Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer J ; 28(4): 257-262, 2022.
Article in English | MEDLINE | ID: mdl-35880934

ABSTRACT

ABSTRACT: Adoptive cellular therapies are making major strides in the treatment of cancer, both for hematologic and solid tumors. These cellular products include chimeric antigen receptor T cells and T-cell receptor-modified T cells, tumor-infiltrating lymphocytes, marrow-infiltrating T cells, natural killer cells as well as macrophage-based therapeutics. Advancement in genomics, computational biology, immunology, and cell therapy manufacturing has facilitated advancement of adoptive T cell therapies into the clinic, whereas clinical efficacy has driven Food and Drug Administration approvals. The growth of adoptive cellular therapy has, in turn, led to innovation in the preclinical models available, from ex vivo cell-based models to in vivo xenograft models of treatment. This review focuses on the development and application of in vitro models and in vivo models (cell line xenograft, humanized mice, and patient-derived xenograft models) that directly evaluate these human cellular products.


Subject(s)
Immunotherapy, Adoptive , Neoplasms , Animals , Cell Line, Tumor , Cell- and Tissue-Based Therapy , Humans , Mice , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes
3.
Oncotarget ; 8(58): 98853-98864, 2017 Nov 17.
Article in English | MEDLINE | ID: mdl-29228732

ABSTRACT

Mechanisms underlying the progression of Chronic Myeloid Leukemia (CML) from chronic phase to myeloid blast crisis are poorly understood. Our previous studies have suggested that overexpression of SETBP1 can drive this progression by conferring unlimited self-renewal capability to granulocyte macrophage progenitors (GMPs). Here we show that overexpression of Hoxa9 or Hoxa10, both transcriptional targets of Setbp1, is also sufficient to induce self-renewal of primary myeloid progenitors, causing their immortalization in culture. More importantly, both are able to cooperate with BCR/ABL to consistently induce transformation of mouse GMPs and development of aggressive leukemias resembling CML myeloid blast crisis, suggesting that either gene can drive CML progression by promoting the self-renewal of GMPs. We further identify Myb as a common critical target for Hoxa9 and Hoxa10 in inducing self-renewal of myeloid progenitors as Myb knockdown significantly reduced colony-forming potential of myeloid progenitors immortalized by the expression of either gene. Interestingly, Myb is also capable of immortalizing primary myeloid progenitors in culture and cooperating with BCR/ABL to induce leukemic transformation of mouse GMPs. Significantly increased levels of MYB transcript also were detected in all human CML blast crisis samples examined over chronic phase samples, further suggesting the possibility that MYB overexpression may play a prevalent role in driving human CML myeloid blast crisis development. In summary, our results identify overexpression of HOXA9, HOXA10, and MYB as critical drivers of CML progression, and suggest MYB as a key therapeutic target for inhibiting the self-renewal of leukemia-initiating cells in CML myeloid blast crisis patients.

4.
Oncotarget ; 7(52): 86300-86312, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27863435

ABSTRACT

SETBP1 missense mutations have been frequently identified in multiple myeloid neoplasms; however, their oncogenic potential remains unclear. Here we show that expression of Setbp1 mutants carrying two such mutations in mouse bone marrow progenitors efficiently induced development of acute myeloid leukemias (AMLs) in irradiated recipient mice with significantly shorter latencies and greater penetrance than expression of wild-type Setbp1, suggesting that these mutations are highly oncogenic. The increased oncogenicity of Setbp1 missense mutants could be due in part to their capability to drive significantly higher target gene transcription. We further identify Myb as a critical mediator of Setbp1-induced self-renewal as its knockdown caused efficient differentiation of myeloid progenitors immortalized by wild-type Setbp1 and Setbp1 missense mutants. Interestingly, Myb is also a direct transcriptional target of Setbp1 and Setbp1 missense mutants as they directly bind to the Myb locus in immortalized cells and dramatically activate a critical enhancer/promoter region of Myb in luciferase reporter assays. Furthermore, Myb knockdown in Setbp1 and Setbp1 missense mutations-induced AML cells also efficiently induced their differentiation in culture and significantly prolonged the survival of their secondary recipient mice, suggesting that targeting MYB pathway could be a promising strategy for treating human myeloid neoplasms with SETBP1 activation.


Subject(s)
Carrier Proteins/physiology , Leukemia, Myeloid, Acute/etiology , Nuclear Proteins/physiology , Proto-Oncogene Proteins c-myb/physiology , Animals , Carrier Proteins/genetics , Female , Homeobox A10 Proteins , Homeodomain Proteins/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Mutation, Missense , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-myb/antagonists & inhibitors
5.
Nat Genet ; 45(8): 942-6, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23832012

ABSTRACT

Here we report whole-exome sequencing of individuals with various myeloid malignancies and identify recurrent somatic mutations in SETBP1, consistent with a recent report on atypical chronic myeloid leukemia (aCML). Closely positioned somatic SETBP1 mutations encoding changes in Asp868, Ser869, Gly870, Ile871 and Asp880, which match germline mutations in Schinzel-Giedion syndrome (SGS), were detected in 17% of secondary acute myeloid leukemias (sAML) and 15% of chronic myelomonocytic leukemia (CMML) cases. These results from deep sequencing demonstrate a higher mutational detection rate than reported with conventional sequencing methodology. Mutant cases were associated with advanced age and monosomy 7/deletion 7q (-7/del(7q)) constituting poor prognostic factors. Analysis of serially collected samples indicated that SETBP1 mutations were acquired during leukemic evolution. Transduction with mutant Setbp1 led to the immortalization of mouse myeloid progenitors that showed enhanced proliferative capacity compared to cells transduced with wild-type Setbp1. Somatic mutations of SETBP1 seem to cause gain of function, are associated with myeloid leukemic transformation and convey poor prognosis in myelodysplastic syndromes (MDS) and CMML.


Subject(s)
Carrier Proteins/genetics , Leukemia, Myeloid/genetics , Mutation , Myeloproliferative Disorders/genetics , Nuclear Proteins/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Animals , Base Sequence , Bone Marrow Cells/metabolism , Cell Transformation, Neoplastic/genetics , Cluster Analysis , Exome , Female , High-Throughput Nucleotide Sequencing , Humans , Leukemia, Myeloid/mortality , Male , Mice , Middle Aged , Molecular Sequence Data , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/pathology , Myeloproliferative Disorders/mortality , T-Lymphocytes/metabolism , Young Adult
6.
Blood ; 119(25): 6099-108, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22566606

ABSTRACT

Acquisition of self-renewal capability by myeloid progenitors to become leukemic stem cells during myeloid leukemia development is poorly understood. Here, we show that Setbp1 overexpression efficiently confers self-renewal capability to myeloid progenitors in vitro, causing their immortalization in the presence of stem cell factor and IL-3. Self-renewal after immortalization requires continuous Setbp1 expression. We also found that Hoxa9 and Hoxa10 mRNA are present at dramatically higher levels in Setbp1-immortalized cells compared with other immortalized cells, and are induced shortly after Setbp1 expression in primary myeloid progenitors. Suppression of either gene in Setbp1-immortalized cells drastically reduces their colony-forming capability. Interestingly, Setbp1 protein associates with Hoxa9 and Hoxa10 promoters in chromatin immunoprecipitation assays in these cells, suggesting that both are direct transcriptional targets of Setbp1. Setbp1 also promotes self-renewal of myeloid progenitors in vivo as its coexpression with BCR/ABL transforms primary mouse myeloid progenitors, generating aggressive leukemias in recipient mice resembling chronic myelogenous leukemia (CML) myeloid blast crisis. Increased SETBP1 mRNA levels were also detected in a subset of CML advanced phase/blast crisis patients with high levels of HOXA9 and HOXA10 expression. Thus, Setbp1 activation represents a novel mechanism conferring self-renewal capability to myeloid progenitors in myeloid leukemia development.


Subject(s)
Carrier Proteins/physiology , Cell Proliferation , Homeodomain Proteins/genetics , Myeloid Progenitor Cells/physiology , Nuclear Proteins/physiology , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Homeobox A10 Proteins , Homeodomain Proteins/metabolism , Homeodomain Proteins/physiology , Humans , Mice , Mice, Inbred C57BL , Models, Biological , Myeloid Progenitor Cells/metabolism , NIH 3T3 Cells , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcriptional Activation/physiology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...