Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 212
Filter
1.
Int J Biol Macromol ; 165(Pt A): 82-92, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-32987084

ABSTRACT

The search for alternatives to antibiotics in aquaculture has focused on the use of vaccines for immune-prophylaxis. The purpose of this study was to examine the feasibility and characteristics of chitosan-alginate microparticles for the oral delivery of immune-prophylactics to finfish. The microparticles, which incorporate fluorescent-labelled lysozyme, were produced by spray-drying method; their structural properties and uptake from the gastrointestinal tract of Tilapia (Oreochromis niloticus) were assessed by microscopy. The main findings show that the microparticles are able to retain their content in an acidic environment and to release it later in slightly alkaline conditions such as those found in the intestines. Moreover, both the microencapsulation procedure and the biopolymers used have no deleterious impact on the lysozyme lytic activity, which is maintained after the protein has been released from the microparticles. Administered in vivo in Tilapia by medicated food, the microparticles transit unaffected through the stomach, and reach the anterior intestines, in particular the villum sectum and the basal lamina of epithelial cells, 2 and 4 h after feeding. Overall, the evidence obtained here supports the potential of these chitosan-alginate microparticles as agents for oral immune-prophylaxis in the management of fish diseases.


Subject(s)
Chitosan/chemistry , Coated Materials, Biocompatible/chemistry , Tilapia/microbiology , Vaccines/pharmacology , Administration, Oral , Alginates/chemistry , Alginates/pharmacology , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/immunology , Anti-Bacterial Agents/pharmacology , Aquaculture , Chitosan/immunology , Chitosan/pharmacology , Coated Materials, Biocompatible/pharmacology , Fish Diseases/immunology , Fish Diseases/microbiology , Fish Diseases/prevention & control , Gastrointestinal Tract/drug effects , Humans , Tilapia/immunology , Vaccines/chemistry , Vaccines/immunology
2.
Folia Med (Plovdiv) ; 62(1): 165-171, 2020 03 31.
Article in English | MEDLINE | ID: mdl-32337904

ABSTRACT

BACKGROUND: The manufacture of medicinal products for human use in the European Economic Area is governed by European Directives and Regulations stipulating the relevant principles and guidelines of Good Manufacturing Practice, describing the minimum standard to be fulfilled in the production processes. AIM: To present analysis of the deficiencies reported following Good Manufacturing Practice inspections in Bulgaria in two consecutive years (2016, 2017) and to compare them with results from similar inspections reported by other EU member states. MATERIALS AND METHODS: A retrospective study was carried out by reviewing the complete Good Manufacturing Practice inspection reports of all manufacturers conducted by the Bulgarian Drug Agency in 2016 and 2017, according to relevant requirements and applicable local legislation. The items reviewed were scope of inspection, type of companies, classification of deficiencies ­ 'critical', 'major' and 'other significant deficiencies', their nature and reference to EU Good Manufacturing Practice. RESULTS: The analyzed data included 55 inspections, revealing 460 various deficiencies, of which 2 were critical and 102 ­ major. Twenty inspections were performed in 2016 vs. 35 inspections in 2017. The pattern of deficiencies was similar to the findings of other EU regulatory agencies, showing that equivalent requirements were applied. Our analysis showed that Bulgarian Drug Agency inspectors rarely raised deficiencies related to Computer Systems, Qualification/Validation, Personnel and Qualification of Suppliers unlike other EU regulators agents. CONCLUSIONS: Our analysis of Good Manufacturing Practice inspection findings in 2016 and 2017 showed that the Bulgarian Drug Agency demonstrated its ability to detect non-compliances and take necessary regulatory actions. Quality related issues constitute the main reasons for non-compliances with the requirements.


Subject(s)
Drug Compounding/standards , Drug Industry/standards , Guideline Adherence/statistics & numerical data , Bulgaria , Drug Recalls , Guidelines as Topic , Humans , Quality Control , Retrospective Studies
3.
J Biol Inorg Chem ; 20(7): 1163-73, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26369538

