Study on the Mechanisms of CML Blast Crisis by Comparative Proteome Analysis.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4770-4770
Author(s):  
Xiaoli Liu ◽  
Qingfeng Du ◽  
Song Zhang ◽  
Rong Li ◽  
Feng Yao ◽  
...  

Abstract The clinical course of chronic myeloid leukemia(CML)is characteristically triphasic, comprising chronic and accelerated phases and blast crisis. Chronic phase(CP) is characterized by the Ph chromosome as the sole genetic abnormality and blast crisis(BC), which is the terminal phase of CML, often associated with additional chromosomal and molecular secondary changes. Although CML is probably the most extensively studied human malignancy, the mechanisms of CML blast crisis are still poorly understood. In current study, we analyzed the changes of protein expression between CML-CP(25 cases) and CML-BC(20 cases) by Two-dimensional polyacrylamide gel electrophoresis(2-D PAGE). Compared with that of CML-CP, 33 proteins’ intensities of CML-BC were found to have significant difference including 23 increasing and 13 decreasing. 15 proteins were identified by peptide mass fingerprint in combination with database searching including proteasome activator complex, hnRNP, annexin A4, serine proteinase inhibitor, annexin A1, GAPDH, RhoGDI, enolase, proteasome subunit 6a, GTP binding protein, leukotriene A4 hydrolase, Rac-RhoGDI, thioredoxin, proteasome subunit 4β and DJ-1 protein, and the functions of these proteins involve cell signal transduction, apoptosis, proliferation and transcription. In conclusion, our study provided a profile of protein expression difference between CML-CP and CML-BC and contributed to understand the mechanisms of CML blast crisis.

2001 ◽  
Vol 19 (11) ◽  
pp. 2915-2926 ◽  
Author(s):  
Razelle Kurzrock ◽  
Carlos E. Bueso-Ramos ◽  
Hagop Kantarjian ◽  
Emil Freireich ◽  
Susan L. Tucker ◽  
...  

PURPOSE: To document the characteristics of patients with major breakpoint cluster region (M-bcr) rearrangement–negative chronic myelogenous leukemia (CML). PATIENTS AND METHODS: The hematopathologist, who was blinded to patients’ molecular status, reviewed the referral bone marrows and peripheral-blood smears from 26 patients with Philadelphia (Ph) translocation–negative CML who lacked Bcr rearrangement (and other evidence of a Bcr-Abl anomaly) and 14 patients (controls) with chronic-phase Ph-positive CML. Clinical data was ascertained by chart review. RESULTS: Among the 26 M-bcr rearrangement–negative CML patients, three pathologic subtypes emerged: (1) patients indistinguishable from classic CML (n = 9), (2) patients with atypical CML (n = 8), and (3) patients with chronic neutrophilic leukemia (n = 9). Among the 14 patients with Ph-positive CML who were included in the blinded review, 13 were classified as classic CML, and one was classified as atypical CML. The only statistically significant difference between M-bcr rearrangement–negative subgroups was in the proportion of patients having karyotypic abnormalities, an observation common only in patients with atypical CML (P = 0.008). However, the small number of patients in each subgroup limited our ability to differentiate between them. Interferon alfa induced complete hematologic remission in five of 14 patients; four of these remissions lasted more than 5 years. Only one of 26 patients developed blast crisis. The median survival of the 26 patients was 37 months. CONCLUSION: Patients with M-bcr rearrangement–negative CML fall into three morphologic subgroups. Disease evolution does not generally involve blastic transformation. Instead, patients show progressive organomegaly, leukocytosis, anemia, and thrombocytosis. Some patients in each subgroup can respond to interferon alfa.


Crustaceana ◽  
2015 ◽  
Vol 88 (7-8) ◽  
pp. 747-765
Author(s):  
P. A. Valentim-Neto ◽  
A. P. M. Fraga ◽  
G. A. S. Müller ◽  
M. R. F. Marques

