scholarly journals Increased mTORC2 Pathway Activation in Lymph Nodes of Imcd-Tafro

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 4316-4316
Author(s):  
Alexis D Phillips ◽  
Joseph K Kakkis ◽  
Patricia Y Tsao ◽  
Sheila K Pierson ◽  
David C Fajgenbaum

Abstract Idiopathic multicentric Castleman disease (iMCD) is a rare and life-threatening disorder involving polyclonal lymphoproliferation and organ dysfunction due to excessive cytokine production, including interleukin-6 (IL-6). Anti-IL-6 therapy is recommended first-line and effective in 34-45% of patients. mTOR, which functions through mTORC1 and mTORC2, is a recently-discovered therapeutic target. The mTOR inhibitor sirolimus, which preferentially inhibits mTORC1, has led to sustained remission in a small cohort of anti-IL-6 refractory iMCD patients with thrombocytopenia, anasarca, fever, renal dysfunction, and organomegaly (TAFRO). However, sirolimus has not shown uniform effect, potentially due to its limited mTORC2 inhibition. To investigate mTORC2 activation in iMCD, we quantified the mTORC2 effector protein pNDRG1 by immunohistochemistry of lymph node tissue from iMCD-TAFRO (N=6) patients and iMCD patients not meeting TAFRO criteria (iMCD-NOS; N=8) (Table 1) as well as autoimmune lymphoproliferative syndrome (ALPS) (N=6), Hodgkin lymphoma (positive controls, N=8), and metastasis-free sentinel lymph nodes (normal controls, N=8). In iMCD-TAFRO, pNDRG1 expression was elevated in the interfollicular space (IF) (P=0.005), germinal centers (GC) (P=0.002), and mantle zones (MZ) (P=0.007) relative to normal controls (Figure 1A). pNDRG1 staining was increased in the IF (P=0.005) of iMCD-NOS relative to normal controls and there was no difference in the GC (P=0.59) and the MZ (P=0.30) (Figure 1B). Next, we compared pNDRG1 expression in iMCD-TAFRO and iMCD-NOS to ALPS, an mTOR-driven, sirolimus-responsive lymphoproliferative disorder.Our results revealed increased pNDRG1 staining in iMCD-TAFRO GC relative to ALPS (P=0.02) (Figure 1C). There were no differences in pNDRG1 expression between iMCD-NOS and ALPS in any region (Figure 1C). Notably, the strongly positive pNDRG1 cells had spindle-shaped morphology resembling stromal cells (Figure 1H). These results suggest increased mTORC2 activity in iMCD and that dual mTORC1/mTORC2 inhibitors may be a rational therapeutic approach for anti-IL-6 treatment refractory patients. Further studies are needed to confirm this finding, uncover cell types showing increased mTORC2 expression, and investigate therapeutic approaches. Figure 1 Figure 1. Disclosures Fajgenbaum: EUSA Pharma: Research Funding; N/A: Other: Holds pending provisional patents for 'Methods of treating idiopathic multicentric Castleman disease with JAK1/2 inhibition' and 'Discovery and validation of a novel subgroup and therapeutic target in idiopathic multicentric Castleman disease'; Pfizer: Other: Study drug for clinical trial of sirolimus.

Blood ◽  
2020 ◽  
Vol 135 (19) ◽  
pp. 1673-1684 ◽  
Author(s):  
Daniel J. Arenas ◽  
Katherine Floess ◽  
Dale Kobrin ◽  
Ruth-Anne Langan Pai ◽  
Maya B. Srkalovic ◽  
...  

