Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Int J Nanomedicine ; 13: 6359-6374, 2018.
Article in English | MEDLINE | ID: mdl-30349255

ABSTRACT

INTRODUCTION: The potential of layered double hydroxide (LDH) as a host of multiple ultraviolet-ray absorbers was investigated by simultaneous intercalation of benzophenone 4 (B4) and Eusolex® 232 (EUS) in Zn/Al LDH. METHODS: The nanocomposites were prepared via coprecipitation method at various molar ratios of B4 and EUS. RESULTS: At equal molar ratios, the obtained nanocomposite showed an intercalation selectivity that is preferential to EUS. However, the selectivity ratio of intercalated anions was shown to be capable of being altered by adjusting the molar ratio of intended guests during synthesis. Dual-guest nanocomposite synthesized with B4:EUS molar ratio 3:1 (ZEB [3:1]) showed an intercalation selectivity ratio of B4:EUS =53:47. Properties of ZEB (3:1) were monitored using powder X-ray diffractometer to show a basal spacing of 21.8 Å. Direct-injection mass spectra, Fourier transform infrared spectra, and ultraviolet-visible spectra confirmed the dual intercalation of both anions into the interlayer regions of dual-guest nanocomposite. The cytotoxicity study of dual-guest nanocomposite ZEB (3:1) on human dermal fibroblast cells showed no significant toxicity until 25 µg/mL. CONCLUSION: Overall, the findings demonstrate successful customization of ultraviolet-ray absorbers composition in LDH host.


Subject(s)
Dermis/drug effects , Fibroblasts/drug effects , Hydroxides/chemistry , Intercalating Agents/administration & dosage , Nanocomposites/administration & dosage , Sunscreening Agents/administration & dosage , Ultraviolet Rays , Cells, Cultured , Dermis/cytology , Dermis/radiation effects , Fibroblasts/cytology , Fibroblasts/radiation effects , Humans , Intercalating Agents/chemistry , Nanocomposites/chemistry , Sunscreening Agents/chemistry
2.
Acta Biomater ; 77: 63-73, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30006312

ABSTRACT

Topical chemotherapy with complementary drugs is one of the most promising strategies to achieve an effective antitumor activity. Herein, a synergistic strategy for hepatoma therapy by the combination of tumor microenvironment-sensitive polymer-doxorubicin (DOX) conjugate thermogel, containing a DNA intercalator DOX, and docetaxel (DTX), a microtubule-interfering agent, was proposed. First, cis-aconitic anhydride-functionalized DOX (CAD) and succinic anhydride-modified DOX (SAD) were conjugated onto the terminal hydroxyl groups of poly(lactide-co-glycolide)-block-poly(ethylene glycol)-block-poly(lactide-co-glycolide) (PLGA-PEG-PLGA), yielding the acid-sensitive CAD-PLGA-PEG-PLGA-CAD and the insensitive SAD-PLGA-PEG-PLGA-SAD conjugates, respectively. The prodrug aqueous solution exhibited a thermoreversible sol-gel transition between room and physiological temperature. Meantime, appropriate mechanical property, biodegradability, as well as a sustained release profile were revealed in such prodrug thermogels. More importantly, the addition of DTX to the DOX-conjugated thermogels (i.e., Gel-CAD and Gel-SAD) was verified with enhanced curative effect against tumor, where the antitumor efficacy of Gel-CAD+DTX was obviously higher than the other groups. A reliable security in vivo was also showed in the Gel-CAD+DTX group. Taken together, such combination of tumor microenvironment-labile prodrug thermogel and a complementary drug exhibited fascinating prospect for local synergistic antineoplastic therapy. STATEMENT OF SIGNIFICANCE: Multidrug chemotherapy with synergistic effect has been proposed recently for hepatoma treatment in the clinic. However, the quick release, fast elimination, and unselectivity of multidrugs in vivo always limit their further application. To solve this problem, a synergistic combination of tumor microenvironment-sensitive polymeric doxorubicin (DOX) prodrug thermogel for DNA intercalation and a microtubule-interfering agent docetaxel (DTX) is developed in the present study for the local chemotherapy of hepatoma. Interestingly, a pH-triggered sustained release behavior, an enhanced antitumor efficacy, and a favorable security in vivo are observed in the combined dual-drug delivery platform. Therefore, effectively combining tumor microenvironment-labile polymeric prodrug thermogel with a complementary drug provides an advanced system and a promising prospect for local synergistic hepatoma chemotherapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Doxorubicin/administration & dosage , Drug Delivery Systems , Liver Neoplasms/drug therapy , Tumor Microenvironment , Administration, Topical , Animals , Docetaxel/administration & dosage , Doxorubicin/chemistry , Endosomes/metabolism , Female , Gels , Humans , Intercalating Agents/administration & dosage , Mice , Microtubules/drug effects , Neoplasm Transplantation , Polyesters/chemistry , Polyethylene Glycols/chemistry , Prodrugs , Rheology , Succinic Anhydrides/chemistry , Tissue Distribution
3.
Nucleic Acids Res ; 46(7): 3284-3297, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29554366

ABSTRACT

Despite All-trans retinoic acid (ATRA) has transformed acute promyelocytic leukemia (APL) from the most fatal to the most curable hematological cancer, there remains a clinical challenge that many high-risk APL patients who fail to achieve a complete molecular remission or relapse and become resistant to ATRA. Herein, we report that 5-(4-methoxyphenethyl)-[1, 3] dioxolo [4, 5-j] phenanthridin-6(5H)-one (ZYH005) exhibits specific anticancer effects on APL and ATRA-resistant APL in vitro and vivo, while shows negligible cytotoxic effect on non-cancerous cell lines and peripheral blood mononuclear cells from healthy donors. Using single-molecule magnetic tweezers and molecule docking, we demonstrate that ZYH005 is a DNA intercalator. Further mechanistic studies show that ZYH005 triggers DNA damage, and caspase-dependent degradation of the PML-RARa fusion protein. As a result, APL and ATRA-resistant APL cells underwent apoptosis upon ZYH005 treatment and this apoptosis-inducing effect is even stronger than that of arsenic trioxide and anticancer agents including 5-fluorouracil, cisplatin and doxorubicin. Moreover, ZYH005 represses leukemia development in vivo and prolongs the survival of both APL and ATRA-resistant APL mice. To our knowledge, ZYH005 is the first synthetic phenanthridinone derivative, which functions as a DNA intercalator and can serve as a potential candidate drug for APL, particularly for ATRA-resistant APL.


Subject(s)
Drug Resistance, Neoplasm/genetics , Intercalating Agents/administration & dosage , Leukemia, Promyelocytic, Acute/drug therapy , Phenanthridines/administration & dosage , Animals , Apoptosis/drug effects , Arsenic Trioxide/administration & dosage , Arsenic Trioxide/chemistry , Caspases/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Intercalating Agents/chemistry , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Mice , Molecular Docking Simulation , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/genetics , Phenanthridines/chemistry , Promyelocytic Leukemia Protein/genetics , Proteolysis/drug effects , Retinoic Acid Receptor alpha/genetics , Tretinoin/administration & dosage , Tretinoin/chemistry , Xenograft Model Antitumor Assays
4.
J Inorg Biochem ; 181: 18-27, 2018 04.
Article in English | MEDLINE | ID: mdl-29353086

ABSTRACT

Dendrimers, which are considered as one of the most promising tools in the field of nanobiotechnology due to their structural organization, showed a great potential in gene therapy, drug delivery, medical imaging and as antimicrobial and antiviral agents. This article is devoted to study interactions between new carbosilane-based metallodendrimers containing ruthenium and anti-cancer small interfering RNA (siRNA). Formation of complexes between anti-cancer siRNAs and Ru-based carbosilane dendrimers was evaluated by transmission electron microscopy, circular dichroism and fluorescence. The zeta-potential and the size of dendriplexes were determined by dynamic light scattering. The internalization of dendriplexes were estimated using HL-60 cells. Results show that ruthenium dendrimers associated with anticancer siRNA have the ability to deliver siRNA as non-viral vectors into the cancer cells. Moreover, dendrimers can protect siRNA against nuclease degradation. Nevertheless, further research need to be performed to examine the therapeutic potential of ruthenium dendrimers as well as dendrimers complexed with siRNA and anticancer drugs towards cancer cells.


Subject(s)
Antineoplastic Agents/metabolism , Dendrimers/metabolism , Intercalating Agents/metabolism , Leukemia, Promyelocytic, Acute/metabolism , Models, Molecular , RNA, Small Interfering/metabolism , Ruthenium/metabolism , Absorption, Physiological , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Circular Dichroism , Dendrimers/administration & dosage , Dendrimers/chemistry , Drug Stability , Dynamic Light Scattering , HL-60 Cells , Humans , Intercalating Agents/administration & dosage , Intercalating Agents/chemistry , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/pathology , Microscopy, Electron, Transmission , Molecular Conformation , Molecular Dynamics Simulation , Molecular Structure , Particle Size , RNA Interference , RNA Stability , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , RNA, Small Interfering/ultrastructure , Ruthenium/administration & dosage , Ruthenium/chemistry , Silanes/chemistry , Silanes/metabolism , Surface Properties
5.
Int J Nanomedicine ; 12: 2657-2672, 2017.
Article in English | MEDLINE | ID: mdl-28435250

ABSTRACT

DNA nanostructures prepared by self-assembly possess good stability, high biocompatibility, and low immunogenicity as drug delivery vehicles. In this work, DNA tetrahedron (TD) was constructed and modified with SL2B aptamer (S) and folic acid (F). TD possessed a small diameter (~6 nm) and entered into the nucleus quickly. SL2B aptamer can inhibit cancer cell growth by disturbing vascular endothelial growth factor/Notch signaling pathways. To explore the effect of SL2B number on colorectal cancer inhibition, SL2B multimers (dimer, trimer, and tetramer) were constructed by functionalization of TD with different numbers of SL2B. One SL2B per TD was the most efficient anticancer strategy and showed significantly better anticancer efficacy than SL2B, probably due to the enhanced stability of SL2B by TD. Doxorubicin (DOX) is a potent anticancer agent that can intercalate into DNA double strands. Results showed that TD could facilitate DOX entrance into the nucleus and the intracellular delivery of DOX was further enhanced by functionalization of SL2B and F. DOX-intercalated TD modified with two F and two S (DOX@TD-2F2S) could cause sufficient HT-29 cell inhibition at a much lower DOX concentration. In sum, DOX@TD-2F2S exhibited a synergic anticancer biological effect with chemotherapy and can be a promising strategy for treating colorectal cancer.


Subject(s)
Aptamers, Nucleotide/chemistry , Colorectal Neoplasms/drug therapy , Folic Acid/administration & dosage , Nanostructures/administration & dosage , Aptamers, Nucleotide/administration & dosage , DNA/chemistry , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Delivery Systems/methods , Drug Stability , Folic Acid/chemistry , Folic Acid/pharmacology , HT29 Cells/drug effects , Humans , Intercalating Agents/administration & dosage , Intercalating Agents/chemistry , Molecular Targeted Therapy , Nanostructures/chemistry
6.
Biomaterials ; 91: 44-56, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26994877

ABSTRACT

By its unique advantages over traditional medicine, nanomedicine has offered new strategies for cancer treatment. In particular, the development of drug delivery strategies has focused on nanoscale particles to improve bioavailability. However, many of these nanoparticles are unable to overcome tumor resistance to chemotherapeutic agents. Recently, new opportunities for drug delivery have been provided by oligonucleotides that can self-assemble into three-dimensional nanostructures. In this work, we have designed and developed functional DNA nanostructures to deliver the chemotherapy drug doxorubicin (Dox) to resistant cancer cells. These nanostructures have two components. The first component is a DNA aptamer, which forms a dimeric G-quadruplex nanostructure to target cancer cells by binding with nucleolin. The second component is double-stranded DNA (dsDNA), which is rich in -GC- base pairs that can be applied for Dox delivery. We demonstrated that Dox was able to efficiently intercalate into dsDNA and this intercalation did not affect the aptamer's three-dimensional structure. In addition, the Aptamer-dsDNA (ApS) nanoparticle showed good stability and protected the dsDNA from degradation in bovine serum. More importantly, the ApS&Dox nanoparticle efficiently reversed the resistance of human breast cancer cells to Dox. The mechanism circumventing doxorubicin resistance by ApS&Dox nanoparticles may be predominantly by cell cycle arrest in S phase, effectively increased cell uptake and decreased cell efflux of doxorubicin. Furthermore, the ApS&Dox nanoparticles could effectively inhibit tumor growth, while less cardiotoxicity was observed. Overall, this functional DNA nanostructure provides new insights into the design of nanocarriers to overcome multidrug resistance through targeted drug delivery.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Aptamers, Nucleotide/chemistry , Breast Neoplasms/drug therapy , Doxorubicin/administration & dosage , Drug Carriers/chemistry , Drug Delivery Systems , Nanoparticles/chemistry , Animals , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Aptamers, Nucleotide/metabolism , Base Sequence , Breast/drug effects , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Carriers/metabolism , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Female , G-Quadruplexes , Humans , Intercalating Agents/administration & dosage , Intercalating Agents/pharmacology , Intercalating Agents/therapeutic use , MCF-7 Cells , Mice, Inbred BALB C , Models, Molecular , Nanoparticles/metabolism , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Nucleolin
7.
Chem Rev ; 116(5): 3436-86, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26865551

ABSTRACT

The platinum drugs, cisplatin, carboplatin, and oxaliplatin, prevail in the treatment of cancer, but new platinum agents have been very slow to enter the clinic. Recently, however, there has been a surge of activity, based on a great deal of mechanistic information, aimed at developing nonclassical platinum complexes that operate via mechanisms of action distinct from those of the approved drugs. The use of nanodelivery devices has also grown, and many different strategies have been explored to incorporate platinum warheads into nanomedicine constructs. In this Review, we discuss these efforts to create the next generation of platinum anticancer drugs. The introduction provides the reader with a brief overview of the use, development, and mechanism of action of the approved platinum drugs to provide the context in which more recent research has flourished. We then describe approaches that explore nonclassical platinum(II) complexes with trans geometry or with a monofunctional coordination mode, polynuclear platinum(II) compounds, platinum(IV) prodrugs, dual-threat agents, and photoactivatable platinum(IV) complexes. Nanoparticles designed to deliver platinum(IV) complexes will also be discussed, including carbon nanotubes, carbon nanoparticles, gold nanoparticles, quantum dots, upconversion nanoparticles, and polymeric micelles. Additional nanoformulations, including supramolecular self-assembled structures, proteins, peptides, metal-organic frameworks, and coordination polymers, will then be described. Finally, the significant clinical progress made by nanoparticle formulations of platinum(II) agents will be reviewed. We anticipate that such a synthesis of disparate research efforts will not only help to generate new drug development ideas and strategies, but also will reflect our optimism that the next generation of approved platinum cancer drugs is about to arrive.


