Combination Treatment with RG7112 and Pegylated Interferon α 2a Specifically Targets JAK2V617F Positive Progenitor Cells From Patients with Myeloproliferative Neoplasms.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2865-2865
Author(s):  
Min Lu ◽  
Xiaoli Wang ◽  
Goar Mosoyan ◽  
Yan Li ◽  
Ronald Hoffman

Abstract Abstract 2865 We have recently reported that MDM2 levels are increased in PV CD34+ cells as a consequence of JAK2617F, while the p53 levels are reduced in CD34+ cells from both patients with PV and PMF. These observations led us to explore therapeutic approaches by which to up-regulate p53 as a strategy to treat MPN patients. We previously reported that combination treatment with low doses of Peg IFNα 2a and Nutlin-3, an antagonist of MDM2, induced PV CD34+ apoptosis and inhibited PV colony formation significantly. The combination of these agents also decreased the proportion of JAK2V617F-positive HPCs. We demonstrated that these two drugs affect p53 through different pathways with Peg IFNα 2a activating p38 MAP kinase and STAT1 leading to increased p53 transcription while nutlin-3 prevents the degradation of p53 (Lu et al, Blood, 2012; In Press). RG7112 is an orally bioactivity small molecule inhibitor of p53-MDM2 binding which activates the p53 pathway. RG7112 is currently being evaluated in several phase 1 cancer trials. Based on our previous data, we hypothesized that RG7112 would be an effective drug to combine with low doses of IFN alpha to treat MPN patients. In order to evaluate the therapeutic effects of RG7112 alone or in combination with Peg IFNα 2a on hematopoietic progenitor cells from MPN patients, CD34+ cells were isolated from patients with PV and PMF patients and were treated with varying of doses of RG7112 alone or/and in combination with a low dose of Peg IFNα 2a and its effects on PV CD34+ cell proliferation, apoptosis and colony formation was evaluated. The effects of this drug on MPN CD34+ cells was compared to its effects on cord blood CD34+ cells After treatment with RG7112 at doses ranging from 100 nM to 5000 nM for 4 days, the numbers of PV CD34+ cells were reduced to a far greater degree (about 60%) than that observed with cord blood CD34+ cells (90% and 80%) at the doses between 200 and 500 nM indicating that low doses of RG7112 selectively affects MPN HPCs. 200nM of RG7112 and 200 U/ml of Peg-IFNα 2a alone induced cell cycle arrest of PV CD34+ cells, and combination treatment significantly promoted apoptosis of MPN CD34+ cells (2.5 folds). This observation leads us to examine the possibility that low doses of RG7112 might be used in combination with Peg-IFNα 2a to treat MPN patients. Treatment with 200 nM of RG7112 alone decreased PV CFU-GM and BFU-E-derived colony formation by 20% while combination treatment with 200 U/ml of Peg-IFNα 2a decreased PV CFU-GM and BFU-E-derived colony formation by 60% and 70%, respectively. Similarly, treatment with 200 nM of RG7112 alone decreased PMF CFU-GM and BFU-E-derived colony formation by 20% and 40%, respectively, while combination treatment with 200 U/ml of Peg-IFNα 2a decreased PMF CFU-GM and BFU-E-derived colony formation by 60% and 80%, respectively. We also found that treatment with both agents in combination also decreased the size of the hematopoietic colonies. By contrast, treatment with the same low doses of RG7112 and Peg-IFNα 2a alone or in combination did not significantly affect the ability of normal CD34+ cells to generate CFU-GM and BFU-E-derived colonies. Combined treatment with low doses of RG7112 and Peg-IFNα 2a decreased the total number of JAKV617F-positive PV HPC by 9% - 25% (p=0.014) leading to an increased proportion of colonies with wild type JAK2. In conclusion, these data demonstrate that IFNα plus RG7112 specifically target malignant MPN HPC by activating p53 in vitro, which we propose can be exploited therapeutically to eliminate JAK2V617F positive HPC in both PV and PMF patients. These results strongly suggest that combinations of low doses of IFNα and RG7112 might serve as a novel therapeutic strategy for the long term treatment of MPN patients. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2012 ◽  
Vol 120 (15) ◽  
pp. 3098-3105 ◽  
Author(s):  
Min Lu ◽  
Xiaoli Wang ◽  
Yan Li ◽  
Joseph Tripodi ◽  
Goar Mosoyan ◽  
...  

Abstract Interferon (IFN-α) is effective therapy for polycythemia vera (PV) patients, but it is frequently interrupted because of adverse events. To permit the long-term use of IFN, we propose combining low doses of IFN with Nutlin-3, an antagonist of MDM2, which is also capable of promoting PV CD34+ cell apoptosis. Combination treatment with subtherapeutic doses of Peg IFN-α 2a and Nutlin-3 inhibited PV CD34+ cell proliferation by 50% while inhibiting normal CD34+ cells by 30%. Combination treatment with Nutlin-3 and Peg IFN-α 2a inhibited PV colony formation by 55%-90% while inhibiting normal colony formation by 22%-30%. The combination of these agents also decreased the proportion of JAK2V617F-positive hematopoietic progenitor cells in 6 PV patients studied. Treatment with low doses of Peg IFN-α 2a combined with Nutlin-3 increased phospho-p53 and p21 protein levels in PV CD34+ cells and increased the degree of apoptosis. These 2 reagents affect the tumor suppressor p53 through different pathways with Peg IFN-α 2a activating p38 MAP kinase and STAT1, leading to increased p53 transcription, whereas Nutlin-3 prevents the degradation of p53. These data suggest that treatment with low doses of both Nutlin-3 combined with Peg IFN-α 2a can target PV hematopoietic progenitor cells, eliminating the numbers of malignant hematopoietic progenitor cells.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2718-2718
Author(s):  
Rieko Sekine ◽  
Toshio Kitamura ◽  
Takashi Tsuji ◽  
Arinobu Tojo

Abstract Pax5 is a key regulator of B cell commitment and is indispensable throughout the early stages of B cell differentiation. Human B lymphopoiesis can be simulated by cultivating cord blood (CB) progenitor cells on the murine bone marrow stromal cell line such as HESS-5. When CB CD34+ cells are seeded onto HESS-5 cells and supplemented with SCF and G-CSF, the number of non-adherent myeloid cells initially increases, reaching a maximum during the 3rd week, and decreases rapidly thereafter. On the other hand, foci of small B-lymphoid cells beneath the stromal layer appear during the 2nd week and increase in size until the 6th week. In this culture system, we have identified two C-terminal splice variants of Pax5; one skips exon 9 without subsequent frameshift (del9), and the other has a frameshift insert between exon 8 and 9, resulting in novel C-terminal sequences (ins8′). By quantitative RT-PCR analysis, del9 mRNA was barely detected in CB CD34+ cells and thereafter upregulated in progenitor B cells during the culture. In contrast, wild-type Pax5 (wt) and ins8′ mRNA could not be detected until the 3rd week. Then, wt mRNA was rapidly upregulated 100-fold or more abundantly than del9 and ins8′ mRNA (Fig. 1). Transactivation potential of Pax5 isoforms was examined with a reporter plasmid containing high-affinity Pax5 binding sites from the CD19 gene linked to the minimal TATA-like promoter. As a result, del9 showed constantly higher (up to 3.5-fold) activity, but ins8′ had slightly lower activity than wt Pax5. Furthermore, TLE4, a Groucho co-repressor family member and a known Pax5 partner, inhibited the transcriptional activity of wt Pax5 but not del9 or ins8′ isoform, suggesting its interaction with the exon 9 sequences of Pax5. We next examined the impact of Pax5 on growth and differentiation of CB CD34+ cells by retroviral transduction of each isoform as well as EGFP. After infection, sorted EGFP + cells were subjected to the colony-forming assay and the co-culture assay on HESS-5 cells. Erythroid or mixed colony formation was almost completely inhibited by either isoform. GM-colony formation was moderately inhibited by del9 and ins8′ isoforms, but profoundly suppressed by wt Pax5. In coculture with HESS-5 cells, the number of non-adherent myeloid cells was reduced by Pax5 isoforms in a similar manner to the results of GM-colony formation. Unexpectedly, after 6 weeks of culture, the number of CD19+ adherent lymphoid cells decreased more profoundly in the culture transduced with wt Pax5 than those with del9 and ins8′ isoforms (Fig 2). These results suggest that constitutive expression of Pax5 in cord blood progenitor cells substantially inhibits not only myelopoiesis but also B lymphopoiesis through the exon 9-dependent and independent mechanism. We also raise the possibility that developmentally regulated concentration of Pax5 isoforms may be crucial for human B lymphopoiesis. Figure Figure


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 514-514
Author(s):  
Wei Xiang ◽  
Jit Kong Cheong ◽  
Shi Hui Ang ◽  
Kartini Asari ◽  
Bryan Teo ◽  
...  

