levels will define the severity based on symptoms (the magnitude of dyspnea will be determined and scored by using the NYHA stratification) and progression of the respiratory symptoms analyzed with ancillary tests (PFTs) or alternatively with the chest HRCT. Patients who fall in the high-activity group may have a worse outcome. In a large cohort of 921 pSS patients, the pulmonary domain of ESSDAI scoring at the time of the diagnosis revealed any pulmonary activity in 6.1% of them, 94% of patients had no pulmonary symptoms. At the end of 75 months, 15% had any pulmonary activity, and sixty percent of the cumulated score corresponded to a new activity [5]. The pulmonary activity had the highest mean cumulated score at the last visit along with the renal and muscular domains, revealing the higher incidence as longer the disease progresses in these three domains [5].

DomainActivity levelDescription
Pulmonary Rate as ‘No activity’ stable long-lasting features related to damage, or respiratory involvement not related to the disease (tobacco use, etc.)No = 0Absence of currently active pulmonary involvement
Low = 5Persistent cough due to bronchial involvement with no radiographic abnormalities on radiography or radiological or HRCT evidence of interstitial lung disease with no breathlessness and normal lung function test
Moderate = 10Moderately active pulmonary involvement, such as interstitial lung disease shown by HRCT with shortness of breath on exercise (NHYA II) or abnormal lung function tests restricted to: 70% > DLCO ≥ 40% or 80% > FVC ≥ 60%
High = 15Highly active pulmonary involvement, such as interstitial lung disease shown by HRCT with shortness of breath at rest (NHYA III, IV) or with abnormal lung function tests: DLCO <40% or FVC <60%

Table 1.

EULAR Sjogren’s syndrome disease activity index (ESSDAI). Pulmonary domain [114].*

The pulmonary domain is subdivided into four category levels based on the severity of symptoms (NYHA scoring system) and findings in the HRCT and the pulmonary function tests. Cough should be part of the disease and not related with tobacco use. Persistent dry cough and long-lasting cough due to damage should be scored as 0. Interpretation of HRCT should be linked to the activity to ground-glass and not honeycombing aspects. Shortness of breath should be attributable to the disease, and confounders (tobacco use, cardiac insufficiency, arterial pulmonary embolism, or infection) should be excluded.


FVC, forced vital capacity; HRCT, high-resolution CT; NYHA, New York Heart Association.

Sjogren’s syndrome may selectively affect different compartments within the respiratory system, but it seems it affects almost all and frequently simultaneously. They may present in an overlap complicating their identification for the subsequent therapeutic intervention. For a better understanding, however, the following compartments should be studied separately.

13. The upper airways

The upper airways seem to be a continuity of the mucosal invasion of inflammatory mononuclear cells, or sialadenitis, affecting the mucosal surfaces in the sinuses, larynx, and ears. The cumulative incidence of chronic rhinosinusitis in pSS has a HR of 2.5 as compared to controls [116].

Dysfunction of salivary glands with the resultant dry mucosa changes the microenvironment in the oral and distant mucosae and thus, promotes a chronic inflammatory state. One good example highlights that pSS patients are at 2.5 times higher risk of developing chronic rhinosinusitis [116]. In addition, the consequent dryness alters or sets off the mucociliary clearance, as seen in the tracheobronchial tree [117]. Furthermore, the histopathology of mucosal surfaces shows infiltrates, similar to what it is seen in the minor salivary glands, where lymphocytic cells surround the mucosal acini configuring the focal lymphocytic sialadenitis [24]. In a study comparing bronchial biopsies of patients with pSS vs. controls, the former showed higher number of infiltrating neutrophils, mast cells, and T lymphocytes. The epithelial damage and structural changes in the subepithelium resembled changes seen in atopic asthma [118]. Moreover, lymphocytic infiltrates are present in BAL of both symptomatic and even asymptomatic patients, representing the continuous and silent inflammatory state along the airways [84, 107, 108, 119].

14. Oral microbiome and Sjogren’s syndrome

Sicca mouth in SS has an impact on the oral microbiome, favoring the growth of a dysbiotic environment that replaces the normal flora compared with controls [120]. It is unclear if this hostile environment predisposes this microbiome shifting, favoring its growth. Also, it is unclear if the dysbiosis may impact on the disease establishment and/or progression. Supporting possible impact on glands by the microbiome, experiments on animal models, revealed an association of dysbiotic oral microbiota with the development of lymphocytic sialadenitis [121]. This hypothesis was evaluated in a recent study that demonstrated the immunomodulatory properties of commensal bacteria (Haemophilus Parainfluenzae). This bacterium keeps the regulatory immune homeostasis, explored at the cellular level. In a study of salivary microbiome, patients with pSS had lower amounts of H. Parainfluenzae. The analized A253 cells, once primed with H. Parainfluenzae exposure, induced suppression of CD4 T cell proliferation and induction of PD-L1 expression [122]. Moreover, treatment with low-dose doxycycline normalized the levels of some salivary metabolites associated with the dysbiotic microenvironment in patients with pSS to levels comparable with healthy controls [123]. These findings support the role of the microbiome on pSS pathogenesis and mucosal dysfunction.

15. Dry mucosae in the mouth and upper respiratory airways and SS

Sicca mucosae in SS may impact on dental and periodontal health. It is common to see gum retraction in most of the teeth and specific dental caries at the neck of them. The sicca environment predisposes patients to develop the growth of opportinistic infections, like candida growth. Candida colonization presents in various forms, ranging from asymptomatic, including leukoplakia, and even as burning mouth syndrome [124]. Many patients may lose their teeth and have a significant decrease in their quality of life. This later is in part attributable to the dysphagia and dysphonia, both related with xerostomia [125]. Hoarseness presents in a frequency between 26% and 33% [125, 126]. The laryngeal mucus and vocal folds will harden causing morphological revealing distinctive vessels and/or edema on the exam. In the video-assisted swallowing test abnormal motility will be seen [125]. Gastric reflux may account for this finding predisposing the dysfunctional esophageal motility [127]. The bamboo node represents a chronic inflammatory state of vocal cords reported in SS and other CTDs [126]. Although not fully recognized as a common finding in pSS, hearing dysfunction and loss were reported in 80% of patients (24/30), with severe hearing loss in 10% of them. However, most of the pathology seemed to be linked with vestibulocochlear (cranial) neuropathy [128].

Cough is common in pSS, and especially dry cough is representative of, mainly but not only, airway inflammation. The term xerotrachea defines dry tracheal mucosa with the inflammatory background that extends distally, causing significant morbidity [84]. Again, difficulties in clearance will prolong inflammation and promote further functional and anatomic changes such as atelectasis, bronchiectasis, bronchitis, peribronchial and peribronchiolar scarring and airway narrowing [70, 83, 119, 129, 130, 131].

16. Lower airways: epidemiology

Patients with SS have a frequent hyperreactive tracheobronchial response in 42–60% [132, 133], and sustained and extended cough following persistent stimuli (dust, tobacco, etc.). They will have abnormally bronchial hyperreaction to the methacholine challenge test, but become inert to the adenosine monophosphate, cold, or hyperventilation. Under the chronic inflammatory state, the pulmonary function test will reveal a decline in the different lung volumes [134, 135]. The pathogenic background of bronchial hyperreaction is unclear, making it difficult to interpret and treat [132]. As previously described, the mucosal chronic inflammation will interfere with clearance, perpetuating and aggravating the dysfunctional hyperresponsiveness [117]. The chest HRCT will be of utility to identify lower airway disease. Findings will be: peripheral bronchiectasis in 5-46%, bronchial wall thickening in 68-85%, nodules in 6-29%, and air trapping in 32%. Together with ground-glass attenuation as the representative parenchymal disease, these are the most common features identified with this modality [87, 107, 136].

16.1 Bronchiolitis

Inflammation in bronchioles has predilection for SS and presenting in different types, such as obliterative bronchiolitis, chronic bronchiolitis, lymphocytic bronchiolitis, constrictive bronchiolitis, and panbronchiolitis [11]. However, the most representative type in SS is follicular bronchiolitis. In biopsies, it may associate with interstitial pneumonia (especially NSIP) [97, 109], and in some cases, along LIP, they may form a continuum, the former limited to peribronchiolar area while the latter to the alveolar septa [119, 137]. Frequencies increase from 12% to 24% when radiographic images accompany the pathological interpretation [109]. Follicular bronchiolitis has a bronchovascular distribution, and the hyperplastic lymphoid follicles with reactive germinal centers run along the bronchovascular bundles [117, 138, 139]. The CT scan will define changes of bronchial thickening in 8–22%, and bronchiolar nodules in 6–24% [82, 84, 133, 140]. Many studies have shown a decline in the DLco that is attributable to bronchiolitis, a fact that needs to be fully proven [99]. Symptoms are represented by dry cough, wheezing, dyspnea, and overlapping infection. Treatment for bronchiolitis challenges the clinician since most of the therapies only seem to be partially responsive. Starting with antibiotic therapy aimed at preventing infectious overlap and especially with use of macrolides, given their anti-inflammatory properties, the mainstream therapy remains on inhaler and/or systemic glucocorticoids, if the disease progresses or becomes seriously symptomatic [141, 142]. Applied disease-modifying drug therapies include azathioprine (AZA), mycophenolate mofetil (MMF), and even rituximab [142]. In the presence of comorbid immune deficiencies (i.e., immune globulin deficiencies, mainly IgG), replacement therapy is recommended [143]. Overall, the treatment intensity should correspond the severity of each case. In complex cases combination of therapies might show better outcomes.

17. Bronchus-associated lymphoid tissue (BALT)

The chronic antigenic stimulus will drive the follicular bronchiolitis to conform a bronchus-associated lymphoid tissue (BALT), which is a benign inflammatory state of polyclonal lymphoid hyperplasia [144], and dense cluster of lymphocytes with follicular structures. These cells follow an antigen-driven stimulus. Well-defined aggregation within a network will separate B from T cells. The B cell compartment encloses follicular dendritic cells (FDCs), which is related with the vascular structures (venules and lymphatics) [142]. BALT is equivalent to the gastric mucosal-associated lymphoid tissue (MALT). In addition, the perivascular compartments are encased by lymphocytic aggregates, labeled as perivascular cuffing. They may extend to the small airways and run parallel to the vessels. This organized lymphocytic aggregation is known as induced BALT that is a chronic inflammatory state ready to get reactivated after a second insult [145].

18. Chronic obstructive pulmonary disease

Patients with SS have significantly decreased PFTs showing a composite of decreased VC, TLC, FEV1, FEV1/VC, and DLco but high RV. This pattern fulfills criteria for COPD even in nonsmokers, a fact that may be explained by the presence of chronic tracheobronchitis [146].

18.1 Bronchiectasis

Structural damage with dilatation in distal bronchi and bronchioles may associate with dry mucosa, poor clearance, and superimposed infectious processes [147, 148]. In SS, the cylindrical pattern seen on the HRCT seems to be the prevailing finding [149]. Frequencies vary depending on the cohorts, from 7% to 54% [11]. The clinical presentation is frequently seen in women with chronic sinusitis, with age at the time of diagnosis, and comorbid gastroesophageal reflux. Antismooth muscle but unfrequent anti-SSA/Ro antibodies were detected in this group. The HRCT will describe cylindrical bronchiectasis localized preferentially in the lower lobes (Figure 4) [11]. A plethora of symptoms may ensue, especially chronic cough, dyspnea, and even recurrent remitting hemoptysis. The concomitant recurrent superimposed infections worsens the prognosis, reported in 10–35% [70, 71, 150]. Multifactorial etiologies play a role, such as gastroesophageal reflux, dysfunction in the tracheobronchial mucociliary clearance, chronic sinusitis, immune suppressor drug therapy, climate, and presence of bronchiectasis [148, 151, 152]. Combination of bronchodilators, secretagogues, chronic antibiotic use as preventative means for flares seem to be the mainstay of therapy [11]. The use of immune suppressor therapy favors higher risks for infections as bronchi may already be chronically colonized with abnormal and pathogenic flora [153].

Figure 4.

HRCT. Scattered, ectatic airways more visible in right middle lung and lingula where there is also bronchiectasis and scarring/volume loss. They represent airway-related disease (bronchiectasis).

As described previously, follicular bronchiolitis may be seen in association with parenchymal disease, mainly interstitial pneumonia. The following sections will display the various types of parenchymal disease in SS.

19. Parenchymal lung disease: epidemiology and patterns

Most of the studies report a prevalence of interstitial lung disease associated with primary Sjogren’s syndrome (ILD-pSS) of around 20% [85, 89, 95, 101, 102, 151, 154, 155, 156, 157, 158]. Other cohorts report variable frequency, ranging from 3% up to 60% [99, 152]. Furthermore, the EULAR task force reported a prevalence of 49% in 526 group of patients with SS, and based on chest HRCT [9], which appears a real-life frequency. Incident cases range between 8% and 17% [85, 101]. Lymphocytic interstitial pneumonia (LIP) was thought to be the most common ILD type [159], but recent large cohorts reveal the following patterns in decreasing order of frequency, based on the HRCT: NSIP in 41.7%, UIP in 10.7%, OP in 3.9%, and LIP in 3.9% of cases out of a total of 124 patients [99]. Other large cohorts with similar frequencies of the various ILD types include amyloidosis in 11%. Admixed patterns were also present in 82 cases reviewed, with combinations of NISP/OP (43.9%), NSIP/UIP (35.4%), and NSIP/LIP (19.5%). Biopsies confirm NSIP to be the most frequent pathology. Symptoms and findings associated with ILD were found to be dry cough, clubbing, elevated lactate dehydrogenase, and positive anti-SSA/Ro antibodies [99, 103, 160]. Interstitial lung disease associates with older age at the disease onset, longer SS duration, fever, xerostomia, xerophthalmia, and neuropathy [161]. It may be the first presentation in a third to a half of patients [162]. Subsequently, not aware of SS’s features, the disease may run undetected. The contribution of lip biopsy in such cases is crucial for this goal especially in cases with negative serology [163]. Laboratories might help and are important for a full evaluation. Hypergammaglobulinemia, lymphopenia, low C4, and high acute-phase reactants are common findings [160]. The PFTs will disclose a restrictive pattern, and low DLco is frequently reported (even up to 64%) [101, 102, 162]. It is common to see an admixture with an obstructive pattern (25%), as a reflection of the association with lower airway disease [162]. The chest X-ray will be of great utility to determine any possible finding in the lung parenchyma, such as linear and reticular patterns, but it has the limitations in sensitivity to detect fine changes [98, 103, 109]. In the chest HRCT, up to 90% of findings will be disclosed [11]. Frequent findings are bilateral infiltrates in almost 99% of cases and predominance in lower lobes and subpleural spaces. Also, lesions distribute in perihilar areas in 9% [99]. Other common findings are the reticular pattern, ground-glass attenuation (92%), non-septal linear opacities (75%), interlobular septal thickening (55%), cystic formation (30%), reticulation, and fibrosis [109, 164]. Honeycombing and features of UIP are unusual [165] (Figure 4). Cystic lesions may present at different sizes and distributed along and imbibed withing the parenchyma. They have thin-walled demarcations and may be a consequence of a valve phenomenon [166]. They are associated with LIP, lymphoma, and even amyloidosis [167].

