Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 57
1.
Molecules ; 25(8)2020 Apr 19.
Article En | MEDLINE | ID: mdl-32325815

In this study, a family of porphyrins based on 5,10,15,20-Tetrakis(4-ethylphenyl)porphyrin (1, Ph) and six metallo-derivatives (Zn2+(2, Ph-Zn), Sn4+(3, Ph-Sn), Mn2+ (4, Ph-Mn), Ni2+ (5, Ph-Ni), Al3+ (6, Ph-Al), and V3+ (7, Ph-V)) were tested as photosensitizers for photodynamic therapy against Leishmania braziliensis and panamensis. The singlet oxygen quantum yield value (ΦΔ) for (1-7) was measured using 1,3-diphenylisobenzofuran (DPBF) as a singlet oxygen trapping agent and 5,10,15,20-(tetraphenyl)-porphyrin (H2TPP) as a reference standard; besides, parasite viability was estimated by the MTT assay. After metal insertion into the porphyrin core, the ΦΔ increased from 0.76-0.90 and cell viability changed considerably. The ΦΔ and metal type changed the cytotoxic activity. Finally, (2) showed both the highest ΦΔ (0.90) and the best photodynamic activity against the parasites studied (IC50 of 1.2 µM).


Leishmania braziliensis/drug effects , Leishmania/drug effects , Metalloporphyrins/chemistry , Metalloporphyrins/toxicity , Photosensitizing Agents/chemistry , Porphyrins/chemistry , Singlet Oxygen/chemistry , Benzofurans/chemistry , Cell Survival/drug effects , Cell Survival/radiation effects , Inhibitory Concentration 50 , Leishmania/radiation effects , Leishmania braziliensis/radiation effects , Light , Metalloporphyrins/chemical synthesis , Photochemotherapy , Porphyrins/chemical synthesis , Porphyrins/toxicity
2.
Int J Toxicol ; 38(4): 291-302, 2019.
Article En | MEDLINE | ID: mdl-31333066

Since our earlier publication (Gad et al, 2013), BioMimetix has advanced BMX-010 (Manganese (III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin or MnTE020PyP; CASRN 219818-60-7) into clinical development as a topical agent for the treatment of psoriasis, atopic dermatitis, and pruritus (idiopathic nonspecific itch). A multiple dose phase I study has been completed in 64 patients without any serious adverse effects. During the course of development, the formulation was initially a gel but has been modified to a cream formulation. The nonclinical safety program has been carried onward to assess preclinical risk to patients. Additional studies completed and reported here include dermal sensitization in a Guinea Pig maximization test study, 2 rabbit phototoxicity studies, a 28-day oral toxicity study in juvenile mice, a 28-day topical systemic toxicity study in Gottingen minipigs, range-finding studies, and complete embryo-fetal developmental toxicity (Segment II) studies in mice and rabbits, an ICH M7 compliant qualification of impurities using 2 (Q)SAR in silico methods, and a 14-day subcutaneous toxicity study of mice to qualify an impurity. All studies (except the (Q)SAR evaluations) were performed in accordance with Good Laboratory Practices (GLP) using Good Manufacturing Practices (GMP) drug substance. The systemic toxicity studies, with the exception of the juvenile toxicity study, included toxicokinetic evaluations, which are reported here. The phase I clinical study had 67 patient participants who received topically applied BMX-010, and there were no notable safety findings and included pharmacokinetic determinations on these patients which are also reported here. Chronic GLP toxicity studies have been initiated in the mouse (6-month oral) and minipig (9-month dermal). To date, the only observed nonclinical toxicity remains a reversible hypertension seen in mice in response to Cmax levels with a no effect threshold, and there have been no drug-related adverse effects.


Metalloporphyrins/pharmacokinetics , Metalloporphyrins/toxicity , Administration, Cutaneous , Animals , Dermatitis, Atopic/drug therapy , Female , Guinea Pigs , Humans , Hypertension/chemically induced , Male , Mice , Psoriasis/drug therapy , Rabbits , Swine , Swine, Miniature , Toxicity Tests
3.
Eur J Med Chem ; 174: 66-75, 2019 Jul 15.
Article En | MEDLINE | ID: mdl-31029945

Two Zn(II) nitro porphyrin derivatives bearing combinations of meso-4-nitrophenyl and meso-4-methylpyridinium moieties and their free-base precursors were synthesized through one-pot microwave process, purified and characterized. The biological activity of these nitroporphyrins was assessed under both photodynamic and non-photodynamic conditions to correlate their structure-activity relationship (SAR). Unlike, the free-base precursors, Zn(II) complexes of these nitroporphyrins displayed nearly complete inhibition in the entry of lentiviruses such as HIV-1 and SIVmac under non-photodynamic conditions. In addition, the Zn(II) complexes also exhibited a higher in vitro photodynamic activity towards human lung cancer cell-line A549 than their free-base precursors. Our results strongly suggest that incorporation of Zn(II) has improved the antiviral and anticancer properties of the nitroporphyrins. To the best of our knowledge, this is the first report demonstrating the dual activity of nitroporphyrin-zinc complexes as antiviral and anti-cancer, which will aid in their development as therapeutics in clinics.


Antineoplastic Agents/pharmacology , HIV Fusion Inhibitors/pharmacology , Metalloporphyrins/pharmacology , Photosensitizing Agents/pharmacology , Zinc/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/radiation effects , Antineoplastic Agents/toxicity , CHO Cells , Cell Line, Tumor , Cricetulus , Fluorescence , HEK293 Cells , HIV Fusion Inhibitors/chemical synthesis , HIV Fusion Inhibitors/radiation effects , HIV Fusion Inhibitors/toxicity , HIV-1/drug effects , Humans , Light , Metalloporphyrins/chemical synthesis , Metalloporphyrins/radiation effects , Metalloporphyrins/toxicity , Molecular Structure , Nitrobenzenes/chemical synthesis , Nitrobenzenes/pharmacology , Nitrobenzenes/radiation effects , Nitrobenzenes/toxicity , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/radiation effects , Photosensitizing Agents/toxicity
4.
Cancer Chemother Pharmacol ; 80(2): 421-431, 2017 Aug.
Article En | MEDLINE | ID: mdl-28685347

PURPOSE: Manganese porphyrins are redox-active drugs and superoxide dismutase mimics, which have been shown to chemosensitize lymphoma, a cancer which frequently occurs in dogs. This study aimed to identify critical information regarding the pharmacokinetics and toxicity of Mn(III) meso-tetrakis (N-n-butoxyetylpyridium-2-yl) porphyrin, (MnTnBuOE-2-PyP5+, MnBuOE) in dogs as a prelude to a clinical trial in canine lymphoma patients. METHODS: A single-dose pharmacokinetic (PK) study in normal dogs was performed to determine the plasma half-life (t 1/2) of MnBuOE. A dose reduction study was performed to establish the maximum tolerated dose (MTD) of MnBuOE. The safety and PK of a multi-dosing protocol was assessed. RESULTS: Peak plasma drug concentration occurred 30 min post-injection. The t 1/2 was defined as 7 h. MnBuOE induced an anaphylactic reaction and prolonged tachycardia. The MTD was defined as 0.25 mg/kg. The dogs were given MTD 3×/week for 2-3 weeks. The highest recorded tissue drug levels were in the lymph nodes (4-6 µM), followed by kidney and liver (2.5, 2.0 uM, respectively). CONCLUSIONS: We obtained critical information regarding the PK and toxicity of MnBuOE in dogs. The acute drug reaction and tachycardia post-injection have not been described in other species and may be specific to canines. The high tissue drug levels in lymph nodes have not been previously reported. MnBuOE accumulation in lymph nodes has important implications for the utility of adjuvant MnBuOE to treat lymphoma. With MnBuOE lymph node accumulation, reduction in the dose and/or administration frequency could be possible, leading to reduced toxicity.


Antineoplastic Agents/administration & dosage , Kidney/metabolism , Liver/metabolism , Lymph Nodes/metabolism , Metalloporphyrins/administration & dosage , Anaphylaxis/chemically induced , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Dog Diseases/drug therapy , Dogs , Half-Life , Lymphoma/drug therapy , Lymphoma/veterinary , Male , Maximum Tolerated Dose , Metalloporphyrins/pharmacokinetics , Metalloporphyrins/toxicity , Species Specificity , Tachycardia/chemically induced , Tissue Distribution
5.
J Photochem Photobiol B ; 173: 412-422, 2017 Aug.
Article En | MEDLINE | ID: mdl-28662468

A longstanding obstacle to cancer eradication centers on the heterogeneous nature of the tissue that manifests it. Variations between cancer cell resistance profiles often result in a survival percentage following classic therapeutics. As an alternative, photodynamic therapys' (PDT) unique non-specific cell damage mechanism and high degree of application control enables it to potentially deliver an efficient treatment regime to a broad range of heterogeneous tissue types thereby overcoming individual resistance profiles. This study follows on from previous design, characterization and solubility analyses of three novel carbohydrate-ligated zinc-porphyrazine (Zn(II)Pz) derivatives. Here we report on their PDT application potential in the treatment of five common cancer tissue types in vitro. Following analyses of metabolic homeostasis, toxicity and cell death induction, overall Zn(II)Pz-PDT proved comparably efficient between all cancer tissue populations. Differential localization patterns of Zn(II)Pz derivatives between cell types did not appear to influence the overall PDT effect. All cell types exhibited significant disruptions to mitochondrial activity and associated ATP production levels. Toxicity and chromatin structure profiles revealed indiscernible patterns of damage between Zn(II)Pz derivatives and cell type. The subtle differences observed between individual Zn(II)Pz derivatives is most likely due to a combination of carbohydrate moiety characteristics on energy transfer processes and associated dosage optimization requirements per tissue type. Collectively, this indicates that resistance profiles are negated to a significant extent by Zn(II)Pz-PDT making these derivatives attractive candidates for PDT applications across multiple tissue types and subtypes.


