Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
J Pharm Belg ; (2): 10-17, 2016 Jun.
Article in French | MEDLINE | ID: mdl-30281226

ABSTRACT

Background Medication safety and adherence are important aspects of quality of life, especially for frail elderly in our society. However, many elderly living at home may experience difficulties when taking their medication, and are therefore at risk for medication- related problems. The current living lab was implemented within the framework of the Flanders Care Living Labs and set the objective to develop a care pathway to improve medication safety and adherence for elderly living at home. Automated Individual Medication Preparation [IMP] was provided by a community-pharmacist in order to support frail elderly in taking their medication more correctly and thereby improving adherence. Methods In a before and after study, each patient was supported for 13 weeks through a multidisciplinary care pathway. Key in this pathway was the shared medication schedule (between GP and pharmacist) as the starting point for IMP and medication counselling. The increased medication support based on IMP was evaluated in frail elderly (age a 65 years, complex medication scheme a 5 medicines and a 3 intakes / day) before and after the study, by measuring adherence and satisfaction with counselling and repackaging the medication in sachets. Results The preliminary results of the current study showed that the self-reported adherence rate in frail elderly was significantly higher (p <0.05) after implementation of the pharmacists' intervention with automated IMP. At the end of the study every patient indicated that the repackaging of the medication by dose was helpful in order to take their medication correctly. Conclusion Increased medication counselling with IMP by the community-pharmacist may be a useful aid and supporting tool for frail elderly living at home with a complex medication scheme.


Subject(s)
Counseling , Medication Therapy Management , Aged , Aged, 80 and over , Community Pharmacy Services , Humans , Medication Adherence , Pharmacists
2.
Nucleosides Nucleotides Nucleic Acids ; 33(4-6): 394-402, 2014.
Article in English | MEDLINE | ID: mdl-24940697

ABSTRACT

Mycoplasmas are opportunistic parasites and some species are suggested to preferentially colonize tumor tissue in cancer patients. We could demonstrate that the annotated thymidine phosphorylase (TP) gene in the genome of Mycoplasma hyorhinis encodes a pyrimidine nucleoside phosphorylase (PyNPHyor) that not only efficiently catalyzes thymidine but also uridine phosphorolysis. The kinetic characteristics of PyNPHyor-catalyzed nucleoside and nucleoside analogue (NA) phosphorolysis were determined. We demonstrated that the expression of such an enzyme in mycoplasma-infected cell cultures dramatically alters the activity of various anticancer/antiviral NAs such as 5-halogenated pyrimidine nucleosides, including 5-trifluorothymidine (TFT). Due to their close association with human cancers, the presence of mycoplasmas may markedly influence the therapeutic efficiency of nucleoside-based drugs.


Subject(s)
Antiviral Agents/pharmacology , Mycoplasma hyorhinis/enzymology , Pyrimidine Phosphorylases/metabolism , Trifluridine/pharmacology , Cell Line, Tumor , Humans , MCF-7 Cells , Mycoplasma hyorhinis/physiology , Pyrimidine Phosphorylases/genetics
3.
Invest New Drugs ; 32(1): 200-10, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23733228

ABSTRACT

Cytostatic agents often do not discriminate in their cytostatic potential between different tumor cell types in vitro. In this study, several 2-aminothiophene-3-carboxylic acid ester derivatives were discovered that show an unusual cytostatic selectivity for several T-cell (but not B-cell) lymphoma, prostate cancer, kidney carcinoma and hepatoma cell lines. Their 50 % cytostatic concentrations were generally in the higher nanomolar range and were approximately 20- to 50-fold lower for these tumor cell types than for any other tumor cell line or non-tumorigenic cells. The tumor-selective compounds caused a more preferential suppression of protein synthesis than DNA or RNA synthesis and the prototype compound 3 resulted in an accumulation of prostate cancer cells in the G1 phase of their cell cycle. Compound 3 was also shown to induce apoptosis in prostate cancer cells. The 2-aminothiophene-3-carboxylic acid ester derivatives represent novel candidate cytostatic agents to be further explored for their tumor-selective potential.


Subject(s)
Amino Acids/therapeutic use , Cytostatic Agents/therapeutic use , Esters/therapeutic use , Thiophenes/therapeutic use , Amino Acids/chemistry , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cytostatic Agents/chemistry , Esters/chemistry , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Structure-Activity Relationship , Thiophenes/chemistry
4.
J Pharm Belg ; (4): 18-27, 2013 Dec.
Article in French | MEDLINE | ID: mdl-24449969

ABSTRACT

Medication adherence in chronic conditions such as asthma, type 2 diabetes, heart failure, HIV and cancer appears to be a frequent problem. However, the literature on adherence in patients who use inhaled corticosteroids (ICS), oral hypoglycemic agents, drugs for heart failure, antiretrovirals or oral chemotherapy, contains little or no relevant data for Belgium. In the context of a Master thesis in Pharmaceutical care at KU Leuven, a quantitative study was performed to determine the prevalence of adherence to chronic medication in Belgium. This retrospective, cross-sectional study used a database containing refill data of a regional pharmacists' association (KLAV). Out of the 603 pharmacies affiliated with this association, all 50 pharmacies where HIV medication was delivered, were selected. Dispensing data from the selected pharmacies were collected from 01/07/2008 to 31/12/2009 for five pathologies, i.e.; asthma, type 2 diabetes, heart failure, HIV and cancer. Adherence (TT) was calculated with the Medication Refill Adherence (MRA) method. In order to determine whether there were associations between age, gender, drug class and adherence, Chi-square tests were used. Compared with the other patients, cancer patients were the most adherent in taking their drugs (median adherence rate = 88%). In addition, this was the only group in which the median adherence rate was above the set limit of 80%. The patients who were prescribed inhaled corticosteroids were the least adherent (median adherence rate = 38%). More than 50% of patients with asthma/COPD, heart failure and diabetes were classified as "under-users". Furthermore, the results showed a significant association within asthma patients between gender and adherence. In asthma, type 2 diabetes, heart failure and HIV patients there was a significant relationship between age and adherence and drug class and adherence. As the current study has some limitations, the results should be handled with caution. Nevertheless, the current study shows that also in Belgium there is a problem with medication adherence in chronic conditions, especially in asthma patients.


Subject(s)
Chronic Disease/drug therapy , Patient Compliance/statistics & numerical data , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Belgium/epidemiology , Cross-Sectional Studies , Databases, Factual , Drug Prescriptions/statistics & numerical data , Female , Humans , Male , Middle Aged , Retrospective Studies , Young Adult
5.
Eur Psychiatry ; 27(7): 528-35, 2012 Oct.
Article in English | MEDLINE | ID: mdl-21392945

ABSTRACT

OBJECTIVE: The aim of the present study was to measure stigma among community pharmacists regarding patients with depression, using social distance as a proxy measure. Furthermore, factors influencing stigmatization were identified. METHODS: A survey consisting of questions regarding social distance, cognitive attitude components, stereotypical beliefs and behavioural tendencies towards pharmaceutical care for patients with depression was carried out. The survey was completed by 149 community pharmacists with a response rate of 82%. Multiple linear regression analysis was applied to the results of the survey. RESULTS: Most pharmacists endorsed social distance towards patients with depression. The social distance further increased the more the situation described implied "social closeness". Age, depression care attitude, the stereotypical belief "danger to others" and the level of involvement pharmacists showed towards patients with depression were found to predict social distance. The present model explains 32.3% of the variance in social distance. CONCLUSION: The results showed that community pharmacists have a stigmatizing attitude towards patients with depression. The determinants found to influence these stigmatizing attitudes might be addressed when trying to reduce stigmatization of patients with depression.


Subject(s)
Attitude of Health Personnel , Depressive Disorder/psychology , Pharmacists , Psychological Distance , Social Stigma , Adult , Female , Humans , Male , Middle Aged , Pharmacies , Surveys and Questionnaires
6.
Curr Pharm Des ; 16(35): 3903-20, 2010.
Article in English | MEDLINE | ID: mdl-21158728

ABSTRACT

Chemokines are key players in the attraction and activation of leukocytes and are thus implicated in the recruitment of immune cells at sites of infection and/or inflammation. They exert their action by binding to seven-transmembrane G protein-coupled receptors. The chemokine stromal cell-derived factor-1 (SDF-1)/CXCL12 represents the single natural ligand for the chemokine receptor CXCR4. CXCL12 possesses angiogenic properties and is involved in the outgrowth and metastasis of CXCR4-expressing tumors and in certain inflammatory autoimmune disorders, such as rheumatoid arthritis. CXCR4 expression on tumor cells is upregulated by hypoxia and angiogenic factors, such as vascular endothelial growth factor (VEGF). CXCR4 also acts as a co-receptor for entry of human immunodeficiency virus (HIV) in CD4(+) T cells. Finally, CXCL12/CXCR4 interactions were shown to play an important role in the migration of hematopoietic stem cells and their progenitors from, and their retention within, the bone marrow, a site characterized by high CXCL12 expression. As such, CXCR4 inhibitors may be utilized to inhibit HIV-1 infection, tumor growth and metastasis and to mobilize hematopoietic stem cells from the bone marrow in the circulation, where they can be collected for autologous stem cell transplantation. Here, we discuss the different aspects of CXCL12/CXCR4 biology as well as the development and anti-cancer/stem cell mobilizing activity of CXCR4 antagonists.