Subject(s)
Antineoplastic Agents/chemical synthesis , Drug Carriers/chemical synthesis , Organoplatinum Compounds/chemical synthesis , Prodrugs/chemical synthesis , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Humans , Intercalating Agents/administration & dosage , Intercalating Agents/chemical synthesis , Intercalating Agents/pharmacology , Nanoparticles/chemistry , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/pharmacology , Prodrugs/administration & dosage , Prodrugs/pharmacology
8.
Bioelectrochemistry ; 109: 1-8, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26764570

ABSTRACT

Composites consisting of ss- and ds-DNA strands and polyacrylamide (PAM) hydrogel have been synthesized. DNA was entrapped non-covalently. The obtained DNA biomaterial exhibited a strong increase in guanine and adenine anodic currents when temperature reached the physiological level. This increase was related to the unique oligonucleotide structural changes in the composite. The structural alterations in the PAM lattices were employed for the release of the drug accumulated in the composite. Doxorubicin (Dox) was selected as the drug; it was accumulated by intercalation to dsDNA and was slowly released from the dsDNA/PAM system by using a minor temperature increase (up to 40÷45 °C) as it is routinely done in hyperthermia. The applied release temperature was either constant or oscillating. The binding strength, the rate of Dox release and the properties of the composite were examined using voltammetry, SEM and ICP-MS.


Subject(s)
Acrylic Resins/chemistry , Antibiotics, Antineoplastic/administration & dosage , DNA/chemistry , Delayed-Action Preparations/chemistry , Doxorubicin/administration & dosage , Hydrogels/chemistry , Biocompatible Materials/chemistry , Guanine/chemistry , Intercalating Agents/administration & dosage , Oxidation-Reduction , Temperature
9.
Clin Cancer Res ; 21(14): 3196-203, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25829397

ABSTRACT

PURPOSE: Blood-based circulating-free (cf) tumor DNA may be an alternative to tissue-based EGFR mutation testing in NSCLC. This exploratory analysis compares matched tumor and blood samples from the FASTACT-2 study. EXPERIMENTAL DESIGN: Patients were randomized to receive six cycles of gemcitabine/platinum plus sequential erlotinib or placebo. EGFR mutation testing was performed using the cobas tissue test and the cobas blood test (in development). Blood samples at baseline, cycle 3, and progression were assessed for blood test detection rate, sensitivity, and specificity; concordance with matched tumor analysis (n = 238), and correlation with progression-free survival (PFS) and overall survival (OS). RESULTS: Concordance between tissue and blood tests was 88%, with blood test sensitivity of 75% and a specificity of 96%. Median PFS was 13.1 versus 6.0 months for erlotinib and placebo, respectively, for those with baseline EGFR mut(+) cfDNA [HR, 0.22; 95% confidence intervals (CI), 0.14-0.33, P < 0.0001] and 6.2 versus 6.1 months, respectively, for the EGFR mut(-) cfDNA subgroup (HR, 0.83; 95% CI, 0.65-1.04, P = 0.1076). For patients with EGFR mut(+) cfDNA at baseline, median PFS was 7.2 versus 12.0 months for cycle 3 EGFR mut(+) cfDNA versus cycle 3 EGFR mut(-) patients, respectively (HR, 0.32; 95% CI, 0.21-0.48, P < 0.0001); median OS by cycle 3 status was 18.2 and 31.9 months, respectively (HR, 0.51; 95% CI, 0.31-0.84, P = 0.0066). CONCLUSIONS: Blood-based EGFR mutation analysis is relatively sensitive and highly specific. Dynamic changes in cfDNA EGFR mutation status relative to baseline may predict clinical outcomes.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , DNA Mutational Analysis/methods , DNA, Neoplasm/blood , Lung Neoplasms/genetics , Aged , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/mortality , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Disease-Free Survival , Double-Blind Method , Erlotinib Hydrochloride/administration & dosage , Female , Genes, erbB-1 , Humans , Intercalating Agents/administration & dosage , Kaplan-Meier Estimate , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Male , Middle Aged , Mutation , Platinum Compounds/administration & dosage , Treatment Outcome , Gemcitabine
10.
J Control Release ; 195: 11-20, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25135792

ABSTRACT

In the development of potent polymeric gene carriers for gene therapy, a good interaction between the polymer and the nucleotide is indispensable to form small and stable polyplexes. Polymers with relatively high cationic charge density are frequently used to provide these interactions, but high cationic charge is usually associated with severe cytotoxicity. In this study an alternative, nucleotide specific binding interaction based on intercalation was investigated to improve polymer/pDNA complex formation. For this purpose bioreducible poly(amido amine) copolymers (p(CBA-ABOL/Nic)) were synthesized with different degrees of intercalating quaternary nicotinamide (Nic) groups and amide-substituted derivatives in their side chains. The quaternary nicotinamide group was chosen as intercalating moiety because this group is part of the naturally occurring NAD+ coenzyme and is therefore expected to be non-toxic and non-carcinogenic. The presence of the quaternary nicotinamide moieties in the poly(amido amine) copolymers showed to effectively promote self-assembled polyplex formation already at low polymer/DNA ratios and results in decreased polyplex size and increased stability of the polyplexes. Furthermore, in contrast to the primary amine functionalized analogs the quaternary nicotinamide polymers showed to be non-hemolytic, indicating their compatibility with cell membranes. Polymers with 25% of Nic in the side chains induced GFP expressions of about 4-5 times that of linear PEI, which is comparable with p(CBA-ABOL), the parent PAA without Nic, but at a two- to fourfold lower required polymer dose. N-phenylation of the nicotinamide functionality even further reduces the required polymer dose to form stable polyplexes, which is a major improvement for these kinds of cationic polymers.


Subject(s)
Gene Transfer Techniques , Intercalating Agents , Niacinamide , Polymers , Animals , COS Cells , Cell Line, Tumor , Cell Survival/drug effects , Chlorocebus aethiops , DNA/administration & dosage , DNA/chemistry , Erythrocytes/drug effects , Gene Expression , Genes, Reporter , Green Fluorescent Proteins/genetics , Hemolysis/drug effects , Humans , Intercalating Agents/administration & dosage , Intercalating Agents/chemistry , Niacinamide/administration & dosage , Niacinamide/chemistry , Plasmids , Polymers/administration & dosage , Polymers/chemistry
11.
Invest Ophthalmol Vis Sci ; 55(6): 3809-18, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24854854

ABSTRACT

PURPOSE: Uveitis is a common cause of vision loss. The renin angiotensin system (RAS), which plays a vital role in cardiovascular system, is a potent mediator of inflammation and has been implicated in the pathogenesis of uveitis. A newly identified axis of RAS, ACE2/Ang-(1-7)/Mas, has emerged as a novel target because it counteracts the deleterious effect of angiotensin II. The purpose of this study was to investigate the effect of endogenous ACE2 activation in preventing endotoxin-induced uveitis (EIU) in mice. METHODS: ACE2 activator diminazene aceturate (DIZE) was administered both systemically and locally. For systemic administration, female BALB/c mice received intraperitoneal injection of DIZE (60 mg/kg body weight [BW]) for 2 days prior to lipopolysaccharide (LPS) intravitreal injection (125 ng) to induce uveitis. For local study, DIZE was given at 0.5, 0.1, and 0 mg/mL as eyedrops six times per day for 2 days before LPS injection. The anterior segment of the mice was examined at 12, 24, 48, and 72 hours after LPS injection, and clinical scores were determined at the same time. Morphology and infiltrating inflammatory cells were evaluated after 24 hours. The mRNA levels of inflammatory cytokines were analyzed by real-time RT-PCR. ACE2 activity was determined using a self-quenching fluorescent substrate. RESULTS: At 24 hours, the clinical score of mice treated with DIZE systemically was significantly lower (mean, ∼1.75) than the saline vehicle group (mean, ∼4) (P < 0.001). Histological examination showed 63.4% reduction of infiltrating inflammatory cells in the anterior segment and 57.4% reduction in the posterior segment of DIZE-treated eyes. The number of CD45(+) inflammatory cells in the vitreous of the DIZE-treated group was decreased (43.3%) compared to the vehicle group (P < 0.01). The mRNA levels of inflammatory cytokines were significantly reduced in the DIZE-treated group (P < 0.01, P < 0.001). The number of infiltrating inflammatory cells was also significantly reduced in eyes that received topical administration of DIZE: 73.8% reduction in the 0.5 mg/mL group and 51.7% reduction in the 0.1mg/mL group compared to the control group. DIZE treatment resulted in significantly increased ACE2 activity in the retina (P < 0.001). CONCLUSIONS: Endogenous ACE2 activation by DIZE has a preventive effect on LPS-induced ocular inflammation in the EIU mouse model. These results support the notions that RAS plays a role in modulating ocular immune response and that enhancing ACE2 provides a novel therapeutic strategy for uveitis.


Subject(s)
Anterior Eye Segment/pathology , Diminazene/analogs & derivatives , Enzyme Activation/drug effects , Peptidyl-Dipeptidase A/drug effects , Renin-Angiotensin System/drug effects , Uveitis/drug therapy , Angiotensin-Converting Enzyme 2 , Animals , Anterior Eye Segment/drug effects , Anterior Eye Segment/metabolism , Diminazene/administration & dosage , Disease Models, Animal , Female , Follow-Up Studies , Intercalating Agents/administration & dosage , Mice , Mice, Inbred BALB C , Ophthalmic Solutions , Peptidyl-Dipeptidase A/metabolism , Renin-Angiotensin System/physiology , Uveitis/chemically induced , Uveitis/enzymology
12.
Bioorg Khim ; 37(3): 408-13, 2011.
Article in Russian | MEDLINE | ID: mdl-21899057

ABSTRACT

For a series of 1,10-phenantroline tris-beta-diketonate europium complexes (EuC), cytotoxic activity on the HBL-100 human breast carcinoma cells was determined. Liposomal preparation of the most active EuC, V12, was also tested for cytotoxicity. Testing of this preparation in vivo on starting lethal murine model of T cell leukemic lymphoma ASF-LL showed that the inclusion of V12 in liposomes did not increase its antitumour activity in a local mode of administration.


Subject(s)
Antineoplastic Agents/administration & dosage , Europium/administration & dosage , Intercalating Agents/administration & dosage , Phenanthrolines/administration & dosage , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Europium/chemistry , Female , Intercalating Agents/chemistry , Liposomes , Mice , Phenanthrolines/chemistry
13.
J Drug Target ; 19(9): 846-55, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21692679

ABSTRACT

BACKGROUND: Increasing attention is currently focussed on the issue of finding strategies for the delivery of Auger-electron-emitting radionuclides into tumor cell nuclei. PURPOSE: In this study, we investigated tumor-cell uptake and cell-killing ability in vitro as well as in vivo biodistribution of an (125)I-labelled anthracycline derivative administered by means of HER2-targeted liposomes. METHODS: Anthracycline derivative Comp1 was radiolabelled with Auger-emitting (125)I and encapsulated in liposomes (DSPC:Chol:DSPE-PEG) using pH-gradient loading. Single-chain fragment F5 was anchored to the liposomes as targeting device for HER2. Uptake and specificity of (125)I-Comp1 delivered via targeting and non-targeting liposomes were analysed in cultured HER2-overexpressing cells. Cell-killing efficacy was evaluated in SKOV3 cells and biodistribution for up to 48 h was studied after intraperitoneal injection in tumor-bearing female BALB/c nu/nu mice. RESULTS: (125)I-Comp1 was specifically taken up by the cultured cells when administered by means of HER2-targeted liposomes and a clear dose-effect correlation in survival of cells was seen with increasing specific activity. The biodistribution studies revealed that (125)I-Comp1 accumulated in tumors when distributed using HER2-targeted liposomes and that this effect was absent when using non-targeting liposomes. CONCLUSION: The HER2-targeted liposomes possess the properties needed to bring about tumor-specific delivery and therapeutic effect of (125)I-Comp1.


Subject(s)
Daunorubicin/analogs & derivatives , Drug Delivery Systems , Intercalating Agents/administration & dosage , Ovarian Neoplasms/drug therapy , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Daunorubicin/administration & dosage , Daunorubicin/pharmacokinetics , Dose-Response Relationship, Drug , Female , Humans , Injections, Intraperitoneal , Intercalating Agents/pharmacokinetics , Iodine Radioisotopes , Liposomes , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/pathology , Receptor, ErbB-2/metabolism , Tissue Distribution
14.
Mutat Res ; 609(1): 47-59, 2006 Oct 10.
Article in English | MEDLINE | ID: mdl-16857419

ABSTRACT

We have previously noted that the Physicians' Desk Reference (PDR) contains over 80 instances in which a drug elicited a positive genotoxic response in one or more in vitro assays, despite having no obvious structural features predictive of covalent drug/DNA interactive potential or known mechanistic basis. Furthermore, in most cases, these drugs were "missed" by computational genotoxicity-predicting models such as DEREK, MCASE and TOPKAT. We have previously reported the application of a V79 cell-based model and a 3D DNA docking model for predicting non-covalent chemical/DNA interactions. Those studies suggested that molecules that are very widely structurally diverse may be capable of intercalating into DNA. To determine whether such non-covalent drug/DNA interactions might be involved in unexpected drug genotoxicity, we evaluated, using both models where possible, 56 marketed pharmaceuticals, 40 of which were reported as being clastogenic in in vitro cytogenetics assays (chromosome aberrations/mouse lymphoma assay). As seen before, the two approaches showed good concordance (62%) and 26 of the 40 (65%) drugs exhibiting in vitro clastogenicity were predicted as intercalators by one or both methods. This finding provides support for the hypothesis that non-covalent DNA interaction may be a common mechanism of clastogenicity for many drugs having no obvious structural alerts for covalent DNA interaction.


Subject(s)
Intercalating Agents/toxicity , Micronuclei, Chromosome-Defective/chemically induced , Pharmaceutical Preparations/administration & dosage , Animals , Cell Line , Computer Simulation , DNA/chemistry , DNA/drug effects , DNA/genetics , Hydrogen Bonding , Intercalating Agents/administration & dosage , Intercalating Agents/chemistry , Micronucleus Tests , Models, Chemical , Molecular Structure , Pharmaceutical Preparations/chemistry , Product Surveillance, Postmarketing , Software , Static Electricity
15.
Cancer Res ; 65(9): 3815-22, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15867379

ABSTRACT

Histone acetylation and deacetylation are crucial in the regulation of gene expression. Dynamic changes in gene expression may affect chromatin structure and, consequently, the interaction of chromatin with regulatory factors. In this study, the effects of the antiseizure drug valproic acid (VPA) on the expression of genes that regulate the structure of chromatin and the access of macromolecules to the DNA were investigated. Exposure of breast cancer cells to VPA resulted in rapid dose-dependent hyperacetylation of the histones H3 and H4. VPA further induced a depletion of several members of the structural maintenance of chromatin (SMC) proteins, SMC-associated proteins, DNA methyltransferase, and heterochromatin proteins. Down-regulation of these proteins was associated with chromatin decondensation. The observed alterations of chromatin structure correlated with enhanced sensitivity of DNA to nucleases and increased interaction of DNA with intercalating agents. VPA-induced chromatin decondensation led to a sequence-specific potentiation of DNA-damaging agents in cell culture and xenograft models. Modulation of heterochromatin maintenance proteins was not a direct, but a downstream, effect of histone acetylation. The effects on the chromatin structure were reversible upon drug withdrawal, but obligatory for the potentiation of DNA-damaging agents. In addition to their antitumor activity as single agents, the chromatin decondensation induced by histone deacetylase inhibitors may enhance the efficacy of cytotoxic agents that act by targeting DNA. The proposed mechanism of action suggests an effect of drug sequencing on the antitumor activity of these drugs.


Subject(s)
Breast Neoplasms/genetics , Chromatin/drug effects , Valproic Acid/pharmacology , Acetylation/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Chromatin/genetics , Chromatin/metabolism , DNA Damage , DNA, Neoplasm/drug effects , DNA, Neoplasm/metabolism , Down-Regulation/drug effects , Drug Synergism , Female , Histones/metabolism , Humans , Intercalating Agents/administration & dosage , Intercalating Agents/pharmacology , Mice , Mice, Nude , Valproic Acid/administration & dosage , Xenograft Model Antitumor Assays
16.
Ann Oncol ; 14(5): 775-82, 2003 May.
Article in English | MEDLINE | ID: mdl-12702533

ABSTRACT

BACKGROUND: The aim of this study was to assess the feasibility of administering bizelesin, a cyclopropylpyrroloindole with extraordinarily high potency as a bifunctional DNA-damaging agent and selectivity for specific AT-rich DNA sequences, as a single i.v. bolus injection every 4 weeks in patients with advanced solid malignancies. The study also sought to determine the maximum tolerated dose (MTD) of bizelesin, characterize its pharmacokinetic behavior, and seek preliminary evidence of anticancer activity. PATIENTS AND METHODS: Patients with advanced solid malignancies were treated with escalating doses of bizelesin as an i.v. bolus injection every 4 weeks. The selection of the specific starting dose, 0.1 micro g/m(2), which was equivalent to one-tenth the toxic dose low in dogs, factored in large interspecies differences in myelotoxicity as gauged using an ex vivo hematopoietic colony-forming assay. Due to concerns about the high potency of bizelesin and the large interspecies differences in toxicity, a conservative dose-escalation scheme was used for dose-level assignment to determine the MTD levels for both minimally pretreated (MP) and heavily pretreated (HP) patients. A variety of analytical assays were assessed to reliably measure bizelesin concentrations in plasma. RESULTS: Sixty-two patients were treated with 185 courses of bizelesin at eight dose levels ranging from 0.1 to 1.5 micro g/m(2). Myelosuppression, principally neutropenia that was always brief, was the most common toxicity observed. Thrombocytopenia and anemia were uncommon and severe non-hematological effects were not observed. Severe neutropenia alone and/or associated with fever was consistently experienced by HP and MP patients at doses exceeding 0.71 and 1.26 micro g/m(2), respectively. These doses also resulted in functionally non-cumulative myelosuppression as repetitive treatment was well-tolerated. A 40% reduction in measurable disease lasting 24 months was noted in a patient with advanced ovarian carcinoma. Various analytical methods were evaluated but none demonstrated the requisite sensitivity to reliably quantify the minute plasma concentrations of bizelesin and metabolites resulting from administering microgram quantities of drug. CONCLUSIONS: The highly potent and unique cytotoxic agent, bizelesin can be feasibly administered to patients with advanced solid malignancies. The recommended doses for phase II studies of bizelesin as a bolus i.v. injection every 4 weeks are 0.71 and 1.26 micro g/m(2) in HP and MP patients, respectively. The characteristics of the myelosuppression, the paucity of severe toxicities with repetitive treatment, the preliminary antitumor activity noted, and, above all, its unique mechanism of action as a selective DNA-damaging agent and high potency, warrant disease-directed evaluations of bizelesin in solid and hematopoietic malignancies and consideration of its use as a cytotoxic in targeted conjugated therapeutics.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , DNA/metabolism , Indoles/administration & dosage , Indoles/metabolism , Neoplasms/drug therapy , Urea/analogs & derivatives , Urea/administration & dosage , Urea/metabolism , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Alkylating/chemistry , Antineoplastic Agents, Alkylating/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Duocarmycins , Female , Humans , Indoles/chemistry , Intercalating Agents/administration & dosage , Intercalating Agents/chemistry , Intercalating Agents/metabolism , Male , Middle Aged , Neoplasms/metabolism , Urea/chemistry
18.
Cancer Chemother Pharmacol ; 50(2): 151-4, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12172981

ABSTRACT

PURPOSE: The Gynecologic Oncology Group performed a phase II study to determine the response rate to pyrazoloacridine (PZA) in patients with advanced, persistent or recurrent squamous carcinoma of the cervix. METHODS: PZA was administered intravenously over 3 h every 3 weeks. A dose of 760 mg/m(2) was given to the first 11 patients and was reduced to 560 mg/m(2) for subsequent patients. The dose reduction was undertaken because of unexpected severe neutropenia among the initial patients. RESULTS: Among 24 evaluable patients, 21 of whom had prior chemotherapy, there was one, brief, complete response (4.2%) and no partial responses. The major toxicity was neutropenia. CONCLUSION: PZA at the dose and schedule employed, has insignificant activity in this population.


Subject(s)
Acridines/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Pyrazoles/therapeutic use , Uterine Cervical Neoplasms/drug therapy , Acridines/administration & dosage , Acridines/adverse effects , Adult , Aged , Carcinoma, Squamous Cell/pathology , Drug Administration Schedule , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/therapeutic use , Female , Humans , Infusions, Intravenous , Intercalating Agents/administration & dosage , Intercalating Agents/adverse effects , Intercalating Agents/therapeutic use , Middle Aged , Neutropenia/chemically induced , Nucleic Acid Synthesis Inhibitors/administration & dosage , Nucleic Acid Synthesis Inhibitors/adverse effects , Nucleic Acid Synthesis Inhibitors/therapeutic use , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Remission Induction , Salvage Therapy , Treatment Outcome , Uterine Cervical Neoplasms/pathology , Vomiting/chemically induced
19.
Adv Drug Deliv Rev ; 49(1-2): 175-87, 2001 Jul 02.
Article in English | MEDLINE | ID: mdl-11377810

ABSTRACT

Numerous drugs are known to deplete mitochondrial DNA (mtDNA) from mammalian cells. These include DNA polymerase gamma and type II topoisomerase inhibitors, lipophilic cationic compounds, and DNA intercalating and non intercalating agents. The effects of these drugs on mtDNA metabolism will be discussed and potential mechanisms underlying their depletion of mtDNA presented.


Subject(s)
DNA, Mitochondrial/drug effects , Drug Delivery Systems/methods , Nucleic Acid Synthesis Inhibitors , Topoisomerase I Inhibitors , Animals , Anti-Infective Agents/administration & dosage , Anti-Infective Agents/chemistry , Anti-Infective Agents, Local/administration & dosage , Anti-Infective Agents, Local/chemistry , DNA Polymerase gamma , DNA Topoisomerases, Type I/metabolism , DNA, Mitochondrial/metabolism , DNA-Directed DNA Polymerase , Dequalinium/administration & dosage , Dequalinium/chemistry , Fluoroquinolones , Humans , Intercalating Agents/administration & dosage , Intercalating Agents/chemistry , Oxidative Phosphorylation/drug effects , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/chemistry
20.
Cancer Chemother Pharmacol ; 43(2): 162-4, 1999.
Article in English | MEDLINE | ID: mdl-9923823

ABSTRACT

PURPOSE: We completed a phase II trial of CI-958 (NSC 635371) in patients with advanced colorectal cancer given at a dose of 700 mg/m2 every 21 days. METHODS: All 15 patients had metastatic disease and had been previously treated with one 5-fluorouracil-based regimen in an adjuvant (six) or metastatic (nine) setting. RESULTS: None of the patients treated with CI-958 had an objective response to treatment. Median survival was 4.8 months after the start of treatment. Leukopenia was the major toxicity, but no patient experienced febrile neutropenia. An acute febrile reaction was seen after infusion in four of the first nine patients treated. This was abrogated by pretreatment with dexamethasone in the remaining patients. CONCLUSIONS: CI-958 was not effective at this dose and schedule in patients with previously treated advanced colorectal cancer.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Indazoles/therapeutic use , Intercalating Agents/therapeutic use , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Drug Administration Schedule , Female , Humans , Indazoles/administration & dosage , Indazoles/adverse effects , Intercalating Agents/administration & dosage , Intercalating Agents/adverse effects , Leukopenia/chemically induced , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...