Carbohydrates/chemistry , Cell Proliferation/drug effects , Metalloporphyrins/chemistry , Photosensitizing Agents/toxicity , A549 Cells , Adenosine Triphosphate/metabolism , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Cell Proliferation/radiation effects , Chromatin/chemistry , Chromatin/drug effects , Humans , Light , MCF-7 Cells , Metalloporphyrins/therapeutic use , Metalloporphyrins/toxicity , Microscopy, Fluorescence , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasms/drug therapy , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Zinc/chemistry
6.
Int J Toxicol ; 35(4): 438-53, 2016 07.
Article En | MEDLINE | ID: mdl-27098749

BMX-001, a manganese porphyrin that has anti-inflammatory, antioxidant, and antitumor properties, is being developed as a potential therapeutic for high-grade glioma (HGG) and head and neck (H&N) cancer. An IND has been opened for BMX-001 in the treatment of HGG (NCT02655601) and another is in preparation for H&N. The safety of BMX-001 has been evaluated in a battery of nonclinical Good Laboratory Practice (GLP)-compliant studies. Systemic toxicity has been evaluated using the intended cGMP product administered subcutaneously for periods of up to 5 weeks in both the mouse and the monkey and included toxicokinetic evaluations to characterize systemic exposure and tissue distribution and clearance of BMX-001. In additional GLP studies, BMX-001 was not irritating to the skin or eye and caused no changes in cardiac rate or rhythm or blood pressure. Mixed results for genotoxicity were seen with the weight of evidence indicating that BMX-001 poses no genotoxic risk in humans. In systemic mouse and monkey studies, loading/maintenance dose no observed adverse effect levels were 12/2 mg/kg/dose and 6/2 mg/kg/dose, respectively, with maintenance doses administered every 3 days after the initial loading dose. Systemic data were used to determine a Food and Drug Administration-approved safe starting dose for the initial clinical study in patients with HGG. BMX-001 was detected in analyzed tissues, including the brain, persisting well past the short plasma clearance period. The highest levels of BMX-001 were seen in the liver and kidneys, with amounts in these tissues returning to close to undetectable levels after a 2-week cessation of dosing.


Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Metalloporphyrins/pharmacokinetics , Metalloporphyrins/toxicity , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Brain/metabolism , Eye/drug effects , Female , Guinea Pigs , Heart Rate/drug effects , Kidney/metabolism , Liver/metabolism , Macaca fascicularis , Male , Maximum Tolerated Dose , Metalloporphyrins/administration & dosage , Metalloporphyrins/blood , Mice , Mutagenicity Tests , Rabbits , Skin/drug effects
7.
Mol Nutr Food Res ; 59(9): 1690-700, 2015 Sep.
Article En | MEDLINE | ID: mdl-26047311

SCOPE: We have studied if curcumin can protect glial cells under an oxidative stress and inflammatory environment, which is known to be deleterious in neurodegeneration. METHODS AND RESULTS: Primary rat glial cultures exposed to the combination of an oxidative (rotenone/oligomycin A) and a proinflammatory LPS stimuli reduced by 50% glial viability. Under these experimental conditions, curcumin afforded significant glial protection and reduction of reactive oxygen species; these effects were blocked by the HO-1 inhibitor tin protoporphyrin-IX (SnPP). These findings correlate with the observation that curcumin induced the antioxidative protein HO-1. Most interesting was the observation that the glial protective effects related to HO-1 induction were microglial specific as shown in glial cultures from LysM(Cre) Hmox(∆/∆) mice where curcumin lost its protective effect. Under LPS conditions, curcumin reduced the microglial proinflammatory markers iNOS and tumor necrosis factor, but increased the anti-inflammatory cytokine IL4. Analysis of the microglial phenotype showed that curcumin favored a ramified morphology toward a microglial alternative activated state against LPS insult also by a HO-1-dependent mechanism. CONCLUSION: The curry constituent curcumin protects glial cells and promotes a microglial anti-inflammatory phenotype by a mechanism that implicates HO-1 induction; these effects may have impact on brain protection under oxidative and inflammatory conditions.


Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Curcumin/pharmacology , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Microglia/drug effects , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Metalloporphyrins/toxicity , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Protoporphyrins/toxicity , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
Cutan Ocul Toxicol ; 34(4): 265-70, 2015.
Article En | MEDLINE | ID: mdl-25373486

CONTEXT: Intravitreal (ITV) dosing has become a clinically important route of administration for the treatment of uveitis, endophthalmitis, retinal vein occlusion, diabetic macular edema and age-related macular degeneration. Despite this, there are no validated non-clinical models of phototoxicity for ITV products. OBJECTIVE: The objective of this study was to develop an ITV rabbit model of phototoxicity for use in assessing the photosafety of small molecules therapeutics. MATERIALS AND METHODS: Dutch Belted rabbits were intravitreally injected bilaterally with four known phototoxicants: 8-methoxypsoralen, lomefloxacin, doxycycline and stannsoporfrin. Triescence(®), a non-phototoxic triamcinolone acetonide steroid formulation designed for ITV administration, was used as a negative control. One eye was then irradiated with solar-simulated ultraviolet radiation for 30 min, 1 h after dosing, while the other eye was occluded, serving as a non-irradiated control. RESULTS: Despite the direct administration of known phototoxicants into the vitreous, no evidence of ocular phototoxicity was observed in any dose group. Direct (non-phototoxic) retinal toxicity was observed in the doxycycline dose group only. CONCLUSION: These data suggest that the posterior segment of the rabbit eye is protected against phototoxicity by anatomical and/or physiological mechanisms, and is not a useful model for the assessment of phototoxicity of intravitreally administered molecules.


