Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 152
1.
J Pharmacol Exp Ther ; 377(1): 1-10, 2021 04.
Article En | MEDLINE | ID: mdl-33500265

Oxidative stress plays a crucial role in the pathogenesis of Parkinson disease (PD), and one strategy for neuroprotective therapy for PD is to scavenge reactive species using a catalytic antioxidant. Previous studies in our laboratory revealed that pretreatment of lipophilic metalloporphyrins showed protective effects in a mouse PD model. In this study, we optimized the formulations of these metalloporphyrins to deliver them orally and tested their efficacy on disease outcomes in a second species after initiation of an insult (i.e., disease modification). In this study, a pharmaceutical formulation of two metalloporphyrin catalytic antioxidants, AEOL11207 and AEOL11114, was tested for oral drug delivery. Both compounds showed gastrointestinal absorption, achieved high plasma concentrations, and readily penetrated the blood-brain barrier after intravenous or oral delivery. AEOL11207 and AEOL11114 bioavailabilities were calculated to be 24% and 25%, respectively, at a dose of 10 mg/kg via the oral route. In addition, both compounds significantly attenuated 6-hydroxydopamine (6-OHDA)-induced neurotoxic damage, including dopamine depletion, cytokine production, and microglial activation in the striata; dopaminergic neuronal loss in the substantia nigra; oxidative/nitrative stress indices (glutathione disulfide and 3-nitrotyrosine) in the ventral midbrain; and rotation behavioral abnormality in rats. These results indicate that AEOL11207 and AEOL11114 are orally active metalloporphyrins and protect against 6-OHDA neurotoxicity 1-3 days postlesioning, suggesting disease-modifying properties and translational potential for PD. SIGNIFICANCE STATEMENT: Two catalytic antioxidants showed gastrointestinal absorption, achieved high plasma concentrations, and readily penetrated the blood-brain barrier. Both compounds significantly attenuated dopamine depletion, cytokine production, microglial activation, dopaminergic neuronal loss, oxidative/nitrative stress indices, and behavioral abnormality in a Parkinson disease rat model. The results suggest that both metalloporphyrins possess disease-modifying properties that may be useful in treating Parkinson disease.


Antioxidants/pharmacokinetics , Metalloporphyrins/pharmacokinetics , Neuroprotective Agents/pharmacokinetics , Parkinsonian Disorders/drug therapy , Administration, Oral , Animals , Antioxidants/administration & dosage , Antioxidants/therapeutic use , Blood-Brain Barrier/metabolism , Male , Metalloporphyrins/administration & dosage , Metalloporphyrins/therapeutic use , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Rats , Rats, Sprague-Dawley , Tissue Distribution
2.
Invest New Drugs ; 39(1): 89-97, 2021 02.
Article En | MEDLINE | ID: mdl-32833137

Photodynamic therapy (PDT) is gradually becoming an alternative method in the treatment of several diseases. Here, we investigated the role of oxygen in photodynamically treated cervical cancer cells (HeLa). The effect of PDT on HeLa cells was assessed by exposing cultured cells to disulphonated zinc phthalocyanine (ZnPcS2) and tetrasulphonated zinc tetraphenylporphyrin (ZnTPPS4). Fluorescence microscopy revealed their different localizations within the cells. ZnTPPS4 seems to be mostly limited to the cytosol and lysosomes, whereas ZnPcS2 is most likely predominantly attached to membrane structures, including plasmalemma and the mitochondrial membrane. Phototoxicity assays of PDT-treated cells carried out under different partial pressures of oxygen showed dose-dependent responses. Interestingly, ZnPcS2 was also photodynamically effective at a minimal level of oxygen, under a nitrogen atmosphere. On the other hand, hyperbaric oxygenation did not lead to a higher PDT efficiency of either photosensitizer. Although both photosensitizers can induce a significant drop in mitochondrial membrane potential, ZnPcS2 has a markedly higher effect on mitochondrial respiration that was completely blocked after two short light cycles. In conclusion, our observations suggest that PDT can be effective even in hypoxic conditions if a suitable sensitizer is chosen, such as ZnPcS2, which can inhibit mitochondrial respiration.


Indoles/pharmacology , Metalloporphyrins/pharmacology , Organometallic Compounds/pharmacology , Oxygen/pharmacology , Photochemotherapy/methods , Cell Survival/drug effects , Dose-Response Relationship, Drug , HeLa Cells , Humans , Indoles/administration & dosage , Membrane Potential, Mitochondrial/drug effects , Metalloporphyrins/administration & dosage , Mitochondria/drug effects , Organometallic Compounds/administration & dosage , Oxygen/administration & dosage , Partial Pressure , Photosensitizing Agents/pharmacology , Singlet Oxygen/analysis
3.
Pharm Res ; 37(11): 220, 2020 Oct 13.
Article En | MEDLINE | ID: mdl-33051728

PURPOSE: Gold porphyrin (AuP) is a complex that has been shown to be potent against various tumors. A biocompatible interpenetrating network (IPN) system comprised of polyethyleneglycol diacrylate (PEGdA) and chemically-modified gelatin has been shown to be an effective implantable drug depot to deliver AuP locally. Here we designed IPN microparticles complexed with AuP to facilitate intravenous administration and to diminish systemic toxicity. METHODS: We have synthesized and optimized an IPN microparticle formulation complexed with AuP. Tumor cell cytotoxicity, antitumor activity, and survival rate in lung cancer bearing nude mice were analyzed. RESULTS: IPN microparticles maintained AuP bioactivity against lung cancer cells (NCI-H460). In vivo study showed no observable systemic toxicity in nude mice bearing NCI-H460 xenografts after intravenous injection of 6 mg/kg AuP formulated with IPN microparticles. An anti-tumor activity level comparable to free AuP was maintained. Mice treated with 6 mg/kg AuP in IPN microparticles showed 100% survival rate while the survival rate of mice treated with free AuP was much less. Furthermore, microparticle-formulated AuP significantly reduced the intratumoral microvasculature when compared with the control. CONCLUSION: AuP in IPN microparticles can reduce the systemic toxicity of AuP without compromising its antitumor activity. This work highlighted the potential application of AuP in IPN microparticles for anticancer chemotherapy.


Angiogenesis Inhibitors/pharmacology , Gold/pharmacology , Lung Neoplasms/drug therapy , Metalloporphyrins/pharmacology , Administration, Intravenous , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/chemistry , Animals , Cell Line, Tumor , Drug Compounding , Gold/administration & dosage , Gold/chemistry , Humans , Lung Neoplasms/pathology , Metalloporphyrins/administration & dosage , Metalloporphyrins/chemistry , Mice, Inbred BALB C , Mice, Nude , Particle Size , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
Respir Physiol Neurobiol ; 282: 103545, 2020 11.
Article En | MEDLINE | ID: mdl-32927098

BACKGROUND: Oxygen therapy and mechanical ventilation are important predisposing factors for the development of bronchopulmonary dysplasia (BPD), leading to increased morbidity and mortality in premature infants. Oxygen toxicity mediated by reactive oxygen species (ROS) may play an important part in the development of BPD. We studied the effects of MnTBAP, a catalytic antioxidant on airway responsiveness and alveolar simplification in adult mice following neonatal hyperoxia. METHODS: Mice litters were randomized to 85 %O2 or room air (RA) on D3 for 12 days to receive either MnTBAP (10 mg/kg/d) or saline intraperitoneally. Methacholine challenge (MCC) performed at 8 and 12 weeks of age by whole-body plethysmography to assess airway reactivity. Alveolarization quantified on lung sections by radial alveolar count (RAC) and mean linear intercept (MLI). Cell counts assessed from bronchoalveolar lavage (BAL) performed at 15 weeks. RESULTS: Mice exposed to hyperoxia and MnTBAP (OXMN) had significantly higher airway reactivity post-MCC at 8 weeks compared to RA and O2 groups. At 12 weeks, airway reactivity was higher post-MCC in both hyperoxia and OXMN groups. MnTBAP did not attenuate hyperoxia-induced airway reactivity in adult mice. Hyperoxia exposed mice demonstrated large and distended alveoli on histopathology at 2 and 15 weeks. MnTBAP did not ameliorate hyperoxia-induced lung injury as assessed by RAC/MLI. Absolute lymphocyte count was significantly higher in BAL in the hyperoxia and OXMN groups. CONCLUSIONS: MnTBAP, a catalytic antioxidant, did not afford protection from hyperoxia-induced lung injury in adult mice.


Antioxidants/pharmacology , Bronchopulmonary Dysplasia/etiology , Bronchopulmonary Dysplasia/prevention & control , Hyperoxia/complications , Metalloporphyrins/pharmacology , Animals , Animals, Newborn , Antioxidants/administration & dosage , Bronchoconstrictor Agents/administration & dosage , Disease Models, Animal , Female , Male , Metalloporphyrins/administration & dosage , Methacholine Chloride/administration & dosage , Mice , Mice, Inbred C57BL , Plethysmography, Whole Body , Pregnancy
5.
Clin J Am Soc Nephrol ; 15(5): 633-642, 2020 05 07.
Article En | MEDLINE | ID: mdl-32291269

