Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 19220, 2021 09 28.
Article in English | MEDLINE | ID: mdl-34584159

ABSTRACT

Targeted pharmacologic activation of antigen-specific (AgS) T cells may bypass limitations inherent in current T cell-based cancer therapies. We describe two immunotherapeutics platforms for selective delivery of costimulatory ligands and peptide-HLA (pHLA) to AgS T cells. We engineered and deployed on these platforms an affinity-attenuated variant of interleukin-2, which selectively expands oligoclonal and polyfunctional AgS T cells in vitro and synergizes with CD80 signals for superior proliferation versus peptide stimulation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Neoplasms/therapy , Recombinant Fusion Proteins/immunology , Animals , B7-1 Antigen/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , HLA-A Antigens/genetics , HLA-A Antigens/immunology , Humans , Lymphocyte Activation , Mice , Mice, Transgenic , Mutation , Neoplasms/immunology , Peptides/genetics , Peptides/immunology , Primary Cell Culture , Protein Engineering , Recombinant Fusion Proteins/genetics
2.
Clin Cancer Res ; 26(8): 1953-1964, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31964784

ABSTRACT

PURPOSE: To assess the potential for CUE-101, a novel therapeutic fusion protein, to selectively activate and expand HPV16 E711-20-specific CD8+ T cells as an off-the shelf therapy for the treatment of HPV16-driven tumors, including head and neck squamous cell carcinoma (HNSCC), cervical, and anal cancers. EXPERIMENTAL DESIGN: CUE-101 is an Fc fusion protein composed of a human leukocyte antigen (HLA) complex, an HPV16 E7 peptide epitope, reduced affinity human IL2 molecules, and an effector attenuated human IgG1 Fc domain. Human E7-specific T cells and human peripheral blood mononuclear cells (PBMC) were tested to demonstrate cellular activity and specificity of CUE-101, whereas in vivo activity of CUE-101 was assessed in HLA-A2 transgenic mice. Antitumor efficacy with a murine surrogate (mCUE-101) was tested in the TC-1 syngeneic tumor model. RESULTS: CUE-101 demonstrates selective binding, activation, and expansion of HPV16 E711-20-specific CD8+ T cells from PBMCs relative to nontarget cells. Intravenous administration of CUE-101 induced selective expansion of HPV16 E711-20-specific CD8+ T cells in HLA-A2 (AAD) transgenic mice, and anticancer efficacy and immunologic memory was demonstrated in TC-1 tumor-bearing mice treated with mCUE-101. Combination therapy with anti-PD-1 checkpoint blockade further enhanced the observed efficacy. CONCLUSIONS: Consistent with its design, CUE-101 demonstrates selective expansion of an HPV16 E711-20-specific population of cytotoxic CD8+ T cells, a favorable safety profile, and in vitro and in vivo evidence supporting its potential for clinical efficacy in an ongoing phase I trial (NCT03978689).


Subject(s)
CD8-Positive T-Lymphocytes/immunology , HLA-A2 Antigen/immunology , Immunoglobulin Fc Fragments/immunology , Interleukin-2/immunology , Neoplasms/therapy , Papillomavirus E7 Proteins/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Animals , Cells, Cultured , Disease Models, Animal , Female , Healthy Volunteers , Humans , Leukocytes, Mononuclear , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/immunology , Neoplasms/virology
4.
Blood Adv ; 2(21): 3012-3024, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30425065

ABSTRACT

Although the treatment paradigm for chronic lymphocytic leukemia (CLL) is rapidly changing, the disease remains incurable, except with allogeneic bone marrow transplantation, and resistance, relapsed disease, and partial responses persist as significant challenges. Recent studies have uncovered roles for epigenetic modification in the regulation of mechanisms contributing to malignant progression of CLL B cells. However, the extent to which epigenetic modifiers can be targeted for therapeutic benefit in CLL patients remains poorly explored. We report for the first time that expression of epigenetic modifier histone deacetylase 6 (HDAC6) is upregulated in CLL patient samples, cell lines, and euTCL1 transgenic mouse models compared with HDAC6 in normal controls. Genetic silencing of HDAC6 conferred survival benefit in euTCL1 mice. Administration of isoform-specific HDAC6 inhibitor ACY738 in the euTCL1 aging and adoptive transfer models deterred proliferation of CLL B cells, delayed disease onset via disruption of B-cell receptor signaling, and sensitized CLL B cells to apoptosis. Furthermore, coadministration of ACY738 and ibrutinib displayed synergistic cell kill against CLL cell lines and improved overall survival compared with either single agent in vivo. These results demonstrate for the first time the therapeutic efficacy of selective HDAC6 inhibition in preclinical CLL models and suggest a rationale for the clinical development of HDAC6 inhibitors for CLL treatment, either alone or in combination with Bruton tyrosine kinase inhibition.


Subject(s)
Gene Silencing , Histone Deacetylase 6/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Adenine/analogs & derivatives , Animals , Antigens, CD19/metabolism , Apoptosis/drug effects , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase 6/genetics , Humans , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Piperidines , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins/metabolism , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Survival Rate
5.
Cancer Res ; 78(13): 3709-3717, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29760044

ABSTRACT

Small-cell lung cancer (SCLC) has the highest malignancy among all lung cancers, exhibiting aggressive growth and early metastasis to distant sites. For 30 years, treatment options for SCLC have been limited to chemotherapy, warranting the need for more effective treatments. Frequent inactivation of TP53 and RB1 as well as histone dysmodifications in SCLC suggest that transcriptional and epigenetic regulations play a major role in SCLC disease evolution. Here we performed a synthetic lethal screen using the BET inhibitor JQ1 and an shRNA library targeting 550 epigenetic genes in treatment-refractory SCLC xenograft models and identified HDAC6 as a synthetic lethal target in combination with JQ1. Combined treatment of human and mouse SCLC cell line-derived xenograft tumors with the HDAC6 inhibitor ricolinostat (ACY-1215) and JQ1 demonstrated significant inhibition of tumor growth; this effect was abolished upon depletion of NK cells, suggesting that these innate immune lymphoid cells play a role in SCLC tumor treatment response. Collectively, these findings suggest a potential new treatment for recurrent SCLC.Significance: These findings identify a novel therapeutic strategy for SCLC using a combination of HDAC6 and BET inhibitors. Cancer Res; 78(13); 3709-17. ©2018 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azepines/pharmacology , Histone Deacetylase 6/antagonists & inhibitors , Killer Cells, Natural/immunology , Lung Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Proteins/antagonists & inhibitors , Small Cell Lung Carcinoma/drug therapy , Triazoles/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azepines/therapeutic use , Cell Line, Tumor , Cell Proliferation , Drug Synergism , Histone Deacetylase 6/genetics , Humans , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , RNA, Small Interfering/metabolism , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Synthetic Lethal Mutations/genetics , Treatment Outcome , Triazoles/therapeutic use , Xenograft Model Antitumor Assays
6.
Cancer Discov ; 7(8): 852-867, 2017 08.
Article in English | MEDLINE | ID: mdl-28408401

