scholarly journals Effective Targeting of Quiescent Chronic Myelogenous Leukemia Stem Cells by Histone Deacetylase Inhibitors in Combination with Imatinib Mesylate

Cancer Cell ◽  
2010 ◽  
Vol 17 (5) ◽  
pp. 427-442 ◽  
Author(s):  
Bin Zhang ◽  
Adam C. Strauss ◽  
Su Chu ◽  
Min Li ◽  
Yinwei Ho ◽  
...  
Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 190-190
Author(s):  
Bin Zhang ◽  
Adam Campbell Strauss ◽  
Su Chu ◽  
Yin Wei Ho ◽  
David S. Snyder ◽  
...  

Abstract Abstract 190 The BCR-ABL tyrosine kinase inhibitor imatinib mesylate (IM) is highly effective in inducing remissions and improving survival in CML patients but fails to eliminate leukemia stem cells, which remain a potential source of relapse. Quiescent leukemia stem cells resist apoptosis following BCR-ABL kinase inhibition, and other strategies are required for their elimination. Histone deacetylase inhibitors (HDACi) have shown promise in the treatment of several cancers, and it is of particular interest that reports suggest that they are also capable of inducing apoptosis in non-proliferating cells. It is known that the potent pan-HDACi LAQ824 (LAQ) and LBH589 (LBH) can induce apoptosis in CML cell lines and blast crisis CML cells. However, the effect of HDACi on quiescent leukemia stem cells from chronic phase CML patients is not known. Here we investigated the effects of LAQ and LBH, alone and in combination with IM, on CML stem and progenitor cells. CML and normal CD34+ cells were cultured with LAQ (10–100nM) or LBH (25–50nM) alone, IM (1mM) alone, and LAQ or LBH with IM for 96 hours. HDACi treatment effectively enhanced Histone H3 and H4 acetylation in CML CD34+ cells. HDACi treatment by itself induced less apoptosis in CML compared to normal CD34+CD38- primitive and CD34+CD38+ committed progenitors, but was highly effective in inducing apoptosis in CML progenitors when combined with IM. In addition, the combination induced significantly higher apoptosis in CML compared with normal CD34+ cells, and unlike IM, also induced apoptosis in non-dividing cells. Combination treatment also inhibited the proliferation of CML progenitor cells as measured by CFSE and growth of CML CFC in methylcellulose progenitor assays. Treatment of CML CD34+ cells with IM resulted in modest reduction in levels of engraftment in NSG mice. In contrast treatment with LAQ824 plus IM resulted in abrogation of engraftment of BCR-ABL+ CML cells (p<0.001). Significantly less inhibition of normal compared to CML cell engraftment was seen following LAQ824 and IM treatment (p=0.006). We used a transgenic Scl-tTa-BCR-ABL mouse model to investigate the effect of HDACi treatment on CML stem cells in vivo. SCLtTA/BCR-ABL transgenic mice were crossed with GFP transgenic mice to allow tracking of transplanted cells. Recipient mice developed CML-like disease 3–4 weeks after transplantation. Mice were treated with IM (200mg/kg daily by gavage), LBH (30 mg/kg IP 3 times a week), LBH with IM, or vehicle alone (control) for 4 weeks. LBH combined with IM resulted in greater reduction in WBC, neutrophils and GFP+ cells than LBH or IM alone. Significantly increased apoptosis and a profound reduction of Lin- Sca-1+ Kit+ (LSK) stem cells were seen in mice treated with IM plus LBH (p<0.001) but not IM or LBH alone. LBH plus IM treatment did not significantly inhibit LSK cells in normal mice. BCR-ABL-transgenic mice treated with IM plus LBH demonstrated prolonged leukemia-free survival after discontinuing treatment compared with mice treated with IM or LBH alone (p<0.05). Combined IM and LAQ824 treatment resulted in marked inhibition of the anti-apoptotic protein MCL-1 in CML CD34+ cells (p<0.001). RNAi mediated inhibition of MCL-1 expression resulted in increased apoptosis in CML compared with normal CD34+ cells that was further enhanced by IM treatment. Our results indicate that treatment with HDACi plus IM effectively and selectively targets leukemia stem cells in CML, suggest a potential role for MCL-1 inhibition in HDACi induced apoptosis, and support ongoing clinical trials of LBH combined with IM to eliminate residual leukemia stem cells in IM-treated CML patients. Disclosures: Bhatia: Novartis: Consultancy.