BACKGROUND AND OBJECTIVES: Oxidative stress is a hallmark and mediator of CKD. Diminished antioxidant defenses are thought to be partly responsible. However, there is currently no way to prospectively assess antioxidant defenses in humans. Tin protoporphyrin (SnPP) induces mild, transient oxidant stress in mice, triggering increased expression of select antioxidant proteins (e.g., heme oxygenase 1 [HO-1], NAD[P]H dehydrogenase [quinone] 1 [NQO1], ferritin, p21). Hence, we tested the hypothesis that SnPP can also variably increase these proteins in humans and can thus serve as a pharmacologic "stress test" for gauging gene responsiveness and antioxidant reserves. DESIGN: , setting, participants, & measurementsA total of 18 healthy volunteers and 24 participants with stage 3 CKD (n=12; eGFR 30-59 ml/min per 1.73 m2) or stage 4 CKD (n=12; eGFR 15-29 ml/min per 1.73 m2) were injected once with SnPP (9, 27, or 90 mg). Plasma and/or urinary antioxidant proteins were measured at baseline and for up to 4 days post-SnPP dosing. Kidney safety was gauged by serial measurements of BUN, creatinine, eGFR, albuminuria, and four urinary AKI biomarkers (kidney injury molecule 1, neutrophil gelatinase-associated lipocalin, cystatin C, and N-acetyl glucosaminidase). RESULTS: Plasma HO-1, ferritin, p21, and NQO1 were all elevated at baseline in CKD participants. Plasma HO-1 and urine NQO1 levels each inversely correlated with eGFR (r=-0.85 to -0.95). All four proteins manifested statistically significant dose- and time-dependent elevations after SnPP injection. However, marked intersubject differences were observed. p21 responses to high-dose SnPP and HO-1 responses to low-dose SnPP were significantly suppressed in participants with CKD versus healthy volunteers. SnPP was well tolerated by all participants, and no evidence of nephrotoxicity was observed. CONCLUSIONS: SnPP can be safely administered and, after its injection, the resulting changes in plasma HO-1, NQO1, ferritin, and p21 concentrations can provide information as to antioxidant gene responsiveness/reserves in subjects with and without kidney disease. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER: A Study with RBT-1, in Healthy Volunteers and Subjects with Stage 3-4 Chronic Kidney Disease, NCT0363002 and NCT03893799.


Kidney Function Tests , Metalloporphyrins/administration & dosage , Oxidative Stress , Protoporphyrins/administration & dosage , Renal Insufficiency, Chronic/diagnosis , Adult , Aged , Biomarkers/blood , Biomarkers/urine , Case-Control Studies , Cyclin-Dependent Kinase Inhibitor p21/blood , Cyclin-Dependent Kinase Inhibitor p21/urine , Female , Ferritins/blood , Ferritins/urine , Glomerular Filtration Rate , Heme Oxygenase-1/blood , Heme Oxygenase-1/urine , Humans , Infusions, Intravenous , Male , Middle Aged , NAD(P)H Dehydrogenase (Quinone)/blood , NAD(P)H Dehydrogenase (Quinone)/urine , Predictive Value of Tests , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/physiopathology , Renal Insufficiency, Chronic/urine
6.
Proc Natl Acad Sci U S A ; 117(13): 7021-7029, 2020 03 31.
Article En | MEDLINE | ID: mdl-32179677

Described here is the development of gadolinium(III) texaphyrin-platinum(IV) conjugates capable of overcoming platinum resistance by 1) localizing to solid tumors, 2) promoting enhanced cancer cell uptake, and 3) reactivating p53 in platinum-resistant models. Side by side comparative studies of these Pt(IV) conjugates to clinically approved platinum(II) agents and previously reported platinum(II)-texaphyrin conjugates demonstrate that the present Pt(IV) conjugates are more stable against hydrolysis and nucleophilic attack. Moreover, they display high potent antiproliferative activity in vitro against human and mouse cell cancer lines. Relative to the current platinum clinical standard of care (SOC), a lead Gd(III) texaphyrin-Pt(IV) prodrug conjugate emerging from this development effort was found to be more efficacious in subcutaneous (s.c.) mouse models involving both cell-derived xenografts and platinum-resistant patient-derived xenografts. Comparative pathology studies in mice treated with equimolar doses of the lead Gd texaphyrin-Pt(IV) conjugate or the US Food and Drug Administration (FDA)-approved agent oxaliplatin revealed that the conjugate was better tolerated. Specifically, the lead could be dosed at more than three times (i.e., 70 mg/kg per dose) the tolerable dose of oxaliplatin (i.e., 4 to 6 mg/kg per dose depending on the animal model) with little to no observable adverse effects. A combination of tumor localization, redox cycling, and reversible protein binding is invoked to explain the relatively increased tolerability and enhanced anticancer activity seen in vivo. On the basis of the present studies, we conclude that metallotexaphyrin-Pt conjugates may have substantial clinical potential as antitumor agents.


Antineoplastic Agents/administration & dosage , Metalloporphyrins/administration & dosage , Oxaliplatin/administration & dosage , A549 Cells , Animals , Antineoplastic Agents/pharmacokinetics , Drug Resistance, Neoplasm , Female , HCT116 Cells , Humans , Metalloporphyrins/pharmacokinetics , Mice, Nude , Oxaliplatin/pharmacokinetics , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Tissue Distribution , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
7.
Naunyn Schmiedebergs Arch Pharmacol ; 392(11): 1435-1445, 2019 11.
Article En | MEDLINE | ID: mdl-31273394

Oxidative stress and persistent inflammation play crucial role in the progression of diabetic wound complications. Hemeoxgenase-1 (HO-1) by degrading hemin has been shown to display anti-oxidant and anti-inflammatory effects. Further, hemin is a potent HO-1 inducer. Thus, the current study was aimed to evaluate the effect of topical application of hemin on diabetic wound in rats. Four hundred square millimeter open excision wound were created 2 weeks after induction of diabetes with single intraperitoneal injection of streptozotocin (60 mg/kg), and the diabetic rats were divided into three groups namely diabetic control, hemin, and tin protoporphyrin (SnPPIX). Ointment base, hemin (0.5% in ointment base), and SnPPIX (0.5% in ointment base) were applied topically to wounded area in diabetic control, hemin, and SnPPIX group rats, respectively, twice daily for 19 days. Hemin significantly increased the wound contraction in comparison to control and SnPPIX-treated rats. Time-dependent analysis revealed significant increase in anti-oxidants with concomitant decrease in oxidants in hemin-treated rats as compared to diabetic control rats. Further, mRNA expression decreased for inflammatory cytokine and increased for anti-inflammatory cytokine in hemin group as compared to diabetic control rats. Expression of HO-1 also increased in hemin group as compared to diabetic control rats. However, SnPPIX group results were in disagreement with results of hemin which is clearly reflected in histopathology. Results indicate the ability of hemin to accelerate wound healing in diabetic rats by combating inflammation and oxidative stress probably via HO-1.


