Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Invest Ophthalmol Vis Sci ; 58(12): 5142-5150, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28986592

ABSTRACT

Purpose: Intravitreal injection of antiangiogenic agents is becoming a standard treatment for neovascular retinal diseases. Sustained release of therapeutics by injecting colloidal carriers is a promising approach to reduce the injection frequency, which reduces treatment burdens and the risk of complications on patients. Such sustained release often requires carriers to have micrometer-scale dimension that, however, can potentially promote glaucoma and inflammation. Small, polycationic particles can be immobilized in vitreous through multiple cooperative ionic interactions with hyaluronic acid of the vitreous interior, but such particles are generally toxic. Here, we synthesized and examined a biocompatible dextran-based nanocarrier (<50 nm in diameter) conjugated with cationic peptides containing L-arginine with minimal toxicity, aiming to provide sustained release of therapeutic drugs in vitreous. Methods: We synthesized the nanocarriers with condensed cholesteryl dextran (CDEX) as core material. Cationic peptides containing 1 to 4 arginine groups, along with fluorescence tags, were conjugated to the CDEX surface. We monitored the carrier diffusion rate ex vivo and half-lives in vivo in rodent vitreous using fluorescence imaging. We evaluated the toxicity by histological examinations at the second, third, eighth, and thirty-sixth week. Results: The diffusion rate of nanocarriers was inversely related to zeta potential values in freshly isolated vitreous humor. We observed increased half-lives in vivo with increasing zeta potential (up to 240 days). Histological examinations confirmed no adverse effects on ocular morphology and organization. Conclusions: We demonstrated the potential of L-arginine peptide-conjugated nanocarriers toward safe and sustained therapeutic release system for posterior eye diseases.


Subject(s)
Antimicrobial Cationic Peptides/pharmacokinetics , Arginine/chemistry , Cholesterol Esters/chemistry , Dextrans/chemistry , Drug Carriers/pharmacokinetics , Vitreous Body/metabolism , Animals , Antimicrobial Cationic Peptides/chemistry , Biocompatible Materials , Drug Carriers/chemistry , Fluorescein Angiography , Half-Life , Intravitreal Injections , Nanoparticles , Rats , Rats, Long-Evans , Rats, Sprague-Dawley
2.
Mol Pharm ; 11(9): 3112-22, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25072100

ABSTRACT

Curcumin (CUR) is a unique natural compound with promising anticancer and anti-inflammatory activities. However, the therapeutic efficacy of curcumin was challenged in clinical trials, mostly due to its low bioavailability, rapid metabolism, and elimination. We designed a nanodrug form of curcumin, which makes it stable and substantially enhances cellular permeability and anticancer activity at standard oral administration. Curcumin was conjugated as an ester to cholesteryl-hyaluronic acid (CHA) nanogel that is capable of targeted delivery to CD44-expressing drug-resistant cancer cells. CHA-CUR nanogels demonstrated excellent solubility and sustained drug release in physiological conditions. It induced apoptosis in cancer cells, suppressing the expression of NF-κB, TNF-α, and COX-2 cellular targets similar to free curcumin. Pharmacokinetic/pharmacodynamic (PK/PD) studies also revealed improved circulation parameters of CHA-CUR at oral, i.p. and i.v. administration routes. CHA-CUR showed targeted tumor accumulation and effective tumor growth inhibition in human pancreatic adenocarcinoma MiaPaCa-2 and aggressive orthotropic murine mammary carcinoma 4T1 animal models. CHA-CUR treatment was well-tolerated and resulted in up to 13-fold tumor suppression, making this nanodrug a potential candidate for cancer prevention and therapeutic treatment.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Curcumin/chemistry , Curcumin/pharmacokinetics , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Polyethyleneimine/chemistry , Polyethyleneimine/pharmacology , Adenoma/drug therapy , Adenoma/metabolism , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Line, Tumor , Curcumin/pharmacology , Cyclooxygenase 2/metabolism , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Drug Delivery Systems/methods , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor/methods , Esters/chemistry , Female , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/chemistry , Mammary Neoplasms, Animal/diet therapy , Mammary Neoplasms, Animal/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/metabolism , Nanogels , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Permeability , Polyethylene Glycols/pharmacokinetics , Polyethyleneimine/pharmacokinetics , Solubility , Tumor Necrosis Factor-alpha/metabolism
3.
Pharm Res ; 31(6): 1605-15, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24452808