Subject(s)
Chemokine CXCL12/physiology , Neoplasm Metastasis/physiopathology , Neovascularization, Pathologic/physiopathology , Receptors, CXCR4/physiology , Stem Cells/physiology , Humans
7.
Biochem Pharmacol ; 78(3): 231-40, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19389384

ABSTRACT

Thymidine phosphorylase (TP) is a catabolic enzyme in thymidine metabolism that is frequently upregulated in many solid tumors. Elevated TP levels are associated with tumor angiogenesis, metastasis and poor prognosis. Therefore, the use of TP inhibitors might offer a promising strategy for cancer treatment. The tritylated inosine derivative 5'-O-tritylinosine (previously designated KIN59) is a non-competitive inhibitor of TP which was previously found to be instrumental for the crystallization of human TP. A combination of computational studies including normal mode analysis, automated ligand docking and molecular dynamics simulations were performed to define a plausible binding site for 5'-O-tritylinosine on human TP. A cavity in which 5'-O-tritylinosine could fit was identified in the vicinity of the Gly405-Val419 loop at a distance of about 11A from the substrate-binding site. In the X-ray crystal structure, this pocket is characterized by an intricate hydrogen-bonding network in which Asp203 was found to play an important role to afford the loop stabilization that is required for efficient enzyme catalysis. Site-directed mutagenesis of this amino acid residue afforded a mutant enzyme with a severely compromised catalytic efficiency (V(max)/K(m) of mutant enzyme approximately 50-fold lower than for wild-type TP) and pronounced resistance to the inhibitory effect of 5'-O-tritylinosine. In contrast, the D203A mutant enzyme kept full sensitivity to the competitive inhibitors 6-aminothymine and 6-amino-5-bromouracil, which is in line with the kinetic properties of these inhibitors. Our findings reveal the existence of a previously unrecognized site in TP that can be targeted by small molecules to inhibit the catalytic activity of TP.


Subject(s)
Aspartic Acid/metabolism , Enzyme Inhibitors/pharmacology , Inosine/analogs & derivatives , Thymidine Phosphorylase/metabolism , Trityl Compounds/pharmacology , Amino Acid Sequence , Animals , Aspartic Acid/antagonists & inhibitors , Catalytic Domain , Crystallization , Crystallography, X-Ray , Humans , Hydrogen Bonding , Inosine/pharmacology , Kinetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Sequence Homology, Amino Acid , Thymidine Phosphorylase/antagonists & inhibitors , Thymidine Phosphorylase/chemistry , Thymidine Phosphorylase/genetics
8.
Verh K Acad Geneeskd Belg ; 70(3): 175-91, 2008.
Article in English | MEDLINE | ID: mdl-18669159

ABSTRACT

Potent thymidine phosphorylase (TP) inhibitors with anti-angiogenic activity are required to improve the prognosis of patients with TP-positive tumors. We have designed and developed novel structural and functional classes of TP inhibitors that also inhibit TP-induced angiogenesis, i.e. (i) the first purine analogue inhibitor of TP (7DX), (ii) the first multifunctional TP inhibitor (TP65), and (iii) the purine riboside analogue KIN59, which inhibits TP without interacting with the substrate-binding sites. The latter finding indicates that a, yet unidentified, allosteric site in TP contributes to its biological activities. In order to identify the amino acid residues in TP that interact with KIN59, we performed co-crystallograpy of the KIN59-TP complex. Our preliminary data suggest the existence of a putative KIN59 binding site. Identification of the allosteric site(s) in TP is important to gain more insight into the different biological activities of TP and may aid in the design of novel TP inhibitors. The nucleotide analogue cidofovir, which is being used clinically for the treatment of cytomegalovirus-induced retinitis in AIDS patients, emerged as a promising antitumor agent. So far, cidofovir has only been evaluated clinically for the treatment of HPV-associated tumors. Our results demonstrate the potent activity of cidofovir against tumors that are not associated with a virus. Cidofovir inhibits the growth of strongly vascularized tumors via inhibition of the growth factor FGF2, and by increasing the expression of the tumor suppressor p53, leading to apoptosis. These exciting results open new perspectives for the use of cidofovir as an anti-tumor agent in the therapy of tumors that are not associated with an oncogenic virus.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis , Enzyme Inhibitors/pharmacology , Thymidine Phosphorylase/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cidofovir , Cytosine/analogs & derivatives , Humans , Organophosphonates , Prognosis
9.
Nucleosides Nucleotides Nucleic Acids ; 25(9-11): 975-80, 2006.
Article in English | MEDLINE | ID: mdl-17065049

ABSTRACT

We found that 5'-O-trityl-inosine (KIN59) inhibits recombinant bacterial (E. coli) and human thymidine phosphorylase (TPase) with an IC50 of 44 microM and 67 microM, respectively. In contrast to previously described TPase inhibitors, KIN59 does not compete with thymidine (dThd) at the pyrimidine nucleoside-binding site or with inorganic phosphate (Pi) at the phosphate-binding site of the enzyme. These findings are strongly suggestive for the presence of an allosteric binding site at the enzyme. TPase is identical to the angiogenic protein platelet-derived endothelial cell growth factor (PD-ECGF). As such, PD-ECGF stimulates angiogenesis in the chick chorioallantoic membrane (CAM) assay. This angiogenic response was completely inhibited by KIN59. Inosine did not inhibit the enzyme or the angiogenic effect of TPase, confirming that the 5'-O-trityl group in KIN59 is essential for the observed effect. Our observations indicate that allosteric sites in TPase may regulate its biological activity.


Subject(s)
Enzyme Inhibitors/pharmacology , Inosine/analogs & derivatives , Thymidine Phosphorylase/antagonists & inhibitors , Trityl Compounds/pharmacology , Allosteric Site , Animals , Chickens , Chorion/metabolism , Escherichia coli/enzymology , Humans , Inhibitory Concentration 50 , Inosine/pharmacology , Neoplasms/drug therapy , Neovascularization, Pathologic , Phosphates , Pyrimidines/chemistry , Thymidine Phosphorylase/metabolism
10.
Curr Pharm Des ; 9(7): 553-66, 2003.
Article in English | MEDLINE | ID: mdl-12570803

ABSTRACT

Angiogenesis is the process of generating new capillary blood vessels. Uncontrolled endothelial cell proliferation is observed in tumor neovascularization and in angioproliferative diseases. Tumors cannot growth as a mass above few mm(3) unless a new blood supply is induced. It derives that the control of the neovascularization process may affect tumor growth and may represent a novel approach to tumor therapy. Angiogenesis is controlled by a balance between proangiogenic and antiangiogenic factors. The angiogenic switch represents the net result of the activity of angiogenic stimulators and inhibitors, suggesting that counteracting even a single major angiogenic factor could shift the balance towards inhibition. Heparan sulfate proteoglycans are involved in the modulation of the neovascularization that takes place in different physiological and pathological conditions. This modulation occurs through the interaction with angiogenic growth factors or with negative regulators of angiogenesis. Thus, the study of the biochemical bases of this interaction may help to design glycosaminoglycan analogs endowed with angiostatic properties. The purpose of this review is to provide an overview of the structure/function of heparan sulfate proteoglycans in endothelial cells and to summarize the angiostatic properties of synthetic heparin-like compounds, chemically modified heparins, and biotechnological heparins.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Heparin/analogs & derivatives , Heparin/pharmacology , Neovascularization, Pathologic/drug therapy , Angiogenesis Inducing Agents/antagonists & inhibitors , Animals , Cell Adhesion/physiology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Heparan Sulfate Proteoglycans/metabolism , Heparan Sulfate Proteoglycans/physiology , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic/physiopathology
11.
Verh K Acad Geneeskd Belg ; 64(3): 197-224, 2002.
Article in English | MEDLINE | ID: mdl-12238243

ABSTRACT

Hemangiomas represent the most frequent tumors of infancy. However, the pathogenesis of these tumors is still largely unknown, and current treatment of juvenile hemangiomas remains unsatisfactory. We have presented a novel animal model for hemangiomas. Induction of hemangioma development was achieved by intraperitoneal (i.p.) infection of 4-day-old rats with the mouse polyoma virus (PyV). This led to the development of multiple hemangiomas, which caused death of the untreated animals within 3 weeks p.i. The hemangiomas had the histological and immunohistochemical features reminiscent of human hemangiomas. Moreover, the angiogenesis inhibitor TNP-470 afforded a protective effect in this model. Tumor growth is determined by the balance between cell proliferation and apoptosis and is modulated by angiogenesis. Angiogenesis is a complex process, involving extensive interplay between cells, extracellular matrix components and soluble factors. Each of these factors represents a possible target for pharmacological intervention to inhibit blood vessel formation and subsequently tumor growth. We have focused on specific inhibitors of the angiogenesis inducers basic fibroblast growth factor and Thymidine Phosphorylase and studied their mechanism of action and anti-angiogenic activity. In addition, we have shown that the apoptosis-inducer cidofovir inhibits PyV-induced hemangioma development in rats and the growth of virus-independent, vascular tumors in mice. So far, cidofovir has only been evaluated clinically for the treatment of human papillomavirus (HPV)-associated tumors. Our findings open new perspectives for the use of cidofovir as an anti-tumor agent in the therapy of hemangiomas and other tumors that are not associated with an oncogenic virus.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Apoptosis , Cytosine/analogs & derivatives , Hemangioma/prevention & control , Neovascularization, Pathologic , Organophosphonates , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Division , Cidofovir , Cyclohexanes , Cytosine/pharmacology , Cytosine/therapeutic use , Disease Models, Animal , Fibroblast Growth Factor 2/antagonists & inhibitors , Growth Substances , Hemangioma/blood supply , Hemangioma/pathology , Humans , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/prevention & control , O-(Chloroacetylcarbamoyl)fumagillol , Organophosphorus Compounds/pharmacology , Organophosphorus Compounds/therapeutic use , Rats , Sesquiterpenes/pharmacology , Sesquiterpenes/therapeutic use , Thymidine Phosphorylase/antagonists & inhibitors , Tumor Virus Infections/prevention & control
12.
Blood ; 98(13): 3554-61, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11739156

ABSTRACT

The interferon (IFN)-inducible chemokines, specifically, IFN-gamma-inducible protein-10 (IP-10), monokine induced by IFN-gamma (Mig), and IFN-inducible T-cell alpha-chemoattractant (I-TAC), share a unique CXC chemokine receptor (CXCR3). Recently, the highly specific membrane-bound protease and lymphocyte surface marker CD26/dipeptidyl peptidase IV (DPP IV) was found to be responsible for posttranslational processing of chemokines. Removal of NH(2)-terminal dipeptides by CD26/DPP IV alters chemokine receptor binding and signaling, and hence inflammatory and anti-human immunodeficiency virus (HIV) activities. CD26/DPP IV and CXCR3 are both markers for Th1 lymphocytes and, moreover, CD26/DPP IV is present in a soluble, active form in human plasma. This study reports that at physiologic enzyme concentrations CD26/DPP IV cleaved 50% of I-TAC within 2 minutes, whereas for IP-10 and Mig the kinetics were 3- and 10-fold slower, respectively. Processing of IP-10 and I-TAC by CD26/DPP IV resulted in reduced CXCR3-binding properties, loss of calcium-signaling capacity through CXCR3, and more than 10-fold reduced chemotactic potency. Moreover, IP-10 and I-TAC cleaved by CD26/DPP IV acted as chemotaxis antagonists and CD26/DPP IV-truncated IP-10 and Mig retained their ability to inhibit the angiogenic activity of interleukin-8 in the rabbit cornea micropocket model. These data demonstrate a negative feedback regulation by CD26/DPP IV in CXCR3-mediated chemotaxis without affecting the angiostatic potential of the CXCR3 ligands IP-10 and Mig.


Subject(s)
Angiogenesis Inhibitors , Chemokines, CXC/chemistry , Chemotaxis, Leukocyte/drug effects , Intercellular Signaling Peptides and Proteins , Lymphocytes/physiology , Receptors, Chemokine/agonists , Signal Transduction/drug effects , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Calcium/metabolism , Chemokine CXCL10 , Chemokine CXCL11 , Chemokine CXCL9 , Chemokines, CXC/metabolism , Chemokines, CXC/pharmacology , Dipeptidyl Peptidase 4/metabolism , Receptors, CXCR3 , Receptors, Chemokine/physiology , Structure-Activity Relationship
13.
Cancer Res ; 61(13): 5057-64, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11431341

ABSTRACT

Cidofovir [(S)-HPMPC; (S)-1-(3-hydroxy-2-phosphonylmethoxypropyl)cytosine] is an antiviral drug that has been approved for the treatment of cytomegalovirus retinitis in AIDS patients. Cidofovir also possesses potent inhibitory activity against various human papillomavirus-induced tumors in animal models and patients. In addition, cidofovir inhibits the development of murine polyomavirus-induced hemangiomas in rats by an as-yet-uncharacterized, antivirus-independent mechanism. Here we report the inhibitory effect of cidofovir on the development of virus-independent vascular tumors originated by basic fibroblast growth factor (FGF2)-overexpressing endothelial cells (FGF2-T-MAE cells). In vitro, cidofovir was cytostatic to FGF2-T-MAE cells at a 50% cytostatic concentration of 6.7 microg/ml. Cidofovir concentrations >25 microg/ml resulted in cytotoxicity because of induction of apoptosis. Cidofovir did not affect FGF2-T-MAE cell sprouting in three-dimensional fibrin gel and morphogenesis on Matrigel at noncytotoxic concentrations. In vivo, cidofovir (100 microg/egg) completely suppressed hemangioma formation on the chick chorioallantoic membrane (CAM) induced by intra-allantoic injection of FGF2-T-MAE cells, without affecting the formation of normal CAM vessels. Accordingly, cidofovir applied locally at 200 microg/disc, reduced neovascularization on the CAM by only 35%. Intratumoral or systemic administration of cidofovir caused a significant inhibition of the growth of s.c., i.p., or intracerebral FGF2-T-MAE xenografts in nude mice and severe combined immunodeficient mice. Drug-induced apoptosis was observed in FGF2-T-MAE tumors as soon as 2 days after the beginning of treatment. In conclusion, cidofovir appears to inhibit the growth of endothelium-derived tumors via induction of apoptosis without exerting a direct antiangiogenic activity.


Subject(s)
Antineoplastic Agents/pharmacology , Cytosine/pharmacology , Fibroblast Growth Factor 2/antagonists & inhibitors , Hemangioendothelioma/drug therapy , Organophosphonates , Organophosphorus Compounds/pharmacology , Animals , Antiviral Agents/pharmacology , Apoptosis/drug effects , Brain Neoplasms/blood supply , Brain Neoplasms/drug therapy , Cell Division/drug effects , Chick Embryo , Cidofovir , Cytosine/analogs & derivatives , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Female , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/physiology , Hemangioendothelioma/blood supply , Hemangioendothelioma/pathology , Mice , Mice, Nude , Mice, SCID , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic/drug effects , Transfection , Tumor Cells, Cultured
14.
Int J Cancer ; 92(2): 161-7, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11291040

ABSTRACT

The acyclic nucleoside phosphonate analogue cidofovir is a broad-spectrum anti-DNA virus agent, which also possesses potent inhibitory activity against various tumors associated with papillomaviruses in animal models and patients. Moreover, we recently described the potent inhibition of polyomavirus (PyV)-induced hemangioma formation in rats by cidofovir. This activity could not be explained by an antiviral mechanism. We have now evaluated the effect of cidofovir on the growth of hemangiosarcomas originating from PyV-transformed (PV/2b/35) cells, which do not produce polyomavirus. In vitro, cidofovir proved to be cytostatic for PV/2b/35 cells at a 50% cytostatic concentration (CC(50)) of 2.3 microg/ml. At cidofovir concentrations > or =20 microg/ml, cytotoxicity due to induction of apoptosis was observed. In vivo, intratumoral therapy with cidofovir, at 100 mg/kg 3 times a week, completely inhibited the development and even caused regression of established PV/2b/35 hemangiosarcomas in nude mice. Five days after the start of treatment, few proliferating cells were noted in the cidofovir-treated tumors, whereas control tumors were characterized by high expression of proliferating cell nuclear antigen (PCNA). Moreover, cidofovir induced apoptosis in the hemangiosarcomas, as evidenced by Tunel (terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling) staining. Also after intraperitoneal administration, cidofovir afforded a prominent protection against the growth of intraperitoneally or intracerebrally inoculated hemangiosarcoma cells in SCID mice. In conclusion, cidofovir possesses a direct antitumor activity, which is mediated by induction of tumor cell apoptosis. Cidofovir should be further explored for its potential in the treatment of fast-growing vascular tumors, like hemangiomas and hemangiosarcomas.


Subject(s)
Antineoplastic Agents/therapeutic use , Cytosine/therapeutic use , Hemangiosarcoma/prevention & control , Organophosphonates , Organophosphorus Compounds/therapeutic use , Animals , Apoptosis/drug effects , Brain Neoplasms/pathology , Brain Neoplasms/prevention & control , Cell Division/drug effects , Cell Line, Transformed , Cidofovir , Cytosine/analogs & derivatives , Female , Hemangiosarcoma/pathology , Mice , Mice, Nude , Mice, SCID , Neoplasm Transplantation , Peritoneal Neoplasms/pathology , Peritoneal Neoplasms/prevention & control , Survival Rate
15.
Cancer Invest ; 19(1): 35-40, 2001.
Article in English | MEDLINE | ID: mdl-11291554

ABSTRACT

Potential anticancer therapy with the fumagillin analog TNP-470 was investigated in the present project using subcutaneously growing rhabdomyosarcomas in rats. Specifically, influences of different tumor sizes at the start of treatment as well as dose/schedules were evaluated with this angiogenesis inhibitor. The results show a significant (p = < or = 0.01) reduction of the growth rate, even for relatively large-sized (> 7 cm3) tumors, when 50 mg/kg TNP-470 was used every other day for up to 3 or 5 injections. With 30 mg/kg TNP-470 injections, effects were seen only with tumors measuring < 7 cm3. The histologic examinations demonstrate an increase in necrosis, both in the center and in the peripheral part of TNP-470-treated tumors. Overall, both tumor volume and drug dose determine treatment outcome with the rat rhabdomyosarcoma. The results suggest that angiogenesis inhibitors could represent a valid component in the treatment of progressive tumor growth, also of large tumors as often encountered in clinics. The antivasculature therapy might also improve hypoxia/necrosis-related therapeutic approaches.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Rhabdomyosarcoma/drug therapy , Sesquiterpenes/therapeutic use , Angiogenesis Inhibitors/administration & dosage , Animals , Cell Survival , Cyclohexanes , Drug Administration Schedule , Male , Necrosis , O-(Chloroacetylcarbamoyl)fumagillol , Rats , Rhabdomyosarcoma/pathology , Sesquiterpenes/administration & dosage
16.
Biochem Pharmacol ; 61(3): 253-70, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11172729

ABSTRACT

Angiogenesis is a fundamental process in reproduction and wound healing. Under these conditions, neovascularization is tightly regulated. Unregulated angiogenesis may lead to several angiogenic diseases and is thought to be indispensable for solid tumor growth and metastasis. The construction of a vascular network requires different sequential steps including the release of proteases from "activated" endothelial cells with subsequent degradation of the basement membrane surrounding the existing vessel, migration of endothelial cells into the interstitial space, endothelial cell proliferation, and differentiation into mature blood vessels. These processes are mediated by a wide range of angiogenic inducers, including growth factors, chemokines, angiogenic enzymes, endothelial specific receptors, and adhesion molecules. Finally, when sufficient neovascularization has occurred, angiogenic factors are down-regulated or the local concentration of inhibitors increases. As a result, the endothelial cells become quiescent, and the vessels remain or regress if no longer needed. Thus, angiogenesis requires many interactions that must be tightly regulated in a spatial and temporal manner. Each of these processes presents possible targets for therapeutic intervention. Synthetic inhibitors of cell invasion (marimastat, Neovastat, AG-3340), adhesion (Vitaxin), or proliferation (TNP-470, thalidomide, Combretastatin A-4), or compounds that interfere with angiogenic growth factors (interferon-alpha, suramin, and analogues) or their receptors (SU6668, SU5416), as well as endogenous inhibitors of angiogenesis (endostatin, interleukin-12) are being evaluated in clinical trials against a variety of solid tumors. As basic knowledge about the control of angiogenesis and its role in tumor growth and metastasis increases, it may be possible in the future to develop specific anti-angiogenic agents that offer a potential therapy for cancer and angiogenic diseases.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/physiology , Angiogenesis Inducing Agents/physiology , Humans , Neoplasms/drug therapy , Neoplasms/physiopathology , Neovascularization, Physiologic/drug effects
17.
Gen Pharmacol ; 34(2): 107-16, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10974418

ABSTRACT

Osmotic cell swelling activates an outwardly rectifying Cl(-) current in endothelial cells that is mediated by volume-regulated anion channels (VRACs). In the past, we have shown that serum-induced proliferation of endothelial cells is arrested in the presence of compounds that potently block the endothelial VRACs. Here we report on the effects of four chemically distinct VRAC blockers [5-nitro-2-(3-phenylpropylamino)benzoic acid] (NPPB), mibefradil, tamoxifen, and clomiphene-on several models of experimental angiogenesis. Mibefradil (20 microM), NPPB (100 microM), tamoxifen (20 microM), and clomiphene (20 microM) inhibited tube formation by rat microvascular endothelial cells plated on matrigel by 42.9 +/- 8.8%, 25.3 +/- 10.4%, 32.2 +/- 4.5%, and 20 +/- 5.8%, respectively (p < 0.05). Additionally, NPPB (50-100 microM) and mibefradil (10-30 microM) significantly inhibited bFGF (10 ng/ml) + TNFalpha (2.5 ng/ml)-stimulated microvessel formation by human microvascular endothelial cells plated on fibrin by 30-70%. Furthermore, NPPB, mibefradil, and clomiphene concentration dependently inhibited spontaneous microvessel formation in the rat aorta-ring assay and vessel development in the chick chorioallantoic membrane assay. These results suggest that VRAC blockers are potent inhibitors of angiogenesis and thus might serve as therapeutic tools in tumor growth and other angiogenesis-dependent diseases.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Ion Channels/antagonists & inhibitors , Neovascularization, Physiologic/drug effects , Allantois/blood supply , Allantois/drug effects , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/growth & development , Cattle , Cells, Cultured , Chick Embryo , Chorion/blood supply , Chorion/drug effects , Clomiphene/pharmacology , Collagen , Drug Combinations , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Fibrin , Humans , In Vitro Techniques , Laminin , Male , Mibefradil/pharmacology , Nitrobenzoates/pharmacology , Proteoglycans , Rats , Rats, Wistar , Tamoxifen/pharmacology
18.
Nucleosides Nucleotides ; 18(4-5): 759-71, 1999.
Article in English | MEDLINE | ID: mdl-10432678

ABSTRACT

Acyclic nucleoside phosphonates such as HPMPC (cidofovir) and PMEA (adefovir) have been identified as broad-spectrum antiviral agents that are effective against herpes-, retro- and hepadnavirus infections (PMEA) and herpes-, pox-, adeno-, polyoma-, and papillomavirus infections (HPMPC). Here we show that HPMPC and PMEA also offer great potential as antitumor agents, through the induction of tumor cell differentiation (PMEA), inhibition of angiogenesis (HPMPC) and induction of apoptosis (HPMPC). In vivo tumor regressions have been noted for choriocarcinoma (PMEA) in rats, hemangioma (HPMPC) in rats and papillomatous lesions (HPMPC) in humans. Acyclic nucleoside phosphonates can be considered as a new dimension to the discipline of chemotherapy. They have a unique mode of action that is targeted at (viral or tumoral) DNA synthesis. They exhibit a pronounced and prolonged anti-viral and/or tumoral activity that can persist for days or weeks after a single administration. Most importantly, they have a uniquely broad spectrum of indications for clinical use, encompassing both DNA- and retrovirus infections, as well as various forms of cancer of both viral and non-viral origin.


Subject(s)
Adenine/analogs & derivatives , Antineoplastic Agents/pharmacology , Cytosine/analogs & derivatives , Organophosphonates , Organophosphorus Compounds/pharmacology , Adenine/pharmacology , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Choriocarcinoma/pathology , Cidofovir , Cytosine/pharmacology , Humans , Neoplasms/pathology , Neoplasms/virology , Neovascularization, Pathologic/prevention & control , Papillomaviridae/pathogenicity , Rats , Tumor Cells, Cultured
19.
Mol Pharmacol ; 56(1): 204-13, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10385702

ABSTRACT

Basic fibroblast growth factor (FGF-2) interacts with high-affinity tyrosine-kinase fibroblast growth factor receptors (FGFRs) and low-affinity heparan sulfate proteoglycans (HSPGs) in target cells. Both interactions are required for FGF-2-mediated biological responses. Here we report the FGF-2 antagonist activity of novel synthetic sulfonic acid polymers with distinct chemical structures and molecular masses (MMs). PAMPS [poly(2-acrylamido-2-methyl-1-propanesulfonic acid)], (MM approximately 7,000-10,000), PAS [poly(anetholesulfonic acid)], (MM approximately 9,000-11,000), PSS [poly(4-styrenesulfonic acid)], (MM = 70,000), and poly(vinylsulfonic acid) (MM = 2,000), inhibited FGF-2 binding to HSPGs and FGFRs in fetal bovine aortic endothelial GM 7373 cells. They also abrogated the formation of the HSPG/FGF-2/FGFR ternary complex, as evidenced by their capacity to prevent FGF-2-mediated cell-cell attachment of FGFR-1-overexpressing, HSPG-deficient Chinese hamster ovary cells to wild-type HSPG-bearing cells. Direct interaction of the polysulfonates with FGF-2 was demonstrated by their ability to protect the growth factor from proteolytic cleavage. Accordingly, molecular modeling, based on the crystal structure of the interaction of FGF-2 with a heparin hexamer, showed the feasibility of docking PAMPS into the heparin-binding domain of FGF-2. In agreement with their FGF-2-binding capacity, PSS, PAS, and PAMPS inhibited FGF-2-induced cell proliferation in GM 7373 cells and murine brain microvascular endothelial cells. The antiproliferative activity of these compounds was associated with the abrogation of FGF-2-induced tyrosine phosphorylation of FGFR-1. Moreover, the polysulfonates PSS and PAS inhibited FGF-2-induced activation of mitogen-activated protein kinase-1/2, involved in FGF-2 signal transduction. In conclusion, sulfonic acid polymers bind FGF-2 by mimicking heparin interaction. These compounds may provide a tool to inhibit FGF-2-induced endothelial cell proliferation in angiogenesis and tumor growth.


Subject(s)
Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Sulfur Compounds/pharmacology , Animals , Binding, Competitive , CHO Cells , Cattle , Cells, Cultured , Computer Simulation , Cricetinae , Endothelium/drug effects , Endothelium/metabolism , Heparin/chemistry , Heparin/pharmacology , Models, Molecular , Molecular Mimicry , Polymers/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/drug effects , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/drug effects , Signal Transduction/drug effects , Sulfonic Acids/pharmacology , Sulfur Compounds/chemistry , Trypsin/metabolism
20.
Cancer Res ; 59(10): 2376-83, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10344747

ABSTRACT

Hemangiomas represent the most frequent tumors of infancy. However, the pathogenesis of these tumors is still largely unknown, and current treatment of juvenile hemangiomas remains unsatisfactory. Here we present a novel animal model to study proliferating hemangiomas and to evaluate the effect of angiostatic compounds on their growth. Intraperitoneal (i.p.) infection of 4-day-old rats with murine polyomavirus resulted in the development of multiple cutaneous, intramuscular (i.m.), and cerebral hemangiomas with 100% frequency. Histological examination of the brain revealed the formation of immature lesions as soon as 4 days postinfection (p.i.). The subsequent exponential growth of the hemangiomas, both in number and size, was associated with severe hemorrhage and anemia. The cerebral, cutaneous, and i.m. lesions consisted of blood-filled cysts, histologically similar to human cavernous hemangiomas and stained positive for proliferating cell nuclear antigen, urokinase-type plasminogen activator, and vascular endothelial growth factor. Mature cerebral hemangiomas also expressed von Willebrand factor. Cerebral lesions caused death of the untreated animals within 19.2 +/- 1.1 days p.i. Remarkably fewer and smaller hemangiomas developed in animals that had been treated s.c. with the angiogenesis inhibitor TNP-470. Accordingly, TNP-470 (50 mg/kg), administered twice a week from 3 days p.i., significantly delayed tumor-associated mortality [mean day of death, 28.2 +/- 3.3 (P < 0.001)]. Even if therapy was initiated when cerebral hemangiomas were already macroscopically visible (i.e., 9 days p.i.), a significant delay in hemangioma-associated mortality was observed. Also, the IFN-inducer polyinosinic-polycytidylic acid caused a delay of 9 days (P < 0.005) in tumor-associated mortality when administered i.p. at 5 mg/kg, twice a week, starting at day 3 p.i. The model described here may be useful for investigating (a) the angiogenic mechanism(s) underlying hemangioma progression; and (b) the effect of anti-angiogenic compounds on vascular tumor growth.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Brain Neoplasms/prevention & control , Cell Transformation, Viral , Disease Models, Animal , Hemangioma/prevention & control , Neovascularization, Pathologic/drug therapy , Polyomavirus Infections/drug therapy , Polyomavirus/pathogenicity , Sesquiterpenes/therapeutic use , Tumor Virus Infections/drug therapy , Animals , Animals, Newborn , Body Weight/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/virology , Cyclohexanes , Disease Progression , Endothelium, Vascular/drug effects , Female , Hemangioma/drug therapy , Hemangioma/etiology , Hemangioma/virology , Humans , Infant , Infant, Newborn , Injections, Subcutaneous , Interferon Inducers/therapeutic use , Male , O-(Chloroacetylcarbamoyl)fumagillol , Poly I-C/therapeutic use , Rats , Sex Factors , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL
...