To better understand the molecular pathogenesis of white spot syndrome virus (WSSV) inLitopenaeus vannamei(Boone, 1931), the protein expression profile in gills was characterized. Farmed shrimp WSSV positive were randomly sorted based on nested PCR. The proteomic analysis of gill proteins was performed using two-dimensional electrophoresis (2-DE), with isofocalisation on an immobilized linear gradient (pH 3-10), followed by separation based on molecular weight using 12.5% denaturating polyacrylamide gel electrophoresis (SDS-PAGE). The comparative analysis of the 2-DE profile between the two groups revealed eight differentially expressed spots in gills of naturally infected shrimp. The spots were successfully identified using MALDI-TOF mass spectrometry peptide mass fingerprint. The up-regulated proteins unique to infected shrimp were identified as peptidyl-prolyl isomerase, mortality factor 4-like protein 1, calreticulin, recombination activating protein, failed axon connection protein, 40S ribosomal S2 and N-deacetylase/N-sulfotransferase. The down-regulated protein unique to non-infected shrimp (control group) was identified as an inhibitor of apoptosis. The differentially expressed proteins are involved in several important cellular processes, such as host defence and protein metabolism. The present work contributes to a better understanding of the overall molecular responses elicited by WSSV infection inL. vannamei, as well as to point out potential molecular biomarkers to evaluate the susceptibility to the virus and the sanitary status in farmed shrimp.


2007 ◽  
Vol 292 (5) ◽  
pp. G1272-G1282 ◽  
Author(s):  
Qilie Luo ◽  
Linda Siconolfi-Baez ◽  
Pallavi Annamaneni ◽  
Mark T. Bielawski ◽  
Phyllis M. Novikoff ◽  
...  

Protein expression patterns were analyzed in a rat model of hepatic neoplasia to detect changes reflecting biological mechanism or potential therapeutic targets. The rat resistant hepatocyte model of carcinogenesis was studied, with a focus on the earliest preneoplastic lesion visible in the liver, the preneoplastic hyperplastic nodule. Expression differences were shown by two-dimensional polyacrylamide gel electrophoresis and image analysis. Polypeptide masses were measured by peptide mass fingerprinting using matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF) and their sequences were obtained by tandem mass spectrometry. Alterations in expression of cytoskeletal and functional proteins were demonstrated, consistent with biological changes known to occur in the preneoplastic cells. Of particular interest was the differential expression of a serine protease inhibitor (serpin) with a role implicated in angiogenesis. Serpin, implicated in the inhibition of angiogenesis, is present in normal liver but has greatly reduced expression at the preneoplastic stage of liver cancer development. Immunofluorescence microscopy with antibodies to this serpin, kallistatin, supports the proteomic identification. Immunofluorescence microscopy with antibodies to the blood vessel marker von Willebrand factor provides evidence for neovascularization in the liver containing multiple preneoplastic nodules. These observations suggest that at an early stage of liver carcinogenesis reduction or loss of angiogenesis inhibitors may contribute to initiation of neoangiogenesis. A number of other identified proteins known to be associated with hepatomas are also present at early-stage neoplasia.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 27-27 ◽  
Author(s):  
Francois Guilhot ◽  
Richard A. Larson ◽  
Stephen G. O’Brien ◽  
Insa Gathmann ◽  
Brian J. Druker

Abstract Background: Results from the International Randomized Study of Interferon and STI571 (IRIS) trial showed that achievement of a CCyR is prognostically relevant for long-term survival in patients (pts) with chronic myeloid leukemia in chronic phase (CML-CP). With the advent of next generation tyrosine kinase inhibitors (TKIs), there is a need to understand factors that may influence long term outcomes. Here we analyze whether time to achievement of a CCyR affects long term outcomes. Methods: The relationship between time to CCyR and long-term outcomes was examined for 551 imatinib-treated patients with newly diagnosed CML-CP at the 6-year follow up of the IRIS trial. As evaluations included non-responders, landmark analyses were conducted in which only pts who were treated for ≥1 year were included (n=509). Patients were stratified according to time to achieving a CCyR as follows: ≤6 months (n=265), >6≤12 months (n=99), >12≤18 months (n=34), >18 months (n=49), or no CCyR during imatinib treatment (n=62). In each category patients were assessed for duration of cytogenetic response, event free survival (EFS; any event while on study), freedom from progression to accelerated phase (AP) or blast crisis (BC), and overall survival (OS). Results: In the 447 patients who were treated for at least 1 year and achieved a CCyR (<1% Ph+) during therapy, the durability of major cytogenetic response (1%–35% Ph+) did not differ significantly regardless of when CCyR was achieved (P=0.76). For the overall population, estimated 6-year rates were 88% for OS, 83% for EFS, and 93% for freedom from progression to AP/BC. No statistically significant difference was observed between the responders when categorized according to time to response. However, patients who did not achieve a CCyR had significantly worse outcomes than those who achieved CCyR (P<0.001). Estimated 6-year OS rates were 94%, 95%, 91% and 98% for patients who first achieved a CCyR within 6, 12, 18 months and after 18 months, respectively (P=0.55 for overall comparison of the response categories), compared with 63% for pts without CCyR during imatinib therapy. Estimated EFS rates at 6 years were 93%, 90%, 87% and 89% (P=0.58) and 33% for patients who do not achieve a CCyR, respectively. At 6 years the estimated rates of freedom from progression to AP/BC were 97%, 97%, 97% and 98% (P=0.98), respectively, but only 63% for pts who do not achieve a CCyR. Overall Survival by Time to CCyR. Conclusion: Long-term outcomes on imatinib for patients in CML-CP are independent of time to achieve CCyR. Therefore, achievement of a “late” CCyR does not increase the potential for progression or portend a worse overall survival for patients treated with imatinib. Figure Figure


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4229-4229
Author(s):  
Liu Xiaoli ◽  
Hongqian Zhu ◽  
Song Zhang ◽  
Qingfeng Du ◽  
Junmei Gong ◽  
...  

Abstract The blast crisis(BC) is terminal phase of chronic myeloid leukemia (CML), which is accompanied by an increase in both BCR/ABL mRNA and protein level and imatinib(IM) resistance. The BCR/ABL oncoprotein is responsible for inducing and sustaining the leukemic phenotype through its deregulated tyrosine kinase activity which is essential for recruitment and induction of signaling pathways leading to cytokine-independent proliferation, resistance to apoptosis, and impaired differentiation of BCR/ABL-expressing myeloid and lymphoid progenitors. However, the molecular mechanisms responsible for blastic transformation remain poorly understood, although a reasonable assumption is that the unrestrained activity of BCR/ABL in hematopoietic stem/progenitor cells is the primary determinant of disease progression. We had analyzed the changes of protein expression between the bone marrow mononuclear cells in CML-CP and that in CML-BC by the technique of proteome. Compared to CML-CP, the protein expression of heterogeneous nuclear ribonucleoprotein K(hnRNPK) increases in CML-BC. In present study, the over-expression of hnRNPK in CML-BC was verified by western blotting. Furthermore, the expression of hnRNPK increases in a new imatinib-resistant BCR/ABL-positive cell line, K562-R, than in the wild-type K562. After K562 and K562-R were treated by different inhibitors such as PD98059, LY294002, AG490 and imatinib, the protein expression and mRNA level of hnRNPK were detected by western blotting and QRT-PCR. Inhibition of BCR-ABL by imatinib significantly decreased hnRNPK expression in K562 and inhibition of MEK by PD98059, downstream of the BCR-ABL signaling cascade, decreased hnRNPK expression in both K562 and K562-R cells. In addition, down-regulation of hnRNPK by small double-stranded RNA specifically complementary to hnRNPK (siRNA-hnRNPK) can inhibite cell growth and induce maximal G2/M block at 48 hours in both K562 and K562-R cells, whereas cell appototis was observed only at 72 hours by AnnexinV-PI assay. The combination of siRNA-hnRNPK and imatinib induce more cell death in K562-R cells than either treatment alone. Our data therefore suggest that hnRNPK is regulated by the BCR-ABL/MAPK cascade in Ph+ CML. The results that down-regulating hnRNPK expression induced cell-growth arrest and subsequent cell death suggest the potential therapeutic utility of this strategy in patients with CML.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1074-1074
Author(s):  
Daniel Jacob Goff ◽  
Annelie Abrahamsson ◽  
Ifat Geron ◽  
Catriona Jamieson

