scholarly journals Mastocytosis-derived extracellular vesicles exhibit a mast cell signature, transfer KIT to stellate cells, and promote their activation

2018 ◽  
Vol 115 (45) ◽  
pp. E10692-E10701 ◽  
Author(s):  
Do-Kyun Kim ◽  
Young-Eun Cho ◽  
Hirsh D. Komarow ◽  
Geethani Bandara ◽  
Byoung-Joon Song ◽  
...  

Extracellular vesicles (EVs) have been implicated in the development and progression of hematological malignancies. We thus examined serum samples from patients with systemic mastocytosis (SM) and found EVs with a mast cell signature including the presence of tryptase, FcεRI, MRGX2, and KIT. The concentration of these EVs correlated with parameters of disease including levels of serum tryptase, IL-6, and alkaline phosphatase and physical findings including hepatosplenomegaly. Given reports that EVs from one cell type may influence another cell’s behavior, we asked whether SM-EVs might affect hepatic stellate cells (HSCs), based on the abnormal liver pathology associated with mastocytosis. We found that KIT was transferred from SM-EVs into an HSC line eliciting proliferation, cytokine production, and differentiation, processes that have been associated with liver pathology. These effects were reduced by KIT inhibition or neutralization and recapitulated by enforced expression of KIT or constitutively active D816V-KIT, a gain-of-function variant associated with SM. Furthermore, HSCs in liver from mice injected with SM-EVs had increased expression of α-SMA and human KIT, particularly around portal areas, compared with mice injected with EVs from normal individuals, suggesting that SM-EVs can also initiate HSC activation in vivo. Our data are thus consistent with the conclusion that SM-EVs have the potential to influence cells outside the hematological compartment and that therapeutic approaches for treatment of SM may be effective in part through inhibition of effects of EVs on target tissues, findings important both to understanding complex disease pathology and in developing interventional agents for the treatment of hematologic diseases.

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1964-1964 ◽  
Author(s):  
Mohamad Jawhar ◽  
Nicole Naumann ◽  
Sebastian Kluger ◽  
Juliana Schwaab ◽  
Georgia Metzgeroth ◽  
...  