Antioxidants/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Hemin/pharmacology , Inflammation Mediators/metabolism , Oxidative Stress/drug effects , Wound Healing/drug effects , Administration, Cutaneous , Animals , Antioxidants/administration & dosage , Blood Glucose/analysis , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase (Decyclizing)/biosynthesis , Hemin/administration & dosage , Male , Metalloporphyrins/administration & dosage , Metalloporphyrins/pharmacology , Ointments , Oxidative Stress/immunology , Protoporphyrins/administration & dosage , Protoporphyrins/pharmacology , Rats , Skin/drug effects , Skin/immunology , Skin/injuries , Streptozocin , Wound Healing/immunology
8.
Ocul Surf ; 17(2): 257-264, 2019 04.
Article En | MEDLINE | ID: mdl-30807830

PURPOSE: To determine the efficacy of the superoxide dismutase mimetic, manganese(III) tetrakis(1-methyl-4-pyridyl) porphyrin (Mn-TM-2-PyP), in vitro in human corneal epithelial (HCE-T) cells and in vivo in a preclinical mouse model for dry-eye disease (DED). METHODS: In vitro, HCE-T cultures were exposed either to tert-butylhydroperoxide (tBHP) to generate oxidative stress or to hyperosmolar conditions modeling cellular stress during DED. Cells were pre-treated with Mn-TM-2-PyP or vehicle. Mn-TM-2-PyP permeability across stratified HCE-T cells was assayed. In vivo, Mn-TM-2-PyP (0.1% w/v in saline) was delivered topically as eye drops in a desiccating stress/scopolamine model for DED. Preclinical efficacy was compared to untreated, vehicle- and ophthalmic cyclosporine emulsion-treated mice. RESULTS: Mn-TM-2-PyP protected HCE-T cells in a dose-dependent manner against tBHP-induced oxidative stress as determined by calculating the IC50 for tBHP in the resazurin, MTT and lactate dehydrogenase release cell viability assays. Mn-TM-2-PyP did not protect HCE-T cells from hyperosmolar insult. Its permeability coefficient across a barrier of HCE-T cells was 1.1 ±â€¯0.05 × 10-6 cm/s and the mass balance was 62 ±â€¯0.6%. In vivo, topical dosing with Mn-TM-2-PyP resulted in a statistically significant reduction of corneal fluorescein staining, similar to ophthalmic cyclosporine emulsion. Furthermore, Mn-TM-2-PyP significantly reduced leukocyte infiltration into lacrimal glands and prevented degeneration of parenchymal tissue. No protective effect against loss of conjunctival goblet cells was observed. Notably, Mn-TM-2-PyP did not produce ocular toxicity when administered topically. DISCUSSION: Our data suggest that Mn-TM-2-PyP, a prototypic synthetic metalloporphyrin compound with potent catalytic antioxidant activity, can improve signs of DED in vivo by reducing oxidative stress in corneal epithelial cells.


Dry Eye Syndromes/drug therapy , Goblet Cells/pathology , Metalloporphyrins/administration & dosage , Oxidative Stress , Animals , Antioxidants , Cell Count , Disease Models, Animal , Dry Eye Syndromes/metabolism , Dry Eye Syndromes/pathology , Goblet Cells/drug effects , Humans , Mice , Mice, Inbred C57BL , Ophthalmic Solutions/administration & dosage , Severity of Illness Index
9.
Redox Biol ; 20: 307-320, 2019 01.
Article En | MEDLINE | ID: mdl-30390545

Negative Pressure Wound Therapy (NPWT), a widely used modality in the management of surgical and trauma wounds, offers clear benefits over conventional wound healing strategies. Despite the wide-ranging effects ascribed to NPWT, the precise molecular mechanisms underlying the accelerated healing supported by NPWT remains poorly understood. Notably, cellular redox status-a product of the balance between cellular reactive oxygen species (ROS) production and anti-oxidant defense systems-plays an important role in wound healing and dysregulation of redox homeostasis has a profound effect on wound healing. Here we investigated potential links between the use of NPWT and the regulation of antioxidant mechanisms. Using patient samples and a rodent model of acute injury, we observed a significant accumulation of MnSOD protein as well as higher enzymatic activity in tissues upon NPWT. As a proof of concept and to outline the important role of SOD activity in wound healing, we replaced NPWT by the topical application of a MnSOD mimetic, Mn(III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin (MnTE-2-PyP5+, MnE, BMX-010, AEOl10113) in the rodent model. We observed that MnE is a potent wound healing enhancer as it appears to facilitate the formation of new tissue within the wound bed and consequently advances wound closure by two days, compared to the non-treated animals. Taken together, these results show for the first time a link between NPWT and regulation of antioxidant mechanism through the maintenance of MnSOD activity. Additionally this discovery outlined the potential role of MnSOD mimetics as topical agents enhancing wound healing.


Negative-Pressure Wound Therapy , Superoxide Dismutase/metabolism , Wound Healing , Administration, Topical , Animals , Antioxidants/metabolism , Biological Mimicry , Biomimetics , Combined Modality Therapy , Disease Management , Disease Models, Animal , Enzyme Activation , Humans , Metalloporphyrins/administration & dosage , Rats , Superoxide Dismutase/administration & dosage , Treatment Outcome , Wound Healing/drug effects
10.
J Biomed Mater Res A ; 106(11): 2955-2962, 2018 11.
Article En | MEDLINE | ID: mdl-30260555

To enhance anticancer efficacy and decrease side effects, the synergistic multi-agent therapy has now increasingly gotten great attention. Herein, the flexible polycaprolactone had been modified by histidine and formed assembles with PEGylated metallo-porphyrin via metal-coordinated supramolecular interaction. This supramolecular assembles showed excellent acid sensitivity. At neutral environment, the hydrophobic anticancer drug could be effectively co-encapsulated with photosensitizer Fe-TPP to improve the water solubility. While at the intracellular microenvironment, the changed acid environment would trigger the drug and Fe-TPP release due to the reduction of metal-coordinated interaction between histidine and metallo-porphyrin leading to the disintegration of assembles. The in vivo anticancer experiments toward HeLa and MCF-7 cells disclosed that this co-delivery system of anticancer drug and photosensitizer showed enhanced anticancer efficacy, implying that the synergistic chemo-photodynamic therapy could improve cellular proliferation inhibition. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 2955-2962, 2018.


Antineoplastic Agents/administration & dosage , Delayed-Action Preparations/chemistry , Doxorubicin/administration & dosage , Metalloporphyrins/administration & dosage , Photosensitizing Agents/administration & dosage , Polyesters/chemistry , Acids/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Coordination Complexes/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , HeLa Cells , Histidine/analogs & derivatives , Humans , Hydrogen-Ion Concentration , MCF-7 Cells , Metalloporphyrins/chemistry , Metalloporphyrins/pharmacokinetics , Metalloporphyrins/pharmacology , Neoplasms/drug therapy , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/pharmacology , Polyethylene Glycols/chemistry
11.
Cancer Sci ; 109(5): 1330-1345, 2018 May.
Article En | MEDLINE | ID: mdl-29575297

Sonodynamic therapy (SDT), which is based on photodynamic therapy (PDT), is a new cancer treatment modality. Unlike PDT, which has poor tissue penetration, ultrasound can penetrate deeply into tissues and largely target tumor tissue to mediate the cytotoxicity of sonosensitizers. We hypothesize that, similar to PDT, SDT may perform effectively as a cancer vaccine. Thus, we developed a therapeutic strategy to explore whether SDT can eliminate primary tumors, inhibit metastases, and prevent tumor relapse. In the present study, we found that HiPorfin (HPD)-induced SDT killed tumor cells, promoted calreticulin expression on the cell surface and elicited immune responses. Meanwhile, we observed that SDT induced functional antitumor vaccination and abscopal effects in H22 tumor-bearing mice. Furthermore, this strategy conferred an immunological memory, which could protect against tumor recurrence after the elimination of the initial tumor. These results showed important effects of SDT on immune responses.


Calreticulin/metabolism , Liver Neoplasms/therapy , Metalloporphyrins/administration & dosage , Ultrasonic Therapy/methods , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Combined Modality Therapy , Humans , Immunotherapy , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Metalloporphyrins/pharmacology , Mice , Photochemotherapy , Photosensitizing Agents
12.
J Colloid Interface Sci ; 522: 163-173, 2018 Jul 15.
Article En | MEDLINE | ID: mdl-29601958

We designed novel polymer-free cubic bicontinuous liquid crystalline dispersions (cubosomes) using monoolein as molecular building block, phospholipids as stabilizers, propylene glycol as hydrotrope. Their kinetic stability was evaluated by analysing the backscattering profiles upon ageing, and the most stable formulation was chosen as potential photosensitizers delivery vehicle for photodynamic therapy (PDT) of human skin melanoma cells. Morphological and topological features of such formulation alternatively loaded with Chlorin e6 or meso-Tetraphenylporphine-Mn(III) chloride photosensitizing dyes were investigated by cryo-TEM, DLS, and SAXS. Bioimaging studies demonstrated that Me45 and MeWo cell lines effectively internalized these cubosomes formulations. Particularly, photodynamic activity experiments proved both the very low cytotoxicity of the cubosomes formulation loaded with Chlorin e6 dye in the "dark" condition, and its significant cytotoxic effect after photoirradiation. The toxic effect recorded when the photosensitizer was encapsulated within the cubosomes was shown to be one order of magnitude higher than that caused by the free photosensitizer. This is the first report of biocompatible polymer-free cubosomes for potential application in both PDT and bioimaging of skin malignant melanoma.


Liquid Crystals/chemistry , Melanoma/diagnostic imaging , Melanoma/therapy , Photosensitizing Agents/chemistry , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/therapy , Cell Line, Tumor , Cell Proliferation , Cell Survival , Chlorophyllides , Drug Carriers/chemistry , Glycerides/chemistry , Humans , Kinetics , Manganese , Metalloporphyrins/administration & dosage , Metalloporphyrins/chemistry , Optical Imaging , Particle Size , Photochemotherapy , Photosensitizing Agents/administration & dosage , Porphyrins/administration & dosage , Porphyrins/chemistry , Propylene Glycol/chemistry , Surface Properties , Melanoma, Cutaneous Malignant
13.
Nano Lett ; 17(11): 6916-6921, 2017 11 08.
Article En | MEDLINE | ID: mdl-29019240

Structurally controlled nanoparticles, such as core-shell nanocomposite particles by combining two or more compositions, possess enhanced or new functionalities that benefited from the synergistic coupling of the two components. Here we report new nanocomposite particles with self-assembled porphyrin arrays as the core surrounded by amorphous silica as the shell. The synthesis of such nanocomposite nanoparticles was conducted through a combined surfactant micelle confined self-assembly and silicate sol-gel process using optically active porphyrin as a functional building block. Depending on kinetic conditions, these particles exhibit structure and function at multiple length scales and locations. At the molecular scale, the porphyrins as the building blocks provide well-defined macromolecular structures for noncovalent self-assembly and unique chemistry for high-yield generation of singlet oxygen for photodynamic therapy (PDT). On the nanoscale, controlled noncovalent interactions of the porphyrin building block result in an extensive self-assembled porphyrin network that enables efficient energy transfer and impressive fluorescence for cell labeling, evidenced by absorption and photoluminescence spectra. Finally, the thin silicate shell on the nanoparticle surface allows easy functionalization, and the resultant targeting porphyrin-silica nanocomposites can selectively destroy tumor cells upon receiving light irradiation.


Delayed-Action Preparations/chemistry , Metalloporphyrins/administration & dosage , Nanocomposites/chemistry , Photosensitizing Agents/administration & dosage , Silicon Dioxide/chemistry , Energy Transfer , HeLa Cells , Humans , Hydrogen-Ion Concentration , Metalloporphyrins/chemistry , Metalloporphyrins/pharmacology , Nanocomposites/ultrastructure , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Singlet Oxygen/chemistry
14.
Angew Chem Int Ed Engl ; 56(44): 13752-13756, 2017 10 23.
Article En | MEDLINE | ID: mdl-28856780

Photodynamic therapy (PDT) has emerged as an important minimally invasive tumor treatment technology. The search for an effective photosensitizer to realize selective cancer treatment has become one of the major foci in recent developments of PDT technology. Controllable singlet-oxygen release based on specific cancer-associated events, as another major layer of selectivity mode, has attracted great attention in recent years. Here, for the first time, we demonstrated that a novel mixed-metal metal-organic framework nanoparticle (MOF NP) photosensitizer can be activated by a hydrogen sulfide (H2 S) signaling molecule in a specific tumor microenvironment for PDT against cancer with controllable singlet-oxygen release in living cells. The effective removal of tumors in vivo further confirmed the satisfactory treatment effect of the MOF NP photosensitizer.


Hydrogen Sulfide/metabolism , Metalloporphyrins/therapeutic use , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Photosensitizing Agents/therapeutic use , Singlet Oxygen/metabolism , Animals , Cell Line, Tumor , HCT116 Cells , Hep G2 Cells , Humans , Metalloporphyrins/administration & dosage , Metalloporphyrins/metabolism , Mice, Nude , Nanoparticles/administration & dosage , Nanoparticles/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Photochemotherapy , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/metabolism , Tumor Microenvironment/drug effects
15.
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
16.
Transl Res ; 186: 1-18, 2017 08.
Article En | MEDLINE | ID: mdl-28586635

Tin protoporphyrin (SnPP), a heme oxygenase (HO) inhibitor, can paradoxically protect against diverse forms of acute kidney injury (AKI). This study sought potential underlying mechanisms. CD-1 mice received intravenous SnPP, followed 4-18 hours later by a variety of renal biochemical, histologic, and genomic assessments. Renal resistance to ischemic-reperfusion injury (IRI) was also sought. SnPP was rapidly taken up by kidney and was confined to proximal tubules. Transient suppression of renal heme synthesis (decreased δ aminolevulinic acid synthase expression), a 2.5-fold increase in "catalytic" Fe levels and oxidant stress resulted (decreased glutathione; increased malondialdehyde, and protein carbonyl content). Nrf2 nuclear translocation (∼2x Nrf2 increase; detected by enzyme-linked immunosorbent assay, Western blotting), with corresponding activation of ∼20 Nrf2-sensitive genes (RNA-Seq) were observed. By 18 hours after SnPP injection, marked protection against IRI emerged. This represented "preconditioning", not a direct SnPP effect, given that SnPP administered at the time of IRI exerted no protective effect. The importance of transient oxidant stress in SnPP "preconditioning" was exemplified by the following: (1) oxidant stress induced by a different mechanism (myoglobin injection) recapitulated SnPP's protective action; (2) GSH treatment blunted SnPP's protective influence; (3) SnPP raised cytoprotective heavy chain ferritin (Fhc), a response enhanced by exogenous Fe injection; and (4) SnCl2, a ∼35- to 50-fold HO-1 inducer (not inhibitor) evoked neither oxidant stress nor mitigated IRI (seemingly excluding HO-1 activity in SnPP's protective effect). SnPP specifically accumulates within proximal tubule cells; transient "catalytic" Fe overload and oxidative stress result; Nrf2-cytoprotective pathways are upregulated; and these changes help protect against ischemic AKI.


Acute Kidney Injury/prevention & control , Ferric Compounds/pharmacology , Glucaric Acid/pharmacology , Metalloporphyrins/pharmacology , NF-E2-Related Factor 2/metabolism , Protoporphyrins/pharmacology , Animals , Ferric Compounds/administration & dosage , Ferric Oxide, Saccharated , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Glucaric Acid/administration & dosage , Male , Metalloporphyrins/administration & dosage , Mice , NF-E2-Related Factor 2/genetics , Oxidants , Protein Binding , Protoporphyrins/administration & dosage , Reperfusion Injury
17.
Radiat Res ; 188(1): 94-104, 2017 07.
Article En | MEDLINE | ID: mdl-28517962

Normal tissue damage after head and neck radiotherapy involves a continuum of pathologic events to the mucosa, tongue and salivary glands. We examined the radioprotective effects of MnBuOE, a redox-active manganese porphyrin, at three stages of normal tissue damage: immediate (leukocyte endothelial cell [L/E] interactions), early (mucositis) and late (xerostomia and fibrosis) after treatment. In this study, mice received 0 or 9 Gy irradiation to the oral cavity and salivary glands ± MnBuOE treatment. Changes in leukocyte-endothelial cell interactions were measured 24 h postirradiation. At 11 days postirradiation, mucositis was assessed with a cathepsin-sensitive near-infrared optical probe. Stimulated saliva production was quantified at 11 weeks postirradiation. Finally, histological analyses were conducted to assess the extent of long-term effects in salivary glands at 12 weeks postirradiation. MnBuOE reduced oral mucositis, xerostomia and salivary gland fibrosis after irradiation. Additionally, although we have previously shown that MnBuOE does not interfere with tumor control at high doses when administered with radiation alone, most head and neck cancer patients will be treated with the combinations of radiotherapy and cisplatin. Therefore, we also evaluated whether MnBuOE would protect tumors against radiation and cisplatin using tumor growth delay as an endpoint. Using a range of radiation doses, we saw no evidence that MnBuOE protected tumors from radiation and cisplatin. We conclude that MnBuOE radioprotects normal tissue at both early and late time points, without compromising anti-tumor effects of radiation and cisplatin.


Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/radiotherapy , Metalloporphyrins/administration & dosage , Radiation Injuries/pathology , Radiation Injuries/prevention & control , Radiation-Protective Agents/administration & dosage , Animals , Cell Line, Tumor , Female , Mice , Mice, Inbred C57BL , Oxidation-Reduction/drug effects , Radiation Injuries/etiology , Treatment Outcome
18.
Redox Biol ; 12: 864-871, 2017 08.
Article En | MEDLINE | ID: mdl-28454069

Although radiation therapy can be effective against cancer, potential damage to normal tissues limits the amount that can be safely administered. In central nervous system (CNS), radiation damage to normal tissues is presented, in part, as suppressed hippocampal neurogenesis and impaired cognitive functions. Mn porphyrin (MnP)-based redox active drugs have demonstrated differential effects on cancer and normal tissues in experimental animals that lead to protection of normal tissues and radio- and chemo-sensitization of cancers. To test the efficacy of MnPs in CNS radioprotection, we first examined the tissue levels of three different MnPs - MnTE-2-PyP5+(MnE), MnTnHex-2-PyP5+(MnHex), and MnTnBuOE-2-PyP5+(MnBuOE). Nanomolar concentrations of MnHex and MnBuOE were detected in various brain regions after daily subcutaneous administration, and MnBuOE was well tolerated at a daily dose of 3mg/kg. Administration of MnBuOE for one week before cranial irradiation and continued for one week afterwards supported production and long-term survival of newborn neurons in the hippocampal dentate gyrus. MnP-driven S-glutathionylation in cortex and hippocampus showed differential responses to MnP administration and radiation in these two brain regions. A better understanding of how preserved hippocampal neurogenesis correlates with cognitive functions following cranial irradiation will be helpful in designing better MnP-based radioprotection strategies.


Central Nervous System/chemistry , Cranial Irradiation/adverse effects , Metalloporphyrins/administration & dosage , Neurogenesis/drug effects , Radiation-Protective Agents/administration & dosage , Animals , Biological Availability , Central Nervous System/drug effects , Central Nervous System/radiation effects , Cerebral Cortex/chemistry , Cerebral Cortex/drug effects , Cerebral Cortex/radiation effects , Female , Hippocampus/chemistry , Hippocampus/drug effects , Hippocampus/radiation effects , Male , Metalloporphyrins/pharmacokinetics , Mice, Inbred C57BL , Neurogenesis/radiation effects , Oxidative Stress/drug effects , Radiation-Protective Agents/pharmacokinetics
19.
Toxicol Appl Pharmacol ; 326: 34-42, 2017 07 01.
Article En | MEDLINE | ID: mdl-28400118

Reactive oxygen species are a well-defined therapeutic target for Parkinson's disease (PD) and pharmacological agents that catalytically scavenge reactive species are promising neuroprotective strategies for treatment. Metalloporphyrins are synthetic catalytic antioxidants that mimic the body's own antioxidant enzymes i.e. superoxide dismutases and catalase. The goal of this study was to determine if newly designed metalloporphyrins have enhanced pharmacodynamics including oral bioavailability, longer plasma elimination half-lives, penetrate the blood brain barrier, and show promise for PD treatment. Three metalloporphyrins (AEOL 11216, AEOL 11203 and AEOL 11114) were identified in this study as potential candidates for further pre-clinical development. Each of these compounds demonstrated blood brain barrier permeability by the i.p. route and two of three compounds (AEOL 11203 and AEOL 11114) were orally bioavailable. All of these compounds protected against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity, including dopamine depletion in the striatum, dopaminergic neuronal loss in the substantial nigra, and increased oxidative/nitrative stress indices (glutathione disulfide and 3-nitrotyrosine) in the ventral midbrain of the mice without inhibiting MPTP metabolism. Daily therapeutic dosing of these metalloporphyrins were well tolerated without accumulation of brain manganese levels or behavioral alterations assessed by open field and rotarod tests. The study identified two orally active metalloporphyrins and one injectable metalloporphyrin as clinical candidates for further development in PD.


Antioxidants/pharmacology , Antiparkinson Agents/pharmacology , Brain/drug effects , MPTP Poisoning/prevention & control , Metalloporphyrins/pharmacology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Administration, Oral , Animals , Antioxidants/administration & dosage , Antioxidants/pharmacokinetics , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/pharmacokinetics , Behavior, Animal/drug effects , Biological Availability , Biomarkers/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Brain/physiopathology , Capillary Permeability , Disease Models, Animal , Dopamine/metabolism , Drug Design , Drug Evaluation, Preclinical , Half-Life , Injections, Intraperitoneal , MPTP Poisoning/etiology , MPTP Poisoning/metabolism , MPTP Poisoning/physiopathology , Male , Metalloporphyrins/administration & dosage , Metalloporphyrins/pharmacokinetics , Mice, Inbred C57BL , Motor Activity/drug effects , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacokinetics , Rotarod Performance Test
20.
Antioxid Redox Signal ; 27(14): 1067-1082, 2017 Nov 10.
Article En | MEDLINE | ID: mdl-28358581

AIMS: Cationic manganese (Mn)-substituted N-pyridylporphyrin-based potent mimics of the family of superoxide dismutases (SODs) protect normal tissues from injury related to ionizing radiation (IR) by reducing levels of reactive oxygen and nitrogen species (ROS/RNS). Furthermore, Mn-porphyrins have demonstrated antitumor and radiosensitizing effects on cancer cells by promoting IR-induced tumor vasculature damage and apoptotic processes. In this study, we explored the underlying mechanisms of Mn-porphyrin-mediated tumor radiosensitization using murine mammary carcinoma 4T1 and melanoma B16 cells in vitro and in vivo. RESULTS: Combination treatment with MnTnHex-2-PyP and IR substantially reduced cell viability, clonogenic cell survival, and DNA damage repair and synergistically increased IR-induced apoptosis of 4T1 and B16 cells. MnTnHex-2-PyP in combination with IR caused a significant delay in growth of 4T1 and B16 xenograft tumors. MnTnHex-2-PyP dose-dependently enhanced IR-mediated production of H2O2-derived species, but not superoxide. Catalase overexpression reversed MnTnHex-2-PyP-enhanced ROS production and apoptosis. Demonstrated suppression of phosphorylation of several mitogen-activated protein (MAP) kinases and activation of NF-κB by MnTnHex-2-PyP/IR, which presumably inhibited activation of the antiapoptotic pathway, are in agreement with our other data on the apoptosis of cancer cells. Innovation and Conclusions: MnTnHex-2-PyP exerted a radiosensitizing effect on 4T1 and B16 tumor models in vitro and in vivo via pro-oxidative actions and therefore bears a large therapeutic potential. When combined with IR, it attenuated DNA damage repair and triggered a shift from prosurvival pathways to apoptotic cell death, likely due to increased ROS production and disturbed cellular redox balance, acting at the level of nuclear factor κB (NF-κB). Antioxid. Redox Signal. 27, 1067-1082.


Breast Neoplasms/therapy , Melanoma, Experimental/therapy , Metalloporphyrins/administration & dosage , Radiation-Sensitizing Agents/administration & dosage , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Signaling System/drug effects , Melanoma, Experimental/metabolism , Metalloporphyrins/pharmacology , Mice , Phosphorylation/drug effects , Phosphorylation/radiation effects , Radiation-Sensitizing Agents/pharmacology , Xenograft Model Antitumor Assays
...