Abstract Introduction: A growing body of evidence suggests that a relatively rare subset of cells within a cancer subverts properties normally ascribed to stem cells in regenerating tissues, such as enhanced self-renewal and survival capacity. Recent studies suggest that these cancer stem cells (CSC) are resistant to treatments that target rapidly dividing cells. In blast crisis chronic myeloid leukemia (BC CML) and some forms of acute myelogenous leukemia (AML), research performed by ourselves and others indicates that CSC originate from the CD34+CD38+lineage- compartment of hematopoietic cells and can serially transplant blast crisis leukemia in immunodeficient mice. Despite abundant data indicating that Bcl-2 family proteins are involved in CML progression, the importance of these proteins in CSC survival remains to be elucidated. Clinical data have shown that CML stem cells become more resistant to therapies targeting BCR-ABL with progression to blast crisis. As BCR-ABL targeted therapy initiates apoptosis, these results suggest that CML CSC may become increasingly resistant to apoptosis with progression. Based on these findings and the results from our serial transplantation experiments, we hypothesized that CML CSC deregulate apoptosis pathways by differential expression of Bcl-2 family molecules and that these changes contribute to CSC ability to survive serial transplantation. Methods: Quantitative FACS Aria analysis of Bcl-2 protein levels was compared in blast crisis (n=5) and chronic phase CML (n=3) patient samples. Mean fluorescence intensity (MFI) of Bcl-2 staining was compared between different hematopoietic populations within the patient samples. For gene expression analysis, cDNA was made from RNA isolated from sorted progenitor populations (CD34+CD38+Lin −). Isoform specific RT-PCR was used to determine expression levels of Bcl-2, Bcl-X, and Mcl-1 isoforms. Mcl-1 expression was confirmed using qPCR. In addition, preliminary experiments were performed (n=2) to determine if CSC engraftment could be reduced in vivo by targeted inhibition of Bcl-2 family molecules using Apogossypol, a clinically tested Bcl-2-family inhibitor. Briefly, immunocompromised neonatal mice were transplanted intrahepatically with luciferase GFP transduced granulocytic sarcomas from mice transplanted with BC CSC using our previously published methodology (Geron et al, Cancer Cell 2008). Transplanted mice were treated for 15 days with Apogossypol by oral gavage and engraftment was monitored by weekly bioluminescent imaging. Engraftment levels were determined by FACS analysis of human CD45+ expression in mouse livers on week 11 post-transplant. Results: Comparing the MFI of Bcl-2 staining in the entire live mononuclear cell population, we detected no statistically significant difference in levels between the blast crisis and chronic phase samples. However, when we gated on separate cell populations, differences in the Bcl-2 MFI emerged. There was a statistically significant increase (P&lt;0.03) in Bcl-2 MFI exclusively in the CD34+CD38+lineage- population of the blast crisis samples indicative of cell type and context specific deregulation of apoptosis in the CSC population. Further, we were interested in whether there were differences in Bcl-2 family expression at the transcriptional level. Notably, while we detected no difference in the levels of the isoforms of Bcl-2 and Bcl-X, splice isoform specific RT-PCR and qPCR revealed a decrease in the expression of the short isoform of Mcl-1, which encodes a pro-apoptotic protein, in serially transplanted BC CSC (CD34+CD38+). Together these results indicate that CML CSC may indeed deregulate the expression of several Bcl-2 family proteins. To test the therapeutic potential of inhibiting these deregulated apoptotic pathways in CML, we treated mice engrafted with CML CSC with Apogossypol, a broad-spectrum inhibitor of pro-survival Bcl-2 molecules. We saw a statistically significant decrease (P&lt;0.05) in the number of CD45+ cells engrafted in the mouse liver after 3 weeks of Apogossypol treatment. Overall, our results suggest that the subversion of apoptosis plays an important role in allowing CML CSC to be serially transplanted and that apoptotic pathways may be a useful target for therapeutics aimed at inhibiting these cells.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4532-4532
Author(s):  
Young Y. Lee ◽  
Kwang-Sung Ahn ◽  
Sung-Soo Yoon ◽  
Jung H. Choi ◽  
Byoung B. Park ◽  
...  

Abstract To identify a gene signature for prognostic markers at transition from chronic phase to blast crisis of chronic myeloid leukemia (CML), we have applied Affymetrix Genechips of 22,000 transcripts to analyze total RNA of CML cells from 12 patients with chronic phase and 12 patients with blast crisis. Data analysis using GeneSpring 6.0 generated a list of 143 differentially expressed genes. A total of 89 genes were up-regulated and 54 genes were down-regulated in blast crisis of CML, and vice versa in chronic phase of CML. Array data for 32 genes was validated using quantitative realtime PCR analysis. The expression levels of HSA6591, FLT3, NTE5, RSG1, LAF4, CPA3, ATF, FCGR3A, MYD88, IFIT1, TP73L, DTNA, MDA, and IL18R1 showed statistically significant difference (p < 0.05) between chronic phase and blast crisis. Since CML cells of blast crisis were generally unresponsive to STI571, we further analyzed roles of FLT3 which is known to be a poor prognositic marker in acute myeloid leukemia. For this experiment, K562 cells (CML blast cells) were transfected with small hairpin RNAs (shRNAs), also referred to as small interfering RNAs, to target human FLT3, resulting in the significant inhibition of FLT3 expression at mRNA and protein levels. MTT assay demonstrated that FLT3 knockdown K562 cells by shRNAs were more sensitive to STI571 compared to wild type of K562, although there was no difference at high concentration of STI571 (320 nM) between FLT3 knockdown K562 cells and wild type of K562 cells. The higher expression levels of apoptosis related genes (PARP, caspase-3, Bax) were observed in FLT3 knockdown K562 cells compared to wild type of K562 cells. Thus, RNA interference-directed targeting of FLT3 might be a novel treatment modality in STI571 refractory CML patients.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4508-4508
Author(s):  
Su Chu ◽  
YinWei Ho ◽  
Guisen Zhao ◽  
Tessa L. Holyoake ◽  
Samuel Waxman ◽  
...  

Abstract Tyrosine kinase inhibitors (TKI) are highly effective in the treatment of CML but do not eliminate primitive, quiescent leukemia stem cells (LSC), which persist as a potential source of leukemia relapse. Additional strategies to enhance eradication of LSC are required to increase the possibility of treatment free remissions for CML patients. Glutathione S-transferase P1-1 (GSTP1-1) is a Phase II detoxifying enzyme which is overexpressed in several cancers and causes drug resistance. The diuretic Ethacrynic acid (EA) is a GSTP1-1 activity inhibitor and also induces celldeath in malignant cells at high concentrations. We have synthesized two EAoxadiazole analogs, 6U and 6S, which demonstrate enhancedpro-apoptotic effects in CML K562 cells expressing high levels of GSTP1-1. Previously we found that 6U and 6S induced apoptosis through downregulation of anti-apoptotic protein MCL-1 in addition to their GSTP1-1 activity inhibition. We extended these observations to primary CML stem/progenitor cells. We observed increased expression of GSTP1-1 mRNA and protein, associated with increased expression of MCL-1, BCL2 and BCL-xL, in chronic phase (CP) and blast crisis (BC) CML compared to normal CD34+ cells. Treatment of CP CML CD34+ cells with 6U or 6S (1 to 6µM) for 24 to 48 hours resulted in a significant dose-dependent induction of apoptosis, inhibition of cell growth, and reduction in colony forming cell (CFC) numbers, with 6U demonstrating greater efficacy than 6S. Treatment with 6U did not induce significant apoptosis of normal (NL) CD34+ cells at doses below 4µM. 6U induced significantly less apoptosis in NL compared with CML CD34+ cells (2µM, p<0.05). We further tested the activity of 6U against purified CML and normal CD34+CD38- stem/primitive progenitors and CD34+CD38+ committed progenitors with or without the BCR-ABL TKI Dasatinib (DAS). 6U treatment induced apoptosis of CML, but not normal, CD34+CD38- and CD34+CD38+ cells (Table). Combination of 6U with DAS (50nM) selectively enhanced apoptosis of CML compared to normal cells, including quiescent, slowly dividing CML LSC that are resistant to TKI-induced apoptosis (p≤0.01). Treatment with 6U alone or with DAS, significantly increased G1, and decreased S/G2/M phase of CML, but not in normal CD34+ cells, and reduced CFC growth from CML CD34+CD38+ cells (Table). CML, but not normal CD34+ cells, treated with 6U, with or without DAS, prior to transplant, showed significantly reduced engraftment in NSG mice, indicating selective inhibition of in vivo repopulating CML LSC (Table). Treatment with 6U was also effective in inducing apoptosis and inhibiting CFC growth in BC CML progenitor cells (Table). 6U treatment resulted in down-regulation of GSTPI1-1 and MCL-1 protein expression in CP and BC CML, but not in normal CD34+ cells. Interestingly 6U treatment also reduced BCR-ABL protein expression in CP and BC CML CD34+ cells. We conclude that CML CP and BC LSC express high levels of GSTP1-1 and anti-apoptotic proteins, which can be targeted by the novel EA derivative 6U through a new mechanism. Since 6U has significantly lesser effects on normal stem cells, it may offer a promising and innovative approach to selectively target CP and BC CML LSC in combination with TKI inhibitors. Abstract 4508. Table CML CP Normal CML BC Ctrl 6U DAS DAS+ 6U Ctrl 6U DAS DAS+ 6U Ctrl 6U DAS DAS+ 6U Apoptosis (normal, CP CML: CD34+CD38-; CML BC CD34+) 3.4± 0.9 15.9±6.7 9.4± 2.6 47.4±13.6 ** 3.3± 0.9 5.1± 1.0 1.6± 0.2 7.0± 1.2 * 3.4± 0.7 30±12.7 10.6±1.8 43.3±14.1 ** CFU-GM (normal, CP CML: CD34+CD38+; CML BC CD34+) 71.3± 7.8 7± 3.2 ** 21± 7.3 ** 5 ± 2.3 ** 121±19.3 102.7±6.2 134.3±15.9 103±5.1 288.5±89.4 26.5±11.3 *** 82.7±33.1 ** 8 ± 3.6 *** NSG engraftment (CD34+) 1.8± 0.3 0.4± 0.1 *** 0.8± 0.3 ** 0.4± 0.04 *** 68.2± 4.9 61± 2.2 68.1± 2.9 64.2± 3.9 Data shown are mean ± SEM of 3-6 samples. Significance, compared to controls. *p≤0.05,**p≤0.01, ***p≤0.001 Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 897-897
Author(s):  
Rüdiger Hehlmann ◽  
Michael Lauseker ◽  
Susanne Saussele ◽  
Markus Pfirrmann ◽  
Stefan W. Krause ◽  
...  

Abstract Background Chronic myeloid leukemia (CML)-study IV was designed to explore whether treatment with imatinib (IM) at 400mg/day (n=400) could be optimized by doubling the dose (n=420), adding IFN (n=430) or cytarabine (n=158) or using IM after IFN-failure (n=128). Methods From July 2002 to March 2012, 1551 newly diagnosed patients in chronic phase were randomized into a 5-arm study. The study was powered to detect a survival difference of 5% at 5 years. The impact of patients' and disease factors on survival was prospectively analyzed. At the time of evaluation, at least 62% of patients still received imatinib, 26.2% were switched to 2nd generation tyrosine kinase inhibitors. Results After a median observation time of 9.5 years, 10-year overall survival was 82%, 10-year progression-free survival 80% and 10-year relative survival 92%. In spite of a faster response with IM800mg, the survival difference between IM400mg and IM800mg was only 3% at 5 years. In a multivariate analysis, the influence on survival of risk-group, major-route chromosomal aberrations, comorbidities, smoking and treatment center (academic vs. other) was significant in contrast to any form of initial treatment optimization. Patients that reached the response milestones 3, 6 and 12 months, had a significant survival advantage of about 6% after 10 years regardless of therapy. The progression probability to blast crisis was 5.8%. Blast crisis was proceeded by high-risk additional chromosomal aberrations. Conclusions For responders, monotherapy with IM400mg provides a close to normal life expectancy independent of the time to response. Survival is more determined by patients' and disease factors than by initial treatment selection. Although improvements are also needed for refractory disease and blast crisis, more life-time can currently be gained by carefully addressing non-CML determinants of survival. Disclosures Hehlmann: Novartis: Research Funding; BMS: Consultancy. Saussele: Pfizer: Honoraria; Incyte: Honoraria; Novartis: Honoraria, Research Funding; BMS: Honoraria, Research Funding. Pfirrmann: BMS: Honoraria; Novartis: Honoraria. Krause: Novartis: Honoraria. Baerlocher: Novartis: Honoraria; BMS: Honoraria; Pfizer: Honoraria. Bruemmendorf: Novartis: Research Funding. Müller: Novartis: Honoraria, Research Funding; BMS: Honoraria, Research Funding; Ariad: Honoraria, Research Funding; Pfizer: Honoraria, Research Funding. Jeromin: MLL Munich Leukemia Laboratory: Employment. Hänel: Roche: Honoraria; Novartis: Honoraria. Burchert: BMS: Honoraria. Waller: Mylan: Consultancy, Honoraria. Mayer: Eisai: Research Funding; Novartis: Research Funding. Link: Novartis: Honoraria. Scheid: Novartis: Honoraria. Schafhausen: Novartis: Honoraria; BMS: Honoraria; Pfizer: Honoraria; Ariad: Honoraria. Hochhaus: Incyte: Research Funding; MSD: Research Funding; Pfizer: Research Funding; Novartis: Research Funding; BMS: Research Funding; ARIAD: Research Funding.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 739-739 ◽  
Author(s):  
Mhairi Copland ◽  
Linda Richmond ◽  
Ashley Hamilton ◽  
Elaine K. Allan ◽  
Junia V. Melo ◽  
...  