ABSTRACT

PURPOSE: Current cancer chemotherapy is gradually shifting to the application of drug combinations that prevent development of drug resistance. Many anticancer drugs have poor solubility and limited oral bioavailability. Using an innovative approach, we developed dual-drug nanoformulations of a polymeric nanogel conjugate with anticancer 5-FU nucleoside analog, floxuridine (FLOX), and the second anticancer drugs, paclitaxel (PCL), or a geldanamycin analog, 17-AAG, for combination therapy. METHODS: PCL or 17-AAG had been encapsulated in the cholesteryl-polyvinyl alcohol-floxuridine nanogel (CPVA-FLOX) by simple solution mixing and sonication. Dual nanodrugs formed particles with diameter 180 nm and either drug content (5-20%) that were stable and could be administered orally. Their cytotoxicity in human and mouse cancer cells was determined by MTT assay, and cellular target inhibition - by Western blot analysis. Tumor growth inhibition was evaluated using an orthotopic mouse mammary 4T1 cancer model. RESULTS: CPVA-FLOX was more potent than free drug in cancer models including drug-resistant ones; while dual nanodrugs demonstrated a significant synergy (CPVA-FLOX/PCL), or showed no significant synergy (CPVA-FLOX/17-AAG) compared to free drugs (PCL or 17-AAG). Dual nanodrug CPVA-FLOX/17-AAG effect on its cellular target (HSP70) was similar to 17-AAG alone. In animal model, however, both dual nanodrugs effectively inhibited tumor growth compared to CPVA-FLOX after oral administration. CONCLUSION: Oral dual-drug nanoformulations of poorly-soluble drugs proved to be a highly efficient combination anticancer therapy in preclinical studies.


Subject(s)
Antineoplastic Agents/administration & dosage , Polymers/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Chemistry, Pharmaceutical , Drug Compounding , Female , HSP70 Heat-Shock Proteins/biosynthesis , Humans , Mice , Mice, Inbred BALB C , Nanoparticles , Solubility
4.
Nanomedicine ; 10(1): 177-85, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23845925

ABSTRACT

Antiviral therapy using nucleoside reverse transcriptase inhibitors (NRTIs) is neurotoxic and has low efficiency in eradication of HIV-1 harbored in central nervous system (CNS). Previously, we reported that active 5'-triphosphates of NRTIs encapsulated in cationic nanogels (nano-NRTIs) suppress HIV-1 activity more efficiently than NRTIs and exhibit reduced mitochondrial toxicity [Vinogradov SV, Poluektova LY, Makarov E, Gerson T, Senanayake MT. Nano-NRTIs: efficient inhibitors of HIV type-1 in macrophages with a reduced mitochondrial toxicity. Antivir Chem Chemother. 2010; 21:1-14. Makarov E, Gerson T, Senanayake T, Poluektova LY, Vinogradov. Efficient suppression of Human Immunodeficiency Virus in Macrophages by Nano-NRTIs. Antiviral Res. 2010; 86(1):A38-9]. Here, we demonstrated low neurotoxicity and excellent antiviral activity of nano-NRTIs decorated with the peptide (AP) binding brain-specific apolipoprotein E receptor. Nano-NRTIs induced lower levels of apoptosis and formation of reactive oxygen species, a major cause of neuron death, than free NRTIs. Optimization of size, surface decoration with AP significantly increased brain accumulation of nano-NRTIs. The efficient CNS delivery of nano-NRTIs resulted in up to 10-fold suppression of retroviral activity and reduced virus-associated inflammation in humanized mouse model of HIV-1 infection in the brain. Our data provide proof of the advanced efficacy of nano-NRTIs as safer alternative of current antiviral drugs. FROM THE CLINICAL EDITOR: This team of investigators demonstrated low neurotoxicity and excellent anti-HIV activity of nano-nucleoside reverse transcriptase inhibitors decorated with the peptide (AP) binding brain-specific apolipoprotein E receptor, providing proof of enhanced efficacy and a safer alternative compared with current antiviral drugs.


