scholarly journals The Akt pathway regulates survival and homing in Waldenstrom macroglobulinemia

Blood ◽  
2007 ◽  
Vol 110 (13) ◽  
pp. 4417-4426 ◽  
Author(s):  
Xavier Leleu ◽  
Xiaoying Jia ◽  
Judith Runnels ◽  
Hai T. Ngo ◽  
Anne-Sophie Moreau ◽  
...  

Waldenstrom macroglobulinemia (WM) is an incurable low-grade lymphoplasmacytic lymphoma. We demonstrate up-regulated Akt activity in WM, and that Akt down-regulation by Akt knockdown and the inhibitor perifosine leads to significant inhibition of proliferation and induction of apoptosis in WM cells in vitro, but not in normal donor peripheral blood and hematopoietic progenitors. Importantly, down-regulation of Akt induced cytotoxicity of WM cells in the bone marrow microenvironment (BMM) context. Perifosine induced significant reduction in WM tumor growth in vivo in a subcutaneous xenograft model through inhibition of Akt phosphorylation and downstream targets. We also demonstrated that Akt pathway down-regulation inhibited migration and adhesion in vitro and homing of WM tumor cells to the BMM in vivo. Proteomic analysis identified other signaling pathways modulated by perifosine, such as activation of ERK MAPK pathway, which induces survival of tumor cells. Interestingly, MEK inhibitor significantly enhanced perifosine-induced cytotoxicity in WM cells. Using Akt knockdown experiments and specific Akt and PI3K inhibitors, we demonstrated that ERK activation is through a direct effect, rather than feedback activation, of perifosine upstream ERK pathway. These results provide understanding of biological effects of Akt pathway in WM and provide the framework for clinical evaluation of perifosine in WM patients.

Blood ◽  
2010 ◽  
Vol 115 (3) ◽  
pp. 559-569 ◽  
Author(s):  
Aldo M. Roccaro ◽  
Antonio Sacco ◽  
Emanuel N. Husu ◽  
Costas Pitsillides ◽  
Steven Vesole ◽  
...  

Abstract We have previously shown clinical activity of a mammalian target of rapamycin (mTOR) complex 1 inhibitor in Waldenstrom macroglobulinemia (WM). However, 50% of patients did not respond to therapy. We therefore examined mechanisms of activation of the phosphoinositide 3-kinase (PI3K)/Akt/mTOR in WM, and mechanisms of overcoming resistance to therapy. We first demonstrated that primary WM cells show constitutive activation of the PI3K/Akt pathway, supported by decreased expression of phosphate and tensin homolog tumor suppressor gene (PTEN) at the gene and protein levels, together with constitutive activation of Akt and mTOR. We illustrated that dual targeting of the PI3K/mTOR pathway by the novel inhibitor NVP-BEZ235 showed higher cytotoxicity on WM cells compared with inhibition of the PI3K or mTOR pathways alone. In addition, NVP-BEZ235 inhibited both rictor and raptor, thus abrogating the rictor-induced Akt phosphorylation. NVP-BEZ235 also induced significant cytotoxicity in WM cells in a caspase-dependent and -independent manner, through targeting the Forkhead box transcription factors. In addition, NVP-BEZ235 targeted WM cells in the context of bone marrow microenvironment, leading to significant inhibition of migration, adhesion in vitro, and homing in vivo. These studies therefore show that dual targeting of the PI3K/mTOR pathway is a better modality of targeted therapy for tumors that harbor activation of the PI3K/mTOR signaling cascade, such as WM.


2008 ◽  
Vol 99 (4) ◽  
pp. 810-815 ◽  
Author(s):  
Dong Yu ◽  
Emiko Sekine ◽  
Akira Fujimori ◽  
Takahiro Ochiya ◽  
Ryuichi Okayasu

Tumor Biology ◽  
2015 ◽  
Vol 37 (4) ◽  
pp. 4979-4990 ◽  
Author(s):  
Yiqun Yao ◽  
Yongjian Liu ◽  
Xiupeng Lv ◽  
Bin Dong ◽  
Feng Wang ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2417-2417 ◽  
Author(s):  
Xavier Leleu ◽  
Hai Ngo ◽  
Judy Runnels ◽  
Costas Pitsillides ◽  
Joel Spencer ◽  
...  

Abstract Background: Waldenstrom Macroglobulinemia (WM) is a low-grade lymphoma characterized by widespread involvement of the bone marrow (BM) and involvement of lymph nodes and hepatosplenomegaly (HSM) in about 20% of the patients. We have recently demonstrated that the presence of HSM is one of the most important adverse prognostic factors in WM. The mechanisms of trafficking of WM cells to and from the BM and lymphoid organs is not well defined. The PI3k/Akt pathway is constitutively activated in WM, and regulates migration/homing in cancer cells and B-cells. We hypothesized that the Akt inhibitor perifosine (NSC 639966; Keryx Biopharmaceuticals, NY) modulates homing of WM cells to the BM. Methods: WM cell line (BCWM.1) was treated with perifosine 2 to 5uM for 2 hours. Surface adhesion receptors were studied using flow cytometry. The adhesion assay coated with fibronectin, a ligand of VLA-4 (EMD Biosciences, CA) was used to test in vitro adhesion. Migration was determined using the transwell migration assay (Costar, NY). We then studied homing of WM into the BM niches using in vivo flow cytometry and confocal microscopy in Balb/c mice. In brief, BCWM.1 were incubated with 5uM perifosine for 2 hrs (or control PBS). The cells were fluorescently labeled by incubation with the dialkylcarbocyanine membrane dye, “DiD” (Molecular Probes), 0.5uM dye for 30 minutes. Cells were then injected in the tail vein of the Balb/c mice, and in vivo confocal flow cytometry was performed on an artery from the ear lobe of the mice. Cell counts were obtained every 5 min. from the time of injection. In vivo confocal microscopy and two-photon microscopy was performed to study cells homing to BM vasculature of the skull (BM niches). Results: WM cells expressed very high levels of VLA-4, with a median 95% expression. BCWM.1 demonstrated increased adhesion to fibronectin-coated wells as compared to BSA-coated wells. Perifosine inhibited adhesion in a dose dependent manner with 50% decrease in adhesion at 2uM. Perifosine 10uM did not change the level of surface expression of VLA-4 after 6 and 24 hrs, indicating an effect on intracellular signaling but not on surface adhesion molecules. Perifosine 5uM significantly inhibited migration of BCWM.1 in response to SDF-1, a ligand that induces migration of WM cells. Perifosine also demonstrated significant inhibition of in vivo homing of WM cells to the BM niches. The number of cells in the peripheral circulation decreased dramatically (75% decrease) after 1 hr in the control, indicating homing, whereas there was a 40% reduction in the cells at 1hr after perifosine treatment (p=0.001). We then looked at the images obtained from the BM niches. The number of cells that homed and adhered to these areas was lower in the perifosine-treated mouse compared to the control mouse, indicating that fewer cells homed to the BM of the treated mouse. Conclusion: These results confirm that the PI3K/Akt pathway is important for migration, adhesion and homing of WM in vitro and in vivo.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2488-2488 ◽  
Author(s):  
Xavier Leleu ◽  
Xiaoying Jia ◽  
Anne-Sophie Moreau ◽  
Evdoxia Hatjiharisi ◽  
Hai Ngo ◽  
...  

Abstract Background: Waldenstrom’s Macroglobulinemia (WM) is a low-grade lymphoplasmacytic lymphoma with limited options of therapy. The PI3k/Akt pathway is a critical regulator of cell survival. Our previous studies using proteomic analysis have demonstrated upregulation of members of the PI3k/Akt pathway in WM. We examined whether the new Akt inhibitor perifosine (NSC 639966; Keryx, NY) induces cytotoxicity in WM. Methods: WM cell lines (BCWM1 and WSU-WM) and IgM secreting low-grade lymphoma cell lines (MEC1, RL) were used. Primary CD19+ malignant cells were obtained from patients after informed consent. Inhibition of proliferation was measured using the MTT assay; DNA synthesis was measured using the thymidine uptake assay and apoptosis using Apo2.7 flow cytometry. Bone marrow stromal cells (BMSC) confer growth and resistance to conventional treatments. We therefore, tested the effect of perifosine on WM cells co-cultured with BMSC. Immunoblotting for signaling pathways was performed at different time (30 minutes to 16 hrs) and doses of therapy. In vivo activity of perifosine was assessed using a SCID-irradiated model with subcutaneous tumors in which perifosine was administered by oral gavage daily (35 mg/kg/day). A two-sided t-test was used to determine statistical differences. Results: Perifosine inhibited phosphorylation of Akt in a dose- and time- dependent fashion, as well as downstream GSK3a/b and ribosomal phospho-S6. Perifosine inhibited Akt activity as confirmed by Akt kinase assay. Perifosine induced significant cytotoxicity and inhibition of DNA synthesis with an IC50 of 5-20uM in all cell lines tested. Similar effects were observed in primary CD19+ patient WM cells. Perifosine induced apoptosis in WM cells as demonstrated by flow cytometry. The mechanism of apoptosis induced by perifosine was through activation of SAPK/JNK pathway, followed by caspase-8, -9 and PARP cleavage. The JNK inhibitor SP600125 abrogated perifosine-induced apoptosis. The growth inhibitory effects of perifosine were significant even in the presence of BMSC, IL-6 and IGF-1, which induce resistance to conventional therapies. Importantly, perifosine did not induce cytotoxicity in healthy donor peripheral blood mononuclear cells or in hematopietic stem cells in a methylcellulose colony forming cell assay, indicating lack of toxicity on normal cells. Interestingly, MAPK members such as MEK/ERK were activated by perifosine. The MEK inhibitor U0126 significantly enhanced perifosine-induced cytotoxicity in WM cells, indicating that this combination may be synergistic in vivo. Finally, perifosine induced significant reduction in WM tumor growth in vivo, as compared to control cohort treated with vehicle only at week 6 (p=0.05). Conclusion: Perifosine has significant antitumor activity in WM both in vitro and in vivo. These results provide the framework for clinical evaluation of perifosine in WM. Supported in part by the Leukemia and Lymphoma Society, the Lymphoma Research Foundation and an American Society of Hematology Scholar Award. * XL and XJ are co-first authors.


Blood ◽  
2009 ◽  
Vol 113 (18) ◽  
pp. 4391-4402 ◽  
Author(s):  
Aldo M. Roccaro ◽  
Antonio Sacco ◽  
Changzhong Chen ◽  
Judith Runnels ◽  
Xavier Leleu ◽  
...  

AbstractMultilevel genetic characterization of Waldenström macroglobulinemia (WM) is required to improve our understanding of the underlying molecular changes that lead to the initiation and progression of this disease. We performed microRNA-expression profiling of bone marrow–derived CD19+ WM cells, compared with their normal cellular counterparts and validated data by quantitative reverse-transcription–polymerase chain reaction (qRT-PCR). We identified a WM-specific microRNA signature characterized by increased expression of microRNA-363*/-206/-494/-155/-184/-542-3p, and decreased expression of microRNA-9* (ANOVA; P < .01). We found that microRNA-155 regulates proliferation and growth of WM cells in vitro and in vivo, by inhibiting MAPK/ERK, PI3/AKT, and NF-κB pathways. Potential microRNA-155 target genes were identified using gene-expression profiling and included genes involved in cell-cycle progression, adhesion, and migration. Importantly, increased expression of the 6 miRNAs significantly correlated with a poorer outcome predicted by the International Prognostic Staging System for WM. We further demonstrated that therapeutic agents commonly used in WM alter the levels of the major miRNAs identified, by inducing downmodulation of 5 increased miRNAs and up-modulation of patient-downexpressed miRNA-9*. These data indicate that microRNAs play a pivotal role in the biology of WM; represent important prognostic marker; and provide the basis for the development of new microRNA-based targeted therapies in WM.


2019 ◽  
Vol 26 (3) ◽  
pp. 1879-1892 ◽  
Author(s):  
Andrea Angelo Pierluigi Tripodi ◽  
Ivan Ranđelović ◽  
Beáta Biri-Kovács ◽  
Bálint Szeder ◽  
Gábor Mező ◽  
...  

AbstractAmong various homing devices, peptides containing the NGR tripeptide sequence represent a promising approach to selectively recognize CD13 receptor isoforms on the surface of tumor cells. They have been successfully used for the delivery of various chemotherapeutic drugs to tumor vessels. Here, we report on the murine plasma stability, in vitro and in vivo antitumor activity of our recently described bioconjugates containing daunorubicin as payload. Furthermore, CD13 expression of KS Kaposi’s Sarcoma cell line and HT-29 human colon carcinoma cell line was investigated. Flow cytometry studies confirm the fast cellular uptake resulting in the rapid delivery of the active metabolite Dau = Aoa-Gly-OH to tumor cells. The increased in vitro antitumor effect might be explained by the faster rearrangement from NGR to isoDGR in case of conjugate 2 (Dau = Aoa-GFLGK(c[NleNGRE]-GG)-NH2) in comparison with conjugate 1 (Dau = Aoa-GFLGK(c[KNGRE]-GG)-NH2). Nevertheless, results indicated that both conjugates showed significant effect on inhibition of proliferation in the primary tumor and also on blood vessel formation making them a potential candidate for targeting angiogenesis processes in tumors where CD13 and integrins are involved.


Blood ◽  
2007 ◽  
Vol 109 (11) ◽  
pp. 4964-4972 ◽  
Author(s):  
Anne-Sophie Moreau ◽  
Xiaoying Jia ◽  
Hai T. Ngo ◽  
Xavier Leleu ◽  
Garrett O'Sullivan ◽  
...  

AbstractWaldenström macroglobulinemia (WM) is an incurable lymphoplasmacytic lymphoma with limited options of therapy. Protein kinase Cβ (PKCβ) regulates cell survival and growth in many B-cell malignancies. In this study, we demonstrate up-regulation of PKCβ protein in WM using protein array techniques and immunohistochemistry. Enzastaurin, a PKCβ inhibitor, blocked PKCβ activity and induced a significant decrease of proliferation at 48 hours in WM cell lines (IC50, 2.5-10 μM). Similar effects were demonstrated in primary CD19+ WM cells, without cytotoxicity on peripheral blood mononuclear cells. In addition, enzastaurin overcame tumor cell growth induced by coculture of WM cells with bone marrow stromal cells. Enzastaurin induced dose-dependent apoptosis at 48 hours mediated via induction of caspase-3, caspase-8, caspase-9, and PARP cleavage. Enzastaurin inhibited Akt phosphorylation and Akt kinase activity, as well as downstream p-MARCKS and ribosomal p-S6. Furthermore, enzastaurin demonstrated additive cytotoxicity in combination with bortezomib, and synergistic cytotoxicity in combination with fludarabine. Finally, in an in vivo xenograft model of human WM, significant inhibition of tumor growth was observed in the enzastaurin-treated mice (P = .028). Our studies therefore show that enzastaurin has significant antitumor activity in WM both in vitro and in vivo, providing the framework for clinical trials to improve patient outcome in WM.


Sign in / Sign up

Export Citation Format

Share Document