Novel Multistage Nanoparticle Drug Delivery to Ablate Leukemia Stem Cells in Their Niche.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2631-2631
Author(s):  
Hongliang Zong ◽  
Siddhartha Sen ◽  
Guodong Zhang ◽  
David G Gorenstein ◽  
Xuewu Liu ◽  
...  

Abstract Abstract 2631 Most patients with acute myelogenous leukemia (AML) die from their disease. Although up to 75% of AML patients achieve remission after initial induction therapy, most of them will relapse and it has been proposed that relapse is the result of ineffective ablation of leukemia stem cells (LSCs) by chemotherapy. Elevated levels of phenotypically defined LSCs at diagnosis are predictive of minimal residual disease (MRD) which, in turn, predicts leukemic relapse, even after myeloablative stem cell transplantation. Therefore, in order to improve AML therapy, it is imperative to identify therapeutic strategies that effectively eliminate LSCs and we hypothesize that effective novel therapeutics in AML must be able to penetrate the protective environment of the BM niche. Thus, we investigated the use of nanotechnology as a “Trojan horse” to deliver anti-LSC drugs to the bone marrow niche. We have previously demonstrated that the plant derived compound parthenolide (PTL), which have sub-optimal bioavilability, can effectively ablate LSCs in vitro. We sought to encapsulate PTL in a novel multi-stage delivery vector system (MSV) comprised of two delivery carriers: (1) degradable porous silicon (pSi) and (2) nanoparticles containing drug of interest. To optimize specific delivery to the BM, we conjugated E-selectin thioaptamer ligand (ESTA-1) to the surface of the particles. First, we performed feasibility studies of the system in AML-xenotransplanted mice. In order to demonstrate the delivery of MSV nanoparticles to the BM, we encapsulated Alexa Fluor 555-conjugated scramble-siRNA as a control. Nanoparticles were injected into established primary-AML xenotransplants. We found that 19.5% of the human cells from were positive for Alexa Fluor-555, thus demonstrating effective delivery to the BM of the xenotransplanted mice. Notably, we confirmed, using a single i.v. injection of MSV-PTL nanoparticles, that active PTL was delivered to the BM, were we observed a 4-fold decrease in viable human cells compared to controls. To assess the in vivo efficacy of the MSV nanoparticles, established primary AML-xenotransplants were treated with either MSV-PTL, MSV-empty, PTL loaded micelles (third stage component only) or PBS. MSV-PTL treated mice demonstrated significantly decreased tumor burden (61.9 % human AML cells) compared to controls (75.8% PBS, 75.6% MSV-empty and 72.9 % micelle-PTL; p<0.05). To evaluate the anti-LSC activity of PTL, secondary transplants with equal numbers of human AML cells were performed. There was a 3.6 fold reduction in AML engraftment in the secondary xenotransplants for MSV-PTL treatment compared to MSV-empty controls (p<0.05), demonstrating that LSCs were targeted by MSV-PTL. Inhibition of NF-κB and activation of γH2AX, two intracellular events triggered specifically by PTL, were consistently identified in the human AML cells obtained from the BM of MSV-PTL treated mice. Also, MSV-PTL enhanced ablation of AML cells not eliminated by ara-C in vivo. Secondary transplants further confirmed that treatment with MSV-PTL after ara-C treatment resulted in a significant decrease of engraftment of human AML cells, suggesting that PTL can eradicate ara-C-resistant LSCs. Taken together, our data show that encapsulation of potent anti-LSC agents, such as PTL, into MSV effectively protects and delivers active drug to the BM of xenotransplanted mice and targets both AML blasts and LSCs. MSV-mediated drug delivery provides a novel method to deliver promising anti-LSC compounds, including those with poor bioavilability, to the BM niche, where they can act directly on LSCs. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2168-2168
Author(s):  
Takayuki Ikezoe ◽  
Chie Nishioka ◽  
Jing Yang ◽  
Satoshi Serada ◽  
Tetsuji Naka ◽  
...  

