Incorporation of the Non-Human Sialic Acid Neu5Gc into Human Leukemic Cells and Targeting by Natural Human Anti-Neu5Gc Antibodies.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3866-3866
Author(s):  
Dzung H. Nguyen ◽  
Pam Tangvoranuntakul ◽  
Ajit Varki

Abstract Humans are incapable of synthesizing the common mammalian cell surface sialic acid N-glycolylneuraminic acid (Neu5Gc), due to an irreversible genetic mutation that occurred after our last common ancestor with great apes. Despite this, we found trace levels of Neu5Gc in certain normal human tissues, and higher levels in fetal and malignant tissues, apparently due to incorporation from dietary sources. Circulating anti-Neu5Gc antibodies also occur in most normal humans, with marked individual variations in levels. We now show that while normal human blood cells metabolically incorporate very little free Neu5Gc from the culture medium, human leukemic cell lines do so efficiently, displaying it on their cell surfaces, as detected by flow cytometry using a monospecific polyclonal chicken antibody, and confirmed chemically by HPLC. Specific deposition of IgG from Neu5Gc-reactive human sera onto Neu5Gc-expressing leukemic cells could be demonstrated. This was associated with lytic cell killing, apoptosis, and antibody-dependent cell-mediated cytotoxicity. Human sera with low levels of anti-Neu5Gc antibodies did not mediate such effects. These data show for the first time that the “natural” anti-Neu5Gc antibodies found in normal humans can be functionally active. The selective incorporation of Neu5Gc into leukemic cells could provide an approach for targeting leukemic cells in vivo, via naturally occurring Neu5Gc-specific antibodies. However, in contrast to unstimulated blood cells, activated human T-lymphocytes also incorporated some Neu5Gc, albeit to a lesser degree than leukemic cells, allowing IgG binding and complement deposition. Thus, exposure of rapidly dividing activated T cells to Neu5Gc could potentially affect ongoing immune responses. Since incorporation into leukemic cells was most efficient, it may still be possible to define a therapeutic window to selectively target malignant cells.

1982 ◽  
Vol 28 (10) ◽  
pp. 1127-1132 ◽  
Author(s):  
Ching Y. Lo ◽  
Hugh B. Fackrell ◽  
Gary M. Barei

When rabbits were injected with the heat-denatured alpha toxin (toxoid) of Staphylococcus aureus, the immune response was demonstrated by an increase in antitoxin that fixed complement. Such antitoxin was detected in 72% of normal human sera. After fractionation of the antitoxin into two types (the antibinding antibodies and the indirect hemagglutinating antibodies), both types of antibodies were found to fix complement in the standard serological complement fixation test. In addition, the indirect hemagglutinating antibodies were capable of fixing complement when the antigen (alpha toxin or toxoid) was covalently or noncovalently bound to erythrocyte membranes. The fixation of complement by membrane-bound immune complexes did not result in lysis of the carrier erythrocytes. The prevalence of complement-fixing antitoxin in normal humans and animals raised the concern that the outcome of in vivo experiments involving alpha toxin could be influenced by the immune status of the host.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 566-566
Author(s):  
Ifat Geron ◽  
Angela Maria Savino ◽  
Noa Tal ◽  
John Brown ◽  
Virginia A Turati ◽  
...  

Abstract Philadelphia-like (Ph-like) B-cell precursor acute lymphoblastic leukemia (BCP-ALL) is a subgroup of BCP-ALL with an expression pattern similar to BCR-ABL+ BCP-ALL that is associated with poor prognosis. Aberrant expression of CRLF2 in BCP-ALL constitutes the majority of Ph-like BCP-ALL cases. CRLF2 is a receptor subunit that together with the IL7RA subunit comprises the receptor of the proinflammatory cytokine TSLP. Though activation of the IL7R pathway is commonly associated with T-cell malignancies, we previously described IL7RA-activating mutations in BCP-ALL predominantly in the context of CRLF2 aberrant expression (Shochat C. et al. J. Exp. Med. 2011). Here we aimed to test the role of aberrations in CRLF2 and IL7RA in the development of Ph-Like BCP-ALL. Both CRLF2 and TSLP differ extensively between mice and human in amino acid sequence and in lineage expression pattern; loss-of-function germline mutations in IL7RA are associated with lack of B and T cells in mice but with lack of only T cells in humans. Hence, we chose to test the hypothesis that activation of CRLF2/IL7RA contributes to the development of Ph-like BCP-ALL in the context of human lymphopoiesis by using a human xenograft system. To aberrantly activate TSLP/IL7 signaling, we transduced cord-blood (CB) CD34+ hematopoietic progenitors with a set of lentiviral vectors carrying CRLF2 and/or IL7RA [(wild type (IL7RAwt) or IL7R bearing an activating mutation (IL7RAins)] under a B-cell promoter/enhancer (to accentuate B-cell lineage expression). The backbone vector (BB) expressing GFP was used as a control. Transduced CB cells were transplanted into NOD/LtSz-scid IL2Rγnull (NSG) mice and engrafted cells were analyzed 24-30 weeks after transplantation. To test for self-renewal capacity, BM cells from primary engrafted mice were serially transplanted into secondary recipients and the occurrence of human engraftment was tested 24-30 weeks after transplantation. Enforced expression of activated IL7RA with or without CRLF2 led to a significant block in B-cell development at the B-cell progenitor stage (CD19+CD10+sIgM-) in vivo resembling the differentiation stage of leukemic cells (figure Bi). Repertoire sequencing of CD10+CD19+-transduced cells that were sorted from BM of transplanted mice revealed a significantly higher population of DJ-rearranged cells in the CRLF2-IL7RAins-transduced population than in BB-transduced cells (mean ratio of DJ/total rearrangement: BB:0.35+/-0.024, CRLF2-IL7RAins:0.76+/- 0.07, p=0.039, n=3 paired cord blood), in agreement with the early differentiation block phenotype measured by immunophenotyping. These cells furthermore exhibited a Ph-like gene expression pattern when compared to BB-transduced cells in gene set enrichment analysis. Overexpression of IL7RA alone significantly enhanced the early-B fraction (CD19+CD10+CD34+) in the BM of transplanted mice (figure Bii). Additionally, aberrant expression of IL7RA enhanced self-renewal capacity as was evident by an increased ability of the transduced cells to engraft in secondary recipients (number of mice with detectable human engraftment out of secondary transplanted mice: BB:0/6, CRLF2-IL7RAwt:0/3, CRLF2-IL7RAinst:0/3, IL7RAwt-GFP:3/6, IL7RAins-GFP:5/8). Notably, in one case, secondary transplantation of IL7RAins-transduced CB triggered the development of acute BCP-ALL. The leukemic cells (CD19+CD10+CD34+sIgM-) were clonal as validated by V(D)J rearrangement (figure Cii), had the ability to further propagate in serial transplantations and gained secondary Ph-like BCP-ALL-characteristic chromosomal deletions in the short arm of chromosome 9 (in the region including the genes for CDKN2A/B, PAX5 and JAK2) and the short arm of chromosome 7 (the region including IKZF1) (figure Ciii). These results support the hypothesis that aberrant activation of the CRLF2/IL7RA pathway in human B-cell lineage progenitors creates a pre-leukemic state by arresting differentiation of B-cell progenitors, instating Ph-like expression pattern and inducing self-renewal. This is the first model of de novo Ph-like BCP-ALL development from normal human hematopoietic progenitors in vivo. Additionally, we present here a first direct in vivo demonstration of a role for IL7 in human B-cell development. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1996 ◽  
Vol 88 (5) ◽  
pp. 1788-1795 ◽  
Author(s):  
Q Zhao ◽  
X Song ◽  
T Waldschmidt ◽  
E Fisher ◽  
AM Krieg

Abstract The use of antisense oligonucleotides as tools for modulating gene expression represents a novel strategy for designing drugs to treat a variety of diseases. Several factors, including cellular uptake and internalization of the oligonucleotides, are important parameters in determining the effectiveness of antisense agents such as therapeutic drugs. We have studied oligonucleotides uptake in normal and leukemic human hematopoietic cells, such as peripheral blood, bone marrow (BM), and HL-60 cell line; and have found that, in normal human blood and BM, myeloid cells and B cells preferably took up more oligonucleotides than T cells. There was no marked difference in oligonucleotide uptake between CD4+ helper T cells and CD8+ cytolytic T cells. Leukemic cells had greater oligonucleotide uptake than their normal counterparts. Furthermore, oligonucleotide uptake was closely related to cell activation status and can be modulated by growth factors or inhibitors. These studies provide a basis for using oligonucleotides as therapeutic drugs both in vitro and in vivo.


2012 ◽  
Vol 88 (1) ◽  
pp. 44-51 ◽  
Author(s):  
Robert S. Franco ◽  
M. Estela Puchulu-Campanella ◽  
Latorya A. Barber ◽  
Mary B. Palascak ◽  
Clinton H. Joiner ◽  
...  

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1466-1466
Author(s):  
Nancy Hurtado-Ziola ◽  
Justin L. Sonnenburg ◽  
Ajit Varki

Abstract The Siglecs (Sialic acid-binding Immunoglobulin Superfamily Lectins) are a recently discovered family of mammalian glycan-binding proteins that have been shown to recognize the terminal sialic acids of glycoproteins and glycolipids. The CD33-Related Siglecs (CD33rSiglecs, namely Siglec-3, -5 through -11 and -XII in humans) are a subgroup of these molecules, which are thought to be primarily expressed on cells of the innate immune system. All CD33rSiglecs are type-1 transmembrane proteins with an N-terminal sialic acid-recognizing V-set domain followed by a variable number of C-2 set domains, a transmembrane region and a cytosolic C-terminal domain that usually has two tyrosine-based signaling motifs, one of which conforms to a canonical negative regulatory ITIM motif. Although the true function of the CD33rSiglecs has yet to be discovered, available data are most consistent with an inhibitory signaling role in the innate immune response, mediated by recognition of host sialic acids as “self”. CD33rSiglecs also interact with sialic acids on the same cell surface, typically resulting in “masking” of their sialic acid-binding sites. Our recent studies have shown that humans and non-human primates have a similar clustered localization of CD33rSiglec genes, and that true orthologs can generally be identified within each cluster (Angata et al., PNAS, in press). However, humans no longer express CMP-sialic acid hydroxylase (CMAH) the enzyme required to generate one of the potential CD33rSiglec sialic acid ligands called N-glycolylneuraminic acid (Neu5Gc), from its precursor N-acetylneuraminic acid (Neu5Ac). This genetic change occurred after our last common ancestor with the great apes, and dramatically altered the “Sialome” (the sialic acid makeup of a specific species) of humans when compared to that of the great apes. While great ape blood cells express about equal amounts of Neu5Ac and Neu5Gc, human blood cells express almost exclusively Neu5Ac. We also recently discovered that preferential recognition of Neu5Gc is the ancestral condition of most or all of the great ape (chimpanzee and gorilla) CD33rSiglecs (Sonnenburg JL, Altheide TK, Varki A. Glycobiology.14:339–46, 2004). We therefore reasoned that the sudden and major change in the sialome of our hominid ancestors could have had a significant impact on the evolution, binding specificities and expression patterns of CD33rSiglecs. Indeed, we have found that all human CD33rSiglecs can recognize both Neu5Ac and Neu5Gc. This presumably represents an evolutionarily-selected “relaxation” in binding specificity that was necessary to “remask” the Siglecs that had lost their Neu5Gc ligands. Also, there are differences in CD33rSiglec expression on monocytes and neutrophils between humans and great apes (chimp, bonobo, gorilla and orangutan). Furthermore, while great ape cells often show multiple populations with different signal intensities, humans express a single bright peak for each Siglec in flow cytometry. Surprisingly, while humans showed almost no CD33rSiglec expression on lymphocytes, the great apes show a moderate to high expression of some Siglecs on these cells. Total leukocyte expression of some CD33rSiglecs also shows differences between humans and great apes. Overall, CD33rSiglecs appear to be rapidly evolving in primates, with an apparent further acceleration of changes in humans. Additional studies are needed to define the mechanistic details, as well as the implications for human health and disease.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2960-2960
Author(s):  
Oliver Christ ◽  
Wolfgang Eisterer ◽  
Xiaoyan Jiang ◽  
Emily Pang ◽  
Karen Leung ◽  
...  

Abstract Transplantation of sublethally irradiated NOD/SCID or NOD/SCID-β2microglobulin (β2m) null mice with cells from most chronic phase chronic myeloid leukemia (CML) patients results in the regeneration in the mice of primarily normal human hematopoietic cells. This is due to the usual predominance of normal cells within the most primitive subsets of bone marrow or blood cells in these patients. To date, no markers that allow the most primitive normal and leukemic cells to be differentially isolated from chronic phase CML samples have been identified except those reflecting an increased turnover of the leukemic cells. As an alternative approach to characterizing chronic phase CML stem cells, we have identified particular patient samples that contain predominantly leukemic LTC-ICs and have found that transplants of these samples regenerate a predominance of leukemic cells in both NOD/SCID and NOD/SCID-β2m null mice. To investigate the biological and phenotypic properties of CML cells that have short- and longterm repopulating activity, we transplanted sublethally irradiated NOD/SCID and NOD/SCID-β2m null mice with FACS-sorted subsets of lin- CML cells from 2 such samples and then monitored their output of cells in the bone marrow of the mice for up to 12 weeks. The CD34+CD38+ CML cells produced a rapid but transient wave of mainly myeloid progeny that peaked at 3 weeks whereas the CD34+CD38− cells produced a more delayed but persistent wave of cells in both types of mice that included some lymphoid progeny although the latter represented a markedly reduced proportion of the total relative to the cells produced by normal human bone marrow. These patterns were seen in both recipient genotypes but cell output was enhanced in NOD/SCID-β2m null mice as expected for short-term repopulating cells. In additional studies with 3 patients’ samples, both types of repopulating cells were found primarily in the aldehyde dehydrogenase-positive fraction based on their staining with BODIPY-labeled amino acetaldehyde. To test the feasibility of the CML xenograft model for evaluating novel treatments in vivo, groups of NOD/SCID mice repopulated to high levels with leukemic cells (49±8%) 7 weeks after being transplanted with 3x107 CD34+ CML cells, were injected with 50 mg/kg imatinib mesylate (or not) i.p. twice daily for 10 days. Bone marrow samples obtained from the imatinib mesylate-treated mice 2, 4, 12 and 22 weeks after initiation of this treatment, initially showed a more rapid and greater decline of human leukemic cells (>2-fold as assessed by both FACS and quantitative real-time PCR); however by 5 months after completion of the treatment, the level of human cells in the bone marrow of both the imatinib mesylate-treated and untreated mice was the same. Taken together, these findings demonstrate that the CML clone in chronic phase patients contains a similar hierarchy of short and longterm repopulating cells as is found in normal adult bone marrow, and that the CML repopulating cells have, in addition to their ability to sustain the clone, a greater innate resistance to the toxic effects that imatinib mesylate has in vivo on the majority population of more differentiated CML cells.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 982-982
Author(s):  
Shih-Shih Chen ◽  
Joseph J. Buggy ◽  
Betty Y Chang ◽  
Jan A. Burger ◽  
Nicholas Chiorazzi

Abstract Abstract 982 Bruton's tyrosine kinase (Btk) is critical for B-lymphocyte function, due to its involvement in key functions in B-cells, e.g., maturation, activation, and trafficking. These functions are mediated by several receptors, including the B-cell antigen receptor (BCR) and the chemokine receptor CXCR4. Both BCR and CXCR4 play important roles in chronic lymphocytic leukemia (CLL), providing leukemic cells with survival and proliferation advantages. Initial studies in CLL suggest that inhibiting Btk with PCI-32765 (Pharmacyclics, Inc) is an effective therapeutic, reducing the size of solid lymphoid tissues leading initially to lymphocytosis and eventually to