Abstract Idiopathic multicentric Castleman disease (iMCD) is a rare and poorly understood hematologic disorder characterized by lymphadenopathy, systemic inflammation, cytopenias, and life-threatening multiorgan dysfunction. Interleukin-6 (IL-6) inhibition effectively treats approximately one-third of patients. Limited options exist for nonresponders, because the etiology, dysregulated cell types, and signaling pathways are unknown. We previously reported 3 anti-IL-6 nonresponders with increased mTOR activation who responded to mTOR inhibition with sirolimus. We investigated mTOR signaling in tissue and serum proteomes from iMCD patients and controls. mTOR activation was increased in the interfollicular space of iMCD lymph nodes (N = 26) compared with control lymph nodes by immunohistochemistry (IHC) for pS6, p4EBP1, and p70S6K, known effectors and readouts of mTORC1 activation. IHC for pS6 also revealed increased mTOR activation in iMCD compared with Hodgkin lymphoma, systemic lupus erythematosus, and reactive lymph nodes, suggesting that the mTOR activation in iMCD is not just a product of lymphoproliferation/inflammatory lymphadenopathy. Further, the degree of mTOR activation in iMCD was comparable to autoimmune lymphoproliferative syndrome, a disease driven by mTOR hyperactivation that responds to sirolimus treatment. Gene set enrichment analysis of serum proteomic data from iMCD patients (n = 88) and controls (n = 42) showed significantly enriched mTORC1 signaling. Finally, functional studies revealed increased baseline mTOR pathway activation in peripheral monocytes and T cells from iMCD remission samples compared with healthy controls. IL-6 stimulation augmented mTOR activation in iMCD patients, which was abrogated with JAK1/2 inhibition. These findings support mTOR activation as a novel therapeutic target for iMCD, which is being investigated through a trial of sirolimus (NCT03933904).


2013 ◽  
Vol 61 (4) ◽  
pp. 737-739 ◽  
Author(s):  
Lucie M. Turcotte ◽  
Colleen K. Correll ◽  
Robyn C. Reed ◽  
Christopher L. Moertel

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2709-2709
Author(s):  
Freda R Coren ◽  
Mateo Sarmiento Bustamante ◽  
Sheila K Pierson ◽  
David C Fajgenbaum

Abstract Unicentric Castleman disease (UCD) is one of several subtypes of Castleman disease that share characteristic histopathology. While UCD involves one region of enlarged lymph nodes (LN) and milder symptoms, idiopathic multicentric CD (iMCD) involves multiple regions of enlarged lymph nodes and cytokine-driven organ dysfunction. UCD symptoms can occur due to compression of neighboring structures or inflammatory cytokine production. Complete excision of the enlarged LN is reported in the literature to be curative in 84-96% of UCD patients. However, many UCD patients describe persistence or worsening of symptoms post excision despite normal laboratory values and absence of measurable disease. To better characterize and describe the experience of individuals diagnosed with UCD, we administered surveys to UCD patients to capture information about general quality of life (QOL), specific health measures, and ongoing UCD symptoms. All patients enrolled in the ACCELERATE Natural History registry who either self-reported a UCD diagnosis or were suspected to have a UCD diagnosis were invited to participate in the Rand36-item Short Form survey, the EQ-5D-5L, the MCD Symptom Score survey, all of which are validated instruments, as well as an additional form regarding ongoing symptoms. Among the 107 UCD patients invited to participate, 51 (48%) responded. Descriptive analyses were conducted on all 51 respondents with a self-reported UCD diagnosis. Subsequently, a subset consisting of 25 respondents, who had been reviewed by a panel of physicians that confirmed a UCD diagnosis (confirmed subcohort, CS), was analyzed. Mean (SD) EQ-5D-5L health index score (100 being perfect health, 0 being worst health imaginable) of the 51 patients was 67.6 (19.9), compared to a representative sample of the US population whose score was 80.4. Patients reported having poor health over the prior 4 weeks, with low scores indicating high levels of fatigue (38.8) and poor general health (47.7). These are notable and comparable to scores for fatigue (52.2) and general health (57.0) among a separate cohort of patients with diabetes, hypertension, coronary heart disease, and/or depression. Strikingly, 57% of patients reported continuation of symptoms post-LN excision, with an additional 16% being unsure. Only 29% reported complete symptom resolution, with the remainder reporting partial resolution (29%), stable disease (20%), worsening disease (12%), or unsure (10%). Nearly all 51 respondents underwent their LN-excision >1 year prior (93%). Fatigue (61%) and night sweats (39%) were most commonly reported in patients with continued symptoms. In fact, 27 patients (53%) reported ongoing symptoms on the day of survey completion with 93% of those patients reporting fatigue that day. Of note, sub-group analyses of the CS revealed similar findings, with a potential trend towards slightly better overall health with mean (SD) health index score 72.6 (14.5), fatigue (44%) and night sweats (36%), and ongoing symptoms reported in 40%. Of the patients in the CS, all had a complete resection, with 3 demonstrating subsequent lymphadenopathy in new regions ranging from 6 months to 5 years after their initial excision. Overall, these data suggest that UCD patients who have had resection of disease continue to experience symptoms that affect QOL. Patients reported lower QOL than a representative national average, as well as health measures comparable to a separate cohort of individuals with chronic health conditions. Of note, the full cohort of self-reported individuals consistently reported lower scores compared to the subset of patients with confirmed UCD (not statistically tested). Of the 26 non-confirmed cases, 8 did not achieve criteria to meet UCD and 18 have not yet been reviewed by our physician panel. A different undiagnosed disorder may be the root cause of symptoms in a portion of these patients and others may be experiencing symptoms due to a co-occurring disorder. These results may be confounded by reporting bias and may not be representative of the full UCD population. Nevertheless, these data suggest that perceived-to-be curative excision does not result in symptom-free outcomes in a substantial proportion of patients and that symptom management may be required beyond excision. Future work is needed to correlate these findings with clinical, laboratory, and experimental data to further elucidate mechanisms and treatment options. Disclosures Fajgenbaum: EUSA Pharma: Research Funding; Pfizer: Other: Study drug for clinical trial of sirolimus; N/A: Other: Holds pending provisional patents for 'Methods of treating idiopathic multicentric Castleman disease with JAK1/2 inhibition' and 'Discovery and validation of a novel subgroup and therapeutic target in idiopathic multicentric Castleman disease'.


2021 ◽  
Vol 5 (17) ◽  
pp. 3445-3456
Author(s):  
Sheila K. Pierson ◽  
Sushila Shenoy ◽  
Ana B. Oromendia ◽  
Alexander M. Gorzewski ◽  
Ruth-Anne Langan Pai ◽  
...  

Abstract Idiopathic multicentric Castleman disease (iMCD) is a poorly understood hematologic disorder involving cytokine-induced polyclonal lymphoproliferation, systemic inflammation, and potentially fatal multiorgan failure. Although the etiology of iMCD is unknown, interleukin-6 (IL-6) is an established disease driver in approximately one-third of patients. Anti–IL-6 therapy, siltuximab, is the only US Food and Drug Administration–approved treatment. Few options exist for siltuximab nonresponders, and no validated tests are available to predict likelihood of response. We procured and analyzed the largest-to-date cohort of iMCD samples, which enabled classification of iMCD into disease categories, discovery of siltuximab response biomarkers, and identification of therapeutic targets for siltuximab nonresponders. Proteomic quantification of 1178 analytes was performed on serum of 88 iMCD patients, 60 patients with clinico-pathologically overlapping diseases (human herpesvirus-8–associated MCD, N = 20; Hodgkin lymphoma, N = 20; rheumatoid arthritis, N = 20), and 42 healthy controls. Unsupervised clustering revealed iMCD patients have heterogeneous serum proteomes that did not cluster with clinico-pathologically overlapping diseases. Clustering of iMCD patients identified a novel subgroup with superior response to siltuximab, which was validated using a 7-analyte panel (apolipoprotein E, amphiregulin, serum amyloid P-component, inactivated complement C3b, immunoglobulin E, IL-6, erythropoietin) in an independent cohort. Enrichment analyses and immunohistochemistry identified Janus kinase (JAK)/signal transducer and activator of transcription 3 signaling as a candidate therapeutic target that could potentially be targeted with JAK inhibitors in siltuximab nonresponders. Our discoveries demonstrate the potential for accelerating discoveries for rare diseases through multistakeholder collaboration.


Blood ◽  
2018 ◽  
Vol 132 (22) ◽  
pp. 2323-2330 ◽  
Author(s):  
David C. Fajgenbaum

Castleman disease (CD) describes a heterogeneous group of hematologic disorders that share characteristic lymph node histopathology. Patients of all ages present with either a solitary enlarged lymph node (unicentric CD) or multicentric lymphadenopathy (MCD) with systemic inflammation, cytopenias, and life-threatening multiple organ dysfunction resulting from a cytokine storm often driven by interleukin 6 (IL-6). Uncontrolled human herpesvirus-8 (HHV-8) infection causes approximately 50% of MCD cases, whereas the etiology is unknown in the remaining HHV-8-negative/idiopathic MCD cases (iMCD). The limited understanding of etiology, cell types, and signaling pathways involved in iMCD has slowed development of treatments and contributed to historically poor patient outcomes. Here, recent progress for diagnosing iMCD, characterizing etio-pathogenesis, and advancing treatments are reviewed. Several clinicopathological analyses provided the evidence base for the first-ever diagnostic criteria and revealed distinct clinical subtypes: thrombocytopenia, anasarca, fever, reticulin fibrosis/renal dysfunction, organomegaly (iMCD-TAFRO) or iMCD-not otherwise specified (iMCD-NOS), which are both observed all over the world. In 2014, the anti-IL-6 therapy siltuximab became the first iMCD treatment approved by the US Food and Drug Administration, on the basis of a 34% durable response rate; consensus guidelines recommend it as front-line therapy. Recent cytokine and proteomic profiling has revealed normal IL-6 levels in many patients with iMCD and potential alternative driver cytokines. Candidate novel genomic alterations, dysregulated cell types, and signaling pathways have also been identified as candidate therapeutic targets. RNA sequencing for viral transcripts did not reveal novel viruses, HHV-8, or other viruses pathologically associated with iMCD. Despite progress, iMCD remains poorly understood. Further efforts to elucidate etiology, pathogenesis, and treatment approaches, particularly for siltuximab-refractory patients, are needed.


2021 ◽  
Vol 12 ◽  
Author(s):  
Frederik Staels ◽  
Albrecht Betrains ◽  
Sherida Woei-A-Jin ◽  
Nancy Boeckx ◽  
Marielle Beckers ◽  
...  

Recently, a novel disorder coined VEXAS (vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic) syndrome was identified in patients with adult-onset inflammatory syndromes, often accompanied by myelodysplastic syndrome1. All patients had myeloid lineage-restricted somatic mutations in UBA1 affecting the Met41 residue of the protein and resulting in decreased cellular ubiquitylation activity and hyperinflammation. We here describe the clinical disease course of two VEXAS syndrome patients with somatic UBA1 mutations of which one with a mild phenotype characterized by recurrent rash and symmetric polyarthritis, and another who was initially diagnosed with idiopathic multicentric Castleman disease and developed macrophage activation syndrome as a complication of the VEXAS syndrome. The latter patients was treated with anti-IL6 therapy (siltuximab) leading to a resolution of systemic symptoms and reduction of transfusion requirements.


Hematology ◽  
2018 ◽  
Vol 2018 (1) ◽  
pp. 318-325 ◽  
Author(s):  
David C. Fajgenbaum

Abstract Castleman disease (CD) describes a heterogeneous group of hematologic disorders that share characteristic lymph node histopathology. Patients of all ages present with either a solitary enlarged lymph node (unicentric CD) or multicentric lymphadenopathy (MCD) with systemic inflammation, cytopenias, and life-threatening multiple organ dysfunction resulting from a cytokine storm often driven by interleukin 6 (IL-6). Uncontrolled human herpesvirus-8 (HHV-8) infection causes approximately 50% of MCD cases, whereas the etiology is unknown in the remaining HHV-8-negative/idiopathic MCD cases (iMCD). The limited understanding of etiology, cell types, and signaling pathways involved in iMCD has slowed development of treatments and contributed to historically poor patient outcomes. Here, recent progress for diagnosing iMCD, characterizing etio-pathogenesis, and advancing treatments are reviewed. Several clinicopathological analyses provided the evidence base for the first-ever diagnostic criteria and revealed distinct clinical subtypes: thrombocytopenia, anasarca, fever, reticulin fibrosis/renal dysfunction, organomegaly (iMCD-TAFRO) or iMCD-not otherwise specified (iMCD-NOS), which are both observed all over the world. In 2014, the anti-IL-6 therapy siltuximab became the first iMCD treatment approved by the US Food and Drug Administration, on the basis of a 34% durable response rate; consensus guidelines recommend it as front-line therapy. Recent cytokine and proteomic profiling has revealed normal IL-6 levels in many patients with iMCD and potential alternative driver cytokines. Candidate novel genomic alterations, dysregulated cell types, and signaling pathways have also been identified as candidate therapeutic targets. RNA sequencing for viral transcripts did not reveal novel viruses, HHV-8, or other viruses pathologically associated with iMCD. Despite progress, iMCD remains poorly understood. Further efforts to elucidate etiology, pathogenesis, and treatment approaches, particularly for siltuximab-refractory patients, are needed.


2020 ◽  
Vol 21 (1) ◽  
Author(s):  
Yuejuan Pan ◽  
Zhuan Cui ◽  
Song Wang ◽  
Danxia Zheng ◽  
Zhenling Deng ◽  
...  

Abstract Background Idiopathic multicentric Castleman disease (iMCD) is an uncommon lymphoproliferative disorder and lacks treatment consensus. Herein, we report a case of iMCD complicated with Sjögren’s syndrome (SS) and secondary membranous nephropathy (SMN). Case presentation A 45-year-old female with dry mouth for 3 months and anasarca and proteinuria for 2 months was admitted. She also experienced chest tightness, wheezing, fever, weight loss, moderate proteinuria and hypoalbuminemia. A computed tomography (CT) scan revealed a tissue mass in the thymus area and enlarged multiple lymph nodes. Her symptoms did not improve after resection of the thymus mass. The pathological findings were “reactive hyperplasia of the mediastinal lymph nodes and thymic hyperplasia”. Lymph node biopsy findings confirmed iMCD with human herpes virus-8 (HHV-8) negativity. Based on anti-nuclear antibody (ANA) 1:320, anti-SSA and anti-SSB antibody positivity, salivary flow less than 0.1 ml/min and lip biopsy with focal lymphocytic sialadenitis, SS was diagnosed. Kidney biopsy showed secondary membranous nephropathy with endocapillary cell proliferation and infiltration of plasma cells and lymphocytes in the tubulointerstitium. Serum interleukin-6 (IL-6) levels were significantly increased, and therapy with tocilizumab (anti-IL-6 receptor antibody) worked well. The combination of cyclophosphamide (CyS) with methylprednisolone (MP) maintained satisfactory remission. Conclusions Our case of iMCD with SS and SMN is rare. There is a need for increased awareness of the disease to avoid unnecessary procedures and misdiagnoses. IL-6 was extremely high, and there was a rapid response to anti-IL-6 receptor agents. The combination of CyS with MP maintained complete remission.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2693-2693
Author(s):  
Sheila K Pierson ◽  
David C Fajgenbaum

Abstract Idiopathic multicentric Castleman disease (iMCD) is a heterogeneous and life-threatening hematologic disorder with unknown etiology. It involves extensive lymphadenopathy with characteristic histopathology, inflammatory symptoms, and life-threatening cytokine-driven multi-organ failure, if inadequately treatment. Diagnosis requires the presence of histopathological, clinical, laboratory, and radiologic findings as well as exclusion of other related diseases including infectious, autoimmune, and lymphoproliferative disorders. Despite these guidelines, definitive diagnosis remains challenging as histopathological changes, clinical features, and laboratory abnormalities are non-specific. Given that iMCD can be aggressive and life-threatening, timely diagnosis is crucial, and specific diagnostic biomarkers are needed. Interleukin-6 (IL-6) is a known disease driver in a portion of patients, but it is not consistently elevated at diagnosis and characterization of other cytokines has not been systematically performed. Previously, a plasma proteomics study identified upregulation of multiple cytokines, including several chemokines, such as C-X-C Motif Chemokine Ligand 13 (CXCL13) and C-C motif ligand 21 (CCL21), during disease flare in a small cohort. We sought to investigate whether these findings would be replicated in a larger cohort and to identify cytokines that could help to improve diagnosis of iMCD. Herein, we analyzed serum samples from 88 iMCD patients, 42 healthy donors, and 20 patients each with rheumatoid arthritis (RA), Hodgkin lymphoma (HL), and a subtype of multicentric Castleman disease caused by infection with human herpes virus 8 (HHV8+MCD). 1300 analytes were quantified by SomaLogic SOMASCAN; Uniprot database was used to select the 159 quantified cytokines for analysis. Data were log2 normalized and capped at the 2.5 th and 97.5 th percentiles. Linear models were used to identify significant differences among disease groups, with p values adjusted by Benjamini & Hochberg. Hierarchical clustering was performed using complete linkage. Of the 159 cytokines quantified, 54 (34%) were identified as significantly different between iMCD and healthy donors. The most upregulated cytokine was CXCL13 with a log2 fold-change (FC) of 1.7 (p<0.0001). The chemokines CCL21 (FC: 1.3, p<0.0001) and CCL23 (FC: 0.87, p<0.0001) were also noted among the top 10 most upregulated cytokines, consistent with the earlier plasma proteomic study. Of note, IL-6 (FC: 0.35, p=0.039) was also found, but it ranked 31 out of 54 in log2 FC magnitude. We next sought to compare cytokine elevation in iMCD to related disorders. Hierarchical clustering of all 159 cytokines appeared to differentiate iMCD from RA, HL, and HHV8+MCD (Figure 1), revealing a cluster with 72/88 (82%) iMCD and 4/60 (6%) related disease patients versus another with 16/88 (18%) iMCD and 56/60 (93%) related disease patients. Given this, we used linear models to test for significant differences in protein expression between iMCD and related diseases. We identified 12 cytokines that were significantly different between iMCD and each comparator disease (Table 1). Of these, 10 cytokines all trended in the positive direction in iMCD versus each comparator disease. CCL21 was again noted among this group and had the highest average FC for all three groups. Herein we validate earlier findings that cytokines, particularly chemokines are significantly upregulated in iMCD. While previous results compared disease flare to disease remission in a small pilot cohort, our patient sample is >10 times larger, represents a broader spectrum of disease severity, and is compared to a set of healthy donors and related diseases. Unbiased hierarchical clustering was able to separate iMCD from other diseases, and linear models identified specific cytokines that are significantly different in iMCD compared to each RA, HL, and HHV8+MCD. Further validation is need to assess whether a cytokine panel can differentiate iMCD from other related diseases, which would be crucial for timely diagnosis and treatment. Of note, CXCL13 and CCL21 are primarily produced by follicular dendritic cells and fibroblastic reticular cells, stromal cells responsible for coordinating B cell and T cell trafficking in the lymph node, respectively. Dysregulation of these cells may contribute to the dysmorphic appearance of lymph nodes and pathogenesis in iMCD. Figure 1 Figure 1. Disclosures Fajgenbaum: EUSA Pharma: Research Funding; Pfizer: Other: Study drug for clinical trial of sirolimus; N/A: Other: Holds pending provisional patents for 'Methods of treating idiopathic multicentric Castleman disease with JAK1/2 inhibition' and 'Discovery and validation of a novel subgroup and therapeutic target in idiopathic multicentric Castleman disease'.


Sign in / Sign up

Export Citation Format

Share Document