Carboplatin Plus Paclitaxel Versus Carboplatin Plus Pegylated Liposomal Doxorubicin As First-Line Treatment for Patients With Ovarian Cancer: The MITO-2 Randomized Phase III Trial

  1. Francesco Perrone
  1. Author affiliations appear at the end of this article.
  1. Corresponding author: Sandro Pignata, MD, PhD, Istituto Nazionale Tumori, via Mariano Semmola, 80131 Napoli, Italy; e-mail: sandro.pignata{at}gmail.com.
  1. Presented in part at the 45th Annual Meeting of the American Society of Clinical Oncology, May 29-June 2, 2009, Orlando, FL; the 46th Annual Meeting of the American Society of Clinical Oncology, June 4-8, 2010, Chicago, IL; and the 35th Congress of the European Society of Medical Oncology, October 8-12, 2010, Milan, Italy.

Abstract

Purpose Carboplatin/paclitaxel is the standard first-line chemotherapy for patients with advanced ovarian cancer. Multicentre Italian Trials in Ovarian Cancer-2 (MITO-2), an academic multicenter phase III trial, tested whether carboplatin/pegylated liposomal doxorubicin (PLD) was more effective than standard chemotherapy.

Patients and Methods Chemotherapy-naive patients with stage IC to IV ovarian cancer (age ≤ 75 years; Eastern Cooperative Oncology Group performance status ≤ 2) were randomly assigned to carboplatin area under the curve (AUC) 5 plus paclitaxel 175 mg/m2 or to carboplatin AUC 5 plus PLD 30 mg/m2, every 3 weeks for six cycles. Primary end point was progression-free survival (PFS). With 632 events in 820 enrolled patients, the study would have 80% power to detect a 0.80 hazard ratio (HR) of PFS.

Results Eight hundred twenty patients were randomly assigned. Disease stages III and IV were prevalent. Occurrence of PFS events substantially slowed before obtaining the planned number. Therefore, in concert with the Independent Data Monitoring Committee, final analysis was performed with 556 events, after a median follow-up of 40 months. Median PFS times were 19.0 and 16.8 months with carboplatin/PLD and carboplatin/paclitaxel, respectively (HR, 0.95; 95% CI, 0.81 to 1.13; P = .58). Median overall survival times were 61.6 and 53.2 months with carboplatin/PLD and carboplatin/paclitaxel, respectively (HR, 0.89; 95% CI, 0.72 to 1.12; P = .32). Carboplatin/PLD produced a similar response rate but different toxicity (less neurotoxicity and alopecia but more hematologic adverse effects). There was no relevant difference in global quality of life after three and six cycles.

Conclusion Carboplatin/PLD was not superior to carboplatin/paclitaxel, which remains the standard first-line chemotherapy for advanced ovarian cancer. However, given the observed CIs and the different toxicity, carboplatin/PLD could be considered an alternative to standard therapy.

INTRODUCTION

Ovarian cancer is the fourth leading cause of cancer-related death in women.1 Intensive surgical staging and cytoreduction, followed by chemotherapy with carboplatin/paclitaxel, represent the standard treatment approach.26 However, even after optimal debulking surgery and response to systemic therapy, the risk of recurrence is high, and long-term survival remains poor. Furthermore, standard medical treatment of ovarian cancer negatively impacts on quality of life (QoL) as a result of frequent toxicity, such as alopecia, neurotoxicity, and fatigue.

Anthracyclines were used in the first-line treatment of advanced ovarian cancer before the introduction of taxanes, with data from meta-analyses suggesting that their addition to the platinum-based treatment might be advantageous.7 Pegylated liposomal doxorubicin (PLD) is a formulation of doxorubicin encapsulated in liposomes that are coated with methoxypolyethylene glycol, promoting the prolongation of circulation of the drug in the blood and its concentration in the tumor. This formulation results in a different pharmacokinetic and toxicity profile, as compared with conventional anthracyclines, with less myelotoxicity, alopecia, nausea, vomiting, and cardiotoxicity, but more skin and mucosal toxicity, namely palmer-plantar erythrodysesthesia and stomatitis.8 In a randomized phase III trial, compared with standard topotecan in second-line treatment, PLD produced better overall survival (OS) in platinum-sensitive patients and was characterized by a favorable tolerability profile, with less severe hematologic toxicity.9 On this basis, PLD is considered a treatment of choice for relapsed ovarian cancer. PLD was also compared to paclitaxel in the treatment of relapsed ovarian cancer.10 No significant efficacy difference was found between the two drugs, but PLD caused significantly less alopecia and neurotoxicity. This evidence encouraged the use of PLD in combination with carboplatin. Several phase I and II studies showed the possibility of easily combining PLD with standard doses and schedules of carboplatin.11,12 A Group d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens phase II study13 in patients with relapsed platinum-sensitive ovarian cancer demonstrated that the combination of carboplatin at area under the curve (AUC) 5 with PLD 30 mg/m2, given every 4 weeks, was active, with a 63% response rate and median progression-free survival (PFS) and OS times of 9.4 and 32 months, respectively. The combination was well tolerated; although nearly half of the patients experienced severe neutropenia, it was complicated by fever in only 3% of patients, and there was only mild to moderate nonhematologic toxicity, including palmer-plantar erythrodysesthesia (32%) and neuropathy (28%).

