Network


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

Hotspot


Dive into the research topics where Jean Louis Pujol is active.

Publication


Featured researches published by Jean Louis Pujol.


Lancet Oncology | 2012

Maintenance therapy with pemetrexed plus best supportive care versus placebo plus best supportive care after induction therapy with pemetrexed plus cisplatin for advanced non-squamous non-small-cell lung cancer (PARAMOUNT): a double-blind, phase 3, randomised controlled trial

Luis Paz-Ares; Filippo De Marinis; Mircea Dediu; Michael Thomas; Jean Louis Pujol; P. Bidoli; Olivier Molinier; Tarini Prasad Sahoo; Eckart Laack; Martin Reck; Jesus Corral; Symantha Melemed; William J. John; Nadia Chouaki; Annamaria Zimmermann; Carla Visseren-Grul; Cesare Gridelli

BACKGROUND Patients with advanced non-squamous non-small-cell lung cancer (NSCLC) benefit from pemetrexed maintenance therapy after induction therapy with a platinum-containing, non-pemetrexed doublet. The PARAMOUNT trial investigated whether continuation maintenance with pemetrexed improved progression-free survival after induction therapy with pemetrexed plus cisplatin. METHODS In this double-blind, multicentre, phase 3, randomised placebo-controlled trial, patients with advanced non-squamous NSCLC aged 18 years or older, with no previous systemic chemotherapy for lung cancer, with at least one measurable lesion, and an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1 participated. Before randomisation, patients entered an induction phase which consisted of four cycles of induction pemetrexed (500 mg/m(2)) plus cisplatin (75 mg/m(2)) on day 1 of a 21-day cycle. Patients who did not progress after completion of four cycles of induction and who had an ECOG performance status of 0 or 1 were stratified according to disease stage (IIIB or IV), ECOG performance status (0 or 1), and induction response (complete or partial response, or stable disease), and randomly assigned (2:1 ratio) to receive maintenance therapy with either pemetrexed (500 mg/m(2) every 21 days) plus best supportive care or placebo plus best supportive care until disease progression. Randomisation was done with the Pocock and Simon minimisation method. Patients and investigators were masked to treatment assignment. The primary endpoint was progression-free survival in the intention-to-treat population. This study is registered with ClinicalTrials.gov, NCT00789373. FINDINGS Of the 1022 patients enrolled, 939 participated in the induction phase. Of these, 539 patients were randomly assigned to receive continuation maintenance with pemetrexed plus best supportive care (n=359) or with placebo plus best supportive care (n=180). Among the 359 patients randomised to continuation maintenance with pemetrexed, there was a significant reduction in the risk of disease progression over the placebo group (HR 0·62, 95% CI 0·49-0·79; p<0·0001). The median progression-free survival, measured from randomisation, was 4·1 months (95% CI 3·2-4·6) for pemetrexed and 2·8 months (2·6-3·1) for placebo. Possibly treatment-related laboratory grade 3-4 adverse events were more common in the pemetrexed group (33 [9%] of 359 patients) than in the placebo group (one [<1%] of 180 patients; p<0·0001), as were non-laboratory grade 3-5 adverse events (32 [9%] of 359 patients in the pemetrexed group; eight [4%] of 180 patients in the placebo group; p=0·080); one possibly treatment-related death was reported in each group. The most common adverse events of grade 3-4 in the pemetrexed group were anaemia (16 [4%] of 359 patients), neutropenia (13 [4%]), and fatigue (15 [4%]). In the placebo group, these adverse events were less common: anaemia (one [<1%] of 180 patients), neutropenia (none), and fatigue (one <1%]). The most frequent serious adverse events were anaemia (eight [2%] of 359 patients in the pemetrexed group vs none in the placebo group) and febrile neutropenia (five [1%] vs none). Discontinuations due to drug-related adverse events occurred in 19 (5%) patients in the pemetrexed group and six (3%) patients in the placebo group. INTERPRETATION Continuation maintenance with pemetrexed is an effective and well tolerated treatment option for patients with advanced non-squamous NSCLC with good performance status who have not progressed after induction therapy with pemetrexed plus cisplatin. FUNDING Eli Lilly and Company.


