scholarly journals A mouse model of disseminated mantle cell lymphoma highlights a lack of activity of estrogen receptor β agonists toward tumor burden

2017 ◽  
Vol 59 (7) ◽  
pp. 1726-1729
Author(s):  
Simon Body ◽  
Anna Esteve-Arenys ◽  
Clara Recasens-Zorzo ◽  
Xavier Troussard ◽  
Gaël Roué ◽  
...  
2012 ◽  
Vol 19 (1) ◽  
pp. 85-95 ◽  
Author(s):  
Anil Prasad ◽  
Ashutosh Shrivastava ◽  
Evangelos Papadopoulos ◽  
Paula M. Kuzontkoski ◽  
M.V. Ramana Reddy ◽  
...  

Haematologica ◽  
2015 ◽  
Vol 100 (11) ◽  
pp. e458-e461 ◽  
Author(s):  
M. Rudelius ◽  
H. Rauert-Wunderlich ◽  
E. Hartmann ◽  
E. Hoster ◽  
M. Dreyling ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3728-3728
Author(s):  
Lapo Alinari ◽  
Qing Liu ◽  
Ching-Shih Chen ◽  
Fengting Yan ◽  
James T Dalton ◽  
...  

Abstract Abstract 3728 Poster Board III-664 Over-expression of Cyclin D1 and constitutive phosphorylation of Akt has been implicated in the pathogenesis of mantle cell lymphoma (MCL). Here we describe FTY720 (fingolimod), an immunosuppressive agent currently being explored in phase III studies in renal transplantation and multiple sclerosis patients, to mediate time- and dose-dependent cell death in primary MCL cells (6 patients) and MCL cell lines, Jeko and Mino. FTY720-induced apoptosis was associated with reactive oxygen species (ROS) generation, Bax up-regulation but not associated with caspase 3 activation in MCL. FTY720 treatment resulted in time-dependent down-modulation of Cyclin D1 and phospho Akt (p-Akt) protein level, two critical disease-relevant molecules in the pathogenesis of MCL. Consistent with the modulation of Cyclin D1, FTY720-induced cell cycle arrest with accumulation of cells in G0/G1 and G2/M phases of the cell cycle with concomitant decrease in S phase entry. Importantly, FTY720 treatment was also associated with a time-dependent phospho Erk (p-Erk) induction in Mino and Jeko cells. To determine the in vivo efficacy of FTY720, we developed a preclinical, in vivo xenograft model of human MCL where MCL cell lines (Jeko, Mino and SP53) were engrafted into severe combined immune deficient (SCID) mice. Cell dose titration trials identified 4 × 107 Mino or Jeko cells injected intravenously via tail vein to result in consistent engraftment and fatal tumor burden in all mice. All mice engrafted with 4 × 107 Jeko cells developed a disseminated disease within 3 weeks and had a median survival of 28 days (compared to 43 days for Mino and 51 days for SP53). Because the Jeko cell line was established from the peripheral blood of a patient with blastic variant MCL and demonstrated a more resistant phenotype to several immuno-chemoterapeutic compounds, this cell line was chosen to create a more stringent in vivo preclinical model. SCID mice were treated with the monoclonal antibody TMβ1 to deplete murine NK cells, engrafted with 4 × 107 Jeko cells and observed daily for signs of tumor burden. Ten mice/group were treated starting at day 15 post-engraftment with intraperitoneal injection of 100 μl of saline or FTY720 (5 mg/kg resuspended in 100 μl of saline), every day, for two weeks. The median survival for FTY720-treated mice (N=10) was 38 days (95% CI:30-39) compared to 26.5 days (95% CI: 26-27 days) for the control group mice (N=10). The results from the log-rank test indicated an overall statistical significant difference in survival functions between the FTY720 treatment and the control group (p=0.001). These results provide the first evidence for a potential use of FTY720 in targeting key pathways that are operable in the pathogenesis of MCL and warrant the further investigation of FTY720 in combination with other agents in clinical trials treating patients with MCL. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 645-645 ◽  
Author(s):  
Xiaoxian Zhao ◽  
Juraj Bodo ◽  
Danyu Sun ◽  
Jeffrey J. Lin ◽  
Lisa Durkin ◽  
...  

Abstract Background Mantle cell lymphoma (MCL) is an aggressive subtype of Non-Hodgkin Lymphoma associated with poor prognosis. Constitutive activation of B-cell receptor (BCR) signaling plays an essential role for the survival and proliferation of malignant B-cells. Targeting Bruton’s tyrosine kinase (BTK), a component of BCR signaling pathway, with ibrutinib is a promising strategy. As a single agent, complete and partial response rates of 21% and 47%, respectively, were observed in a phase 2 study for relapsed or refractory MCL. Simultaneous inhibition of multiple biologic pathways has the potential to result in a synergism. We combined ibrutinib with ABT-199, a BH3 mimetic that selectively targets the BCL-2 pathway, and tested their in vitroefficacy against MCL. Experimental design A novel MCL cell line, CCMCL1, and four other MCL cell lines (Jeko-1, Mino, JVM2, Rec-1) were used for flow cytometry-based apoptosis and cell cycle analyses to evaluate the combinational effect of ibrutinib with ABT-199 (ChemieTek. Indianpolis. IN). The interaction between drugs was examined with Calcusyn software and combination index values served to determine the combined effect as synergistic (<1), additive (=1), or antagonistic (>1). Immunoblotting was performed to investigate signaling pathways of MCL cells exposed to these agents. Results CCMCL1 was derived from primary leukemic MCL cells. Cells were initially directly injected via tail vein into an NSG mouse. Engrafted cells were then isolated at 10 weeks from spleen and placed into routine cell culture. Immunophenotyping showed CCMCL1 cells have similar characteristics as the primary patient MCL cells, which expressed CD5, CD19, CD20, FMC7 and monotypic kappa light chain. Immunohistochemical staining of engrafted mouse spleen tissue showed expression of cyclin D1 and SOX11. The karyotype is highly complex with an IGH@/CCND1 fusion by metaphase FISH. In addition to spleen, MCL cell infiltration was observed in mouse liver, bone marrow, blood, brain, lung, kidney and intestine. In vitro cultured CCMCL1 cells underwent apoptosis upon expose to ibrutinib and ABT-199 as single agents. Combination of these two drugs resulted in synergistic induction of apoptosis (Table 1). Synergism was also observed with Jeko-1, Mino, JVM-2 and Rec-1 cells. Immunoblotting showed CCMCL1 cells have constitutive expression of cyclin D1, SOX11, PAX5 and MCL1. Ibrutinib as a single agent induced a rapid down-regulation of SOX11 and MCL1, while combination of ibrutinib with ABT-199 further enhanced down regulation of SOX11, followed by down-regulation of PAX5 at a later time point. We are currently testing the in vivo efficacy of combining these two drugs in a CCMCL1 NSG mouse model. Conclusion Our CCMCL1/NSG mouse model is a new model for pre-clinical assessment of MCL treatment approaches. Combination of ibrutinib with ABT-199 has synergistic effect of apoptotic induction in CCMCL1 as well as four other MCL cell lines tested. Ibrutinib or ibruitnib/ABT-199 combination induced apoptosis of MCL is associated with down-regulation of SOX11 and PAX5. Simultaneous down regulation of MCL1 via ibrutinib and targeting of BCL2 may contribute to the in vitrosynergism observed. These data support further investigation of this novel therapeutic strategy. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1748-1748
Author(s):  
Tim Pieters ◽  
Steven Goossens ◽  
Sara T'Sas ◽  
Geert Berx ◽  
Jody J. Haigh ◽  
...  

Abstract Mantle cell lymphoma (MCL) is a highly aggressive subtype of B-cell lymphoma that is characterized by a poor response to current treatment regimens. Most MCLs carry a prototypical translocation, t(11;14), which juxtaposes the CCND1 gene towards the immunoglobulin heavy chain (IGH) locus, resulting in cyclin D1 overexpression. Strikingly, MCL has not been recapitulated in transgenic mouse models of Ccnd1 overexpression alone. Notably, a subset of MCL patients are cyclin D1 negative but instead overexpress cyclin D2 (encoded by CCND2)as a consequence of recurrent genomic rearrangements involving the CCND2 locus. Here, we developed a novel conditional ROSA26-driven Ccnd2 overexpression mouse model and showed that hematopoietic-specific Ccnd2 activation is sufficient to drive MCL formation in mice. Starting from 36 weeks, these mice develop huge B-cell lymphomas and these tumor cells have the typical MCL morphology, are Sox11 positive and disseminate into other organs, all typical features of MCL. In addition, preliminary shallow sequencing analysis revealed a somatic Crlf2 deletion as a cooperative genetic event in one of the murine Ccnd2-driven MCL tumors. In this study, we want to further validate and characterize this novel Ccnd2-driven mouse model for MCL and test putative synergisms between Ccnd2 overexpression and other recurrent cooperating genetic lesions that occur in human MCL, such as loss of p53 or SOX11 activation. Noteworthy, the MCL cells from this mouse model also contain a luciferase reporter, allowing accurate in vivo tracing of tumor cells in xenograft experiments. These xenograft experiments can be used as preclinical models, in which bioluminescence is used to asses the tumor burden and to monitor tumor regression upon drug treatment. In conclusion, we have developed a novel mouse model in which Ccnd2 overexpression faithfully recapitulates MCL and this model will allow us to understand the molecular mechanisms that drive MCL and identify and test novel drugs to treat this aggressive and until now incurable disease. Disclosures No relevant conflicts of interest to declare.


Author(s):  
Archana Vijaya Kumar ◽  
Carmen Donate ◽  
Beat A. Imhof ◽  
Thomas Matthes

2019 ◽  
Vol 73 ◽  
pp. 303-309 ◽  
Author(s):  
Monika Długosz-Danecka ◽  
Katarzyna Krawczyk ◽  
Bogdan Ochrem ◽  
Michał Szymczyk ◽  
Monika Joks ◽  
...  

Aim: Mantle cell lymphoma (MCL) patients have poor prognosis, due to development of chemoresistance in relapsing patients. Therefore, despite the features of indolent lymphoma at presentation, consolidation and/or maintenance strategies to achieve minimal tumor burden and postpone subsequent relapses remain the standard of care. Material/Methods: In the retrospective analysis of Polish Lymphoma Research Group (PLRG), post-induction strategies were assessed in 355 MCL patients. Those in complete or partial remission (CR or PR) after induction regimen (n = 263, 74.08%) were consolidated with autologous stem cell transplantation (ASCT) (n = 71, 20%) or radioimmunotherapy (RIT, n = 37, 10.42%), subjected to rituximab maintenance (MR, n = 17, 4.79%) or had none post-induction treatment (NPI, n = 138, 38.87%). Responses to therapy, progression and overall survival (PFS and OS) were compared. Results: CR after induction was significantly increased by consolidation strategies, from 68% to 95% in ASCT and from 43% to 90% in RIT cohort. Median PFS in patients subjected to ASCT and RIT was significantly higher compared to NPI group (2.3, 3.8 and 1.8 years respectively, p <0.05). Consolidation strategies also prolonged median OS (not reached in ASCT, 7.3 in RIT and 4 years in NPI cohort, p <0.005 ). Conclusions: Our data confirms the efficacy of ASCT consolidation in selected patients and demonstrates that RIT may be regarded as an alternative consolidation strategy for patients not eligible for ASCT. At the time the study was performed, there were few possibilities to retreat elderly and comorbid patients; therefore, despite PFS benefit of RIT, younger, transplanted patients experienced a longer median OS.


2021 ◽  
Vol 11 ◽  
Author(s):  
Lixia Zhou ◽  
Tenghua Yu ◽  
Fei Yang ◽  
Jingjing Han ◽  
Bin Zuo ◽  
...  

Mantle cell lymphoma (MCL) is an aggressive form of non-Hodgkin’s B-cell lymphoma with poor prognosis. Despite recent advances, resistance to therapy and relapse remain significant clinical problems. G-protein-coupled estrogen receptor (GPER)-mediated estrogenic rapid signaling is implicated in the development of many cancers. However, its role in MCL is unknown. Here we report that GPER activation with selective agonist G-1 induced cell cycle arrest, DNA damage, mitochondria membrane potential abnormality, and eventually apoptosis of MCL cell lines. We found that G-1 induced DNA damage and apoptosis of MCL cells by promoting the expression of nicotinamide adenine dinucleotide phosphate oxidase and the generation of reactive oxygen species. In addition, G-1 inhibited MCL cell proliferation by inactivation of NF-κB signaling and exhibited anti-tumor functions in MCL xenografted mice. Most significantly, G-1 showed synergistic effect with ibrutinib making it a potential candidate for chemotherapy-free therapies against MCL.


2021 ◽  
Vol 218 (10) ◽  
Author(s):  
Anna E. Beaudin

Therapeutic discovery for mantle cell lymphoma (MCL) has been hindered by a lack of preclinical mouse models that recapitulate human disease. In this issue, Pieters and colleagues (2021. J. Exp. Med.https://doi.org/10.1084/jem.20202280) establish a novel mouse model of MCL driven by overexpression of cyclin D2 and identify fetal-derived B1a cells as putative cell of origin for MCL.


Sign in / Sign up

Export Citation Format

Share Document