The rGel/BLyS fusion toxin inhibits STAT3 signaling via down-regulation of interleukin-6 receptor in diffuse large B-cell lymphoma

2010 ◽  
Vol 80 (9) ◽  
pp. 1335-1342 ◽  
Author(s):  
Mi-Ae Lyu ◽  
Bokyung Sung ◽  
Lawrence H. Cheung ◽  
John W. Marks ◽  
Bharat B. Aggarwal ◽  
...  
2015 ◽  
Vol 44 (3) ◽  
pp. 265-278 ◽  
Author(s):  
Roba M. Talaat ◽  
Amal M. Abdel-Aziz ◽  
Eman A. El-Maadawy ◽  
Naser Abdel-Bary

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 926-926
Author(s):  
Y. Lynn Wang ◽  
Jiao Ma ◽  
Wei Xing ◽  
Pin Lu ◽  
Karen Dresser ◽  
...  

Abstract Non-Hodgkin Lymphoma (NHL) represents about 5 percent of all cancers diagnosed in the United States. While incidence of NHL has increased slightly over the past decade, death rates have been declining steadily. These declines in mortality can be attributed to improvements in treatment that are based on an increased understanding of the biology of the disease. Diffuse large B-cell lymphoma (DLBCL) accounts for ~30% of NHLs and greater than 80% of aggressive NHLs. Recent studies including large-scale genetic analyses have demonstrated the critical roles of the B-cell receptor’s (BCR) and JAK/STAT pathways in DLBCL. Herein, we investigated the anti-lymphoma activity of cerdulatinib (aka PRT062070), a novel compound that dually targets both SYK and JAK/STAT signaling pathways. To determine whether targeting both SYK and JAK/STAT is relevant in DLBCL, we examined the expression of p-SYK (pY525/526) and p-STAT3 (pY705) on a tissue microarray of 62 DLBCL primary tumors, including 41 GCB and 21 non-GCB cases. p-SYK expression was detected in 29 (47%) cases with a characteristic peri-membrane staining pattern. Of those 29 p-SYK positive cases, 17 were GCB type (17/41, 41%) and 12 were non-GCB type (12/21, 57%). p-STAT3 exhibits a characteristic nuclear staining pattern in DLBCL cases. A total of 26 (42%) stained positive for p-STAT3; 16 were GCB type (16/41, 39%) and 10 were non-GCB type (10/21, 48%). Interestingly, there are 19 cases (31%) with reactivity for both p-SYK and p-STAT3, among which, 11 were GCB type (27%) and 8 were non-GCB type (38%). SYK and STAT3 are also phosphorylated in a panel of nine DLBCL cell lines. Immunoblotting analyses showed that ABC and GCB subtypes of DLBCL cells appear to exhibit different JAK/STAT and BCR signaling profiles. For instance, p-AKT was highly expressed in GCB cells, whereas p-STAT3 was more strongly expressed in ABC cells. Overall, the DLBCL cells are more sensitive to the dual inhibitor than to the SYK-specific inhibitor alone. In both GCB and ABC cell lines, cerdulatinib induced apoptosis via down-regulation of MCL1 protein and PARP cleavage. The compound also blocked G1/S transition and caused cell cycle arrest through inhibition of RB phosphorylation and down-regulation of cyclin E. Further analyses of the cell signaling activities showed that STAT3 phosphorylation was sensitive to inhibition by cerdulatinib in ABC cell lines while phosphorylation of SYK, PLCg2, AKT and ERK was sensitive to inhibition by cerdulatinib in GCB cell lines. Importantly, JAK/STAT and BCR signaling can be blocked by cerdulatinib in GCB and non-GCB primary human DLBCL cells, which led to cell death of these cells. Our work provided mechanistic insights into the actions of SYK/JAK dual inhibitor cerdulatinib, suggesting that the drug may be a potent treatment of DLBCL with a broader anti-tumor activity in both ABC and GCB subtypes of the lymphoma. Disclosures Pandey: Portola Pharmaceuticals: Employment. Conley:Portola Pharmaceuticals: Employment. Coffey:Portola Pharmaceuticals: Employment.


