Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
ACS Appl Mater Interfaces ; 15(5): 6312-6325, 2023 Feb 08.
Article in English | MEDLINE | ID: mdl-36701696

ABSTRACT

Precision nanomedicine can be employed as an alternative to chemo- or radiotherapy to overcome challenges associated with the often narrow therapeutic window of traditional treatment approaches, while safely inducing effective, targeted antitumor responses. Herein, we report the formulation of a therapeutic nanocomposite comprising a hyaluronic acid (HA)-coated gold nanoframework (AuNF) delivery system and encapsulated IT848, a small molecule with potent antilymphoma and -myeloma properties that targets the transcriptional activity of nuclear factor kappa B (NF-κB). The porous AuNFs fabricated via a liposome-templated approach were loaded with IT848 and surface-functionalized with HA to formulate the nanotherapeutics that were able to efficiently deliver the payload with high specificity to myeloma and lymphoma cell lines in vitro. In vivo studies characterized biodistribution, pharmacokinetics, and safety of HA-AuNFs, and we demonstrated superior efficacy of HA-AuNF-formulated IT848 vs free IT848 in lymphoma mouse models. Both in vitro and in vivo results affirm that the AuNF system can be adopted for targeted cancer therapy, improving the drug safety profile, and enhancing its efficacy with minimal dosing. HA-AuNF-formulated IT848 therefore has strong potential for clinical translation.


Subject(s)
Lymphoma , Multiple Myeloma , Nanoparticles , Mice , Animals , Tissue Distribution , Gold , Drug Delivery Systems/methods , Lymphoma/drug therapy , Hyaluronic Acid/pharmacology , Hyaluronan Receptors/metabolism
2.
Mol Cancer Ther ; 21(12): 1798-1809, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36190955

ABSTRACT

Multiple myeloma is a plasma cell malignancy that is still largely incurable, despite considerable progress in recent years. NF-κB is a well-established therapeutic target in multiple myeloma, but none of the currently available treatment options offer direct, specific pharmacologic targeting of NF-κB transcriptional activity. Thus, we designed a novel direct NF-κB inhibitor (IT848) as a drug candidate with strong potential for clinical translation and conducted comprehensive in vitro and in vivo mechanistic studies in multiple myeloma cell lines, primary multiple myeloma cells, xenograft models, and immunocompetent mouse models of multiple myeloma. Here, we show that IT848 inhibits NF-κB activity through inhibition of DNA binding of all five NF-κB subunits. IT848 treatment of multiple myeloma cell lines and patient samples inhibited proliferation and induced caspase-dependent and independent apoptosis. In addition to direct NF-κB inhibitory effects, IT848 treatment altered the redox homeostasis of multiple myeloma cells through depletion of the reduced glutathione pool, selectively inducing oxidative stress in multiple myeloma but not in healthy cells. Multiple myeloma xenograft studies confirmed the efficacy of IT848 as single agent and in combination with bortezomib. Furthermore, IT848 significantly improved survival when combined with programmed death protein 1 inhibition, and correlative immune studies revealed that this clinical benefit was associated with suppression of regulatory T-cell infiltration of the bone marrow microenvironment. In conclusion, IT848 is a potent direct NF-κB inhibitor and inducer of oxidative stress specifically in tumor cells, displaying significant activity against multiple myeloma cells in vitro and in vivo, both as monotherapy as well as in combination with bortezomib or immune checkpoint blockade.


Subject(s)
Multiple Myeloma , Mice , Animals , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , NF-kappa B/metabolism , Bortezomib/pharmacology , Bortezomib/therapeutic use , Tumor Microenvironment , Apoptosis , I-kappa B Proteins/metabolism , Oxidation-Reduction , DNA/metabolism , Cell Line, Tumor
3.
Haematologica ; 102(11): 1878-1889, 2017 11.
Article in English | MEDLINE | ID: mdl-28860341

ABSTRACT

IT-901 is a novel and selective NF-κB inhibitor with promising activity in pre-clinical models. Here we show that treatment of chronic lymphocytic leukemia cells (CLL) with IT-901 effectively interrupts NF-κB transcriptional activity. CLL cells exposed to the drug display elevated mitochondrial reactive oxygen species, which damage mitochondria, limit oxidative phosphorylation and ATP production, and activate intrinsic apoptosis. Inhibition of NF-κB signaling in stromal and myeloid cells, both tumor-supportive elements, fails to induce apoptosis, but impairs NF-κB-driven expression of molecules involved in cell-cell contacts and immune responses, essential elements in creating a pro-leukemic niche. The consequence is that accessory cells do not protect CLL cells from IT-901-induced apoptosis. In this context, IT-901 shows synergistic activity with ibrutinib, arguing in favor of combination strategies. IT-901 is also effective in primary cells from patients with Richter syndrome (RS). Its anti-tumor properties are confirmed in xenograft models of CLL and in RS patient-derived xenografts, with documented NF-κB inhibition and significant reduction of tumor burden. Together, these results provide pre-clinical proof of principle for IT-901 as a potential new drug in CLL and RS.


Subject(s)
Antineoplastic Agents/pharmacology , Energy Metabolism/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , NF-kappa B/antagonists & inhibitors , Adenine/analogs & derivatives , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival , Disease Models, Animal , Drug Synergism , Gene Silencing , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Targeted Therapy , NF-kappa B/genetics , NF-kappa B/metabolism , Piperidines , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
4.
Cancer Res ; 76(2): 377-89, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26744524

ABSTRACT

NF-κB plays a variety of roles in oncogenesis and immunity that may be beneficial for therapeutic targeting, but strategies to selectively inhibit NF-κB to exert antitumor activity have been elusive. Here, we describe IT-901, a bioactive naphthalenethiobarbiturate derivative that potently inhibits the NF-κB subunit c-Rel. IT-901 suppressed graft-versus-host disease while preserving graft-versus-lymphoma activity during allogeneic transplantation. Further preclinical assessment of IT-901 for the treatment of human B-cell lymphoma revealed antitumor properties in vitro and in vivo without restriction to NF-κB-dependent lymphoma. This nondiscriminatory, antilymphoma effect was attributed to modulation of the redox homeostasis in lymphoma cells resulting in oxidative stress. Moreover, NF-κB inhibition by IT-901 resulted in reduced stimulation of the oxidative stress response gene heme oxygenase-1, and we demonstrated that NF-κB inhibition exacerbated oxidative stress induction to inhibit growth of lymphoma cells. Notably, IT-901 did not elicit increased levels of reactive oxygen species in normal leukocytes, illustrating its cancer selective properties. Taken together, our results provide mechanistic insight and preclinical proof of concept for IT-901 as a novel therapeutic agent to treat human lymphoid tumors and ameliorate graft-versus-host disease.


Subject(s)
NF-kappa B/metabolism , Proto-Oncogene Proteins c-rel/antagonists & inhibitors , Animals , Female , Hematologic Neoplasms , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Oxidative Stress , Proto-Oncogene Proteins c-rel/genetics , Proto-Oncogene Proteins c-rel/metabolism , Reactive Oxygen Species , Signal Transduction
5.
Cancer Discov ; 4(5): 578-91, 2014 May.
Article in English | MEDLINE | ID: mdl-24550032

ABSTRACT

Preventing unfavorable GVHD without inducing broad suppression of the immune system presents a major challenge of allogeneic hematopoietic stem cell transplantation (allo-HSCT). We developed a novel strategy to ameliorate GVHD while preserving graft-versus-tumor (GVT) activity by small molecule-based inhibition of the NF-κB family member c-Rel. Underlying mechanisms included reduced alloactivation, defective gut homing, and impaired negative feedback on interleukin (IL)-2 production, resulting in optimal IL-2 levels, which, in the absence of competition by effector T cells, translated into expansion of regulatory T cells. c-Rel activity was dispensable for antigen-specific T-cell receptor (TCR) activation, allowing c-Rel-deficient T cells to display normal GVT activity. In addition, inhibition of c-Rel activity reduced alloactivation without compromising antigen-specific cytotoxicity of human T cells. Finally, we were able to demonstrate the feasibility and efficacy of systemic c-Rel inhibitor administration. Our findings validate c-Rel as a promising target for immunomodulatory therapy and demonstrate the feasibility and efficacy of pharmaceutical inhibition of c-Rel activity.


