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











Publication year range
1.
Front Immunol ; 11: 590072, 2020.
Article in English | MEDLINE | ID: mdl-33329575

ABSTRACT

Development of chronic lymphocytic leukemia (CLL) is associated with severe immune dysfunction. T-cell exhaustion, immune checkpoint upregulation, and increase of regulatory T cells contribute to an immunosuppressive tumor microenvironment. As a result, CLL patients are severely susceptible to infectious complications that increase morbidity and mortality. CLL B-cell survival is highly dependent upon interaction with the supportive tumor microenvironment. It has been postulated that the reversal of T-cell dysfunction in CLL may be beneficial to reduce tumor burden. Previous studies have also highlighted roles for histone deacetylase 6 (HDAC6) in regulation of immune cell phenotype and function. Here, we report for the first time that HDAC6 inhibition exerts beneficial immunomodulatory effects on CLL B cells and alleviates CLL-induced immunosuppression of CLL T cells. In the Eµ-TCL1 adoptive transfer murine model, genetic silencing or inhibition of HDAC6 reduced surface expression of programmed death-ligand 1 (PD-L1) on CLL B cells and lowered interleukin-10 (IL-10) levels. This occurred concurrently with a bolstered T-cell phenotype, demonstrated by alteration of coinhibitory molecules and activation status. Analysis of mice with similar tumor burden indicated that the majority of T-cell changes elicited by silencing or inhibition of HDAC6 in vivo are likely secondary to decrease of tumor burden and immunomodulation of CLL B cells. The data reported here suggest that CLL B cell phenotype may be altered by HDAC6-mediated hyperacetylation of the chaperone heat shock protein 90 (HSP90) and subsequent inhibition of the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway. Based on the beneficial immunomodulatory activity of HDAC6 inhibition, we rationalized that HDAC6 inhibitors could enhance immune checkpoint blockade in CLL. Conclusively, combination treatment with ACY738 augmented the antitumor efficacy of anti-PD-1 and anti-PD-L1 monoclonal antibodies in the Eµ-TCL1 adoptive transfer murine model. These combinatorial antitumor effects coincided with an increased cytotoxic CD8+ T-cell phenotype. Taken together, these data highlight a role for HDAC inhibitors in combination with immunotherapy and provides the rationale to investigate HDAC6 inhibition together with immune checkpoint blockade for treatment of CLL patients.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , T-Lymphocytes/drug effects , Aged , Animals , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Disease Models, Animal , Female , Histone Deacetylase 6/genetics , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Male , Mice, Transgenic , Middle Aged , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , T-Lymphocytes/immunology
3.
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
4.
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
5.
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
6.
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
7.
Clin Cancer Res ; 23(13): 3307-3315, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28053023

ABSTRACT

Purpose: Histone deacetylase (HDAC) inhibition improves the efficacy of proteasome inhibition for multiple myeloma but adds substantial toxicity. Preclinical models suggest that the observed synergy is due to the role of HDAC6 in mediating resistance to proteasome inhibition via the aggresome/autophagy pathway of protein degradation.Experimental Design: We conducted a phase I/II trial of the HDAC6-selective inhibitor ricolinostat to define the safety, preliminary efficacy, and recommended phase II dose in combination with standard proteasome inhibitor therapy. Patients with relapsed or refractory multiple myeloma received oral ricolinostat on days 1-5 and 8-12 of each 21-day cycle.Results: Single-agent ricolinostat therapy resulted in neither significant toxicity nor clinical responses. Combination therapy with bortezomib and dexamethasone was well-tolerated during dose escalation but led to dose-limiting diarrhea in an expansion cohort at a ricolinostat dose of 160 mg twice daily. Combination therapy at a ricolinostat dose of 160 mg daily in a second expansion cohort was well tolerated, with less severe hematologic, gastrointestinal, and constitutional toxicities compared with published data on nonselective HDAC inhibitors. The overall response rate in combination with daily ricolinostat at ≥160 mg was 37%. The response rate to combination therapy among bortezomib-refractory patients was 14%. Samples taken during therapy showed dose-dependent increases of acetylated tubulin in peripheral blood lymphocytes.Conclusions: At the recommended phase II dose of ricolinostat of 160 mg daily, the combination with bortezomib and dexamethasone is safe, well-tolerated, and active, suggesting that selective inhibition of HDAC6 is a promising approach to multiple myeloma therapy. Clin Cancer Res; 23(13); 3307-15. ©2017 AACR.