ABSTRACT

Effective therapies for non-small cell lung cancer (NSCLC) remain challenging despite an increasingly comprehensive understanding of somatically altered oncogenic pathways. It is now clear that therapeutic agents with potential to impact the tumor immune microenvironment potentiate immune-orchestrated therapeutic benefit. Herein, we evaluated the immunoregulatory properties of histone deacetylase (HDAC) and bromodomain inhibitors, two classes of drugs that modulate the epigenome, with a focus on key cell subsets that are engaged in an immune response. By evaluating human peripheral blood and NSCLC tumors, we show that the selective HDAC6 inhibitor ricolinostat promotes phenotypic changes that support enhanced T-cell activation and improved function of antigen-presenting cells. The bromodomain inhibitor JQ1 attenuated CD4+FOXP3+ T regulatory cell suppressive function and synergized with ricolinostat to facilitate immune-mediated tumor growth arrest, leading to prolonged survival of mice with lung adenocarcinomas. Collectively, our findings highlight the immunomodulatory effects of two epigenetic modifiers that, together, promote T cell-mediated antitumor immunity and demonstrate their therapeutic potential for treatment of NSCLC.Significance: Selective inhibition of HDACs and bromodomain proteins modulates tumor-associated immune cells in a manner that favors improved T-cell function and reduced inhibitory cellular mechanisms. These effects facilitated robust antitumor responses in tumor-bearing mice, demonstrating the therapeutic potential of combining these epigenetic modulators for the treatment of NSCLC. Cancer Discov; 7(8); 852-67. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 783.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Histone Deacetylase Inhibitors/administration & dosage , Hydroxamic Acids/administration & dosage , Pyrimidines/administration & dosage , Aged , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Female , Histone Deacetylases/genetics , Histone Deacetylases/immunology , Humans , Hydroxamic Acids/adverse effects , Immunotherapy , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Middle Aged , Pyrimidines/adverse effects , Xenograft Model Antitumor Assays
8.
PLoS One ; 12(3): e0173507, 2017.
Article in English | MEDLINE | ID: mdl-28264055

ABSTRACT

Thalidomide-based Immunomodulatory Drugs (IMiDs®), including lenalidomide and pomalidomide, are effective therapeutics for multiple myeloma. These agents have been approved with, or are under clinical development with, other targeted therapies including proteasome inhibitors, αCD38 monoclonal antibodies, as well as histone deacetylase (HDAC) inhibitors for combination therapy. HDAC inhibitors broadly targeting Class I and IIb HDACs have shown potent preclinical efficacy but have frequently demonstrated an undesirable safety profile in combination therapy approaches in clinical studies. Therefore, development of more selective HDAC inhibitors could provide enhanced efficacy with reduced side effects in combination with IMiDs® for the treatment of B-cell malignancies, including multiple myeloma. Here, the second generation selective HDAC6 inhibitor citarinostat (ACY-241), with a more favorable safety profile than non-selective pan-HDAC inhibitors, is shown to synergize with pomalidomide in in vitro assays through promoting greater apoptosis and cell cycle arrest. Furthermore, utilizing a multiple myeloma in vivo murine xenograft model, combination treatment with pomalidomide and ACY-241 leads to increased tumor growth inhibition. At the molecular level, combination treatment with ACY-241 and pomalidomide leads to greater suppression of the pro-survival factors survivin, Myc, and IRF4. The results presented here demonstrate synergy between pomalidomide and ACY-241 in both in vitro and in vivo preclinical models, providing further impetus for clinical development of ACY-241 for use in combination with IMiDs for patients with multiple myeloma and potentially other B-cell malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Thalidomide/analogs & derivatives , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Drug Synergism , Female , Histone Deacetylase 6 , Humans , Mice , Thalidomide/pharmacology , Xenograft Model Antitumor Assays
9.
PLoS One ; 12(1): e0169128, 2017.
Article in English | MEDLINE | ID: mdl-28060870

ABSTRACT

Acute myeloid leukemia (AML) is a heterogeneous group of hematopoietic stem cell disorders characterized by defects in myeloid differentiation and increased proliferation of neoplastic hematopoietic precursor cells. Outcomes for patients with AML remain poor, highlighting the need for novel treatment options. Aberrant epigenetic regulation plays an important role in the pathogenesis of AML, and inhibitors of DNA methyltransferase or histone deacetylase (HDAC) enzymes have exhibited activity in preclinical AML models. Combination studies with HDAC inhibitors plus DNA methyltransferase inhibitors have potential beneficial clinical activity in AML, however the toxicity profiles of non-selective HDAC inhibitors in the combination setting limit their clinical utility. In this work, we describe the preclinical development of selective inhibitors of HDAC1 and HDAC2, which are hypothesized to have improved safety profiles, for combination therapy in AML. We demonstrate that selective inhibition of HDAC1 and HDAC2 is sufficient to achieve efficacy both as a single agent and in combination with azacitidine in preclinical models of AML, including established AML cell lines, primary leukemia cells from AML patient bone marrow samples and in vivo xenograft models of human AML. Gene expression profiling of AML cells treated with either an HDAC1/2 inhibitor, azacitidine, or the combination of both have identified a list of genes involved in transcription and cell cycle regulation as potential mediators of the combinatorial effects of HDAC1/2 inhibition with azacitidine. Together, these findings support the clinical evaluation of selective HDAC1/2 inhibitors in combination with azacitidine in AML patients.


Subject(s)
Antineoplastic Agents/pharmacology , Azacitidine/pharmacology , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Leukemia, Myeloid, Acute/metabolism , Animals , Biomarkers , Bone Marrow Cells , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Drug Synergism , Female , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Gene Expression Profiling , Gene Expression Regulation, Leukemic/drug effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mice , Molecular Targeted Therapy , Xenograft Model Antitumor Assays
10.
Oncotarget ; 8(2): 2694-2707, 2017 Jan 10.
Article in English | MEDLINE | ID: mdl-27926524

ABSTRACT

ACY-241 is a novel, orally available and selective histone deacetylase (HDAC) 6 inhibitor in Phase 1b clinical development in multiple myeloma (NCT 02400242). Like the structurally related drug ACY-1215 (ricolinostat), ACY-241 has the potential for a substantially reduced side effect profile versus current nonselective HDAC inhibitor drug candidates due to reduced potency against Class I HDACs while retaining the potential for anticancer effectiveness. We now show that combination treatment of xenograft models with paclitaxel and either ricolinostat or ACY-241 significantly suppresses solid tumor growth. In cell lines from multiple solid tumor lineages, combination treatment with ACY-241 and paclitaxel enhanced inhibition of proliferation and increased cell death relative to either single agent alone. Combination treatment with ACY-241 and paclitaxel also resulted in more frequent occurrence of mitotic cells with abnormal multipolar spindles and aberrant mitoses, consistent with the observed increase of aneuploid cells. At the molecular level, multipolar mitotic spindle formation was observed to be NuMA-dependent and γ-tubulin independent, suggesting that treatment-induced multipolar spindle formation does not depend on centrosomal amplification. The significantly enhanced efficacy of ACY-241 plus paclitaxel observed here, in addition to the anticipated superior safety profile of a selective HDAC6 inhibitor versus pan-HDAC inhibitors, provides a strong rationale for clinical development of this combination in patients with advanced solid tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Paclitaxel/pharmacology , Acetylation , Animals , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Drug Synergism , Female , Histone Deacetylase Inhibitors/chemistry , Humans , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Tubulin/metabolism , Xenograft Model Antitumor Assays
11.
Breast Cancer Res ; 17(1): 149, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26643555

ABSTRACT

INTRODUCTION: Inflammatory breast cancer (IBC) is the most lethal form of breast cancers with a 5-year survival rate of only 40 %. Despite its lethality, IBC remains poorly understood which has greatly limited its therapeutic management. We thus decided to utilize an integrative functional genomic strategy to identify the Achilles' heel of IBC cells. METHODS: We have pioneered the development of genetic tools as well as experimental and analytical strategies to perform RNAi-based loss-of-function studies at a genome-wide level. Importantly, we and others have demonstrated that these functional screens are able to identify essential functions linked to certain cancer phenotypes. Thus, we decided to use this approach to identify IBC specific sensitivities. RESULTS: We identified and validated HDAC6 as a functionally necessary gene to maintain IBC cell viability, while being non-essential for other breast cancer subtypes. Importantly, small molecule inhibitors for HDAC6 already exist and are in clinical trials for other tumor types. We thus demonstrated that Ricolinostat (ACY1215), a leading HDAC6 inhibitor, efficiently controls IBC cell proliferation both in vitro and in vivo. Critically, functional HDAC6 dependency is not associated with genomic alterations at its locus and thus represents a non-oncogene addiction. Despite HDAC6 not being overexpressed, we found that its activity is significantly higher in IBC compared to non-IBC cells, suggesting a possible rationale supporting the observed dependency. CONCLUSION: Our finding that IBC cells are sensitive to HDAC6 inhibition provides a foundation to rapidly develop novel, efficient, and well-tolerated targeted therapy strategies for IBC patients.


Subject(s)
Histone Deacetylases/metabolism , Inflammatory Breast Neoplasms/enzymology , Cell Line, Tumor , Cell Survival , Female , Gene Expression , Gene Knockdown Techniques , Gene Ontology , Histone Deacetylase 6 , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Humans , Inflammatory Breast Neoplasms/pathology
12.
Br J Haematol ; 169(3): 423-34, 2015 May.
Article in English | MEDLINE | ID: mdl-25709080

ABSTRACT

Proteasome inhibition induces the accumulation of aggregated misfolded/ubiquitinated proteins in the aggresome; conversely, histone deacetylase 6 (HDAC6) inhibition blocks aggresome formation. Although this rationale has been the basis of proteasome inhibitor (PI) and HDAC6 inhibitor combination studies, the role of disruption of aggresome formation by HDAC6 inhibition has not yet been studied in multiple myeloma (MM). The present study aimed to evaluate the impact of carfilzomib (CFZ) in combination with a selective HDAC6 inhibitor (ricolinostat) in MM cells with respect to the aggresome-proteolysis pathway. We observed that combination treatment of CFZ with ricolinostat triggered synergistic anti-MM effects, even in bortezomib-resistant cells. Immunofluorescent staining showed that CFZ increased the accumulation of ubiquitinated proteins and protein aggregates in the cytoplasm, as well as the engulfment of aggregated ubiquitinated proteins by autophagosomes, which was blocked by ricolinostat. Electron microscopy imaging showed increased autophagy triggered by CFZ, which was inhibited by the addition of ACY-1215. Finally, an in vivo mouse xenograft study confirmed a decrease in tumour volume, associated with apoptosis, following treatment with CFZ in combination with ricolinostat. Our results suggest that ricolinostat inhibits aggresome formation, caused by CFZ-induced inhibition of the proteasome pathway, resulting in enhanced apoptosis in MM cells.


Subject(s)
Apoptosis/drug effects , Hydroxamic Acids/pharmacology , Multiple Myeloma/metabolism , Oligopeptides/pharmacology , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Drug Synergism , Endoplasmic Reticulum Stress/drug effects , Female , Heterografts , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Phagosomes/metabolism , Proteasome Inhibitors/pharmacology
13.
Oncotarget ; 6(7): 4863-87, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25605023

ABSTRACT

Gain-of-function mutations in the catalytic site of EZH2 (Enhancer of Zeste Homologue 2), is observed in about 22% of diffuse large B-cell lymphoma (DLBCL) cases. Here we show that selective inhibition of histone deacetylase 1,2 (HDAC1,2) activity using a small molecule inhibitor causes cytotoxic or cytostatic effects in EZH2 gain-of-function mutant (EZH2GOF) DLBCL cells. Our results show that blocking the activity of HDAC1,2 increases global H3K27ac without causing a concomitant global decrease in H3K27me3 levels. Our data shows that inhibition of HDAC1,2 is sufficient to decrease H3K27me3 present at DSBs, decrease DSB repair and activate the DNA damage response in these cells. In addition to increased H3K27me3, we found that the EZH2GOF DLBCL cells overexpress another chemotherapy resistance factor - B-lymphoma and BAL-associated protein (BBAP). BBAP monoubiquitinates histone H4K91, a residue that is also subjected to acetylation. Our results show that selective inhibition of HDAC1,2 increases H4K91ac, decreases BBAP-mediated H4K91 monoubiquitination, impairs BBAP-dependent DSB repair and sensitizes the refractory EZH2GOF DLBCL cells to treatment with doxorubicin, a chemotherapy agent. Hence, selective HDAC1,2 inhibition provides a novel DNA repair mechanism-based therapeutic approach as it can overcome both EZH2- and BBAP-mediated DSB repair in the EZH2GOF DLBCL cells.


Subject(s)
DNA Repair , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Polycomb Repressive Complex 2/metabolism , Ubiquitin-Protein Ligases/metabolism , Adult , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein , HeLa Cells , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Humans , Lymphoma, Large B-Cell, Diffuse/metabolism , Male , Polycomb Repressive Complex 2/genetics , Transfection , Ubiquitin-Protein Ligases/genetics
14.
Mol Cancer Ther ; 13(12): 2886-97, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25239935

ABSTRACT

Interactions between the HDAC6 inhibitor ricolinostat (ACY1215) and the irreversible proteasome inhibitor carfilzomib were examined in non-Hodgkin lymphoma (NHL) models, including diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), and double-hit lymphoma cells. Marked in vitro synergism was observed in multiple cell types associated with activation of cellular stress pathways (e.g., JNK1/2, ERK1/2, and p38) accompanied by increases in DNA damage (γH2A.X), G2-M arrest, and the pronounced induction of mitochondrial injury and apoptosis. Combination treatment with carfilzomib and ricolinostat increased reactive oxygen species (ROS), whereas the antioxidant TBAP attenuated DNA damage, JNK activation, and cell death. Similar interactions occurred in bortezomib-resistant and double-hit DLBCL, MCL, and primary DLBCL cells, but not in normal CD34(+) cells. However, ricolinostat did not potentiate inhibition of chymotryptic activity by carfilzomib. shRNA knockdown of JNK1 (but not MEK1/2), or pharmacologic inhibition of p38, significantly reduced carfilzomib-ricolinostat lethality, indicating a functional contribution of these stress pathways to apoptosis. Combined exposure to carfilzomib and ricolinostat also markedly downregulated the cargo-loading protein HR23B. Moreover, HR23B knockdown significantly increased carfilzomib- and ricolinostat-mediated lethality, suggesting a role for this event in cell death. Finally, combined in vivo treatment with carfilzomib and ricolinostat was well tolerated and significantly suppressed tumor growth and increased survival in an MCL xenograft model. Collectively, these findings indicate that carfilzomib and ricolinostat interact synergistically in NHL cells through multiple stress-related mechanisms, and suggest that this strategy warrants further consideration in NHL.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Lymphoma, Non-Hodgkin/metabolism , Proteasome Inhibitors/pharmacology , Pyrimidines/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Drug Interactions , Drug Synergism , Female , Gene Knockdown Techniques , Histone Deacetylase 6 , Histone Deacetylases/genetics , Humans , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/pathology , Oligopeptides/pharmacology , Oxidative Stress/drug effects , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
PLoS One ; 7(11): e49466, 2012.
Article in English | MEDLINE | ID: mdl-23166678

ABSTRACT

The phosphoinositide 3-kinase (PI3K) pathway is targeted for frequent alteration in glioblastoma (GBM) and is one of the core GBM pathways defined by The Cancer Genome Atlas. Somatic mutations of PIK3R1 are observed in multiple tumor types, but the tumorigenic activity of these mutations has not been demonstrated in GBM. We show here that somatic mutations in the iSH2 domain of PIK3R1 act as oncogenic driver events. Specifically, introduction of a subset of the mutations identified in human GBM, in the nSH2 and iSH2 domains, increases signaling through the PI3K pathway and promotes tumorigenesis of primary normal human astrocytes in an orthotopic xenograft model. Furthermore, we show that cells that are dependent on mutant P85α-mediated PI3K signaling exhibit increased sensitivity to a small molecule inhibitor of AKT. Together, these results suggest that GBM patients whose tumors carry mutant PIK3R1 alleles may benefit from treatment with inhibitors of AKT.


Subject(s)
Astrocytes/metabolism , Cell Transformation, Neoplastic/genetics , Class Ia Phosphatidylinositol 3-Kinase/genetics , Glioblastoma/genetics , Signal Transduction/genetics , Analysis of Variance , Cell Survival/drug effects , Cell Transformation, Neoplastic/metabolism , Dimethyl Sulfoxide/toxicity , Dose-Response Relationship, Drug , Heterocyclic Compounds, 3-Ring/toxicity , Humans , Immunoblotting , Mutagenesis , Mutation/genetics , Plasmids/genetics
16.
Neuro Oncol ; 14(11): 1325-31, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23074196

ABSTRACT

Large-scale cancer genomics efforts are identifying hundreds of somatic genomic alterations in glioblastoma (GBM). Distinguishing between active driver and neutral passenger alterations requires functional assessment of each gene; therefore, integrating biological weight of evidence with statistical significance for each genomic alteration will enable better prioritization for downstream studies. Here, we demonstrate the feasibility and potential of in vitro functional genomic screens to rapidly and systematically prioritize high-probability candidate genes for in vivo validation. Integration of low-complexity gain- and loss-of-function screens designed on the basis of genomic data identified 6 candidate GBM oncogenes, and RINT1 was validated as a novel GBM oncogene based on its ability to confer tumorigenicity to primary nontransformed murine astrocytes in vivo. Cancer genomics-guided low-complexity genomic screens can quickly provide a functional filter to prioritize high-value targets for further downstream mechanistic and translational studies.


Subject(s)
Brain Neoplasms/genetics , Cell Cycle Proteins/genetics , Cell Transformation, Neoplastic/genetics , Glioblastoma/genetics , Oncogenes/genetics , Animals , Comparative Genomic Hybridization , Genomics , Humans , Immunoblotting , Immunohistochemistry , Mice , Mice, Nude
17.
Cancer Discov ; 2(8): 736-49, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22750848

ABSTRACT

UNLABELLED: Leveraging The Cancer Genome Atlas (TCGA) multidimensional data in glioblastoma, we inferred the putative regulatory network between microRNA and mRNA using the Context Likelihood of Relatedness modeling algorithm. Interrogation of the network in context of defined molecular subtypes identified 8 microRNAs with a strong discriminatory potential between proneural and mesenchymal subtypes. Integrative in silico analyses, a functional genetic screen, and experimental validation identified miR-34a as a tumor suppressor in proneural subtype glioblastoma. Mechanistically, in addition to its direct regulation of platelet-derived growth factor receptor-alpha (PDGFRA), promoter enrichment analysis of context likelihood of relatedness-inferred mRNA nodes established miR-34a as a novel regulator of a SMAD4 transcriptional network. Clinically, miR-34a expression level is shown to be prognostic, where miR-34a low-expressing glioblastomas exhibited better overall survival. This work illustrates the potential of comprehensive multidimensional cancer genomic data combined with computational and experimental models in enabling mechanistic exploration of relationships among different genetic elements across the genome space in cancer. SIGNIFICANCE: We illustrate here that network modeling of complex multidimensional cancer genomic data can generate a framework in which to explore the biology of cancers, leading to discovery of new pathogenetic insights as well as potential prognostic biomarkers. Specifically in glioblastoma, within the context of the global network, promoter enrichment analysis of network edges uncovered a novel regulation of TGF-ß signaling via a Smad4 transcriptomic network by miR-34a.


Subject(s)
Glioblastoma/genetics , MicroRNAs/genetics , Transforming Growth Factor beta/metabolism , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genes, Tumor Suppressor , Glioblastoma/metabolism , Humans , Mice , MicroRNAs/metabolism , Prognosis , Signal Transduction , Transforming Growth Factor beta/genetics
18.
Genes Dev ; 26(8): 756-84, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22508724

ABSTRACT

Glioblastoma is both the most common and lethal primary malignant brain tumor. Extensive multiplatform genomic characterization has provided a higher-resolution picture of the molecular alterations underlying this disease. These studies provide the emerging view that "glioblastoma" represents several histologically similar yet molecularly heterogeneous diseases, which influences taxonomic classification systems, prognosis, and therapeutic decisions.


Subject(s)
Brain Neoplasms/classification , Brain Neoplasms/genetics , Glioblastoma/classification , Glioblastoma/genetics , Brain Neoplasms/pathology , Gene Expression Profiling , Genes, Tumor Suppressor , Genomics , Glioblastoma/pathology , Humans , Neovascularization, Pathologic/genetics , Transcription, Genetic
19.
Proc Natl Acad Sci U S A ; 107(25): 11501-6, 2010 Jun 22.
Article in English | MEDLINE | ID: mdl-20534551

ABSTRACT

Glioblastoma multiforme (GBM) is a fatal primary brain tumor harboring myriad genetic and epigenetic alterations. The recent multidimensional analysis of the GBM genome has provided a more complete view of the landscape of such alterations and their linked pathways. This effort has demonstrated that certain pathways are universally altered, but that the specific genetic events altered within each pathway can vary for each particular patient's tumor. With this atlas of genetic and epigenetic events, it now becomes feasible to assess how the patterns of mutations in a pathway influence response to drugs that are targeting such pathways. This issue is particularly important for GBM because, in contrast to other tumor types, molecularly targeted therapies have failed to alter overall survival substantially. Here, we combined functional genetic screens and comprehensive genomic analyses to identify CDK6 as a GBM oncogene that is required for proliferation and viability in a subset of GBM cell lines and tumors. Using an available small molecule targeting cyclin-dependent kinases (CDKs) 4 and 6, we sought to determine if the specific pattern of retinoblastoma pathway inactivation dictated the response to CDK4/6 inhibitor therapy. We showed that codeletion of CDKN2A and CDKN2C serves as a strong predictor of sensitivity to a selective inhibitor of CDK4/6. Thus, genome-informed drug sensitivity studies identify a subset of GBMs likely to respond to CDK4/6 inhibition. More generally, these observations demonstrate that the integration of genomic, functional and pharmacologic data can be exploited to inform the development of targeted therapy directed against specific cancer pathways.


Subject(s)
Central Nervous System Neoplasms/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Retinoblastoma Protein/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Central Nervous System Neoplasms/drug therapy , Cyclin-Dependent Kinase Inhibitor p18/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic , Glioblastoma/drug therapy , Humans , Inhibitory Concentration 50 , Mice , Neoplasm Transplantation , Piperazines/pharmacology , Pyridines/pharmacology
20.
Cancer Cell ; 17(5): 497-509, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20478531

ABSTRACT

A hallmark feature of glioblastoma is its strong self-renewal potential and immature differentiation state, which contributes to its plasticity and therapeutic resistance. Here, integrated genomic and biological analyses identified PLAGL2 as a potent protooncogene targeted for amplification/gain in malignant gliomas. Enhanced PLAGL2 expression strongly suppresses neural stem cell (NSC) and glioma-initiating cell differentiation while promoting their self-renewal capacity upon differentiation induction. Transcriptome analysis revealed that these differentiation-suppressive activities are attributable in part to PLAGL2 modulation of Wnt/beta-catenin signaling. Inhibition of Wnt signaling partially restores PLAGL2-expressing NSC differentiation capacity. The identification of PLAGL2 as a glioma oncogene highlights the importance of a growing class of cancer genes functioning to impart stem cell-like characteristics in malignant cells.


Subject(s)
Cell Differentiation/physiology , DNA-Binding Proteins/physiology , Glioblastoma/pathology , RNA-Binding Proteins/physiology , Signal Transduction/physiology , Stem Cells/cytology , Transcription Factors/physiology , Wnt Proteins/metabolism , Animals , Cell Transformation, Neoplastic , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...