Your browser doesn't support javascript.
loading
Development and validation of AI-assisted transcriptomic signatures to personalize adjuvant chemotherapy in patients with pancreatic ductal adenocarcinoma.
Fraunhoffer, N; Hammel, P; Conroy, T; Nicolle, R; Bachet, J-B; Harlé, A; Rebours, V; Turpin, A; Abdelghani, M B; Mitry, E; Biagi, J; Chanez, B; Bigonnet, M; Lopez, A; Evesque, L; Lecomte, T; Assenat, E; Bouché, O; Renouf, D; Lambert, A; Monard, L; Mauduit, M; Cros, J; Iovanna, J; Dusetti, N.
Afiliação
  • Fraunhoffer N; Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix- Marseille Université and Institut Paoli-Calmettes; Parc Scientifique et Technologique de Luminy, Marseille, France.
  • Hammel P; Digestive and Medical Oncology, Paul Brousse Hospital, Assistance Publique - Hôpitaux de Paris (AP- HP), Université of Paris-Saclay, Villejuif, France.
  • Conroy T; Medical Oncology department, Institut de cancérologie de Lorraine, Vandoeuvre-lès-Nancy, France and Université de Lorraine, APEMAC, équipe MICS, Nancy, France.
  • Nicolle R; Université Paris Cité, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018 Paris, France.
  • Bachet JB; Service d'Hépato - Gastro - Entérologie, Hôpital Pitié Salpêtrière, Assistance Publique - Hôpitaux de Paris (APHP), Sorbonne Université, Paris, France.
  • Harlé A; Service de Biopathologie, Institut de Cancérologie de Lorraine, Université de Lorraine, CNRS UMR 7039 CRAN, 54519 Vandœuvre-lès-Nancy CEDEX, France.
  • Rebours V; Pancreatology and Digestive Oncology Department, Beaujon Hospital - APHP, Clichy- INSERM - UMR 1149 - Université Paris-Cité, France.
  • Turpin A; Department of Oncology, Lille University Hospital; CNRS UMR9020, INSERM UMR1277, University of Lille, Institut Pasteur, Lille, France.
  • Abdelghani MB; Institut de Cancérologie Strasbourg Europe, Strasbourg, France.
  • Mitry E; Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix- Marseille Université and Institut Paoli-Calmettes; Parc Scientifique et Technologique de Luminy, Marseille, France; Department of Medical Oncology, Paoli-Calmettes Institute, Marseille, France.
  • Biagi J; Department of Oncology, Queen's University, Canada.
  • Chanez B; Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix- Marseille Université and Institut Paoli-Calmettes; Parc Scientifique et Technologique de Luminy, Marseille, France; Department of Medical Oncology, Paoli-Calmettes Institute, Marseille, France.
  • Bigonnet M; PredictingMed, Luminy Science and Technology Park, Marseille, France.
  • Lopez A; Hepatogastroenterology department, University Hospital, Nancy, France.
  • Evesque L; Department of Medical Oncology, Antoine Lacassagne Center, Nice.
  • Lecomte T; Hepatogastroenterology department, Hôpital Trousseau, Tours, France and INSERM UMR 1069, Tours University, Tours, France.
  • Assenat E; Medical oncology department, Centre Hospitalier Universitaire de Saint-Eloi, Montpellier, France.
  • Bouché O; Digestive oncology department, Centre Hospitalier Universitaire Robert Debré, Reims, France.
  • Renouf D; Division of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada; Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
  • Lambert A; Medical Oncology department, Institut de cancérologie de Lorraine, Vandoeuvre-lès-Nancy, France and Université de Lorraine, APEMAC, équipe MICS, Nancy, France.
  • Monard L; R&D Unicancer, Paris, France.
  • Mauduit M; R&D Unicancer, Paris, France.
  • Cros J; Université Paris Cité, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018 Paris, France; Université Paris Cité, Department of Pathology, FHU MOSAIC, Beaujon/Bichat University Hospital (APHP), Clichy/Paris, France.
  • Iovanna J; Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix- Marseille Université and Institut Paoli-Calmettes; Parc Scientifique et Technologique de Luminy, Marseille, France; Hospital de Alta Complejidad El Cruce, Florencio Varela, Buenos Aires, Argentina; University
  • Dusetti N; Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix- Marseille Université and Institut Paoli-Calmettes; Parc Scientifique et Technologique de Luminy, Marseille, France. Electronic address: nelson.dusetti@inserm.fr.
Ann Oncol ; 2024 Jun 19.
Article em En | MEDLINE | ID: mdl-38906254
ABSTRACT

BACKGROUND:

After surgical resection of pancreatic ductal adenocarcinoma (PDAC), patients are predominantly treated with adjuvant chemotherapy, commonly consisting of gemcitabine-based regimens or the modified FOLFIRINOX regimen (mFFX). While mFFX has been shown to be more effective than gemcitabine-based regimens, it is also associated with higher toxicity. Current treatment decisions are based on patient performance status rather than on the molecular characteristics of the tumor. To address this gap, the goal of this study was to develop drug-specific transcriptomic signatures for personalized chemotherapy treatment. PATIENTS AND

METHODS:

We used PDAC datasets from preclinical models, encompassing chemotherapy response profiles for the mFFX-regimen components. From them we identified specific gene transcripts associated with chemotherapy response. Three transcriptomic AI-signatures were obtained by combining Independent Component Analysis, Least Absolute Shrinkage and the Selection Operator-Random Forest approach. We integrated a previously developed gemcitabine signature with three newly developed ones. The machine learning strategy employed to enhance these signatures incorporates transcriptomic features from the tumor microenvironment, leading to the development of the Pancreas-View tool ultimately clinically validated in a cohort of 343 patients from the PRODIGE-24/CCTG PA6 trial.

RESULTS:

Patients who were predicted to be sensitive to the administered drugs (n=164; 47.8%) had longer disease-free survival (DFS) than the other patients. The median DFS in the mFFX sensitive group treated with mFFX was 50.0 months (stratified HR 0.31; 95% CI, 0.21-0.44; p<0.001) and 33.7 months (stratified HR 0.40; 95% CI, 0.17-0.59; p<0.001) in the gemcitabine sensitive group when treated with gemcitabine. Comparatively patients with signature predictions unmatched with the treatments (n=86; 25.1%) or those resistant to all drugs (n=93; 27.1%) had shorter DFS (10.6 and 10.8 months, respectively).

CONCLUSIONS:

This study presents a transcriptome-based tool that was developed using preclinical models and machine learning to accurately predict sensitivity to mFFX and gemcitabine.
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Ano de publicação: 2024 Tipo de documento: Article