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 Thoracic Oncology | 2007

Comparison of docetaxel- and vinca alkaloid-based chemotherapy in the first-line treatment of advanced non-small cell lung cancer: A meta-analysis of seven randomized clinical trials

Jean-Yves Douillard; Silvy Laporte; Frank V. Fossella; Vassilis Georgoulias; Jean Louis Pujol; Kaoru Kubota; Alain Monnier; Shinzoh Kudoh; Jaime Ernesto Rubio; Michel Cucherat

Introduction: To compare the impact on overall survival (OS) of docetaxel-based chemotherapy versus vinca alkaloid–based regimens for first-line therapy of advanced non-small cell lung cancer. Methods: A meta-analysis of all randomized, controlled trials comparing docetaxel- and vinca alkaloid–based chemotherapy was undertaken using MEDLINE, CANCERLIT, MEDSCAPE, Google Scholar, the Cochrane Library, the National Institutes of Health randomized, controlled trials register, and conference proceedings, supplemented by information from clinical study reports. All published and unpublished randomized, controlled trials (in any language) were included. Analysis was based on pooling individual logarithms of the hazard ratio for OS and the odds ratio (OR) for safety. Results: From eight potentially eligible trials, seven were selected (n = 2867). Docetaxel was administered with a platinum agent (three trials), with gemcitabine (two trials), or as monotherapy (two trials). Vinca alkaloid (vinorelbine [six trials] and vindesine [one trial]) was administered with cisplatin (six trials) or alone (one trial). The pooled estimate for OS showed an 11% improvement in favor of docetaxel (hazard ratio = 0.89; 95% confidence interval: 0.82–0.96; p = 0.004). Sensitivity analyses considering only vinorelbine as a comparator or only the doublet regimens showed similar improvements. Grade 3/4 neutropenia and grade 3/4 serious adverse events were less frequent with docetaxel- versus vinca alkaloid–based regimens (OR = 0.59; 95% confidence interval: 0.38–0.89; p = 0.013 and OR = 0.68; 95% confidence interval: 0.55–0.84; p < 0.001, respectively). Conclusion: According to this meta-analysis, docetaxel is superior to vinca alkaloid–based regimens in terms of OS and safety for first-line therapy of advanced non-small cell lung cancer.


Journal of Thoracic Oncology | 2007

Survival without Common Toxicity Criteria Grade 3/4 Toxicity for Pemetrexed Compared with Docetaxel in Previously Treated Patients with Advanced Non-small Cell Lung Cancer (NSCLC): A Risk-Benefit Analysis

Jean Louis Pujol; Sofia Paul; Nadia Chouaki; Patrick Peterson; Patti Moore; Donald A. Berry; Marc Salzberg

Background: In a recent large phase III study, previously treated patients with advanced non-small cell lung cancer who received pemetrexed demonstrated a survival time similar to patients who received docetaxel (median, 8.3 months with pemetrexed versus 7.9 months with docetaxel), with a more favorable toxicity profile, and significantly fewer Common Toxicity Criteria grade 3/4 toxicities. This is a retrospective risk-benefit analysis of survival without grade 3/4 toxicity, defined as the time to the first occurrence of Common Toxicity Criteria grade 3 or 4 toxicity or death, in the prospective phase III study comparing pemetrexed with docetaxel. Methods: A total of 541 patients (of 571 randomized) received either pemetrexed (500 mg/m2 intravenously [IV]) supplemented with vitamin B12 injections and oral folic acid or docetaxel (75 mg/m2 IV) on day 1 of 21-day cycles. Survival without grade 3/4 toxicity was analyzed using Kaplan-Meier and Cox methods. Results: Pemetrexed demonstrated a statistically significantly longer survival without grade 3/4 toxicity compared with docetaxel (hazard ratio = 0.60, 95% confidence interval: 0.50–0.72; p < 0.0001). A supportive analysis based on selected grade 3/4 toxicities (neutropenia lasting >5 days, febrile neutropenia, infection with neutropenia, anemia, thrombocytopenia, fatigue, nausea, vomiting, diarrhea, stomatitis, and neurosensory events) also demonstrated an advantage for pemetrexed (hazard ratio = 0.53; 95% confidence interval: 0.44–0.64; p < 0.0001). Conclusion: This analysis of survival without grade 3/4 toxicity suggests a benefit-to-risk profile that favors pemetrexed over docetaxel in the second-line treatment of patients with non-small cell lung cancer.


Journal of Thoracic Oncology | 2012

Safety, resource use, and quality of life in paramount: a phase III study of maintenance pemetrexed versus placebo after induction pemetrexed plus cisplatin for advanced nonsquamous non-small-cell lung cancer

Cesare Gridelli; Filippo De Marinis; Jean Louis Pujol; Martin Reck; Rodryg Ramlau; B. Parente; Gary Middleton; Jesus Corral; Katherine B. Winfree; Symantha Melemed; Anna Zimmermann; William J. John; Julie Beyrer; Nadia Chouaki; Carla Visseren-Grul; Luis Paz-Ares

Introduction: In a phase III, randomized, double-blind study (PARAMOUNT), maintenance pemetrexed demonstrated significant benefit in advanced non–small-cell lung cancer (NSCLC). We present safety, resource use, and quality of life (QoL) results. Methods: After four 21-day cycles of pemetrexed-cisplatin (N = 939), patients with advanced nonsquamous NSCLC, whose disease had not progressed and who had a performance status of 0/1, were randomized 2:1 (N = 539) to maintenance pemetrexed 500 mg/m2 plus best supportive care or placebo plus best supportive care every 21 days until disease progression or unacceptable toxicity. QoL was measured using the EuroQol 5-dimensional questionnaire (EQ-5D). Results: Frequently reported grade 3 to 4 drug-related toxicities with maintenance pemetrexed versus placebo were anemia (4.5% versus 0.6%; p = 0.016), fatigue (4.2% versus 0.6%; p = 0.016), and neutropenia (3.6% versus 0.0%; p < 0.006). No significant differences in drug-related grade 3 to 5 toxicities were observed with long-term pemetrexed exposure (>6 cycles), except grade 3 to 4 neutropenia, which did not result in increased infections. Patients on maintenance pemetrexed required more transfusions (13.4% versus 5.0%; p = 0.003), granulocyte colony- or granulocyte-macrophage colony-stimulating factors (5.3% versus 0.0%; p <0.001), anti-infectives (25.3% versus 16.7%; p = 0.028), and hospitalizations because of study drug (8.4% versus 3.3%, p = 0.028) than placebo-treated patients did. No significant treatment-by-time interactions, overall treatment differences, or clinically relevant changes from baseline were observed in EQ-5D scores during treatment. Conclusions: Long-term use of continuation maintenance pemetrexed was well tolerated; resource use was low, corresponding with known pemetrexed toxicities. The EQ-5D results demonstrate that patients tolerate long-term maintenance pemetrexed without worsening QoL.


BMJ Open | 2013

Prediction of survival benefits from progression-free survival benefits in advanced non-small-cell lung cancer: evidence from a meta-analysis of 2334 patients from 5 randomised trials

Silvy Laporte; Pierre Squifflet; Noémie Baroux; Frank V. Fossella; Vassilis Georgoulias; Jean Louis Pujol; Jean-Yves Douillard; Shinzohy Kudoh; Jean-Pierre Pignon; E. Quinaux; Marc Buyse

Objectives To investigate whether progression-free survival (PFS) can be considered a surrogate endpoint for overall survival (OS) in advanced non-small-cell lung cancer (NSCLC). Design Meta-analysis of individual patient data from randomised trials. Setting Five randomised controlled trials comparing docetaxel-based chemotherapy with vinorelbine-based chemotherapy for the first-line treatment of NSCLC. Participants 2331 patients with advanced NSCLC. Primary and secondary outcome measures Surrogacy of PFS for OS was assessed through the association between these endpoints and between the treatment effects on these endpoints. The surrogate threshold effect was the minimum treatment effect on PFS required to predict a non-zero treatment effect on OS. Results The median follow-up of patients still alive was 23.4 months. Median OS was 10 months and median PFS was 5.5 months. The treatment effects on PFS and OS were correlated, whether using centres (R²=0.62, 95% CI 0.52 to 0.72) or prognostic strata (R²=0.72, 95% CI 0.60 to 0.84) as units of analysis. The surrogate threshold effect was a PFS hazard ratio (HR) of 0.49 using centres or 0.53 using prognostic strata. Conclusions These analyses provide only modest support for considering PFS as an acceptable surrogate for OS in patients with advanced NSCLC. Only treatments that have a major impact on PFS (risk reduction of at least 50%) would be expected to also have a significant effect on OS. Whether these results also apply to targeted therapies is an open question that requires independent evaluation.


Journal of Thoracic Oncology | 2014

Final Efficacy and Safety Results of Pemetrexed Continuation Maintenance Therapy in the Elderly from the PARAMOUNT Phase III Study

Cesare Gridelli; Filippo De Marinis; Mike Thomas; Kumar Prabhash; Claude El Kouri; Fiona Blackhall; Frederique Bustin; Jean Louis Pujol; William J. John; Belen San Antonio; Annamaria Zimmermann; Nadia Chouaki; Carla Visseren-Grul; Luis Paz-Ares

Introduction: The PARAMOUNT Phase III trial showed that maintenance pemetrexed after pemetrexed plus cisplatin induction was well tolerated and effective for patients with advanced nonsquamous non–small-cell lung cancer. Approximately 17% of patients receiving maintenance therapy in this study were 70 years of age or older. Here we report efficacy and safety results from the PARAMOUNT study for elderly (≥70 years) and non-elderly (<70 years) patients. Methods: Final efficacy and safety data from the PARAMOUNT study were analyzed post hoc using subgroup analyses for elderly and non-elderly patients. Results: The median age was 73 years in the elderly subgroup (n = 92) and 60 years in the non-elderly subgroup (n = 447). Subgroups had similar baseline characteristics, except for a higher percentage of males and patients with a performance status of one in the elderly subgroup. For elderly patients, the median PFS was 6.4 months for pemetrexed and 3.0 months for placebo; the median OS was 13.7 months for pemetrexed and 12.1 months for placebo. For non-elderly patients, the median PFS was 4.0 months for pemetrexed and 2.8 months for placebo; the median OS was 13.9 months for pemetrexed and 10.8 months for placebo. Elderly patients experienced similar levels of low-grade toxicities, but had a higher percentage of grade 3/4 anemia and neutropenia than non-elderly patients, although importantly, this did not translate into increased febrile neutropenia. Conclusions: Continuation maintenance pemetrexed had comparable survival and toxicity profiles in the elderly and non-elderly subgroups. However, grade 3/4 anemia and neutropenia were numerically higher for elderly patients.