Subject(s)
Antiviral Agents/administration & dosage , HIV Infections/drug therapy , Polyethylene Glycols/administration & dosage , Polyethyleneimine/administration & dosage , Reverse Transcriptase Inhibitors/administration & dosage , Animals , Antiviral Agents/adverse effects , Antiviral Agents/chemistry , Apoptosis/drug effects , Brain/drug effects , Central Nervous System/drug effects , Central Nervous System/pathology , HIV Infections/virology , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , HIV-1/pathogenicity , Humans , Mice , Mice, Transgenic , Nanogels , Polyethylene Glycols/chemistry , Polyethyleneimine/chemistry , Reactive Oxygen Species/metabolism , Reverse Transcriptase Inhibitors/adverse effects , Reverse Transcriptase Inhibitors/chemistry
6.
Bioconjug Chem ; 24(4): 658-68, 2013 Apr 17.
Article in English | MEDLINE | ID: mdl-23547842

ABSTRACT

Many drug-resistant tumors and cancer stem cells (CSC) express elevated levels of CD44 receptor, a cellular glycoprotein binding hyaluronic acid (HA). Here, we report the synthesis of nanogel-drug conjugates based on membranotropic cholesteryl-HA (CHA) for efficient targeting and suppression of drug-resistant tumors. These conjugates significantly increased the bioavailability of poorly soluble drugs with previously reported activity against CSC, such as etoposide, salinomycin, and curcumin. The small nanogel particles (diameter 20-40 nm) with a hydrophobic core and high drug loads (up to 20%) formed after ultrasonication and demonstrated a sustained drug release following the hydrolysis of biodegradable ester linkage. Importantly, CHA-drug nanogels demonstrated 2-7 times higher cytotoxicity in CD44-expressing drug-resistant human breast and pancreatic adenocarcinoma cells compared to that of free drugs and nonmodified HA-drug conjugates. These nanogels were efficiently internalized via CD44 receptor-mediated endocytosis and simultaneous interaction with the cancer cell membrane. Anchoring by cholesterol moieties in the cellular membrane after nanogel unfolding evidently caused more efficient drug accumulation in cancer cells compared to that in nonmodified HA-drug conjugates. CHA-drug nanogels were able to penetrate multicellular cancer spheroids and displayed a higher cytotoxic effect in the system modeling tumor environment than both free drugs and HA-drug conjugates. In conclusion, the proposed design of nanogel-drug conjugates allowed us to significantly enhance drug bioavailability, cancer cell targeting, and the treatment efficacy against drug-resistant cancer cells and multicellular spheroids.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Hyaluronan Receptors/metabolism , Hyaluronic Acid/chemistry , Neoplastic Stem Cells/drug effects , Polyethylene Glycols/chemistry , Polyethyleneimine/chemistry , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cholesterol/chemistry , Drug Design , Humans , MCF-7 Cells , Models, Molecular , Molecular Structure , Nanogels , Neoplastic Stem Cells/metabolism , Structure-Activity Relationship
7.
J Control Release ; 167(2): 200-9, 2013 Apr 28.
Article in English | MEDLINE | ID: mdl-23385032

ABSTRACT

A majority of nanoencapsulated drugs that have shown promise in cancer chemotherapy are administered intravenously. Development of effective oral nanoformulations presents a very challenging medical goal. Here, we describe successful applications of innovative polymeric nanogels in the form of conjugates with activated nucleoside analogs for oral administration in cancer chemotherapy. Previously, we reported the synthesis of amphiphilic polyvinyl alcohol and dextrin-based nanogel conjugates with the phosphorylated 5-FU nucleoside Floxuridine and demonstrated their enhanced activity against regular and drug-resistant cancers (T.H. Senanayake, G. Warren, S.V. Vinogradov, Novel anticancer polymeric conjugates of activated nucleoside analogs, Bioconjug. Chem. 22 (2011) 1983-1993). In this study, we synthesized and evaluated oral applications of nanogel conjugates of a protected Gemcitabine, the drug never used in oral therapies. These conjugates were able to quickly release an active form of the drug (Gemcitabine 5'-mono-, di- and triphosphates) by specific enzymatic activities, or slowly during hydrolysis. Gemcitabine conjugates demonstrated up to 127 times higher in vitro efficacy than the free drug against various cancer cells, including the lines resistant to nucleoside analogs. Surprisingly, these nanogel-drug conjugates were relatively stable in gastric conditions and able to actively penetrate through the gastrointestinal barrier based on permeability studies in Caco-2 cell model. In tumor xenograft models of several drug-resistant human cancers, we observed an efficient inhibition of tumor growth and extended the life-span of the animals by 3 times that of the control with orally treated Gemcitabine- or Floxuridine-nanogel conjugates. Thus, we have demonstrated a potential of therapeutic nanogel conjugates with the activated and stabilized Gemcitabine as a successful oral drug form against Gemcitabine-resistant and other drug-resistant tumors.


Subject(s)
Antineoplastic Agents/administration & dosage , Deoxycytidine/analogs & derivatives , Nanostructures/administration & dosage , Neoplasms/drug therapy , Administration, Oral , Alkaline Phosphatase/chemistry , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Cholesterol/chemistry , Deoxycytidine/administration & dosage , Deoxycytidine/chemistry , Drug Resistance, Neoplasm , Female , Humans , Hydrolysis , Mice , Mice, Nude , Nanostructures/chemistry , Neoplasms/pathology , Particle Size , Permeability , Phosphoric Diester Hydrolases/chemistry , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
8.
Medchemcomm ; 3(1): 14-21, 2012.
Article in English | MEDLINE | ID: mdl-23293738

ABSTRACT

Chromatin remodelling enzymes such as the histone deacetylases (HDACs) and histone demethylases such as lysine-specific demethylase 1 (LSD1) have been validated as targets for cancer drug discovery. Although a number of HDAC inhibitors have been marketed or are in human clinical trials, the search for isoform-specific HDAC inhibitors is an ongoing effort. In addition, the discovery and development of compounds targeting histone demethylases are in their early stages. Epigenetic modulators used in combination with traditional antitumor agents such as 5-azacytidine represent an exciting new approach to cancer chemotherapy. We have developed multiple series of HDAC inhibitors and LSD1 inhibitors that promote the re-expression of aberrantly silenced genes that are important in human cancer. The design, synthesis and biological activity of these analogues is described herein.

9.
Bioconjug Chem ; 22(10): 1983-93, 2011 Oct 19.
Article in English | MEDLINE | ID: mdl-21863885

ABSTRACT

Inherent or therapy-induced drug resistance is a major clinical setback in cancer treatment. The extensive usage of cytotoxic nucleobases and nucleoside analogues in chemotherapy also results in the development of specific mechanisms of drug resistance, such as nucleoside transport or activation deficiencies. These drugs are prodrugs; and being converted into the active mono-, di-, and triphosphates inside cancer cells following administration, they affect nucleic acid synthesis, nucleotide metabolism, or sensitivity to apoptosis. Previously, we actively promoted the idea that the nanodelivery of active nucleotide species, e.g., 5'-triphosphates of nucleoside analogues, can enhance drug efficacy and reduce nonspecific toxicity. In this study, we report the development of a novel type of drug nanoformulations, polymeric conjugates of nucleoside analogues, which are capable of the efficient transport and sustained release of phosphorylated drugs. These drug conjugates have been synthesized, starting from cholesterol-modified mucoadhesive polyvinyl alcohol or biodegradable dextrin, by covalent attachment of nucleoside analogues through a tetraphosphate linker. Association of cholesterol moieties in aqueous media resulted in intramolecular polymer folding and the formation of small nanogel particles containing 0.5 mmol/g of a 5'-phosphorylated nucleoside analogue, e.g., 5-fluoro-2'-deoxyuridine (floxuridine, FdU), an active metabolite of anticancer drug 5-fluorouracyl (5-FU). The polymeric conjugates demonstrated rapid enzymatic release of floxuridine 5'-phosphate and much slower drug release under hydrolytic conditions (pH 1.0-7.4). Among the panel of cancer cell lines, all studied polymeric FdU-conjugates demonstrated an up to 50× increased cytotoxicity in human prostate cancer PC-3, breast cancer MCF-7, and MDA-MB-231 cells, and more than 100× higher efficacy against cytarabine-resistant human T-lymphoma (CEM/araC/8) and gemcitabine-resistant follicular lymphoma (RL7/G) cells as compared to free drugs. In the initial in vivo screening, both PC-3 and RL7/G subcutaneous tumor xenograft models showed enhanced sensitivity to sustained drug release from polymeric FdU-conjugate after peritumoral injections and significant tumor growth inhibition. All these data demonstrate a remarkable clinical potential of novel polymeric conjugates of phosphorylated nucleoside analogues, especially as new therapeutic agents against drug-resistant tumors.


Subject(s)
Antimetabolites, Antineoplastic/chemistry , Fluorouracil/chemistry , Nucleosides/chemistry , Polymers/chemistry , Prodrugs/chemistry , Animals , Antimetabolites, Antineoplastic/therapeutic use , Cell Line, Tumor , Cholesterol/chemistry , Female , Fluorouracil/therapeutic use , Humans , Male , Mice , Neoplasms/drug therapy , Neoplasms/pathology , Prodrugs/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology
11.
J Cell Mol Med ; 13(8B): 1877-85, 2009 Aug.
Article in English | MEDLINE | ID: mdl-20141611

ABSTRACT

The histone deacetylase (HDAC) inhibitor trichostatin A (TSA) has recently been shown to inhibit deleterious effects of cytokines on beta-cells, but it is unable to protect beta-cells from death due to its own cytotoxicity. Herein, we investigated novel HDAC inhibitors for their cytoprotective effects against IL-1beta-induced damage to isolated beta-cells. We report that three novel compounds (THS-73-44, THS-72-5 and THS-78-5) significantly inhibited HDAC activity and increased the acetylation of histone H4 in isolated beta-cells. Further, these compounds exerted no toxic effects on metabolic cell viability in these cells. However, among the three compounds tested, only THS-78-5 protected against IL-1beta-mediated loss in beta-cell viability. THS-78-5 was also able to attenuate IL-1beta-induced inducible nitric oxide synthase expression and subsequent NO release. Our data also indicate that the cytoprotective properties of THS-78-5 against IL-1beta-mediated effects may, in part, be due to inhibition of IL-1beta-induced transactivation of nuclear factor kappaB (NF-kappaB) in these cells. Together, we provide evidence for a novel HDAC inhibitor with a significant potential to prevent IL-1beta-mediated effects on isolated beta-cells. Potential implications of these findings in the development of novel therapeutics to prevent deleterious effects of cytokines and the onset of autoimmune diabetes are discussed.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Islets of Langerhans/drug effects , Animals , Cell Line , Humans , Interleukin-1beta/physiology , Islets of Langerhans/metabolism
12.
J Med Chem ; 51(8): 2447-56, 2008 Apr 24.
Article in English | MEDLINE | ID: mdl-18348516

ABSTRACT

A series of polyaminohydroxamic acids (PAHAs) and polyaminobenzamides (PABAs) were synthesized and evaluated as isoform-selective histone deacetylase (HDAC) inhibitors. These analogues contain a polyamine chain to increase affinity for chromatin and facilitate cellular import. Seven PAHAs inhibited HDAC >50% (1 microM), and two PABAs inhibited HDAC >50% (5 microM). Compound 17 increased acetylated alpha-tubulin in HCT116 colon tumor cells 253-fold but only modestly increased p21 (waf1) and acetylated histones 3 and 4, suggesting that 17 selectively inhibits HDAC 6. PABA 22 alone minimally increased p21 (waf1) and acetylated histones 3 and 4 but caused dose-dependent increases in p21 (waf1) in combination with 0.1 microM 5-azadeoxycytidine. Finally, 22 appeared to be a substrate for the polyamine transport system. None of these compounds were cytotoxic at 100 microM. PAHAs and PABAs exhibit strikingly different cellular effects from SAHA and have the potential for use in combination antitumor therapies with reduced toxicity.


Subject(s)
Benzamides/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Isoenzymes/antagonists & inhibitors , Cell Line, Tumor , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Humans , Magnetic Resonance Spectroscopy , Spectrophotometry, Infrared
SELECTION OF CITATIONS
SEARCH DETAIL