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.
Recent Pat Anticancer Drug Discov ; 18(2): 224-240, 2022.
Article in English | MEDLINE | ID: mdl-35593340

ABSTRACT

BACKGROUND: SV-BR-1-GM, derived from a patient with grade 2 (moderately differentiated) breast cancer, is a GM-CSF-secreting breast cancer cell line with properties of antigen-presenting cells. SV-BR-1-GM and next-generation versions are covered by several pending and granted patents. METHODS: We report findings from an open-label phase I, single-arm pilot study with irradiated SV-BR-1-GM cells in 3 breast and 1 ovarian cancer subjects. Inoculations were preceded by lowdose intravenous cyclophosphamide and followed by interferon-alpha2b injections into the SVBR- 1-GM inoculation sites. We assessed both cellular and humoral immune responses, and measured expression levels of SV-BR-1-GM HLA alleles. RESULTS: Treatment was generally safe and well tolerated. Immune responses were elicited universally. Overall survival was more than 33 months for three of the four patients. As previously reported, one patient had prompt regression of metastases in lung, breast, and soft tissue. Following cessation of treatment, the patient relapsed widely, including in the brain. Upon retreatment, rapid tumor response was again seen, including complete regression of brain metastases. Consistent with a role of Class II HLA in contributing to SV-BR-1-GM's mechanism of action, this patient allele-matched SV-BR-1-GM at the HLA-DRB1 and HLA-DRB3 loci. We are in the process of developing next-generation SV-BR-1-GM, expressing patient-specific HLAs. Patent applications were filed in various jurisdictions. Thus far, one is granted, in Japan. CONCLUSION: A whole-cell immunotherapy regimen with SV-BR-1-GM cells induced regression of metastatic breast cancer. We develop intellectual property based on SV-BR-1-GM's predicted mechanism of action to develop additional whole-cell immunotherapies for cancer patients.


Subject(s)
Breast Neoplasms , Cancer Vaccines , Neoplasms, Second Primary , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Pilot Projects , Patents as Topic , Biomarkers , Cell Line , Melanoma, Cutaneous Malignant
2.
Blood Adv ; 3(20): 3020-3032, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31648327

ABSTRACT

The sphingosine-1-phosphate (S1P) receptor S1PR2 and its downstream adaptor Gα13 are recurrently mutationally inactivated in the germinal center B-cell subtype of diffuse large B-cell lymphoma (DLBCL) and are silenced by the S1PR2 repressor FOXP1 in the activated B-cell like subtype of the disease. Loss of S1PR2 signaling relieves the germinal center confinement that is maintained by an S1P gradient and allows cells to resist S1P-induced apoptosis. We have shown previously that S1PR2 expression is induced in normal B cells through a newly described transforming growth factor-ß (TGF-ß)/TGF-ßRII/SMAD1 signaling axis that is inactivated in >85% of DLBCL patients. DLBCL cell lines lacking S1PR2, TGFBRII, or SMAD1 as the result of genomic editing all have a strong growth advantage in vitro, as well as in subcutaneous and orthotopic xenotransplantation models. Here, we show that the TGF-ß signaling pathway in DLBCL is blocked at the level of SMAD1 in DLBCL cell lines and patient samples by hypermethylation of CpG-rich regions surrounding the SMAD1 transcription start site. The pharmacologic restoration of SMAD1 expression by the demethylating agent decitabine (DAC) sensitizes cells to TGF-ß-induced apoptosis and reverses the growth of initially SMAD1- cell lines in ectopic and orthotopic models. This effect of DAC is reduced in a SMAD1-knockout cell line. We further show that DAC restores SMAD1 expression and reduces the tumor burden in a novel patient-derived orthotopic xenograft model. The combined data lend further support to the concept of an altered epigenome as a major driver of DLBCL pathogenesis.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Demethylation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Signal Transduction/drug effects , Smad1 Protein/genetics , Smad1 Protein/metabolism , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Disease Models, Animal , Gene Silencing , Humans , Immunohistochemistry , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Xenograft Model Antitumor Assays
3.
EMBO Mol Med ; 11(10): e10576, 2019 10.
Article in English | MEDLINE | ID: mdl-31515941

ABSTRACT

Interleukin-6 (IL-6) is a growth factor for normal B cells and plasma cell-derived malignancies. Here, we show that the IL-6 signaling pathway is also active in a subset of diffuse large B-cell lymphoma (DLBCL) patients with particularly poor prognosis. Primary DLBCL cells and DLBCL cell lines expressing IL-6R engraft and form orthotopic lymphomas in humanized mice that ectopically produce human IL-6, and in mice reconstituted with a human immune system. We show that a subset of DLBCL cases have evolved mechanisms that ensure constitutive activation of the IL-6 signaling pathway, i.e., the expression of both chains of the IL-6R, the expression of the cytokine itself, and the mutational inactivation of a negative regulator of IL-6 signaling, SOCS1. IL-6 signaling promotes MYC-driven lymphomagenesis in a genetically engineered model, and treatment with the IL-6R-specific antibody tocilizumab reduces growth of primary DLBCL cells and of DLBCL cell lines in various therapeutic settings. The combined results uncover the IL-6 signaling pathway as a driver and negative prognosticator in aggressive DLBCL that can be targeted with a safe and well-tolerated biologic.


Subject(s)
Lymphoma, Large B-Cell, Diffuse/physiopathology , Receptors, Interleukin-6/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein/metabolism , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Disease Models, Animal , Humans , Lymphoma, Large B-Cell, Diffuse/therapy , Mice, SCID , Tumor Cells, Cultured
4.
Blood ; 131(20): 2235-2246, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29615404

ABSTRACT

The sphingosine-1-phosphate receptor S1PR2 and its downstream signaling pathway are commonly silenced in diffuse large B-cell lymphoma (DLBCL), either by mutational inactivation or through negative regulation by the oncogenic transcription factor FOXP1. In this study, we examined the upstream regulators of S1PR2 expression and have newly identified the transforming growth factor-ß (TGF-ß)/TGF-ßR2/SMAD1 axis as critically involved in S1PR2 transcriptional activation. Phosphorylated SMAD1 directly binds to regulatory elements in the S1PR2 locus as assessed by chromatin immunoprecipitation, and the CRISPR-mediated genomic editing of S1PR2, SMAD1, or TGFBR2 in DLBCL cell lines renders cells unresponsive to TGF-ß-induced apoptosis. DLBCL clones lacking any 1 of the 3 factors have a clear growth advantage in vitro, as well as in subcutaneous xenotransplantation models, and in a novel model of orthotopic growth of DLBCL cells in the spleens and bone marrow of MISTRG mice expressing various human cytokines. The loss of S1pr2 induces hyperproliferation of the germinal center (GC) B-cell compartment of immunized mice and accelerates MYC-driven lymphomagenesis in spontaneous and serial transplantation models. The specific loss of Tgfbr2 in murine GC B-cell phenocopies the effects of S1pr2 loss on GC B-cell hyperproliferation. Finally, we show that SMAD1 expression is aberrantly downregulated in >85% of analyzed DLBCL patients. The combined results uncover an important novel tumor suppressive function of the TGF-ß/TGF-ßR2/SMAD1/S1PR2 axis in DLBCL, and show that DLBCL cells have evolved to inactivate the pathway at the level of SMAD1 expression.


Subject(s)
Lymphoma, Large B-Cell, Diffuse/metabolism , Receptors, Lysosphingolipid/metabolism , Signal Transduction , Smad1 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Apoptosis , CRISPR-Cas Systems , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Gene Targeting , Germinal Center/pathology , Humans , Mice , Mice, Knockout , Models, Biological , Sphingosine-1-Phosphate Receptors
5.
Leuk Lymphoma ; 59(7): 1710-1716, 2018 07.
Article in English | MEDLINE | ID: mdl-29043881

ABSTRACT

Investigation of cancer cell lines is important for oncology research to characterize and understand mechanisms of cellular signaling and survival strategies in cancer. We analyzed the mutational profile of 11 diffuse large B-cell lymphoma (DLBCL) cell lines using a customized high throughput sequencing panel. We compared our data to previously published mutation data to better characterize these cell lines and establish consensus on the mutational status of some functionally relevant genes. With our targeted sequencing approach we detected 61 somatic mutations. The most frequently affected gene was TP53. MYD88 mutations were only seen in activated B-cell like cell lines. Overall comparison across different datasets revealed that just around 38% of mutations are reliable and can be validated by at least two independent observations whereas 24% of mutations could not be validated. Our analysis reveals considerable discrepancies regarding the mutational profile of some well-established cell lines.


Subject(s)
Biomarkers, Tumor , Lymphoma, Large B-Cell, Diffuse/genetics , Mutation , Cell Line, Tumor , Computational Biology/methods , DNA Mutational Analysis/methods , Databases, Genetic , Genetic Association Studies , Genetic Predisposition to Disease , Humans
6.
Proc Natl Acad Sci U S A ; 114(36): 9701-9706, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28831000

ABSTRACT

The genes encoding the histone acetyl-transferases (HATs) CREB binding protein (CREBBP) and EP300 are recurrently mutated in the activated B cell-like and germinal center (GC) B cell-like subtypes of diffuse large B cell lymphoma (DLBCL). Here, we introduced a patient mutation into a human DLBCL cell line using CRISPR and deleted Crebbp and Ep300 in the GC B cell compartment of mice. CREBBP-mutant DLBCL clones exhibited reduced histone H3 acetylation, expressed significantly less MHCII, and grew faster than wild-type clones in s.c. and orthotopic xenograft models. Mice lacking Crebbp in GC B cells exhibited hyperproliferation of their GC compartment upon immunization, had reduced MHCII surface expression on GC cells, and developed accelerated MYC-driven lymphomas. Ep300 inactivation reproduced some, but not all, consequences of Crebbp inactivation. MHCII deficiency phenocopied the effects of CREBBP loss in spontaneous and serial transplantation models of MYC-driven lymphomagenesis, supporting the idea that the mutational inactivation of CREBBP promotes immune evasion. Indeed, the depletion of CD4+ T cells greatly facilitated the engraftment of lymphoma cells in serial transplantation models. In summary, we provide evidence that both HATs are bona fide tumor suppressors that control MHCII expression and promote tumor immune control; mutational inactivation of CREBBP, but not of EP300, has additional cell-intrinsic engraftment and growth-promoting effects.


Subject(s)
CREB-Binding Protein/antagonists & inhibitors , CREB-Binding Protein/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CREB-Binding Protein/deficiency , CREB-Binding Protein/immunology , CRISPR-Cas Systems , Cell Line, Tumor , Cell Proliferation/genetics , Down-Regulation , E1A-Associated p300 Protein/antagonists & inhibitors , E1A-Associated p300 Protein/deficiency , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/immunology , Gene Deletion , Genes, MHC Class II , Germinal Center/immunology , Germinal Center/pathology , HLA Antigens/genetics , Heterografts , Histone Code/genetics , Humans , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...