Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
J Immunother Cancer ; 12(8)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39214650

ABSTRACT

BACKGROUND: Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with a poor prognosis particularly in the metastatic setting. Treatments with anti-programmed cell death protein-1/programmed death-ligand 1 (PD-L1) immune checkpoint inhibitors (ICI) in combination with chemotherapies have demonstrated promising clinical benefit in metastatic TNBC (mTNBC) but there is still an unmet need, particularly for patients with PD-L1 negative tumors. Mechanisms of resistance to ICIs in mTNBC include the presence of immunosuppressive tumor-associated macrophages (TAMs) in the tumor microenvironment (TME). Eganelisib is a potent and selective, small molecule PI3K-γ inhibitor that was shown in preclinical studies to reshape the TME by reducing myeloid cell recruitment to tumors and reprogramming TAMs from an immune-suppressive to an immune-activating phenotype and enhancing activity of ICIs. These studies provided rationale for the clinical evaluation of eganelisib in combination with the anti-PD-L1 atezolizumab and nab-paclitaxel in firstline mTNBC in the phase 2 clinical trial MAcrophage Reprogramming in Immuno-Oncology-3 (MARIO-3, NCT03961698). We present here for the first time, in-depth translational analyses from the MARIO-3 study and supplemental data from eganelisib monotherapy Ph1/b study in solid tumors (MARIO-1, NCT02637531). METHODS: Paired pre-treatment and post-treatment tumor biopsies were analyzed for immunophenotyping by multiplex immunofluorescence (n=11), spatial transcriptomics using GeoMx digital spatial profiling (n=12), and PD-L1 immunohistochemistry, (n=18). Peripheral blood samples were analyzed using flow cytometry and multiplex cytokine analysis. RESULTS: Results from paired tumor biopsies from MARIO-3 revealed gene signatures of TAM reprogramming, immune activation and extracellular matrix (ECM) reorganization. Analysis of PD-L1 negative tumors revealed elevated ECM gene signatures at baseline that decreased after treatment. Gene signatures of immune activation were observed regardless of baseline PD-L1 status and occurred in patients having longer progression-free survival. Peripheral blood analyses revealed systemic immune activation. CONCLUSIONS: This is the first report of translational analyses including paired tumor biopsies from a phase 2 clinical study of the first-in-class PI3K-γ inhibitor eganelisib in combination with atezolizumab and nab-paclitaxel in frontline mTNBC. These results support the mechanism of action of eganelisib as a TAM-reprogramming immunotherapy and support the rationale for combining eganelisib with ICI and chemotherapy in indications with TAM-driven resistance to ICI.


Subject(s)
Immune Checkpoint Inhibitors , Triple Negative Breast Neoplasms , Tumor Microenvironment , Female , Humans , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Neoplasm Metastasis , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/drug effects , Clinical Trials, Phase I as Topic
2.
ACS Med Chem Lett ; 13(7): 1137-1143, 2022 Jul 14.
Article in English | MEDLINE | ID: mdl-35859865

ABSTRACT

SETD2, a lysine N-methyltransferase, is a histone methyltransferase that plays an important role in various cellular processes and was identified as a target of interest in multiple myeloma that features a t(4,14) translocation. We recently reported the discovery of a novel small-molecule SETD2 inhibitor tool compound that is suitable for preclinical studies. Herein we describe the conformational-design-driven evolution of the advanced chemistry lead, which resulted in compounds appropriate for clinical evaluation. Further optimization of this chemical series led to the discovery of EZM0414, which is a potent, selective, and orally bioavailable inhibitor of SETD2 with good pharmacokinetic properties and robust pharmacodynamic activity in a mouse xenograft model.