Therefore, we planned a randomized phase III clinical trial, Multicentre Italian Trials in Ovarian Cancer-2 (MITO-2), to evaluate whether carboplatin/PLD is superior in terms of PFS to the standard carboplatin/paclitaxel as first-line therapy of patients with advanced ovarian cancer. To plan the same dose-intensity of carboplatin in both arms, we chose an every-3-week schedule of carboplatin and PLD.

PATIENTS AND METHODS

Study Design

MITO-2 was an open-label, randomized, phase III study (Clinicaltrials.gov identifier: NCT00326456). The primary end point was PFS. Secondary end points included OS, treatment activity, toxicity, and QoL.

Overall, 820 patients were planned to be enrolled, and 632 events were needed to have 80% power of detecting a 0.80 hazard ratio (HR) of progression, with two-tailed α = .05 (East Software; Cytel, Cambridge, MA). This would represent an increase in median PFS from 18 to 22.5 months. No interim analyses were planned.

Patients were randomly assigned 1:1 to the standard or experimental arm. Telephone random assignment was performed centrally (Clinical Trials Unit, National Cancer Institute, Napoli, Italy) by a computer-driven minimization procedure. Stratification variables were center, residual disease after surgery (absent, ≤ 1 cm, > 1 cm, or no primary surgery), stage (IC, II, III, or IV), Eastern Cooperative Oncology Group performance status (0 to 1 or 2).

Study Population

Women younger than age 75 years, with a cytologic or histologic diagnosis of epithelial ovarian cancer (stage IC to IV according to International Federation of Gynecology and Obstetrics staging system), an Eastern Cooperative Oncology Group performance status ≤ 2, and a life expectancy of ≥ 3 months were eligible. Patients were excluded if they had received previous chemotherapy. Patients with a history of clinically relevant heart disease, with other concomitant diseases representing contraindication to treatment drugs, or with previous or concomitant other malignancy (except nonmelanoma skin cancer or carcinoma in situ of the uterine cervix) were not eligible. Adequate bone marrow, kidney, and liver functions were required. The study was approved by local ethics committees of each participating institution, and all patients gave written informed consent.

Study Treatment

Patients in the standard arm received carboplatin AUC 5 (dosed according to the Calvert formula, with creatinine clearance estimated according to the Cockcroft formula) and paclitaxel 175 mg/m2. Patients randomly assigned to the experimental arm received carboplatin AUC 5 and PLD (Caelyx; Schering-Plough, Kenilworth, NJ) 30 mg/m2. In both arms, chemotherapy was given intravenously on day 1 every 3 weeks. Carboplatin was diluted in 250 mL of glucose 5% and infused over 30 minutes. Paclitaxel was diluted in 250 mL of physiologic saline and infused over 3 hours. PLD was diluted in 250 mL of glucose 5% and infused over 60 minutes, following completion of carboplatin infusion. Treatment was initially administered for three cycles, and patients with stable or responding disease continued treatment for further a three cycles.

Conditions required for re-treatment were leukocytes more than 3,000/μL, neutrophils more than 1,500/μL, platelets ≥ 100,000/μL, and absence of organ toxicity (excluding alopecia) ≥ grade 2. Treatment was discontinued as a result of prolonged toxicity in patients requiring a treatment delay of ≥ 2 weeks.

A 20% dose reduction for all drugs was planned in patients with neutrophils less than 500/μL or platelets less than 50,000/μL for more than 7 days. Carboplatin dose was reduced to AUC 4 in patients with creatinine clearance less than 60 mL. In the event of ≥ grade 2 skin toxicity, PLD was delayed for up to 2 weeks or until toxicity resolved to ≤ grade 1; otherwise, PLD was interrupted. Subsequent doses were reduced by 25% if grade 3 or 4 skin toxicity cleared within 2 weeks. In the standard arm, doses of carboplatin and paclitaxel were reduced by 20% in presence of neuropathy.

Patient Evaluation

Computed tomography scan or nuclear magnetic resonance of the abdomen and pelvis and any other tests that gave positive results during staging were performed after three and six cycles of chemotherapy. Response evaluation was in accordance with Response Evaluation Criteria in Solid Tumors (RECIST) version 1.0.14 During the follow-up, the first among the following events defined progression: increase of more than 20% in the sum of largest diameters of known lesions; appearance of a new lesion; confirmed increase of more than 25% in CA-125; or death without clinical or instrumental signs of disease progression.

QoL was assessed using the European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire C30 (QLQ-C30).15 Toxicity was graded according to the National Cancer Institute Common Toxicity Criteria (version 2.0).

Statistical Analysis

Efficacy analyses were performed on an intent-to-treat basis. PFS was defined as the time interval between random assignment and progression or death, whichever occurred first, or last follow-up for patients alive without progression. OS was defined as the time interval between random assignment and death or date of last follow-up for patients still alive. Median follow-up was calculated according to the inverted Kaplan-Meier method.16 PFS and OS curves were estimated using the Kaplan-Meier product-limit method17 and compared using the log-rank test.18 For PFS, the Cox proportional hazards model19 was used to assess treatment effect adjusted by baseline prognostic variables.

A preplanned early activity analysis was performed in 2006, using data from the first 50 patients eligible for response assessment in the experimental arm.20 For the final analysis, objective response rate (ORR) was defined as the proportion of complete and partial responses among patients with at least one target lesion. Patients who died or stopped treatment because of toxicity or refusal before restaging were conservatively defined as nonresponders. The statistical significance of the difference in ORR between arms was assessed using the χ2 test. Activity was also described in women with nontarget lesions only and in women without any tumor lesion but with elevated CA-125 levels before starting treatment.

A preplanned toxicity analysis of the experimental combination was performed in 2004, based on the first 50 patients receiving experimental treatment.21 For the final toxicity analysis, all patients who received chemotherapy at least once were eligible. The worst grade of toxicity experienced was computed for each patient. For each toxicity, two statistical tests were performed to compare study arms; patterns of toxicity (considering all grades) were compared using an exact linear rank test, whereas rates of severe toxicity (grade ≥ 3 v grade 0 to 2) were compared using χ2 or Fisher's exact tests as appropriate.

QoL analysis was performed according to the EORTC manual.22 Multi-item scales were computed by calculating the mean raw scores and transforming them linearly, in scales ranging from 0 to 100. For single items, only linear transformation was performed. Changes from baseline after three and six cycles were calculated for each domain and compared between arms by a linear model, using baseline values as a covariate.

Statistical analyses were performed using S-Plus version 6.1 (Insightful, Seattle, WA). Exact tests were performed using StatXact 7 (Cytel).

RESULTS

Patient Characteristics

Between January 2003 and November 2007, 820 patients were randomly assigned, as planned (Fig 1). Baseline characteristics were balanced between the two arms (Table 1). Median age was 57 years (range, 21 to 77 years), and 97% of the patients had a performance status of 0 to 1. The majority of patients had advanced disease (60% with stage III and 21% with stage IV), and 45% of patients were enrolled after suboptimal surgical debulking.

Fig 1.

Flow of study procedures and data collection according to CONSORT diagram. QoL, quality of life; RECIST, Response Evaluation Criteria in Solid Tumors.

Table 1.

Baseline Clinical Characteristics of Patients by Treatment Arm

Treatment Compliance

Information on treatment received was not available in six patients. Of the remaining 814 patients, 10 patients did not start treatment (Table 2).

Table 2.

Compliance With Chemotherapy

Eighty-six percent and 81.1% of the patients completed six cycles in the standard and experimental arms, respectively. The proportion of patients delaying treatment because of toxicity, usually hematologic, was higher in the experimental arm at all cycles. Overall, 11.5% and 34.5% of cycles were delayed as a result of toxicity in the standard and experimental arms, respectively. Considering patients receiving six cycles, median times between first and last administration of chemotherapy were 16.3 and 18.1 weeks in the standard and experimental arms, respectively.

Efficacy

All randomly assigned patients were included in the intent-to-treat efficacy analysis. By the end of 2009, the PFS curve of the whole study population (not scattered by arm) showed that event occurrence substantially slowed before the required number of events was attained. More than half of the patients had optimal debulking, and more than one third did not have residual disease at all. This was a result of the significant proportion of patients with early-stage disease. The better prognosis of these subgroups of patients clearly conditioned the overall event rate (Appendix Fig A1, online only) and the time required to attain the planned events. Therefore, in concert with the Independent Data Monitoring Committee, the final analysis was anticipated at 556 events as of December 31, 2009. This number of events still allowed the detection of an HR for PFS of 0.79 with 80% power.

With a median follow-up time of 40 months, 282 events (68.8%) were recorded in the standard arm, and 274 events (66.8%) were recorded in the experimental arm. Median PFS was 16.8 months (95% CI, 15.2 to 19.4 months) in the standard arm and 19.0 months (95% CI, 16.3 to 24.0 months) in the experimental arm (HR, 0.95; 95% CI, 0.81 to 1.13; log-rank test, P = .58). PFS curves are shown in Figure 2A. In multivariable analysis adjusted by stage, performance status, residual disease, age, and size of the institution, the difference between treatments remained not significant (HR, 0.97; 95% CI, 0.82 to 1.14; P = .70), whereas residual disease and stage were independent predictors of PFS (Table 3). Exploratory analysis by subgroups according to International Federation of Gynecology and Obstetrics stage, performance status, age category, residual disease after surgery, and tumor histology showed no heterogeneity of treatment effect (Fig 3). With 313 deaths (38.2%) recorded, the median OS (Fig 2B) was 53.2 months (95% CI, 42.3 months to not available) in the standard arm compared with 61.6 months (95% CI, 50.4 months to not available) in the experimental arm (HR, 0.89; 95% CI, 0.72 to 1.12; log-rank test, P = .32).

Fig 2.

(A) Progression-free survival and (B) overall survival curves by treatment arm. HR, hazard ratio; NA, not available.

Table 3.

Cox Proportional Hazard Model for Progression-Free Survival

Fig 3.

Treatment effect on progression-free survival within major patient subgroups. Vertical dotted line represents hazard ratio (experimental arm v standard arm) in the overall study population. ECOG PS, Eastern Cooperative Oncology Group performance status.

Two-hundred ninety-seven patients (36.2%) were eligible for response analysis according to RECIST criteria (≥ one target lesion), 160 patients (39.0%) in the standard and 137 patients (33.4%) in the experimental arm. ORR was 59% in the standard arm (24 complete responses and 71 partial responses) and 57% in the experimental arm (23 complete responses and 55 partial responses; P = .76). In 184 patients with nontarget lesions only, complete response was 33% and 29% in the standard and experimental arms, respectively (P = .69). In 173 patients with elevated CA-125 only, CA-125 normalization was obtained in 82% and 86% of patients in the standard and experimental arms, respectively (P = .70; Appendix Table A1, online only).

Toxicity

All patients who received at least one dose were eligible for toxicity analysis (n = 803). Worst toxicities by treatment arm are listed in Table 4. There were six potentially treatment-related deaths—four in the standard arm (allergy during paclitaxel infusion followed by sudden death at home, n = 1; febrile neutropenia, n = 1; non-neutropenic fever, n = 1; intestinal necrosis with septic shock, n = 1) and two in the experimental arm (bleeding, n = 1; disseminated intravascular coagulation, n = 1).

Table 4.

Worst Grade (according to NCI-CTC) of Toxicity According to Treatment Arm

Thrombocytopenia and anemia were significantly more frequent and severe in the experimental arm; RBC transfusions were more frequently required in the experimental arm than the standard arm (6% v 2%, respectively; P = .001). There were no significant differences in leukopenia, neutropenia, febrile neutropenia, infections, platelet transfusions, and bleeding. The nonhematologic adverse effects profile was significantly different between the arms. Hair loss, diarrhea, and neuropathy were significantly worse in the standard arm, whereas skin toxicity and stomatitis were significantly worse in the experimental arm.

QoL

Overall, 620 patients (76%; 309 patients in the standard arm v 311 patients in the experimental arm) completed the valid baseline QoL questionnaire. Of these, 336 patients completed the questionnaire after three cycles (165 patients in the standard arm v 171 patients in the experimental arm), and 247 patients completed the questionnaire after six cycles (130 patients in the standard arm v 117 patients in the experimental arm). Mean difference from baseline in global QoL (EORTC QLQ-C30 items 29 and 30) was 3.70 in the standard arm versus 4.09 in the experimental arm after three cycles, and 8.07 in the standard arm versus 10.56 in the experimental arm after six cycles. Both comparisons were not statistically significant. No relevant differences were observed in all functional domains and in most symptom scales. Statistically significant differences were reported in loss of appetite after three cycles, favoring the standard treatment, and in diarrhea after three cycles, favoring the experimental arm (Appendix Fig A2, online only).

