Network


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

Hotspot


Dive into the research topics where Libor Havel is active.

Publication


Featured researches published by Libor Havel.


The New England Journal of Medicine | 2015

Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer.

Julie R. Brahmer; Karen L. Reckamp; P. Baas; Lucio Crinò; Wilfried Eberhardt; Elena Poddubskaya; Scott Antonia; Adam Pluzanski; Everett E. Vokes; Esther Holgado; David Waterhouse; Neal Ready; Justin F. Gainor; Osvaldo Arén Frontera; Libor Havel; Martin Steins; Marina C. Garassino; Joachim Aerts; Manuel Domine; Luis Paz-Ares; Martin Reck; Christine Baudelet; Christopher T. Harbison; Brian Lestini; David R. Spigel

BACKGROUND Patients with advanced squamous-cell non-small-cell lung cancer (NSCLC) who have disease progression during or after first-line chemotherapy have limited treatment options. This randomized, open-label, international, phase 3 study evaluated the efficacy and safety of nivolumab, a fully human IgG4 programmed death 1 (PD-1) immune-checkpoint-inhibitor antibody, as compared with docetaxel in this patient population. METHODS We randomly assigned 272 patients to receive nivolumab, at a dose of 3 mg per kilogram of body weight every 2 weeks, or docetaxel, at a dose of 75 mg per square meter of body-surface area every 3 weeks. The primary end point was overall survival. RESULTS The median overall survival was 9.2 months (95% confidence interval [CI], 7.3 to 13.3) with nivolumab versus 6.0 months (95% CI, 5.1 to 7.3) with docetaxel. The risk of death was 41% lower with nivolumab than with docetaxel (hazard ratio, 0.59; 95% CI, 0.44 to 0.79; P<0.001). At 1 year, the overall survival rate was 42% (95% CI, 34 to 50) with nivolumab versus 24% (95% CI, 17 to 31) with docetaxel. The response rate was 20% with nivolumab versus 9% with docetaxel (P=0.008). The median progression-free survival was 3.5 months with nivolumab versus 2.8 months with docetaxel (hazard ratio for death or disease progression, 0.62; 95% CI, 0.47 to 0.81; P<0.001). The expression of the PD-1 ligand (PD-L1) was neither prognostic nor predictive of benefit. Treatment-related adverse events of grade 3 or 4 were reported in 7% of the patients in the nivolumab group as compared with 55% of those in the docetaxel group. CONCLUSIONS Among patients with advanced, previously treated squamous-cell NSCLC, overall survival, response rate, and progression-free survival were significantly better with nivolumab than with docetaxel, regardless of PD-L1 expression level. (Funded by Bristol-Myers Squibb; CheckMate 017 ClinicalTrials.gov number, NCT01642004.).


The New England Journal of Medicine | 2017

First-Line Nivolumab in Stage IV or Recurrent Non–Small-Cell Lung Cancer

David P. Carbone; Martin Reck; Luis Paz-Ares; Benjamin C. Creelan; Leora Horn; Martin Steins; Enriqueta Felip; Michel M. van den Heuvel; Tudor-Eliade Ciuleanu; Firas Benyamine Badin; Neal Ready; T. Jeroen N. Hiltermann; Suresh R. Nair; Rosalyn A. Juergens; Solange Peters; Elisa Minenza; John Wrangle; Delvys Rodriguez-Abreu; Hossein Borghaei; George R. Blumenschein; Liza C. Villaruz; Libor Havel; J. Krejčí; Jesus Corral Jaime; Han Chang; William J. Geese; Prabhu Bhagavatheeswaran; Allen C. Chen; Mark A. Socinski