2016 ◽  
Vol 23 (11) ◽  
pp. 2842-2855 ◽  
Author(s):  
Yanli Jin ◽  
Danian Nie ◽  
Juan Li ◽  
Xin Du ◽  
Yuhong Lu ◽  
...  

2016 ◽  
Vol 126 (10) ◽  
pp. 3961-3980 ◽  
Author(s):  
Yanli Jin ◽  
Jingfeng Zhou ◽  
Fang Xu ◽  
Bei Jin ◽  
Lijing Cui ◽  
...  

2012 ◽  
Vol 30 (15_suppl) ◽  
pp. e17022-e17022
Author(s):  
Eugene McPherson

e17022 Background: Histone deacetylase (HDAC) are enzymes involved remodeling chromatin and epigenetic regulation of gene expression. Histone deacetylase inhibitors (HDACI) can reverse aberrant epigenetic changes in CML stem cells. BCR-abl tyrosine kinase inhibitors IM is effective in inducing remission in CML-cp patients but residual leukemia stem cells (LSC) may give rise to resistance and relapse. Pan-HDACI may arrest this with downregulation of IL-6, and NFkb activation when adjuvant Se-Met is combined with IM. Methods: CML-cp , CML-bc cases were presented with evidence of overexpression of BCR-abl fusion protein reported previously in elderly patients. A 91 YOF with CML-cp , subclinical hypothyroidism with elevated IL-6, CRP, sIL-2R, VEGF, NFkb, and IGF-1 was treated for seven years with adjuvant Se-Met, IM and levothyroxine ith normalization, minmal toxicity and extended trending down of IL-6, CRP,sIL-2R. Results: After seven years of adjuvant therapy with Se-Met, IM, and Levothyroxine a significant reduction of IL-6, CRP of 9.8075 down from 30.783 mg/L. Subsequuent normalization of the other pro-inflammatory cytokines were normalized. Conclusions: HDACI activity expressed in synergism of Se-Met with IM and Levothyroxine may lower the apoptotic threshold in CML-cp stem cells decreasing BCR-abl expression and rescuing quiescent progenitor cells that may prolong remission and decrease disease relapse.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2181-2181 ◽  
Author(s):  
King-Pan Ng ◽  
Tuang Yeow Poh ◽  
Wen Tian Sun ◽  
Charles Chuah ◽  
S. Tiong Ong

Abstract Abstract 2181 Poster Board II-158 The use of Abl tyrosine kinase inhibitors (TKI), such as imatinib mesylate (IM), has seen a major advance in the control of chronic myelogenous leukemia (CML). However, since TKIs do not seem to eliminate CML stem cells, the use of TKIs may not represent a curative approach. Recent reports have shown that the bone marrow (BM) microenvironment, which facilitates the proliferation and renewal of resident hematopoietic stem cells (HSC), is hypoxic. The average BM O2 tension has been measured at 6-7% O2 in humans (physiologic hypoxia), including in patients with leukemia (Fiegl et al. Blood, 2009), while levels in the HSC niche are estimated to be even lower (<1% O2). In light of the above, it is likely that CML stem cells also reside in the HSC niche, and are similarly adapted for survival and self-renewal in this environment. Furthermore, although hypoxia is known to be associated with both radio- and chemo-resistance in solid tumors, its role in leukemia has has not been thoroughly investigated, particularly in the context of resistance to targeted therapies. Accordingly, we hypothesized that hypoxia protects CML progenitor cells from elimination by IM, a phenomenon which may contribute to disease persistence. To test the above, we examined if hypoxia modified the response of CML progenitor cells toward IM. Primary chronic phase (CP) CML samples “from either BM or peripheral blood (PB)” were obtained at the time of diagnosis from seven individuals with clinically-defined IM-sensitive (IM-S) or IM-resistant (IM-R) disease. Cells were then incubated with IM (0, 0.25, 1 or 5μM) under hypoxic (0.5% O2) or normoxic (21% O2) conditions for 96 hours. The treated cells were then harvested and plated in methylcellulose for 14 days (under 21% O2), after which the number of colony-forming cells (CFCs) were counted by two independent observers. In the absence of IM, four of the seven samples had a significant increase in CFCs (1.5-2.4 fold) when cultured in 0.5% O2 vs 21% O2, suggesting that primary CML CFCs may be better maintained in hypoxia. Furthermore, when treated with IM, six out of seven samples cultured under 0.5% O2 demonstrated dramatic increases in CFCs compared to those treated under 21% O2: a 2.3 to 9.0-fold increase at 1μM, and a 4.4 to 35.0-fold increase at 5μM IM. We also found that the protective effect of hypoxia was independent of the original source of the CFCs (BM or PB). In addition, we found that progenitors from both IM-S and IM-R patients were protected from IM under 0.5% O2, suggesting that hypoxia-induced protection is a general feature of CML progenitors. In order to establish a model for further study, we also tested if hypoxia elicits similar responses in four human CML cell lines (K562, AR230, LAMA84, and BV173). In contrast to primary CML cells, we found that hypoxia actually impaired the ability of all four cell lines to form colonies, and also did not confer protection from IM. These results show that CML cell lines have adapted to conditions of normoxia, and are thus inappropriate models to study the hypoxic response in CML. Additional data will be presented describing the mechanisms that may underlie the protective effect of hypoxia, as well as compounds that can counteract such effects. Ongoing experiments using the LT-CIC assay will also determine the response of primitive CML progenitors to growth under conditions of physiologic hypoxia vs normoxia. In conclusion, our results show that physiologic hypoxia protects CML progenitors from IM, and suggest that blocking of hypoxia-induced survival pathway(s) in CML progenitor cells may facilitate the elimination of residual CML progenitors. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 512-512 ◽  
Author(s):  
Bin Zhang ◽  
Yin Wei Ho ◽  
Tessa L. Holyoake ◽  
Ravi Bhatia

Abstract BCR-ABL tyrosine kinase inhibitors (TKI), although highly effective in inducing remission and improving survival in chronic myelogenous leukemia (CML) patients, fail to eliminate leukemia stem cells (LSC), which remain a potential source of relapse. Most CML patients need continued TKI treatment to prevent disease relapse, and new strategies to eliminate residual leukemia stem cells are required to enhance possibility of achieving treatment-free remission. In previous studies we have shown that increased several cytokines expressed by leukemia cells may provide a selective growth advantage to CML compared with normal long term hematopoietic stem cells (LTHSC) within the CML BM microenvironment. Studies evaluating the effects of individual factors indicated that exposure to Interleukin-1α/β (IL-1α/β) at concentrations similar to those observed in CML BM resulted in significantly increased growth of CML compared with normal LTHSC (Cancer Cell 2012, 21:577). Consistent with previous reports (PNAS 2010, 107:16280), we observed that expression of the IL-1 receptor-associated protein (IL-1RAP), an important IL-1 signaling component, was increased in primitive CML cells, potentially explaining enhanced IL-1 sensitivity. To further evaluate the role of microenvironmental IL-1 in maintenance of CML LTHSC, we used recombinant IL-1 receptor antagonist (IL-1RA) to block IL-1 receptor signaling. IL-1RA is clinically approved for the treatment of rheumatoid arthritis. Purified LTHSC (Lin-Sca-1+Kit+Flt3-CD150+CD48- cells) from the SCL-tTA/BCR-ABL inducible mouse model of CML (CD45.1) and from congenic FVBN mice (CD45.2) were mixed in a 1:1 ratio and cultured with CML BM plasma, with and without IL-1RA. Culture with CML BM plasma for 7 days results in significantly increased growth of CML compared to normal LTHSC. The ratio of CML to normal cells was significantly reduced in the presence of IL-1RA (2.5μg/ml) (3.6:1 without IL-1RA, 1.7:1 with IL-1RA, p=0.0002), indicating that inhibition of IL-1 signaling reduced the growth advantage of CML LTHSC cultured in CML BM plasma. We next investigated the effect of IL-1RA on CML hematopoiesis in vivo. BM cells from CML mice (CD45.1) were transplanted into congenic FVBN mice (CD45.2) to generate CML-like disease in recipient mice. Four weeks after transplantation mice were treated with Nilotinib (NIL, 50mg/kg/d, gavage), IL-1RA (150mg/kg/d s.c.), the combination of NIL and IL-1RA, or vehicle (control) for 3 weeks. Treatment with NIL plus IL-1RA resulted in significantly greater reduction in CD45.1+ CML cells in blood, and in CML LTHSC, MPP, CMP and GMP in BM, compared with NIL alone (CML LTHSC/2 femurs: control 738±122, NIL 486±94, IL-1RA 525±49, combination 360±33, P=0.01 combination vs. Nilotinib). Mice treated with NIL plus IL-1RA also showed significantly prolonged survival after completion of treatment compared to mice treated with NIL alone (median survival 6 days for NIL alone versus 45 days for combination, p=0.02). Following transplantation of BM cells from treated mice into 2nd recipients (CD45.2), significantly lower CML cell engraftment in BM and reduced development of leukemia was seen after transplantation of cells from mice treated with the combination compared with NIL or untreated controls (8 out of 8 mice developed leukemia for control, 6 out of 8 for NIL, 5 out of 8 for IL-1RA, 3 out of 8 for the combination). We also studied the effect of treatment with NIL (5μm), IL-1RA (5μg/ml), NIL+IL-1RA, or vehicle for 72 hours on human CML and normal CD34+CD38+ and CD34+CD38- cells cultured with CML BM conditioned medium (CM). The combination of NIL and IL-1RA significantly reduced CML CD34+CD38+ and CD34+CD38- cell growth compared to Nilotinib alone (CD38- cells: NIL 23.7±10.1%, combination 13.1±8.9% of control, p<0.05), cell division (measured by CFSE labeling) (CD38- proliferation index: NIL 3.3±1.0, combination 2.4±0.6, p=0.06) and CFC frequency in methylcellulose progenitor assays (CD38- cells: NIL 67±22 per 1000 cells, combination 39±26, p<0.05); and moderately increased apoptosis of CML CD34+CD38- cells. We conclude that inhibition of microenvironmental IL-1 signaling using IL-1RA significantly increases inhibition of self-renewing murine and human CML stem cells in combination with NIL. Our results support further evaluation of IL-1 inhibition as a strategy to enhance elimination of CML LSC in TKI-treated patients. Disclosures: Holyoake: Novartis: Membership on an entity’s Board of Directors or advisory committees; Bristol Myers Squibb: Membership on an entity’s Board of Directors or advisory committees; Ariad: Membership on an entity’s Board of Directors or advisory committees.


2016 ◽  
Vol 126 (3) ◽  
pp. 975-991 ◽  
Author(s):  
Bin Zhang ◽  
Ling Li ◽  
Yinwei Ho ◽  
Min Li ◽  
Guido Marcucci ◽  
...  

2018 ◽  
Vol 24 (4) ◽  
pp. 450-462 ◽  
Author(s):  
Bin Zhang ◽  
Le Xuan Truong Nguyen ◽  
Ling Li ◽  
Dandan Zhao ◽  
Bijender Kumar ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document