Abstract Abstract 2168 To identify molecular targets in leukemia stem cells (LSCs), this study compared the protein expression profile of freshly isolated LSCs (CD34+/CD38- compartment) with that of non-LSC (CD34+/CD38+ compartment) counterparts from individuals with acute myelogenous leukemia (AML) using isobaric tags for relative and absolute quantitation (iTRAQ). A total of 98 proteins were overexpressed, while six proteins were underexpressed in LSCs compared with their non-LSC counterparts. Proteins overexpressed in LSCs included a number of proteins involved in DNA repair, cell cycle arrest, gland differentiation, anti-apoptosis, adhesion, and drug resistance. Aberrant expression of CD82, a family of adhesion molecules, in LSCs was noted in additional clinical samples (n=6) by flow cytometry. In addition, we found that imatinib-resistant chronic eosinophilic leukemi EOL-1R cells expressed a greater amount of CD82 and remained in a dormant state compared to the parental EOL-1 cells. Interestingly, down-regulation of CD82 in EOL-1R cells by a small interfering RNA stimulated their migration capacity, as assessed by the transwell assay. These observations suggested that the aberrant expression of CD82 probably played a role in adhesion of hematopoietic cells to bone marrow microenvironment. Targeting CD82 could detach LSCs from bone marrow niche and sensitized these cells to anti-leukemia agents. 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.


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

Leukemia ◽  
2015 ◽  
Vol 30 (7) ◽  
pp. 1582-1586 ◽  
Author(s):  
H Zong ◽  
S Sen ◽  
G Zhang ◽  
C Mu ◽  
Z F Albayati ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3153-3153 ◽  
Author(s):  
Ronan Quere ◽  
Göran Karlsson ◽  
Falk Hertwig ◽  
Marianne Rissler ◽  
Beata Lindqvist ◽  
...  

Abstract Abstract 3153 Leukemia stem cells (LSCs) are capable of limitless self-renewal and are responsible for the maintenance of leukemia. Since elimination of LSCs will be of therapeutic benefit, it is important to identify regulatory pathways that control their development. We studied LSCs in a Smad4-/- mouse model of acute myelogenous leukemia (AML) induced either by the HOXA9 gene or by the fusion oncogene NUP98-HOXA9. We first serially replated hematopoietic colonies from BM cells transduced with these oncogenes to analyze if loss of SMAD4 can increase immortalization of HSPCs. We observed that the immortalizing function of HOXA9 and NUP98-HOXA9 was increased in Smad4-/- HSPCs. The enhanced immortalization of SMAD4 deficient cells should be related to their incapacity to activate TGFβ signaling. However when we administrated a TGFβ receptor kinase inhibitor, the immortalization of Smad4-/- cells was still increased, compared to TGFb receptor-blocked Wt cells. Thereby, we conclude that the activation of the canonical TGFb signaling pathway is not required for the growth-reducing effect of HOXA9/NUP98-HOXA9 transduced progenitors mediated by SMAD4. We discovered that HOXA9/SMAD4 complexes accumulate in the cytoplasm of normal HSPCs transduced with HOXA9 or NUP98-HOXA9. In contrast there is no cytoplasmic accumulation of HOXA9 in SMAD4-/- HSPCs and as a consequence increased levels of HOXA9 accumulate in the nucleus leading to increased immortalization of HSPCs. Next, we investigated whether Smad4-/- HSPCs might exhibit increased frequency of BM transformation to induce leukemia in mice. Smad4 deficiency induces expansion of primitive LSK hematopoietic cells expressing HOXA9 or NUP98-HOXA9 in vivo, increases the frequency of leukemia stem cells and bone marrow transformation in mice. Accordingly, a larger fraction of mice transplanted with Smad4-/- transduced-HSPCs succumbed more rapidly to AML. Next, we developed an approach to activate the TGFβ pathway by restoring the subcellular distribution of the endogenous SMAD4 protein accumulated in the cytoplasm of HSPCs transduced with HOXA9 or NUP98-HOXA9. To this end, we first identified the best competitor by generating different constructs that enable expression of diverse portions of the MH1 domain of SMAD4 that bind HOXA9 (Wang et al., EMBO, 2007). All the retroviral vectors (RFP+) were tested by co-expression together with the NUP98-HOXA9 oncogene (GFP+). Co-transduced GFP+ RFP+ cells were sorted by FACS to assess the effect of MH1 overexpression by CFC assay. Interestingly, one specific portion of SMAD4 (MH1-c encoding a peptide of 20aa) dramatically reduced immortalization capacity of NUP98-HOXA9. The insensitivity of Smad4-/- cells shows notably that the specificity of this effect is SMAD4 dependent. Compared to the other truncated MH1 portions tested, the increased effect of MH1-c was correlated to its capacity to bind HOXA9 and to induce apoptosis. To understand the mechanism behind the effect of MH1-c, we looked at the localization of SMAD4 by immunostaining and confocal microscopy. Expression of MH1-c was observed to induce a robust nuclear translocation of the SMAD4 protein, while SMAD4 remained accumulated in the cytoplasm of cells transduced with the empty control vector (MH1-e). This clearly shows that MH1-c disrupts the physical interaction between HOXA9 and SMAD4. The SMAD4 subcellular distribution pattern was altered and the nuclear translocation clearly activated the TGFβ pathway and apoptosis of cells. Next, we asked whether MH1-c might affect the development of leukemia in vivo. When HSPCs were co-transduced with HOXA9 and the empty control vector and transplanted into the tail vein of lethally irradiated recipient mice, the transduced HSPCs developed AML. In contrast, HSPCs co-transduced with HOXA9 and MH1-c remained healthy over one year, without any detectable GFP+ RFP+ cells in BM when they were sacrificed. Taken together, these findings show that active SMAD4 that can freely translocate to the nucleus of HSPCs in vivo can prevent or profoundly delay the initiation of malignant transformation. In conclusion, targeting the association between SMAD4 and HOXA9 prevents/reduces immortalization in vitro and transformation to leukemia in vivo. These findings demonstrate how activation of a negative regulatory pathway that prevents leukemogenesis may be a feasible approach to improve leukemia treatment. Disclosures: Slovak: PerkinElmer: Employment.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5257-5257
Author(s):  
Wenbin Zhong ◽  
Vesa Olkkonen ◽  
Xu Bing ◽  
Biying Zhu ◽  
Guoping Li ◽  
...  

Abstract Acute myelogenous leukemia (AML) is one of the deadliest hematological malignancies and there is at present no efficient strategy to defeat it. New detailed insight into AML leukemia stem cells (LSCs) survival will facilitate the identification of targets for the development of new therapeutic approaches. Recent work has provided evidence that LSCs are defective in their ability to employ glycolysis, but are highly reliant on oxidative phosphorylation, and the maintenance of mitochondrial function is essential for LSCs survival. It is increasingly clear that Ca2+ released constitutively from endoplasmic reticulum (ER) is taken up by mitochondria to sustain optimal bioenergetics and cell survival. Here we report three striking findings: 1) oxysterol-binding protein (OSBP)-related protein 4 (ORP4L) is expressed in LSCs but not in normal hematopoietic stem cells (HSCs). 2) ORP4L is essential for LSC bioenergetics; It forms a complex with PLCβ3 and IP3 receptor 1 (ITPR1) to control Ca2+ release from the ER and subsequent cytosolic and mitochondrial parallel Ca2+ spike oscillations that sustain pyruvate dehydrogenase (PDH) activation and oxidative phosphorylation. 3) ORP4L inhibition eradicates LSCs in vitro and in vivo through impairment of Ca2+-dependent bioenergetics. These results suggest a novel role of ORP4L in governing Ca2+ release to sustain mitochondrial function and survival of LSCs and identify ORP4L as a putative new oncoprotein and therapeutic target for LSCs elimination. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 57-57
Author(s):  
Vincenzo Giambra ◽  
Catherine E Jenkins ◽  
Sonya H Lam ◽  
Catherine Hoofd ◽  
Miriam Belmonte ◽  
...  

Abstract Prior work has shown that NOTCH1 is a prominent oncogene in T-cell acute lymphoblastic leukemia (T-ALL) with activating NOTCH1 mutations occurring in over 50% of cases (Weng et al, Science 2004) and loss-of-function mutations in its negative regulator FBXW7 occurring in 8-15% of cases (O’Neil et al, J Exp Med 2007; Thompson et al, J Exp Med 2007). Subsequent work has shown that continued Notch signaling is required for maintenance of T-ALL leukemia stem cells (Armstrong et al, Blood 2009; Tatarek et al, Blood 2011; Giambra et al, Nat Med 2012). Several lines of evidence have substantiated genetic interactions between the Notch and Wnt signaling pathways in various contexts, and Wnt signaling has been shown to play important roles in hematopoietic stem cell biology and also in hematopoietic cancers such as acute myelogenous leukemia (AML) and chronic myelogenous leukemia (CML) (reviewed in Luis et al, Leukemia 2012). To address what role if any Wnt signaling may play in T-ALL, we generated primary murine leukemias by viral transduction of bone marrow progenitors with activated NOTCH1, then delivered a fluorescent Wnt reporter construct (7TGP; Fuerer & Nusse, PLoS ONE 2010) by lentiviral transduction, and retransplanted the leukemias to interrogate Wnt signaling activity in vivo. We report here that active Wnt signaling is restricted to minor subpopulations within bulk T-ALL tumors, and that these Wnt-active subsets are highly enriched for leukemia-initiating cell (LIC) activity. Moreover, using Ctnnb1loxP/loxP animals we show that inducible Cre-mediated deletion of β-catenin or enforced expression of a dominant-negative TCF construct severely compromises LIC activity. We also show that β-catenin levels are upregulated by hypoxia through Hif1a stabilization, and that deletion of Hif1a also severely compromises LIC activity. Interestingly, Wnt-active subsets are distributed diffusely throughout the marrow interstitial space suggesting that tumor infiltration induces formation of local hypoxic niches as opposed to taking up residence in pre-existing anatomic compartments with low oxygen tensions. Taken together, these results suggest a model in which hypoxic niches in vivo facilitate Hif1a-dependent accumulation of β-catenin which drives Wnt signaling and self-renewal of leukemia stem cells. Finally, we show using patient-derived xenografts that antagonism of Hif1a or Wnt signaling also compromises human LIC activity, suggesting that pharmacologic targeting of these pathways could have therapeutic application in patients with T-ALL. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (7) ◽  
pp. 2578-2585 ◽  
Author(s):  
Sarah J. Neering ◽  
Timothy Bushnell ◽  
Selcuk Sozer ◽  
John Ashton ◽  
Randall M. Rossi ◽  
...  

Myeloid leukemia arises from leukemia stem cells (LSCs), which are resistant to standard chemotherapy agents and likely to be a major cause of drug-resistant disease and relapse. To investigate the in vivo properties of LSCs, we developed a mouse model in which the biologic features of human LSCs are closely mimicked. Primitive normal hematopoietic cells were modified to express the BCR/ABL and Nup98/HoxA9 translocation products, and a distinct LSC population, with the aberrant immunophenotype of lineage−, Kit+/−, Flt3+, Sca+, CD34+, and CD150−, was identified. In vivo studies were then performed to assess the response of LSCs to therapeutic insult. Treatment of animals with the ABL kinase inhibitor imatinib mesylate induced specific modulation of blasts and progenitor cells but not stem- cell populations, thereby recapitulating events inferred to occur in human chronic myelogenous leukemia (CML) patients. In addition, challenge of leukemic mice with total body irradiation was selectively toxic to normal hematopoietic stem cells (HSCs), suggesting that LSCs are resistant to apoptosis and/or senescence in vivo. Taken together, the system provides a powerful means by which the in vivo behavior of LSCs versus HSCs can be characterized and candidate treatment regimens can be optimized for maximal specificity toward primitive leukemia cells.


Blood ◽  
2011 ◽  
Vol 118 (26) ◽  
pp. 6909-6919 ◽  
Author(s):  
Shailaja Hegde ◽  
Naveen Kaushal ◽  
Kodihalli C. Ravindra ◽  
Christopher Chiaro ◽  
Kelsey T. Hafer ◽  
...  

Abstract Targeting cancer stem cells is of paramount importance in successfully preventing cancer relapse. Recently, in silico screening of public gene-expression datasets identified cyclooxygenase-derived cyclopentenone prostaglandins (CyPGs) as likely agents to target malignant stem cells. We show here that Δ12-PGJ3, a novel and naturally produced CyPG from the dietary fish-oil ω-3 polyunsaturated fatty acid eicosapentaenoic acid (EPA; 20:5) alleviates the development of leukemia in 2 well-studied murine models of leukemia. IP administration of Δ12-PGJ3 to mice infected with Friend erythroleukemia virus or those expressing the chronic myelogenous leukemia oncoprotein BCR-ABL in the hematopoietic stem cell pool completely restored normal hematologic parameters, splenic histology, and enhanced survival. More importantly, Δ12-PGJ3 selectively targeted leukemia stem cells (LSCs) for apoptosis in the spleen and BM. This treatment completely eradicated LSCs in vivo, as demonstrated by the inability of donor cells from treated mice to cause leukemia in secondary transplantations. Given the potency of ω-3 polyunsaturated fatty acid–derived CyPGs and the well-known refractoriness of LSCs to currently used clinical agents, Δ12-PGJ3 may represent a new chemotherapeutic for leukemia that targets LSCs.


2016 ◽  
Vol 13 (2) ◽  
pp. 248-259 ◽  
Author(s):  
Hong-Sheng Zhou ◽  
Hong-Sheng Zhou ◽  
Bing Z. Carter ◽  
Michael Andreeff ◽  
Bing Z. Carter ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document