Inhibition by CTP and UTP analogues of uridine kinase from mouse leukemic cells

1982 ◽  
Vol 47 (12) ◽  
pp. 3464-3469 ◽  
Author(s):  
Jiří Veselý ◽  
Ivan Rosenberg ◽  
Antonín Holý

The new phosphonate analogues of CTP and UTP (CTPc and UTPc) inhibit the phosphorylation of uridine catalysed by uridine kinase in the presence of ATP and Mg2+-ions. The inhibition is competitive with respect to phosphate donor, and non-competitive with respect to phosphate acceptor. With respect to uridine the Ki constants for CTPc and UTPc are 7.5 . 10-5 mol l-1 and 1.0 . 10-4 mol l-1, respectively. With respect to ATP the Ki value for CTPc (3.6 . 10-6 mol l-1) is 3x lower than that for CTP. The novel analogues could be useful for further study of uridine kinase.

1983 ◽  
Vol 48 (6) ◽  
pp. 1783-1787 ◽  
Author(s):  
Jiří Veselý ◽  
Ivan Rosenberg ◽  
Antonín Holý

The phosphonate analogues of ATP and GTP can function as phosphate donors in uridine kinase reaction. The Km constants for ATP and its analogue ATPc (I) are identical, the Vmax value for ATP is five times higher than that for ATPc. Also the Vmax constants with respect to uridine follow the same pattern (250 nmol for ATP, 35.7 nmol for ATPc). The optimum Mg2+ concentration for the phosphonate is 3 times higher compared with ATP.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2846-2846
Author(s):  
Yoko Tabe ◽  
Yuanyuan Xu ◽  
Teresa McQueen ◽  
Michael Andreeff ◽  
Marina Konopleva

Abstract Transforming growth factor β1 (TGF-β1) is an essential regulator of cell proliferation, survival, and apoptosis, depending on the cellular context. TGF-β1 is also known to affect cell-to-cell interactions between tumor and stromal cells through production of the extracellular matrix and stimulation of integrin receptors. We investigated the role of TGF-β1 in the survival of human leukemic cells growing in the context of bone marrow (BM) microenvironment, and the anti-leukemia effects of the novel TGF-β receptor inhibitor LY2109761. BM-derived mesenchymal stem cells (MSC) produced TGF-β in an autocrine fashion. Treatment with rhTGF-β1 (2ng/mL) inhibited the spontaneous and Ara-C-induced apoptosis in U937 cells (% AnnexinV(+), control 34.5±8.4; TGF-β1 18.4±4.5; Ara-C 88.6±3.0; Ara-C/TGF-β1 60.4±8.0, p=0.04). These effects were more prominent in U937 cells co-cultured with MSC (% AnnexinV(+), control 19.4±2.8; TGF-β1 3.5±1.0; Ara-C 69.0±3.6; Ara-C+TGF-β1 24.9±3.3; p=0.01). In U937 cells co-cultured with MSC, rhTGF-β1 conferred higher cell protective effects on leukemia cells attached to MSC than on floating cells. Conversely, the pro-survival effects of TGF-β1 were inhibited by 5mM LY2109761 (%AnnexinV(+); MSC(−), control 31.8±2.3, TGF-β1 19.5±3.0, LY 28.4±4.4, TGF-β1+LY 37.7±2.0; MSC(+), control 22.1±1.7, TGF-β1 7.8±0.9, LY16.1±2.6, TGF-β1+ LY 18.0±1.1, p<0.01). Similar results were obtained using TGF-β1 neutralizing antibody. TGF-β1 induced pro-survival phosphorylation of Akt in U937 cells cultured alone or co-cultured with MSC, which was abrogated by LY2109761. Further, rhTGF-β1 induced a moderate increase in C/EBPβ gene and LAP isoform (cell cycle arrest inducing) of C/EBPβ protein in U937 cells cultured without MSCs, while markedly upregulating C/EBPβ gene and protein, both LIP (cell proliferation inducing) and LAP isoforms, under MSCs co-culture condition, suggesting the novel role of C/EBPβ in TGF-β1-mediated U937 cell survival. In summary, these results indicate that TGF-β1-secreting BM stromal cells promote the survival of U937 leukemia cells via direct cell-to-cell interaction and promote chemoresistance of leukemia cells through the activation of Akt signaling and upregulation of C/EBPβ. The blockade of TGF-β signaling by LY2109761, which effectively inhibited the pro-survival signaling, may enhance the efficacy of chemotherapy against leukemia cells in the BM microenvironment.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1789-1789
Author(s):  
Joachim Rudolf Göthert ◽  
Roze Imsak ◽  
Michael Möllmann ◽  
Stefanie Weber ◽  
Mandy Necke ◽  
...  

Abstract Abstract 1789 Active agents for the treatment of CLL are available. However, CLL eventually progresses to refractory disease. Defects in apoptosis, cytogenetic abnormalities and alterations in non-malignant cells of the micro-environment are likely to be the major causes of therapeutic resistance. Therefore, novel agents are required to abrogate apoptosis blocks and to alter the micro-environment of CLL. One approach targets cyclin-dependent kinases (Cdk). Specifically, Cdk9 promotes initiation and elongation steps in transcription maintaining expression levels of anti-apoptotic proteins with inherently rapid turnover rates. Inhibitors of cyclin-dependent kinases (Cdks) have been previously shown to exert cytotoxic effects on CLL cells by inhibiting Cdk7 and Cdk9. However, inhibitors used in these studies suffer from lack of selectivity, making unambiguous interpretation of results difficult, and leading to unspecific toxicity. Here, we studied the novel Cdk inhibitor LDC-9-A, which exhibits potent and selective inhibitory activity against Cdk9. The in vitro cytotoxicity of LDC-9-A was evaluated after 24 and 48 hours in CLL cases (n=8) and the CLL derived prolymphocytic cell line MEC-1. After a culture period of 48 hours LD50 values ranged between 0.1 and 1.9 μM for primary CLL cells and MEC-1 cells, respectively. Western blot analysis of protein extracts from MEC-1 cells showed that LDC-9-A down-modulates the anti-apoptotic proteins Bcl-2 and Mcl-1 in a dose- and time-dependent manner. In order to study the in vivo anti-CLL activity of LDC-9-A 9–12 month-old transgenic TCL1 mice (n=4) were treated for two consecutive days with an orally bioavailable LDC-9-A formulation (50 mg/kg body weight). Control groups consisted of TCL1 mice receiving vehicle only (n=3) and wild-type control littermates (n=4) receiving LDC-9-A. Numbers of peripheral blood leukemic cells (CD19+CD5+CD3−) and non-malignant T-cells (CD19−CD5+CD3+) were longitudinally evaluated by flow cytometry. Remarkably, LDC-9-A treatment of TCL1 transgenic mice led to a decrease of blood leukemic cells to 6.3±2.0% (mean±SEM) of baseline levels (p<0.0001) two days post treatment cessation. The numbers of T-cells decreased to 15.5±6.6% (p<0.01) in TCL1 mice while the blood T-cells of LDC-9-A treated wild-type controls did only decrease to 49.5±11.6% of baseline levels. Because LDC-9-A was expected to alter cellular Cdk9-mediated anti-apoptotic protein replenishment Bcl-2 levels were determined by flow cytometry analysis. Interestingly, cellular Bcl-2 levels of surviving leukemic cells as well as surviving non-malignant T-cells were significantly higher compared to levels before treatment (1.6- and 2.6-fold, respectively, p<0.05). These data implied that either LDC-9-A was not able to clear cells with high Bcl-2 expression or that LDC-9-A led to a compensatory up-regulation of Bcl-2 in residual cells. Finally, the anti-CLL activity of LDC-9-A was further validated by employing a recently developed adoptive transfer model of primary human CLL cells. In this model NOD/SCID/γcnull (NSG) mice were transplanted with CLL cells and treated with different doses of LDC-9-A for 3–5 sequential days. In line with the data generated with the TCL1 murine CLL model the LDC-9-A treatment of xenografted animals with 60 and 90 mg/kg body weight resulted in an 80–90% reduction of human CLL cell recovery from murine spleens as compared to vehicle treated controls. In summary, we demonstrated effective anti-CLL activity of the novel Cdk9 inhibitor LDC-9-A in two independent preclinical mouse models. The activity of LDC-9-A against CLL cells was achieved at least in part by modulating cellular anti-apoptotic protein levels. Furthermore, LDC-9-A interfered with non-malignant T-cells, which represent an important component of the CLL micro-environment. Disclosures: Eickhoff: Lead Discovery Center GmbH (LDC): Employment. Baumann:Lead Discovery Center GmbH (LDC): Employment. Unger:Lead Discovery Center GmbH (LDC): Employment. Schultz-Fademrecht:Lead Discovery Center GmbH (LDC): Employment. Dürig:Roche: Speakers Bureau; GSK: Speakers Bureau; Santaris Pharma: Consultancy, Research Funding; Celgene: Research Funding.


2006 ◽  
Vol 30 (12) ◽  
pp. 1541-1546 ◽  
Author(s):  
Uichi Nishiyama ◽  
Tetsuya Yoshino ◽  
Masako Ozai ◽  
Rieko Yoshioka ◽  
Motoko Fujisawa ◽  
...  

Leukemia ◽  
2013 ◽  
Vol 27 (11) ◽  
pp. 2220-2228 ◽  
Author(s):  
N Garnier ◽  
L A Petruccelli ◽  
M F Molina ◽  
M Kourelis ◽  
S Kwan ◽  
...  
Keyword(s):  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1958-1958
Author(s):  
Naoko Watanabe-Okochi ◽  
Jiro Kitaura ◽  
Toshio Kitamura ◽  
Mineo Kurokawa

Abstract Abstract 1958 Poster Board I-981 Background: Evi1 gene is located on chromosome 3q26 and aberrantly expressed in acute myeloid leukemia (AML) patients with or without 3q26 abnormalities, and inappropriate expression of Evi1 associates with poor prognosis. Evi-1 is originally identified in a common integration site of murine leukemia retrovirus and enhanced expression of Evi1 by retrovirus integration is thought to be responsible for leukemogenesis in mouse models. However, retroviral expression of marker genes such as GFP has not induced leukemia even if some clones possessing integration at Evi1 site have been identified. These data indicate that Evi1 requires cooperative factors to induce progressive leukemia, whereas overexpression of Evi1 is enough to lead to clonal expansion of hematopoietic cells. Therefore, identifying genes collaborating with Evi1 is one of the key issues of understanding Evi1-related leukemogenesis. Recently, we demonstrated that a point mutation of the transcription factor AML1 (AML1-D171N) can induce myelodysplastic syndrome (MDS) that progresses to AML in association with overexpression of Evi1 through a mouse bone marrow transplantation model. In that work, we analyzed mice transplanted with BM cells transduced Evi1 alone as control and surprisingly confirmed that all of the mice developed leukemia within 6-11 months after the transplant. In this report, we will describe interesting findings in the novel mouse model of Evi1-induced leukemia. Result: C57BL6/Ly-5.1 murine BM cells infected with retroviruses harboring Evi1 were transplanted into irradiated syngeneic Ly-5.2 mice. The mice looked fine until 5 months, but GFP-positive-Evi1 expressing cells were gradually increased in the peripheral blood (PB), and then the mice died at 6-11 months after the transplantation. The mice showed dysplastic features in myeloid and erythroid cells, increase of blasts in the PB and the BM, hepatosplenomegaly, slight anemia, and some of the mice showed severe leukocytosis. The mice were thought to die of multiple organ failure due to invasion of leukemic cells not due to anemia. The phenotype is different from that of the mouse BMT model expressing Evi1 by retrovirus reported by another group, in which the mice died about 10 months with severe peripheral cytopenia and finally the disease did not progress to AML. Therefore, we considered that Evi1 might have collaborated with unknown genes near retrovirus integration sites in our case and analyzed integration sites by the bubble PCR method. Interestingly, frequent integration at 3' side of C/EBPb gene was found in six mice out of eight mice transplanted with Evi1-transduced BM cells. The integrations were located at 62.5-86.7kb downstream of C/EBPb gene. Next, we examined the expression level of C/EBPb, Tmem189, and Ptpn1, all of which are located near the integration site, and confirmed that C/EBPb showed elevated expression although neither Tmem189 nor Ptpn1 did. We also identified Bcas1, Rps6ka1, and Rapgef4 genes at the retroviral integration site in the other two mice without integration near C/EBPb. Discussion: C/EBPb, also known as NF-IL6, is a transcription factor that specifically binds to an IL1-responsive element in the IL-6 gene and has a role in regulation not only for the IL-6 gene but also for several cytokine genes such as TNF, IL-8, and G-CSF. The hematopoietic progenitor cells of C/EBPb-deficient mice have been reported to respond imperfectly to GM-CSF and G-CSF. Furthermore, C/EBPb is a downstream target of the Ras-Raf pathway. The locus of C/EBPb gene has been reported as a common integration site in the Retrovirus Tagged Cancer Gene Database (RTCGD), which is a database of retroviral insertional mutagenesis in mouse tumors. AKxD mice, Cdkn2a-KO mice, NUP98/HOXD13 transgenic mice, and MYC/Runx2 transgenic mice were reported to develop myeloid or lymphoid leukemia by retroviral insertion into 3' side of C/EBPb gene. In this study, we identified frequent integration at 3' side of C/EBPb gene in Evi1-transduced leukemic cells, whereas we have not identified this locus in AML1-mutants-transduced leukemic cells. Based on these findings and our results, C/EBPb is supposed to be a candidate gene to collaborate with Evi1 in leukemogenesis. Conclusion: We identified involvement of C/EBPb in Evi1-induced leukemogenesis. The novel mouse model that we generated in this study could help understanding the molecular basis of Evi1-related leukemia. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2906-2906
Author(s):  
Helena Mistry ◽  
Grace Hsieh ◽  
Sara Buhrlage ◽  
Min Huang ◽  
Eunmi Park ◽  
...  

Abstract ID1 (inhibitor of DNA-binding-1) is a member of the helix-loop-helix family of transcriptional regulatory proteins. The ID-family of proteins (ID1-ID4) inhibit the DNA binding of transcription factors which regulate cellular differentiation and proliferation. Accordingly, deregulation of ID proteins has been observed in many cancer types including leukemia. High levels of ID1 expression are found in primary acute myeloid leukemia (AML) samples and correlate with poor prognosis. ID1 is also identified as a common downstream target of the oncogenic tyrosine kinases, BCR-ABL, TEL-ABL and FLT3-ITD. In addition, Id1 has been shown to promote a myeloproliferative disease in mice, and knockdown of ID1 expression inhibits leukemic cell growth. Therefore, ID1 is an excellent candidate for targeted therapy in leukemia. However, suitable drugs to target ID1 have not been developed to date. ID1 is normally polyubiquitinated and degraded by the proteasome. Recently, it has been shown that USP1, a ubiquitin specific protease, deubiquitinates ID1 and rescues it from proteasome degradation. Inhibition of USP1 therefore offers a new avenue to target ID1 in cancer. Here, using a Ubiquitin-Rhodamine-based high throughput screen, we identified small molecule inhibitors of USP1 and investigated their therapeutic potential for leukemia. These inhibitors blocked the deubiquitinating enzyme activity of USP1 in vitro in a dose-dependent manner with an IC50 in the nanomolar range, and also targeted the enzyme activity of native USP1. To determine the cellular consequences of USP1 inhibition, we exposed leukemic cells to micromolar concentrations of the inhibitors and evaluated ID1 levels and survival. USP1 inhibitors promoted the degradation of ID1 and, concurrently, inhibited the growth (>90% inhibition in 24 hrs) of chronic myelogenous leukemia (CML) and AML cell lines with induction of apoptosis in a dose dependent manner. The EC50 of the inhibitors for the leukemic cell growth inhibition was approximately 1.07 μM ± 0.08 (95% Confidence Limits). Interestingly, exposure to low doses of USP1 inhibitor for 5 days in culture resulted in erythroid differentiation of K562 leukemic cells. A known USP1 inhibitor, Pimozide, also promoted ID1 degradation and inhibited growth of leukemic cells (>90% inhibition in 48 hrs), though at a higher drug concentrations as compared to the novel USP1 inhibitors. Importantly, the novel USP1 inhibitors promoted ID1 degradation and exhibited cytotoxicity (>90% death in 48 hrs) in primary AML patient-derived leukemic cells. Notably, siRNA-mediated knockdown of USP1 in K562 leukemic cells resulted in growth inhibition, increased apoptosis and cell cycle arrest. Collectively, our results demonstrate that the novel small molecule inhibitors of USP1 promote ID1 degradation and are cytotoxic to leukemic cells. The identification of USP1 inhibitors therefore opens up a new approach for leukemia therapy. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 125 (26) ◽  
pp. 4010-4016 ◽  
Author(s):  
Elias Jabbour ◽  
Susan O’Brien ◽  
Farhad Ravandi ◽  
Hagop Kantarjian

Abstract With modern intensive combination polychemotherapy, the complete response (CR) rate in adults with acute lymphoblastic leukemia (ALL) is 80% to 90%, and the cure rate is 40% to 50%. Hence, there is a need to develop effective salvage therapies and combine novel agents with standard effective chemotherapy. ALL leukemic cells express several surface antigens amenable to target therapies, including CD20, CD22, and CD19. Monoclonal antibodies target these leukemic surface antigens selectively and minimize off-target toxicity. When added to frontline chemotherapy, rituximab, an antibody directed against CD20, increases cure rates of adults with Burkitt leukemia from 40% to 80% and those with pre-B ALL from 35% to 50%. Inotuzumab ozogamicin, a CD22 monoclonal antibody bound to calicheamicin, has resulted in marrow CR rates of 55% and a median survival of 6 to 7 months when given to patients with refractory-relapsed ALL. Blinatumomab, a biallelic T cell engaging the CD3-CD19 monoclonal antibody, also resulted in overall response rates of 40% to 50% and a median survival of 6.5 months in a similar refractory-relapsed population. Other promising monoclonal antibodies targeting CD20 (ofatumumab and obinutuzumab) or CD19 or CD20 and bound to different cytotoxins or immunotoxins are under development. Combined modalities of chemotherapy and the novel monoclonal antibodies are under investigation.


Sign in / Sign up

Export Citation Format

Share Document