Bone marrow microenvironment confers imatinib resistance to chronic myelogenous leukemia and oroxylin A reverses the resistance by suppressing Stat3 pathway

2014 ◽  
Vol 89 (1) ◽  
pp. 121-136 ◽  
Author(s):  
X. Li ◽  
H. Miao ◽  
Y. Zhang ◽  
W. Li ◽  
Z. Li ◽  
...  
Blood ◽  
1995 ◽  
Vol 85 (12) ◽  
pp. 3636-3645 ◽  
Author(s):  
R Bhatia ◽  
PB McGlave ◽  
GW Dewald ◽  
BR Blazar ◽  
CM Verfaillie

The bone marrow microenvironment supports and regulates the proliferation and differentiation of hematopoietic cells. Dysregulated hematopoiesis in chronic myelogenous leukemia (CML) is caused, at least in part, by abnormalities in CML hematopoietic progenitors leading to altered interactions with the marrow microenvironment. The role of the microenvironment itself in CML has not been well characterized. We examined the capacity of CML stroma to support the growth of long-term culture-initiating cells (LTC-IC) obtained from normal and CML marrow. The growth of normal LTC-IC on CML stroma was significantly reduced compared with normal stroma. This did not appear to be related to abnormal production of soluble factors by CML stroma because normal LTC-IC grew equally well in Transwells above CML stroma as in Transwells above normal stroma. In addition, CML and normal stromal supernatants contained similar quantities of both growth-stimulatory (granulocyte colony-stimulating factor (CSF), interleukin-6, stem cell factor, granulocyte-macrophage CSF, and interleukin-1 beta) and growth-inhibitory cytokines (transforming growth factor-beta, macrophage inflammatory protein-1 alpha, and tumor necrosis factor-alpha). The relative proportion of different cell types in CML and normal stroma was similar. However, polymerase chain reaction and fluorescence in situ hybridization studies showed the presence of bcr-abl-positivo cells in CML stroma, which were CD14+ stromal macrophages. To assess the effect of these malignant macrophages on stromal function, CML and normal stromal cells were separated by fluorescence-activated cell sorting into stromal mesenchymal cell (CD14-) and macrophage (CD14+) populations. CML and normal CD14-cells supported the growth of normal LTC-IC equally well. However, the addition of CML macrophages to normal or CML CD14-mesenchymal cells resulted in impaired progenitor support. This finding indicates that the abnormal function of CML bone marrow stroma is related to the presence of malignant macrophages. In contrast to normal LTC-IC, the growth of CML LTC-IC on allogeneic CML stromal layers was not impaired and was significantly better than that of normal LTC-IC cocultured with the same CML stromal layers. These studies demonstrate that, in addition to abnormalities in CML progenitors themselves, abnormalities in the CML marrow microenvironment related to the presence of malignant stromal macrophages may contribute to the selective expansion of leukemic progenitors and suppression of normal hematopoiesis in CML.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1670-1670
Author(s):  
Daniela S. Krause ◽  
Keertik Fulzele ◽  
Andre Catic ◽  
Michael Hurley ◽  
Sanon Lezeau ◽  
...  

Abstract Abstract 1670 It is known that osteoblastic cells regulate the normal hematopoietic stem cell (HSC) niche and control its size. Parathyroid hormone (PTH) is an important regulator of osteoblasts and osteoclasts maintaining calcium homeostasis and, additionally, increasing HSC number in transplant recipients and protecting HSCs from repeated exposure to cytotoxic chemotherapy. We, therefore, hypothesized, that PTH-treatment may allow normal HSCs to outcompete leukemic stem cells (LSCs) in a murine model of chronic myelogenous leukemia. Mice with osteoblastic cell-specific constitutive activation of the receptor for PTH and PTH-related protein (Col1-caPPR mice) are characterized by activation of osteoblastic cells and increases in osteoclast and osteoblast number, trabecular bone, bone turnover and cortical porosity. Col1-caPPR mice have significantly prolonged survival and reduced leukemic mortality compared to wildtype (wt) littermates in a murine retroviral transduction/transplantation model of BCR-ABL1-induced CML-like disease (p=0.002) and B-cell acute lymphoblastic leukemia (B-ALL) (p=0.0004). However, a leukemogenic fusion transcription factor, MLL-AF9, known to cause acute myeloid leukemia in this model, led to more rapid death in the Col1-caPPR recipients compared with their wt counterparts (p<0.0001), indicating that the increased survival of Col1-caPPR recipients is specific for BCR-ABL1-induced leukemia. Continuous infusion of human PTH(1–34) into wt mice with BCR-ABL1-induced CML led to a statistically significant decrease in spleen weights and decreased bone marrow infiltration by BCR-ABL+ cells. Limiting dilution secondary transplantation of BM cells from saline- or PTH-treated primary animals with fully established CML into wt recipients revealed a 15-fold reduction of LSCs in a PTH-treated environment. Secondary mice who received BM from saline-treated donors had an overall survival that was 1/4 that of recipients of marrow from a PTH-treated BM microenvironment. Transforming growth factor beta-1 (TGFβ-1), whose largest and most concentrated tissue source is bone, was increased in the bones of Col1-caPPR mice. TGFβ-1 significantly decreased the in-vitro growth of the BCR-ABL+ cell line K562, but not the MLL-AF9+ cell line THP-1 suggesting that TGFβ-1, increased in the bone marrow microenvironment of Col1-caPPR mice, may be actively suppressing the growth of the BCR-ABL+ diseases, but not of MLL-AF9+ AML. Conversely, blockade of TGFβ-1, -2, and -3 by anti- TGFβ antibody treatment increased the incidence of CML in Col1-caPPR mice from 50% to 75%. Knockdown of TGF Receptor I in transplanted BCR-ABL+ BM in the CML model increased the percentage of BCR-ABL+ myeloid cells in peripheral blood in wt and, more strikingly, in Col1-caPPR recipient mice and increased the overall incidence of CML in Col1-caPPR mice. These results argue that reduction in TGFβ-1 signaling may rescue the CML phenotype in Col1-caPPR mice. In conclusion, these studies suggest that modulation of the bone marrow microenvironment by PTH reduces the frequency of LSCs in CML, possibly by suppression of LSCs via TGFβ-1. Consequently, a clinical trial on the combined use of imatinib and PTH in patients with CML has been initiated at our institution. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 937-937
Author(s):  
Daniela S. Krause ◽  
Sanon Lezeau ◽  
Michael Hurley ◽  
Ernestina Schipani ◽  
David T. Scadden

Abstract Abstract 937 The role of the bone marrow microenvironment, and in particular of osteoblastic cells, for normal hematopoiesis has recently been described. However, the role of elements of the bone marrow microenvironment on the initiation, maintenance or progression of leukemia is less clear. To test the influence of activated osteoblasts on the progression of chronic myelogenous leukemia (CML) we used the well-described murine retroviral transduction/transplantation model of BCR-ABL1-induced CML-like disease and mice with osteoblastic cell-specific constitutive activation of the parathyroid-hormone (PTH) receptor (PPR mice) as recipients. Compared to wildtype (wt) littermate control mice PPR mice had significantly prolonged survival (p=0.002) and reduced leukemic mortality with splenic leukemopoiesis contributing to leukemic fatality. Analysis of distinct proviral integration sites in splenic tissue by Southern blotting showed no difference in engraftment of viral clones in wt versus PPR mice. Survival of PPR recipients in the BCR-ABL1-induced model of B-cell acute lymphoblastic leukemia using non-5-fluorouracil-treated donor bone marrow was also significantly prolonged compared to wt mice (p=0.0004). However, a leukemogenic allele known to result in acute myeloid leukemia (AML), MLL-AF9, led to more rapid death in the PPR recipients compared with their wt counterparts arguing that the prolongation of survival in the BCR-ABL1-induced diseases was oncogene-specific. In-vitro assays including the Whitlock-Witte assay to test the role of PPR stroma or a cobblestone colony formation assay in osteogenic medium revealed no difference in the growth of plated BCR-ABL1+ or BCR-ABL1- B-lymphoid progenitors or lin- c-kit+ Sca-1+ cells on wt versus PPR stroma, respectively. In vivo, treatment of mice with the phosphodiesterase inhibitor forskolin, which increases intracellular cyclic adenosine monophosphate (cAMP) levels, similar to increased signaling from the PTH receptor, did not lead to prolonged survival in the murine model of CML-like disease. Prior splenectomy of wt, as well as PPR recipients of BCR-ABL1-induced CML-like disease did not significantly prolong survival, but drastically reduced the efficiency of induction of CML-like disease with the great majority of wt and PPR animals succumbing to non-CML causes. Secondary transplantation of CML-like disease from bone marrow or spleen from a wt or PPR microenvironment revealed, firstly, less efficient induction of secondary disease in wt recipients of PPR bone marrow compared to wt bone marrow and, secondly, superiority of CML-induction in secondary recipients of spleen compared to bone marrow from a PPR microenvironment. In order to test for an osteoblast-extrinsic cause of the prolonged survival of PPR recipients in the CML-model and to test for the role of bone remodeling, wt and PPR recipients of BCR-ABL1-transduced bone marrow were treated with saline or osteoprotegerin, an inhibitor of osteoclast differentiation and proliferation. Surprisingly, 100% of PPR and wt control mice succumbed to CML-like disease sooner than the control mice treated with saline. Additionally, continuous infusion of human PTH(1-34) into wt mice with BCR-ABL1-induced CML-like disease by minipump led to prolonged survival compared to saline-treated animals arguing that PTH may be an intervention beneficial in human CML. This, to our knowledge, represents first evidence that modulation of the bone marrow microenvironment may improve the outcome in leukemia. Disclosures: Scadden: Fate Therapeutics: Consultancy, Equity Ownership, Patents & Royalties.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4517-4517
Author(s):  
Luke Truitt ◽  
Catherine Hutchinson ◽  
Karen Mochoruk ◽  
John F. DeCoteau ◽  
C. Ronald Geyer

Abstract Chronic myelogenous leukemia (CML) is maintained by a minor population of leukemic stem cells (LSCs) that exhibit innate resistance to tyrosine kinase inhibitors (TKIs) targeting BCR-ABL. Innate resistance can be induced by cytokines and growth factors secreted by bone marrow stromal cells (BMSFs) that protect CML-LSCs from TKIs, resulting in minimal residual disease. Developing therapies that can be combined with TKIs to eradicate TKI-insensitive CML-LSCs, is critical for disrupting innate TKI resistance and preventing disease relapse. Cancer cells balance reactive oxygen species (ROS) and antioxidants at higher than normal levels, which promotes their proliferation and survival, but also makes them susceptible to damage by ROS-generating agents. BCR-ABL expression increases cellular ROS levels, whereas, TKI inhibition of BCR–ABL reduces ROS. Furthermore, BMSFs, which are implicated in innate TKI resistance, can increase ROS levels in CML cells. Thus, we postulated that BMSF mediated increases in ROS might enhance triggering of ROS-mediated damage in TKI treated CML-LSCs by chaetocin, a mycotoxin with anticancer properties that imposes oxidative stress by inhibiting thioredoxin reductase-1. To investigate chaetocin effects on innate TKI resistance, we first compared its activity with imatinib against TonB210, a murine hematopoietic cell line with inducible BCR-ABL expression, in response to BMSFs. Imatinib did not affect the growth of BCR-ABL(-) TonB210 cells but suppressed BCR-ABL(+) Ton-B210 growth, and BMSFs protected against imatinib growth suppression. In contrast, chaetocin significantly suppressed the growth of both BCR-ABL(-) and BCR-ABL(+) TonB210 cells, and these effects were potentiated by BMSFs. We then compared the effects of chaetocin as a single agent, and in combination with imatinib, on the growth of CML-LSCs derived from an established murine retroviral transduction/transplantation model of CML blast crisis, in response to BMSFs. The presence of BMSFs reduced cytotoxicity and apoptosis induction by imatinib, but potentiated these effects in chaetocin treated CML-LSCs. Colony formation by CML-LSCs was significantly inhibited by treatment with either imatinib or chaetocin. However, BMSFs conferred significant protection from colony inhibition by imatinib, whereas, no colony formation was observed in cells exposed to chaetocin and BMSFs. Both BMSFs and chaetocin increased ROS in CML-LSCs and the addition of BMSFs and chaetocin resulted in significantly higher levels compared to chaetocin or BMSFs alone. Pretreatment of CML-LSCs with the anti-oxidant N-acetyl-cysteine blocked chaetocin cytotoxicity, even in the presence of BMSFs. Chaetocin effects on CML-LSC self-renewal in vivo were assessed by transplanting CML-LSCs into secondary recipients following in vitro exposure to chaetocin, in the presence or absence of BMSFs. Disease latency in mice transplanted with CML-LSCs following chaetocin treatment more than doubled compared to mice transplanted with untreated CML-LSCs or CML-LSCs exposed to BMSFs. Mice transplanted with CML-LSCs following chaetocin treatment in the presence of BMSFs had significantly extended survival time compared to mice transplanted with CML-LSCs treated with chaetocin alone. Our findings indicate that chaetocin activity against leukemia initiating cells is significantly enhanced in the presence of BMSFs and suggest that chaetocin may be effective as a co-drug to complement TKIs in CML treatment by disrupting the innate resistance of CML-LSCs through an ROS dependent mechanism. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document