Subject(s)
Histone Deacetylase 6/genetics , Histone Deacetylase Inhibitors/administration & dosage , Hydroxamic Acids/administration & dosage , Multiple Myeloma/drug therapy , Pyrimidines/administration & dosage , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bortezomib/administration & dosage , Dexamethasone/administration & dosage , Drug Resistance, Neoplasm/genetics , Female , Histone Deacetylase 6/antagonists & inhibitors , Humans , Hydroxamic Acids/adverse effects , Male , Maximum Tolerated Dose , Middle Aged , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Pyrimidines/adverse effects
8.
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
9.
Lancet Oncol ; 17(11): 1569-1578, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27646843

ABSTRACT

BACKGROUND: Histone deacetylase (HDAC) inhibitors are an important new class of therapeutics for treating multiple myeloma. Ricolinostat (ACY-1215) is the first oral selective HDAC6 inhibitor with reduced class I HDAC activity to be studied clinically. Motivated by findings from preclinical studies showing potent synergistic activity with ricolinostat and lenalidomide, our goal was to assess the safety and preliminary activity of the combination of ricolinostat with lenalidomide and dexamethasone in relapsed or refractory multiple myeloma. METHODS: In this multicentre phase 1b trial, we recruited patients aged 18 years or older with previously treated relapsed or refractory multiple myeloma from five cancer centres in the USA. Inclusion criteria included a Karnofsky Performance Status score of at least 70, measureable disease, adequate bone marrow reserve, adequate hepatic function, and a creatinine clearance of at least 50 mL per min. Exclusion criteria included previous exposure to HDAC inhibitors; previous allogeneic stem-cell transplantation; previous autologous stem-cell transplantation within 12 weeks of baseline; active systemic infection; malignancy within the last 5 years; known or suspected HIV, hepatitis B, or hepatitis C infection; a QTc Fridericia of more than 480 ms; and substantial cardiovascular, gastrointestinal, psychiatric, or other medical disorders. We gave escalating doses (from 40-240 mg once daily to 160 mg twice daily) of oral ricolinostat according to a standard 3 + 3 design according to three different regimens on days 1-21 with a conventional 28 day schedule of oral lenalidomide (from 15 mg [in one cohort] to 25 mg [in all other cohorts] once daily) and oral dexamethasone (40 mg weekly). Primary outcomes were dose-limiting toxicities, the maximum tolerated dose of ricolinostat in this combination, and the dose and schedule of ricolinostat recommended for further phase 2 investigation. Secondary outcomes were the pharmacokinetics and pharmacodynamics of ricolinostat in this combination and the preliminary anti-tumour activity of this treatment. The trial is closed to accrual and is registered at ClinicalTrials.gov, number NCT01583283. FINDINGS: Between July 12, 2012, and Aug 20, 2015, we enrolled 38 patients. We observed two dose-limiting toxicities with ricolinostat 160 mg twice daily: one (2%) grade 3 syncope and one (2%) grade 3 myalgia event in different cohorts. A maximum tolerated dose was not reached. We chose ricolinostat 160 mg once daily on days 1-21 of a 28 day cycle as the recommended dose for future phase 2 studies in combination with lenalidomide 25 mg and dexamethasone 40 mg. The most common adverse events were fatigue (grade 1-2 in 14 [37%] patients; grade 3 in seven [18%]) and diarrhoea (grade 1-2 in 15 [39%] patients; grade 3 in two [5%]). Our pharmacodynamic studies showed that at clinically relevant doses, ricolinostat selectively inhibits HDAC6 while retaining a low and tolerable level of class I HDAC inhibition. The pharmacokinetics of ricolinostat and lenalidomide were not affected by co-administration. In a preliminary assessment of antitumour activity, 21 (55% [95% CI 38-71]) of 38 patients had an overall response. INTERPRETATION: The findings from this study provide preliminary evidence that ricolinostat is a safe and well tolerated selective HDAC6 inhibitor, which might partner well with lenalidomide and dexamethasone to enhance their efficacy in relapsed or refractory multiple myeloma. FUNDING: Acetylon Pharmaceuticals.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Aged , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Female , Histone Deacetylase Inhibitors/administration & dosage , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/adverse effects , Lenalidomide , Male , Middle Aged , Multiple Myeloma/mortality , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Thalidomide/administration & dosage , Thalidomide/adverse effects , Thalidomide/analogs & derivatives
10.
PLoS One ; 11(4): e0153767, 2016.
Article in English | MEDLINE | ID: mdl-27073918

ABSTRACT

Therapeutic intervention aimed at reactivation of fetal hemoglobin protein (HbF) is a promising approach for ameliorating sickle cell disease (SCD) and ß-thalassemia. Previous studies showed genetic knockdown of histone deacetylase (HDAC) 1 or 2 is sufficient to induce HbF. Here we show that ACY-957, a selective chemical inhibitor of HDAC1 and 2 (HDAC1/2), elicits a dose and time dependent induction of γ-globin mRNA (HBG) and HbF in cultured primary cells derived from healthy individuals and sickle cell patients. Gene expression profiling of erythroid progenitors treated with ACY-957 identified global changes in gene expression that were significantly enriched in genes previously shown to be affected by HDAC1 or 2 knockdown. These genes included GATA2, which was induced greater than 3-fold. Lentiviral overexpression of GATA2 in primary erythroid progenitors increased HBG, and reduced adult ß-globin mRNA (HBB). Furthermore, knockdown of GATA2 attenuated HBG induction by ACY-957. Chromatin immunoprecipitation and sequencing (ChIP-Seq) of primary erythroid progenitors demonstrated that HDAC1 and 2 occupancy was highly correlated throughout the GATA2 locus and that HDAC1/2 inhibition led to elevated histone acetylation at well-known GATA2 autoregulatory regions. The GATA2 protein itself also showed increased binding at these regions in response to ACY-957 treatment. These data show that chemical inhibition of HDAC1/2 induces HBG and suggest that this effect is mediated, at least in part, by histone acetylation-induced activation of the GATA2 gene.


Subject(s)
Anemia, Sickle Cell/metabolism , Erythroid Cells/drug effects , Fetal Hemoglobin/metabolism , GATA2 Transcription Factor/metabolism , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 2/antagonists & inhibitors , Anemia, Sickle Cell/genetics , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Erythroid Cells/metabolism , GATA2 Transcription Factor/genetics , Histone Deacetylase Inhibitors/pharmacology , Humans , beta-Globins/genetics , beta-Globins/metabolism
11.
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
12.
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
14.
Blood ; 119(11): 2579-89, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22262760

ABSTRACT

Histone deacetylase (HDAC) enzymatic activity has been linked to the transcription of DNA in cancers including multiple myeloma (MM). Therefore, HDAC inhibitors used alone and in combination are being actively studied as novel therapies in MM. In the present study, we investigated the preclinical activity of ACY-1215, an HDAC6-selective inhibitor, alone and in combination with bortezomib in MM. Low doses of ACY-1215 combined with bortezomib triggered synergistic anti-MM activity, resulting in protracted endoplasmic reticulum stress and apoptosis via activation of caspase-3, caspase-8, and caspase-9 and poly (ADP) ribosome polymerase. In vivo, the anti-MM activity of ACY-1215 in combination with bortezomib was confirmed using 2 different xenograft SCID mouse models: human MM injected subcutaneously (the plasmacytoma model) and luciferase-expressing human MM injected intravenously (the disseminated MM model). Tumor growth was significantly delayed and overall survival was significantly prolonged in animals treated with the combination therapy. Pharmacokinetic data showed peak plasma levels of ACY-1215 at 4 hours after treatment coincident with an increase in acetylated α-tubulin, a marker of HDAC6 inhibition, by immunohistochemistry and Western blot analysis. These studies provide preclinical rationale for acetylated α-tubulin use as a pharmacodynamic biomarker in future clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/pharmacokinetics , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Hydroxamic Acids/pharmacokinetics , Plasmacytoma/drug therapy , Pyrazines/pharmacology , Pyrimidines/pharmacology , Pyrimidines/pharmacokinetics , Animals , Antineoplastic Combined Chemotherapy Protocols , Apoptosis/drug effects , Blotting, Western , Bortezomib , Cell Proliferation/drug effects , Drug Synergism , Endoplasmic Reticulum Stress/drug effects , Fluorescent Antibody Technique , Histone Deacetylase 6 , Histone Deacetylases/genetics , Humans , Immunoenzyme Techniques , Male , Mice , Mice, SCID , Plasmacytoma/metabolism , Plasmacytoma/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Psychopharmacology (Berl) ; 220(4): 687-96, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21989804

ABSTRACT

RATIONALE: Accumulating evidence indicates that schizophrenia and autism spectrum disorder patients are marked by cognitive deficits in working memory and strategy switching. There is accumulating evidence that 5-hydroxytryptamine (5-HT)(6) receptors may serve as a useful target to improve cognitive functioning. OBJECTIVES: In the present experiments, the novel 5-HT(6) antagonist, PRX-07034, was examined for its selectivity of the 5-HT(6) receptor, as well as its effect on delayed spontaneous alternation and strategy switching. METHODS: The binding affinity of PRX-07034 to the 5-HT(6) receptor, other 5-HT receptors, as well as other G-protein coupled receptors, ion channels, and transporters was evaluated. Cyclic AMP production was measured from transfected HEK-293 cells. In separate behavioral experiments, rats received different doses of PRX-07034 (0.1, 1, or 3 mg/kg, i.p.) 30 min prior to delayed spontaneous alternation testing or prior to the acquisition and switch phases in a place-response switch test. RESULTS: The results indicated that PRX-07034 is both a potent (Ki = 4-8 nM) and highly selective 5-HT(6) receptor antagonist (≥100-fold selectivity for the 5-HT(6) receptor compared to 68 other GPCRs, ion channels, and transporters, except D(3) (Ki = 71 nM) and 5-HT(1B) (Ki = 260 nM) receptors. For cyclic AMP quantification, PRX-07034 demonstrated antagonist activity (IC(50) = 19 nM) without an effect on basal levels and did not show any agonist activity up to 10 µM. PRX-07034 at 1 and 3 mg/kg (but not 0.1 mg/kg) significantly enhanced delayed spontaneous alternation. The drug at 1 and 3 mg/kg also enhanced switching between a place and response strategy, but did not affect initial learning of either a place or response discrimination. CONCLUSIONS: These findings demonstrate that PRX-07034 is a selective 5-HT(6) receptor antagonist that may represent a novel treatment for enhancing working memory and cognitive flexibility.


Subject(s)
Cognition/drug effects , Memory, Short-Term/drug effects , Receptors, Serotonin/metabolism , Serotonin Antagonists/pharmacology , Animals , Cognition/physiology , HEK293 Cells , Humans , Male , Memory, Short-Term/physiology , Protein Binding/drug effects , Random Allocation , Rats , Rats, Long-Evans , Reaction Time/drug effects , Reaction Time/physiology
16.
Biotechnol Prog ; 21(1): 300-8, 2005.
Article in English | MEDLINE | ID: mdl-15903269

ABSTRACT

A facile and cost-effective process for screening synthetic libraries for an affinity ligand is described. A high throughput 96-well plate filtration method was designed to screen both discrete compounds and mixtures of compounds attached to a solid support. Human serum albumin (HSA) was used as a target protein to demonstrate the proof of concept. Detection and quantitation by fluorescence was accomplished with the use of fluorescamine to conjugate the protein in the filtrate. It is found that mixtures demonstrating low average binding reflect an overall lower hit rate of the components, whereas deconvolution of mixtures with high protein binding consistently provides a high hit rate. This differs from many of the previous experiences screening solid-phase mixtures in which high false positive rates are noted to occur. A total of 100K compounds were tested: 25K as discrete samples and 75K as mixtures. An overall hit rate of 8% was observed. Secondary screening of compounds measured specificity, recovery, and dynamic binding capacity. The effectiveness of the method is illustrated using an affinity column made with a representative lead compound. A similar purity was achieved in a single-step purification of HSA from serum as compared to that obtained by two steps of ion-exchange chromatography. The process for primary screening of a large number of compounds is simple, inexpensive, and applicable to any soluble target protein of known or unknown function from crude mixtures and may have additional utility as a generic chemical affinity tool for the functional characterization of novel proteins emerging from proteomics work.


Subject(s)
Chromatography, Affinity , Combinatorial Chemistry Techniques/methods , Serum Albumin/chemistry , Combinatorial Chemistry Techniques/economics , Humans , Ligands , Protein Binding/physiology , Sensitivity and Specificity
17.
J Biomol Screen ; 8(5): 544-54, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14567781

ABSTRACT

A simple and flexible setup for conducting drug metabolism studies is described in this report. A heating block was designed for the Multimek liquid handler platform for incubation of multiple samples at 37 degrees C in a 96-well format. This setup enables the rapid performance of drug metabolism experiments on a large number of samples. In this report, the authors present the validation of the system by 1) showing reproducible and consistent determination of the in vitro half-life of midazolam in every well across the entire plate and 2) determination of metabolic parameter values of midazolam, testosterone, diclofenac, warfarin, and dextromethorphan and inhibition parameter values of quinidine and ketoconazole, all comparable to literature values. In addition, the authors demonstrate the application of the setup to determining the metabolic stability of a set of proprietary compounds, the inhibition of activity of cytochrome P450 (CYP) enzymes, and the conduct of a single combination experiment that can simultaneously determine the metabolic stability and CYP inhibition activity. Overall, the system represents a simple, high-throughput and useful tool for drug metabolism screening in drug discovery.


Subject(s)
Enzyme Inhibitors/pharmacology , Enzymes/metabolism , Molecular Biology/methods , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Chromatography, Liquid/methods , Cytochrome P-450 CYP2C9 , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP2D6 Inhibitors , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Dextromethorphan/metabolism , Dextromethorphan/pharmacology , Diclofenac/metabolism , Diclofenac/pharmacology , Drug Evaluation, Preclinical/instrumentation , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/metabolism , Enzymes/drug effects , Half-Life , Humans , Kinetics , Mass Spectrometry/methods , Midazolam/metabolism , Midazolam/pharmacology , Molecular Biology/instrumentation , Sensitivity and Specificity , Testosterone/metabolism , Testosterone/pharmacology , Warfarin/metabolism , Warfarin/pharmacology
18.
Kidney Int ; 61(1): 51-60, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11786084

ABSTRACT

BACKGROUND: Proximal tubule epithelial cells (PTEC) play a central role in the response of the kidney to insult by virtue of their production of chemokines and cytokines that signal an inflammatory response. Bone morphogenic protein-7 (BMP-7/OP-1), a member of the transforming growth factor-beta (TGF-beta) superfamily, has previously been demonstrated to reduce macrophage infiltration and tissue damage in animal models of acute and chronic renal failure. The present study was designed to define the molecular mechanism of BMP-7 action in human PTEC. METHODS: Expression of BMP-7 in the adult mouse kidney was determined indirectly through X-gal staining of heterozygous BMP-7/lacZ mice in combination with cell-type specific markers. Primary human PTEC were cultured in the presence of the pro-inflammatory cytokine, tumor necrosis factor-alpha (TNF-alpha), with and without BMP-7. RNA isolated from these two populations was then used to identify differentially regulated genes via gene-array analysis. Modulation of potential target genes was subsequently confirmed through ELISA and/or quantitative PCR. RESULTS: Expression from the BMP-7/lacZ transgene was detected in the collecting duct, thick ascending limb, distal convoluted tubule, and podocytes within glomeruli. No expression was detected within PTEC; however, these cells were found to express mRNA for BMP receptors including, ActR-I, BMPR-IA, ActR-II, ActR-IIB, and BMPR-II. BMP-7 significantly reduced TNF-alpha stimulated increases in mRNA for the pro-inflammatory genes, interleukin-6 (IL-6) and interleukin-1beta (IL-1beta), and the chemoattractants monocyte chemotactic protein-1 (MCP-1) and interleukin-8 (IL-8) in primary human PTEC. In addition, BMP-7 also reduced the expression of mRNA for endothelin-2 (ET-2), a vasoconstrictor, and increased the expression of mRNA for heme oxygenase-1 (HO-1), a vasodilator, although the latter was not statistically significant. In experiments designed to examine MCP-1 and IL-6 protein levels in response to additional TGF-beta superfamily members, TGF-beta1 was unable to mimic the effects of BMP-7 in reducing IL-6 production. However, the closely related BMP-6 exhibited similar properties to those of BMP-7. Each of the factors reduced MCP-1 expression. CONCLUSIONS: BMP-7 represses the basal and TNF-alpha-stimulated expression of the pro-inflammatory cytokines IL-6 and IL-1beta, the chemokines MCP-1 and IL-8, and the vasoconstrictor ET-2 in PTEC. This data are consistent with the in vivo observations that BMP-7 administration in a model of chronic and acute renal failure results in a reduction in the infiltration of macrophages in the renal interstitium. Taken together, these observations suggest that BMP-7 may be a novel therapeutic agent for kidney disorders involving inflammation and ischemic damage of PTEC.


Subject(s)
Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Chemokines/genetics , Kidney Tubules, Proximal/physiology , Neoplasm Proteins , Receptors, Growth Factor , Renal Circulation/physiology , Animals , Bone Morphogenetic Protein 7 , Bone Morphogenetic Protein Receptors , Cells, Cultured , Chemokine CCL2/genetics , DNA Primers , DNA-Binding Proteins/genetics , Endothelin-2/genetics , Gene Expression/physiology , Growth Differentiation Factor 5 , Growth Substances/genetics , Humans , Inhibitor of Differentiation Proteins , Interleukin-1/genetics , Interleukin-6/genetics , Interleukin-8/genetics , Kidney Tubules, Proximal/cytology , Lac Operon , Mice , Mice, Transgenic , Neutrophils/immunology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/analysis , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Transcription Factors/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1 , Vasoconstriction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL