Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Pierluigi Orioli is active.

Publication


Featured researches published by Pierluigi Orioli.


Bioorganic & Medicinal Chemistry | 1999

Acid-sensitive polyethylene glycol conjugates of doxorubicin: preparation, in vitro efficacy and intracellular distribution.

Paula C.A. Rodrigues; Ulrich Beyer; Peter Schumacher; Thomas Roth; Heinz H. Fiebig; Clemens Unger; Luigi Messori; Pierluigi Orioli; D. H. Paper; Rolf Mülhaupt; Felix Kratz

Coupling anticancer drugs to synthetic polymers is a promising approach of enhancing the antitumor efficacy and reducing the side-effects of these agents. Doxorubicin maleimide derivatives containing an amide or acid-sensitive hydrazone linker were therefore coupled to alpha-methoxy-poly(ethylene glycol)-thiopropionic acid amide (MW 20000 Da), alpha,omega-bis-thiopropionic acid amide poly(ethylene glycol) (MW 20000 Da) or alpha-tert-butoxy-poly(ethylene glycol)-thiopropionic acid amide (MW 70000 Da) and the resulting polyethylene glycol (PEG) conjugates isolated through size-exclusion chromatography. The polymer drug derivatives were designed as to release doxorubicin inside the tumor cell by acid-cleavage of the hydrazone bond after uptake of the conjugate by endocytosis. The acid-sensitive PEG conjugates containing the carboxylic hydrazone bonds exhibited in vitro activity against human BXF T24 bladder carcinoma and LXFL 529L lung cancer cells with IC70 values in the range 0.02-1.5 microm (cell culture assay: propidium iodide fluorescence or colony forming assay). In contrast, PEG doxorubicin conjugates containing an amide bond between the drug and the polymer showed no in vitro activity. Fluorescence microscopy studies in LXFL 529 lung cancer cells revealed that free doxorubicin accumulates in the cell nucleus whereas doxorubicin of the acid-sensitive PEG doxorubicin conjugates is primarily localized in the cytoplasm. Nevertheless, the acid-sensitive PEG doxorubicin conjugates retain their ability to bind to calf thymus DNA as shown by fluorescence and visible spectroscopy studies. Results regarding the effect of an acid-sensitive PEG conjugate of molecular weight 20000 in the chorioallantoic membrane (CAM) assay indicate that this conjugate is significantly less embryotoxic than free doxorubicin although antiangiogenic effects were not observed.


British Journal of Cancer | 2003

Antiangiogenic properties of selected ruthenium(III) complexes that are nitric oxide scavengers.

Lucia Morbidelli; Sandra Donnini; Sandra Filippi; Luigi Messori; Francesca Piccioli; Pierluigi Orioli; Gianni Sava; Marina Ziche

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 1H-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.


Inorganica Chimica Acta | 2000

Crystal structure and solution chemistry of the cytotoxic complex 1,2-dichloro(o-phenanthroline)gold(III) chloride

Francesco Abbate; Pierluigi Orioli; Bruno Bruni; Giordana Marcon; Luigi Messori

Abstract The crystal structure of the cytotoxic complex 1,2-dichloro(o-phenanthroline)gold(III) chloride ([AuphenCl2]Cl) has been solved through single crystal X-ray diffraction methods. The complex is square planar and exhibits a quite regular geometry. Crystals of the compound belong to the space group P21/n with a=12.632(5), b=16.916(3), c=12.902(6) A, β=91.31(3)° and Z=8. The coordination of the two gold(III) ions in the asymmetric unit is completed by two chloride ions at 2.972(3) and 3.043(3) A, respectively, forming a distorted square pyramid. The behavior in solution of [AuphenCl2]Cl was further analyzed through 1H NMR spectroscopy. Results point out that the [Au(III)phen]3+ molecular fragment is stable in solution for several hours, even under physiological conditions, whereas the two chloride ligands are released within approximately 30 min after dissolution in the buffer, at 25°C. The gold(III) chromophore is easily and quickly reduced by addition of stoichiometric amounts of sodium ascorbate; metallic gold is formed and free phenanthroline liberates. The implications of these findings for the biological properties of the [Au(III)phen]3+ species are discussed.


Inorganica Chimica Acta | 1998

Gold(III) compounds as potential antitumor agents: Cytotoxicity and DNA binding properties of some selected polyamine-gold(III) complexes

Stefania Carotti; Annalisa Guerri; Teresita Mazzei; Luigi Messori; Enrico Mini; Pierluigi Orioli

Abstract Three gold(III)-polyamine complexes of different structure — ([Au(en) 2 Cl 3 , [AuCl(dien)]Cl 2 and [Au(cyclam)](ClO 4 ) 2 Cl) — were synthesized and tested for cytotoxic properties against a human tumor cell line (A2780) either sensitive (A2780/S) or resistant (A2780/R) to cisplatin. Significant cytotoxic activities were found for both [AuCl(dien)]Cl 2 and [Au(en) 2 ]Cl 3 , whereas [Au(cyclam)](ClO 4 ) 2 Cl was poorly cytotoxic. The stability of these gold(III) complexes under physiological conditions was investigated through absorption spectroscopy; their ability to bind DNA, the presumed final target for the cytotoxic action, and modify its conformation was studied through atomic absorption and circular dichroism spectroscopies. Attempts are made to define preliminary structure-function relationships for cytotoxic gold(III) complexes.


Journal of Inorganic Biochemistry | 2001

Interactions of two cytotoxic organotin(IV) compounds with calf thymus DNA

Angela Casini; Luigi Messori; Pierluigi Orioli; Marcel Gielen; Martine Kemmer; Rudolph Willem

The reactions with DNA of two antitumor active organotin(IV) compounds, the dimer of bis[(di-n-butyl 3,6-dioxaheptanoato)tin] (C(52)H(108)Sn(4)O(1) x 2H(2)O), compound 1, and tri-n-butyltin 3,6,9-trioxodecanoate (C(19)H(40)SnO(5) x 1/2H(2)O), compound 2, were analysed by circular dichroism, DNA melting experiments and gel mobility shift assays. It is found that both complexes modify only slightly the B-type circular dichroism spectroscopy (CD) spectrum of calf thymus DNA. On the other hand, both complexes were found to affect significantly the parameters of the thermally induced helix-to-coil transition. Addition of 1 or 2 to calf thymus DNA samples does not favor DNA renaturation after melting ruling out formation of interstrand crosslinks. Moreover, the effects of both compounds on plasmid DNA gel mobility were investigated. From the analysis of the present results it is inferred that both organotin(IV) compounds do interact with DNA, probably at the level of the phosphate groups.


Journal of Inorganic Biochemistry | 1997

Biological properties of two gold(III) complexes: AuCl3(Hpm) and AuCl2(pm).

Paola Calamai; Stefania Carotti; Annalisa Guerri; Luigi Messori; Enrico Mini; Pierluigi Orioli; Gian Paolo Speroni

The reactivity in solution of two recently characterized gold(III) complexes, AuCl3(Hpm) and AuCl2(pm), has been investigated in view of their potential use as anti-cancer agents. In water, both compounds undergo relatively fast hydrolysis of the bound chlorides without loss of the heterocycle ligand; the process is much faster within a physiological buffer. When the two gold(III) complexes react with proteins like albumin or transferrin, reduction of gold(III) to gold(I) and/or hydrolysis is observed. On the other hand, both complexes bind rapidly and tightly to either polynucleotides or calf thymus DNA, with gold remaining in the +3 oxidation state. Circular dichroism investigations reveal a large perturbation of DNA conformation upon gold(III) binding; preferential binding to GC sequences is shown. Cytotoxicity studies on a number of tumor cell lines demonstrate a good activity of these gold(III) complexes compared to cisplatin. However, quick hydrolysis and/or reduction of these compounds under physiological conditions may represent a severe limitation to their use.


FEBS Letters | 1999

Formation of titanium(IV) transferrin by reaction of human serum apotransferrin with titanium complexes

Luigi Messori; Pierluigi Orioli; Vivien Banholzer; Istvan Pais; Paolo Zatta

The reaction of human serum apotransferrin with titanium(IV) citrate under physiological conditions results in the formation of a specific bis‐titanium(IV) transferrin adduct (Ti2Tf hereafter) with two titanium(IV) ions loaded at the iron binding sites. The same specific Ti2Tf complex is formed by reacting apotransferrin with titanium(III) chloride and exposing the sample to air. The derivative thus obtained was characterized by spectroscopic techniques, including absorption, UV difference, circular dichroism and 13C NMR spectroscopies, and shown to be stable within the pH range 5.5–9.0. Surprisingly, the reaction of apoTf with titanium(IV) nitrilotriacetate (NTA) does not lead to formation of appreciable amounts of Ti2Tf, even after long incubation times, although some weak interactions of Ti(IV)NTA with apoTf are spectroscopically detected. Implications of the present results for a role of transferrin in the uptake, transport and delivery of soluble titanium(IV) compounds under physiological conditions are discussed.


Bioinorganic Chemistry and Applications | 2003

Gold(III) Compounds as New Family of Anticancer Drugs

Luigi Messori; Giordana Marcon; Pierluigi Orioli

Gold(III) complexes are emerging as a new class of metal complexes with outstanding cytotoxic properties and are presently being evaluated as potential antitumor agents. This renewed interest is the result of recent studies in which various gold(III) complexes have been shown to be stable under physiological conditions and to manifest relevant antiproliferative properties against selected human tumor cell lines. The pharmacological investigation of some representative gold(III) complexes has been extended to consider their effects on the cell cycle and to reveal induction of apoptosis. Remarkably, preliminary studies suggest that the interactions in vitro of gold(Ill) complexes with calf thymus DNA are weak whereas significant binding to model proteins takes place. Our findings imply that the mechanism of action of cytotoxic gold(Ill) complexes might be substantially different from that of clinically established platinum compounds.


Chemico-Biological Interactions | 2000

Cytotoxicity and DNA binding properties of a chloro glycylhistidinate gold(III) complex (GHAu)

Stefania Carotti; Giordana Marcon; Maja Marussich; Teresita Mazzei; Luigi Messori; Enrico Mini; Pierluigi Orioli

The chloro glycylhistidinate gold(III) complex (GHAu) is shown to be fairly cytotoxic towards the established A2780 ovarian carcinoma human cell line either sensitive or resistant to cisplatin. Remarkably, GHAu is far more cytotoxic than the corresponding zinc(II), palladium(II), platinum(II) and cobalt(II) complexes implying that cytotoxicity is essentially to be ascribed to the presence of a gold(III) center. Circular dichroism (CD) spectra, atomic absorption measurements and DNA melting profiles suggest that GHAu in vitro is able to bind DNA, the presumed target for several antitumor metal complexes, and to modify its conformation, even if the observed changes are generally small. Implications of these findings for the mechanism of action of cytotoxic gold(III) complexes are discussed.


Journal of Inorganic Biochemistry | 2003

Molecular structure, solution chemistry and biological properties of the novel [ImH][trans-IrCl4(Im)(DMSO)], (I) and of the orange form of [(DMSO)2H][trans-IrCl4(DMSO)2], (II), complexes

Luigi Messori; Giordana Marcon; Pierluigi Orioli; Marco Fontani; Piero Zanello; Alberta Bergamo; Gianni Sava; Pasquale Mura

The new iridium(III) complex, imidazolium[trans(DMSO,imidazole)tetrachloroiridate(III)], (I) (DMSO=dimethyl sulfoxide), and the orange form of [(DMSO)(2)H][trans(DMSO)(2)tetrachloroiridate(III)], (II) have been prepared and characterized, both in the solid state and in solution, by X-ray diffraction and by various physicochemical techniques. Single crystal X-ray diffraction studies point out that complex (II) is isomorphous to the ruthenium(III) analogue, [(DMSO)(2)H][trans-RuCl(4)(DMSO)(2)], (III). Crystallographic data are the following: a=16.028(2) A, b=24.699(3) A, c=8.262(1) A, in space group Pbca (Z=8) for (imidazolium)[trans(DMSO,imidazole)tetrachloroiridate(III)], (I); and a=9.189(2) A, b=16.511(4) A, c=14.028(3) A, beta=100.82(2) degrees in space group P2/n (Z=4) for [(DMSO)(2)H][trans(DMSO)(2)tetrachloroiridate(III)], (II). Visible absorption spectra show that both complexes are stable for several days, at pH 7.4, at room temperature. No significant chloride hydrolysis is observed, even at high temperature (70 degrees C), over 24 h. The extreme stability of these iridium(III) complexes within a physiological buffer was further assessed by (1)H NMR; in addition, cyclic voltammetry measurements evidenced a high stability of the oxidation state +3. Preliminary biological studies show that both complexes do not bind appreciably bovine serum albumin nor inhibit significantly the proliferation of representative human tumor cell lines, suggesting that hydrolysis of coordinated chlorides is a crucial feature for the biological properties and the antitumor activity of the parent ruthenium(III) complexes.

Collaboration


Dive into the Pierluigi Orioli's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bruno Bruni

University of Florence

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Enrico Mini

University of Florence

View shared research outputs
Top Co-Authors

Avatar

L. Sacconi

University of Florence

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge