scholarly journals CD123-Targeted Nano-Curcumin Molecule Enhances Cytotoxic Efficacy in Leukemic Stem Cells

Nanomaterials ◽  
2021 ◽  
Vol 11 (11) ◽  
pp. 2974
Author(s):  
Wariya Nirachonkul ◽  
Siriporn Ogonoki ◽  
Tarika Thumvijit ◽  
Supanimit Chiampanichayakul ◽  
Pawaret Panyajai ◽  
...  

Acute myeloblastic leukemia (AML) is a disease with a high rate of relapse and drug resistance due to the remaining leukemic stem cells (LSCs). Therefore, LSCs are specific targets for the treatment of leukemia. CD123 is specifically expressed on LSCs and performs as a specific marker. Curcumin is the main active compound of a natural product with low toxicity for humans. It has been reported to inhibit leukemic cell growth. However, curcumin is practically insoluble in water and has low bioavailability. In this study, we aimed to formulate curcumin nanoparticles and conjugate with the anti-CD123 to overcome the low water solubility and improve the targeting of LSCs. The cytotoxicity of both curcumin-loaded PLGA/poloxamer nanoparticles (Cur-NPs) and anti-CD123-curcumin-loaded PLGA/poloxamer nanoparticles (anti-CD123-Cur-NPs) were examined in KG-1a cells. The results showed that Cur-NPs and Cur-NPs-CD123 exhibited cytotoxic effects on KG-1a cells with the IC50 values of 74.20 ± 6.71 and 41.45 ± 5.49 µM, respectively. Moreover, anti-CD123-Cur-NPs induced higher apoptosis than Cur-NPs. The higher uptake of anti-CD123-Cur-NPs in KG-1a cells was confirmed by using flow cytometry. In conclusion, the anti-CD123-Cur-NPs formulation improved curcumin’s bioavailability and specific targeting of LSCs, suggesting that it is a promising drug delivery system for improving the therapeutic efficacy against AML.

Blood ◽  
2014 ◽  
Vol 123 (25) ◽  
pp. 3951-3962 ◽  
Author(s):  
Harald Herrmann ◽  
Irina Sadovnik ◽  
Sabine Cerny-Reiterer ◽  
Thomas Rülicke ◽  
Gabriele Stefanzl ◽  
...  

Key Points DPPIV (CD26) is a new specific marker of CML LSC that aids CML diagnostics and the measurement, characterization, and purification of LSC. DPPIV on CML LSC degrades SDF-1 and thereby promotes the niche-escape of LSC, which may contribute to extramedullary myeloproliferation in CML.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1637-1637
Author(s):  
Huiyu Li ◽  
Yi-Mei Du ◽  
Linlin Guo ◽  
Tiannan Guo ◽  
Shenghua Jie ◽  
...  

Abstract Background: Recent studies suggest that HERG K+ channel is an important regulator of non excitable cell proliferation and migration, and has been found in tumor cells including acute myeloid leukemia(AML), where HERG K+ channel is generally considered to be absent from their healthy counterparts. Bone marrow stromal cells constitutively secrete the stromal cell-derived factor-1 (SDF-1) which is a homeostatic chemokine that signals through CXCR4, SDF-1/CXCR4 axis and plays an important role in hematopoiesis development and leukemic cells migration. In this study, we investigated whether SDF-1-induced leukemic cell migration associated with HERG K+ channel. Methods: primary CD34+/CD38− leukemic stem cells (LSCs) were isolated by cell sorting using a FACS Vantage. Transwell was used to assess the effect of E-4031, a specific HERG K+ channel inhibitor, on leukemic cell migration, the lower chamber was filled with serum-free RPMI-1640 with 100ng/ml SDF-1. Flow cytometry was used to analyze the CXCR4 expression as well as phenotypical analysis of leukemia samples. HERG K+ channels were expressed in Xenopus oocyte by microinjection and the resulting currents were measured using the standard two microelectrode voltage clamp techniques. Results: numbers of HL-60 cells with and without E-4031 treatment migrated towards SDF-1 in the lower chamber were 1.58±0.98 ×104 and 3.47±0.81 ×104 respectively, indicating E-4031 significantly blocked the cell migration induced by SDF-1. The similar results were also observed in primary leukemic cells (n=7) and leukemic stem cells(n=3). From a holding potential of −80 mV varying potentials from −70 mV to +50 mV in 10 mV increments (2s) were applied to elicit activating currents. Each pulse was followed by a constant return pulse to −50 mV (2s) to evoke outward tail currents. 100 ng/ml SDF-1 increased HERG K+ current expressed in oocytes, for example, at +50 mV, HERG current increased about 30% (n=5). The HERG K+ current increase by SDF-1 might contribute to the mechanism of SDF-1 induced leukemic cell migration. There were no significant changes of CXCR4 expression on both HL-60 cells and primary leukemic cells regardless of untreated and treated with E-4031 for 24 hours (p>0.05), suggesting that the leukemic cell migration induced by SDF-1 were specifically associated with HERG K+ channel, not by regulating CXCR4 expression. Conclusion: the data showed that HERG K+ channel was essential for leukemic cell migration induced by SDF-1. SDF-1 enhanced herg current suggested that SDF-1 promotes leukemic cell migration. Blocking HERG K+ channel with specific inhibitor could decrease leukemic cell and leukemic stem cell migration caused by SDF-1. Prospectively, HERG K+ channel may be a potential therapeutic target with specific inhibitors in leukemia treatment.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2899-2899
Author(s):  
Thierry Guillaume ◽  
Virginie Dehame ◽  
Patrice Chevallier ◽  
Pierre Peterlin ◽  
Marc Grégoire ◽  
...  

Abstract Monocytic neoplasms comprise a heterogeneous group of hematologic malignancies including chronic myelomonocytic leukemia (CMML), juvenile myelomonocytic leukemia (JMML), acute myelomonocytic and monocytic leukemia (AML-M4 and AML-M5), and monocytic sarcoma. Monocytic or granulomonocytic hyperplasia is a finding frequently-if not invariably-shared by these different entities, as is a poor therapeutic outcome in the absence of hematopoietic stem cell transplantation. Cell surface molecules aberrantly expressed or overexpressed by leukemic cells represent potential disease-specific therapeutic targets. MUC1, a polymorphic type I high molecular weight glycoprotein represents such a molecule. MUC1 consists of an extracellular domain containing 20 to 125 tandem repeats of a 20 amino acid-long sequence, followed by a transmembrane domain and a short cytoplasmic tail leading to intracellular signaling. Cleavage of MUC1 yields two unequal chains: a large extracellular alpha subunit containing the tandem repeat array bound in a strong non-covalent interaction to a smaller beta subunit containing the transmembrane and cytoplasmic domains. Essentially all anti-MUC1 antibodies reported to date target the highly immunogenic tandem repeat of the MUC1 alpha chain. Because the alpha chain binds the cell-bound domains of MUC1 only intermittently in an 'on-and-off' manner, agents directed against the alpha chain will not effectively target MUC1+ cells. In contrast, the MUC1 SEA domain represents a stable structure fixed to the cell surface at all times. We therefore generated mAbs that specifically recognize the cell-bound MUC1 SEA domain. One of them, a partially humanized murine mAb termed DMB-5F3 was used to examine the expression of MUC1 on AML cells by flow cytometry. A series of twenty-two AML samples (blood-derived n=12; bone marrow-derived n=10; AML0=2, AML1=2, AML2=10, AML4=1, AML5=5, AML6=2) collected either at the time of diagnosis or at relapse were analysed for MUC1 expression by flow cytometry. A murine mammary tumor cell line stably transfected with human MUC1 DNA served as control. Blasts cells from 5 AML samples highly expressed MUC1, and significantly, all were of monocytic or myelomonocytic lineage (AML4=1, AML5=4). Leukemic stem cells (CD34pos or CD34neg linneg) from the MUC1+ AMLs were examined and likewise found to express MUC1. In addition, AML cell lines MV411, MOLM14, and SHI-1 derived from monocytic leukemic lineage clearly expressed cell surface MUC1, while non- monocytic leukemic cell lines U937, K562, and HL60 had little or no expression. Normal monocytes and monocytes derived from patients with activated monocytosis were also found to express MUC1. Based on these findings we examined MUC1 expression in a series of myelomonocytic leukemia (CMML and JMML). In fifteen CMML samples examined (type 1 n=11, type 2 n=4) (blood n=7, BM n=7) 92%-100% (median 99.7%) of CD14+CD56+ CMML cells bound mAb DMB-5F3 to cell-surface MUC1. CD14+CD16+CD56+ blast cells from 2 pts with JMML were also found to express MUC1 (between 64% and 71 % positive). Based on these findings we conclude that expression of MUC1 is restricted to monocytic and myelomonocytic leukemias and that MUC1 represents an effective target for leukemic immunotherapy. Significantly, anti-MUC1 mAb also targets monocytic leukemic stem cells, reinforcing its therapeutic potential. The fact that the anti-MUC1 antibody DMB-5F3 can enter cells and thereby ferry Ab-bound toxin opens the way for us to demonstrate leukemic cell killing with anti-MUC1 mAb-immunotoxin conjugates. Disclosures Moreau: Bristol-Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen-Cilag: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees; Millennium: Honoraria, Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4836-4836
Author(s):  
Gyeongsin Park ◽  
Michael Heuser ◽  
Tobias Berg ◽  
R. Keith Humphries