Journal of Thoracic Oncology | 2015

Safety and Immunogenicity of MAGE-A3 Cancer Immunotherapeutic with or without Adjuvant Chemotherapy in Patients with Resected Stage IB to III MAGE-A3-Positive Non-Small-Cell Lung Cancer

Jean Louis Pujol; Johan Vansteenkiste; Tommaso De Pas; Djordje Atanackovic; Martin Reck; Michiel Thomeer; Jean-Yves Douillard; Gianpiero Fasola; Vanessa Potter; Paul Taylor; Lionel Bosquée; Robert Scheubel; Silvija Jarnjak; Muriel Debois; Pedro de Sousa Alves; Jamila Louahed; Vincent Brichard; Frederic Lehmann

Introduction: To assess the safety and immunogenicity of MAGE-A3 immunotherapeutic in patients with stage IB–III MAGE-A3-positive non–small-cell lung cancer (NSCLC) who were or were not undergoing standard cisplatin/vinorelbine chemotherapy. Methods: This open, prospective, multicenter, parallel-group phase I study (NCT00455572) enrolled patients with resected (cohorts 1–3) or unresectable (cohort 4) MAGE-A3-positive NSCLC. MAGE-A3 immunotherapeutic (300 &mgr;g recombinant MAGE-A3 formulated with AS15) was administered (eight doses, 3 weeks apart) concurrent with (cohort 1), after (cohort 2), or without (cohort 3) standard-adjuvant chemotherapy, or after standard radiotherapy and/or chemotherapy (cohort 4). Results: Sixty-seven patients received greater than or equal to 1 dose of MAGE-A3 immunotherapeutic. Grade 3/4 adverse events (AEs) were reported for 16 out of 19 (84%), 2 out of 18 (11%), 5 out of 18 (28%), and 1 out of 12 (8%) patients in cohorts 1, 2, 3, and 4, respectively. Many grade 3/4 AEs in cohort 1 (e.g., neutropenia) were typical of chemotherapy. Six patients, including three in cohort 1, reported study treatment–related grade 3/4 AEs (injection-site reactions or musculoskeletal/back pain, which resolved within 5 days). One patient (in cohort 4) died, but this and the other serious adverse events were not study treatment related. MAGE-A3-specific antibody responses to immunotherapy were induced in all patients evaluated in all cohorts. MAGE-A3-specific CD4+ T-cell responses to immunotherapy were detected in 4 out of 11 (36%), 4 out of 15 (27%), 2 out of 8 (25%), and 5 out of 6 (83%) evaluated patients in cohorts 1, 2, 3, and 4, respectively; and CD8+ T-cell responses were only detected in four patients. Conclusion: In resected and unresectable NSCLC patients and irrespective of whether standard chemotherapy was concurrent or not, MAGE-A3 immunotherapeutic is well tolerated and induces MAGE-A3-specific immune responses.


Journal of Thoracic Oncology | 2008

Circulating Serum Vascular Endothelial Growth Factor is Not a Prognostic Factor of Non-small Cell Lung Cancer

Mohammad Chakra; Jean Louis Pujol; Pierre-Jean Lamy; Marie Cécile Bozonnat; Xavier Quantin; William Jacot; Jean-Pierre Daurès

Introduction: High circulating serum vascular endothelial growth factor (VEGF) levels might reflect enhanced angiogenesis in patients suffering from non-small cell lung cancer (NSCLC). This study aimed at determining the prognostic significance of circulating VEGF as a prognostic factor in NSCLC. Methods: Four hundred fifty-one histologically or cytologically proven and previously untreated NSCLC patients have been studied. Median follow-up was 13 years and 9 months. Eleven clinical and biologic variables were recorded. The levels of circulating VEGF were measured in the serum by quantitative immunoassay. Patients have had received conventional treatment (without anti-VEGF therapy) according to the international guidelines. All statistical tests were two-sided. Results: Receiver operating characteristic curves (area under the ROC curve: 0.66 ± 0.05) showed that circulating VEGF serum level did not demonstrate a high sensitivity-specificity relationship, and therefore, demonstrated a low ability to differentiate NSCLC from benign lung diseases. A 600 pg/mL level of circulating VEGF serum was considered as threshold with 40.8% of NSCLC patients presenting with a high level. The circulating VEGF distribution differed significantly according to disease stage, nodal status, and performance status (PS), with the highest levels observed in metastatic stage, positive mediastinal nodal status, and poor PS. In univariate survival analysis, patients with a high pretreatment circulating VEGF serum level proved to have a shorter overall survival when compared with patients presenting with a circulating VEGF serum level ≤600 pg/mL. However, in the Cox proportional hazard model, this variable was not included in the panel of independent determinants of a poor outcome that was as follows: advanced or metastatic diseases according to the 6th edition of the staging system, PS ≥2, nodal status N2-3, metastatic disease, neuron-specific enolase >12.5 ng/mL, CYFRA 21-1 >3.6 ng/mL. Conclusion: The prognostic information given by a high circulating VEGF serum level is not an independent determinant of survival owing to a high relationship with main prognostic variables such as PS, stage of the disease, and nodal status. This finding does not preclude a putative prognostic impact of in situ detection of VEGF and VEGF receptors in tumor specimen.


Journal of Thoracic Oncology | 2009

Cardiorespiratory Fitness in Patients with Advanced Non-small Cell Lung Cancer: Why is this Feature Important to Evaluate? Can it be Improved?

Jean Louis Pujol; Xavier Quantin; Mohammad Chakra

Patients suffering from non-small cell lung cancer (NSCLC) are frequently affected by a high level of symptom burden.1 Consequently, these patients also have a poor quality of life, insofar as symptom score is one of the five dimensions of quality of life (aside, functional, psychologic, social, and spiritual dimensions2). A high symptom score, by itself might require specific interventions, palliation or supportive therapies to achieve symptoms alleviation. Consequences of a high symptom score could be regarded from two major points of view. (i) some symptoms can indicate a severe patient disability and therefore might modify anticancer treatment program by limiting indication for some important treatment modalities such as surgery. (ii) high symptom score is reducing patient ability to achieve a normal familial and social activity. This interference between symptoms score and social life, referred to as global health status in quality of life scale, has been demonstrated as indicating a poor outcome.3 Among the most frequently observed symptoms in NSCLC, two could be regarded as directly involved in reducing patient daily activity living: dyspnoea and fatigue. Both symptoms have major impact in cardiopulmonary fitness on exercise. Studies that aim at measuring cardiopulmonary fitness in NSCLC might therefore be regarded as a global investigation exploring main host-tumor relationship features that could have major impact in both treatment decision and quality of life.4 Conversely, studies that aim at improving cardiopulmonary fitness on exercise might be considered as interesting approaches at improving quality of life. In this issue of the journal, Kasymjanova et al.5 presented a noninterventional study evaluating exercise capacity in patients with newly diagnosed advanced NSCLC and its relationship with survival. Temel et al.,6 also in this issue report an interventional study performed in an effort at improving functional outcome and symptoms in a similar advanced NSCLC population by mean of a structured exercise program. In this brief editorial we would like to summarize and comment on results of both studies by highlighting three different issues: (1) methodological aspects of the evaluation of cardiopulmonary fitness in oncologic patients; (2) multidimensional aspect of fatigue and dyspnoea in patients receiving treatment for advanced NSCLC; and (3) meaning of the evaluation and intervention on cardiopulmonary fitness for these patients in daily practice. Briefly, in the study by Kasymjanova et al., patients receiving chemotherapy for a NSCLC have underwent repeated evaluation by exercise tests (by means of the submaximum 6-minute walk test [6MW]): One before chemotherapy (preceded by a training test) and the second after the second cycle of chemotherapy. The main findings are as follows: a poor performance during the prestudy 6MW test (6MW 400 m) at presentation by itself, is an independent unfavorable prognostic factor; patients who did not achieve the complete program (but were only able to perform the prestudy test) were at higher risk of death. Among patients who completed both preand posttreatment 6MW tests, patients for whom performance on exercise had decreased, were at higher risk of disease progression

Collaboration


Dive into the Jean Louis Pujol's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Luis Paz-Ares

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Filippo De Marinis

European Institute of Oncology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xavier Quantin

University of Montpellier

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Frank V. Fossella

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge