TGFβ1 Induces MLL-Rearranged Acute Lymphoblastic Leukemia into Chemoresistant Quiescence in a Coordinated Manner with FLT3 Ligand

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3972-3972
Author(s):  
Minori Tamai ◽  
Yoshiyuki Furuichi ◽  
Shin Kasai ◽  
Norie Ando ◽  
Kumiko Goi ◽  
...  

Abstract Background Acute lymphoblastic leukemias with rearrangements of mixed-lineage leukemia gene (MLL+ALL) frequently develop in infants and have a dismal prognosis. The refractory nature of this leukemia is known to be associated with persistence of high levels of minimal residual disease (MRD). We previously reported that FLT3 ligand (FL) stimulation of MLL+ALL cells, which express FLT3 at high levels, induced quiescence resistant to anti-leukemic agents, to the process of which up-regulation of p27, one of cyclin-dependent kinase inhibitors (CDKIs), was closely attributable. Because a vast amount of FL is secreted from bone marrow stromal cells, it was postulated that this FLT3/FL interaction should contribute, at least in part, to the high levels of MRD in MLL+ALL (Furuichi et. al. Cancer Res 2007). Of interest, in B-cell precursor ALLs other than MLL+ALL, FL stimulated their proliferation. Therefore, we performed the microarray analysis using the MLL+ALL cell line and the ETV6-RUNX1-positive cell line for comparing changes in the gene expression profiles at 24h after FL stimulation, and picked up the transforming growth factor β1 (TGFβ1) as a gene whose mRNA level was conversely regulated between two cell lines. TGFβ1 is a growth inhibitory cytokine for some cancer cells and known to have a potential to induce hematopoietic stem cell quiescence. In the present study, we examined the effects of TGFβ1 and/or FL on MLL+ALL cells particularly in terms of induction of quiescence and chemosensitivity. Materials and Methods The MLL+ALL cell line KOCL58 (MLL-AF4+) was used throughout the study to examine TGFβ1 mRNA, cell cycle progression, induction of apoptosis, and changes in chemosensitivity in the presence of TGFβ1 and/or FL. Results 1. We first examined changes in TGFβ1 mRNA after FL (20ng/ml) stimulation by real time RT-PCR in KOCL58 cells, and confirmed that TGFβ1 mRNA expression level was significantly increased (1.5 fold at 24h). Of note, when the cells were stimulated with FL and TGFβ1 (10ng/ml), TGFβ1 mRNA expression level was further increased (3.5 fold) between 7 and 24h. 2. We next evaluated the effect of TGFβ1 on proliferation. When KOCL58 cells were incubated with a low concentration of TGFβ1 (0.33ng/ml) in the presence or absence of FL (20ng/m), flow cytometric analysis using the BrdU/PI double staining showed that the G0/G1 population (35.2%; before stimulation) was increased in the presence of TGFβ1 (48.4%) or FL (53.0%), and further in the presence of TGFβ1+FL (75.1%). We also examined expression levels of cell cycle regulating molecules, and found that p27 was significantly increased in the presence of FL and further in the presence of TGFβ1+FL. This p27 up-regulation was due to post-transcriptional mechanism, because p27 mRNA remained unchanged but the half-life of p27 protein was elongated in the presence of FL and/or TGFβ1. 3. To elucidate whether TGFβ1 affects the sensitivity of MLL+ALL cells to a cell cycle dependent anti-leukemic agent AraC, we pre-incubated KOCL58 cells in the presence of TGFβ1 and/or FL for 72h and then exposed to AraC (500nM) for 24h. Flow cytometric analysis using FITC-conjugated Annexin V demonstrated that the Annexin V-negative viable population (%) was decreased by AraC exposure (25.6±1.4%), but it was significantly restored by pre-incubation with FL (39.4±1.7%, P<0.0005) and further by pre-incubation with TGFβ1+FL (49.3±5.9%, P<0.003), although not significantly different by pre-incubation with TGFβ1 alone. Flow cytometric analysis using anti-active caspase 3 antibody also demonstrated that the positive population was increased to 50.8% by AraC exposure, but it was decreased by pre-incubation with either TGFβ1 (27.5%) or FL (24.8%), and further by pre-incubation with TGFβ1+FL (10.2%) (Figure). 4.We performed chmosensitivity experiments to AraC (800nM) using KOCL58 cells adhering to bone marrow stromal cells, which secrete FL and TGFβ1, in the presence of FLT3 inhibitor (CEP701, 5nM) and/or TGFβ-receptor 1 inhibitor (HTS, 5mM), and found that KOCL58 cells adhering to stromal cells showed a marked resistance to AraC (60% increase in Annexin V-negative population), but this restoring effect was partially (~20%) alleviated by each of inhibitors. [Discussion] TGFβ1 should be involved in the development of MRD in MLL+ALL in synergy with FL, and a combined use of inhibitors against FLT3 and TGFβ1 might be effective for eradicating MRD in MLL+ALL patients. Figure Figure. Disclosures No relevant conflicts of interest to declare.

Neuroreport ◽  
2005 ◽  
Vol 16 (6) ◽  
pp. 581-584 ◽  
Author(s):  
Min Ye ◽  
Shengdi Chen ◽  
Xijin Wang ◽  
Chen Qi ◽  
Guoqiang Lu ◽  
...  

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2494-2494
Author(s):  
Lauren C. Wallis ◽  
Matthew J. Streetly ◽  
Rebecca Auer ◽  
John Gribben ◽  
Dean Zhang ◽  
...  

Abstract Conventional techniques for assessing drug response and apoptosis induction rely on static assessment of cellular changes at predetermined time points (e.g. detection of exposed membrane phospholipids by Annexin V). The Kinetics of Optical Response assay (KOR) is a new technique that detects induction of apoptosis dynamically. It employs a spectrophotometric methodology to detect changes in optical density associated with membrane blebbing related to growth and death, allowing detection of apoptosis in real time. The KOR assay has already predicted the response to cytotoxic agents of AML cell lines and primary samples. This study uses the KOR assay in lymphoid malignancy and shows sensitivity to apoptosis induction by conventional and novel agents including bortezomib. The lymphoma cell line DOHH2 (t(14;18)), U266 (myeloma), K562 (CML) and primary CLL cells were used in this study with HL60 (AML) as a control. Cells were seeded in 96 well plates and treated with a variety of drugs alone or in combination (cytarabine, fludarabine, doxorubicin, daunorubicin, etoposide, melphalan, bortezomib) at multiple concentrations. Measurements were made at 5 min. intervals for up to 48 hrs and analysed using KORSoft™ software to generate apoptotic response curves. To validate this approach conventional techniques were used for comparison (Alamar Blue for cytotoxicity and flow cytometric analysis of cell cycle and apoptosis using propidium iodide and Annexin V staining respectively). The KOR assay can show changes in growth characteristics, induction of apoptosis and necrosis in response to drugs permitting a continuous analysis for maximum sensitivity (Smax). DOHH2 was found to be dose responsive to four of the drugs used, with the Smax for 10μM daunorubicin at 6 hours (48%), 1μM doxorubicin at 8 hours (38%), 100μM etoposide at 8 hours (52%), and minimally to 100μM cytarabine at 16 hours (21%). There was no effect from fludarabine. The addition of bortezomib increased Smax to 89% with etoposide and to a lesser degree with the other cytotoxic drugs. U266 showed a similar spectrum of results with greatest Smax with 100μM melphalan at 9 hours (57%) enhanced to 78% with the addition of bortezomib. There was minimal response to cytarabine and fludarabine. Parallel flow cytometric analysis using Annexin V and PI showed similar results to those from the KOR assay confirming the assessment of apoptosis to be valid. Cell cycle analysis showed an increased sub-G1 peak in keeping with apoptosis at times of Smax assessed by the KOR assay. The Alamar Blue cytotoxicity assay showed a dose dependent decrease in cell proliferation in response to increasing drug dose again paralleling other apoptosis measurements implying an apoptotic effect due to drug action and correlate well with those from the KOR assay. Primary CLL samples following CD19 selection were cultured with and without IL4 and exposed to the KOR assay with cytotoxics and bortezomib. Culture with IL4 alone gave good growth characteristics and revealed the combination of etoposide and bortezomib to provide the best induction of apoptosis (Smax 82%) compared to etoposide (26%) or bortezomib (32%) alone. The KOR assay is a microtitre approach to the assessment in real time of apoptosis. This study suggests the combination of bortezomib and etoposide is effective for lymphoma. Such approaches can accelerate the development of effective clinical trials.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1519-1519 ◽  
Author(s):  
Douglas W. McMillin ◽  
Jake Delmore ◽  
Joseph M. Negri ◽  
Patrick Hayden ◽  
Nicholas Mitsiades ◽  
...  

Abstract Context: Conceptually, targeting proteins involved in cell cycle regulation is an appealing therapeutic strategy for multiple myeloma (MM), given the increased proliferative rate of biologically aggressive multi-drug resistant MM cells compared to not only their normal plasma cell counterparts, but also MM cells from earlier stages of the disease. Within the intricate network of signaling cascades and regulatory checkpoints involved in cell cycle progression and survival of neoplastic cells, cyclin-dependent kinases (CDKs) have emerged as intriguing therapeutic targets. We report the results of preclinical studies on the anti-MM activity of the selective CDK1/2 small molecule inhibitor NVP-LCQ195/AT9311 (Novartis Pharma/Astex Therapeutics). Methods/Results: In MTT colorimetric survival assays against a panel of 28 human MM cell lines, NVP-LCQ195 exhibited dose-dependent anti-MM activity, with IC50 values at or below ~0.5 μM (a pharmacologically relevant concentration) for 12 of 28 cell lines many of which are resistant to conventional or novel anti-MM therapies (e.g. dexamethasone, melphalan, thalidomide derivatives). NVP-LCQ195 was also active against primary MM cells isolated from heavily-pretreated/drug-resistant cases of MM. In marked contrast, the IC50 values of NVP-LCQ195 against non-malignant cells such as bone marrow stromal cells (BMSCs) and immortalized hepatocyte cells was &gt;4 μM. In addition, both PHA-stimulated and unstimulated normal donor PBMCs were less sensitive than the majority of MM cell lines of our panel. Treatment with NVP-LCQ195 was able to overcome the protective effects conferred on MM cells by exogenous IL-6 (10ng/mL) or IGF-1 (50ng/mL), as well as by co-culture of MM cells with bone marrow stromal cells (BMSCs). NVP-LCQ195 was observed to trigger a distinct pattern of S-phase arrest, followed by eventual induction of apoptotic cell death. Mechanistic studies revealed that hsp90, B-raf, cyclin D1, and cyclin E2 levels decrease in response to NVP-LCQ195 treatment, while caspases- 3, -8 and PARP are cleaved within 16 and 24 hrs of drug treatment. We evaluated a series of combinations of this agent with conventional (e.g. dexamethasone, doxorubicin) and novel (e.g. bortezomib) anti-MM agents. No evidence of antagonism with any of these anti-MM agents was observed, indicating that combinations of NVP-LCQ195 with the aforementioned anti-MM agents can be feasible in clinical settings. Conclusion: Functional inhibition of proteins involved in cell cycle regulation remains an attractive approach for the treatment of MM. Treatment of MM cells with the CDK1/2 inhibitor NVP-LCQ195 induces, in a subset of MM cell lines, a distinct pattern of initial cell cycle arrest, anti-proliferative/pro-apoptotic molecular sequleae and eventual induction of cell death. Our results determine intriguing potential combination regimens for MM treatment. Further in vitro and in vivo studies are attempting to delineate molecular markers of MM cell responsiveness vs. resistance to NVP-LCQ195 and provide a framework for individualized treatment of select MM patients with this interesting class of compounds.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 20105-20105
Author(s):  
Y. Chen ◽  
X. Yu

20105 Background: To observe the effect and mechanism of gemcitabine-induced radiosensitization on CNE-1 nasopharyngeal carcinoma cell line. Methods: The cell survival curves were obtained with clonogenic assay. The cell cycle phases were measured with flow cytometric analysis. Results: No cytotoxic effect was observed in 0.1 μM and 2 μM gemcitabine alone group. The radiosensitivities were observed in the groups treated with radiation and gemcitabine exposed in different consistencies and times. The SER were 1.35,1.82; 1.55 and 2.16 respectively. The G1 cell cycle phase block was observed in the two consistency groups after exposing to gemcitabine for 24 hours. Dq were rather lower in 2 μM groups especially that exposed for 24 hours. Conclusions: The radiosensitivity was obviously observed in the groups treated with two consistencies of gemcitabine. The effect was more obviously as the consistency and the exposing time increased. The block of cell cycle from G1 to S and the decrease of sublethal damage repair might be contributed to the mechanism. No significant financial relationships to disclose.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 683-683
Author(s):  
Maria Rita Pitari ◽  
Marco Rossi ◽  
Cirino Botta ◽  
Eugenio Morelli ◽  
Annamaria Gullà ◽  
...  

Abstract Multiple Myeloma (MM)-related osteolytic lesions of the skeleton result from asynchronous bone turnover wherein increased bone resorption by osteoclasts (OCLs) is associated to suppression of bone formation due to inhibition of osteoblasts (OBLs). In this context, the balance between osteoprotegerin (OPG) and the receptor activator for nuclear factor-κB ligand (RANKL) plays a relevant role, because RANKL signaling induces differentiation, activation and survival of OCLs, whereas OPG acts as a decoy receptor of RANKL, thus blocking bone resorption. In the MM milieu, the interaction of malignant plasma cells (PCs) and bone marrow stromal cells (BMSCs) favors the production of RANKL and reduces OPG secretion by BMSCs. The resulting severe imbalance in RANKL/OPG ratio is the main trigger of MM-related bone disease (BD). Emerging evidence indicates that microRNAs (miRNAs) play a crucial role in bone remodelling as they can act on bone effectors (OCLs, OBLs, BMSCs) and PCs. Indeed, several findings suggest that miR-29 family support OBL differentiation, while we have previously demonstrated that miR-29b is down-regulated during osteoclastogenesis and its enforced expression functionally inhibits terminally differentiated OCLs. Moreover, also miR-21 plays an important role in OCL differentiation and is up-regulated by IL-6 via STAT3 that, in turn, promotes survival and proliferation of PCs, favoring bone damage. On these bases, we investigated whether modulation of miR-21 expression in BMSCs may have an impact on MM microenviroment, focusing our attention on OPG production, taking into account that different bioinformatics resources indicate OPG mRNA as a predicted target of miR-21. To this end, we established a co-culture system where primary BMSCs and HS-5, a human BMSC cell line, were exposed to either RPMI 8226 MM cells or primary CD138+ PCs isolated from MM patients. Then, we isolated BMSCs and evaluated miR-21 and OPG expression. We observed strong up-regulation of miR-21 in BMSCs and HS-5 together with a significant reduction of OPG levels in the presence of MM cells. These preliminary data suggested that antagonizing miR-21 in BMSCs exposed to MM cells could restore RANKL/OPG ratio through the up-regulation of OPG. To address this hypothesis, we transduced HS-5 cell line with a lentiviral vector carrying anti-miR-21 sequence (a21 HS-5) or with an empty lentiviral vector carrying only the GFP gene (GFP CNT HS-5). OPG expression was determined by RT-PCR, Western Blotting and ELISA assays after 48h and 72h of exposure to RPMI 8226 cells or primary PCs. We observed a significant increase in OPG production in a21 HS-5 cells compared to controls (Fig. 1, panel A and panel B). We also evaluated RANKL expression in the same samples. Interestingly, constitutive inhibition of miR-21 in HS-5 cell line upon exposure to MM cells induced a significant RANKL down-regulation, thus restoring a physiological RANKL/OPG ratio. We hypothesized that this surprising result may be due to the activity of the protein inhibitor of activated STAT3 (PIAS3). Indeed, PIAS3 binds to STAT3 and blocks its DNA binding ability, thereby inhibiting STAT3-mediated gene activation. It is already known that PIAS3 acts as a negative regulator of the RANKL expression and it is a direct and validated target of miR-21. Therefore, the inhibition of miR-21 induces PIAS3 up-regulation and a decreased RANKL transcription. To confirm the involvement of PIAS3 in RANKL suppression in our system, we performed a WB assay, showing that in a21 HS-5 cells, PIAS3 is indeed over-expressed compared to controls. Taken together, these findings provide the first evidence that inhibition of miR-21 in MM microenviroment restores RANKL/OPG balance, which might result in prevention of skeletal- related events. These findings support the design of innovative miR-21 inhibition-based approaches against MM-related BD. Supported by the Italian Association for Cancer Research (AIRC) “Special Program Molecular Clinical Oncology - 5 per mille”, PI:P.T., n. 9980, 2010/15.”Fig.1Fig.1. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5216-5216
Author(s):  
Laura M Desbourdes ◽  
Adam J Guess ◽  
Suheyla Hasgur ◽  
Kathleen M Overholt ◽  
Minjun Yu ◽  
...  

Abstract Introduction The 5-year survival for patients with acute myeloid leukemia (AML) has stagnated for over two decades at about 60% for children, 40% for young adults, and <15% for elderly patients. While most patients achieve remission, approximately 50% will relapse which is generally attributed to the persistence of leukemic stem cells. Interferon α (IFNα) is an effective therapy for patients with AML due to multiple mechanisms of action. However, high serum levels are associated with many adverse effects. In this proof-of-concept study, we used engineered mesenchymal stem/stromal cells (MSC) to deliver high concentrations of IFNα locally to an AML chloroma, potentially diminishing the poorly tolerated systemic side-effects. Methods Bone marrow MSCs from C57BL/6 mouse were isolated and transduced with a lentiviral vector expressing murine IFNα (IFNα-MSCs) and/or GFP (GFP MSCs). After measuring IFNα secretion by ELISA and confirming activity by the induction of the MHC I expression on the transduced cells, the anti-AML activity of these transduced MSCs was assessed by co-culture with the C57BL/6 AML cell line c1498 which expresses DsRed and firefly luciferase (FFluc). Apoptotic cell frequencies and cell cycle phase distributions of leukemia cells with or without MSCs were assessed by flow cytometry. The in vivo validation has been performed by subcutaneous injection of c1498 cells (chloroma) with or without GFP MSCs or IFNα MSCs in C57BL/6 mice. Tumor growth was monitored by bioluminescence imaging every 3 or 4 days. Results Flow cytometric analysis and ELISA confirmed the secretion of bio-active of IFNα by transduced MSCs (41.5 ng/1E06 MSCs/24h). In co-cultures, the presence of IFNα MSCs at the ratio 100:1 (c1498: MSC) significantly decreased the population of c1498 cells mainly by inducing apoptosis compared to MSC-free or GFP MSC co-cultures while no effect was observed on cell cycle distribution. The pro-apoptotic effect of IFNα MSCs was then investigated in vivo by subcutaneous injection of c1498 cells with or without MSCs (ratio 10:1) in C57BL/6 mice.The presence of IFNα MSCs significantly decreased leukemic cell mass as shown by the bioluminescence of the DsRed+ FFLuc+ c1498 cells. This result was confirmed by flow cytometric analysis of the percentage of DsRed + cells in the chloroma. Conclusions This study shows that IFNα MSCs present a strong anti-leukemic effect in vitro and in vivo promoting apoptosis and thus decreasing the leukemic burden. Further experiments will focus on a potential synergetic effect with Cytarabine treatment and a preclinical study using human IFNα MSCs in a xenograft murine model. Disclosures No relevant conflicts of interest to declare.


2008 ◽  
Vol 84 (8) ◽  
pp. 659-667 ◽  
Author(s):  
Jean Marc Bertho ◽  
Christelle Demarquay ◽  
Moubarak Mouiseddine ◽  
Noémie Douenat ◽  
Johanna Stefani ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document