DISCUSSION

The MITO-2 trial compared an experimental regimen of carboplatin/PLD versus standard carboplatin/paclitaxel as first-line treatment of patients with advanced ovarian cancer. The experimental treatment did not significantly prolong PFS, the primary end point of the trial, and no differences were apparent in OS, ORR, and QoL. Nonhematologic toxicity significantly differed between the arms, with hair loss and neurotoxicity being drastically less frequent in the experimental arm, but with this arm having more skin toxicity and stomatitis. Hematologic toxicity was also worse with experimental treatment but within acceptable limits for clinical practice. Overall, given these results and considering that CIs of both PFS and OS completely lie within limits typically considered acceptable for noninferiority, the experimental treatment might be considered as an alternative to the standard treatment. This choice, of course, should take into account patient's will and consider limitations as a result of the lack of regulatory approval of PLD for first-line treatment and its cost.

The dramatic reduction of hair loss and neurotoxicity seen with the experimental treatment is extremely important. Approximately three fourths of patients experience some degree of peripheral neuropathy while on carboplatin/paclitaxel, and 7% have ≥ grade 3 peripheral neuropathy.5 We previously showed that residual neurotoxicity occurs frequently in patients after completion of carboplatin/paclitaxel, with a long-lasting pattern of recovery and 14% of patients still suffering residual neuropathy 1 year after treatment.23 In addition, such toxicity may still persist in a significant proportion of patients who experience relapse and, therefore, affects second-line treatment choice. However, in the MITO-2 trial, differences in toxicity pattern did not translate into relevant QoL differences. This might eventually depend on the fact that we limited QoL data collection to the treatment period, therefore missing late neurotoxicity. Furthermore, we only used the EORTC QLQ-C30 general questionnaire and not a more specific QoL questionnaire (eg, EORTC QLQ Ovarian Cancer Module).

The MITO-2 study, which studied the substitution of paclitaxel with PLD in combination with carboplatin, adds important evidence about the role of anthracyclines in first-line treatment of ovarian cancer. Meta-analysis of trials performed before the introduction of taxanes showed that the addition of doxorubicin prolonged survival7; however, on the contrary, in the taxane era, the addition of epirubicin to carboplatin/paclitaxel did not prolong OS and PFS in two phase III trials.24,25 In a recent phase III trial26 comparing carboplatin/PLD with carboplatin/paclitaxel in platinum-sensitive recurrent ovarian cancer, a statistically significant advantage in PFS in favor of carboplatin/PLD was found (HR of PFS, 0.821; P = .005; median PFS, 11.3 v 9.4 months, respectively). Carboplatin/PLD was associated with less alopecia and neurotoxicity.

To our knowledge, the only study testing PLD in first-line treatment of ovarian cancer is a phase III trial performed by the Gynecology Oncology Group comparing standard carboplatin/paclitaxel with two triplet regimens (standard carboplatin/paclitaxel plus gemcitabine or PLD) and two sequential doublet regimens (carboplatin plus topotecan or gemcitabine, both followed by carboplatin/paclitaxel).27 Although efficacy was similar across the five arms, hematologic toxicity increased with three-drug combinations. However, the information regarding PLD efficacy in this trial is limited because PLD was given in association with carboplatin/paclitaxel every other cycle, and the overall dose-intensity was 5 mg/m2/wk, much lower than that considered optimal.

Final MITO-2 analysis was performed with fewer events than planned (556 instead of 632 events), but it is unlikely that this affected the results. The reason was a dramatic decrease in the incidence of PFS events, which was not a result of flaws in follow-up procedures, consistent with plans, but rather a result of the favorable prognostic characteristics of the enrolled patients (significant proportion of patients with early-stage disease and of patients without residual disease after surgery). Therefore, an extremely longer time would have been required to get the planned number of events, and non–ovarian cancer deaths might dilute PFS differences. The Independent Data Monitoring Committee blindly advised to perform final analysis with the available events, considering that statistical power was reduced just to 75% and that there was still 80% power in detecting an HR for PFS of 0.79.

In conclusion, the MITO-2 trial shows that the combination of carboplatin/PLD does not prolong PFS compared with standard carboplatin/paclitaxel. However, given the observed difference in toxicity, it can be considered as a reasonable alternative for first-line treatment of advanced ovarian cancer, particularly in patients at high risk of neurotoxicity or wishing to avoid alopecia.

AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST

Although all authors completed the disclosure declaration, the following author(s) indicated a financial or other interest that is relevant to the subject matter under consideration in this article. Certain relationships marked with a “U” are those for which no compensation was received; those relationships marked with a “C” were compensated. For a detailed description of the disclosure categories, or for more information about ASCO's conflict of interest policy, please refer to the Author Disclosure Declaration and the Disclosures of Potential Conflicts of Interest section in Information for Contributors.

Employment or Leadership Position: None Consultant or Advisory Role: Alba Brandes, Schering-Plough (C) Stock Ownership: None Honoraria: Sandro Pignata, Schering-Plough; Roberto Sorio, Schering-Plough; Alba Brandes, Schering-Plough, Roche; Francesco Perrone, Schering-Plough Research Funding: Sandro Pignata, Schering-Plough; Francesco Perrone, Schering-Plough Expert Testimony: None Other Remuneration: None

AUTHOR CONTRIBUTIONS

Conception and design: Sandro Pignata, Giovanni Scambia, Ciro Gallo, Francesco Perrone

Collection and assembly of data: Sandro Pignata, Giovanni Scambia, Gabriella Ferrandina, Antonella Savarese, Roberto Sorio, Enrico Breda, Vittorio Gebbia, Pietro Musso, Luigi Frigerio, Pietro Del Medico, Alessandra Vernaglia Lombardi, Antonio Febbraro, Paolo Scollo, Antonella Ferro, Stefano Tamberi, Alba Brandes, Alberto Ravaioli, Maria Rosaria Valerio, Enrico Aitini, Donato Natale, Laura Scaltriti, Stefano Greggi, Carmela Pisano, Domenica Lorusso, Vanda Salutari, Francesco Legge, Massimo Di Maio, Alessandro Morabito, Francesco Perrone

Data analysis and interpretation: Sandro Pignata, Massimo Di Maio, Alessandro Morabito, Ciro Gallo, Francesco Perrone

Manuscript writing: All authors

Final approval of manuscript: All authors

Affiliations

Sandro Pignata, Stefano Greggi, Carmela Pisano, Massimo Di Maio, Alessandro Morabito, and Francesco Perrone, Istituto Nazionale Tumori; Ciro Gallo, Seconda Università di Napoli, Napoli; Giovanni Scambia, Domenica Lorusso, and Vanda Salutari, Policlinico Gemelli, Università Cattolica del Sacro Cuore; Antonella Savarese, Istituto Nazionale Tumori Regina Elena; Enrico Breda, Ospedale S. Giovanni Calibita Fatebenefratelli, Roma; Gabriella Ferrandina and Francesco Legge, Università Cattolica del Sacro Cuore, Campobasso; Roberto Sorio, Centro di Riferimento Oncologico, Aviano; Vittorio Gebbia, Casa di Cura La Maddalena; Pietro Musso, Azienda Ospedaliera di Rilievo Nazionale e di Alta Specializzazione Civico Di Cristina Benfratelli; Maria Rosaria Valerio, Policlinico Giaccone, Università, Palermo; Luigi Frigerio, Ospedali Riuniti, Bergamo; Pietro Del Medico, Ospedale Bianchi Melacrino Morelli, Reggio Calabria; Alessandra Vernaglia Lombardi, Casa di Cura Malzoni Villa dei Platani, Avellino; Antonio Febbraro, Ospedale Fatebenefratelli, Benevento; Paolo Scollo, Ospedale Cannizzaro, Catania; Antonella Ferro, Ospedale S. Chiara, Trento; Stefano Tamberi, Ospedale Civile, Faenza; Alba Brandes, Ospedale Bellaria, Bologna; Alberto Ravaioli, Ospedale degli Infermi, Rimini; Enrico Aitini, Ospedale Carlo Poma, Mantova; Donato Natale, Ospedale S. Massimo, Penne; and Laura Scaltriti, Ospedale Ramazzini, Carpi, Italy.

Appendix

MITO-2 Investigators

Sandro Pignata, Carmela Pisano, Gaetano Facchini, Giovanni Salvatore Bruni, Lucia Cannella, Davide Leopardo, Stefano Greggi, Francesco Iodice, Gennaro Casella (Dipartimento Uro-Ginecologico, Istituto Nazionale Tumori, Napoli); Giovanni Scambia, Domenica Lorusso, Vanda Salutari, Antonia Testa, Rosa De Vincenzo, Alessia di Legge, Claudia Masi, Valeria Masciullo, Mirella Di Stefano (Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, Roma); Gabriella Ferrandina, Francesco Legge, Giacomo Corrado, Marco Petrillo, Aida Distefano, Manuela Ludovisi (Centro di Ricerca e Formazione ad Alta Tecnologia nelle Scienze Biomediche, Università Cattolica del Sacro Cuore, Campobasso); Antonella Savarese, Francesco Cognetti, Gianluigi Ferretti, Enrico Vizza, Emanuela Mancini (Istituto Nazionale Tumori Regina Elena, Roma); Roberto Sorio, Simona Scalone, Giorgio Giorda, Elio Campagnutta (Centro di Riferimento Oncologico, Aviano [PN]); Enrico Breda, Antonella Mecozzi, Emanuela Proietti, Loredana Rossi, Angelo Fedele Scinto (Ospedale S. Giovanni Calibita Fatebenefratelli, Roma); Vittorio Gebbia, Eugenia Baiardi, Carlo Arcara, Antonio Testa (Casa di Cura La Maddalena, Palermo); Pietro Musso, Rosa Demma, Francesco Maria Re (Azienda Ospedaliera di Rilievo Nazionale e di Alta Specializzazione Civico Di Cristina Benfratelli, Palermo); Luigi Frigerio, Luisa Busci, Antonella Villa (present address, Clinica Mangiagalli, Milano), Laura Carlini (present address, Ospedale Bolognini, Segrate [BG]) (Ospedali Riuniti, Bergamo); Pietro Del Medico, Mario Nardi, Mario Raffaele, Domenico Azzarello (Ospedale Bianchi Melacrino Morelli, Reggio Calabria); Alessandra Vernaglia Lombardi, Carmine Malzoni, Mario Malzoni, Giuseppina Farnetano (Casa di Cura Malzoni Villa dei Platani, Avellino); Antonio Febbraro, Claudia Corbo, Ilaria Spagnoletti (Ospedale Fatebenefratelli, Benevento); Paolo Scollo, Giuseppe Scibilia, Gabriella D'Agate (Ospedale Cannizzaro, Catania); Enzo Galligioni, Antonella Ferro, Viviana Murgia (Ospedale S. Chiara, Trento); Stefano Tamberi, Laura Amaducci, Carmelo Bucolo (Ospedale Civile, Faenza); Alba Brandes, Simonetta Rimondini, Stefania Bartolini (Ospedale Bellaria, Bologna); Alberto Ravaioli, Giovanni Oliverio, Enzo Pasquini (Ospedale degli Infermi, Rimini, Ospedale Cervesi, Cattolica [RN]); Maria Rosaria Valerio, Mario Lomauro, Nicola Gebbia (Policlinico Giaccone, Università, Palermo); Enrico Aitini, Giovanna Cavazzini, Francesca Adami (Ospedale Carlo Poma, Mantova); Donato Natale, Bruna Fornarini, Dante Orlando (Ospedale S. Massimo, Penne [PE]); Fabrizio Artioli, Laura Scaltriti, Lorenzo Aguzzoli (Ospedale Ramazzini, Carpi [MO]); Emanuele Naglieri, Agnese Latorre, Sabina Delcuratolo (Istituto Tumori “Giovanni Paolo II,” Bari); Cesare Gridelli, Filomena Del Gaizo (Ospedale S.Giuseppe Moscati, Avellino); Alfonso M. D'Arco, Antonietta Fabbrocini (Ospedale Civile Umberto I, Nocera Inferiore [SA]); Isabel Henriques, Ondina Campos (CHC Maternidade Bissaya-Barreto, Coimbra, Portugal); Bruno Massidda, Valeria Pusceddu (Policlinico Universitario Monserrato, Cagliari); Rossella Lauria, Valeria Forestieri (Università Federico II, Napoli); Marina Marzola, Pamela Pizzutilo (Arcispedale Sant'Anna, Ferrara); Rocco De Vivo (Ospedale S. Bortolo ULSS 6, Vicenza); Giovanni Lore (Ospedale S. Maria degli Angeli, Pordenone); Saverio Danese (Ospedale S. Anna, Torino); Nada Riva (Istituto Romagnolo per lo Studio e la Cura dei Tumori, Meldola [FC]); Vito Lorusso (Ospedale Vito Fazzi, Lecce); Gabriella Landi (Oncologia Medica Senologica, Istituto Nazionale Tumori, Napoli); Giuseppe Nettis (Ospedale Miulli, Acquaviva delle Fonti [BA]); Filomena Narducci (Ospedale SS. Trinità Sora [FR]); Rosalbino Biamonte (Ospedale Mariano Santo, Cosenza); Vincenzo Montesarchio (Ospedale Cotugno, Napoli); Giancarlo Balbi (Clinica Ostetrica e Ginecologica, Seconda Università, Napoli); Cosimo Sacco (Azienda Ospedaliero Universitaria, Udine); Vincenzo Arigliano (Ospedale di Bentivoglio [BO]); Simona Losito (Anatomia Patologica, Istituto Nazionale Tumori, Napoli); Ciro Gallo, Simona Signoriello, Paolo Chiodini, Giuseppe Signoriello (Statistica Medica, Seconda Università, Napoli); Francesco Perrone, Aldo Vecchione, Massimo Di Maio, Alessandro Morabito, Maria Carmela Piccirillo, Ermelinda De Maio (present address, Ospedali Riuniti, Livorno), Antonia Del Giudice, Fabiano Falasconi, Jane Bryce, Gianfranco De Feo, Giuliana Canzanella, Alfonso Savio (Unità Sperimentazioni Cliniche, Istituto Nazionale Tumori, Napoli, Italy).

Data Monitoring Committee

Nicoletta Colombo (Istituto Europeo di Oncologia, Milano); Paolo Bruzzi (Istituto Nazionale per la Ricerca sul Cancro, Genova); Roldano Fossati (Istituto di Ricerche Farmacologiche Mario Negri, Milano), Italy.

Data Managers

Giuliana Canzanella, Federika Crudele, Manuela Florio, Fiorella Romano, Giovanni de Matteis, Balbina Apice, Rosa Nunziata (Napoli); Agnese Provenziani (Roma); Gianna Tabaro (Aviano [PN]); Paolo Russo (Palermo); Antonella Falzea (Reggio Calabria); Alessandra Piancastelli, Emanuela Montanari (Faenza [RA]); Giuliana Drudi, Barbara Venturini (Rimini, Cattolica [RN]); Patrizia Morselli, Beatrice Vivorio (Mantova); Roberta Guerzoni, Giorgia Razzini (Carpi [MO]); Valentina Barbato, Rita Ambrosio (Avellino); Sabrina Prisco (Nocera Inferiore [SA]); Elena Raisi (Ferrara); Bruna Stupar (Vicenza); Michela Ballardini (Meldola [FC]); Luciana Petrucelli (Lecce); and Marica Gaiardo (Udine), Italy.

Research Nurses

Jane Bryce, Marzia Falanga, Immacolata Gargiulo (Napoli); Ilaria Orsini, Tonia Marra, Chiara Frediani, Alessandra Focaccetti (Roma); Annamaria Colussi, Sara Rosalen (Aviano [PN]); Michela Pilati (Rimini, Cattolica [RN]); Paola Costantini (Udine), Italy.

Fig A1.

Progression-free survival curves of patients enrolled onto the trial by (A) residual disease after surgery and (B) tumor stage.

Fig A2.

Quality-of-life (QoL) analysis for standard (blue bars) versus experimental (gold bars) arms. Bars correspond to mean changes from baseline after three and after six cycles. (A) For global QoL and functional domains, improvement is indicated by positive changes. (B) For symptoms, improvement is indicated by negative changes. (*) Statistically significant difference between arms, P < .05.

Table A1.

Intent-to-Treat Analysis of Treatment Activity

Footnotes

  • Written on behalf of the Multicentre Italian Trials in Ovarian Cancer-2 (MITO-2) Investigators.

  • Supported in part by Integrated Therapeutics Group (ITGI) and Schering-Plough Italy, which supplied the experimental drug, and by the nonprofit Italian Association for Cancer Research.

  • ITGI and Schering-Plough Italy had no role in trial design and data interpretation.

  • Authors' disclosures of potential conflicts of interest and author contributions are found at the end of this article.

  • Clinical trial information can be found for the following: NCT00326456.

  • Received November 18, 2010.
  • Accepted May 27, 2011.

REFERENCES

| Table of Contents

This Article

  1. JCO vol. 29 no. 27 3628-3635

Classifications

  • Advertisement
  • Advertisement
  • Advertisement