scholarly journals Immunotherapy Targeting ST2/IL-33 Signaling in Myeloid Leukemia Stem Cells

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 23-23
Author(s):  
Denggang Fu ◽  
Hua Jiang ◽  
Alan Long ◽  
Hong fen Guo ◽  
Maegan L. Capitano ◽  
...  

Abstract Therapies for acute myeloid leukemia (AML) has barely changed over 30 years, while treatment for other blood cancers have made remarkable leaps forward. Recent advances in genomics have allowed molecular targeted therapies (i.e. FLT3-ITD, IDH, c-KIT inhibitors) extending survival, but most patients still succumb (Burd et al. Nat Med 2020). Therefore, developing more efficient, less toxic, and immune-based therapies for AML is an urgent unmet need. Previous studies showed that stromal cell-derived IL-33 stimulates myeloproliferative neoplasms (Mager et al. J Clin Invest 2015). Stimulation-2 (ST2), IL-33 receptor, contributes to leukemia stem cells (LSCs) survival in Cbfb-MYH11 mice (Wang et al . Sci Rep 2019). We showed that ST2 blockade enhanced graft versus leukemia activity against MLL-AF9 egfp AML after hematopoietic cell transplantation (Zhang et al . Sci Transl Med 2015). We and others, also, found that ST2 is expressed on normal murine and human hematopoietic stem cells (HSCs), respectively (Capitano et al. Blood Cells Mol Dis 2020; Alt et al. Biol Blood Marrow Transplant 2019). These data suggest a leukemia-promoting role of ST2/IL-33 signaling. To determine clinical relevance of ST2 in AML, we generated Kaplan-Meier curves using TCGA (n=173) and TARGET AML (n=187) databases. Decreased survival was observed in patients with high IL1RL1 (ST2 gene) which was validated in an independent database (AMLCG 1999 trial, n=417) (Fig. 1A). Since ST2 is expressed on HSCs, we interrogated if ST2 is expressed on LSCs defined as CD34 +CD38 - in the Princess Margaret Leukemia biobank (n=192), and found ST2 is higher on LSCs vs CD34 -CD38 +/- cells (Fig. 1B). We then sought to analyze ST2 on bone marrow samples comparing complete responders vs refractory patients to note that ST2 expression was increased in refractory patients' LSCs (Fig. 1C). To scrutinize the role of ST2 in initiating leukemogenesis, we performed limiting dilution transplantation using 500, 200, and 50 Lin -Sca-1 +c-KIT +-sorted LSCs from WT vs ST2 -/- MLL-AF9 egfp transduced cells. Frequency of LSCs in ST2 -/- cells was decreased by ~15-fold as opposed to WT cells [1:2141 (1:546-1:8405) vs 1:145 (1:75-1:283), p=3.37e-05] (Fig. 2A). We also tested leukemia maintenance, secondary transplantations from the primary recipients resulted in leukemia growth delay in ST2 -/- vs WT cells which was confirmed in tertiary transplantations (Fig. 2B-E). Self-renewal ability of LSCs is correlated to reactive oxygen species (ROS) (Testa et al. Exp Hematol 2016), and we found that ROS levels in ST2 -/- leukemic cells are markedly diminished in contrast to WT leukemic cells (Fig. 2F). ST2 deficiency in leukemic cells arrests G2/S/M cell cycle progression in LSCs (Fig. 2G-J). These data indicated that ST2 is indispensable for initiating and maintaining LSCs in MLL-AF9 AML. We next developed murine and human Fc-silenced-bispecific antibodies engaging mouse or human ST2 and CD3 (BsAb) built on the IgG[L]-scFv platform with proven ability to drive T cells into human tumors for effective tumor ablation (Santich et al. Sci Transl Med 2020; Park et al. J Immunother Cancer 2021) (Fig. 3A, 3E). Both BsAbs showed >90% purity by HPLC, stability under heat stress and low endotoxin. Animals did not exhibit any in vivo toxicity at BsAb doses of 0.4, 2, 5, 10 μg ip q 3 days x 6 doses (not shown). In the immunocompetent MLL-AF9 mice, murine anti-ST2 BsAb (BC281) treatment (10 μg i.p, 4 days post-AML challenge and given every three days for a total of 6 injections) resulted in extended survival compared to isotype control mice (Fig. 3B). Leukemic cells and LSCs were accordingly decreased in treated vs control group (Fig. 3C-D). We modeled humanized leukemic mice with MOLM-14 egfp cells and weekly injection of human CD8 + T cells in NOD.Cg-Prkdc scid Il2rg tm1Wjl/SzJ (NSG) mice (Fig. 3F). Animals treated with human anti-ST2 BsAb (BC282), using a similar regimen as for the immunocompetent model, led to better survival when compared to animals