Network


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

Hotspot


Dive into the research topics where Elena Marangon is active.

Publication


Featured researches published by Elena Marangon.


European Journal of Cancer | 2013

A first in human phase I study of the proteasome inhibitor CEP-18770 in patients with advanced solid tumours and multiple myeloma

E. Gallerani; Massimo Zucchetti; Dario Brunelli; Elena Marangon; Cristina Noberasco; Dagmar Hess; Angelo Delmonte; Giovanni Martinelli; Steffen Böhm; Christopher Driessen; Filippo de Braud; Silvia Marsoni; Roberta Cereda; Federica Sala; Maurizio D’Incalci; Cristiana Sessa

BACKGROUND The safety, pharmacokinetics (PK) and pharmacodynamics of CEP-18770, a new peptide boronic acid proteasome inhibitor, have been investigated after intravenous administration on days 1, 4, 8 and 11 of every 21d cycle in patients with solid tumours and multiple myeloma (MM). PATIENTS AND METHODS Thirty-eight patients were treated with CEP-18770 at escalating doses from 0.1 to 1.8mg/m(2) where 2 out of 5 patients showed dose limiting toxicities. The maximum tolerated/recommended dose (MTD/RD) of 1.5mg/m(2) was tested in 12 additional patients. Skin rash was dose-limiting and occurred in 53% of patients; other frequent toxicities were asthenia (29%), stomatitis (21%) and pyrexia (16%). No significant peripheral neuropathy was observed. PK in plasma was linear with a half-life of the elimination phase of 62.0±43.5h. Proteasome inhibition in peripheral blood mononuclear cells was dose related in MM patients; it was of 45.4±11.5% at the RD. CONCLUSIONS CEP-18770 showed a favourable safety profile with lack of neurotoxicity and linear plasma PK. The definition of the optimal biological dose and schedule of treatment is actively pursued because of the high incidence of skin toxicity of the twice a week schedule.


PLOS ONE | 2015

Development and validation of a high-performance liquid chromatography-tandem mass spectrometry method for the simultaneous determination of irinotecan and its main metabolites in human plasma and its application in a clinical pharmacokinetic study.

Elena Marangon; Bianca Posocco; Elisa Mazzega; Giuseppe Toffoli

Irinotecan is currently used in several cancer regimens mainly in colorectal cancer (CRC). This drug has a narrow therapeutic range and treatment can lead to side effects, mainly neutropenia and diarrhea, frequently requiring discontinuing or lowering the drug dose. A wide inter-individual variability in irinotecan pharmacokinetic parameters and pharmacodynamics has been reported and associated to patients’ genetic background. In particular, a polymorphism in the UGT1A1 gene (UGT1A1*28) has been linked to an impaired detoxification of SN-38 (irinotecan active metabolite) to SN-38 glucuronide (SN-38G) leading to increased toxicities. Therefore, therapeutic drug monitoring of irinotecan, SN-38 and SN-38G is recommended to personalize therapy. In order to quantify simultaneously irinotecan and its main metabolites in patients’ plasma, we developed and validated a new, sensitive and specific HPLC–MS/MS method applicable to all irinotecan dosages used in clinic. This method required a small plasma volume, addition of camptothecin as internal standard and simple protein precipitation. Chromatographic separation was done on a Gemini C18 column (3 μM, 100 mm x 2.0 mm) using 0.1% acetic acid/bidistilled water and 0.1% acetic acid/acetonitrile as mobile phases. The mass spectrometer worked with electrospray ionization in positive ion mode and selected reaction monitoring. The standard curves were linear (R2 ≥0.9962) over the concentration ranges (10–10000 ng/mL for irinotecan, 1–500 ng/mL for SN-38 and SN-38G and 1–5000 ng/mL for APC) and had good back-calculated accuracy and precision. The intra- and inter-day precision and accuracy, determined on three quality control levels for all the analytes, were always <12.3% and between 89.4% and 113.0%, respectively. Moreover, we evaluated this bioanalytical method by re-analysis of incurred samples as an additional measure of assay reproducibility. This method was successfully applied to a pharmacokinetic study in metastatic CRC patients enrolled in a genotype-guided phase Ib study of irinotecan administered in combination with 5-fluorouracil/leucovorin and bevacizumab.