3.
Sci Transl Med ; 7(277): 277ra30, 2015 Mar 04.
Article in English | MEDLINE | ID: mdl-25739764

ABSTRACT

Antitumor T cells either avoid or are inhibited in hypoxic and extracellular adenosine-rich tumor microenvironments (TMEs) by A2A adenosine receptors. This may limit further advances in cancer immunotherapy. There is a need for readily available and safe treatments that weaken the hypoxia-A2-adenosinergic immunosuppression in the TME. Recently, we reported that respiratory hyperoxia decreases intratumoral hypoxia and concentrations of extracellular adenosine. We show that it also reverses the hypoxia-adenosinergic immunosuppression in the TME. This, in turn, stimulates (i) enhanced intratumoral infiltration and reduced inhibition of endogenously developed or adoptively transfered tumor-reactive CD8 T cells, (ii) increased proinflammatory cytokines and decreased immunosuppressive molecules, such as transforming growth factor-ß (TGF-ß), (iii) weakened immunosuppression by regulatory T cells, and (iv) improved lung tumor regression and long-term survival in mice. Respiratory hyperoxia also promoted the regression of spontaneous metastasis from orthotopically grown breast tumors. These effects are entirely T cell- and natural killer cell-dependent, thereby justifying the testing of supplemental oxygen as an immunological coadjuvant to combine with existing immunotherapies for cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/immunology , Oxygen/therapeutic use , Adenosine/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Hyperoxia/complications , Hyperoxia/pathology , Hypoxia/complications , Hypoxia/immunology , Hypoxia/pathology , Immunosuppression Therapy , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplasms/pathology , Oxygen/pharmacology , Remission Induction , Respiration/drug effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/drug effects
4.
J Mol Med (Berl) ; 92(12): 1283-92, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25120128

ABSTRACT

UNLABELLED: Intratumoral hypoxia and hypoxia inducible factor-1α (HIF-1-α)-dependent CD39/CD73 ectoenzymes may govern the accumulation of tumor-protecting extracellular adenosine and signaling through A2A adenosine receptors (A2AR) in tumor microenvironments (TME). Here, we explored the conceptually novel motivation to use supplemental oxygen as a treatment to inhibit the hypoxia/HIF-1α-CD39/CD73-driven accumulation of extracellular adenosine in the TME in order to weaken the tumor protection. We report that hyperoxic breathing (60 % O2) decreased the TME hypoxia, as well as levels of HIF-1α and downstream target proteins of HIF-1α in the TME according to proteomic studies in mice. Importantly, oxygenation also downregulated the expression of adenosine-generating ectoenzymes and significantly lowered levels of tumor-protecting extracellular adenosine in the TME. Using supplemental oxygen as a tool in studies of the TME, we also identified FHL-1 as a potentially useful marker for the conversion of hypoxic into normoxic TME. Hyperoxic breathing resulted in the upregulation of antigen-presenting MHC class I molecules on tumor cells and in the better recognition and increased susceptibility to killing by tumor-reactive cytotoxic T cells. Therapeutic breathing of 60 % oxygen resulted in the significant inhibition of growth of established B16.F10 melanoma tumors and prolonged survival of mice. Taken together, the data presented here provide proof-of principle for the therapeutic potential of systemic oxygenation to convert the hypoxic, adenosine-rich and tumor-protecting TME into a normoxic and extracellular adenosine-poor TME that, in turn, may facilitate tumor regression. We propose to explore the combination of supplemental oxygen with existing immunotherapies of cancer. KEY MESSAGES: Oxygenation decreases levels of tumor protecting hypoxia. Oxygenation decreases levels of tumor protecting extracellular adenosine. Oxygenation decreases expression of HIF-1alpha dependent tumor-protecting proteins. Oxygenation increases MHC class I expression and enables tumor regression.


Subject(s)
Adenosine/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/complications , Hypoxia/therapy , Neoplasms/complications , Neoplasms/therapy , Oxygen/therapeutic use , Animals , Cell Hypoxia , Cell Line, Tumor , Female , Hypoxia/metabolism , Mice, Inbred C57BL , Neoplasms/metabolism , Tumor Microenvironment
5.
Blood ; 118(17): 4666-73, 2011 Oct 27.
Article in English | MEDLINE | ID: mdl-21873545

ABSTRACT

Large chromosomal deletions are among the most common molecular abnormalities in cancer, yet the identification of relevant genes has proven difficult. The 5q- syndrome, a subtype of myelodysplastic syndrome (MDS), is a chromosomal deletion syndrome characterized by anemia and thrombocytosis. Although we have previously shown that hemizygous loss of RPS14 recapitulates the failed erythroid differentiation seen in 5q- syndrome, it does not affect thrombocytosis. Here we show that a microRNA located in the common deletion region of 5q- syndrome, miR-145, affects megakaryocyte and erythroid differentiation. We find that miR-145 functions through repression of Fli-1, a megakaryocyte and erythroid regulatory transcription factor. Patients with del(5q) MDS have decreased expression of miR-145 and increased expression of Fli-1. Overexpression of miR-145 or inhibition of Fli-1 decreases the production of megakaryocytic cells relative to erythroid cells, whereas inhibition of miR-145 or overexpression of Fli-1 has a reciprocal effect. Moreover, combined loss of miR-145 and RPS14 cooperates to alter erythroid-megakaryocytic differentiation in a manner similar to the 5q- syndrome. Taken together, these findings demonstrate that coordinate deletion of a miRNA and a protein-coding gene contributes to the phenotype of a human malignancy, the 5q- syndrome.


Subject(s)
Anemia, Macrocytic/genetics , MicroRNAs/genetics , Open Reading Frames/genetics , Anemia, Macrocytic/etiology , Animals , Case-Control Studies , Cell Differentiation/genetics , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Erythroid Cells/metabolism , Erythropoiesis/genetics , Erythropoiesis/physiology , Humans , Loss of Heterozygosity , Megakaryocytes/metabolism , Megakaryocytes/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , MicroRNAs/metabolism , MicroRNAs/physiology , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , Proto-Oncogene Protein c-fli-1/physiology , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomal Proteins/physiology , Tumor Cells, Cultured
6.
Proc Natl Acad Sci U S A ; 107(1): 252-7, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-20018760

ABSTRACT

The prognosis for adults with precursor B-cell acute lymphoblastic leukemia (B-ALL) remains poor, in part from a lack of therapeutic targets. We identified the type I cytokine receptor subunit CRLF2 in a functional screen for B-ALL-derived mRNA transcripts that can substitute for IL3 signaling. We demonstrate that CRLF2 is overexpressed in approximately 15% of adult and high-risk pediatric B-ALL that lack MLL, TCF3, TEL, and BCR/ABL rearrangements, but not in B-ALL with these rearrangements or other lymphoid malignancies. CRLF2 overexpression can result from translocation with the IGH locus or intrachromosomal deletion and is associated with poor outcome. CRLF2 overexpressing B-ALLs share a transcriptional signature that significantly overlaps with a BCR/ABL signature, and is enriched for genes involved in cytokine receptor and JAK-STAT signaling. In a subset of cases, CRLF2 harbors a Phe232Cys gain-of-function mutation that promotes constitutive dimerization and cytokine independent growth. A mutually exclusive subset harbors activating mutations in JAK2. In fact, all 22 B-ALLs with mutant JAK2 that we analyzed overexpress CRLF2, distinguishing CRLF2 as the key scaffold for mutant JAK2 signaling in B-ALL. Expression of WT CRLF2 with mutant JAK2 also promotes cytokine independent growth that, unlike CRLF2 Phe232Cys or ligand-induced signaling by WT CRLF2, is accompanied by JAK2 phosphorylation. Finally, cells dependent on CRLF2 signaling are sensitive to small molecule inhibitors of either JAKs or protein kinase C family kinases. Together, these findings implicate CRLF2 as an important factor in B-ALL with diagnostic, prognostic, and therapeutic implications.


Subject(s)
Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Receptors, Cytokine/genetics , Signal Transduction/physiology , Adult , Child , Cytokines/metabolism , DNA Mutational Analysis , Female , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Male , Mutation , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Prognosis , Receptors, Cytokine/metabolism , Survival Rate , Transcription, Genetic
7.
Cell ; 139(3): 573-86, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19879843

ABSTRACT

Previous work has shown that mature B cells depend upon survival signals delivered to the cells by their antigen receptor (BCR). To identify the molecular nature of this survival signal, we have developed a genetic approach in which ablation of the BCR is combined with the activation of specific, BCR dependent signaling cascades in mature B cells in vivo. Using this system, we provide evidence that the survival of BCR deficient mature B cells can be rescued by a single signaling pathway downstream of the BCR, namely PI3K signaling, with the FOXO1 transcription factor playing a central role.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Survival , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Antigen, B-Cell/metabolism , Animals , Mice , Mice, Knockout , Signal Transduction
8.
Dev Cell ; 16(2): 314-20, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19217432