Abstract Abstract 4836 Engraftment is a process including homing to bone marrow, implantation and proliferation. Implantation implies interactions with specialized microenvironments, niches, in which hematopoietic stem cells (HSCs) live and are regulated. Studies have demonstrated the possibility that leukemic stem cells (LSCs) interact with niches in a similar manner to HSCs. We investigated whether HSCs and LSCs compete with each other in their engraftment. We employed a mouse transplantation assay with unmanipulatated bone marrow cells (BMCs) as a source of normal HSCs and LSCs generated by transduction of BMCs with Meningioma 1 (MN1), a potent oncogene causing myeloid leukemia in mice. In irradiated recipients (750 cGy), cotransplantation of leukemic cells (1×105) with various numbers of BMCs (1×105, 1×106 and 1×107) demonstrated that the engraftment level of leukemic cells is influenced by BMCs in a dose dependant manner (5.2%, 41.3% and 82.2% at 2-weeks; 52.3%, 69.5% and 86.9% at 4weeks; mice died before the 5 weeks bleeding, 94.9% and 97.5% at 5weeks, respectively). Cotransplantation of various numbers of leukemic cells (1×104, 1×105 and 1×106) with a fixed number of BMCs (1×106) demonstrated a similar pattern of leukemic engraftment (7.0%, 59.5% and 87.1% at 2weeks; 62.0%, 85.7% at 4 weeks, and mice died before the four week bleeding, respectively). To further elucidate the competition between HSCs and LSCs, we transplanted the cells at different time intervals. Transplantation of normal BMCs (1×106) 2 days prior to transplantation of LSCs (1×105) resulted in much reduced levels of leukemic engraftment compared to that seen in mice simultaneously transplanted (3.5% vs 59.5% at 2 weeks; 73.1% vs 85.76% at 4weeks). This competitive suppression of leukemic engraftment was further enhanced by transplanting larger numbers of normal BMCs (2×107) as little as 12 hours prior LSC transplantation (5×105) compared to simultaneous injection (0% vs 7.26% at 2weeks, 0.9% vs 35.3% at 3 weeks, and 6.0% vs 60.6% at 4 weeks). When BMCs (1×105) or leukemic cells (1×105) were transplanted at equal doses of 1×105 together with normal helper cells (1×106) the leukemic cells expanded 280-fold compared to only 7.3 fold for normal BMCs at 2 weeks (total cell count from two femurs and two tibias per 1×105 transplanted cells). Thus the competitive suppression of leukemic cell growth seen upon sequential transplantation of normal BMCs is not readily explained by enhanced kinetics of normal BMC growth but rather by competition at the level of initial engraftment. In conclusion, our data demonstrate that there is a competition between normal and leukemic cells during the engraftment process, suggesting niche competition of HSCs and LSCs. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (9) ◽  
pp. 3334-3344 ◽  
Author(s):  
Lin Wang ◽  
Heather O'Leary ◽  
James Fortney ◽  
Laura F. Gibson

Abstract Although leukemic stem cells (LSCs) show a symbiotic relationship with bone marrow microenvironmental niches, the mechanism by which the marrow microenvironment contributes to self-renewal and proliferation of LSCs remains elusive. In the present study, we identified a unique subpopulation of Philadelphia chromosome–positive (Ph+) acute lymphoblastic leukemia (ALL) cells coexpressing markers of endothelial cells (including VE-cadherin, PECAM-1, and Flk-1) and committed B-lineage progenitors. After long-term coculture with bone marrow stromal cells, tumor cells formed hematopoietic colonies and cords, expressed early stem- cell markers, and showed endothelial sprouting. Gene expression profiles of LSCs were altered in the presence of stromal cell contact. Stromal cell contact promoted leukemic cell VE-cadherin expression, stabilized β-catenin, and up-regulated Bcr-abl fusion gene expression. Our study indicates that these specific tumor cells are uniquely positioned to respond to microenvironment-derived self-renewing and proliferative cues. Ph+/VE-cadherin+ tumor subpopulation circumvents the requirement of exogenous Wnt signaling for self-renewal through stromal cell support of leukemic cell VE-cadherin expression and up-regulated Bcr-abl tyrosine kinase activity. These data suggest that strategies targeting signals in the marrow microenvironment that amplify the Bcr-abl/VE-cadherin/β-catenin axis may have utility in sensitizing drug-resistant leukemic stem cells.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5151-5151
Author(s):  
Lawrence G. Lum ◽  
Ewa Kubicka ◽  
Bliemeister T Edwin ◽  
Archana Thakur ◽  
Manley Huang

Background: Acute myeloid leukemia (AML) is a heterogeneous disease characterized by the accumulation of hematopoietic stem/progenitor cells that poses a significant therapeutic challenge. Recurrence of the disease is attributed to leukemia-initiating cells, also referred to as leukemic stem cells (LSCs), which are thought to be spared from chemotherapy and capable of reinitiating the disease. Thus, novel non-toxic therapeutic strategies to target and eradicate LSCs and AML blasts are urgently needed. Immunotherapy (IT) is an attractive strategy to improve outcomes for patients with AML, as it does not rely on the cytotoxic mechanisms employed by conventional chemotherapies. Methods: Bispecific antibody was produced by chemical hetero-conjugation of anti-CD33 monoclonal antibody (gemtuzumab ozogamicin [GO]) linked to calicheamicin to anti-CD3 monoclonal antibody (anti-CD3 x GO bispecific antibody, CD33Bi) or anti-CD3 and anti-CD123 (CD123Bi). We tested four AML cell lines (EOL-1, KG-1, NoMo-1 and TF-1) as targets for cytotoxicity by CD33Bi and CD123Bi armed ATC using flow cytometry-based cytotoxicity assays. Cytokines and chemokines released during CD33Bi or CD123Bi armed ATC mediated killing of targets were analyzed by multiplex luminex assay. A xenogeneic NOD/SCID/gamma chain KO (NSG) mouse model was used for the evaluation of in vivo activity of Bispecific antibody (50ng/million cells) Armed activated T cells (BATs). Engraftment of AML cells-KG1 was tracked by quantifying the human CD45+ AML cells using flow cytometry. Two patient samples were analyzed for blast and LSC population before and after incubation with CD123-BATs. Results: In a non-radioactive quantitative flow cytometry-based cytotoxicity assay Bispecific antibody (50ng/million cells) Armed activated T cells (BATs) show substantial killing by both CD33-BATs and CD123-BATs against AML cell lines (EOL1, KG1, NoMo1 and TF-1) at 1:1 and 2:1 E/T ratios ranging from 60-90% cytotoxicity and produced increased levels of Th1 cytokines (TNF-alpha, GM-CSF and IFN-gamma). The KG1 engrafted 6- to 8-week-old NSG mice (n=8 mice/group) injected IV with 20 x 106 CD33-BATs (3x/week for 4 weeks; 100ml/injection) was able to inhibit leukemic cell expansion significantly compared to 0.06 mg/kg gemtuzumab ozogamicin [GO] antibody (p<0.0006); likewise significant inhibition of leukemic cell expansion was observed with CD123-BATs compared to PBS control mice (p<0.009). One patient sample incubated with CD123-BATs showed significant reduction in blast after CD123-BATs treatment compared to before treatment (13.2% vs. 30%) or CD34+CD38- LSC after CD123-BATs treatment (5.2%) compared to before treatment (20%). Second patient sample also showed 50% reduction in blast population, no LSC were detected in this sample. Conclusions: ATC armed with CD33Bi or CD123Bi at 50 ng/106 cells showed comparable cytotoxicity directed at either high or low leukemic blast populations, able to inhibit leukemic cell expansion significantly compared to the control mice. This approach may provide a potent and non-toxic strategy to target leukemic blasts and leukemic stem cells. Disclosures Huang: TransTarget Inc.: Other: Co-Founder.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4394-4394
Author(s):  
Janna Neudenberger ◽  
Marc Hotfilder ◽  
Annegret Rosemann ◽  
Claudia Langebrake ◽  
Dirk Reinhardt ◽  
...  

Abstract Monitoring of minimal residual disease in childhood leukemia is widely used to determine treatment response and to detect persisting leukemic blasts for relapse prediction. In this context, it is increasingly recognized that only few leukemic blasts possess self-renewal capability and that only these self-renewing leukemic stem cells are able to initiate relapses. Therefore, the leukemic stem cells as the relapse-initiating cells should be the target cells of minimal residual disease detection. The human homologue of the rat chondroitin sulfate proteoglycan NG2 protein (detected by the monoclonal antibody 7.1) is aberrantly expressed in a high percentage of leukemias harboring 11q23 abnormalities and its expression has been proposed as a potential marker for MRD detection. To evaluate NG2 as a marker for leukemic stem cells, bone marrow or peripheral blood samples of 4 AML/t(9;11), 1 AML/t(10;11), 1 AML/t(?;11) and 2 ALL/t(4;11) patients with NG2-positive leukemia were analyzed by four color flow cytometry for NG2 expression on the cell surface of primitive cell populations. Immature cells were defined by the antigen profiles CD34+CD38− (CD33−) in AML and CD34+(CD117+)CD19− in ALL. AML stem cells are known to be CD34+CD38−. In the ALL patients, 32% resp. 81% of CD34+CD19− cells were leukemic by FISH analysis for MLL/AF4. For this rare event analysis, 5 x 10e6 cells were incubated with saturating amounts of the respective antibodies. Data acquisition and analysis was performed on a 2-laser FACSCalibur (BDIS). 1 x 10e6 events were acquired but only CD34+CD38− (in AML) or CD34+CD19− cells (in ALL) were stored (storage gate). Matched isotype controls were analyzed using the same storage gate to exclude amplification of unspecific events. In-between each acquisition, the cytometer was vigorously flushed to prevent carry-over of cells from previous acquisitions. In addition, 10.000 ungated events were acquired to compare the CD34+Lin− stem/progenitor cells with the dominant leukemic cell clone. In all eight patients with NG2-positve leukemia, cells with a stem cell-like immunophenotype were shown to lack expression of NG2. Instead, flow cytometry analyses revealed a close correlation between expression of the myeloid differentiation marker CD33 and increasing levels of NG2 on maturing AML cells. Similarly, by LightCycler RT-PCR CD34+CD19+ cells showed higher expression of NG2-RNA compared with CD34+CD19− cell in both ALL samples. Our results demonstrate that NG2 is not expressed on primitive leukemic cells but is upregulated with differentiation within the leukemic cell clone. This hampers the use of NG2 expression as a sensitive marker for MRD detection as expression of NG2 does not allow detection of relapse-initiating leukemic stem cells.


Pharmaceutics ◽  
2020 ◽  
Vol 12 (5) ◽  
pp. 401 ◽  
Author(s):  
Cinzia Borghese ◽  
Naike Casagrande ◽  
Giuseppe Corona ◽  
Donatella Aldinucci

Adipose-derived stem cells (ADSCs) primed with paclitaxel (PTX) are now hypothesized to represent a potential Trojan horse to vehicle and deliver PTX into tumors. We analyzed the anticancer activity of PTX released by ADSCs primed with PTX (PTX-ADSCs) (~20 ng/mL) in a panel of ovarian cancer (OvCa) cells sensitive or resistant to PTX. We used two (2D) and three dimensional (3D) in vitro models (multicellular tumor spheroids, MCTSs, and heterospheroids) to mimic tumor growth in ascites. The coculture of OvCa cells with PTX-ADSCs inhibited cell viability in 2D models and in 3D heterospheroids (SKOV3-MCTSs plus PTX-ADSCs) and counteracted PTX-resistance in Kuramochi cells. The cytotoxic effects of free PTX and of equivalent amounts of PTX secreted in PTX-ADSC-conditioned medium (CM) were compared. PTX-ADSC-CM decreased OvCa cell proliferation, was more active than free PTX and counteracted PTX-resistance in Kuramochi cells (6.0-fold decrease in the IC50 values). Cells cultivated as 3D aggregated MCTSs were more resistant to PTX than 2D cultivation. PTX-ADSC-CM (equivalent-PTX) was more active than PTX in MCTSs and counteracted PTX-resistance in all cell lines. PTX-ADSC-CM also inhibited OvCa-MCTS dissemination on collagen-coated wells. In conclusion, PTX-ADSCs and PTX-MSCs-CM may represent a new option with which to overcome PTX-resistance in OvCa.


Sign in / Sign up

Export Citation Format

Share Document