Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Immunol Immunother ; 71(2): 473-489, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34247273

ABSTRACT

Cluster of differentiation 47 (CD47) is a transmembrane protein ubiquitously expressed on human cells but overexpressed on many different tumor cells. The interaction of CD47 with signal-regulatory protein alpha (SIRPα) triggers a "don't eat me" signal to the macrophage, inhibiting phagocytosis. Thus, overexpression of CD47 enables tumor cells to escape from immune surveillance via the blockade of phagocytic mechanisms. We report here the development and characterization of CC-90002, a humanized anti-CD47 antibody. CC-90002 is unique among previously reported anti-CD47 bivalent antibodies that it does not promote hemagglutination while maintaining high-affinity binding to CD47 and inhibition of the CD47-SIRPα interaction. Studies in a panel of hematological cancer cell lines showed concentration-dependent CC-90002-mediated phagocytosis in acute lymphoblastic leukemia, acute myeloid leukemia (AML), lenalidomide-resistant multiple myeloma (MM) cell lines and AML cells from patients. In vivo studies with MM cell line-derived xenograft models established in immunodeficient mice demonstrated significant dose-dependent antitumor activity of CC-90002. Treatment with CC-90002 significantly prolonged survival in an HL-60-disseminated AML model. Mechanistic studies confirmed the binding of CC-90002 to tumor cells and concomitant recruitment of F4-80 positive macrophages into the tumor and an increase in expression of select chemokines and cytokines of murine origin. Furthermore, the role of macrophages in the CC-90002-mediated antitumor activity was demonstrated by transient depletion of macrophages with liposome-clodronate treatment. In non-human primates, CC-90002 displayed acceptable pharmacokinetic properties and a favorable toxicity profile. These data demonstrate the potential activity of CC-90002 across hematological malignancies and provided basis for clinical studies CC-90002-ST-001 (NCT02367196) and CC-90002-AML-001 (NCT02641002).


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, Differentiation/immunology , CD47 Antigen/immunology , Immunoglobulin Fc Fragments/immunology , Leukemia, Promyelocytic, Acute/drug therapy , Macrophages/immunology , Receptors, Immunologic/immunology , Animals , Antibodies, Monoclonal/immunology , Antigens, Differentiation/metabolism , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/pharmacology , Apoptosis , CD47 Antigen/metabolism , Cell Differentiation , Cell Proliferation , Female , Humans , Leukemia, Promyelocytic, Acute/immunology , Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Phagocytosis , Prognosis , Receptors, Immunologic/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Blood ; 121(20): 4175-83, 2013 May 16.
Article in English | MEDLINE | ID: mdl-23543457

ABSTRACT

Genomic instability is a hallmark of chronic myeloid leukemia in chronic phase (CML-CP) resulting in BCR-ABL1 mutations encoding resistance to tyrosine kinase inhibitors (TKIs) and/or additional chromosomal aberrations leading to disease relapse and/or malignant progression. TKI-naive and TKI-treated leukemia stem cells (LSCs) and leukemia progenitor cells (LPCs) accumulate high levels of reactive oxygen species (ROS) and oxidative DNA damage. To determine the role of TKI-refractory LSCs in genomic instability, we used a murine model of CML-CP where ROS-induced oxidative DNA damage was elevated in LSCs, including quiescent LSCs, but not in LPCs. ROS-induced oxidative DNA damage in LSCs caused clinically relevant genomic instability in CML-CP-like mice, such as TKI-resistant BCR-ABL1 mutations (E255K, T315I, H396P), deletions in Ikzf1 and Trp53, and additions in Zfp423 and Idh1. Despite inhibition of BCR-ABL1 kinase, imatinib did not downregulate ROS and oxidative DNA damage in TKI-refractory LSCs to the levels detected in normal cells, and CML-CP-like mice treated with imatinib continued to accumulate clinically relevant genetic aberrations. Inhibition of class I p21-activated protein kinases by IPA3 downregulated ROS in TKI-naive and TKI-treated LSCs. Altogether, we postulate that genomic instability may originate in the most primitive TKI-refractory LSCs in TKI-naive and TKI-treated patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Drug Resistance, Neoplasm/genetics , Genomic Instability , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Neoplastic Stem Cells/physiology , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Animals , Cells, Cultured , DNA Damage/drug effects , Genomic Instability/drug effects , Genomic Instability/physiology , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mice, Transgenic , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oxidative Stress/drug effects , Oxidative Stress/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Reactive Oxygen Species/metabolism
3.
BMJ Case Rep ; 20122012 Feb 25.
Article in English | MEDLINE | ID: mdl-22665563

ABSTRACT

Meningioma is the benign, unencapsulated neoplasm arising from meningo-epithelial arachnoid cells of cerebellopontine angle-internal auditory canal dura but extracranial meningiomas are very rare. In making the diagnosis of invasive meningioma, both changes of benign meningioma and invasive growth should be present. A case of the neglected invasive meningioma is described here which invaded approximately half of the fronto-temporo-parietal skull vault. The diagnosis was established and confirmed by the histopathology and immunohistochemistry, the cells were positive for epithelial membrane antigen, vimetin and progesterone receptors. The patient underwent surgical resection.


Subject(s)
Meningeal Neoplasms/pathology , Meningioma/pathology , Skull Neoplasms/secondary , Adult , Diagnosis, Differential , Frontal Bone/diagnostic imaging , Frontal Bone/pathology , Humans , Male , Meningeal Neoplasms/diagnosis , Meningeal Neoplasms/diagnostic imaging , Meningioma/diagnosis , Meningioma/diagnostic imaging , Parietal Bone/diagnostic imaging , Parietal Bone/pathology , Skull Neoplasms/diagnosis , Skull Neoplasms/diagnostic imaging , Skull Neoplasms/pathology , Temporal Bone/diagnostic imaging , Temporal Bone/pathology , Tomography, X-Ray Computed
4.
Blood ; 119(26): 6187-97, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22493298

ABSTRACT

Chromosomal aberrations are an important consequence of genotoxic exposure and contribute to pathogenesis and progression of several malignancies. We investigated the susceptibility to chromosomal aberrations in chronic myelogenous leukemia (CML) progenitors after exposure to ionizing radiation. In normal progenitors, ionizing radiation induced both stable and unstable chromosomal lesions, but only stable aberrations persisted after multiple divisions. In contrast, radiation of chronic phase CML progenitors resulted in enhanced generation of unstable lesions that persisted after multiple divisions. CML progenitors demonstrated active cell cycle checkpoints and increased nonhomologous end joining DNA repair, suggesting that persistence of unstable aberrations was the result of continued generation of these lesions. CML progenitors demonstrated enhanced susceptibility to repeated cycles of chromosome damage, repair, and damage through a breakage-fusion-bridge mechanism. Perpetuation of breakage-fusion-bridge cycles in CML progenitors was mediated by classic nonhomologous end joining repair. These studies reveal a previously unrecognized mechanism of chromosomal instability in leukemia progenitors because of continued generation of unstable chromosomal lesions through repeated cycles of breakage and repair of such lesions.


Subject(s)
Chromosomal Instability/genetics , Chromosome Breakage , DNA End-Joining Repair/physiology , Gene Fusion/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/metabolism , Antigens, CD34/metabolism , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/physiology , Cell Cycle Checkpoints/radiation effects , Cell Line, Tumor , Chromosomal Instability/radiation effects , Chromosome Breakage/radiation effects , DNA Damage/physiology , DNA End-Joining Repair/genetics , DNA End-Joining Repair/radiation effects , Gene Fusion/radiation effects , Humans , In Situ Hybridization, Fluorescence , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Models, Biological , Neoplastic Stem Cells/radiation effects , Radiation, Ionizing , Recurrence
5.
Blood ; 119(8): 1904-14, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-22207735

ABSTRACT

The tyrosine kinase inhibitor imatinib is highly effective in the treatment of chronic myelogenous leukemia (CML), but primary and acquired resistance of CML cells to the drug offset its efficacy. Molecular mechanisms for resistance of CML to tyrosine kinase inhibitors are not fully understood. In the present study, we show that BCR-ABL activates the expression of the mammalian stress response gene SIRT1 in hematopoietic progenitor cells and that this involves STAT5 signaling. SIRT1 activation promotes CML cell survival and proliferation associated with deacetylation of multiple SIRT1 substrates, including FOXO1, p53, and Ku70. Imatinib-mediated inhibition of BCR-ABL kinase activity partially reduces SIRT1 expression and SIRT1 inhibition further sensitizes CML cells to imatinib-induced apoptosis. Knockout of SIRT1 suppresses BCR-ABL transformation of mouse BM cells and the development of a CML-like myeloproliferative disease, and treatment of mice with the SIRT1 inhibitor tenovin-6 deters disease progression. The combination of SIRT1 gene knockout and imatinib treatment further extends the survival of CML mice. Our results suggest that SIRT1 is a novel survival pathway activated by BCR-ABL expression in hematopoietic progenitor cells, which promotes oncogenic transformation and leukemogenesis. Our findings suggest further exploration of SIRT1 as a therapeutic target for CML treatment to overcome resistance.


Subject(s)
Cell Transformation, Neoplastic/genetics , Fusion Proteins, bcr-abl/genetics , Sirtuin 1/genetics , Transcriptional Activation , Animals , Antineoplastic Agents/pharmacology , Benzamides , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/metabolism , Humans , Imatinib Mesylate , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Piperazines/pharmacology , Pyrimidines/pharmacology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Sirtuin 1/metabolism , Stress, Physiological/genetics , Transfection
6.
Blood ; 118(6): 1504-15, 2011 Aug 11.
Article in English | MEDLINE | ID: mdl-21670473

ABSTRACT

SHP2, a cytoplasmic protein-tyrosine phosphatase encoded by the PTPN11 gene, plays a critical role in developmental hematopoiesis in the mouse, and gain-of-function mutations of SHP2 are associated with hematopoietic malignancies. However, the role of SHP2 in adult hematopoiesis has not been addressed in previous studies. In addition, the role of SHP2 in human hematopoiesis has not been described. These questions are of considerable importance given the interest in development of SHP2 inhibitors for cancer treatment. We used shRNA-mediated inhibition of SHP2 expression to investigate the function of SHP2 in growth factor (GF) signaling in normal human CD34(+) cells. SHP2 knockdown resulted in markedly reduced proliferation and survival of cells cultured with GF, and reduced colony-forming cell growth. Cells expressing gain-of-function SHP2 mutations demonstrated increased dependency on SHP2 expression for survival compared with cells expressing wild-type SHP2. SHP2 knockdown was associated with significantly reduced myeloid and erythroid differentiation with retention of CD34(+) progenitors with enhanced proliferative capacity. Inhibition of SHP2 expression initially enhanced and later inhibited STAT5 phosphorylation and reduced expression of the antiapoptotic genes MCL1 and BCLXL. These results indicate an important role for SHP2 in STAT5 activation and GF-mediated proliferation, survival, and differentiation of human progenitor cells.


Subject(s)
Antigens, CD34/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Hematopoietic Cell Growth Factors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , STAT5 Transcription Factor/metabolism , Animals , Blotting, Western , Cell Survival/drug effects , Cell Transplantation/methods , Cells, Cultured , Fetal Blood/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , RNA Interference , STAT5 Transcription Factor/genetics , Transplantation, Heterologous
7.
Exp Hematol ; 37(2): 206-14, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19100678

ABSTRACT

OBJECTIVE: This study investigated two approaches, short hairpin RNA (shRNA) and the potent ABL inhibitor, dasatinib, alone and together, to achieve complete inhibition of BCR-ABL activity in Philadelphia-positive (Ph(+)) cells. MATERIALS AND METHODS: shRNA specific for BCR-ABL b3a2 were delivered, by lentiviral transduction or electroporation, to K562 cells, with or without dasatinib. mRNA and protein knockdown were measured by quantitative reverse transcriptase polymerase chain reaction, flow cytometry, and Western blotting. BCR-ABL activity was assessed by intracellular flow cytometry for pCrkL. Cell death and apoptosis were assayed using trypan blue exclusion, Annexin-V, and active caspase-3 staining. RESULTS: Forty-eight hours after transduction or electroporation of shRNA, BCR-ABL mRNA, and protein were reduced by 75% and >90%, respectively, and sustained for 5 days. Lentiviral delivery and electroporation were equally effective. pCrkL was inhibited in association with cell death. By 5 days after transduction or electroporation, viable cells represented 50% of input, with a 12-fold reduction vs control, which expanded 6-fold. When shRNA, titrated by green fluorescent protein into low and high, was combined with dasatinib (concentration range, 0-10 nM), low shRNA was additive with low dasatinib (0.6 and 1 nM), leading to inhibition of pCrkL, induction of activated caspase-3, expression of Annexin-V, and marked reduction in viable cells. CONCLUSION: These results confirm that by lowering BCR-ABL levels with shRNA, complete inhibition of oncoprotein activity can be achieved with a lower concentration of dasatinib, thus providing a rationale for combining these approaches in the setting of high target expression, such as found in advanced phase disease and in the stem cell compartment.


Subject(s)
Apoptosis/drug effects , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/metabolism , Lentivirus , Philadelphia Chromosome , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology , Annexin A5/biosynthesis , Annexin A5/genetics , Apoptosis/genetics , Blotting, Western , Caspase 3/biosynthesis , Caspase 3/genetics , Dasatinib , Dose-Response Relationship, Drug , Flow Cytometry , Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Leukemic/genetics , Humans , K562 Cells , RNA/genetics , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
8.
Blood ; 109(12): 5411-21, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17347407

ABSTRACT

Increased levels of Bcr-Abl expression in chronic myelogenous leukemia (CML) cells are associated with disease progression and imatinib (IM) resistance. However, it is not clear if these associations are a direct result of elevated Bcr-Abl expression. We used a human transduction model of CML to directly investigate the role of varying Bcr-Abl expression levels in determining the phenotype and IM sensitivity of hematopoietic cells. CD34(+) cells were transduced with vectors coexpressing Bcr-Abl and GFP, and cells expressing low and high levels of GFP and Bcr-Abl (BA(lo) and BA(hi)) were selected. BA(hi) cells demonstrated enhanced activation of downstream proliferative and antiapoptotic signaling and enhanced proliferation and survival compared to BA(lo) cells. Freshly isolated BA(hi) CD34(+) cells and cell lines demonstrated increased IM-mediated growth inhibition likely reflecting Bcr-Abl dependence for growth and survival. CD34(+) cells expressing BCR/ABL kinase-mutant genes demonstrated resistance to IM-mediated inhibition of proliferation and viability, which was not enhanced by increased expression of BCR/ABL kinase-mutant genes. We conclude that Bcr-Abl overexpression results in increased proliferation and antiapoptotic signaling in CD34(+) cells, but may not play a direct role in IM resistance in progenitor cells expressing either wild-type or mutant BCR/ABL genes.


Subject(s)
Drug Resistance, Neoplasm , Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Neoplastic , Hematopoietic Stem Cells/pathology , Piperazines/pharmacology , Pyrimidines/pharmacology , Benzamides , Cell Line, Transformed , Cell Proliferation , Cell Survival , Humans , Imatinib Mesylate , Phenotype , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...