treated with mutated non-functional anti-ST2 BsAb (BC283) (Fig. 3G). Frequency of MOLM-14 egfp cells was lower in the BC282 vs BC283 group (Fig. 3H). These results suggested that anti-ST2 BsAbs can inhibit AML growth to improve survival. We concluded that ST2 is a potential therapeutic target, and ST2-specific T cell engaging BsAbs represent promising immunotherapeutics for AML. Figure 1 Figure 1. Disclosures Cheung: Medical University of South Carolina: Patents & Royalties: inventor on the ST2 bispecific antibody patent application; Y-mabs Therapeutics and Abpro-Labs Inc: Patents & Royalties: inventor on multiple patents filed by MSK, including those licensed to Ymabs Therapeutics, Biotec Pharmacon, and Abpro-labs; Eureka Therapeutics: Membership on an entity's Board of Directors or advisory committees. Paczesny: Medical University of South Carolina: Patents & Royalties: inventor on the ST2 bispecific antibody patent application.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1634-1634
Author(s):  
Michael A Amrein ◽  
Elias D Bührer ◽  
Stefan Forster ◽  
Stephan Isringhausen ◽  
Christian M Schürch ◽  
...  

Chronic myeloid leukemia (CML) is a typical stem-cell driven malignancy, driven by leukemia stem cells (LSCs). LSCs are resistant to conventional therapies. This resistance is mediated by cell-intrinsic mechanisms and interactions with the microenvironment. LSCs depend on signals from a specialized microenvironment, a so called niche, to maintain their stem cell characteristics. In CML the bone marrow (BM) as a niche is well-investigated and several therapeutic targets, which aim at LSCs by interrupting their interaction with the BM-niche are under investigation. However, even though splenomegaly is a hallmark of CML the contribution of the splenic microenvironment to CML development has not been studied so far. This project aims to investigate the role of the splenic microenvironment as an independent secondary LSC niche and its contribution to disease development. To induce a CML-like disease in mice we retrovirally transduced FACS-sorted Lineage- Sca-1+ cKit+ BM cells with pMSCV-p210BCR/ABL-IRES-GFP and injected the transduced cells into non-irradiated mice. To find out if the spleen contributes to disease development we induced CML in splenectomized and sham operated mice. Splenectomized mice survived significantly longer compared to sham operated controls (median survival 31 vs. 22 days; p=0.0006) with 20% of the splenectomized mice surviving longer than 90 days. Moreover, the number of LSCs in the BM of splenectomized mice was reduced 3.7-fold (p=0.002). Flowcytometric analysis of the spleen and BM compartments of CML bearing mice revealed that the majority of the leukemic stem and progenitor cells (LSPCs) were located in the spleen (19-fold more LSCs in the spleen; p =0.007). Moreover we found the leukemic compartment in the spleen to be enriched for LSPCs compared to the BM (20 % spleen vs. 10 % BM; % LSPCs of total leukemic cells; p=0.01). To confirm this phenotypic observation functionally we performed a limiting dilution transplantation of leukemic cells from spleen and BM. In line with the phenotypic observation we found a higher frequency of LSCs in the spleen compared to the BM (1/41'703 vs. 1/432'594; p=0.02). We next analyzed the gene expression of LSPCs from spleen and BM. We found that the gene expression profile of splenic LSPCs showed higher expression of stemness-related genes and reduced expression of myeloid differentiation genes compared to BM LSPCs, indicating that the spleen is more supportive of primitive LSPCs. Knowing that the spleen contributes to disease development by providing an alternate niche for LSCs we next analyzed the spleens using confocal microscopy. We found that the LSCs resided exclusively in the red pulp. Previous studies have shown that HSCs reside in direct contact with red pulp macrophages (RPMs) during extramedullary hematopoiesis (Dutta et al., JEM, 2015). In addition we found that in spleens from human CML patients CD34+ leukemia cells localized together with macrophages (p=0.001). Furthermore we could show that RPMs are capable of producing both SCF and G-CSF. To test the role of RPMs as a potential niche component in vitro we co-incubated LSCs and RPMs overnight before plating the LSCs in a colony formation assay. We found that the co-incubation with RPMs improved the colony formation capacity of LSCs (CFUs 166 vs. 138; p=0.0356). To test the role of RPMs in vivo we depleted macrophages in CML mice using clodronate liposomes. This resulted in significantly reduced splenomegaly (867mg vs. 249mg; p<0.0001) and reduced numbers of splenic LSCs (23-fold; p=0.001). This was further confirmed in a genetically engineered mouse model lacking RPMs (Spic-/-) (735mg vs. 450mg; p=0.0112 and 22-fold fewer LSCs; p<0.0001). Finally, while in naïve mice RPMs can be differentiated into a CD24low and CD24high population, the CD24high population is lost in CML bearing mice. In summary, we found that the spleen provides an alternate niche for LSCs, thereby contributing to CML development. Compared to the BM niche the splenic niche is more supportive of primitive LSPCs, as shown by the higher frequency of LSCSs found in the spleen and the higher expression of stemness related genes in splenic LSCs compared to BM LSCs. Moreover we identified CD24low RPMs as a unique and central component of the splenic LSC niche. Even though we could show that RPMs are capable of producing SCF and G-CSF the exact mechanisms by which RPMs support LSCs remains to be investigated. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1079-1079
Author(s):  
Biniam Adane ◽  
Haobin Ye ◽  
Shanshan Pei ◽  
Nabilah Khan ◽  
Mohammad Minhajuddin ◽  
...  

Abstract NADPH dependent oxidase 2 (NOX2) is the founding member of a family of multimeric, oxido-reductase enzymes that catalyze the production of superoxides by transferring a single electron from the cofactor NADPH to molecular oxygen. It is primarily utilized in neutrophils and macrophages to generate copious amount of reactive oxygen species (ROS) to facilitate the neutralization of engulfed particulates during phagocytosis. In sharp contrast to this specialized function however, recent evidence implies a non-phagocytic role for NADPH oxidases in which physiologic levels of ROS generated by these enzymes modulate key signaling proteins and transcription factors to exert profound biological effects. Based on this information we decided to investigate the potential role of NOX2 in normal and leukemic stem cells. Using transgenic NOX2 knock out mice, genetically defined murine models of myeloid leukemia and primary human acute myeloid leukemia (AML) specimens, we show that NOX2 is critical for the proper function of normal and malignant hematopoietic stem cells. In silico analysis using published transcriptional profiles of hematopoietic populations revealed that multiple subunits of the NOX2 complex are expressed at low levels in hematopoietic stem cells (HSCs) and at relatively higher levels in multipotent progenitors (MPPs). Next, we characterized the different hematopoietic compartments from age and sex matched wild type (WT) and transgenic NOX2 knock out (KO) mice. Our studies revealed that in the bone marrow of KO mice, a subset of multipotent progenitor populations (MPP2 & MPP3), which often have biased myelo-erythroid output are markedly expanded relative to their wild type counterparts. Consistently, we found increased levels of granulocytes and monocytes in the peripheral circulation of NOX2 KO mice. To test whether NOX2 has a functional, biological role in the self-renewal of HSCs, we performed competitive transplantation assays using equal numbers of whole BM cells from WT and KO mice to co-repopulate lethally irradiated hosts. Analysis of engrafted mice showed that the contribution from NOX2 KO HSCs was severely compromised in all lineages and developmental stages of hematopoiesis examined. Collectively, these results suggest a critical biological role for NOX2 in maintaining the quiescence and long term self-renewal of HSCs. Similar to normal hematopoiesis, we found out that NOX2 is also widely expressed by functionally defined leukemic stem cells in a murine model of myeloid leukemia generated by expressing the oncogenic translocations BCR-ABL and NUP98-HOXA9. To evaluate the role of NOX2 in leukemogenesis, we established the BCR-ABL/NUP98-HOXA9 model using primitive cells derived from either WT or KO. Intriguingly, NOX2 KO leukemic cells generated a much less aggressive disease upon transplantation into primary and subsequently into secondary recipients. Furthermore, leukemic cells in which NOX2 is suppressed displayed aberrant mitotic activity and altered developmental potential marked by loss of quiescence, enhanced entry into cycle and terminal differentiation. To gain mechanistic insight into the observed phenotype, we isolated leukemic stem cells and performed whole genome expression analysis. The data showed that deficiency of NOX2 leads to downregulation of the cell cycle inhibitor CDKN2C (p18) and robust activation of the granulocyte fate determining transcription factor CEBPε. Thus we conclude that loss of NOX2 impacts leukemogenesis through rewiring of the cell cycle machinery and developmental programs in leukemic stem cells. Finally, we found that in CD34+ primary human AML cells, NOX2 and the other subunits of the complex are abundantly expressed. Furthermore, pharmacologic inhibition of NOX2 with VAS2870, a selective NADPH oxidase inhibitor, reduced the level of ROS and limited the in vitro proliferation and survival of leukemic cells. Next we genetically suppressed the expression of NOX2 in primary human AML cells using sh-RNAs and transplanted these cells into immune compromised mice. Consistent with the murine leukemia, NOX2 knocked down AML cells failed to engraft and expand in vivo. Taken together, our results firmly establish a hitherto unrecognized, prominent regulatory role for NOX2 in the biology of normal and malignant hematopoietic stem cells and imply a potential therapeutic opportunity that can get exploited to treat AML. Disclosures Pollyea: Celgene: Other: advisory board, Research Funding; Ariad: Other: advisory board; Pfizer: Other: advisory board, Research Funding; Glycomimetics: Other: DSMB member; Alexion: Other: advisory board.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 16-17
Author(s):  
Qiang Liu ◽  
Olga I. Gan ◽  
Gabriela Krivdova ◽  
Aaron Trotman-Grant ◽  
Stephanie M. Dobson ◽  
...  

Acute myeloid leukemia (AML) is an aggressive hematologic malignancy with poor survival, especially in older patients. Despite high remission rates after chemotherapy, relapse and death are frequent due to persistence of leukemia stem cells (LSCs), which possess properties linked to therapy resistance. Thus, there is an urgent need for a deeper understanding of the unique properties of LSCs. MicroRNAs (miRNAs) are non-coding RNAs that decrease expression of their target mRNAs by post-translational silencing. miRNA profiling of human AML samples fractionated based on LSC activity revealed that miR-125b is expressed at significantly higher levels on cell fractions enriched in LSCs. To evaluate the role of miR-125b in LSCs, expression of miR-125b was enforced in a hierarchical AML model cell line (OCI-AML-8227). miR-125b overexpression (OE) resulted in a significantly lower percentage of CD14+CD15+ differentiated myeloblasts (Figure 1A) and enhanced clonogenic potential in vitro (Figure 1B). Xenotransplantation of four AML patient samples with miR-125b OE revealed a significant increase in the proportion of CD117+ cells, a marker of hematopoietic and leukemic progenitors (Figure 1C). Secondary transplantation of cells harvested from primary engrafted mice at limiting dilution demonstrated a marked increase in LSC frequency with miR-125b OE compared to controls for the two AML samples tested (Figure 1D). Together, these data strongly suggest that miR-125b enhances the self-renewal of LSCs. To investigate the mechanisms by which miR-125b enhances self-renewal, proteomic analysis of miR-125b-OE Ba/F3 cells as well as in silico target prediction were performed and identified PTPN18 as a top putative target for miR-125b. PTPN18 is a tyrosine phosphatase that has been reported to dephosphorylate auto-phosphorylated kinases such as Her2 and Abl to prevent their activation. To evaluate whether PTPN18 OE can rescue the effects miR-125b on LSCs, we carried out transduction of an AML patient sample with control, miR-125b OE, PTPN18 OE, or both miR-125b and PTPN18 OE vectors followed by xenotransplantation. Similar to previous findings, miR-125b OE alone significantly reduced the frequency of CD11b+CD15+ differentiated myeloblasts. Co-transduction of miR-125b/PTPN18 