Aurora A Kinase Is a Novel Therapeutic Target In The Myeloproliferative Neoplasms

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 109-109 ◽  
Author(s):  
Qiang Jeremy Wen ◽  
Benjamin Goldenson ◽  
Sebastien Malinge ◽  
Brady L Stein ◽  
Terra L Lasho ◽  
...  

Abstract We recently reported that the induction of polyploidization of malignant megakaryocytes shows great promise as a new therapy for acute leukemia. Polyploidization inducers such as dimethylfasudil (diMF) and MLN8237, both of which target Aurora A kinase (AURKA), induce proliferation arrest, polyploidization, expression of megakaryocyte differentiation markers and apoptosis of leukemic megakaryocytes in vitro and in vivo. Since megakaryocytes in primary myelofibrosis (PMF) show impaired polyploidization and maturation, and likely directly contribute to the disease, we predicted that polyploidization inducers would provide a new therapeutic strategy. To determine the effect of these compounds on the growth of MPN cells, we first treated the JAK2 V617F mutant megakaryocytic SET2 cell line with varying doses of MLN8237 and diMF. Both compounds effectively and dose dependently inhibited proliferation, induced polyploidization and upregulation of lineage specific markers CD41 and CD42, and increased apoptosis. Furthermore, MLN8237 synergized with ruxolitinib to induce apoptosis of the SET2 cells and also potently induced growth arrest of JAK2 inhibitor persistent SET2 cells. We observed a similar polyploidization and differentiating activity of MLN8237 and diMF on megakaryocytes derived from primary human PMF progenitors. The ability of these agents to induce polyploidization was specific, as the non-megakaryocyte fractions of the cultures were not affected. Next, we assayed the activity of polyploidization inducers on progression of MPNs in two mouse models: JAK2V617F conditional knockin mice and mice engrafted with MPLW515L expressing bone marrow progenitors. Of note, spleens from both mouse models displayed a robust increase in both total and phosphorylated forms of AURKA relative to control animals, further suggesting that AURKA is a rational target in this disease. We first assayed the activities of MLN8237 and diMF in the MPLW515L bone marrow transplantation model. Recipient mice develop a rapid MPN characterized by leukocytosis, thrombocytosis and bone marrow fibrosis. Both MLN8237 and diMF reduced the disease burden, as evidenced by significant reductions in the liver and spleen weights, white cell counts and platelet counts. Both compounds also led to a significant decrease of fibrosis in the bone marrow, diminished infiltration of megakaryocytes and granulocytes in the liver, and a profound reduction in the numbers of megakaryocytes within the spleen. Moreover, plasma levels of TGF-β a known myelofibrogenic cytokine, were decreased by more than 3-fold by the drug treatment. Both diMF and MLN8237 led to selective polyploidization of megakaryocytes in the spleen as well as marked reductions in the levels of p-AURKA. Of note, neither agent affected the extent of phosphorylation of STAT3 or STAT5. Therefore, we tested whether the combined use of a JAK inhibitor and a polyploidy inducer would show enhanced activity in vivo. Indeed, the combination of MLN8237 and ruxolitinib led to greater reductions in tumor burden in the MPLW515L mouse model than either agent alone. Similar results were obtained using the JAK2V617F knock-in model. To further validate our conclusion that AURKA is a target in PMF, we infected Aurkafl/fl floxed bone marrow progenitors with MPLW515L and transplanted the cells to irradiated recipients. Excision of both alleles of Aurka by Cre mediated recombination completely resolved the disease, while heterozygous deletion of Aurka significantly reduced the disease burden, in a manner similar to treatment with MLN8237. Given that heterozygous deletion of Aurka does not alter normal hematopoiesis in mice, the fact that a 50% reduction in kinase expression was associated with a significant decrease in disease burden suggests that there is an effective therapeutic window in which AURKA inhibitors will be effective against MPN while sparing normal hematopoiesis. Although JAK inhibitors provide symptomatic relief, it is becoming clear that they are not curative. Thus, there is an urgent need to develop new agents to use in combination with JAK inhibitors. Our data reveal that inducing polyploidization and differentiation of dysplastic megakaryocytes in PMF ameliorates features of the disease both in vitro and in vivo. These results support the initiation of clinical studies that combine a JAK inhibitor with an AURKA inhibitor. Disclosures: Crispino: Sanofi: Research Funding.

2010 ◽  
Vol 8 (3) ◽  
pp. 373-384 ◽  
Author(s):  
Jessica J. Huck ◽  
Mengkun Zhang ◽  
Alice McDonald ◽  
Doug Bowman ◽  
Kara M. Hoar ◽  
...  

2010 ◽  
Vol 83 (Suppl_1) ◽  
pp. 344-344
Author(s):  
Patricia Y. Akinfenwa ◽  
Nonna V. Kolomeyevskaya ◽  
Claire M. Mach ◽  
Zhen Li ◽  
Matthew L. Anderson

2003 ◽  
Vol 162 (5) ◽  
pp. 757-764 ◽  
Author(s):  
Yasuhiko Terada ◽  
Yumi Uetake ◽  
Ryoko Kuriyama

A mitosis-specific Aurora-A kinase has been implicated in microtubule organization and spindle assembly in diverse organisms. However, exactly how Aurora-A controls the microtubule nucleation onto centrosomes is unknown. Here, we show that Aurora-A specifically binds to the COOH-terminal domain of a Drosophila centrosomal protein, centrosomin (CNN), which has been shown to be important for assembly of mitotic spindles and spindle poles. Aurora-A and CNN are mutually dependent for localization at spindle poles, which is required for proper targeting of γ-tubulin and other centrosomal components to the centrosome. The NH2-terminal half of CNN interacts with γ-tubulin, and induces cytoplasmic foci that can initiate microtubule nucleation in vivo and in vitro in both Drosophila and mammalian cells. These results suggest that Aurora-A regulates centrosome assembly by controlling the CNN's ability to targeting and/or anchoring γ-tubulin to the centrosome and organizing microtubule-nucleating sites via its interaction with the COOH-terminal sequence of CNN.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2833-2833
Author(s):  
Amanda Przespolewski ◽  
Scott Portwood ◽  
Jason Den Haese ◽  
Demi Lewis ◽  
Eunice S. Wang

Abstract Background: Successful immunotherapeutic approaches for acute myeloid leukemia (AML) have yet to be developed. We hypothesized that targeting both the innate and adaptive immune responses in leukemic hosts would elicit significant anti-tumor activity with lesser toxicities than chemotherapy. To test this, we evaluated the efficacy of immune checkpoint inhibition (murine anti-PD-1 antibody (ab)) alone and in combination with 5,6-dimethylxanthenone-4-acetic acid (DMXAA), an innate immune agonist and anti-vascular agent, in an immunocompetent model of murine AML. Methods: Expression of PD-L1 was assessed by flow cytometry on the murine AML cell line, C1498, alone and following treatment with vehicle, DMXAA or interferon-gamma (positive control). A LEGEND MAX mouse ELISA kit was utilized to measure IL-6 and IFN-β. C57BL/6 mice were inoculated with stably transfected C1498 murine AML cells expressing luciferase and the fluorescent protein DSRed2. Once disease was established, animals were treated with vehicle, DMXAA (20 mg/kg every four days x 7 weeks), anti-murine PD-1 antibody (10 mg/kg every 3 days x 4 doses) or DMXAA + anti-PD-1 antibody (same doses). Animals underwent weekly clinical assessments, weights, and bioluminescent imaging for disease burden. Overall study endpoints were time to morbidity and differences in leukemia disease burden as compared with vehicle-treated controls. Mice were euthanized on day 15 after injection of C1498 cells (8 days following treatment) for collection of plasma, bone marrow, liver and spleen samples for tumor burden, activated T-cells. Results: DMXAA doses (ranging from 1-100 μg/ml) inhibited C1498 in vitro cell growth at 48 hours (48h) in a dose dependent manner. Treatment of C1498 cells in culture with escalating doses of DMXAA (1-100μg/ml) or IFN-gamma (positive control) induced higher PD-L1 expression on these AML cells consistent with direct immunomodulatory effects. Furthermore, C1498 cells exposed to higher doses of DMXAA (10-100μg/ml) for 48h produced measurably higher levels of IL-6 and IFN-β expression in cell supernatants. We then examined the effects of DMXAA, anti-PD-1 ab, or the combination of DMXAA + anti-PD-1 ab treatment in vivo in C57BL/6 mice systemically engrafted with C1498-luciferase AML cells. Treatment overall was well tolerated and resulted in significantly decreased disease burden as measured by total body bioluminescence vs. vehicle controls (p<0.05). Median time to morbidity was significantly decreased in all treatment arms as compared with controls: vehicle = 28 days, DMXAA = 32 days, anti-PD-1 ab = 39 days, and combination DMXAA + anti-PD-1 ab = 53 days (p<0.05). Combination therapy resulted in significantly longer overall survival than single agent therapy (DMXAA vs. DMXAA+anti-PD-1 ab, p=0.032; anti-PD1 ab vs. DMXAA+antii-PD-1 ab p=0.038)(n=total 13-16 mice per group) (representative data shown in Figure 1). Therapy with DMXAA alone and in combination with anti-PD-1 ab was associated with markedly higher PD-1, PD-L1, and PD-L2 expression levels in bone marrow cells harvested from leukemic mice 48h after treatment. Significantly higher numbers of activated T cells were also identified in the bone marrow and spleen of leukemic mice following two weeks of DMXAA therapy alone or in combination with anti-PD-1 ab. Additional in vivo measurements of systemic cytokine levels following therapy are underway. Conclusions: Here we demonstrate that the combination of an innate immune agonist (DMXAA) with an immune checkpoint inhibitor (anti-PD-1 ab) improved anti-leukemic effects in a preclinical AML model. In vitro DMXAA therapy inhibited murine AML growth in a dose dependent manner, enhanced PD-L1 expression, and induced leukemic production of cytokines (IL-6, IFN-β). In vivo combination DMXAA and anti-PD-1 ab therapy in an immunocompetent murine AML model increased activated host T cell numbers and marrow PD-1/L1/L2 expression in conjunction with decreased tumor burden and prolonged overall survival. These studies may pave the way for future clinical trials evaluating this novel immunomodulatory strategy in AML patients. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 5 (16) ◽  
pp. 3134-3146
Author(s):  
Burcu Aslan ◽  
Gorkem Kismali ◽  
Lisa S. Chen ◽  
LaKesla R. Iles ◽  
Mikhila Mahendra ◽  
...  

Abstract Although ibrutinib improves the overall survival of patients with chronic lymphocytic leukemia (CLL), some patients still develop resistance, most commonly through point mutations affecting cysteine residue 481 (C481) in Bruton’s tyrosine kinase (BTKC481S and BTKC481R). To enhance our understanding of the biological impact of these mutations, we established cell lines that overexpress wild-type or mutant BTK in in vitro and in vivo models that mimic ibrutinib-sensitive and -resistant CLL. MEC-1 cell lines stably overexpressing wild-type or mutant BTK were generated. All cell lines coexpressed GFP, were CD19+ and CD23+, and overexpressed BTK. Overexpression of wild-type or mutant BTK resulted in increased signaling, as evidenced by the induction of p-BTK, p-PLCγ2, and p-extracellular signal–related kinase (ERK) levels, the latter further augmented upon IgM stimulation. In all cell lines, cell cycle profiles and levels of BTK expression were similar, but the RNA sequencing and reverse-phase protein array results revealed that the molecular transcript and protein profiles were distinct. To mimic aggressive CLL, we created xenograft mouse models by transplanting the generated cell lines into Rag2−/−γc−/− mice. Spleens, livers, bone marrow, and peripheral blood were collected. All mice developed CLL-like disease with systemic involvement (engraftment efficiency, 100%). We observed splenomegaly, accumulation of leukemic cells in the spleen and liver, and macroscopically evident necrosis. CD19+ cells accumulated in the spleen, bone marrow, and peripheral blood. The overall survival duration was slightly lower in mice expressing mutant BTK. Our cell lines and murine models mimicking ibrutinib-resistant CLL will serve as powerful tools to test reversible BTK inhibitors and novel, non–BTK-targeted therapeutics.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 378-378 ◽  
Author(s):  
Alba Matas-Céspedes ◽  
Anna Vidal Crespo ◽  
Vanina Rodriguez ◽  
Gael Roue ◽  
Armando Lopez-Guillermo ◽  
...  

Abstract Daratumumab (DARA) is a human CD38 antibody with broad-spectrum killing activity. DARA induces killing of tumor cells, mainly via complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC) (de Weers M, J Immunol 2011). DARA is currently being evaluated in phase I/II clinical trials in patients with multiple myeloma. In these clinical studies the adverse events have been manageable and marked reductions in paraprotein and bone marrow plasma cells have been observed. We have previously reported (Blood (ASH annual meeting abstracts). Nov 2012, 120 (21): 3935) that DARA induces cytotoxic activity in vitro via ADCC in primary cells and cell lines from Mantle Cell Lymphoma (MCL), Follicular Lymphoma (FL) and Chronic Lymphoctic Leukemia (CLL). CDC induction was low, which is associated to high expression of the complement inhibitors and reduced number of CD38 molecules per cell in these indications. This suggests a threshold for CD38-targeted CDC lysis. In addition, based on known interactions between CD38-CXCR4, we also demonstrated that in the CLL subtype, with high CD38 and more migratory capacity, DARA (10-30 µg/mL) inhibited in vitro CXCL12/SDF1α-mediated migration up to 70%. Here, we present the first preclinical in vivo results of DARA in mouse models of MCL, transformed FL(tFL) and CLL. We generated heterotopic and systemic mouse models of these entities by subcutaneous or intravenous inoculation of tumor cell lines in SCID mice, that retain both NK cells and macrophages as potential effector cells. In MCL (REC cell line) and tFL (RL cell line) subcutaneous mouse models, we tested DARA activity in both prophylactic (treatment initiation simultaneous to lymphoma cell inoculation) and therapeutic settings(treatment initiation one week after lymphoma cell inoculation, when tumors were about 100 mm3 in size). In the prophylactic setting, mice received 3 doses of DARA or control IgG bi-weekly (20/10/10 mg/kg). In the therapeutic setting, treatment was intensified to 4 doses of DARA or control IgG weekly (20/10/10/10 mg/kg). In both schedules, mice were sacrificed one week after the last dose. DARA completely abrogated tumor growth of REC or RL cells in the prophylactic setting. Moreover, in the therapeutic setting, DARA induced total tumor regression of REC tumors in 4 out of 6 mice, and prevented the splenomegaly observed in control IgG treated mice. In the case of tFL and therapeutic setting, DARA reduced 60% of tumor growth compared to control IgG treated mice at day 32, when experiment was finished. In CLL, we analyzed the effect of DARA on cell homing to lymphoid organs together with its therapeutic properties in a systemic CLL mouse model. Using NOD/SCID/gamma null mice (lacking NK cells and effective macrophages), we analyzed the effect of DARA on primary CLL cell migration from Peripheral Blood (PB) to bone marrow (BM) and Spleen. In this system, NSG mice were pretreated (day 0) with DARA, control IgG or anti-CXCR4 as positive control for inhibition of cell homing, followed by fresh CLL cell inoculation (50×106 cells/per mice) on day 1. PB, BM and spleen cells were isolated on day 2 and CLL cells were identified by staining for CD45/CD19/CD5 and counted using a flow cytometer. Cell counting showed that CLL cells mainly migrate to the spleen, and that DARA significantly reduced this migration (55% inhibition on average, p<0.05). Migration of CLL cells to BM was limited and was not affected by pretreatment of mice with DARA. Finally, we tested DARA therapeutic activity in a systemic CLL mouse model (MEC2 cell line), following the schedule described before (4 doses of control IgG/ DARA weekly (20/10/10/10 mg/kg)), and assessed efficacy on mice overall survival. Mice treated with control IgG started to lose weight and showed signs of disease between days 30-40 and were sacrificed for ethical reasons. In the DARA treated group, in 7 out of 8 mice survival was extended up to day 90, when the experiment was stopped. In conclusion, DARA demonstrated strong in vivo activity in immunocompromised mouse models of MCL, tFL and CLL cell lines and interfered with homing of primary CLL cells to the spleen. These results warrant further investigation of DARA in clinical trials for these indications. Disclosures: Lopez-Guillermo: Roche: Membership on an entity’s Board of Directors or advisory committees. Lammerts van Bueren:Genmab: Employment, Stocks Other. Bakker:Genmab: Employment, Stocks Other. Parren:Genmab: Employment, Stocks Other. Perez-Galan:Genmab: Research Funding.


Blood ◽  
2010 ◽  
Vol 115 (25) ◽  
pp. 5202-5213 ◽  
Author(s):  
Güllü Görgün ◽  
Elisabetta Calabrese ◽  
Teru Hideshima ◽  
Jeffrey Ecsedy ◽  
Giulia Perrone ◽  
...  

Abstract Aurora-A is a mitotic kinase that regulates mitotic spindle formation and segregation. In multiple myeloma (MM), high Aurora-A gene expression has been correlated with centrosome amplification and proliferation; thus, inhibition of Aurora-A in MM may prove to be therapeutically beneficial. Here we assess the in vitro and in vivo anti-MM activity of MLN8237, a small-molecule Aurora-A kinase inhibitor. Treatment of cultured MM cells with MLN8237 results in mitotic spindle abnormalities, mitotic accumulation, as well as inhibition of cell proliferation through apoptosis and senescence. In addition, MLN8237 up-regulates p53 and tumor suppressor genes p21 and p27. Combining MLN8237 with dexamethasone, doxorubicin, or bortezomib induces synergistic/additive anti-MM activity in vitro. In vivo anti-MM activity of MLN8237 was confirmed using a xenograft-murine model of human-MM. Tumor burden was significantly reduced (P = .007) and overall survival was significantly increased (P < .005) in animals treated with 30 mg/kg MLN8237 for 21 days. Induction of apoptosis and cell death by MLN8237 were confirmed in tumor cells excised from treated animals by TdT-mediated dUTP nick end labeling assay. MLN8237 is currently in phase 1 and phase 2 clinical trials in patients with advanced malignancies, and our preclinical results suggest that MLN8237 may be a promising novel targeted therapy in MM.


Author(s):  
Mariya Stavnichuk ◽  
Svetlana V. Komarova

Over the years, numerous studies demonstrated reciprocal communications between processes of bone marrow hematopoiesis and bone remodeling. Megakaryocytes, rare bone marrow cells responsible for platelet production, were demonstrated to be involved in bone homeostasis. Myelofibrosis, characterized by an increase in pleomorphic megakaryocytes in the bone marrow, commonly leads to the development of osteosclerosis. In vivo, an increase in megakaryocyte number was shown to result in osteosclerosis in GATA-1low, NF-E2-/-, TPOhigh, Mpllf/f;PF4cre, Lnk-/-, Mpig6b-/-, Mpig6bfl/fl;Gp1ba-Cr+/KI, Pt-vWD mouse models. In vitro, megakaryocytes stimulate osteoblast proliferation and have variable effects on osteoclast proliferation and activity through soluble factors and direct cell-cell communications. Intriguingly, new studies revealed that the ability of megakaryocytes to communicate with bone cells is affected by the age and sex of animals. This mini-review summarises changes seen in bone architecture and bone cell function in mouse models with an elevated number of megakaryocytes and the effects megakaryocytes have on osteoblasts and osteoclasts in vitro, and discusses potential molecular players that can mediate these effects.


2002 ◽  
Vol 22 (3) ◽  
pp. 874-885 ◽  
Author(s):  
Claudia Crosio ◽  
Gian Maria Fimia ◽  
Romain Loury ◽  
Masashi Kimura ◽  
Yukio Okano ◽  
...  

ABSTRACT Phosphorylation at a highly conserved serine residue (Ser-10) in the histone H3 tail is considered to be a crucial event for the onset of mitosis. This modification appears early in the G2 phase within pericentromeric heterochromatin and spreads in an ordered fashion coincident with mitotic chromosome condensation. Mutation of Ser-10 is essential in Tetrahymena, since it results in abnormal chromosome segregation and extensive chromosome loss during mitosis and meiosis, establishing a strong link between signaling and chromosome dynamics. Although mitotic H3 phosphorylation has been long recognized, the transduction routes and the identity of the protein kinases involved have been elusive. Here we show that the expression of Aurora-A and Aurora-B, two kinases of the Aurora/AIK family, is tightly coordinated with H3 phosphorylation during the G2/M transition. During the G2 phase, the Aurora-A kinase is coexpressed while the Aurora-B kinase colocalizes with phosphorylated histone H3. At prophase and metaphase, Aurora-A is highly localized in the centrosomic region and in the spindle poles while Aurora-B is present in the centromeric region concurrent with H3 phosphorylation, to then translocate by cytokinesis to the midbody region. Both Aurora-A and Aurora-B proteins physically interact with the H3 tail and efficiently phosphorylate Ser10 both in vitro and in vivo, even if Aurora-A appears to be a better H3 kinase than Aurora-B. Since Aurora-A and Aurora-B are known to be overexpressed in a variety of human cancers, our findings provide an attractive link between cell transformation, chromatin modifications and a specific kinase system.


1999 ◽  
Vol 43 (5) ◽  
pp. 1285-1288 ◽  
Author(s):  
Jason V. Brooks ◽  
Synthia K. Furney ◽  
Ian M. Orme

ABSTRACT The capacity of metronidazole to inhibit the growth ofMycobacterium tuberculosis was tested in in vitro and in vivo mouse models. In vitro addition of metronidazole to cultures of infected bone marrow-derived macrophages had no effect, nor did it increase the reduction in bacterial load due to isoniazid. In vivo, metronidazole did not reduce bacterial numbers in the lungs of aerosol-infected mice during the active stage of the disease, during a phase of containment, or after prolonged isoniazid therapy (Cornell model). A small but significant reduction was seen if metronidazole therapy was given during an established chronic disease state 100 days after aerosol administration. These data indicate that under most conditions M. tuberculosis organisms are not in a metabolic state in which they are susceptible to the action of metronidazole and, hence, that this drug would be of limited clinical value.


Sign in / Sign up

Export Citation Format

Share Document