ABSTRACT

Fanconi anemia (FA) is a human genetic disease characterized by chromosome instability, cancer predisposition, and cellular hypersensitivity to DNA crosslinking agents. The FA pathway regulates the repair of DNA crosslinks. A critical step in this pathway is the monoubiquitination and deubiquitination of FANCD2. Deubiquitination of FANCD2 is mediated by the ubiquitin protease, USP1. Here, we demonstrate that targeted deletion of mouse Usp1 results in elevated perinatal lethality, male infertility, crosslinker hypersensitivity, and an FA phenotype. Usp1(-/-) mouse embryonic fibroblasts had heightened levels of monoubiquitinated Fancd2 in chromatin. Usp1(-/-) cells exhibited impaired Fancd2 foci assembly and a defect in homologous recombination repair. Double knockout of Usp1 and Fancd2 resulted in a more severe phenotype than either single knockout. Our results indicate that mouse Usp1 functions downstream in the FA pathway. Deubiquitination is a critical event required for Fancd2 nuclear foci assembly, release from chromatin, and function in DNA repair.


Subject(s)
Endopeptidases/genetics , Endopeptidases/physiology , Fanconi Anemia/genetics , Genomic Instability , Animals , Arabidopsis Proteins , Crosses, Genetic , DNA Repair , Fanconi Anemia Complementation Group D2 Protein/metabolism , Female , Fibroblasts/metabolism , Gene Deletion , Lymphocytes/metabolism , Male , Mice , Mice, Knockout , Phenotype , Ubiquitin-Specific Proteases
9.
Proc Natl Acad Sci U S A ; 105(47): 18472-7, 2008 Nov 25.
Article in English | MEDLINE | ID: mdl-19011096

ABSTRACT

The human fetus is not rejected by the maternal immune system despite expressing paternal antigens. Natural killer cells, the major lymphocyte population of the human decidua (dNKs), express genes with immunomodulatory potential. These include galectin-1 (gal1), a lectin with apoptotic activity on activated CD8(+) T cells, Th1 and Th17 CD4(+) cells. Although many cell types at the maternal-fetal interface also produce gal1, its production by dNKs has been used here to study its function in pregnancy. Media conditioned by dNKs containing gal1 induced apoptosis of activated T cells. This effect was blocked by anti-gal1 antibodies. Decidual T (dT) cells but not peripheral T (pT) cells bound gal1 and presented a distinct glycophenotype compatible with sensitivity to gal1. Annexin V staining, TUNEL, and hypodiploidy showed a substantial proportion of apoptotic dT cells. Immunohistochemistry revealed widespread expression of gal1 as well as periglandular apoptotic dT foci that colocalized with dNKs. Thus, secretion of gal1 by dNKs and other decidual cells contributes to the generation of an immune-privileged environment at the maternal-fetal interface.


Subject(s)
Apoptosis/physiology , Galectin 1/physiology , Maternal-Fetal Exchange , T-Lymphocytes/cytology , Culture Media, Conditioned , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoprecipitation , In Situ Nick-End Labeling , Killer Cells, Natural/immunology , Pregnancy
10.
Clin Cancer Res ; 14(17): 5626-34, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18765557

ABSTRACT

PURPOSE: CpG oligodeoxynucleotides (CpG-ODN) are being investigated as cancer vaccine adjuvants because they mature plasmacytoid dendritic cells (PDC) into potent antigen-presenting cells. CpG-ODN also induce PDC to secrete chemokines that alter lymphocyte migration. Whether CpG-ODN TLR signals enhance antigen-specific immunity and/or trafficking in humans is unknown. EXPERIMENTAL DESIGN: We conducted a phase I study of CpG-ODN (1018 ISS) given as a vaccine adjuvant with granulocyte-macrophage colony-stimulating factor (GM-CSF) to induce T-cell immunity to a peptide vaccine from the tumor-associated antigen hTERT. RESULTS: The adjuvant effect was limited; only 1 of 16 patients showed a high-frequency hTERT-specific tetramer CD8(+) T-cell response. However, CpG-ODN induced marked, transient peripheral blood lymphopenia. Biopsies showed dense lymphocytic infiltration at the vaccine site clustered around activated PDC. In vitro, CpG-ODN-treated PDC induced T-cell migration, showing that CpG-ODN stimulation of human PDC was sufficient to chemoattract T cells. CONCLUSIONS: Our results show that (a) CpG-ODN with GM-CSF may not be an effective adjuvant strategy for hTERT peptide vaccines but (b) GM-CSF/CpG-ODN causes a PDC-mediated chemokine response that recruits T-cell migration to the peripheral tissues. These findings suggest a novel therapeutic role for targeted injections of CpG-ODN to direct lymphocyte migration to specific sites such as the tumor bed.


Subject(s)
Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Oligodeoxyribonucleotides/therapeutic use , T-Lymphocytes/immunology , Telomerase/immunology , Vaccines, Subunit/therapeutic use , Adjuvants, Immunologic , Cancer Vaccines/adverse effects , Cell Movement , Humans , Oligodeoxyribonucleotides/immunology , Toll-Like Receptor 9/antagonists & inhibitors
11.
Leuk Lymphoma ; 49(5): 902-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18464109

ABSTRACT

Follicular dendritic cells (FDCs) support the survival of follicular lymphoma (FL). Tumor necrosis factor alpha (TNFalpha) is overexpressed by FL cells and is critical in the development and maintenance of FDCs. We hypothesised that TNFalpha might be an ideal therapeutic target. We treated seven patients with relapsed/refractory FL with 8 weeks of etanercept, 25 mg SC on day 1 and 4 of each week. Patients without progression received 16 additional weeks of etanercept. All patients completed at least 8 weeks of etanercept and two patients completed 24 weeks. At the 8 week evaluation five patients had SD. Of the five with SD, two progressed at 9 and 12 weeks on therapy and the remaining three progressed between 12 and 24 weeks after initiating therapy. Minimal toxicity was observed. FDG-PET imaging demonstrated decreases in standardized uptake value (SUV) following treatment with etanercept in five patients. Further studies in FL targeting the microenvironment in conjunction with standard cytotoxic therapy are warranted.


Subject(s)
Immunoglobulin G/administration & dosage , Lymphoma, Follicular/drug therapy , Receptors, Tumor Necrosis Factor/administration & dosage , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adult , Aged , Drug Delivery Systems/methods , Drug Monitoring , Etanercept , Humans , Immunoglobulin G/toxicity , Middle Aged , Positron-Emission Tomography , Salvage Therapy , Treatment Outcome
12.
Nat Immunol ; 9(4): 405-14, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18327259

ABSTRACT

The genomic region encoding the miR-17-92 microRNA (miRNA) cluster is often amplified in lymphoma and other cancers, and cancer cells carrying this amplification have higher expression of miRNA in this cluster. Retroviral expression of miR-17-92 accelerates c-Myc-induced lymphoma development, but precisely how higher expression of miR-17-92 promotes lymphomagenesis remains unclear. Here we generated mice with higher expression of miR-17-92 in lymphocytes. These mice developed lymphoproliferative disease and autoimmunity and died prematurely. Lymphocytes from these mice showed more proliferation and less activation-induced cell death. The miR-17-92 miRNA suppressed expression of the tumor suppressor PTEN and the proapoptotic protein Bim. This mechanism probably contributed to the lymphoproliferative disease and autoimmunity of miR-17-92-transgenic mice and contributes to lymphoma development in patients with amplifications of the miR-17-92 coding region.


Subject(s)
Autoimmune Diseases/genetics , Lymphocytes/immunology , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/immunology , MicroRNAs/biosynthesis , MicroRNAs/genetics , Animals , Autoimmune Diseases/pathology , Cell Death/genetics , Cell Death/immunology , Cell Proliferation , Cells, Cultured , Gene Amplification , Gene Expression Regulation, Neoplastic/immunology , Humans , Jurkat Cells , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocytes/metabolism , Lymphoma/genetics , Lymphoma/immunology , Lymphoproliferative Disorders/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/physiology
13.
Blood ; 109(11): 4964-72, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17284528

ABSTRACT

Waldenström macroglobulinemia (WM) is an incurable lymphoplasmacytic lymphoma with limited options of therapy. Protein kinase Cbeta (PKCbeta) regulates cell survival and growth in many B-cell malignancies. In this study, we demonstrate up-regulation of PKCbeta protein in WM using protein array techniques and immunohistochemistry. Enzastaurin, a PKCbeta inhibitor, blocked PKCbeta activity and induced a significant decrease of proliferation at 48 hours in WM cell lines (IC(50), 2.5-10 muM). Similar effects were demonstrated in primary CD19(+) WM cells, without cytotoxicity on peripheral blood mononuclear cells. In addition, enzastaurin overcame tumor cell growth induced by coculture of WM cells with bone marrow stromal cells. Enzastaurin induced dose-dependent apoptosis at 48 hours mediated via induction of caspase-3, caspase-8, caspase-9, and PARP cleavage. Enzastaurin inhibited Akt phosphorylation and Akt kinase activity, as well as downstream p-MARCKS and ribosomal p-S6. Furthermore, enzastaurin demonstrated additive cytotoxicity in combination with bortezomib, and synergistic cytotoxicity in combination with fludarabine. Finally, in an in vivo xenograft model of human WM, significant inhibition of tumor growth was observed in the enzastaurin-treated mice (P = .028). Our studies therefore show that enzastaurin has significant antitumor activity in WM both in vitro and in vivo, providing the framework for clinical trials to improve patient outcome in WM.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Protein Kinase C/antagonists & inhibitors , Waldenstrom Macroglobulinemia/drug therapy , Antigens, CD19/biosynthesis , Biopsy , Dose-Response Relationship, Drug , Enzyme Activation , Humans , In Vitro Techniques , Inhibitory Concentration 50 , Leukocytes, Mononuclear/immunology , Oligonucleotide Array Sequence Analysis , Protein Kinase C/metabolism , Protein Kinase C beta , Time Factors , Waldenstrom Macroglobulinemia/enzymology
14.
Blood ; 104(5): 1298-305, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15142873

ABSTRACT

The study of hematopoiesis has been greatly facilitated by transplantation of blood cell populations into recipient animals. Efficient engraftment of donor cells generally requires ablation of the host hematopoietic system. The zebrafish has recently emerged as a developmental and genetic system to study hematopoiesis. To enable the study of hematopoietic stem cell (HSC) biology, immune cell function, and leukemogenesis in zebrafish, we have developed hematopoietic cell transplantation (HCT) into adult recipient animals conditioned by gamma irradiation. Dose-response experiments showed that the minimum lethal dose (MLD) of 40 Gy led to the specific ablation of hematolymphoid cells and death by 14 days after irradiation. Sublethal irradiation doses of 20 Gy predominantly ablated lymphocytes and permitted transplantation of a lethal T-cell leukemia. Finally, transplantation of hematopoietic cells carrying transgenes yielding red fluorescent erythrocytes and green fluorescent leukocytes showed that HCT is sufficient to rescue the MLD, that recipient hematolymphoid tissues were repopulated by donor-derived cells, and that donor blood cell lineages can be independently visualized in living recipients. Together, these results establish transplantation assays to test for HSC function and oncogenic transformation in zebrafish.


Subject(s)
Hematopoiesis/radiation effects , Hematopoietic Stem Cell Transplantation , Lymphoid Tissue/radiation effects , Animals , Animals, Genetically Modified , Female , Gamma Rays , Green Fluorescent Proteins , Hematopoietic Stem Cell Transplantation/mortality , Kidney/cytology , Leukemia, T-Cell/etiology , Leukemia, T-Cell/pathology , Luminescent Proteins/genetics , Lymphoid Tissue/pathology , Male , Transplantation Conditioning , Whole-Body Irradiation , Zebrafish
15.
Leuk Lymphoma ; 45(2): 315-20, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15101717

ABSTRACT

The marginal zone non-Hodgkin's lymphomas are a recently defined group of related low-grade B cell malignancies whose natural history is heterogeneous. The optimal therapy is often unclear, particularly for the subset of patients with disseminated disease that behaves aggressively. We have retrospectively analyzed the outcomes of 11 patients with chemosensitive but disseminated marginal zone lymphomas who underwent uniform conditioning with cyclophosphamide and total body irradiation followed by bone marrow transplantation (BMT) with anti-B cell monoclonal antibody-purged autologous bone marrow between January 1994 and September 1999. All patients had stage IV disease and received multiple chemotherapy regimens prior to autologous BMT. Only 36% were in complete remission at the time of bone marrow harvest, and 36% had overt bone marrow infiltration at that time. Two treatment-related deaths occurred between 100 days and 6 months. Three patients relapsed and died of disease. One patient developed and died of myelodysplasia. Five patients remain in continuous complete remission at a median follow-up of 52 months (45%). The median progression-free survival for these patients was 56 months, with median overall survival 58 months. The only significant predictor of disease-free and overall survival was age at the time of transplant; no patient under 45 at the time of transplant has relapsed or died of any cause (P = 0.003). Outcomes of autologous BMT in patients with disseminated marginal zone NHL are similar to those in follicular NHL, and suggest that certain patients may experience prolonged disease-free survival.


Subject(s)
Bone Marrow Transplantation/methods , Lymphoma, Non-Hodgkin/therapy , Transplantation, Autologous/methods , Adult , Age Factors , Bone Marrow Cells/metabolism , Disease-Free Survival , Female , Humans , Lymphoma, Non-Hodgkin/mortality , Male , Middle Aged , Remission Induction , Time Factors , Treatment Outcome
16.
N Engl J Med ; 348(13): 1201-14, 2003 Mar 27.
Article in English | MEDLINE | ID: mdl-12660384

ABSTRACT

BACKGROUND: Idiopathic hypereosinophilic syndrome involves a prolonged state of eosinophilia associated with organ dysfunction. It is of unknown cause. Recent reports of responses to imatinib in patients with the syndrome suggested that an activated kinase such as ABL, platelet-derived growth factor receptor (PDGFR), or KIT, all of which are inhibited by imatinib, might be the cause. METHODS: We treated 11 patients with the hypereosinophilic syndrome with imatinib and identified the molecular basis for the response. RESULTS: Nine of the 11 patients treated with imatinib had responses lasting more than three months in which the eosinophil count returned to normal. One such patient had a complex chromosomal abnormality, leading to the identification of a fusion of the Fip1-like 1 (FIP1L1) gene to the PDGFRalpha (PDGFRA) gene generated by an interstitial deletion on chromosome 4q12. FIP1L1-PDGFRalpha is a constitutively activated tyrosine kinase that transforms hematopoietic cells and is inhibited by imatinib (50 percent inhibitory concentration, 3.2 nM). The FIP1L1-PDGFRA fusion gene was subsequently detected in 9 of 16 patients with the syndrome and in 5 of the 9 patients with responses to imatinib that lasted more than three months. Relapse in one patient correlated with the appearance of a T674I mutation in PDGFRA that confers resistance to imatinib. CONCLUSIONS: The hypereosinophilic syndrome may result from a novel fusion tyrosine kinase - FIP1L1-PDGFRalpha - that is a consequence of an interstitial chromosomal deletion. The acquisition of a T674I resistance mutation at the time of relapse demonstrates that FIP1L1-PDGFRalpha is the target of imatinib. Our data indicate that the deletion of genetic material may result in gain-of-function fusion proteins.


Subject(s)
Chromosome Deletion , Enzyme Inhibitors/therapeutic use , Hypereosinophilic Syndrome/genetics , Piperazines/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Pyrimidines/therapeutic use , Recombination, Genetic , Adult , Base Sequence , Benzamides , Chromosomes, Human, Pair 4/genetics , Female , Humans , Hypereosinophilic Syndrome/drug therapy , Imatinib Mesylate , Male , Middle Aged , Molecular Sequence Data , Remission Induction/methods
17.
Proc Natl Acad Sci U S A ; 99(12): 8283-8, 2002 Jun 11.
Article in English | MEDLINE | ID: mdl-12060771

ABSTRACT

Acute promyelocytic leukemia (APL) cells invariably express aberrant fusion proteins involving the retinoic acid receptor alpha (RARalpha). The most common fusion partner is promyelocytic leukemia protein (PML), which is fused to RARalpha in the balanced reciprocal chromosomal translocation, t(15;17)(q22:q11). Expression of PML/RARalpha from the cathepsin G promoter in transgenic mice causes a nonfatal myeloproliferative syndrome in all mice; about 15% go on to develop APL after a long latent period, suggesting that additional mutations are required for the development of APL. A candidate target gene for a second mutation is FLT3, because it is mutated in approximately 40% of human APL cases. Activating mutations in FLT3, including internal tandem duplication (ITD) in the juxtamembrane domain, transform hematopoietic cell lines to factor independent growth. FLT3-ITDs also induce a myeloproliferative disease in a murine bone marrow transplant model, but are not sufficient to cause AML. Here, we test the hypothesis that PML/RARalpha can cooperate with FLT3-ITD to induce an APL-like disease in the mouse. Retroviral transduction of FLT3-ITD into bone marrow cells obtained from PML/RARalpha transgenic mice results in a short latency APL-like disease with complete penetrance. This disease resembles the APL-like disease that occurs with long latency in the PML/RARalpha transgenics, suggesting that activating mutations in FLT3 can functionally substitute for the additional mutations that occur during mouse APL progression. The leukemia is transplantable to secondary recipients and is ATRA responsive. These observations document cooperation between PML/RARalpha and FLT3-ITD in development of the murine APL phenotype.


Subject(s)
Leukemia, Promyelocytic, Acute/genetics , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/genetics , Animals , Cathepsin G , Cathepsins/genetics , Crosses, Genetic , Humans , Immunophenotyping , Leukemia, Promyelocytic, Acute/immunology , Leukemia, Promyelocytic, Acute/pathology , Mice , Mice, Inbred C3H , Mice, Inbred Strains , Mice, Transgenic , Neoplasm Transplantation , Serine Endopeptidases , Tretinoin/pharmacology , Tumor Stem Cell Assay
18.
Blood ; 99(1): 310-8, 2002 Jan 01.
Article in English | MEDLINE | ID: mdl-11756186

ABSTRACT

FLT3 receptor tyrosine kinase is expressed on lymphoid and myeloid progenitors in the hematopoietic system. Activating mutations in FLT3 have been identified in approximately 30% of patients with acute myelogenous leukemia, making it one of the most common mutations observed in this disease. Frequently, the mutation is an in-frame internal tandem duplication (ITD) in the juxtamembrane region that results in constitutive activation of FLT3, and confers interleukin-3 (IL-3)-independent growth to Ba/F3 and 32D cells. FLT3-ITD mutants were cloned from primary human leukemia samples and assayed for transformation of primary hematopoietic cells using a murine bone marrow transplantation assay. FLT3-ITDs induced an oligoclonal myeloproliferative disorder in mice, characterized by splenomegaly and leukocytosis. The myeloproliferative phenotype, which was associated with extramedullary hematopoiesis in the spleen and liver, was confirmed by histopathologic and flow cytometric analysis. The disease latency of 40 to 60 days with FLT3-ITDs contrasted with wild-type FLT3 and enhanced green fluorescent protein (EGFP) controls, which did not develop hematologic disease (> 200 days). These results demonstrate that FLT3-ITD mutant proteins are sufficient to induce a myeloproliferative disorder, but are insufficient to recapitulate the AML phenotype observed in humans. Additional mutations that impair hematopoietic differentiation may be required for the development of FLT3-ITD-associated acute myeloid leukemias. This model system should be useful to assess the contribution of additional cooperating mutations and to evaluate specific FLT3 inhibitors in vivo.


Subject(s)
Bone Marrow Transplantation , Leukemia, Myeloid, Acute/genetics , Mutation , Myeloproliferative Disorders/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Amino Acid Sequence , Animals , Cell Line , Cloning, Molecular , Disease Models, Animal , Flow Cytometry , Gene Expression , Humans , Immunophenotyping , Leukocytosis , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Myeloproliferative Disorders/pathology , Proto-Oncogene Proteins/chemistry , Receptor Protein-Tyrosine Kinases/chemistry , Splenomegaly , Tandem Repeat Sequences , Transfection , Tumor Cells, Cultured , Vascular Endothelial Growth Factor Receptor-1
SELECTION OF CITATIONS
SEARCH DETAIL