Drug Evaluation, Preclinical/methods , Small Molecule Libraries/toxicity , Toxicity Tests/methods , Vitreous Body/drug effects , Animals , Doxycycline/toxicity , Fluoroquinolones/toxicity , Intravitreal Injections , Male , Metalloporphyrins/toxicity , Methoxsalen/toxicity , Rabbits
9.
Genesis ; 52(4): 309-14, 2014 Apr.
Article En | MEDLINE | ID: mdl-24616213

Photodynamic therapy utilizes light, a photosensitizer, and molecular oxygen as a treatment modality for a variety of cancers. We have recently combined ruthenium(II) polypyridyl groups with a zinc(II) centered porphyrin as a new photosensitizer for the treatment of melanoma. In-vitro studies have indicated that this photosensitizer is toxic to melanoma cells when irradiated with low energy light; however, it is nontoxic to normal cells under similar conditions. To determine the toxicity and cell viability of this compound in-vivo we present, herein, a study using Drosophila melanogaster. In the absence of light, the new photosensitizer shows no discernible effects to fly larvae at various concentrations of compound and stages of larval development. When the larvae were fed the photosensitizer it was observed, by fluorescence microscopy, that the compound passes through the cell membrane and localizes in the cytosol at lower concentrations and the nucleus at slightly higher concentrations indicating that the compound is not immediately metabolized.


Metalloporphyrins/toxicity , Photosensitizing Agents/toxicity , Ruthenium/toxicity , Zinc/toxicity , Animals , Brain/metabolism , Drosophila melanogaster , Drug Screening Assays, Antitumor , Larva/drug effects , Larva/metabolism , Metalloporphyrins/pharmacokinetics , Photochemotherapy , Photosensitizing Agents/pharmacokinetics , Ruthenium/pharmacokinetics , Zinc/pharmacokinetics
10.
Cell Stress Chaperones ; 18(6): 719-31, 2013 Nov.
Article En | MEDLINE | ID: mdl-23595218

Two porphyrins, CoTPPS and MnTMPyPCl5, were tested for their photodynamic activity and potential novel use in a therapy of human cancers. We investigated an effect of photodynamic reaction (PDR), electroporation (EP) and their combination (electro-photodynamic reaction [EP-PDR]) on human colon adenocarcinoma cell lines (LoVo and resistant to doxorubicin LoVoDX), human breast adenocarcinoma (wild type MCF-7/WT and resistant to doxorubicin MCF-7/DOX), and human melanoma (Me45). The efficiency of macromolecules transport was examined with cytofluorymetry by assessing the degree of propidium iodide (PI) penetration. Additionally, cellular ultrastructure after EP was evaluated. We determined cyto- and photo-cytotoxic effect on the cells viability (MTT assay) after standard PDR and PDR combined with EP. Intracellular distribution and mitochondrial colocalization of both porphyrins was also performed. The experiments proved that both complexes exhibit desirable photodynamic properties on LoVo LoVoDX cells, and EP effectively supports photodynamic method in this type of cancer. The application of EP provided shorter time of incubation (only 10 min) and enhanced effect of applied therapy. The porphyrins did not affect the MCF-7 and Me45 cell lines.


Coordination Complexes/toxicity , Metalloporphyrins/toxicity , Organometallic Compounds/toxicity , Oxidative Stress/drug effects , Photosensitizing Agents/toxicity , Porphyrins/toxicity , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Cell Line, Tumor , Cell Survival/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Coordination Complexes/chemistry , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Electroporation , Humans , Light , MCF-7 Cells , Metalloporphyrins/chemistry , Organometallic Compounds/chemistry , Photochemotherapy , Porphyrins/chemistry , Propidium/chemistry , Propidium/metabolism
11.
Toxicol Appl Pharmacol ; 266(3): 419-29, 2013 Feb 01.
Article En | MEDLINE | ID: mdl-23231920

In the current study C57BL/6J mice were injected intraperitoneally with Hg(2+) in the absence and presence of TCDD. After 6 and 24h the liver was harvested and the expression of Cyps was determined. In vitro, isolated hepatocytes were incubated with TCDD in the presence and absence of Hg(2+). At the in vivo level, Hg(2+) significantly decreased the TCDD-mediated induction of Cyps at 6h while potentiating their levels at 24h. In vitro, Hg(2+) significantly inhibited the TCDD-mediated induction of Cyp1a1 in a concentration- and time-dependent manner. Interestingly, Hg(2+) increased the serum hemoglobin (Hb) levels in mice treated for 24h. Upon treatment of isolated hepatocytes with Hb alone, there was an increase in the AhR-dependent luciferase activity with a subsequent increase in Cyp1a1 protein and catalytic activity levels. Importantly, when hepatocytes were treated for 2h with Hg(2+) in the presence of TCDD, then the medium was replaced with new medium containing Hb, there was potentiation of the TCDD-mediated effect. In addition, Hg(2+) increased heme oxygenase-1 (HO-1) mRNA, which coincided with a decrease in the Cyp1a1 activity level. When the competitive HO-1 inhibitor, tin mesoporphyrin was applied to the hepatocytes there was a partial restoration of Hg(2+)-mediated inhibition of Cyp1a1 activity. In conclusion, we demonstrate for the first time that there is a differential modulation of the TCDD-mediated induction of Cyp1a1 by Hg(2+) in C57BL/6J mice livers and isolated hepatocytes. Moreover, this study implicates Hb as an in vivo specific modulator of Cyp1 family.