ABSTRACT

The ruthenium-based drug NAMI-A, characterised by its selectivity against solid tumour metastases, promotes TGF-ß1-dependent fibrosis and the reduction of the release of MMPs in the primary tumour. The aim of the study was to examine the interaction of NAMI-A with TGF-ß1 in the process of metastasis formation. NAMI-A (1) affects the secretion of TGF-ß1 in metastatic MDA-MB-231 cells rather than in non-tumorigenic HBL-100 cells, (2) prevails over TGF-ß1 with regard to the invasive capacity of the treated cells, and (3) contrasts integrin-dependent migration stimulated by TGF-ß1. It, thus, appears that the effects of NAMI-A on cell invasion and migration are best summarised as an interference with TGF-ß1 and a reduction of its activity in these events. At a molecular level, the similar activity of NAMI-A and TGF-ß1 on RhoA GTPase supports its interaction with cell surface integrins while TGF-ß1 can activate it by interaction with its TGFßR receptor. The inhibition of TGF-ß1-induced migration of MDA-MB-231 cells by NAMI-A cannot simply be attributed to a modulation of the Smad2 and p38MAPK pathways. In conclusion, the effects of NAMI-A on the biological role of TGF-ß1 in cancer metastasis are insufficient to attribute the responsibility for the anti-metastatic activity of the ruthenium-based drug to this target alone.


Subject(s)
Dimethyl Sulfoxide/analogs & derivatives , Organometallic Compounds/pharmacology , Ruthenium/pharmacology , Transforming Growth Factor beta1/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dimethyl Sulfoxide/chemistry , Dimethyl Sulfoxide/pharmacokinetics , Dimethyl Sulfoxide/pharmacology , Humans , Molecular Structure , Neoplasm Metastasis/drug therapy , Neoplasms/drug therapy , Neoplasms/physiopathology , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacokinetics , Ruthenium/chemistry , Ruthenium Compounds
4.
J Biol Inorg Chem ; 20(5): 831-40, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25982099

ABSTRACT

The ruthenium-based drug imidazolium trans-imidazoledimethylsulphoxidetetrachlorido ruthenate (NAMI-A) is a novel antitumour drug under clinical evaluation. In this study, NAMI-A is tested on aortic rings in vitro and on the systolic blood pressure in vivo with the aim of evaluating its effects on smooth muscle cells and, more in general, on the vascular system. Pre-incubation of aortic rings with 10 µM NAMI-A for 10 min potentiates the contraction induced by phenylephrine (PE). The reduction of the B max value of [(3)H]-prazosin bound to NAMI-A-treated aortic rings and the ability of NAMI-A to displace [(3)H]-prazosin and [(3)H]-IP3 binding by 25 and 42%, respectively, suggest the involvement of α1-adrenoceptor in mediating the effects on smooth muscle cells. NAMI-A also decreases the number of maximal sites of [(3)H]-prazosin bound to kidney membrane preparation from 34 to 24 fmol/mg proteins. A single i.p. dose (105 mg/kg) or a repeated treatment for 6 consecutive days (17 mg/kg/day) in Wistar rats increases the systolic blood pressure, respectively, 1 h and 3 days after treatment, and the responsiveness of rat aortic rings to PE. Atomic absorption spectroscopy confirms the presence of ruthenium in the aortic rings excised from the treated rats. These findings suggest monitoring the cardiovascular parameters when the drug is used in humans for treating cancer patients, particularly if the drug is associated with chemicals that are potentially active at the cardiovascular level.


Subject(s)
Antineoplastic Agents/pharmacology , Aorta/drug effects , Blood Pressure/drug effects , Dimethyl Sulfoxide/analogs & derivatives , Muscle Contraction/drug effects , Myocytes, Smooth Muscle/drug effects , Organometallic Compounds/pharmacology , Phenylephrine/pharmacology , Animals , Antineoplastic Agents/chemistry , Aorta/cytology , Dimethyl Sulfoxide/chemistry , Dimethyl Sulfoxide/pharmacology , Dose-Response Relationship, Drug , Male , Myocytes, Smooth Muscle/cytology , Organometallic Compounds/chemistry , Phenylephrine/chemistry , Rats , Rats, Wistar , Ruthenium Compounds , Structure-Activity Relationship
6.
Dalton Trans ; 44(4): 1905-13, 2015 Jan 28.
Article in English | MEDLINE | ID: mdl-25489765

ABSTRACT

NAMI-A is a ruthenium-based drug endowed with the unique property of selectively targeting solid tumour metastases. Although two clinical studies had already been completed, limited information exists on the behavior of NAMI-A after injection into the bloodstream. PK data in humans informs us of a rather low free drug concentration, of a relatively high half-life time of elimination and of a linear relationship between the administered dose and the corresponding AUC for up to toxic doses. In the present study, we examined the chemical kinetics of albumin binding with or without the presence of reducing agents, and we evaluated how these chemical aspects might influence the in vivo PK and the in vitro ability of NAMI-A to inhibit cell migration, which is a bona fide, rapid and easy way to suggest anti-metastatic properties. The experimental data support the binding of NAMI-A to serum albumin. The reaction is facilitated when the drug is in its reduced form and, in agreement with already reported data, the adduct formed with albumin maintains the biological activity of the ruthenium drug. The formation of the adduct is favored by low ratios of NAMI-A : HSA and by the reduction of the drug with ascorbic acid. The difference in in vivo PK and the faster binding to albumin of the reduced NAMI-A seem to suggest that the drug is not rapidly reduced immediately upon injection, even at low doses. Most probably, cell and protein binding prevail over the reduction of the drug. This observation supports the thesis that the reduction of the drug before injection must be considered relevant for the pharmacological activity of NAMI-A against tumour metastases.


Subject(s)
Antineoplastic Agents , Dimethyl Sulfoxide/analogs & derivatives , Organometallic Compounds , Serum Albumin/chemistry , Serum Albumin/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Ascorbic Acid/chemistry , Cell Adhesion/drug effects , Cell Line, Tumor , Dimethyl Sulfoxide/chemistry , Dimethyl Sulfoxide/pharmacokinetics , Dimethyl Sulfoxide/pharmacology , Humans , Kidney/metabolism , Liver/metabolism , Lung/metabolism , Male , Mice, Inbred ICR , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacokinetics , Organometallic Compounds/pharmacology , Oxidation-Reduction , Rhodamines/metabolism , Ruthenium/blood , Ruthenium/metabolism , Ruthenium Compounds
7.
J Inorg Biochem ; 106(1): 90-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22112845

ABSTRACT

The study of metal complexes for the treatment of cancer diseases has resulted in the identification of some unique properties of ruthenium-based compounds. Among these inorganic-based agents, two of them, namely the ruthenium(III) drugs NAMI-A and KP1019 have undertaken with some success the clinical evaluations of phase I and preliminary phase II trials in patients. Here we highlight the strategies that have led to the discovery of metal-based (NAMI-A and KP1019) and of organometallic (RM175, RAPTA-T, RDC11 and DW1/2) ruthenium-based complexes, and we report their main biological/pharmacological characteristics and expectations for further development.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Organometallic Compounds/therapeutic use , Ruthenium/chemistry , Animals , Antineoplastic Agents/chemistry , Cell Survival/drug effects , Clinical Trials as Topic , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Neoplasms/pathology , Organometallic Compounds/chemistry
8.
J Inorg Biochem ; 104(1): 79-86, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19906432

ABSTRACT

We have compared the organometallic arene complexes [(eta(6)-biphenyl)M(ethylenediamine)Cl](+) RM175 (M=Ru(II)) and its isostructural osmium(II) analogue AFAP51 (M=Os(II)) for their ability to induce cell detachment resistance from fibronectin, collagen IV and poly-l-lysine, and cell re-adhesion after treatment, their effects on cell migration and cell viability, on matrix metalloproteinases production, and on primary tumour growth of MCa mammary carcinoma, the effect of human serum albumin on their cytotoxicity. There are differences between ruthenium and osmium. The Os complex is up to 6x more potent than RM175 towards highly-invasive breast MDA-MB-231, human breast MCF-7 and human epithelial HBL-100 cancer cells, but whereas RM175 was active against MCa mammary carcinoma in vivo and caused metastasis reduction, AFAP51 was not. Intriguingly the presence of human serum albumin in the growth medium enhanced the cytotoxicity of both compounds. RM175 increased the resistance of MDA-MB-231 cells to detachment from substrates and both compounds inhibited the production of MMP-2. These data confirm the key role of ruthenium itself in anti-metastatic activity. It will be interesting to explore the activity of osmium arene complexes in other tumour models and the possibility of changing the non-arene ligands to tune the anticancer activity of osmium in vivo.


Subject(s)
Breast Neoplasms/drug therapy , Carcinoma/drug therapy , Organometallic Compounds/therapeutic use , Osmium/therapeutic use , Ruthenium/therapeutic use , Animals , Breast Neoplasms/pathology , Carcinoma/secondary , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Humans , Matrix Metalloproteinases/drug effects , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Organometallic Compounds/chemistry , Osmium/chemistry , Ruthenium/chemistry
9.
Met Based Drugs ; 2009: 681270, 2009.
Article in English | MEDLINE | ID: mdl-19789639

ABSTRACT

The effects of indazolium trans-[tetrachlorobis(1H-indazole)ruthenate(III)] (KP1019, or FFC14A), the second ruthenium compound that entered clinical trials, in an in vitro model of tumour invasion and metastasis show that the antitumour effects of this compound might include also the modulation of cell behaviour although its cytotoxicity appears to be predominant over these effects. The comparison with its imidazole analogue KP418 shows however its superiority, being able to control in vitro cell growth and in some instances also in vivo tumour development. These results suggest that the activity of KP1019 is predominantly due to direct cytotoxic effects for tumour cells, evident also in vivo on primary tumour growth and that the effects on modulation of the biological behaviour of the cancer cell can be present but might have only a partial role.

10.
Diabetes Metab ; 34(6 Pt 1): 587-94, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18926757

ABSTRACT

AIM: Diabetic nephropathy is the leading cause of end-stage kidney disease in developed countries and is related to chronic hyperglycaemia. The increased production and tissue deposition of advanced glycation end products (AGE) are known to play a major role in the pathogenesis of diabetic kidney damage. This study was undertaken to determine if lysozyme (LZ), microencapsulated in orally administrable chitosan-coated alginate microspheres (MS), is effective against the early changes seen in the initial stages of diabetic nephropathy. METHODS: LZ-containing MS (MSLZ) and an equivalent dose (equidose) of nonencapsulated LZ were given as oral treatments. LZ was administered to Wistar rats for seven weeks after diabetes induction with streptozotocin. RESULTS: The results showed that microencapsulated LZ treatment significantly reduced the concentration of serum AGE in the circulation and their deposition in the kidneys. Likewise, MSLZ significantly prevented the development of microalbuminuria compared with untreated diabetic rats. Furthermore, MSLZ significantly prevented the development of glomerular and renal hypertrophy as well as overexpression of AGE receptors (RAGE). An equidose of free LZ had little or no effect whatsoever. CONCLUSION: Our study supports a relationship between serum AGE and nephropathy in diabetes, and suggests that orally administered microencapsulated LZ can exert kidney-protective activity in a diabetic animal model.


Subject(s)
Diabetic Nephropathies/prevention & control , Glycation End Products, Advanced/blood , Muramidase/therapeutic use , Albuminuria , Animals , Blood Glucose , Body Weight/drug effects , Capsules , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/blood , Glycosuria , Muramidase/administration & dosage , Rats , Rats, Wistar
11.
Dalton Trans ; (13): 1267-72, 2007 Apr 07.
Article in English | MEDLINE | ID: mdl-17372640

ABSTRACT

Metastases are more decisive for tumour prognosis than primary lesions, because of their multiple locations, low accessibility to surgery and/or radiotherapy, and generally poor responsiveness to chemotherapy. The metastasis should therefore be the primary target for drug therapy. Among ruthenium complexes, NAMI-A is a leading compound that shows selective effects for solid tumour metastases related to a mechanism of action involving the inhibition of the processes of tumour invasiveness. NAMI-A opens an avenue to new perspectives in cancer chemotherapy. This includes novel compounds directed at targets selectively expressed by tumour metastases, thus reducing the typical side effects of the current metal-based drugs that are active via their unselective DNA interaction.


Subject(s)
Antineoplastic Agents/chemistry , Dimethyl Sulfoxide/analogs & derivatives , Drug Design , Neoplasm Metastasis/drug therapy , Organometallic Compounds/chemistry , Ruthenium/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Dimethyl Sulfoxide/chemistry , Dimethyl Sulfoxide/therapeutic use , Humans , Mice , Molecular Structure , Organometallic Compounds/therapeutic use , Ruthenium Compounds
12.
Leukemia ; 20(5): 785-92, 2006 May.
Article in English | MEDLINE | ID: mdl-16525489

ABSTRACT

All-trans retinoic acid (ATRA) represents the therapy of choice for patients with acute promyelocytic leukemia (APL). However, patients often relapse due to ATRA-resistance. The molecular basis of APL alterations indicates that addition of a histone deacetylase inhibitor to ATRA may restore the sensitivity to retinoids. We explored the in vitro and in vivo effects of a novel retinoic/butyric hyaluronan ester (HBR) on a retinoic acid (RA)-sensitive human myeloid cell line, NB4, and on its RA-resistant subclone, NB4.007/6. In vitro, HBR induced growth arrest and terminal differentiation in RA-sensitive NB4 cells (as confirmed by an increased expression of CD11 family members and nitroblue tetrazolium assay), whereas it inhibited the growth of RA-resistant cells by apoptosis, paralleled by an increase in the levels of caspase 3 and 7. In vivo, HBR treatment of NB4-inoculated severe combined immunodeficient mice resulted in a statistically significant increase in survival time (P<0.0001), comparable to that induced by a maximum tolerated dose of RA alone. Also on P388-inoculated mice, HBR was active in contrast to RA that was completely ineffective. Present findings suggest that, owing to the simultaneous presence of RA and an histone deacetylases inhibitor, HBR might be useful in controlling the proliferation of RA-resistant cells and the differentiation of RA-sensitive cells.


Subject(s)
Butyric Acid/pharmacology , Esters/pharmacology , Hyaluronic Acid/analogs & derivatives , Hyaluronic Acid/pharmacology , Leukemia, Promyelocytic, Acute/drug therapy , Tretinoin/pharmacology , Apoptosis/drug effects , Butyric Acid/chemistry , Butyric Acid/therapeutic use , CCAAT-Enhancer-Binding Protein-alpha/drug effects , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-beta/drug effects , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Esters/chemical synthesis , Esters/therapeutic use , Histones/drug effects , Histones/metabolism , Humans , Hyaluronic Acid/therapeutic use , In Vitro Techniques , Neoplasm Proteins/drug effects , Oncogene Proteins, Fusion/drug effects , Protein Binding , Tretinoin/chemistry , Tretinoin/therapeutic use , Tumor Cells, Cultured
13.
J Pharmacol Exp Ther ; 317(1): 284-91, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16368900

ABSTRACT

The antimetastatic ruthenium complex imidazolium trans-imidazoledimethylsulfoxide-tetrachlorouthenate (NAMI-A) is tested in the B16 melanoma model in vitro and in vivo. Treatment of B6D2F1 mice carrying intra-footpad B16 melanoma with 35 mg/kg/day NAMI-A for 6 days reduces metastasis weight independently of whether NAMI-A is given before or after surgical removal of the primary tumor. Metastasis reduction is unrelated to NAMI-A concentration, which is 10-fold lower than on primary site (1 versus 0.1 mM), and is correlated to the reduction of plasma gelatinolitic activity and to the decrease of cells expressing CD44, CD54, and integrin-beta(3) adhesion molecules. Metastatic cells also show the reduction of the S-phase cells with accumulation in the G(0)/G(1) phase. In vitro, on the highly metastatic B16F10 cell line, NAMI-A reduces cell Matrigel invasion and its ability to cross a layer of endothelial cells after short exposure (1 h) to 1 to 100 microM concentrations. In these conditions, NAMI-A reduces the gelatinase activity of tumor cells, and it also increases cell adhesion to poly-L-lysine and, in particular, to fibronectin, and this effect is associated to the increase of F-actin condensation. This work shows the selective effectiveness of NAMI-A on the metastatic melanoma and suggests that metastasis inhibition is due to the negative modulation of tumor cell invasion processes, a mechanism in which the reduction of the gelatinolitic activity of tumor cells plays a crucial role.


Subject(s)
Antineoplastic Agents/therapeutic use , Dimethyl Sulfoxide/analogs & derivatives , Lung Neoplasms/drug therapy , Melanoma, Experimental/drug therapy , Organometallic Compounds/therapeutic use , Skin Neoplasms/drug therapy , Animals , Cell Adhesion Molecules/biosynthesis , Cell Cycle/drug effects , Cell Line, Tumor , Dimethyl Sulfoxide/therapeutic use , Down-Regulation , Female , Foot , Lung Neoplasms/secondary , Melanoma, Experimental/secondary , Mice , Mice, Inbred Strains , Neoplasm Invasiveness , Ruthenium Compounds , Skin Neoplasms/pathology
14.
J Pharmacol Exp Ther ; 313(1): 227-33, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15579494

