Differential Gene Expression in CLL Cells from Bone Marrow and Peripheral Blood Suggests a Role of Bone Marrow Stroma in Leukemic Cell Proliferation.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 708-708
Author(s):  
Adrian Wiestner ◽  
Gerald E. Marti ◽  
Eric M. Billings ◽  
Hui Liu ◽  
Elinor Lee ◽  
...  

Abstract Chronic lymphocytic leukemia (CLL) has been described as an accumulative disease of mature lymphocytes. In the peripheral blood (PB) CLL cells are in Go/G1-phase, and gene expression is most consistent with resting B-cells. However, a recent study demonstrated that a surprisingly large fraction of CLL cells are constantly turned over and that proliferation contributes significantly to the expansion of the clone. The sites where proliferation occurs are not well defined but likely include the bone marrow (BM) and/or lymphoid organs. Likewise, the signals governing proliferation of the leukemic cells are ill defined but there appears to be a role for CLL extrinsic factors including stroma cell interactions or antigen stimulation. We hypothesized that gene expression in BM-CLL cells differs from that in PB-CLL cells. Here we report our analysis of 8 pairs of matched CLL samples derived from 7 individuals in whom we simultaneously obtained PB and BM. All patients were untreated, 3 female, 4 male; in 2 the CLL cells expressed unmutated IgVH genes, and 4 were ZAP70 positive. After informed consent, we obtained a dedicated research aspirate. CD19 selection resulted in >98% purity in all samples. PB-CLL cells express high levels of CXCR4, the receptor for SDF-1. As SDF-1 is expressed by BM stroma cells and CLL cells internalize CXCR4 after binding SDF-1 we measured CXCR4 expression by flow cytometry as an indicator of recent contact between leukemic cells and stroma. In PB, a mean of 63% (range 29–90%) of CLL cells expressed CXCR4 above isotype as compared to 30% (3–62%) of cells from the BM (p=0.007). Conversely, we followed CD69 expression as a marker of activation. In 5 of the 8 pairs CD69 was more highly expressed in BM than in PB derived CD19+ cells (p=0.005) indicating activation of the leukemic cells in the BM microenvironment. We performed gene expression analysis of total mRNA of all matched pairs on Affymetrix U133A 2.0 arrays according to standard protocols. We considered all genes with present calls in either PB-CLL or BM-CLL. The samples were normalized to correct for the individual to individual variance by first normalizing each individual’s PB and BM expression values by their PB value, and then averaging over the 8 individuals using GeneSpring software (Agilent). There were 543 genes with at least 1.5x higher expression in BM vs PB and 192 genes with at least 1.5x higher expression in PB versus BM at p<0.05. Genes more highly expressed in the BM derived CD19-selected cells included topoisomerase II alpha, several cyclins, including cyclin D1, signal transduction components such as PI-3 kinase and components of the Wnt pathway, transcription factors and enhancers such as c-Fos and Sox-4 and several chemokines and chemokine receptors. Of note, there was no difference in the expression of ZAP70, LPL, ADAM29, Bcl-2 and Mcl-1 between the two sites. Our findings are consistent with a model in which CLL cells migrate along an SDF-1 chemokine gradient to the BM where they are stimulated in contact with BM-stroma cells. The higher expression of cell cycle genes in the BM resident CLL cells supports recent findings of a sizeable proliferating fraction of CLL cells and suggests that at least part of this proliferating pool resides in the BM. Ongoing analysis is directed at identifying signaling pathways contributing such proliferation signals.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2927-2927 ◽  
Author(s):  
Jerome Paggetti ◽  
Franziska Haderk ◽  
Martina Seiffert ◽  
Bassam Janji ◽  
Yeoun Jin Kim ◽  
...  