BACKGROUND Nivolumab has been associated with longer overall survival than docetaxel among patients with previously treated non–small‐cell lung cancer (NSCLC). In an open‐label phase 3 trial, we compared first‐line nivolumab with chemotherapy in patients with programmed death ligand 1 (PD‐L1)–positive NSCLC. METHODS We randomly assigned, in a 1:1 ratio, patients with untreated stage IV or recurrent NSCLC and a PD‐L1 tumor‐expression level of 1% or more to receive nivolumab (administered intravenously at a dose of 3 mg per kilogram of body weight once every 2 weeks) or platinum‐based chemotherapy (administered once every 3 weeks for up to six cycles). Patients receiving chemotherapy could cross over to receive nivolumab at the time of disease progression. The primary end point was progression‐free survival, as assessed by means of blinded independent central review, among patients with a PD‐L1 expression level of 5% or more. RESULTS Among the 423 patients with a PD‐L1 expression level of 5% or more, the median progression‐free survival was 4.2 months with nivolumab versus 5.9 months with chemotherapy (hazard ratio for disease progression or death, 1.15; 95% confidence interval [CI], 0.91 to 1.45; P=0.25), and the median overall survival was 14.4 months versus 13.2 months (hazard ratio for death, 1.02; 95% CI, 0.80 to 1.30). A total of 128 of 212 patients (60%) in the chemotherapy group received nivolumab as subsequent therapy. Treatment‐related adverse events of any grade occurred in 71% of the patients who received nivolumab and in 92% of those who received chemotherapy. Treatment‐related adverse events of grade 3 or 4 occurred in 18% of the patients who received nivolumab and in 51% of those who received chemotherapy. CONCLUSIONS Nivolumab was not associated with significantly longer progression‐free survival than chemotherapy among patients with previously untreated stage IV or recurrent NSCLC with a PD‐L1 expression level of 5% or more. Overall survival was similar between groups. Nivolumab had a favorable safety profile, as compared with chemotherapy, with no new or unexpected safety signals. (Funded by Bristol‐Myers Squibb and others; CheckMate 026 ClinicalTrials.gov number, NCT02041533.)


Lancet Oncology | 2014

Tecemotide (L-BLP25) versus placebo after chemoradiotherapy for stage III non-small-cell lung cancer (START): a randomised, double-blind, phase 3 trial

Charles Butts; Mark A. Socinski; Paul Mitchell; Nick Thatcher; Libor Havel; Maciej Krzakowski; Sergiusz Nawrocki; Tudor-Eliade Ciuleanu; Lionel Bosquée; José Manuel Trigo; Alexander Spira; Lise Tremblay; Jan Nyman; Rodryg Ramlau; Gun Wickart-Johansson; Peter M. Ellis; Oleg Gladkov; Jose R. Pereira; Wilfried Eberhardt; Christoph Helwig; Andreas Schröder; Frances A. Shepherd

BACKGROUND Effective maintenance therapies after chemoradiotherapy for lung cancer are lacking. Our aim was to investigate whether the MUC1 antigen-specific cancer immunotherapy tecemotide improves survival in patients with stage III unresectable non-small-cell lung cancer when given as maintenance therapy after chemoradiation. METHODS The phase 3 START trial was an international, randomised, double-blind trial that recruited patients with unresectable stage III non-small-cell lung cancer who had completed chemoradiotherapy within the 4-12 week window before randomisation and received confirmation of stable disease or objective response. Patients were stratified by stage (IIIA vs IIIB), response to chemoradiotherapy (stable disease vs objective response), delivery of chemoradiotherapy (concurrent vs sequential), and region using block randomisation, and were randomly assigned (2:1, double-blind) by a central interactive voice randomisation system to either tecemotide or placebo. Injections of tecemotide (806 μg lipopeptide) or placebo were given every week for 8 weeks, and then every 6 weeks until disease progression or withdrawal. Cyclophosphamide 300 mg/m(2) (before tecemotide) or saline (before placebo) was given once before the first study drug administration. The primary endpoint was overall survival in a modified intention-to-treat population. This study is registered with ClinicalTrials.gov, number NCT00409188. FINDINGS From Feb 22, 2007, to Nov 15, 2011, 1513 patients were randomly assigned (1006 to tecemotide and 507 to placebo). 274 patients were excluded from the primary analysis population as a result of a clinical hold, resulting in analysis of 829 patients in the tecemotide group and 410 in the placebo group in the modified intention-to-treat population. Median overall survival was 25.6 months (95% CI 22.5-29.2) with tecemotide versus 22.3 months (19.6-25.5) with placebo (adjusted HR 0.88, 0.75-1.03; p=0.123). In the patients who received previous concurrent chemoradiotherapy, median overall survival for the 538 (65%) of 829 patients assigned to tecemotide was 30.8 months (95% CI 25.6-36.8) compared with 20.6 months (17.4-23.9) for the 268 (65%) of 410 patients assigned to placebo (adjusted HR 0.78, 0.64-0.95; p=0.016). In patients who received previous sequential chemoradiotherapy, overall survival did not differ between the 291 (35%) patients in the tecemotide group and the 142 (35%) patients in the placebo group (19.4 months [95% CI 17.6-23.1] vs 24.6 months [18.8-33.0], respectively; adjusted HR 1.12, 0.87-1.44; p=0.38). Grade 3-4 adverse events seen with a greater than 2% frequency with tecemotide were dyspnoea (49 [5%] of 1024 patients in the tecemotide group vs 21 [4%] of 477 patients in the placebo group), metastases to central nervous system (29 [3%] vs 6 [1%]), and pneumonia (23 [2%] vs 12 [3%]). Serious adverse events with a greater than 2% frequency with tecemotide were pneumonia (30 [3%] in the tecemotide group vs 14 [3%] in the placebo group), dyspnoea (29 [3%] vs 13 [3%]), and metastases to central nervous system (32 [3%] vs 9 [2%]). Serious immune-related adverse events did not differ between groups. INTERPRETATION We found no significant difference in overall survival with the administration of tecemotide after chemoradiotherapy compared with placebo for all patients with unresectable stage III non-small-cell lung cancer. However, tecemotide might have a role for patients who initially receive concurrent chemoradiotherapy, and further study in this population is warranted. FUNDING Merck KGaA (Darmstadt, Germany).


Lung Cancer | 2003

Gemcitabine plus cisplatin vs. gemcitabine plus carboplatin in stage IIIb and IV non-small cell lung cancer: a phase III randomized trial

Petr Zatloukal; Luboš Petruželka; Milada Zemanová; Vítězslav Kolek; Jana Skřičková; Miloš Pešek; Hana Fojtů; Ivona Grygárková; Dimka Sixtová; Jaromír Roubec; Eva Hořenková; Libor Havel; Petr Průša; Leona Nováková; Tomáš Skácel; Milan Kůta

PURPOSE This randomized, multicenter, phase III trial was conducted to compare the tolerability of gemcitabine plus cisplatin (GP) vs. gemcitabine plus carboplatin (GC) in chemonaive patients with stage IIIb and IV non-small cell lung carcinoma (NSCLC). Secondary objectives were to evaluate response, duration of response, time to progressive disease (TTPD), and survival. PATIENTS AND METHODS Eligible patients were required to have stage IIIb or IV NSCLC, no previous chemotherapy, Karnofsky performance status of at least 70, bidimensionally measurable disease, and age 18-75 years. Randomized patients in both arms were given gemcitabine 1200 mg/m(2) on days 1 and 8, followed on day 1 by cisplatin 80 mg/m(2) (GP) or carboplatin AUC=5 (GC). Treatment cycles were repeated every 21 days for a maximum of six cycles, or until disease progression or unacceptable toxicity occurred. RESULTS Enrolled patients in both arms, 87 in GP and 89 in GC, were well balanced for demographics and disease characteristics. Dose intensity was 93.8 and 92.7% for gemcitabine in GP/GC arms, respectively; 97.7% for cisplatin and 99.9% for carboplatin. Patients with at least one grade 3/4 toxicity excluding nausea, vomiting or alopecia, were 44% in GP arm and 54% in GC arm. The only significantly different toxicities were, nausea and vomiting in GP and thrombocytopenia in GC group. The overall response rates, median TTPD, response duration and survival were, 41/29%, 5.87/4.75 months, 7.48/5.15 months, and 8.75/7.97 months for GP and GC arms, respectively. CONCLUSION GP and GC are effective and feasible regimens for advanced NSCLC, and are comparable in efficacy and toxicity. GC may offer acceptable option to patients with advanced NSCLC, especially those who are unable to receive cisplatin.


Lancet Oncology | 2016

Efficacy of the MAGE-A3 cancer immunotherapeutic as adjuvant therapy in patients with resected MAGE-A3-positive non-small-cell lung cancer (MAGRIT): a randomised, double-blind, placebo-controlled, phase 3 trial

Johan Vansteenkiste; Byoung Chul Cho; Tonu Vanakesa; Tommaso De Pas; Marcin Zieliński; Moon Soo Kim; Jacek Jassem; Masahiro Yoshimura; Jubrail Dahabreh; Haruhiku Nakayama; Libor Havel; Haruhiko Kondo; Tetsuya Mitsudomi; Konstantinos Zarogoulidis; Oleg Gladkov; Katalin Udud; Hirohito Tada; Hans Hoffman; Anders Bugge; Paul Taylor; Emilio Esteban González; Mei Lin Liao; Jianxing He; Jean Louis Pujol; Jamila Louahed; Muriel Debois; Vincent Brichard; Channa Debruyne; Patrick Therasse; Nasser K. Altorki

BACKGROUND Fewer than half of the patients with completely resected non-small-cell lung cancer (NSCLC) are cured. Since the introduction of adjuvant chemotherapy in 2004, no substantial progress has been made in adjuvant treatment. We aimed to assess the efficacy of the MAGE-A3 cancer immunotherapeutic in surgically resected NSCLC. METHODS In this randomised, double-blind, placebo-controlled trial, we recruited patients aged at least 18 years with completely resected stage IB, II, and IIIA MAGE-A3-positive NSCLC who did or did not receive adjuvant chemotherapy from 443 centres in 34 countries (Europe, the Americas, and Asia Pacific). Patients were randomly assigned (2:1) to receive 13 intramuscular injections of recMAGE-A3 with AS15 immunostimulant (MAGE-A3 immunotherapeutic) or placebo during 27 months. Randomisation and treatment allocation at the investigator site was done centrally via internet with stratification for chemotherapy versus no chemotherapy. Participants, investigators, and those assessing outcomes were masked to group assignment. A minimisation algorithm accounted for the number of chemotherapy cycles received, disease stage, lymph node sampling procedure, performance status score, and lifetime smoking status. The primary endpoint was broken up into three co-primary objectives: disease-free survival in the overall population, the no-chemotherapy population, and patients with a potentially predictive gene signature. The final analyses included the total treated population (all patients who had received at least one treatment dose). This trial is registered with ClinicalTrials.gov, number NCT00480025. FINDINGS Between Oct 18, 2007, and July 17, 2012, we screened 13 849 patients for MAGE-A3 expression; 12 820 had a valid sample and of these, 4210 (33%) had a MAGE-A3-positive tumour. 2312 of these patients met all eligibility criteria and were randomly assigned to treatment: 1515 received MAGE-A3 and 757 received placebo and 40 were randomly assigned but never started treatment. 784 patients in the MAGE-A3 group also received chemotherapy, as did 392 in the placebo group. Median follow-up was 38·1 months (IQR 27·9-48·4) in the MAGE-A3 group and 39·5 months (27·9-50·4) in the placebo group. In the overall population, median disease-free survival was 60·5 months (95% CI 57·2-not reached) for the MAGE-A3 immunotherapeutic group and 57·9 months (55·7-not reached) for the placebo group (hazard ratio [HR] 1·02, 95% CI 0·89-1·18; p=0·74). Of the patients who did not receive chemotherapy, median disease-free survival was 58·0 months (95% CI 56·6-not reached) in those in the MAGE-A3 group and 56·9 months (44·4-not reached) in the placebo group (HR 0·97, 95% CI 0·80-1·18; p=0·76). Because of the absence of treatment effect, we could not identify a gene signature predictive of clinical benefit to MAGE-A3 immunotherapeutic. The frequency of grade 3 or worse adverse events was similar between treatment groups (246 [16%] of 1515 patients in the MAGE-A3 group and 122 [16%] of 757 in the placebo group). The most frequently reported grade 3 or higher adverse events were infections and infestations (37 [2%] in the MAGE-A3 group and 19 [3%] in the placebo group), vascular disorders (30 [2%] vs 17 [3%]), and neoplasm (benign, malignant, and unspecified (29 [2%] vs 16 [2%]). INTERPRETATION Adjuvant treatment with the MAGE-A3 immunotherapeutic did not increase disease-free survival compared with placebo in patients with MAGE-A3-positive surgically resected NSCLC. Based on our results, further development of the MAGE-A3 immunotherapeutic for use in NSCLC has been stopped. FUNDING GlaxoSmithKline Biologicals SA.


Journal of Clinical Oncology | 2016

Phase III Randomized Trial of Ipilimumab Plus Etoposide and Platinum Versus Placebo Plus Etoposide and Platinum in Extensive-Stage Small-Cell Lung Cancer

Martin Reck; Alexander Luft; Aleksandra Szczesna; Libor Havel; Sang We Kim; Wallace Akerley; Maria Catherine Pietanza; Yi-Long Wu; Christoph C. Zielinski; Michael Thomas; Enriqueta Felip; Kathryn A. Gold; Leora Horn; Joachim Aerts; Kazuhiko Nakagawa; Paul Lorigan; Anne Pieters; Teresa Sanchez; Justin Fairchild; David R. Spigel

Purpose Patients with extensive-stage disease small-cell lung cancer (SCLC) have poor survival outcomes despite first-line chemotherapy with etoposide and platinum. This randomized, double-blind phase III study evaluated the efficacy and safety of ipilimumab or placebo plus etoposide and platinum in patients with newly diagnosed extensive-stage disease SCLC. Patients and Methods Patients were randomly assigned at a ratio of one to one to receive chemotherapy with etoposide and platinum (cisplatin or carboplatin) plus ipilimumab 10 mg/kg or placebo every 3 weeks for a total of four doses each in a phased induction schedule (chemotherapy in cycles one to four; ipilimumab or placebo beginning in cycle three up to cycle six), followed by ipilimumab or placebo maintenance every 12 weeks. Primary end point was overall survival (OS) among patients receiving at least one dose of blinded study therapy. Results Of 1,132 patients randomly assigned, 954 received at least one dose of study therapy (chemotherapy plus ipilimumab, n = 478; chemotherapy plus placebo, n = 476). Median OS was 11.0 months for chemotherapy plus ipilimumab versus 10.9 months for chemotherapy plus placebo (hazard ratio, 0.94; 95% CI, 0.81 to 1.09; P = .3775). Median progression-free survival was 4.6 months for chemotherapy plus ipilimumab versus 4.4 months for chemotherapy plus placebo (hazard ratio, 0.85; 95% CI, 0.75 to 0.97). Rates and severity of treatment-related adverse events were similar between arms, except for diarrhea, rash, and colitis, which were more frequent with chemotherapy plus ipilimumab. Rate of treatment-related discontinuation was higher with chemotherapy plus ipilimumab (18% v 2% with chemotherapy plus placebo). Five treatment-related deaths occurred with chemotherapy plus ipilimumab and two with chemotherapy plus placebo. Conclusion Addition of ipilimumab to chemotherapy did not prolong OS versus chemotherapy alone in patients with newly diagnosed extensive-stage disease SCLC. No new or unexpected adverse events were observed with chemotherapy plus ipilimumab. Several ongoing studies are evaluating ipilimumab in combination with programmed death-1 inhibitors in SCLC.


Annals of Oncology | 2014

1173OMAGRIT, A DOUBLE-BLIND, RANDOMIZED, PLACEBO-CONTROLLED PHASE III STUDY TO ASSESS THE EFFICACY OF THE RECMAGE-A3 + AS15 CANCER IMMUNOTHERAPEUTIC AS ADJUVANT THERAPY IN PATIENTS WITH RESECTED MAGE-A3-POSITIVE NON-SMALL CELL LUNG CANCER (NSCLC)

Johan Vansteenkiste; Byoung Chul Cho; Tonu Vanakesa; T. De Pas; Mark R. Zielinski; M.S. Kim; Jacek Jassem; Masahiro Yoshimura; Jubrail Dahabreh; H. Nakayama; Libor Havel; Haruhiko Kondo; Tetsuya Mitsudomi; Konstantinos Zarogoulidis; Oleg Gladkov; B. Spiessens; Vincent Brichard; Channa Debruyne; Patrick Therasse; Nasser K. Altorki

ABSTRACT Aim: Adjuvant chemotherapy (ACT) is the standard of care for Stage II and IIIA NSCLC, and for high risk Stage IB NSCLC. However, the 5-year disease-free survival remains poor (35-50%) and about half of the patients will not receive ACT for various reasons. This Phase III trial investigated whether the recMAGE-A3 + AS15 cancer immunotherapeutic (MAGE-A3 CI) as adjuvant therapy improved disease-free survival (DFS) in patients with resected NSCLC. Methods: MAGRIT was a randomized, double-blind, placebo-controlled trial in patients with completely resected MAGE-A3-positive NSCLC Stages IB, II, and IIIA (TNM version 6) and who did or did not receive ACT. Patients were randomly assigned (2:1) to receive 13 intramuscular injections of MAGE-A3 CI or placebo over a 27-month (m) treatment period. The three co-primary endpoints were DFS in the overall and in the no-ACT population and DFS in patients with a potentially predictive gene signature (GS). Results: Out of 13,849 patients screened, 4,210 patients had a MAGE-A3 positive tumour sample and 2,272 patients were randomised and treated. Overall, 52% of the patients received ACT; 47%, 36% and 17% were Stage IB, II and IIIA, respectively. Median age was 63 years and 24% of patients were females. Mean relative dose intensity was above 98% in both groups throughout the treatment period. Median follow-up at the time of final analysis was 38.8m. Median DFS was 60.5m and 57.9m respectively for MAGE-A3 CI and placebo (HR 1.024, 95% CI 0.891-1.177; p = 0.7379). In patients who did not receive ACT, median DFS was 58.0m and 56.9m for MAGE-A3 CI and placebo groups, respectively (HR 0.970, 95% CI 0.797-1.179; p = 0.7572). The rate of grade ≥ 3 adverse events (16%) did not differ between treatment groups. Conclusions: Treatment of NSCLC patients with MAGE-A3 CI did not increase DFS compared to placebo in either the overall population or in patients who did not receive ACT. Due to the absence of treatment effect, a GS predictive of clinical benefit to MAGE-A3 CI could not be identified. Funding Source: GlaxoSmithKline Biologicals SA. Disclosure: J.F. Vansteenkiste: Pr Vansteenkiste received GSK fees as Primary investigator for the MAGRIT study; B. Cho: Dr Cho received consultancy fees from Novartis and Boehringer-ingelheim; T. De Pas: No conflicts of interest. Fee received from GSK as member of steering committee of the study; J. Jassem: Dr Jassem received grant and personal fees for Consultancy from GSK; M. Yoshimura: Pr Yoshimura received GSK fees as Primary investigator for the MAGRIT and PEARL study; H. Nakayama: Dr Nakayama received GSK fees as Primary investigator for the MAGRIT and PEARL study; T. Mitsudomi: Dr Mitsudomi received personal fees from GSK for an advisory role; B. Spiessens: Bart Spiessens is employee of GSK and do own stock options of GSK; V. Brichard: Dr Birchart is GSK employee and do own Stock options from GSK; C. Debruyne: Dr Debruyne is GSK employee and do own GSK stock options; P. Therasse: GSK employee and Stock options owner; N. Altorki: Dr Altorki received GSK fees for trial conduct of GSK studies. All other authors have declared no conflicts of interest.


Lung Cancer | 2001

A case-control study of lung cancer among Czech women

Antonin Kubik; Petr Zatloukal; Peter Boyle; Chris Robertson; Sara Gandini; Ladislav Tomasek; Nigel Gray; Libor Havel

Few data are available to explain the continuing increase in lung cancer mortality among Czech women. The study was designed to examine the role of active smoking and other known or suspected factors. Data collected by personal interviews during the 15 months of a hospital-based case control study are reported. A total of 140 microscopically confirmed cases and 280 frequency-matched controls were analysed using multiple logistic regression. Cigarette smoking was the most important factor associated with excess risk for lung cancer among women. Significantly increased risk was found both among current smokers (OR=11.20, 95% CI 5.9-21.2), and ex-smokers (OR=10.02, 95% CI 5.5-18.4). Positive dose-response gradients (P<0.001) were observed between lung cancer risk and the daily number of cigarettes, duration of smoking, number of pack-years, inhaling, and grade of nicotine dependence assessed by the Fagerström test (Heatherton TF, Kozlowski LT, Frecker RC, Fagerström KO. Br J Addict 1991;86:1119-1470; Pomerleau OF. In: Bolliger CT, Fagerström KO, editors. The Tobacco Epidemic. Basle: Karger, 1997: 122-131). Exposure to environmental smoke was associated with elevated lung cancer risk (OR=3.58, for lifetime non-smokers exposed both in childhood and in adult age). Physical exercise and body mass index were inversely associated with lung cancer risk. For the category of physical exercise of more than 5 h per week, the odds ratio was 0.38, compared to subjects admitting no physical exercise. For body mass index, the odds ratio for the highest (compared to the lowest) quartile was 0.50. Chronic cough and phlegm (at least 3 months per year) were associated with excess risk (OR=6.07) only if their duration was less than 2 years before diagnosis of lung cancer, and, therefore, they were suspected of being more likely early symptoms of preclinical lung cancer rather than its cause. Our results support the statement that cigarette smoking is by far the most important cause of the on-going epidemic of lung cancer among Czech women, and are consistent with the concept of a balance between risk and protective factors whose eventual maintenance or alteration determine the development of disease (as suggested by Rylander R, Axelsson G, Andersson L, Liljequist T, Bergman B. Lung Cancer 1996;14(Suppl 1): S75-S83). Concerted control of smoking appears to be an urgent priority in lung cancer prevention among women, including specific approaches targeted on the female population.


Journal of Thoracic Oncology | 2016

P2.36: Nivolumab (nivo) in Patients (pts) With Advanced (adv) NSCLC and Central Nervous System (CNS) Metastases (mets): Track: Immunotherapy

Jonathan W. Goldman; Lucio Crinò; Everett E. Vokes; Esther Holgado; Karen L. Reckamp; Adam Pluzanski; David R. Spigel; Martin Kohlhaeufl; Marina C. Garassino; Laura Q. Chow; Scott N. Gettinger; David E. Gerber; Libor Havel; Suresh S. Ramalingam; Grace K. Dy; William J. Geese; Ang Li; Anne Blackwood-Chirchir; Diane Healey; Julie R. Brahmer; Gilberto Lopes

Marseille/FRANCE, H. Lee Moffitt Cancer Center & Research Institute, Tampa/FL/UNITED STATES OF AMERICA, Centre Léon Bérard, Lyon/FRANCE, Memorial Sloan Kettering Cancer Center, New York/NY/ UNITED STATES OF AMERICA, Ospedale di Perugia, Perugia/ITALY, LungenClinic Grosshansdorf, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Grosshansdorf/GERMANY, University Hospital Essen, West German Cancer Centre, Ruhrlandklinik, University Duisburg-Essen, Essen/ GERMANY, Bristol-Myers Squibb, Princeton/NJ/UNITED STATES OF AMERICA, Sarah Cannon Research Institute/ Tennessee Oncology, PLLC, Nashville/TN/UNITED STATES OF AMERICA, Núcleo de Oncologia da Bahia, Salvador/ BRAZIL


Clinical Lung Cancer | 2014

Oral vinorelbine plus cisplatin as first-line chemotherapy in nonsquamous non-small-cell lung cancer: final results of an International randomized phase II study (NAVotrial 01).

Jaafar Bennouna; Libor Havel; Maciej Krzakowski; Jens Kollmeier; Radj Gervais; Eric Dansin; Monika Serke; Adolfo Favaretto; Aleksandra Szczesna; Manuel Cobo; L. Ciuffreda; Jacek Jassem; Mario Nicolini; Rodryg Ramlau; Domenico Amoroso; Barbara Melotti; Teresa Almodovar; Marcello Riggi; Noël Raphaël Caux; Nathalie Vaissiere; Eng Huat Tan

BACKGROUND The combination of oral vinorelbine plus cisplatin has been studied in numerous trials as first-line treatment of patients with non-small cell lung cancer (NSCLC) regardless of histologic subtype. NAVoTrial 01 is the first study that explores this combination specifically in nonsquamous (NS) NSCLC by assessing the feasibility of this doublet (ratio 1:2) in an investigational approach. A reference arm with pemetrexed plus cisplatin was included. Maintenance therapy with single-agent therapy after 4 cycles of combination therapy was included in the study schedules because it reflected a trend in first-line treatment of NSCLC. PATIENTS AND METHODS Stage IIIB/IV untreated/relapsed patients with NS NSCLC received a 3-week cycle of pemetrexed 500 mg/m(2) and cisplatin 75 mg/m(2) on day 1 (arm A) or oral vinorelbine 80 mg/m(2) on days 1 and 8 (first cycle 60 mg/m(2)) and cisplatin 80 mg/m(2) on day 1 (arm B). After 4 cycles, patients without disease progression received single-agent maintenance treatment with pemetrexed or oral vinorelbine. RESULTS Overall, 153 patients were randomized (arm A/arm B: 51/102). Disease control rate (%) for arm A was 76.5 (95% confidence interval [CI], 62.5-87.2) and for arm B it was 75.0 (95% CI, 65.3-83.1), Response rates for arm A were 31.4% (95% CI, 19.1-45.9) and for arm B were 24.0% (95% CI, 16.0-33.6). Median progression-free survival for arm A was 4.3 months (95% CI, 3.8-5.6) and for arm B it was 4.2 months (95% CI, 3.6-4.7). Median survival for arm A was 10.8 months (95% CI, 7.0-16.4) and for arm B it was 10.2 months (95% CI, 7.8-11.9). Main grade 3/4 hematologic toxicities were neutropenia 18.3% (arm A) and 44.0% (arm B), whereas febrile neutropenia was reported in 2% of patients in each arm. CONCLUSION Oral vinorelbine and cisplatin had an efficacy in line with that achieved with a standard treatment such as pemetrexed and cisplatin, coupled with an acceptable safety profile.

Collaboration


Dive into the Libor Havel's collaboration.

Top Co-Authors

Avatar

Miloš Pešek

Charles University in Prague

View shared research outputs
Top Co-Authors

Avatar

František Salajka

Charles University in Prague

View shared research outputs
Top Co-Authors

Avatar

Leona Koubková

Charles University in Prague

View shared research outputs
Top Co-Authors

Avatar

Dimka Sixtová

Charles University in Prague

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karel Hejduk

Charles University in Prague

View shared research outputs
Top Co-Authors

Avatar

Petr Zatloukal

Charles University in Prague

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

M. Hrnčiarik

Charles University in Prague

View shared research outputs
Researchain Logo
Decentralizing Knowledge