ABSTRACT

The duration of cell proadhesive effects induced by imidazolium trans-imidazole dimethyl sulfoxide tetrachlororuthenate (NAMI-A), a compound endowed with in vivo antimetastatic properties, was tested in vitro on the human epithelial tumor cell line KB. The intensity of proadhesive effects continues to increase up to 48 to 72 h after NAMI-A withdrawal and declines only after 96 h. The proadhesive effect on cells seeded on fibronectin is greater than on plastic, since it already reaches its maximum after 24 h. This effect suggests a role for integrin activation, which is further stressed by the inhibitory activity of the disintegrin molecule echistatin. The intensity and duration of NAMI-A's proadhesive effects are correlated to cell exposure time and to the rapid release of NAMI-A metabolites in the culture medium in the first 5 min after drug withdrawal. These metabolites are probably neutral species with ruthenium-bound bioligands to allow for the rapid exchange between cells and extracellular medium. These data suggest a long-lasting effect of NAMI-A in biological systems, even at very low concentrations, and stress the low and reversible effects on kidney, where it naturally concentrates. These data on proadhesive effects are, further, relevant for in vivo antimetastatic effects, as this adhesion is associated to cell motility and invasion, which in turn are related to tumor malignancy and metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Imidazoles/pharmacology , Organometallic Compounds/pharmacology , Peptides/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Cell Adhesion/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Fibronectins/metabolism , Humans , Intercellular Signaling Peptides and Proteins , KB Cells , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Ligands , Neoplasm Metastasis , Rhodamines , Spectrophotometry, Atomic
15.
Eur J Cancer ; 40(9): 1383-96, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15177498

ABSTRACT

Imidazolium trans-imidazoledimethylsulphoxidetrachlororuthenate (NAMI-A) was tested in vitro on the pro-adhesive properties, evaluated as resistance to trypsin treatment, which is a bona fide measure of adhesion strength, of KB and HeLa carcinoma cell lines and on human polymorphonuclear neutrophils (HPMN). NAMI-A increased the pro-adhesive activity of KB cells at 0.001 mM concentration, after few minutes incubation and this effect was not influenced by the vehicle used for cell challenge, neither did it depend on NAMI-A concentration or on temperature. The same effect occurred on HeLa cells at 0.01 mM NAMI-A. This effect, detected at concentrations up to 100 times lower than those necessary to block cells at the G(2)-M premitotic phase of cell cycle, or to inhibit matrix metalloproteinase release or cell invasion, was not related to ruthenium uptake by tumour cells. HeLa cells and healthy HPMN, following short exposure to 0.1 mM NAMI-A, assumed a different shape, with the extrusion of filopodia (HeLa) and of large lamellopodia (HPMN), which increased their interactions with the substrate. This effect was attributed to stabilisation, altered turnover and sensitivity to cytochalasin D of actin filaments. Provided that adhesion is associated with cell motility and invasion, these data suggest that NAMI-A may exert antimetastatic properties at concentrations lower than those observed in the lungs at the end of a conventional intraperitoneal treatment in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Dimethyl Sulfoxide/analogs & derivatives , Dimethyl Sulfoxide/pharmacology , Neoplasms/drug therapy , Neutrophils/drug effects , Organometallic Compounds/pharmacology , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/ultrastructure , Antibodies, Blocking/pharmacology , Antineoplastic Agents/analysis , Cell Adhesion/drug effects , Cell Line , Dimethyl Sulfoxide/analysis , HeLa Cells , Humans , Integrins/immunology , Microscopy, Confocal , Microscopy, Electron, Scanning , Neoplasms/pathology , Neutrophils/chemistry , Neutrophils/ultrastructure , Organometallic Compounds/analysis , Ruthenium/analysis , Ruthenium Compounds , Trypsin
16.
Invest New Drugs ; 21(4): 401-11, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14586207

ABSTRACT

NAMI-A is an innovative ruthenium(III) complex with a very encouraging preclinical profile of metastasis inhibition, which is undergoing initial phases of clinical trials. To assess the pharmacological relevance of the drug fraction associated to plasma proteins, adducts of NAMI-A with either serum albumin or serum transferrin were prepared and their biological effects tested in vitro and in vivo. Specifically, adducts of NAMI-A with either serum albumin or serum transferrin, prepared and characterized at a ruthenium-to-protein molar ratio of 4:1, were evaluated in vitro on the KB human tumor cell line and in vivo on the MCa mammary carcinoma tumor. The effects of NAMI-A/protein adducts on cell viability and on cell cycle progression were found to be far smaller than those produced by free NAMI-A. GFAAS measurements point out that the amount of ruthenium that gets into cells is drastically reduced when NAMI-A is presented in its protein-bound form. In vivo use of NAMI-A adducts with albumin and transferrin resulted markedly less effective on lung metastasis reduction, than free NAMI-A. Overall, the present results suggest that binding to plasma proteins causes a drastic decrease of NAMI-A bioavailability and a subsequent reduction of its biological activity, implying that association to plasma proteins essentially represents a mechanism of drug inactivation.


Subject(s)
Dimethyl Sulfoxide/analogs & derivatives , Dimethyl Sulfoxide/metabolism , Organometallic Compounds/metabolism , Serum Albumin/metabolism , Transferrin/metabolism , Animals , Antigens, CD/biosynthesis , Antigens, Differentiation, B-Lymphocyte/biosynthesis , Cell Line, Tumor , Cell Survival/physiology , Dimethyl Sulfoxide/chemistry , Dimethyl Sulfoxide/pharmacology , Female , Humans , Mice , Mice, Inbred CBA , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Protein Binding/physiology , Receptors, Transferrin , Ruthenium Compounds , Serum Albumin/pharmacology , Transferrin/pharmacology , Xenograft Model Antitumor Assays/methods
17.
Br J Cancer ; 88(9): 1484-91, 2003 May 06.
Article in English | MEDLINE | ID: mdl-12778081

ABSTRACT

The nitric oxide synthase (NOS) pathway has been clearly demonstrated to regulate angiogenesis. Increased levels of NO correlate with tumour growth and spreading in different experimental and human cancers. Drugs interfering with the NOS pathway may be useful in angiogenesis-dependent tumours. The aim of this study was to pharmacologically characterise certain ruthenium-based compounds, namely NAMI-A, KP1339, and RuEDTA, as potential NO scavengers to be used as antiangiogenic/antitumour agents. NAMI-A, KP1339 and RuEDTA were able to bind tightly and inactivate free NO in solution. Formation of ruthenium-NO adducts was documented by electronic absorption, FT-IR spectroscopy and (1)H-NMR. Pretreatment of rabbit aorta rings with NAMI-A, KP1339 or RuEDTA reduced endothelium-dependent vasorelaxation elicited by acetylcholine. This effect was reversed by 8-Br-cGMP. The key steps of angiogenesis, endothelial cell proliferation and migration stimulated by vascular endothelial growth factor (VEGF) or NO donor drugs, were blocked by NAMI-A, KP1339 and RuEDTA, these compounds being devoid of any cytotoxic activity. When tested in vivo, NAMI-A inhibited angiogenesis induced by VEGF. It is likely that the antitumour properties previously observed for ruthenium-based NO scavengers, such as NAMI-A, are related to their NO-related antiangiogenic properties.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Dimethyl Sulfoxide/analogs & derivatives , Dimethyl Sulfoxide/pharmacology , Endothelium, Vascular/physiology , Nitric Oxide/physiology , Organometallic Compounds/pharmacology , Ruthenium Compounds/pharmacology , Cell Line , Cell Survival/drug effects , Chemotaxis/drug effects , Coronary Vessels , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Free Radical Scavengers , Humans , Nitric Oxide Synthase/antagonists & inhibitors , Spectroscopy, Fourier Transform Infrared , Venules/drug effects , Venules/physiology
18.
Int J Pharm ; 248(1-2): 247-59, 2002 Nov 06.
Article in English | MEDLINE | ID: mdl-12429478

ABSTRACT

This paper describes the development of a stable pharmaceutical dosage form for NAMI-A, a novel antimetastatic ruthenium complex, for Phase I testing. NAMI-A drug substance was characterized using several spectrometric and chromatographic techniques. In preformulation studies, it was found that NAMI-A in aqueous solution was not stable enough to allow sterilization by moist heat. The effect of several excipients on the stability of the formulation solution was investigated. None of them provided sufficient stability to allow long-term storage of an aqueous solution of NAMI-A. Therefore, a lyophilized product was developed. Five different formulations were prepared and subjected to thermogravimetric (TG) analysis and stability studies at various conditions for 1 year. Minimal degradation during the production process is achieved with a formulation solution of pH 3-4. Of the acids tested, only hydrochloric acid (HCl 0.1 mM) both stabilized the formulation solution and was compatible with the lyophilized product. This product was stable for at least 1 year when stored at -20 degrees C, 25 degrees C/60% relative humidity (RH) and 40 degrees C/75% RH, and was also photostable.


Subject(s)
Antineoplastic Agents/chemistry , Dimethyl Sulfoxide/analogs & derivatives , Dimethyl Sulfoxide/chemistry , Neoplasm Metastasis/prevention & control , Organometallic Compounds/chemistry , Technology, Pharmaceutical/methods , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Chemistry, Pharmaceutical , Dimethyl Sulfoxide/administration & dosage , Dimethyl Sulfoxide/pharmacokinetics , Freeze Drying , Infusions, Parenteral , Organometallic Compounds/administration & dosage , Organometallic Compounds/pharmacokinetics , Ruthenium/administration & dosage , Ruthenium/chemistry , Ruthenium/pharmacokinetics , Ruthenium Compounds
19.
Br J Cancer ; 86(6): 993-8, 2002 Mar 18.
Article in English | MEDLINE | ID: mdl-11953835

ABSTRACT

NAMI-A is a ruthenium-based compound with selective anti-metastasis activity in experimental models of solid tumours. We studied whether this activity was dependent on anti-angiogenic ability of NAMI-A. We thus investigated its in vitro effects on endothelial cell functions necessary for angiogenesis to develop, as well as its in vivo effects in the chick embryo chorioallantoic membrane model. Endothelial cell proliferation, chemotaxis, and secretion of the matrix-degrading enzyme metalloproteinase-2 were inhibited by NAMI-A in a dose-dependent manner, and without morphologic signs of cell apoptosis or necrosis. Lastly, NAMI-A displayed a dose-dependent in vivo anti-angiogenic activity in the chorioallantoic membrane model. These data suggest that the anti-angiogenic activity of NAMI-A can contribute to its anti-metastatic efficacy in mice bearing malignant solid tumours.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Dimethyl Sulfoxide/analogs & derivatives , Dimethyl Sulfoxide/pharmacology , Endothelium, Vascular/drug effects , Neoplasm Metastasis/prevention & control , Organometallic Compounds/pharmacology , Cells, Cultured , Chemotaxis/drug effects , Endothelium, Vascular/cytology , Humans , Matrix Metalloproteinase Inhibitors , Ruthenium Compounds
20.
Eur J Cancer ; 38(3): 427-35, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11818210

ABSTRACT

The influence of chemical stability on the antimetastatic ruthenium(III) compound imidazolium trans-imidazoletetrachlorodimethylsulphoxideruthenium(III) (NAMI-A) in aqueous solution was studied both in vitro and in vivo. The loss of dimethyl-sulphoxide (DMSO) ligand from the compound was tested by using a NAMI-A solution acidified with HCl at pH 3.0 and aged for 0, 4, 8 and 24 h prior to intraperitoneal (i.p.) injection into CBA mice bearing advanced MCa mammary carcinoma. The activity of NAMI-A on lung metastases showed no change even after the loss of DMSO ligand from up to 50% of the molecules. The reduction of NAMI-A did not modify the number of KB cells blocked in the S+G2M phases, independent of whether the reduction occurred outside the cells or after loading the cells with the compound prior to treatment with the reductants (ascorbic acid, glutathione or cysteine). In vivo, the complete reduction of NAMI-A with equivalent amounts of ascorbic acid, glutathione or cysteine prior to administration to mice bearing advanced MCa mammary carcinoma was more active than NAMI-A alone. The data show that NAMI-A, although undergoing a series of chemical modifications, maintains its antimetastatic activity in a broad range of experimental conditions.


Subject(s)
Antineoplastic Agents/chemistry , Dimethyl Sulfoxide/analogs & derivatives , Dimethyl Sulfoxide/chemistry , Mammary Neoplasms, Experimental/drug therapy , Organometallic Compounds/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cell Division , Dimethyl Sulfoxide/metabolism , Dimethyl Sulfoxide/pharmacokinetics , Dimethyl Sulfoxide/therapeutic use , Female , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred CBA , Neoplasm Transplantation , Organometallic Compounds/pharmacokinetics , Organometallic Compounds/therapeutic use , S Phase , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...