Malignant hematopoietic cell lines: In vitro models for the study of Waldenström's macroglobulinemia

2008 ◽  
Vol 32 (11) ◽  
pp. 1669-1673 ◽  
Author(s):  
Hans G. Drexler ◽  
Roderick A.F. MacLeod
2015 ◽  
Vol 39 (1) ◽  
pp. 18-29 ◽  
Author(s):  
Hans G. Drexler ◽  
Stefan Ehrentraut ◽  
Stefan Nagel ◽  
Sonja Eberth ◽  
Roderick A.F. MacLeod

Blood ◽  
2020 ◽  
Author(s):  
Antonio Sacco ◽  
Cinzia Federico ◽  
Arianna Giacomini ◽  
Cinzia Caprio ◽  
Federica Maccarinelli ◽  
...  

The human fibroblast growth factor/fibroblast growth factor-receptor (FGF/FGFR) axis deregulation is largely involved in supporting the pathogenesis of hematologic malignancies, including Waldenström's Macroglobulinemia (WM). WM is still an incurable disease, and patients succumb due to disease progression. Therefore, novel therapeutics designed to specifically target deregulated signaling pathways in WM are required. We aimed to investigate the role of FGF/FGFR system blockade in WM by using a pan-FGF trap molecule (NSC12). Wide-transcriptome profiling confirmed inhibition of FGFR-signaling in NSC12-treated WM cells; unveiling a significant inhibition of MYD88 also confirmed at protein level. Importantly, the NSC12-dependent silencing of MYD88 was functionally active, as it led to inhibition of MYD88-driven pathways, such as BTK and SYK, as well as the MYD88-downstream target HCK. Of note, both canonical and non-canonical NFkB cascades were down-regulated in WM cells upon NSC12 treatment. Functional sequelae exerted by NSC12 in WM cells were studied, demonstrating significant inhibition of WM cell growth, induction of WM cell apoptosis, halting MAPK, JAK/STAT3 and PI3K-Akt pathways. Importantly, NSC12 exerted an anti-WM effect even in the presence of bone marrow microenvironment, both in vitro and in vivo. Our studies provide the evidence for using NSC12 as a specific FGF/FGFR system inhibitor, thus representing a novel therapeutic strategy in WM.


2004 ◽  
Vol 28 (12) ◽  
pp. 1243-1251 ◽  
Author(s):  
Hans G. Drexler ◽  
Yoshinobu Matsuo ◽  
Roderick A.F. MacLeod

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4929-4929 ◽  
Author(s):  
Olivier Tournilhac ◽  
Daniel Ditzel Santos ◽  
Lian Xu ◽  
Evdoxia Hatjiharissi ◽  
Yu Tsu ◽  
...  

Abstract Recently, we demonstrated that in Waldenstrom’s macroglobulinemia (WM), clonal lymphoplasmacytic cells (LPC) appear to derive growth and survival signals from excess mast cells (MC) present in the bone marrow (BM). (Tournilhac et al, JCO 2004 22:571S). We therefore have sought agents which could target both LPC and MC. One such candidate is imatinib mesylate which has shown activity in certain mast cell disorders likely on the basis of its ability to target the tyrosine kinases, CD117 (Stem Cell Factor Receptor, c-kit), and Platelet Derived Growth Factor Receptor (PDGFa-R). By flow cytometric analysis, we demonstrated CD117 expression on sorted LPC from 17/22 (76.2%) WM patients, above 20% in 13 cases, whose expressionwe confirmed by RT-PCR analysis. Moreover, we also demonstrated expression of PDGFa-R along with its ligand PDGFa in BM LPC from 16/18 (88.9%) and 7/10 (70%) WM patients.. In addition to CD117, we found by RT-PCR analysis a PDGFa expression in LAD and HMC-1 MC lines and in KU the basophilic cell lines KU as well as in sorted BM MC (FcER1+ CD117+) from 3/10 (30%) WM patients. Importantly, co-culture of sorted LPC from WM patients along with 0.5% paraformaldehyde fixed LAD cells or ex vivo expanded (EVE) BM MC induced proliferation of sorted LPC from WM patients, which was inhibited at pharmacologically achievable levels of imatinib mesylate with an IC50 of 0.5–10 μmoles/L. Moreover, imatinib mesylate inhibited the LAD, HMC-1 and KU cell lines as well as EVE cord blood MC or EVE BM MC at an IC50 of 0.01–1 μmoles/L. Lastly, imatinib mesylate also inhibited proliferation of both MC and LPC when unfixed LAD or ex vivo MC were incubated with WM LPC with an IC50 of 1–10 μmoles/L. These studies therefore demonstrate that both LPC and MC are therapeutic targets for imatinib mesylate, and provide the framework for clinical studies evaluating its use in WM.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2688-2688
Author(s):  
Lucy S. Hodge ◽  
Steven C. Ziesmer ◽  
Frank J Secreto ◽  
Zhi-Zhang Yang ◽  
Anne J. Novak ◽  
...  

Abstract Abstract 2688 Members of the signal transducers and activators of transcription (STAT) family of proteins function as secondary messengers mediating cellular responses to various cytokines. Aberrant activation of STAT5 protein has been implicated in the pathogenesis of hematologic malignancies due to the ability of these transcription factors to regulate genes involved in cellular proliferation and survival. The two highly homologous transcription factors collectively known as STAT5, namely STAT5A and STAT5B, display both redundant and distinct physiologic functions in non-malignant B cells. Yet, despite evidence suggesting independent functions for STAT5A and STAT5B in the pathophysiology of solid tumors, the precise roles of these isoforms in hematologic malignancies such as Waldenstrom's macroglobulinemia (WM) are not known. Initial immunohistochemical staining of WM bone marrow biopsies (n=6) revealed higher levels of STAT5 phosphorylation compared to bone marrows from normal controls (n=6). Additionally, phosphorylated STAT5 was highly expressed in CD19/CD138+ sorted cells obtained from the bone marrow of patients with WM (average ΔMFI compared to isotype control, 4.64 +/− 1.7, n=6) as determined by FACS analysis. Furthermore, western blotting demonstrated baseline STAT5 phosphorylation in both WM and IgM-secreting cell lines (MWCL-1 and BCWM-1, respectfully). The expression of other phosphorylated STAT proteins (1, 3, 4, and 6) was not detected. To better understand the overall role of STAT5 biologically in WM, MWCL-1 and BCWM.1 cells were treated with a specific inhibitor of STAT5, N′-((4-Oxo-4H-chromen-3-yl)methylene) nicotinohydrazide. Following 72 hours in culture, this inhibitor significantly decreased IgM secretion at lower doses when compared to controls (10 μM, MWCL-1: 9413 +/− 511 ng/mL vs. 6166 +/− 160 ng/mL, p<0.001; BCWM.1: 13,380 +/− 393 ng/mL +/− 10970 +/− 240 ng/mL, p<0.001) and nearly abolished IgM secretion at higher doses. Significant effects on proliferation and viability were also observed, but only at higher concentrations of drug where the specificity toward STAT5 may have diminished. To characterize the roles of the individual STAT5 isoforms in WM, we have developed doxycycline-inducible STAT5A-shRNA and STAT5B-shRNA and used these to create stable knockdowns in the MWCL-1 cell line. Addition of doxycycline to these cell lines for 72h nearly abolishes STAT5A or STAT5B protein expression, respectively, without affecting expression of the other isoform, while no change in STAT5 expression is detected in cells transduced with the empty vector (EV) control. The functional effects of isoform-specific STAT5 inhibition on WM tumor biology were assessed through proliferation assays and ELISA to determine the utility of therapeutically targeting a single STAT5 isoform. No difference in proliferation was observed in the STAT5A- or STAT5B-shRNA cells following 72h induction with doxycycline as compared to the EV control cells. However, specific knockdown of STAT5A following doxycycline induction significantly decreased IgM secretion as compared to the EV control (EV, 91.8 +/− 10.2% of baseline secretion, STAT5A, 66.7 +/− 15.1% of baseline, p<0.05), whereas IgM secretion was decreased by knockdown of STAT5B but not significantly. Similarly, IL-6 secretion was only diminished in cells lacking STAT5A as compared to EV cells (EV, 94 +/− 14.16% of baseline secretion; STAT5A, 64% +/− 7.6% of baseline, p<0.001). Overall these initial data indicate a role for isoform-specific STAT5 activation in the maintenance of WM tumors, and future studies will further our understanding of the specific roles of STAT5A and STAT5B in WM tumors. Disclosures: Ansell: Seattle Genetics, Inc.: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document