Abstract The use of Bcr-Abl tyrosine kinase inhibitors (TKIs) has led to excellent clinical responses in patients with chronic phase chronic myeloid leukaemia (CML). However these TKIs have been less effective as single agents in blast phase (BP) CML and this represents an urgent unmet need. A number of novel agents are now being investigated but most have not been translated to the clinics yet. Pyrvinium, a FDA-approved anthelminthic drug, was reported to selectively inhibit growth of a number of tumour cell types including myeloma and erythroleukaemia. We investigated the effect and mechanism of action of pyrvinium in BP-CML. Our results show that pyrvinium selectively targeted BP-CML CD34+ progenitor cells. It induced apoptosis in CD34+ cells from TKI-resistant BP-CML patients who harbour Bcr-Abl kinase mutations, while sparing normal cord blood CD34+ cells. In addition, pyrvinium was more effective in inhibiting colony formation and self-renewal capacity in BP-CML CD34+ cells than cord blood CD34+ cells. Further increase in apoptosis, decrease in colony formation and self-renewal were seen when dasatinib was combined with pyrvinium in BP-CML but not cord blood CD34+ cells (Figure 1 A-C). We also showed that pyrvinium was synergistic in combination with dasatinib in inhibiting proliferation and inducing apoptosis in CML cell lines. We next tested the effects of pyrvinium and its combination with dasatinib in a CML xenograft model and showed that pyrvinium significantly delayed tumour growth with no signs of toxicity in the mice. When combined with dasatinib, the tumour growth was completely inhibited (Figure 1D). These results indicate that pyrvinium is active in BP-CML in vitro and in vivo. The anti-cancer effects of pyrvinium have been reported to due to allosteric activation of casein kinase 1α (CK1α) and suppression of Wnt/β-catenin signalling (Thorne et al, Nature Chemical Biology 2010). However we showed that despite the efficient knockdown of CK1α and overexpression of β-catenin, the proliferation inhibitory effect of pyrvinium was not abolished, indicating a CK1α and β-catenin-independent mechanism of action in BP-CML. Instead, we observed that pyrvinium preferentially localized to mitochondria in CML cells and that pyrvinium inhibited oxygen consumption of CML cells within 5 minutes of treatment. In mitochondrial respiratory chain-deficient CML ρ0 cells which lack mitochondrial DNA and have undetectable oxygen consumption, we showed that the effect of pyrvinium in reducing ATP levels and inducing apoptosis was abolished. These results indicate that pyrvinium acts in CML through the inhibition of mitochondrial respiration. We have shown that pyrvinium alone and in combination with Bcr-Abl TKI selectively targets BP-CML progenitor cells and pyrvinium acts in BP-CML through the inhibition of mitochondrial respiration. Given that pyrvinium is already clinically available, our pre-clinical findings can be translated rapidly into the clinics. Our data also suggests that targeting mitochondrial respiration may be a potential therapeutic strategy in aggressive leukaemia. Figure 1 (A) Pyrvinium induces apoptosis of BP-CML CD34+ but not CB CD34+ cells and combination of pyrvinium and dasatinib is superior in inducing apoptosis. Pyrvinium is more effective in decreasing colony formation (B) and serial replating capacity (C) in BP-CML CD34+ than CB CD34+ cells. The combination is significantly more effective than single drug treatment (* p<0.01). (D) Pyrvinium is effective in vivo and significantly enhances the tumour growth inhibition of dasatinib (* p<0.01). Figure 1. (A) Pyrvinium induces apoptosis of BP-CML CD34+ but not CB CD34+ cells and combination of pyrvinium and dasatinib is superior in inducing apoptosis. Pyrvinium is more effective in decreasing colony formation (B) and serial replating capacity (C) in BP-CML CD34+ than CB CD34+ cells. The combination is significantly more effective than single drug treatment (* p<0.01). (D) Pyrvinium is effective in vivo and significantly enhances the tumour growth inhibition of dasatinib (* p<0.01). Disclosures Off Label Use: The use of pyrvinium in leukaemia. Chuah:Novartis: Honoraria; Bristol-Myers Squibb: Honoraria.


Blood ◽  
2011 ◽  
Vol 117 (6) ◽  
pp. 1840-1850 ◽  
Author(s):  
Kam Tong Leung ◽  
Kathy Yuen Yee Chan ◽  
Pak Cheung Ng ◽  
Tze Kin Lau ◽  
Wui Man Chiu ◽  
...  

Abstract The stromal cell–derived factor-1 (SDF-1)/chemokine C-X-C receptor 4 (CXCR4) axis plays a critical role in homing and engraftment of hematopoietic stem/progenitor cells (HSCs) during bone marrow transplantation. To investigate the transcriptional regulation provided by this axis, we performed the first differential transcriptome profiling of human cord blood CD34+ cells in response to short-term exposure to SDF-1 and identified a panel of genes with putative homing functions. We demonstrated that CD9, a member of the tetraspanin family of proteins, was expressed in CD34+CD38−/lo and CD34+CD38+ cells. CD9 levels were enhanced by SDF-1, which simultaneously down-regulated CXCR4 membrane expression. Using specific inhibitors and activators, we demonstrated that CD9 expression was modulated via CXCR4, G-protein, protein kinase C, phospholipase C, extracellular signal-regulated kinase, and Janus kinase 2 signals. Pretreatment of CD34+ cells with the anti-CD9 monoclonal antibody ALB6 significantly inhibited SDF-1–mediated transendothelial migration and calcium mobilization, whereas adhesion to fibronectin and endothelial cells was enhanced. Pretreatment of CD34+ cells with ALB6 significantly impaired their homing to bone marrow and spleen of sublethally irradiated NOD/SCID (nonobese diabetic/severe combined immune-deficient) mice. Sorted CD34+CD9− cells displayed lower bone marrow homing capacity compared with that of total CD34+ cells. CD9 expression on homed CD34+ cells was significantly up-regulated in vivo. Our results indicate that CD9 might possess specific functions in HSC homing.


2013 ◽  
Vol 22 (8) ◽  
pp. 1501-1506 ◽  
Author(s):  
Pascale Duchez ◽  
Jean Chevaleyre ◽  
Philippe Brunet De La Grange ◽  
Marija Vlaski ◽  
Jean-Michel Boiron ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 794-794
Author(s):  
Fan Liu ◽  
Xinyang Zhao ◽  
Fabiana Perna ◽  
Lan Wang ◽  
Priya Koppikar ◽  
...  

Abstract Abstract 794 Background: The cytoplasmic, non-receptor JAK2 tyrosine kinase is mutated at amino acid residue 617 (from valine to phenylalanine) in most patients with myeloproliferative neoplasms (MPNs), resulting in a constitutively activated kinase that phosphorylates STAT proteins in the absence of upstream signals. Overexpression of JAK2V617F leads to cytokine-independent growth of Ba/F3 cells and the JAK2V617F transgenic and knockin mice develop a disease phenotype resembling human polycythemia vera. Results: We hypothesized that the JAK2V617F occurs so consistently in MPNs because it gains some functional property. The type II arginine methyltransferase PRMT5 was initially identified because of its interaction with JAK2 in a yeast two hybrid screen. We examined the interaction between JAK2 and PRMT5 and found that JAK2V617F and JAK2K539L (another active JAK2 kinase) bound PRMT5 more strongly than did wild-type JAK2. PRMT5 mediates the symmetrical dimethylation of arginine residues within histones H2A and H4 and methylates other cellular proteins as well, such as p53. The oncogenic forms of JAK2 acquire the ability to phosphorylate PRMT5, which greatly impaired its methyltransferase activity. We have shown the in vivo importance of this post-translation modification as treating JAK2V617F-positive cells (but not the wild-type JAK2-harboring cells) with different JAK2 inhibitors significantly increased histone arginine methylation levels. To define the effect of inhibiting PRMT5 activity on hematopoiesis, we knocked down PRMT5 in human cord blood derived CD34+ cells using shRNA and observed increased colony formation and erythroid differentiation; In contrast, PRMT5 overexpression in these cells led to reduced colony formation and inhibition of erythroid differentiation. Furthermore, overexpression of PRMT5, especially a phosphorylation site mutant form of PRMT5 (PRMT5M6), diminishes the proliferative and erythroid generating capacity of JAK2V617F+ CD34+ cells isolated from MPN patients to a greater degree than normal cord blood CD34+ cells. Importantly, we found marked increase in PRMT5 phosphorylation in JAK2V617F-positive MPN patents relative to normal cord blood CD34+ cells, suggesting that this phosphorylation is important for the myeloproliferation phenotype. Conclusion: we show that the oncogenic mutant forms of JAK2 kinase, such as JAK2V617F and JAK2K539L, are not simply constitutively active forms of wild-type JAK2, rather they have specific gains-of-function that allow them to phosphorylate PRMT5 and down-regulate its enzymatic activity. Inhibition of PRMT5 contributes to the myeloproliferation and erythroid differentiation promoting effects of JAK2V617F. This gain-of –function mutation results in cross-talk between oncogenic kinases and histone arginine methylation. Taken together, we demonstrate a novel link between the mutant JAK2 kinases and PRMT5 methyltransferase activity, which contributes to MPN pathogenesis. Further insights about the shared gene expression profile of JAK2 inhibition vs. PRMT5 knockdown will be presented to understand the basics for the behavior change in hematopoietic stem/progenitor cells brought about by these two interventions. Disclosures: No relevant conflicts of interest to declare.


1999 ◽  
Vol 45 (4, Part 2 of 2) ◽  
pp. 143A-143A
Author(s):  
Sameer Bakhshi ◽  
Hsiao-Nan Hao ◽  
Jiun Zhao ◽  
Nahed Abdel-Haq ◽  
William D Lyman

Sign in / Sign up

Export Citation Format

Share Document