19.1 Nonspecific interstitial pneumonia

This subtype is the most frequent form of ILD-pSS and presents with variable degrees of symptoms, including cough, dyspnea or, rarely, may run asymptomatic [9]. Alveolitis may correlate with NSIP, and one way to identify it is with the BAL. This later study will disclose lymphocytic cells [108]. Frequencies vary depending on the cohorts and methodology used. In a series of 33 cases of ILD, the lung biopsy yielded NSIP pattern in 20/33 61% with the fibrotic type in 19 (57%) [109]. In a cohort of 263 patients with pSS, 8% were identified with ILD with a third of them having NISP pattern [85]. In another study, 19.3% had ILD-pSS, and almost half to them had NSIP pattern based on on HRCT [96]. On the chest HRCT, relevant features in this subtype are the ground-glass opacities, mainly in lower lung fields and subpleural predominance. Other findings are reticular abnormalities, traction bronchiectasis, peri-bronchovascular extension, and pulmonary consolidation. Sparing between pleura-parenchyma interface is a hallmark along with tracking of opacities along lower-zone bronchovascular bundles (Figure 5) [168]. The biopsy will reveal preservation of architecture and a composite of inflammatory cells. The distribution characterizes by the lymphocytic expansion of alveolar septa. Fibrosis is also seen and associates with traction bronchiectasis (Figure 3A). Honeycombing is rarely seen. Depending on the cellular/fibrotic predominance, NSIP is subdivided into the cellular or fibrotic types. As said, this later is the most frequent presentation in pSS [97].

Figure 5.

Parenchyma with reticulation, ground-glass attenuation, and traction bronchiectasis in the lower lobes, middle lobe, and lingula. This process is peribronchial in distribution and extends into peripheral portions of the lung parenchyma. Portions of the subpleural lung parenchyma in these regions are spared. There is no honeycombing. Bilateral pleural effusions larger in right than left lower lobes. Findings are consistent with nonspecific interstitial pneumonia.

The 5-year survival rate was of 83–87.4% [97, 109]. Low PaO2 and presence of microscopic honeycombing were associated with worse survival [109]. In another study, worse survival was associated with PaCo2, extent of reticular abnormality on HRCT, and severity of fibroblastic foci on the biopsy [97]. No differences between the NSIP and UIP patterns in terms of prognosis were identified [97], although this is controversial. The clinical course will vary, but this pattern usually is responsive to the immune modulation.

19.2 Usual interstitial pneumonia

This subtype is infrequent in SS, but prevalence varies between 10% and 17% [9, 99, 102]. Main differences between UIP and NSIP patterns are based on the HRCT features and interpretation. Intralobular reticulation, honeycombing, traction bronchiectasis, cystic lesions, and temporal heterogeneity may prevail as patterns. The hallmark for UIP is the honeycombing appearance (Figures 2A, 3B and C and 6) [168]. Although rare, UIP needs to be recognized as treatment usually is unresponsive to the therapy [103].

Figure 6.

There is extensive subpleural reticulation, honeycombing, and traction bronchiectasis/bronchiolectasis that predominate in the posterior basal lower lobes. There is no normal lung parenchyma between the fibrotic lung and the adjacent pleural surfaces. There are scattered foci of mild mosaicism indicative of air trapping. Findings are consistent with usual interstitial pneumonia.

19.3 Lymphocytic interstitial pneumonia

Main histopathologic feature is the polymorphous lymphoid infiltrate involving diffusely the alveolar septa of lymphocytes (T and B cells), plasma cells, and histiocytes [169]. Plasma cells show a polyclonal pattern [170]. As described, lesions may present with follicular bronchiolitis along the bronchovascular structures [171], and occasionally will present with foci of BALT hyperplasia (Figure 5) [144]. Also amyloid deposits may overlap [159]. Lymphocytic Interstitial pneumonia (LIP) is a benign lymphoproliferative disease. Frequencies range between 3% and 15% [9, 99]. Cough, dyspnea, and inspiratory crackles are common. The CT studies will disclose a diffuse ground-glass opacity and consolidation as the most common features. Thin-walled cysts can be present, along with combination of thickened bronchovascular bundles and nodularity in association with follicular bronchiolitis [168]. Frequently, the biopsy is necessary to differentiate from lymphoma. In the presence of germinal centers (GC), this differential has a more relevant importance since both LIP and lymphoma can portray this distinction (Figure 2B and C). B-cell lymphoma arising from BALT lesions presents with monomorphous B-cell infiltrates with invasion of lymphatics, vessel walls, pleura, and subsequent destruction of alveolar architecture. Monoclonal plasma cells, Immunohistochemistry and gene rearrangements will help define this differential from LIP [172, 173, 174]. Differential from NSIP relies on the more intense lymphocyte density seen in LIP [173]. Early and aggressive pharmacologic approach with high-dose glucocorticoids, followed by immune modulators/suppressors, seems to halt the disease progression [11].

19.4 Organizing pneumonia

It is an unusual presentation more common in rheumatoid arthritis, with a frequency of 3.9–11% [9, 75, 160]. Symptoms reveal severe dyspnea, cough, and oxygen dependency. A restrictive pattern will prevail, but a mixed combination is seen. HRCT features consist of diffuse or multifocal patchy bilateral ground-glass opacities and/or consolidation without extensive reticulation or honeycombing [175, 176]. Other features are the reversed halo opacity and bronchial wall thickening [177, 178]. Histopathology will reveal plugs of granulation tissue within small airways (Masson bodies) and chronic inflammatory cell infiltration in alveolar walls [179, 180]. Case reports document the favorable response to glucocorticoids [180, 181], but others may be fatal [182]. Differences of OP with cryptogenic OP are the more frequency in women, positive serology, and relapse presentations. Mortality is linked with progressive dyspnea [183].

20. Cystic lung disease

Cystic lesions may have different dimensions ranging from 0.5 cm to 7 cm, in internal structure within cysts, and frequently associate with ground-glass opacities and nodules [167]. They typically tend to localize in lower lobes, but distribution may be diffuse. Underlying amyloidosis, LIP and lymphomas should be explored [167, 184]. Thin-walled cysts can also occur in the absence of other parenchymal lesions (Figure 7). Frequently, they have peribronchovascular distribution [167].

Figure 7.

Multiple thin-walled cysts noted on CT imaging-central predominance. Surrounding lung appears otherwise grossly unremarkable.

20.1 Lymphoma

Sjogren’s syndrome has a higher risk for lymphoma as compared with other CTDs, with a standardized incidence ratio of 37.5, 95%,CI 20.7–67.6 [185]. An estimate of 5% of all SS patients may develop lymphoma with the low-grade extranodal marginal zone B-cell lymphoma (MZL) type being the most relevant histological type [186]. Chronic inflammation adjacent to epithelial cells may predispose to the generation of mucosal-associated lymphocyte tissue (MALT) hyperplasia and in the lungs labeled as bronchial-associated lymphocyte tissue, BALT [144]. The ongoing, relentless, and uncontrolled antigenic stimulus may promote activation of pro-oncogenic genes within lymph nodes or in extranodal lymphocytic aggregates (such as in the lungs), particularly under presence of germinal centers, driving them to endure a monoclonal transformation [187, 188]. MALT lymphomas surge from the marginal zone of B-cells that are localized surrounding the mantle zone and germinal centers, with the denomination of non-Hodgkin’s lymphoma, arising from the extranodal marginal zone B-cell. Other types of B-cell lymphomas are present as well. Frequency of pulmonary lymphomas is of 1%–2% [189], and predictors are difficult to define due to the scarcity of cases. Symptoms reveal a dry, chronic cough, and slowly progressive dyspnea or may run undetected [190]. Few patients may have constitutional symptoms (B-symptoms, lymphadenopathy, fever, weight loss, sweats, malaise, etc.) [191]. Findings on the chest HRCT are bronchial wall thickening and bronchiectasis, preferably in lower lobes. Lung parenchyma surrounding the abnormal airways may associate with confluent alveolar opacifications or ground-glass changes. Nodular densities are common [191]. The biopsies will disclose lymphoepithelial lesions involving the bronchial and bronchiolar epithelium, positive CD20 stain, clonal kappa/lambda distribution, Ki-67 proliferation, and abnormal gene rearrangement. Prominent plasma cell proliferation was observed on flow cytometry [191]. Therapy is based on use of alkylating agents and a combination of chemotherapy drugs. Rituximab seems to be the current standard of care as most of plasma cells express CD20 marker [191]. Most of the combination therapies set lymphoma in remission.

21. Amyloidosis in pulmonary Sjogren’s syndrome

This is a disorder caused by fibrillary plasma protein deposits on different tissues [192]. Rare cases of amyloidosis in SS are present, mainly in the skin and lungs, and very unusual, systemic amyloidosis. Other sites are reported including the tracheobronchial walls, kidneys, lacrimal glands, tongue, and mammary glands. When it affects the lungs or the tracheobronchial wall, symptoms may ensue, such as cough, dyspnea, pleuritic pain, and hemoptysis [193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203]. Amyloid composition in SS is usually of AL type (lambda or kappa) light chains, or less commonly, AA amyloid type [192]. Women are more frequently affected [195]. Radiographic images will reveal nodules, either calcified or not, and of different heterogeneity. They associate with cystic lesions. Histopathology will reveal infiltration of lymphocytes, plasma cells and amyloid deposits. Amorphous eosinophilic material and Congo red staining will reveal apple-green birefringence. MALT lymphoma should be explored as both associate frequently. Lymphocytic interstitial pneumonia is also frequently present with related cystic formation. The epithelium of cystic lesions may contain amyloid deposits [167]. Symptomatic therapy and glucocorticoids may be of help [192].

22. Pulmonary arterial hypertension, PAH

This is a rare condition in SS; however, the epidemiology reveals new data. Studies in East Asian ethnic group demonstrated a high frequency, such as systemic lupus erythematosus, in nearly half of a large group of 129 patients with confirmed PAH [204]. Women of ages between 30 and 40 are the most affected group [205]. Serology might be of great utility, as well as the biopsy of minor salivary glands. Prognosis at 1, 3, and 5 years is 80.2%, 74.8%, and 67.4%, respectively [206], being the anti-SSB/La antibodies poor predictors [207]. Of course, anti-phospholipid antibody syndrome always should be ruled out among other hypercoagulable risk factors to cause PAH [208].

23. Treatment of Sjogren syndrome and its pulmonary manifestations

23.1 Brief history

Therapy initially relied on symptom management, based on cholinergic pharmacologic drugs (e.g., Pilocarpine), mucolytics, but also radiation therapy (mostly targeting the parotid glands) [61]. The pivotal management, however, and since the 1950s, was based on glucocorticoids [209, 210]. Introduction of hydroxychloroquine in SS was not until the late 1980s and 1990s, and immune modulators have been extensively tried in case reports and case series [211, 212]. In 1998, Schnabel introduced IV cyclophosphamide for pulmonary disease and reported in cases of pseudolymphoma and associated lymphoma with pulmonary compromise [213, 214, 215]. Other immune modulators have been used, mainly on cases with advanced disease. Among the therapy, azathioprine [79, 216] and mycophenolate mofetil [217, 218] have been reported. Biologic therapy, such as rituximab, a humanized monoclonal antibody targeting mature B-cells (CD20+), was tried in case series and reports with promising results [219], in reparatory cases [220] and also associated with lymphoproliferative diseases [221, 222, 223], in special cases, such as in shrinking lung syndrome associated with SS [224]. Off-the-label cases reported other biologicals, such as tocilizumab [225] and abatacept [226].

24. The overall approach

Pulmonary complications of SS primarily comprise airway mucosal dryness (Xerotrachea), a range of interstitial lung diseases (ILDs), non-Hodgkin lymphomas, pleural effusion and/or thickening, and very rarely it can also cause pulmonary hypertension and/or thromboembolic phenomenon [12, 227].

We will layout an overview of symptomatic treatment of SS and sicca symptoms before discussing treatment of SS-associated pulmonary pathologies. Sicca symptoms are a common feature in most patients with SS, and its treatment can lead to dramatic symptomatic improvement in patients’ health and understanding of the disease. Basic measures for prevention of dry eyes and dry mouth should be advised in all patients [228]. These include maintenance of good oral hygiene [229], avoidance of coffee, alcohol, and sugar-filled liquids [230]. Use of artificial tears and avoidance of medications with anticholinergic properties should be stressed, especially drugs used for urinary incontinence such as oxybutynin [231]. Furthermore, this strategy can be compounded with addition of muscarinic agonists such as cevimeline and pilocarpine. These medications are collectively called sialagogues and have been shown to increase salivary flow and improvement in xerostomia in several randomized trials [232, 233, 234].

There have been investigative developments in oral electrostimulators, which induce salivary production and flow [235, 236]. However, their usage is vastly limited due to lack of larger trials, greater efficacy of medications, and cumbersome device management.

Dry eyes (Xerophthalmia) can often be the main feature of SS presentation and is usually the most frustrating symptom faced by most patients. Daily use of artificial tears and nightly use of oral lubricant are highly advised. In patients who still complain of dry and itchy eyes after these measures, topical cyclosporine and/or lifitegrast can be utilized [237]. Topical cyclosporine emulsions can be used with daily use of artificial tears, its efficacy is shown to increase with more frequent daily applications [238, 239, 240].

Managing nasal dryness is an important consideration as nasal congestion can lead to mouth breathing and worsening of xerostomia. Nasal dryness can be effectively managed with intermittent nasal saline sprays and room humidifiers. Laryngopharyngeal reflux (LPR) is a known manifestation of SS affecting aerodigestive tract. This should be treated with anti-GERD therapy to mitigate the erosive effects of gastric acid on laryngeal structures [241].

25. Treatment of interstitial lung disease

Management of most SS-related ILDs is based on empiric treatment options as no randomized controlled trials (RCTs) have yet been performed. It is also important to understand that treatment of SS-ILD is based on longitudinal worsening in HRCT/PFT results and overall symptomatic nature of the disease. Asymptomatic patients with mild ILD based on HRCT/PFT may not need a lung biopsy for confirmation of exact ILD type and may be monitored with HRCT/PFT every 6–12 months to assess disease progression. NSIP is often the most common histopathology found with SS-ILD, with UIP being second most common. LIP is rarely observed, but it is classically associated with SS-ILD [85].

Patients with symptomatic SS-related NSIP need treatment and are usually started on prednisone 1 mg/kg up to a total of 60 mg per day [75]. Patients should be assessed in 4–6 weeks with a PFT and symptom evaluation. If improvement is observed, low-dose prednisone is usually continued for at least 6 months with subsequent PFT/HRCT and symptom evaluations. Specific details on caution to use glucocorticoids overall are highlighted on the recently published consensus guidelines for the evaluation and management of pulmonary disease in Sjogren’s syndrome [45].

Immunosuppressive regimen with azathioprine (AZA) or mycophenolate mofetil (MMF) should be considered for patients as a glucocorticoid sparing therapy to prevent developing side effects to it. Both AZA and MMF have been shown to stabilize FVC decline in patients with CTD-ILDs (these cohorts had patients with SS-ILD as well) [79, 242]. In a recently published retrospective study, patients with pSS-ILD (19 cases) had a modest FVC% and DLco% slope improvement over time with use of both AZA and more favorably MMF, revealing the efficacy of both immune modulators on this condition [243]. In the same study, utility of rituximab was controversial.

Rituximab is left as an emerging option for patients who have refractory lung disease after use of above mentioned strategies. The benefit to target CD20 receptors in mature B-cell lineage has the advantage to blockade any antibody-mediated autoimmune disease. Its usage has been demonstrated in few case reports and case series in SS [244, 245]; however, there is still need of larger controlled trials to formally evaluate its efficacy. Experiences on rituximab in systemic sclerosis and ILD (SSc-ILD) added to the conventional treatment (methotrexate, AZA, or MMF), showed improvement in FVC after 2 years of use [246], and it may not only be a drug as a rescue strategy over MMF [247], but also possibly a line of standard of care therapy in the future when used upfront of other immune modulators (MMF) [248].

Cyclophosphamide has been used in few cases of SS-related ILD. Experiences from cyclophosphamide use in patients with SSc-ILD show efficacy to prevent FCV decline [249, 250]; however, their usage is markedly limited due to toxic effects and loss of efficacy once the drug is stopped [250]. Substitutes for cyclophosphamide after the induction phase were proposed. During the maintenance phase, azathioprine [251] and MMF [252] have been shown to preserve FVC. In the later study, comparing MMF vs. cyclophosphamide during the induction phase, MMF showed similar results in efficacy but lower toxic drug effects, encouraging providers to consider MMF as the drug of choice [252].

Other conventional drugs, including cyclosporine, have shown to prevent the progression of ILD in SS, but owing to their systemic side effects, their use is limited [110].

Tocilizumab, an IL-6 receptor inhibitor, has been tested in patients with SSc-ILD. Results are promising, especially in those individuals with active disease (alveolitis, high Rodnan skin score, high acute-phase reactants, and early stages of the disease) [253]. The decrease in the FVC slope at 24 and 48 weeks comparing with the placebo group that did worse contrasted the positive findings not described on the primary goal as was the prevention of further cutaneous fibrosis. The following trials of this drug confirmed the efficacy to preserve FVC [254], prompting for its FDA approval for SSc-ILD in early 2021. The utility in SS-ILD has not been systemically explored; yet the evidence reveals good outcomes in SS patients presenting with arthritis [255]. It seems tocilizumab to be a promising therapy for active inflammatory lung disease in SS. As an example, in a case report of refractory organizing pneumonia associated with SS, the use of tocilizumab showed to be very effective [225]. Contrasting with IL-6 blockers, TNF-alpha blocker therapy did not seem to be effective in SS overall [256, 257].

Abatacept is another biological therapy. It encompasses the fusion of a cytotoxic T-lymphocyte-associated protein 4 to the FC portion of an IgG1, with high binding affinity to the CD80 and CD86 receptors of the antigen-presenting cell (APC). This way it blocks costimulatory interaction of CD80/CD86 receptors with the T cell receptor (CD28) necessary for T cell activation and proliferation [258]. Approved for rheumatoid arthritis, the experience reveals stabilization of RA-ILD based on HRCT and FVC prospective evaluations [259, 260, 261, 262, 263, 264, 265]. Most of the impact was seen in carriers of the NSIP subtype [263], a finding that follows UIP in frequency in RA. Despite the subtype, the overall evidence reveals benefits of abatacept in this disease [261, 264, 266]. The impact of methotrexate on patients on abatacept seemed not to cause any worse pulmonary function deterioration; however, methotrexate’s use in ILD raises questions on safety. The same may be considered with other medication class, such as the TNF-alpha blockers with frequent reports of worse ILD in the setting of CTDs [267]. In SS, the efficacy of abatacept has shown in salivary gland inflammatory findings and extraglandular manifestations [268]. A case report revealed improvement in pneumonitis while combining abatacept and tacrolimus [226], suggesting that synergy among both drugs potentiates efficacy on ILD settings.

Belimumab, a specific monoclonal antibody targeting the B-cell activating factor (BlyS), restores circulating B cell numbers, composition, and activity in patients with SS [269]. The BELISS open-label trial of 30 patients with pSS revealed a decline in ESSDAI at 28 weeks, with main improvement in fatigue, but not in sicca symptoms [270]. However, this study did not mention effects on the respiratory system. Main changes in extraglandular manifestations are expected to be observed when its application is sequentially combined with rituximab therapy [271]. Current data regarding the impact of such therapy on the respiratory system are awaited.

Even the backbone in the pathogenesis of SS has a cytokine signature orchestrated by interferon I and II [272, 273], trials on SS of anti-interferon therapies are missing. As compared with lupus trials with this type of treatment modality [274], results of several trials in SS are still pending.

26. Future treatment perspectives for SS-ILD

Two promising antifibrotic drugs, pirfenidone and nintedanib, initially indicated and approved for idiopathic pulmonary fibrosis (IPF), with clear benefits in retarding the annual FVC decline and stabilization, have been recently explored in non-IPF pulmonary fibrotic progressive phenotype. This latter group encompasses different diseases that include sarcoidosis, interstitial pneumonitis, idiopathic NSIP, and unclassifiable idiopathic interstitial pneumonia [275]. Considering this group to share similar pathogenic pathways as in IPF, in that the pulmonary function declines in time along with ominous outcomes, the application of these drugs in this group is reasonable.

Nintedanib, an indolione derivate, has tyrosine kinase inhibitory activity and initially tested as an anticancer drug [276], blocks different profibrotic pathways: fibroblast growth factor receptors, vascular endothelial growth factor receptors, platelet-derived growth factors, and other tyrosine kinases, cytokines and chemokines (CCL18) [277]. With properties to reduce the proliferation and migration of lung fibrocytes and few adverse effects, it has been an effective drug in IPF. Likewise, pirfenidone, a small molecule, a pyridine derivate, inhibits PDGF, transforming growth factor β1 (TGFβ1) [278, 279] and promotes the balance between profibrotic and antifibrotic metalloproteinases [280], in addition to inhibiting proinflammatory cytokines [281].

In the INBUILD trial, 633 patients who had non-IPF progressive fibrosing ILD and that included CTD-ILD patients (i.e., RA patients 89/633, 13%) [282] were tested either with nintedanib or conventional therapy (placebo). Annual FVC showed decreased slope decline in the nintedanib group across all etiologies, including the CTD-ILD group, and without differences even in the UIP-like subgroup [283]. Patients with CTD-ILD group stopped their immune suppressor drugs prior to enrolling in the trial, to avoid confounders [282]. These findings were the ground to establish evidence for nintedanib efficacy in non-IPF progressive fibrosing ILDs independently of the diagnosis, enabling the FDA approval for its use. Even most of the experience falls into patients with SSc-ILD and RA-ILD, based on this evidence, SS-ILD may benefit from nintedanib as well. It is worth to mention the phase III SENSCIS trial on 576 patients with SSc-ILD testing nintedanib vs. placebo. Results showed a lower annual rate of decline in FVC (primary endpoint) with nintedanib than with placebo, but with more gastrointestinal adverse events in the nintedanib group [284]. Safety on the use of pirfenidone in SSc-ILD showed similar adverse events as in the placebo group, and moreover, combination of pirfenidone and MMF showed adequate tolerability and safety (LOTUSS trial) [285]. Further ongoing trials will reveal more information regarding safety and efficacy. Similarly, data are pending for RA-ILD and on pirfenidone (TRAIL-1, on phase II trial).

An anecdotal report on SS-ILD with UIP subtype showed promising experience in FVC preservation [286] as single evidence of the experience in SS patients. In this case series, pirfenidone was the applied drug to treat ILD. Again, further studies will reveal much more information on these promising drugs [287].

Until more trials show documented efficacy of the abovementioned regimens, treatment for SS-ILD will be subjected to individual clinical scenarios and physician preferences.

27. Conclusions

Extraglandular expression of SS may encompass many organs, with the respiratory system as one of the most frequently affected systems, carrying significant morbidity. With the lower airways being the most common manifestations in SS, the upper airways may associate with multiple presentations, including sinusitis. The airway disease should be acknowledged to be part of the syndrome, particularly when patients have poor response to therapy and behave differently to bronchial asthma. Airway disease treatment may challenge conventional strategies offered. Although less frequent, ILD may carry most of the main problems. Mortality associates with severe parenchymal disease, shortening life expectations in vulnerable groups (older age, smokers, males, and longer disease duration). Many other parenchymal manifestations are associated with SS such as LIP, amyloidosis, cystic lesions, lymphoma, and pulmonary hypertension and should be contemplated in the differentials. Clinicians should follow up patients with SS keeping in mind that the manifestations of the respiratory system may present at any point in time. Conventional therapies are available with variable results such as mycophenolate mofetil, azathioprine, cyclophosphamide, cyclosporine, and biological therapy. Among the latter, IL-6 inhibitors, costimulatory receptor antagonists, B-cell antagonist therapy, and other cytokine blocker therapy, including interferon blockers, seem to offer promising and safe profiles for the treatment of SS-ILD. New promising antifibrotic therapy (e.g., nintedanib and pirfenidone) will probably change the outcome panorama in SS-ILD. Combination of therapies seems to be an excellent modality to treat difficult cases.

