Next Article in Journal
Assessing the Accuracy of Diagnostic Capabilities of Large Language Models
Previous Article in Journal
Nordic Orofacial Test-Screening Protocol as a Tool for Assessment of Orofacial Dysfunction in Pediatric and Adult Patients
Previous Article in Special Issue
Diagnostic Value of Comprehensive Echocardiographic Assessment Including Speckle-Tracking in Patients with Sarcoidosis Versus Healthy Controls: A Systematic Review and Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Drug-Induced Sarcoid-like Reactions Associated to Targeted Therapies and Biologic Agents

1
Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy
2
Respiratory Disease Unit, University Hospital of Modena—Policlinico, 41124 Modena, Italy
3
Center for Rare Lung Disease, University Hospital of Modena—Policlinico, 41124 Modena, Italy
*
Author to whom correspondence should be addressed.
Diagnostics 2025, 15(13), 1658; https://doi.org/10.3390/diagnostics15131658
Submission received: 23 May 2025 / Revised: 22 June 2025 / Accepted: 25 June 2025 / Published: 29 June 2025
(This article belongs to the Special Issue Sarcoidosis: From Diagnosis to Management)

Abstract

Background: Sarcoidosis is a multisystem inflammatory disease characterized by the immune-mediated formation of non-necrotizing epithelioid granulomas. Several commonly used medications can induce similar granulomatous reactions, known as drug-induced sarcoid-like reactions (DISRs), which closely mimic sarcoidosis. Despite their specificity in targeting molecular pathways, certain therapies—particularly targeted treatments—have increasingly been linked to DISRs. Methods: This narrative review was based on a PubMed search using the terms “SARCOID LIKE REACTION” and “DRUG”. A cross-check was performed with “SARCOID” combined with each identified drug to identify misclassified cases. Drugs with limited evidence or weak pathogenetic plausibility were excluded, leaving only molecularly targeted therapies for consideration. Sources included case reports, case series, and reviews selected based on their clinical and scientific relevance, without any restrictions on time or language. Results: In light of the available data, five main pharmacological groups were found to be associated to DISR: immune checkpoint inhibitors, TNF-α antagonists, BRAF inhibitors, monoclonal antibodies, and miscellaneous agents. Each group has distinct mechanisms of action and clinical indications, which likely affect the frequency, presentation, and timing of DISRs. Conclusions: Diagnosing DISRs is challenging, and a structured approach is crucial for differentiating them from other conditions. To support clinicians, we propose a diagnostic algorithm to guide decision-making in suspected cases. Management should be individualized, as most DISRs either resolve spontaneously or improve after the discontinuation of the causative drug. Important factors influencing therapeutic decisions include the severity of the underlying disease, the availability of alternative treatments, and the extent of DISR manifestations.

1. Introduction

Sarcoidosis is a multisystem inflammatory disease with an immune-mediated pathogenesis, characterized by a broad spectrum of clinical manifestations [1].
The key feature of the disease is the formation of epithelioid non-necrotizing granulomas (Figure 1), which result from a complex interplay between the innate and adaptive immune systems in genetically predisposed individuals [2].
Interestingly, several commonly used drugs can induce the formation of similar granulomas, leading to clinical manifestations that closely resemble sarcoidosis. These reactions are known as drug-induced sarcoid-like reactions (DISRs) [3].
Despite the specificity of targeted therapies in modulating precise molecular pathways, these treatments, among others, have been increasingly implicated in the development of DISRs.
DISRs are characterized by the formation of non-caseating granulomas, which, upon histological examination, resemble those observed in classical sarcoidosis [4].
Although the exact immunological mechanisms driving the formation of these structures remain unclear, one widely supported hypothesis suggests that they result from an aberrant immune response in genetically predisposed individuals. In particular, in DISRs associated with targeted therapies, this immune response appears to be triggered by two main mechanisms:
  • Excessive immune response: Certain drugs can induce a significant and uncontrolled increase in immune activity, as observed with checkpoint inhibitors and BRAF inhibitors. These medications can lead to excessive activation of T cells, including Th1 and Th17, ultimately promoting granuloma formation [5,6].
  • Paradoxical immune activation: Some anti-inflammatory medications can alter the immune profile, leading to the unexpected activation of pathways involved in granuloma formation. For example, despite their role in treating inflammatory conditions, both anti-TNF agents and Rituximab have been reported to induce granuloma development in some patients [7,8].
No specific immunological traits have been identified that clearly distinguish sarcoid-like reactions from sarcoidosis at the histological level, and little is known about the personal factors that predispose individuals to the development of the disease. Several studies suggest that circulating Th17 lymphocytes prior to disease onset may play a pivotal role in pathogenesis, although the evidence supporting this remains limited [8]. This histological similarity is mirrored by clinical resemblance, further complicating the diagnostic process. Both conditions are typically diagnoses of exclusion, and clinical suspicion often arises from radiological findings and characteristic signs that closely mimic sarcoidosis. Consequently, the lack of definitive diagnostic markers and overlapping clinical and histopathological features hinder the collection of accurate epidemiological data, making it difficult to estimate true incidence and prevalence. Given the rising use of targeted therapies and the frequent misdiagnosis of these reactions, often resulting in unnecessary or inappropriate management, this review aims to provide a summary of current knowledge on sarcoid-like reactions specifically associated with targeted therapies.

2. Materials and Methods

This narrative review was conducted using the PubMed database. The initial search was performed using a primary string composed of the keywords “SARCOID LIKE REACTION” and “DRUG”, which yielded 520 records and provided a base of relevant articles and a list of associated drugs. A cross-check was then conducted by entering the keyword “SARCOID” in combination with the name of each selected drug, in order to retrieve cases that had been mistakenly classified as sarcoidosis. An initial screening was performed to exclude articles involving drug classes for which only a very limited number of cases had been reported, as well as those lacking a plausible pathogenetic link to sarcoid-like reactions. This was performed to avoid including instances where a causal association between the drug and the reaction could not be reasonably supported, or where the presence of concomitant sarcoidosis appeared more likely. A second screening was then conducted, leaving only molecularly targeted therapies. Following duplicate removal, 294 articles were retained for the final review, including single case reports, case series, and review articles. Data were obtained in the form of single case reports, case series, and reviews. No temporal or language filters were applied. Articles were selected based on their clinical and scientific relevance to the topic.

3. Targeted Therapies Associated with Drug-Induced Sarcoid-like Reaction

3.1. Immune Checkpoint Inhibitors

We identified five agent groups linked to DISR. The first group in the order of importance consisted of immune checkpoint inhibitors (ICIs). ICIs are monoclonal antibodies with antitumor activity. They exert their effect by modulating the immune system through the blockade of inhibitory pathways that downregulate immune system activities. In healthy individuals, these regulatory mechanisms help maintaining immune responses within a physiologic range, protecting the host from autoimmunity. By blocking immune checkpoints, ICIs eliminate the inhibitory signals that regulate T-cell activation, enabling tumor-reactive T lymphocytes to bypass these regulatory mechanisms and initiate an effective antitumor response [9].
ICIs target two main pathways: the cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and the programmed death-1 (PD-1) (Figure 2). The former is inhibited by Ipilimumab, the first drug in this class, which was approved in 2011 for metastatic melanoma. The latter is targeted by Nivolumab and Pembrolizumab, which bind to the PD-1 receptor expressed by T lymphocytes, while Atezolizumab, Durvalumab, and Avelumab bind to the PD-L1 ligand that is present on cancer cells [10].
While ICIs offer a significant therapeutic advantage by enhancing the immune system’s ability to target and destroy cancer cells, this heightened immune response can also lead to a range of immune-related adverse events (irAEs). The most frequent irAEs for CTLA-4 inhibitors include colitis, hypophysis, and skin rash, while PD-1 inhibitors are more commonly associated with pneumonitis, hypothyroidism, and vitiligo. Typically, PD-1 and PD-L1 inhibitors are better tolerated than CTLA-4 inhibitors [11].
Among these side effects, DISRs are also described (109 cases). Pembrolizumab is the most reported ICI associated with DISRs. Of the 42 cases analyzed for this review, 24 involved Pembrolizumab administered for the treatment of metastatic melanoma and 7 for non-small cell lung cancer. The average time of onset for DISRs was 7.32 months; the earliest case occurred two weeks after drug administration, while the latest appeared 24 months post-treatment. Symptoms were present in just over half of the cases (54%), with the most common being subcutaneous nodules, erythematous papules, dry cough, and fatigue. In 19 cases, no symptoms were reported, and in some reports, symptoms were unknown. Radiological findings were abnormal in nearly all cases, showing lymphadenopathy, primarily hilar and mediastinal, but also affecting other regions such as gastric, retroperitoneal, and cervical lymph nodes, as well as multiple bilateral pulmonary nodules. It is reasonable to hypothesize that, in asymptomatic cases (46%), the detection of radiological abnormalities may have been due to the routine radiological follow-up of patients undergoing ICI therapy for malignancies. This aspect may pose a challenge, as it raises the issue of differential diagnosis between DISRs and disease progression/recurrence.
In terms of treatment, 6 cases were managed with corticosteroids, while in 14 cases, the ICI was simply discontinued. In three cases, steroid therapy was combined with the discontinuation of the drug. In most cases (76% of the cases with known outcome), these therapeutic strategies led to the resolution of the sarcoid-like reactions [12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45].
Other ICIs associated with sarcoid-like reactions include Nivolumab (21 cases) and Ipilimumab (20 cases), both used alone or in combination (16 cases). In general, these cases present similar radiological findings, diagnostic modalities, treatment, and resolution to those observed with Pembrolizumab. Regarding the timing of onset, Nivolumab and the combination of Nivolumab and Ipilimumab showed a shorter average onset time, specifically 3.96 months (standard deviation: 2.82) and 4.15 months (standard deviation: 3.99), respectively. DISRs related to Nivolumab and Ipilimumab also included the onset of dyspnea in five and six cases, respectively, a symptom not present in reactions associated with Pembrolizumab or the combination of the two drugs [4,19,22,36,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89].
Additionally, the literature reports a few cases of DISRs caused by Atezolizumab (four cases), Avelumab (one case), Durvalumab (three cases), Sintilimab (one case), and the combination of Dabrafenib/Trametinib (one case) [36,90,91,92,93,94,95,96,97,98] (Table 1).

3.2. Tumor Necrosis Factor Alpha Antagonists

The second group identified included tumor necrosis factor alpha (TNF-α) antagonists. TNF-α is a cytokine primarily produced by macrophages that is involved in systemic inflammation and part of a group of mediators that stimulates the acute phase reaction. It can also be produced by CD4+ T lymphocytes, neutrophils, mast cells, eosinophils, and neurons. In physiologic quantities, TNF-α regulates cell proliferation and apoptosis and enhances the activity and recruitment of numerous inflammatory cells.
Anti-TNF-α drugs are biological agents that antagonize this acute phase molecule through various mechanisms. Introduced in 1999, key examples include infliximab, adalimumab, etanercept, golimumab, and certolizumab [99]. These medications are used to treat rheumatologic conditions such as rheumatoid arthritis, ankylosing spondylitis, and psoriatic arthritis, as well as chronic inflammatory diseases like Crohn’s disease, ulcerative colitis, and psoriasis. They selectively bind to TNF-α, forming stable complexes that prevent its interaction with membrane receptors TNFR1 (p55) and TNFR2 (p75), thereby modulating the biological responses it induces or regulates [99,100]. Despite being highly selective agents, these drugs have been associated with various adverse events, including granulomatous infections (such as tuberculosis, leprosy, and leishmaniasis), malignancies (such as lymphoma), hematologic disorders (anemia and pancytopenia), demyelinating conditions, worsening congestive heart failure, and hypersensitivity reactions [101,102]. (Figure 3).
Even if anti-TNF-α biologics are recommended as third-line therapy for sarcoidosis, paradoxically, some patients have experienced worsening of pre-existing sarcoidosis, such as uveitis or cutaneous sarcoidosis [8]. Moreover, numerous reports have described the development of the same reactions in patients without a prior history of sarcoidosis following treatment with TNF-α inhibitors, that resolved after the discontinuation of the therapy [8]. These manifestations can be classified as drug-induced sarcoid-like reactions (DISRs).
In this review, we identified 294 reports including a total of 111 cases of TNF-α induced sarcoid-like reactions, with etanercept being the most frequently represented drug (57 cases, including 2 cases associated with adalimumab and 1 case associated with infliximab), followed by adalimumab (30 cases) and infliximab (21 cases). Two cases associated with certolizumab and golimumab are also described.
The etiology of anti-TNF-α sarcoid-like reaction remains unclear and challenging to explain. It is hypothesized that anti-TNF-α-induced sarcoidosis may result from imbalances involving TNF receptor 2 (TNFR2) and T-regulatory cells. Normally, the activation of TNFR2 (p75) by TNF-α initiates an inhibitory pathway that modulates Treg cells. Consequently, the administration of an anti-TNF-α therapy blocks the TNFR2 pathway, leading to a significant increase in Treg cells, which are crucial regulators of T-cell differentiation. Furthermore, the imbalance between Th1 and Th2 responses contributes to granuloma formation [8].
In the case of Etanercept, the pathogenesis can be linked to its ability to bind lymphotoxin-α (LT-α). It has been demonstrated that this molecule can independently interact with TNF-α receptors, contributing to the formation of infectious granulomas. Combined with the fact that polymorphisms in LT-α have also been linked to an increased incidence of erythema nodosum in sarcoidosis patients, these factors may explain how etanercept-induced alterations in LT-α signaling could play a role in the pathogenesis of DISRs [8].
Among the patients treated with etanercept, rheumatoid arthritis was the most common underlying condition (30 cases), followed by ankylosing spondylitis (10 cases) and psoriatic arthritis and psoriasis (9 cases) [103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135]. Other identified conditions included juvenile idiopathic arthritis and Sjögren’s syndrome. Symptoms typically manifested 2.5 years after starting anti-TNF-α therapy. The onset of sarcoid-like symptoms varied widely, ranging generally from 3 weeks to 6 years. One case was also reported where symptoms appeared after 10 years of treatment, and another where they emerged after 17 years of etanercept therapy.
Nearly half of the cases (25 patients) of DISRs associated with etanercept presented with respiratory symptoms, such as dry cough, dyspnea, and fatigue. Twelve patients exhibited cutaneous manifestations, including erythematous nodules or erythroderma, while two cases presented solely with general symptoms such as malaise, low-grade fever, and weight loss. Etanercept, similar to adalimumab and infliximab, has been closely associated with ocular manifestations, including unilateral or bilateral uveitis (12 cases). Additionally, three asymptomatic patients experienced exclusively progressive renal function deterioration.
Radiological changes were observed in 64.9% of patients on chest X-ray or CT scans, including bilateral hilar mediastinal lymphadenopathy, with or without parenchymal involvement. These changes included nodular or reticulonodular infiltrates, ground-glass opacities, and crazy-paving patterns. One case involved pleural effusion with a reticulonodular pattern. Notably, radiological alterations were also present in patients that did not manifest specific respiratory symptoms. In just nine cases, no radiological alterations were found.
Our review revealed that the majority of patients treated with anti-TNF-α who developed DISR exhibited pulmonary involvement, in contrast to other drug categories, such as BRAF inhibitors, where cutaneous involvement was the most common symptom. This observation could be of interest given that, in pulmonary sarcoidosis, alveolar macrophages spontaneously produce excessive amounts of TNF-α, along with interleukins IL-1, IL-2, IL-6, and IL-10, implicating these cells in the formation of non-caseating granulomas. Further investigation is needed to confirm this and eventually demonstrate how it correlates with the local activation of paradoxical mechanisms that drive granulomatous inflammation [8].
The diagnosis of DIRS is typically confirmed by the histopathological identification of non-caseating granulomas, except in cases of ocular uveitis, where diagnosis is based on clinical and fundoscopic evaluation. Transbronchial biopsy or lymph node sampling via EBUS or mediastinoscopy confirmed 27 diagnoses, while skin biopsies provided diagnostic confirmation in 12 cases. Renal, muscle, and salivary gland biopsies have also been reported.
The vast majority of cases were managed by discontinuing the drug (75%, 45 cases), while in 12 cases etanercept was continued. Corticosteroid therapy was the most commonly employed treatment, used in 34 cases, administered either systemically or topically in cases of uveitis. Additionally, the use of other immunosuppressive therapies is also reported, such as methotrexate (six cases), as well as azathioprine, cyclosporine, and leflunomide (one case each).
Almost all cases achieved clinical resolution following treatment, with only one case showing persistent mediastinal lymphadenopathy on radiological follow-up at 3 months.
DIRS associated with other anti-TNFα agents, such as adalimumab [126,128,132,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153] and infliximab [129,154,155,156,157,158,159,160,161,162,163,164,165], exhibited similar clinical and radiological characteristics to those observed with etanercept. Pulmonary and/or mediastinal involvement, along with cutaneous manifestations, were the most reported features. Infliximab was associated with only two cases of uveitis and adalimumab with seven cases of uveitis. The onset of symptoms occurred after an average of 3.6 years for infliximab and 1.3 years for adalimumab.
A case of DIRS secondary to golimumab therapy for ankylosing spondylitis was also reported, presenting with bilateral granulomatous anterior uveitis and a case secondary to certolizumab therapy administered for rheumatoid arthritis, characterized by cutaneous papules. Both cases were resolved following the discontinuation of the biologic therapy and initiation of corticosteroid treatment [126,166] (Table 2).

