51 results on '"Montaldo PG"'
Search Results
2. ANTITUMOR ACTIVITY OF SODIUM ASCORBATE AGAINST NEUROBLASTOMA CELL LINES IN VITRO
- Author
-
Carosio, R, Zuccari, G, Orienti, I, Montaldo, Pg., R. Carosio, G. Zuccari, I. Orienti, and P.G. Montaldo
- Subjects
DRUG DISCOVERY ,Neuroblastoma ,IN VITRO TEST - Abstract
Sodium ascorbate, at millimolar concentrations, induces apoptosis of neuroblastoma cell lines in vitro. The caspase cascade is activated following treatment with ascorbate, as assessed by fluorescence assay with the pan-caspase substrate z-VAD-fmk. Sodium ascorbate down-modulates CD71 (transferrin receptor) membrane expression, although overall (membrane and cytoplasmic) expression is not significantly affected. Since NB cells are strongly dependent on iron supply, it is likely that redistribution of CD71 within cellular compartments interferes with a correct iron metabolism, giving rise to apoptosis. The time- and dose-dependence, as well as the extent of the effects of sodium ascorbate on NB cells is likely to depend upon differences in its catabolism and the intensity of CD71 recycling and membrane re-expression among the different NB cell lines. Due to its lack of overt toxicity, high-dose sodium ascorbate can be considered as an additional drug in the traditional therapeutic schedules for NB.
- Published
- 2006
3. Phase I trial and pharmacokinetics of fenretinide in children with neuroblastoma
- Author
-
Garaventa, A., Luksch, R., Lo Piccolo, Ms, Cavadini, E., Montaldo, Pg, Pizzitola, MR, Luca Boni, Ponzoni, M., Decensi, A., Bernardi, B., Bellani, Ff, and Formelli, F.
4. Preparation and Evaluation of Polyvinyl alcohol-co-oleylvinyl ether Derivatives as Tumor-Specific Cytotoxic Systems
- Author
-
Isabella Orienti, Guendalina Zuccari, R. Carosio, P. G. Montaldo, Orienti I., Zuccari G., Carosio R., and Montaldo PG.
- Subjects
DYNAMIC AND STATIC LIGHT SCATTERING ,TUMOR CELL LINES NORMAL RESTING LYMPHOCYTES ,Light ,Polymers and Plastics ,Polymers ,Biocompatible Materials ,Polyvinyl alcohol ,Polyethylene Glycols ,Scattering ,chemistry.chemical_compound ,Biopolymers ,Models ,Neoplasms ,Materials Chemistry ,Scattering, Radiation ,Lymphocytes ,Drug Carriers ,Tumor ,Radiation ,Calorimetry, Differential Scanning ,Temperature ,Monomer ,Membrane ,SELECTIVE CYTOTOXICITY TOWARDS TUMOR CELLS ,Thermodynamics ,Ethers ,Vinyl alcohol ,Cell Survival ,Substituent ,Antineoplastic Agents ,Chemical ,Bioengineering ,Ether ,Calorimetry ,Differential Scanning ,Cell Line ,Biomaterials ,POLYVINYL ALCOHOL AMPHIPHILIC DERIVATIVES ,Cell Line, Tumor ,Polymer chemistry ,Humans ,Cell Membrane ,Aqueous two-phase system ,THERMODYNAMIC INSTABILITY ,Models, Chemical ,Molecular Weight ,Polyvinyl Alcohol ,Solubility ,chemistry ,Ethylene glycol - Abstract
A series of poly(vinyl alcohol) amphiphilic derivatives have been prepared to obtain polymeric aggregates in aqueous phase holding thermodynamic instability. The aim was to evaluate their ability to interact with tumor cells eliciting selective cytotoxicity. The poly(vinyl alcohol) derivatives were prepared by partial substitution of poly(vinyl alcohol) (MW 10 kDa) with both oleyl chains and poly(ethylene glycol) monoethyl ethers (PEGMEE) of different molecular weights. The substitution degree was 1.5% for the oleyl chains and 1% for the PEGMEE chains (moles of substituent per 100 mol of hydroxyvinyl monomer). The polyvinyl derivatives obtained easily dissolved in water. Dynamic and static light scattering measurements on the polymer aqueous solutions indicated the formation of polymeric aggregates characterized by low polydispersity (0.232-0.299) and mean size (218-382 nm) in the range suitable for intravenous administration. Moreover, they were characterized by different packing densities and thermodynamic instabilities driving the polymers to interact with hydrophobic membranes. Among the analyzed polymers, the poly(vinyl alcohol)-co-oleylvinyl ether substituted with triethylene glycol monoethyl ether (P10(4)) provided in solution the highest affinity for hydrophobic membranes. P10(4), moreover, was the most cytotoxic toward the tumor cell lines analyzed (neuroblastoma: SH-SY5Y, IMR-32, HTLA-230. melanoma: MZ2-MEL, RPMI7932.), while it did not appreciably alter the viability of the normal resting lymphocytes. The peculiar behavior of the P10(4) aggregates has been correlated to their high thermodynamic instability in solution due to the high packing density that triggers the polymeric aggregates to interact with hydrophobic membranes such as the tumor cell membranes, thus eliciting cytotoxicity.
- Published
- 2005
- Full Text
- View/download PDF
5. Modified polyvinylalcohol for encapsulation of all-trans-retinoic acid in polymeric micelles
- Author
-
R. Carosio, Guendalina Zuccari, Adamo Fini, Paolo G. Montaldo, Isabella Orienti, ZUCCARI G, CAROSIO R, FINI A., MONTALDO PG, and ORIENTI I.
- Subjects
Drug ,Cell Survival ,medicine.drug_class ,Drug Compounding ,media_common.quotation_subject ,Retinoic acid ,Pharmaceutical Science ,Antineoplastic Agents ,Tretinoin ,Pharmacology ,Micelle ,polyvinylalcohol substituted with oleylamine ,all-trans-retinoic acid ,spray-dried drug-polymer complexes ,solubility studies ,cytotoxicity ,chemistry.chemical_compound ,Drug Stability ,Cell Line, Tumor ,medicine ,Humans ,Retinoid ,Amines ,Particle Size ,ATRA ,Cytotoxicity ,neoplasms ,Micelles ,media_common ,Chemistry ,organic chemicals ,biological factors ,Bioavailability ,MODIFIED PVA ,Solubility ,Targeted drug delivery ,Biochemistry ,POLYMER MICELLES ,Polyvinyl Alcohol ,medicine.drug - Abstract
All-trans-retinoic acid (ATRA) is now included in many antitumor therapeutic schemes for the treatment of acute promyelocytic leukaemia, Kaposi's sarcoma, head and neck squamous cell carcinoma, ovarian carcinoma, bladder cancer and neuroblastoma. Unfortunately its poor aqueous solubility hampers its parenteral formulation. To date, there is no parenteral formulation of ATRA commercially available and oral administration of ATRA is associated with progressively diminishing ATRA levels in plasma, which is related to induction of retinoic acid-binding protein and increased drug catabolism by cytochrome P-450-mediated reaction. An ATRA formulation, obtained by complexation of the drug into polymeric micelles, might be suitable for parenteral administration overcoming these unwanted effects. To this purpose we prepared an amphiphilic polymer by polyvinylalcohol (PVA) substitution with oleyl amine at 1.5% substitution degree (mol substituent per 100 mol hydroxyvinylmonomer) and evaluated its functional properties with regard to ATRA complexation. The substituted polymer displayed ability to interact with ATRA both in aqueous solution and in the solid state following spray-drying of drug-polymer hydro-alcoholic solutions. The spray-dried complexes rapidly dissolved in water providing high levels of ATRA solubilization as a function of the drug-polymer weight ratio. The complexes characterized by 1:5 drug-polymer weight ratio provided higher levels of ATRA solubilization than 1:3 and 1:10 drug-polymer weight ratios respectively. Pre-formed polymeric micelles in water equilibrated in the presence of excess solid ATRA provided the lowest levels of solubilization. The drug release from the complexes was very slow in PBS, indicating their suitability in antitumor drug targeting where a fundamental requirement is stability towards drug release for at least 24 h, corresponding to the average circulation time period of macromolecular carriers. The cytotoxicity studies against neuroblastoma cell lines outlined increased cytotoxicity of complexed ATRA with respect to free ATRA, likely due to the increased bioavailability of the hydrophobic drug from the complex. We conclude that ATRA entrapped into self-assembling polymer micelles may be a useful parenteral ATRA formulation overcoming the unwanted pharmacological mechanism that lead to acquired retinoid resistance.
- Published
- 2005
- Full Text
- View/download PDF
6. In vitro and in vivo antitumor activity of the novel derivatized polyvinyl alcohol-based polymer P10(4)
- Author
-
Paolo G. Montaldo, Isabella Orienti, R. Carosio, Lizzie Raffaghello, Guendalina Zuccari, Raffaghello L, Zuccari G, Carosio R, Orienti I, and Montaldo PG.
- Subjects
Cancer Research ,Time Factors ,Nude ,Apoptosis ,Animals ,Antineoplastic Agents ,Cell Line, Tumor ,Disease Models, Animal ,Dose-Response Relationship, Drug ,Drug Screening Assays, Antitumor ,Female ,Humans ,Injections, Intravenous ,Melanoma ,Mice ,Mice, Nude ,Neuroblastoma ,Polyethylene Glycols ,Polyvinyl Alcohol ,Structure-Activity Relationship ,Tissue Distribution ,Transplantation, Heterologous ,Xenograft Model Antitumor Assays ,Drug Screening Assays ,Caspase ,Heterologous ,Tumor ,biology ,Chemistry ,Biological activity ,Oncology ,Biochemistry ,DNA fragmentation ,Drug ,Intravenous ,Cell Line ,Injections ,Dose-Response Relationship ,In vivo ,medicine ,Clonogenic assay ,Transplantation ,Animal ,Antitumor ,medicine.disease ,In vitro ,Disease Models ,Cancer research ,biology.protein - Abstract
Purpose: The major limitation to successful chemotherapy of neuroblastoma is the toxicity of traditional antitumor drugs. Hence, less toxic and more effective drugs are to be found, and novel formulations of conventional compounds allowing a more favorable biodistribution should be sought for. In an attempt to pursue this task, we recently synthesized an amphiphilic polymer based on a polyvinyl alcohol backbone [P10(4)]. Experimental Design: The cytotoxic activity of P10(4) was evaluated both in vitro on neuroblastoma and melanoma cell lines and in vivo in pseudometastatic neuroblastoma models. Apoptosis was assessed by morphology, cytofluorimetric analysis of DNA content, and DNA fragmentation assay. Caspases activation was investigated by kits specific for caspase-1, caspase-2, caspase-3, caspase-4, caspase-6, caspase-7, caspase-8, caspase-9, caspase-10, and caspase-13. Colony formation was evaluated by soft agar assay. Results: P10(4) exerted a potent cytotoxic activity on different neuroblastoma and melanoma cell lines through induction of both extrinsic and intrinsic caspase cascades and subsequent apoptosis. Moreover, the clonogenic potential of cells that survived P10(4) treatment was strongly reduced. Next, we tested the effects of P10(4) in nude mice injected with both a human and a murine neuroblastoma cell lines i.v. P10(4) significantly increased the life span and the long-term survival of treated mice over controls. No side effects were observed, even at doses higher than those used for therapeutic purposes. Conclusions: Our data suggest that P10(4) holds promise as an anticancer compound and, because of its lack of interaction with DNA, is unlikely to give rise to drug resistance.
- Published
- 2006
7. Enhanced anti-neuroblastoma activity of a fenretinide complexed form after intravenous administration.
- Author
-
Carosio R, Pistoia V, Orienti I, Formelli F, Cavadini E, Mangraviti S, Montaldo PG, Ognio E, Emionite L, and Zuccari G
- Subjects
- Animals, Antineoplastic Agents pharmacokinetics, Biological Availability, Cell Division drug effects, Cell Line, Tumor, Disease Models, Animal, Female, Fenretinide pharmacokinetics, Humans, Infusions, Intravenous, Mice, Mice, Nude, Neuroblastoma metabolism, Neuroblastoma pathology, Antineoplastic Agents administration & dosage, Apoptosis drug effects, Drug Delivery Systems, Fenretinide administration & dosage, Neuroblastoma drug therapy
- Abstract
Objectives: The major limitation to successful chemotherapy of neuroblastoma (NB) is the toxicity and the poor bioavailability of traditional drugs., Methods: We synthesised an amphiphilic dextrin derivative (DX-OL) able to host fenretinide (4-HPR) by complexation. In this study, we have investigated the effects of 4-HPR-loaded amphipilic dextrin (DX-OL/4-HPR) in comparison with 4-HPR alone both in vitro on human NB cells and in vivo in pseudometastatic NB models. The haemolysis assay was used as a measure of the potential damage caused by the pharmaceutical formulation in vivo. Pharmacokinetic experiments were performed to assess drug plasma levels in mice treated with free or complexed 4-HPR., Key Findings: DX-OL/4-HPR exerted a more potent cytotoxic activity on NB cells. Complexed 4-HPR significantly increased the proportion of sub-G1 cells with respect to free 4-HPR. Dextrin derivatives showed no haemolytic activity, indicating their suitability for parenteral administration. DX-OL/4-HPR increased the lifespan and the long-term survival of treated mice over controls. The analysis of drug plasma levels indicates that the complexed drug has a higher AUC due to a reduced clearance from the blood., Conclusions: Our data suggest that DX-OL/4-HPR is an injectable formulation that is able to improve drug aqueous solubility and bioavailability., (© 2011 The Authors. JPP © 2011 Royal Pharmaceutical Society.)
