Skip to main content
Erschienen in: BMC Pulmonary Medicine 1/2020

Open Access 01.12.2020 | Research article

Clinical characteristics and organ system involvement in sarcoidosis: comparison of the University of Minnesota Cohort with other cohorts

verfasst von: Hok Sreng Te, David M. Perlman, Chetan Shenoy, Daniel J. Steinberger, Rebecca J. Cogswell, Henri Roukoz, Erik J. Peterson, Lin Zhang, Tadashi L. Allen, Maneesh Bhargava

Erschienen in: BMC Pulmonary Medicine | Ausgabe 1/2020

Abstract

Background

Sarcoidosis is a systemic granulomatous disease of unknown etiology. Clinical cohort studies of different populations are important to understand the high variability in clinical presentation and disease course of sarcoidosis. The aim of the study is to evaluate clinical characteristics, including organ involvement, pulmonary function tests, and laboratory parameters, in a sarcoidosis cohort at the University of Minnesota. We compare the organ system involvement of this cohort with other available cohorts.

Methods

We conducted a retrospective data collection and analysis of 187 subjects with biopsy-proven sarcoidosis seen at a tertiary center. Organ system involvement was determined using the WASOG sarcoidosis organ assessment instrument. Clinical phenotype groups were classified using the Genomic Research in Alpha-1 Antitrypsin Deficiency and Sarcoidosis criteria.

Results

Mean subject age at diagnosis was 45.8 ± 12.4, with a higher proportion of males (55.1%), and a higher proportion of blacks (17.1%) compared to the racial distribution of Minnesota residents (5.95%). The majority (71.1%) of subjects required anti-inflammatory therapy for at least 1 month. Compared to the A Case Control Etiologic Study of Sarcoidosis cohort, there was a higher frequency of extra-thoracic lymph node (34.2% vs. 15.2%), eye (20.9% vs. 11.8%), liver (17.6% vs. 11.5%), spleen (20.9% vs. 6.7%), musculoskeletal (9.6% vs. 0.5%), and cardiac (10.7% vs. 2.3%) involvement in our cohort. A multisystem disease with at least five different organs involved was identified in 13.4% of subjects. A restrictive physiological pattern was observed in 21.6% of subjects, followed by an obstructive pattern in 17.3% and mixed obstructive and restrictive pattern in 2.2%. Almost half (49.2%) were Scadding stages II/III. Commonly employed disease activity markers, including soluble interleukin-2 receptor and angiotensin-converting enzyme, did not differ between treated and untreated groups.

Conclusions

This cohort features a relatively high frequency of high-risk sarcoidosis phenotypes including cardiac and multiorgan disease. Commonly-utilized serum biomarkers do not identify subpopulations that require or do better with treatment. Findings from this study further highlight the high-variability nature of sarcoidosis and the need for a more reliable biomarker to predict and measure disease severity and outcomes for better clinical management of sarcoidosis patients.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ACE
Angiotensin converting enzyme
ACCESS
A Case Control Etiologic Study of Sarcoidosis
ATS
American Thoracic Society
CBD
Chronic beryllium disease
CPET
Cardiac -positron emission tomography
CVID
Common variable immunoglobulin deficiency
DLCO
Diffusing capacity of the lungs for carbon monoxide
FDG
Fluorodeoxyglucose
FEV1
Forced expiratory volume in 1 s
FVC
Forced vital capacity
GRADS
Genomic Research in Alpha-1 Antitrypsin Deficiency and Sarcoidosis
HRS
Heart Rhythm Society
JMHW
Japanese Ministry of Health and Welfare
LGE
Late gadolinium enhancement
NASH
Nonalcoholic steatohepatitis
sIL-2R
Soluble interleukin 2 receptor
TLC
Total lung capacity
UCSF
University of California San Francisco
UMN
University of Minnesota
WASOG
World Association of Sarcoidosis and Other Granulomatous Disorders
WBC
White blood cell

Background

Sarcoidosis is a multi-system disease of unknown etiology characterized by the presence of non-caseating granulomas in involved organs [1]. Although lungs, mediastinal and hilar lymph nodes are most commonly involved, sarcoidosis is associated with heterogeneous manifestations, disease severity, and outcomes [14]. The incidence of sarcoidosis is at least 45–300/100,000 individuals in the US [5], of both sexes, and all ages and races [1, 68]. The annual mortality is 2.8/million people [3, 9] and is rising [10] with a more aggressive course and more extrapulmonary involvement reported in black patients [11].
Recently, four high-risk sarcoidosis manifestations were recognized as highly associated with greater morbidity and mortality [12]. These include treatment-resistant pulmonary disease, neuro-sarcoidosis, cardiac sarcoidosis, and multisystem organ involvement. In the US and Europe, respiratory failure is the most common cause of sarcoidosis-related mortality [9, 10]. In contrast, Japanese cohorts have a higher incidence of cardiac disease, and cardiac involvement is the most frequent case of death due to sarcoidosis in Japan [13]. Lofgren’s syndrome, an acute presentation of sarcoidosis associated with fever, erythema nodosum, arthralgias and mediastinal or hilar adenopathy, is more frequent in Scandinavian countries and is associated with a good prognosis [14]. The heterogeneity in sarcoidosis presents challenges in predicting disease course, optimizing treatments, and establishing a uniform standard for organ involvement assessment.
There is significant geographic variability in sarcoidosis manifestations. The variability in sarcoidosis manifestations could be explained by regional variability in sarcoidosis but also could be due to the tools employed for assessment of organ involvement. A number of reports have used the tool developed by the A Case Control Etiologic Study of Sarcoidosis (ACCESS) investigators [15] to assess disease manifestation [11, 16, 17]. This tool was further modified by the World Association of Sarcoidosis and Other Granulomatous Disorders (WASOG) task force to address additional manifestations and possible organs involved with sarcoidosis. The WASOG sarcoidosis organ assessment instrument classifies organ involvement as “highly probable”, “probable”, “possible”, or “no consensus” [18]. Various other assessment tools for sarcoidosis have been proposed. More recently, the Genomic Research in Alpha-1 Antitrypsin Deficiency and Sarcoidosis (GRADS) study classified clinical phenotypes into nine clinical phenotype groups [19], including efforts to define multiorgan, cardiac defining therapy, and other phenotypes.
Our objective is to describe the clinical characteristics of the sarcoidosis cases seen at the University of Minnesota, and to compare the characteristics of our cohort to the ACCESS and other cohorts. The racial distribution of Minnesota residents includes White (83.75%), Black or African American (5.95%), Asian (4.66%), two or more races (2.81%), other race (1.74%), Native American (1.05%), and Native Hawaiian or Pacific Islander (0.04%) [20]. A unique feature of the Minnesota population is the substantially higher percentage of residents of Scandinavian origin which may influence the disease manifestations. While only 0.2% of US population is of Scandinavian origin, up to 1.5% of Minnesota residents self-report a Scandinavian origin, and we presume that our cohort may have this representation [21].

Methods

This cohort includes 187 sarcoidosis subjects consented between March 2015 and May 2019 at a tertiary sarcoidosis referral center in Minnesota. The diagnosis of sarcoidosis was made per American Thoracic Society (ATS) statement [1], meeting the following criteria: 1) a compatible clinical and radiologic finding; 2) histological evidence of non-caseating granulomas; and 3) exclusion of other diseases such as infections, common variable immunoglobulin deficiency (CVID), chronic beryllium disease (CBD), and malignancy. Diagnosis dates were determined per biopsy dates and estimated to be June 15th when days and/or months were not available. Subjects without histological evidence were excluded from this cohort. Subjects with a history of malignancy diagnosed within two years prior to or after sarcoidosis were excluded to exclude the possibility of sarcoid-like reactions to malignancies.
We conducted a chart review to collect data on demographics, diagnostics, imaging, past medical history, treatment program, and pulmonary function tests. Racial demographics were collected per patient self-report through the electronic medical record. With limited data, Hispanic whites and non-Hispanic whites were both categorized as “White” in this study. Results from laboratory within one year prior to the initial visit were also collected, including angiotensin converting enzyme (ACE), white blood count (WBC) with differential, soluble interleukin-2 receptor (sIL-2R), C-reactive protein (CRP), immunoglobulin G (IgG), albumin, total protein, liver function tests, calcium, parathyroid hormone (PTH), 25-OH vitamin D, and 1,25-DiOH vitamin D. Scadding stage was established using the closest available imaging study - chest x-ray (CXR) and chest CT topogram if CXR was not available - by an expert independent chest radiologist. The treatment groups, organ involvement, and clinical phenotype were assessed based on the characteristics at enrollment and prior history.
In the absence of histological evidence, the organ system involvement was determined using the ‘highly probable“ and “probable” criteria per the WASOG sarcoidosis organ assessment instrument [18]. Per the WASOG instrument, thoracic involvement includes both pulmonary involvement and thoracic lymph node involvement. One modification we made from the instrument was that although arthralgia was listed under “possible” criteria, we included it for bone-joint involvement when it was associated with Lofgren’s syndrome. Additionally, small fiber neuropathy with positive skin biopsy findings was included for nervous system involvement. In addition to PET, subjects with abnormal signal intensity on magnetic resonance imaging (MRI) of bone marrow were also included for bone marrow involvement.
We established the clinical phenotype groups using the GRADS criteria [19] with minor modifications. Subjects with Lofgren’s syndrome but treated for > 3 months were nonetheless assigned to Acute Sarcoidosis (Group 7). Untreated or treated subjects who were off treatment for at least one year from the visit date were assigned to the Remitting Disease group (Group 8). Subjects with five or more involved organs including cardiac involvement and on systemic anti-inflammatory therapy without remission (not assigned to Group 8) were assigned to Cardiac Defining Therapy (Group 9) as opposed to Multiorgan Disease (Group 1). We identified unclassifiable cases that did not fit any of the GRADS phenotypes, including Stage 0 treated or untreated and Stage I treated.
We grouped the PFT abnormalities into obstructive, restrictive, or mixed obstructive and restrictive ventilatory defects. Obstructive ventilatory defect was defined using Global Initiative for Chronic Obstructive Lung Disease criteria as forced expiratory volume in 1 s (FEV1) / forced vital capacity (FVC) ratio (FEV1/FVC) < 70% and a reduction of FEV1 [22]. Restriction ventilatory defect was defined as FEV1/FVC ratio ≥ 70% and a reduction in FVC. A mixed obstructive and restrictive pattern was defined as FEV1/FVC ratio < 70%, a decrease in FEV1 and total lung capacity (TLC). To simplify the interpretation, the FEV1, FVC, TLC and diffusing capacity of the lungs for carbon monoxide (DLCO) were considered reduced if they were below 80% of predicted values.
Statistical analysis was conducted using R version 3.6.1. Categorical variables were analyzed using Chi-square test or Fisher’s exact test when expected values are less than five, while continuous variables were conducted using two-sample Student’s t-test and analysis of variance (ANOVA) when more than two groups were compared, with p < 0.05 considered statistically significant.

Results

Demographics

We included 187 consecutive sarcoidosis subjects with histological evidence of noncaseating granulomas and a clinical presentation consistent with sarcoidosis (Table 1). The majority of the subjects were white (70.6%) or African American (17.1%). The mean age at diagnosis was 45.8 ± 12.4 years, with no significant difference between males (44.7 ± 12.5 years) and females (47.1 ± 12.1 years), and whites (47.1 ± 12.6 years) and blacks (43.3 ± 10.9 years). More than half of the subjects were males (55.1%). The mean body mass index (BMI) was 31.2 ± 7.1 kg/m2, with no significant difference between males (31.4 ± 6.4 kg/m2) and females (31.0 ± 7.8 kg/m2), and Whites (31.6 ± 6.8 kg/m2) and Blacks (29.6 ± 6.4 kg/m2). The use of anti-inflammatory medications for at least one month for sarcoidosis was present in 71.1% of subjects. The mean age at diagnosis for the treated group (44.2 ± 12.0 years) who received immunosuppression for ≥1 month was lower compared to cases (50.0 ± 12.4 years) that were never treated or treated < 1 month. The treated group also had a higher mean BMI (32.0 ± 7.5 kg/m2 vs 29.4 ± 5.7 kg/m2, p = 0.01). Only a minority of the subjects (9%) reported a family history of sarcoidosis. Thirty eight percent of subjects were former cigarette smokers, and 8% were current smokers.
Table 1
Demographic characteristics of the cohort (n = 187)
Characteristics
mean ± SD or n (%)
Age at enrollment (years)
53.0 ± 12.6
Age at diagnosis (years)
45.8 ± 12.4
Sex
 Male
103 (55.1%)
 Female
84 (44.9%)
Race
 White
132 (70.6)
 African American
32 (17.1)
 Asian
3 (1.6)
 American Indian or Alaska Native
1 (0.5)
 Choose not to answer
19 (10.2)
Body mass index (kg/m2)
31.2 ± 7.1
Smoking status
 Never
101 (54.0)
 Former
71 (38.0)
 Current
15 (8.0)
Family history of sarcoidosis
16 (9.0)
Use of anti-inflammatory agents
 Treated ≥1 month
133 (71.1)
 Treated < 1 month or never
54 (28.9)
Pulmonary function tests
 FEV1 (% predicted)
80.0 ± 23.5
 FVC (% predicted)
85.2 ± 21.2
 FEV1:FVC ratio
74.4 ± 11.5
 TLC (% predicted)
92.7 ± 17.3
 DLCO (% predicted)
89.5 ± 26.0

Organ involvement

Organ involvement was assessed using the WASOG sarcoidosis organ assessment instrument [18]. Thoracic involvement was detected in 184 (98.4%) subjects by histology or imaging studies with chest x-ray (CXR) and/or CT scan, with the majority of the cases (85.9%) having histological evidence of noncaseating granulomas (Table 2). Extra-thoracic involvement was observed in 128 (68.4%) subjects, 59 (31.6%) subjects had thoracic involvement only, and 3 (1.6%) subjects had extra-thoracic involvement only. Bone and joint involvement were more frequent for whites (8.2% vs 0% for blacks, p = 0.04). Otherwise, there was no statistically significant association between each of the organ involvement and sex, race, or treatment group.
Table 2
Organ involvement and histological evidence indicating subjects with evidence of noncaseating granulomas
Characteristics
Organ Involvement per WASOG Tool
Histological Evidence
n/187
%
95% CI
n/total
%
Lung
184
98.4
95.4–99.7
158/184
85.9
Skin
27
14.4
9.7–20.3
22/27
81.5
Liver
33
17.6
12.5–23.9
12/33
36.4
Eye
39
20.9
15.3–27.4
5/39
12.8
Spleen
39
20.9
15.3–27.4
2/39
5.1
Salivary Gland
4
2.1
0.6–5.4
1/4
25.0
ENT
4
2.1
0.6–5.4
3/4
75.0
Calcium-VitD
10
5.3
2.6–9.6
0/10a
0.0
Bone-Joint
18b
9.6
5.8–14.8
3/18
16.7
Bone Marrow
6
3.2
1.2–6.9
4/6
66.7
Muscle
2
1.1
0.1–3.8
1/2
50.0
Extra-Thoracic Lymph Node
64
34.2
27.5–41.5
18/64
28.1
Kidney
2
1.1
0.1–3.8
2/2
100.0
Nervous System
14
7.5
4.1–12.2
1/14c
7.1
Cardiac
20
10.7
6.7–16.0
3/20
15.0
Other Organs
9
4.8
2.2–8.9
6/9
66.7
 Testis
3
1.6
0.3–4.6
2/3
66.7
 Large intestine
1
0.5
0.0–2.9
1/1
100.0
 Stomach
2
1.1
0.1–3.8
2/2
100.0
 Thyroid
2
1.1
0.1–3.8
0/2
0.0
 Appendix
1
0.5
0.0–2.9
1/1
100.0
a10 subjects with evidence of calcium stones by stone analysis for calcium-vitD involvement
b4 subjects with arthralgia associated with Lofgren’s syndrome was included for bone-joint involvement
c3 subjects with evidence of small fiber neuropathy by skin biopsy for nervous system involvement
Other organs that were commonly involved included extra-thoracic lymph nodes (34.2%), eye (20.9%), and spleen (20.9%). Extra-thoracic adenopathy was detected by imaging in at least two sites, including cervical, supraclavicular, axillary, paraoesophageal, mesenteric, retroperitoneal, portocaval, hepatogastric, paraaortic, porta hepatis, inguinal, or iliac chain node stations; 28.1% of those with adenopathy underwent biopsy that provided evidence of granulomatous lymphadenitis. Among those with ocular disease, uveitis (61.5%) and lacrimal gland enlargement (18.0%) were the primary manifestations. In cases with splenic involvement, splenomegaly was present in 12 (30.8%), and splenic lesions were detected in 28 (71.8%) by imaging studies. In five subjects, splenomegaly was secondary to portal hypertension from non-alcoholic steatohepatitis (NASH), and these cases were not considered to have sarcoidosis splenic involvement as there was an alternative explanation for the splenomegaly. The majority of subjects (67.9%) had more than one organ involved (Table 3). Multiorgan involvement (at least five different organs involved) was identified in 13.4% of subjects. The mean number of organs involved was higher in treated vs. untreated cases in our cohort (2.7 vs 2.1, p = 0.004). Otherwise, there was no significant difference in the mean number of organs involved across gender and racial groups.
Table 3
Number of organs involved in each subject
Number of organs
Cases %
n
%
1
60
32.1
2
50
26.7
3
35
18.7
4
17
9.1
5
14
7.5
6
7
3.7
7
2
1.1
8
2
1.1
High-risk sarcoidosis with cardiac or neurologic involvement was observed in 20 (10.7%) and 14 (7.5%) subjects, respectively. Among subjects with cardiac involvement, three had endomyocardial biopsies demonstrating granulomatous inflammation; eight had a history of ventricular tachycardia and/or high-degree atrioventricular (AV) block such as Morbitz type II or complete heart block found in five subjects; ten had left ventricular ejection fraction below 50%. Nine patients received automatic implantable cardioverter defibrillators and two cases had pacemaker implantation. Advanced cardiac imaging was abnormal with suggestion of myocardial involvement in 16 cases. 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) performed with a cardiac protocol demonstrated abnormal uptake in 13 subjects, while late gadolinium enhancement (LGE) on cardiac MRI was detected in 9 subjects [23]. One subject underwent heart transplant. For nervous system involvement, three subjects had small fiber neuropathy, followed by three pituitary, two meninges, two spinal cord, one cranial nerve, one white matter, one sensory hearing loss, and one cranio-facial with peripheral neuropathy. Two subjects had both cardiac and neurological involvement.

Pulmonary function tests and Scadding stage

Pulmonary function tests were available for 185 subjects. Normal pulmonary function tests were observed in 109 (58.9%) subjects, a restrictive pattern in 40 subjects (21.6%), an obstructive pattern in 32 subjects (17.3%), and mixed obstructive and restrictive pattern in 4 (2.2%) subjects. For FVC, the median and 25th percentile were 88% predicted and 72% predicted, respectively. For FEV1, they were 84% predicted and 67% predicted, and for DLCO, they were 93% predicted and 75% predicted, respectively. Lung volumes were available for 129 subjects, and the median and 25th percentile of TLC were 95% predicted and 82% predicted, respectively. Twenty-nine (22.5%) cases had decreased TLC (below 80% predicted), and 22 (17.1%) had restriction on spirometry and decreased TLC. Decreased TLC with normal spirometry was seen in only 4 (3.1%) cases. Fifty-five (30.4%) subjects had reduced DLCO, while 12 (6.6%) had reduced DLCO with normal spirometry. Eighteen (14.0%) subjects had abnormal DLCO with normal TLC, and 4 (2.2%) had an isolated decrease in DLCO with normal spirometry and lung volumes. Black subjects had lower mean FVC, FEV1, and DCLO than white subjects (p < 0.001) (Fig. 1, panel a, b, d). Likewise, subjects treated with anti-inflammatory medications for at least ≥1 month had lower mean FVC, FEV1, and DLCO (p < 0.001), and lower FEV1/FVC ratio (p = 0.006) compared to untreated subjects (Fig. 1, panel e, f, g, h).
Ninety-two (49.2%) cases had parenchymal lung involvement with Scadding stage II or III disease. Seventy cases (37.4%) had Scadding stage 0 disease, and 2.1% had primarily fibrotic stage IV (Table 4). Difference in the mean FVC, FEV1, FEV1/FVC and DLCO were observed based on Scadding stage (p < 0.001, Fig. 2). The mean FVC was 92 ± 19% for stage 0, 95 ± 17% for stage I, 80 ± 20 for stage II, 79 ± 21 for stage III, and 48 ± 18 for stage IV. The mean FVC was lower in stage IV disease compared to other Scadding stages, suggesting a restrictive pattern in these cases. Cases with parenchymal involvement (stage II or III) had lower mean FVC than those without parenchymal disease (stage 0 or I). The mean DLCO was 97 ± 23% for stage 0, 99 ± 28% for stage I, 86 ± 21% for stage II, 81 ± 26% for stage III, and 39 ± 19% for stage IV. Similar to FVC, the mean DLCO was lower for the Scadding stage IV cases compared to those of other Scadding stages, and although in normal range, the mean DLCO of stage II and II combined was lower than cases with no parenchymal lung disease. The mean FEV1 was 90 ± 20 for stage 0, 94 ± 13 for stage I, 69 ± 21 for stage II, 74 ± 22 for stage III, and 31 ± 10 for stage IV. Of the 32 cases with obstructive disease, the mean FEV1 was 69 ± 10 for stage 0 (n = 5), 53 ± 18 for stage II (n = 15), 55 ± 14 for stage III (n = 10), and 27 ± 6 for stage IV (n = 2). No subject with obstructive pattern had stage I disease.
Table 4
Demographics by Scadding stage
Stage
0
I
II
III
IV
p value
Number of subjects
70 (37.4%)
21 (11.2%)
47 (25.1%)
45 (24.1%)
4 (2.1%)
 
Age at enrollment (years)
52.5 ± 12.2
49.0 ± 12.5
53.9 ± 13.2
54.5 ± 12.7
53.5 ± 9.7
0.55
Age at diagnosis (years)
47.1 ± 12.3
43.2 ± 11.7
45.9 ± 13.9
45.0 ± 11.6
41.5 ± 4.2
0.67
BMI (kg/m2)
31.5 ± 7.9
30.3 ± 5.0
31.2 ± 5.2
31.6 ± 8.5
29.3 ± 4.1
0.94
Sex
 Male
30 (16.0)
12 (6.4)
31 (16.6)
26 (13.9)
4 (2.1)
0.04
 Female
40 (21.4)
9 (4.8)
16 (8.6)
19 (10.2)
0 (0.0)
 
Race
 White
52 (27.8)
12 (6.4)
38 (20.3)
29 (15.5)
1 (0.5)
0.04
 Black
9 (4.8)
3 (1.6)
7 (3.7)
11 (5.9)
2 (1.1)
 
 Asian
3 (1.6)
0 (0.0)
0 (0.0)
0 (0.0)
0 (0.0)
 
 American Indian or Alaska Native
1 (0.5)
0 (0.0)
0 (0.0)
0 (0.0)
0 (0.0)
 
 Choose not to answer
5 (2.7)
6 (3.2)
2 (1.1)
5 (2.7)
1 (0.5)
 
Smoking status
 Current
6 (3.2)
2 (1.1)
3 (1.6)
3 (1.6)
1 (0.5)
0.27
 Former
28 (15.0)
3 (1.6)
18 (9.6)
20 (10.7)
2 (1.1)
 
 Never
36 (19.3)
16 (8.6)
26 (13.9)
22 (11.8)
1 (0.5)
 

Treatment groups

The majority (71.1%) of subjects reported treatment for at least 1 month for sarcoidosis. Systemic anti-inflammatory agents for greater than three months were needed in 123 (65.8%) cases, and only 22 (11.8%) were off therapy at one-year anniversary of start treatment (Table 5). Prednisone was the first agent for a majority (70.1%) of the cases, and methotrexate and Imuran were used in 59 (31.6%) and 9 (4.8%) cases respectively. Fourteen cases received infliximab with a preferred dose of 5 mg/kg for induction (0, 2 and 4 weeks) and a maintenance dose given every 4 weeks. Solumedrol 1 g once a month was used in cases with cardiac disease and when there were adverse effects or intolerance of conventional oral or biologic agents. 31.0% of subjects were treated with only one anti-inflammatory agent, while 23.5% were treated with two and 10.7% with three anti-inflammatory agents (Table 6) for management of sarcoidosis over the duration of illness.
Table 5
Treatment groups by duration and agents
Treatment Groups
n
%
Treated < 1 month or never
54
28.9
Treatment ≥1 month
133
71.1
Treatment > 3 months
123
65.8
Off treatment ≥12 months
22
11.8
Prednisone
131
70.1
Methylprednisolone
13
7.0
Methotrexate
59
31.6
Azathioprine
9
4.8
Mycophenolic acid
13
7.0
Hydroxychloroquine
16
8.6
Infliximab
14
7.5
Table 6
Number of immunosuppression agents used in each subject
Number of agents used
n (%)
0
54 (28.9%)
1
58 (31.0%)
2
44 (23.5%)
3
20 (10.7%)
4
7 (3.7%)
5
4 (2.1%)

Clinical phenotypes per GRADS

Clinical phenotypes were determined using the GRADS study criteria. Cases with remitting disease, i.e., subjects who were never treated or were off treatment for greater than 12 months, were the largest group (n = 45 [24.1%]) in this cohort (Table 7). Of those classified into Group 8, five subjects had multiorgan involvement while four subjects had cardiac involvement. We then evaluated the proportion of cases with parenchymal disease based on GRADS phenotyping. Forty-four (23.5%) subjects were classified into Group 3 Stage II-III treated, with 23 (12.3%) subjects classified into Group 4 Stage II-III untreated. Meanwhile, only 1.1 and 0.5% of subjects were classified into Group 5 Stage IV treated and Group 6 Stage IV untreated, respectively. A substantial number of cases were not classifiable based on GRADS phenotyping (n = 37 [19.8%]), including 4.3% with Stage I who were treated, 14.4% with Stage 0 treated, and 1.1% with Stage 0 untreated. Some of the cases who met criteria for Group 1 (multiorgan disease) or Group 9 (cardiac defining therapy) were not included for the respective groups if they met criteria for remitting disease. For instance, while 25 subjects had multiorgan involvement, only 16 of them were classified into Group 1 Multiorgan since six of them were classified into Group 8 Remitting and three of them were classified into Group 9 Cardiac defining therapy. Likewise, while 20 subjects had cardiac involvement, only seven of them who were treated for cardiac involvement and not remitting were classified into Group 9 Cardiac defining therapy.
Table 7
Clinical phenotype study groups per GRADS
Clinical Phenotype Study Groups
n
%
Group 1: Multiorgan
16
8.6
Group 2: Nonacute, Stage I, untreated
8
4.3
Group 3: Stage II-III, treated
44
23.5
Group 4: Stage II-III, untreated
23
12.3
Group 5: Stage IV, treated
2
1.1
Group 6: Stage IV, untreated
1
0.5
Group 7: Acute sarcoidosis
4
2.1
Group 8: Remitting
45
24.1
Group 9: Cardiac defining therapy
7
3.7
Unclassifiable per GRADS
37
19.8
 Stage 1, treated
8
4.3
 Stage 0, treated
27
14.4
 Stage 0, untreated
2
1.1

Laboratory values

The sarcoidosis relevant laboratory studies for this cohort are shown in Table 8. A higher WBC count (p = 0.003), and lower albumin (p = 0.04) was seen in cases who were treated. White cases had lower IgG (p = 0.01), higher albumin (p = 0.01), and lower total protein levels (p = 0.002) compared to blacks. No significant difference between treated and untreated groups, and between whites and blacks was found for ACE, percent lymphocytes, soluble IL2 receptor, CRP, total bilirubin, alkaline phosphatase, alanine aminotransferase, aspartate aminotransferase, calcium, parathyroid hormone, 25-OH Vitamin D, and 1,25 Di OH Vitamin D.
Table 8
Lab values between treated vs untreated, and whites and blacks
Characteristics
Total
Treated
Untreated
 
White
Black
 
n
mean ± SD
n
mean ± SD
n
mean ± SD
p-value
n
mean ± SD
n
mean ± SD
p-value
ACE (U/L)
124
43.6 ± 40.6
85
42.7 ± 43.1
39
45.7 ± 35.0
0.68
90
43.8 ± 39.8
19
50.9 ± 51.7
0.58
WBC (109/L)
176
6.7 ± 2.6
128
7.0 ± 2.7
48
5.9 ± 1.9
0.003
127
6.7 ± 2.4
29
6.1 ± 2.2
0.17
Lymphocytes (%)
169
21.4 ± 20.1
123
20.8 ± 22.9
46
23.2 ± 8.9
0.32
120
21.2 ± 22.7
29
23.5 ± 11.1
0.43
sIL-2R (unit/mL)
47
599.8 ± 306.9
33
580.5 ± 317.4
14
645.5 ± 286.5
0.50
32
600.9 ± 273.1
6
597.8 ± 452.1
0.99
CRP (mg/L)
96
12.8 ± 29.0
65
13.3 ± 31.2
31
11.8 ± 24.1
0.80
64
8.3 ± 13.6
17
28.8 ± 59.1
0.17
IgG (mg/dL)
91
1171.9 ± 379.8
60
1136.5 ± 362.9
31
1240.6 ± 407.7
0.22
60
1070.8 ± 277.4
16
1492.3 ± 555.5
0.01
Albumin (g/dL)
172
3.8 ± 0.4
123
3.8 ± 0.4
49
3.9 ± 0.3
0.04
122
3.9 ± 0.4
28
3.7 ± 0.3
0.01
Total protein (g/dL)
172
7.6 ± 0.6
123
7.6 ± 0.6
49
7.7 ± 0.5
0.09
122
7.5 ± 0.5
28
7.9 ± 0.6
0.002
Total bilirubin (mg/dL)
172
0.6 ± 0.3
123
0.6 ± 0.4
49
0.6 ± 0.3
0.86
122
0.6 ± 0.3
28
0.6 ± 0.4
0.65
ALP (U/L)
172
91.1 ± 44.7
123
93.6 ± 44.4
49
84.7 ± 45.3
0.25
122
86.9 ± 39.5
28
112.3 ± 66.3
0.06
ALT (U/L)
172
35.8 ± 16.1
123
36.9 ± 17.1
49
33.0 ± 12.8
0.11
122
36.4 ± 15.7
28
32.8 ± 18.0
0.33
AST (U/L)
172
24.4 ± 11.2
123
25.2 ± 12.2
49
22.4 ± 7.8
0.08
122
23.8 ± 9.4
28
24.7 ± 13.0
0.72
Calcium (mg/dL)
177
9.1 ± 0.6
128
9.1 ± 0.6
49
9.2 ± 0.5
0.18
125
9.1 ± 0.6
30
9.2 ± 0.6
0.39
PTH (pg/mL)
46
53.5 ± 38.5
32
58.5 ± 44.2
14
42.1 ± 16.3
0.07
30
55.7 ± 35.6
8
48.9 ± 62.1
0.77
25, OH Vit D (ug/L)
99
29.5 ± 18.0
70
28.8 ± 19.2
29
31.1 ± 14.9
0.52
67
28.4 ± 16.2
15
31.7 ± 21.4
0.57
1, 25 DiOH Vitamin D (pg/mL)
88
53.1 ± 21.1
61
51.4 ± 21.8
27
56.9 ± 19.2
0.24
59
50.7 ± 19.4
12
59.0 ± 24.2
0.28

Discussion

Our cohort has similarities and crucial differences compared to previously described cohorts (Table 9). This cohort is primarily composed of cases from upper Midwest and has a higher proportion of blacks and lower proportion of whites compared to the racial distribution of Minnesota residents. With respect to the ACCESS cohort, the age at diagnosis in our cohort was similar, but we observed a higher proportion of males and a lower proportion of black subjects in our cohort. We also observed a higher proportion of extra-thoracic lymph node, eye, liver, spleen, and bone-joint involvement. Other cohorts also reported similarities and differences compared to the ACCESS cohort. A cohort of 166 sarcoidosis cases in Israel was older [17]. Similar to ACCESS cases, more patients in the Israeli cohort were females, and the majority of patients had lung involvement. However, fewer sarcoidosis cases in this cohort had extra thoracic lymph node, liver, CNS, and joints involvement. A small cohort of 21 cases form Mexico was different than the ACCESS cohort with a lower proportion of women and lower median age [16]. This group had more frequent skin and bone marrow involvement and less frequent pulmonary involvement. In contrast, a cohort from the University of California [24] found a higher proportion of cases with neurologic involvement, and another cohort in Olmsted County in Minnesota [25] had fewer cases with eye involvement, but more bone and joint disease. In our cohort, in addition to higher proportion of extra-thoracic lymph node, eye, liver, spleen, and bone-joint involvement, cardiac sarcoidosis, a high-risk manifestation of sarcoidosis, was also more frequent. Given the interaction between the environmental factors and genetics in the pathogenesis of sarcoidosis, regional variations in disease phenotypes may be due to differences in the genetics of the population and/or the nature of the exposure(s) triggering the disease. Because our cohort is drawn from a referral center for sarcoidosis, there may be enrichment for more complicated and advanced cases.
Table 9
Demographics and organ involvement characteristics of various cohorts
Characteristics
UMN
(n = 187)
ACCESS
(n = 736)
Israel (n = 166)
Mexico City
(n = 21)
UCSF
(n = 126)
Olmsted County (n = NA)
Mean age ± SD (years)
45.8 ± 12.4a
(46% < 40 yr old, 54% ≥ 40 yr old)a
62 ± 14
NA
50 ± 12b
NA
Median age (years)
45 (22–75)a
42.1 (18–83)a
63 (26–94)
31 (18–72)a
51 (30–76)b
NA
Male, n (%)
103 (55.1)
268 (36.4)
54 (32.5)
10 (47.6)
49 (39)
NA
Female, n (%)
84 (44.9)
468 (63.6)
112 (67.5)
11 (52.4)
77 (61)
NA
White, n (%)
132 (70.6)
393 (53.4)
NA
NA
97 (77)
NA
Black, n (%)
32 (17.1)
325 (44.2)
NA
NA
18 (14)
NA
Other, n (%)
23 (12.3)
18 (2.4)
Jew: 111 (67)
Arab: 51 (31)
Ethiopian 4 (2)
NA
11 (9)
NA
Lungs, n (%)
184 (98.4)
699 (95.0)
151 (91.0)
14 (66.6)
NA
97%
Skinc, n (%)
27 (14.4)
178 (24.2)
5 (3.0)
9 (42.8)
NA
18%
Non-thoracic lymph node, n (%)
64 (34.2)
112 (15.2)
5 (3.0)
4 (19.0)
NA
3%
Eye, n (%)
39 (20.9)
87 (11.8)
6 (3.6)
4 (19.0)
NA
7%
Liver, n (%)
33 (17.6)
85 (11.5)
6 (3.6)
4 (19)
NA
6%
Spleen, n (%)
39 (20.9)
49 (6.7)
6 (3.6)
0 (0.0)
NA
4%
Neurologic, n (%)
14 (7.5)
34 (4.6)
12 (7.2)
0 (0.0)
23 (18.3%)
3%
Parotid/salivary, n (%)
4 (2.1)
29 (3.9)
0 (0.0)
1 (4.7)
NA
NA
Bone marrow, n (%)
6 (3.2)
29 (3.9)
1 (0.6)
5 (23.8)
NA
NA
Calcium, n (%)
10 (5.3)
27 (3.7)
1 (0.6)
1 (4.7)
NA
NA
ENT, n (%)
4 (2.1)
22 (3.0)
0 (0.0)
1 (4.7)
NA
NA
Cardiac, n (%)
20 (10.7)
17 (2.3)
1 (0.6)
0 (0.0)
5 (4.0%)
1%
Renal, n (%)
2 (1.1)
5 (0.7)
4 (2.4)
0 (0.0)
NA
3%
Bone/joint, n (%)
18 (9.6)
4 (0.5)
6 (3.6)
0 (0.0)
NA
12%
Muscle, n (%)
2 (1.1)
3 (0.4)
0 (0.0)
0 (0.0)
NA
NA
ENT ear, nose, and throat, UMN University of Minnesota, ACCESS A Case Control Etiologic Study of Sarcoidosis, UCSF University of California San Francisco
aAge at diagnosis
bAge at visit
cIncluding erythema nodosum
Pulmonary sarcoidosis continues to result in significant morbidity and mortality with rising death rates [10], hospital admissions, and health care costs [2]. It is associated with significantly reduced health related quality of life and symptoms portending the need for treatment and for disease progression [26]. A large proportion of our cases were either stage II/III disease or stage 0 disease, and stage IV primarily fibrotic disease is less common. A substantial proportion of our cases had near normal spirometry and DLCO, and restrictive ventilatory defect was the most frequent finding on pulmonary function testing compared to obstructive and mixed patterns. A large proportion of subjects in our cohort required treatment with anti-inflammatory agents. In the ACCESS incident cohort, lung function was normal in many with FVC > 80% in 69%, yet ~ 47% cases had Scadding stage II/III disease while stage IV disease was present in only ~ 5%. In our cohort, the median and 25th percentile of FVC is 88% predicted and 72% predicted, findings similar to ACCESS. Similar results were also seen for FEV1, and obstructive changes were common with an FEV1/FVC ratio of < 70% in more than 20% of patients. An isolated gas exchange abnormality is not uncommon in sarcoidosis and thus an important clinical outcome. In our cohort, 14.0% had an abnormal DLCO and normal TLC, while 6.6% had abnormal DLCO and normal spirometry, and 17% demonstrated normal DLCO and abnormal spirometry. Thus, patients may demonstrate obstructive, restrictive, mixed patterns or an isolated DLCO. Researchers have commonly used spirometry, DLCO, and chest radiography, to track disease progression in studies [27, 28]. Lung volumes or formal cardiopulmonary exercise testing (CPET) is not routinely used nor is a standard of care in sarcoidosis clinical practice or research as it is more invasive, expensive and normal in many [29]. Patient-reported outcome measures and the six-minute walk test have been recommended in disease assessment [30]. A recent international Delphi study supported multi-faceted outcomes to assess disease severity for clinical and research purposes, including spirometry/DLCO, quality of life, assessment of progression as well as a biomarker [31]. These data support the importance of defining a comprehensive biomarker of disease progression.
A recent NHLBI workshop recognized cardiac involvement as a high-risk phenotype [12, 32]. The proportion of sarcoidosis cases with abnormal advanced cardiac imaging was substantially higher in our cohort than the ACCESS and other cohorts (Table 9), but less than the reported incidence in post mortem studies [3335]. Similar rates of increased detection of cardiac sarcoidosis has also been reported by other investigators employing contemporary imaging studies [3638]. The utility of these imaging modalities is now widely recognized and incorporated in the diagnostic criteria for cardiac sarcoidosis. The ACCESS criteria did not have advanced imaging criteria, and for the 2006 Japanese Ministry of Health and Welfare (JMHW) criteria, late gadolinium enhancement (LGE) on cardiac MRI was a minor criterion [39] and needed additional criteria to be fulfilled for a diagnosis of cardiac sarcoidosis. In contrast, in the current recommendations by Heart Rhythm Society (HRS), LGE on CMR is sufficient for an adequate clinical diagnosis of cardiac disease in the presence of histologic extracardiac evidence of sarcoidosis [40]. Similarly, for the WASOG organ assessment tool that we used for this cohort [18] and the Japanese Circulation Society Guidelines [41], LGE on cardiac MRI makes the likelihood of cardiac involvement at least probable. Similarly, patchy uptake on a dedicated cardiac PET scan has also been incorporated into the HRS and the WASOG criteria. A crucial element in establishing a diagnosis of cardiac sarcoidosis, even with histology proven extra-cardiac disease, is excluding other causes of LGE, such as coronary artery disease or other non-ischemic cardiomyopathies such as genetic arrhythmogenic cardiomyopathies which could also demonstrate increased myocardial uptake on FDG-PET imaging [42]. Challenges also remain regarding the ideal dietary preparation to suppress normal glucose uptake by the myocardium and avoid false positive FDG-PET. A diffuse uptake of FDG is suggestive of incomplete suppression of physiological glucose uptake in contrast to a patchy or patchy on diffuse uptake that may suggest myocardial inflammatory process [43, 44]. In the absence of a gold standard, as suggested by other groups [45], a multidisciplinary approach is likely to improve the accuracy of diagnosis. The long-term outcomes of cases with abnormal cardiac imaging, especially in cases with no or minimal symptoms and normal heart function, remains unknown.
Extra-pulmonary involvement is presumed to be more common in blacks with sarcoidosis. However despite being a cohort composing of predominantly white cases, our cohort had frequent multisystem disease (using the GRADS criteria and also the WASOG organ assessment tool). We observed more frequent involvement of the skin, extra thoracic lymph nodes, liver, spleen, neurologic, cardiac and bone and joint disease. Of these, neurologic, cardiac, and multisystem disease were considered high-risk manifestations of sarcoidosis due to higher morbidity and mortality. Bone and joint involvement is also a frequent cause of impaired quality of life and often requires systemic anti-inflammatory therapy. We included cases with Lofgren’s syndrome in this category due to our observation of significant morbidity during the acute presentation related to arthralgias, although most of these cases had improvement in symptoms and did not need chronic therapy. As the size of our cohort grows, it will be ideally suited for future studies of high-risk sarcoidosis phenotypes.
We also examined laboratory studies obtained at the clinic visits. Routine laboratory tests were performed for surveillance of drug-related side effects to assess the disease activity with biomarkers and for asymptomatic organ involvement [46]. We identified differences in several analytes in the treated vs. untreated groups and in blacks compared to whites. A higher leukocyte count in treated sarcoidosis cases could reflect an appropriate response to therapy including improved hematopoiesis [47]. Leukopenia as a manifestation of sarcoidosis could be due to bone marrow involvement [48, 49], hypersplenism [50], or lymphocyte redistribution. The T cell redistribution is reported to cause peripheral lymphopenia and is linked to worse severity of the sarcoidosis [51, 52]. We did not observe any difference in the lymphocyte count in the treated vs. untreated and blacks vs. whites. Our cohort is larger in size than the cohort where peripheral lymphopenia was observed and has a larger proportion of cases with lung involvement [52]. Other potential biomarkers of disease activity such as angiotensin converting enzyme [53, 54] and soluble IL-2R levels [55, 56] were also not different in treated vs untreated groups or in whites vs. blacks. The higher level of albumin observed in whites and a higher level of total protein in blacks has been reported previously [57] in these racial groups. Difference in protein fractional patterns could explain a higher IgG level. Various reasons including frequent infections or persistent chronic inflammation could account for the differences in albumin and immunoglobulin levels [58, 59]. Overall, these findings are in line with the findings of other reports that current biomarkers have limited utility for assessing disease activity in sarcoidosis, and highlight the urgent need to develop biomarkers for prognostication and measuring response to therapy [32]. Concurrent to the search for useful serum biomarkers for disease activity, methods for incorporating comprehensive data including other assessments of disease activity, extent of organ dysfunction, and patient reported outcomes, are needed to improve disease severity assessment, prognosis assessment, and guidelines for treatment.
We observed a high proportion of cases that needed systemic anti-inflammatory therapy. Oral corticosteroids are the first-line agents with methotrexate and Imuran as the preferred steroid sparing agent. The dosing of TNF blockers used in sarcoidosis at our center is different than the typical dose for rheumatologic conditions with a maintenance dose of every four weeks (5 mg/kg) for infliximab, in line with the current experience-based recommendations [60].
Despite the retrospective nature of our study, this cohort description provides new insights into regional variability of sarcoidosis as it represents the clinical manifestation in residents of the upper Midwest. Our cohort is a mixed population of incident and prevalent cases and might be a more appropriate representation of sarcoid-related morbidity as opposed to a cohort of newly diagnosed cases. For assessment of pulmonary function testing, we used percent predicted values to assess lung function abnormalities. The other option of using percentile cutoff might be more appropriate, but we do not have this information on all the participants in our study. We also acknowledge that not all the cases in our cohort had all the data points that we presented. As our cohort size grows, we expect the missing values to have a lesser impact on the study findings.