Abstract Recently, we have shown that BMS-214662, a cytotoxic farnesyltransferase inhibitor (FTI), targets primitive progenitor cells (PPC) in chronic myeloid leukaemia (CML) [Copland et al, Blood2005;106:204a]. These PPC are believed to be responsible for the molecular persistence which occurs following treatment with imatinib mesylate (IM) in CML. We have also shown that neither dasatinib (BMS-354825) nor nilotinib (AMN107) targets these PPC in vitro [Copland et al, Blood2006;107:4532; Jorgensen et al, Blood2005;106:314a]. To further investigate the efficacy of BMS-214662 against CML stem cells we performed long term culture-initiating cell (LTC-IC) assays with both chronic phase CML and normal CD34+ progenitors to assess drug selectivity. The CD34+ cells were treated for 72 hours under the following conditions no drug control, IM 5μM, dasatinib 150nM, BMS-214662 250nM, IM + BMS-214662, dasatinib + BMS-214662 before LTC-IC assay. Compared to the no drug control, CD34+ CML cells showed increased colony formation in the IM and dasatinib arms (181 and 178% respectively) indicating that, by inhibiting proliferation, these drugs exert a protective effect on CML PPC. The addition of BMS-214662 to either IM or dasatinib significantly reduced the number of colonies compared to either agent alone (P=0.045 and P=0.029 respectively). The BMS-214662 containing arms showed a dramatic reduction in total colony numbers to &lt; 2.5% of the no drug control (P=0.038). In 2/3 experiments, FISH showed that, after treatment with BMS-214662, the majority of cells from residual colonies were BCR-ABL negative. In addition, BMS-214662 was less toxic to normal LTC-IC from healthy donors. Using CFSE-based flow cytometry to track cell division, we assessed BMS-214662 alone and in combination with IM or dasatinib in blast crisis (BC) CML. BMS-214662 reduced the number of PPC compared to the no drug control and acted synergistically with IM or dasatinib to reduce the total number of viable cells and PPC in BC CML. To determine if the effects of BMS-214662 in CML were due to inhibition of Ras, we used the CFSE method, caspase-3 activity to measure apoptosis and Ki-67/7AAD cell cycle analysis to assess quiescence to compare BMS-214662 with another FTI with equivalent Ras inhibition, BMS-225975. As previously, after 6 days culture, not only had BMS-214662 significantly reduced the number of CML PPC compared to the no drug control (P=0.018), but also compared to BMS-225975 containing arms (P=0.024). There was no significant difference between the no drug control and BMS-225975 arms. Caspase-3 activity was highest in the BMS-214662-containing arms. After 72 hours, Ki-67/7AAD analysis showed reduced G0 cells in the BMS-214662 arms. The disparate effects of BMS-214662 and BMS-225975 suggest that the efficacy of BMS-214662 in CML is not via inhibition of Ras. We then determined the efficacy of BMS-214662 in Ba/F3 cells expressing different BCR-ABL kinase mutations (WT BCR-ABL, T315I, M351T and H396P) using viable cell counts and 3H thymidine proliferation assays. BMS-214662 was equipotent in both WT BCR-ABL and mutant BCR-ABL kinase expressing cells. These results provide further evidence that BMS-214662 selectively targets CML stem cells, acts in synergy with IM or dasatinib and may prove useful in the management of IM-resistant and BC CML.


Sign in / Sign up

Export Citation Format

Share Document