OE vectors resulted in generation of significantly more CD11b+CD15+ cells compared to miR-125b OE alone (Figure 1E), suggesting that suppression of PTPN18 contributes to miR-125b-mediated enhancement of LSC self-renewal. To identify putative phosphotyrosines that might be altered through the miR-125b-PTPN18 signalling axis, we performed immunoprecipitation of phosphotyrosines followed by mass spectrometry in miR-125b-OE Ba/F3 cells and identified increased GSK3 tyrosine phosphorylation as a top target. Additionally, miR-125b OE was confirmed to enhance GSK3 tyrosine phosphorylation, whereas PTPN18 OE reduced it (Figure 1F), together strongly suggesting that miR-125b could enhance tyrosine phosphorylation of GSK3 by silencing PTPN18. GSK3A and GSK3B (GSK3A/B) are paralogous genes that share a high degree of sequence homology and belong to the glycogen synthase kinase 3 (GSK3) family. Tyrosine phosphorylation activates the kinase activity of GSK3, whereas serine phosphorylation inactivates it. We recently identified GSK inhibitors as top candidates targeting LSCs in a stemness-based drug screen using OCI-AML-8227 cells (data not shown). Treatment of OCI-AML-8227 cells with two selective inhibitors of GSK3 selectively reduced the proportion of CD34+ cells while concomitantly increasing expression of myeloid markers CD14 and CD15 (Figure 1G). Overall, our results support an important functional role for PTPN18 and GSK3 in LSC function, and present a potential novel therapeutic target against LSCs. This study highlights the importance of understanding the role of miRNAs and may identify a new druggable vulnerability in LSCs that could lead to the development of new treatment options for AML patients. Figure 1 Disclosures Dick: Bristol-Myers Squibb/Celgene: Research Funding. Wang:Trilium Therapeutics: Patents & Royalties.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 240-240 ◽  
Author(s):  
Jonathan M. Gerber ◽  
B. Douglas Smith ◽  
Brownhilda Ngwang ◽  
Hao Zhang ◽  
Milada Vala ◽  
...  

Abstract Abstract 240 Relapse of acute myeloid leukemia (AML) is hypothesized to reflect the failure of current therapies to adequately target leukemia stem cells (LSCs) - the rare, resistant cells presumed responsible for maintenance of the leukemia. These cells have generally been reported to have a similar phenotype to normal hematopoietic stem cells (HSCs). However, despite the considerable research on LSCs over the past two decades, the clinical significance of these cells remains uncertain. We hypothesized that any minimal residual disease (MRD) present after therapy would be enriched for LSCs and that such persistence of LSCs would predict relapse. CD34+ cell populations from 27 AML patients and 10 normal donors were analyzed by flow cytometry for CD38 expression and for aldehyde dehydrogenase (ALDH) activity by Aldefluor. A total of 16 AML patients who achieved morphologic complete remission (CR) after induction chemotherapy were followed throughout their treatment course, and the flow cytometric staining patterns of their CD34+ cells at follow-up intervals were correlated with clinical outcomes. Cell subpopulations were sorted and then analyzed by fluorescence in situ hybridization (FISH) for leukemia-specific cytogenetic abnormalities (when applicable) and by transplantation into NOD/SCID-IL2Rgnull (NSG) mice to determine their in vivo self-renewal capacity. Normal bone marrow CD34+CD38− cells consistently exhibited two, discrete subpopulations by ALDH activity: one with low ALDH activity levels (CD34+CD38−ALDHlow) and another with high levels (CD34+CD38−ALDHhigh). As few as 1000 CD34+CD38−ALDHhigh cells generated normal hematopoiesis after transplantation into NSG mice. An additional population of CD34+CD38− cells with intermediate (int) levels of ALDH activity was found in the AML patients, but not in any of the normal donors studied. When present, even in patients in cytogenetic CR, this CD34+CD38−ALDHint population was at least 89% leukemic by FISH; and 1000 of these cells generated AML when transplanted into NSG mice. The CD34+CD38−ALDHhigh cells were invariably present in small numbers in newly-diagnosed AML patients. These cells did not harbor the AML-specific FISH abnormality, and 1000 generated normal hematopoiesis when transplanted into NSG mice, consistent with a residual population of normal HSCs. In those AML patients who achieved CR, any detectable MRD was enriched for the CD34+CD38−ALDHint leukemic cells. This population comprised 34% (range 9–51%) of the total leukemic burden when detectable in patients in cytogenetic CR, as compared to just 3% (range 0.5–4%) at initial diagnosis (p=0.02). Six of the seven CR patients with a detectable CD34+CD38−ALDHint population ultimately relapsed; the lone exception underwent allogeneic transplantation in first CR. Conversely, all nine of the patients with a consistently undetectable CD34+CD38−ALDHint population have remained in CR (p<0.01), with a median follow-up of 688 days. Putative AML LSCs have a unique CD34+CD38−ALDHint phenotype, which distinguishes them from normal (CD34+CD38−ALDHhigh) HSCs. These LSCs appear to be more resistant to therapy than the bulk leukemic cells, as reflected by their relative enrichment in MRD after therapy. Furthermore, the persistence of these putative LSCs in patients after therapy was highly predictive of subsequent clinical relapse. Analysis of the ALDH activity of CD34+CD38− cells potentially offers a patient-specific means by which to clinically assess response at the level of the LSC. The ability to separate LSCs from normal HSCs within the same individual may also facilitate better identification of therapeutic targets and resistance mechanisms. These data provide some of the first evidence supporting the clinical relevance of LSCs. Disclosures: Sharkis: Aldagen: Patents & Royalties. Jones:Aldagen: Patents & Royalties.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3520-3520
Author(s):  
Takeshi Yamada ◽  
Chun Shik Park ◽  
Ye Shen ◽  
H. Daniel Lacorazza

Abstract Abstract 3520 The G0/G1 switch gene 2 (G0S2) was originally identified in human blood mononuclear cells as an early response gene. We recently reported that G0S2 maintains quiescence in hematopoietic stem cells (Yamada et al., Plos One, 2012). In addition to hematopoietic stem cells, G0S2 also inhibits proliferation of lymphocytes and hematopoietic progenitor cells. Methylation of the G0S2 gene has been reported in lung, and head and neck cancer, suggesting that G0S2 has potential tumor suppressor function in solid tumors. However, the role of G0S2 in hematological malignancies has not been studied yet. To evaluate the impact of DNA demetylation of the G0S2 gene in human leukemia, we cultured a panel of myeloid (HEL, K562, HL-60, Kasumi) and lymphoid human leukemia cell lines (Jurkat, DND41, and H9) with 10 μM of 5-Azacytidine (5-Aza). Treatment with 5-Aza led to a significant increase of G0S2 expression in K562 cells (24.1-fold) and HL-60 cells (4.9-fold) and reduced proliferation. We show that retroviral overexpression of G0S2 in K562 cells caused inhibition of proliferation. Conversely, gene silencing of G0S2 in 5-Aza-treated K562 cells increased proliferation. Differentiation of K562 with hemin and HL-60 with all-trans retinoic acid was associated with an increase of G0S2 expression of 6.7-fold and 9.9-fold, respectively. Taken together, gain- and loss-of function studies revealed that G0S2 regulates cell proliferation and differentiation in human myeloid leukemia cells. Since G0S2 does not have a direct known function on the cell cycle, we hypothesized that G0S2 interacts with proteins involved in the control of cell proliferation. Mass spectrometric analysis of proteins pulled down with G0S2 in hematopoietic cells revealed that nucleolin, a molecule known to regulate ribosome biogenesis, physically interacts with G0S2 resulting in a perinuclear retention of nucleolin. This interaction was confirmed in K562 cells overexpressing G0S2 by reciprocal co-immunoprecipitation. This data suggest that silencing of G0S2 in leukemic cells prevents sequestration of nucleolin in the cytosol inhibiting its pro-proliferation functions. Collectively, our studies uncovered an important inhibitory role of G0S2 in the proliferation of leukemic cells, suggesting a possible tumor suppressor function in myeloid malignancies. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5782-5782 ◽  
Author(s):  
Mia Eriksson ◽  
Pablo Peña ◽  
Marion Chapellier ◽  
Carl Högberg ◽  
Thoas Fioretos ◽  
...  

Abstract Acute myeloid leukemia (AML) is a fatal disease that contains rare immature cells with self-renewal and leukemia-initiating capacity, known as leukemia stem cells (LSCs). Because current therapies are inefficient in eradicating LSCs, new therapies are warranted that efficiently target this cell population. One strategy being explored towards new therapies is identification of novel therapeutic targets on the cell surface of AML stem cells. To identify a cell surface protein upregulated on LSC, we used flow cytometry to measure the expression level of 13 cell surface proteins on immature leukemic cells from 18 AML patients and bone marrow cells from 7 healthy controls. We identified Toll-like receptor 1 (TLR1) as significantly upregulated in the immature CD34+CD38-compartment compared to corresponding normal cells that were almost devoid of TLR1 expression. These findings are consistent with elevated TLR1 mRNA levels observed in MDS patients (Wei et al, Leukemia, 2013). To evaluate the role of Tlr1 on immature leukemic cells, we performed shRNA-mediated inhibition of Tlr1 in MLL-AF9-expressing murine c-Kit+ leukemic cells. By using lentiviral vectors expressing the Tlr1-shRNAs along with a puromycin resistance gene, we identified two unique shRNAs that successfully suppressed the Tlr1 transcript and protein expression in comparison to a shRNA control. We next co-expressed the two Tlr1-shRNAs along with GFP in leukemia cells and monitored the percentage of GFP positive cells over time. Expression of the Tlr1-shRNAs resulted in strong depletion of the leukemic cells both in vitro and in vivo relative to the control shRNA. These findings suggest that TLR1 is important for the growth and survival of leukemic cells. To further address the role of Tlr1 on leukemic cells, we stimulated the leukemic cells with Pam3CSK4, a specific Tlr1/2 agonist. Pam3CSK4 alone was added to in vitro cultures of leukemic cells for three days, leading to increased survival and a slight increased number of leukemic cells. However, flow cytometric analysis revealed a differentiation shift of cells stimulated with Pam3CSK4 indicated by a decreased expression of the immature cell surface marker c-Kit and an increased expression of the myeloid linage marker Mac-1. To evaluate how Pam3CSK4 affects LSCs, we added Pam3CSK4 to ex vivo-cultures of leukemic cells for 3 days and then transplanted the cells into sublethally irradiated mice. Blood samples after two weeks showed a decreased leukemic burden in mice receiving Pam3CSK4-stimulated cells compared to controls. These findings suggest that enforced TLR1/TLR2-signaling causes differentiation of LSCs. In summary, this study demonstrates that TLR1 is upregulated on AML-stem cell enriched patient cells and that TLR1 expression is finely balanced to maintain LSCs. More specifically, our data suggest that the leukemic cells require Tlr1-expression for survival, but enhanced Tlr1/Tlr2-activation force the LSCs into differentiation. Hence, our study suggests that approaches aiming either for inhibition or enforced activation of TLR1 in AML should be explored further towards a potential new AML therapy. Collectively, we here identify TLR1 as a novel and promising candidate therapeutic target in AML. Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Vol 20 (23) ◽  
pp. 5826 ◽  
Author(s):  
Tobias Gluexam ◽  
Alexander M. Grandits ◽  
Angela Schlerka ◽  
Chi Huu Nguyen ◽  
Julia Etzler ◽  
...  

The neuropeptide CGRP, acting through the G-protein coupled receptor CALCRL and its coreceptor RAMP1, plays a key role in migraines, which has led to the clinical development of several inhibitory compounds. Recently, high CALCRL expression has been shown to be associated with a poor prognosis in acute myeloid leukemia (AML). We investigate, therefore, the functional role of the CGRP-CALCRL axis in AML. To this end, in silico analyses, human AML cell lines, primary patient samples, and a C57BL/6-based mouse model of AML are used. We find that CALCRL is up-regulated at relapse of AML, in leukemic stem cells (LSCs) versus bulk leukemic cells, and in LSCs versus normal hematopoietic stem cells. CGRP protects receptor-positive AML cell lines and primary AML samples from apoptosis induced by cytostatic drugs used in AML therapy, and this effect is inhibited by specific antagonists. Furthermore, the CGRP antagonist olcegepant increases differentiation and reduces the leukemic burden as well as key stem cell properties in a mouse model of AML. These data provide a basis for further investigations into a possible role of CGRP-CALCRL inhibition in the therapy of AML.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3878-3878
Author(s):  
Yajing Chu ◽  
Yangpeng Chen ◽  
Huidong Guo ◽  
Mengke Li ◽  
Jun Shi ◽  
...  

Abstract Acute myeloid leukemia (AML) is the most frequent and heterogeneous malignancy in adult leukemic patients. Genome-wide analyses revealed that genes involved in epigenetic modifications are among the most often re-occurring mutations in AML, suggesting a crucial role of epigenetic regulation in leukemogenesis and leukemia relapse. As a mammalian lysine methyltransferase, SUV39H1 catalyzes di- and tri-methylation of histone 3 lysine 9, and is the predominant H3K9 methyltransferase expressed in hematopoietic stem cells (HSCs). Previous studies have shown that in MLL-rearranged leukemic cells, the normal localization of Suv39h1 and Sirt1 was interrupted due to the DNA binding of Dot1L to DNA. However, the biological role of SUV39H1 in MLL-rearranged leukemia remains unexplored. In this study, we investigated the role and the underlying mechanism of Suv39h1 during leukemia progression. By analyzing the clinical databases, we found a significantly reduced expression of SUV39H1 in AML cells in comparison with normal bone marrow (BM) cells. More importantly, we found that low expression of SUV39H1 predicts poorer survival in AML patients. In MLL-fusion induced AML mouse models (MLL-AF9/MA9 and MLL-NRIP3/MN3), Suv39h1 also exhibited lower expression in leukemia stem cells (LSCs, defined as c-Kit+ or Lin-Sca1-IL-7R-c-Kit+CD34+CD16/32+ L-GMP cells) when compared with normal HSPCs. These data suggest a potential role of SUV39H1 in leukemic progression and/or maintenance. To explore if Suv39h1 functions as a tumor suppressor in MLL-fusion driven leukemogenesis, we overexpressed Suv39h1 in MA9 BM AML cells. Western blotting analysis confirmed the overexpression of Suv39h1 with a moderate increase in global H3K9me3 levels in Suv39h1-overexpressed (SUV-OE) MA9 AML cells. Interestingly, Suv39h1 overexpression prolonged the survival of recipient AML mice in both secondary and tertiary transplantation groups. Both the frequency and the absolute number of phenotypic LSCs in BM and SP were significantly reduced in SUV-OE groups as manifested by flow cytometry. Furthermore, limiting dilution assays revealed a significant six-fold decrease of functional LSCs in SUV-OE AML cells (1/314 LSCs in SUV-OE AML cells vs 1/56 in controls). Cell cycle analysis of control and SUV-OE LSCs from BM revealed a significantly decreased proportion of SUV-OE cells in the S/G2/M phase concordant by an increased proportion of G0/G1 phases when compared with control cells. In contrast, a similar apoptotic ratio of L-GMPs in BM was observed between control and SUV-OE groups. Taken together, these data demonstrated that overexpressing Suv39h1 in AML cells reduces the frequency of functional LSCs by suppression its proliferation. To explore the underlying mechanisms, gene expression profiles were assessed by RNA-Seq of SUV-OE and control mouse AML c-Kit+ cells. A total of 69 genes were differentially expressed with fold change ≥ 4. Among these genes, Hoxb13 was of particular interesting since it was reported to be recurrently mutated in several types of cancers including leukemia. ChIP-qPCR revealed a two-fold increase of H3K9m3 distribution at the promoter of Hoxb13 in SUV-OE groups, indicating Hoxb13 may be a direct downstream target of Suv39h1. Restoring the expression of Hoxb13 in SUV-OE AML cells diminished the effect of SUV-OE-mediated prolonged survival of SUV-OE AML mice. Interestingly, overexpression of Hoxb13 alone in MA9 cells had no significant effect on the survival of MA9 AML mice, indicating that Hoxb13 is a downstream effector of Suv39h1, rather than MA9, and Suv39h1 itself is a downstream mediator of MA9. To summarize, we here for the first time, demonstrate that Suv39h1 is significantly down-regulated in AMLs and could function as a tumor suppressor in MLL-rearranged leukemia by epigenetically inhibiting the Hoxb13 expression. The molecular mechanism mediated by Suv39h1-Hoxb13 axis in tumor suppression could potentially provide us novel therapeutic strategies for MLL-rearranged leukemia. YJ.C, YP.C and HD.G contributed equally to this work. Corresponding authors: WP.Y and MJ.X. Figure. Figure. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document