Skip to main content
The Oncologist logoLink to The Oncologist
. 2025 May 11;30(5):oyaf017. doi: 10.1093/oncolo/oyaf017

Sarcoid-like reactions in patients treated with checkpoint inhibitors for advanced solid tumors

Ian Nykaza 1,#, Yonina R Murciano-Goroff 2,3,#, Antoine Desilets 4, Guilherme Harada 5, Michael A Postow 6,7, Margaret K Callahan 8,9, Chung-Han Lee 10,11, Charles M Rudin 12,13, David Paul Kelsen 14,15, Zsofia K Stadler 16, Andreas G Wibmer 17, Jaclyn F Hechtman 18, Alexander Drilon 19,20, Claire F Friedman 21,22,
PMCID: PMC12065934  PMID: 40349135

Abstract

Importance

While new intrathoracic adenopathy in a patient with cancer can represent progression of disease, the differential diagnosis is broad. Sarcoid-like reactions (SLR) remain an underreported source of lymphadenopathy in patients treated with immune checkpoint inhibitors (ICI), with limited reports in patients with cancers other than melanoma.

Objective

To characterize SLRs among patients treated with ICI for advanced solid tumors.

Methods

Data were collected on the clinical, pathologic, and radiographic presentation of patients treated with ICI who developed clinical or imaging findings suggestive of an SLR, including the presence of hilar or mediastinal lymphadenopathy, cutaneous/subcutaneous involvement, and/or worsening of existing sarcoidosis on ICI.

Results

Twelve patients were identified as having experienced an SLR. While 6 patients had melanoma, SLRs were also observed among patients with lung, gynecologic, and genitourinary cancers, including high-grade serous ovarian carcinoma, and an angiomyolipoma. Median time from initiation of ICI to diagnosis of an SLR was 3.4 months (range: 1.8-9.1). All but one patient (92%) were deemed to have had a radiographic response to ICI.

Conclusions and relevance

Clinicians should maintain the awareness of the possibility of SLRs in patients receiving ICI, particularly in patients whose scans show evidence of “mixed” response, with decreases in certain lesions coupled with new/increasing intrathoracic lymphadenopathy and/or other systemic signs of sarcoid.

Keywords: sarcoidosis, sarcoid-like reaction, immune checkpoint inhibitors, immunotherapy


Implications for Practice.

This study emphasizes the importance of recognizing sarcoid-like reactions (SLRs) as a potential immune-related adverse event that occurs across a variety of tumor types beyond melanoma, including lung, gynecologic, and genitourinary cancers. Misdiagnosis of SLRs as disease progression risks premature discontinuation of effective immune checkpoint inhibitor (ICI) treatment. Clinicians should be aware of key diagnostic features, including a relatively early onset (between 1.8 and 9.1 months) after treatment initiation, mixed radiographic responses (eg, shrinkage of known tumors alongside new lymphadenopathy), and associated extra-thoracic symptoms such as uveitis or subcutaneous nodules. Although biomarkers such as ACE and eosinophilia may raise suspicion, biopsy remains the gold standard, particularly in cases with intrathoracic manifestations that may mimic disease progression. This is especially relevant in lung cancer patients, where distinguishing SLRs from progression is further complicated by comorbidities such as chronic obstructive pulmonary disease.

Notably, SLRs may correlate with improved response to ICIs, as all but one patient in this series demonstrated radiographic benefit without disease progression at the time of SLR diagnosis. Corticosteroids or immunosuppression were effective in managing SLRs in most cases. However, clinicians should balance the risks of immunosuppression against the benefits of continuing ICIs. This underscores the importance of integrating clinical, radiographic, and pathological data into decision-making processes to optimize patient outcomes. Further research is needed to identify more specific biomarkers for SLRs and to explore the underlying immune mechanisms, ensuring that this phenomenon, now recognized as pan-cancer, is managed effectively without compromising antineoplastic therapy.

Introduction

Immune checkpoint inhibitors (ICI) have revolutionized the treatment of many advanced solid tumor types.1,2 Immune checkpoint inhibitors are associated with a diverse set of immune-related adverse effects (irAEs), as well as a unique pattern of disease response known as “pseudo-progression.”2-5 These features of ICI response create diagnostic challenges, as clinicians must distinguish radiographically between inflammatory reactions and true progression of malignancy in order to avoid taking patients off of efficacious therapies prematurely.6,7 Increased lymphadenopathy can be especially difficult to distinguish due to the wide differential of causative etiologies which include inflammatory reactions and disease progression among many other processes. Sarcoid-like reactions (SLRs), which are defined as the development of lesions that clinically and pathologically mirror those seen in patients with sarcoid following the receipt of immunologically active therapies, can lead to the development of benign mediastinal, hilar, or intrapulmonary inflammation. Sarcoid-like reactions have been described both in patients receiving ICI8-29 as well as other immunologically active therapies, including highly active antiretroviral therapy, tumor necrosis factor-α antagonists, and interferon therapy.30-36

Differentiating SLRs from progressive disease is challenging without biopsies. Drug-induced SLRs may also be localized to various organ systems; SLRs attributed to ICI treatment have most commonly been identified in the lung and skin.34-36 Whether SLRs in additional locations are truly less common with ICI or whether other areas are simply harder to biopsy and/or more likely to be confused with disease progression remains uncertain.34-37

Most cases of SLRs reported following receipt of ICI have been in patients with melanoma.36 While the preponderance of melanoma cases in the literature may reflect the early adoption of ICI for this disease, it is also notable that there have been several cases of SLRs reported in patients with melanoma who never received ICI, potentially pointing to unique features of the disease’s biology.35,38,39 In recent years, SLRs have been identified following receipt of ICI for additional malignancies,.8,11,22,27,29,31,40–54 but the number of reported cases remains small and additional investigation is needed to facilitate diagnosis in patients who may have pre-existing malignant hilar and/or mediastinal adenopathy on imaging.

In this paper, we present clinicopathologic data from a series of patients with SLRs following treatment with ICI for a variety of solid tumor types with the aim of investigating distinguishing features of SLRs that can aid clinicians in recognizing the occurrence of these reactions pan-cancer.

Methods

Patients were eligible for inclusion in this case series if they had been seen at Memorial Sloan Kettering Cancer Center between 2015 and 2022 and were identified by their treating physicians as having experienced a possible ICI-associated SLR based on clinical and/or imaging findings. Patients were considered to have an SLR if they had biopsy-proven evidence of sarcoid-like inflammation, including noncaseating granulomas, following receipt of ICI confirmed by a certified pathologist (J.H.). One patient had no tissue available for histopathological assessment; however, this patient had an outside biopsy that was consistent with SLR. Additionally, a patient with a remote diagnosis of sarcoidosis was included in this series given progression of their sarcoidosis in the setting of ICI. This patient had never endorsed any symptoms or required any treatment for their sarcoid prior to receipt of ICI. All scans were reviewed by a certified radiologist (A.W.) specializing in cancer care, including for evidence of hilar and/or mediastinal adenopathy. Clinical and pathologic data were compiled via manual chart review with a data collection cutoff of 7/31/2023. This study was approved by the Institutional Review Board of the Memorial Sloan Kettering Cancer Center.

Results

Patients

We identified 12 patients with SLRs following treatment with ICI, with 11 patients having available tissue for pathologic confirmation (Tables 1-4; Figure 1). Six of these patients were treated for advanced or unresectable melanoma. Additional patients were treated for gynecologic tumors (n = 2), including high-grade serous ovarian carcinoma and primary peritoneal carcinosarcoma; nonsmall cell lung carcinoma (NSCLC) (n = 2) and genitourinary malignancies (n = 2), including clear cell renal carcinoma and epithelioid angiomyolipoma. Treatment regimens included combined PD-1/CTLA4 blockade (n = 7), PD-1 blockade monotherapy (n = 1), PD-1 blockade with carboplatin and paclitaxel (n = 3), or CTLA4 blockade followed by PD-1 blockade (n = 1).

Table 1.

Characteristics of patients with melanoma.

ID Age/Sex Melanoma subtype /Stage Tumor involves media-stinum Immuno-therapy target/clinical response Time from first IO to sarcoid diagnosis (months) Sarcoid involvement/Biopsy indicative of SLR Symptoms of sarcoid ACE AFB Other irAEs Disease status at last follow-up Eosinophilia within 1 month of diagnosis (%) Received further IO given after sarcoid diagnosis Received immune suppression for sarcoid involvement?
1 51 F Unknown primary/M1b /BRAF V600E Yes CTLA4/PD1 × 4/Yes 1.8 Cutaneous, Hilum/mediastinum, lung/Yes Cutaneous plaque N/A (+) Pneumonitis, hypothyroid, colitis No evidence of disease. No further treatment received. Yes (18.8) Yes Received steroids for cutaneous sarcoid
2 60 M Cutaneous/M1a/BRAF V600K, TP53 Yes CTLA4/PD1 × 2/Yes 5.4 Hilum/mediastinum, lung
/Yes
Chronic night sweats 168 (−) Hepatitis, vitiligo Died; cause unknown. No evidence of disease with no further treatment at last follow-up. No No No; received steroids and mycophenolate mofetil for hepatitis
3 76 M Cutaneous/IIIC/NRAS No CTLA4/PD1 × 4/Yes 3.7 Hilum/mediastinum, lung, spleen/Yes Night sweats, blurred vision/uveitis 58 (−) Vitiligo, uveitis, arthralgias No evidence of disease. No further treatment received. No No Received steroids for pulmonary sarcoid involvement
4 58 M Cutaneous/M1b/Unknown mutations Yes CTLA4/PD1 × 1/Yes 3.4 Hilum/mediastinum/Yes Dry cough following steroid taper for pneumo-nitis N/A (−) Pneumonitis Surgical management of disease recurrence 1.6 years after the sarcoid diagnosis. No evidence of disease since. No No Received steroids for granulomatous pneumonitis and pulmonary sarcoid involvement
5 66 M Unknown primary/IIIC/BRAF D594N No PD1 × 4/Adjuvant 3.3 Hilum/mediastinum, enlarged cervical lymph nodes
/Yes
None 42 (+) Arthralgias Died, cause unknown. POD-spine and brain lesions detected roughly 1.3-year postsarcoid diagnosis. Yes (5.2) No No; later received steroids for vertebral compression in context of vertebral metastasis
6 54 F Cutaneous/IIIC/NRAS No CTLA4 × 4, CTLA4 reinduction, PD1 thereafter (sarcoid diagnosed after first cycle)/response assessment not available Unknown Subcutaneous nodules, pulmonary inflammation
/N/A—no biopsy specimen available but outside pathology indicated SLR
Hospitalized for respiratory symptoms. Had subcutaneous nodules. N/A N/A Hepatobiliary toxicity, thyroiditis Increased thoracic lymphadenopathy that cannot be easily biopsied; unclear whether from sarcoid or melanoma. No Yes Received steroids for pulmonary involvement and later for hepatitis

Abbreviations: AFB= acid fast bacilli testing; IO = immunotherapy; irAE = immune-related adverse events; N/A = not available; POD= progression of disease.

Table 4.

Characteristics of patients with pulmonary malignancies.

ID Age/Sex Tumor type/Stage Tumor involves media-stinum Immuno-therapy target/clinical response Time from first IO to sarcoid diagnosis (months) Sarcoid involvement/Biopsy indicative of SLR Symptoms of sarcoid ACE AFB Other irAEs Disease status at last follow-up Eosinophilia within 1 month of diagnosis (%) Received further IO given after sarcoid diagnosis Received immune suppression for sarcoid involvement?
11 61 M Lung adenocarcinoma/IV Yes PD1 × 2 (alone)/Radiographic progression
PD1 × 8 (with chemotherapy), PD1 × 16 (with anti-VEGF), PD1 × 4 (alone)/Yes
9.1 Mediastinal/hi-lar lymph nodes
/Yes
Developed worsening SOB and fatigue over baseline COPD, required hospitalization N/A (−) Pruritis Continues on systemic therapy with stable disease. No Yes Received steroids for COPD exacerbation in setting of pulmonary sarcoid
12 57 F Lung adenocarcinoma/IV Yes CTLA4/PD1 × 2/Yes 4.9 Mediastinal lymph nodes, diffuse subcutaneous nodules
/Yes
Mild cough, fatigue, skin changes, hospitalized due to hypercalce-mia (16mg/dL) and AKI 132 (IO = immunotherapy;) Colitis No evidence of disease, no further treatment received. Yes (6.9) No Received hydroxy-choloroquine for sarcoid management

Abbreviations: AFB= acid fast bacilli testing; IO = immunotherapy; irAE = immune-related adverse events; N/A = not available.

Figure 1.

This figure depicts histopathological and imaging findings of sarcoid-like reactions in patients receiving immune checkpoint inhibitor (ICI) therapy.

Fifty-one y/o F (subject ID 1) with melanoma, showing representative biopsy specimens from (A) tumor and (B) granuloma, as well as (C) Hilar adenopathy on imaging, and 61 y/o M (subject ID 9) with clear cell renal cell carcinoma, with biopsies showing (D) tumor, (E) noncaseating granulomatous inflammation, and (F) imaging showing hilar adenopathy, and 71 y/o F (subject 10) with epithelioid angiomyolipoma with biopsies showing (G) Tumor, (H) Granuloma, and (I) imaging showing adenopathy.

Table 2.

Characteristics of patients with gynecological malignancies.

ID Age/Sex Tumor type/Stage Tumor involves media-stinum Immuno-therapy target/clinical response Time from first IO to sarcoid diagnosis (months) Sarcoid involvement /
Biopsy indicative of SLR
Symptoms of sarcoid ACE AFB Other irAEs Disease status at last follow-up Eosinophilia within 1 month of diagnosis (%) Received further IO given after sarcoid diagnosis Received immune suppression for sarcoid involvement?
7 59 F High grade serous ovarian cancer/IVb Yes PD1 × 7 (with chemotherapy), with maintenance PD1 × 3 thereafter/Yes 3.0 Mediastinal nodes
/Yes
Cough and dyspnea N/A Un-known Lichenoid rash Died from disease No Yes Later received steroids for shortness of breath of unclear etiology
8 67 F History of breast cancer; now with peritoneal high grade carcinosarcoma/IV Borderline mediastinal nodes present at baseline PD1 × 4 (with chemotherapy), and maintenance PD1 × 12 thereafter/Yes -> eventual progression on maintenance Baseline history of asymptomatic sarcoid (previously untreated) Mediastinal/hilar nodes present from diagnosis, pulmonary ground glass opacities.
/Yes
History of shortness of breath; also had large pleural effusions N/A (-) Kidney injury Died from disease *Yes (6.1)—measured after imaging of ground glass opacities attributed to sarcoid reactivation vs SLR Yes No; received steroid taper for kidney injury

Abbreviations: AFB= acid fast bacilli testing; IO = immunotherapy; irAE = immune-related adverse events.

Table 3.

Characteristics of patients with genitourinary malignancies.

ID Age/Sex Tumor type/Stage Tumor involves media-stinum Immuno-therapy target/clinical response Time from first IO to sarcoid diagnosis (months) Sarcoid involvement /
Biopsy indicative of SLR
Symptoms of sarcoid ACE AFB Other irAEs Disease status at last follow-up Eosinophilia within 1 month of diagnosis (%) Received further IO given after sarcoid diagnosis Received immune suppression for sarcoid involvement?
9 61 M Clear cell renal cell carcinoma/IV Yes CTLA4/PD1 × 3/Yes 2.6 Hilum/mediastinum, lung, pleura
/Yes
Baseline pretreatment cough, and was on oxygen at night; developed worsening SOB 73 (−) Pneumonitis Died, cause unknown in setting of COPD and HFpEF exacerbation. Underwent cytoreductive surgery with no pathologic evidence of disease. No further treatment with no radiographic evidence of disease. No No Received steroids for pulmonary sarcoid involvement and pneumonitis
10 71 F Epithelioid Angiomyolipoma Yes CTLA4/PD1 × 4, PD1 × 1/clinical improve-ment, with worsening scans 3.3 Mediastinum, skin, kidney. At the time, noted to have pleural nodularity and peri-splenic soft tissue
/Yes
Worsening SOB, fatigue, skin changes. 72 (−) None Believed to have radiologic progression of disease, but no viable tumor seen on pathology. Yes (5.5) No Received steroids for pulmonary sarcoid involvement

Abbreviations: AFB= acid fast bacilli testing; IO = immunotherapy; irAE = immune-related adverse events; N/A = not available.

One patient had a pre-existing diagnosis of sarcoidosis, identified via lymph node biopsy, in the setting of lymphadenopathy of unknown cause, 15 years prior to receipt of ICI and never previously required treatment. This patient exhibited clinical progression of sarcoid on ICI with the development of pulmonary punctate nodularity and ground glass opacities (GGOs), which had an atypical appearance for metastatic disease. Eleven of the patients who developed an SLR (92%) experienced additional irAEs during treatment, including pneumonitis (n = 3), hepatic and/or biliary inflammation (n = 2), vitiligo (n = 2), arthralgias (n = 2), thyroiditis/hypothyroidism (n = 2), lichenoid rash (n = 1), colitis (n = 2), uveitis (n = 1), pruritis (n = 1), and acute kidney injury (AKI, n = 1).

Clinical features of SLRs

Median time from initiation of ICI to SLR diagnosis was 3.4 months (range: 1.8-9.1) (Figure 2). Eleven of the patients (92%) had symptoms attributable to an SLR, including respiratory symptoms (shortness of breath or cough; n = 8), cutaneous or subcutaneous lesions (n = 3), night sweats (n = 2), and uveitis (n = 2).

Figure 2.

This swimmer plot illustrates the clinical characteristics of sarcoid-like reactions (SLR) in patients undergoing immunotherapy. The X-axis represents time in months since the first immunotherapy dose, while the Y-axis lists individual patients by ID.

Swimmer’s plot showing time from first immunotherapy to sarcoid diagnosis, immune suppressive treatment, and first progression of disease postimmunotherapy. Note that patient ID 4 only received 1 dose of immunotherapy. Subjects who received unpublished experimental therapy, care at an outside facility for which exact dates were unavailable, or who were initially diagnosed with sarcoid prior to commencing immunotherapy are not shown.

On imaging, mediastinal and/or hilar lesions were seen in 11 of the 12 patients (92%), with the remaining patient exhibiting evidence of pneumonitis (Table 1). Nine patients (75%) had baseline pre-ICI treatment imaging findings suggestive of mediastinal and/or hilar tumor involvement and 3 patients (25%) had biopsy-proven mediastinal and/or hilar tumor involvement. All these patients had evolving intrathoracic changes on imaging at the time of their sarcoid diagnosis, including increased pulmonary nodularity, GGOs, and/or more prominent adenopathy. In 7 of these cases, intrathoracic changes correlating with an SLR were first seen on scans showing a decrease in disease burden in other areas, that is, a mixed pattern. An additional patient with a known diagnosis of sarcoidosis at baseline developed increased GGOs during treatment that were not biopsied but were suspected to be inflammatory due to the timing of ICI treatment.

Angiotensin converting enzyme (ACE) levels were available for 6 patients; levels ranged from 42 to 168 (median: 72.5; upper limit of normal [ULN] 67 U/L) and were elevated in 4 of the 6 patients. One patient was hospitalized due to hypercalcemia with a serum calcium concentration of 16 mg/dL. This patient also had an AKI at admission, attributed to hypercalcemia and possible SLR involvement. Eosinophilia, defined as a level greater than the ULN (4.9% at our institution), was present within 1 month of diagnosis in 4 of 10 patient (40%) with available data and without prior history of sarcoidosis. Additionally, the patient with a prior diagnosis of asymptomatic sarcoidosis exhibited eosinophilia after development of GGOs. Four patients had a positive acid-fast bacillus (AFB) stain on bronchoscopy, while 6 had a negative AFB stain and 2 patients had no AFB stain results available.

Management of SLRs

In most cases (58%; n = 7), immunotherapy was permanently discontinued after diagnosis of an SLR. Six patients (50%) had ICI discontinued due to pulmonary SLRs and associated symptoms. One patient (8%) had therapy withheld due to hepatotoxicity at the time of SLR diagnosis and believed to be a separate irAE. All patients who discontinued ICI treatment exhibited radiographic responses to ICI treatment and were placed on active observation after treatment discontinuation. Five patients (42%) continued ICI treatment following the SLR diagnosis.

The median follow-up from SLR diagnosis to the end of study was 43.6 months (range 13.0-94.3 months). Four patients (33%), 3 with melanoma and 1 with NSCLC, exhibited no signs of progression at last follow-up and required no further cancer-directed therapy. One patient with melanoma received surgical management for recurrence 1.6 years after SLR diagnosis and subsequently exhibited no evidence of disease. The 2 patients with genitourinary malignancy also underwent surgical management for possible disease recurrence; however, no pathologic evidence of disease was found in both these cases.

Five patients (42%) eventually developed radiographic progression of disease following ICI. However, one of the patients with suspected radiographic progression was later found to have no evidence of disease on biopsy, with no viable tumor seen on pathology. Four patients (33%) were ultimately determined to have definitive progression of their cancer and 2 patients (17%) remained on systemic chemotherapy as of last follow-up. Two patients died from their disease, while 3 additional patients passed away from unknown causes with limited available records. Two of these patients had no known evidence of disease at the time of death and 1 had known brain and spine metastases.

Overall, 7 patients (67%) received steroids for known, tissue proven SLRs or for pulmonary symptoms and inflammation attributed to an SLR. This included 3 of the 5 patients who continued ICI after the SLR diagnosis. One of the patients who received steroids for shortness of breath had received both ICI and gemcitabine, the latter of which can be associated with pneumonitis as well.55 Additional indications for immunosuppressive treatment included hepatitis (n = 1), immune-mediated AKI (n = 1), vertebral compression fracture (n = 1), and an exacerbation of existing chronic obstructive pulmonary disease (COPD), believed to be triggered by an SLR (n = 1). One patient who continued ICI treatment after SLR diagnosis also received hydroxychloroquine for SLR management after eventual discontinuation of ICI.

Discussion

We present clinicopathologic data from patients with a variety of different tumor types who experienced SLRs following receipt of ICI. Given that SLRs can be radiographically mistaken for progression of disease, this case series affirms the importance of maintaining a high index of suspicion for SLRs. Because the preponderance of the literature on SLRs has focused on patients with melanoma,8-21,23-26,28,29 there has been a suggestion that patients with melanoma may be especially prone to SLRs.35,38,56 The present series highlights that SLRs can occur across a variety of tumor types following ICI and offers additional information on the disease course of patients who develop this treatment-related complication.

Sarcoid-like reactions following ICI are likely underreported. In a single center series of 908 patients who received anti-PD-1 or PD-L1 therapy, with or without CTLA-4 blockade, SLR incidence was estimated to be as low as 0.2% (n = 2).57 However, in a separate single center study analyzing radiographic irAEs in 147 patients with advanced melanoma, up to 5% patients treated with the CTLA-4 blocking agent ipilimumab demonstrated sarcoid-like adenopathy, suggesting a higher incidence of SLRs than previously recognized.12

The present series highlights that SLRs can occur following ICI in gynecologic and genitourinary tumors as well as in primary lung tumors. Sarcoid-like reactions due to ICI treatment have previously been reported in renal cell carcinoma, urothelial, uterine, ovarian, and prostate cancers.22,40–46 This series expands upon this existing landscape by providing descriptions of ICI-associated SLRs in 2 histologies that, to our knowledge, have not previously been reported, namely: epithelioid angiomyolipoma and high-grade serous ovarian carcinoma. We also describe the first case of ICI-associated sarcoid flare in a patient with primary peritoneal carcinosarcoma who had a baseline of asymptomatic sarcoidosis.

In this series, we reported 2 SLRs in patients with lung cancer. It is notable that few lung cases have been previously reported,8,11,58,59 despite the frequent use of ICI for the disease. Moreover, those previously reported have tended to exhibit extrathoracic manifestations of SLR.8,60 Given that both patients with lung cancer in this cohort exhibited intrathoracic manifestations, confirmed by biopsy, it is possible that the dearth of reported SLRs in lung cancers may partly be due to the challenges of differentiating between an SLR and malignant progression in these patients, including due to comorbidities such as COPD in this population.8-11

For clinicians, differentiating between an SLR and progression of disease is critical. Our series highlights several key clinical features that can help make this distinction. First, in our series, cases of an SLR were diagnosed between 1.8 and 9.1 months after starting treatment. Several patients had simultaneous development of extrathoracic symptoms such as uveitis and/or subcutaneous nodules that increased suspicion for an SLR. Patients that solely exhibit intrathoracic manifestations of an SLR are at risk for being missed and/or labeled as having progressive disease. For these patients, mixed radiographic response, such as shrinking of known tumors in the context of new lymphadenopathy, may help raise suspicion.58

Blood-based biomarkers were imperfect for diagnosis in our cohort. For classic sarcoidosis, the most widely used biomarker is serum ACE, which is elevated in 30%-80% of patients.61 A number of other biomarkers, including levels of lysozyme, neopterin, and soluble Interleukin-2 receptor, have been explored but have limited sensitivity and specificity.61 In our series, 4 of 6 patients with available testing had elevated ACE levels. Although elevated ACE levels may raise suspicion for an SLR, biopsy remains the gold standard for SLR diagnosis, as ACE can be elevated in other inflammatory conditions leading to intrathoracic adenopathy such as tuberculosis.61,62 We also looked at eosinophilia as a potential biomarker, as this has been reported in association with sarcoidosis.63 However, excluding the patient with a previous sarcoid diagnosis, eosinophilia was only present in 4 patients. Furthermore, eosinophilia secondary to ICI treatment is well documented, limiting the utility of this measure in identifying SLRs.64 Additional studies are needed to refine biomarkers of SLRs.

In addition to examining diagnostic clues, we also analyzed the disease course of patients with SLRs following ICI. Prior research has established that patients who develop certain irAEs, such as vitiligo, are more likely to benefit from ICI.65 In keeping with a recently published series in which sarcoid-like granulomatosis was associated with improved overall survival, all but one patient in our series was deemed to have radiographic benefit from ICI.26 None of the patients in our series had progression of disease at the time of their SLR. In terms of management, most patients in our cohort received treatment with corticosteroids for SLRs or symptoms thought to be related to the SLR. Many patients also received immunosuppression for additional irAEs at some point following ICI. Such treatments for other irAEs may have also damped down further sarcoid-like inflammation.

This study has several limitations. First, it represents a retrospective, single-center case series. While the number of patients reported is modest, our cohort constitutes one of the largest series of patients with SLRs after immunotherapy reported to date and includes the first reports of SLRs in epithelioid angiomyolipoma and high-grade serous ovarian carcinoma. Second, several patients had pretreatment staging scans with mediastinal and/or hilar adenopathy. While we cannot exclude the possibility that some of these patients had undiagnosed sarcoid prior to receiving immunotherapy, it is also possible that SLRs occurred in areas with contemporaneous tumor involvement. Indeed, in one case, a mediastinal node was excised post-ICI treatment and demonstrated sarcoid-like noncaseating granulomatous inflammation co-existent with malignant cells. Lastly, we cannot rule out the possibility that these patients had other predisposing factors, since even certain chemotherapies have been associated with SLRs and since at least one patient in our series had a prior negative work-up for sarcoid before starting treatment.36

Overall, our pan-cancer series underscores that SLRs can occur in a variety of tumor types beyond melanoma. It also highlights the fact that maintaining an awareness of the possible involvement of SLRs in patients with new or progressive intrathoracic lesions on imaging who otherwise respond to ICI is critical to in order to avoid unnecessary discontinuation of potentially effective antineoplastic therapy. By identifying patients who developed SLRs during treatment for a variety of advanced solid tumors, this report emphasizes that sarcoidosis associated with immunotherapy is a more widespread phenomenon pan-cancer than previously recognized. Further work is necessary to identify potential biomarkers for the development of SLRs and to elucidate underlying immune-mediated mechanisms.

Contributor Information

Ian Nykaza, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.

Yonina R Murciano-Goroff, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States.

Antoine Desilets, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.

Guilherme Harada, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.

Michael A Postow, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States.

Margaret K Callahan, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States.

Chung-Han Lee, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States.

Charles M Rudin, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States.

David Paul Kelsen, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States.

Zsofia K Stadler, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.

Andreas G Wibmer, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.

Jaclyn F Hechtman, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.

Alexander Drilon, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States.

Claire F Friedman, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States.

Author contributions

Ian Nykaza (Data curation, Formal Analysis, Investigation, Writing—review & editing), Yonina R. Murciano-Goroff (Conceptualization, Data curation, Investigation, Methodology, Project administration, Validation, Writing—original draft, Writing—review & editing), Antoine Desilets (Data curation, Formal analysis, Writing—review & editing), Guilherme Harada (Data curation, Investigation, Writing—review & editing), Michael A. Postow (Data curation, Investigation, Writing—review & editing), Margaret K. Callahan (Data curation, Formal analysis, Investigation, Writing—review & editing), Chung-Han Lee (Investigation, Writing—review & editing), Charles M Rudin (Investigation, Methodology, Resources, Writing—review & editing), David Paul Kelsen (Conceptualization, Investigation, Resources, Validation, Writing—review & editing), Zsofia K Stadler (Investigation, Writing—review & editing), Andreas G Wibmer (Investigation, Validation, Writing—review & editing), Jacyln F Hechtman (Investigation, Writing—review & editing), Alexander Drilon (Data curation, Investigation, Resources, Supervision, Writing—review & editing), and Claire F. Friedman (Conceptualization, Data curation, Investigation, Resources, Supervision, Writing—review & editing)

Funding

This work was supported by the National Cancer Institute/National Institutes of Health Cancer Center Support grant to Memorial Sloan Kettering Cancer Center (P30 CA008748).

Ethics approval and consent to participate

The study was conducted in accordance with the Declaration of Helsinki. Study conduct was approved by the Institutional review committee.

Consent for publication

Not required.

Conflicts of interest

Y.R.M.-G. reports travel, accommodation, and expenses from AstraZeneca and Loxo Oncology/Eli Lilly. She acknowledges honoraria from Virology Education and Projects in Knowledge (for a CME program funded by an educational grant from Amgen). She acknowledges associated research funding to the institution from Mirati Therapeutics, Bristol Myers Squibb, Loxo Oncology at Eli Lilly, Elucida Oncology, Taiho Oncology, Hengrui USA, Ltd/Jiangsu Hengrui Pharmaceuticals, Luzsana Biotechnology, Endeavor Biomedicines, and AbbVie. She is an employee of Memorial Sloan Kettering Cancer Center, which has an institutional interest in Elucida. She acknowledges royalties from Rutgers University Press and Wolters Kluwer. She acknowledges food/beverages from Endeavor Biomedicines. Y.R.M.-G. acknowledges receipt of training through an institutional K30 grant from the National Institutes of Health (CTSA UL1TR00457). She has received funding from a Kristina M. Day Young Investigator Award from Conquer Cancer, the ASCO Foundation, endowed by Dr Charles M. Baum and Carol A. Baum. She is also funded by the Fiona and Stanley Druckenmiller Center for Lung Cancer Research, the Andrew Sabin Family Foundation, the Society for MSK, the Squeri Grant for Drug Development, and a Paul Calabresi Career Development Award for Clinical Oncology (NIH/NCI K12 CA184746) as well as through NIH/NCI R01 CA279264.

C.M.R. has consulted regarding oncology drug development with AbbVie, Amgen, Astra Zeneca, D2G, Daiichi Sankyo, Epizyme, Genentech/Roche, Ipsen, Jazz, Kowa, Merck, and Syros. He serves on the scientific advisory boards of Auron, Bridge Medicines, DISCO, Earli, and Harpoon Therapeutics.

C.F.F. reports personal/consultancy fees from AstraZeneca and Bristol Myers Squibb, as well as participation in steering committees (compensation waived) for Merck and Genentech. These are outside the scope of the submitted work. She also reports institutional research funding from Genentech, Merck, Bristol Myers Squibb, Daiichi, and AstraZeneca.

A.D. reports: honoraria/advisory boards: Ignyta/Genentech/Roche, Loxo/Bayer/Lilly, Takeda/Ariad/Millenium, TP Therapeutics, AstraZeneca, Pfizer, Blueprint Medicines, Helsinn, Beigene, BergenBio, Hengrui Therapeutics, Exelixis, Tyra Biosciences, Verastem, MORE Health, Abbvie, 14ner/Elevation Oncology, Remedica Ltd., ArcherDX, Monopteros, Novartis, EMD Serono, Melendi, Liberum, Repare RX, Chugai, Merus, Chugai Pharmaceutical, Nuvalent, mBrace, AXIS, EPG Health, Harborside Nexus, Liberum, RV More, Ology. He reports associated research paid to institution: Pfizer, Exelixis, GlaxoSmithKlein, Teva, Taiho, PharmaMar. He reports royalties from: Wolters Kluwer; He reports other relationships with: Merck, Puma, Merus, Boehringer Ingelheim. He reports CME honoraria from: Medscape, OncLive, PeerVoice, Physicians Education Resources, Targeted Oncology, Research to Practice, Axis,Peerview Institute, Paradigm Medical Communications, WebMD, MJH Life Sciences, AXIS, EPG Health, JNCC/Harborside.

J.D.W. is a consultant for: Amgen; Apricity; Ascentage Pharma; Arsenal IO; Astellas; AstraZeneca; Bayer; Bicara Therapeutics; Boehringer Ingelheim; Bristol Myers Squibb; Chugai; Daiichi Sankyo, Dragonfly; Eli Lilly; F Star; Georgiamune; Idera; Imvaq; Kyowa Hakko Kirin; Maverick Therapeutics; Merck; Neon Therapeutics; Psioxus; Recepta; Tizona; Trieza; Truvax; Trishula; Sellas; Surface Oncology; Syndax; Syntalogic, Werewolf Therapeutics. J.D.W. has Grant/Research Support from: Bristol Myers Squibb; Sephora. J.D.W. has Equity in: Tizona Pharmaceuticals; Adaptive Biotechnologies; Imvaq; Beigene; Linneaus, Apricity, Arsenal IO; Georgiamune; Trieza; Maverick; Ascentage.

C.H.L. is a consultant for: Amgen, BMS, Exelixis, Eisai, Merck, Pfizer, and EMD Serono. Research funds to the institute from: BMS, Calithera, Eisai, Eli Lilly, Exelixis, Merck, and Pfizer. Honoria from: AiCME, Intellisphere, and Research to Practice.

M.A.P. has participated in ad hoc consultancy for: BMS, Merck, Novartis, Eisai, Pfizer, Chugai and receives institutional support from RGenix, Infinity, BMS, Merck, and Novartis.

Data Availability

Data are available on reasonable request. All data relevant to the study are included in the article. The corresponding author may be contacted with any requests.

References

  • 1. Vaddepally RK, Kharel P, Pandey R, Garje R, Chandra AB.. Review of indications of FDA-approved immune checkpoint inhibitors per NCCN guidelines with the level of evidence. Cancers. 2020;12:738. https://doi.org/ 10.3390/cancers12030738 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Murciano-Goroff YR, Warner AB, Wolchok JD.. The future of cancer immunotherapy: microenvironment-targeting combinations. Cell Res. 2020;30:507-519. https://doi.org/ 10.1038/s41422-020-0337-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Postow MA, Callahan MK, Wolchok JD.. Immune checkpoint blockade in cancer therapy. J Clin Oncol. 2015;33:1974-1982. https://doi.org/ 10.1200/JCO.2014.59.4358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Postow MA, Sidlow R, Hellmann MD.. Immune-related adverse events associated with immune checkpoint blockade. N Engl J Med. 2018;378:158-168. https://doi.org/ 10.1056/NEJMra1703481 [DOI] [PubMed] [Google Scholar]
  • 5. Friedman CF, Proverbs-Singh TA, Postow MA.. Treatment of the immune-related adverse effects of immune checkpoint inhibitors: a review. JAMA Oncol. 2016;2:1346-1353. https://doi.org/ 10.1001/jamaoncol.2016.1051 [DOI] [PubMed] [Google Scholar]
  • 6. Chiou VL, Burotto M.. Pseudoprogression and immune-related response in solid tumors. J Clin Oncol. 2015;33:3541-3543. https://doi.org/ 10.1200/JCO.2015.61.6870 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. de Miguel M, Calvo E.. Clinical challenges of immune checkpoint inhibitors. Cancer Cell. 2020;38:326-333. https://doi.org/ 10.1016/j.ccell.2020.07.004 [DOI] [PubMed] [Google Scholar]
  • 8. Suozzi KC, Stahl M, Ko CJ, et al. Immune-related sarcoidosis observed in combination ipilimumab and nivolumab therapy. JAAD Case Rep. 2016;2:264-268. https://doi.org/ 10.1016/j.jdcr.2016.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. 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. https://doi.org/ 10.1183/09031936.00107912 [DOI] [PubMed] [Google Scholar]
  • 10. Murphy KP, Kennedy MP, Barry JE, O’Regan KN, Power DG.. 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. https://doi.org/ 10.1159/000362614 [DOI] [PubMed] [Google Scholar]
  • 11. Nishino M, Sholl LM, Awad MM, et al. Sarcoid-like granulomatosis of the lung related to immune-checkpoint inhibitors: distinct clinical and imaging features of a unique immune-related adverse event. Cancer Immunol Res. 2018;6:630-635. https://doi.org/ 10.1158/2326-6066.CIR-17-0715 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Tirumani SH, Ramaiya NH, Keraliya A, et al. Radiographic profiling of immune-related adverse events in advanced melanoma patients treated with ipilimumab. Cancer Immunol Res. 2015;3:1185-1192. https://doi.org/ 10.1158/2326-6066.CIR-15-0102 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Tetzlaff MT, Nelson KC, Diab A, 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. https://doi.org/ 10.1186/s40425-018-0323-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. 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. https://doi.org/ 10.1177/1078155216667635 [DOI] [PubMed] [Google Scholar]
  • 15. Danlos FX, Pagès C, Baroudjian B, et al. Nivolumab-induced sarcoid-like granulomatous reaction in a patient with advanced melanoma. Chest. 2016;149:e133-e136. https://doi.org/ 10.1016/j.chest.2015.10.082 [DOI] [PubMed] [Google Scholar]
  • 16. Eckert A, Schoeffler A, Dalle S, et al. Anti-CTLA4 monoclonal antibody induced sarcoidosis in a metastatic melanoma patient. Dermatology. 2009;218:69-70. https://doi.org/ 10.1159/000161122 [DOI] [PubMed] [Google Scholar]
  • 17. Vogel WV, Guislain A, Kvistborg P, et al. Ipilimumab-induced sarcoidosis in a patient with metastatic melanoma undergoing complete remission. J Clin Oncol. 2012;30:e7-e10. https://doi.org/ 10.1200/JCO.2011.37.9693 [DOI] [PubMed] [Google Scholar]
  • 18. Reule RB, North JP.. Cutaneous and pulmonary sarcoidosis-like reaction associated with ipilimumab. J Am Acad Dermatol. 2013;69:e272-e273. https://doi.org/ 10.1016/j.jaad.2013.07.028 [DOI] [PubMed] [Google Scholar]
  • 19. Reddy SB, Possick JD, Kluger HM, Galan A, Han D.. Sarcoidosis following anti-PD-1 and anti-CTLA-4 therapy for metastatic melanoma. J Immunother. 2017;40:307-311. https://doi.org/ 10.1097/CJI.0000000000000181 [DOI] [PubMed] [Google Scholar]
  • 20. Berthod G, Lazor R, Letovanec I, et al. Pulmonary sarcoid-like granulomatosis induced by ipilimumab. J Clin Oncol. 2012;30:e156-e159. https://doi.org/ 10.1200/JCO.2011.39.3298 [DOI] [PubMed] [Google Scholar]
  • 21. Cousin S, Toulmonde M, Kind M, et al. Pulmonary sarcoidosis induced by the anti-PD1 monoclonal antibody pembrolizumab. Ann Oncol. 2016;27:1178-1179. https://doi.org/ 10.1093/annonc/mdw125 [DOI] [PubMed] [Google Scholar]
  • 22. Zhang M, Schembri G.. Nivolumab-induced development of pulmonary sarcoidosis in renal cell carcinoma. Clin Nucl Med. 2017;42:728-729. https://doi.org/ 10.1097/RLU.0000000000001758 [DOI] [PubMed] [Google Scholar]
  • 23. Chorti E, Kanaki T, Zimmer L, et al. Drug-induced sarcoidosis-like reaction in adjuvant immunotherapy: Increased rate and mimicker of metastasis. Eur J Cancer. 2020;131:18-26. https://doi.org/ 10.1016/j.ejca.2020.02.024 [DOI] [PubMed] [Google Scholar]
  • 24. Garanzini EM, 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. BJR Case Rep. 2020;6:20190065. https://doi.org/ 10.1259/bjrcr.20190065 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Chanson N, Ramos-Casals M, Pundole X, et al. ; ICIR. Immune checkpoint inhibitor-associated sarcoidosis: a usually benign disease that does not require immunotherapy discontinuation. Eur J Cancer. 2021;158:208-216. https://doi.org/ 10.1016/j.ejca.2021.05.041 [DOI] [PubMed] [Google Scholar]
  • 26. Cabanié C, Ammari S, Hans S, et al. Outcomes of patients with cancer and sarcoid-like granulomatosis associated with immune checkpoint inhibitors: a case-control study. Eur J Cancer. 2021;156:46-59. https://doi.org/ 10.1016/j.ejca.2021.07.015 [DOI] [PubMed] [Google Scholar]
  • 27. Yousuf H, Mekki R, Khan K, Hussain A.. Pembrolizumab-induced sarcoid-like reaction in a patient with lung cancer. Cureus. 2020;12:e12395. https://doi.org/ 10.7759/cureus.12395 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Yasin H, Yadala V, Khan NAJ, et al. Immunotherapy-induced sarcoid-like reaction: a shrewd imitator. J Investig Med High Impact Case Rep. 2021;9:23247096211009400. https://doi.org/ 10.1177/23247096211009400 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Khalid T, Patil A, Ahmed A, Yousif A.. Sarcoid-like reaction: a unique response to immunotherapy in malignant melanoma. BMJ Case Rep. 2021;14:e243723. https://doi.org/ 10.1136/bcr-2021-243723 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Mitchell MA, Hogan K, Amjadi K.. Atezolizumab-induced sarcoid-like granulomatous reaction in a patient with urothelial cell carcinoma. Immunotherapy. 2018;10:1189-1192. https://doi.org/ 10.2217/imt-2018-0035 [DOI] [PubMed] [Google Scholar]
  • 31. Nishino M, Sholl LM, Awad MM, et al. Sarcoid-like granulomatosis of the lung related to immune-checkpoint inhibitors: distinct clinical and imaging features of a unique immune-related adverse event. Cancer Immunol Res. 2018;6:630-635. https://doi.org/ 10.1158/2326-6066.cir-17-0715 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Murthi M, Yoshioka K, Cho JH, et al. Presence of concurrent sarcoid-like granulomas indicates better survival in cancer patients: a retrospective cohort study. ERJ Open Res. 2020;6:00061-02020. https://doi.org/ 10.1183/23120541.00061-2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Kim MH, Lee K, Kim KU, et al. Sarcoidosis mimicking cancer metastasis following chemotherapy for ovarian cancer (Murthi et al. 2020). Cancer Res Treat. 2013;45:354-358. https://doi.org/ 10.4143/crt.2013.45.4.354 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Ravaglia C, Gurioli C, Casoni GL, et al. Sarcoid-like lesion is a frequent benign cause of lymphadenopathy in neoplastic patients. Eur Respir J. 2013;41:754-755. https://doi.org/ 10.1183/09031936.00141212 [DOI] [PubMed] [Google Scholar]
  • 35. Dimitriou F, Frauchiger AL, Urosevic-Maiwald M, et al. Sarcoid-like reactions in patients receiving modern melanoma treatment. Melanoma Res. 2018;28:230-236. https://doi.org/ 10.1097/CMR.0000000000000437 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Chopra A, Nautiyal A, Kalkanis A, Judson MA.. Drug-induced sarcoidosis-like reactions. Chest. 2018;154:664-677. https://doi.org/ 10.1016/j.chest.2018.03.056 [DOI] [PubMed] [Google Scholar]
  • 37. El Jammal T, Pavic M, Gerfaud-Valentin M, Jamilloux Y, Sève P.. Sarcoidosis and cancer: a complex relationship. Front Med (Lausanne). 2020;7:594118. https://doi.org/ 10.3389/fmed.2020.594118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Lomax AJ, McGuire HM, McNeil C, 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. https://doi.org/ 10.1111/1756-185X.13076 [DOI] [PubMed] [Google Scholar]
  • 39. Beutler BD, Cohen PR.. Sarcoidosis in melanoma patients: case report and literature review. Cancers (Basel). 2015;7:1005-1021. https://doi.org/ 10.3390/cancers7020821 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. 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. https://doi.org/ 10.1002/ccr3.5960 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. 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. 2021;5:15-18. https://doi.org/ 10.1002/iju5.12372 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Charkviani M, Herrmann SM.. Immune checkpoint inhibitor-associated sarcoidosis reaction in the kidney: case report. Kidney Med. 2023;5:100626. https://doi.org/ 10.1016/j.xkme.2023.100626 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. 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. https://doi.org/ 10.1155/2019/6702870 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. 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. https://doi.org/ 10.1136/bcr-2021-247278 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. van den Eertwegh AJ, Versluis J, van den Berg HP, et al. Combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol. 2012;13:509-517. https://doi.org/ 10.1016/S1470-2045(12)70007-4 [DOI] [PubMed] [Google Scholar]
  • 46. Cousin S, Toulmonde M, Kind M, et al. Pulmonary sarcoidosis induced by the anti-PD1 monoclonal antibody pembrolizumab. Ann Oncol. 2016;27:1178-1179. https://doi.org/ 10.1093/annonc/mdw125 [DOI] [PubMed] [Google Scholar]
  • 47. Cotliar J, Querfeld C, Boswell WJ, et al. Pembrolizumab-associated sarcoidosis. JAAD Case Rep. 2016;2:290-293. https://doi.org/ 10.1016/j.jdcr.2016.06.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Torrecilla-Vall-Llossera C, Jucglà Serra A, Molinero Caturla J, et al. Sarcoid-like reactions to immune checkpoint inhibitors. Actas Dermosifiliogr. 2024;115:80-83. https://doi.org/ 10.1016/j.ad.2023.03.010 [DOI] [PubMed] [Google Scholar]
  • 49. Paydas S. Sarcoid-like reaction in cases treated by checkpoint inhibitors. Med Oncol. 2021;38:29. https://doi.org/ 10.1007/s12032-021-01477-y [DOI] [PubMed] [Google Scholar]
  • 50. 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. https://doi.org/ 10.14740/jmc4038 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Sirgi Y, Krochmal R, Fleury CM, et al. Pembrolizumab-associated cutaneous and pulmonary sarcoidosis in non-small cell lung cancer treatment. Clin Lung Cancer. 2022;23:542-546. https://doi.org/ 10.1016/j.cllc.2022.05.011 [DOI] [PubMed] [Google Scholar]
  • 52. Zhao X, Yue D, Qian J, 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. https://doi.org/ 10.3389/fimmu.2022.794217 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Torres-Zurita A, Vázquez-Montero L, Gallego-López L, Mediano-Rambla MD, 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. https://doi.org/ 10.3389/fimmu.2023.1150128 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Takamori S, Furubayashi N, Taguchi K, 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. https://doi.org/ 10.1111/1759-7714.14011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Sahin IH, Geyer AI, Kelly DW, O’Reilly EM.. Gemcitabine-related pneumonitis in pancreas adenocarcinoma--an infrequent event: elucidation of risk factors and management implications. Clin Colorectal Cancer. 2016;15:24-31. https://doi.org/ 10.1016/j.clcc.2015.08.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Kim ST, Pundole X, Dadu R, et al. Use of immune checkpoint inhibitors in cancer patients with pre-existing sarcoidosis. Immunotherapy. 2021;13:465-475. https://doi.org/ 10.2217/imt-2020-0272 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Le Burel S, Champiat S, Mateus C, et al. Prevalence of immune-related systemic adverse events in patients treated with anti-programmed cell death 1/anti-programmed cell death-ligand 1 agents: a single-centre pharmacovigilance database analysis. Eur J Cancer. 2017;82:34-44. https://doi.org/ 10.1016/j.ejca.2017.05.032 [DOI] [PubMed] [Google Scholar]
  • 58. Shinomiya S, Kaira K, Mouri A, Kagamu H.. Synchronous dilemma of sarcoid-like reaction and drastic response after PD-1 blockade administration in lung cancer. Jpn J Clin Oncol. 2021;51:1179-1180. https://doi.org/ 10.1093/jjco/hyab043 [DOI] [PubMed] [Google Scholar]
  • 59. Cascone T, William WN, Weissferdt A, et al. Neoadjuvant nivolumab or nivolumab plus ipilimumab in operable non-small cell lung cancer: the phase 2 randomized NEOSTAR trial. Nat Med. 2021;27:504-514. https://doi.org/ 10.1038/s41591-020-01224-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Birnbaum MR, Ma MW, Fleisig S, et al. Nivolumab-related cutaneous sarcoidosis in a patient with lung adenocarcinoma. JAAD Case Rep. 2017;3:208-211. https://doi.org/ 10.1016/j.jdcr.2017.02.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Kraaijvanger R, Janssen Bonás M, Vorselaars ADM, Veltkamp M.. Biomarkers in the diagnosis and prognosis of sarcoidosis: current use and future prospects. Front Immunol. 2020;11:1443. https://doi.org/ 10.3389/fimmu.2020.01443 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Brice EAW, Friedlander W, Bateman ED, Kirsch BE.. Serum angiotensin-converting enzyme activity, concentration, and specific activity in granulomatous interstitial lung disease, tuberculosis, and COPD. Chest. 1995;107:706-710. https://doi.org/ 10.1378/chest.107.3.706 [DOI] [PubMed] [Google Scholar]
  • 63. Renston JP, Goldman ES, Hsu RM, Tomashefski JF Jr. Peripheral blood eosinophilia in association with sarcoidosis. Mayo Clin Proc. 2000;75:586-590. https://doi.org/ 10.4065/75.6.586 [DOI] [PubMed] [Google Scholar]
  • 64. Scanvion Q, Béné J, Gautier S, et al. Moderate-to-severe eosinophilia induced by treatment with immune checkpoint inhibitors: 37 cases from a national reference center for hypereosinophilic syndromes and the French pharmacovigilance database. Oncoimmunology. 2020;9:1722022. https://doi.org/ 10.1080/2162402X.2020.1722022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Hua C, Boussemart L, Mateus C, et al. Association of vitiligo with tumor response in patients with metastatic melanoma treated with pembrolizumab. JAMA Dermatol. 2016;152:45-51. https://doi.org/ 10.1001/jamadermatol.2015.2707 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data are available on reasonable request. All data relevant to the study are included in the article. The corresponding author may be contacted with any requests.


Articles from The Oncologist are provided here courtesy of Oxford University Press

RESOURCES