Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Discov ; 10(9): 1296-1311, 2020 09.
Article in English | MEDLINE | ID: mdl-32371478

ABSTRACT

The molecular mechanisms leading to resistance to PD-1 blockade are largely unknown. Here, we characterize tumor biopsies from a patient with melanoma who displayed heterogeneous responses to anti-PD-1 therapy. We observe that a resistant tumor exhibited a loss-of-function mutation in the tumor suppressor gene FBXW7, whereas a sensitive tumor from the same patient did not. Consistent with a functional role in immunotherapy response, inactivation of Fbxw7 in murine tumor cell lines caused resistance to anti-PD-1 in immunocompetent animals. Loss of Fbxw7 was associated with altered immune microenvironment, decreased tumor-intrinsic expression of the double-stranded RNA (dsRNA) sensors MDA5 and RIG1, and diminished induction of type I IFN and MHC-I expression. In contrast, restoration of dsRNA sensing in Fbxw7-deficient cells was sufficient to sensitize them to anti-PD-1. Our results thus establish a new role for the commonly inactivated tumor suppressor FBXW7 in viral sensing and sensitivity to immunotherapy. SIGNIFICANCE: Our findings establish a role of the commonly inactivated tumor suppressor FBXW7 as a genomic driver of response to anti-PD-1 therapy. Fbxw7 loss promotes resistance to anti-PD-1 through the downregulation of viral sensing pathways, suggesting that therapeutic reactivation of these pathways could improve clinical responses to checkpoint inhibitors in genomically defined cancer patient populations.This article is highlighted in the In This Issue feature, p. 1241.


Subject(s)
Drug Resistance, Neoplasm/genetics , F-Box-WD Repeat-Containing Protein 7/genetics , Immune Checkpoint Inhibitors/pharmacology , Skin Neoplasms/drug therapy , Aged , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Cell Line, Tumor/transplantation , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Disease Models, Animal , F-Box-WD Repeat-Containing Protein 7/metabolism , Gene Expression Regulation, Neoplastic/immunology , HeLa Cells , Humans , Immune Checkpoint Inhibitors/therapeutic use , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/metabolism , Loss of Function Mutation , Male , Mice , Mutagenesis, Site-Directed , Programmed Cell Death 1 Receptor/antagonists & inhibitors , RNA, Double-Stranded/immunology , RNA, Double-Stranded/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
2.
Cell Cycle ; 11(16): 2985-90, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22825249

ABSTRACT

Induced pluripotent stem cells (iPSC) hold significant promise for advancing biomedical research. In the case of monogenic diseases, patient-iPSC and their derivatives contain the disease-causing mutation, suggesting the possibility of recapitulating salient disease features in vitro. Fanconi anemia (FA) is the most common inherited bone marrow failure syndrome. The etiology of bone marrow failure in FA remains largely unclear, but limited studies on patient bone marrow cells indicate cell intrinsic defects as causative. We examined the feasibility of modeling FA in a system based on hematopoietic differentiation of patient-specific iPSC. An informative iPSC-based model is predicated on the ability to derive disease-specific (uncorrected) patient iPSC that contain the disease-causing mutation, are pluripotent, maintain a normal karyotype and are capable of hematopoietic differentiation. Careful analysis of hematopoietic differentiation of such iPSC holds the promise of uncovering new insights into bone marrow failure and may enable high-throughput screening with the goal of identifying compounds that ameliorate hematopoietic failure. Ultimately, genetic correction, molecular characterization and successful engraftment of iPSC-derived cells may provide an attractive alternative to current hematopoietic stem cell-targeted gene therapy in some monogenic diseases, including FA.


Subject(s)
Fanconi Anemia/pathology , Hematopoiesis , Hematopoietic Stem Cells/pathology , Induced Pluripotent Stem Cells/pathology , Animals , Cellular Reprogramming , Chromosome Aberrations , Chromosomes, Human/genetics , Embryoid Bodies/pathology , Fanconi Anemia/genetics , Fibroblasts/pathology , Humans , Metaphase , Mice , Mice, SCID , Mutation , Xenograft Model Antitumor Assays/methods
3.
Blood ; 119(23): 5449-57, 2012 Jun 07.
Article in English | MEDLINE | ID: mdl-22371882

ABSTRACT

Fanconi anemia (FA) is a recessive syndrome characterized by progressive fatal BM failure and chromosomal instability. FA cells have inactivating mutations in a signaling pathway that is critical for maintaining genomic integrity and protecting cells from the DNA damage caused by cross-linking agents. Transgenic expression of the implicated genes corrects the phenotype of hematopoietic cells, but previous attempts at gene therapy have failed largely because of inadequate numbers of hematopoietic stem cells available for gene correction. Induced pluripotent stem cells (iPSCs) constitute an alternate source of autologous cells that are amenable to ex vivo expansion, genetic correction, and molecular characterization. In the present study, we demonstrate that reprogramming leads to activation of the FA pathway, increased DNA double-strand breaks, and senescence. We also demonstrate that defects in the FA DNA-repair pathway decrease the reprogramming efficiency of murine and human primary cells. FA pathway complementation reduces senescence and restores the reprogramming efficiency of somatic FA cells to normal levels. Disease-specific iPSCs derived in this fashion maintain a normal karyotype and are capable of hematopoietic differentiation. These data define the role of the FA pathway in reprogramming and provide a strategy for future translational applications of patient-specific FA iPSCs.


Subject(s)
Fanconi Anemia/genetics , Genetic Therapy/methods , Hematopoiesis , Induced Pluripotent Stem Cells/cytology , Animals , Cells, Cultured , DNA Damage , Fanconi Anemia/metabolism , Fanconi Anemia/therapy , Fanconi Anemia Complementation Group A Protein/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Deletion , Humans , Induced Pluripotent Stem Cells/metabolism , Karyotype , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...