2020 ◽  
Vol 59 (23) ◽  
pp. 3061-3065
Author(s):  
Kohei Shiroshita ◽  
Taku Kikuchi ◽  
Mikio Okayama ◽  
Hidenori Kasahara ◽  
Takahiro Kamiya ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 817-817 ◽  
Author(s):  
Mohamed Rahmani ◽  
Mandy Aust ◽  
LaShanale Wallace ◽  
Steven Grant

Abstract Dysregulation of the PI3K/AKT/mTOR pathway and histone deacetylases (HDACs) has been described in diffuse large B-cell lymphoma (DLBCL). Previous studies demonstrated that combined treatment with PI3K/AKT pathway signaling inhibitors (e.g., LY294002, or perifosine) and histone deacetylase inhibitors (HDACis, e.g., sodium butyrate, vorinostat) resulted in a dramatic induction of apoptosis in human myeloid leukemia cells (Rahmani et al., Oncogene 22:6231-42, 2003; Rahmani et al., Cancer Res 65:2422-32, 2005). Here we examined interactions between the clinically relevant dual PI3K/mTOR inhibitor BEZ235 and the pan-HDACi panobinostat (both Novartis) in DLBCL cells. Notably, combined treatment with BEZ235 (25-200 nM) and panobinostat (5-15 nM) resulted in sharp decreases in cell growth and viability and profound induction of mitochondrial dysfunction and apoptosis. These events were observed in various DLBCL cell lines including both GC-DLBCL (SUDHL4, SUDHL16, OCI-LY7) and ABC-DLBCL (HBL-1, TMD8) as well as in mantle cell lymphoma cells (JeKo-1). Enhanced lethality of this regimen was accompanied by a marked increase in cytochrome c and AIF release into the cytosol, caspase-3 activation, and PARP cleavage. It was also associated with down-regulation of Mcl-1, a pronounced increase in H3 and H4 acetylation, and up-regulation of phospho- gH2AX, an indicator of DNA damage (e.g., DNA double-strand breaks). In addition, panobinostat robustly induced p21CIP1 accumulation in DLBCL cells, an event that was largely abrogated by BEZ235. Of note, treatment with BEZ235 alone or in combination with panobinostat triggered a decrease in GSK3 phosphorylation and levels of its downstream target, b-catenin consistent with GSK3 activation. Interestingly, inhibition of GSK3 by CHIR-98014 or the GSK3 inhibitor IX (BIO), but not its inactive analogue MeBIO, significantly diminished BEZ235/panobinostat lethality, arguing that GSK3 activation plays a significant functional role in lethality. Immunoprecipitation studies revealed that down-regulation of Mcl-1 was associated with enhanced binding of Bim to Bcl-xL and Bcl-2 and a marked decrease in Bak binding to Bcl-xL. In addition, knockdown of Bak also markedly diminished BEZ235/panobinostat-mediated lethality, as assessed by Annexin V/PI positivity. Together, these findings suggest that Bak plays a key functional role in the pronounced activity of BEZ235/panobinostat toward DLBCL cells. They also raise the possibility that BEZ235 may enhance panobinostat lethality by increasing Bim binding to Bcl-xL/Bcl-2, leading to the release of Bak/Bax from Bcl-2/Bcl-xL, culminating in apoptosis. Significantly, HS-5 stromal cell-conditioned media failed to protect DLBCL cells from combined panobinostat/BEZ235 treatment, suggesting that this strategy may be effective in circumventing microenvironmental forms of resistance. Combined treatment also exhibited robust activity against primary DLBCL cells, whereas exposure to the same regimens did not significantly reduce the viability of normal CD34+ progenitor cells nor did it reduce their clonogenic potential. Finally, in vivo studies utilizing a murine xenograft model bearing SUDHL4 cells revealed that co-treatment with BEZ235 and panobinostat markedly reduced in vivo tumor growth, whereas agents administered individually exhibited only modest effects. In addition, Kaplan-Meier analysis revealed that combined treatment significantly prolonged the survival of mice; in contrast, single agents were ineffective in increasing survival. Together, these findings suggest that the anti-DLBCL activities of combined BEZ235 and panobinostat treatment may involve multiple mechanisms, including Mcl-1 down-regulation, increased Bim binding to Bcl-2/Bcl-xL, release of Bak from Bcl-xL, abrogation of p21CIP1 accumulation, induction of DNA damage, and GSK3 activation, culminating in Bax/Bak activation and apoptosis. These findings raise the possibility that combining PI3K/AKT/mTOR inhibitors (e.g., BEZ235) and HDACis (e.g., panobinostat), may represent a novel and effective strategy against various DLBCL subtypes and possibly other hematologic malignancies. Disclosures: No relevant conflicts of interest to declare.


Neoplasia ◽  
2010 ◽  
Vol 12 (5) ◽  
pp. 366-375 ◽  
Author(s):  
Mi-Ae Lyu ◽  
Deepak Rai ◽  
Kwang Seok Ahn ◽  
Bokyung Sung ◽  
Lawrence H. Cheung ◽  
...  

Author(s):  
Shunfeng Hu ◽  
Shuai Ren ◽  
Yiqing Cai ◽  
Jiarui Liu ◽  
Yang Han ◽  
...  

Abstract Glycoprotein prostaglandin D2 synthase (PTGDS) is a member of the lipocalin superfamily and plays dual roles in prostaglandins metabolism and lipid transport. PTGDS has been involved in various cellular processes including the tumorigenesis of solid tumors, yet its role in carcinogenesis is contradictory and the significance of PTGDS in hematological malignancies is ill-defined. Here, we aimed to explore the expression and function of PTGDS in diffuse large B-cell lymphoma (DLBCL), especially the potential role of PTGDS inhibitor, AT56, in lymphoma therapy. Remarkable high expression of PTGDS was found in DLBCL, which was significantly correlated with poor prognosis. PTGDS overexpression and rhPTGDS were found to promote cell proliferation. Besides, in vitro and in vivo studies indicated that PTGDS knockdown and AT56 treatment exerted an anti-tumor effect by regulating cell viability, proliferation, apoptosis, cell cycle, and invasion, and enhanced the drug sensitivity to adriamycin and bendamustine through promoting DNA damage. Moreover, the co-immunoprecipitation-based mass spectrum identified the interaction between PTGDS and MYH9, which was found to promote DLBCL progression. PTGDS inhibition led to reduced expression of MYH9, and then declined activation of the Wnt-β-catenin-STAT3 pathway through influencing the ubiquitination and degradation of GSK3-β in DLBCL. The rescue experiment demonstrated that PTGDS exerted an oncogenic role through regulating MYH9 and then the Wnt-β-catenin-STAT3 pathway. Based on point mutation of glycosylation sites, we confirmed the N-glycosylation of PTGDS in Asn51 and Asn78 and found that abnormal glycosylation of PTGDS resulted in its nuclear translocation, prolonged half-life, and enhanced cell proliferation. Collectively, our findings identified for the first time that glycoprotein PTGDS promoted tumorigenesis of DLBCL through MYH9-mediated regulation of Wnt-β-catenin-STAT3 signaling, and highlighted the potential role of AT56 as a novel therapeutic strategy for DLBCL treatment.


PLoS Genetics ◽  
2014 ◽  
Vol 10 (1) ◽  
pp. e1004105 ◽  
Author(s):  
Krystyna Mazan-Mamczarz ◽  
X. Frank Zhao ◽  
Bojie Dai ◽  
James J. Steinhardt ◽  
Raymond J. Peroutka ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document