Subject(s)
Graft vs Host Disease/prevention & control , Lymphocyte Activation/drug effects , Proto-Oncogene Proteins c-rel/antagonists & inhibitors , Small Molecule Libraries/pharmacology , T-Lymphocytes/drug effects , Animals , Female , Gene Expression Regulation , Graft vs Host Disease/immunology , Graft vs Tumor Effect/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Proto-Oncogene Proteins c-rel/genetics , Proto-Oncogene Proteins c-rel/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Transplantation, Homologous
6.
Cancer Res ; 72(6): 1494-503, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22266222

ABSTRACT

Continued reliance on the androgen receptor (AR) is now understood as a core mechanism in castration-resistant prostate cancer (CRPC), the most advanced form of this disease. While established and novel AR pathway-targeting agents display clinical efficacy in metastatic CRPC, dose-limiting side effects remain problematic for all current agents. In this study, we report the discovery and development of ARN-509, a competitive AR inhibitor that is fully antagonistic to AR overexpression, a common and important feature of CRPC. ARN-509 was optimized for inhibition of AR transcriptional activity and prostate cancer cell proliferation, pharmacokinetics, and in vivo efficacy. In contrast to bicalutamide, ARN-509 lacked significant agonist activity in preclinical models of CRPC. Moreover, ARN-509 lacked inducing activity for AR nuclear localization or DNA binding. In a clinically valid murine xenograft model of human CRPC, ARN-509 showed greater efficacy than MDV3100. Maximal therapeutic response in this model was achieved at 30 mg/kg/d of ARN-509, whereas the same response required 100 mg/kg/d of MDV3100 and higher steady-state plasma concentrations. Thus, ARN-509 exhibits characteristics predicting a higher therapeutic index with a greater potential to reach maximally efficacious doses in man than current AR antagonists. Our findings offer preclinical proof of principle for ARN-509 as a promising therapeutic in both castration-sensitive and castration-resistant forms of prostate cancer.


Subject(s)
Androgen Antagonists/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Prostatic Neoplasms/drug therapy , Thiohydantoins/therapeutic use , Androgen Antagonists/pharmacokinetics , Anilides/pharmacokinetics , Anilides/therapeutic use , Animals , Antineoplastic Agents, Hormonal/blood , Antineoplastic Agents, Hormonal/pharmacokinetics , Benzamides , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Nitriles/pharmacokinetics , Nitriles/therapeutic use , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/blood , Phenylthiohydantoin/pharmacokinetics , Phenylthiohydantoin/therapeutic use , Rats , Receptors, Androgen/drug effects , Thiohydantoins/blood , Thiohydantoins/chemical synthesis , Thiohydantoins/pharmacokinetics , Tosyl Compounds/pharmacokinetics , Tosyl Compounds/therapeutic use , Xenograft Model Antitumor Assays
9.
Science ; 324(5928): 787-90, 2009 May 08.
Article in English | MEDLINE | ID: mdl-19359544

ABSTRACT

Metastatic prostate cancer is treated with drugs that antagonize androgen action, but most patients progress to a more aggressive form of the disease called castration-resistant prostate cancer, driven by elevated expression of the androgen receptor. Here we characterize the diarylthiohydantoins RD162 and MDV3100, two compounds optimized from a screen for nonsteroidal antiandrogens that retain activity in the setting of increased androgen receptor expression. Both compounds bind to the androgen receptor with greater relative affinity than the clinically used antiandrogen bicalutamide, reduce the efficiency of its nuclear translocation, and impair both DNA binding to androgen response elements and recruitment of coactivators. RD162 and MDV3100 are orally available and induce tumor regression in mouse models of castration-resistant human prostate cancer. Of the first 30 patients treated with MDV3100 in a Phase I/II clinical trial, 13 of 30 (43%) showed sustained declines (by >50%) in serum concentrations of prostate-specific antigen, a biomarker of prostate cancer. These compounds thus appear to be promising candidates for treatment of advanced prostate cancer.


Subject(s)
Androgen Antagonists/therapeutic use , Antineoplastic Agents/therapeutic use , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms/drug therapy , Androgen Antagonists/metabolism , Androgen Antagonists/pharmacokinetics , Androgen Antagonists/pharmacology , Anilides/metabolism , Anilides/pharmacology , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Benzamides , Biological Availability , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/drug effects , DNA/metabolism , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Nitriles/metabolism , Nitriles/pharmacology , Phenylthiohydantoin/metabolism , Phenylthiohydantoin/pharmacokinetics , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms/pathology , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Tosyl Compounds/metabolism , Tosyl Compounds/pharmacology , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays
10.
Future Med Chem ; 1(9): 1683-707, 2009 Dec.
Article in English | MEDLINE | ID: mdl-21425986

ABSTRACT

The Rel/NF-κB transcription factors have emerged as novel therapeutic targets for a variety of human diseases and pathological conditions, including inflammation, autoimmune diseases, cancer, ischemic injury, osteoporosis, transplant rejection and neurodegeneration. Several US FDA-approved drugs may, in part, attribute their therapeutic effects to the inhibition of the Rel/NF-κB pathway. Strategies for blocking the Rel/NF-κB signaling pathway have inspired the pharmaceutical industry to develop inhibitors for I-κB kinase, however, this article focuses instead on identifying natural compounds that directly target and inhibit DNA binding and transcription activity of Rel/NF-κB. These include compounds containing a quinone core, an α,ß unsaturated carbonyl and a benzene diamine. By investigating the mechanisms of action of existing natural inhibitors, novel strategies and synthetic approaches can be devised that will facilitate the development of novel and selective Rel/NF-κB inhibitors with better safety profiles.


Subject(s)
NF-kappa B/antagonists & inhibitors , Transcription Factor RelA/antagonists & inhibitors , Animals , Autoimmune Diseases/drug therapy , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/metabolism , Ketones/chemistry , Ketones/therapeutic use , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Quinones/chemistry , Quinones/therapeutic use , Signal Transduction , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
11.
Am J Physiol Gastrointest Liver Physiol ; 289(4): G753-9, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15905414

ABSTRACT

The cystic fibrosis (CF) transmembrane conductance regulator (CFTR) plays a crucial role in mediating duodenal bicarbonate (HCO(3)(-)) secretion (DBS). Although impaired DBS is observed in CF mutant mice and in CF patients, which would predict increased ulcer susceptibility, duodenal injury is rarely observed in CF patients and is reduced in CF mutant mice. To explain this apparent paradox, we hypothesized that CFTR dysfunction increases cellular [HCO(3)(-)] and buffering power. To further test this hypothesis, we examined the effect of a novel, potent, and highly selective CFTR inhibitor, CFTR(inh)-172, on DBS and duodenal ulceration in rats. DBS was measured in situ using a standard loop perfusion model with a pH stat under isoflurane anesthesia. Duodenal ulcers were induced in rats by cysteamine with or without CFTR(inh)-172 pretreatment 1 h before cysteamine. Superfusion of CFTR(inh)-172 (0.1-10 microM) over the duodenal mucosa had no effect on basal DBS but at 10 microM inhibited acid-induced DBS, suggesting that its effect was limited to CFTR activation. Acid-induced DBS was abolished at 1 and 3 h and was reduced 24 h after treatment with CFTR(inh)-172, although basal DBS was increased at 24 h. CFTR(inh)-172 treatment had no effect on gastric acid or HCO(3)(-) secretion. Duodenal ulcers were observed 24 h after cysteamine treatment but were reduced in CFTR(inh)-172-pretreated rats. CFTR(inh)-172 acutely produces CFTR dysfunction in rodents for up to 24 h. CFTR inhibition reduces acid-induced DBS but also prevents duodenal ulcer formation, supporting our hypothesis that intracellular HCO(3)(-) may be an important protective mechanism for duodenal epithelial cells.


Subject(s)
Benzoates/pharmacology , Bicarbonates/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Duodenal Ulcer/prevention & control , Thiazoles/pharmacology , Animals , Chromatography, High Pressure Liquid , Cystamine/toxicity , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Duodenal Ulcer/chemically induced , Duodenum/drug effects , Duodenum/metabolism , Gastric Acid/metabolism , Male , Rats , Rats, Sprague-Dawley , Sulfhydryl Reagents/toxicity , Thiazolidines
SELECTION OF CITATIONS
SEARCH DETAIL
...