Cytochrome P-450 CYP1A1/metabolism , Heme Oxygenase-1/metabolism , Liver/drug effects , Mercury/toxicity , Polychlorinated Dibenzodioxins/toxicity , Animals , Blotting, Western , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/genetics , Hemoglobins/metabolism , Hepatocytes/drug effects , Hepatocytes/enzymology , Hepatocytes/metabolism , Liver/enzymology , Liver/metabolism , Male , Metalloporphyrins/toxicity , Mice , Mice, Inbred C57BL , RNA/chemistry , RNA/genetics , Real-Time Polymerase Chain Reaction
12.
Angew Chem Int Ed Engl ; 51(50): 12482-5, 2012 Dec 07.
Article En | MEDLINE | ID: mdl-23139178

Highly efficient apoptotic hyperthermia is achieved using a double-effector nanoparticle that can generate reactive oxygen species (ROS) and heat. ROS render cancer cells more susceptible to subsequent heat treatment, which remarkably increases the degree of apoptotic cell death. Xenograft tumors (100 mm(3)) in mice are completely eliminated within 8 days after a single mild magnetic hyperthermia treatment at 43 °C for 30 min.


Apoptosis/drug effects , Hyperthermia, Induced , Metal Nanoparticles/toxicity , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Cell Line, Tumor , Humans , Magnetic Fields , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Metalloporphyrins/chemistry , Metalloporphyrins/therapeutic use , Metalloporphyrins/toxicity , Mice , Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Temperature , Transplantation, Heterologous
13.
Eur J Pharm Biopharm ; 81(3): 540-7, 2012 Aug.
Article En | MEDLINE | ID: mdl-22576132

We reported previously the antitumor effect of heme oxygenase-1 (HO-1) inhibition by zinc protoporphyrin IX (ZnPP). ZnPP per se is poorly water soluble and thus cannot be used as anticancer chemotherapeutic. Subsequently, we developed water-soluble micelles of ZnPP using styrene-maleic acid copolymer (SMA), which encapsulated ZnPP (SMA-ZnPP). In this report, the in vitro and in vivo therapeutic effects of SMA-ZnPP are described. In vitro experiments using 11 cultured tumor cell lines and six normal cell lines revealed a remarkable cytotoxicity of SMA-ZnPP against various tumor cells; average IC(50) is about 11.1 µM, whereas the IC(50) to various normal cells is significantly higher, that is, more than 50 µM. In the pharmacokinetic study, we found that SMA-ZnPP predominantly accumulated in the liver tissue after i.v. injection, suggesting its applicability for liver cancer. As expected, a remarkable antitumor effect was achieved in the VX-2 tumor model in the liver of rabbit that is known as one the most difficult tumor models to cure. Antitumor effect was also observed in murine tumor xenograft, that is, B16 melanoma and Meth A fibrosarcoma. Meanwhile, no apparent side effects were found even at the dose of ∼7 times higher concentration of therapeutics dose. These findings suggest a potential of SMA-ZnPP as a tool for anticancer therapy toward clinical development, whereas further investigations are warranted.


Antineoplastic Agents/pharmacology , Heme Oxygenase-1/antagonists & inhibitors , Maleates/pharmacology , Metalloporphyrins/pharmacology , Neoplasms/drug therapy , Polystyrenes/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Cell Line , Cell Line, Tumor , Chick Embryo , Chlorocebus aethiops , Female , Humans , Inhibitory Concentration 50 , Injections, Intravenous , Liver/metabolism , Male , Maleates/administration & dosage , Maleates/toxicity , Metalloporphyrins/administration & dosage , Metalloporphyrins/toxicity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Micelles , Neoplasms/pathology , Polystyrenes/administration & dosage , Polystyrenes/toxicity , Rabbits , Tissue Distribution , Xenograft Model Antitumor Assays
14.
Free Radic Biol Med ; 53(3): 464-72, 2012 Aug 01.
Article En | MEDLINE | ID: mdl-22588110

Sonodynamic therapy (SDT) with low-intensity ultrasound combined with a sonosensitizer may be a promising approach to cancer therapy. Use of ultrasound has the advantage of being noninvasive, with deep-penetration properties, and convenient because of the low or no sensitivity of sonosensitizers to light. In this study, SDT with a novel sonosensitizer (a porphyrin derivative) was evaluated in vitro and in vivo. Ultrasound irradiation with a sonosensitizer elicited potent sonotoxicity in vitro without the danger of phototoxicity. The sonotoxic effect was mediated by reactive oxygen species (ROS) and was reduced by ROS scavengers. Cell membrane lipid peroxidation increased significantly just after ultrasound irradiation with a sonosensitizer, but there was no increase in apoptosis. In an in vivo mouse xenograft model, SDT with a sonosensitizer markedly inhibited tumor cell growth. The skin hypersensitivity after light exposure was not observed in a sonosensitizer-treatment group, consistent with the in vitro findings. These results suggest that ROS generated by SDT with a sensitizer can damage tumor cells, resulting in necrosis and prevention of tumor growth. This noninvasive treatment with no adverse effects such as skin sensitivity is therefore promising for therapy of cancers located deep within patients.


Antineoplastic Agents/therapeutic use , Metalloporphyrins/therapeutic use , Neoplasms/drug therapy , Photosensitizing Agents/therapeutic use , Porphyrins/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Apoptosis , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Humans , Lipid Peroxidation , Malondialdehyde/metabolism , Metalloporphyrins/toxicity , Mice , Mice, SCID , Necrosis/chemically induced , Neoplasms/pathology , Photochemotherapy , Photosensitizing Agents/pharmacology , Photosensitizing Agents/toxicity , Porphyrins/pharmacology , Porphyrins/toxicity , Reactive Oxygen Species/metabolism , Sonication , Sound , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
Free Radic Biol Med ; 52(9): 1828-34, 2012 May 01.
Article En | MEDLINE | ID: mdl-22336516

The Mn porphyrins of k(cat)(O(2)(.-)) as high as that of a superoxide dismutase enzyme and of optimized lipophilicity have already been synthesized. Their exceptional in vivo potency is at least in part due to their ability to mimic the site and location of mitochondrial superoxide dismutase, MnSOD. MnTnHex-2-PyP(5+) is the most studied among lipophilic Mn porphyrins. It is of remarkable efficacy in animal models of oxidative stress injuries and particularly in central nervous system diseases. However, when used at high single and multiple doses it becomes toxic. The toxicity of MnTnHex-2-PyP(5+) has been in part attributed to its micellar properties, i.e., the presence of polar cationic nitrogens and hydrophobic alkyl chains. The replacement of a CH(2) group by an oxygen atom in each of the four alkyl chains was meant to disrupt the porphyrin micellar character. When such modification occurs at the end of long alkyl chains, the oxygens become heavily solvated, which leads to a significant drop in the lipophilicity of porphyrin. However, when the oxygen atoms are buried deeper within the long heptyl chains, their excessive solvation is precluded and the lipophilicity preserved. The presence of oxygens and the high lipophilicity bestow the exceptional chemical and physical properties to Mn(III) meso-tetrakis(N-n-butoxyethylpyridinium-2-yl)porphyrin, MnTnBuOE-2-PyP(5+). The high SOD-like activity is preserved and even enhanced: log k(cat)(O(2)(.-))=7.83 vs 7.48 and 7.65 for MnTnHex-2-PyP(5+) and MnTnHep-2-PyP(5+), respectively. MnTnBuOE-2-PyP(5+) was tested in an O(2)(.-) -specific in vivo assay, aerobic growth of SOD-deficient yeast, Saccharomyces cerevisiae, where it was fully protective in the range of 5-30 µM. MnTnHep-2-PyP(5+) was already toxic at 5 µM, and MnTnHex-2-PyP(5+) became toxic at 30 µM. In a mouse toxicity study, MnTnBuOE-2-PyP(5+) was several-fold less toxic than either MnTnHex-2-PyP(5+) or MnTnHep-2-PyP(5+).


Metalloporphyrins/pharmacology , Molecular Mimicry , Superoxide Dismutase/metabolism , Animals , Catalysis , Electrochemical Techniques , Male , Metalloporphyrins/toxicity , Mice , Mice, Inbred C57BL , Micelles , Oxidative Stress , Saccharomyces cerevisiae/drug effects , Spectrometry, Mass, Electrospray Ionization , Spectrophotometry, Ultraviolet
16.
Acta Biol Hung ; 62(4): 426-40, 2011 Dec.
Article En | MEDLINE | ID: mdl-22119871

In this study, the effects of a porphyrazine derivative, [octakis (2-trimethylammoniumethyl thio) porphyrazinatocobalt] octaiodide (CoPzq), were explored on the developmental characteristics, sex ratio and egg fertility of Drosophila melanogaster. CoPzq was preferred in the study due to its probability to be used as a chemotherapeutic agent in medicine. As a result of the application of CoPzq, a phenotypic abnormality was revealed within the individuals of F1 and F2 generation of Drosophila. Although individuals of F2 generation did not directly come into contact with the chemical, the rate of abnormality is high. An increase was observed within both generations in terms of the egg fertility. Besides, a deviation emerged in the sex-ratio in some concentrations.


Drosophila melanogaster/drug effects , Drosophila melanogaster/growth & development , Metalloporphyrins/toxicity , Animals , Cadmium Chloride/toxicity , Ovum/drug effects , Reproduction/drug effects , Sex Ratio
17.
J Inorg Biochem ; 105(8): 1035-43, 2011 Aug.
Article En | MEDLINE | ID: mdl-21726765

The synthesis, structural aspects, pharmacological assays, and in vitro photoinduced cytotoxic properties of [Ru(NO)(ONO)(pc)] (pc=phthalocyanine) are described. Its biological effect on the B16F10 cell line was studied in the presence and absence of visible light irradiation. At comparable irradiation levels, [Ru(NO)(ONO)(pc)] was more effective than [Ru(pc)] at inhibiting cell growth, suggesting that occurrence of nitric oxide release following singlet oxygen production upon light irradiation may be an important mechanism by which the nitrosyl ruthenium complex exhibits enhanced biological activity in cells. Following visible light activation, the [Ru(NO)(ONO)(pc)] complex displayed increased potency in B16F10 cells upon modifications to the photoinduced dose; indeed, enhanced potency was detected when the nitrosyl ruthenium complex was encapsulated in a drug delivery system. The liposome containing the [Ru(NO)(ONO)(pc)] complex was over 25% more active than the corresponding ruthenium complex in phosphate buffer solution. The activity of the complex was directly proportional to the ruthenium amount present inside the cell, as determined by inductively coupled plasma mass spectroscopy. Flow cytometry analysis revealed that the photocytotoxic activity was mainly due to apoptosis. Furthermore, the vasorelaxation induced by [Ru(NO)(ONO)(pc)], proposed as NO carrier, was studied in rat isolated aorta. The observed vasodilation was concentration-dependent. Taken together, the present findings demonstrate that the [Ru(NO)(ONO)(pc)] complex induces vascular relaxation and could be a potent anti-tumor agent. Nitric oxide release following singlet oxygen production upon visible light irradiation on a nitrosyl ruthenium complex produces two radicals and may elicit phototoxic responses that may find useful applications in photodynamic therapy.


Metalloporphyrins/chemistry , Metalloporphyrins/toxicity , Nitric Oxide/biosynthesis , Organometallic Compounds/chemistry , Organometallic Compounds/toxicity , Singlet Oxygen/metabolism , Animals , Aorta/drug effects , Aorta/metabolism , Cells, Cultured , Flow Cytometry , Male , Mice , Rats , Rats, Wistar , Vasodilation/drug effects
18.
Dalton Trans ; 40(16): 4111-21, 2011 Apr 28.
Article En | MEDLINE | ID: mdl-21384047

Cationic Mn(III) N-alkylpyridylporphyrins (MnPs) are potent SOD mimics and peroxynitrite scavengers and diminish oxidative stress in a variety of animal models of central nervous system (CNS) injuries, cancer, radiation, diabetes, etc. Recently, properties other than antioxidant potency, such as lipophilicity, size, shape, and bulkiness, which influence the bioavailability and the toxicity of MnPs, have been addressed as they affect their in vivo efficacy and therapeutic utility. Porphyrin bearing longer alkyl substituents at pyridyl ring, MnTnHex-2-PyP(5+), is more lipophilic, thus more efficacious in vivo, particularly in CNS injuries, than the shorter alkyl-chained analog, MnTE-2-PyP(5+). Its enhanced lipophilicity allows it to accumulate in mitochondria (relative to cytosol) and to cross the blood-brain barrier to a much higher extent than MnTE-2-PyP(5+). Mn(III) N-alkylpyridylporphyrins of longer alkyl chains, however, bear micellar character, and when used at higher levels, become toxic. Recently we showed that meta isomers are ∼10-fold more lipophilic than ortho species, which enhances their cellular accumulation, and thus reportedly compensates for their somewhat inferior SOD-like activity. Herein, we modified the alkyl chains of the lipophilic meta compound, MnTnHex-3-PyP(5+) via introduction of a methoxy group, to diminish its toxicity (and/or enhance its efficacy), while maintaining high SOD-like activity and lipophilicity. We compared the lipophilic Mn(III) meso-tetrakis(N-(6'-methoxyhexyl)pyridinium-3-yl)porphyrin, MnTMOHex-3-PyP(5+), to a hydrophilic Mn(III) meso-tetrakis(N-(2'-methoxyethyl)pyridinium-3-yl)porphyrin, MnTMOE-3-PyP(5+). The compounds were characterized by uv-vis spectroscopy, mass spectrometry, elemental analysis, electrochemistry, and ability to dismute O(2)˙(-). Also, the lipophilicity was characterized by thin-layer chromatographic retention factor, R(f). The SOD-like activities and metal-centered reduction potentials for the Mn(III)P/Mn(II)P redox couple were similar-to-identical to those of N-alkylpyridyl analogs: log k(cat) = 6.78, and E(1/2) = +68 mV vs. NHE (MnTMOHex-3-PyP(5+)), and log k(cat) = 6.72, and E(1/2) = +64 mV vs. NHE (MnTMOE-3-PyP(5+)). The compounds were tested in a superoxide-specific in vivo model: aerobic growth of SOD-deficient E. coli, JI132. Both MnTMOHex-3-PyP(5+) and MnTMOE-3-PyP(5+) were more efficacious than their alkyl analogs. MnTMOE-3-PyP(5+) is further significantly more efficacious than the most explored compound in vivo, MnTE-2-PyP(5+). Such a beneficial effect of MnTMOE-3-PyP(5+) on diminished toxicity, improved efficacy and transport across the cell wall may originate from the favorable interplay of the size, length of pyridyl substituents, rotational flexibility (the ortho isomer, MnTE-2-PyP(5+), is more rigid, while MnTMOE-3-PyP(5+) is a more flexible meta isomer), bulkiness and presence of oxygen.


Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Escherichia coli/enzymology , Manganese/chemistry , Metalloporphyrins/chemistry , Metalloporphyrins/metabolism , Superoxide Dismutase/metabolism , Ascorbic Acid/metabolism , Biomimetic Materials/chemical synthesis , Biomimetic Materials/toxicity , Electrochemistry , Escherichia coli/drug effects , Hydrophobic and Hydrophilic Interactions , Isomerism , Metalloporphyrins/chemical synthesis , Metalloporphyrins/toxicity , Oxidation-Reduction , Superoxide Dismutase/deficiency
19.
Eur J Pharmacol ; 586(1-3): 1-8, 2008 May 31.
Article En | MEDLINE | ID: mdl-18395709

Peroxynitrite is well-recognized as being capable of inducing damaging cellular effects and has been identified as a mediator of cell damage in numerous disease states, including cardiovascular diseases. Metalloporphyrins are a class of molecule that represents an exciting new pharmacological approach to reducing peroxynitrite levels. These compounds catalyze the conversion of the harmful peroxynitrite molecules into less toxic derivatives and can be considered the reasonable intervention to reduce the toxicity of peroxynitrite. Several compounds have been synthesized and tested with promising results. Differences in the metalloporphyrin structure affect their reactivity. Iron-based metalloporphyrins display the highest rate of peroxynitrite decomposition with the narrowest scope of side reactions, whereas manganese-based metalloporphyrins react slower and with more pronounced secondary reactions, notably functioning as superoxide dismutase mimetics. This review examines the evidence that peroxynitrite is operative in patients with cardiovascular disease focusing on metalloporphyrin peroxynitrite decomposition catalysts and the evidence for their utility in the pharmacological treatment of major cardiovascular diseases. The data suggest that modification of peroxynitrite-induced cardiovascular injury is an intriguing and useful treatment approach.


Cardiovascular Diseases/drug therapy , Metalloporphyrins/therapeutic use , Myocardial Reperfusion Injury/drug therapy , Peroxynitrous Acid/antagonists & inhibitors , Animals , Catalysis , Free Radical Scavengers/pharmacology , Humans , Metalloporphyrins/chemistry , Metalloporphyrins/toxicity , Peroxynitrous Acid/metabolism , Peroxynitrous Acid/physiology
20.
Toxicol Lett ; 177(2): 97-107, 2008 Mar 15.
Article En | MEDLINE | ID: mdl-18289802

We investigate the cytotoxic effect of metal protoporphyrins including ferric protoporphyrin (FePP; hemin), cobalt protoporphyrin (CoPP), and tin protoporphyrin (SnPP) in glioblastoma cells C6 and GBM8401. Data of MTT assay show that FePP and CoPP, but not SnPP, significantly reduce the viability of glioma cells C6 and GBM8401 in the absence of serum. In the condition with fetal bovine serum (FBS) or bovine serum albumin (BSA), the cytotoxic effect of FePP and CoPP was completely inhibited. Binding of FePP, CoPP, and SnPP with BSA was examined via spectrophotometer analysis, and the protective effect of serum against FePP and CoPP-induced cell death was abolished by BSA depletion. A loss in the integrity of DNA with an occurrence of apoptotic events including DNA ladders, caspase 3 and PARP protein cleavage, and chromatin-condensed cells is observed in FePP-treated or CoPP-treated C6 cells. An increase in intracellular peroxide level was examined in FePP, but not CoPP, -treated C6 cells, and N-acetyl-l-cysteine (NAC) addition significantly protected C6 cells from FePP, but not CoPP, -induced cell death with reducing FePP-stimulated reactive oxygen species (ROS) production. Activation of extracellular regulated kinases (ERKs) and c-Jun-N-terminal kinases (JNKs) with an increase in the heme oxygenase-1 (HO-1) protein was observed in FePP-treated or CoPP-treated C6 cells in the absence of FBS or BSA, and adding JNKs inhibitor SP600125 (SP), but not ERKs inhibitor PD98059 (PD), significantly attenuated FePP-induced or CoPP-induced HO-1 protein expression in accordance with reducing JNKs protein phosphorylation. However, PD98059, SP600125, or transfection of C6 cells with antisense HO-1 oligonucleotides show no effect on the cytotoxicity elicited by FePP and CoPP in C6 cells. Effect of serum and BSA on the cytotoxicity of metal protoporphyrins in glioma cells is first demonstrated in the present study, and the roles of ROS, MAPKs, and HO-1 were elucidated.


Albumins/metabolism , Apoptosis , Heme Oxygenase-1/metabolism , Metalloporphyrins/toxicity , Protoporphyrins/toxicity , Reactive Oxygen Species/metabolism , Animals , Cattle , Cell Line, Tumor , Cell Survival/drug effects , Chromatin/metabolism , DNA/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Glioblastoma/metabolism , MAP Kinase Kinase 4/metabolism , Metalloporphyrins/antagonists & inhibitors , Metalloporphyrins/metabolism , Phosphorylation , Protoporphyrins/antagonists & inhibitors , Protoporphyrins/metabolism , Rats , Serum Albumin, Bovine/metabolism , Spectrometry, Fluorescence
...