Robust patient-derived xenografts of MDS/MPN overlap syndromes capture the unique characteristics of CMML and JMML

A Yoshimi, ME Balasis, A Vedder… - Blood, The Journal …, 2017 - ashpublications.org
A Yoshimi, ME Balasis, A Vedder, K Feldman, Y Ma, H Zhang, SCW Lee, C Letson…
Blood, The Journal of the American Society of Hematology, 2017ashpublications.org
Chronic myelomonocytic leukemia (CMML) and juvenile myelomonocytic leukemia (JMML)
are myelodysplastic syndrome (MDS)/myeloproliferative neoplasm (MPN) overlap disorders
characterized by monocytosis, myelodysplasia, and a characteristic hypersensitivity to
granulocyte-macrophage colony-stimulating factor (GM-CSF). Currently, there are no
available disease-modifying therapies for CMML, nor are there preclinical models that fully
recapitulate the unique features of CMML. Through use of immunocompromised mice with …
Abstract
Chronic myelomonocytic leukemia (CMML) and juvenile myelomonocytic leukemia (JMML) are myelodysplastic syndrome (MDS)/myeloproliferative neoplasm (MPN) overlap disorders characterized by monocytosis, myelodysplasia, and a characteristic hypersensitivity to granulocyte-macrophage colony-stimulating factor (GM-CSF). Currently, there are no available disease-modifying therapies for CMML, nor are there preclinical models that fully recapitulate the unique features of CMML. Through use of immunocompromised mice with transgenic expression of human GM-CSF, interleukin-3, and stem cell factor in a NOD/SCID-IL2Rγnull background (NSGS mice), we demonstrate remarkable engraftment of CMML and JMML providing the first examples of serially transplantable and genetically accurate models of CMML. Xenotransplantation of CD34+ cells (n = 8 patients) or unfractionated bone marrow (BM) or peripheral blood mononuclear cells (n = 10) resulted in robust engraftment of CMML in BM, spleen, liver, and lung of recipients (n = 82 total mice). Engrafted cells were myeloid-restricted and matched the immunophenotype, morphology, and genetic mutations of the corresponding patient. Similar levels of engraftment were seen upon serial transplantation of human CD34+ cells in secondary NSGS recipients (2/5 patients, 6/11 mice), demonstrating the durability of CMML grafts and functionally validating CD34+ cells as harboring the disease-initiating compartment in vivo. Successful engraftments of JMML primary samples were also achieved in all NSGS recipients (n = 4 patients, n = 12 mice). Engraftment of CMML and JMML resulted in overt phenotypic abnormalities and lethality in recipients, which facilitated evaluation of the JAK2/FLT3 inhibitor pacritinib in vivo. These data reveal that NSGS mice support the development of CMML and JMML disease-initiating and mature leukemic cells in vivo, allowing creation of genetically accurate preclinical models of these disorders.
ashpublications.org