Although it is well-established that many malignancies can be transplanted, there is little evidence to demonstrate that a pre-malignant disease, such as myelodysplastic syndrome (MDS), can be transplanted and subsequently undergo malignant transformation in vivo. We used NUP98-HOXD13 (NHD13) mouse bone marrow to demonstrate that MDS could be transferred by a long-term repopulating cell. Recipients of the NHD13 MDS bone marrow displayed all of the features typical of MDS, including peripheral blood cytopenias, dysplasia, and transformation to AML. We identified the myelodysplastic syndrome-initiating cell (M-IC) in the lineage negative (linneg) BM population, and subsequently further defined the M-IC cell in terms of cell surface markers. In normal hematopoiesis, the long term hematopoietic repopulating cell is contained within the LinnegSca1+Kit+ (LSK) population. Our experiments demonstrated that the M-IC resides in the LSK long term repopulating compartment. One reason for the difficulty in developing effective treatments for myelodysplastic syndrome is that there are no myelodysplastic syndrome cell lines which can be used to model or study the disease. Although numerous investigators have attempted to develop xenograft models for myelodysplastic syndrome, these attempts have met with little success. Given that the NHD13 mice develop a highly penetrant myelodysplastic syndrome that closely resembles the human disease, we have begun studies to determine if these mice are a useful pre-clinical model for myelodysplastic syndrome. Our initial studies have used the DNA-methyltransferase inhibitor 5-azacytidine. Our pilot study included 3 groups of mice [NHD13 mice injected with 5-azacytidine (n=6), NHD13 mice injected with saline (n=4), and WT mice injected with 5-azacytidine (n=4)]. After 16 weeks of therapy, the results appeared promising, as the treated NHD13 mice showed a significant increase in hemoglobin compared to the saline treated NHD13 mice (2.21 +/- 1.47 g/dL vs 0.13 +/- 0.66 g/dL, p=0.02). Unfortunately, three of the NHD13 treated mice were found dead in the following two months, and we were not able to determine the cause of death for these mice. The three remaining mice had gradual decreases in hemoglobin over the next 12 weeks, and were close to baseline hemoglobin levels. Nonetheless the observation that these mice had stable hemoglobin levels after 28 weeks was encouraging. To determine whether we were achieving levels of 5-azacytidine adequate to cause cytosine demethylation, we examined global and gene-specific methylation status, in collaboration with Dr. J.P. Issa (M.D. Anderson);these experiments demonstrated hypermethylation in the NHD13 mice compared to controls, and partial reversal of the hypermethylation with 5-azacytidine treatment. Because the experiments discussed above used transgenic mice, effective treatment with 5-azacytidine could not replace the MDS bone marrow with completely normal (ie, wildtype or WT) bone marrow, since all of the bone marrow was transgenic. Therefore, in order to distinguish improvement in peripheral blood cytopenia due to differentiation of the MDS clone from elimination of the MDS clone, we have repeated the experiments using chimeric mice, that have both WT and NHD13 bone marrow. These repeat experiments have been performed using Decitabine (DAC), a related DNA methyltransferase inhibitor. Preliminary studies are very encouraging. After 4 monthly cycles of therapy, 5/5 control mice (treated with saline only) have died due to leukemia or MDS, whereas 7/7 treated mice have survived. Furthermore, the DAC-treated mice have shown improvements in peripheral blood counts. The major portion of this data is unpublished, a small amount has been published in 2010. Despite the recent FDA approval of three drugs for MDS, the only curative option for patients with MDS is allogeneic hematopoietic stem cell transplantation (HSCT). Allogeneic HSCT has two essential components, high-dose cytotoxic chemo-radiotherapy, and an immune-mediated graft versus host (GVH), or graft versus tumor (GVT) effect . However, the relative contributions of high dose cytotoxic therapy and GVT are not well established in MDS. We propose to use transplantation of the NHD13 mice as a means to investigate this question. The goals of our initial experiments are two-fold. First, we plan to determine if NHD13 mice with MDS can enter a long term remission through the use of lethal irradiation followed by transplantation with WT syngeneic hematopoietic stem cells (HSC). If mice enter a long term remission through lethal irradiation and HSC transfer, we will conclude that long term disease remission in this model can be achieved via high dose cytotoxic therapy alone, and that the contribution of GVT is not required. Our second goal is to identify a radiation dose that is effective at inducing a brief or partial remission, but ineffective at inducing a long term remission. This dose schedule will then be used to investigate a role for GVT effect in the treatment of MDS. Remission will be defined as normal peripheral blood counts, and no evidence of host-derived hematopoiesis, for 26-52 weeks post BMT. The radiation dose schedule effective at inducing a brief or partial remission will be employed for mismatched HSC transplants that will allow for a GVT effect. C3H.SW mice are identical to C57Bl6 mice at the major histocompatibility loci, but have numerous mismatches at minor histocompatibility loci108. For this reason, transplantation of C3H.SW donor cells into C57Bl6 host mice has been used to study GVH and GVT108. We will transplant T-cell depleted or whole C3H.SW BMNC into irradiated NHD13 C57Bl6 mice. Mice will be evaluated at timed intervals as described above for engraftment and peripheral blood hematopoiesis. In addition, mice will be weighed weekly as a means of assessing GVH. Mice that develop clinical signs consistent with severe GVH (weight loss, ruffled fur, lethargy), without signs of leukemia or MDS will be euthanized and evaluated for histologic signs of GVH. Our initial experiments, using either 650 or 1000 CGy of radiation, indicated that 650 CGy was ineffective, but that 1000 CGy could induce a remission of 26-38 weeks. However, despite this period of prolonged remission, all of the mice have ultimately relapsed, indicating that this myeloablative therapy was not curative. We have begun experiments with the minor histo-compatibility mismatched BM. The initial experiments showed little GVH effect, and subsequent experiments using higher doses of BM and T cells showed severe GVH. We are now repeating experiments with a lower dose of T cells, in an effort to induce a GVT effect with minimal GVH. These studies have not yet been published. In addition to the experiments outlined above, we have transferred NHD13 mice to colleagues at several academic institutions, and have licensed NHD13 mice to at least two separate biotech companies for pre-clinical studies. These colleagues have plans to treat NHD13 mice with a variety of agents, including histone deacetylase inhibitors, apoptosis inhibitors, and angiogenesis inhibitors. One of these companies has an abstract that will be presented in Dec. 2009.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Investigator-Initiated Intramural Research Projects (ZIA)
Project #
1ZIABC010983-03
Application #
8157558
Study Section
Project Start
Project End
Budget Start
Budget End
Support Year
3
Fiscal Year
2010
Total Cost
$379,492
Indirect Cost
Name
National Cancer Institute Division of Basic Sciences
Department
Type
DUNS #
City
State
Country
Zip Code
Monlish, Darlene A; Bhatt, Sima T; Duncavage, Eric J et al. (2018) Loss of Toll-like receptor 2 results in accelerated leukemogenesis in the NUP98-HOXD13 mouse model of MDS. Blood 131:1032-1035
Bhagat, Tushar D; Chen, Si; Bartenstein, Matthias et al. (2017) Epigenetically Aberrant Stroma in MDS Propagates Disease via Wnt/?-Catenin Activation. Cancer Res 77:4846-4857
Ma, Hayley S; Greenblatt, Sarah M; Shirley, Courtney M et al. (2016) All-trans retinoic acid synergizes with FLT3 inhibition to eliminate FLT3/ITD+ leukemia stem cells in vitro and in vivo. Blood 127:2867-78
Maegawa, Shinji; Gough, Sheryl M; Watanabe-Okochi, Naoko et al. (2014) Age-related epigenetic drift in the pathogenesis of MDS and AML. Genome Res 24:580-91
Santos, Margarida A; Faryabi, Robert B; Ergen, Aysegul V et al. (2014) DNA-damage-induced differentiation of leukaemic cells as an anti-cancer barrier. Nature 514:107-11
Gough, Sheryl M; Lee, Fan; Yang, Fan et al. (2014) NUP98-PHF23 is a chromatin-modifying oncoprotein that causes a wide array of leukemias sensitive to inhibition of PHD histone reader function. Cancer Discov 4:564-77
Gough, Sheryl M; Chung, Yang Jo; Aplan, Peter D (2012) Depletion of cytotoxic T-cells does not protect NUP98-HOXD13 mice from myelodysplastic syndrome but reveals a modest tumor immunosurveillance effect. PLoS One 7:e36876
Beachy, Sarah H; Aplan, Peter D (2010) Mouse models of myelodysplastic syndromes. Hematol Oncol Clin North Am 24:361-75
Estécio, Marcos R H; Gallegos, Juan; Vallot, Céline et al. (2010) Genome architecture marked by retrotransposons modulates predisposition to DNA methylation in cancer. Genome Res 20:1369-82
Lin, Ying-Wei; Beharry, Zanna M; Hill, Elizabeth G et al. (2010) A small molecule inhibitor of Pim protein kinases blocks the growth of precursor T-cell lymphoblastic leukemia/lymphoma. Blood 115:824-33

Showing the most recent 10 out of 13 publications