decreased blood absolute lymphocyte counts (ALCs). To understand the effect of blocking Btk-mediated signaling in CLL, we utilized an accelerated adoptive transfer CLL mouse model, injecting 5×106 TCL1 leukemia cells into SCID mice that succumb 5–6 weeks after cell transfer. Total 45 mice were injected, separated into 3 groups, and then treated with PCI-32765, at either 2, 3 or 4 weeks after cell transfer, with 5 mice from each group receiving either vehicle control or PCI-32765 (5 or 25 mg/kg/day) in daily drinking water. Mice were bled to track changes in ALCs. Animals treated at 2-weeks post cell transfer with the suboptimal (5mg/kg/day) and optimal (25mg/kg/day) doses exhibited a transient lymphocytosis at day 4, with a 7- and 10-fold increase in circulating TCL1 leukemia cells, respectively (p=0.002). By day 7, these levels had fallen to those of untreated mice. Until week 6, mice receiving the optimal dose of PCI-32765 at week 2 and 3 but not week 4, appeared healthy and had significantly reduced ALCs (p<0.001). These mice had small or no lymph nodes (LNs) and significantly smaller livers and spleens with markedly reduced leukemic infiltration. In contrast, mice receiving vehicle control or sub-optimal dose of PCI-32765 exhibited lethargy, weight loss, and hunched posture; these animals had massive lymphocytosis, huge hepatosplenomegaly, and lymphadenopathy. Surprisingly, the delayed disease progression by PCI-32765 correlates with blocked CXCR4 surface membrane recycling in CLL cells. In addition, there were significantly repressed levels of phosphorylated phospholipase C-gamma 2 (PLCg2) in optimally treated mice. This is relevant because phosphorylation of PLCg2 by BTK influences chemokine-controlled cell migration, at least partially through levels of CXCL12 or its receptor CXCR4. In vivo studies of CLL patients suggest that peripheral blood cells bearing a CXCR4BRCD5DIM (R4BR) surface phenotype are more likely to re-enter lymphoid tissues, while cells expressing CXCR4DIMCD5BR (R4dim) have more likely recently left solid tissues. Here, we found spleen cells in optimally treated mice had significantly lower percentages of R4BR; while in the blood, there was an increased population of R4DIM cells. These data suggest that the smaller spleen and LN sizes of treated mice are due to promoted migration of CLL cells out of lymphoid tissues and blocked return from blood. In vivo effect of PCI-32765 on TCL1 cell proliferation was also evaluated. BrdU was injected in mice 24 hours prior to sacrifice. As TCL1 cells proliferate in lymphoid organs, significantly repressed BrdU incorporation was demonstrated in spleen and LN cells of optimally treated mice, consistent with PCI-32765 inhibiting proliferation. In addition, in support of enhanced migration of cells out of lymphoid tissues, there was an ∼3-4 fold increase in BrdU-labeled cells in blood after optimal PCI-32765 treatment. Finally, the effects of PCI-32765 on TCL1 cell homing was assessed by injecting SCID mice with 2.5×106 R4DIM blood cells from either controls or optimally treated mice. 24 hours later, mice engrafted with R4DIM cells from PCI-32765 treated animals had ∼30–50 fold fewer leukemic cells in spleen (p=0.018); and ∼1.5 fold increased CLL cells in blood (p=0.021), further supporting blocked homing by PCI-32765. Collectively, our data suggest that targeting Btk, its receptors, and the downstream targets that require its use delays CLL progression. This effect is, at least partially, due to repressed surface CXCR4 expression and blocked cell proliferation, mediated either directly in CLL cells or indirectly, by minimizing the likelihood of receiving trophic stimuli via the BCR or CXCR4 in a solid tissue microenvironment. Disclosures: Buggy: Pharmacyclics, Inc.: Employment. Chang:Pharmacyclics Inc: Employment. Burger:Pharmacyclics, Inc: Research Funding.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 286-286 ◽  
Author(s):  
John H. Bushweller ◽  
Anuradha Illendula ◽  
John Anto Pulikkan ◽  
Jolanta Grembecka ◽  
Paul Bradley ◽  
...  

Abstract Abstract 286 Emerging literature suggests that inability to cure cancers with current therapies may be attributed to a population of so-called cancer stem cells or cancer initiating cells that have long term self-renewal potential and can fully recapitulate tumor phenotype at time of relapse. The use of targeted therapies that inhibit the activity of oncoproteins directly present the potential for specificity in ablating tumor cells, including cancer stem cells, while having minimal impact on normal tissues. The gene encoding CBFβ (CBFB) is disrupted by the chromosome 16 inversion [inv(16)(p13q22)], associated with ∼12% of acute myeloid leukemia (AML) in humans, resulting in a fusion protein containing most of CBFβ fused to the coiled-coil tail region of smooth muscle myosin heavy chain (SMMHC). The CBFβ-SMMHC fusion protein acts as a dominant repressor of CBF function, binding RUNX1 and dysregulating the expression of multiple target genes required for normal hematopoiesis. Current AML treatment utilizing cytotoxic chemotherapy results in 45–65% five year overall survival but only 20% for patients older than 60. These data clearly indicate that the development of targeted therapy that can improve the therapeutic response for inv(16) AML patients is essential. The targeting of transcription factors in cancer therapy is a relatively new approach with tremendous potential. We developed a small molecule inhibitor, AI-10-49, that binds to the aberrant transcription factor CBFβ-SMMHC and disrupts its interaction with RUNX proteins. We have taken advantage of the oligomeric nature of CBFβ-SMMHC by developing dimeric compounds to achieve selectivity toward the fusion protein. Optimization of the linker length in these dimeric compounds shows a clear linker length dependence and a dramatic enhancement in activity for the dimeric versus monomeric inhibitors, providing validation for this approach. AI-10-49 is a result of this optimization and modifications to improve pharmacokinetic properties, resulting in a potent inhibitor with a half-life of ∼3 hours in mice. This compound displays selective toxicity to human leukemia cell lines with the inv(16) at a submicromolar dose (IC50=0.4μM). We have also shown no effect on growth of normal mouse or human bone marrow mononuclear cells. Using RT-PCR, we have shown dramatic derepression of the well-validated target genes CSF1R (9-fold) and RUNX3 (18-fold) in ME-1 (inv(16)) cells and little to no effect in Kasumi-1 (t(8;21)) and U937 cells. We have also shown increased apoptosis of the mouse preleukemic myeloid progenitor cells expressing CBFβ-SMMHC upon treatment ex vivo. The toxicity of AI-10-49 was tested in mice at the 25 mg/kg dosage employed for efficacy studies. Mice were dosed twice a day at 12 hour intervals for 1 week. No change in counts of white blood cells, red blood cells, or platelets was observed compared to formulation control. No changes were observed in granulocytes, monocytes, T cell, and B cell frequency by flow cytometry. Analysis of tissues after treatment showed no defects in spleen, kidney, liver, lungs, GI tract, heart, and bone marrow. A maximum tolerated dose (MTD) study showed no toxicity at doses up to 500 mg/kg, indicating the compound is very well tolerated. The efficacy of AI-10-49 was tested in vivo using the Cbfb+/MYH11/Mx1Cre/NrasG12D mouse model of inv(16) AML. Mice were transplanted with 5×105 leukemic cells, allowed 5 days for engraftment, and treated between days 5 and 15 post transplantation with 2 doses of vehicle or of AI-10-49 per day (25mg/kg/day). The control group (n=4) developed leukemia with a median latency of 4.6 weeks and full penetrance, while the test group (n=4) developed leukemia with a median latency of 8.4 weeks and incomplete penetrance. One mouse (25%) from the test group showed progressive decrease of leukemic cells to background levels at week 12 in peripheral blood, and remains healthy with no evidence of disease to date (week 16). Current efforts are focused on testing the compound efficacy of AI-10-49 with first line therapy drug Ara-C in mice. This work presents strong evidence of the identification of a small molecule that specifically targets CBFβ-SMMHC function and ablates cells expressing the leukemia fusion protein in vitro and in mice. This study provides proof of principle that oncogenic transcription factors can be targeted and may have significant impact on the treatment of inv(16) acute myeloid leukemia. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 390-390 ◽  
Author(s):  
John Anto Pulikkan ◽  
Anuradha Illendula ◽  
Jolanta Grembecka ◽  
Liting Xue ◽  
Roger Rajewski ◽  
...  

Abstract The leukemia fusion protein CBFβ-SMMHC, associated with acute myeloid leukemia (AML) with chromosome inversion inv(16)(p13q22), is a driver mutation in leukemia development. Studies by our laboratory and others have established that CBFβ-SMMHC outcompetes CBFβ for binding to RUNX1, deregulates RUNX1 transcription factor activity in hematopoiesis, and induces AML. Studies in mice and patient AML cells support the concept that CBFβ-SMMHC generates pre-leukemic myeloid progenitors, which acquire cooperating mutations to progress to leukemia. Current inv(16) AML treatment using non-selective cytotoxic chemotherapy results in a good initial response, but long-term survival is approximately 60%. This suggests that additional efforts are necessary for the development of improved therapeutic response for CBF AML patients. We have identified AI-4-57 as the active compound that inhibits CBFβ-SMMHC/RUNX1 binding (IC50= 22 uM), using a screen of the NCI diversity set library. AI-4-57 specifically binds to the CBFβ portion of CBFβ-SMMHC, as determined by NMR. This compound was modified in order to improve its potency and stability, and identified the divalent derivative AI-10-49 for further characterization. AI-10-49 showed increased potency (IC50= 0.26 µM) improved in vivo pharmacokinetics (serum t1/2 = 380 min), and enhanced activity in inv(16) positive ME-1 cells (IC50 = 0.6 uM). Importantly, AI-10-49 showed negligible activity (IC50>25 μM) in normal human bone marrow, defining a robust potential therapeutic window. Co-immunoprecipitation assays of ME-1 cells demonstrated that AI-10-49 (1µM for six hours) effectively and specifically dissociated RUNX1/CBFβ-SMMHC when compared to CBFβ/RUNX1 binding (Meandiss: 90% and 15%, respectively). Expression of RUNX1 target genes RUNX3, CSF1R, and CEBPA is repressed by CBFβ-SMMHC in inv(16) AML. The occupancy of RUNX1 in their promoters was significantly increased by chromatin-immunoprecipitation (8, 2.2, and 8 fold, respectively) in 6 hour treated (1µM AI-10-49) ME-1 cells, suggesting that CBFβ-SMMHC represses RUNX1 targets by blocking RUNX1 binding to target regulatory sites. In addition, RUNX3, CSF1R, and CEBPA expression increased 2 to 8 fold when compared to DMSO treated ME-1 cells. Importantly, RUNX1 occupancy and target expression changes were not observed in inv(16)-negative U937 cells. These data establish AI-10-49 selectivity in inhibiting CBFβ-SMMHC binding to RUNX1 and validate our approach of using bivalent inhibitors to achieve this specificity. To test AI-10-49 activity in vivo, mice were transplanted with leukemic cells expressing CBFβ-SMMHC and NrasG12D (from Cbfb+/MYH11:Ras+/G12Dknock-in mice), and treated, starting at day five post transplantation, with vehicle (DMSO) or 200 mg/kg AI-10-49 for ten days. The median latency of leukemia was delayed one fold in AI-10-49 treated mice (MLAI-10-49= 61 days, MLDMSO= 33.5 days, P=2.7x10-6; Log-rank test). In addition, toxicity assays revealed no detectable cumulative toxicity in mice treated with AI-10-49 for seven days. To test the efficacy of AI-10-49 in human inv(16) AML, the survival of four inv(16) and four normal karyotype AML patient samples were tested in 48 hour dose response assays. The viability of inv(16) patient cells was clearly reduced by AI-10-49 (viability: 50%, 10 μM AI-10-49/DMSO). In contrast, the viability of normal karyotype AML samples was unaffected at concentrations below 20µM. These studies show that AI-10-49 selectively inhibits viability in inv(16) AML blasts, while having negligible effects on AML blasts with normal karyotype or on normal human hematopoietic progenitors. Dysregulated gene expression is a hallmark of cancer and is particularly important for the maintenance of cancer stem cells, such as self-renewal, leading to relapse. The targeting of proteins that drive transcriptional dysregulation, so called “transcription therapy”, represents an avenue for drug development with immense potential. This study reports the development of a small molecule with high efficacy and specific in the inhibition of CBFβ-SMMHC activity while having a minimal effect on CBFβ function. In summary, AI-10-49 is a potent first generation CBFβ-SMMHC inhibitor that induces cell death in inv(16) AML cells and establishes a proof-of-principle that transcription factor fusion oncoproteins can be directly targeted for leukemia treatment. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document