Abstract Chronic lymphocytic leukemia (CLL), the most common hematologic malignancy in Western countries, is mostly affecting the elderly over 65 year-old. CLL is characterized by the accumulation of mature but non-functional B lymphocytes of clonal origin in the blood and the primary lymphoid organs. CLL was previously considered as a relatively static disease resulting from the accumulation of apoptosis-resistant but quiescent B lymphocytes. However, recent studies using heavy water labeling indicated that CLL is in fact a very dynamic disease with alternation of proliferation phases and peripheral circulation. A focus on the trafficking of CLL cells in vivo has shown that leukemic cells circulate between the blood and the lymphoid organs but have a preference for the bone marrow. Recent next-generation sequencing of CLL cells indicated the presence of different genetic subclones. This intraclonal heterogeneity observed in CLL subpopulations may be in part determined by the interactions that leukemic cells entertain with their microenvironment when B cells migrate into the lymph nodes and the bone marrow. Indeed, tumor-stroma interactions are not only providing signals necessary for leukemic cells survival but may also influence the clonal architecture and evolution. One of these interactions involves CLL-derived exosomes. Here, we show that CLL-exosomes efficiently transfer nucleic acids, including functional microRNAs, and proteins, including MHC-Class II molecules and B-cell specific proteins, to bone marrow mesenchymal stem cells and endothelial cells. CLL-exosomes also activate signaling pathways, including PI3K and NF-κB pathways, in these stromal cells. As a consequence, gene expression is strongly modified indicating a switch towards a cancer-associated fibroblast phenotype. Functionally, exosome-stimulated stromal cells show a striking actin cytoskeleton remodeling characterized by the formation of stress fibers, and enhanced proliferation, motility and angiogenic properties. We also identified several proteins synthesized and secreted by stromal cells that promote leukemic cell adhesion and survival ex vivo. To confirm the involvement of CLL-exosomes in CLL pathology in vivo, MEC-1-eGFP cells were subcutaneously injected into immunocompromised NSG mice together with CLL-exosomes. We observed a significant increase in tumor size and a reduction in survival of exosome-treated animals. Flow cytometry analysis of selected organs indicated an enrichment in leukemic cells in the kidney, providing a potential explanation to the renal failures observed in CLL patients. In conclusion, the communication between CLL cells and stromal cells may be a critical factor influencing CLL progression by promoting leukemic cell survival. This study demonstrates the crucial role of exosomes as mediators of the communication between leukemic cells and their microenvironment. Exosomes could thus represent a suitable target for therapeutic intervention in CLL. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5065-5065
Author(s):  
Tamara Sorokina ◽  
Irina Shipounova ◽  
Alexey Bigildeev ◽  
Nina I. Drize ◽  
Larisa A. Kuzmina ◽  
...  

Abstract Background In patients with acute leukemia the stromal microenvironment is deeply modified. Disturbances in signaling pathways, genetic abnormalities and functional changes in mesenchymal cells of these patients have been previously described. Chemotherapy also affect stromal progenitor cells. A damaged microenvironment might impair hematopoiesis in acute leukemia patients. Aims To investigate the relative expression level in MMSCs and CFU-Fs, derived from the bone marrow (BM) of acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) patients before and over the course of chemotherapy. Methods 54 newly diagnosed cases (33 AML, 21 ALL) were involved in the study after informed consent. BM was aspirated prior to any treatment (time-point 0) and at days 37, 100 and 180 since the beginning of treatment of acute leukemia. MMSCs were cultured in aMEM with 10% fetal calf serum, CFU-Fs, in aMEM with 20% fetal calf serum. The relative expression level (REL) of different genes was measured by TaqMan RQ-PCR. As a control MMSCs and CFU-Fs from 88 healthy donors were used. Results At the time of the disease manifestation the analysis of gene expression in MMSCs from acute leukemia patients revealed a significant increase in the REL of genes which regulate immune system responses and thereby can influence on the leukemic cell proliferation and migration (IL-6, IL-8, IL-1b and IL-1R1) (Pic.1). Also at the time of the diagnosis an increase in REL of genes, that are responsible for hematopoiesis regulation, was observed. For example, the REL of CSF1 that can influence on leukemic cells proliferation was increased at the disease manifestation and became normal during the treatment. The same dynamics was observed in the REL of JAG1 that has an antiapoptotic effect on leukemic cells. The REL of LIF had been also significantly increased at the disease manifestation, reflecting the efforts of MMSCs to inhibit leukemic proliferation. Chemotherapy affected REL of the studied genes differently. The treatment lead to the downregulation of IGF, TGFB1 and TGFB2 (Pic.2). As far asTGFB1 and 2 inhibit the differentiation of mesenchymal stem cells, and IGF is associated with myelodysplastic changes in elderly bone marrow, so their downregulation may refer to the effectiveness of therapy. The REL of genes regulating MMSC proliferation (PDGFRa and PDGFRb, FGF2, FGFR1 and 2) increased during chemotherapy. Exploring cell adhesion molecules, the decrease in the REL of their encoding genes was observed. As far as VCAM facilitate the leukemic cell extravasation and ICAM was shown to depress the Th17 cell differentiation, the down-regulation of their genes may reflect the microenvironment restoration. The influence of chemotherapy lead to decrease in REL of genes, associated with MMSCs differentiation (BGLAP and SOX9 (Pic.3)), reflecting the mechanism of the blocking of MMSCs migration and differentiation under the stress conditions. The alterations of bone marrow stroma were more pronounced in patients who didn't achieve remission. The REL of 9 genes was studied in CFU-F colonies. There were no differences in gene expression in CFU-Fs before the treatment, except for an increase in the REL of PPARg in acute leukemia CFU-Fs. During the treatment, a decrease in the REL of SPP1 and an increase in the REL of FGFR1 and 2 were observed. Conclusion Therefore, chemotherapy used does not impair the functional ability of MMSCs and CFU-Fs, but influence on their gene expression profile. The two types of precursors are affected differently, indicating their different differentiation level and functions. Figure 1 Figure 1. Figure 2 Figure 2. Figure 3 Figure 3. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4592-4592
Author(s):  
Julius Juarez ◽  
John Hewson ◽  
Adam Cisterne ◽  
Rana Baraz ◽  
Kenneth F. Bradstock ◽  
...  

Abstract The role of CXCL12 in the growth of B cell progenitor acute lymphoblastic leukemia (ALL) and the homing of these cells to the bone marrow has been well established. However the effect of modulating CXCL12/CXCR4 interactions on the growth of ALL cells in vivo has not been examined. In this study we used specific peptide and small molecule antagonists of CXCR4 to examine the importance of CXCL12/CXCR4 interactions in the development of leukemia in an in-vivo murine model of ALL. CXCR4 antagonists induced mobilization of human and murine B cell progenitor ALL cells into the peripheral blood, with a 3.8±1.9 and 6.5±3.3 fold increase in leukemic cells/ml one hour after administration of the antagonist respectively, similar to that observed for normal progenitors. Daily administration of AMD3100 commencing the day following the injection of cells and continuing for 21 days resulted in a mean reduction in peripheral blood white cell count of 50±12% and the leukemic cell count of 63±4%. There was also a significant reduction in both the total cells in the spleen of 58±1% and the leukemic cell number in this organ of 75±11%. A significant reduction in leukemic cell numbers in the bone marrow was observed in one (44% reduction) case. There was reduced infiltration of other organs including kidney, liver and skeletal muscle. This study demonstrates that disrupting the CXCL12/CXCR4 axis in B cell progenitor ALL reduces the tumor burden. Whether this is due to direct inhibitory effects on proliferation and survival, or results from disruption of the leukemic cell interactions within the bone marrow remains to be determined.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3746-3746
Author(s):  
Anna Paczulla ◽  
Martina Konantz ◽  
Sarah Grzywna ◽  
Lothar Kanz ◽  
Claudia Lengerke

Abstract Introduction The caudal-type homeobox (Cdx) gene family has been mainly studied during early development for its role in axial elongation and antero-posterior patterning. More recently, Cdx genes were shown to regulate embryonic hematopoiesis by interactions with the canonical Wnt pathway and Hox genes. The role of Cdx genes in adult hematopoiesis remains poorly understood. Adult hematopoietic stem and progenitor cells derived from healthy murine bone marrow (BM) express low levels of Cdx1 and Cdx4 but not Cdx2. However, the majority (>80%) of human acute myeloid (AML) and lymphoid leukemias (ALL) were shown to express the human homologue CDX2, and ectopic induction of Cdx2 expression was sufficient to robustly induce myeloid leukemia in murine bone marrow cells. On the molecular level, the leukemogenic activity of Cdx2 was associated with modulation of Hox and Klf4 gene expression (Faber et al, 2013). The current study further explores the role of CDX2 in leukemogenesis by analyzing the effects of CDX2 expression induction or repression on human healthy and malignant hematopoietic cells and its molecular effects on the Wnt signaling pathway known to regulate Cdx genes during embryonic development. Methods Human bone marrow or mobilized peripheral blood derived CD34+ cells as well as the human leukemic cell lines SKM-1, NOMO-1, EOL-1 and NALM16 were exposed to lentiviruses containing CDX2 overexpression, shRNAs against CDX2 or control constructs. Efficient modulation of CDX2 expression was verified on gene expression level by qRT-PCR and on protein level by immunoblot analysis. CDX2 modified and control cells were subjected to growth, colony forming (CFU), cell cycle, flow cytometry and qRT-PCR gene expression analysis assays and analyzed in vivo upon xenotransplantation in NOD/SCID/IL2Rγnull (NSG) mice. To explore the effect of Dickkopf-1 (DKK1), recombinant human DKK1 protein or carrier was supplemented to the methylcellulose in CFU assays. Results shRNA-mediated knockdown of CDX2 in leukemic cell lines lead to reduced growth (SKM-1, NALM16) and CFU formation (SKM-1 cells). Consistently, CDX2 knockdown SKM-1 cells showed lower ability to repopulate NSG mice and, upon subcutaneous injection in the flank, gave rise to much smaller tumors when compared to control cells, supporting the notion that CDX2 plays roles in human leukemogenesis. In contrast to the data published in mice, healthy human CD34+ cells transduced to overexpress CDX2 were unable to induce leukemia upon transplantation in NSG mice within an observation period of 5 months. On the molecular level, CDX2 modified cells showed differential expression of Klf4 and Hox but also Wnt pathway associated genes. Notably, robust induction of the canonical Wnt-inhibitory molecule DKK1 was observed in both healthy CD34+ stem/progenitor and leukemic cells upon CDX2 induction, while CDX2 suppression showed opposite effects. Analysis of the DKK1 promotor region revealed an interspecies conserved putative binding site for CDX2 as well as multiple HOX gene binding sites, suggesting that CDX2 can modulate DKK1 expression directly but also via its downstream HOX genes. Importantly, CFU assays performed on CDX2-knockdown cells showed a rescue of colony formation upon stimulation with DKK1 protein as compared to treatment with carrier only, demonstrating that the observed molecular interaction is functionally relevant in human leukemic cells. In contrast, control leukemic cells treated with DKK1 showed reduced CFU formation, indicating that CDX2 might act through DKK1 activation to fine-tune Wnt signal activation to the dosage that best promotes leukemogenesis and leukemic cell growth and survival. Conclusion Taken together, our data indicate that CDX2 employs DKK1 activation to modulate the Wnt signaling pathway and thereby growth, clonogenic capacity as well as in vivo tumorigenicity of human leukemia cells. In contrast to murine cells, CDX2 activation requires cooperative molecular events in order to induce leukemia in human healthy stem and progenitor cells. Disclosures: No relevant conflicts of interest to declare.


Cancers ◽  
2019 ◽  
Vol 11 (12) ◽  
pp. 1939 ◽  
Author(s):  
Severin ◽  
Frezzato ◽  
Visentin ◽  
Martini ◽  
Trimarco ◽  
...  