Conclusion

Among sarcoidosis cases in our predominantly white Minnesota population, we found a relatively high proportion of advanced and multisystem disease. Lofgren’s syndrome was not common in our cohort despite high percentage of presumed north-European ancestry in our population. Stage II/III pulmonary disease, cardiac disease, multisystem diseases and joint and bone diseases are more frequently detected in this cohort. The analysis of our cohort highlights the lack of utility of serum biomarkers and limitations of pulmonary function tests in assessing prognosis, need for treatment, and disease severity. The higher incidence of cardiac disease, compared to previous cohorts, suggests that cardiac involvement may be under-recognized and highlights the importance of incorporating advanced imaging and a multidisciplinary approach into the evaluation of patients suspected of cardiac involvement. Our cohort is well suited to investigate high-risk sarcoidosis phenotypes, and studying regional variations in disease phenotypes may shed light on disease mechanisms. Incorporating patient reported outcomes in routine care could provide additional insights into indications and outcomes of treatment.

Acknowledgements

Not applicable.
The study was approved by University of Minnesota Institutional Review Board with protocol number 1501 M60321 for collection of biospecimens and clinical data.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Statement on sarcoidosis. Joint Statement of the American Thoracic Society (ATS), the European Respiratory Society (ERS) and the World Association of Sarcoidosis and Other Granulomatous Disorders (WASOG) adopted by the ATS Board of Directors and by the ERS Executive Committee, February 1999. Am J Respir Crit Care Med. 1999;160(2):736–55. Statement on sarcoidosis. Joint Statement of the American Thoracic Society (ATS), the European Respiratory Society (ERS) and the World Association of Sarcoidosis and Other Granulomatous Disorders (WASOG) adopted by the ATS Board of Directors and by the ERS Executive Committee, February 1999. Am J Respir Crit Care Med. 1999;160(2):736–55.
3.
Zurück zum Zitat Gerke AK, Judson MA, Cozier YC, Culver DA, Koth LL. Disease Burden and Variability in Sarcoidosis. Ann Am Thorac Soc. 2017;14(Supplement_6):S421–8.PubMedPubMedCentralCrossRef Gerke AK, Judson MA, Cozier YC, Culver DA, Koth LL. Disease Burden and Variability in Sarcoidosis. Ann Am Thorac Soc. 2017;14(Supplement_6):S421–8.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Valeyre D, Prasse A, Nunes H, Uzunhan Y, Brillet PY, Muller-Quernheim J. Sarcoidosis. Lancet. 2014;383(9923):1155–67.PubMedCrossRef Valeyre D, Prasse A, Nunes H, Uzunhan Y, Brillet PY, Muller-Quernheim J. Sarcoidosis. Lancet. 2014;383(9923):1155–67.PubMedCrossRef
5.
Zurück zum Zitat Erdal BS, Clymer BD, Yildiz VO, Julian MW, Crouser ED. Unexpectedly high prevalence of sarcoidosis in a representative U.S. metropolitan population. Respir Med. 2012;106(6):893–9.PubMedCrossRef Erdal BS, Clymer BD, Yildiz VO, Julian MW, Crouser ED. Unexpectedly high prevalence of sarcoidosis in a representative U.S. metropolitan population. Respir Med. 2012;106(6):893–9.PubMedCrossRef
7.
Zurück zum Zitat Rybicki BA, Major M, Popovich J Jr, Maliarik MJ, Iannuzzi MC. Racial differences in sarcoidosis incidence: a 5-year study in a health maintenance organization. Am J Epidemiol. 1997;145(3):234–41.PubMedCrossRef Rybicki BA, Major M, Popovich J Jr, Maliarik MJ, Iannuzzi MC. Racial differences in sarcoidosis incidence: a 5-year study in a health maintenance organization. Am J Epidemiol. 1997;145(3):234–41.PubMedCrossRef
8.
9.
Zurück zum Zitat Mirsaeidi M, Machado RF, Schraufnagel D, Sweiss NJ, Baughman RP. Racial difference in sarcoidosis mortality in the United States. Chest. 2015;147(2):438–49.PubMedCrossRef Mirsaeidi M, Machado RF, Schraufnagel D, Sweiss NJ, Baughman RP. Racial difference in sarcoidosis mortality in the United States. Chest. 2015;147(2):438–49.PubMedCrossRef
10.
Zurück zum Zitat Swigris JJ, Olson AL, Huie TJ, Fernandez-Perez ER, Solomon J, Sprunger D, Brown KK. Sarcoidosis-related mortality in the United States from 1988 to 2007. Am J Respir Crit Care Med. 2011;183(11):1524–30.PubMedPubMedCentralCrossRef Swigris JJ, Olson AL, Huie TJ, Fernandez-Perez ER, Solomon J, Sprunger D, Brown KK. Sarcoidosis-related mortality in the United States from 1988 to 2007. Am J Respir Crit Care Med. 2011;183(11):1524–30.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Baughman RP, Teirstein AS, Judson MA, Rossman MD, Yeager H Jr, Bresnitz EA, DePalo L, Hunninghake G, Iannuzzi MC, Johns CJ, et al. Clinical characteristics of patients in a case control study of sarcoidosis. Am J Respir Crit Care Med. 2001;164(10 Pt 1):1885–9.PubMedCrossRef Baughman RP, Teirstein AS, Judson MA, Rossman MD, Yeager H Jr, Bresnitz EA, DePalo L, Hunninghake G, Iannuzzi MC, Johns CJ, et al. Clinical characteristics of patients in a case control study of sarcoidosis. Am J Respir Crit Care Med. 2001;164(10 Pt 1):1885–9.PubMedCrossRef
12.
Zurück zum Zitat Sauer WH, Stern BJ, Baughman R, Culver DA, Royal W. High risk Sarcoidosis: current concepts and research imperatives. Ann Am Thorac Soc. 2017;14 (Supplement_6):S437–S444. Sauer WH, Stern BJ, Baughman R, Culver DA, Royal W. High risk Sarcoidosis: current concepts and research imperatives. Ann Am Thorac Soc. 2017;14 (Supplement_6):S437–S444.
13.
Zurück zum Zitat Iwai K, Sekiguti M, Hosoda Y, DeRemee RA, Tazelaar HD, Sharma OP, Maheshwari A, Noguchi TI. Racial difference in cardiac sarcoidosis incidence observed at autopsy. Sarcoidosis. 1994;11(1):26–31.PubMed Iwai K, Sekiguti M, Hosoda Y, DeRemee RA, Tazelaar HD, Sharma OP, Maheshwari A, Noguchi TI. Racial difference in cardiac sarcoidosis incidence observed at autopsy. Sarcoidosis. 1994;11(1):26–31.PubMed
14.
Zurück zum Zitat Brown F, Tanner LS. Lofgren Syndrome. In StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2020. PMID: 29493940. Brown F, Tanner LS. Lofgren Syndrome. In StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2020. PMID: 29493940.
15.
Zurück zum Zitat Judson MA, Baughman RP, Teirstein AS, Terrin ML, Yeager H Jr. Defining organ involvement in sarcoidosis: the ACCESS proposed instrument. ACCESS research group. A Case control etiologic study of Sarcoidosis. Sarcoidosis Vasc Diffuse Lung Dis. 1999;16(1):75–86.PubMed Judson MA, Baughman RP, Teirstein AS, Terrin ML, Yeager H Jr. Defining organ involvement in sarcoidosis: the ACCESS proposed instrument. ACCESS research group. A Case control etiologic study of Sarcoidosis. Sarcoidosis Vasc Diffuse Lung Dis. 1999;16(1):75–86.PubMed
16.
Zurück zum Zitat Carrillo-Perez DL, Apodaca-Chaveza EI, Carrillo-Maravilla E, Mejia-Avila M, Hernandez-Oropeza JL, Reyes E, Atisha-Fregoso Y, Hernandez-Molina G. Sarcoidosis: a single hospital-based study in a 24-year period. Rev Investig Clin. 2015;67(1):33–8. Carrillo-Perez DL, Apodaca-Chaveza EI, Carrillo-Maravilla E, Mejia-Avila M, Hernandez-Oropeza JL, Reyes E, Atisha-Fregoso Y, Hernandez-Molina G. Sarcoidosis: a single hospital-based study in a 24-year period. Rev Investig Clin. 2015;67(1):33–8.
17.
Zurück zum Zitat Markevitz N, Epstein Shochet G, Levi Y, Israeli-Shani L, Shitrit D. Sarcoidosis in Israel: clinical outcome status, organ involvement, and long-term follow-up. Lung. 2017;195(4):419–24.PubMedCrossRef Markevitz N, Epstein Shochet G, Levi Y, Israeli-Shani L, Shitrit D. Sarcoidosis in Israel: clinical outcome status, organ involvement, and long-term follow-up. Lung. 2017;195(4):419–24.PubMedCrossRef
18.
Zurück zum Zitat Judson MA, Costabel U, Drent M, Wells A, Maier L, Koth L, Shigemitsu H, Culver DA, Gelfand J, Valeyre D, et al. The WASOG Sarcoidosis organ assessment instrument: an update of a previous clinical tool. Sarcoidosis Vasc Diffuse Lung Dis. 2014;31(1):19–27.PubMed Judson MA, Costabel U, Drent M, Wells A, Maier L, Koth L, Shigemitsu H, Culver DA, Gelfand J, Valeyre D, et al. The WASOG Sarcoidosis organ assessment instrument: an update of a previous clinical tool. Sarcoidosis Vasc Diffuse Lung Dis. 2014;31(1):19–27.PubMed
19.
Zurück zum Zitat Moller DR, Koth LL, Maier LA, Morris A, Drake W, Rossman M, Leader JK, Collman RG, Hamzeh N, Sweiss NJ, et al. Rationale and Design of the Genomic Research in Alpha-1 antitrypsin deficiency and Sarcoidosis (GRADS) study. Sarcoidosis Protocol. Ann Am Thorac Soc. 2015;12(10):1561–71.PubMedPubMedCentralCrossRef Moller DR, Koth LL, Maier LA, Morris A, Drake W, Rossman M, Leader JK, Collman RG, Hamzeh N, Sweiss NJ, et al. Rationale and Design of the Genomic Research in Alpha-1 antitrypsin deficiency and Sarcoidosis (GRADS) study. Sarcoidosis Protocol. Ann Am Thorac Soc. 2015;12(10):1561–71.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Vogelmeier CF, Criner GJ, Martinez FJ, Anzueto A, Barnes PJ, Bourbeau J, Celli BR, Chen R, Decramer M, Fabbri LM, et al. Global Strategy for the Diagnosis, Management, and Prevention of Chronic Obstructive Lung Disease 2017 Report. GOLD executive summary. Am J Respir Crit Care Med. 2017;195(5):557–82.PubMedCrossRef Vogelmeier CF, Criner GJ, Martinez FJ, Anzueto A, Barnes PJ, Bourbeau J, Celli BR, Chen R, Decramer M, Fabbri LM, et al. Global Strategy for the Diagnosis, Management, and Prevention of Chronic Obstructive Lung Disease 2017 Report. GOLD executive summary. Am J Respir Crit Care Med. 2017;195(5):557–82.PubMedCrossRef
23.
Zurück zum Zitat Kazmirczak F, Chen KA, Adabag S, von Wald L, Roukoz H, Benditt DG, Okasha O, Farzaneh-Far A, Markowitz J, Nijjar PS, et al. Assessment of the 2017 AHA/ACC/HRS guideline recommendations for implantable Cardioverter-defibrillator implantation in cardiac Sarcoidosis. Circ Arrhythm Electrophysiol. 2019;12(9):e007488.PubMedCrossRef Kazmirczak F, Chen KA, Adabag S, von Wald L, Roukoz H, Benditt DG, Okasha O, Farzaneh-Far A, Markowitz J, Nijjar PS, et al. Assessment of the 2017 AHA/ACC/HRS guideline recommendations for implantable Cardioverter-defibrillator implantation in cardiac Sarcoidosis. Circ Arrhythm Electrophysiol. 2019;12(9):e007488.PubMedCrossRef
24.
Zurück zum Zitat Ungprasert P, Crowson CS, Matteson EL. Epidemiology and clinical characteristics of sarcoidosis: an update from a population-based cohort study from Olmsted County, Minnesota. Reumatismo. 2017;69(1):16–22.PubMedPubMedCentralCrossRef Ungprasert P, Crowson CS, Matteson EL. Epidemiology and clinical characteristics of sarcoidosis: an update from a population-based cohort study from Olmsted County, Minnesota. Reumatismo. 2017;69(1):16–22.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Benn BS, Lehman Z, Kidd SA, Ho M, Sun S, Ramstein J, Arger NK, Nguyen CP, Su R, Gomez A, et al. Clinical and biological insights from the University of California san Francisco prospective and longitudinal cohort. Lung. 2017;195(5):553–61.PubMedCrossRef Benn BS, Lehman Z, Kidd SA, Ho M, Sun S, Ramstein J, Arger NK, Nguyen CP, Su R, Gomez A, et al. Clinical and biological insights from the University of California san Francisco prospective and longitudinal cohort. Lung. 2017;195(5):553–61.PubMedCrossRef
26.
Zurück zum Zitat Judson MA, Preston S, Hu K, Zhang R, Jou S, Modi A, Sukhu I, Ilyas F, Rosoklija G, Yucel R. Quantifying the relationship between symptoms at presentation and the prognosis of sarcoidosis. Respir Med. 2019;152:14–9.PubMedCrossRef Judson MA, Preston S, Hu K, Zhang R, Jou S, Modi A, Sukhu I, Ilyas F, Rosoklija G, Yucel R. Quantifying the relationship between symptoms at presentation and the prognosis of sarcoidosis. Respir Med. 2019;152:14–9.PubMedCrossRef
27.
Zurück zum Zitat Su R, Li MM, Bhakta NR, Solberg OD, Darnell EP, Ramstein J, Garudadri S, Ho M, Woodruff PG, Koth LL. Longitudinal analysis of sarcoidosis blood transcriptomic signatures and disease outcomes. Eur Respir J. 2014;44(4):985–93.PubMedCrossRef Su R, Li MM, Bhakta NR, Solberg OD, Darnell EP, Ramstein J, Garudadri S, Ho M, Woodruff PG, Koth LL. Longitudinal analysis of sarcoidosis blood transcriptomic signatures and disease outcomes. Eur Respir J. 2014;44(4):985–93.PubMedCrossRef
28.
Zurück zum Zitat Arger NK, Ho M, Woodruff PG, Koth LL. Serum CXCL11 correlates with pulmonary outcomes and disease burden in sarcoidosis. Respir Med. 2019;152:89–96.PubMedPubMedCentralCrossRef Arger NK, Ho M, Woodruff PG, Koth LL. Serum CXCL11 correlates with pulmonary outcomes and disease burden in sarcoidosis. Respir Med. 2019;152:89–96.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Chenivesse C, Boulanger S, Langlois C, Wemeau-Stervinou L, Perez T, Wallaert B. Oxygen desaturation during a 6-minute walk test as a predictor of maximal exercise-induced gas exchange abnormalities in sarcoidosis. J Thorac Dis. 2016;8(8):1995–2003.PubMedPubMedCentralCrossRef Chenivesse C, Boulanger S, Langlois C, Wemeau-Stervinou L, Perez T, Wallaert B. Oxygen desaturation during a 6-minute walk test as a predictor of maximal exercise-induced gas exchange abnormalities in sarcoidosis. J Thorac Dis. 2016;8(8):1995–2003.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Baughman RP, Sparkman BK, Lower EE. Six-minute walk test and health status assessment in sarcoidosis. Chest. 2007;132(1):207–13.PubMedCrossRef Baughman RP, Sparkman BK, Lower EE. Six-minute walk test and health status assessment in sarcoidosis. Chest. 2007;132(1):207–13.PubMedCrossRef
31.
Zurück zum Zitat Kampstra NA, Grutters JC, van Beek FT, Culver DA, Baughman RP, Renzoni EA, Wuyts W, Kouranos V, Wijsenbeek MS, Biesma DH, et al. First patient-centred set of outcomes for pulmonary sarcoidosis: a multicentre initiative. BMJ Open Respir Res. 2019;6(1):e000394.PubMedPubMedCentralCrossRef Kampstra NA, Grutters JC, van Beek FT, Culver DA, Baughman RP, Renzoni EA, Wuyts W, Kouranos V, Wijsenbeek MS, Biesma DH, et al. First patient-centred set of outcomes for pulmonary sarcoidosis: a multicentre initiative. BMJ Open Respir Res. 2019;6(1):e000394.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Maier LA, Crouser ED, Martin WJ, Eu J: Executive summary of the NHLBI Workshop Report: Leveraging Current Scientific Advances to Better Understand Sarcoidosis Variability and Improve Outcomes. Ann Am Thorac Soc 2017. Maier LA, Crouser ED, Martin WJ, Eu J: Executive summary of the NHLBI Workshop Report: Leveraging Current Scientific Advances to Better Understand Sarcoidosis Variability and Improve Outcomes. Ann Am Thorac Soc 2017.
33.
Zurück zum Zitat Matsui Y, Iwai K, Tachibana T, Fruie T, Shigematsu N, Izumi T, Homma AH, Mikami R, Hongo O, Hiraga Y, et al. Clinicopathological study of fatal myocardial sarcoidosis. Ann N Y Acad Sci. 1976;278:455–69.PubMedCrossRef Matsui Y, Iwai K, Tachibana T, Fruie T, Shigematsu N, Izumi T, Homma AH, Mikami R, Hongo O, Hiraga Y, et al. Clinicopathological study of fatal myocardial sarcoidosis. Ann N Y Acad Sci. 1976;278:455–69.PubMedCrossRef
34.
Zurück zum Zitat Roberts WC, McAllister HA Jr, Ferrans VJ. Sarcoidosis of the heart. A clinicopathologic study of 35 necropsy patients (group 1) and review of 78 previously described necropsy patients (group 11). Am J Med. 1977;63(1):86–108.PubMedCrossRef Roberts WC, McAllister HA Jr, Ferrans VJ. Sarcoidosis of the heart. A clinicopathologic study of 35 necropsy patients (group 1) and review of 78 previously described necropsy patients (group 11). Am J Med. 1977;63(1):86–108.PubMedCrossRef
35.
Zurück zum Zitat Silverman KJ, Hutchins GM, Bulkley BH. Cardiac sarcoid: a clinicopathologic study of 84 unselected patients with systemic sarcoidosis. Circulation. 1978;58(6):1204–11.PubMedCrossRef Silverman KJ, Hutchins GM, Bulkley BH. Cardiac sarcoid: a clinicopathologic study of 84 unselected patients with systemic sarcoidosis. Circulation. 1978;58(6):1204–11.PubMedCrossRef
36.
Zurück zum Zitat Patel AR, Klein MR, Chandra S, Spencer KT, Decara JM, Lang RM, Burke MC, Garrity ER, Hogarth DK, Archer SL, et al. Myocardial damage in patients with sarcoidosis and preserved left ventricular systolic function: an observational study. Eur J Heart Fail. 2011;13(11):1231–7.PubMedCrossRef Patel AR, Klein MR, Chandra S, Spencer KT, Decara JM, Lang RM, Burke MC, Garrity ER, Hogarth DK, Archer SL, et al. Myocardial damage in patients with sarcoidosis and preserved left ventricular systolic function: an observational study. Eur J Heart Fail. 2011;13(11):1231–7.PubMedCrossRef
37.
Zurück zum Zitat Patel AR, Kramer CM. Role of Cardiac Magnetic Resonance in the Diagnosis and Prognosis of Nonischemic Cardiomyopathy. JACC Cardiovasc Imaging. 2017;10(10 Pt A):1180–93.PubMedPubMedCentralCrossRef Patel AR, Kramer CM. Role of Cardiac Magnetic Resonance in the Diagnosis and Prognosis of Nonischemic Cardiomyopathy. JACC Cardiovasc Imaging. 2017;10(10 Pt A):1180–93.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Patel MR, Cawley PJ, Heitner JF, Klem I, Parker MA, Jaroudi WA, Meine TJ, White JB, Elliott MD, Kim HW, et al. Detection of myocardial damage in patients with sarcoidosis. Circulation. 2009;120(20):1969–77.PubMedPubMedCentralCrossRef Patel MR, Cawley PJ, Heitner JF, Klem I, Parker MA, Jaroudi WA, Meine TJ, White JB, Elliott MD, Kim HW, et al. Detection of myocardial damage in patients with sarcoidosis. Circulation. 2009;120(20):1969–77.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Soejima K, Yada H. The work-up and management of patients with apparent or subclinical cardiac sarcoidosis: with emphasis on the associated heart rhythm abnormalities. J Cardiovasc Electrophysiol. 2009;20(5):578–83.PubMedCrossRef Soejima K, Yada H. The work-up and management of patients with apparent or subclinical cardiac sarcoidosis: with emphasis on the associated heart rhythm abnormalities. J Cardiovasc Electrophysiol. 2009;20(5):578–83.PubMedCrossRef
40.
Zurück zum Zitat Birnie DH, Sauer WH, Bogun F, Cooper JM, Culver DA, Duvernoy CS, Judson MA, Kron J, Mehta D, Cosedis Nielsen J, et al. HRS expert consensus statement on the diagnosis and management of arrhythmias associated with cardiac sarcoidosis. Heart Rhythm. 2014;11(7):1305–23.PubMedCrossRef Birnie DH, Sauer WH, Bogun F, Cooper JM, Culver DA, Duvernoy CS, Judson MA, Kron J, Mehta D, Cosedis Nielsen J, et al. HRS expert consensus statement on the diagnosis and management of arrhythmias associated with cardiac sarcoidosis. Heart Rhythm. 2014;11(7):1305–23.PubMedCrossRef
41.
Zurück zum Zitat Fumio Terasaki KY. New guidelines for diagnosis of cardiac Sarcoidosis in Japan. Ann Nucl Cardiol. 2017. Fumio Terasaki KY. New guidelines for diagnosis of cardiac Sarcoidosis in Japan. Ann Nucl Cardiol. 2017.
42.
Zurück zum Zitat Okasha O, Kazmirczak F, Chen KA, Farzaneh-Far A, Shenoy C. Myocardial involvement in patients with histologically diagnosed cardiac Sarcoidosis: a systematic review and meta-analysis of gross pathological images from autopsy or cardiac transplantation cases. J Am Heart Assoc. 2019;8(10):e011253.PubMedPubMedCentralCrossRef Okasha O, Kazmirczak F, Chen KA, Farzaneh-Far A, Shenoy C. Myocardial involvement in patients with histologically diagnosed cardiac Sarcoidosis: a systematic review and meta-analysis of gross pathological images from autopsy or cardiac transplantation cases. J Am Heart Assoc. 2019;8(10):e011253.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Cerqueira M, Case J, Maddahi J. Proceedings of the ASNC cardiac PET summit meeting, May 12, 2014, Baltimore MD : 5. Advances in radiopharmaceutical availability and development of cardiac PET tracers. J Nucl Cardiol. 2015;22(4):730–9.PubMedCrossRef Cerqueira M, Case J, Maddahi J. Proceedings of the ASNC cardiac PET summit meeting, May 12, 2014, Baltimore MD : 5. Advances in radiopharmaceutical availability and development of cardiac PET tracers. J Nucl Cardiol. 2015;22(4):730–9.PubMedCrossRef
44.
Zurück zum Zitat Slart R, Glaudemans A, Lancellotti P, Hyafil F, Blankstein R, Schwartz RG, Jaber WA, Russell R, Gimelli A, Rouzet F, et al. A joint procedural position statement on imaging in cardiac sarcoidosis: from the cardiovascular and Inflammation & Infection Committees of the European Association of Nuclear Medicine, the European Association of Cardiovascular Imaging, and the American society of nuclear cardiology. J Nucl Cardiol. 2017. Slart R, Glaudemans A, Lancellotti P, Hyafil F, Blankstein R, Schwartz RG, Jaber WA, Russell R, Gimelli A, Rouzet F, et al. A joint procedural position statement on imaging in cardiac sarcoidosis: from the cardiovascular and Inflammation & Infection Committees of the European Association of Nuclear Medicine, the European Association of Cardiovascular Imaging, and the American society of nuclear cardiology. J Nucl Cardiol. 2017.
45.
Zurück zum Zitat Ribeiro Neto ML, Jellis CL, Joyce E, Callahan TD, Hachamovitch R, Culver DA. Update on Cardiac Sarcoidosis. Ann Am Thorac Soc. 2019. Ribeiro Neto ML, Jellis CL, Joyce E, Callahan TD, Hachamovitch R, Culver DA. Update on Cardiac Sarcoidosis. Ann Am Thorac Soc. 2019.
46.
Zurück zum Zitat Judson MA. The three tiers of screening for sarcoidosis organ involvement. Respir Med. 2016;113:42–9.PubMedCrossRef Judson MA. The three tiers of screening for sarcoidosis organ involvement. Respir Med. 2016;113:42–9.PubMedCrossRef
47.
Zurück zum Zitat Pena-Garcia JI, Shaikh S, Barakoti B, Papageorgiou C, Lacasse A. Bone marrow involvement in sarcoidosis: an elusive extrapulmonary manifestation. J Community Hosp Intern Med Perspect. 2019;9(2):150–4.PubMedPubMedCentralCrossRef Pena-Garcia JI, Shaikh S, Barakoti B, Papageorgiou C, Lacasse A. Bone marrow involvement in sarcoidosis: an elusive extrapulmonary manifestation. J Community Hosp Intern Med Perspect. 2019;9(2):150–4.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Yanardag H, Pamuk GE, Karayel T, Demirci S. Bone marrow involvement in sarcoidosis: an analysis of 50 bone marrow samples. Haematologia (Budap). 2002;32(4):419–25. Yanardag H, Pamuk GE, Karayel T, Demirci S. Bone marrow involvement in sarcoidosis: an analysis of 50 bone marrow samples. Haematologia (Budap). 2002;32(4):419–25.
50.
Zurück zum Zitat Pavlovic-Popovic Z, Zaric B, Kosjerina Z, Petrovic D. Splenomegaly in Sarcoidosis: frequency, treatment, prognosis and long-term follow-up. Srp Arh Celok Lek. 2015;143(5–6):279–83.PubMedCrossRef Pavlovic-Popovic Z, Zaric B, Kosjerina Z, Petrovic D. Splenomegaly in Sarcoidosis: frequency, treatment, prognosis and long-term follow-up. Srp Arh Celok Lek. 2015;143(5–6):279–83.PubMedCrossRef
51.
Zurück zum Zitat Jones NP, Tsierkezou L, Patton N. Lymphopenia as a predictor of sarcoidosis in patients with uveitis. Br J Ophthalmol. 2016;100(10):1393–6.PubMedCrossRef Jones NP, Tsierkezou L, Patton N. Lymphopenia as a predictor of sarcoidosis in patients with uveitis. Br J Ophthalmol. 2016;100(10):1393–6.PubMedCrossRef
52.
Zurück zum Zitat Sweiss NJ, Salloum R, Gandhi S, Alegre ML, Sawaqed R, Badaracco M, Pursell K, Pitrak D, Baughman RP, Moller DR, et al. Significant CD4, CD8, and CD19 lymphopenia in peripheral blood of sarcoidosis patients correlates with severe disease manifestations. PLoS One. 2010;5(2):e9088.PubMedPubMedCentralCrossRef Sweiss NJ, Salloum R, Gandhi S, Alegre ML, Sawaqed R, Badaracco M, Pursell K, Pitrak D, Baughman RP, Moller DR, et al. Significant CD4, CD8, and CD19 lymphopenia in peripheral blood of sarcoidosis patients correlates with severe disease manifestations. PLoS One. 2010;5(2):e9088.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Duan J, Xu Y, Zhu H, Zhang H, Sun S, Sun H, Wang W, Xie S. Relationship between CT activity score with lung function and the serum angiotensin converting enzyme in pulmonary sarcoidosis on chest HRCT. Medicine (Baltimore). 2018;97(36):e12205.CrossRef Duan J, Xu Y, Zhu H, Zhang H, Sun S, Sun H, Wang W, Xie S. Relationship between CT activity score with lung function and the serum angiotensin converting enzyme in pulmonary sarcoidosis on chest HRCT. Medicine (Baltimore). 2018;97(36):e12205.CrossRef
54.
Zurück zum Zitat Ungprasert P, Carmona EM, Crowson CS, Matteson EL. Diagnostic utility of angiotensin-converting enzyme in Sarcoidosis: a population-based study. Lung. 2016;194(1):91–5.PubMedCrossRef Ungprasert P, Carmona EM, Crowson CS, Matteson EL. Diagnostic utility of angiotensin-converting enzyme in Sarcoidosis: a population-based study. Lung. 2016;194(1):91–5.PubMedCrossRef
55.
Zurück zum Zitat Ziegenhagen MW, Benner UK, Zissel G, Zabel P, Schlaak M, Muller-Quernheim J. Sarcoidosis: TNF-alpha release from alveolar macrophages and serum level of sIL-2R are prognostic markers. Am J Respir Crit Care Med. 1997;156(5):1586–92.PubMedCrossRef Ziegenhagen MW, Benner UK, Zissel G, Zabel P, Schlaak M, Muller-Quernheim J. Sarcoidosis: TNF-alpha release from alveolar macrophages and serum level of sIL-2R are prognostic markers. Am J Respir Crit Care Med. 1997;156(5):1586–92.PubMedCrossRef
56.
Zurück zum Zitat Miyoshi S, Hamada H, Kadowaki T, Hamaguchi N, Ito R, Irifune K, Higaki J. Comparative evaluation of serum markers in pulmonary sarcoidosis. Chest. 2010;137(6):1391–7.PubMedCrossRef Miyoshi S, Hamada H, Kadowaki T, Hamaguchi N, Ito R, Irifune K, Higaki J. Comparative evaluation of serum markers in pulmonary sarcoidosis. Chest. 2010;137(6):1391–7.PubMedCrossRef
57.
Zurück zum Zitat Koh ET, Chi MS, Lowenstein FW. Comparison of selected blood components by race, sex, and age. Am J Clin Nutr. 1980;33(8):1828–35.PubMedCrossRef Koh ET, Chi MS, Lowenstein FW. Comparison of selected blood components by race, sex, and age. Am J Clin Nutr. 1980;33(8):1828–35.PubMedCrossRef
58.
Zurück zum Zitat Lichtman MA, Hames CG, McDonough JR. Serum protein electrophoretic fractions among negro and White subject in Evans County, Georgia. Am J Clin Nutr. 1965;16:492–508.PubMedCrossRef Lichtman MA, Hames CG, McDonough JR. Serum protein electrophoretic fractions among negro and White subject in Evans County, Georgia. Am J Clin Nutr. 1965;16:492–508.PubMedCrossRef
59.
Zurück zum Zitat Lichtman MA, Vaughan JH, Hames CG. The distribution of serum immunoglobulins, anti-gamma-G globulins ("rheumatoid factors") and antinuclear antibodies in White and negro subjects in Evans County. Georgia. Arthritis Rheum. 1967;10(3):204–15.PubMedCrossRef Lichtman MA, Vaughan JH, Hames CG. The distribution of serum immunoglobulins, anti-gamma-G globulins ("rheumatoid factors") and antinuclear antibodies in White and negro subjects in Evans County. Georgia. Arthritis Rheum. 1967;10(3):204–15.PubMedCrossRef
60.
Zurück zum Zitat Drent M, Cremers JP, Jansen TL, Baughman RP. Practical eminence and experience-based recommendations for use of TNF-alpha inhibitors in sarcoidosis. Sarcoidosis Vasc Diffuse Lung Dis. 2014;31(2):91–107.PubMed Drent M, Cremers JP, Jansen TL, Baughman RP. Practical eminence and experience-based recommendations for use of TNF-alpha inhibitors in sarcoidosis. Sarcoidosis Vasc Diffuse Lung Dis. 2014;31(2):91–107.PubMed
Metadaten
Titel
Clinical characteristics and organ system involvement in sarcoidosis: comparison of the University of Minnesota Cohort with other cohorts
verfasst von
Hok Sreng Te
David M. Perlman
Chetan Shenoy
Daniel J. Steinberger
Rebecca J. Cogswell
Henri Roukoz
Erik J. Peterson
Lin Zhang
Tadashi L. Allen
Maneesh Bhargava
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
BMC Pulmonary Medicine / Ausgabe 1/2020
Elektronische ISSN: 1471-2466
DOI
https://doi.org/10.1186/s12890-020-01191-x

Weitere Artikel der Ausgabe 1/2020

BMC Pulmonary Medicine 1/2020 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.