Abstract Recent data have highlighted that the molecular pathogenesis of advanced systemic mastocytosis (advSM) is complex. In addition to the phenotypically most important mutations in KIT, e.g. KIT D816V in 80-90% of patients, one or more additional mutations, e.g. in SRSF2, ASXL1, RUNX1, CBL, JAK2 and others, are present in 60-70% of patients (Jawhar et al., Leukemia 30, 2016). In individual patients, a complex mutational profile is detected not only in mature mast cells (MCs) but also in myeloid progenitors derived from granulocyte-macrophage colony-forming progenitor cells (CFU-GM), indicating multi-lineage involvement of all identified mutations in the vast majority of patients (Jawhar et al., Leukemia 29, 2015). Midostaurin, a multi-targeted kinase inhibitor, has demonstrated an overall response rate of 60% in advSM patients (Gotlib et al., NEJM 374, 2016). BLU-285 is a highly selective KIT D816V kinase inhibitor which has demonstrated biochemical activity on the mutated KIT enzyme (KIT D816V IC50 = 0.27 nM). In the current study, we sought to a) investigate the inhibitory effects of midostaurin and BLU-285 on single-cell-derived CFU-GM from bone marrow mononuclear cells derived from multi-mutated KIT D816V+ advSM patients and b) correlate the midostaurin CFU-GM data with clinical and various response parameters in midostaurin-treated advSM patients. The mutational status of CFU-GM colonies (median colonies per patient, n=20; range 10-30) was analyzed for KIT D816V and additional mutations by PCR followed by Sanger Sequencing. In 10 multi-mutated advSM patients (aggressive SM [n=8] or mast cell leukemia [n=2] with an associated hematological neoplasm), CFU-GM colonies were screened prior to midostaurin (month 0, n=10) and, if available, at month 6 on midostaurin (n=8). At month 0, a median of 90% (range, 40-100) CFU-GM colonies were KIT D816V+, while at month 6 a median of 70% (range, 5-100) CFU-GM colonies were KIT D816V+. A significant relative reduction (≥50%) in the proportion of KIT D816V+ colonies at month 6 was observed in 4/8 (50%) patients. Midostaurin-naïve CFU-GM were incubated with midostaurin at concentrations up to 1000 nM and showed a dose-dependent significant reduction (≥50%) of KIT D816V+ colonies in 1/7 (14%) patients. Overall, the in vitro effects correlated with the in vivo effects of midostaurin on CFU-GM and established IWG-MRT-ECNM response criteria (e.g. mast cell infiltration in BM, serum tryptase level) and KIT D816V allele burden in peripheral blood. Midostaurin-naïve CFU-GM from 7/10 (70%) patients were also incubated with different concentrations of BLU-285 ranging from 0 to 75 nM. A dose-dependent, significant relative reduction (≥50%) of KIT D816V+ CFU-GM colonies was observed at concentrations between 45 and 75nM in 5/7 (71%) patients. Of interest, 3/5 (60%) in vitro responders to BLU-285 were resistant to midostaurin (in vivo and in vitro) while CFU-GM colonies from 2 patients resistant to BLU-285 were also resistant to midostaurin. In addition to KIT D816V, recurrent molecular aberrations (median 2/patient, range 1-3) were identified in all patients, most frequently in SRSF2 (n=9), TET2 (n=7) and ASXL1 (n=4). Neither drug had an effect on the relative frequency of additional mutations in CFU-GM colonies. In summary, we conclude that a) the relative reduction of KIT D816V+ CFU-GM colonies between month 0 and month 6 on midostaurin correlates with clinical response, b) the CFU-GM colony assays may provide useful information for prediction of response to midostaurin, c) the highly selective KIT D816V inhibitor BLU-285 has significant activity against KIT D816V, even in cases which are resistant to midostaurin, and d) neither drug had an effect on the prognostically relevant additional mutations. Disclosures Evans: Blueprint Medicines: Employment, Equity Ownership. Gardino:Blueprint Medicines Corporation: Employment. Lengauer:Blueprint Medicines Corporation: Employment.


2019 ◽  
Vol 2019 ◽  
pp. 1-7
Author(s):  
Jason M. Springer ◽  
Vineesh V. Raveendran ◽  
Mingcai Zhang ◽  
Ryan Funk ◽  
Donald D. Smith ◽  
...  

Mast cells play an important role in immunomodulation and in the maintenance of vascular integrity. Interleukin-6 (IL-6) is one of the key biomarkers and therapeutic target in systemic vasculitis. The objective of the current study is to describe the role of mast cells in arterial IL-6 homeostasis. Eight- to ten-week-old male C57BL/6 (wild-type) mice were injected with either (a) saline, (b) compound 48/80 (a systemic mast cell degranulating agent), (c) lipopolysaccharide (LPS), or (d) a combination of C48/80 and LPS. Twenty-four hours after the injections, mice were sacrificed and serum samples and aortic tissues were analyzed for determining inflammatory response and cytokine expression profile. The results revealed that induction of mast cell degranulation significantly lowers serum IL-6 levels and aortic expression of IL-6 in LPS-treated mice. Significantly higher aortic expression of toll-like receptor-2 (TLR-2) and TNF-α was seen in the LPS and LPS+C48/80 groups of mice compared to controls. Aortic expression of TLR-4 was significantly decreased in LPS+C48/80 compared to C48/80 alone. LPS+C48/80-treated mice presented with a 3-fold higher aortic expression of suppressor of cytokine signaling (SOCS-1) compared to saline-injected groups. The inhibition of LPS-induced increase in serum IL-6 levels by mast cell degranulation was not seen in H1R knockout mice which suggests that mast cell-derived histamine acting through H1R may participate in the regulatory process. To examine whether the mast cell-mediated downregulation of LPS-induced IL-6 production is transient or cumulative in nature, wild-type mice were injected serially over a period of 10 days (5 injections) and serum cytokine levels were quantified. We found no significant differences in serum IL-6 levels between any of the groups. While mice injected with C48/80 or LPS had higher IL-10 compared to vehicle-injected mice, there was no difference between C48/80- and LPS+C48/80-injected mice. In conclusion, in an in vivo setting, mast cells appear to partially and transiently regulate systemic IL-6 homeostasis. This effect may be regulated through increased systemic IL-10 and/or aortic overexpression of SOCS-1.


2017 ◽  
Vol 47 (7) ◽  
pp. 909-917 ◽  
Author(s):  
T. Gülen ◽  
C. Möller Westerberg ◽  
K. Lyberg ◽  
M. Ekoff ◽  
J. Kolmert ◽  
...  

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2426-2426 ◽  
Author(s):  
Shadmehr Demehri ◽  
Amie S. Corbin ◽  
Marc Loriaux ◽  
Brian J. Druker ◽  
Michael W. Deininger

Abstract Background : Systemic mastocytosis (SM) is characterized by infiltration of extracutanous tissues by neoplastic mast cells. Primary target organs are liver, spleen and bone marrow. In some instances, SM progresses to aggressive systemic mastocytosis (ASM) or mast cell leukemia (MCL), which are associated with extensive mast cell infiltration into various organs and their failure. Almost all cases of SM exhibit point mutations at codon 816 of Kit, a receptor tyrosine kinase. These mutations (most commonly D816V) lead to constitutive activation of the kinase and are the causative lesion of SM. Here, we describe a novel murine model of SM/ASM that shares many characteristics with the human disease and may be useful for in vivo drug testing, including targeted therapy of D816 mutant Kit with small molecule inhibitors. Materials and methods : P815 cells, a cell line expressing D814Y Kit (homologous to human D816V kit) that was established in DBA2 mice (Dunn T, Bap M. J Natl Cancer Inst1957;18:587–95) were injected retro-orbitally into two groups (n = 4) of 8–10 week-old syngeneic mice, at a dose of 1x102 and 5x104 cells. Using an automated animal cell counter, the mice were monitored at 48h intervals with full blood counts, including white cell differential and platelets. The presence of mast cells was assessed by FACS for mast cell markers (CD117-PE and CD45-APC) as well as Giemsa staining. The animals were evaluated daily for signs of morbidity. Moribund mice were sacrificed and subjected to autopsy. Liver, spleen and bone marrow were analyzed by histopathology, and the expression and phophorylation status of Kit was assessed by FACS and immunoblotting. Results : Both cell doses induced an aggressive disease, with all animals reaching a moribund stage on day 9 (5x104 cells) and 16 (1x102 cells). A significant (p<0.001, student’s t test) drop in the platelet count regularly accompanied the appearance of mast cells in the peripheral blood (PB) (figure 1A). Subsequently, the animals developed marked granulocytosis. Autopsy demonstrated gross enlargement of liver and spleen, while lungs and kidneys were unaffected. Histopathology and FACS showed extensive infiltration of spleen, liver and bone marrow by Kit-positive cells (figure 1B). Immunoblotting revealed high levels of tyrosine phosphorylated Kit protein in whole cell lysates from PB, BM and spleen. Conclusion : We have established a highly reproducible model of SM/ASM that resembles the human disease. A particular advantage of this model is that the onset of disease can conveniently be monitored by serial PB counts. In addition, the latency of the disease can be modified by the size of the initial inoculum. Its extremely predictable course together with the parameters it provides for monitoring disease progression should make this model useful for the study of small molecules that target D816 mutant Kit. Figure 1. The infiltration of Kit-positive P815 cells into various organs of DBA/2 mouse. (A) PB over the course of the disease progression. (B) Hematopoietic organs at death. Figure 1. The infiltration of Kit-positive P815 cells into various organs of DBA/2 mouse. (A) PB over the course of the disease progression. (B) Hematopoietic organs at death.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3747-3747 ◽  
Author(s):  
Ghaith Wedeh ◽  
Emir Hadzijusufovic ◽  
Sabine Cerny-Reiterer ◽  
Harald Herrmann ◽  
Katharina Blatt ◽  
...  

Abstract Advanced mast cell (MC) neoplasms are characterized by uncontrolled growth and rapid expansion of neoplastic MC in various organ systems and a poor prognosis. In human patients, advanced systemic mastocytosis (SM) is rare and usually presents as aggressive SM (ASM) or mast cell leukemia (MCL). In canines, advanced MC tumors (MCT) are considered one of the most frequent skin tumors. Hence, so far, no effective treatment concept has been established for advanced MC neoplasms in humans or canines. This is due to the fact that neoplastic MC in advanced SM usually are resistant against various cytoreductive agents. Therefore, research is currently focusing on potential drug targets and the effects of novel targeted drugs on neoplastic MC. We have recently identified the bromodomain-containing protein-4 (BRD4) as a promising new drug target expressed by neoplastic cells in acute myeloid leukemia (Zuber et al, Nature 2011;478:524-528). In the present study, we examined BRD4 as a potential therapeutic target in human ASM and MCL, and canine MCT. As assessed by immunohistochemistry, neoplastic MC expressed substantial amounts of cytoplasmic and nuclear BRD4 in ASM and MCL, whereas in human indolent SM (ISM), MC expressed lower amounts of BRD4 or did not express cytoplasmic BRD4. The human MCL lines HMC-1.1 and HMC-1.2 as well as the canine mastocytoma cell lines NI-1 and C2, also expressed BRD4. In order to study the potential biological function of BRD4, we applied a BRD4-specific shRNA. The shRNA-induced knockdown of BRD4 was found to be associated with a markedly reduced proliferation of HMC-1 cells. Correspondingly, the BRD4-targeting drug JQ1 induced dose-dependent growth inhibition in all MC lines examined, with higher IC50 values obtained in HMC-1.1 cells (IC50: 1.0 µM) and HMC-1.2 cells (IC50: 1.0 µM) compared to NI-1 cells (0.2 µM) and C2 cells (0.5 µM). As assessed by morphology and AnnexinV/PI staining, the growth-inhibitory effects of JQ1 on neoplastic human and canine MC lines were accompanied by apoptosis (Figure). The apoptosis-inducing effects of JQ1 was also demonstrable by staining for active caspase 3. Furthermore, JQ1 was found to downregulate the expression of the activation-related MC antigen CD63 and the proliferation-related antigen CD71 (transferrin receptor) in HMC-1 cells. In addition, JQ1 was found to inhibit proliferation in primary neoplastic MC obtained from patients with ASM or MCL (IC50 0.1-0.5 µM). We next screened for potential drug partners that would potentiate the anti-neoplastic effects of JQ1. Of all drugs tested, all-trans retinoic acid (ATRA) and PKC412 were identified as most potent drug partners of JQ1. These drugs were found to act highly synergistic with JQ1 in producing apoptosis and growth inhibition in HMC-1.1 and HMC-1.2 cells (Figure). Together, BRD4 is a novel marker and promising target in advanced MC neoplasms. Whether JQ1 or other BET bromodomain inhibitors are effective in vivo in patients with advanced SM or canine MCT remains to be elucidated. HMC-1.2 cells were incubated in control medium (Co) or in various concentrations of JQ1 (black bars), ATRA (gray bars), or a combination of both agents at a ratio of 1:5 (white bars) at 37°C for 48 hours. Then, AnnexinV/propidium iodide (PI) staining was performed. The percentage of AnnexinV+ cells was determined by flow cytometry. Results represent the mean±S.D. of 3 independent experiments. Asterisk (*): p<0.05 compared to control (Co). Disclosures: Bradner: Tensha Therapeutics: Dr. Bradner is a scientific founder of Tensha Therapeutics, which is developing drug-like derivatives of the JQ1 bromodomain inhibitor as cancer therapeutics, through a license from the Dana-Farber Cancer Institute. Other. Horny:Novartis : Consultancy, Honoraria.


2015 ◽  
Vol 4 (1) ◽  
pp. 64-69 ◽  
Author(s):  
Catherine Cho ◽  
Anna Nguyen ◽  
Katherine J. Bryant ◽  
Sean G. O'Neill ◽  
H. Patrick McNeil

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2538-2538
Author(s):  
Anne Kaiser ◽  
Renier Myburgh ◽  
Laura Volta ◽  
Christian Edoardo Pellegrino ◽  
Markus G Manz

Abstract Mastocytosis is a malignant disease resulting from oncogenic transformed mast cells. Up to 80% of malignant cells harbor a D816V mutation in the KIT-receptor (CD117), leading to constitutive kinase activation and proliferation and survival of mast cells. Advanced forms of mastocytosis (aggressive systemic mastocytosis: ASM, systemic mastocytosis with associated hematological disease: SM-AHN, mast cell leukemia: MCL) present as a therapeutic challenge. Although the recently approved poly tyrosine kinase inhibitor Midostaurin provides some improvement, the median overall survival ranges from 3.5 years (ASM) to less than six months (MCL). The reduced life expectancy is frequently due to mast cell infiltration resulting in multi organ failure. Additionally, there are patients who do not benefit from the treatment with Midostaurin (overall response 60%) or suffer from side effects, which lead to reduction or termination of therapy. Currently, the only available curative approach is conditioning poly-chemotherapy followed by allogenic stem cell transplantation (allo-HSCT). However, allo-HSCT is associated with substantial side-effects and, also due to high rates of relapse, only leads to an overall survival of 43% for ASM and 17% for MCL after three years. Thus, better therapeutic options are needed. Recently, we demonstrated that CD117 (KIT-receptor) positive human AML can be efficiently eradicated by anti-CD117 CAR T-cells in vitro and in vivo (Myburgh et al., Leukemia 2020). As mast cells, and also transformed mast cells, highly express CD117, we here tested if anti-CD117 CAR T-cells would equally efficiently eliminate this malignant cell population. We thus co-cultured various established mast cell lines (partly harboring the oncogenic driver mutation KIT D816V) with anti-CD117-CAR T-cells in a 1:1 effector to target ratio in vitro. After 24 hours of co-culturing, the tumor cells were effectively killed, and this was still observed despite increasing the effector to target ratio to 1:4. Also, within 28 days of co-culture, the longest time followed in vitro, tumor cells were controlled and did not outgrow. Increased proliferation of anti-CD117-CAR T-cells in the presence of mast cells was observed and tracked throughout the 28-day experiment. In conclusion, we demonstrate that the human mast cell lines HMC-1.1 KIT V560G, HMC-1.2 KIT V560G, KIT D816V, ROSA KIT WT, ROSA KIT D816V, LAD2 and MCPV-1 can be efficiently targeted and killed in vitro by allogeneic anti-CD117-CAR T-cells. Given that CD117 is expressed on healthy hematopoietic stem and progenitor cells (HSPCs) on a substantially lower level, there might be a therapeutic window for anti-CD117 immunotherapy in advanced forms of mastocytosis. However, as CAR T-cells are highly efficient, collateral damage on healthy HSPCs will likely need to be compensated by subsequent HSC transplantation. We are currently translating these promising in vitro immunotherapeutic settings into surrogate xenogeneic in vivo models. Disclosures Myburgh: University of Zurich: Patents & Royalties: CD117xCD3 TEA. Manz: CDR-Life Inc: Consultancy, Current holder of stock options in a privately-held company; University of Zurich: Patents & Royalties: CD117xCD3 TEA.


Sign in / Sign up

Export Citation Format

Share Document