References

  1. 1. Parke AL, Buchanan WW. Sjogren’s syndrome: History, clinical and pathological features. Inflammopharmacology. 1998;6(4):271-287
  2. 2. Fox RI, Howell FV, Bone RC, Michelson P. Primary Sjogren syndrome: Clinical and immunopathologic features. Seminars in Arthritis and Rheumatism. 1984;14(2):77-105
  3. 3. Haldorsen K, Bjelland I, Bolstad AI, Jonsson R, Brun JG. A five-year prospective study of fatigue in primary Sjogren’s syndrome. Arthritis Research & Therapy. 2011;13(5):R167
  4. 4. Arends S, Meiners PM, Moerman RV, Kroese FG, Brouwer E, Spijkervet FK, et al. Physical fatigue characterises patient experience of primary Sjogren’s syndrome. Clinical and Experimental Rheumatology. 2017;35(2):255-261
  5. 5. Ramos-Casals M, Brito-Zeron P, Solans R, Camps MT, Casanovas A, Sopena B, et al. Systemic involvement in primary Sjogren’s syndrome evaluated by the EULAR-SS disease activity index: Analysis of 921 Spanish patients (GEAS-SS registry). Rheumatology (Oxford, England). 2014;53(2):321-331
  6. 6. Retamozo S, Acar-Denizli N, Rasmussen A, Horvath IF, Baldini C, Priori R, et al. Systemic manifestations of primary Sjogren’s syndrome out of the ESSDAI classification: Prevalence and clinical relevance in a large international, multi-ethnic cohort of patients. Clinical and Experimental Rheumatology. 2019;118(3):97-106
  7. 7. Asmussen K, Andersen V, Bendixen G, Schiodt M, Oxholm P. A new model for classification of disease manifestations in primary Sjogren’s syndrome: Evaluation in a retrospective long-term study. Journal of Internal Medicine. 1996;239(6):475-482
  8. 8. Ramos-Casals M, Acar-Denizli N, Vissink A, Brito-Zeron P, Li X, Carubbi F, et al. Childhood-onset of primary Sjogren’s syndrome: Phenotypic characterization at diagnosis of 158 children. Rheumatology (Oxford, England). 2021;60(10):4558-4567
  9. 9. Ramos-Casals M, Brito-Zeron P, Seror R, Bootsma H, Bowman SJ, Dorner T, et al. Characterization of systemic disease in primary Sjogren’s syndrome: EULAR-SS task force recommendations for articular, cutaneous, pulmonary and renal involvements. Rheumatology (Oxford, England). 2015;54(12):2230-2238
  10. 10. Dias C, Isenberg DA. Susceptibility of patients with rheumatic diseases to B-cell non-Hodgkin lymphoma. Nature Reviews Rheumatology. 2011;7(6):360-368
  11. 11. Flament T, Bigot A, Chaigne B, Henique H, Diot E, Marchand-Adam S. Pulmonary manifestations of Sjogren’s syndrome. European Respiratory Review: An Official Journal of the European Respiratory Society. 2016;25(140):110-123
  12. 12. Gupta S, Ferrada MA, Hasni SA. Pulmonary manifestations of primary Sjögren’s syndrome: Underlying immunological mechanisms, clinical presentation, and management. Frontiers in Immunology. 2019;10:1327
  13. 13. Fox RI. Sjogren’s syndrome. Lancet. 2005;366(9482):321-331
  14. 14. Mavragani CP, Moutsopoulos HM. The geoepidemiology of Sjogren’s syndrome. Autoimmunity Reviews. 2010;9(5):A305-A310
  15. 15. Qin B, Wang J, Yang Z, Yang M, Ma N, Huang F, et al. Epidemiology of primary Sjogren’s syndrome: A systematic review and meta-analysis. Annals of the Rheumatic Diseases. 2015;74(11):1983-1989
  16. 16. Patel R, Shahane A. The epidemiology of Sjogren’s syndrome. Clinical Epidemiology. 2014;6:247-255
  17. 17. Birlik M, Akar S, Gurler O, Sari I, Birlik B, Sarioglu S, et al. Prevalence of primary Sjogren’s syndrome in Turkey: A population-based epidemiological study. International Journal of Clinical Practice. 2009;63(6):954-961
  18. 18. Anagnostopoulos I, Zinzaras E, Alexiou I, Papathanasiou AA, Davas E, Koutroumpas A, et al. The prevalence of rheumatic diseases in Central Greece: A population survey. BMC Musculoskeletal Disorders. 2010;11:98
  19. 19. Goransson LG, Haldorsen K, Brun JG, Harboe E, Jonsson MV, Skarstein K, et al. The point prevalence of clinically relevant primary Sjogren’s syndrome in two Norwegian counties. Scandinavian Journal of Rheumatology. 2011;40(3):221-224
  20. 20. Peri Y, Agmon-Levin N, Theodor E, Shoenfeld Y. Sjögren’s syndrome, the old and the new. Best Practice & Research. Clinical Rheumatology. 2012;26(1):105-117
  21. 21. Cornec D, Devauchelle-Pensec V, Tobon GJ, Pers JO, Jousse-Joulin S, Saraux A. B cells in Sjogren’s syndrome: From pathophysiology to diagnosis and treatment. Journal of Autoimmunity. 2012;39(3):161-167
  22. 22. Fayyaz A, Kurien BT, Scofield RH. Autoantibodies in Sjogren’s Syndrome. Rheumatic Diseases Clinics of North America. 2016;42(3):419-434
  23. 23. Daniels TE, Cox D, Shiboski CH, Schiodt M, Wu A, Lanfranchi H, et al. Associations between salivary gland histopathologic diagnoses and phenotypic features of Sjogren’s syndrome among 1,726 registry participants. Arthritis and Rheumatism. 2011;63(7):2021-2030
  24. 24. Greenspan JS, Daniels TE, Talal N, Sylvester RA. The histopathology of Sjogren’s syndrome in labial salivary gland biopsies. Oral Surgery, Oral Medicine, and Oral Pathology. 1974;37(2):217-229
  25. 25. Fisher BA, Jonsson R, Daniels T, Bombardieri M, Brown RM, Morgan P, et al. Standardisation of labial salivary gland histopathology in clinical trials in primary Sjogren’s syndrome. Annals of the Rheumatic Diseases. 2017;76(7):1161-1168
  26. 26. Brito-Zeron P, Acar-Denizli N, Ng WF, Zeher M, Rasmussen A, Mandl T, et al. How immunological profile drives clinical phenotype of primary Sjogren’s syndrome at diagnosis: Analysis of 10,500 patients (Sjogren big data project). Clinical and Experimental Rheumatology. 2018;112(3):102-112
  27. 27. Ramos-Casals M, Brito-Zeron P, Siso-Almirall A, Bosch X. Primary Sjogren syndrome. BMJ. 2012;344:e3821
  28. 28. Shiboski CH, Shiboski SC, Seror R, Criswell LA, Labetoulle M, Lietman TM, et al. 2016 American College of Rheumatology/European league against rheumatism classification criteria for primary Sjogren’s syndrome: A consensus and data-driven methodology involving three international patient cohorts. Arthritis & Rheumatology. 2017;69(1):35-45
  29. 29. Defendenti C, Atzeni F, Spina MF, Grosso S, Cereda A, Guercilena G, et al. Clinical and laboratory aspects of Ro/SSA-52 autoantibodies. Autoimmunity Reviews. 2011;10(3):150-154
  30. 30. Jordan-Gonzalez P, Gago-Pinero R, Varela-Rosario N, Perez-Rios N, Vila LM. Characterization of a subset of patients with primary Sjogren’s syndrome initially presenting with C3 or C4 hypocomplementemia. European Journal of Rheumatology. 2020;7(3):112-117
  31. 31. Baldini C, Pepe P, Quartuccio L, Priori R, Bartoloni E, Alunno A, et al. Primary Sjogren’s syndrome as a multi-organ disease: Impact of the serological profile on the clinical presentation of the disease in a large cohort of Italian patients. Rheumatology (Oxford, England). 2014;53(5):839-844
  32. 32. Retamozo S, Brito-Zeron P, Ramos-Casals M. Prognostic markers of lymphoma development in primary Sjogren syndrome. Lupus. 2019;28(8):923-936
  33. 33. Theander E, Jonsson R, Sjostrom B, Brokstad K, Olsson P, Henriksson G. Prediction of Sjogren’s syndrome years before diagnosis and identification of patients with early onset and severe disease course by autoantibody profiling. Arthritis & Rheumatology. 2015;67(9):2427-2436
  34. 34. Arbuckle MR, McClain MT, Rubertone MV, Scofield RH, Dennis GJ, James JA, et al. Development of autoantibodies before the clinical onset of systemic lupus erythematosus. The New England Journal of Medicine. 2003;349(16):1526-1533
  35. 35. Aoshiba K, Tsuji T, Yamaguchi K, Itoh M, Nakamura H. The danger signal plus DNA damage two-hit hypothesis for chronic inflammation in COPD. The European Respiratory Journal. 2013;42(6):1689-1695
  36. 36. Beckman KA, Luchs J, Milner MS. Making the diagnosis of Sjogren’s syndrome in patients with dry eye. Clinical Ophthalmology. 2016;10:43-53
  37. 37. Hernandez-Molina G, Nunez-Alvarez C, Avila-Casado C, Llorente L, Hernandez-Hernandez C, Calderillo ML, et al. Usefulness of IgA anti-alpha-fodrin antibodies in combination with rheumatoid factor and/or antinuclear antibodies as substitute immunological criterion in Sjogren syndrome with negative anti-SSA/SSB antibodies. The Journal of Rheumatology. 2016;43(10):1852-1857
  38. 38. He J, Guo JP, Ding Y, Li YN, Pan SS, Liu Y, et al. Diagnostic significance of measuring antibodies to cyclic type 3 muscarinic acetylcholine receptor peptides in primary Sjogren’s syndrome. Rheumatology (Oxford, England). 2011;50(5):879-884
  39. 39. Routsias JG, Tzioufas AG. Sjogren’s syndrome—Study of autoantigens and autoantibodies. Clinical Reviews in Allergy & Immunology. 2007;32(3):238-251
  40. 40. Karakus S, Baer AN, Akpek EK. Clinical correlations of novel autoantibodies in patients with dry eye. Journal of Immunology Research. 2019;2019:7935451
  41. 41. Shen L, Suresh L, Lindemann M, Xuan J, Kowal P, Malyavantham K, et al. Novel autoantibodies in Sjogren’s syndrome. Clinical Immunology. 2012;145(3):251-255
  42. 42. Brito-Zeron P, Acar-Denizli N, Zeher M, Rasmussen A, Seror R, Theander E, et al. Influence of geolocation and ethnicity on the phenotypic expression of primary Sjogren’s syndrome at diagnosis in 8310 patients: A cross-sectional study from the big data Sjogren project consortium. Annals of the Rheumatic Diseases. 2017;76(6):1042-1050
  43. 43. Kassan SS, Moutsopoulos HM. Clinical manifestations and early diagnosis of Sjogren syndrome. Archives of Internal Medicine. 2004;164(12):1275-1284
  44. 44. Vitali C, Bombardieri S, Jonsson R, Moutsopoulos HM, Alexander EL, Carsons SE, et al. Classification criteria for Sjogren’s syndrome: A revised version of the European criteria proposed by the American-European consensus group. Annals of the Rheumatic Diseases. 2002;61(6):554-558
  45. 45. Lee AS, Scofield RH, Hammitt KM, Gupta N, Thomas DE, Moua T, et al. Consensus guidelines for evaluation and management of pulmonary disease in Sjogren’s. Chest. 2021;159(2):683-698
  46. 46. Rasmussen A, Ice JA, Li H, Grundahl K, Kelly JA, Radfar L, et al. Comparison of the American-European consensus group Sjogren’s syndrome classification criteria to newly proposed American College of Rheumatology criteria in a large, carefully characterised sicca cohort. Annals of the Rheumatic Diseases. 2014;73(1):31-38
  47. 47. Fox RI, Robinson CA, Curd JG, Kozin F, Howell FV. Sjogren’s syndrome. Proposed criteria for classification. Arthritis and Rheumatism. 1986;29(5):577-585
  48. 48. Vitali C, Bombardieri S, Moutsopoulos HM, Balestrieri G, Bencivelli W, Bernstein RM, et al. Preliminary criteria for the classification of Sjogren’s syndrome. Results of a prospective concerted action supported by the European Community. Arthritis and Rheumatism. 1993;36(3):340-347
  49. 49. Bloch KJ, Buchanan WW, Wohl MJ, Bunim JJ. Sjoegren’s syndrome. A clinical, pathological, and serological study of sixty-two cases. Medicine (Baltimore). 1965;44:187-231
  50. 50. Bombardieri M, Argyropoulou OD, Ferro F, Coleby R, Pontarini E, Governato G, et al. One year in review 2020: Pathogenesis of primary Sjogren’s syndrome. Clinical and Experimental Rheumatology. 2020;38 Suppl 126(4):3-9
  51. 51. Leber T. Praparate zu dem Vortag uber Entstnhung der Netzhautablosung und uber verschiedene Hornhautaffecttionen. Ber Ophthalmol Ges Heidelberg. 1882;14:165-166
  52. 52. Hadden WB. On “dry mouth,” or suppression of the salivary and buccal secretions. Transactions of the Clinical Society of London. 1888;21:176-179
  53. 53. Bloch KJ, Wohl MJ, Ship II, Oglesby BB, Bunim JJ, Sjogren’s syndrome. 1. Serologic reactions in patients with Sjogren’s syndrome with and without rheumatoid arthritis. Arthritis and Rheumatism. 1960;3:287-297
  54. 54. Fischer E. Uber Fadchenkeratitis. Graefe’s Archive. 1889;35:201
  55. 55. Houwer AM. Keratitis filamentosa and chronic arthritis. Transactions of the Ophthalmological Societies of the United Kingdom. 1927;47:88-96
  56. 56. Holm S, Sjogren H, et al. Studies on keratoconjunctivitis sicca, based on examination of 500 subjects affected with rheumatism and an equally large control material. Acta Ophthalmologica. 1948;26(2):269-273
  57. 57. Hill LC. Systemic lupus erythematosus. British Medical Journal. 1957;2(5047):726-732
  58. 58. Jones BR. Lacrimal and salivary precipitating antibodies in Sjogren’s syndrome. Lancet. 1958;2(7050):773-776
  59. 59. Delaleu N, Jonsson R, Koller MM. Sjogren’s syndrome. European Journal of Oral Sciences. 2005;113(2):101-113
  60. 60. Bunim JJ. A broader spectrum of Sjogren’s syndrome and its pathogenetic implications. Annals of the Rheumatic Diseases. 1961;20:1-10
  61. 61. Weber FP. Sjogren’s syndrome, especially its non-ocular features. The British Journal of Ophthalmology. 1945;29(6):299-312
  62. 62. Baruch HH, Firooznia H, Sackler JP, Genieser NB, Rafii M, Golimbu C. Pulmonary disorders associated with Sjogren’s syndrome. Revista Interamericana de Radiología. 1977;2(2):77-81
  63. 63. Bucher UG, Reid L. Sjogren’s syndrome: Report of a fatal case with pulmonary and renal lesions. British Journal of Diseases of the Chest. 1959;53:237-252
  64. 64. Ellman P, Weber FP, Goodier TE. A contribution to the pathology of Sjogren’s disease. The Quarterly Journal of Medicine. 1951;20(77):33-42
  65. 65. Leventhal BG, Waldorf DS, Talal N. Impaired lymphocyte transformation and delayed hypersensitivity in Sjogren’s syndrome. The Journal of Clinical Investigation. 1967;46(8):1338-1345
  66. 66. Talal N, Bunim JJ. The development of malignant lymphoma in the course of Sjoegren’s syndrome. The American Journal of Medicine. 1964;36:529-540
  67. 67. Talal N, Sokoloff L, Barth WF. Extrasalivary lymphoid abnormalities in Sjogren’s syndrome (reticulum cell sarcoma, “pseudolymphoma,” macroglobulinemia). The American Journal of Medicine. 1967;43(1):50-65
  68. 68. Anderson LG, Talal N. The spectrum of benign to malignant lymphoproliferation in Sjogren’s syndrome. Clinical and Experimental Immunology. 1972;10(2):199-221
  69. 69. Bonner H Jr, Ennis RS, Geelhoed GW, Tarpley TM Jr. Lymphoid infiltration and amyloidosis of lung in Sjogren’s syndrome. Archives of Pathology. 1973;95(1):42-44
  70. 70. Fairfax AJ, Haslam PL, Pavia D, Sheahan NF, Bateman JR, Agnew JE, et al. Pulmonary disorders associated with Sjogren’s syndrome. The Quarterly Journal of Medicine. 1981;50(199):279-295
  71. 71. Strimlan CV, Rosenow EC 3rd, Weiland LH, Brown LR. Lymphocytic interstitial pneumonitis. Review of 13 cases. Annals of Internal Medicine. 1978;88(5):616-621
  72. 72. Yamamoto Y, Otsuka Y, Katsuyama T, Nishimura Y, Oka K, Hasegawa K, et al. An elderly male with primary Sjogren’s syndrome presenting Pleuritis as the initial manifestation. Acta Medica Okayama. 2021;75(4):539-542
  73. 73. Teshigawara K, Kakizaki S, Horiya M, Kikuchi Y, Hashida T, Tomizawa Y, et al. Primary Sjogren’s syndrome complicated by bilateral pleural effusion. Respirology. 2008;13(1):155-158
  74. 74. Sato T, Hatano M, Iwasaki Y, Maki H, Saito A, Minatsuki S, et al. Prevalence of primary Sjogren’s syndrome in patients undergoing evaluation for pulmonary arterial hypertension. PLoS One. 2018;13(5):e0197297
  75. 75. Parambil JG, Myers JL, Lindell RM, Matteson EL, Ryu JH. Interstitial lung disease in primary Sjögren syndrome. Chest. 2006;130(5):1489-1495
  76. 76. Sambataro G, Ferro F, Orlandi M, Sambataro D, Torrisi SE, Quartuccio L, et al. Clinical, morphological features and prognostic factors associated with interstitial lung disease in primary Sjgren’s syndrome: A systematic review from the Italian Society of Rheumatology. Autoimmunity Reviews. 2020;19(2):102447
  77. 77. Natalini JG, Johr C, Kreider M. Pulmonary involvement in Sjogren syndrome. Clinics in Chest Medicine. 2019;40(3):531-544
  78. 78. Davidson BK, Kelly CA, Griffiths ID. Ten year follow up of pulmonary function in patients with primary Sjogren’s syndrome. Annals of the Rheumatic Diseases. 2000;59(9):709-712
  79. 79. Deheinzelin D, Capelozzi VL, Kairalla RA, Barbas Filho JV, Saldiva PH, de Carvalho CR. Interstitial lung disease in primary Sjögren’s syndrome. Clinical-pathological evaluation and response to treatment. American Journal of Respiratory and Critical Care Medicine. 1996;154(3 Pt 1):794-799
  80. 80. Yazisiz V, Arslan G, Ozbudak IH, Turker S, Erbasan F, Avci AB, et al. Lung involvement in patients with primary Sjogren’s syndrome: What are the predictors? Rheumatology International. 2010;30(10):1317-1324
  81. 81. Garcia-Carrasco M, Ramos-Casals M, Rosas J, Pallares L, Calvo-Alen J, Cervera R, et al. Primary Sjogren syndrome: Clinical and immunologic disease patterns in a cohort of 400 patients. Medicine (Baltimore). 2002;81(4):270-280
  82. 82. Uffmann M, Kiener HP, Bankier AA, Baldt MM, Zontsich T, Herold CJ. Lung manifestation in asymptomatic patients with primary Sjogren syndrome: Assessment with high resolution CT and pulmonary function tests. Journal of Thoracic Imaging. 2001;16(4):282-289
  83. 83. Strimlan CV, Rosenow EC 3rd, Divertie MB, Harrison EG Jr. Pulmonary manifestations of Sjogren’s syndrome. Chest. 1976;70(03):354-361
  84. 84. Papiris SA, Maniati M, Constantopoulos SH, Roussos C, Moutsopoulos HM, Skopouli FN. Lung involvement in primary Sjogren’s syndrome is mainly related to the small airway disease. Annals of the Rheumatic Diseases. 1999;58(1):61-64
  85. 85. Roca F, Dominique S, Schmidt J, Smail A, Duhaut P, Levesque H, et al. Interstitial lung disease in primary Sjogren’s syndrome. Autoimmunity Reviews. 2017;16(1):48-54
  86. 86. Kurumagawa T, Kobayashi H, Motoyoshi K. Potential involvement of subclinical Sjogren’s syndrome in various lung diseases. Respirology. 2005;10(1):86-91
  87. 87. Egashira R, Kondo T, Hirai T, Kamochi N, Yakushiji M, Yamasaki F, et al. CT findings of thoracic manifestations of primary Sjogren syndrome: Radiologic-pathologic correlation. Radiographics: A review publication of the Radiological Society of North America. Inc. 2013;33(7):1933-1949
  88. 88. Nakanishi M, Fukuoka J, Tanaka T, Demura Y, Umeda Y, Ameshima S, et al. Small airway disease associated with Sjogren’s syndrome: Clinico-pathological correlations. Respiratory Medicine. 2011;105(12):1931-1938
  89. 89. Nannini C, Jebakumar AJ, Crowson CS, Ryu JH, Matteson EL. Primary Sjogren’s syndrome 1976-2005 and associated interstitial lung disease: A population-based study of incidence and mortality. BMJ Open. 2013;3(11):e003569
  90. 90. Palm O, Garen T, Berge Enger T, Jensen JL, Lund MB, Aalokken TM, et al. Clinical pulmonary involvement in primary Sjogren’s syndrome: Prevalence, quality of life and mortality—A retrospective study based on registry data. Rheumatology (Oxford, England). 2013;52(1):173-179
  91. 91. Belenguer R, Ramos-Casals M, Brito-Zeron P, del Pino J, Sentis J, Aguilo S, et al. Influence of clinical and immunological parameters on the health-related quality of life of patients with primary Sjogren’s syndrome. Clinical and Experimental Rheumatology. 2005;23(3):351-356
  92. 92. Kim HJ, Kim KH, Hann HJ, Han S, Kim Y, Lee SH, et al. Incidence, mortality, and causes of death in physician-diagnosed primary Sjogren’s syndrome in Korea: A nationwide, population-based study. Seminars in Arthritis and Rheumatism. 2017;47(2):222-227
  93. 93. Huang H, Xie W, Geng Y, Fan Y, Zhang Z. Mortality in patients with primary Sjogren’s syndrome: A systematic review and meta-analysis. Rheumatology (Oxford, England). 2021;60(9):4029-4038
  94. 94. Singh AG, Singh S, Matteson EL. Rate, risk factors and causes of mortality in patients with Sjogren’s syndrome: A systematic review and meta-analysis of cohort studies. Rheumatology (Oxford, England). 2016;55(3):450-460
  95. 95. Lin DF, Yan SM, Zhao Y, Zhang W, Li MT, Zeng XF, et al. Clinical and prognostic characteristics of 573 cases of primary Sjogren’s syndrome. Chinese Medical Journal. 2010;123(22):3252-3257
  96. 96. Gao H, Sun Y, Zhang XY, Xie L, Zhang XW, Zhong YC, et al. Characteristics and mortality in primary Sjogren syndrome-related interstitial lung disease. Medicine (Baltimore). 2021;100(35):e26777
  97. 97. Enomoto Y, Takemura T, Hagiwara E, Iwasawa T, Fukuda Y, Yanagawa N, et al. Prognostic factors in interstitial lung disease associated with primary Sjogren’s syndrome: A retrospective analysis of 33 pathologically-proven cases. PLoS One. 2013;8(9):e73774
  98. 98. Matsuyama N, Ashizawa K, Okimoto T, Kadota J, Amano H, Hayashi K. Pulmonary lesions associated with Sjogren’s syndrome: Radiographic and CT findings. The British Journal of Radiology. 2003;76(912):880-884
  99. 99. Dong X, Zhou J, Guo X, Li Y, Xu Y, Fu Q, et al. A retrospective analysis of distinguishing features of chest HRCT and clinical manifestation in primary Sjogren’s syndrome-related interstitial lung disease in a Chinese population. Clinical Rheumatology. 2018;37(11):2981-2988
  100. 100. Kampolis CF, Fragkioudaki S, Mavragani CP, Zormpala A, Samakovli A, Moutsopoulos HM. Prevalence and spectrum of symptomatic pulmonary involvement in primary Sjogren’s syndrome. Clinical and Experimental Rheumatology. 2018;112(3):94-101
  101. 101. Manfredi A, Sebastiani M, Cerri S, Cassone G, Bellini P, Casa GD, et al. Prevalence and characterization of non-sicca onset primary Sjogren syndrome with interstitial lung involvement. Clinical Rheumatology. 2017;36(6):1261-1268
  102. 102. Wang Y, Hou Z, Qiu M, Ye Q. Risk factors for primary Sjogren syndrome-associated interstitial lung disease. Journal of Thoracic Disease. 2018;10(4):2108-2117
  103. 103. Parambil JG, Myers JL, Lindell RM, Matteson EL, Ryu JH. Interstitial lung disease in primary Sjogren syndrome. Chest. 2006;130(5):1489-1495
  104. 104. Chen MH, Chou HP, Lai CC, Chen YD, Chen MH, Lin HY, et al. Lung involvement in primary Sjogren’s syndrome: Correlation between high-resolution computed tomography score and mortality. Journal of the Chinese Medical Association. 2014;77(2):75-82
  105. 105. Yan S, Li M, Wang H, Yang X, Zhao J, Wang Q, et al. Characteristics and risk factors of pulmonary arterial hypertension in patients with primary Sjogren’s syndrome. International Journal of Rheumatic Diseases. 2018;21(5):1068-1075
  106. 106. Fox RI. The incidence of pulmonary hypertension is higher in systemic lupus and Sjogren’s patients than in scleroderma patients in China. Lupus. 2018;27(7):1051-1052
  107. 107. Hatron PY, Wallaert B, Gosset D, Tonnel AB, Gosselin B, Voisin C, et al. Subclinical lung inflammation in primary Sjogren’s syndrome. Relationship between bronchoalveolar lavage cellular analysis findings and characteristics of the disease. Arthritis and Rheumatism. 1987;30(11):1226-1231
  108. 108. Papiris SA, Saetta M, Turato G, La Corte R, Trevisani L, Mapp CE, et al. CD4-positive T-lymphocytes infiltrate the bronchial mucosa of patients with Sjogren’s syndrome. American Journal of Respiratory and Critical Care Medicine. 1997;156(2 Pt 1):637-641
  109. 109. Ito I, Nagai S, Kitaichi M, Nicholson AG, Johkoh T, Noma S, et al. Pulmonary manifestations of primary Sjogren’s syndrome: A clinical, radiologic, and pathologic study. American Journal of Respiratory and Critical Care Medicine. 2005;171(6):632-638
  110. 110. Shi JH, Liu HR, Xu WB, Feng RE, Zhang ZH, Tian XL, et al. Pulmonary manifestations of Sjogren’s syndrome. Respiration; International Review of Thoracic Diseases. 2009;78(4):377-386
  111. 111. Kelly C, Gardiner P, Pal B, Griffiths I. Lung function in primary Sjogren’s syndrome: A cross sectional and longitudinal study. Thorax. 1991;46(3):180-183
  112. 112. Seror R, Ravaud P, Bowman SJ, Baron G, Tzioufas A, Theander E, et al. EULAR Sjogren’s syndrome disease activity index: Development of a consensus systemic disease activity index for primary Sjogren’s syndrome. Annals of the Rheumatic Diseases. 2010;69(6):1103-1109
  113. 113. Seror R, Bootsma H, Bowman SJ, Dorner T, Gottenberg JE, Mariette X, et al. Outcome measures for primary Sjogren’s syndrome. Journal of Autoimmunity. 2012;39(1-2, 102):97
  114. 114. Seror R, Bowman SJ, Brito-Zeron P, Theander E, Bootsma H, Tzioufas A, et al. EULAR Sjogren’s syndrome disease activity index (ESSDAI): A user guide. RMD Open. 2015;1(1):e000022
  115. 115. Seror R, Bootsma H, Saraux A, Bowman SJ, Theander E, Brun JG, et al. Defining disease activity states and clinically meaningful improvement in primary Sjogren’s syndrome with EULAR primary Sjogren’s syndrome disease activity (ESSDAI) and patient-reported indexes (ESSPRI). Annals of the Rheumatic Diseases. 2016;75(2):382-389
  116. 116. Chang GH, Chen YC, Lin KM, Yang YH, Liu CY, Lin MH, et al. Real-world database examining the association between Sjogren’s syndrome and chronic rhinosinusitis. Journal of Clinical Medicine. 2019;8(2):155-166
  117. 117. Mathieu A, Cauli A, Pala R, Satta L, Nurchis P, Loi GL, et al. Tracheo-bronchial mucociliary clearance in patients with primary and secondary Sjogren’s syndrome. Scandinavian Journal of Rheumatology. 1995;24(5):300-304
  118. 118. Amin K, Ludviksdottir D, Janson C, Nettelbladt O, Gudbjornsson B, Valtysdottir S, et al. Inflammation and structural changes in the airways of patients with primary Sjogren’s syndrome. Respiratory Medicine. 2001;95(11):904-910
  119. 119. Gardiner P, Ward C, Allison A, Ashcroft T, Simpson W, Walters H, et al. Pleuropulmonary abnormalities in primary Sjogren’s syndrome. The Journal of Rheumatology. 1993;20(5):831-837
  120. 120. Rusthen S, Kristoffersen AK, Young A, Galtung HK, Petrovski BE, Palm O, et al. Dysbiotic salivary microbiota in dry mouth and primary Sjogren’s syndrome patients. PLoS One. 2019;14(6):e0218319
  121. 121. Lee J, Alam J, Choi E, Ko YK, Lee A, Choi Y. Association of a dysbiotic oral microbiota with the development of focal lymphocytic sialadenitis in IkappaB-zeta-deficient mice. NPJ Biofilms Microbiomes. 2020;6(1):49
  122. 122. Tseng YC, Yang HY, Lin WT, Chang CB, Chien HC, Wang HP, et al. Salivary dysbiosis in Sjogren’s syndrome and a commensal-mediated immunomodulatory effect of salivary gland epithelial cells. NPJ Biofilms Microbiomes. 2021;7(1):21
  123. 123. Herrala M, Turunen S, Hanhineva K, Lehtonen M, Mikkonen JJW, Seitsalo H, et al. Low-dose doxycycline treatment normalizes levels of some salivary metabolites associated with oral microbiota in patients with primary Sjogren’s syndrome. Metabolites. 2021;11(9):595-608
  124. 124. Aljanobi H, Sabharwal A, Krishnakumar B, Kramer JM. Is it Sjogren’s syndrome or burning mouth syndrome? Distinct pathoses with similar oral symptoms. Oral Surgery, Oral Medicine, Oral Pathology and Oral Radiology. 2017;123(4):482-495
  125. 125. Graf S, Kirschstein L, Knopf A, Mansour N, Jeleff-Wolfler O, Buchberger AMS, et al. Systematic evaluation of laryngeal impairment in Sjogren’s syndrome. European Archives of Oto-Rhino-Laryngology. 2021;278(7):2421-2428
  126. 126. Stojan G, Baer AN, Danoff SK. Pulmonary manifestations of Sjogren’s syndrome. Current Allergy and Asthma Reports. 2013;13(4):354-360
  127. 127. Volter F, Fain O, Mathieu E, Thomas M. Esophageal function and Sjogren’s syndrome. Digestive Diseases and Sciences. 2004;49(2):248-253
  128. 128. Seeliger T, Bonig L, Witte T, Thiele T, Lesinski-Schiedat A, Stangel M, et al. Hearing dysfunction in patients with neuro-Sjogren: A cross-sectional study. Annals of Translational Medicine. 2020;8(17):1069
  129. 129. Quismorio FP Jr. Pulmonary involvement in primary Sjogren’s syndrome. Current Opinion in Pulmonary Medicine. 1996;2(5):424-428
  130. 130. Papathanasiou MP, Constantopoulos SH, Tsampoulas C, Drosos AA, Moutsopoulos HM. Reappraisal of respiratory abnormalities in primary and secondary Sjogren’s syndrome. A controlled study. Chest. 1986;90(3):370-374
  131. 131. Constantopoulos SH, Tsianos EV, Moutsopoulos HM. Pulmonary and gastrointestinal manifestations of Sjogren’s syndrome. Rheumatic Diseases Clinics of North America. 1992;18(3):617-635
  132. 132. Gudbjornsson B, Hedenstrom H, Stalenheim G, Hallgren R. Bronchial hyperresponsiveness to methacholine in patients with primary Sjogren’s syndrome. Annals of the Rheumatic Diseases. 1991;50(1):36-40
  133. 133. La Corte R, Potena A, Bajocchi G, Fabbri L, Trotta F. Increased bronchial responsiveness in primary Sjogren’s syndrome. A sign of tracheobronchial involvement. Clinical and Experimental Rheumatology. 1991;9(2):125-130
  134. 134. Stalenheim G, Gudbjornsson B. Anti-inflammatory drugs do not alleviate bronchial hyperreactivity in Sjogren’s syndrome. Allergy. 1997;52(4):423-427
  135. 135. Ludviksdottir D, Valtysdottir ST, Hedenstrom H, Hallgren R, Gudbjornsson B. Eight-year follow-up of airway hyperresponsiveness in patients with primary Sjogren’s syndrome. Upsala Journal of Medical Sciences. 2017;122(1):51-55
  136. 136. Bellido-Casado J, Plaza V, Diaz C, Geli C, Dominguez J, Margarit G, et al. Bronchial inflammation, respiratory symptoms and lung function in primary Sjogren’s syndrome. Archivos de Bronconeumología. 2011;47(7):330-334
  137. 137. Fortoul TI, Cano-Valle F, Oliva E, Barrios R. Follicular bronchiolitis in association with connective tissue diseases. Lung. 1985;163(5):305-314
  138. 138. Yousem SA, Colby TV, Carrington CB. Follicular bronchitis/bronchiolitis. Human Pathology. 1985;16(7):700-706
  139. 139. Ryu JH, Myers JL, Swensen SJ. Bronchiolar disorders. American Journal of Respiratory and Critical Care Medicine. 2003;168(11):1277-1292
  140. 140. Franquet T, Gimenez A, Monill JM, Diaz C, Geli C. Primary Sjogren’s syndrome and associated lung disease: CT findings in 50 patients. AJR American Journal of Roentgenology. 1997;169(3):655-658
  141. 141. Borie R, Schneider S, Debray MP, Adle-Biasssette H, Danel C, Bergeron A, et al. Severe chronic bronchiolitis as the presenting feature of primary Sjogren’s syndrome. Respiratory Medicine. 2011;105(1):130-136
  142. 142. Tashtoush B, Okafor NC, Ramirez JF, Smolley L. Follicular bronchiolitis: A literature review. Journal of Clinical and Diagnostic Research. 2015;9(9):OE01-OE05
  143. 143. Camarasa Escrig A, Amat Humaran B, Sapia S, Leon Ramirez JM. Follicular bronchiolitis associated with common variable immunodeficiency. Archivos de Bronconeumología. 2013;49(4):166-168
  144. 144. Travis WD, Galvin JR. Non-neoplastic pulmonary lymphoid lesions. Thorax. 2001;56(12):964-971
  145. 145. Halle S, Dujardin HC, Bakocevic N, Fleige H, Danzer H, Willenzon S, et al. Induced bronchus-associated lymphoid tissue serves as a general priming site for T cells and is maintained by dendritic cells. The Journal of Experimental Medicine. 2009;206(12):2593-2601
  146. 146. Mandl T, Diaz S, Ekberg O, Hesselstrand R, Piitulainen E, Wollmer P, et al. Frequent development of chronic obstructive pulmonary disease in primary SS—Results of a longitudinal follow-up. Rheumatology (Oxford, England). 2012;51(5):941-946
  147. 147. Takenaka S, Ogura T, Oshima H, Izumi K, Hirata A, Ito H, et al. Development and exacerbation of pulmonary nontuberculous mycobacterial infection in patients with systemic autoimmune rheumatic diseases. Modern Rheumatology. 2020;30(3):558-563
  148. 148. Chung A, Wilgus ML, Fishbein G, Lynch JP 3rd. Pulmonary and bronchiolar involvement in Sjogren’s syndrome. Seminars in Respiratory and Critical Care Medicine. 2019;40(2):235-254
  149. 149. Stenmark KR, Fagan KA, Frid MG. Hypoxia-induced pulmonary vascular remodeling: Cellular and molecular mechanisms. Circulation Research. 2006;99(7):675-691
  150. 150. Newball HH, Brahim SA. Chronic obstructive airway disease in patients with Sjogren’s syndrome. The American Review of Respiratory Disease. 1977;115(2):295-304
  151. 151. Kakugawa T, Sakamoto N, Ishimoto H, Shimizu T, Nakamura H, Nawata A, et al. Lymphocytic focus score is positively related to airway and interstitial lung diseases in primary Sjogren’s syndrome. Respiratory Medicine. 2018;137:95-102
  152. 152. Soto-Cardenas MJ, Perez-De-Lis M, Bove A, Navarro C, Brito-Zeron P, Diaz-Lagares C, et al. Bronchiectasis in primary Sjogren’s syndrome: Prevalence and clinical significance. Clinical and Experimental Rheumatology. 2010;28(5):647-653
  153. 153. Di Pasquale M, Aliberti S, Mantero M, Gramegna A, Blasi F. Pharmacotherapeutic management of bronchial infections in adults: Non-cystic fibrosis bronchiectasis and chronic obstructive pulmonary disease. Expert Opinion on Pharmacotherapy. 2020;21(16):1975-1990
  154. 154. Li X, Xu B, Ma Y, Li X, Cheng Q, Wang X, et al. Clinical and laboratory profiles of primary Sjogren’s syndrome in a Chinese population: A retrospective analysis of 315 patients. International Journal of Rheumatic Diseases. 2015;18(4):439-446
  155. 155. Liu C, Zhang H, Yao G, Hu Y, Qi J, Wang Y, et al. Characteristics of primary Sjogren’s syndrome patients with IgG4 positive plasma cells infiltration in the labial salivary glands. Clinical Rheumatology. 2017;36(1):83-88
  156. 156. Strevens Bolmgren V, Olsson P, Wollmer P, Hesselstrand R, Mandl T. Respiratory symptoms are poor predictors of concomitant chronic obstructive pulmonary disease in patients with primary Sjogren’s syndrome. Rheumatology International. 2017;37(5):813-818
  157. 157. Taouli B, Brauner MW, Mourey I, Lemouchi D, Grenier PA. Thin-section chest CT findings of primary Sjogren’s syndrome: Correlation with pulmonary function. European Radiology. 2002;12(6):1504-1511
  158. 158. Ter Borg EJ, Kelder JC. Development of new extra-glandular manifestations or associated auto-immune diseases after establishing the diagnosis of primary Sjogren’s syndrome: A long-term study of the Antonius Nieuwegein Sjogren (ANS) cohort. Rheumatology International. 2017;37(7):1153-1158
  159. 159. Liebow AA, Carrington CB. Diffuse pulmonary lymphoreticular infiltrations associated with dysproteinemia. The Medical Clinics of North America. 1973;57(3):809-843
  160. 160. Lopez Velazquez M, Highland KB. Pulmonary manifestations of systemic lupus erythematosus and Sjogren’s syndrome. Current Opinion in Rheumatology. 2018;30(5):449-464
  161. 161. Zhang T, Yuan F, Xu L, Sun W, Liu L, Xue J. Characteristics of patients with primary Sjogren’s syndrome associated interstitial lung disease and relevant features of disease progression. Clinical Rheumatology. 2020;39(5):1561-1568
  162. 162. Reina D, Roig Vilaseca D, Torrente-Segarra V, Cerda D, Castellvi I, Diaz Torne C, et al. Sjogren’s syndrome-associated interstitial lung disease: A multicenter study. Reumatologia Clinica. 2016;12(4):201-205
  163. 163. Fischer A, Swigris JJ, du Bois RM, Groshong SD, Cool CD, Sahin H, et al. Minor salivary gland biopsy to detect primary Sjogren syndrome in patients with interstitial lung disease. Chest. 2009;136(4):1072-1078
  164. 164. Aerni MR, Vassallo R, Myers JL, Lindell RM, Ryu JH. Follicular bronchiolitis in surgical lung biopsies: Clinical implications in 12 patients. Respiratory Medicine. 2008;102(2):307-312
  165. 165. Desai SR, Veeraraghavan S, Hansell DM, Nikolakopolou A, Goh NS, Nicholson AG, et al. CT features of lung disease in patients with systemic sclerosis: Comparison with idiopathic pulmonary fibrosis and nonspecific interstitial pneumonia. Radiology. 2004;232(2):560-567
  166. 166. Gupta N, Wikenheiser-Brokamp KA, Fischer A, McCormack FX. Diffuse cystic lung disease as the presenting manifestation of Sjogren syndrome. Annals of the American Thoracic Society. 2016;13(3):371-375
  167. 167. Gupta N, Vassallo R, Wikenheiser-Brokamp KA, McCormack FX. Diffuse cystic lung disease. Part I. American Journal of Respiratory and Critical Care Medicine. 2015;191(12):1354-1366
  168. 168. Elicker B, Pereira CA, Webb R, Leslie KO. High-resolution computed tomography patterns of diffuse interstitial lung disease with clinical and pathological correlation. Jornal Brasileiro de Pneumologia: Publicacao Oficial da Sociedade Brasileira de Pneumologia e Tisilogia. 2008;34(9):715-744
  169. 169. Arcadu A, Moua T, Yi ES, Ryu JH. Lymphoid interstitial pneumonia and other benign lymphoid disorders. Seminars in Respiratory and Critical Care Medicine. 2016;37(3):406-420
  170. 170. Tashiro K, Ohshima K, Suzumiya J, Yoneda S, Yahiro M, Sugihara M, et al. Clonality of primary pulmonary lymphoproliferative disorders; using in situ hybridization and polymerase chain reaction for immunoglobulin. Leukemia & Lymphoma. 1999;36(1-2):157-167
  171. 171. Guinee DG Jr. Update on nonneoplastic pulmonary lymphoproliferative disorders and related entities. Archives of Pathology & Laboratory Medicine. 2010;134(5):691-701
  172. 172. Katzenstein AL, Doxtader E, Narendra S. Lymphomatoid granulomatosis: Insights gained over 4 decades. The American Journal of Surgical Pathology. 2010;34(12):e35-e48
  173. 173. Swigris JJ, Berry GJ, Raffin TA, Kuschner WG. Lymphoid interstitial pneumonia: A narrative review. Chest. 2002;122(6):2150-2164
  174. 174. Nicholson AG. Lymphocytic interstitial pneumonia and other lymphoproliferative disorders in the lung. Seminars in Respiratory and Critical Care Medicine. 2001;22(4):409-422
  175. 175. American Thoracic S, European Respiratory S. American Thoracic Society/European Respiratory Society International Multidisciplinary Consensus Classification of the Idiopathic Interstitial Pneumonias. This joint statement of the American Thoracic Society (ATS), and the European Respiratory Society (ERS) was adopted by the ATS board of directors, June 2001 and by the ERS Executive Committee, June 2001. American Journal of Respiratory and Critical Care Medicine. 2002;165(2):277-304
  176. 176. Nicholson AG. Classification of idiopathic interstitial pneumonias: Making sense of the alphabet soup. Histopathology. 2002;41(5):381-391
  177. 177. Baque-Juston M, Pellegrin A, Leroy S, Marquette CH, Padovani B. Organizing pneumonia: What is it? A conceptual approach and pictorial review. Diagnostic and Interventional Imaging. 2014;95(9):771-777
  178. 178. Bouchardy LM, Kuhlman JE, Ball WC Jr, Hruban RH, Askin FB, Siegelman SS. CT findings in bronchiolitis obliterans organizing pneumonia (BOOP) with radiographic, clinical, and histologic correlation. Journal of Computer Assisted Tomography. 1993;17(3):352-357
  179. 179. Lazor R, Vandevenne A, Pelletier A, Leclerc P, Court-Fortune I, Cordier JF. Cryptogenic organizing pneumonia. Characteristics of relapses in a series of 48 patients. The Groupe d’Etudes et de Recherche sur les Maladles “Orphelines” Pulmonaires (GERM”O”P). American Journal of Respiratory and Critical Care Medicine. 2000;162(2 Pt 1):571-577
  180. 180. Cordier JF, Cottin V, Lazor R, Thivolet-Bejui F. Many faces of bronchiolitis and organizing pneumonia. Seminars in Respiratory and Critical Care Medicine. 2016;37(3):421-440
  181. 181. Matteson EL, Ike RW. Bronchiolitis obliterans organizing pneumonia and Sjogren’s syndrome. The Journal of Rheumatology. 1990;17(5):676-679
  182. 182. Vourlekis JS, Brown KK, Cool CD, Young DA, Cherniack RM, King TE, et al. Acute interstitial pneumonitis. Case series and review of the literature. Medicine (Baltimore). 2000;79(6):369-378
  183. 183. Yoo JW, Song JW, Jang SJ, Lee CK, Kim MY, Lee HK, et al. Comparison between cryptogenic organizing pneumonia and connective tissue disease-related organizing pneumonia. Rheumatology (Oxford, England). 2011;50(5):932-938
  184. 184. Takahashi H, Tsuboi H, Yokosawa M, Asashima H, Hirota T, Kondo Y, et al. Diffusion-weighted magnetic resonance imaging of parotid glands before and after abatacept therapy in patients with Sjogren’s syndrome associated with rheumatoid arthritis: Utility to evaluate and predict response to treatment. Modern Rheumatology. 2018;28(2):300-307
  185. 185. Lazarus MN, Robinson D, Mak V, Moller H, Isenberg DA. Incidence of cancer in a cohort of patients with primary Sjogren’s syndrome. Rheumatology (Oxford, England). 2006;45(8):1012-1015
  186. 186. Kovacs L, Szodoray P, Kiss E. Secondary tumours in Sjogren’s syndrome. Autoimmunity Reviews. 2010;9(4):203-206
  187. 187. Papageorgiou A, Voulgarelis M, Tzioufas AG. Clinical picture, outcome and predictive factors of lymphoma in Sjgren syndrome. Autoimmunity Reviews. 2015;14(7):641-649
  188. 188. Ramos-Casals M, De Vita S, Tzioufas AG. Hepatitis C virus, Sjogren’s syndrome and B-cell lymphoma: Linking infection, autoimmunity and cancer. Autoimmunity Reviews. 2005;4(1):8-15
  189. 189. Hansen LA, Prakash UB, Colby TV. Pulmonary lymphoma in Sjogren’s syndrome. Mayo Clinic Proceedings. 1989;64(8):920-931
  190. 190. Graham BB, Mathisen DJ, Mark EJ, Takvorian RW. Primary pulmonary lymphoma. The Annals of Thoracic Surgery. 2005;80(4):1248-1253
  191. 191. Yachoui R, Leon C, Sitwala K, Kreidy M. Pulmonary MALT lymphoma in patients with Sjogren’s syndrome. Clinical Medicine & Research. 2017;15(1-2):6-12
  192. 192. Milani P, Basset M, Russo F, Foli A, Palladini G, Merlini G. The lung in amyloidosis. European Respiratory Review: An Official Journal of the European Respiratory Society. 2017;26(145):170046-170054
  193. 193. Baqir M, Kluka EM, Aubry MC, Hartman TE, Yi ES, Bauer PR, et al. Amyloid-associated cystic lung disease in primary Sjogren’s syndrome. Respiratory Medicine. 2013;107(4):616-621
  194. 194. Simsek E, Caliskan A, Tutun U, Sahin S. Cause of a rare acute renal insufficiency: Rupture aortocaval fistula. Vascular. 2014;22(4):290-292
  195. 195. Rajagopala S, Singh N, Gupta K, Gupta D. Pulmonary amyloidosis in Sjogren’s syndrome: A case report and systematic review of the literature. Respirology. 2010;15(5):860-866
  196. 196. Wong BC, Wong KL, Ip MS, Wang EP, Chan KW, Cheng LC. Sjogren’s syndrome with amyloid a presenting as multiple pulmonary nodules. The Journal of Rheumatology. 1994;21(1):165-167
  197. 197. Nakamura N, Yamada G, Itoh T, Suzuki A, Morita-Ichimura S, Teramoto S, et al. Pulmonary MALT lymphoma with amyloid production in a patient with primary Sjogren’s syndrome. Internal medicine. 2002;41(4):309-311
  198. 198. Polansky SM, Ravin CE. Nodular pulmonary infiltrate in a patient with Sjogren’s syndrome. Chest. 1980;77(3):411-412
  199. 199. Carbone R, Cosso C, Cimmino MA. Pulmonary nodular amyloidosis in Sjogren syndrome. The Journal of Rheumatology. 2015;42(1):134
  200. 200. Kobayashi H, Matsuoka R, Kitamura S, Tsunoda N, Saito K. Sjogren’s syndrome with multiple bullae and pulmonary nodular amyloidosis. Chest. 1988;94(2):438-440
  201. 201. Rodrigues K, Neves FS, Stoeterau KB, Werner Castro GR, Nobre LF, Zimmermann AF, et al. Pulmonary amyloidosis in Sjogren’s syndrome: A rare diagnosis for nodular lung lesions. International Journal of Rheumatic Diseases. 2009;12(4):358-360
  202. 202. Kluka EM, Bauer PR, Aubry MC, Ryu JH. Enlarging lung nodules and cysts in a 53-year-old woman with primary Sjogren syndrome. Chest. 2013;143(1):258-261
  203. 203. Seguchi T, Kyoraku Y, Saita K, Ihi T, Nagai M, Akiyama Y, et al. Human T-cell lymphotropic virus type I (HTLV-1) associated myelopathy and Sjogren’s syndrome representing pulmonary nodular amyloidosis and multiple bullae: Report of an autopsy case. Virchows Archiv. 2006;448(6):874-876
  204. 204. Jing ZC, Xu XQ, Han ZY, Wu Y, Deng KW, Wang H, et al. Registry and survival study in chinese patients with idiopathic and familial pulmonary arterial hypertension. Chest. 2007;132(2):373-379
  205. 205. Teruuchi S, Bando M, Hironaka M, Ohno S, Sugiyama Y. Sjogren’s syndrome with multiple bullae and pulmonary nodular amyloidosis. Nihon Kokyūki Gakkai Zasshi. 2000;38(12):918-922
  206. 206. Liu Z, Yang X, Tian Z, Qian J, Wang Q, Zhao J, et al. The prognosis of pulmonary arterial hypertension associated with primary Sjogren’s syndrome: A cohort study. Lupus. 2018;27(7):1072-1080
  207. 207. Zhao Y, Wang H, Chen M, Zhang N, Yang ZW, Li D, et al. Primary Sjogren’s syndrome associated pulmonary arterial hypertension: 20 new cases. Zhonghua Yi Xue Za Zhi. 2019;99(37):2921-2925
  208. 208. Zuily S, Domingues V, Suty-Selton C, Eschwege V, Bertoletti L, Chaouat A, et al. Antiphospholipid antibodies can identify lupus patients at risk of pulmonary hypertension: A systematic review and meta-analysis. Autoimmunity Reviews. 2017;16(6):576-586
  209. 209. Fox PC, Datiles M, Atkinson JC, Macynski AA, Scott J, Fletcher D, et al. Prednisone and piroxicam for treatment of primary Sjogren’s syndrome. Clinical and Experimental Rheumatology. 1993;11(2):149-156
  210. 210. Fauci AS. Corticosteroids in autoimmune disease. Hospital Practice (Office ed.). 1983;18(10):99-103, 7-18, 13-4
  211. 211. Fox RI, Chan E, Benton L, Fong S, Friedlaender M, Howell FV. Treatment of primary Sjogren’s syndrome with hydroxychloroquine. The American Journal of Medicine. 1988;85(4A):62-67
  212. 212. Kruize AA, Hene RJ, Kallenberg CG, van Bijsterveld OP, van der Heide A, Kater L, et al. Hydroxychloroquine treatment for primary Sjogren’s syndrome: A two year double blind crossover trial. Annals of the Rheumatic Diseases. 1993;52(5):360-364
  213. 213. Schnabel A, Reuter M, Gross WL. Intravenous pulse cyclophosphamide in the treatment of interstitial lung disease due to collagen vascular diseases. Arthritis and Rheumatism. 1998;41(7):1215-1220
  214. 214. Tsuzaka K, Akama H, Yamada H, Akizuki M, Tojo T, Homma M. Pulmonary pseudolymphoma presented with a mass lesion in a patient with primary Sjogren’s syndrome: Beneficial effect of intermittent intravenous cyclophosphamide. Scandinavian Journal of Rheumatology. 1993;22(2):90-93
  215. 215. Yum HK, Kim ES, Ok KS, Lee HK, Choi SJ. Lymphocytic interstitial pneumonitis associated with Epstein-Barr virus in systemic lupus erythematosus and Sjogren’s syndrome. Complete remission with corticosteriod and cyclophosphamide. The Korean Journal of Internal Medicine. 2002;17(3):198-203
  216. 216. Oldham JM, Lee C, Valenzi E, Witt LJ, Adegunsoye A, Hsu S, et al. Azathioprine response in patients with fibrotic connective tissue disease-associated interstitial lung disease. Respiratory Medicine. 2016;121:117-122
  217. 217. Saketkoo LA, Espinoza LR. Experience of mycophenolate mofetil in 10 patients with autoimmune-related interstitial lung disease demonstrates promising effects. The American Journal of the Medical Sciences. 2009;337(5):329-335
  218. 218. Swigris JJ, Olson AL, Fischer A, Lynch DA, Cosgrove GP, Frankel SK, et al. Mycophenolate mofetil is safe, well tolerated, and preserves lung function in patients with connective tissue disease-related interstitial lung disease. Chest. 2006;130(1):30-36
  219. 219. Klinowski G, Gozzi F, Trentacosti F, Andrisani D, Sebastiani M, Clini EM. Rituximab for the treatment of acute onset interstitial lung disease in primary Sjogren’s syndrome. Pulmonology. 2021;27(6):575-578
  220. 220. Benad M, Koschel D, Herrmann K, Wiefel K, Kleymann A, Aringer M. Effects of cyclophosphamide and rituximab in patients with connective tissue diseases with severe interstitial lung disease. Clinical and Experimental Rheumatology. 2021. Epub ahead of print
  221. 221. Chen MH, Chen CK, Chou HP, Chen MH, Tsai CY, Chang DM. Rituximab therapy in primary Sjogren’s syndrome with interstitial lung disease: A retrospective cohort study. Clinical and Experimental Rheumatology. 2016;34(6):1077-1084
  222. 222. Watanabe Y, Koyama S, Miwa C, Okuda S, Kanai Y, Tetsuka K, et al. Pulmonary mucosa-associated lymphoid tissue (MALT) lymphoma in Sjogren’s syndrome showing only the LIP pattern radiologically. Internal Medicine. 2012;51(5):491-495
  223. 223. Wise LM, Arkfeld DG. A patient with primary Sjogren’s syndrome, cystic lung disease, and MALT lymphoma treated successfully with rituximab: A case-based review. Clinical Rheumatology. 2020;39(4):1357-1362
  224. 224. Blanco Perez JJ, Perez Gonzalez A, Guerra Vales JL, Melero Gonzalez R, Pego Reigosa JM. Shrinking lung in primary Sjogren syndrome successfully treated with rituximab. Archivos de Bronconeumología. 2015;51(9):475-476
  225. 225. Justet A, Ottaviani S, Dieude P, Taille C. Tocilizumab for refractory organising pneumonia associated with Sjogren’s disease. BML Case Reports. 2015;2015
  226. 226. Thompson G, McLean-Tooke A, Wrobel J, Lavender M, Lucas M. Sjogren syndrome with associated lymphocytic interstitial pneumonia successfully treated with tacrolimus and Abatacept as an alternative to rituximab. Chest. 2018;153(3):e41-ee3
  227. 227. Lee AS, Scofield RH, Hammitt KM, Gupta N, Thomas DE, Moua T, et al. Consensus guidelines for evaluation and management of pulmonary disease in Sjögren’s. Chest. 2021;159(2):683-698
  228. 228. Plemons JM, Al-Hashimi I, Marek CL. Managing xerostomia and salivary gland hypofunction: Executive summary of a report from the American Dental Association Council on Scientific Affairs. Journal of the American Dental Association (1939). 2014;145(8):867-873
  229. 229. Furness S, Worthington HV, Bryan G, Birchenough S, McMillan R. Interventions for the management of dry mouth: Topical therapies. The Cochrane Database of Systematic Reviews. 2011;(12):Cd008934, 1-93
  230. 230. Salum FG, Medella-Junior FAC, Figueiredo MAZ, Cherubini K. Salivary hypofunction: An update on therapeutic strategies. Gerodontology. 2018;35(4):305-316
  231. 231. Tan ECK, Lexomboon D, Sandborgh-Englund G, Haasum Y, Johnell K. Medications that cause dry mouth as an adverse effect in older people: A systematic review and Metaanalysis. Journal of the American Geriatrics Society. 2018;66(1):76-84
  232. 232. Papas AS, Sherrer YS, Charney M, Golden HE, Medsger TA Jr, Walsh BT, et al. Successful treatment of dry mouth and dry eye symptoms in Sjögren’s syndrome patients with oral pilocarpine: A randomized, placebo-controlled, dose-adjustment study. Journal of Clinical Rheumatology: Practical Reports on Rheumatic & Musculoskeletal Diseases. 2004;10(4):169-177
  233. 233. Wu CH, Hsieh SC, Lee KL, Li KJ, Lu MC, Yu CL. Pilocarpine hydrochloride for the treatment of xerostomia in patients with Sjögren’s syndrome in Taiwan—A double-blind, placebo-controlled trial. Journal of the Formosan Medical Association. 2006;105(10):796-803
  234. 234. Tsifetaki N, Kitsos G, Paschides CA, Alamanos Y, Eftaxias V, Voulgari PV, et al. Oral pilocarpine for the treatment of ocular symptoms in patients with Sjögren’s syndrome: A randomised 12 week controlled study. Annals of the Rheumatic Diseases. 2003;62(12):1204-1207
  235. 235. Rao RS, Akula R, Satyanarayana TSV, Indugu V. Recent advances of pacemakers in treatment of xerostomia: A systematic review. Journal of International Society of Preventive and Community Dentistry. 2019;9(4):311-315
  236. 236. Ami S, Wolff A. Implant-supported electrostimulating device to treat xerostomia: A preliminary study. Clinical Implant Dentistry and Related Research. 2010;12(1):62-71
  237. 237. Holland EJ, Whitley WO, Sall K, Lane SS, Raychaudhuri A, Zhang SY, et al. Lifitegrast clinical efficacy for treatment of signs and symptoms of dry eye disease across three randomized controlled trials. Current Medical Research and Opinion. 2016;32(10):1759-1765
  238. 238. Foulks GN. Treatment of dry eye disease by the non-ophthalmologist. Rheumatic Diseases Clinics of North America. 2008;34(4):987-1000 x
  239. 239. Sall K, Stevenson OD, Mundorf TK, Reis BL. Two multicenter, randomized studies of the efficacy and safety of cyclosporine ophthalmic emulsion in moderate to severe dry eye disease. CsA phase 3 study group. Ophthalmology. 2000;107(4):631-639
  240. 240. Kim EC, Choi JS, Joo CK. A comparison of vitamin a and cyclosporine a 0.05% eye drops for treatment of dry eye syndrome. American Journal of Ophthalmology. 2009;147(2):206-13.e3
  241. 241. Belafsky PC, Postma GN. The laryngeal and esophageal manifestations of Sjögren’s syndrome. Current Rheumatology Reports. 2003;5(4):297-303
  242. 242. Fischer A, Brown KK, Du Bois RM, Frankel SK, Cosgrove GP, Fernandez-Perez ER, et al. Mycophenolate mofetil improves lung function in connective tissue disease-associated interstitial lung disease. The Journal of Rheumatology. 2013;40(5):640-646
  243. 243. Amlani B, Elsayed G, Barvalia U, Kanne JP, Meyer KC, Sandbo N, et al. Treatment of primary sjogren’s syndrome-related interstitial lung disease: A retrospective cohort study. Sarcoidosis, Vasculitis, and Diffuse Lung Diseases: Official Journal of WASOG. 2020;37(2):136-147
  244. 244. Isaksen K, Jonsson R, Omdal R. Anti-CD20 treatment in primary Sjögren’s syndrome. Scandinavian Journal of Immunology. 2008;68(6):554-564
  245. 245. Seror R, Sordet C, Guillevin L, Hachulla E, Masson C, Ittah M, et al. Tolerance and efficacy of rituximab and changes in serum B cell biomarkers in patients with systemic complications of primary Sjögren’s syndrome. Annals of the Rheumatic Diseases. 2007;66(3):351-357
  246. 246. Daoussis D, Melissaropoulos K, Sakellaropoulos G, Antonopoulos I, Markatseli TE, Simopoulou T, et al. A multicenter, open-label, comparative study of B-cell depletion therapy with rituximab for systemic sclerosis-associated interstitial lung disease. Seminars in Arthritis and Rheumatism. 2017;46(5):625-631
  247. 247. Narvaez J, Luch JL, Molina-Molina M, Vicens-Zygmunt V, Luburich P, Yanez MA, et al. Rituximab as a rescue treatment added on mycophenolate mofetil background therapy in progressive systemic sclerosis associated interstitial lung disease unresponsive to conventional immunosuppression. Seminars in Arthritis and Rheumatism. 2020;50(5):977-987
  248. 248. Rimar D, Rosner I, Slobodin G. Upfront combination therapy with rituximab and mycophenolate Mofetil for progressive systemic sclerosis. The Journal of Rheumatology. 2021;48(2):304-305
  249. 249. Nadashkevich O, Davis P, Fritzler M, Kovalenko W. A randomized unblinded trial of cyclophosphamide versus azathioprine in the treatment of systemic sclerosis. Clinical Rheumatology. 2006;25(2):205-212
  250. 250. Tashkin DP, Elashoff R, Clements PJ, Goldin J, Roth MD, Furst DE, et al. Cyclophosphamide versus placebo in scleroderma lung disease. The New England Journal of Medicine. 2006;354(25):2655-2666
  251. 251. Iudici M, Cuomo G, Vettori S, Bocchino M, Sanduzzi Zamparelli A, Cappabianca S, et al. Low-dose pulse cyclophosphamide in interstitial lung disease associated with systemic sclerosis (SSc-ILD): Efficacy of maintenance immunosuppression in responders and non-responders. Seminars in Arthritis and Rheumatism. 2015;44(4):437-444
  252. 252. Tashkin DP, Roth MD, Clements PJ, Furst DE, Khanna D, Kleerup EC, et al. Mycophenolate mofetil versus oral cyclophosphamide in scleroderma-related interstitial lung disease (SLS II): A randomised controlled, double-blind, parallel group trial. The Lancet Respiratory Medicine. 2016;4(9):708-719
  253. 253. Khanna D, Denton CP, Jahreis A, van Laar JM, Frech TM, Anderson ME, et al. Safety and efficacy of subcutaneous tocilizumab in adults with systemic sclerosis (faSScinate): A phase 2, randomised, controlled trial. Lancet. 2016;387(10038):2630-2640
  254. 254. Khanna D, Denton CP, Lin CJF, van Laar JM, Frech TM, Anderson ME, et al. Safety and efficacy of subcutaneous tocilizumab in systemic sclerosis: Results from the open-label period of a phase II randomised controlled trial (faSScinate). Annals of the Rheumatic Diseases. 2018;77(2):212-220
  255. 255. Felten R, Meyer N, Duffaut P, Saadoun D, Hachulla E, Hatron P, et al. IL-6 receptor inhibition in primary sjögren syndrome: Results from a randomized multicenter academic double blind placebo-controlled trial of tocilizumab in 110 patients [abstract]. Arthritis and Rheumatism. 2019;71. Available from: https://acrabstracts.org/abstract/il-6-receptor-inhibition-in-primary-sjogren-syndrome-results-from-a-randomized-multicenter-academic-double-blind-placebo-controlled-trial-of-tocilizumab-in-110-patients/
  256. 256. Sankar V, Brennan MT, Kok MR, Leakan RA, Smith JA, Manny J, et al. Etanercept in Sjogren’s syndrome: A twelve-week randomized, double-blind, placebo-controlled pilot clinical trial. Arthritis and Rheumatism. 2004;50(7):2240-2245
  257. 257. Moutsopoulos NM, Katsifis GE, Angelov N, Leakan RA, Sankar V, Pillemer S, et al. Lack of efficacy of etanercept in Sjogren syndrome correlates with failed suppression of tumour necrosis factor alpha and systemic immune activation. Annals of the Rheumatic Diseases. 2008;67(10):1437-1443
  258. 258. Herrero-Beaumont G, Martinez Calatrava MJ, Castaneda S. Abatacept mechanism of action: Concordance with its clinical profile. Reumatologia Clinica. 2012;8(2):78-83
  259. 259. Nakashita T, Ando K, Takahashi K, Motojima S. Possible effect of abatacept on the progression of interstitial lung disease in rheumatoid arthritis patients. Respiratory Investigation. 2016;54(5):376-379
  260. 260. Mochizuki T, Yano K, Ikari K, Hiroshima R, Takaoka H, Kawakami K, et al. The efficacy of abatacept in Japanese patients with rheumatoid arthritis: 104 weeks radiographic and clinical results in clinical practice. Modern Rheumatology. 2016;26(4):499-506
  261. 261. Kurata I, Tsuboi H, Terasaki M, Shimizu M, Toko H, Honda F, et al. Effect of biological disease-modifying anti-rheumatic drugs on airway and interstitial lung disease in patients with rheumatoid arthritis. Internal Medicine. 2019;58(12):1703-1712
  262. 262. Kang EH, Jin Y, Desai RJ, Liu J, Sparks JA, Kim SC. Risk of exacerbation of pulmonary comorbidities in patients with rheumatoid arthritis after initiation of abatacept versus TNF inhibitors: A cohort study. Seminars in Arthritis and Rheumatism. 2020;50(3):401-408
  263. 263. Cassone G, Manfredi A, Atzeni F, Venerito V, Vacchi C, Picerno V, et al. Safety of Abatacept in Italian patients with rheumatoid arthritis and interstitial lung disease: A Multicenter retrospective study. Journal of Clinical Medicine. 2020;9(1):277-287
  264. 264. Mena-Vazquez N, Godoy-Navarrete FJ, Manrique-Arija S, Aguilar-Hurtado MC, Romero-Barco CM, Urena-Garnica I, et al. Non-anti-TNF biologic agents are associated with slower worsening of interstitial lung disease secondary to rheumatoid arthritis. Clinical Rheumatology. 2021;40(1):133-142
  265. 265. Fernandez-Diaz C, Castaneda S, Melero-Gonzalez RB, Ortiz-Sanjuan F, Juan-Mas A, Carrasco-Cubero C, et al. Abatacept in interstitial lung disease associated with rheumatoid arthritis: National multicenter study of 263 patients. Rheumatology (Oxford, England). 2020;59(12):3906-3916
  266. 266. Ferandez-Díaz C, Castaneda S, Melero R, Loricera J, Ortiz-Sanju’an F, Juan-Mas A, et al. Poster #SAT0035. Response to abatacept of different patterns of interstitial lung disease in rheumatoid arthritis: National multicenter study of 263 patients. Annals of the Rheumatic Diseases. 2020;79(Suppl. 1):943-944. DOI: 10.1136/annrheumdis-2020-eular.1741
  267. 267. Mori S. Management of rheumatoid arthritis patients with interstitial lung disease: Safety of biological antirheumatic drugs and assessment of pulmonary fibrosis. Clinical Medicine Insights: Circulatory, Respiratory and Pulmonary Medicine. 2015;9(Suppl 1):41-49
  268. 268. Tsuboi H, Matsumoto I, Hagiwara S, Hirota T, Takahashi H, Ebe H, et al. Effectiveness of abatacept for patients with Sjogren’s syndrome associated with rheumatoid arthritis. An open label, multicenter, one-year, prospective study: ROSE (Rheumatoid Arthritis with Orencia Trial toward Sjogren’s syndrome Endocrinopathy) trial. Modern Rheumatology. 2016;26(6):891-899
  269. 269. Pontarini E, Fabris M, Quartuccio L, Cappeletti M, Calcaterra F, Roberto A, et al. Treatment with belimumab restores B cell subsets and their expression of B cell activating factor receptor in patients with primary Sjogren’s syndrome. Rheumatology (Oxford, England). 2015;54(8):1429-1434
  270. 270. Mariette X, Seror R, Quartuccio L, Baron G, Salvin S, Fabris M, et al. Efficacy and safety of belimumab in primary Sjogren’s syndrome: Results of the BELISS open-label phase II study. Annals of the Rheumatic Diseases. 2015;74(3):526-531
  271. 271. Gandolfo S, De Vita S. Double anti-B cell and anti-BAFF targeting for the treatment of primary Sjogren’s syndrome. Clinical and Experimental Rheumatology. 2019;118(3):199-208
  272. 272. Hall JC, Baer AN, Shah AA, Criswell LA, Shiboski CH, Rosen A, et al. Molecular subsetting of interferon pathways in Sjogren’s syndrome. Arthritis & Rheumatology. 2015;67(9):2437-2446
  273. 273. Nezos A, Gravani F, Tassidou A, Kapsogeorgou EK, Voulgarelis M, Koutsilieris M, et al. Type I and II interferon signatures in Sjogren’s syndrome pathogenesis: Contributions in distinct clinical phenotypes and Sjogren’s related lymphomagenesis. Journal of Autoimmunity. 2015;63:47-58
  274. 274. Furie R, Khamashta M, Merrill JT, Werth VP, Kalunian K, Brohawn P, et al. Anifrolumab, an anti-interferon-alpha receptor monoclonal antibody, in moderate-to-severe systemic lupus erythematosus. Arthritis Rheumatology. 2017;69(2):376-386
  275. 275. Wells AU, Brown KK, Flaherty KR, Kolb M, Thannickal VJ, Group IPFCW. What’s in a name? That which we call IPF, by any other name would act the same. The European Respiratory Journal. 2018;51(5):1800692-1800704
  276. 276. Wollin L, Wex E, Pautsch A, Schnapp G, Hostettler KE, Stowasser S, et al. Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. The European Respiratory Journal. 2015;45(5):1434-1445
  277. 277. Hilberg F, Roth GJ, Krssak M, Kautschitsch S, Sommergruber W, Tontsch-Grunt U, et al. BIBF 1120: Triple angiokinase inhibitor with sustained receptor blockade and good antitumor efficacy. Cancer Research. 2008;68(12):4774-4782
  278. 278. Gurujeyalakshmi G, Hollinger MA, Giri SN. Pirfenidone inhibits PDGF isoforms in bleomycin hamster model of lung fibrosis at the translational level. The American Journal of Physiology. 1999;276(2):L311-L318
  279. 279. Corbel M, Lanchou J, Germain N, Malledant Y, Boichot E, Lagente V. Modulation of airway remodeling-associated mediators by the antifibrotic compound, pirfenidone, and the matrix metalloproteinase inhibitor, batimastat, during acute lung injury in mice. European Journal of Pharmacology. 2001;426(1-2):113-121
  280. 280. Ruwanpura SM, Thomas BJ, Bardin PG. Pirfenidone: Molecular mechanisms and potential clinical applications in lung disease. American Journal of Respiratory Cell and Molecular Biology. 2020;62(4):413-422
  281. 281. Liu H, Drew P, Cheng Y, Visner GA. Pirfenidone inhibits inflammatory responses and ameliorates allograft injury in a rat lung transplant model. The Journal of Thoracic and Cardiovascular Surgery. 2005;130(3):852-858
  282. 282. Flaherty KR, Wells AU, Brown KK. Nintedanib in progressive fibrosing interstitial lung diseases. Reply. The New England Journal of Medicine. 2020;382(8):781
  283. 283. Wells AU, Flaherty KR, Brown KK, Inoue Y, Devaraj A, Richeldi L, et al. Nintedanib in patients with progressive fibrosing interstitial lung diseases-subgroup analyses by interstitial lung disease diagnosis in the INBUILD trial: A randomised, double-blind, placebo-controlled, parallel-group trial. The Lancet Respiratory Medicine. 2020;8(5):453-460
  284. 284. Distler O, Highland KB, Gahlemann M, Azuma A, Fischer A, Mayes MD, et al. Nintedanib for systemic sclerosis-associated interstitial lung disease. The New England Journal of Medicine. 2019;380(26):2518-2528
  285. 285. Khanna D, Albera C, Fischer A, Khalidi N, Raghu G, Chung L, et al. An open-label, phase II study of the safety and tolerability of Pirfenidone in patients with scleroderma-associated interstitial lung disease: The LOTUSS trial. The Journal of Rheumatology. 2016;43(9):1672-1679
  286. 286. Enomoto Y, Nakamura Y, Colby TV, Inui N, Suda T. Pirfenidone for primary Sjogren’s syndrome-related fibrotic interstitial pneumonia. Sarcoidosis, Vasculitis, and Diffuse Lung Diseases: Official Journal of WASOG. 2017;34(1):91-96
  287. 287. Fischer A, Distler J. Progressive fibrosing interstitial lung disease associated with systemic autoimmune diseases. Clinical Rheumatology. 2019;38(10):2673-2681

Written By

Moiz Ehtesham, Anupama Tiwari, Rose Sneha George and Ruben A. Peredo

Submitted: 25 October 2021 Reviewed: 21 January 2022 Published: 26 August 2022