Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
Ophthalmol Ther ; 11(2): 869-879, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35235173

ABSTRACT

INTRODUCTION: Pilocarpine hydrochloride (pilo) ophthalmic solution has traditionally been used for the treatment of glaucoma, with opportunities to improve the tolerability profile experienced by patients. Pilocarpine hydrochloride ophthalmic solution 1.25% (Vuity™, Allergan, an AbbVie company) was approved in late 2021 for the treatment of adults with presbyopia. This publication describes the properties of the optimized, proprietary vehicle of this new ophthalmic solution developed with the aim of improving tolerability upon instillation. METHODS: An in vitro method determined the time required for the pH of pilo 1.25% in the proprietary vehicle (Optimized Formulation) and a commercially available 1% pilo ophthalmic solution (Generic Formulation) to equilibrate with the pH of simulated tear fluid (STF). In a pilot study, five of the six screened participants received one drop of the Optimized Formulation in one eye and Generic Formulation in the other. Ocular discomfort and vision blur were evaluated for each eye just prior to and at multiple times after drop instillation using visual analog scales (VAS), and adverse events were assessed. RESULTS: The in vitro method showed that the Optimized Formulation achieved faster pH equilibration than the Generic Formulation. The pilot study revealed that the Optimized Formulation demonstrated less ocular discomfort, vision blur, and adverse events compared to the Generic. CONCLUSION: The in vitro and pilot study of the Optimized Formulation indicated that it rapidly equilibrates to the physiologic pH of the tear film, providing greater comfort and tolerability while also minimizing vision blur. Overall, the proprietary vehicle is expected to improve comfort, result in less vision blur, and provide a well-tolerated alternative method to deliver pilo for the treatment of presbyopia when compared to what is commercially available.

2.
PLoS One ; 8(1): e54346, 2013.
Article in English | MEDLINE | ID: mdl-23349858

ABSTRACT

BACKGROUND: Pancreatic cancer is one of the deadliest cancers with a 5-year survival rate of 6%. Therapeutic options are very limited and there is an unmet medical need for safe and efficacious treatments. Cancer cell metabolism and mitochondria provide unexplored targets for this disease. We recently identified a novel class of triphenylphosphonium salts, TP compounds, with broad- spectrum anticancer properties. We examined the ability of our prototypical compound TP421- chosen for its fluorescent properties - to inhibit the growth of pancreatic cancer cells and further investigated the molecular mechanisms by which it exerts its anticancer effects. METHODOLOGY/PRINCIPAL FINDINGS: TP421 exhibited sub-micromolar IC(50) values in all the pancreatic cancer cell lines tested using MTT and colony formation assays. TP421 localized predominantly to mitochondria and induced G(0)/G(1) arrest, ROS accumulation, and activation of several stress-regulated kinases. Caspase and PARP-1 cleavage were observed indicating an apoptotic response while LC3B-II and p62 were accumulated indicating inhibition of autophagy. Furthermore, TP421 induced de-phosphorylation of key signaling molecules involved in FAK mediated adhesion that correlated with inhibition of cell migration. CONCLUSIONS/SIGNIFICANCE: TP421 is a representative compound of a new promising class of mitochondrial-targeted agents useful for pancreatic cancer treatment. Because of their unique mechanism of action and efficacy further development is warranted.


Subject(s)
Apoptosis/drug effects , Coumarins/administration & dosage , Drug Discovery , Mitochondria , Organophosphorus Compounds/administration & dosage , Pancreatic Neoplasms/drug therapy , Autophagy/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Fluorescence , Humans , Inhibitory Concentration 50 , Mitochondria/drug effects , Mitochondria/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation , Signal Transduction/drug effects
3.
ChemMedChem ; 7(10): 1825-39, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22952012

ABSTRACT

Apurinic/apyrimidinic (AP) endonuclease 1 (APE1) is a multifaceted protein with an essential role in the base excision repair (BER) pathway. Its implication in tumor development, progression, and resistance has been confirmed in multiple cancers, making it a viable target for intensive investigation. In this work, we designed and synthesized different classes of small-molecule inhibitors of the catalytic endonuclease function of APE1 that contain a 3-carbamoylbenzoic acid scaffold. Further structural modifications were made with the aim of increasing the activity and cytotoxicity of these inhibitors. Several of our compounds were shown to inhibit the catalytic endonuclease function of APE1 with potencies in the low-micromolar range in vitro, and therefore represent novel classes of APE1 inhibitors worthy of further development.


Subject(s)
Benzoic Acid/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , Drug Design , Methylurea Compounds/chemistry , Benzoic Acid/chemical synthesis , Benzoic Acid/toxicity , Cell Line , Cell Survival/drug effects , DNA Repair/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Deoxyuridine/analogs & derivatives , Deoxyuridine/toxicity , Humans
4.
Curr Mol Pharmacol ; 5(1): 14-35, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22122462

ABSTRACT

APE1 is a multifaceted protein that orchestrates multiple activities in the cell, one of which is the preservation of genomic integrity; a vital process that takes place in the context of the base excision repair (BER) pathway. Studies have implicated APE1 in rendering cancerous cells less vulnerable to the effects of DNA-damaging agents that are commonly used for the treatment of cancer. Furthermore, suppression of APE1 expression in cancer cell lines is accompanied by the potentiation of the activity of cytotoxic agents. As a result, major efforts have been directed towards the identification of small-molecule inhibitors of this DNA-repair enzyme. Herein, we review all patented small-molecule APE1 inhibitors reported prior to 2011. Unfortunately, the potency and selectivity of many of the reported inhibitors were not disclosed by the original authors, and at present it is unclear if APE1 is a bona fide target for many of the purported inhibitors. Moreover, cellular activity and toxicity of many inhibitors remain to be established. Since this is the first comprehensive review of small molecule APE1 inhibitors, we present all compounds reported to inhibit APE1 activity with an IC50 value ≤ 25 µM. Efforts towards a careful validation and optimization of these compounds are warranted. Furthermore, we explore potential allosteric drug-binding sites on the protein as an alternative approach for modulating the activity of this multifunctional protein. In addition, we give an overview of APE2, as well as other APE1 homologues in some disease-causing pathogens. Finally, given the universal importance of DNA repair, as well as the considerable conservation of repair proteins across all living organisms, we propose targeting the AP endonuclease activity of pathogens by the compounds discussed in this review, thereby expanding their therapeutic potential and application.


Subject(s)
DNA Repair/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Small Molecule Libraries/chemistry , Catalytic Domain , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Endonucleases , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Humans , Multifunctional Enzymes , Neoplasms/drug therapy , Neoplasms/enzymology , Small Molecule Libraries/therapeutic use , Structure-Activity Relationship
5.
Bioorg Med Chem ; 19(16): 4935-52, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21778063

ABSTRACT

HIV-1 integrase (IN) is a validated therapeutic target for antiviral drug design. However, the emergence of viral strains resistant to clinically studied IN inhibitors demands the discovery of novel inhibitors that are structurally as well mechanistically different. Herein, we describe the design and discovery of novel IN inhibitors targeting the catalytic domain as well as its interaction with LEDGF/p75, which is essential for the HIV-1 integration as an IN cofactor. By merging the pharmacophores of salicylate and catechol, the 2,3-dihydroxybenzamide (5a) was identified as a new scaffold to inhibit the strand transfer reaction efficiently. Further structural modifications on the 2,3-dihydroxybenzamide scaffold revealed that the heteroaromatic functionality attached on the carboxamide portion and the piperidin-1-ylsulfonyl substituted at the phenyl ring are beneficial for the activity, resulting in a low micromolar IN inhibitor (5p, IC(50)=5 µM) with more than 40-fold selectivity for the strand transfer over the 3'-processing reaction. More significantly, this active scaffold remarkably inhibited the interaction between IN and LEDGF/p75 cofactor. The prototype example, N-(cyclohexylmethyl)-2,3-dihydroxy-5-(piperidin-1-ylsulfonyl) benzamide (5u) inhibited the IN-LEDGF/p75 interaction with an IC(50) value of 8 µM. Using molecular modeling, the mechanism of action was hypothesized to involve the chelation of the divalent metal ions inside the IN active site. Furthermore, the inhibitor of IN-LEDGF/p75 interaction was properly bound to the LEDGF/p75 binding site on IN. This work provides a new and efficient approach to evolve novel HIV-1 IN inhibitors from rational integration and optimization of previously reported inhibitors.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Catalytic Domain/drug effects , Catechols/chemical synthesis , HIV Integrase Inhibitors/chemical synthesis , HIV-1/drug effects , Receptor, Nerve Growth Factor/antagonists & inhibitors , Salicylates/chemical synthesis , Transcription Factors/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/analysis , Adaptor Proteins, Signal Transducing/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Catalytic Domain/genetics , Catechols/chemistry , Cell Line, Tumor , Drug Design , Drug Resistance, Multiple, Viral , Drug Screening Assays, Antitumor , HIV Integrase Inhibitors/chemistry , HIV Integrase Inhibitors/pharmacology , HIV-1/genetics , Humans , Metals/chemistry , Models, Molecular , Molecular Structure , Molecular Targeted Therapy , Receptor, Nerve Growth Factor/analysis , Receptor, Nerve Growth Factor/drug effects , Receptor, Nerve Growth Factor/metabolism , Salicylates/chemistry , Transcription Factors/analysis , Transcription Factors/drug effects , Transcription Factors/metabolism
6.
PLoS One ; 5(10)2010 Oct 04.
Article in English | MEDLINE | ID: mdl-20957228

ABSTRACT

BACKGROUND: Recently, there has been a surge of interest in developing compounds selectively targeting mitochondria for the treatment of neoplasms. The critical role of mitochondria in cellular metabolism and respiration supports this therapeutic rationale. Dysfunction in the processes of energy production and metabolism contributes to attenuation of response to pro-apoptotic stimuli and increased ROS production both of which are implicated in the initiation and progression of most human cancers. METHODOLOGY/PRINCIPAL FINDINGS: A high-throughput MTT-based screen of over 10,000 drug-like small molecules for anti-proliferative activity identified the phosphonium salts TP187, 197 and 421 as having IC50 concentrations in the submicromolar range. TP treatment induced cell cycle arrest independent of p53 status, as determined by analysis of DNA content in propidium iodide stained cells. In a mouse model of human breast cancer, TP-treated mice showed significantly decreased tumor growth compared to vehicle or paclitaxel treated mice. No toxicities or organ damage were observed following TP treatment. Immunohistochemical staining of tissue sections from TP187-treated tumors demonstrated a decrease in cellular proliferation and increased caspase-3 cleavage. The fluorescent properties of analog TP421 were exploited to assess subcellular uptake of TP compounds, demonstrating mitochondrial localization. Following mitochondrial uptake cells exhibited decreased oxygen consumption and concomittant increase in mitochondrial superoxide production. Proteomics analysis of results from a 600 target antibody microarray demonstrated that TP compounds significantly affected signaling pathways relevant to growth and proliferation. CONCLUSIONS/SIGNIFICANCE: Through our continued interest in designing compounds targeting cancer-cell metabolism, the Warburg effect, and mitochondria we recently discovered a series of novel, small-molecule compounds containing a triphenylphosphine moiety that show remarkable activity in a panel of cancer cell lines as well as in a mouse model of human breast cancer. The mechanism of action includes mitochondrial localization causing decreased oxygen consumption, increased superoxide production and attenuated growth factor signaling.


Subject(s)
Antineoplastic Agents/pharmacology , Organophosphorus Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspase 3/metabolism , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Drug Screening Assays, Antitumor , Female , Humans , Immunohistochemistry , Mice , Mitochondria/metabolism , Organophosphorus Compounds/pharmacokinetics , Proteomics , Superoxides/metabolism , Transplantation, Heterologous
7.
J Pharm Sci ; 98(10): 3562-74, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19199298

ABSTRACT

The objective of this work was to evaluate the solution stability of the EC1 domain of E-cadherin under various conditions. The EC1 domain was incubated at various temperatures (4, 37, and 70 degrees C) and pH values (3.0, 7.0, and 9.0). At pH 9.0 and 37 or 70 degrees C, a significant loss of EC1 was observed due to precipitation and a hydrolysis reaction. The degradation was suppressed upon addition of dithiothreitol (DTT), suggesting that the formation of EC1 dimer facilitated the EC1 degradation. At 4 degrees C and various pH values, the EC1 secondary and tertiary showed changes upon incubation up to 28 days, and DTT prevented any structural changes upon 28 days of incubation. Molecular dynamics simulations indicated that the dimer of EC1 has higher mobility than does the monomer; this higher mobility of the EC1 dimer may contribute to instability of the EC1 domain.


Subject(s)
Cadherins/chemistry , Amino Acid Sequence , Chromatography, High Pressure Liquid , Circular Dichroism , Computer Simulation , Dimerization , Dithiothreitol/analysis , Drug Stability , Escherichia coli/genetics , Humans , Hydrogen-Ion Concentration , Mass Spectrometry , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Spectrometry, Fluorescence , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL