scholarly journals FGF2-FGFR1 signaling regulates release of Leukemia-Protective exosomes from bone marrow stromal cells

eLife ◽  
2019 ◽  
Vol 8 ◽  
Author(s):  
Nathalie Javidi-Sharifi ◽  
Jacqueline Martinez ◽  
Isabel English ◽  
Sunil K Joshi ◽  
Renata Scopim-Ribeiro ◽  
...  

Protective signaling from the leukemia microenvironment leads to leukemia cell persistence, development of resistance, and disease relapse. Here, we demonstrate that fibroblast growth factor 2 (FGF2) from bone marrow stromal cells is secreted in exosomes, which are subsequently endocytosed by leukemia cells, and protect leukemia cells from tyrosine kinase inhibitors (TKIs). Expression of FGF2 and its receptor, FGFR1, are both increased in a subset of stromal cell lines and primary AML stroma; and increased FGF2/FGFR1 signaling is associated with increased exosome secretion. FGFR inhibition (or gene silencing) interrupts stromal autocrine growth and significantly decreases secretion of FGF2-containing exosomes, resulting in less stromal protection of leukemia cells. Likewise, Fgf2 -/- mice transplanted with retroviral BCR-ABL leukemia survive significantly longer than their +/+ counterparts when treated with TKI. Thus, inhibition of FGFR can modulate stromal function, reduce exosome secretion, and may be a therapeutic option to overcome resistance to TKIs.Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (<xref ref-type="decision-letter" rid="SA1">see decision letter</xref>).

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 5407-5407
Author(s):  
Gaya Narendran ◽  
Matthew F. Clarkson ◽  
Randal Johnston

Abstract Introduction Acute lymphoblastic leukemia (ALL) is the most common malignant disease in children, constituting more than a quarter of all childhood cancers. The survival rate of standard/good risk ALL now exceeds 80%. In spite of the significant improvements in outcome, the emergence of resistant disease remains the most common cause of death. Escalation of the intensity of combination chemotherapy with the introduction of second-line drugs is accompanied by cumulative toxicity, with marginal incremental benefits. Hence novel therapeutic approaches are urgently needed to improve the outcome in these patients. In addition to the inherent molecular alterations, recent reports have suggested an active role for the bone marrow stromal microenvironment in promoting both leukemia cell viability and drug resistance. Previous studies have shown a pivotal role for the stromal chemokine CXCL12 (SDF-1) and its receptor CXCR4 in this process. In the multiple myeloma model, inhibition of CXCR4 by the targeted agent AMD3100 (Plerixafor) abrogates the chemotherapy protection conferred by stromal cells. Recent years have seen evidence for the effectiveness of oncolytic viruses as cytotoxic agents against refractory tumors. The strategy behind this approach is to develop viruses that can replicate and cause cell lysis specifically in cancer cells while leaving non-malignant cells unaffected. In this study we explore the utility of reovirus mediated anti-leukemic therapy in an experimental model of pediatric leukemia in the context of co-culture with bone marrow stromal cells. Methods A panel of leukemia cell lines (n=4) representing pediatric ALL and acute myeloid leukemia (AML) were evaluated when co-cultured in the presence of stromal conditioned medium, live bone marrow stromal cells, stromal cells fixed with 0.5% glutaraldehyde, or control human skin fibroblasts (1Br3 cells). After three days in culture, cell proliferation kinetics under each condition was measured by WST-1 assay. The influence of nonspecific leukemia cell attachment was evaluated by the culture of leukemia cells in poly-L-lysine coated plates. Next the leukemic cells were treated with oncolytic reovirus, at various concentrations with and without co-cultures conditions to evaluate the potential of leukemia cells to escape reovirus mediated cytolysis. Finally, the ability of stromal cells to modulate reovirus anti-leukemic activity was evaluated in the presence of various concentrations of AMD3100. Results and Discussion An average of a 2-fold increase in leukemia cell proliferation (in the absence of reovirus) was seen with live stromal co-cultures compared to conditioned media or fixed stromal cells, indicating the requirement of live cell-cell contact with stromal cells in this process. Poly-L lysine coated plates did not increase leukemia cell growth. Our data reveals that, as previously seen with chemotherapeutic drug exposure, the presence of stromal cells is able to decrease the cytolytic activity of reovirus on leukemic cells. Leukemia cells alone exhibit a survival of 10% following a three-day exposure to reovirus (50 MOI). However, in the presence of stromal the survival rate was increased to 40% (p < 0.05). Furthermore, the addition of AMD3100 provided partial restoration of the oncolytic activity (mean 15% survival, n=3 experiments). Stromal conditioned media also decreased reovirus infectivity, although to a lower extent (mean 17% cell survival). Co-culture of leukemia cells with control 1br3 human skin fibroblast cells provided no protection from reoviral cytolysis (mean 10% survival). In this proof-of-concept study, we provide evidence for the first time that the bone marrow niche may provide at least partial protection from oncolytic virotherapy. This indicates the importance of evaluating the efficacy of oncolytic viruses in the context of the tumor microenvironment in future studies. Furthermore, we provide evidence for the consideration of combination therapies with targeted agents such as AMD3100, which may disrupt the stromal protective activities, to increase the effectiveness virotherapy in the treatment of refractory leukemia. Disclosures: No relevant conflicts of interest to declare.


2019 ◽  
Author(s):  
Sana Usmani ◽  
Urmila Sivagnanalingam ◽  
Olena Tkachenko ◽  
Leti Nunez ◽  
Jessica Shand ◽  
...  

PLoS ONE ◽  
2012 ◽  
Vol 7 (5) ◽  
pp. e37203 ◽  
Author(s):  
Patricia Macanas-Pirard ◽  
Andrea Leisewitz ◽  
Richard Broekhuizen ◽  
Kelly Cautivo ◽  
Francisco M. Barriga ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3078-3078
Author(s):  
Shamim Lotfi ◽  
Aarthi Jayanthan ◽  
Victor A. Lewis ◽  
Greg Guilcher ◽  
Matthew S Squires ◽  
...  

Abstract Abstract 3078 Poster Board III-15 Leukemia in children less than 1 year of age confers a poor prognosis, despite intensification of therapy. These leukemias possess unique biologic characteristics including the presence of mixed-lineage leukemia (MLL) gene rearrangement and high expression of Fms-like tyrosine kinase 3 (FLT3). AT9283, a potent inhibitor of Aurora A and B kinases, JAK2, JAK3, and mutant Abl Kinase, has demonstrated inhibition of multiple solid tumor cell lines in vitro and in mouse xenograft models. Aurora kinase inhibition has been shown to inhibit cancer cell growth by interfering with the mitotic apparatus. We investigated the activity of AT9283 against cell lines derived from refractory infant leukemia cells to identify its efficacy in a future treatment protocol. Method Five cell lines derived from infant leukemia cells were used (ALL: BEL1, KOPN8, KCCF2, B1 and AML: TIB202). We also included the cell line SEM that was derived from a 5 year old child with t (4;11) MLL-AF4 preB-ALL. Normal bone marrow stromal cells were used to evaluate cytotoxicity against non-malignant cells. AT9283 was provided by Astex Therapeutics Ltd. (Cambridge, UK). Approximately 1×104 cells per well were seeded in 96-well plates and incubated with increasing concentrations of AT9283, alone or in combination with a panel of conventional and novel therapeutic agents. After four days, cell survival was measured by Alamar blue assay and IC50 values and combination indices were calculated. Stem-like cells were quantified by the distribution of ALDH bright cells by Aldefluor assay (Stem cell technologies) and characterized by conventional clonogenic assays. Alterations in cell-signaling pathways and survival proteins were measured by Western blot analysis using total and phospho-specific antibodies. Results AT9283 inhibited the growth of all five cell lines with a 10 fold variation in IC50 within cell lines (IC50 range, 0.1 to 0.01 μM). There was a corresponding increase in the number of cells displaying a polyploid phenotype, an effect of aurora kinase inhibition. No significant cytotoxicity against bone marrow stromal cells was seen under the experimental conditions used in this study (IC50 > 10 μM). Changes in the activation and expression of a variety of intracellular proteins were noted, including the down regulation of activated ERK1/2, MYC and AKT within 10 minutes of exposure to the agent. An increase in the activated form of RAF and ATF2 was observed immediately after drug exposure. Importantly, a significant decrease in the level of constitutive pFLT-3 was demonstrated. A concurrent increase in cleaved PARP was also noted, indicating the initiation of apoptosis. In combination studies, the HDAC inhibitor Apicidin showed synergy across all cell lines (CI range: 0.07 to 0.62). A decrease in ALDH bright stem-like cells was observed in a dose dependent manner, up to 50% over 24 hours at IC50 concentrations. Conclusions Our in vitro studies show that AT9283 significantly decreases the growth and survival of infant leukemia cell lines. Importantly, AT9283 potently induces FLT3 de-phosphorylation, inhibiting a critical growth stimulatory pathway of infant ALL cells. We have identified changes in a number of signaling and apoptotic molecules that can provide a panel of markers for biological correlative analysis for drug activity in vivo. Also, the drug combination studies demonstrate the potential of HDAC inhibition to synergize with the activity of this agent. Finally, the effect on stem-like cells provides a rationale and critical preclinical data for the formulation of an effective clinical trial for the treatment of infants with refractory ALL. Disclosures Squires: AstexTherapeutics Ltd: Employment.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1027-1027
Author(s):  
Zhongbo Hu ◽  
Xiaomiao Li ◽  
David Ostrov ◽  
William Slayton

Abstract Abstract 1027 Integrin VLA-5 (α5β1, CD49e/CD29) plays an important role in hematopoietic cells functioning as well as in promoting tumor angiogenesis and tumor metastasis. Molecules targeting VLA-5 can be rapidly developed into anti-inflammatory and anti-tumor pharmaceuticals. VLA-5 is highly expressed on Ph+ leukemia cells and VLA-5 inhibitory antibodies can significantly inhibit the adhesion of Ph+ leukemia cells to human fibronectin. We generated an atomic homology model of VLA-5 based on the crystal structure of the extracellular segment of integrin αVβ3 in complex with a cyclic peptide presenting the Arg-Gly-Asp sequence and utilized this structure-based approach to identify VLA-5 binding drug-like small molecules. We selected the Arg-Gly-Asp binding residues and the epitopes of VLA-5 antibody as the target for small molecule binding using SPHERE_SELECT in DOCK6. The grid-based scoring system was used for scoring with the non-bonded force field energy function. The 100 highest scoring small molecules were assayed in an in vitro adhesion assay using leukemia cell lines and solid phase assay. This approach identified several leading small-molecule compounds, V10, V20, V37 and L4. Their IC50 are respectively 22.5μM, 23.7μM, 32.0μM and 28.9μM. These compounds can inhibit the adhesion of VLA-5 expressing Philadelphia chromosome positive leukemia to both human fibronectin and bone marrow stromal cells. Compounds V10 and V20 also significantly inhibited the growth of Ph+ leukemia cells. These compounds can enhance the effect of imatinib and dasatinib to kill Ph+ leukemia cells when cultured contacting with bone marrow stromal cells. We are currently testing the synergistic effect of these compounds with tyrosine kinase inhibitors to treat the Ph+ acute lymphoblastic leukemia in NOD/SCID animal model. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 40 (11) ◽  
Author(s):  
Lanxia Zhou ◽  
Hong Guo ◽  
Fang Jia ◽  
Xuan Chen ◽  
Xiaowei Zhang ◽  
...  

Abstract The core of the tumor microenvironment in the hematological system is formed by bone marrow stromal cells (BMSCs). In the present study, we explored the interaction between the urokinase plasminogen activator (uPA) system and the leukemia bone marrow microenvironment (BMM). We established BMSCs–HL60 and HS-5–K562 co-culture models in direct contact mode to simulate the BMM in leukemia. In BMSCs-HL60 co-culture model, the expression levels of uPA, uPA receptor (uPAR), plasminogen activator inhibitor 1 (PAI-1) and vascular endothelial growth factor (VEGF) in BMSCs were higher than those in mono-cultured BMSCs. Matrix metalloproteinase (MMP)-9 (MMP-9) was up-regulated in co-cultured HL60 cells. In HS-5–K562 co-culture model, only uPA, PAI-1, and VEGF-A were up-regulated in HS-5 cells. The levels of the uPA protein in the co-culture supernatant were significantly higher than that of mono-cultured BMSCs or HS-5 cells. Our findings demonstrate that the co-culture stimulates the production of uPA, uPAR, PAI-1, MMP-9, and VEGF-A by BMSCs. It could further explain how the uPA system in leukemia cells is involved in the growth, development, and prognosis of leukemia.


Sign in / Sign up

Export Citation Format

Share Document