The bone marrow microenvironment promotes proliferation and drug resistance in chronic lymphocytic leukemia (CLL). Although ibrutinib is active in CLL, it is rarely able to clear leukemic cells protected by bone marrow mesenchymal stromal cells (BMSCs) within the marrow niche. We investigated the modulation of JAK2/STAT3 pathway in CLL by BMSCs and its targeting with AG490 (JAK2 inhibitor) or Stattic (STAT3 inhibitor). B cells collected from controls and CLL patients, were treated with medium alone, ibrutinib, JAK/Signal Transducer and Activator of Transcription (STAT) inhibitors, or both drugs, in the presence of absence of BMSCs. JAK2/STAT3 axis was evaluated by western blotting, flow cytometry, and confocal microscopy. We demonstrated that STAT3 was phosphorylated in Tyr705 in the majority of CLL patients at basal condition, and increased following co-cultures with BMSCs or IL-6. Treatment with AG490, but not Stattic, caused STAT3 and Lyn dephosphorylation, through re-activation of SHP-1, and triggered CLL apoptosis even when leukemic cells were cultured on BMSC layers. Moreover, while BMSCs hamper ibrutinib activity, the combination of ibrutinib+JAK/STAT inhibitors increase ibrutinib-mediated leukemic cell death, bypassing the pro-survival stimuli derived from BMSCs. We herein provide evidence that JAK2/STAT3 signaling might play a key role in the regulation of CLL-BMSC interactions and its inhibition enhances ibrutinib, counteracting the bone marrow niche.


2019 ◽  
Vol 20 (2) ◽  
pp. 292 ◽  
Author(s):  
Paolo Giannoni ◽  
Franco Fais ◽  
Giovanna Cutrona ◽  
Daniela Totero

Chronic lymphocytic leukemia (CLL) is characterized by the progressive expansion of B lymphocytes CD5+/CD23+ in peripheral blood, lymph-nodes, and bone marrow. The pivotal role played by the microenvironment in disease pathogenesis has become increasingly clear. We demonstrated that bone marrow stromal cells and trabecular bone cells sustain survival of leukemic B cells through the production of hepatocyte growth factor (HGF). Indeed the trans-membrane kinase receptor for HGF, c-MET, is expressed on CLL cells and STAT3 TYR705 or AKT phosphorylation is induced after HGF/c-MET interaction. We have further observed that c-MET is also highly expressed in a peculiar type of cells of the CLL-microenvironment showing nurturing features for the leukemic clone (nurse-like cells: NLCs). Since HGF treatment drives monocytes toward the M2 phenotype and NLCs exhibit features of tumor associated macrophages of type 2 we suggested that HGF, released either by cells of the microenvironment or leukemic cells, exerts a double effect: i) enhances CLL cells survival and ii) drives differentiation of monocytes-macrophages to an oriented immune suppressive phenotype. We here discuss how paracrine, but also autocrine production of HGF by malignant cells, may favor leukemic clone expansion and resistance to conventional drug treatments in CLL, as well as in other hematological malignancies. Novel therapeutic approaches aimed to block HGF/c-MET interactions are further proposed.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 5-5
Author(s):  
Shaker A Mousa ◽  
Noureldien Darwish ◽  
Thangirala Sudha

Acute myeloid leukemia is one of the most aggressive malignant hematological disorders on worldwide basis. More than 19,000 new cases are estimated in the United States in 2020 (1.1% of all new cancer cases). The estimated death rate from AML is 11,180 that represent 56% of the new cases (1.8% of all cancer death), with an overall 5-year survival rate of 27.4% [1]. Current treatment regimens for AML include traditional chemotherapy, radiotherapy, allogeneic hematopoietic cell transplantation and targeted therapies for specific mutations in limited numbers of AML patients, all of whom still suffer from adverse effects and relapse. New broad spectrum effective and safe treatment options are urgently needed for the different types of AML. Our strategy focused on developing a novel targeted therapy for the thyrointegrin αvβ3 receptors that are over-expressed in leukemic cells. This receptors support adhesion, engraftment, proliferation, invasion/metastasis, and angiogenesis functions of leukemic cells. The identified thyrointegrin αvβ3 antagonist fluorobenzyl Polyethylene glycol Mono-Triazole Tetraiodothyroacetic Acid (fb-PMT) binds with high affinity and specificity. Our study showed that fb-PMT effectively suppresses the functions of AML cell line and primary cell harboring FLT3-ITD after successful engraftment in transgenic NSG-S xenograft mouse models. Daily treatment with fb-PMT at doses ranging from 1-10 mg/kg, subcutaneously for 3-4 weeks were associated with leukemogenesis regression, suppression of cancer invasion and extended survival in both models. These findings were verified using IVIS scanning and histopathological examination to evaluate the engraftment of leukemic cells in the bone marrow and other organs including, spleen, liver, lung, and brain. Furthermore, fb-PMT at 3 and 10 mg/kg, s.c. daily for 3-4 weeks exhibited significant reduction (P<0.001) of leukemic cell burden 74% and >95%, respectively. Peripheral blood smears from fb-PMT treated animals were reversed back to normal with no blast cells along with normal cell counts. Bone marrow regain its normal maturation with abundant segmented neutrophil and megakaryocytes, representing complete hematological remission along with complete suppression of leukemic cell metastasis and invasion into different organs. To evaluate the relapse after treatment, 40 mice were maintained off treatment for further 2 weeks and IVIS scans and peripheral blood smear examination were performed on animals at end of first week and at sacrifice. The fb-PMT (10 mg/kg) off treatment has shown successful maintained remission after stopping the daily treatment as confirmed using blood smear examination, IVIS scan, and histopathological examination. Additionally, safety assessments in rodents and monkeys demonstrated safety and tolerability at multiple folds above the anticipated human doses. Lastly, our genome-wide microarray screen demonstrated that fb-PMT works through the Hedgehog pathway and Il-21 receptor down-regulation as well as downregulation of several genes including, IGF2, TWIST1 and FYN oncogene, angiopoietin 1 (ANGPT1), angiopoietin-like 2 (ANGPTL2) and PIM1 oncogene KIT, HRAS, INH2, BCL, AKT1, IDH2, CDK4/6, TIMP1, VEGF, EGFR and PD-L1, KDM6A, EZH2. Collectively, preclinical findings of fb-PMT warrant its clinical investigation for the effective and safe management of AML. Reference 1. Key Statistics for Acute Myeloid Leukemia (AML). 2020; Available from: https://www.cancer.org/cancer/acute-myeloid-leukemia/about/key-statistics.html. Figure 1 Disclosures Mousa: Vascular Vision Pharma Co.: Patents & Royalties.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3457-3457
Author(s):  
Maria Norberg ◽  
Marie Sevov ◽  
Mahmoud Mansouri ◽  
Gerard Tobin ◽  
Mats Merup ◽  
...  

Abstract Abstract 3457 Poster Board III-345 One of the hallmarks of the clinically heterogeneous disease chronic lymphocytic leukemia (CLL) is defective apoptosis, which is considered to contribute not only to cell accumulation but also to disease progression and resistance to therapy. In an effort to further investigate the ‘apoptotic profile’ in CLL, we have utilized the multiplex ligation-dependent probe amplification (MLPA) method to analyze the expression profile of 35 apoptosis-related genes in 67 CLL cases including 49 peripheral blood, 9 lymph node, 7 bone marrow and 2 spleen samples. CLL cells derived from peripheral blood displayed a different overall expression profile when compared to peripheral blood mononuclear cells (PBMC) from five healthy controls, with several genes including the anti-apoptotic genes BCL2A1 and MCL1 and the pro-apoptotic genes BAX and BMF all being significantly differently expressed (>2 fold difference in median expression). Few differences were detected when comparing peripheral blood CLL samples (25/49 IGHV unmutated/IGHV3-21) to lymph node samples (6/9 unmutated/IGHV3-21) or to bone marrow samples (5/7 unmutated/IGHV3-21), with lower relative expression of BIK and BAX in peripheral blood samples. Furthermore, we correlated the expression profiles of all genes included in the assay with IGHV mutation status, IGHV3-21 gene usage and genomic aberrations, but overall few differences were evident between prognostically different subgroups. An exception to this was the BIK gene which showed significantly higher expression in unmutated/IGHV3-21 compared to mutated CLL. Additionally, MCL1 and BNIP3L displayed higher expression in 17p-deleted cases compared to 13q deletion/no aberration cases. High expression of HRK and BAX indicated a correlation with poor overall survival and shorter time to treatment. However, when performing real-time quantitative PCR on an extended patient cohort (n=112) only prediction of time to treatment could be confirmed for HRK and BAX (p=0.0035 and p=0.024, respectively). Finally, in order to validate our MLPA approach, we compared the gene expression levels of four genes in 19 CLL samples using both MLPA and real-time quantitative PCR, which revealed a significant correlation for all genes tested. In summary, MLPA is a quick and cost-effective method for pathway specific gene expression analysis and the present analysis of 67 CLL cases highlights the rather homogenous apoptosis gene expression profiles among different clinical subsets of the disease. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 5018-5018
Author(s):  
Henry Qixiang Li ◽  
Jingping Liu ◽  
Xiaoyu An ◽  
Na Wang ◽  
Liang Huang ◽  
...  

Abstract We have recently successfully engrafted leukemic cells from bone marrow of several AML patients into immunocompromised NOD/SCID mice. One of them, AM7577, was reported in the last year ASH1. This model displayed typical aggressive AML disease of M5 subtype, which starts at bone marrow and gradually expand to peripheral (spleen, lymphonode and peripheral blood…). AM7577 also harbors interesting genotypes of mutations for IDH2-R140Q, FLT3-ITD, DNMT3A R882H and NPM1. Our second AML PDX model, AM8096, was established similarly using bone marrow from a recurrent patient with AML-M2 disease. AML8096 displayed similarly aggressive disease and the same 100% mortality, including typical symptoms (BW loss, hunched, inactivity, labored breathing, ruffled coat and eventual mortality) and with abundant leukemic cells in bone, etc. The leukemic cells can serially be passed in mice with 100% take-rate and cause consistent disease (even with < 1e4 cells). This also creates a renewable and potentially unlimited source of leukemia cells. The leukemic cells in mice are identical to those of the original patient leukemic cells (CD45+, CD33+, CD13+, CD123+, and CD19-. However, some aspects of disease presentations are vastly different from that of AM7577. First, the peripheral symptom is significant less characterized by lower leukemic counts in peripheral blood and only slightly enlarged spleen and smaller lymphonode. Second, the leukemic cell morphology is also rather different with AM8096 demonstrating less differentiated phenotype. Third, the genotype, in contrast to AM7577, is wild-types for all the above oncogenes. While we have not identified the likely leukemogenesis driver gene for this model, we are currently performing RNAseq of AM8096, along with AM7577, in order to explore the underlying molecular mechanisms that drive both diseases. In addition, we are also investigating the drug response to standard of care (SOC), as compared to those AM7577. In summary, the two AML models could serve as useful experimental models to investigate the diverse leukemogenesis, including molecular mechanisms by genetic profiling. Ultimately, they can be used to help to identify new treatment strategies for AML. 1. Liu, e.a. A unique leukemia mouse model established from AML patient with IDH2 R140Q and FLT3-ITD mutations among other common AML mutations. ASH-2012 Annual Meeting (2012). Disclosures: Li: Crown Bioscience: Employment. Liu:Crown Bioscience: Employment. An:Crown Bioscience: Employment. Wu:Crown Bioscience: Employment. Cai:Crown Bioscience: Employment. Wery:Crown Bioscience: Employment.


Sign in / Sign up

Export Citation Format

Share Document