- Published
- 2012
- Full Text
- View/download PDF
8. Recent advances in targeted anti-vasculature therapy: the neuroblastoma model.
- Author
-
Pastorino F, Di Paolo D, Loi M, Becherini P, Caffa I, Zorzoli A, Marimpietri D, Carosio R, Perri P, Montaldo PG, Brignole C, Pagnan G, Ribatti D, Allen TM, and Ponzoni M
- Subjects
- Angiogenesis Inhibitors therapeutic use, Animals, Antineoplastic Agents therapeutic use, Disease Progression, Drug Delivery Systems, Humans, Neovascularization, Pathologic drug therapy, Neuroblastoma blood supply, Neuroblastoma pathology, Prognosis, Angiogenesis Inhibitors pharmacology, Antineoplastic Agents pharmacology, Neuroblastoma drug therapy
- Abstract
Novel anti-vasculature strategies that are emerging for the treatment of cancer and for the inhibition of angiogenesis may be a promising new tool for the adjuvant therapy of malignant tumours. Over the last fifteen years, several reports have been published concerning the relationship between tumour progression and angiogenesis in experimental models of neuroblastoma in vitro and in vivo. Moreover, a high vascular index in neuroblastoma correlates with poor prognosis, suggesting dependence of aggressive tumour growth on active angiogenesis. Here, we present an overview of the most recent advances in anti-vasculature therapy of neuroblastoma, and describe some preclinical results as well as future perspectives.
- Published
- 2009
- Full Text
- View/download PDF
9. Improvement of aqueous solubility of fenretinide and other hydrophobic anti-tumor drugs by complexation with amphiphilic dextrins.
- Author
-
Orienti I, Zuccari G, Carosio R, and Montaldo PG
- Subjects
- Antineoplastic Agents pharmacology, Cell Line, Tumor, Dextrins pharmacology, Drug Carriers chemistry, Drug Carriers pharmacology, Fenretinide pharmacology, Humans, Solubility, Surface-Active Agents pharmacology, Antineoplastic Agents chemistry, Dextrins chemistry, Fenretinide chemistry, Hydrophobic and Hydrophilic Interactions, Surface-Active Agents chemistry
- Abstract
This study relates to the preparation of a series of amphiphilic dextrins and their evaluation as complexing agents for anti-tumor hydrophobic drugs such as fenretinide, paclitaxel, etoposide, and camptothecin. The amphiphilic dextrins were obtained by conjugation of low molecular weight dextrin (average molecular weight 1670, average polymerization degree 9.33 glucose monomer) with hydrocarbon chains at substitution degree of about 0.1 mole hydrocarbon chain per mole of glucose monomer, as confirmed by 1H-NMR spectra. The conjugates were highly soluble in water and dissolved with formation of nano-aggregates endowed with hydrophobic inner cores able to host hydrophobic drugs by complexation. Complexation raised hydrophobic drugs aqueous solubility; the best results were obtained with fenretinide. Solid complexes with fenretinide were prepared by using three different approaches: the kneading method, the co-solubilisation method, and the co-precipitation method. Kneading method provided the complexes endowed with the best functional properties. Thermogravimetric analysis on solid samples suggested a notable thermal stability up to 300 degrees C for both the conjugated dextrins and the solid complexes. In differential scanning calorimetry profiles no significant differences were observed among amphiphilic dextrins and complexed drug, indicating that the guest molecule exists in an amorphous state in the solid matrices. Particle size analysis confirmed the dimensional suitability of the complexes for parenteral administration. Moreover, sustained drug release, in vitro, has been observed from all the complexes analyzed. Regarding the biological effects, the cytotoxicity of complexed fenretinide towards HTLA-230 neuroblastoma cell line was always higher than the free drug, suggesting that complexation increased drug bioavailability. These findings, taken together, indicated that these biodegradable, self-assembling dextrin conjugates may be regarded as new potential complexing agents for hydrophobic drugs and, in particular, for fenretinide, to increase drug solubility, bioavailability, and thus therapeutic efficacy.
- Published
- 2009
- Full Text
- View/download PDF
10. Micellar complexes of all-trans retinoic acid with polyvinylalcohol-nicotinoyl esters as new parenteral formulations in neuroblastoma.
- Author
-
Zuccari G, Bergamante V, Carosio R, Gotti R, Montaldo PG, and Orienti I
- Subjects
- Chemistry, Pharmaceutical, Drug Administration Routes, Drug Carriers administration & dosage, Drug Carriers chemistry, Esters chemistry, Humans, Micelles, Neuroblastoma pathology, Polyvinyl Alcohol chemistry, Solubility, Tretinoin chemistry, Tumor Cells, Cultured, Antineoplastic Agents administration & dosage, Cell Proliferation drug effects, Infusions, Parenteral, Neuroblastoma therapy, Tretinoin administration & dosage
- Abstract
All-trans-retinoic acid (ATRA) is now included in many antitumor therapeutic schemes for the treatment of acute promyelocytic leukemia, Kaposi's sarcoma, head and neck squamous cell carcinoma, ovarian carcinoma, bladder cancer, and neuroblastoma. Unfortunately, its poor aqueous solubility hampers its parenteral formulation, whereas oral administration of ATRA is associated with progressively diminishing drug levels in plasma, which is related to induction of retinoic acid-binding proteins and increased drug catabolism by cytochrome P450-mediated reactions. An ATRA formulation, obtained by complexation of the drug into polymeric micelles, might be suitable for parenteral administration overcoming these unwanted effects. To this purpose, amphiphilic polymers were prepared by polyvinylalcohol (PVA) partial esterification with nicotinoyl moieties and their functional properties evaluated with regard to ATRA complexation. The physicochemical characteristics of the polymers and the complexes were analyzed by 1H-NMR, Dynamic Light Scattering (DLS), Capillary Electophoresis (CE), and were correlated with the complex ability to improve the drug solubilization and release the free drug in an aqueous environment. Subsequently, the best complex, providing the highest ATRA solubilization and release, was evaluated in vitro to test its cytotoxicity towards neuroblastoma cell lines. The PVA substitution degree calculated from 1H-NMR was found to be 5.0%, 8.2%, 15.3% (nicotinoyl moiety:PVA monomer molar ratio), while capillary electrophoresis analysis on the complexes revealed that the drug loadings were 0.95%, 1.20%, 4.76% (ATRA:polymer w:w) for PVA substitution degrees of 5.0%, 8.2%, and 15.3%, respectively. Complexation strongly increased ATRA aqueous solubility, which reached 1.20 +/- 0.25 mg/mL. The DLS measurements of the polymers and the complexes in aqueous solutions revealed mean sizes always below 400 nm, low polydispersity (min 0.202 +/- 0.013, max 0.450 +/- 0.032), and size almost unaffected by concentration. Drug fractional release did not exceed 8% after 48 h. The cytotoxicity studies against neuroblastoma cell lines outlined a significant growth inhibition effect of complexed ATRA with respect to free ATRA. These data suggest that the systems analyzed may be suitable carriers for parenteral administration of ATRA and other hydrophobic antitumor drugs, where the carriers are required to improve drug aqueous solubility and delay drug release almost after their accumulation in solid tumors.
- Published
- 2009
- Full Text
- View/download PDF
11. Fenretinide-polyvinylalcohol conjugates: new systems allowing fenretinide intravenous administration.
- Author
-
Orienti I, Zuccari G, Bergamante V, Carosio R, Gotti R, Cilli M, and Montaldo PG
- Subjects
- Animals, Biological Availability, Cell Line, Tumor, Cell Proliferation, Dimethyl Sulfoxide chemistry, Humans, Infusions, Intravenous, Mice, Mice, Nude, Models, Chemical, Neoplasm Metastasis, Neuroblastoma metabolism, Solubility, Antineoplastic Agents administration & dosage, Drug Delivery Systems, Fenretinide administration & dosage, Fenretinide chemistry, Polyvinyl Alcohol chemistry
- Abstract
N-(4-hydroxyphenyl)retinamide (fenretinide, 4-HPR) has been shown to be active toward many tumors without appreciable side effects. However its in vitro activity does not match a correspondent efficacy in vivo. The main reason is that the drug's hydrophobicity hinders its bioavailability in the body fluids. Even if the drug is previously dissolved in organic solvents, such as ethanol or DMSO, the subsequent dilution in body fluids trigger its precipitation in fine aggregates characterized by very low dissolution efficiency, never reaching amounts suitable for therapeutic response. To date no intravenous formulation of 4-HPR exists on the market. The 4-HPR linkage to a hydrophilic polymer by a covalent bond easily hydrolyzable in aqueous environment is expected to increase the drug's aqueous solubility, providing the free drug after hydrolysis of the covalent bond. This may be a useful tool for the preparation of aqueous intravenous formulations of 4-HPR. For this purpose, we linked 4-HPR to polyvinylalcohol (PVA) by a carbonate bond at different drug/hydroxy vinyl monomer molar ratios. We demonstrated that conjugation increased 4-HPR aqueous solubility and strongly inhibited neuroblastoma cell proliferation. In addition, in an in vivo neuroblastoma metastatic model, we obtained a significant antitumor effect as a consequence of the improved drug bioavailability.
- Published
- 2007
- Full Text
- View/download PDF
12. Sodium ascorbate induces apoptosis in neuroblastoma cell lines by interfering with iron uptake.
- Author
-
Carosio R, Zuccari G, Orienti I, Mangraviti S, and Montaldo PG
- Subjects
- Annexin A5 metabolism, Caspases metabolism, Cell Cycle drug effects, Cell Line, Tumor drug effects, Cell Proliferation drug effects, Humans, Membrane Potential, Mitochondrial drug effects, Neuroblastoma metabolism, Receptors, Transferrin genetics, Receptors, Transferrin metabolism, Trypan Blue, Antioxidants pharmacology, Apoptosis drug effects, Ascorbic Acid pharmacology, Iron metabolism, Neuroblastoma pathology
- Abstract
Background: Neuroblastoma (NB) is an extra-cranial solid tumour of childhood. In spite of the good clinical response to first-line therapy, complete eradication of NB cells is rarely achieved. Thus, new therapeutic strategies are needed to eradicate surviving NB cells and prevent relapse. Sodium ascorbate has been recently reported to induce apoptosis of B16 melanoma cells through down-regulation of the transferrin receptor, CD71. Since NB and melanoma share the same embryologic neuroectodermal origin, we used different human NB cell lines to assess whether the same findings occurred., Results: We could observe dose- and time-dependent induction of apoptosis in all NB cell lines. Sodium ascorbate decreased the expression of CD71 and caused cell death within 24 h. An increase in the global and specific caspase activity took place, as well as an early loss of the mitochondrial transmembrane potential. Moreover, intracellular iron was significantly decreased after exposure to sodium ascorbate. Apoptotic markers were reverted when the cells were pretreated with the iron donor ferric ammonium citrate (FAC), further confirming that iron depletion is responsible for the ascorbate-induced cell death in NB cells., Conclusion: Sodium ascorbate is highly toxic to neuroblastoma cell lines and the specific mechanism of vitamin C-induced apoptosis is due to a perturbation of intracellular iron levels ensuing TfR-downregulation.
- Published
- 2007
- Full Text
- View/download PDF
13. Enhancement of oleyl alcohol anti tumor activity through complexation in polyvinylalcohol amphiphilic derivatives.
- Author
-
Orienti I, Zuccari G, Bergamante V, Fini A, Carosio R, and Montaldo PG
- Subjects
- Antineoplastic Agents chemistry, Calorimetry, Differential Scanning, Cell Line, Tumor, Cell Survival drug effects, Chemistry, Pharmaceutical, Dose-Response Relationship, Drug, Fatty Alcohols chemistry, Humans, Light, Microscopy, Electron, Scanning, Pharmaceutical Solutions, Scattering, Radiation, Solubility, Solvents chemistry, Surface-Active Agents chemistry, Time Factors, Water chemistry, Antineoplastic Agents pharmacology, Fatty Alcohols pharmacology, Polyvinyl Alcohol chemistry
- Abstract
Oleyl alcohol was complexed with new amphiphilic polyvinylalcohol derivatives with the aim of increasing its aqueous solubility, thus improving bioavailability and favoring its antitumor activity. Water-soluble amphiphilic polymers were prepared by polyvinyl alcohol (PVA) substitution with oleyl chains through a succinyl spacer at 2% and 3% substitution degree. The complexes were obtained by spray-drying hydroalcoholic solutions of the substituted polymers and free oleyl alcohol at different weight ratios (3:1; 5:1; 10:1 w/w). The main physicochemical characteristics of the complexes were analyzed and correlated to the cytotoxic activity of oleyl alcohol toward tumor cell lines. The complexes strongly increased the aqueous solubility of oleyl alcohol and provided oleyl alcohol release in the presence of extractive conditions (simulating in vivo absorption). The complexes obtained by 10:1 polymer:fatty alcohol weight ratio offered higher release rates than the 5:1 and 3:1 ratios, respectively. Complexation also increased oleyl alcohol cytotoxicity toward tumor cells due to increased availability of the active molecule in the aqueous phase. Pure polymers were found to be biocompatible and no toxic effect was detected up to the highest concentration used in the present study (500 mu g/ml). The complexation of oleyl alcohol with the polymers analyzed here efficiently increased the availability of the fatty alcohol in aqueous environment. The enhanced cytotoxicity toward tumor cells of the complexed oleyl alcohol and the polymer biocompatibility make these amphiphilic PVA derivatives interesting candidates for soluble pharmaceutical formulations containing hydrophobic drugs whose therapeutic potential is often underestimated due to unsuitable levels of their aqueous solubilization.
- Published
- 2007
- Full Text
- View/download PDF
14. Amphiphilic poly(vinyl alcohol) derivatives as complexing agents for fenretinide.
- Author
-
Orienti I, Zuccari G, Bergamante V, Mileo E, Lucarini M, Carosio R, and Montaldo PG
- Subjects
- Animals, Antineoplastic Agents pharmacology, Cell Survival drug effects, Dose-Response Relationship, Drug, Electron Spin Resonance Spectroscopy, Fenretinide pharmacology, Humans, Mice, Mice, Nude, Tumor Cells, Cultured, Antineoplastic Agents chemistry, Fenretinide chemistry, Polyvinyl Alcohol chemistry
- Abstract
Poly(vinyl alcohol) (PVA) substituted with oleyl chains and tetraethyleneglycol monoethyl ether chains (TEGMEE) at 1.5% and 1% degrees of substitution respectively (mol of substituent to mol of hydroxyvinyl monomer) has previously been shown to self-assemble in water, providing aggregates selectively cytotoxic toward tumor cells vs normal cells. These polymers have also been shown to increase the long-term survival of nude mice injected with both human and murine neuroblastoma cell lines. In the present work, we changed the substitution degree of the oleyl chains on the poly(vinyl alcohol) backbone and maintained constant at 1% the degree of TEGMEE substitution. We evaluated the main physicochemical characteristics of the final polymers, their cytotoxicity toward tumor cells, and their complexing ability for hydrophobic molecules. The aim was to investigate the possibility of improving intrinsic antitumor efficacy of the polymer by changing the degree of oleyl chain substitution and further increase activity by complexation with antitumor drugs. The polymers were prepared at oleyl chain substitution degrees ranging from 0.5 to 3% (mol of substituent to mol of hydroxyvinyl monomer). The most active was again the 1.5% substituted polymer. It was further characterized by exhibiting the highest complexing ability toward hydrophobic molecules allowing the formation of a complex with fenretinide (HPR). The polymer-HPR complex was stable in aqueous environment and released the free drug prevalently in the presence of fluid hydrophobic phases. It was cytotoxic toward tumor cells with minimal activity toward normal cells. Antitumor activity exceeded that of the separate complex components resulting from the concomitant effect of the polymer and the HPR solubilized by complexation.
- Published
- 2006
- Full Text
- View/download PDF
15. In vitro and in vivo antitumor activity of the novel derivatized polyvinyl alcohol-based polymer P10(4).
- Author
-
Raffaghello L, Zuccari G, Carosio R, Orienti I, and Montaldo PG
- Subjects
- Animals, Antineoplastic Agents administration & dosage, Antineoplastic Agents chemistry, Apoptosis drug effects, Cell Line, Tumor, Disease Models, Animal, Dose-Response Relationship, Drug, Drug Screening Assays, Antitumor, Female, Humans, Injections, Intravenous, Mice, Mice, Nude, Neuroblastoma secondary, Polyethylene Glycols chemistry, Polyvinyl Alcohol administration & dosage, Polyvinyl Alcohol chemistry, Structure-Activity Relationship, Time Factors, Tissue Distribution, Transplantation, Heterologous, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Melanoma drug therapy, Neuroblastoma drug therapy, Polyethylene Glycols pharmacology, Polyvinyl Alcohol analogs & derivatives, Polyvinyl Alcohol pharmacology
- Abstract
Purpose: The major limitation to successful chemotherapy of neuroblastoma is the toxicity of traditional antitumor drugs. Hence, less toxic and more effective drugs are to be found, and novel formulations of conventional compounds allowing a more favorable biodistribution should be sought for. In an attempt to pursue this task, we recently synthesized an amphiphilic polymer based on a polyvinyl alcohol backbone [P10(4)]., Experimental Design: The cytotoxic activity of P10(4) was evaluated both in vitro on neuroblastoma and melanoma cell lines and in vivo in pseudometastatic neuroblastoma models. Apoptosis was assessed by morphology, cytofluorimetric analysis of DNA content, and DNA fragmentation assay. Caspases activation was investigated by kits specific for caspase-1, caspase-2, caspase-3, caspase-4, caspase-6, caspase-7, caspase-8, caspase-9, caspase-10, and caspase-13. Colony formation was evaluated by soft agar assay., Results: P10(4) exerted a potent cytotoxic activity on different neuroblastoma and melanoma cell lines through induction of both extrinsic and intrinsic caspase cascades and subsequent apoptosis. Moreover, the clonogenic potential of cells that survived P10(4) treatment was strongly reduced. Next, we tested the effects of P10(4) in nude mice injected with both a human and a murine neuroblastoma cell lines i.v. P10(4) significantly increased the life span and the long-term survival of treated mice over controls. No side effects were observed, even at doses higher than those used for therapeutic purposes., Conclusions: Our data suggest that P10(4) holds promise as an anticancer compound and, because of its lack of interaction with DNA, is unlikely to give rise to drug resistance.
- Published
- 2006
- Full Text
- View/download PDF
16. Preparation and evaluation of polyvinyl alcohol-co-oleylvinyl ether derivatives as tumor-specific cytotoxic systems.
- Author
-
Orienti I, Zuccari G, Carosio R, and Montaldo PG
- Subjects
- Antineoplastic Agents administration & dosage, Antineoplastic Agents pharmacology, Biocompatible Materials chemistry, Calorimetry, Differential Scanning, Cell Line, Tumor, Cell Membrane metabolism, Cell Survival, Drug Carriers chemistry, Ethers chemistry, Humans, Light, Lymphocytes metabolism, Models, Chemical, Molecular Weight, Polymers chemistry, Scattering, Radiation, Solubility, Temperature, Thermodynamics, Biopolymers chemistry, Neoplasms drug therapy, Polyethylene Glycols chemistry, Polyvinyl Alcohol chemistry
- Abstract
A series of poly(vinyl alcohol) amphiphilic derivatives have been prepared to obtain polymeric aggregates in aqueous phase holding thermodynamic instability. The aim was to evaluate their ability to interact with tumor cells eliciting selective cytotoxicity. The poly(vinyl alcohol) derivatives were prepared by partial substitution of poly(vinyl alcohol) (MW 10 kDa) with both oleyl chains and poly(ethylene glycol) monoethyl ethers (PEGMEE) of different molecular weights. The substitution degree was 1.5% for the oleyl chains and 1% for the PEGMEE chains (moles of substituent per 100 mol of hydroxyvinyl monomer). The polyvinyl derivatives obtained easily dissolved in water. Dynamic and static light scattering measurements on the polymer aqueous solutions indicated the formation of polymeric aggregates characterized by low polydispersity (0.232-0.299) and mean size (218-382 nm) in the range suitable for intravenous administration. Moreover, they were characterized by different packing densities and thermodynamic instabilities driving the polymers to interact with hydrophobic membranes. Among the analyzed polymers, the poly(vinyl alcohol)-co-oleylvinyl ether substituted with triethylene glycol monoethyl ether (P10(4)) provided in solution the highest affinity for hydrophobic membranes. P10(4), moreover, was the most cytotoxic toward the tumor cell lines analyzed (neuroblastoma: SH-SY5Y, IMR-32, HTLA-230. melanoma: MZ2-MEL, RPMI7932.), while it did not appreciably alter the viability of the normal resting lymphocytes. The peculiar behavior of the P10(4) aggregates has been correlated to their high thermodynamic instability in solution due to the high packing density that triggers the polymeric aggregates to interact with hydrophobic membranes such as the tumor cell membranes, thus eliciting cytotoxicity.
- Published
- 2005
- Full Text
- View/download PDF
17. Modified polyvinylalcohol for encapsulation of all-trans-retinoic acid in polymeric micelles.
- Author
-
Zuccari G, Carosio R, Fini A, Montaldo PG, and Orienti I
- Subjects
- Amines chemistry, Antineoplastic Agents pharmacology, Cell Line, Tumor, Cell Survival drug effects, Drug Stability, Humans, Micelles, Particle Size, Solubility, Tretinoin pharmacology, Antineoplastic Agents chemistry, Drug Compounding, Polyvinyl Alcohol chemistry, Tretinoin chemistry
- Abstract
All-trans-retinoic acid (ATRA) is now included in many antitumor therapeutic schemes for the treatment of acute promyelocytic leukaemia, Kaposi's sarcoma, head and neck squamous cell carcinoma, ovarian carcinoma, bladder cancer and neuroblastoma. Unfortunately its poor aqueous solubility hampers its parenteral formulation. To date, there is no parenteral formulation of ATRA commercially available and oral administration of ATRA is associated with progressively diminishing ATRA levels in plasma, which is related to induction of retinoic acid-binding protein and increased drug catabolism by cytochrome P-450-mediated reaction. An ATRA formulation, obtained by complexation of the drug into polymeric micelles, might be suitable for parenteral administration overcoming these unwanted effects. To this purpose we prepared an amphiphilic polymer by polyvinylalcohol (PVA) substitution with oleyl amine at 1.5% substitution degree (mol substituent per 100 mol hydroxyvinylmonomer) and evaluated its functional properties with regard to ATRA complexation. The substituted polymer displayed ability to interact with ATRA both in aqueous solution and in the solid state following spray-drying of drug-polymer hydro-alcoholic solutions. The spray-dried complexes rapidly dissolved in water providing high levels of ATRA solubilization as a function of the drug-polymer weight ratio. The complexes characterized by 1:5 drug-polymer weight ratio provided higher levels of ATRA solubilization than 1:3 and 1:10 drug-polymer weight ratios respectively. Pre-formed polymeric micelles in water equilibrated in the presence of excess solid ATRA provided the lowest levels of solubilization. The drug release from the complexes was very slow in PBS, indicating their suitability in antitumor drug targeting where a fundamental requirement is stability towards drug release for at least 24 h, corresponding to the average circulation time period of macromolecular carriers. The cytotoxicity studies against neuroblastoma cell lines outlined increased cytotoxicity of complexed ATRA with respect to free ATRA, likely due to the increased bioavailability of the hydrophobic drug from the complex. We conclude that ATRA entrapped into self-assembling polymer micelles may be a useful parenteral ATRA formulation overcoming the unwanted pharmacological mechanism that lead to acquired retinoid resistance.
- Published
- 2005
- Full Text
- View/download PDF
18. Modified doxorubicin for improved encapsulation in PVA polymeric micelles.
- Author
-
Orienti I, Zuccari G, Fini A, Rabasco AM, Carosio R, Raffaghello L, and Montaldo PG
- Subjects
- Doxorubicin chemistry, Micelles, Doxorubicin administration & dosage, Polyvinyl Alcohol administration & dosage
- Abstract
Polyvinylalcohol, partially substituted with lipophilic acyl chains, generates polymeric micelles in aqueous phase, containing a hydrophobic core able to encapsulate lipophilic drugs. Two types of polymers were obtained by conjugation of polyvinylalcohol with oleoyl or linoleoyl chains as pendant groups. The polymers, at a substitution degree of approximately 1%, are soluble in water and form polymeric micelles whose size increases with polymer concentration. Doxorubicin was hydrophobized, by linking an oleoyl chain via amide bond, to make the drug more similar to the substituted polymers and promote its encapsulation into the inner core of the micelles. The properties of the drug-polymer systems were evaluated in solution by dynamic light scattering technique and correlated to the physicochemical characteristics of the drug and the substituted polymers. Solubilization tests revealed that the similarity of the chain, in both the polymer and the drug, promotes better drug encapsulation in the oleoyl than linoleoyl derivative. The drug-polymer systems are stable in phosphate buffer saline (pH 7.4) at 37 degrees C, and the release of the drug is activated by the presence of the proteolytic enzyme pronase-E. The enzyme activated drug release and the size of the polymeric micelles, compatible with the pore dimensions of the tumor vessels, make these systems interesting for targeting lipophilic drugs to solid tumors, where the proteolytic enzyme concentration strongly raises with respect to the other body compartments.
- Published
- 2005
- Full Text
- View/download PDF
19. Animal age-, dose- and cell line-dependent growth of human neuroblastoma in nude mice. A statistical analysis.
- Author
-
Mariottini GL, Montaldo PG, and Copello F
- Subjects
- Age Factors, Animals, Cell Line, Tumor, Humans, Mice, Mice, Nude, Neoplasm Transplantation, Neoplasms, Experimental pathology, Neuroblastoma pathology
- Abstract
Cells lines from human neuroblastoma (NB) and T/lymphoma (T-L) were injected subcutaneously (sc) in female CD1 nu/nu athymic nude mice. Results obtained after the observation of tumour growth were statistically analyzed by SAS. The following four parameters were considered: 1) dose of injected cells, 2) type of injected tumour (NB or T-L), 3) age of mice after individuation of three groups of animals (group A, 4-9 weeks old, group B, 9-20 weeks old, group C, > 20 weeks old), 4) injected cell line within the same tumour type. Latency time (LT), corresponding to the interval between cell inoculum and the appearance of a 5 mm diameter subcutaneous mass, and survival time (ST), corresponding to the interval between cell inoculum and the appearance of a 20 mm diameter subcutaneous tumour mass, were considered to evaluate tumour growth. Results showed that mass progression is affected by the number of injected cells and both LT and ST are age- and dose-dependent; furthermore, significant differences were recorded by using different NB and T-L cell lines. Group C showed longer LT than other groups; group B animals showed a statistically significant longer ST than groups A and C (p < 0.001). Our results indicate that growth of human NB in athymic mice is faster in young animals, which also show a significantly poorer prognosis, while better ST was observed in old and middle-aged animals. Results show statistically significant differences of both LT and ST in animals differing in age and in animals inoculated with different cell amounts. These results seem not to be related with biological properties of NB cells too, since neither the occurrence of MYCN amplification nor chromosome 1p deletion significantly modified such behaviour.
- Published
- 2004
20. Development of a real-time polymerase chain reaction assay for prediction of the uptake of meta-[(131)I]iodobenzylguanidine by neuroblastoma tumors.
- Author
-
Carlin S, Mairs RJ, McCluskey AG, Tweddle DA, Sprigg A, Estlin C, Board J, George RE, Ellershaw C, Pearson AD, Lunec J, Montaldo PG, Ponzoni M, van Eck-Smit BL, Hoefnagel CA, van den Brug MD, Tytgat GA, and Caron HN
- Subjects
- Biopsy, Brain Neoplasms drug therapy, Breast Neoplasms drug therapy, Cell Line, Tumor, DNA Primers pharmacology, DNA, Complementary metabolism, Humans, Norepinephrine Plasma Membrane Transport Proteins, Polymerase Chain Reaction, Prognosis, RNA, Messenger metabolism, Symporters metabolism, 3-Iodobenzylguanidine pharmacokinetics, Antineoplastic Agents pharmacokinetics, Neuroblastoma drug therapy, Reverse Transcriptase Polymerase Chain Reaction methods
- Abstract
Purpose: The suitability of neuroblastoma patients for therapy using radiolabeled meta-iodobenzylguanidine (MIBG) is determined by scintigraphy after the administration of a tracer dose of radioiodinated MIBG whose uptake is dependent upon the cellular expression of the noradrenaline transporter (NAT). As a possible alternative to gamma camera imaging, we developed a novel molecular assay of NAT expression. mRNA extracted from neuroblastoma biopsy samples, obtained retrospectively, was reverse transcribed, and NAT-specific cDNA was quantified by real-time PCR, referenced against the expression of the housekeeping gene glyceraldehyde-3-phosphate dehydrogenase. EXPERMENTAL DESIGN: Tumor specimens from 54 neuroblastoma patients were analyzed using real-time PCR, and NAT expression was compared with the corresponding diagnostic scintigrams., Results: Forty-eight of 54 (89%) of tumors showed MIBG uptake by scintigraphy. NAT expression was found to be significantly associated with MIBG uptake (P < 0.0001, Fisher's exact test). None of the samples from the six tumors that failed to concentrate MIBG expressed detectable levels of the NAT (specificity = 1.0). However, of the 48 MIBG uptake-positive tumors, only 43 (90%) expressed NAT (sensitivity = 0.9). The real-time PCR test has a positive predictive value of 1.0 but a negative predictive value of 0.55., Conclusions: The results indicate that whereas this method has substantial ability to predict the capacity of neuroblastoma tumors to accumulate MIBG, confirmation is required in prospective studies to determine more accurately the predictive strength of the test and its role in the management of patients with neuroblastoma.
- Published
- 2003
21. Anti-GD2 monoclonal antibody immunotherapy: a promising strategy in the prevention of neuroblastoma relapse.
- Author
-
Raffaghello L, Marimpietri D, Pagnan G, Pastorino F, Cosimo E, Brignole C, Ponzoni M, and Montaldo PG
- Subjects
- Animals, Humans, Mice, Neoplasm Recurrence, Local immunology, Neoplasm Recurrence, Local prevention & control, Neuroblastoma prevention & control, Survival Rate, Tumor Cells, Cultured, Antibodies, Monoclonal therapeutic use, Gangliosides immunology, Immunotherapy, Neoplasm Recurrence, Local therapy, Neuroblastoma therapy
- Abstract
In spite of the satisfactory frequency of clinical response to first-line therapy in neuroblastoma (NB), complete eradication of NB cells is rarely achieved. As a consequence, the majority of patients with advanced stage NB undergo relapse, which is often resistant to conventional treatment and rapidly overwhelming. Thus, after induction of the apparent remission, new therapeutic strategies are needed to completely eradicate the small number of surviving NB cells and to prevent relapse. We explored the potential of different doses of the anti-GD2 monoclonal antibody (mAb) 14G2a in an experimental metastatic model where a limited number of HTLA-230 human NB cells are injected i.v. into nude mice, leading to extensive metastases and death of animals within 7-8 weeks. Treatment with 14G2a mAb (1-4 mg/kg cumulative dose given as five i.v. daily administrations) dramatically reduced the metastatic spread of NB cells and prolonged the long-term survival of treated mice in a dose-dependent manner. Neither macrophages nor NK cells appeared to contribute to the protective effect of antibody treatment in vivo, suggesting either an involvement of granulocytes or a complement-mediated cytotoxicity towards NB cells. Whatever the effecting mechanism(s) involved, these results strongly support the clinical use of anti-GD2 mAbs after first-line induction regimens.
- Published
- 2003
- Full Text
- View/download PDF
22. Fenretinide as an anti-angiogenic agent in neuroblastoma.
- Author
-
Ribatti D, Raffaghello L, Marimpietri D, Cosimo E, Montaldo PG, Nico B, Vacca A, and Ponzoni M
- Subjects
- Humans, Neuroblastoma drug therapy, Angiogenesis Inhibitors therapeutic use, Antineoplastic Agents therapeutic use, Fenretinide therapeutic use, Neovascularization, Pathologic prevention & control, Neuroblastoma blood supply
- Abstract
Angiogenesis is a critical event in the progression of human neuroblastoma. This mini-review summarizes our literature and experimental data concerning the use of anti-angiogenic molecules, such as TNP-470 and fenretinide, in neuroblastoma treatment.
- Published
- 2003
- Full Text
- View/download PDF
23. Immunoliposomal fenretinide: a novel antitumoral drug for human neuroblastoma.
- Author
-
Raffaghello L, Pagnan G, Pastorino F, Cosimo E, Brignole C, Marimpietri D, Bogenmann E, Ponzoni M, and Montaldo PG
- Subjects
- Animals, Cell Division, Disease Models, Animal, Humans, Liposomes, Mice, Neuroblastoma pathology, Tumor Cells, Cultured, Antineoplastic Agents administration & dosage, Apoptosis drug effects, Fenretinide administration & dosage, Gangliosides immunology, Neuroblastoma drug therapy
- Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor of childhood. In advanced disease stages, prognosis is poor and treatments have limited efficacy, thus novel strategies are warranted. The synthetic retinoid fenretinide (HPR) induces apoptosis in NB and melanoma cell lines. We reported an in vitro potentiation of HPR effects on melanoma cells when the drug is incorporated into GD2-targeted immunoliposomes (anti-GD2-SIL-HPR). Here, we investigated the antitumor activity of anti-GD2-SIL-HPR against NB cells, both in vitro and in vivo. Anti-GD2-immunoliposomes (anti-GD2-SIL) showed specific, competitive binding to, and uptake by, various NB cell lines. Moreover, anti-GD2-SIL-HPR presented increased selectivity and efficacy in inhibiting NB cell proliferation through the induction of apoptosis, compared to free drug and SL-HPR. In an in vivo NB metastatic model, we demonstrated that anti-GD2-SIL-HPR completely inhibited the development of macroscopic and microscopic metastases in comparison to controls. However, similar, but significantly less potent antitumor effect was observed also in mice treated with anti-GD2 immunoliposomes without HPR (anti-GD2-SIL-blank) or anti-GD2 mAb alone (P=0.0297 and P=0.0294, respectively, vs. anti-GD2-SIL-HPR). Moreover, our results clearly demonstrated that, although anti-GD2 mAb had a strong antitumor effect in this in vivo NB model, 100% curability was obtained only following treatment with anti-GD2-SIL-HPR (P<0.0001). Anti-GD2 liposomal HPR should receive clinical evaluation as adjuvant therapy of neuroblastoma.
- Published
- 2003
- Full Text
- View/download PDF
24. Phase I trial and pharmacokinetics of fenretinide in children with neuroblastoma.
- Author
-
Garaventa A, Luksch R, Lo Piccolo MS, Cavadini E, Montaldo PG, Pizzitola MR, Boni L, Ponzoni M, Decensi A, De Bernardi B, Bellani FF, and Formelli F
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Female, Fenretinide administration & dosage, Fenretinide adverse effects, Humans, Male, Neuroblastoma metabolism, Vitamin A blood, Antineoplastic Agents pharmacokinetics, Fenretinide pharmacokinetics, Neuroblastoma drug therapy
- Abstract
Purpose: Fenretinide (4HPR), a synthetic retinoid, induces apoptosis in neuroblastoma cells. A Phase I study in children with neuroblastoma was designed to determine maximum tolerated dose, toxicity, and pharmacokinetics., Experimental Design: Fifty-four patients received oral 4HPR, once daily, for 28 days, followed by a 7-day interruption, for up to 6 courses. The starting dose was 100 mg/m(2)/day. At least 3 patients were entered at each escalating 4HPR dose level. Pharmacokinetic sampling was performed on days 1 and 28 of the first course., Results: Fifty-four patients, of whom 53 were evaluable, received doses between 100 and 4000 mg/m(2)/day for a total of 168 courses. Additional dose escalation was precluded by capsule number intake. A total of 34 of 53 evaluable patients showed manageable, reversible toxicities, which were not dose related. One dose-limiting toxicity (nyctalopia grade 3) occurred after the 1000 mg/m(2)/day dose. Twelve patients showed grade 2 toxicity: skin xerosis (6 cases); nyctalopia (3 cases); hepatic toxicity (1 case); diarrhea (1 case); and headache (1 case). Stable disease was observed in 41 patients for a median period of 23 months (range 2-35+). After first administration, average 4HPR peak plasma levels ranged from 0.6 to 6 micro M (after 100 and 4000 mg/m(2)/day, respectively) and increased 2-fold (to 1.3 and 12.9 micro M, respectively) after the 28-day treatment. 4HPR half-life increased from 17 h after the first administration to 25 h after the 28(th) administration. Incidence of grade 2-3 toxicity was 0 of 12 (0%), 7 of 22 (31%), and 4 of 8 (50%) with peak 4HPR concentrations <3 micro M, 3-10 micro M, and >10 micro M, respectively. After repeated treatment, retinol levels decreased from 20 to 10% of pretreatment levels after all of the doses., Conclusions: In children, 4HPR administration up to 4000 mg/m(2)/day over 28 days, followed by a 7-day interruption, results in manageable toxicity and in drug plasma concentrations comparable with those that induce apoptosis in neuroblastoma cell lines.