3.3. BRAF Inhibitors

The third group consisted of B-Raf proto-oncogene, serine/threonine kinase (BRAF) inhibitors. BRAF inhibitors are targeted therapies that exert antitumor effects by specifically inhibiting the mutant BRAF protein, which is produced as a result of activating mutations in the BRAF gene. The most prevalent of these mutations is V600E, commonly found in metastatic melanoma, non-small cell lung cancer, and anaplastic thyroid carcinoma. This mutation leads to the continuous activation of the MAPK (mitogen-activated protein kinase) signaling pathway, driving uncontrolled cellular growth and proliferation. By targeting this pathway, BRAF inhibitors block the aberrant signaling that is central to tumor progression [167].
To further inhibit this pathway and prevent reactivation via MEK, a kinase enzyme that phosphorylates and activates ERK (another kinase downstream in the pathway), BRAF inhibitors are often combined with MEK inhibitors such as trametinib and cobimetinib. This combination provides a more comprehensive blockade of the MAPK cascade, enhancing therapeutic efficacy [168].
BRAF inhibitors are associated with various adverse effects, including fever, skin reactions, and liver toxicity [169]. The frequent occurrence of cutaneous manifestations, which can range from photosensitivity to keratoacanthomas and squamous cell carcinoma, highlights the critical role of the MAPK signaling pathway in maintaining skin homeostasis. The inhibition of this pathway, particularly due to the activation of germline mutations in RAS, disrupts normal skin function, resulting in a higher incidence of skin-related side effects [170,171]. In addition to these more common toxicities, BRAF inhibitors have also been linked to immune dysregulation reactions, particularly DISRs. As noted in several recent studies, these reactions can be attributed to the immunomodulatory effects of BRAF and MEK inhibitors (Figure 4). These agents create an immune-stimulatory microenvironment by altering cytokine profiles and enhancing the expression of immune-stimulatory molecules, such as CD40L and IFNγ, while concurrently reducing immunosuppressive factors, including regulatory T cells and myeloid-derived suppressor cells (MDSCs) [6].
Dabrafenib in combination with Trametinib is the most frequently reported treatment associated with DISRs in this category. Of the 36 cases of DISRs analyzed in this review [36,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187], 22 occurred in patients receiving this combination therapy. Among these, 21 patients were being treated for metastatic melanoma, while 1 case involved a chiasmatic glioma. The median time to DISR onset was 8.9 months, with a range spanning from 1 to 24 months after the initiation of treatment.
Clinical manifestations were observed in 73% of cases (N = 16), with the most frequent being mild cutaneous involvement (13 cases), including erythematous papules and panniculitis lesions. In six cases, patients were asymptomatic. This tendency toward cutaneous involvement mirrors patterns seen with other side effects associated with BRAF and MEK inhibitors, suggesting that skin may represent a primary site of immune-related adverse reactions to these therapies. Notably, there was also a report of one case of uveitis and one case with elevated liver function enzymes [170,171].
Regarding radiological findings, chest CT scans were performed in 9 of the 22 cases. Mediastinal lymphadenopathy was observed in all but one case, which showed isolated parenchymal involvement. It is worth noting that, in the remaining 13 cases, there was no mention of whether radiological investigations were performed. While it is unclear if these imaging studies were carried out, it is likely they were part of routine oncological follow-up protocols. The absence of significant findings could explain why they were not reported.
Diagnoses were confirmed through histopathological analysis, including skin biopsies and EBUS-TBNA, both revealing non-caseating granulomas consistent with DISRs. In the only case without histopathological examination, involving a patient with uveitis, the diagnosis was based on the clinical, laboratory, and instrumental criteria from the 2014 International Workshop on Ocular Sarcoidosis (IWOS). Approaches to patient management differed across cases, with the most common being topical corticosteroids for eight patients and clinical observation without intervention for seven patients. Oral corticosteroids and temporary discontinuation of therapy were used as alternative strategies in selected cases. Only one patient required the permanent discontinuation of therapy. All patients achieved resolution of their DISRs, except for one who experienced partial remission.
While there is no standardized approach to managing these reactions, the frequent spontaneous resolution monitored through follow-up or following treatment with topical corticosteroids suggests that an initial conservative management strategy may be a reasonable choice.
In addition to Dabrafenib and Trametinib, several other BRAF inhibitors have also been associated with DISRs. These include Vemurafenib (eight cases), alone or in combination with Cobimetinib (three cases), Dabrafenib without any MEKi (two cases), and Encorafenib combined with Binimetinib (one case). No significant differences have been noted between these medications and the data concerning Dabrafenib, whether in terms of clinical presentations or management strategies. In addition to the previously reported cases, we identified several case reports in the literature where, although not explicitly labeled as DISRs, histological examinations showed non-necrotizing granulomas [173]. In particular, three cases of interstitial nephritis were identified, all associated with non-necrotizing granulomas found on histological examination. These findings indicate that the occurrence of DISRs related to BRAF inhibitors might be more widespread than what is currently documented in the literature. Increasing awareness of these manifestations could lead to better management strategies and a deeper understanding of their implications (Table 3).

3.4. Monoclonal Antibodies

The fourth group was made up of various monoclonal antibodies. Over the past decade, monoclonal antibodies have become widely used, especially in the treatment of chronic inflammatory diseases and lymphomas, because of their precise targeted action. Nonetheless, cases of DISR have been reported in the literature following their use, highlighting how such reactions can occur, even with therapies specifically developed to minimize off-target effects. Specifically, DISR have been observed after the administration of Natalizumab, Tocilizumab, Rituximab, Ustekinumab, Trastuzumab, and Dupilumab [7,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213,214,215]. Among these, Rituximab accounts for the highest number of reported cases. Despite this association, rituximab is also known to be used as a third-line treatment for sarcoidosis by targeting CD20 proteins on B cells, which play a crucial role in the pathogenesis of this disease. In particular, B cells may contribute by producing autoantibodies, presenting antigens to T cells, and releasing inflammatory cytokines that promote granuloma formation and chronic inflammation [216]. Rituximab-induced sarcoid reactions may result from a disrupted balance in the immune system after B-cell depletion, leading to changes in cytokine regulation and increased T-cell activity. Approximately 6–7 months after Rituximab treatment, as B cells begin to repopulate, the immune environment shifts, with an increase in naive and transitional B cells, higher BAFF (B-cell activating factor) levels, more IL-10-producing B cells, and fewer memory B cells. This shift mimics the immune profile seen in sarcoidosis, which is characterized by similar changes in B-cell subpopulations and cytokine production. As a result, the recovering immune system may inadvertently recreate the inflammatory conditions that contribute to sarcoidosis, potentially triggering DISR [7,201].
In the literature, rituximab has been associated with a total of sixteen patients with sarcoid-like reaction. Twelve of them were treated for non-Hodgkin lymphoma (NHL) as part of a chemotherapy regimen, while four were treated for autoimmune diseases. The average time of the DISR onset over all is fourteen months. However, this value is heavily driven by one outlier exhibiting an onset of 95 months. When dropping the outlier, the average time becomes eight months, which approximately corresponds with the median value too. The most frequent DISR manifestations linked to the administration of monoclonal antibodies are cutaneous and pulmonary. Interestingly, all six patients who developed DISR after being treated with Tocilizumab only exhibited skin involvement. Slightly more than half of the patients experienced general symptoms, primarily exertional dyspnea, joint pain, and skin lesions. The most common imaging finding was hilar lymphadenopathy, although few patients exhibited also a nodular or micronodular pattern.
The treatment of DIRS varied across cases, ranging from the discontinuation of the offending drug to the use of corticosteroids (administered systemically or locally), or clinical follow-up alone. In most cases, the treatment led to a resolution: 28 out of 38 (Table 4).

3.5. Miscellaneous Drugs

The fifth and final group encompassed miscellaneous agents not falling into the previous categories. While the number of cases in the literature is small, it is worth noticing that DISR has been also associated with other drug classes, such as interleukin inhibitors (Abatacept) and ALK inhibitors (Lorlatinib) [132,201,217,218,219]. Therefore, it is important to take into consideration the possibility of DISR when administering these drugs (Table 5).

4. Diagnosis

Diagnosing DISRs poses significant challenges due to their resemblance to sarcoidosis, other granulomatous diseases, and serious conditions such as malignancies and tumor recurrence or progression, which may necessitate urgent treatment. Misdiagnosis in these cases can lead to inappropriate management and serious consequences. Therefore, a structured approach is essential to differentiate DISRs from these conditions.
Since DISRs are diagnosed by exclusion, the goal is not necessarily to reach a definitive conclusion, but rather to assess the likelihood of the diagnosis by either reinforcing or weakening the initial suspicion. This is performed by identifying supporting evidence while ruling out factors that point to other conditions.
Suspicion of DISRs typically arises from the recognition of characteristic clinical and radiological findings. As observed in the analyzed case reports, these reactions often present with features such as cutaneous lesions, granulomatous infiltration of scars, uveitis, bilateral hilar adenopathy, and increased 18F fluorodeoxyglucose uptake on PET scans. As mentioned earlier, these symptoms may differ based on the specific drug involved, with certain classes of medications being more closely linked to particular clinical presentations.
These manifestations are also common in sarcoidosis, underscoring the importance of a thorough clinical history. For DISRs, this process involves three key steps: establishing a clear temporal association between the start of the drug and the onset of symptoms; reviewing any previous episodes that might suggest undiagnosed sarcoidosis; and ruling out other granulomatous diseases. Examples of such conditions include infectious granulomas, which can result from mycobacterial or fungal infections, and acute granulomatous interstitial lung disease, sometimes associated with methotrexate or Bacillus Calmette–Guérin therapy [220,221,222,223,224]. To facilitate this process, we propose a diagnostic algorithm that aims to clarify the decision-making pathway when confronted with a suspected DISR. The proposed framework is the product of integrating insights derived from previously published algorithms [3] as well as the analysis of the previously discussed case reports. Depending on how well the clinical picture fits, we can classify the likelihood of DISR as unlikely, probable, or highly probable. Figure 5 outlines our proposed diagnostic approach to establishing a DISR.

5. Management and Therapy

The effective management of DISRs should consider a variety of clinical and therapeutic factors. As outlined earlier, these conditions often resolve on their own or after stopping the offending drug. In our review, 88% of cases (259 out of 294) showed partial or complete resolution of symptoms and radiological findings spontaneously or after treatment. Only in the remaining 35 cases, no resolution was observed.
These findings suggest that stopping the medication or initiating treatment may not always be necessary. The decision should be based on several key factors, with the most influential being:
  • The severity of the primary condition;
  • The availability of other therapeutic options;
  • The severity of DISR symptoms.
Primary conditions may differ greatly in terms of severity, progression, and prognosis. For example, in cases where the underlying condition is mild and has a favorable prognosis, it may be simpler for the clinician to consider stopping the drug linked to the DISR. In contrast, for more severe conditions, continuing the current treatment may take precedence.
Another key factor is the availability of alternative treatments; switching to a similar drug in the same class might be ideal in some instances, although the risk of recurrence with a new medication must be carefully evaluated—especially with TNF-alpha inhibitors. When no alternatives are available, clinicians may lean toward maintaining the current regimen. The severity of DISR symptoms, especially in cases involving multiple organs, also plays a significant role. Some DISRs cause minimal or no symptoms, while others show a wider range of clinical manifestations, occasionally affecting multiple organ systems. If the primary drug is effective and DISR symptoms are mild, continuation might be feasible. If the drug is beneficial but the DISR presents significant symptoms, corticosteroids or other treatments used for sarcoidosis could be considered.
Managing DISRs involves several therapeutic options, including careful follow-up, drug discontinuation, topical corticosteroids for localized symptoms, systemic corticosteroids for more widespread involvement, and, when needed, immunomodulatory therapies.
When corticosteroids or other anti-sarcoidosis agents are required for the treatment of a DISR, we suspect that similar doses as those used for sarcoidosis may be effective. However, there is insufficient data to recommend a specific dosing regimen for the disease. Typically, an initial dose of at least 20 mg of daily prednisone equivalent is used. The corticosteroid dose and other immunosuppressants may be adjusted based on the patient’s underlying condition.
Despite these considerations, the current evidence does not yet offer clear recommendations on optimal treatment strategies, such as the recommended duration of clinical observation to allow for spontaneous resolution before initiating additional therapy.
When it comes to follow-up, it is advisable to follow the recommendations from the latest ERS/ATS guidelines on sarcoidosis published in 2020. If future research provides stronger evidence linking specific drug classes to particular clinical manifestations, this may pave the way for developing a more tailored follow-up model for patients with DISRs.
Further studies are crucial to address these gaps in knowledge.

6. Conclusions

Drug-induced sarcoid-like reactions (DISRs) represent a rare and poorly understood complication of targeted therapies. They are almost indistinguishable from sarcoidosis in their clinical, radiological, and histopathological presentation, with resolution upon withdrawal of the causative drug remaining the key distinguishing feature.
This close resemblance may, paradoxically, offer an opportunity: drugs known to induce DISRs could provide valuable insights into the immunopathogenesis of sarcoidosis. In this regard, DISRs represent a unique model through which to explore the immune pathways involved in granuloma formation.
Although the current evidence is limited, our case review suggests a possible association between clinical presentation, disease trajectory, and the pharmacological class of the agent involved. These patterns may ultimately contribute to a better understanding of the diverse phenotypes observed in sarcoidosis itself.
Despite this heterogeneity, our analysis confirms that DISRs usually follow a reassuringly benign trajectory: most patients require no specific therapy, and, with careful risk–benefit appraisal, continuation of the targeted agent is feasible. Heightened clinical awareness is therefore essential to avoid misdiagnosis and unnecessary investigations or therapeutic interventions.
Looking ahead, future studies may pave the way for a more standardized yet tailored approach to treatment and follow-up strategies, considering patient-specific factors as well as the characteristics of the causative drug.

Author Contributions

Conceptualization, F.A. and S.C.; writing—original draft preparation, F.A., L.C., M.N., S.R. and S.C.; writing—review and editing, F.A., L.C., M.N., S.R., S.C., D.A., F.G., B.B., E.C. and R.T.; visualization, F.A., S.R., L.C. and M.N.; supervision, S.C. and B.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
BAFFB-cell-activating factor
BRAFB-Raf proto-oncogene, serine/threonine kinase
CTLA-4Cytotoxic T-lymphocyte-associated protein 4
DISRsDrug-induced sarcoid-like reactions
ICIsImmune checkpoint inhibitors
irAEsImmune-related adverse events
IWOSInternational Workshop on Ocular Sarcoidosis
MAPKMitogen-activated protein kinase
PD-1Programmed death-1
TNF-alfaTumor necrosis factor-alpha

References

  1. Sakthivel, P.; Bruder, D. Mechanism of granuloma formation in sarcoidosis. Curr. Opin. Hematol. 2017, 24, 59–65. [Google Scholar] [CrossRef]
  2. Broos, C.E.; Van Nimwegen, M.; Hoogsteden, H.C.; Hendriks, R.W.; Kool, M.; Van Den Blink, B. Granuloma Formation in Pulmonary Sarcoidosis. Front. Immunol. 2013, 4, 437. [Google Scholar] [CrossRef] [PubMed]
  3. Chopra, A.; Nautiyal, A.; Kalkanis, A.; Judson, M.A. Drug-Induced Sarcoidosis-Like Reactions. Chest 2018, 154, 664–677. [Google Scholar] [CrossRef]
  4. Danlos, F.-X.; Pagès, C.; Baroudjian, B.; Vercellino, L.; Battistella, M.; Mimoun, M.; Jebali, M.; Bagot, M.; Tazi, A.; Lebbé, C. Nivolumab-Induced Sarcoid-Like Granulomatous Reaction in a Patient with Advanced Melanoma. Chest 2016, 149, e133–e136. [Google Scholar] [CrossRef] [PubMed]
  5. Apalla, Z.; Kemanetzi, C.; Papageorgiou, C.; Bobos, M.; Manoli, M.; Fotiadou, C.; Hatzibougias, D.; Boukovinas, I.; Stergiou, E.; Levva, S.; et al. Challenges in sarcoidosis and sarcoid-like reactions associated to immune checkpoint inhibitors: A narrative review apropos of a case. Dermatol. Ther. 2021, 34, e14618. [Google Scholar] [CrossRef] [PubMed]
  6. Kuske, M.; Westphal, D.; Wehner, R.; Schmitz, M.; Beissert, S.; Praetorius, C.; Meier, F. Immunomodulatory effects of BRAF and MEK inhibitors: Implications for Melanoma therapy. Pharmacol. Res. 2018, 136, 151–159. [Google Scholar] [CrossRef]
  7. Galimberti, F.; Fernandez, A.P. Sarcoidosis following successful treatment of pemphigus vulgaris with rituximab: A rituximab-induced reaction further supporting B-cell contribution to sarcoidosis pathogenesis? Clin. Exp. Dermatol. 2016, 41, 413–416. [Google Scholar] [CrossRef]
  8. Amber, K.T.; Bloom, R.; Mrowietz, U.; Hertl, M. TNF-α: A treatment target or cause of sarcoidosis? J. Eur. Acad. Dermatol. Venereol. 2015, 29, 2104–2111. [Google Scholar] [CrossRef]
  9. Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [Google Scholar] [CrossRef]
  10. Wei, S.C.; Duffy, C.R.; Allison, J.P. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov. 2018, 8, 1069–1086. [Google Scholar] [CrossRef]
  11. Ramos-Casals, M.; Brahmer, J.R.; Callahan, M.K.; Flores-Chávez, A.; Keegan, N.; Khamashta, M.A.; Lambotte, O.; Mariette, X.; Prat, A.; Suárez-Almazor, M.E. Immune-related adverse events of checkpoint inhibitors. Nat. Rev. Dis. Primers 2020, 6, 38. [Google Scholar] [CrossRef] [PubMed]
  12. Marcoval, J.; Bauer-Alonso, A.; Fornons-Servent, R.; Jiménez-Colomo, L.; Sabaté-Llobera, A.; Penín, R.M. Subcutaneous sarcoidosis induced by pembrolizumab in a melanoma patient mimicking subcutaneous metastasis at 18F-FDG PET/CT. Rev. Esp. Med. Nucl. E Imagen Mol. Engl. Ed. 2021, 40, 255–256. [Google Scholar] [CrossRef] [PubMed]
  13. Keukeleire, S.D.; Schwarze, J.; Awada, G.; Everaert, H.; Van Binst, A.M.; Cras, L.; Neyns, B.; Aspeslagh, S. An atypical sarcoid-like reaction during anti-protein death 1 treatment in a patient with metastatic melanoma. Melanoma Res. 2020, 30, 524–527. [Google Scholar] [CrossRef]
  14. Jespersen, H.; Bjursten, S.; Ny, L.; Levin, M. Checkpoint inhibitor-induced sarcoid reaction mimicking bone metastases. Lancet Oncol. 2018, 19, e327. [Google Scholar] [CrossRef]
  15. Gambichler, T.; Philippou, S.; Scheel, C.H.; Susok, L. Development of thoracic sarcoid reactions associated with complete response to anti-PD-1 therapy in a patient with advanced cutaneous squamous cell carcinoma. J. Eur. Acad. Dermatol. Venereol. 2022, 36, 35–40. [Google Scholar] [CrossRef] [PubMed]
  16. Grosse, A.; Grosse, C. Diagnostic yield of broncho-alveolar lavage for pembrolizumab induced sarcoid-like reaction of the lung. Cytopathology 2019, 30, 686–687. [Google Scholar] [CrossRef]
  17. Lu, Y. FDG PET/CT Course of Pembrolizumab-Associated Multiorgan Sarcoidosis. Clin. Nucl. Med. 2019, 44, 167–168. [Google Scholar] [CrossRef]
  18. Woodbeck, R.; Metelitsa, A.I.; Naert, K.A. Granulomatous Tumoral Melanosis Associated with Pembrolizumab Therapy: A Mimicker of Disease Progression in Metastatic Melanoma. Am. J. Dermatopathol. 2018, 40, 523–526. [Google Scholar] [CrossRef]
  19. Hiraki, T.; Hatanaka, M.; Arimura, A.; Kawahira, H.; Kirishima, M.; Kitazono, I.; Horinouchi, M.; Higashi, M.; Kanekura, T.; Tanimoto, A. Granulomatous/sarcoid-like reactions in the setting of programmed cell death-1 inhibition: A potential mimic of disease recurrence. J. Cutan. Pathol. 2020, 47, 154–160. [Google Scholar] [CrossRef]
  20. Keane, F.; Yogiaveetil, E.; Kezlarian, B.; Lagratta, M.; Segal, N.H.; Abou-Alfa, G.; O’Reilly, E.M.; Saltz, L.; El Dika, I. Immune checkpoint blockade induced sarcoid-like reaction mimicking progression of disease in a patient with microsatellite instable colorectal cancer: Case report and review of the literature. J. Gastrointest. Oncol. 2024, 15, 500–507. [Google Scholar] [CrossRef]
  21. Mazumder, A.; Mehrmal, S.; Chaudhry, S. Immunotherapy-induced exclusively cutaneous sarcoid-like reaction. BMJ Case Rep. 2023, 16, e252766. [Google Scholar] [CrossRef] [PubMed]
  22. Lomax, A.J.; McGuire, H.M.; McNeil, C.; Choi, C.J.; Hersey, P.; Karikios, D.; Shannon, K.; Van Hal, S.; Carr, U.; Crotty, A.; et al. Immunotherapy-induced sarcoidosis in patients with melanoma treated with PD -1 checkpoint inhibitors: Case series and immunophenotypic analysis. Int. J. Rheum. Dis. 2017, 20, 1277–1285. [Google Scholar] [CrossRef] [PubMed]
  23. Chahin, M.; Stack, A.; Siddiqi, A.; Shaikh, M. Is it metastatic melanoma or is it sarcoidosis? Non-caseating granulomas due to pembrolizumab. BMJ Case Rep. 2020, 13, e240701. [Google Scholar] [CrossRef] [PubMed]
  24. Chau, B.T.C.; McKelvie, P.; Bazargan, A.; Mohamed, M. Pembrolizumab associated sarcoid-like lymphadenopathy mimicking progressive disease in Hodgkin lymphoma. Pathology 2021, 53, 675–678. [Google Scholar] [CrossRef]
  25. Sirgi, Y.; Krochmal, R.; Fleury, C.M.; Holmes, M.; Dewitt, C.A.; Cardis, M.; Kim, C. Pembrolizumab-Associated Cutaneous and Pulmonary Sarcoidosis in Non–Small Cell Lung Cancer Treatment. Clin. Lung Cancer 2022, 23, 542–546. [Google Scholar] [CrossRef]
  26. Yousuf, H.; Mekki, R.; Khan, K.; Hussain, A. Pembrolizumab-Induced Sarcoid-Like Reaction in a Patient with Lung Cancer. Cureus 2020, 12, e12395. [Google Scholar] [CrossRef]
  27. Malaty, S.; Bastian, C.M.; Ramirez-Cibes, I.; Shahlapour, M.; Dhillon, W. Pembrolizumab-Induced Sarcoid-Like Reaction: FDG-PET Scan Interpretation in the Era of Immunotherapy. Cureus 2020, 12, e9449. [Google Scholar] [CrossRef]
  28. Cheshire, S.C.; Board, R.E.; Lewis, A.R.; Gudur, L.D.; Dobson, M.J. Pembrolizumab-induced Sarcoid-like Reactions during Treatment of Metastatic Melanoma. Radiology 2018, 289, 564–567. [Google Scholar] [CrossRef]
  29. McKenna, M.C.; Molloy, K.; Crowther, S.; Feeney, J.; Gillis, A.; Connolly, M.; Kelleher, F. Pembrolizumab-Related Sarcoid-Like Reaction Presenting as Reactivation of Quiescent Scars. J. Oncol. Pract. 2018, 14, 200–201. [Google Scholar] [CrossRef]
  30. Honigman, A.D.; Lai, F.; Elakis, J.; Prall, O.; Goh, M.; McCormack, C. Pembrolizumab-induced sarcoid granulomatous panniculitis and bullous pemphigoid in a single patient. Clin. Case Rep. 2019, 7, 773–775. [Google Scholar] [CrossRef]
  31. Smith, R.J.; Mitchell, T.C.; Chu, E.Y. Pembrolizumab-induced sarcoidal infusion site reaction. J. Cutan. Pathol. 2018, 45, 727–729. [Google Scholar] [CrossRef]
  32. Fakhri, G.; Akel, R.; Salem, Z.; Tawil, A.; Tfayli, A. Pulmonary Sarcoidosis Activation following Neoadjuvant Pembrolizumab plus Chemotherapy Combination Therapy in a Patient with Non-Small Cell Lung Cancer: A Case Report. Case Rep. Oncol. 2017, 10, 1070–1075. [Google Scholar] [CrossRef] [PubMed]
  33. Cousin, S.; Toulmonde, M.; Kind, M.; Cazeau, A.-L.; Bechade, D.; Coindre, J.-M.; Italiano, A. Pulmonary sarcoidosis induced by the anti-PD1 monoclonal antibody pembrolizumab. Ann. Oncol. 2016, 27, 1178–1179. [Google Scholar] [CrossRef] [PubMed]
  34. Park, S.-D.; Kim, M.-S.; Han, M.-H.; Kim, Y.-J.; Jung, H.-Y.; Choi, J.-Y.; Cho, J.-H.; Park, S.-H.; Kim, C.-D.; Kim, Y.-L.; et al. Renal Sarcoidosis-like Reaction Induced by PD-1 Inhibitor Treatment in Non-Small Cell Lung Cancer: A Case Report and Literature Review. Medicina 2023, 59, 991. [Google Scholar] [CrossRef]
  35. Shrateh, O.N.; Abugharbieh, Y.; Asbeh, Y.A.; Hour, H.; Awad, I.; Bannoura, S. Sarcoid-like reaction and hypothyroidism induced by PD-1 inhibitor treatment in metastatic renal cell carcinoma: A case report and literature review. BMC Pulm. Med. 2024, 24, 123. [Google Scholar] [CrossRef] [PubMed]
  36. Dimitriou, F.; Frauchiger, A.L.; Urosevic-Maiwald, M.; Naegeli, M.C.; Goldinger, S.M.; Barysch, M.; Franzen, D.; Kamarachev, J.; Braun, R.; Dummer, R.; et al. Sarcoid-like reactions in patients receiving modern melanoma treatment. Melanoma Res. 2018, 28, 230–236. [Google Scholar] [CrossRef]
  37. Takamori, S.; Furubayashi, N.; Taguchi, K.; Matsubara, T.; Fujishita, T.; Ito, K.; Yamaguchi, M.; Toyozawa, R.; Seto, T.; Negishi, T.; et al. Sarcoid-like reaction of the extrathoracic lymph node in a patient with lung adenocarcinoma treated with pembrolizumab. Thorac. Cancer 2021, 12, 2122–2125. [Google Scholar] [CrossRef]
  38. Soto, F.; Torre-Sada, L.F.; Mott, F.E.; Kim, S.T.; Nurieva, R.; Nagarajan, P.; Guo, M.; Shannon, V.R.; Faiz, S.A.; Casal, R.F.; et al. Sarcoidosis and Airway Disease After Immune Checkpoint Inhibitor Therapy: Case Study and Review of the Literature. J. Immunother. Precis. Oncol. 2023, 6, 111–116. [Google Scholar] [CrossRef]
  39. Yatim, N.; Mateus, C.; Charles, P. Sarcoidosis post-anti-PD-1 therapy, mimicking relapse of metastatic melanoma in a patient undergoing complete remission. Rev. Médecine Interne 2018, 39, 130–133. [Google Scholar] [CrossRef]
  40. Gayá García-Manso, I.; García Ródenas, M.D.M.; Barroso Medel, M.E.; Illán Gambín, F.J. Sarcoidosis-Like Granulomatous Reaction Associated with Pembrolizumab Immunotherapy. Arch. Bronconeumol. Engl. Ed. 2018, 54, 592–593. [Google Scholar] [CrossRef]
  41. Shi, Y.; Li, J.; Chen, M.; Liu, H.; Ma, D.; Lin, Y.; Wang, M.; Xu, Y. Sarcoidosis-like reaction after neoadjuvant pembrolizumab combined with chemotherapy mimicking disease progression of NSCLC induced encouraging discovery of pathological complete response. Thorac. Cancer 2021, 12, 3433–3436. [Google Scholar] [CrossRef]
  42. Wang, L.L.; Patel, G.; Chiesa-Fuxench, Z.C.; McGettigan, S.; Schuchter, L.; Mitchell, T.C.; Ming, M.E.; Chu, E.Y. Timing of Onset of Adverse Cutaneous Reactions Associated with Programmed Cell Death Protein 1 Inhibitor Therapy. JAMA Dermatol. 2018, 154, 1057. [Google Scholar] [CrossRef] [PubMed]
  43. Tetzlaff, M.T.; Nelson, K.C.; Diab, A.; Staerkel, G.A.; Nagarajan, P.; Torres-Cabala, C.A.; Chasen, B.A.; Wargo, J.A.; Prieto, V.G.; Amaria, R.N.; et al. Granulomatous/sarcoid-like lesions associated with checkpoint inhibitors: A marker of therapy response in a subset of melanoma patients. J. Immunother. Cancer 2018, 6, 14. [Google Scholar] [CrossRef]
  44. Firwana, B.; Ravilla, R.; Raval, M.; Hutchins, L.; Mahmoud, F. Sarcoidosis-like syndrome and lymphadenopathy due to checkpoint inhibitors. J. Oncol. Pharm. Pract. 2017, 23, 620–624. [Google Scholar] [CrossRef] [PubMed]
  45. Rodriguez, E.F.; Lipson, E.; Suresh, K.; Cappelli, L.C.; Monaco, S.E.; Maleki, Z. Immune checkpoint blocker-related sarcoid-like granulomatous inflammation: A rare adverse event detected in lymph node aspiration cytology of patients treated for advanced malignant melanoma. Hum. Pathol. 2019, 91, 69–76. [Google Scholar] [CrossRef] [PubMed]
  46. Berthod, G.; Lazor, R.; Letovanec, I.; Romano, E.; Noirez, L.; Mazza Stalder, J.; Speiser, D.E.; Peters, S.; Michielin, O. Pulmonary Sarcoid-Like Granulomatosis Induced by Ipilimumab. J. Clin. Oncol. 2012, 30, e156–e159. [Google Scholar] [CrossRef]
  47. Eckert, A.; Schoeffler, A.; Dalle, S.; Phan, A.; Kiakouama, L.; Thomas, L. Anti-CTLA4 Monoclonal Antibody Induced Sarcoidosis in a Metastatic Melanoma Patient. Dermatology 2009, 218, 69–70. [Google Scholar] [CrossRef]
  48. Reule, R.B.; North, J.P. Cutaneous and pulmonary sarcoidosis-like reaction associated with ipilimumab. J. Am. Acad. Dermatol. 2013, 69, e272–e273. [Google Scholar] [CrossRef]
  49. Martínez Leboráns, L.; Esteve Martínez, A.; Victoria Martínez, A.M.; Alegre De Miquel, V.; Berrocal Jaime, A. Cutaneous sarcoidosis in a melanoma patient under Ipilimumab therapy: Sarcoidosis induced by Ipilimumab therapy. Dermatol. Ther. 2016, 29, 306–308. [Google Scholar] [CrossRef]
  50. Mobini, N.; Dhillon, R.; Dickey, J.; Spoon, J.; Sadrolashrafi, K. Exclusive Cutaneous and Subcutaneous Sarcoidal Granulomatous Inflammation due to Immune Checkpoint Inhibitors: Report of Two Cases with Unusual Manifestations and Review of the Literature. Case Rep. Dermatol. Med. 2019, 2019, 6702870. [Google Scholar] [CrossRef]
  51. Cervantes, J.; Rosen, A.; Dehesa, L.; Dickinson, G.; Alonso-Llamazares, J. Granulomatous Reaction in a Patient with Metastatic Melanoma Treated with Ipilimumab: First Case Reported with Isolated Cutaneous Findings. Actas Dermo-Sifiliográficas Engl. Ed. 2019, 110, 43–49. [Google Scholar] [CrossRef] [PubMed]
  52. Vogel, W.V.; Guislain, A.; Kvistborg, P.; Schumacher, T.N.M.; Haanen, J.B.A.G.; Blank, C.U. Ipilimumab-Induced Sarcoidosis in a Patient with Metastatic Melanoma Undergoing Complete Remission. J. Clin. Oncol. 2012, 30, e7–e10. [Google Scholar] [CrossRef]
  53. Andersen, R.; Nørgaard, P.; Al-Jailawi, M.K.M.; Svane, I.M. Late development of splenic sarcoidosis-like lesions in a patient with metastatic melanoma and long-lasting clinical response to ipilimumab. OncoImmunology 2014, 3, e954506. [Google Scholar] [CrossRef] [PubMed]
  54. González-Cruz, C.; Bodet, D.; Muñoz-Couselo, E.; García-Patos, V. Mediastinal FDG-positive lymph nodes simulating melanoma progression: Drug-induced sarcoidosis like/lymphadenopathy related to ipilimumab. BMJ Case Rep. 2021, 14, e237310. [Google Scholar] [CrossRef]
  55. Murphy, K.P.; Kennedy, M.P.; Barry, J.E.; O’Regan, K.N.; Power, D.G. New-Onset Mediastinal and Central Nervous System Sarcoidosis in a Patient with Metastatic Melanoma Undergoing CTLA4 Monoclonal Antibody Treatment. Oncol. Res. Treat. 2014, 37, 351–353. [Google Scholar] [CrossRef]
  56. Tissot, C.; Carsin, A.; Freymond, N.; Pacheco, Y.; Devouassoux, G. Sarcoidosis complicating anti-cytotoxic T-lymphocyte-associated antigen-4 monoclonal antibody biotherapy. Eur. Respir. J. 2013, 41, 246–247. [Google Scholar] [CrossRef] [PubMed]
  57. Reddy, S.B.; Possick, J.D.; Kluger, H.M.; Galan, A.; Han, D. Sarcoidosis Following Anti-PD-1 and Anti-CTLA-4 Therapy for Metastatic Melanoma. J. Immunother. 2017, 40, 307–311. [Google Scholar] [CrossRef]
  58. Wilgenhof, S.; Morlion, V.; Seghers, A.C.; Vandenbroucke, F.; Everaert, H.; Neyns, B. Sarcoidosis in a Patient with Metastatic Melanoma Sequentially Treated with Anti-CTLA-4 Monoclonal Antibody and Selective BRAF Inhibitor. Anticancer Res. 2012, 32, 1355–1359. [Google Scholar]
  59. Garanzini, E.M.; Scaramuzza, D.; Spadarella, G.; Di Guardo, L.; Marchianò, A. Sarcoidosis-like disease mimicking metastases during adjuvant ipilimumab therapy in advanced melanoma patient: CT scan and MRI help in managing difficult clinical decision. BJRcase Rep. 2020, 6, 20190065. [Google Scholar] [CrossRef]
  60. Koelzer, V.H.; Rothschild, S.I.; Zihler, D.; Wicki, A.; Willi, B.; Willi, N.; Voegeli, M.; Cathomas, G.; Zippelius, A.; Mertz, K.D. Systemic inflammation in a melanoma patient treated with immune checkpoint inhibitors—An autopsy study. J. Immunother. Cancer 2016, 4, 13. [Google Scholar] [CrossRef]
  61. Katagiri, A.; Yamazaki, H.; Ikeda, T. A case of sarcoidosis-like reaction associated with immune checkpoint inhibitors in metastatic renal cell carcinoma. IJU Case Rep. 2022, 5, 15–18. [Google Scholar] [CrossRef] [PubMed]
  62. Zhao, X.; Yue, D.; Qian, J.; Zhang, L.; Song, J.; Zhang, B.; Zhang, C.; Sun, L.; Ma, Y.; Zhang, H.; et al. Case Report: Sarcoid-Like Reactions and Tertiary Lymphoid Structures Following Dual Checkpoint Inhibition in a Patient with Early-Stage Lung Adenocarcinoma. Front. Immunol. 2022, 13, 794217. [Google Scholar] [CrossRef]
  63. Pham, J.P.; Star, P.; Wong, S.; Damian, D.L.; Saw, R.P.M.; Whitfeld, M.J.; Menzies, A.M.; Joshua, A.M.; Smith, A. Cutaneous Sarcoidosis Due to Immune-Checkpoint Inhibition and Exacerbated by a Novel BRAF Dimerization Inhibitor. Skin. Health Dis. 2021, 1, e71. [Google Scholar] [CrossRef]
  64. Tan, I.; Malinzak, M.; Salama, A.K.S. Delayed onset of neurosarcoidosis after concurrent ipilimumab/nivolumab therapy. J. Immunother. Cancer 2018, 6, 77. [Google Scholar] [CrossRef] [PubMed]
  65. Sagawa, T.; Sato, Y.; Nagashima, H.; Takada, K.; Takahashi, M.; Hirakawa, M.; Hamaguchi, K.; Tamura, F.; Fujikawa, K.; Okamoto, K.; et al. Hilar/mediastinal and cutaneous drug-induced sarcoidosis-like reaction associated with immune checkpoint inhibitors in metastatic colorectal cancer: A case report. Front. Immunol. 2023, 14, 1203621. [Google Scholar] [CrossRef]
  66. Charkviani, M.; Herrmann, S.M. Immune Checkpoint Inhibitor–Associated Sarcoidosis Reaction in the Kidney: Case Report. Kidney Med. 2023, 5, 100626. [Google Scholar] [CrossRef]
  67. Suozzi, K.C.; Stahl, M.; Ko, C.J.; Chiang, A.; Gettinger, S.N.; Siegel, M.D.; Bunick, C.G. Immune-related sarcoidosis observed in combination ipilimumab and nivolumab therapy. JAAD Case Rep. 2016, 2, 264–268. [Google Scholar] [CrossRef] [PubMed]
  68. Yasin, H.; Yadala, V.; Khan, N.A.J.; Graffeo, V.; Denning, K.; Lebowicz, Y. Immunotherapy-Induced Sarcoid-Like Reaction: A Shrewd Imitator. J. Investig. Med. High Impact Case Rep. 2021, 9, 23247096211009400. [Google Scholar] [CrossRef]
  69. Reuss, J.E.; Kunk, P.R.; Stowman, A.M.; Gru, A.A.; Slingluff, C.L.; Gaughan, E.M. Sarcoidosis in the setting of combination ipilimumab and nivolumab immunotherapy: A case report & review of the literature. J. Immunother. Cancer 2016, 4, 94. [Google Scholar] [CrossRef]
  70. Torres-Zurita, A.; Vázquez-Montero, L.; Gallego-López, L.; Mediano-Rambla, M.D.; De La Cruz-Merino, L. Sarcoidosis-like reaction induced by immune checkpoint inhibitor in a patient with hepatocellular carcinoma: A case report. Front. Immunol. 2023, 14, 1150128. [Google Scholar] [CrossRef]
  71. Purcell, V.; Preti, B.; Fernandes, R. Suspected immune checkpoint inhibitor-induced pulmonary sarcoid reaction in metastatic renal cell carcinoma. Clin. Case Rep. 2022, 10, e5960. [Google Scholar] [CrossRef]
  72. Kim, C.; Gao, J.; Shannon, V.R.; Siefker-Radtke, A. Systemic sarcoidosis first manifesting in a tattoo in the setting of immune checkpoint inhibition. BMJ Case Rep. 2016, 2016, bcr2016216217. [Google Scholar] [CrossRef] [PubMed]
  73. DeCarli, K.; Masel, R.; Hsu, A.; Lopresti, M. Treatment-induced sarcoidosis in a patient with metastatic clear cell ovarian cancer. BMJ Case Rep. 2021, 14, e247278. [Google Scholar] [CrossRef]
  74. Chiang, J.; Hebroni, F.; Bedayat, A.; Pourzand, L. Case 286: Sarcoidlike Granulomatosis and Lymphadenopathy—Thoracic Manifestations of Nivolumab Drug Toxicity. Radiology 2021, 298, 471–475. [Google Scholar] [CrossRef]
  75. Ung, C.; Gragoudas, E. Checkpoint inhibitor-induced sarcoid choroidal granulomas. Am. J. Ophthalmol. Case Rep. 2020, 18, 100652. [Google Scholar] [CrossRef]
  76. Urrego-Callejas, T.; Sandoval-Álvarez, S.; Gómez-Wolff, R.; Vásquez, G. Cutaneous and Pulmonary Sarcoid-Like Reaction Induced by Nivolumab: Case Report and Brief Literature Review. JCR J. Clin. Rheumatol. 2021, 27, S460–S464. [Google Scholar] [CrossRef] [PubMed]
  77. Laroche, A.; Alarcon Chinchilla, E.; Bourgeault, E.; Doré, M.-A. Erythema Nodosum as the Initial Presentation of Nivolumab-Induced Sarcoidosis-Like Reaction. J. Cutan. Med. Surg. 2018, 22, 627–629. [Google Scholar] [CrossRef] [PubMed]
  78. Lainez, S.; Tissot, C.; Cottier, M.; Vergnon, J.-M. EBUS-TBNA Can Distinguish Sarcoid-Like Side Effect of Nivolumab Treatment from Tumor Progression in Non-Small Cell Lung Cancer. Respiration 2017, 94, 518–521. [Google Scholar] [CrossRef]
  79. Diaz-Perez, J.A.; Beveridge, M.G.; Victor, T.A.; Cibull, T.L. Granulomatous and lichenoid dermatitis after IgG4 anti-PD-1 monoclonal antibody therapy for advanced cancer. J. Cutan. Pathol. 2018, 45, 434–438. [Google Scholar] [CrossRef]
  80. Jain, P.V.; Squires, C.; Gasparri, M.; Sheinin, Y. Nivolumab induced pulmonary sarcoid-like granulomas with a concurrent pleural schwannoma: A pathologic-radiologic correlation of a rare condition mimicking metastatic melanoma. Ann. Diagn. Pathol. 2022, 57, 151880. [Google Scholar] [CrossRef]
  81. Tulbah, R.I.; Rowe, S.P.; Solnes, L.B.; Javadi, M.S. Nivolumab-Associated Pulmonary and Bone Sarcoidosis in a Patient with Melanoma of Unknown Primary. Clin. Nucl. Med. 2019, 44, e519–e521. [Google Scholar] [CrossRef] [PubMed]
  82. Zhang, M.; Schembri, G. Nivolumab-Induced Development of Pulmonary Sarcoidosis in Renal Cell Carcinoma. Clin. Nucl. Med. 2017, 42, 728–729. [Google Scholar] [CrossRef]
  83. Birnbaum, M.R.; Ma, M.W.; Fleisig, S.; Packer, S.; Amin, B.D.; Jacobson, M.; McLellan, B.N. Nivolumab-related cutaneous sarcoidosis in a patient with lung adenocarcinoma. JAAD Case Rep. 2017, 3, 208–211. [Google Scholar] [CrossRef]
  84. Beer, L.; Hochmair, M.; Kifjak, D.; Haug, A.R.; Prayer, F.; Mayerhoefer, M.E.; Herold, C.; Prosch, H. Particular findings on lung CT in patients undergoing immunotherapy for bronchogenic carcinoma. Wien. Klin. Wochenschr. 2020, 132, 467–474. [Google Scholar] [CrossRef]
  85. Ogawa, T.; Ishitsuka, Y.; Iwamoto, K.; Koguchi-Yoshioka, H.; Tanaka, R.; Watanabe, R.; Fujisawa, Y.; Fujimoto, M. Programmed cell death 1 blockade-induced cutaneous sarcoid-like epithelioid granulomas in advanced melanoma: A case report. J. Eur. Acad. Dermatol. Venereol. 2018, 32, pe260. [Google Scholar] [CrossRef] [PubMed]
  86. Montaudié, H.; Pradelli, J.; Passeron, T.; Lacour, J.-P.; Leroy, S. Pulmonary sarcoid-like granulomatosis induced by nivolumab. Br. J. Dermatol. 2017, 176, 1060–1063. [Google Scholar] [CrossRef] [PubMed]
  87. Noguchi, S.; Kawachi, H.; Yoshida, H.; Fukao, A.; Terashita, S.; Ikeue, T.; Horikawa, S.; Sugita, T. Sarcoid-Like Granulomatosis Induced by Nivolumab Treatment in a Lung Cancer Patient. Case Rep. Oncol. 2018, 11, 562–566. [Google Scholar] [CrossRef]
  88. Fukuchi, K.; Hikawa, M.; Sano, Y.; Kasuya, A.; Aoshima, M.; Tatsuno, K.; Nakamura, Y.; Kosugi, I.; Tokura, Y. Sarcoid-like reaction and vitiligo occurring after nivolumab therapy in a patient with metastatic melanoma. J. Dermatol. 2019, 46, E359–E360. [Google Scholar] [CrossRef]
  89. Paolini, L.; Poli, C.; Blanchard, S.; Urban, T.; Croué, A.; Rousselet, M.-C.; Le Roux, S.; Labarrière, N.; Jeannin, P.; Hureaux, J. Thoracic and cutaneous sarcoid-like reaction associated with anti-PD-1 therapy: Longitudinal monitoring of PD-1 and PD-L1 expression after stopping treatment. J. Immunother. Cancer 2018, 6, 52. [Google Scholar] [CrossRef]
  90. Mitchell, M.A.; Hogan, K.; Amjadi, K. Atezolizumab-Induced Sarcoid-Like Granulomatous Reaction in a Patient with Urothelial Cell Carcinoma. Immunotherapy 2018, 10, 1189–1192. [Google Scholar] [CrossRef]
  91. Tsunoda, A.; Mizuno, T.; Iida, S.; Uchida, K.; Yamashita, M.; Sukeno, K.; Oka, H.; Tono, Y.; Ishihara, M.; Saito, K.; et al. Atezolizumab-Induced Sarcoidosis-Like Reaction in a Patient with Metastatic Breast Cancer. Case Rep. Oncol. Med. 2022, 2022, 2709062. [Google Scholar] [CrossRef] [PubMed]
  92. Tam, K.; Wallace, M.; Mulrennan, S. Recommencement of atezolizumab with associated pulmonary sarcoid-like reaction. Respirol. Case Rep. 2024, 12, e01363. [Google Scholar] [CrossRef] [PubMed]
  93. Lafon, M.; Blaye, C.; Kind, M.; Bechade, D.; Chassaigne, F.; Italiano, A.; Grellety, T. Sarcoidosis-like reaction in metastatic triple negative breast cancer treated by anti-PD-L1. Breast J. 2019, 25, 971–973. [Google Scholar] [CrossRef]
  94. Balestra, R.; Benzaquen, S.; Wang, J. Sarcoidosis-like Granulomatous Lung Reaction Associated with Anti–Programmed Death Receptor-1 Ligand Therapy. Ann. Am. Thorac. Soc. 2017, 14, 296–299. [Google Scholar] [CrossRef] [PubMed]
  95. Sanderson, E.; Wimaleswaran, H.; Senko, C.; White, S.; McDonald, C.F. Durvalumab induced sarcoid-like pulmonary lymphadenopathy. Respirol. Case Rep. 2020, 8, e00542. [Google Scholar] [CrossRef]
  96. Minami, S.; Yasuoka, H.; Shoshihara, N.; Ishida, D.; Sakamaki, Y. Sarcoid-Like Granulomatosis of the Lung Related to Durvalumab After Chemoradiation for Pulmonary Squamous Cell Carcinoma. J. Med. Cases 2023, 14, 19–24. [Google Scholar] [CrossRef]
  97. Pantarotto, M.; Barata, R.; Coelho, R.; Carvalheiro, C.; Rolim, I.; Garrido, P.; GIl, N.; Duarte-Ramos, F.; Stumpf Tonin, F.S. The Meaning of Lymphadenopathies During Adjuvant Durvalumab After Chemoradiotherapy for Lung Cancer: Thinking Beyond Disease Progression. Cureus 2022, 14, e26729. [Google Scholar] [CrossRef]
  98. Li, H.; Mu, F.; Zou, B.; Wang, L. Pulmonary sarcoidosis-like reactions induced by sintilimab in esophageal cancer: A case report. Medicine 2023, 102, e34432. [Google Scholar] [CrossRef]
  99. Udalova, I.; Monaco, C.; Nanchahal, J.; Feldmann, M. Anti-TNF Therapy. Microbiol. Spectr. 2016, 4, 4. [Google Scholar] [CrossRef]
  100. Desai, S.B.; Furst, D.E. Problems encountered during anti-tumour necrosis factor therapy. Best Pract. Res. Clin. Rheumatol. 2006, 20, 757–790. [Google Scholar] [CrossRef]
  101. Atzeni, F.; Gianturco, L.; Talotta, R.; Varisco, V.; Ditto, M.C.; Turiel, M.; Sarzi-Puttini, P. Investigating the Potential Side Effects of Anti-TNF Therapy for Rheumatoid Arthritis: Cause for Concern? Immunotherapy 2015, 7, 353–361. [Google Scholar] [CrossRef]
  102. Antoni, C.; Braun, J. Side effects of anti-TNF therapy: Current knowledge. Clin. Exp. Rheumatol. 2002, 20, S152. [Google Scholar] [PubMed]
  103. Dalbeth, N.; Perez-Ruiz, F.; Edwards, N.L.; Schlesinger, N. In Defense of Research into the Crystal Induced Arthropathies. J. Rheumatol. 2008, 35, 2278–2279. [Google Scholar] [CrossRef] [PubMed]
  104. Akiyama, M.; Kaneko, Y.; Hanaoka, H.; Kuwana, M.; Takeuchi, T. Acute Kidney Injury due to Renal Sarcoidosis during Etanercept Therapy: A Case Report and Literature Review. Intern. Med. 2015, 54, 1131–1134. [Google Scholar] [CrossRef]
  105. Lamrock, E.; Brown, P. Development of cutaneous sarcoidosis during treatment with tumour necrosis alpha factor antagonists. Australas. J. Dermatol. 2012, 53, e87–e90. [Google Scholar] [CrossRef] [PubMed]
  106. Vieira, M.A.H.B.; Saraiva, M.I.R.; Silva, L.K.L.D.; Fraga, R.C.; Kakizaki, P.; Valente, N.Y.S. Development of exclusively cutaneous sarcoidosis in patient with rheumatoid arthritis during treatment with etanercept. Rev. Assoc. Médica Bras. 2016, 62, 718–720. [Google Scholar] [CrossRef]
  107. Miyagi, R.; Ideguchi, H.; Soga, T.; Yamakawa, Y.; Otsuki, H.; Niino, H.; Shiina, T.; Ueda, A.; Ishigatsubo, Y. Development of pulmonary and cardiac sarcoidosis during etanercept therapy. Int. J. Rheum. Dis. 2014, 17, 810–812. [Google Scholar] [CrossRef]
  108. González-López, M.A.; Blanco, R.; González-vela, M.C.; Fernández-llaca, H.; Rodríguez-valverde, V. Development of sarcoidosis during etanercept therapy. Arthritis Care Res. 2006, 55, 817–820. [Google Scholar] [CrossRef]
  109. Ishiguro, T.; Takayanagi, N.; Kurashima, K.; Matsushita, A.; Harasawa, K.; Yoneda, K.; Tsuchiya, N.; Miyahara, Y.; Yamaguchi, S.; Yano, R.; et al. Development of Sarcoidosis during Etanercept Therapy. Intern. Med. 2008, 47, 1021–1025. [Google Scholar] [CrossRef]
  110. Skoie, I.M.; Wildhagen, K.; Omdal, R. Development of sarcoidosis following etanercept treatment: A report of three cases. Rheumatol. Int. 2012, 32, 1049–1053. [Google Scholar] [CrossRef]
  111. Verschueren, K.; Van Essche, E.; Verschueren, P.; Taelman, V.; Westhovens, R. Development of sarcoidosis in etanercept-treated rheumatoid arthritis patients. Clin. Rheumatol. 2007, 26, 1969–1971. [Google Scholar] [CrossRef] [PubMed]
  112. Burns, A.M.; Green, P.J.; Pasternak, S. Etanercept-induced cutaneous and pulmonary sarcoid-like granulomas resolving with adalimumab. J. Cutan. Pathol. 2012, 39, 289–293. [Google Scholar] [CrossRef] [PubMed]
  113. Phillips, K.; Weinblatt, M. Granulomatous lung disease occurring during etanercept treatment. Arthritis Rheum. 2005, 53, 618–620. [Google Scholar] [CrossRef]
  114. Saifee, M.; Bansal, A.; Bever, G.J.; Stewart, J.M. Late-onset Etanercept-associated Ocular Sarcoidosis with Profound Vision Loss. Ocul. Immunol. Inflamm. 2022, 30, 2055–2059. [Google Scholar] [CrossRef] [PubMed]
  115. Peno-Green, L.; Lluberas, G.; Kingsley, T.; Brantley, S. Lung Injury Linked to Etanercept Therapy. Chest 2002, 122, 1858–1860. [Google Scholar] [CrossRef]
  116. Thongpooswan, S.; Abrudescu, A. Lung Sarcoidosis in Etanercept Treated Rheumatoid Arthritis Patient: A Case Report and Review of the Literature. Case Rep. Rheumatol. 2014, 2014, 358567. [Google Scholar] [CrossRef]
  117. Eichhoff, G. Management with secukinumab of tumour necrosis factor inhibitor-induced pulmonary sarcoidosis-like reaction in a patient with psoriasis. Clin. Exp. Dermatol. 2020, 45, 455–456. [Google Scholar] [CrossRef]
  118. Ngoufack, C.; Semerano, L.; Podglajen, I.; Bruneval, P.; Meune, C.; Valeyre, D.; Dhote, R.; Boissier, M.-C.; Saidenberg-Kermanac’h, N. Mitral valve granulomatosis: A paradoxical reaction complicating etanercept treatment in rheumatoid arthritis. A case report. Jt. Bone Spine 2021, 88, 105183. [Google Scholar] [CrossRef]
  119. Wendling, D.; Nueffer, J.-P. Muscular sarcoidosis under anti-TNF treatment in ankylosing spondylitis. Jt. Bone Spine 2018, 85, 769. [Google Scholar] [CrossRef]
  120. Tong, D.; Manolios, N.; Howe, G.; Spencer, D. New onset sarcoid-like granulomatosis developing during anti-TNF therapy: An under-recognised complication. Intern. Med. J. 2012, 42, 89–94. [Google Scholar] [CrossRef]
  121. Hübscher, O.; Re, R.; Iotti, R. Pulmonary rheumatoid nodules in an etanercept-treated patient. Arthritis Rheum. 2003, 48, 2077–2078. [Google Scholar] [CrossRef] [PubMed]
  122. Farah, R.E.; Shay, M.D. Pulmonary Sarcoidosis Associated with Etanercept Therapy. Pharmacother. J. Hum. Pharmacol. Drug Ther. 2007, 27, 1446–1448. [Google Scholar] [CrossRef]
  123. Bhamra, K.; Stevens, R. Pulmonary Sarcoidosis following Etanercept Treatment. Case Rep. Rheumatol. 2012, 2012, 724013. [Google Scholar] [CrossRef] [PubMed]
  124. Majjad, A.; Bezza, A.; Biyi, A.; El Ochi, M.R.; El Maghraoui, A. Pulmonary Sarcoidosis following Etanercept Treatment for Ankylosing Spondylitis: A Case Report and Review of the Literature. Case Rep. Rheumatol. 2018, 2018, 9867248. [Google Scholar] [CrossRef] [PubMed]
  125. Louie, G.H.; Chitkara, P.; Ward, M.M. Relapse of sarcoidosis upon treatment with etanercept. Ann. Rheum. Dis. 2008, 67, 896–898. [Google Scholar] [CrossRef] [PubMed]
  126. Daien, C.I.; Monnier, A.; Claudepierre, P.; Constantin, A.; Eschard, J.-P.; Houvenagel, E.; Samimi, M.; Pavy, S.; Pertuiset, E.; Toussirot, E.; et al. Sarcoid-like granulomatosis in patients treated with tumor necrosis factor blockers: 10 cases. Rheumatology 2009, 48, 883–886. [Google Scholar] [CrossRef]
  127. Hashkes, P.J.; Shajrawi, I. Sarcoid-related uveitis occuring during etanercept therapy. Clin. Exp. Rheumatol. 2003, 21, 645–646. [Google Scholar]
  128. Massara, A.; Cavazzini, L.; La Corte, R.; Trotta, F. Sarcoidosis Appearing During Anti-Tumor Necrosis Factor α Therapy: A New “Class Effect” Paradoxical Phenomenon. Two Case Reports and Literature Review. Semin. Arthritis Rheum. 2010, 39, 313–319. [Google Scholar] [CrossRef]
  129. Toussirot, E.; Pertuiset, E.; Kantelip, B.; Wendling, D. Sarcoidosis occuring during anti-TNF-alpha treatment for inflammatory rheumatic diseases: Report of two cases. Clin. Exp. Rheumatol. 2008, 26, 471–475. [Google Scholar]
  130. Unterstell, N.; Bressan, A.L.; Serpa, L.A.; Castro, P.P.D.F.E.; Gripp, A.C. Systemic sarcoidosis induced by etanercept: First Brazilian case report. An. Bras. Dermatol. 2013, 88, 197–199. [Google Scholar] [CrossRef]
  131. Park, S.-K.; Hwang, P.-H.; Yun, S.-K.; Kim, H.-U.; Park, J. Tumor Necrosis Factor Alpha Blocker-Induced Erythrodermic Sarcoidosis in with Juvenile Rheumatoid Arthritis: A Case Report and Review of the Literature. Ann. Dermatol. 2017, 29, 74. [Google Scholar] [CrossRef] [PubMed]
  132. Sobolewska, B.; Baglivo, E.; Edwards, A.O.; Kramer, M.; Miserocchi, E.; Palestine, A.G.; Schwab, I.R.; Zamir, E.; Doycheva, D.; Zierhut, M. Drug-induced Sarcoid Uveitis with Biologics. Ocul. Immunol. Inflamm. 2022, 30, 907–914. [Google Scholar] [CrossRef]
  133. Van Der Stoep, D.; Braunstahl, G.-J.; Van Zeben, J.; Wouters, J. Sarcoidosis During Anti-Tumor Necrosis Factor-α Therapy: No Relapse After Rechallenge. J. Rheumatol. 2009, 36, 2847–2848. [Google Scholar] [CrossRef] [PubMed]
  134. Bewley, A.P.; Maleki, S. Systemic sarcoidosis reactions as a result of tumour necrosis factor-alpha treatment for patients with psoriasis. Clin. Exp. Dermatol. 2021, 46, 1548–1550. [Google Scholar] [CrossRef] [PubMed]
  135. Theunssens, X.; Bricman, L.; Dierckx, S.; Sapart, E.; Sokolova, T.; Avramovska, A.; Durez, P. Anti-TNF Induced Sarcoidosis-Like Disease in Rheumatoid Arthritis Patients: Review Cases from the RA UCLouvain Brussels Cohort. Rheumatol. Ther. 2022, 9, 763–770. [Google Scholar] [CrossRef]
  136. Salvatierra, J.; Magro-Checa, C.; Rosales-Alexander, J.L.; Raya-Alvarez, E. Acute sarcoidosis as parotid fever in rheumatoid arthritis under anti-tumor necrosis factor-α therapy. Rheumatology 2011, 50, 1346–1348. [Google Scholar] [CrossRef]
  137. Bhargava, S.; Perlman, D.M.; Allen, T.L.; Ritter, J.H.; Bhargava, M. Adalimumab induced pulmonary sarcoid reaction. Respir. Med. Case Rep. 2013, 10, 53–55. [Google Scholar] [CrossRef]
  138. Metyas, S.K.; Tadros, R.M.; Arkfeld, D.G. Adalimumab-induced noncaseating granuloma in the bone marrow of a patient being treated for rheumatoid arthritis. Rheumatol. Int. 2009, 29, 437–439. [Google Scholar] [CrossRef]
  139. Rodrigues-Fernandes, C.I.; Migliorati, C.A.; Gueiros, L.A.M.; De Lima Morais, T.M.; Vargas, P.A.; De Almeida, O.P.; Lopes, M.A.; Santos-Silva, A.R. Adalimumab-induced sarcoidosis-like reaction involving oral cavity in rheumatoid arthritis: A case-based review. Clin. Rheumatol. 2021, 40, 3833–3839. [Google Scholar] [CrossRef]
  140. Marcella, S.; Welsh, B.; Foley, P. Development of sarcoidosis during adalimumab therapy for chronic plaque psoriasis: TNF antagonist-associated sarcoidosis. Australas. J. Dermatol. 2011, 52, e8–e11. [Google Scholar] [CrossRef]
  141. Korsten, P.; Sweiss, N.J.; Nagorsnik, U.; Niewold, T.B.; Gröne, H.-J.; Gross, O.; Müller, G.A. Drug-Induced Granulomatous Interstitial Nephritis in a Patient with Ankylosing Spondylitis During Therapy with Adalimumab. Am. J. Kidney Dis. 2010, 56, e17–e21. [Google Scholar] [CrossRef] [PubMed]
  142. Turkowski, Y.; Konnikov, N.; Mahalingam, M. Necrotizing Granulomas in a Patient with Psoriasis and Sarcoidosis After Adalimumab—Medication-Induced Reaction or Reactivation of Latent Disease? Am. J. Dermatopathol. 2019, 41, 661–666. [Google Scholar] [CrossRef] [PubMed]
  143. Wladis, E.J.; Tarasen, A.J.; Roth, Z.J.; Farber, M.G.; Ross, J.; Michaels, V.M. Orbital Sarcoid-Like Granulomatosis After Inhibition of Tumor Necrosis Factor-α. Ophthal. Plast. Reconstr. Surg. 2016, 32, e30–e32. [Google Scholar] [CrossRef] [PubMed]
  144. Korta, D.Z.; Ochieng, P.; Fishman, D.; Katz, S.E. Pulmonary sarcoidosis and latent tuberculosis in a patient with psoriasis treated with adalimumab. Dermatol. Online J. 2015; 21. [Google Scholar] [CrossRef]
  145. Hamada, E.; Yamamoto, Y.; Okuda, Y.; Sakaguchi, K.; Suzuki, K.; Kai, Y.; Takeda, M.; Hontsu, S.; Yamauchi, M.; Yoshikawa, M.; et al. Pulmonary sarcoidosis with a cavitary lesion in the lung caused by a TNF-α inhibitor: A case report. Respirol. Case Rep. 2022, 10, e01065. [Google Scholar] [CrossRef]
  146. Morgane, V.; Jean-Charles, C.; Nicolas, M.; Jocelyne, M.; Nicole, P.; Bruno, B.; Philippe, Z.; Pierre-Louis, C. Renal sarcoid-like granulomatosis during anti-TNF therapy. Kidney Int. 2014, 86, 215. [Google Scholar] [CrossRef]
  147. Wilkins, C.S.; Ashourian, K.T.; Sobol, E.K.; Fink, M.; Saltzman, B.; Teich, S. Sarcoid-Associated Bilateral Multifocal Choroiditis Secondary to Adalimumab. J. Curr. Ophthalmol. 2021, 33, 205–208. [Google Scholar] [CrossRef]
  148. Kanellopoulou, T.; Filiotou, A.; Kranidioti, H.; Dourakis, S.P. Sarcoid-like granulomatosis in patients treated with anti-TNFα factors. A case report and review of the literature. Clin. Rheumatol. 2011, 30, 581–583. [Google Scholar] [CrossRef]
  149. Leventaki, V.; Flerlage, J. Sarcoid-like granulomatosis in a patient receiving anti-TNFα for psoriasis. Am. J. Hematol. 2018, 93, 722–723. [Google Scholar] [CrossRef]
  150. Kotze, P.G.; De Barcelos, I.F.; Da Silva Kotze, L.M. Sarcoidosis during therapy with adalimumab in a Crohn’s disease patient: A paradoxical effect. J. Crohns Colitis 2013, 7, e599–e600. [Google Scholar] [CrossRef]
  151. Christoforidou, A.; Goudakos, J.; Bobos, M.; Lefkaditis, E.; Vital, V.; Markou, K. Sarcoidosis-like granulomatosis of the hypopharynx as a complication of anti-TNF therapy. Am. J. Otolaryngol. 2013, 34, 268–272. [Google Scholar] [CrossRef]
  152. Decock, A.; Van Assche, G.; Vermeire, S.; Wuyts, W.; Ferrante, M. Sarcoidosis-Like Lesions: Another Paradoxical Reaction to Anti-TNF Therapy? J. Crohns Colitis 2016, 11, 378–383. [Google Scholar] [CrossRef] [PubMed]
  153. Dhaille, F.; Viseux, V.; Caudron, A.; Dadban, A.; Tribout, C.; Boumier, P.; Clabaut, A.; Lok, C. Cutaneous Sarcoidosis Occurring during Anti-TNF-Alpha Treatment: Report of Two Cases. Dermatology 2010, 220, 234–237. [Google Scholar] [CrossRef] [PubMed]
  154. Kashima, S.; Moriichi, K.; Ando, K.; Ueno, N.; Tanabe, H.; Yuzawa, S.; Fujiya, M. Development of pulmonary sarcoidosis in Crohn’s disease patient under infliximab biosimilar treatment after long-term original infliximab treatment: A case report and literature review. BMC Gastroenterol. 2021, 21, 373. [Google Scholar] [CrossRef]
  155. Numakura, T.; Tamada, T.; Nara, M.; Muramatsu, S.; Murakami, K.; Kikuchi, T.; Kobayashi, M.; Muroi, M.; Okazaki, T.; Takagi, S.; et al. Simultaneous development of sarcoidosis and cutaneous vasculitis in a patient with refractory Crohn’s disease during infliximab therapy. BMC Pulm. Med. 2016, 16, 30. [Google Scholar] [CrossRef] [PubMed]
  156. Fuentes-Valenzuela, E.; Navarro Cañadas, C.; Oyarzún Bahamonde, E.; Moreta Rodriguez, M.; Barrio, J. Severe sarcoidosis-like reaction in a patient with Crohn’s Disease treated with infliximab. Any relationship? Clin. Res. Hepatol. Gastroenterol. 2021, 45, 101696. [Google Scholar] [CrossRef]
  157. Gîlcă, G.-E.; Diaconescu, S.; Bălan, G.G.; Timofte, O.; Ştefănescu, G. Sarcoidosis associated with infliximab therapy in ulcerative colitis: A case report. Medicine 2017, 96, e6156. [Google Scholar] [CrossRef]
  158. Robicheaux Clementine, R.; Lyman, J.; Zakem, J.; Mallepalli, J.; Lindsey, S.; Quinet, R. Tumor Necrosis Factor-Alpha Antagonist-Induced Sarcoidosis. JCR J. Clin. Rheumatol. 2010, 16, 274–279. [Google Scholar] [CrossRef]
  159. O’shea, F.D.; Marras, T.K.; Inman, R.D. Pulmonary sarcoidosis developing during infliximab therapy. Arthritis Care Res. 2006, 55, 978–981. [Google Scholar] [CrossRef]
  160. Josse, S.; Klemmer, N.; Moreno-Swirc, S.; Goëb, V.; Lequerré, T.; Vittecoq, O. Infliximab induced skin and pulmonary sarcoidosis in a rheumatoid arthritis patient. Jt. Bone Spine 2009, 76, 718–719. [Google Scholar] [CrossRef]
  161. Olivier, A.; Gilson, B.; Lafontaine, S.; Pautot, J.-X.; Bindi, P. Sarcoïdose pneumo-rénale apparue sous anti-TNFα. Rev. Médecine Interne 2012, 33, e25–e27. [Google Scholar] [CrossRef]
  162. Takahashi, H.; Kaneta, K.; Honma, M.; Ishida-Yamamoto, A.; Ashida, T.; Kohgo, Y.; Ohsaki, Y.; Iizuka, H. Sarcoidosis during infliximab therapy for Crohn’s disease. J. Dermatol. 2010, 37, 471–474. [Google Scholar] [CrossRef]
  163. Fok, K.C.; Ng, W.W.S.; Henderson, C.J.A.; Connor, S.J. Cutaneous sarcoidosis in a patient with ulcerative colitis on infliximab. J. Crohns Colitis 2012, 6, 708–712. [Google Scholar] [CrossRef]
  164. Almodovar, R.; Izquierdo, M.; Zarco, P.; Quirós, F.J.; Mazzucchelli, R.; Steen, B. Pulmonary sarcoidosis in a patient with ankylosing spondylitis treated with infliximab. Clin. Exp. Rheumatol. 2007, 25, 99. [Google Scholar] [PubMed]
  165. Massara, A.; Cavazzini, L.; La Corte, R.; Trotta, F. Comment on: Sarcoid-like granulomatosis in patients treated with tumour necrosis factor blockers: 10 cases. Rheumatology 2010, 49, 1019–1021. [Google Scholar] [CrossRef] [PubMed]
  166. Sakai, H.; Nomura, W.; Sugawara, M. Certolizumab Pegol-Induced Folliculitis-Like Lichenoid Sarcoidosis in a Patient with Rheumatoid Arthritis. Case Rep. Dermatol. 2017, 9, 158–163. [Google Scholar] [CrossRef]
  167. Poulikakos, P.I.; Sullivan, R.J.; Yaeger, R. Molecular Pathways and Mechanisms of BRAF in Cancer Therapy. Clin. Cancer Res. 2022, 28, 4618–4628. [Google Scholar] [CrossRef]
  168. Mai, R.; Zhou, S.; Zhong, W.; Rong, S.; Cong, Z.; Li, Y.; Xie, Q.; Chen, H.; Li, X.; Liu, S.; et al. Therapeutic efficacy of combined BRAF and MEK inhibition in metastatic melanoma: A comprehensive network meta-analysis of randomized controlled trials. Oncotarget 2015, 6, 28502–28512. [Google Scholar] [CrossRef] [PubMed]
  169. Flaherty, K.T.; Infante, J.R.; Daud, A.; Gonzalez, R.; Kefford, R.F.; Sosman, J.; Hamid, O.; Schuchter, L.; Cebon, J.; Ibrahim, N.; et al. Combined BRAF and MEK Inhibition in Melanoma with BRAF V600 Mutations. N. Engl. J. Med. 2012, 367, 1694–1703. [Google Scholar] [CrossRef]
  170. Lacouture, M.; Sibaud, V. Toxic Side Effects of Targeted Therapies and Immunotherapies Affecting the Skin, Oral Mucosa, Hair, and Nails. Am. J. Clin. Dermatol. 2018, 19, 31–39. [Google Scholar] [CrossRef]
  171. Wan, P.T.C.; Garnett, M.J.; Roe, S.M.; Lee, S.; Niculescu-Duvaz, D.; Good, V.M.; Project, C.G.; Jones, C.M.; Marshall, C.J.; Springer, C.J.; et al. Mechanism of Activation of the RAF-ERK Signaling Pathway by Oncogenic Mutations of B-RAF. Cell 2004, 116, 855–867. [Google Scholar] [CrossRef]
  172. Assan, F.; Schlemmer, F.; Assie, J.-B.; Mahevas, M.; Sustronck, P.; Ortonne, N.; Velter, C.; Fardet, L.; Zehou, O. Atypical systemic sarcoid-like granulomatosis in two patients treated with BRAF and MEK inhibitors. Eur. J. Dermatol. 2019, 29, 556–557. [Google Scholar] [CrossRef]
  173. Pham, J.P.; Star, P.; Phan, K.; Loh, Y.; Joshua, A.M.; Smith, A. BRAF inhibition and the spectrum of granulomatous reactions. J. Am. Acad. Dermatol. 2022, 87, 605–613. [Google Scholar] [CrossRef] [PubMed]
  174. Hui Ong, E.L.; Sinha, R.; Jmor, S.; Fearfield, L. BRAF Inhibitor–Associated Granulomatous Dermatitis: A Report of 3 Cases. Am. J. Dermatopathol. 2019, 41, 214–217. [Google Scholar] [CrossRef]
  175. Boutros, A.; Schiavi, C.; Cecchi, F.; Spagnolo, F.; Guadagno, A.; Tanda, E.T.; Giusti, F.; Murdaca, G.; Queirolo, P. Case Report: Immune-Related Toxicity During Adjuvant Treatment with BRAF Plus MEK Inhibitors in a Melanoma Patient. Front. Immunol. 2020, 11, 579523. [Google Scholar] [CrossRef] [PubMed]
  176. Park, J.J.; Hawryluk, E.B.; Tahan, S.R.; Flaherty, K.; Kim, C.C. Cutaneous Granulomatous Eruption and Successful Response to Potent Topical Steroids in Patients Undergoing Targeted BRAF Inhibitor Treatment for Metastatic Melanoma. JAMA Dermatol. 2014, 150, 307. [Google Scholar] [CrossRef]
  177. Leal, L.; Agut-Busquet, E.; Romani, J.; Sabat, M.; Yebenes, M.; Saez, A.; Luelmo, J. Cutaneous granulomatous panniculitis and sarcoidal granulomatous papular eruption in a patient with metastatic melanoma treated with a BRAF inhibitor. J. Dermatol. 2016, 43, 715–716. [Google Scholar] [CrossRef]
  178. Huynh, S.; Lheure, C.; Franck, N.; Goldman-Lévy, G.; Aractingi, S.; Dupin, N.; Kramkimel, N.; Guégan, S. Induced sarcoid-like reactions in patients with metastatic melanoma treated with dabrafenib and trametinib: A monocentric retrospective study. Melanoma Res. 2020, 30, 317–320. [Google Scholar] [CrossRef] [PubMed]
  179. Bala, V.-M.; Mitsogianni, M.; Laschos, K.; Pliakou, E.; Lazaridi, E.; Lampropoulou, D.-I.; Aravantinos, G. Mediastinal and hilar sarcoid-like reaction in a patient treated with dabrafenib and trametinib for metastatic melanoma: A case report and review of the literature. Mol. Clin. Oncol. 2022, 16, 99. [Google Scholar] [CrossRef] [PubMed]
  180. Green, J.S.; Norris, D.A.; Wisell, J. Novel cutaneous effects of combination chemotherapy with BRAF and MEK inhibitors: A report of two cases. Br. J. Dermatol. 2013, 169, 172–176. [Google Scholar] [CrossRef]
  181. Morgese, F.; Cognigni, V.; Scortichini, L.; Ranallo, N.; Lunerti, V.; Migliore, A.; Tronconi, F.; Berardi, R. Potential immune-related adverse events during dabrafenib and trametinib treatment: A case series of patients with BRAF V600E melanoma. Mol. Clin. Oncol. 2022, 18, 2. [Google Scholar] [CrossRef]
  182. Eser Öztürk, H.; Süllü, Y. Sarcoid-like Granulomatous Intraocular Inflammation Caused by Vemurafenib Treatment for Metastatic Melanoma. Turk. J. Ophthalmol. 2020, 50, 50–52. [Google Scholar] [CrossRef] [PubMed]
  183. Das, A.; Wilhelmsson, M.; Foo, J.C.; Yeung, R.S.M.; Krtizinger, F.; McKeown, T.; Coblentz, A.; Ertl-Wagner, B.; Tabori, U.; Bartels, U. Sarcoid-like reaction in a child following prolonged therapeutic exposure to dabrafenib and trametinib for BRAF V600E mutated hypothalamic/chiasmatic glioma. Pediatr. Blood Cancer, 2023; epub ahead of print. [Google Scholar] [CrossRef]
  184. Adam, A.; Thomas, L.; Bories, N.; Zaharia, D.; Balme, B.; Freymond, N.; Dalle, S. Sarcoidosis associated with vemurafenib. Br. J. Dermatol. 2013, 169, 206–208. [Google Scholar] [CrossRef] [PubMed]
  185. Lheure, C.; Kramkimel, N.; Franck, N.; Laurent-Roussel, S.; Carlotti, A.; Queant, A.; Goldwasser, F.; Avril, M.-F.; Dupin, N. Sarcoidosis in Patients Treated with Vemurafenib for Metastatic Melanoma: A Paradoxical Autoimmune Activation. Dermatology 2015, 231, 378–384. [Google Scholar] [CrossRef]
  186. Rubio-Rivas, M.; Moreira, C.; Marcoval, J. Sarcoidosis related to checkpoint and BRAF/MEK inhibitors in melanoma. Autoimmun. Rev. 2020, 19, 102587. [Google Scholar] [CrossRef] [PubMed]
  187. Anastasopoulou, A.; Diamantopoulos, P.T.; Skalioti, C.; Liapis, G.; Psychogiou, E.; Ziogas, D.C.; Gogas, H. The diagnosis and management of sarcoid-like reactions in patients with melanoma treated with BRAF and MEK inhibitors. A case series and review of the literature. Ther. Adv. Med. Oncol. 2021, 13, 17588359211047349. [Google Scholar] [CrossRef]
  188. Durcan, R.; Heffron, C.; Sweeney, B. Natalizumab induced cutaneous sarcoidosis-like reaction. J. Neuroimmunol. 2019, 333, 476955. [Google Scholar] [CrossRef]
  189. Parisinos, C.A.; Lees, C.W.; Wallace, W.A.H.; Satsangi, J. Sarcoidosis complicating treatment with natalizumab for Crohn’s disease. Thorax 2011, 66, 1109–1110. [Google Scholar] [CrossRef]
  190. Shono, Y.; Kamata, M.; Takeoka, S.; Ikawa, T.; Tateishi, M.; Fukaya, S.; Hayashi, K.; Fukuyasu, A.; Tanaka, T.; Ishikawa, T.; et al. Cutaneous sarcoidosis in a patient with rheumatoid arthritis receiving tocilizumab. J. Dermatol. 2018, 45, e217–e218. [Google Scholar] [CrossRef]
  191. Lambert, N.; Hansen, I.; El Moussaoui, M.; Giot, J.; Vercheval, C.; Lommers, É.; Somja, J.; Moutschen, M.; Maquet, P. Lung and liver sarcoidosis-like reaction induced by tocilizumab. Br. J. Clin. Pharmacol. 2021, 87, 4848–4852. [Google Scholar] [CrossRef]
  192. Del Giorno, R.; Iodice, A.; Mangas, C.; Gabutti, L. New-onset cutaneous sarcoidosis under tocilizumab treatment for giant cell arteritis: A quasi-paradoxical adverse drug reaction. Case report and literature review. Ther. Adv. Musculoskelet. Dis. 2019, 11, 1759720X19841796. [Google Scholar] [CrossRef]
  193. Bustamente, L.; Buscot, M.; Marquette, C.-H.; Roux, C. Sarcoidosis and tocilizumab: Is there a link? Clin. Exp. Rheumatol. 2017, 35, 716. [Google Scholar]
  194. Nutz, A.; Pernet, C.; Combe, B.; Cohen, J.-D. Sarcoidosis Induced by Tocilizumab: A Paradoxical Event? J. Rheumatol. 2013, 40, 1773–1774. [Google Scholar] [CrossRef] [PubMed]
  195. Theodosiou, G.; Luu, H.; Svensson, Å. Tocilizumab-induced sarcoidosis-like reaction in a patient with giant cell arteritis. Clinical implications of a paradoxical phenomenon. Int. J. Dermatol. 2020, 59, 888–889. [Google Scholar] [CrossRef] [PubMed]
  196. Glanowski, C.; Clouzeau, A.; Bialé, L.; Banal, F.; Lechevalier, D. Une sarcoïdose sous tocilizumab: À propos d’un cas. Rev. Médecine Interne 2018, 39, 834. [Google Scholar] [CrossRef]
  197. Pescitelli, L.; Emmi, G.; Tripo, L.; Lazzeri, L.; Urban, M.L.; Silvesri, E.; Vannucchi, M.; Prignano, F. Cutaneous sarcoidosis during rituximab treatment for microscopic polyangiitis: An uncommon adverse effect? Eur. J. Dermatol. 2017, 27, 667–668. [Google Scholar] [CrossRef]
  198. Cordeiro, R.A.; De Oliveira, J.L.; Ferraciolli, S.F.; Guedes, L.K.N.; Pasoto, S.G. Neurosarcoidosis during the treatment of primary Sjögren’s syndrome: Is it a paradoxical effect of rituximab? Clin. Exp. Rheumatol. 2022, 40, 2464–2465. [Google Scholar] [CrossRef]
  199. Kornacker, M.; Kraemer, A.; Leo, E.; Ho, A. Occurrence of sarcoidosis subsequent to chemotherapy for non-Hodgkin’s lymphoma: Report of two cases. Ann. Hematol. 2002, 81, 103–105. [Google Scholar] [CrossRef]
  200. Fakih, O.; Verhoeven, F.; Prati, C.; Wendling, D. Paradoxical Löfgren’s syndrome in a patient treated with rituximab: Interferon is not the key. Rheumatology 2020, 59, 1181–1182. [Google Scholar] [CrossRef]
  201. Mrabet, S.; Dahmene, R.; Fradi, A.; Jaziri, A.; Boukadida, R.; Azzebi, A.; Sahtout, W.; Ben Aicha, N.; Zellama, D.; Achour, A.; et al. Sarcoid-Like Reaction in the Kidney Following Rituximab for Mantle Lymphoma in a 60-Year-Old Man. Am. J. Mens Health 2023, 17, 15579883231159343. [Google Scholar] [CrossRef]
  202. Ozer, O.; Eskazan, A.E.; Ar, M.C.; Beköz, H.; Tabak, F.; Ongen, G.; Ferhanoglu, B. Sarcoidosis mimicking lymphoma on positron emission tomography-computed tomography in two patients treated for lymphoma: Two case reports. J. Med. Case Rep. 2009, 3, 7306. [Google Scholar] [CrossRef]
  203. Li, W.; Holdeman, K.; Laurini, J.A.; Bierman, P.J.; Vose, J.M.; Bociek, R.G.; Armitage, J.O. Sarcoidosis mimicking recurrent lymphoma. Am. J. Hematol. 2012, 87, 711–712. [Google Scholar] [CrossRef] [PubMed]
  204. London, J.; Grados, A.; Fermé, C.; Charmillon, A.; Maurier, F.; Deau, B.; Crickx, E.; Brice, P.; Chapelon-Abric, C.; Haioun, C.; et al. Sarcoidosis Occurring After Lymphoma: Report of 14 Patients and Review of the Literature. Medicine 2014, 93, e121. [Google Scholar] [CrossRef]
  205. Vesely, N.C.; Thomas, R.M.; Rudnick, E.; Longo, M.I. Scar sarcoidosis following rituximab therapy. Dermatol. Ther. 2020, 33, e13693. [Google Scholar] [CrossRef] [PubMed]
  206. Choo, S.P.; Lim, D.W.T.; Lo, C.P.L.; Koong, H.N.; Tao, M.; Lim, C.H. Variable Problems in Lymphomas: CASE 1. Burkitt’s Lymphoma Presenting with Central Airway Obstruction. J. Clin. Oncol. 2005, 23, 8112–8113. [Google Scholar] [CrossRef] [PubMed]
  207. Powell, J.B.; Matthews, P.; Rattehalli, R.; Woodhead, F.; Perkins, P.; Powell, G.; Szczecinska, W.; Gach, J.E. Acute systemic sarcoidosis complicating ustekinumab therapy for chronic plaque psoriasis. Br. J. Dermatol. 2015, 172, 834–836. [Google Scholar] [CrossRef]
  208. Gad, M.M.; Bazarbashi, N.; Kaur, M.; Gupta, A. Sarcoid- like Phenomenon—Ustekinumab induced granulomatous reaction mimicking diffuse metastatic disease: A case report and review of the literature. J. Med. Case Rep. 2019, 13, 257. [Google Scholar] [CrossRef]
  209. Kobak, S.; Semiz, H. Ustekinumab-induced Sarcoidosis in a Patient with Psoriatic Arthritis. Curr. Drug Saf. 2020, 15, 163–166. [Google Scholar] [CrossRef]
  210. Kogan, D.; Newton, K.; Pineda, E.; Evans, A.; Guo, H.; Azumi, N.; Cohen, P.; Reichner, C.; Anderson, E. A Case of Trastuzumab-Induced Pulmonary Sarcoidosis. Chest 2012, 142, 1037A. [Google Scholar] [CrossRef]
  211. Panagiotidis, E.; Paschali, A.; Andreadou, A.; Chatzipavlidou, V. HER-2 breast cancer treatment induced mediastinal sarcoid like reaction depicted on 18F-FDG PET/CT. Hell. J. Nucl. Med. 2021, 24, 159–160. [Google Scholar]
  212. Halabi, R.; Grossman, C. Trastuzumab Induced Sarcoidosis Mimicking Metastatic Carcinoma. Chest 2011, 140, 56A. [Google Scholar] [CrossRef]
  213. Tsitos, S.; Niederauer, L.C.; Albert I Gracenea, P.; Mueller, J.; Straube, A.; Von Baumgarten, L. Case Report: Drug-Induced (Neuro) Sarcoidosis-Like Lesion Under IL4 Receptor Blockade with Dupilumab. Front. Neurol. 2022, 13, 881144. [Google Scholar] [CrossRef] [PubMed]
  214. Belhomme, N.; Gaignon, T.; Jouneau, S.; Misery, L.; Abasq-Thomas, C.; Cador, B.; Lecureur, V.; Cadiou, S.; Ballerie, A.; Polard, E.; et al. Drug-induced granulomatosis: Is dupilumab the new kid on the block? J. Eur. Acad. Dermatol. Venereol. 2020, 34, e312–e313. [Google Scholar] [CrossRef]
  215. Saito, S.; Shimizu, M.; Miyashita, R.; Inoue, T.; Nagano, A.; Kunugi, S.; Hatori, T.; Okano, T.; Seike, M. Successful immunosuppressant-free treatment of a drug-induced sarcoidosis-like reaction caused by dupilumab. Respir. Med. Case Rep. 2023, 44, 101883. [Google Scholar] [CrossRef]
  216. Bauer, L.; Müller, L.J.; Volkers, S.M.; Heinrich, F.; Mashreghi, M.-F.; Ruppert, C.; Sander, L.E.; Hutloff, A. Follicular Helper–like T Cells in the Lung Highlight a Novel Role of B Cells in Sarcoidosis. Am. J. Respir. Crit. Care Med. 2021, 204, 1403–1417. [Google Scholar] [CrossRef] [PubMed]
  217. Gleue, C.A.; Shah, K.; Wentworth, A.; Bridges, A. Cutaneous sarcoid-like drug reaction caused by an anaplastic lymphoma kinase inhibitor. J. Cutan. Pathol. 2021, 48, 425–428. [Google Scholar] [CrossRef] [PubMed]
  218. Facchinetti, F.; Gnetti, L.; Balestra, V.; Silva, M.; Silini, E.M.; Ventura, L.; Majori, M.; Bordi, P.; Tiseo, M. Sarcoid-like reaction mimicking disease progression in an ALK-positive lung cancer patient receiving lorlatinib. Investig. New Drugs 2019, 37, 360–363. [Google Scholar] [CrossRef]
  219. Morin, C.; Villeneuve, T.; Norkowski, E.; Rosencher, L.; Cadranel, J.; Mazières, J. Sarcoid-like reaction related to ALK-ROS inhibitors in lung cancer patients. Respir. Med. Res. 2024, 86, 101138. [Google Scholar] [CrossRef]
  220. Paterson, D.L.; Patel, A. Bacillus Calmette-Guérin (BCG) immunotherapy for bladder cancer: Review of complications and their treatment. Aust. N. Z. J. Surg. 1998, 68, 340–344. [Google Scholar] [CrossRef]
  221. De Diego, A.; Rogado, M.C.; Prieto, M.; Nauffal, D.; Perpiñá, M. Disseminated Pulmonary Granulomas after Intravesical Bacillus Calmette-Guérin Immunotherapy. Respiration 1997, 64, 304–306. [Google Scholar] [CrossRef]
  222. Cannon, G.W. Methotrexate pulmonary toxicity. Rheum. Dis. Clin. N. Am. 1997, 23, 917–937. [Google Scholar] [CrossRef]
  223. Imokawa, S.; Colby, T.V.; Leslie, K.O.; Helmers, R.A. Methotrexate pneumonitis: Review of the literature and histopathological findings in nine patients. Eur. Respir. J. 2000, 15, 373. [Google Scholar] [CrossRef] [PubMed]
  224. Zisman , D.A.; McCune, W.J.; Tino, G.; Lynch, J.P. Drug-induced pneumonitis: The role of methotrexate. Sarcoidosis Vasc. Diffus. Lung Dis. 2001, 18, 243–252. [Google Scholar]
Figure 1. Mechanism of granuloma formation. An unknown antigen (Ag) activates alveolar macrophages (AMs), which begin to produce TNF-α, and dendritic cells (DCs), which migrate to the draining lymph nodes and secrete IL-12 and IL-1. These cytokines promote the differentiation and expansion of Th1 and Th17 lymphocytes, producers of IFN-γ and IL-2, further amplifying the immune response. TNF-α also stimulates natural killer (NK) cells to release IFN-γ, enhancing macrophage activation. Activated macrophages upregulate adhesion molecules (e.g., ICAM-1) and secrete a wide array of pro-inflammatory mediators, including IL-1β, TNF-α, CCL20, IL-12, IL-18, IL-23, IL-17, and TGF-β, contributing to a sustained inflammatory environment. This cytokine-rich environment recruits and organizes specific immune cells, such as Th1 and Th17 cells, B cells, monocytes, and regulatory T cells (Tregs), promoting the formation and maintenance of the granuloma. Created in BioRender. Rubini, S. (2025) https://BioRender.com/c1p8l3p (accessed on 8 May 2025).
Figure 1. Mechanism of granuloma formation. An unknown antigen (Ag) activates alveolar macrophages (AMs), which begin to produce TNF-α, and dendritic cells (DCs), which migrate to the draining lymph nodes and secrete IL-12 and IL-1. These cytokines promote the differentiation and expansion of Th1 and Th17 lymphocytes, producers of IFN-γ and IL-2, further amplifying the immune response. TNF-α also stimulates natural killer (NK) cells to release IFN-γ, enhancing macrophage activation. Activated macrophages upregulate adhesion molecules (e.g., ICAM-1) and secrete a wide array of pro-inflammatory mediators, including IL-1β, TNF-α, CCL20, IL-12, IL-18, IL-23, IL-17, and TGF-β, contributing to a sustained inflammatory environment. This cytokine-rich environment recruits and organizes specific immune cells, such as Th1 and Th17 cells, B cells, monocytes, and regulatory T cells (Tregs), promoting the formation and maintenance of the granuloma. Created in BioRender. Rubini, S. (2025) https://BioRender.com/c1p8l3p (accessed on 8 May 2025).
Diagnostics 15 01658 g001
Figure 2. Mechanism of action of immune checkpoint inhibitors (ICIs). ICIs work by blocking inhibitory pathways that suppress immune responses. Ipilimumab targets CTLA-4, which is expressed on activated T cells in lymphoid organs. Nivolumab and Pembrolizumab target PD-1, which is found on T cells in peripheral tissues. Atezolizumab, Durvalumab, and Avelumab target PD-L1, which is expressed on both tumor cells and immune cells. Created in BioRender. Rubini, S. (2025) https://BioRender.com/0s4w3t6 (accessed on 9 May 2025).
Figure 2. Mechanism of action of immune checkpoint inhibitors (ICIs). ICIs work by blocking inhibitory pathways that suppress immune responses. Ipilimumab targets CTLA-4, which is expressed on activated T cells in lymphoid organs. Nivolumab and Pembrolizumab target PD-1, which is found on T cells in peripheral tissues. Atezolizumab, Durvalumab, and Avelumab target PD-L1, which is expressed on both tumor cells and immune cells. Created in BioRender. Rubini, S. (2025) https://BioRender.com/0s4w3t6 (accessed on 9 May 2025).
Diagnostics 15 01658 g002
Figure 3. Mechanism of action of anti-tumor necrosis factor (Anti-TNF) agents. Anti-TNF agents selectively bind to TNF-α, inhibiting its interaction with TNFR1 (p55) and thereby preventing the activation of NF-kB, MAPK, and apoptotic pathways. Additionally, they block TNF-α from binding to TNFR2 (p75), leading to a significant increase in Treg cells, which can produce TGF-β, contributing to immune regulation and granuloma formation.
Figure 3. Mechanism of action of anti-tumor necrosis factor (Anti-TNF) agents. Anti-TNF agents selectively bind to TNF-α, inhibiting its interaction with TNFR1 (p55) and thereby preventing the activation of NF-kB, MAPK, and apoptotic pathways. Additionally, they block TNF-α from binding to TNFR2 (p75), leading to a significant increase in Treg cells, which can produce TGF-β, contributing to immune regulation and granuloma formation.
Diagnostics 15 01658 g003
Figure 4. Mechanism of action of BRAF and MEK inhibitors. BRAF inhibitors, often combined with MEK inhibitors, target the mutant BRAF^V600E protein, blocking the MAPK signaling pathway. This leads to altered cytokine profiles, increased expression of immune-stimulatory molecules (e.g., CD40L, IFNγ), and reduced levels of immunosuppressive cells, such as regulatory T cells and myeloid-derived suppressor cells (MDSCs). Created in BioRender. Rubini, S. (2025) https://BioRender.com/6eaaz3x (accessed on 9 May 2025).
Figure 4. Mechanism of action of BRAF and MEK inhibitors. BRAF inhibitors, often combined with MEK inhibitors, target the mutant BRAF^V600E protein, blocking the MAPK signaling pathway. This leads to altered cytokine profiles, increased expression of immune-stimulatory molecules (e.g., CD40L, IFNγ), and reduced levels of immunosuppressive cells, such as regulatory T cells and myeloid-derived suppressor cells (MDSCs). Created in BioRender. Rubini, S. (2025) https://BioRender.com/6eaaz3x (accessed on 9 May 2025).
Diagnostics 15 01658 g004
Figure 5. Diagnostic algorithm.
Figure 5. Diagnostic algorithm.
Diagnostics 15 01658 g005
Table 1. Immune checkpoint inhibitors.
Table 1. Immune checkpoint inhibitors.
DrugMechanism of ActionTreatment IndicationDISR Cases
(n)
Median Time of Onset (months)Number of Patients
With Signs and SymptomsRequiring Drug DiscontinuationRequiring TreatmentShowing Resolution
PembrolizumabTargets and blocks PD-1 ligandBCA, ccRCC, CRC, cSCC, EAC, NPC, NSCLC, MM, NHL, SARC, UC, ULMS427.3223
1 NR
17
8 NR
9
8 NR
26
8 NR
NivolumabTargets and blocks PD-1 ligandccRCC, EM, GBM, MM, NSCLC, OM, PC213.913
4 NR
101013
4 NR
IpilumabInhibits CTLA-4 pathwayMM206.3715
1 NR
13
2 NR
9
2 NR
18
1 NR
Nivolumab
+
Ipilumab
CCOC, ccRCC, CRC, HCC, NSCLC, MM, RCC, UC164.1512
3 NR
5
1 NR
11
1 NR
14
1 NR
AtezolizumabBinds PDL-1 ligand, targeting PD-1 pathwayBCA, HCC, UC43.452324
DurvalumabBinds PDL-1 ligand, targeting PD-1 pathwayNSCLC3NS2223
AvelumabBinds PD-L1 and B7-1 receptors on T cellsOM1NS0101
SintilimabTargets and blocks PD-1 ligandESCC1NS1011
Notes: BCA: breast adenocarcinoma, CCOC: clear cell ovarian cancer, ccRCC: clear cell renal carcinoma, CRC: colon rectal adenocarcinoma, cSCC: cutaneous squamous cell carcinoma, EAC: endometrial adenocarcinoma, EM: esophageal melanoma, ESCC: esophageal squamous cell carcinoma, GBM: glioblastoma, HCC: hepatocellular carcinoma, MM: melanoma, NHL: non-Hodgkin lymphoma, NPC: nasopharyngeal carcinoma, NS: not significant, NSCLC: non-small cell lung cancer, NR: not reported, OM: ocular melanoma, PC: pleiotropic carcinoma, RCC: renal cell carcinoma, SARC: sarcoma, UC: urothelial carcinoma, ULMS: uterine leiomyosarcoma.
Table 2. TNF-α inhibitors.
Table 2. TNF-α inhibitors.
DrugMechanism of ActionTreatment IndicationDISR Cases
(n)
Median Time of Onset (months)Number of Patients
With Signs and
Symptoms
Requiring Drug Discontinuation Requiring TreatmentShowing Resolution
EtanerceptBinds TNFR2 (p75)AS, RA, CPP, JIA, JRA, PsA, SS48
1 (+ ADA)
1 (+ IFX)
1 (+ MTX)
6 (+ PDN)
305436
5 *
2 **
1 (+ IFX)
1 NR
42
1 NR
55
AdalimumabBlocks TNF-α–TNFR1/2 interactionRA, AS, BD, CD, CPP, JIA, JRA, PsA, PSO, SAPHO, UC30
1 (+ ETN)
15.242828
1 ***
2731
InfliximabBinds to soluble and transmembrane forms of TNF-αRA, AS, CD, PsA, UC, UP214117
2 NR
1513
2 NR
21
GolimumabBinds and inhibits soluble and transmembrane form of TNF-αAS1NS1111
CertolizumabTarget the activation of TNF-αRA1NS1111
Notes: ADA: Adalimumab, AS: ankylosing spondylitis, BD: Behçet’s disease, CD: Crohn’s disease, CPP: chronic plaque psoriasis, ETN: Etanercept, JIA: juvenile idiopathic arthritis, JRA: juvenile rheumatoid arthritis, IFX: Infliximab, MTX: methotrexate, NR: not reported, NS: not significant, PDN: prednisone, PsA: psoriatic arthritis, PSO: psoriasis, RA: rheumatoid arthritis, SAPHO: synovitis acne pustulosis hyperostosis and osteitis, SS: Sjogren’s syndrome, UC: ulcerative colitis, UP: ulcerative pancolitis. * Switch to adalimumab; ** switch to tocilizumab; *** switch to infliximab.
Table 3. BRAF and MEK inhibitors.
Table 3. BRAF and MEK inhibitors.
DrugMechanism of ActionTreatment IndicationDISR Cases
(n)
Median Time of Onset (months)Number of Patients
With Signs and SymptomsRequiring Drug DiscontinuationRequiring TreatmentShowing Resolution
(A) BRAF inhibitors
VemurafenibCompetitive inhibitor of BRAF V600E mutationMM, LCH8963
1 **
47
DabrafenibSelective BRAF inhibitor targeting ATP-binding siteMM2NS21 **12
(B) BRAF inhibitors and MEK inhibitors
Vemurafenib +
Cobimetinib
Inhibition of MAPK pathways *MM3NS32 **23
Dabrafenib +
Trametinib
Inhibition of MAPK pathways *MM, CG228.9163
2 **
1220
Encorafenib +
Binimetinib
Inhibition of MAPK pathways *MM1NS1011
Notes: CG: chiasmatic glioma, MAPK: mitogen-activated protein kinase, MEK: mitogen-activated protein kinase/extracellular signal-regulated kinase, MM: melanoma, NS: not significant, LCH: Langerhans cell histiocytosis. * Interaction as a competitive inhibitor of BRAF V600E mutation + inhibition of MEK1 and MEK2 proteins. ** Drug stopped and then restarted.
Table 4. Monoclonal antibodies.
Table 4. Monoclonal antibodies.
DrugMechanism of ActionTreatment IndicationDISR Cases
(n)
Median Time of Onset (months)Number of Patients
With Signs and
Symptoms
Requiring Drug DiscontinuationRequiring Treatment Showing Resolution
NatalizumabTargets
the lymphocyte adhesion molecule a4 integrin
CD, MS3NS2221
RituximabTargets CD20 on B lymphocyte cell surfaceMCL, CML, DLBCL, NHL, SS, RA, PV, RMPA161490816
UstekinumabBlocks IL-12 and IL-23Ps3NS3332
TocilizumabBlocks IL-6GCA, RA7132356
DupilumabBlocks IL-4 and IL-13AD, ERS3NS2231
TrastuzumabHER2 receptor inhibitorBreast cancer3NS1111
Notes: AD: atopic dermatitis, CD: Crohn’s disease, CML: chronic myelogenous leukemia, DLBCL: diffuse large B-cell lymphoma, ERS: eosinophilic rhinosinusitis, GCS: giant cell arteritis, MCL: mantle cell lymphoma, MS: multiple sclerosis, NHL: non-Hodgkin lymphoma, NS: not significant, Ps: psoriasis, PV: Pemphigus vulgaris, RMPA: refractory microscopic polyangiitis, SS: Sjögren’s syndrome, RA: rheumatoid arthritis.
Table 5. Miscellaneous drugs.
Table 5. Miscellaneous drugs.
DrugMechanism of ActionTreatment IndicationDISR Cases (n)Median Time of Onset (months)Number of Patients
With Signs and
Symptoms
Requiring Drug DiscontinuationRequiring TreatmentShowing Resolution
(A) Interleukin inhibitor
AbataceptBinds to CD80/D86 on antigen-presenting cells, attenuating T-cell activationRA1NS1011
(B) Tyrosine Kinase inhibitor
LorlatinibBlocks ALK and ROS1
proteins
NSCLC
with ALK
mutation
2NS0100
Notes: RA: rheumatoid arthritis, NS: not significant, NSCLC: non-small cell lung cancer.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Andolfi, F.; Caffarri, L.; Neviani, M.; Rubini, S.; Andrisani, D.; Gozzi, F.; Beghé, B.; Clini, E.; Tonelli, R.; Cerri, S. Drug-Induced Sarcoid-like Reactions Associated to Targeted Therapies and Biologic Agents. Diagnostics 2025, 15, 1658. https://doi.org/10.3390/diagnostics15131658

AMA Style

Andolfi F, Caffarri L, Neviani M, Rubini S, Andrisani D, Gozzi F, Beghé B, Clini E, Tonelli R, Cerri S. Drug-Induced Sarcoid-like Reactions Associated to Targeted Therapies and Biologic Agents. Diagnostics. 2025; 15(13):1658. https://doi.org/10.3390/diagnostics15131658

Chicago/Turabian Style

Andolfi, Federica, Luca Caffarri, Matilde Neviani, Silvia Rubini, Dario Andrisani, Filippo Gozzi, Bianca Beghé, Enrico Clini, Roberto Tonelli, and Stefania Cerri. 2025. "Drug-Induced Sarcoid-like Reactions Associated to Targeted Therapies and Biologic Agents" Diagnostics 15, no. 13: 1658. https://doi.org/10.3390/diagnostics15131658

APA Style

Andolfi, F., Caffarri, L., Neviani, M., Rubini, S., Andrisani, D., Gozzi, F., Beghé, B., Clini, E., Tonelli, R., & Cerri, S. (2025). Drug-Induced Sarcoid-like Reactions Associated to Targeted Therapies and Biologic Agents. Diagnostics, 15(13), 1658. https://doi.org/10.3390/diagnostics15131658

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop