ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Treatment of pulmonary sarcoidosis refractory to initial therapy

Treatment of pulmonary sarcoidosis refractory to initial therapy
Literature review current through: Jan 2024.
This topic last updated: Nov 28, 2022.

INTRODUCTION — Sarcoidosis is a multisystem disease of unknown etiology characterized by tissue infiltration with noncaseating granulomas. The granulomas may occur in any organ, but the most frequently affected sites are the lungs, lymph nodes, skin, eyes, and liver. Spontaneous resolution of the disease is common, but progressive and disabling organ failure can occur in up to 10 percent of patients.

For patients with sarcoidosis who cannot tolerate or do not respond to initial treatments, other immunosuppressive or immunomodulatory agents may be of benefit [1], although no medications have been approved by the US Food and Drug Administration (FDA) for the treatment of sarcoidosis [2-7]. Organ transplantation has been performed successfully for end-stage hepatic, renal, cardiac, and pulmonary sarcoidosis. Unfortunately, limited data exist regarding the indications and efficacy of these approaches in the management of pulmonary sarcoidosis.

The mainstay of initial treatment for pulmonary sarcoidosis is oral glucocorticoid therapy, although patients with poor tolerance of glucocorticoids may use an alternative agent, typically methotrexate. The treatment of pulmonary sarcoidosis that cannot be managed with initial therapeutic approaches will be reviewed here. The diagnosis and initial treatment of pulmonary sarcoidosis, as well as management of sarcoidosis affecting other organs, are discussed separately.

(See "Clinical manifestations and diagnosis of sarcoidosis".)

(See "Treatment of pulmonary sarcoidosis: Initial approach".)

(See "Management and prognosis of cardiac sarcoidosis".)

(See "Neurologic sarcoidosis" and "Neurologic sarcoidosis", section on 'Treatment'.)

(See "Kidney disease in sarcoidosis".)

(See "Gastrointestinal, hepatic, pancreatic, and peritoneal sarcoidosis".)

(See "Sarcoid arthropathy", section on 'Treatment' and "Sarcoidosis of bone", section on 'Treatment' and "Sarcoid myopathy", section on 'Treatment'.)

(See "Cutaneous sarcoidosis: Management".)

APPROACH — Progressive and disabling respiratory impairment complicates pulmonary sarcoidosis despite initial therapy in up to 10 percent of patients. For these patients, we suggest addition of another immunosuppressive agent. Other patients may be intolerant of glucocorticoids and require glucocorticoid-sparing immunosuppressive therapy. Many of these patients may benefit from referral to a sarcoidosis center.

Typical reasons patients require alteration of therapy — Patients with sarcoidosis who may benefit from next line therapies generally have one or more of the following features (see "Treatment of pulmonary sarcoidosis: Initial approach"):

Progression of disease despite adequate initial therapy (eg, at least three months of either prednisone 10 to 20 mg daily or methotrexate 10 to 15 mg weekly) [8].

Intolerable side-effects of initial therapy. Typical glucocorticoid side effects include difficult to control diabetes mellitus, excessive weight gain, myopathy, osteoporosis. Typical methotrexate side effects include hepatic toxicity and leukopenia.

Need for a glucocorticoid-sparing agent in a patient who requires long-term glucocorticoid therapy, has been unable to taper below 10 mg/day, and has had adverse effects from chronic glucocorticoid therapy.

Pretreatment assessment for those with progressive disease — Prior to intensifying therapy for pulmonary sarcoidosis, we investigate possible reasons for lack of clinical improvement, such as noncompliance with initial treatment, comorbid disease, and end-stage fibrosis.

Noncompliance with glucocorticoid therapy is common, due to the numerous side effects of glucocorticoid therapy [6]. Sometimes patients will admit on questioning that they have been missing doses due to fear about the potential side effects. This is helpful information when assessing the likelihood of a response to another potentially toxic agent.

We also assess patients for comorbid diseases that may contribute to their symptoms, pulmonary function impairment, or radiographic abnormalities (eg, infection, heart failure, thromboembolic disease, pulmonary hypertension) [9,10]. Testing typically includes repeat pulmonary function tests (PFTs), a non-contrast high-resolution computed tomography (HRCT) scan, and often an echocardiogram. Dyspnea that is out of proportion to the spirometric findings or a diffusing capacity that is lower than expected based on the amount of interstitial disease on chest imaging might suggest pulmonary hypertension or thromboembolic disease. This is also a good time to ask the patient again about possible occupational exposure to beryllium or any exposures that could cause hypersensitivity pneumonitis. (See "Pulmonary hypertension due to lung disease and/or hypoxemia (group 3 pulmonary hypertension): Epidemiology, pathogenesis, and diagnostic evaluation in adults" and "Hypersensitivity pneumonitis (extrinsic allergic alveolitis): Clinical manifestations and diagnosis" and "Chronic beryllium disease (berylliosis)", section on 'Risk factors'.)

End-stage lung fibrosis is not responsive to immunosuppressive therapy, so it is important to determine whether a patient with apparently refractory disease has end-stage fibrosis or refractory but active inflammation. Chest imaging is not precise enough in most patients to exclude the presence of active disease in a severely scarred lung. On the other hand, PFT abnormalities that have been stable for a few years are more likely due to fibrosis than active inflammation, although the stability of PFT abnormalities does not entirely exclude reversibility. If it is uncertain whether the patient has end-stage fibrosis without active disease, one or two second-line immunosuppressive agents are often tried before deciding that the patient has stable fibrosis and is not in need of ongoing therapy. Patients with advanced lung disease may be candidates for lung transplantation. (See 'Lung transplantation' below.)

Agent selection — The agents that appear to have the greatest likelihood of benefit in pulmonary sarcoidosis with an acceptable side effect profile are methotrexate (MTX), azathioprine, leflunomide, and mycophenolate. Tumor necrosis factor-alpha (TNF-a) antagonists also show promise. Randomized trial data to guide selection among the agents are lacking. All of these agents carry substantial risk for toxicity, particularly myelosuppression, hepatotoxicity, and opportunistic infection. The specific toxicities and ways to reduce their impact should be carefully reviewed with the patient.

MTX is the most frequently used second-line agent, although it cannot be used in patients with underlying liver disease. Patients who fail or do not tolerate MTX are commonly transitioned to another second-line immunosuppressive agent. Occasionally, two agents are used in combination, such as MTX and leflunomide.

If none of the second-line agents, alone or in combination, is effective, the next step is often a TNF-a antagonist, usually infliximab or adalimumab. Data are insufficient to determine whether a second-line agent (eg, MTX) should be used during TNF-a antagonist therapy. Combination therapy (eg, TNF-a inhibitor with MTX) has been used in rheumatoid arthritis to improve efficacy and reduce the risk of development of antibodies to the TNF-a inhibitor, but it has not been formally evaluated in sarcoidosis. Combination therapy is also associated with increased risk of infection and possibly malignancy. (See "Treatment of rheumatoid arthritis in adults resistant to initial conventional synthetic (nonbiologic) DMARD therapy", section on 'Methotrexate plus TNF inhibitor' and "Tumor necrosis factor-alpha inhibitors: Induction of antibodies, autoantibodies, and autoimmune diseases".)

If a TNF-a antagonist is contraindicated (eg, known hepatitis or aspergillus infection) or unsuccessful, the next steps are less clear, but may include hydroxychloroquine or one of the other agents that has been successful in other types of immunologic lung disease but has not been formally studied in sarcoidosis, such as mycophenolate, cyclophosphamide, or rituximab. (See 'Investigational agents' below.)

Assessing response — The response to therapy with any of the agents described above is evaluated in the same manner as that described for initial therapy of pulmonary sarcoidosis and requires ongoing assessment of changes in symptoms (eg, dyspnea, cough, hemoptysis, chest pain, fatigue), PFTs, gas transfer, and radiographic abnormalities. (See "Treatment of pulmonary sarcoidosis: Initial approach", section on 'Assessing response to therapy'.)

The parameters of response are sometimes conflicting and clinical judgment is needed to determine whether changes reflect true worsening or true improvement. Generally, greater weight is attributed to changes in symptoms than to changes in other parameters.

In addition to assessing the response to therapy, patients are monitored for new, extrapulmonary manifestations of sarcoidosis on a regular basis. An approach to monitoring is presented separately. (See "Clinical manifestations and diagnosis of sarcoidosis", section on 'Ongoing monitoring'.)

Opportunistic infection risk — Opportunistic infectious complications (eg, pneumocystis, cryptococcosis, aspergillosis, mycobacterial) are rare in patients with sarcoidosis treated with MTX, azathioprine, leflunomide, and mycophenolate [9,11,12]. However, patients on a combination of glucocorticoids (≥20 mg daily) and another immunosuppressive agent (eg, MTX, azathioprine, leflunomide, TNF-a antagonist) do appear to be at some increased risk and clinicians need to be aware of this possibility. The types of infection associated with these therapies and the indications for preventive therapy are discussed separately. (See "Glucocorticoid effects on the immune system", section on 'Infection risk' and "Treatment and prevention of Pneumocystis pneumonia in patients without HIV", section on 'Indications' and "Tumor necrosis factor-alpha inhibitors: Bacterial, viral, and fungal infections".)

PATIENTS WHO PROGRESS ON OR DO NOT TOLERATE GLUCOCORTICOIDS

Preferred option: Methotrexate — Methotrexate (MTX), an antimetabolite with both immunosuppressive and antiinflammatory properties, is the most commonly used nonglucocorticoid immunosuppressive agent for sarcoidosis [13]. It is also used for the management of other chronic inflammatory or autoimmune disorders, such as rheumatoid arthritis, psoriasis, and Crohn disease. The mechanism of action of MTX and its use in these other diseases are discussed separately. (See "Use of methotrexate in the treatment of rheumatoid arthritis" and "Treatment of psoriasis in adults", section on 'Methotrexate'.)

Alternatives to methotrexate, for those who are not candidates, are discussed below. (See 'Patients who progress on or cannot use methotrexate' below.)

Pretreatment preparation and contraindications — Prior to initiating MTX, complete blood counts (CBC), albumin, aminotransferases, alkaline phosphatase, bilirubin, creatinine, and hepatitis B and C viral testing (ie, HBsAg, anti-HBc, anti-HCV) are obtained. Patients with evidence of underlying liver disease (eg, aminotransferase level above two times the upper limit of normal) or chronic infection with hepatitis B or C are not candidates for MTX therapy [14]. A creatinine clearance less than 30 mL/min is considered a contraindication for methotrexate in patients with rheumatoid arthritis [14]. (See "Use of methotrexate in the treatment of rheumatoid arthritis".)

In addition, ingestion of alcoholic beverages while using MTX is strongly discouraged, and patients who are unwilling to reduce their alcohol consumption to an occasional and minimal level are not treated with MTX.

MTX is contraindicated in pregnancy due to teratogenic effects. It should be discontinued at least three months prior to a planned pregnancy and should not be used during breastfeeding [13]. Women of childbearing age should use a reliable method of contraception while taking MTX. (See "Safety of rheumatic disease medication use during pregnancy and lactation", section on 'Methotrexate'.)

Dosage and administration — MTX can be administered either orally or intramuscularly. We typically begin with oral therapy at a dose of 5 to 7.5 mg weekly. The dose is gradually increased (eg, by increments of 2.5 mg every two weeks) until a dose of 10 to 15 mg per week is achieved [13,15,16]. We switch to intramuscular MTX when patients have refractory nausea or have not achieved a beneficial effect after three to six months of oral therapy at 15 mg per week. A trial of MTX therapy should last at least six months to allow adequate assessment of effectiveness. (See "Treatment of pulmonary sarcoidosis: Initial approach", section on 'Assessing response to therapy'.)

Folic acid at a dose of 1 mg per day or 5 mg weekly is routinely given to patients on chronic methotrexate therapy to reduce the incidence of myelosuppression [13]. A routine peripheral blood count is obtained every four to eight weeks [14].

The optimal frequency for assessing liver function tests during methotrexate therapy for sarcoidosis is unknown. Alanine aminotransferase (ALT) with or without aspartate aminotransferase (AST), creatinine, and a CBC should be obtained every three to six weeks until a stable dose is reached and then every one to three months [13].

If ALT/AST increases without other clear cause, the methotrexate dose should be decreased or the drug stopped [13].

Because hepatic toxicity may initially be clinically occult, some have advocated liver biopsy when the total dose exceeds one gram or after 18 to 24 months of therapy, even without signs of hepatic injury, or if 6 of 12 serum aminotransferase levels are abnormal in a single year. (See "Hepatotoxicity associated with chronic low-dose methotrexate for nonmalignant disease".)

Adverse effects — The most serious side effects of immunosuppressive MTX therapy are hepatic fibrosis (in up to 10 percent of cases when the dose exceeds 5 g), leukopenia, and interstitial pneumonitis, resulting in pulmonary fibrosis [6]. In a study of the long-term use of MTX for sarcoidosis, 50 patients completed two years of therapy; the major adverse effects requiring hospitalization were hepatic toxicity and leukopenia [17]. (See "Major side effects of low-dose methotrexate" and "Methotrexate-induced lung injury", section on 'Clinical manifestations' and "Hepatotoxicity associated with chronic low-dose methotrexate for nonmalignant disease".)

The differential diagnosis of abnormal liver function in patients with sarcoidosis receiving MTX is broad and includes drug toxicity and hepatic sarcoidosis. Liver biopsy is frequently required to establish a definitive diagnosis. In a series of 100 liver biopsies performed in 68 patients with sarcoidosis, 14 biopsies demonstrated significant changes related to MTX; however, evidence of sarcoidosis involving the liver was even more common, noted in 47 biopsies [18]. Toxic reactions to MTX eventually developed in 10 percent of patients treated for more than two years. (See "Evaluation of the adult patient with hepatic granuloma", section on 'Sarcoidosis'.)

MTX-induced interstitial pneumonitis may be difficult to distinguish from progressive interstitial lung changes secondary to sarcoidosis. Characteristic features of MTX-induced lung injury include nonproductive cough, dyspnea, and fever beginning within days to weeks after initiation of MTX therapy. Peripheral blood (but not pulmonary) eosinophilia, when present, is more likely due to MTX than sarcoidosis [19-21]. Typical radiographic findings of MTX-induced lung injury include diffuse or patchy ground glass opacities, centrilobular nodules, and increased reticular opacities; pleural effusion is seen in about 10 percent. Poorly formed granulomas may be seen on lung biopsy. Recovery usually occurs after withdrawal of the drug. (See "Methotrexate-induced lung injury".)

Other toxicities include nausea, alopecia, and skin rash [6]. MTX is known to be teratogenic and may transiently suppress gonadal function. (See "Safety of rheumatic disease medication use during pregnancy and lactation".)

Chronic MTX treatment has been associated with the development of lymphoproliferative disorders, some of which regress after discontinuation of MTX [22,23]. It is not known whether MTX had an etiologic role in the development of the lymphoproliferative disease, or whether the underlying inflammatory condition was the sole contributor. (See "Malignancy and rheumatic disorders", section on 'Antirheumatic medications and risk of malignancy'.)

Efficacy — A small randomized trial, several small series, and case reports suggest that MTX is effective in patients with sarcoidosis affecting the lungs, skin, eyes, and central nervous system [15,17,24-31]. As an example, 24 patients with recent-onset pulmonary sarcoidosis were randomly assigned to low-dose MTX plus glucocorticoids or glucocorticoids alone [15]. At 12 months, those in the MTX group had been able to taper prednisone to a lower dose than those without MTX. However, when the study was analyzed including all the patients who had dropped out, the benefit was no longer statistically significant.

Clinical experience suggests a response rate to methotrexate of 40 to 60 percent among patients with pulmonary sarcoidosis [6,17]. A systematic review did not find convincing evidence of benefit, but this was due to insufficient published data, rather than demonstrated lack of benefit [32].

A two-year, retrospective cohort study of 200 patients with sarcoidosis treated with methotrexate or azathioprine found that the daily prednisone dose decreased a mean of 6.32 mg/year, forced expiratory volume in one second (FEV1) increased 52 mL/year, vital capacity increased 95 mL/year, and diffusing capacity (DLCO) increased 1.23 percent/year [33]. No significant differences were noted between the two treatments except for a higher rate of infection with azathioprine (35 versus 18 percent).

PATIENTS WHO PROGRESS ON OR CANNOT USE METHOTREXATE — A variety of immunosuppressive agents have been used to treat pulmonary sarcoidosis that is insufficiently controlled with an acceptable dose of glucocorticoids. The evidence in support of individual second-line agents is largely observational [34,35].

Azathioprine — Azathioprine is used as second-line therapy for pulmonary sarcoidosis, generally as a supplement to glucocorticoids rather than as a single drug [2,30,36-42]. Due to the greater clinical experience with methotrexate (MTX), azathioprine is usually used in patients who have failed MTX due to side effects or lack of benefit. The exact mechanism by which azathioprine might affect sarcoidosis is not clear.

Pretreatment preparation – The toxicity of azathioprine is largely related to its metabolites and is strongly affected by the presence of genetic polymorphisms of the enzyme thiopurine-S-methyltransferase (TPMT) [43]. TPMT activity can be predicted by genotyping or by measurement of serum TPMT enzyme activity levels. Low TPMT enzyme activity levels are associated with increased toxicity. Pretreatment screening of patients for genetic polymorphisms of TPMT or reduced TPMT enzyme activity is controversial as patients with normal genotypes can still develop myelosuppression and hepatic toxicity. However, many clinicians perform one test or another before initiating azathioprine therapy. It is not known whether a high level of TPMT activity would identify patients who need a higher dose of azathioprine to achieve a therapeutic effect. (See "Pharmacology and side effects of azathioprine when used in rheumatic diseases", section on 'Pharmacogenetics and azathioprine toxicity' and "Thiopurines: Pretreatment testing and approach to therapeutic drug monitoring for adults with inflammatory bowel disease".)

Baseline complete blood counts and serum albumin, aminotransferases, and creatinine are obtained in all patients.

Azathioprine is a teratogen, so women of childbearing age should use a reliable method of contraception during azathioprine therapy. (See "Pharmacology and side effects of azathioprine when used in rheumatic diseases" and "Safety of rheumatic disease medication use during pregnancy and lactation", section on 'Azathioprine and 6-mercaptopurine'.)

Dosage and administration – The usual starting dose of azathioprine is 50 mg per day, given as a single daily oral dose; 25 mg per day may be used initially if TPMT is not assessed. To reduce the likelihood of gastrointestinal side effects, the dose is slowly increased by 50 mg every two to four weeks until the desired dose is reached. The typical maintenance dose is 2 mg/kg per day (up to a maximum of 200 mg/day), unless the patient has renal insufficiency or an adverse effect necessitating a lower dose. We reduce the azathioprine dose if the white blood cell count decreases to ≤4000/microL or the platelet count to ≤150,000/microL. A discernible response to therapy may not be evident until the patient has received three to six months of therapy. (See "Pharmacology and side effects of azathioprine when used in rheumatic diseases", section on 'Dose titration and monitoring'.)

A complete blood count (including hemoglobin, white blood cell count, and platelet count) should be monitored after the first two weeks, then every two weeks during dose escalation, and every four to eight weeks after a stable dose is achieved. There is less consensus on the frequency of liver enzyme testing. A reasonable protocol is to check serum aminotransferases every 8 to 12 weeks during the first several months of AZA therapy and then less often during chronic therapy if prior testing has been normal. (See "Pharmacology and side effects of azathioprine when used in rheumatic diseases", section on 'Pharmacogenetics and azathioprine toxicity' and "Thiopurines: Pretreatment testing and approach to therapeutic drug monitoring for adults with inflammatory bowel disease", section on 'Summary and recommendations'.)

Adverse effects – Gastrointestinal complaints (eg, nausea, vomiting, and diarrhea), rash, fever, and malaise are the most common side effects. Dyspeptic symptoms can be reduced by taking the medication with meals. Hematologic side effects include depression of all cell lines, which may be difficult to distinguish from the bone marrow suppression associated with sarcoidosis. A small percentage of patients demonstrate an increase in liver function tests, but reports of severe hepatitis are rare. Infection is a known complication of azathioprine therapy [33]. An increased risk of subsequent malignancy has been reported in renal transplant patients who are treated with both azathioprine and prednisone. (See "Pharmacology and side effects of azathioprine when used in rheumatic diseases", section on 'Adverse effects'.)

Efficacy – No randomized trials have examined the efficacy of azathioprine for pulmonary sarcoidosis. An open-label series examined the effect of azathioprine (2 mg/kg per day) combined with glucocorticoids in 11 patients with chronic or relapsing pulmonary sarcoidosis [40]. All of the patients experienced improvement in symptoms and/or pulmonary function tests. Therapy was continued for a mean of 20 months (range 8 to 26). Three patients, including the one who discontinued therapy after 8 months, sustained relapses at 8, 18, and 22 months. The remaining eight had stable remissions for at least 4 to 73 months after discontinuing therapy.

In a separate series, 7 of 10 patients with pulmonary sarcoidosis exhibited clinical improvement with azathioprine 150 mg per day without concomitant glucocorticoids [38]. In contrast, a retrospective case review found a benefit in only 2 of 10 patients [42].

As noted above, in a two-year cohort study, modest improvements were noted among patients taking either MTX (n = 145) or azathioprine (n = 55) without significant between group differences, except for a higher infection rate with azathioprine (35 versus 18 percent) [33]. (See 'Efficacy' above.)

Leflunomide — Leflunomide is an antimetabolite similar to MTX but with less gastrointestinal toxicity.

Pretreatment preparation – Prior to initiating leflunomide therapy, complete blood counts, albumin, aminotransferases, creatinine, and hepatitis B and C viral testing are obtained [14]. Patients with evidence of underlying liver disease (eg, aminotransferase level above two times the upper limit of normal) or chronic infection with hepatitis B or C are not candidates for leflunomide therapy.

Alcohol avoidance is recommended while taking leflunomide.

Both females and males (and/or their female partners) of childbearing age should use a reliable method of contraception during leflunomide therapy and for up to two years afterwards, unless they follow a cholestyramine clearance regimen. (See "Safety of rheumatic disease medication use during pregnancy and lactation", section on 'Leflunomide'.)

Dosage and administration – The typical initial leflunomide dose is 20 mg per day, without a loading dose. We do not use a loading dose as a greater incidence of side effects was noted when a loading dose of 100 mg per day for three days was used in the treatment of rheumatoid arthritis. Alternatively, leflunomide can be initiated at 10 mg per day and increased to 20 mg, if the patient is tolerating the medication but has not achieved control of the disease manifestations [44]. (See "Pharmacology, dosing, and adverse effects of leflunomide in the treatment of rheumatoid arthritis", section on 'Pharmacokinetics, drug elimination, and genetic factors'.)

Extrapolating from the experience with rheumatoid arthritis, it may take 6 to 12 weeks for any improvement in lung function to occur.

Adverse effects – The most common adverse effects of leflunomide are nausea, diarrhea, abdominal pain, hepatotoxicity, rash, and peripheral neuropathy [45,46]. This risk of hepatotoxicity appears higher among patients with underlying liver disease (eg, alcohol abuse, viral hepatitis, autoimmune liver disease) who are taking another hepatotoxic agent. The frequency of adverse effects may be influenced by the patient's genotype at the locus for cytochrome P450, CYP1A2, or for dihydroorotate dehydrogenase, an enzyme inhibited by leflunomide [47,48]. However, testing for these genotypes is not clinically available. (See "Pharmacology, dosing, and adverse effects of leflunomide in the treatment of rheumatoid arthritis", section on 'Adverse effects'.)

The American College of Rheumatology suggests that patients taking leflunomide undergo monitoring of serum albumin and serum aspartate and alanine aminotransferase (AST and ALT) levels every two to four weeks for the first three months, followed by monitoring every 8 to 12 weeks for the next three months, and then every 12 weeks thereafter [14]. Alcohol avoidance is recommended during leflunomide therapy. Due to the long half-life of leflunomide, if the ALT increases to two times the upper limit of normal or higher, leflunomide should be stopped and active removal with cholestyramine initiated. (See "Pharmacology, dosing, and adverse effects of leflunomide in the treatment of rheumatoid arthritis", section on 'Warnings and monitoring for liver disease'.)

Efficacy – Evidence in favor of using leflunomide for pulmonary sarcoidosis is limited to case series and to indirect evidence of benefit in a few cases of extrapulmonary sarcoidosis [44,49,50]. In a retrospective case series, 32 patients received leflunomide with or without concomitant MTX for pulmonary or ocular sarcoidosis [44]. Complete or partial response was seen in 12 of 17 patients who received leflunomide alone. Three patients discontinued leflunomide due to nausea, but no other significant side effects were reported. The combination of leflunomide with MTX resulted in a partial or complete response in 13 of 15 patients. In a case report, a patient with pulmonary, sinus, and cutaneous sarcoidosis that was refractory to azathioprine and hydroxychloroquine responded to leflunomide 20 mg per day [50]. He had been intolerant of MTX, but tolerated leflunomide without side effects.

Mycophenolate mofetil — Mycophenolate mofetil (MMF), an inhibitor of lymphocyte proliferation and activity, has been used to treat a variety of interstitial lung diseases associated with rheumatic disease. However, data regarding the use of MMF in sarcoidosis are limited, and its role in pulmonary sarcoid requires further study.

Selection of the initial dose and monitoring of MMF are described separately. (See "Mycophenolate: Overview of use and adverse effects in the treatment of rheumatic diseases", section on 'Mycophenolate dose and administration'.)

Evidence in favor of a benefit to mycophenolate includes the following studies [51-53]:

In a retrospective review of 37 patients with pulmonary sarcoidosis who had failed to respond to a prior immunosuppressive agent or had experienced an adverse event necessitating discontinuation of the agent, addition of MMF was associated with a decrement in prednisone dosing (5.6 mg over one year) [51]. MMF was not associated with improvement in pulmonary function in the group as a whole. In a subgroup analysis, a trend towards improvement in forced vital capacity (FVC) and diffusing capacity for carbon monoxide (DLCO) was noted among patients who were given MMF due to intolerance of a prior immunosuppressive agent, rather than due to therapeutic failure.

In a case series, MMF was added to prednisone in 10 patients with biopsy-proven pulmonary sarcoidosis for a median of 31 months; plasma trough levels of MMF were 1 to 3 mg/dL [52]. Over the course of therapy, symptoms, radiographic abnormalities, and pulmonary function improved in four patients and remained stable in six. The mean improvement in FVC was 8.5 percent (range -2 to 16 percent). Concomitant prednisone was reduced slightly from a mean of 14 to 6 mg/day.

Benefit from MMF has been reported in case series of patients with cutaneous, central nervous system, and renal sarcoidosis [54-57]. In a case report, cutaneous and bone marrow sarcoidosis that were refractory to glucocorticoids and MMF responded to a combination of MMF and methotrexate [58]. (See "Interstitial lung disease in dermatomyositis and polymyositis: Treatment", section on 'Mycophenolate mofetil' and "Neurologic sarcoidosis", section on 'Other immunomodulatory therapies'.)

MMF treatment requires careful dosing and monitoring to avoid toxicity. Neutropenia is less of a problem with MMF than with other immunosuppressive agents, but may occur. Nausea and diarrhea may be dose limiting. The dosing, adverse effects, and monitoring of MMF therapy are discussed in greater detail separately. (See "Mycophenolate: Overview of use and adverse effects in the treatment of rheumatic diseases".)

Combination therapy — For patients who have progressive pulmonary sarcoidosis that is refractory to one or two of these second-line immunosuppressive agents taken individually, a common practice is to combine two of the agents, such as methotrexate and leflunomide [59]. As an example, in an open label study, 15 of 17 patients with sarcoidosis affecting various organs responded to a combination of MTX and leflunomide [44]. If the patient’s condition worsens or toxic effects develop, the clinician may introduce a third-choice therapeutic option, as described below [60].

PATIENTS WITH DISEASE REFRACTORY TO THE ABOVE AGENTS — For patients whose disease is refractory to the above agents or who are unable to tolerate them at a therapeutic dose, the next step is usually a tumor necrosis factor-alpha (TNF-a) antagonist, although sarcoidosis is not a US Food and Drug Administration (FDA) approved indication for these agents. Other alternatives include entering a clinical trial or using an investigational agent as described below. (See 'Investigational agents' below.)

Tumor necrosis factor-alpha antagonists — For patients whose disease is refractory to the above agents, the next step is typically a TNF-a antagonist. The cytokine TNF-a is thought to accelerate the inflammatory process in sarcoidosis via its role in maintenance of granuloma formation. Thus, using agents that block the effect of TNF-a may be beneficial in treating sarcoidosis. Due to the potential toxicity of these agents, we reserve them for patients who have persistent disease and have failed treatment with glucocorticoids (eg, prednisone ≥15 mg/day) AND at least one second-line immunosuppressive agent (eg, methotrexate [MTX], azathioprine, leflunomide) [5,61,62].

Efficacy of individual agents — Studies of the efficacy of TNF-a antagonists in the treatment of sarcoidosis have yielded mixed results.

Infliximab (ie, Remicade) is a chimeric, humanized monoclonal antibody that neutralizes TNF-a. It has been studied in patients with pulmonary and extrapulmonary sarcoidosis refractory to glucocorticoid therapy [63-69]. In a randomized trial, 138 patients with chronic pulmonary sarcoidosis were assigned to receive infusions of placebo, low-dose infliximab (3 mg/kg), or higher-dose infliximab (5 mg/kg) at baseline and weeks 2, 6, 12, 18, and 24 [67]. At evaluations performed at 24 and 52 weeks, low-dose infliximab increased the percent of predicted forced vital capacity (FVC) compared with baseline, whereas placebo did not. The proportion of patients that reported adverse events was similar among the treatment groups; the most common adverse events were upper respiratory infection, cough, dyspnea, and bronchitis.

The infliximab trial described above has been criticized for its small and clinically insignificant effect size, although it did identify subgroups that may derive greater benefit [67,70]. As an example, 92 patients with extrapulmonary sarcoidosis that was refractory to chronic glucocorticoids were randomly assigned to take infliximab or placebo for 24 weeks [71]. A modest improvement was noted in the infliximab group, although the improvement was not maintained during the 24-month follow-up period. Long-term infliximab treatment has not been well studied. Patients with predominantly extrapulmonary sarcoidosis appear to receive the most benefit from long-term treatment [69].

Preliminary evidence suggests that a subset of patients with sarcoidosis who have peripheral blood CD4+ T cell lymphopenia and resistance to conventional immunosuppressants may be more likely to respond to infliximab [72]. Among five consecutive patients with this phenotype, the CD4+ lymphopenia corrected with infliximab and all reported improvement in at least one clinical disease manifestation of sarcoidosis. Additional studies regarding the cause of the CD4+ lymphopenia in these patients and the use of CD4+ lymphopenia as a biomarker for response to infliximab are needed.

Adalimumab (ie, Humira) is a fully human anti-TNF-a antibody. A few case reports and small case series have described improvement in extrapulmonary sarcoidosis with adalimumab [73-77]. In addition, among 18 patients who switched from infliximab to adalimumab due to intolerance of infliximab (allergic reaction with or without antibodies) or disease progression, seven patients showed improved forced vital capacity, six patients remained stable, and five deteriorated [78]. One patient developed a lupus-like syndrome, three had severe infections, and seven had mild infections.

Etanercept (ie, Enbrel) is a soluble TNF-a receptor fusion protein that binds TNF-a and has a longer half-life than the native soluble receptor. It was assessed in a preliminary clinical trial of patients with progressive stage II or III sarcoidosis. The trial was stopped after the enrollment of 17 patients, when interval assessment noted treatment failure in 11 participants [79]. Treatment failure was defined as progressive clinical deterioration, the need for other immunosuppressive medications, or the development of intolerable side effects. In a separate trial, etanercept was not shown to improve chronic ocular disease [80]. On the basis of these results, larger trials of this agent for progressive pulmonary sarcoidosis do not appear indicated.

Pretreatment preparation — Therapy with TNF-a antagonists has been associated with reactivation of a variety of latent infections including tuberculosis and hepatitis B and C, although the latter occurs less often. (See "Tumor necrosis factor-alpha inhibitors: Bacterial, viral, and fungal infections".)

Prior to initiating therapy with a TNF-a antagonist, patients are asked about risk factors for tuberculosis infection and undergo a tuberculin skin test (PPD) or a peripheral blood interferon release assay. For patients who have been previously vaccinated with Bacille Calmette-Guérin (BCG), the interferon release assay can help to differentiate between tuberculosis infection and BCG vaccination. (See "Risk of mycobacterial infection associated with biologic agents and JAK inhibitors", section on 'Screening' and "Use of interferon-gamma release assays for diagnosis of tuberculosis infection (tuberculosis screening) in adults".)

Flares of hepatitis B have been documented with infliximab therapy; progression of hepatitis C has rarely been reported with TNF-a antagonists. We ask patients about risk factors for hepatitis viral infection and obtain serologic testing for hepatitis B and C. Serologic testing should include assessment of HBsAg, anti-HBc, and anti-HCV. If the HBsAg test is positive, further testing should include HBeAg, anti-HBe, and an HBV DNA level. (See "Tumor necrosis factor-alpha inhibitors: An overview of adverse effects", section on 'Hepatotoxicity' and "Tumor necrosis factor-alpha inhibitors: Bacterial, viral, and fungal infections", section on 'Viral infections'.)

Dosage and administration — Infliximab is administered by intravenous infusion 3 to 5 mg/kg at weeks 0, 2, 6, and 12 [66-68,72]. The optimal frequency for subsequent dosing is not known. Among patients with rheumatoid arthritis and Crohn disease, scheduled maintenance therapy (eg, every six to eight weeks) appears more effective than no maintenance therapy. The optimal duration of a therapeutic trial of infliximab in pulmonary sarcoidosis is unknown, but a six-month trial was better than a three-month trial in a group of 244 patients with rheumatoid arthritis [81]. (See "Treatment of Crohn disease in adults: Dosing and monitoring of tumor necrosis factor-alpha inhibitors" and "Treatment of rheumatoid arthritis in adults resistant to initial conventional synthetic (nonbiologic) DMARD therapy", section on 'Methotrexate plus TNF inhibitor'.)

Adalimumab is administered by subcutaneous injection. The optimal dose in pulmonary sarcoidosis is not known, but a dose of 40 mg every week or every other week has been used in case reports [73-75,82]. In other inflammatory conditions, the usual dose is 40 mg every other week; sometimes an initial dose of 80 to 160 mg (given as 4 injections over 24 hours) is used.

Combination with other agents — The combination of infliximab (or another TNF-a inhibitor) with MTX or azathioprine appears to inhibit antibody formation to the monoclonal antibody and to improve disease control among patients with rheumatoid arthritis or Crohn disease. However, the risks of serious infection and malignancy (lymphoma) in these diseases appear greater with combination therapy than with infliximab alone. Data for sarcoidosis are insufficient to make a clear recommendation between these choices. However, we typically initiate infliximab or adalimumab in combination with a low dose of glucocorticoid or low-dose MTX to prevent antibody formation. (See "Treatment of Crohn disease in adults: Dosing and monitoring of tumor necrosis factor-alpha inhibitors" and "Treatment of rheumatoid arthritis in adults resistant to initial conventional synthetic (nonbiologic) DMARD therapy", section on 'Methotrexate plus TNF inhibitor'.)

Adverse effects — The major adverse effects associated with the TNF-a antagonists are increased susceptibility to infection, particularly mycobacterial and invasive fungal infections, and infusion-reactions. Additional side effects reported in separate studies (in one patient each) include alopecia, oral candidiasis, cellulitis, pneumonia, visual field defect, fatal pulmonary embolism, and development of a hypercoagulable state associated with circulating anticardiolipin antibodies [63-66,68]. (See "Tumor necrosis factor-alpha inhibitors: An overview of adverse effects" and "Tumor necrosis factor-alpha inhibitors: Bacterial, viral, and fungal infections" and "Risk of mycobacterial infection associated with biologic agents and JAK inhibitors" and "Tumor necrosis factor-alpha inhibitors: Risk of malignancy".)

The development of noncaseating granulomas consistent with sarcoidosis has been reported during anti-TNF-a therapy for other diseases [83-86]. Granulomas have occurred in the skin, lungs, and lymph nodes. The sarcoid-like disease manifestations have typically resolved with cessation of anti-TNF-a therapy, although some patients have also received glucocorticoid therapy.

INVESTIGATIONAL AGENTS — Several medications have been proposed for use in sarcoidosis based on their mechanism of action, but are not commonly used for pulmonary sarcoidosis due lack of adequate data to support their use or their side effect profile.

Clinical trials — Information about clinical trials of therapies for pulmonary sarcoidosis can be found at Clinicaltrials.gov.

Cyclophosphamide — Cyclophosphamide is used rarely as a glucocorticoid-sparing agent in the treatment of sarcoidosis [87,88]. Due to its toxicity profile, it is reserved for use as a "third-line" drug in patients whose condition is deteriorating despite therapy with glucocorticoids and one of the more commonly used immunosuppressive agents. (See 'Patients who progress on or cannot use methotrexate' above.)

Mechanism of actionCyclophosphamide is an alkylating agent of the nitrogen mustard group. It is an inactive drug that is metabolized by the cytochrome P-450 system into active metabolites. These metabolites decrease lymphocyte numbers and function and may also have antiinflammatory effects. (See "General toxicity of cyclophosphamide in rheumatic diseases".)

Dosage and administrationCyclophosphamide is generally given orally as a single daily dose. We usually begin at a dose of 25 to 50 mg/day and increase gradually, by 25 mg increments, aiming to reduce and maintain the white blood cell count (WBC) to between 4000 and 7000/mm3. The WBC count is measured biweekly for the first 6 to 12 weeks and then at least monthly thereafter. We do not exceed a dose of 150 mg/day, even if the leukocyte count remains about 7000/m3, as it is our clinical impression that this dose is associated with a lower rate of side effects without loss of clinical efficacy. There is little experience in sarcoidosis with the intravenous administration of intermittent ("pulse") therapy with cyclophosphamide. It has been used occasionally in patients with refractory or progressive disease. The usual dose is 500 to 1000 mg given intravenously over 30 to 60 minutes once every two or four weeks; the exact dose is calculated based on the patient's age, creatinine clearance, and peripheral leukocyte counts during treatment. Intravenous administration is discussed in greater detail separately. (See "General principles of the use of cyclophosphamide in rheumatic diseases".)

We do not expect to achieve a favorable response to cyclophosphamide for at least three to six months after initiating therapy, either with or without accompanying low doses of glucocorticoids.

Adverse effects – Reductions in all hematologic cell lines can be seen, and these require an adjustment in dosage. As discussed above, we adjust the cyclophosphamide dose to maintain the WBC count between 4000 and 7000/mm3. Other toxicities include increased risk of infection, hemorrhagic cystitis, stomatitis, nausea, diarrhea, hepatotoxicity (rarely), and carcinoma of the bladder. Strategies to reduce the risk of hemorrhagic cystitis include advising the patient to drink eight or more glasses (eight ounces each) of water daily to create a forced diuresis and monthly monitoring of the urine for red blood cells or other abnormalities. (See "General toxicity of cyclophosphamide in rheumatic diseases".)

Infertility may occur in both males and females. Cyclophosphamide may be teratogenic, so women of childbearing age should use a reliable method of contraception. (See "Safety of rheumatic disease medication use during pregnancy and lactation", section on 'Cyclophosphamide'.)

Tocilizumab — Tocilizumab is a humanized interleukin (IL)-6 receptor antibody approved for use in rheumatoid arthritis. In a case series of four patients with pulmonary sarcoidosis refractory to prednisone and other immunosuppressive agents, tocilizumab was associated with improvement in symptoms and, in some cases, pulmonary function tests [89]. Of note, development of sarcoidosis (mediastinal and hilar lymphadenopathy and cutaneous nodules) has been reported in patients with rheumatoid arthritis being treated with tocilizumab [90,91].

Golimumab and ustekinumab — Golimumab is a humanized tumor necrosis factor-alpha (TNF-a) antibody that has been found effective for rheumatoid arthritis; ustekinumab is a monoclonal antibody to interleukin (IL)-12/IL-23 that is used in psoriasis. In a randomized trial, 132 patients with lung sarcoidosis were assigned to golimumab, ustekinumab, or placebo for 24 weeks [92]. At week 16, the change from baseline in percent predicted forced vital capacity was no different in the active treatment groups compared with placebo.

Rituximab — Rituximab is a monoclonal antibody that targets CD-20 on B lymphocytes, leading to B lymphocyte depletion. As sarcoidosis is associated with hypergammaglobulinemia, it is hypothesized that a B lymphocyte-directed therapy may be of benefit. Several reports describe treatment of refractory sarcoidosis affecting the lungs, heart, and eyes with variable results [93-96]. Further study is needed to determine whether rituximab is beneficial in refractory sarcoidosis.

Hydroxychloroquine and chloroquine — Hydroxychloroquine and chloroquine are antimalarial drugs with immunomodulating properties. Clinical experience with these agents for pulmonary sarcoidosis is limited, and their relative efficacy versus other therapies has not been assessed. Due to the limited benefit and concerns about ocular toxicity, we reserve these medications for circumstances where other options have failed or are contraindicated. Hydroxychloroquine is preferred over chloroquine due to a more favorable adverse effect profile.  

EfficacyChloroquine has been used most often to treat cutaneous sarcoidosis [97,98], but several reports have described its use in patients with pulmonary disease [35,99-105]. (See "Cutaneous sarcoidosis: Management", section on 'Antimalarials'.)

As an example, one trial treated 23 symptomatic patients with biopsy-proven pulmonary sarcoidosis for six months with chloroquine, and then randomized patients to either maintenance therapy with chloroquine or observation [105]. Small but statistically significant advantages in forced expiratory volume in one second (FEV1) and relapse rate were observed in the group maintained on chloroquine during a mean follow-up of 20 months. In cutaneous sarcoidosis, chloroquine appears to have a higher response rate than hydroxychloroquine, although hydroxychloroquine is often preferred due to lower ocular toxicity [106]. (See "Cutaneous sarcoidosis: Management", section on 'Antimalarials'.)

Dosage and administration – Glucose-6-phosphate dehydrogenase (G6PD) levels must be determined prior to initiating therapy; these agents cannot be given if the patient is G6PD deficient. For patients with a normal G6PD level, the dose of hydroxychloroquine is 5 mg/kg real body weight with the dose not to exceed 400 mg/day; the dose of chloroquine is approximately 250 mg/day (maximum daily dose ≤2.3 mg/kg real body weight). Higher doses have been used based upon an observational study [105], but are now avoided due to the risk of retinopathy associated with higher doses. All patients should undergo a baseline eye examination; the primary tests used in follow-up to screen for retinopathy are automated visual fields plus spectral-domain coherence tomography (SD OCT). (See "Diagnosis and management of glucose-6-phosphate dehydrogenase (G6PD) deficiency" and "Antimalarial drugs in the treatment of rheumatic disease", section on 'Routine eye examinations'.)

Adverse effects – The major adverse effects of the antimalarial agents are irreversible retinopathy and blindness. Hydroxychloroquine is associated with a lower rate of retinal toxicity than chloroquine even when the dose is adjusted for lean body weight [107]. Thus, eye examination by an ophthalmologist is required at baseline and every 6 to 12 months during therapy to avoid drug-related toxic retinitis and corneal lesions [6]. Alternate day treatment minimizes the risk of ocular toxicity.

Nausea is a frequent dose-limiting problem.

Antimycobacterial therapy — Data suggestive of a role for an unidentified mycobacterium as a cause of some cases of sarcoidosis led to a study of combination levofloxacin, ethambutol, azithromycin, and rifampin for pulmonary sarcoidosis [108]. Improvement in forced vital capacity (FVC) was reported for patients able to complete the eight weeks of therapy. Approximately half of patients were not able to complete the entire the eight-week protocol mainly because of adverse events, including leucopenia, arthralgias, insomnia, and rash.

LUNG TRANSPLANTATION — For patients with advanced pulmonary fibrosis due to pulmonary sarcoidosis and associated pulmonary hypertension, lung transplantation may offer the only hope for long-term survival [10,109,110]. Patients considered for lung transplantation usually have stage IV radiographic disease (ie, advanced fibrotic changes, honeycombing, hilar retraction, and cystic change) and are New York Heart Association III or IV. Survival rates following lung transplantation for pulmonary sarcoidosis are comparable to those for other indications [111-114]. (See "Lung transplantation: An overview".)

Data are limited regarding the optimal timing of lung transplantation for sarcoidosis. Parameters that would favor a lung transplant evaluation include a forced vital capacity (FVC) less than 1.5 L (or less than 50 percent of predicted) and the development of oxygen dependence or pulmonary hypertension due to sarcoidosis, as these features are associated with increased one-year mortality in the absence of transplantation [115,116]. (See "Lung transplantation: General guidelines for recipient selection".)

Bilateral lung transplantation appears to be associated with slightly better survival than single lung transplantation [114]. When deciding between a single or bilateral lung transplantation, the presence of a mycetoma or significant bronchiectasis may necessitate a bilateral lung procedure [111]. In addition, patients with mycetomas may require specific, individualized antifungal therapy [117,118]. The extent of extrapulmonary sarcoidosis must be carefully assessed prior to surgery. In particular, cardiac involvement may indicate a greater risk from lung transplantation alone or the need for a simultaneous heart-lung transplantation. Heart-lung transplantation increases the risk for short-term posttransplant mortality, although long-term outcomes are comparable to double lung transplantation [111,119]. (See "Lung transplantation: General guidelines for recipient selection", section on 'Cystic fibrosis and bronchiectasis' and "Management and prognosis of cardiac sarcoidosis", section on 'Approach to management'.)

Following lung transplantation, asymptomatic foci of noncaseating granulomas, suggestive of recurrent disease, have been identified in the allografts of sarcoidosis patients [120-124]. However, clinically significant organ dysfunction due to recurrent sarcoidosis is rare, and evidence of recurrence usually disappears within three months, without associated clinical sequelae during the follow-up period [109,110,113,122].

The incidence of acute rejection and bronchiolitis obliterans syndrome appear similar after transplantation among patients with sarcoidosis, idiopathic pulmonary fibrosis, and chronic obstructive pulmonary disease, despite observations to the contrary in the older literature [113,121,122,124,125].

AGENTS WITHOUT CLEAR BENEFIT — A number of agents have substantial toxicity and/or insufficient evidence of benefit for pulmonary sarcoidosis and are consequently avoided. These agents include colchicine, chlorambucil, cyclosporine, nonsteroidal antiinflammatory agents, tetracyclines, and thalidomide.

Colchicine – Colchicine has multiple effects, including the arrest of cell division, inhibition of granulocyte migration, and inhibition of the release of several proteins from cells. It may also interfere with the secretion of collagen from fibroblasts and may increase collagen degradation by enhancing the action of collagenase. Colchicine may be useful for sarcoid arthritis (based on case series) but is not used for pulmonary sarcoidosis.

Side effects of colchicine include nausea, vomiting, abdominal pain, and diarrhea.

Chlorambucil – Chlorambucil is an alkylating agent that is rarely used for the treatment of refractory pulmonary sarcoidosis [36,126-129]. Clinical experience is limited, and the relative efficacy of chlorambucil versus other therapies has not been assessed. Due to the limited benefit and substantial toxicity, we avoid this medication except in circumstances where other options have failed or are contraindicated. (See "General toxicity of cyclophosphamide in rheumatic diseases".)

The evidence in favor of using chlorambucil for sarcoidosis is limited. In one report, 10 patients with progressive disease were treated with chlorambucil alone or in combination with glucocorticoids [126]. Eight had some degree of improvement that may have been attributable to chlorambucil. The responsive patients showed beneficial effects within three months of starting chlorambucil therapy.

Bone marrow function usually recovers rapidly when the drug is discontinued, but irreversible marrow failure has also been reported. An alarming rate of oncogenesis (leukemia and other tumors) limits its potential use in nonneoplastic diseases. Other side effects include nausea, vomiting, hepatotoxicity, dermatitis, and infertility.

Cyclosporine – Cyclosporine, a fungus-derived cyclic peptide that suppresses cell-mediated and humoral immune responses, is primarily used for the prevention of allograft rejection. In addition, it may be effective in T-cell mediated disorders such as uveitis, psoriasis, rheumatoid arthritis, and primary biliary cholangitis. The use of cyclosporine in the treatment of pulmonary sarcoidosis remains experimental, due to limited clinical experience and known toxicities [130-134].

The limited clinical experience with cyclosporine for pulmonary sarcoidosis has not demonstrated significant improvement in control of the disease. As an example, one study randomly assigned 37 patients with biopsy-proven sarcoidosis and deteriorating respiratory function to treatment with prednisone with or without concomitant, open-label cyclosporine, titrated to trough levels of 100 to 200 ng/mL [134]. Combined outcome measures did not differ significantly between the two treatment groups at 3, 9, or 18 months. It is possible that the small trials in which its use has been studied have had insufficient power to detect beneficial effects of cyclosporine treatment or that only selected subgroups of patients will derive a benefit.

The major adverse reactions are renal dysfunction, tremor, hirsutism, hypertension, and gum hyperplasia [134]. There is apparently no myelotoxicity associated with the therapeutic use of cyclosporine. Many drugs interfere with the metabolism of cyclosporine. (See "Cyclosporine and tacrolimus nephrotoxicity" and "Pharmacology of cyclosporine and tacrolimus", section on 'Side effects'.)

Nonsteroidal antiinflammatory agentsIbuprofen, indomethacin, or salicylates may help reduce acute inflammatory or constitutional symptoms of sarcoidosis (eg, arthritis and fever) but are not recommended for the treatment of pulmonary sarcoidosis.

Tetracyclines – Antibiotics in the tetracycline family (eg, doxycycline, minocycline) have been used to treat cutaneous sarcoidosis with variable effect [135]. There is insufficient evidence of benefit to use these agents for the treatment of pulmonary sarcoidosis. (See "Cutaneous manifestations of sarcoidosis".)

Thalidomide – Thalidomide has anti-tumor necrosis factor-alpha (TNF-a) activity and has been used to treat cutaneous sarcoidosis in a few patients [136,137]. However, it does not appear to be of benefit in pulmonary sarcoidosis [138]. In an open label study, 10 patients with pulmonary sarcoidosis dependent on glucocorticoids received thalidomide 200 mg/day for 24 weeks [138]. No significant improvement in dyspnea, pulmonary function, or quality of life occurred. A dose reduction was necessary in 9 of 10 patients due to side effects such as excess somnolence, constipation, and peripheral neuropathy.

Pentoxifylline – Pentoxifylline is a xanthine that has hemorheologic activity (improves blood flow), leading to its use in the treatment of vascular insufficiency. In addition, pentoxifylline inhibits the synthesis of TNF-a by alveolar macrophages from patients with sarcoidosis, potentially inhibiting the formation and maintenance of granulomas [139,140]. As a result, it has been postulated that pentoxifylline might have a beneficial clinical effect in patients with sarcoidosis; however, its use for this indication remains experimental.

In a preliminary study, 23 untreated patients with active sarcoidosis were treated with pentoxifylline; improvement was noted in 11 patients, while 7 patients remained stable and none deteriorated [141]. Three patients discontinued therapy because of gastrointestinal side effects. Three patients whose sarcoidosis was refractory to glucocorticoids had improvement following addition of pentoxifylline to the glucocorticoid regimen. In a separate trial, 27 patients with pulmonary sarcoidosis were randomly assigned to pentoxifylline with prednisone or prednisone alone; no benefit was found in pulmonary function parameters or dyspnea, although a post hoc analysis found a diminution in flares of sarcoidosis and in the prednisone usage with pentoxifylline [142].

The primary side effects of pentoxifylline are gastrointestinal, and are similar to those of other xanthine agents [141,142].

Endothelin receptor antagonists – It has been proposed that the antifibrotic effect of the endothelin receptor antagonists might be beneficial in interstitial lung diseases with a fibrotic component. A trial to assess the efficacy of the endothelin receptor antagonist bosentan in patients with progressive pulmonary sarcoidosis with and without pulmonary hypertension was terminated due to difficulties with recruitment. A separate study of bosentan for sarcoidosis associated pulmonary hypertension is ongoing. (See "Pulmonary hypertension due to lung disease and/or hypoxemia (group 3 pulmonary hypertension): Treatment and prognosis".)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Sarcoidosis".)

PATIENT INFORMATION — UpToDate offers two types of patient education materials, “The Basics” and “Beyond the Basics.” The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on “patient info” and the keyword(s) of interest.)

Basics topics (see "Patient education: Sarcoidosis (The Basics)")

Beyond the Basics topics (see "Patient education: Sarcoidosis (Beyond the Basics)")

SUMMARY AND RECOMMENDATIONS

Indications for altering therapy – The majority of patients with pulmonary sarcoidosis either do not require treatment at all, or their disease responds well to systemic glucocorticoid therapy. However, progressive and disabling organ failure despite initial therapy can occur in up to 10 percent of patients, and some patients do not tolerate initial immunosuppressant regimens. For these patients, changes in treatment approach are needed. (See 'Introduction' above and 'Typical reasons patients require alteration of therapy' above.)

Pretreatment assessment Prior to intensifying therapy for progression of pulmonary sarcoidosis, we investigate possible reasons for lack of clinical improvement during initial therapy, such as noncompliance, comorbid disease, and end-stage fibrosis. (See 'Pretreatment assessment for those with progressive disease' above.)

Patients who progress on or do not tolerate glucocorticoids

For patients with pulmonary sarcoidosis refractory to systemic glucocorticoids, we suggest the addition of methotrexate (Grade 2C). In most instances, we continue low-dose glucocorticoids (0.25 mg/kg per day, usually less than 20 mg per day). (See 'Patients who progress on or do not tolerate glucocorticoids' above.)

For patients with active pulmonary sarcoidosis who need to discontinue glucocorticoids due to adverse effects, we suggest substitution with methotrexate (Grade 2C). (See 'Patients who progress on or do not tolerate glucocorticoids' above.)

Patients who progress on or are not candidates for methotrexateFor patients who progress on or cannot use methotrexate, we suggest azathioprine, leflunomide, or mycophenolate rather than other options (Grade 2C). The choice of therapy should include thiopurine methyltransferase (TPMT) level and other individual patient factors associated with potential toxicities of these agents. (See 'Patients who progress on or cannot use methotrexate' above.)

Later line therapy

For patients who do not respond to a trial of one or two of the second-line immunosuppressive agents given alone or in combination with glucocorticoids, the next step is either to combine two of the second-line immunosuppressive agents or to add a tumor necrosis factor-alpha (TNF-a) antagonist (eg, infliximab or adalimumab). The choice is usually guided by the individual patient's history of medication-related adverse effects, clinician experience with the agents, and patient preference. (See 'Combination therapy' above and 'Tumor necrosis factor-alpha antagonists' above.)

Several additional immunosuppressants have been used to treat sarcoidosis refractory to other agents, but clinical experience is limited for several (eg, rituximab, golimumab, ustekinumab, tocilizumab, antimycobacterial agents) and others are associated with significant adverse effects (eg, cyclophosphamide, hydroxychloroquine, chloroquine). (See 'Investigational agents' above.)

Patients with advanced pulmonary fibrosis or severe pulmonary hypertension associated with sarcoidosis may be appropriate candidates for lung transplantation. We refer patients with stage IV radiographic disease (table 1) or World Health Organization class III or IV pulmonary hypertension (table 2). (See 'Lung transplantation' above.)

  1. Baughman RP, Grutters JC. New treatment strategies for pulmonary sarcoidosis: antimetabolites, biological drugs, and other treatment approaches. Lancet Respir Med 2015; 3:813.
  2. Baughman RP. Pulmonary sarcoidosis. Clin Chest Med 2004; 25:521.
  3. Sharma OP. Pulmonary sarcoidosis and corticosteroids. Am Rev Respir Dis 1993; 147:1598.
  4. du Bois RM. Corticosteroids in sarcoidosis: friend or foe? Eur Respir J 1994; 7:1203.
  5. 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; 160:736.
  6. Baughman RP, Costabel U, du Bois RM. Treatment of sarcoidosis. Clin Chest Med 2008; 29:533.
  7. Melani AS, Bigliazzi C, Cimmino FA, et al. A Comprehensive Review of Sarcoidosis Treatment for Pulmonologists. Pulm Ther 2021; 7:325.
  8. Korsten P, Strohmayer K, Baughman RP, Sweiss NJ. Refractory pulmonary sarcoidosis - proposal of a definition and recommendations for the diagnostic and therapeutic approach. Clin Pulm Med 2016; 23:67.
  9. Baughman RP, Lower EE. Fungal infections as a complication of therapy for sarcoidosis. QJM 2005; 98:451.
  10. Shlobin OA, Nathan SD. Management of end-stage sarcoidosis: pulmonary hypertension and lung transplantation. Eur Respir J 2012; 39:1520.
  11. Girard N, Cottin V, Hot A, et al. [Opportunistic infections and sarcoidosis]. Rev Mal Respir 2004; 21:1083.
  12. Jamilloux Y, Valeyre D, Lortholary O, et al. The spectrum of opportunistic diseases complicating sarcoidosis. Autoimmun Rev 2015; 14:64.
  13. Cremers JP, Drent M, Bast A, et al. Multinational evidence-based World Association of Sarcoidosis and Other Granulomatous Disorders recommendations for the use of methotrexate in sarcoidosis: integrating systematic literature research and expert opinion of sarcoidologists worldwide. Curr Opin Pulm Med 2013; 19:545.
  14. Saag KG, Teng GG, Patkar NM, et al. American College of Rheumatology 2008 recommendations for the use of nonbiologic and biologic disease-modifying antirheumatic drugs in rheumatoid arthritis. Arthritis Rheum 2008; 59:762.
  15. Baughman RP, Winget DB, Lower EE. Methotrexate is steroid sparing in acute sarcoidosis: results of a double blind, randomized trial. Sarcoidosis Vasc Diffuse Lung Dis 2000; 17:60.
  16. Baughman RP, Cremers JP, Harmon M, et al. Methotrexate in sarcoidosis: hematologic and hepatic toxicity encountered in a large cohort over a six year period. Sarcoidosis Vasc Diffuse Lung Dis 2020; 37:e2020001.
  17. Lower EE, Baughman RP. Prolonged use of methotrexate for sarcoidosis. Arch Intern Med 1995; 155:846.
  18. Baughman RP, Koehler A, Bejarano PA, et al. Role of liver function tests in detecting methotrexate-induced liver damage in sarcoidosis. Arch Intern Med 2003; 163:615.
  19. Schnabel A, Richter C, Bauerfeind S, Gross WL. Bronchoalveolar lavage cell profile in methotrexate induced pneumonitis. Thorax 1997; 52:377.
  20. Inokuma S, Kono H, Kohno Y, et al. Methotrexate-induced lung injury in patients with rheumatoid arthritis occurs with peripheral blood lymphocyte count decrease. Ann Rheum Dis 2006; 65:1113.
  21. Chikura B, Sathi N, Lane S, Dawson JK. Variation of immunological response in methotrexate-induced pneumonitis. Rheumatology (Oxford) 2008; 47:1647.
  22. Niitsu N, Okamoto M, Nakamine H, Hirano M. Clinicopathologic correlations of diffuse large B-cell lymphoma in rheumatoid arthritis patients treated with methotrexate. Cancer Sci 2010; 101:1309.
  23. Hoshida Y, Xu JX, Fujita S, et al. Lymphoproliferative disorders in rheumatoid arthritis: clinicopathological analysis of 76 cases in relation to methotrexate medication. J Rheumatol 2007; 34:322.
  24. Lacher MJ. Spontaneous remission or response to methotrexate in sarcoidosis. Ann Intern Med 1968; 69:1247.
  25. Fenton DA, Shaw M, Black MM. Invasive nasal sarcoidosis treated with methotrexate. Clin Exp Dermatol 1985; 10:279.
  26. Lower EE, Baughman RP. The use of low dose methotrexate in refractory sarcoidosis. Am J Med Sci 1990; 299:153.
  27. Suda T, Sato A, Toyoshima M, et al. Weekly low-dose methotrexate therapy for sarcoidosis. Intern Med 1994; 33:437.
  28. Henderson CA, Ilchyshyn A, Curry AR. Laryngeal and cutaneous sarcoidosis treated with methotrexate. J R Soc Med 1994; 87:632.
  29. Kaye O, Palazzo E, Grossin M, et al. Low-dose methotrexate: an effective corticosteroid-sparing agent in the musculoskeletal manifestations of sarcoidosis. Br J Rheumatol 1995; 34:642.
  30. Agbogu BN, Stern BJ, Sewell C, Yang G. Therapeutic considerations in patients with refractory neurosarcoidosis. Arch Neurol 1995; 52:875.
  31. Baughman RP, Lower EE. The effect of corticosteroid or methotrexate therapy on lung lymphocytes and macrophages in sarcoidosis. Am Rev Respir Dis 1990; 142:1268.
  32. Paramothayan S, Lasserson T. Treatments for pulmonary sarcoidosis. Respir Med 2008; 102:1.
  33. Vorselaars AD, Wuyts WA, Vorselaars VM, et al. Methotrexate vs azathioprine in second-line therapy of sarcoidosis. Chest 2013; 144:805.
  34. Paramothayan S, Lasserson TJ, Walters EH. Immunosuppressive and cytotoxic therapy for pulmonary sarcoidosis. Cochrane Database Syst Rev 2006; :CD003536.
  35. Schutt AC, Bullington WM, Judson MA. Pharmacotherapy for pulmonary sarcoidosis: a Delphi consensus study. Respir Med 2010; 104:717.
  36. Gelwan MJ, Kellen RI, Burde RM, Kupersmith MJ. Sarcoidosis of the anterior visual pathway: successes and failures. J Neurol Neurosurg Psychiatry 1988; 51:1473.
  37. Elkin R, Willcox PA. Neurosarcoidosis. A report of 5 cases. S Afr Med J 1985; 67:943.
  38. Pacheco Y, Marechal C, Marechal F, et al. Azathioprine treatment of chronic pulmonary sarcoidosis. Sarcoidosis 1985; 2:107.
  39. O'Reilly BJ, Burrows EH. VIIIth cranial nerve involvement in sarcoidosis. J Laryngol Otol 1995; 109:1089.
  40. Müller-Quernheim J, Kienast K, Held M, et al. Treatment of chronic sarcoidosis with an azathioprine/prednisolone regimen. Eur Respir J 1999; 14:1117.
  41. Bradley B, Branley HM, Egan JJ, et al. Interstitial lung disease guideline: the British Thoracic Society in collaboration with the Thoracic Society of Australia and New Zealand and the Irish Thoracic Society. Thorax 2008; 63 Suppl 5:v1.
  42. Lewis SJ, Ainslie GM, Bateman ED. Efficacy of azathioprine as second-line treatment in pulmonary sarcoidosis. Sarcoidosis Vasc Diffuse Lung Dis 1999; 16:87.
  43. Bakker JA, Drent M, Bierau J. Relevance of pharmacogenetic aspects of mercaptopurine metabolism in the treatment of interstitial lung disease. Curr Opin Pulm Med 2007; 13:458.
  44. Baughman RP, Lower EE. Leflunomide for chronic sarcoidosis. Sarcoidosis Vasc Diffuse Lung Dis 2004; 21:43.
  45. Emery P, Breedveld FC, Lemmel EM, et al. A comparison of the efficacy and safety of leflunomide and methotrexate for the treatment of rheumatoid arthritis. Rheumatology (Oxford) 2000; 39:655.
  46. Savage RL, Highton J, Boyd IW, Chapman P. Pneumonitis associated with leflunomide: a profile of New Zealand and Australian reports. Intern Med J 2006; 36:162.
  47. Bohanec Grabar P, Rozman B, Tomsic M, et al. Genetic polymorphism of CYP1A2 and the toxicity of leflunomide treatment in rheumatoid arthritis patients. Eur J Clin Pharmacol 2008; 64:871.
  48. Grabar PB, Rozman B, Logar D, et al. Dihydroorotate dehydrogenase polymorphism influences the toxicity of leflunomide treatment in patients with rheumatoid arthritis. Ann Rheum Dis 2009; 68:1367.
  49. Modrykamien A, Arrossi A, Reddy A. A 50-year-old man with stage 2 sarcoidosis with pleural involvement. J Hosp Med 2009; 4:E1.
  50. Majithia V, Sanders S, Harisdangkul V, Wilson JG. Successful treatment of sarcoidosis with leflunomide. Rheumatology (Oxford) 2003; 42:700.
  51. Hamzeh N, Voelker A, Forssén A, et al. Efficacy of mycophenolate mofetil in sarcoidosis. Respir Med 2014; 108:1663.
  52. Brill AK, Ott SR, Geiser T. Effect and safety of mycophenolate mofetil in chronic pulmonary sarcoidosis: a retrospective study. Respiration 2013; 86:376.
  53. Papiris S, Stagaki E, Papadaki G, et al. Mycophenolate mofetil as an alternative treatment in sarcoidosis. Pulm Pharmacol Ther 2019; 58:101840.
  54. Moudgil A, Przygodzki RM, Kher KK. Successful steroid-sparing treatment of renal limited sarcoidosis with mycophenolate mofetil. Pediatr Nephrol 2006; 21:281.
  55. Kouba DJ, Mimouni D, Rencic A, Nousari HC. Mycophenolate mofetil may serve as a steroid-sparing agent for sarcoidosis. Br J Dermatol 2003; 148:147.
  56. Pawate S, Moses H, Sriram S. Presentations and outcomes of neurosarcoidosis: a study of 54 cases. QJM 2009; 102:449.
  57. Saketkoo LA, Espinoza LR. Experience of mycophenolate mofetil in 10 patients with autoimmune-related interstitial lung disease demonstrates promising effects. Am J Med Sci 2009; 337:329.
  58. Kalajian AH, Van Meter JR, Callen JP. Sarcoidal anemia and leukopenia treated with methotrexate and mycophenolate mofetil. Arch Dermatol 2009; 145:905.
  59. Baughman RP, Ohmichi M, Lower EE. Combination therapy for sarcoidosis. Sarcoidosis Vasc Diffuse Lung Dis 2001; 18:133.
  60. Pande A, Culver DA. Knowing when to use steroids, immunosuppressants or biologics for the treatment of sarcoidosis. Expert Rev Respir Med 2020; 14:285.
  61. Baughman RP, Lower EE, Drent M. Inhibitors of tumor necrosis factor (TNF) in sarcoidosis: who, what, and how to use them. Sarcoidosis Vasc Diffuse Lung Dis 2008; 25:76.
  62. Bargagli E, Olivieri C, Rottoli P. Cytokine modulators in the treatment of sarcoidosis. Rheumatol Int 2011; 31:1539.
  63. Pritchard C, Nadarajah K. Tumour necrosis factor alpha inhibitor treatment for sarcoidosis refractory to conventional treatments: a report of five patients. Ann Rheum Dis 2004; 63:318.
  64. Ulbricht KU, Stoll M, Bierwirth J, et al. Successful tumor necrosis factor alpha blockade treatment in therapy-resistant sarcoidosis. Arthritis Rheum 2003; 48:3542.
  65. Yee AM, Pochapin MB. Treatment of complicated sarcoidosis with infliximab anti-tumor necrosis factor-alpha therapy. Ann Intern Med 2001; 135:27.
  66. Baughman RP, Lower EE. Infliximab for refractory sarcoidosis. Sarcoidosis Vasc Diffuse Lung Dis 2001; 18:70.
  67. Baughman RP, Drent M, Kavuru M, et al. Infliximab therapy in patients with chronic sarcoidosis and pulmonary involvement. Am J Respir Crit Care Med 2006; 174:795.
  68. Rossman MD, Newman LS, Baughman RP, et al. A double-blinded, randomized, placebo-controlled trial of infliximab in subjects with active pulmonary sarcoidosis. Sarcoidosis Vasc Diffuse Lung Dis 2006; 23:201.
  69. Hostettler KE, Studler U, Tamm M, Brutsche MH. Long-term treatment with infliximab in patients with sarcoidosis. Respiration 2012; 83:218.
  70. White ES. Infliximab in sarcoidosis: more answers or more questions? Am J Respir Crit Care Med 2006; 174:732.
  71. Judson MA, Baughman RP, Costabel U, et al. Efficacy of infliximab in extrapulmonary sarcoidosis: results from a randomised trial. Eur Respir J 2008; 31:1189.
  72. Crouser ED, Lozanski G, Fox CC, et al. The CD4+ lymphopenic sarcoidosis phenotype is highly responsive to anti-tumor necrosis factor-{alpha} therapy. Chest 2010; 137:1432.
  73. Field S, Regan AO, Sheahan K, Collins P. Recalcitrant cutaneous sarcoidosis responding to adalimumab but not to etanercept. Clin Exp Dermatol 2010; 35:795.
  74. Patel SR. Systemic sarcoidosis with bone marrow involvement responding to therapy with adalimumab: a case report. J Med Case Rep 2009; 3:8573.
  75. Heffernan MP, Smith DI. Adalimumab for treatment of cutaneous sarcoidosis. Arch Dermatol 2006; 142:17.
  76. Lahmer T, Knopf A, Lanzl I, et al. Using TNF-alpha antagonist adalimumab for treatment for multisystem sarcoidosis: a case study. Rheumatol Int 2012; 32:2367.
  77. Milman N, Graudal N, Loft A, et al. Effect of the TNF-α inhibitor adalimumab in patients with recalcitrant sarcoidosis: a prospective observational study using FDG-PET. Clin Respir J 2012; 6:238.
  78. Crommelin HA, van der Burg LM, Vorselaars AD, et al. Efficacy of adalimumab in sarcoidosis patients who developed intolerance to infliximab. Respir Med 2016; 115:72.
  79. Utz JP, Limper AH, Kalra S, et al. Etanercept for the treatment of stage II and III progressive pulmonary sarcoidosis. Chest 2003; 124:177.
  80. Baughman RP, Lower EE, Bradley DA, et al. Etanercept for refractory ocular sarcoidosis: results of a double-blind randomized trial. Chest 2005; 128:1062.
  81. Pocock JM, Vasconcelos JC, Ostör AJ. Assessment of anti-TNF-alpha efficacy in rheumatoid arthritis: is 3 months sufficient? Rheumatology (Oxford) 2008; 47:1073.
  82. Minnis PA, Poland M, Keane MP, Donnelly SC. Adalimumab for refractory pulmonary sarcoidosis. Ir J Med Sci 2016; 185:969.
  83. Skoie IM, Wildhagen K, Omdal R. Development of sarcoidosis following etanercept treatment: a report of three cases. Rheumatol Int 2012; 32:1049.
  84. Dhaille F, Viseux V, Caudron A, et al. Cutaneous sarcoidosis occurring during anti-TNF-alpha treatment: report of two cases. Dermatology 2010; 220:234.
  85. Daïen CI, Monnier A, Claudepierre P, et al. Sarcoid-like granulomatosis in patients treated with tumor necrosis factor blockers: 10 cases. Rheumatology (Oxford) 2009; 48:883.
  86. Toussirot E, Berthelot JM, Pertuiset E, et al. Pulmonary nodulosis and aseptic granulomatous lung disease occurring in patients with rheumatoid arthritis receiving tumor necrosis factor-alpha-blocking agent: a case series. J Rheumatol 2009; 36:2421.
  87. Salomon MI, Poon TP, Hsu KC, et al. Membranous glomerulopathy in a patient with sarcoidosis. Arch Pathol 1975; 99:479.
  88. Demeter SL. Myocardial sarcoidosis unresponsive to steroids. Treatment with cyclophosphamide. Chest 1988; 94:202.
  89. Sharp M, Donnelly SC, Moller DR. Tocilizumab in sarcoidosis patients failing steroid sparing therapies and anti-TNF agents. Respir Med X 2019; 1.
  90. Nutz A, Pernet C, Combe B, Cohen JD. Sarcoidosis induced by tocilizumab: a paradoxical event? J Rheumatol 2013; 40:1773.
  91. Shono Y, Kamata M, Takeoka S, et al. Cutaneous sarcoidosis in a patient with rheumatoid arthritis receiving tocilizumab. J Dermatol 2018; 45:e217.
  92. Judson MA, Baughman RP, Costabel U, et al. Safety and efficacy of ustekinumab or golimumab in patients with chronic sarcoidosis. Eur Respir J 2014; 44:1296.
  93. Krause ML, Cooper LT, Chareonthaitawee P, Amin S. Successful use of rituximab in refractory cardiac sarcoidosis. Rheumatology (Oxford) 2016; 55:189.
  94. Cinetto F, Compagno N, Scarpa R, et al. Rituximab in refractory sarcoidosis: a single centre experience. Clin Mol Allergy 2015; 13:19.
  95. Sweiss NJ, Lower EE, Mirsaeidi M, et al. Rituximab in the treatment of refractory pulmonary sarcoidosis. Eur Respir J 2014; 43:1525.
  96. Beccastrini E, Vannozzi L, Bacherini D, et al. Successful treatment of ocular sarcoidosis with rituximab. Ocul Immunol Inflamm 2013; 21:244.
  97. Johns CJ, Zachary JB, MacGregor MI, et al. The longitudinal study of chronic sarcoidosis. Trans Am Clin Climatol Assoc 1983; 94:173.
  98. Zic JA, Horowitz DH, Arzubiaga C, King LE Jr. Treatment of cutaneous sarcoidosis with chloroquine. Review of the literature. Arch Dermatol 1991; 127:1034.
  99. Siltzbach LE, Teirstein AS. Chloroquine therapy in 43 patients with intrathoracic and cutaneous sarcoidosis. Acta Med Scand Suppl 1964; 425:302.
  100. Chloroquine in the treatment of sarcoidosis. A report from the Research Committee of the British Tuberculosis Association. Tubercle 1967; 48:257.
  101. Krasnitz A. Chloroquine therapy in pulmonary sarcoidosis. N Y State J Med 1967; 67:1729.
  102. Brodthagen H. Chloroquine in pulmonary sarcoidosis. Lancet 1968; 1:1157.
  103. Chloroquine in pulmonary sarcoidosis. Lancet 1968; 1:736.
  104. James DG, Barter S, Jash D, et al. Sarcoidosis of the upper respiratory tract (SURT). J Laryngol Otol 1982; 96:711.
  105. Baltzan M, Mehta S, Kirkham TH, Cosio MG. Randomized trial of prolonged chloroquine therapy in advanced pulmonary sarcoidosis. Am J Respir Crit Care Med 1999; 160:192.
  106. Baughman RP, Lower EE. Evidence-based therapy for cutaneous sarcoidosis. Clin Dermatol 2007; 25:334.
  107. Baughman RP, Lower EE. Medical therapy of sarcoidosis. Semin Respir Crit Care Med 2014; 35:391.
  108. Drake WP, Richmond BW, Oswald-Richter K, et al. Effects of broad-spectrum antimycobacterial therapy on chronic pulmonary sarcoidosis. Sarcoidosis Vasc Diffuse Lung Dis 2013; 30:201.
  109. Judson MA. Lung transplantation for pulmonary sarcoidosis. Eur Respir J 1998; 11:738.
  110. Milman N, Burton C, Andersen CB, et al. Lung transplantation for end-stage pulmonary sarcoidosis: outcome in a series of seven consecutive patients. Sarcoidosis Vasc Diffuse Lung Dis 2005; 22:222.
  111. Shorr AF, Helman DL, Davies DB, Nathan SD. Sarcoidosis, race, and short-term outcomes following lung transplantation. Chest 2004; 125:990.
  112. Shah L. Lung transplantation in sarcoidosis. Semin Respir Crit Care Med 2007; 28:134.
  113. Wille KM, Gaggar A, Hajari AS, et al. Bronchiolitis obliterans syndrome and survival following lung transplantation for patients with sarcoidosis. Sarcoidosis Vasc Diffuse Lung Dis 2008; 25:117.
  114. Taimeh Z, Hertz MI, Shumway S, Pritzker M. Lung transplantation for pulmonary sarcoidosis. Twenty-five years of experience in the USA. Thorax 2016; 71:378.
  115. Shorr AF, Davies DB, Nathan SD. Predicting mortality in patients with sarcoidosis awaiting lung transplantation. Chest 2003; 124:922.
  116. Baughman RP, Winget DB, Bowen EH, Lower EE. Predicting respiratory failure in sarcoidosis patients. Sarcoidosis Vasc Diffuse Lung Dis 1997; 14:154.
  117. Hadjiliadis D, Sporn TA, Perfect JR, et al. Outcome of lung transplantation in patients with mycetomas. Chest 2002; 121:128.
  118. Vadnerkar A, Clancy CJ, Celik U, et al. Impact of mold infections in explanted lungs on outcomes of lung transplantation. Transplantation 2010; 89:253.
  119. Fadel E, Mercier O, Mussot S, et al. Long-term outcome of double-lung and heart-lung transplantation for pulmonary hypertension: a comparative retrospective study of 219 patients. Eur J Cardiothorac Surg 2010; 38:277.
  120. Scott J, Higenbottam T. Transplantation of the lungs and heart and lung for patients with severe pulmonary complications from sarcoidosis. Sarcoidosis 1990; 7:9.
  121. Johnson BA, Duncan SR, Ohori NP, et al. Recurrence of sarcoidosis in pulmonary allograft recipients. Am Rev Respir Dis 1993; 148:1373.
  122. Bjørtuft O, Foerster A, Boe J, Geiran O. Single lung transplantation as treatment for end-stage pulmonary sarcoidosis: recurrence of sarcoidosis in two different lung allografts in one patient. J Heart Lung Transplant 1994; 13:24.
  123. Carré P, Rouquette I, Durand D, et al. Recurrence of sarcoidosis in a human lung allograft. Transplant Proc 1995; 27:1686.
  124. Kazerooni EA, Jackson C, Cascade PN. Sarcoidosis: recurrence of primary disease in transplanted lungs. Radiology 1994; 192:461.
  125. Lingaraju R, Pochettino A, Blumenthal NP, et al. Lung transplant outcomes in white and African American recipients: special focus on acute and chronic rejection. J Heart Lung Transplant 2009; 28:8.
  126. Kataria YP. Chlorambucil in sarcoidosis. Chest 1980; 78:36.
  127. Sahgal SM, Sharma OP. Fatal herpes simplex infection during chlorambucil therapy for sarcoidosis. J R Soc Med 1984; 77:144.
  128. Hughes GS Jr, Kataria YP, O'Brien TF Jr. Sarcoidosis presenting as biliary cirrhosis: treatment with chlorambucil. South Med J 1983; 76:1440.
  129. Israel HL, McComb BL. Chlorambucil treatment of sarcoidosis. Sarcoidosis 1991; 8:35.
  130. Bielory L, Frohman LP. Low-dose cyclosporine therapy of granulomatous optic neuropathy and orbitopathy. Ophthalmology 1991; 98:1732.
  131. York EL, Kovithavongs T, Man SF, et al. Cyclosporine and chronic sarcoidosis. Chest 1990; 98:1026.
  132. Martinet Y, Pinkston P, Saltini C, et al. Evaluation of the in vitro and in vivo effects of cyclosporine on the lung T-lymphocyte alveolitis of active pulmonary sarcoidosis. Am Rev Respir Dis 1988; 138:1242.
  133. Kavanaugh AF, Andrew SL, Cooper B, et al. Cyclosporine therapy of central nervous system sarcoidosis. Am J Med 1987; 82:387.
  134. Wyser CP, van Schalkwyk EM, Alheit B, et al. Treatment of progressive pulmonary sarcoidosis with cyclosporin A. A randomized controlled trial. Am J Respir Crit Care Med 1997; 156:1371.
  135. Bachelez H, Senet P, Cadranel J, et al. The use of tetracyclines for the treatment of sarcoidosis. Arch Dermatol 2001; 137:69.
  136. Baughman RP, Judson MA, Teirstein AS, et al. Thalidomide for chronic sarcoidosis. Chest 2002; 122:227.
  137. Nguyen YT, Dupuy A, Cordoliani F, et al. Treatment of cutaneous sarcoidosis with thalidomide. J Am Acad Dermatol 2004; 50:235.
  138. Judson MA, Silvestri J, Hartung C, et al. The effect of thalidomide on corticosteroid-dependent pulmonary sarcoidosis. Sarcoidosis Vasc Diffuse Lung Dis 2006; 23:51.
  139. Marques LJ, Zheng L, Poulakis N, et al. Pentoxifylline inhibits TNF-alpha production from human alveolar macrophages. Am J Respir Crit Care Med 1999; 159:508.
  140. Tong Z, Dai H, Chen B, et al. Inhibition of cytokine release from alveolar macrophages in pulmonary sarcoidosis by pentoxifylline: comparison with dexamethasone. Chest 2003; 124:1526.
  141. Zabel P, Entzian P, Dalhoff K, Schlaak M. Pentoxifylline in treatment of sarcoidosis. Am J Respir Crit Care Med 1997; 155:1665.
  142. Park MK, Fontana Jr, Babaali H, et al. Steroid-sparing effects of pentoxifylline in pulmonary sarcoidosis. Sarcoidosis Vasc Diffuse Lung Dis 2009; 26:121.
Topic 4325 Version 32.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