- Published
- 2003
25. In vitro and in vivo antitumor activity of liposomal Fenretinide targeted to human neuroblastoma.
- Author
-
Raffaghello L, Pagnan G, Pastorino F, Cosimo E, Brignole C, Marimpietri D, Montaldo PG, Gambini C, Allen TM, Bogenmann E, and Ponzoni M
- Subjects
- Animals, Cell Division, Disease Models, Animal, Fenretinide administration & dosage, Humans, Mice, Mice, Nude, Neoplasm Metastasis, Neoplasm Transplantation, Survival Analysis, Time Factors, Tumor Cells, Cultured, Apoptosis drug effects, Fenretinide pharmacology, Fenretinide therapeutic use, Liposomes administration & dosage, Neuroblastoma drug therapy, Neuroblastoma pathology
- Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor of childhood. In advanced disease stages, prognosis is poor and treatments have limited efficacy, thus novel strategies are warranted. The synthetic retinoid Fenretinide (HPR) induces apoptosis in NB and melanoma cell lines. We reported an in vitro potentiation of HPR effects on melanoma cells when the drug is incorporated into GD2-targeted immunoliposomes (anti-GD2-SIL-HPR). We investigated the antitumor activity of anti-GD2-SIL-HPR against NB cells, both in vitro and in vivo. Anti-GD2-SIL showed specific, competitive binding to and uptake by, various NB cell lines. In in vitro cytotoxicity studies, NB cells, incubated with 30 microM HPR entrapped in anti-GD2-immunoliposomes, showed a significant reduction in cellular growth compared to free HPR, HPR entrapped in Ab-free liposomes or anti-GD2 empty liposomes. In an in vivo NB metastatic model, we demonstrated that anti-GD2-SIL-HPR completely inhibited the development of macroscopic and microscopic metastases in comparison to controls. Similar, but significantly less potent, antitumor effect was observed also in mice treated with anti-GD2 immunoliposomes without HPR (anti-GD2-SIL-blank) or anti-GD2 MAb alone (p = 0.0297 and p = 0.0294, respectively, vs. anti-GD2-SIL-HPR). Moreover, our results clearly demonstrated that although anti-GD2 MAb had a strong antitumor effect in this in vivo NB model, 100% curability was obtained only after treatment with anti-GD2-SIL-HPR (p < 0.0001). Anti-GD2 liposomal HPR should receive clinical evaluation as adjuvant therapy of neuroblastoma., (Copyright 2003 Wiley-Liss, Inc.)
- Published
- 2003
- Full Text
- View/download PDF
26. Inhibition of neuroblastoma-induced angiogenesis by fenretinide.
- Author
-
Ribatti D, Alessandri G, Baronio M, Raffaghello L, Cosimo E, Marimpietri D, Montaldo PG, De Falco G, Caruso A, Vacca A, and Ponzoni M
- Subjects
- Adenocarcinoma blood supply, Adrenal Glands blood supply, Animals, Cell Cycle, Cell Division, Cell Line, Cell Movement, Chick Embryo, Chorion, Endometrial Neoplasms blood supply, Endothelial Growth Factors metabolism, Endothelium cytology, Enzyme-Linked Immunosorbent Assay, Female, Fibroblast Growth Factor 2 metabolism, Flow Cytometry, Humans, Immunohistochemistry, Kinetics, Lymphokines metabolism, Reverse Transcriptase Polymerase Chain Reaction, Time Factors, Tumor Cells, Cultured, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Anticarcinogenic Agents pharmacology, Antineoplastic Agents pharmacology, Fenretinide pharmacology, Neovascularization, Pathologic, Neuroblastoma blood supply, Neuroblastoma drug therapy
- Abstract
Retinoids are a class of natural or synthetic compounds that participate in the control of cell proliferation, differentiation and fetal development. The synthetic retinoid fenretinide (HPR) inhibits carcinogenesis in various animal models. Retinoids have also been suggested to be effective inhibitors of angiogenesis. The effects of HPR on certain endothelial cell functions were investigated in vitro, and its effects on angiogenesis was studied in vivo, by using the chorioallantoic membrane (CAM) assay. HPR inhibited vascular endothelial growth factor- (VEGF-) and fibroblast growth factor-2- (FGF-2)-induced endothelial cell proliferation without affecting endothelial motility; moreover, HPR inhibited growth factor-induced angiogenesis in the CAM assay. Furthermore, a significant antiangiogenic potential of HPR has also been observed in neuroblastoma (NB) biopsy-induced angiogenesis in vivo. We previously demonstrated that supernatants derived from NB cell lines stimulated endothelial cell proliferation. In the present study, we found that this effect was abolished when NB cells were incubated in the presence of HPR. VEGF- and FGF-2-specific ELISA assays, performed on both NB cells derived from conditioned medium and cellular extracts, indicated no consistent effect of HPR on the level of these angiogenic cytokines. Moreover, RT-PCR analysis of VEGF and FGF-2 gene expression confirmed the above lack of effect. HPR was also able to significantly repress the spontaneous growth of endothelial cells, requiring at least 48-72 hr of treatment with HPR, followed by a progressive accumulation of cells in G(1) at subsequent time points. Finally, immunohistochemistry experiments performed in the CAM assay demonstrated that endothelial staining of both VEGF receptor 2 and FGF-2 receptor-2 was reduced after implantation of HPR-loaded sponges, as compared to control CAMs. These data suggest that HPR exerts its antiangiogenic activity through both a direct effect on endothelial cell proliferative activity and an inhibitory effect on the responsivity of the endothelial cells to the proliferative stimuli mediated by angiogenic growth factors., (Copyright 2001 Wiley-Liss, Inc.)
- Published
- 2001
- Full Text
- View/download PDF
27. Angiogenesis extent and expression of matrix metalloproteinase-2 and -9 correlate with progression in human neuroblastoma.
- Author
-
Ribatti D, Surico G, Vacca A, De Leonardis F, Lastilla G, Montaldo PG, Rigillo N, and Ponzoni M
- Subjects
- Child, Child, Preschool, Female, Humans, Infant, Male, Neuroblastoma enzymology, Neuroblastoma pathology, Matrix Metalloproteinase 2 genetics, Matrix Metalloproteinase 9 genetics, Neovascularization, Pathologic etiology, Neuroblastoma blood supply, RNA, Messenger analysis
- Abstract
In human tumors changes in angiogenesis and expression of extracellular matrix-degrading enzymes occur simultaneously during invasion and metastasis. Tissues from 20 biopsies of human neuroblastoma (NB) were investigated immunohistochemically by using an antibody against factor VIII to determine their microvessel number, and by in situ hybridisation to determine the expression of mRNA of the matrix metalloproteinase-2 (MMP-2) and MMP-9. The extent of angiogenesis and the expression of the MMP-2 and MMP-9 mRNA were upregulated in advancing stages. These in situ data suggest that angiogenesis and degradation of extracellular matrix occur simultaneously with NB tumor progression.
- Published
- 2001
- Full Text
- View/download PDF
28. Delivery of c-myb antisense oligodeoxynucleotides to human neuroblastoma cells via disialoganglioside GD(2)-targeted immunoliposomes: antitumor effects.
- Author
-
Pagnan G, Stuart DD, Pastorino F, Raffaghello L, Montaldo PG, Allen TM, Calabretta B, and Ponzoni M
- Subjects
- Blotting, Western, Humans, Liposomes, Oligodeoxyribonucleotides, Antisense genetics, Proto-Oncogene Proteins c-myb genetics, Tumor Cells, Cultured, Antibodies, Monoclonal administration & dosage, Antineoplastic Agents administration & dosage, Drug Delivery Systems, Gangliosides, Gene Expression Regulation, Neoplastic drug effects, Neuroblastoma drug therapy, Oligodeoxyribonucleotides, Antisense administration & dosage, Proto-Oncogene Proteins c-myb antagonists & inhibitors
- Abstract
Background: Advanced-stage neuroblastoma resists conventional treatment; hence, novel therapeutic approaches are required. We evaluated the use of c-myb antisense oligodeoxynucleotides (asODNs) delivered to cells via targeted immunoliposomes to inhibit c-Myb protein expression and neuroblastoma cell proliferation in vitro., Methods: Phosphorothioate asODNs and control sequences were encapsulated in cationic lipid, and the resulting particles were coated with neutral lipids to produce coated cationic liposomes (CCLs). Monoclonal antibodies directed against the disialoganglioside GD(2) were covalently coupled to the CCLs. (3)H-labeled liposomes were used to measure cellular binding, and cellular uptake of asODNs was evaluated by dot-blot analysis. Growth inhibition was quantified by counting trypan blue dye-stained cells. Expression of c-Myb protein was examined by western blot analysis., Results: Our methods produced GD(2)-targeted liposomes that stably entrapped 80%-90% of added c-myb asODNs. These liposomes showed concentration-dependent binding to GD(2)-positive neuroblastoma cells that could be blocked by soluble anti-GD(2) monoclonal antibodies. GD(2)-targeted liposomes increased the uptake of asODNs by neuroblastoma cells by a factor of fourfold to 10-fold over that obtained with free asODNs. Neuroblastoma cell proliferation was inhibited to a greater extent by GD(2)-targeted liposomes containing c-myb asODNs than by nontargeted liposomes or free asODNs. GD(2)-targeted liposomes containing c-myb asODNs specifically reduced expression of c-Myb protein by neuroblastoma cells. Enhanced liposome binding and asODN uptake, as well as the antiproliferative effect, were not evident in GD(2)-negative cells., Conclusions: Encapsulation of asODNs into immunoliposomes appears to enhance their toxicity toward targeted cells while shielding nontargeted cells from antisense effects and may be efficacious for the delivery of drugs with broad therapeutic applications to tumor cells.
- Published
- 2000
- Full Text
- View/download PDF
29. Autocrine regulation of volume-sensitive anion channels in airway epithelial cells by adenosine.
- Author
-
Musante L, Zegarra-Moran O, Montaldo PG, Ponzoni M, and Galietta LJ
- Subjects
- Adenine Nucleotides metabolism, Cell Line, Transformed, Chloride Channels metabolism, Chromatography, High Pressure Liquid, Epithelial Cells drug effects, Epithelial Cells metabolism, Humans, Osmotic Pressure, Trachea cytology, Trachea metabolism, Type C Phospholipases metabolism, Adenosine pharmacology, Chloride Channels drug effects, Trachea drug effects
- Abstract
The activity of volume-sensitive Cl- channels was studied in human tracheal epithelial cells (9HTEo-) by taurine efflux experiments. The efflux elicited by a hypotonic shock was partially inhibited by adenosine receptor antagonists, by alpha,beta-methyleneadenosine 5'-diphosphate (alphabetaMeADP), an inhibitor of the 5'-ectonucleotidase, and by adenosine deaminase. On the other hand, dipyridamole, a nucleoside transporter inhibitor, increased the swelling-induced taurine efflux. Extracellular ATP and adenosine increased taurine efflux by potentiating the effect of hypotonic shock. alphabetaMeADP strongly inhibited the effect of extracellular ATP but not that of adenosine. These results suggest that anion channel activation involves the release of intracellular ATP, which is then degraded to adenosine by specific ectoenzymes. Adenosine then binds to purinergic receptors, causing the activation of the channels. To directly demonstrate ATP efflux, cells were loaded with [3H]AMP, and the release of radiolabeled molecules was analyzed by high performance liquid chromatography. During hypotonic shock, cell supernatants showed the presence of ATP, ADP, and adenosine. alphabetaMeADP inhibited adenosine formation and caused the appearance of AMP. Under hypotonic conditions, elevation of intracellular Ca2+ by ionomycin caused an increase of ATP and adenosine in the extracellular solution. Our results demonstrate that volume-sensitive anion channels are regulated with an autocrine mechanism involving swelling-induced ATP release and then hydrolysis to adenosine.
- Published
- 1999
- Full Text
- View/download PDF
30. GD2-mediated melanoma cell targeting and cytotoxicity of liposome-entrapped fenretinide.
- Author
-
Pagnan G, Montaldo PG, Pastorino F, Raffaghello L, Kirchmeier M, Allen TM, and Ponzoni M
- Subjects
- Antibody Specificity, Cell Division drug effects, Drug Carriers, Humans, Liposomes, Melanoma pathology, Sensitivity and Specificity, Tumor Cells, Cultured, Antineoplastic Agents therapeutic use, Fenretinide therapeutic use, Gangliosides therapeutic use, Melanoma drug therapy
- Abstract
Melanoma is a highly malignant and increasingly common neoplasm. Because metastatic melanoma remains incurable, new treatment approaches are needed. Immunoliposomes have been previously shown to enhance the selective localization of immunoliposome-entrapped drugs to solid tumors with improvements in the therapeutic index of the drugs. Previously, we reported that the synthetic retinoid fenretinide (HPR) is an inducer of apoptosis in neuroblastoma (NB) cells, sharing the neuroectodermal origin with melanoma cells. HPR is a strong inducer of apoptosis also in melanoma cells, although at doses 10-fold higher than those achievable clinically. Thus, our purpose was to investigate the in vitro potentiation of its cytotoxic effect on melanoma cells in combination with long-circulating GD2-targeted immunoliposomes. GD2 is a disialoganglioside extensively expressed on tumors of neuroectodermal origin, including melanoma. Murine anti-GD2 antibody (Ab) 14.G2a and its human/mouse chimeric variant ch14.18 have been ligated to sterically stabilized liposomes by covalent coupling of Ab to the polyethylene glycol (PEG) terminus. Ab-bearing liposomes showed specific, competitive binding to and uptake by various melanoma cell lines compared with liposomes bearing non-specific isotype-matched Abs or Ab-free liposomes. Cytotoxicity was evaluated after 2 hr treatment, followed by extensive washing and 72 hr incubation. This treatment protocol was designed to minimize non-specific adsorption of liposomes to the cells, while allowing for maximum Ab-mediated binding. When melanoma cells were incubated with 30 microM HPR entrapped in anti-GD2 liposomes, a significant reduction in cellular growth was observed compared to free HPR, entrapped HPR in Ab-free liposomes or empty liposomes. Cytotoxicity was not evident in tumor cell lines of other origins that did not express GD2. Growth of NB cells was also inhibited by immunoliposomes with entrapped HPR.
- Published
- 1999
- Full Text
- View/download PDF
31. N-(4-hydroxyphenyl) retinamide is cytotoxic to melanoma cells in vitro through induction of programmed cell death.
- Author
-
Montaldo PG, Pagnan G, Pastorino F, Chiesa V, Raffaghello L, Kirchmeier M, Allen TM, and Ponzoni M
- Subjects
- Antibody Specificity, Cell Division drug effects, Humans, Liposomes, Melanoma pathology, Tumor Cells, Cultured, Antineoplastic Agents therapeutic use, Apoptosis drug effects, Fenretinide therapeutic use, Melanoma drug therapy
- Abstract
Melanoma is a highly malignant and increasingly common tumour. Since metastatic melanoma remains incurable, new treatment approaches are needed. Previously, we reported that the synthetic retinoid N-(4-hydroxyphenyl)retinamide (fenretinide, HPR) induces apoptosis in neuroblastoma cells, sharing a neuroectodermal origin with melanoma cells. Since no data exist thus far on the effects of HPR on human melanoma tumours, our purpose was to investigate the in vitro modulation of cell growth and apoptosis by HPR in melanoma cells. Ten human melanoma cell lines were exposed in vitro to increasing concentrations of HPR. Dose-dependent growth inhibition and cytotoxicity were observed. According to cytofluorimetric analysis, propidium iodide staining and TUNEL assay, HPR-treated melanoma cells were shown to undergo apoptosis. However, IC50 values ranged from 5 to 28 microM, while IC90 values were between 10 and 45 microM. These last concentrations are approximately 10-fold higher than those achievable in patients given oral HPR. To explore the potential of new delivery strategies, HPR was loaded at high concentrations into immunoliposomes directed to disialoganglioside GD2, a tumour-specific antigen extensively expressed by neuroectoderma-derived tumours. Treatment of melanoma cells for a short time (2 hr) with HPR-containing immunoliposomes followed by culture in drug-free medium gave rise to apoptosis of target cells, whereas cells treated for 2 hr with equivalent concentrations of the free drug survived. The efficacy of immunoliposomal HPR was strongly dependent on the density of GD2 expression in the different cell lines.
- Published
- 1999
- Full Text
- View/download PDF
32. Induction of differentiation and apoptosis by interferon-gamma in human neuroblastoma cells in vitro as a dual and alternative early biological response.
- Author
-
Montaldo PG, Chiesa V, Bado M, Raffaghello L, Rozzo C, and Ponzoni M
- Abstract
Interferon-gamma (IFN-gamma) has a well known differentiation-promoting activity on several neuroblastoma (NB) cell lines and has also been reported to induce apoptosis in different cellular models. We have investigated the potential of IFN-gamma to trigger, besides differentiation, programmed cell death in NB cells and the relationship between these processes. Nine NB cell lines, characterized by different phenotypic and maturational features, were cultured in the presence of IFN-gamma (1000 IU/ml) for up to 5 days with either only one treatment at the start of the culture or renewing the culture medium (with or without IFN-gamma) every other day. Neuronal differentiation was assessed by evaluation of morphological changes and expression of mature cytoskeletal proteins, while apoptosis was evaluated at the desired times by fluorescent and electronic microscopy, DNA content analysis and DNA fragmentation assay. Our findings show that apoptosis is an early (mainly non post-differentiative) event and is much more evident following a single IFN-gamma administration. Moreover, IFN-gamma-triggered apoptosis is independent of the cellular phenotype (schwannian or neuronal) and appears to be mutually exclusive with respect to differentiation at the single cell level. Our results strengthen the potential of IFN-gamma as a promising therapeutic agent for NB.
- Published
- 1997
- Full Text
- View/download PDF
33. Increase of metaiodobenzylguanidine uptake and intracellular half-life during differentiation of human neuroblastoma cells.
- Author
-
Montaldo PG, Raffaghello L, Guarnaccia F, Pistoia V, Garaventa A, and Ponzoni M
- Subjects
- 3-Iodobenzylguanidine, Biological Transport, Cell Differentiation, Half-Life, Humans, Interferon-gamma pharmacology, Neuroblastoma pathology, RNA, Messenger analysis, Tumor Cells, Cultured, Antineoplastic Agents pharmacokinetics, Iodobenzenes pharmacokinetics, Neuroblastoma metabolism
- Abstract
Iodine-labelled metaiodobenzylguanidine (MIBG) is a radiopharmaceutical used for diagnostic imaging and targeted radiotherapy of neuroendocrine tumors. We previously reported that the ability of a neuroblastoma (NB) cell line, LAN-5, to accumulate MIBG was powerfully stimulated by interferon-gamma (IFN-gamma), a well-known NB differentiation-promoting agent. To extend the above findings, we have investigated 5 NB cell lines for their ability to accumulate 125I-MIBG in basal conditions or after various combinations of differentiative stimuli. Our results show that association of IFN-gamma and tumor necrosis factor-alpha boosts MIBG uptake in the early times of incubation in LAN-5 and GI-LI-N cells, while both SK-N-SH and SK-N-BE(2)c cells are strongly stimulated by co-treatment with IFN-gamma and all-trans retinoic acid. Moreover, although only LAN-5 and GI-LI-N cells are sensitive to IFN-gamma alone, the combination of IFN-gamma and IFN-alpha causes a synergistic increase in MIBG uptake in all the NB cell lines tested. From experiments on MIBG release we conclude that no intracellular storage within specialized structures took place during differentiation. The observed enhancement in MIBG accumulation results from an increased uptake of the drug only. This conclusion was confirmed by analyzing MIBG-transporter gene expression, which was increased in cells subjected differentiative regimens. According to these findings, inducing differentiation of NB cells in vitro appears to improve their MIBG incorporation ability powerfully.
- Published
- 1996
- Full Text
- View/download PDF
34. Differential effects of N-(4-hydroxyphenyl)retinamide and retinoic acid on neuroblastoma cells: apoptosis versus differentiation.
- Author
-
Ponzoni M, Bocca P, Chiesa V, Decensi A, Pistoia V, Raffaghello L, Rozzo C, and Montaldo PG
- Subjects
- Cell Cycle drug effects, Cell Differentiation drug effects, Cell Division drug effects, Cytoskeletal Proteins analysis, Cytoskeletal Proteins drug effects, DNA Damage, DNA, Neoplasm analysis, DNA, Neoplasm drug effects, DNA, Neoplasm metabolism, Fluorescent Antibody Technique, Humans, Neurons cytology, Neurons drug effects, Apoptosis drug effects, Fenretinide pharmacology, Neuroblastoma drug therapy, Neuroblastoma pathology, Tretinoin pharmacology
- Abstract
Retinoids exert various important biological effects in the control of normal growth, differentiation, and fetal development. While retinoic acid (RA) has entered clinical trials as a differentiation-promoting agent, it is only recently that the synthetic retinoid N-(4-hydroxyphenyl)retinamide (HPR) has been shown to be of potential clinical interest in cancer chemoprevention and treatment. Since thus far no data exist on the effects of HPR on neural crest cell-derived tumors, we have examined its in vitro effects on neuroblastoma (NB) cell lines and found that at relevant pharmacological concentrations it induces a dose-dependent growth inhibition. The antiproliferative effects of HPR were, in six of six cell lines tested, drastically more potent that those induced by an equimolar dose of RA. Time course growth analysis showed that HPR at 3 x 10(-6) M induces a very rapid (24-72 h) fall in thymidine uptake (> 90%), whereas at 3 x 10(-7) M it exhibits cytostatic effects. In contrast to RA, HPR did not show morphological changes typical of NB cell maturation nor did it induce the expression of any cytoskeletal protein associated with neuronal differentiation. DNA flow cytofluorimetric analysis revealed that HPR did not induce an arrest in a specific phase of the cell cycle while triggering apoptosis. This phenomenon was evidenced both by the visualization of "DNA ladders" on gel electrophoresis and by a quantitative assay for evaluating programmed cell death based upon the labeling of DNA breaks with tritiated thymidine. With the latter method, apoptotic cells were detectable as early as 3-6 h after treatment of NB cells with 10(-5) M HPR, while more than 50% of cells were apoptotic by 24-72 h following exposure to 3 x 10(-6) M HPR. In contrast, RA induced a low rate of apoptosis in NB cells only after 3-5 days. Time lapse photomicroscopy showed that NB cells treated with HPR underwent a death process highly reminiscent of apoptosis, with progressive condensation of the cytoplasm around the nucleus and intense cell shrinkage. The cells then rounded up and detached from the plate. Furthermore, propidium iodide staining of the DNA showed that a high proportion of cells treated with HPR displayed a small and brightly staining nucleus; chromatin appeared aggregated into dense masses in the nuclear periphery, a typical feature of apoptotic cells. In conclusion, our study demonstrates that contrary to the differentiation-promoting activity of RA, HPR dramatically suppresses NB cell growth by inducing programmed cell death.(ABSTRACT TRUNCATED AT 400 WORDS)
- Published
- 1995
35. Synergistic differentiation-promoting activity of interferon gamma and tumor necrosis factor-alpha: role of receptor regulation on human neuroblasts.
- Author
-
Montaldo PG, Carbone R, Corrias MV, Ferraris PC, and Ponzoni M
- Subjects
- Base Sequence, Drug Synergism, Humans, Molecular Sequence Data, Polymerase Chain Reaction, Recombinant Proteins pharmacology, Tumor Cells, Cultured, Interferon-gamma pharmacology, Neuroblastoma metabolism, Receptors, Interferon drug effects, Receptors, Tumor Necrosis Factor drug effects, Tumor Necrosis Factor-alpha pharmacology
- Abstract
Background: Interferon gamma (IFN-gamma) and tumor necrosis factor-alpha (TNF) synergize in inducing human neuroblastoma cells to differentiate terminally in vitro into mature nonproliferating neurons. The mechanisms by which this synergistic activity takes place are still obscure., Purpose: To understand the basis of IFN-gamma-TNF synergism, we investigated the constitutive equipment of receptors to IFN-gamma and TNF in two human neuroblastoma cell lines (i.e., LAN-5 and GI-LI-N) and their quantitative and functional variations following treatment with IFN-gamma or TNF., Methods: IFN-gamma receptors and TNF receptors were assessed and functionally characterized by radioreceptor-binding assay before and after treatment of the cells with IFN-gamma or TNF. The TNF receptor subtypes were identified by the reverse transcriptase-polymerase chain reaction, chemical cross-linking of receptors to iodinated TNF, and inhibition of TNF binding by type-specific anti-TNF receptor monoclonal antibodies. The effects of cytokines on cell differentiation were assessed by thymidine incorporation inhibition and morphologic maturation., Results: No quantitative or functional modification of IFN-gamma receptors was observed in TNF-treated cells. However, after treatment with IFN-gamma, TNF receptor numbers were enhanced to a different extent in both cell lines. The two neuroblastoma cell lines expressed, both constitutively and after IFN-gamma induction, only one species of TNF receptor, i.e., the p80 form in LAN-5 and the p60 form in GI-LI-N. Sequential treatment with IFN-gamma followed by TNF, but not in the opposite order, could reproduce the early effects of differentiation in neuroblastoma cells, supporting a role for TNF receptor up-regulation as a basis for the cooperation between the two cytokines., Conclusion: The results strongly suggest that receptor regulation can be at least one mechanism by which IFN-gamma and TNF exert their synergistic effects. Moreover, it appears that the two TNF receptor types are redundant in signaling neuroblastoma cell differentiation., Implications: Our findings can provide a guideline for a rational design of experimental differentiation-based therapeutic protocols in patients with neuroblastoma.
- Published
- 1994
- Full Text
- View/download PDF
36. [Neuroblastoma as a model for the in-vitro study of differentiated therapy].
- Author
-
Ponzoni M, Bocca P, Chiesa V, Guarnaccia F, Rozzo C, and Montaldo PG
- Subjects
- Cell Transformation, Neoplastic drug effects, Cell Transformation, Neoplastic metabolism, Cell Transformation, Neoplastic pathology, Child, Drug Screening Assays, Antitumor, Humans, Neuroblastoma metabolism, Neuroblastoma pathology, Neurons drug effects, Neurons metabolism, Neurons pathology, Time Factors, Tumor Cells, Cultured, Neuroblastoma drug therapy
- Abstract
Differentiation of human neuroblastoma (NB) cells is a very interesting biologic event, providing useful insights in both basic neurobiology and clinical management of this malignancy. Investigation of in vitro NB differentiation exploits several NB cell lines that can be induced to differentiate by an array of natural or synthetic chemicals, as well as biological factors such as some cytokines. The hallmarks of neuronal differentiation are represented by a partial or complete block of cell proliferation, morphological alterations and acquisition of biological features typical of mature neurons (for example, induced synthesis and storage of monoamines and neuropeptides, expression of peculiar cytoskeletal proteins and membrane antigens). The possibility to transfer the differentiative approach to the treatment of NB patients opens exciting therapeutic perspectives.
- Published
- 1994
37. Interferon-gamma-induced differentiation of human neuroblastoma cells increases cellular uptake and halflife of metaiodobenzylguanidine.
- Author
-
Montaldo PG, Carbone R, Cornaglia Ferraris P, and Ponzoni M
- Subjects
- 3-Iodobenzylguanidine, Biological Transport, Active drug effects, Biotechnology, Cell Differentiation drug effects, Cell Line, Half-Life, Humans, Iodine Radioisotopes therapeutic use, Iodobenzenes therapeutic use, Neuroblastoma pathology, Neuroblastoma radiotherapy, RNA, Messenger metabolism, Recombinant Proteins, Interferon-gamma pharmacology, Iodine Radioisotopes pharmacokinetics, Iodobenzenes pharmacokinetics, Neuroblastoma metabolism
- Abstract
Iodine labeled metaiodobenzylguanidine (MIBG) is a radiopharmaceutical employed for both diagnosis and metabolic radiotherapy of neuroblastoma (NB). Resistance to the radiotherapeutic effects of MIBG is common, due to lack of MIBG accumulation by NB cells. MIBG enters competent cells via the noradrenaline transporter; this function requires a relative cellular maturation and is missing in most NB cell lines. In vitro differentiation of NB cells can be achieved with gamma-interferon (gamma-IFN) and other agents. We have verified that gamma-IFN-induced differentiation of NB cells is specifically associated with an increase in their ability to incorporate MIBG. This phenomenon is due to enhancement of MIBG transporter activity, according to pharmacological sensitivity and semiquantitative PCR-based analysis of specific MIBG transporter mRNA. New therapeutic strategies based on both differentiation therapy and targeted radiotherapy of NB can so be devised.
- Published
- 1993
38. Interferon-γ-induced differentiation of human neuroblastoma cells increases cellular uptake and halflife of metaiodobenzylguanidine.
- Author
-
Montaldo PG, Carbone R, Cornaglia Ferraris P, and Ponzoni M
- Abstract
Iodine labeled metaiodobenzylguanidine (MIBG) is a radiopharmaceutical employed for both diagnosis and metabolic radiotherapy of neuroblastoma (NB). Resistance to the radiotherapeutic effects of MIBG is common, due to lack of MIBG accumulation by NB cells. MIBG enters competent cells via the noradrenaline transporter; this function requires a relative cellular maturation and is missing in most NB cell lines. In vitro differentiation of NB cells can be achieved with γ-interferon (γ-IFN) and other agents. We have verified that γ-IFN-induced differentiation of NB cells is specifically associated with an increase in their ability to incorporate MIBG. This phenomenon is due to enhancement of MIBG transporter activity, according to pharmacological sensitivity and semiquantitative PCR-based analysis of specific MIBG transporter mRNA. New therapeutic strategies, based on both differentiation therapy and targeted radiotherapy of NB can so be devised.
- Published
- 1993
- Full Text
- View/download PDF
39. Stimulation of receptor-coupled phospholipase A2 by interferon-gamma.
- Author
-
Ponzoni M, Montaldo PG, and Cornaglia-Ferraris P
- Subjects
- Arachidonic Acid metabolism, Chromatography, Thin Layer, Enzyme Activation, Humans, Indomethacin pharmacology, Lysophosphatidylcholines biosynthesis, Masoprocol pharmacology, Neuroblastoma, Phospholipases A2, Signal Transduction, Tumor Cells, Cultured, Interferon-gamma pharmacology, Phospholipases A metabolism
- Abstract
The biomolecular mechanisms that mediate signal transduction by type II (gamma) interferon (IFN) are poorly understood. IFN-gamma is a potent growth inhibitory cytokine also endowed with antiviral, immunomodulatory, and differentiating activities on various cell targets, including neural cells. IFN-gamma induced a rapid and transient activation of phospholipase A2 in LAN-5, a human neuroblastoma cell line. A consequence of phospholipase A2 activation was the release of arachidonic acid and the generation of lysophospholipids from membrane phospholipids. Treatment of pre-labeled LAN-5 cells with a receptor-saturating concentration of IFN-gamma led to a time-dependent release of [3H]arachidonic acid into the culture media and generation of [32P]lysophosphatidylcholine. Pretreatment of cultures with the phospholipase A2 inhibitor, bromophenacyl bromide, markedly inhibited both [3H]arachidonic acid release and lysophosphatidylcholine production induced by IFN-gamma treatment. Pretreatment of LAN-5 cells with nordihydroguaiaretic acid, a lipoxygenase inhibitor, or with indomethacin, a cyclooxygenase inhibitor, amplified the release of [3H]arachidonic acid and production of lysophosphatidylcholine induced by non-saturating concentrations of IFN-gamma. In parallel, and with the same time-dependent effect, a significant decrease in phosphatidylcholine labeling was observed in IFN-gamma-treated cells, further indicating that a potential signal transduction mechanism of IFN-gamma is the hydrolysis of membrane phosphatidylcholine by phospholipase A2.
- Published
- 1992
- Full Text
- View/download PDF
40. gamma-Interferon increases metaiodobenzylguanidine incorporation and retention in human neuroblastoma cells.
- Author
-
Montaldo PG, Carbone R, Ponzoni M, and Cornaglia-Ferraris P
- Subjects
- 3-Iodobenzylguanidine, Base Sequence, Biological Transport, Cell Differentiation, Gene Expression, Humans, In Vitro Techniques, Iodobenzenes chemistry, Molecular Sequence Data, Neuroblastoma, Oligodeoxyribonucleotides chemistry, Polymerase Chain Reaction, RNA, Messenger genetics, Receptors, Cell Surface metabolism, Tumor Cells, Cultured, Interferon-gamma pharmacology, Iodobenzenes metabolism
- Abstract
Iodine-labeled m-iodobenzylguanidine (MIBG) is a widely used radiopharmaceutical for both diagnosis and biologically targeted radiotherapy of neuroblastoma. However, resistance to the radiotherapeutic effects of MIBG is often encountered, mainly due to lack of MIBG accumulation by neoplastic cells. We have investigated whether the induction of neuroblastoma cell differentiation modifies MIBG incorporation and retention. LAN-5 cells were selected, due to their moderate ability to take up MIBG. Treatment of these cells with gamma-interferon (IFN-gamma) resulted in morphological changes accompanied by a significant increase in overall cell-associated MIBG. Desimipramine, but not reserpine, easily depleted IFN-gamma-treated LAN-5 cells of their MIBG content. This suggests that the mechanism involved is an uptake enhancement rather than an improved storage ability. Indeed, IFN-gamma induces de nov synthesis of MIBG receptor-transporters, as demonstrated by polymerase chain reaction amplification and semiquantitative analysis. Our results suggest that pretreating neuroblastoma patients with IFN-gamma before MIBG administration may enhance the efficacy of both biologically targeted radioimaging and therapy of this tumor.
- Published
- 1992
41. A combined evaluation of biochemical and morphological changes during human neuroblastoma cell differentiation.
- Author
-
Lanciotti M, Montaldo PG, Folghera S, Lucarelli E, Cornaglia-Ferraris P, and Ponzoni M
- Subjects
- Biomarkers, Tumor metabolism, Catecholamines metabolism, Cell Differentiation drug effects, Cytarabine pharmacology, Gene Expression Regulation, Neoplastic drug effects, Humans, Interferon-gamma pharmacology, Nerve Growth Factors pharmacology, Neuroblastoma metabolism, Neuropeptides metabolism, Tetradecanoylphorbol Acetate pharmacology, Tretinoin pharmacology, Tumor Cells, Cultured drug effects, Tumor Cells, Cultured metabolism, Tumor Cells, Cultured pathology, Tumor Necrosis Factor-alpha pharmacology, Neoplasm Proteins metabolism, Nerve Tissue Proteins metabolism, Neuroblastoma pathology, Neurotransmitter Agents metabolism
- Abstract
1. The effects of retinoic acid, gamma-interferon, cytosine arabinoside, nerve growth factor, tumor necrosis factor, and 12-O-tetradecanoylphorbol 13-acetate on the human neuroblastoma cell line, LAN-5, were studied. Intracellular levels of acetylcholinesterase, neuron-specific enolase, catecholamines and related neurotransmitters, vasointestinal peptide, and substance P were evaluated after induction. 2. Cell morphology was strongly affected by retinoic acid, gamma-interferon, cytosine arabinoside, and 12-O-tetradecanoylphorbol 13-acetate. The main effects of retinoic acid and gamma-interferon were the loosening of cell clusters and the extension of long neurites; cytosine arabinoside induced cell body swelling and marked neuritogenesis. Following 12-O-tetradecanoylphorbol 13-acetate treatment, the cells became small, round, and neuritic. Conversely, modifications induced by nerve growth factor and tumor necrosis factor were mild. Cell proliferation rate was reduced by retinoic acid, gamma-interferon, cytosine arabinoside, and 12-O-tetradecanoylphorbol 13-acetate, while nerve growth factor and tumor necrosis factor were devoid of effects. 3. Acetylcholinesterase activity was significantly stimulated by retinoic acid and by gamma-interferon. Neuron-specific enolase activity was unaffected by all treatments except 12-O-tetradecanoylphorbol 13-acetate, which enhanced it by 1.6-fold. 4. The cellular catecholamine and related metabolite content was lowered by retinoic acid and gamma-interferon, while cytosine arabinoside and, even more, 12-O-tetradecanoylphorbol 13-acetate showed a stimulatory activity on their intracellular accumulation. 5. Finally, the cell-associated vasointestinal peptide level was strikingly increased by gamma-interferon and, to a lesser extent, by retinoic acid, cytosine arabinoside, and 12-O-tetradecanoylphorbol 13-acetate. 6. It is concluded that the most relevant biochemical changes associated with LAN-5 cells differentiation involve the repertoire of neurotransmitters and neuropeptides. These events vary in quality and in quantity, likely due to the pattern complexity of gene expression triggered by each inducer in determining the diversity of neuronal phenotypes.
- Published
- 1992
- Full Text
- View/download PDF
42. The combination of gamma-interferon and tumor necrosis factor causes a rapid and extensive differentiation of human neuroblastoma cells.
- Author
-
Ponzoni M, Casalaro A, Lanciotti M, Montaldo PG, and Cornaglia-Ferraris P
- Subjects
- Acetylcholinesterase metabolism, Antigens, Surface analysis, Cytoskeletal Proteins analysis, Cytoskeletal Proteins metabolism, DNA Replication drug effects, Dihydroxyphenylalanine metabolism, Dopamine metabolism, Drug Synergism, Fluorescent Antibody Technique, Humans, Hydroxyindoleacetic Acid metabolism, Kinetics, Neuroblastoma metabolism, Norepinephrine metabolism, Phosphopyruvate Hydratase analysis, Recombinant Proteins pharmacology, Serotonin metabolism, Thymidine metabolism, Tumor Cells, Cultured, Cell Differentiation drug effects, Interferon-gamma pharmacology, Neuroblastoma pathology, Tumor Necrosis Factor-alpha pharmacology
- Abstract
Neuroblastoma (NB), a tumor originating from the sympathetic nervous system, is the most common extracranial neurological tumor of childhood. Human NB cells may differentiate in vitro under treatment with biological agents, as gamma-interferon (IFN-gamma) and tumor necrosis factor (TNF). Unfortunately, NB cell lines resistant to the differentiation-inducing effects of both drugs have been observed. Here we demonstrate that a combination of IFN-gamma plus TNF causes extensive and generalized differentiation of NB cells toward a neuronal phenotype. Both IFN-gamma and TNF, given alone, moderately reduced cell growth and induced partial morphological maturation. Their combination further reduced cell proliferation. The combined treatment gave a synergistic rather than additive cytostatic effect, documented also by a dramatically enhanced differentiation toward a neuronal morphology. Membrane immunofluorescence showed a homologous and heterologous up-regulation of IFN-gamma receptor, as well as a marked induction of HLA Class I antigens and, to a lesser extent, of Class II antigens on NB cells induced to differentiate. Treatment of NB cell lines with IFN-gamma/TNF results in the induction of a differentiated phenotype, as indicated by the increased expression of the Mr 160,000 and 200,000 neurofilament proteins and that of microtubule-associated proteins. Evaluation of biochemical markers of neuronal differentiation confirmed the ability of the combined treatment to induce neuroblast maturation. These results suggest that the combination of IFN-gamma and TNF should be considered for experimental clinical trials in neuroblastoma.
- Published
- 1992
43. In vitro pharmacology of metaiodobenzylguanidine uptake, storage and release in human neuroblastoma cells.
- Author
-
Montaldo PG, Carbone R, Lanciotti M, Ponzoni M, and Cornaglia-Ferraris P
- Subjects
- 3-Iodobenzylguanidine, Humans, In Vitro Techniques, Tumor Cells, Cultured, Antineoplastic Agents pharmacokinetics, Iodobenzenes pharmacokinetics, Neuroblastoma metabolism
- Abstract
Four human neuroblastoma (NB) cell lines (LAN-5, SK-N-BE(2)C, GI-LI-N, and GI-CA-N) have been investigated for their ability to take up and store [125I]metaiodobenzylguanidine (125I-MIBG) in vitro. Only SK-N-BE(2)C and LAN-5 cells were able to specifically take up MIBG, with the former cell line showing a more efficient retention of the radiotracer. 125I-MIBG incorporation in both cell lines was inhibited by norepinephrine, desipramine, ouabain and energy depletion. Thus, all the major criteria for specific (type 1) uptake were fulfilled. Conversely, GI-LI-N and GI-CA-N cells did not show any specific uptake. Pharmacological manipulations aimed at defining the intracellular site(s) of 125I-MIBG storage clearly showed that the radiotracer is not accumulated in the reserpine-sensitive neurosecretory granules and vesicles in NB cells, contrary to what has been observed in a chromaffin derived tumor cell line (PC12). Our study provides new and suitable models to investigate in vitro the molecular and cellular pharmacology of MIBG in NB cells.
- Published
- 1991
44. Accumulation of m-iodobenzylguanidine by neuroblastoma cells results from independent uptake and storage mechanisms.
- Author
-
Montaldo PG, Lanciotti M, Casalaro A, Cornaglia-Ferraris P, and Ponzoni M
- Subjects
- 3-Iodobenzylguanidine, Biological Transport, Cell Line, Energy Metabolism, Humans, Kinetics, Neuroblastoma metabolism, Ouabain pharmacology, Sodium metabolism, Time Factors, Antineoplastic Agents metabolism, Iodobenzenes metabolism
- Abstract
The modalities of uptake and storage of iodine-labeled m-iodobenzylguanidine (MIBG) by four human neuroblastoma cell lines have been studied. SK-N-BE(2)C cell line has been shown to possess the specific (type 1) MIBG uptake, as well as an efficient extravesicular storage mechanism. Conversely, LAN-5 cells, which show a nonsaturation kinetic of MIBG incorporation, lack only the ability to efficiently store the MIBG taken up by a mechanism that can be pharmacologically defined as uptake 1. The two other neuroblastoma cell lines tested, GI-LI-N and GI-CA-N, lack both uptake and storage capacity. In view of the fact that the only detailed study on specific MIBG uptake by a human neuroblastoma cell line has been carried out on SK-N-SH, a highly heterogeneous cell line, our report provides new insights on the molecular and cellular pharmacology of radiolabeled MIBG.
- Published
- 1991
45. Gamma-interferon, retinoic acid, and cytosine arabinoside induce neuroblastoma differentiation by different mechanisms.
- Author
-
Ponzoni M, Lanciotti M, Montaldo PG, and Cornaglia-Ferraris P
- Subjects
- Antigens, Neoplasm metabolism, Biomarkers, Tumor metabolism, Cell Differentiation drug effects, Cytoskeletal Proteins metabolism, DNA Polymerase I antagonists & inhibitors, Dose-Response Relationship, Drug, Humans, Inositol Phosphates metabolism, Neoplasm Proteins metabolism, Stimulation, Chemical, Tumor Cells, Cultured drug effects, Tumor Cells, Cultured pathology, Cytarabine pharmacology, Interferon-gamma pharmacology, Neuroblastoma pathology, Tretinoin pharmacology
- Abstract
1. The effects of gamma-interferon (gamma-IFN), retinoic acid (RA), and cytosine arabinoside (ARA-C) on the growth, morphology, and phenotype of the human neuroblastoma (NB) cell lines, LAN-1 and GI-ME-N, have been extensively tested. 2. RA, gamma-IFN, and ARA-C induced a dose-dependent morphological differentiation and growth inhibition, without affecting cell viability. Cells exposed to 10(-6) M RA or 1000 U/ml gamma-IFN significantly decreased their growth rate within the first 24 and 48 hr of culture, respectively. Cells became smaller and polygonal and sprouted long cellular processes with varicosities along their courses. In contrast, ARA-C-differentiated cells were larger and flattened, with few elongated dendritic processes. 3. Analysis of membrane and cytoskeletal markers by immunofluorescence and Western blot showed several changes in NB-specific antigen expression after 5 days of treatment with all inducing agents. Analysis of labeled phosphatidylinositol metabolites from prelabeled cells showed, within 1 min of treatment with RA, a rapid decrease in inositol 1,4,5-trisphosphate and of 1,2-diacylglycerol levels. No changes in inositol phospholipid metabolism were observed in gamma-IFN- or ARA-C-treated cells. 4. We conclude that RA-induced decrease in phosphatidylinositol (PI) hydrolysis is not likely to be a consequence of the acquisition of a different phenotype, as its changes precede the acquisition of neuronal markers. In addition, gamma-IFN and ARA-C, both inducing a mature phenotype, did not affect PI hydrolysis. 5. Decreased PI hydrolysis seems to be sufficient, although not necessary, to commit NB cells to neuronal differentiation. Analysis of molecular mechanisms associated with NB cell differentiation may be helpful to clarify the potential of various biological agents in affecting the development of the neural cell.
- Published
- 1991
- Full Text
- View/download PDF
46. Morphological and biochemical modifications during neuroblastoma cell differentiation.
- Author
-
Lanciotti M, Montaldo PG, Cornaglia-Ferraris P, and Ponzoni M
- Abstract
In order to define some distinctive criteria of differentiation, we studied the effects of retinoic acid (RA), γ-interferon (γ-IFN), cytosine arabinoside (ARA-C), nerve growth factor (NGF), tumor necrosis factor (TNF), and 12-0-tetradecanoylphorbol-13 acetate (TPA) on the human neuroblastoma cell line, LAN-5. Intracellular levels of acetylcholinesterase, neuron specific enolase, catecholamines, and related neurotransmitters were evaluated after 8 days of treatment with various inducers. Our data indicate that the most striking biochemical changes accompanying LAN-5 cell differentiation are a shift from a catecholaminergic to a cholinergic phenotype, and/or a drastic increase in the catecholamines levels. Moreover, small changes may be independent of thein vitro maturation. Thus, biochemical phenotyping, more than morphological evaluation, is a fruitful approach to monitor neuronal cell differentiation.
- Published
- 1991
- Full Text
- View/download PDF
47. High-performance liquid chromatographic determination in human plasma of a new immunomodulatory agent with a peptidyl-hypoxanthine structure (RM 06).
- Author
-
Montaldo PG and Cornaglia-Ferraris P
- Subjects
- Adult, Humans, Molecular Structure, Regression Analysis, Adjuvants, Immunologic blood, Chromatography, High Pressure Liquid, Dipeptides blood, Hypoxanthines blood
- Published
- 1990
- Full Text
- View/download PDF
48. Pharmacokinetics of intrapleural versus intravenous etoposide (VP 16) and teniposide (VM 26) in patients with malignant pleural effusion.
- Author
-
Montaldo PG, Figoli F, Zanette ML, Sorio R, Zucchetti M, Tirelli U, and D'Incalci M
- Subjects
- Aged, Etoposide administration & dosage, Etoposide adverse effects, Female, Humans, Infusions, Intravenous, Lung Neoplasms metabolism, Male, Middle Aged, Pleural Effusion metabolism, Teniposide administration & dosage, Teniposide adverse effects, Etoposide pharmacokinetics, Lung Neoplasms drug therapy, Pleural Effusion drug therapy, Podophyllotoxin analogs & derivatives, Teniposide pharmacokinetics
- Abstract
The pharmacokinetics of etoposide (VP 16) and teniposide (VM 26) were studied after intrapleural administration to 3 patients with lung cancer and malignant pleural effusion. Comparison with the kinetic behavior of intravenously infused VP 16 and VM 26 in the same patients suggests that intrapleural drug delivery achieves higher and longer-lasting pleural concentrations, thus providing a theoretical basis for the palliative treatment of malignant pleural effusions. Although no systemic toxicity was observed after intrapleural administration of either drug, 1 of the 3 patients developed a transient asymptomatic hemorrhagic pleurisy during the first 2 days after the drug, alerting to the possible local toxicity of such treatment.
- Published
- 1990
- Full Text
- View/download PDF
49. High-performance liquid chromatographic determination of 5-bromodeoxyuridine in human plasma.
- Author
-
Montaldo PG and D'Incalci M
- Subjects
- Chromatography, High Pressure Liquid, Humans, Spectrophotometry, Ultraviolet, Bromodeoxyuridine blood
- Abstract
A new method is described for the determination of submicromolar concentrations of 5-bromodeoxyuridine in human plasma. Sample pretreatment involves cold methanol deproteinization, freezing-thawing and lyophilization. The sample is then analysed by reversed-phase high-performance liquid chromatography. This method is very reproducible and has a detection limit of 0.1 microgram/ml (0.32.10(-6) M). Comparison with other procedures indicates that the method is advantageous as regards sensitivity and specificity and can be readily applied in clinical pharmacological investigations.
- Published
- 1989
- Full Text
- View/download PDF
50. Purification to homogeneity and biochemical characterization of two suppressor factors from human malignant T-cells.
- Author
-
Ponzoni M, Montaldo PG, Lanciotti M, Castagnola E, Cirillo C, and Cornaglia-Ferraris P
- Subjects
- Cell Line, Chromatography, Gel, Chromatography, Ion Exchange, Humans, Leukemia, Lymphoid immunology, Molecular Weight, Suppressor Factors, Immunologic isolation & purification, T-Lymphocytes immunology
- Abstract
A cell clone (GI-CO-T-9) derived from a long term T-cell culture (PF-382), established from a patient affected by acute T-lymphoblastic leukemia (T-ALL), was selected for the presence in the culture medium of factors suppressing T-cell proliferation. The crude supernatant has been subjected to a multi-step chromatographic fractioning, including: preparative gel permeation, anion exchange, and hydrophobic interaction High Performance Liquid Chromatography (HPLC). The highly purified material was characterized by polyacrylamide gel electrophoresis in sodium dodecyl sulfate (SDS-PAGE), revealing single bands of 115 Kd and 80 Kd. The isoelectric points (pI), determined by flat-bed isoelectric-focusing, were 7.4 for High Molecular Weight Suppressor Factor (HMWSF) and 3.5-3.6 for Low Molecular Weight Suppressor Factor (LMWSF).
- Published
- 1988
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.