Drug Metabolism and Disposition | 2006

Pharmacokinetics and metabolism in mice of IDN 5390 (13-(N-Boc-3-i- butylisoserinoyl)-C-7,8-seco-10-deacetylbaccatin III), a new oral C-seco-taxane derivative with antiangiogenic property effective on paclitaxel-resistant tumors

Roberta Frapolli; Elena Marangon; Marco Zaffaroni; Tina Colombo; Cristiano Falcioni; Renzo Bagnati; Matteo Simone; Maurizio D'Incalci; Carla Manzotti; Gabriele Fontana; Paolo Morazzoni; Massimo Zucchetti

IDN 5390 (13-(N-Boc-3-i-butylisoserinoyl)-C-7,8-seco-10-deacetylbaccatin III) is a new taxane, derived from 7,8-C-seco-10-deacetylbaccatin, selected for its ability to inhibit angiogenesis, mainly by acting on endothelial cell motility, and for its selective activity on class III β-tubulin. In vivo, IDN 5390 shows activity against paclitaxel-sensitive and -resistant tumors when administered on a prolonged, continuous dosage schedule. We studied the pharmacokinetics and bioavailabilty of the drug in mice after single and repeated oral treatment. IDN 5390 was rapidly absorbed after oral administration, with good bioavailability (43%). After intravenous injection, it was extensively distributed in tissue, mainly the liver, kidney, and heart, with low but persistent levels in brain. The kinetics appear dose-dependent with a clearance of 2.6, 1.4, and 0.9 l/kg at, respectively, 60, 90, and 120 mg/kg, and a half-life 24, 36, and 54 min. After prolonged daily oral doses given for 2 weeks, we found that there was a decrease in drug availability; i.e., the area under the concentration-time curve value after p.o. daily administration on day 14 was 2-fold lower than that on day 1. Metabolism plays a major role in elimination of the drug, and at least 12 metabolites were identified in feces and urine. The percentage excreted as metabolites after an oral dose (42%) was higher than that after the i.v. dose (33%), suggesting a first-pass effect. Four metabolites were found in plasma at detectable levels; one of them, with restored taxane scaffold, is a species 3 times more potent than IDN 5390, possibly contributing to the observed anti-tumor activity.


Journal of Pharmaceutical and Biomedical Analysis | 2014

Quantification of trabectedin in human plasma: Validation of a high-performance liquid chromatography–mass spectrometry method and its application in a clinical pharmacokinetic study

Monique Zangarini; Laura Ceriani; Federica Sala; Elena Marangon; Renzo Bagnati; Maurizio D’Incalci; Federica Grosso; Massimo Zucchetti

A rapid, sensitive and specific HPLC-MS/MS method was developed and validated for the quantification of trabectedin in human plasma after using deuterated trabectedin as Internal Standard (IS). After the addition of ammonium sulphate, the analyte was extracted from human plasma with acidified methanol (0.1 M HCl). Chromatographic separation was done on an Accucore XL C₁₈ column (4 μm; 50 mm × 2.1 mm) using a Mobile Phase (MP) consisting of CH₃COONH₄ 10 mM, pH 6.8 (MP A) and CH₃OH (MP B). The mass spectrometer worked with electrospray ionization in positive ion mode and Selected Reaction Monitoring (SRM), using target ions at [M-H₂O+H]⁺ m/z 744.4 for trabectedin and [M-H₂O+H]⁺m/z 747.5 for the IS. The standard curve was linear (R² ≥ 0.9955) over the concentration range 0.025-1.0 ng/ml and had good back-calculated accuracy and precision. The intra- and inter-day precision and accuracy determined on three quality control samples (0.04, 0.08 and 0.80 ng/ml) were <9.9% and between 98.3% and 105.3%, respectively. The extraction recovery was >81% and the lower limit of quantification 0.025 ng/ml. The method was successfully applied to study trabectedin pharmacokinetics in a patient with a liposarcoma who received 1.3 mg/m² as a 24 h continuous i.v. infusion.


Journal of Mass Spectrometry | 2010

Development and validation of a high-performance liquid chromatography-tandem mass spectrometry method for the determination of the novel proteasome inhibitor CEP-18770 in human plasma and its application in a clinical pharmacokinetic study

Federica Sala; Elena Marangon; Renzo Bagnati; Valeria Livi; Roberta Cereda; Maurizio D'Incalci; Massimo Zucchetti

CEP-18770, [(1R)-1-{[(2S,3R)-3-hydroxy-2-{[(6-phenyl-2-pyridinyl)carbonyl]amino}butanoyl]amino}-3-methylbutyl]boronic acid, is a novel proteasome inhibitor, now under early clinical evaluation as an anticancer agent. To investigate its clinical pharmacokinetics, a high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) method was developed and validated to measure the drug in human plasma, based on simple protein precipitation with acetonitrile after the addition of irbesartan as internal standard. The method requires a small volume of sample (100 µl) and is rapid and selective, allowing good resolution of peaks in 5 min. It is sensitive, precise and accurate, with overall precision, expressed as coefficient of variation (CV%), always < 10.0%, accuracy in the range 93.8-107.7% and high recovery, close to 100%. The limit of detection is 0.01 ng/ml and the lower limit of quantitation (LLOQ) is 0.20 ng/ml. The assay was validated in the range from the LLOQ up to 50.00 ng/ml. This is the first method developed and validated for analyzing a proteasome inhibitor with a boronic-acid-based structure in human plasma. The method was successfully applied to study the pharmacokinetics of CEP-18770 in cancer patients with solid tumors or multiple myeloma who had received the drug as a short intravenous bolus during the initial Phase I trial.


Mass Spectrometry Reviews | 2017

Mass spectrometry in the pharmacokinetic studies of anticancer natural products

Sara Crotti; Bianca Posocco; Elena Marangon; Donato Nitti; Giuseppe Toffoli; Marco Agostini

In the history of medicine, nature has represented the main source of medical products. Indeed, the therapeutic use of plants certainly goes back to the Sumerian and Hippocrates and nowadays nature still represents the major source for new drugs discovery. Moreover, in the cancer treatment, drugs are either natural compounds or have been developed from naturally occurring parent compounds firstly isolated from plants and microbes from terrestrial and marine environment. A critical element of an anticancer drug is represented by its severe toxicities and, after administration, the drug concentrations have to remain in an appropriate range to be effective. Anyway, the drug dosage defined during the clinical studies could be inappropriate for an individual patient due to differences in drug absorption, metabolism and excretion. For this reason, personalized medicine, based on therapeutic drug monitoring (TDM), represents one of most important challenges in cancer therapy. Mass spectrometry sensitivity, specificity and fastness lead to elect this technique as the Golden Standard for pharmacokinetics and drug metabolism studies therefore for TDM. This review focuses on the mass spectrometry-based methods developed for pharmacokinetic quantification in human plasma of anticancer drugs derived from natural sources and already used in clinical practice. Particular emphasis was placed both on the pre-analytical and analytical steps, such as: sample preparation procedures, sample size required by the analysis and the limit of quantification of drugs and metabolites to give some insights on the clinical practice applicability.


Clinical Cancer Research | 2017

Genotype-guided dosing study of FOLFIRI plus bevacizumab in patients with metastatic colorectal cancer

Giuseppe Toffoli; Manish R. Sharma; Elena Marangon; Bianca Posocco; Elizabeth Gray; Quan Mai; Angela Buonadonna; Blase N. Polite; Gianmaria Miolo; Gianna Tabaro; Federico Innocenti

Purpose: UGT1A1*28 confers a higher risk of toxicity in patients treated with irinotecan. Patients with *1/*1 and *1/*28 genotypes might tolerate higher than standard doses of irinotecan. We aimed to identify the MTD of irinotecan in patients with metastatic colorectal cancer (mCRC) with *1/*1 and *1/*28 genotypes treated with FOLFIRI plus bevacizumab, and to determine whether bevacizumab alters irinotecan pharmacokinetics. Experimental Design: Previously untreated patients with mCRC (25 *1/*1; 23 *1/*28) were given FOLFIRI plus bevacizumab every 2 weeks. The irinotecan dose was escalated using a 3 + 3 design in each genotype group as follows: 260, 310, and 370 mg/m2. The MTD was the highest dose at which <4/10 patients had a dose-limiting toxicity (DLT). Pharmacokinetics of irinotecan and SN-38 were measured on days 1 to 3 (without bevacizumab) and 15 to 17 (with bevacizumab). Results: For *1/*1 patients, 2 DLTs were observed among 10 patients at 310 mg/m2, while 370 mg/m2 was not tolerated (2 DLTs in 4 patients). For *1/*28 patients, 2 DLTs were observed among 10 patients at 260 mg/m2, while 310 mg/m2 was not tolerated (4 DLTs in 10 patients). Neutropenia and diarrhea were the most common DLTs. Changes in the AUCs of irinotecan and SN-38 associated with bevacizumab treatment were marginal. Conclusions: The MTD of irinotecan in FOLFIRI plus bevacizumab is 310 mg/m2 for UGT1A1 *1/*1 patients and 260 mg/m2 for *1/*28 patients. Bevacizumab does not alter the pharmacokinetics of irinotecan. The antitumor efficacy of these genotype-guided doses should be tested in future studies of patients with mCRC treated with FOLFIRI plus bevacizumab. Clin Cancer Res; 23(4); 918–24. ©2016 AACR.


European Journal of Mass Spectrometry | 2014

Matrix-assisted laser desorption/ionization, nanostructure-assisted laser desorption/ionization and carbon nanohorns in the detection of antineoplastic drugs. 1. The cases of irinotecan, sunitinib and 6-alpha-hydroxy paclitaxel

Eleonora Calandra; Sara Crotti; Marco Agostini; Donato Nitti; Marco Roverso; Giuseppe Toffoli; Elena Marangon; Bianca Posocco; Pietro Traldi

The development of surface-assisted laser desorption/ionization (SALDI) methodologies in mass spectrometry allows, in principle, the development of new analytical approaches to qualitative and quantitative measurements on small molecules. Some of these methods have been applied to characterize two antineoplastic drugs: irinotecan (1) and sunitinib (2), and also 6-α-hydroxy-paclitaxel (3), the main metabolite of paclitaxel. Three different SALDI approaches have been tested employing nanostructure-assisted laser desorption/ionization (NALDI), carbon nanohorns (NHs) and carbon NHs covered by liquid additives. The results so obtained have been compared to those observed under matrix-assisted laser desorption/ionization (MALDI) conditions. Compounds 1 and 2 show the easy formation of protonated molecular species under all the experimental conditions, but the highest absolute intensity was achieved by NALDI. On the contrary, ionic species of low intensity are present for 3, among which are those that exhibit the highest intensity caused by [M + K]+ ions. After a critical evaluation of the obtained data, the linear response of the [M + H]+ ion intensity of 1 versus different deposited sample amounts was investigated, and the best results (R2 = 0.9889) were obtained under MALDI conditions. The analysis of plasma samples spiked with 1 showed, again, that the MALDI approach was the best one (R2 = 0.9766). The failure of NALDI measurements could be rationalized by the presence of ion-suppression effects.


Journal of Chromatography B | 2009

Development and validation of a LC-MS/MS method for the determination of the novel oral 1,14 substituted taxane derivatives, IDN 5738 and IDN 5839, in mouse plasma and its application to the pharmacokinetic study.

Elena Marangon; Cristiano Falcioni; Carla Manzotti; Gabriele Fontana; Maurizio D'Incalci; Massimo Zucchetti

Two LC-ESI-MS and CID-MS/MS methods were developed and validated for pharmacokinetic studies of the novel oral taxane derivatives IDN 5738 and IDN 5839, used for preclinical evaluation in mice. The analysis requires 100muL of plasma sample, involves the addition of an internal standard and protein precipitation with 0.1% HCOOH in acetonitrile. The HPLC separation was obtained on Sunfire C18 column and Selected Reaction Monitoring technique was used to quantify the taxanes. The recoveries were more than 90%; the methods were linear over the validated concentrations range of 25-1500ng/mL for IDN 5738 and 25-5000ng/mL for IDN 5839 and had a limit of detection of 0.14 and 0.25ng/mL, respectively. The inter-day coefficient of variation (CV%) of the calibration standards ranged between 1.3 and 7.2% for IDN 5738 and between 0.0 and 9.0% for IDN 5839 and the mean accuracy was in the range 85.3-112.0% for IDN 5738 and between 80.0 and 111.0% for IDN 5839. Moreover, analysing quality control plasma samples on three different days, the methods resulted precise and accurate showing intra- and inter-day CV within 12% for both analytes, and accuracy of 92.0-113.3% and 85.9-105.7% for IDN 5738 and IDN 5839, respectively. With these methods, we studied for the first time, the pharmacokinetics of the two taxanes showing for both, good oral bioavailability (>50%).


Cancer Chemotherapy and Pharmacology | 2009

Pharmacokinetic profile of imatinib mesylate and N-desmethyl-imatinib (CGP 74588) in children with newly diagnosed Ph+ acute leukemias.

Elena Marangon; Marco Citterio; Federica Sala; Elena Barisone; Alma Lippi; Carmelo Rizzari; Andrea Biondi; Maurizio D'Incalci; Massimo Zucchetti

Dear Editor, Because of the rarity of childhood Philadelphia chromosome-positive (Ph+) leukemias, data on imatinib pharmacokinetics in children are scant. Imatinib pharmacokinetics were reported in a limited number of leukemic children receiving drug in doses of 260–570 mg/m per day [1]. Despite a wide inter-patient variability, the plasma drug levels were similar to those reported in adult patients treated with standard doses of 400–600 mg/day [2–5]. No data were provided on the pharmacokinetics of the main circulating metabolite of imatinib, N-desmethyl-imatinib (CGP 74588), that had already been determined in adults [2, 6]. Ph+ childhood leukemia is less sensitive to imatinib than adult CML and this is likely to be due to major biological diVerences between these leukemias. However, the diVerent sensitivity may at least partly be related to diVerent bioavailability, metabolism and pharmacokinetic features of imatinib in children and adults that might require adjustment of doses and schedules. In our study, we determined the pharmacokinetics of imatinib and CGP 74588 in three children (two females and one male, aged 11, 15 and 6 years, respectively), with newly diagnosed Ph+ ALL treated according to the induction phase of the EsPhALL (European intergroup study on post-induction treatment of Ph+ ALL) protocol and in one female child (aged 6 years) with CML in lymphoblastic phase. Imatinib was administered at the dosage of 300 mg/m per day in all patients. They were treated in diVerent Associazione Italiana Ematologia Oncologia Pediatrica (AIEOP) centers: Monza, Turin and Florence. The Institutionals Ethical Board approved the study, conducted in accordance with the Helsinky declaration of Principle. Patient four merits more detail: male, 6 years old, Ph+ ALL. Three days after starting the induction phase Ib according to the EsPhALL protocol (cyclophosphamide, cytarabine, imatinib 300 mg/m per day £ 28 days administered orally, dissolved in apple juice), the patient developed a neurologic disorder, with asthenia, drowsiness, slowed speech and aphasia, with negative neuroimaging. Chemotherapy and imatinib were suspended, until full spontaneous clinical recovery; 25 days later, imatinib was restarted, initially at half of full dose (a new pharmacokinetic study was carried out when therapy restarted), then 70% and, Wnally, at full dose. This time the drug was taken orally, by swallowing the whole capsule. The patient underwent stem cell transplant and died due to treatment-related toxicity. For the pharmacokinetic study, blood samples were collected at pretreatment (before the morning dose) and 0.5, 1, 2, 4, 8 and 24 h after the Wrst dose and after at least one week of daily doses, presumably when the plasma imatinib concentration achieved steady-state. E. Marangon (&) · F. Sala · M. D’Incalci · M. Zucchetti Laboratory of Cancer Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche “Mario Negri”, Milan, Italy e-mail: [email protected]

Collaboration


Dive into the Elena Marangon's collaboration.

Top Co-Authors

Avatar

Giuseppe Toffoli

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Massimo Zucchetti

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Federica Sala

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar

Maurizio D'Incalci

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pietro Traldi

National Research Council

View shared research outputs
Top Co-Authors

Avatar

Renzo Bagnati

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge