662 results on '"Earle A. Chiles Research Institute"'
Search Results
2. Personalized TCR-T: Study of Adoptively Transferred T-cell Receptor Gene-engineered T Cells (TCR-T)
- Author
-
Celldex Therapeutics and Providence Cancer Center, Earle A. Chiles Research Institute
- Published
- 2024
3. Encorafenib and Binimetinib With or Without Nivolumab in Treating Patients With Metastatic Radioiodine Refractory BRAF V600 Mutant Thyroid Cancer
- Author
-
Providence Cancer Center, Earle A. Chiles Research Institute
- Published
- 2024
4. Study of MEDI0562 Prior to Surgical Resection in Head and Neck Squamous Cell Carcinoma (HNSCC) or Melanoma
- Author
-
Providence Cancer Center, Earle A. Chiles Research Institute and MedImmune LLC
- Published
- 2024
5. The High-Dose Aldesleukin (IL-2) 'Select' Trial for Patients With Metastatic Renal Cell Carcinoma (SELECT)
- Author
-
City of Hope National Medical Center, Providence Cancer Center, Earle A. Chiles Research Institute, Dartmouth-Hitchcock Medical Center, Indiana University, Loyola University, Our Lady of Mercy Medical Center, Roswell Park Cancer Institute, University of California, Los Angeles, University of Cincinnati, University of Pittsburgh, University of Virginia, Vanderbilt University, Wayne State University, Dana-Farber Cancer Institute, and David McDermott, Principal Investigator
- Published
- 2024
6. Neoadjuvant Immunoradiotherapy in Head & Neck Cancer
- Author
-
Providence Cancer Center, Earle A. Chiles Research Institute
- Published
- 2024
7. GB1211 and Pembrolizumab Versus Pembrolizumab and Placebo in Patients With Metastatic Melanoma and Head and Neck Squamous Cell Carcinoma
- Author
-
Providence Cancer Center, Providence Cancer Center, Earle A. Chiles Research Institute, and Galecto Biotech AB
- Published
- 2024
8. Neoadjuvant Immunoradiotherapy in Head & Neck Cancer (NIRT 2-HNC)
- Author
-
Providence Cancer Center, Earle A. Chiles Research Institute
- Published
- 2024
9. MK-3475 and Gemcitabine in Non-Small Cell Lung Cancer (NSCLC)
- Author
-
Merck Sharp & Dohme LLC and Providence Cancer Center, Earle A. Chiles Research Institute
- Published
- 2023
10. GR-MD-02 + Pembrolizumab Versus Pembrolizumab Monotherapy in Melanoma and Squamous Cell Head and Neck Cancer Patients
- Author
-
Providence Cancer Center, Earle A. Chiles Research Institute
- Published
- 2023
11. Hydroxychloroquine in Patients With Newly Diagnosed COVID-19 Compared to Standard of Care
- Author
-
Center for Outcomes Research and Education and Providence Cancer Center, Earle A. Chiles Research Institute
- Published
- 2020
12. Stereotactic Body Radiation and Monoclonal Antibody to OX40 (MEDI6469) in Breast Cancer Patients With Metastatic Lesions (OX40 Breast)
- Author
-
Robert W. Franz Cancer Center, Providence Cancer Center, Earle A. Chiles Research Institute, and MedImmune LLC
- Published
- 2019
13. Galectin Inhibitor (GR-MD-02) and Ipilimumab in Patients With Metastatic Melanoma
- Author
-
Providence Cancer Center, Earle A. Chiles Research Institute and Galectin Therapeutics Inc.
- Published
- 2019
14. Pilot Study of DRibble Vaccine for Prostate Cancer Patients (DRibble)
- Author
-
Providence Health & Services, Providence Cancer Center, and Providence Cancer Center, Earle A. Chiles Research Institute
- Published
- 2018
15. Immunochemoradiotherapy in Patients With Pancreatic Cancer
- Author
-
Providence Cancer Center, Earle A. Chiles Research Institute and Robert W. Franz Cancer Research Center
- Published
- 2018
16. Combination Vaccine Immunotherapy (DRibbles) for Patients With Definitively-Treated Stage III Non-small Cell Lung Cancer
- Author
-
National Cancer Institute (NCI), Providence Cancer Center, Earle A. Chiles Research Institute, Providence Health & Services, Mayo Clinic, and Louisiana State University Health Sciences Center in New Orleans
- Published
- 2017
17. Study of Everolimus, Pemetrexed, Carboplatin, and Bevacizumab to Treat Stage IV Lung Cancer
- Author
-
University of Arizona, Providence Cancer Center, Earle A. Chiles Research Institute, and Novartis Pharmaceuticals
- Published
- 2014
18. Trial of Neoadjuvant Docetaxel and Cisplatin for Resectable Non-Small Cell Lung Cancer
- Author
-
Providence Cancer Center, Earle A. Chiles Research Institute and Sanofi
- Published
- 2011
19. GVAX in Advanced Prostate Cancer Patients Made Lymphopenic
- Author
-
Providence Cancer Center, Earle A. Chiles Research Institute and Cell Genesys
- Published
- 2009
20. Emerging concepts in biomarker discovery; the US-Japan Workshop on Immunological Molecular Markers in Oncology
- Author
-
A. Karolina Palucka, Anatoli Malyguine, Yutaka Kawakami, Yingdong Zhao, Mai-Britt Zocca, Lisa H. Butterfield, Michele Maio, Hiroya Takeuchi, David F. Stroncek, Damien Chaussabel, John M. Kirkwood, Howard Streicher, Wolf Hervé Fridman, Noriyuki Sato, Oleg Eremin, Lance A. Liotta, Hisashi Wada, Peter P. Lee, Thomas O. Kleen, Hisahiro Matsubara, Tomonori Yaguchi, Mohammed Kashani-Sabet, Emanuel F. Petricoin, Ena Wang, Kohzoh Imai, James W. Jacobson, Paul V Lehmann, Masahisa Jinushi, Antoni Ribas, Marimo Sato, Hiroshi Shiku, Kazunori Kato, Mary L. Disis, Magdalena Thurin, Hideaki Tahara, Julia Wulfkuhle, Yoshihiko Hirohashi, Stefan Ambs, Bernard A. Fox, Xifeng Wu, Shawmarie Mayrand-Chung, Giuseppe Masucci, Minoru Toyota, Hiroyoshi Nishikawa, Yuichiro Doki, Yasunori Akutsu, Akira Kanamoto, Benjamin Zeskind, Samir N. Khleif, Francesco M. Marincola, Michael T. Lotze, Zoltan Pos, Licia Rivoltini, Jon M. Wigginton, Craig L. Slingluff, Kiminori Nakamura, BMC, Ed., Department of Surgery and Bioengineering, The University of Tokyo (UTokyo)-Institute of Medical Science-Advanced Clinical Research Center, Cancer Diagnosis Program, National Institutes of Health (NIH)-National Cancer Institute (NCI), Infectious Disease and Immunogenetics Section (IDIS), National Institutes of Health (NIH)-Department of Transfusion Medicine-Clinical Center and Center for Human Immunology (CHI), Departments of Medicine, Surgery and Immunology, Division of Hematology Oncology-University of Pittsburgh Cancer Institute, Tumor Vaccine Group, University of Washington [Seattle]-Center for Translational Medicine in Women's Health, Department of Molecular Microbiology and Immunology, Oregon Health and Science University [Portland] (OHSU)-Robert W Franz Research Center-Earle A Chiles Research Institute-Providence Portland Medical Center, Stanford School of Medicine [Stanford], Stanford Medicine, Stanford University-Stanford University, Cancer Vaccine Section, National Institutes of Health [Bethesda] (NIH)-National Cancer Institute (NCI), Discovery Medicine-Oncology, Bristol-Myers Squibb Inc., Laboratory of Human Carcinogenesis, National Institutes of Health (NIH)-National Cancer Institute (NCI)-Center of Cancer Research, Department of Frontier Surgery, Chiba University-Graduate School of Medicine, Baylor Institute for Immunology Research (BIIR), Department of Surgery, Osaka University [Osaka]-Graduate School of Medicine, Section of Surgery, Biomedical Research Unit-Nottingham Digestive Disease Centre-University of Nottingham, UK (UON), Centre de Recherche des Cordeliers (CRC (UMR_S 872)), Université Pierre et Marie Curie - Paris 6 (UPMC)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Sapporo Medical University, Melanoma Clinic, University of California (UC), Department of Molecular Medicine, School of Medicine-Sapporo Medical University, Division of Cellular Signaling, Institute for Advanced Medical Research-Keio University School of Medicine [Tokyo, Japan], Cellular Technology Ltd, Shaker Heights, Department of Molecular Pathology and Microbiology, Center for Applied Proteomics and Molecular Medicine-George Mason University [Fairfax], Illman Cancer Center, University of Pittsburgh (PITT), Pennsylvania Commonwealth System of Higher Education (PCSHE)-Pennsylvania Commonwealth System of Higher Education (PCSHE), Medical Oncology and Immunotherapy, Istituto Toscano Tumori-Department. of Oncology-Hospital of Siena, Cancer Bioimmunotherapy Unit, Department of Medical Oncology-Centro di Riferimento Oncologico, IRCCS, Laboratory of Cell Mediated Immunity, SAIC-Frederick-Inc. NCI-Frederick, Department of Oncology-Pathology, karolinska institute, The Biomarkers Consortium (BC), Public-Private Partnership Program-Office of the Director-National Institute of Health (NIH), Department of Cancer Vaccine & Department of Immuno-gene Therapy, Mie University, Department of Medicine, Jonsson Comprehensive Cancer Center, Unit of Immunotherapy of Human Tumors, Istituto Nazionale Tumori-IRCCS Foundation, Department of Pathology, Sapporo Medical University School of Medicine, Sapporo Medical University-Sapporo Medical University, Division of Surgical Oncology-University of Virginia School of Medicine, Cancer Therapy Evaluation Program, National Institutes of Health (NIH)-National Cancer Institute (NCI)-DCTD, Cell Therapy Section (CTS), Department of Transfusion Medicine-Clinical Center-National Institutes of Health, Keio University School of Medicine [Tokyo, Japan], Department of Biochemistry, Department of Epidemiology, The University of Texas Health Science Center at Houston (UTHealth)-The University of Texas M.D. Anderson Cancer Center [Houston], Immuneering Corporation, Biometric Research Branch, DanDritt Biotech A/S, University of California, Keio University School of Medicine [Tokyo, Japan]-Institute for Advanced Medical Research, The University of Tokyo-Institute of Medical Science-Advanced Clinical Research Center, Infectious Disease and Immunogenetics Section ( IDIS ), Oregon Health and Science University-Robert W Franz Research Center-Earle A Chiles Research Institute-Providence Portland Medical Center, Stanford University School of Medicine [Stanford], Stanford University [Stanford], National Cancer Institute (NCI)-, National Institutes of Health (NIH), Baylor Institute for Immunology Research ( BIIR ), Biomedical Research Unit-Nottingham Digestive Disease Centre-University of Nottingham, UK ( UON ), Centre de Recherche des Cordeliers ( CRC (UMR_S 872) ), Université Pierre et Marie Curie - Paris 6 ( UPMC ) -Université Paris Descartes - Paris 5 ( UPD5 ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Centre National de la Recherche Scientifique ( CNRS ), School of Medicine, Keio University School of Medicine-Institute for Advanced Medical Research, University of Pittsburgh, The Biomarkers Consortium ( BC ), Mie University Graduate School of Medicine, Cell Therapy Section ( CTS ), Keio University School of Medicine, and University of Texas at Houston [Houston] ( UTHealth ) -MD Anderson Cancer Center
- Subjects
Oncology ,Standardization ,medicine.medical_treatment ,lcsh:Medicine ,MESH : Japan ,Disease ,0302 clinical medicine ,Cancer immunotherapy ,MESH : Tumor Markers, Biological ,[ SDV.IMM ] Life Sciences [q-bio]/Immunology ,MESH: Neoplasms ,Biomarker discovery ,MESH: Japan ,Medicine(all) ,0303 health sciences ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,General Medicine ,3. Good health ,MESH: Reproducibility of Results ,030220 oncology & carcinogenesis ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,MESH : Biomedical Research ,medicine.medical_specialty ,MESH : National Cancer Institute (U.S.) ,MESH : United States ,[SDV.IMM] Life Sciences [q-bio]/Immunology ,MESH : United States Food and Drug Administration ,MESH: National Cancer Institute (U.S.) ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,[ SDV.MHEP ] Life Sciences [q-bio]/Human health and pathology ,Internal medicine ,MESH: United States Food and Drug Administration ,medicine ,MESH: United States ,030304 developmental biology ,MESH: Humans ,Biochemistry, Genetics and Molecular Biology(all) ,business.industry ,Task force ,MESH : Reproducibility of Results ,MESH: Biomedical Research ,lcsh:R ,MESH : Humans ,Cancer ,Immunotherapy ,medicine.disease ,MESH : Neoplasms ,Clinical trial ,MESH: Tumor Markers, Biological ,Commentary ,business ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
Supported by the Office of International Affairs, National Cancer Institute (NCI), the "US-Japan Workshop on Immunological Biomarkers in Oncology" was held in March 2009. The workshop was related to a task force launched by the International Society for the Biological Therapy of Cancer (iSBTc) and the United States Food and Drug Administration (FDA) to identify strategies for biomarker discovery and validation in the field of biotherapy. The effort will culminate on October 28th 2009 in the "iSBTc-FDA-NCI Workshop on Prognostic and Predictive Immunologic Biomarkers in Cancer", which will be held in Washington DC in association with the Annual Meeting. The purposes of the US-Japan workshop were a) to discuss novel approaches to enhance the discovery of predictive and/or prognostic markers in cancer immunotherapy; b) to define the state of the science in biomarker discovery and validation. The participation of Japanese and US scientists provided the opportunity to identify shared or discordant themes across the distinct immune genetic background and the diverse prevalence of disease between the two Nations. Converging concepts were identified: enhanced knowledge of interferon-related pathways was found to be central to the understanding of immune-mediated tissue-specific destruction (TSD) of which tumor rejection is a representative facet. Although the expression of interferon-stimulated genes (ISGs) likely mediates the inflammatory process leading to tumor rejection, it is insufficient by itself and the associated mechanisms need to be identified. It is likely that adaptive immune responses play a broader role in tumor rejection than those strictly related to their antigen-specificity; likely, their primary role is to trigger an acute and tissue-specific inflammatory response at the tumor site that leads to rejection upon recruitment of additional innate and adaptive immune mechanisms. Other candidate systemic and/or tissue-specific biomarkers were recognized that might be added to the list of known entities applicable in immunotherapy trials. The need for a systematic approach to biomarker discovery that takes advantage of powerful high-throughput technologies was recognized; it was clear from the current state of the science that immunotherapy is still in a discovery phase and only a few of the current biomarkers warrant extensive validation. It was, finally, clear that, while current technologies have almost limitless potential, inadequate study design, limited standardization and cross-validation among laboratories and suboptimal comparability of data remain major road blocks. The institution of an interactive consortium for high throughput molecular monitoring of clinical trials with voluntary participation might provide cost-effective solutions.
- Published
- 2009
- Full Text
- View/download PDF
21. Editorial: Cancer Immunotherapies: From Efficacy to Resistance Mechanisms
- Author
-
Janin Chandra, Morten Hansen, Nathalie Labarriere, Ilaria Marigo, Fernando Souza-Fonseca-Guimaraes, Lazar Vujanovic, Yoshinobu Koguchi, Nicolas Jacquelot, University of Queensland - The Diamantina Institute, University of Queensland [Brisbane], Copenhagen University Hospital, Immunology and New Concepts in ImmunoTherapy (INCIT), Université d'Angers (UA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Nantes Université - UFR de Médecine et des Techniques Médicales (Nantes Univ - UFR MEDECINE), Nantes Université - pôle Santé, Nantes Université (Nantes Univ)-Nantes Université (Nantes Univ)-Nantes Université - pôle Santé, Nantes Université (Nantes Univ)-Nantes Université (Nantes Univ), LabEX IGO Immunothérapie Grand Ouest, Nantes Université (Nantes Univ), Veneto Institute of Oncology IOV-IRCCS [Padua, Italy], Università degli Studi di Padova = University of Padua (Unipd), University of Pittsburgh (PITT), Pennsylvania Commonwealth System of Higher Education (PCSHE), Earle A. Chiles Research Institute [Portland, OR, USA], Providence Portland Medical Center-Robert W. Franz Cancer Research Center, Princess Margaret Cancer Centre [Toronto, Canada], and Pecqueret, Valérie
- Subjects
Biochemical Phenomena ,[SDV]Life Sciences [q-bio] ,Immunology ,adoptive T cell therapy ,cancer ,immune checkpoint blockers ,immunotherapy ,tumor microenvironment ,[SDV] Life Sciences [q-bio] ,Neoplasms ,Immunology and Allergy ,Humans ,ComputingMilieux_MISCELLANEOUS - Abstract
International audience; No abstract available
- Published
- 2022
- Full Text
- View/download PDF
22. Predictors of responses to immune checkpoint blockade in advanced melanoma
- Author
-
Brigitte Dréno, Caroline Flament, Nicolas Jacquelot, B. Balme, Yaron Meirow, Mitchell P. Levesque, Jeffrey S. Weber, Christophe Borg, Lawrence Fong, Amir Khammari, Guido Kroemer, Sylvie Rusakiewicz, Morten Hartvig Hansen, Ignacio Melero, Gorana Tomasic, Henrik Schmidt, Meriem Messaoudene, Andrea Cavalcanti, Maria Paula Roberti, Sarah Jegou, Yoshinobu Koguchi, Stefan Michiels, Moshe Sade-Feldman, Michal Baniyash, Ana C. Anderson, Byoung S. Kwon, Lieping Chen, Aurélien Marabelle, David Enot, Arun Burra, Inge Marie Svane, Stéphane Dalle, A.M. Di Giacomo, Dirk Schadendorf, Nils Ternès, Laurence Zitvogel, Reinhard Dummer, Andressa L. Sodre, Serena S. Kwek, O Beatrix, Vijay K. Kuchroo, Alexander M.M. Eggermont, David M. Woods, Benjamin Weide, Mark J. Smyth, Michele Maio, Maha Ayyoub, François Aubin, Connie P.M. Duong, Michal Lotem, Immunologie des tumeurs et immunothérapie (UMR 1015), Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut Gustave Roussy (IGR)-Université Paris-Sud - Paris 11 (UP11), Université Paris-Saclay, Institut Gustave Roussy (IGR), Plateforme de métabolomique, Direction de la recherche [Gustave Roussy], Institut Gustave Roussy (IGR)-Institut Gustave Roussy (IGR), Centre d'Investigation Clinique en Biotherapie des cancers (CIC 1428 , CBT 507 ), Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM), Service de biostatistique et d'épidémiologie (SBE), Direction de la recherche clinique [Gustave Roussy], Microorganismes et physiopathologie intestinale (ERL INSERM U1157 - CNRS UMR 7203), Laboratoire des biomolécules (LBM UMR 7203), Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Département de Chimie - ENS Paris, École normale supérieure - Paris (ENS Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-École normale supérieure - Paris (ENS Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Département de Chimie - ENS Paris, Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Pierre et Marie Curie - Paris 6 (UPMC), Laura & Isaac Perlmutter Cancer Center [New York, NY, USA], New York University Langone Medical Center (NYU Langone Medical Center), NYU System (NYU)-NYU System (NYU), Department of Hematology and Oncology [Herlev, Denmark], Copenhagen University Hospital, The Lautenberg Center for General and Tumor Immunology [Jerusalem, Israel], The Hebrew University Hadassah Medical School -BioMedical Research institute Israel Canada of the Faculty of Medicine [Jerusalem, Israel], Division of Hematology-Oncology [San Francisco, CA, USA] (Department of Medicine), University of California [San Francisco] (UCSF), University of California-University of California, Department of Immunobiology [New Haven, CT, USA] (Yale School of Medicine), Yale University [New Haven], Eutilex Co., Ltd [Seoul, Korea], Section of Clinical Immunology, Allergy, and Rheumatology [New Orleans, LA, USA] (Department of Medicine), Tulane University Health Sciences Center [New Orleans, LA, USA], Evergrande Center for Immunologic Diseases [Boston, MA, USA] (Ann Romney Center for Neurologic Diseases), Brigham & Women's Hospital, Harvard Medical School, Department of Dermatology [Tubingen, Germany], University Medical Center [Tubingen, Germany], Carcinogénèse épithéliale : facteurs prédictifs et pronostiques - UFC (EA 3181) (CEF2P / CARCINO), Centre Hospitalier Régional Universitaire de Besançon (CHRU Besançon)-Université de Franche-Comté (UFC), Université Bourgogne Franche-Comté [COMUE] (UBFC)-Université Bourgogne Franche-Comté [COMUE] (UBFC), Service d'Oncologie Médicale [CHRU Besançon], Centre d'Investigation Clinique de Besançon (Inserm CIC 1431), Université de Franche-Comté (UFC), Université Bourgogne Franche-Comté [COMUE] (UBFC)-Université Bourgogne Franche-Comté [COMUE] (UBFC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier Régional Universitaire de Besançon (CHRU Besançon)-Etablissement français du sang [Bourgogne-Franche-Comté] (EFS [Bourgogne-Franche-Comté]), Interactions hôte-greffon-tumeur, ingénierie cellulaire et génique - UFC (UMR INSERM 1098) (RIGHT), Institut National de la Santé et de la Recherche Médicale (INSERM)-Etablissement français du sang [Bourgogne-Franche-Comté] (EFS [Bourgogne-Franche-Comté])-Université de Franche-Comté (UFC), Centre Hospitalier Lyon Sud [CHU - HCL] (CHLS), Hospices Civils de Lyon (HCL), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon, Centre de Recherche en Cancérologie de Lyon (UNICANCER/CRCL), Centre Léon Bérard [Lyon]-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Département de Pathologie [CHU Lyon-Sud - HCL], Hospices Civils de Lyon (HCL)-Hospices Civils de Lyon (HCL), Département de biologie et pathologie médicales [Gustave Roussy], Division of Medical Oncology and Immunotherapy [Siena, Italy], University Hospital of Siena, Department of Dermatology [Heidelberg, Germany], University Duisburg-Essen [Germany]-University Hospital [Heidelberg, Germany]-Germany & German Cancer Consortium - DKTZ [Heidelberg, Germany], Division of Hepatology and Gene Therapy (CIMA), Center for Applied Medical Research [Plamplona] (CIMA), Universidad de Navarra [Pamplona] (UNAV)-Universidad de Navarra [Pamplona] (UNAV), Oncology Department [Pamplona, Spain], University Clinic of Navarra - CUN [Pamplona, Spain], Centro de Investigación Biomedica en Red de Oncologia [Pamplona, Spain] (CIBERO), Clinical and Translational Research in Skin Cancer (CRCINA-ÉQUIPE 2), Centre de Recherche en Cancérologie et Immunologie Nantes-Angers (CRCINA), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Centre National de la Recherche Scientifique (CNRS)-Université d'Angers (UA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Centre National de la Recherche Scientifique (CNRS)-Université d'Angers (UA), Department of Dermatology [Zürich, Switzerland], Universität Zürich [Zürich] = University of Zurich (UZH)-University hospital of Zurich [Zurich], Earle A. Chiles Research Institute [Portland, OR, USA], Providence Portland Medical Center-Robert W. Franz Cancer Research Center, Sharett Institute of Oncology [Jerusalem, Israel], Hadassah Medical Organization [Jerusalem, Israel], Department of Oncology [Aarhus, Denmark], Aarhus University Hospital, Centre de Recherche des Cordeliers (CRC), Université Paris Diderot - Paris 7 (UPD7)-École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Apoptose, cancer et immunité (Equipe labellisée Ligue contre le cancer - CRC - Inserm U1138), Institut Gustave Roussy (IGR)-Centre de Recherche des Cordeliers (CRC), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Diderot - Paris 7 (UPD7)-École pratique des hautes études (EPHE), Université Sorbonne Paris Cité (USPC), Pôle de biologie, Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO), Département de chirurgie générale [Gustave Roussy], Service de dermatologie, Département de médecine oncologique [Gustave Roussy], Immunology in Cancer and Infection Laboratory [Herston, QLD, Australia], QIMR Berghofer Medical Research Institute, School of Medicine [Herston, QLD, Australia], University of Southern Queensland (USQ), This work was supported by Institut National du Cancer INCa, ANR, Ligue contre le cancer (équipe labellisée de L.Z.) and Swiss Bridge Foundation, ISREC Foundation, LABEX OncoImmunology, la direction générale de l’offre de soins (DGOS), Université Paris-Sud, SIRIC SOCRATE (INCa/DGOS/INSERM 6043), PACRI network and PIA2 TORINO-LUMIERE. N.J. received a fellowship from Cancéropole Idf. M.J.S. was supported by a National Health and Medical Research Council of Australia Senior Principal Research Fellowship., Bernardo, Elizabeth, Université Paris-Sud - Paris 11 (UP11)-Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Gustave Roussy (IGR)-Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Université Pierre et Marie Curie - Paris 6 (UPMC)-Département de Chimie - ENS Paris, École normale supérieure - Paris (ENS-PSL), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-École normale supérieure - Paris (ENS-PSL), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Département de Chimie - ENS Paris, Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS), University of California [San Francisco] (UC San Francisco), University of California (UC)-University of California (UC), Carcinogénèse épithéliale : facteurs prédictifs et pronostiques - UFC (UR 3181) (CEF2P / CARCINO), Centre Hospitalier Régional Universitaire de Besançon (CHRU Besançon)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Etablissement français du sang [Bourgogne-Franche-Comté] (EFS [Bourgogne-Franche-Comté])-Université de Franche-Comté (UFC), Université d'Angers (UA)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Université d'Angers (UA)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre hospitalier universitaire de Nantes (CHU Nantes), Université Pierre et Marie Curie - Paris 6 (UPMC)-École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Pierre et Marie Curie - Paris 6 (UPMC)-École pratique des hautes études (EPHE), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Franche-Comté (UFC), Université Bourgogne Franche-Comté [COMUE] (UBFC)-Université Bourgogne Franche-Comté [COMUE] (UBFC)-Etablissement français du sang [Bourgogne-Franche-Comté] (EFS [Bourgogne-Franche-Comté]), Centre Hospitalier Régional Universitaire de Besançon (CHRU Besançon)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Etablissement français du sang [Bourgogne-Franche-Comté] (EFS BFC)-Université de Franche-Comté (UFC), Institut National de la Santé et de la Recherche Médicale (INSERM)-Etablissement français du sang [Bourgogne-Franche-Comté] (EFS BFC)-Université de Franche-Comté (UFC), Universität Duisburg-Essen = University of Duisburg-Essen [Essen]-University Hospital [Heidelberg, Germany]-Germany & German Cancer Consortium - DKTZ [Heidelberg, Germany], Université Pierre et Marie Curie - Paris 6 (UPMC)-École Pratique des Hautes Études (EPHE), and Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Pierre et Marie Curie - Paris 6 (UPMC)-École Pratique des Hautes Études (EPHE)
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_treatment ,Medizin ,General Physics and Astronomy ,0302 clinical medicine ,METASTATIC MELANOMA ,TUMOR-INFILTRATING LYMPHOCYTES ,lcsh:Science ,Cancer ,screening and diagnosis ,Multidisciplinary ,CD137 ,3. Good health ,Detection ,030220 oncology & carcinogenesis ,Patient Safety ,BREAST-CANCER PATIENTS ,Nivolumab ,Adjuvant ,CIRCULATING SOLUBLE FAS ,medicine.drug ,medicine.medical_specialty ,CELL LUNG-CANCER ,Science ,PROGNOSTIC IMPACT ,Clinical Trials and Supportive Activities ,Ipilimumab ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Immune system ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Clinical Research ,Internal medicine ,Journal Article ,STAGE-III MELANOMA ,medicine ,CLINICAL-SIGNIFICANCE ,business.industry ,UNTREATED MELANOMA ,General Chemistry ,Immune checkpoint ,Blockade ,4.1 Discovery and preclinical testing of markers and technologies ,030104 developmental biology ,COMBINED NIVOLUMAB ,Immunology ,lcsh:Q ,business ,CD8 - Abstract
Immune checkpoint blockers (ICB) have become pivotal therapies in the clinical armamentarium against metastatic melanoma (MMel). Given the frequency of immune related adverse events and increasing use of ICB, predictors of response to CTLA-4 and/or PD-1 blockade represent unmet clinical needs. Using a systems biology-based approach to an assessment of 779 paired blood and tumor markers in 37 stage III MMel patients, we analyzed association between blood immune parameters and the functional immune reactivity of tumor-infiltrating cells after ex vivo exposure to ICB. Based on this assay, we retrospectively observed, in eight cohorts enrolling 190 MMel patients treated with ipilimumab, that PD-L1 expression on peripheral T cells was prognostic on overall and progression-free survival. Moreover, detectable CD137 on circulating CD8+ T cells was associated with the disease-free status of resected stage III MMel patients after adjuvant ipilimumab + nivolumab (but not nivolumab alone). These biomarkers should be validated in prospective trials in MMel., The clinical management of metastatic melanoma requires predictors of the response to checkpoint blockade. Here, the authors use immunological assays to identify potential prognostic/predictive biomarkers in circulating blood cells and in tumor-infiltrating lymphocytes from patients with resected stage III melanoma.
- Published
- 2017
- Full Text
- View/download PDF
23. The additional facet of immunoscore: immunoprofiling as a possible predictive tool for cancer treatment
- Author
-
Paolo A. Ascierto, Walter J. Urba, Gennaro Ciliberto, Carlo Bifulco, Bernard A. Fox, Mariaelena Capone, Francesco M. Marincola, Jérôme Galon, Gerardo Botti, Alessandro Lugli, BMC, Ed., Melanoma, Cancer Immunotherapy and Innovative Therapy Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS)-Istituto Nazionale dei Tumori di Napoli 'Fondazione G. Pascale', Earle A. Chiles Research Institute, Providence Portland Medical Center-Robert W. Franz Cancer Research Center, Translational Research Unit (TRU), Institute of Pathology-Clinical Pathology Division-University of Bern, Sidra Medical and Research Centre, Weill Medical College of Cornell University [New York], Centre de Recherche des Cordeliers (CRC (UMR_S 872)), Université Pierre et Marie Curie - Paris 6 (UPMC)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Department of Molecular Microbiology and Immunology, Oregon Health and Science University [Portland] (OHSU)-Knight Cancer Institute, Université Paris Descartes - Paris 5 (UPD5)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Translational Research Unit ( TRU ), Centre de Recherche des Cordeliers ( CRC (UMR_S 872) ), Université Pierre et Marie Curie - Paris 6 ( UPMC ) -Université Paris Descartes - Paris 5 ( UPD5 ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Centre National de la Recherche Scientifique ( CNRS ), and Oregon Health and Science University-Knight Cancer Institute
- Subjects
Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Ipilimumab ,610 Medicine & health ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,General Biochemistry, Genetics and Molecular Biology ,Tumor-infiltrating lymphocytes ,[ SDV.CAN ] Life Sciences [q-bio]/Cancer ,03 medical and health sciences ,0302 clinical medicine ,Text mining ,Immune system ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Internal medicine ,Neoplasms ,[ SDV.MHEP ] Life Sciences [q-bio]/Human health and pathology ,medicine ,Humans ,030304 developmental biology ,Medicine(all) ,0303 health sciences ,Tumor microenvironment ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,business.industry ,Biochemistry, Genetics and Molecular Biology(all) ,FOXP3 ,General Medicine ,Immunotherapy ,Immunoprofiling ,Prognosis ,3. Good health ,Editorial ,030220 oncology & carcinogenesis ,Immunology ,570 Life sciences ,biology ,Nivolumab ,business ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology ,medicine.drug - Abstract
International audience; Recent investigations of the tumor microenvironment have shown that many tumors are infiltrated by inflammatory and lymphocytic cells. Increasing evidence suggests that the number, type and location of these tumor-infiltrating lymphocytes in primary tumors has prognostic value, and this has led to the development of an 'immunoscore' As well as providing useful prognostic information, the immunoscore concept also has the potential to help predict response to treatment, thereby improving decision- making with regard to choice of therapy. This predictive aspect of the tumor microenvironment forms the basis for the concept of immunoprofiling, which can be described as 'using an individual's immune system signature (or profile) to predict that patient's response to therapy. The immunoprofile of an individual can be genetically determined or tumor-induced (and therefore dynamic). Ipilimumab is the first in a series of immunomodulating antibodies and has been shown to be associated with improved overall survival in patients with advanced melanoma. Other immunotherapies in development include anti-programmed death 1 protein (nivolumab), anti-PD-ligand 1, anti-CD137 (urelumab), and anti-OX40. Biomarkers that can be used as predictive factors for these treatments have not yet been clinically validated. However, there is already evidence that the tumor microenvironment can have a predictive role, with clinical activity of ipilimumab related to high baseline expression of the immune-related genes FoxP3 and indoleamine 2,3-dioxygenase and an increase in tumor-infiltrating lymphocytes. These biomarkers could represent the first potential proposal for an immunoprofiling panel in patients for whom anti-CTLA-4 therapy is being considered, although prospective data are required. In conclusion, the evaluation of systemic and local immunological biomarkers could offer useful prognostic information and facilitate clinical decision making. The challenge will be to identify the individual immunoprofile of each patient and the consequent choice of optimal therapy or combination of therapies to be used.
- Published
- 2013
- Full Text
- View/download PDF
24. Future perspectives in melanoma research. Meeting report from the 'Melanoma Research: a bridge Naples-USA. Naples, December 6th-7th 2010'
- Author
-
Mario Sznol, Licia Rivoltini, Nicola Mozzillo, Frederic Lehmann, Michele Maio, Alistair J. Cochran, Claudio Dansky Ullmann, Bernard A. Fox, Giuseppe Palmieri, Soldano Ferrone, Stefano Bertuzzi, Antoni Ribas, Eleonora De Maio, David F. Stroncek, John M. Kirkwood, Mohamed Kashani-sabet, Ruth Halaban, Mary J.C. Hendrix, F. M. Marincola, Keith T. Flaherty, Ena Wang, Douglas J Schwartzentruber, Jérôme Galon, Peter P. Lee, Paolo A. Ascierto, James Chih-Hsin Yang, Department of Melanoma, Sarcoma, and Head and Neck Disease, Instituto Nazionale Tumori Fondazione Pascale, Office of Science Policy Analysis, National Institutes of Health, Unit of Cancer Genetics, National Research Council (CNR)-Institute of Biomolecular Chemistry, Department of Dermatology, Yale University School of Medicine, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Center for Melanoma Research and Treatment, California Pacific Medical Center Research Institute, University of Pittsburgh (PITT), Pennsylvania Commonwealth System of Higher Education (PCSHE), Infectious Diseases and Immunogenetics Section (IDIS), Department of Transfusion Medicine-Clinical Center and Center for Human Immunology (CHI), NIH, David Geffen School of Medicine [Los Angeles], University of California [Los Angeles] (UCLA), University of California-University of California, Unit of Immunotherapy of Human Tumors, Istituto Nazionale Tumori-IRCCS Foundation, Stanford School of Medicine [Stanford], Stanford Medicine, Stanford University-Stanford University, Laboratory of Molecular and Tumor Immunology, Providence Portland Medical Center-Robert W. Franz Cancer Research Center-Earle A. Chiles Research Institute, Department of Molecular Microbiology and Immunology, Oregon Health and Science University [Portland] (OHSU), Department of Medicine, Division of Hematology/Oncology-Pittsburgh Cancer Institute-University of Pittsburgh (PITT), Pennsylvania Commonwealth System of Higher Education (PCSHE)-Pennsylvania Commonwealth System of Higher Education (PCSHE), Cancer Therapy Evaluation Program DCTD, National Cancer Institute [Bethesda] (NCI-NIH), National Institutes of Health [Bethesda] (NIH)-National Institutes of Health [Bethesda] (NIH)-Clinical Investigations Branch, Cancer Immunotherapeutics Business Unit, GlaxoSmithKline Biologicals, Yale Cancer Center, Center for Cancer Care, Indiana University System, Medical Oncology and Immunotherapy, Hospital of Siena-Department of Oncology-Instituto Toscano Tumori, Cancer Center, Massachusetts General Hospital [Boston], Centre de Recherche des Cordeliers (CRC (UMR_S 872)), Université Pierre et Marie Curie - Paris 6 (UPMC)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Jonsson Comprehensive Cancer Center, Surgical Brach, National Institute of the Health (NIH), This work was supported by the Italian Ministry of Health 'Progetto Ricerca Corrente Istituto Nazionale Tumori Pascale M2/11 Approccio multidisciplinare dalla ricerca alla cura del melanoma' and by Fondazione Melanoma Onlus., Yale School of Medicine [New Haven, Connecticut] (YSM), University of California (UC)-University of California (UC), Yale School of Medicine, University of Pittsburgh, Cancer Institute, Infectious Diseases and Immunogenetics Section ( IDIS ), David Geffen School of Medicine at UCLA, University of California at Los Angeles [Los Angeles] ( UCLA ), Stanford University School of Medicine [Stanford], Stanford University [Stanford], Oregon Health and Science University, Division of Hematology/Oncology-Pittsburgh Cancer Institute-University of Pittsburgh, National Cancer Institute ( NIH ) -Clinical Investigations Branch, Indiana University Health Goshen, Massachusetts General Hospital, Centre de Recherche des Cordeliers ( CRC (UMR_S 872) ), Université Pierre et Marie Curie - Paris 6 ( UPMC ) -Université Paris Descartes - Paris 5 ( UPD5 ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Centre National de la Recherche Scientifique ( CNRS ), and BMC, Ed.
- Subjects
Pathology ,medicine.medical_specialty ,Library science ,lcsh:Medicine ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Review ,General Biochemistry, Genetics and Molecular Biology ,[ SDV.CAN ] Life Sciences [q-bio]/Cancer ,03 medical and health sciences ,0302 clinical medicine ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Clinical investigation ,International congress ,[ SDV.MHEP ] Life Sciences [q-bio]/Human health and pathology ,melanoma ,Medicine ,030304 developmental biology ,Pharmaceutical industry ,0303 health sciences ,Government ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,Melanoma patient ,business.industry ,lcsh:R ,General Medicine ,3. Good health ,Clinical Practice ,Clinical trial ,030220 oncology & carcinogenesis ,Molecular targets ,business ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
Progress in understanding the molecular basis of melanoma has made possible the identification of molecular targets with important implications in clinical practice. In fact, new therapeutic approaches are emerging from basic science and it will be important to implement their rapid translation into clinical practice by active clinical investigation. The first meeting of Melanoma Research: a bridge Naples-USA, organized by Paolo A. Ascierto (INT, Naples, Italy) and Francesco Marincola (NIH, Bethesda, USA) took place in Naples, on 6-7 December 2010. This international congress gathered more than 30 international and Italian faculty members and was focused on recent advances in melanoma molecular biology, immunology and therapy, and created an interactive discussion across Institutions belonging to Government, Academy and Pharmaceutical Industry, in order to stimulate new approaches in basic, translational and clinical research. Four topics of discussion were identified: New pathways in Melanoma, Biomarkers, Clinical Trials and New Molecules and Strategies.
- Published
- 2011
- Full Text
- View/download PDF
25. Defining the critical hurdles in cancer immunotherapy
- Author
-
Andrea Nicolini, Francesco M. Marincola, William E. Carson, Paolo A. Ascierto, Michele Maio, Jedd D. Wolchok, Michael T. Lotze, Jirina Bartunkova, Weihua Xiao, Hauke Winter, Barbara Seliger, Jon M. Wigginton, Cedrik M. Britten, Ignacio Melero, Guido Kroemer, Neil L. Berinstein, Jill O'Donnell-Tormey, Heinz Zwierzina, Lothar Bergmann, Lloyd J. Old, Christian H. Ottensmeier, Jérôme Galon, Per thor Straten, Koji Kawakami, Michael Papamichail, Yutaka Kawakami, Michael I. Nishimura, Mary L. Disis, Steinar Aamdal, C. J. M. Melief, Pedro Romero, Kristen Hege, Wenru Song, Pawel Kalinski, Jonathan L. Bramson, Harpreet Singh-Jasuja, Jens Peter Marschner, Bernard A. Fox, Samir N. Khleif, Brad H. Nelson, Marij J. P. Welters, Elizabeth M. Jaffee, Patrick Hwu, Rik J. Scheper, Robert C. Rees, Giuseppe Masucci, Hideaki Tahara, Cristina Bonorino, Glenn Dranoff, Ernest C. Borden, William J. Murphy, Zhigang Tian, Michael B. Atkins, Robert O. Dillman, Thomas F. Gajewski, Hiroshi Shiku, Leif Håkansson, Michael J. Mastrangelo, Lisa H. Butterfield, Shukui Qin, Laurence Zitvogel, Harry Dolstra, Michele Guida, George Coukos, Mohamed L. Salem, Xuetao Cao, Giorgio Parmiani, Enrico Proietti, Ena Wang, Sylvia Janetzki, A. Raja Choudhury, Gerd Ritter, Hyam I. Levitsky, Kunle Odunsi, Kohzoh Imai, Paul von Hoegen, Christoph Huber, Réjean Lapointe, Antoni Ribas, Dolores J. Schendel, Pamela S. Ohashi, Beatrix Kotlan, Cécile Gouttefangeas, James H. Finke, Alfred E. Chang, Howard L. Kaufman, Lindy G. Durrant, Sjoerd H. van der Burg, Jared Gollob, Dainius Characiejus, Tara Withington, Padmanee Sharma, Ronald B. Herberman, Cristina Maccalli, Ulrich Keilholtz, Axel Hoos, Graham Pawelec, Fabio Grizzi, Tanja D. de Gruijl, F. Stephen Hodi, Ruggero Ridolfi, James P. Allison, Licia Rivoltini, Carl H. June, Rolf Kiessling, Department of Molecular Microbiology and Immunology, Oregon Health and Science University [Portland] (OHSU)-Knight Cancer Institute, Earle A. Chiles Research Institute, Providence Portland Medical Center-Robert W. Franz Research Center-Providence Cancer Center, Clinical Cooperation Group 'Immune Monitoring', German Research Center for Environmental Health-Helmholtz Centre Munich-Institute of Molecular Immunology, Division of Hematology Oncology, University of Pittsburgh Cancer Institute-Departments of Medicine, Department of Surgery, Cancer Institute-University of Pittsburgh (PITT), Pennsylvania Commonwealth System of Higher Education (PCSHE)-Pennsylvania Commonwealth System of Higher Education (PCSHE), Department of Immunology, University of Pittsburgh Cancer Institute, Department of Clinical Cancer Research, The Norwegian Radium Hospital-Oslo University Hospital [Oslo], Memorial Sloane Kettering Cancer Center [New York], Howard Hughes Medical Institute (HHMI), Medical Oncology and Innovative Therapy, Instituto Nazionale Tumori-Fondazione 'G. Pascale', Beth Israel Deaconess Medical Center, Harvard Medical School [Boston] (HMS), Institute of Immunology, Charles University [Prague] (CU)-FOCIS Center of Excellence-2nd Medical School, Goethe-Universität Frankfurt am Main, IRX Therapeutics, Stanford University-ImmunoVaccine Inc., Instituto Nacional para o Controle do Câncer, Instituto de Pesquisas Biomédicas-PUCRS Faculdade de Biociências, Department of Solid Tumor Oncology, Cleveland Clinic, Department of Translational Hematology and Oncology Research, Department of Pathology, McMaster University [Hamilton, Ontario], University Medical Center Mainz, III. Medical Department, Ribological GmbH, Department of Medicine-University Medical Center of the Johannes Gutenberg-University-Clinical Development, BioNTech AG, Chinese Academy of Medical Sciences, Second Military Medical University-National Key Laboratory of Medical Immunology, Ohio State University [Columbus] (OSU), University of Michigan [Ann Arbor], University of Michigan System-University of Michigan System, Institute of Oncology, Vilnius University [Vilnius]-Faculty of Medicine, Department of Medicine, University of Queensland [Brisbane], Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania [Philadelphia]-University of Pennsylvania [Philadelphia], Department of Medical Oncology, VU Medical Center-Cancer Center Amsterdam, Hoag Institute for Research and Education, Hoag Cancer Institute, Department of Laboratory Medicine, Radboud university [Nijmegen]-Nijmegen Centre for Molecular Life Sciences-Nijmegen Medical Centre [Nijmegen], Brigham and Women's Hospital [Boston], Dana-Farber Cancer Institute [Boston], Academic Department of Clinical Oncology, University of Nottingham, UK (UON), Centre de Recherche des Cordeliers (CRC (UMR_S 872)), Université Paris Descartes - Paris 5 (UPD5)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Alnylam Pharmaceuticals, Inc., Institute for Cell Biology, Istituto Clinico Humanitas [Milan] (IRCCS Milan), Humanitas University [Milan] (Hunimed), Oncology Department, University of Lund, CanImGuide Therapeutics AB, University of California [San Francisco] (UCSF), University of California, Intrexon Corporation, Germantown, Bristol-Myers Squibb Company, Translational Oncology & Immunology, Centre TRON at the Mainz University Medical Center, Department of Melanoma Medical Oncology, The University of Texas M.D. Anderson Cancer Center [Houston], The Institute of Medical Science, The University of Tokyo (UTokyo), Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine [Baltimore]-Johns Hopkins University School of Medicine [Baltimore], ZellNet Consulting, Pathology and Laboratory Medicine, University of Pennsylvania [Philadelphia], Rush University Cancer Center, Rush University Medical Center [Chicago], School of Medicine and Public Health, Kyoto University [Kyoto], Division of Cellular Signaling, Institute for Advanced Medical Research, Dept. of Hematology and Medical Oncology, Charité Comprehensive Cancer Center, Cancer Vaccine Section, NCI, Department of Oncology - Pathology, Cancer Center Karolinska [Karolinska Institutet] (CCK), Karolinska Institutet [Stockholm]-Karolinska Institutet [Stockholm], Department of Molecular Immunology and Toxicology, Center of Surgical and Molecular Tumor pathology, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CR CHUM), Centre Hospitalier de l'Université de Montréal (CHUM), Université de Montréal (UdeM)-Université de Montréal (UdeM), School of Medicine, Johns Hopkins University (JHU)-Oncology Center, Department of Molecular Oncology, Foundation San Raffaele Scientific Institute, Medical Oncology and Immunotherapy, Istituto Toscano Tumori-University Hospital of Siena-Department of Oncology, Merck KGaA, Merck & Co. Inc, Thomas Jefferson University, Department of Oncology-Pathology, karolinska institute, CIMA, CUN and Medical School University of Navarra, Department of Immunohematology and Blood Transfusion, Leiden University Medical Center (LUMC), Davis Medical Center, Sacramento-University of California, Deeley Research Centre, BC Cancer Agency (BCCRC), Department of Internal Medicine, University of Pisa - Università di Pisa, Oncology Institute, Loyola University Medical Center (LUMC)-Cardinal Bernardin Cancer Center, Tumor Immunology and Immunotherapy Program, Roswell Park Cancer Institute [Buffalo]-Department of Gynecologic Oncology, Ontario Cancer Institute, University Health Network, Cancer Immunotherapy Consortium (CIC), Cancer Research Institute, Cancer Research, Ludwig Institute, Experimental Cancer Medicine Centre, University of Southampton-Faculty of Medicine, Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, Unit of Immuno-Biotherapy of Melanoma and Solid Tumors, San Raffaele Scientific Institute, Center for Medical Research, Eberhard Karls Universität Tübingen = Eberhard Karls University of Tuebingen, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanita', Chinese PLA Cancer Center, Department of Oncology-The Eighty-First Hospital, The John van Geest Cancer Research Centre, School of Science and Technology-Nottingham Trent University, Jonsson Comprehensive Cancer Center, Immunoterapia e Terapia Cellulare Somatica, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (I.R.S.T.), Unit of Immunotherapy of Human Tumors, Istituto Nazionale Tumori-IRCCS Foundation, Division of Clinical Onco-Immunology, Université de Lausanne (UNIL)-Ludwig Center for Cancer Research, Immunology and Biotechnology Unit, Faculty of Science-Department of Zoology-Tanta University, VU University Medical Center [Amsterdam], Institute of Medical Immunology, Martin-Luther-Universität Halle Wittenberg (MLU), Departments of Immunology, Department of Cancer Vaccine, Mie University, Department of Immuno-gene Therapy, Immatics Biotechnologies GmbH, Eberhard Karls Universität Tübingen = Eberhard Karls University of Tuebingen-Department of Immunology-Institute for Cell Biology, Millennium: The Takeda Oncology Company, Pfizer Oncology, Center for Cancer Immune Therapy (CCIT), Herlev and Gentofte Hospital-Department of Hematology, Department of Surgery and Bioengineering, The University of Tokyo (UTokyo)-Institute of Medical Science-Advanced Clinical Research Center, School of Life Sciences-University of Science & Technology of China [Suzhou], Institute of Immunopharmacology & Immunotherapy, Shandong University-School of Pharmaceutical Sciences, Experimental Cancer Immunology and Therapy, Leiden University Medical Center (LUMC)-Department of Clinical Oncology, Euraccine Consulting Group, Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine-Clinical Center-National Institute of Health NIH), Center for Human Immunology (CHI), National Institute of Health (NIH), Leiden University Medical Center (LUMC)-Department of Clinical Oncology (K1-P), Ludwig Maximilians University-Klinikum Grosshadern, Biological Therapy of Cancer, Medical and Surgical Services Organizations-International Society For Biological Therapy Of Cancer, School of Life Science-University of Science and Technology of China [Hefei] (USTC), Immunologie des tumeurs et immunothérapie (UMR 1015), Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut Gustave Roussy (IGR)-Université Paris-Sud - Paris 11 (UP11), Department Haematology and Oncology, Innsbruck Medical University [Austria] (IMU), Medical Center, University of Chicago, Discovery Medicine-Oncology, Tumor Vaccine Group, University of Washington [Seattle]-Center for Translational Medicine in Women's Health, The work of CIMT-CIP was supported by a grant from the Wallace Coulter foundation (Florida, USA)., Helmholtz Centre Munich-Institute of Molecular Immunology-Helmholtz Zentrum München = German Research Center for Environmental Health, University of Pennsylvania-University of Pennsylvania, Radboud University [Nijmegen]-Nijmegen Centre for Molecular Life Sciences-Nijmegen Medical Centre [Nijmegen], Université Pierre et Marie Curie - Paris 6 (UPMC)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Lund University [Lund], University of California [San Francisco] (UC San Francisco), University of California (UC), University of Pennsylvania, Kyoto University, Sacramento-University of California (UC), Université de Lausanne = University of Lausanne (UNIL)-Ludwig Center for Cancer Research, Klinikum Grosshadern-Ludwig-Maximilians University [Munich] (LMU), University of Science and Technology of China [Hefei] (USTC)-School of Life Science, Université Paris-Sud - Paris 11 (UP11)-Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM), Innsbruck Medical University = Medizinische Universität Innsbruck (IMU), BMC, Ed., Computer Systems, Medical oncology laboratory, Pathology, CCA - Immuno-pathogenesis, CCA - Innovative therapy, Oregon Health and Science University-Knight Cancer Institute, Cancer Institute-University of Pittsburgh, The Norwegian Radium Hospital-Oslo University Hospital, Memorial Sloan-Kettering Cancer Center, Sloan-Kettering Institute, Howard Hughes Medical Institute, Ludwig Center for Cancer Immunotherapy, A Teaching Hospital of Harvard Medical School, Charles University [Prague]-FOCIS Center of Excellence-2nd Medical School, Stanford University [Stanford]-ImmunoVaccine Inc., Ohio State University [Columbus] ( OSU ), University of Michigan Medical Center, University of Pennsylvania Medical Center, Radboud university [Nijmegen]-Nijmegen Centre for Molecular Life Sciences-Nijmegen Medical Centre, University of Nottingham, UK ( UON ), Cleveland Clinic Foundation, Centre de Recherche des Cordeliers ( CRC (UMR_S 872) ), Université Pierre et Marie Curie - Paris 6 ( UPMC ) -Université Paris Descartes - Paris 5 ( UPD5 ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Centre National de la Recherche Scientifique ( CNRS ), Istituto Clinico Humanitas [Milan] ( IRCCS Milan ), Humanitas University [Milan] ( Hunimed ), University of California [San Francisco] ( UCSF ), Harvard Medical School [Boston] ( HMS ), MD Anderson Cancer Center, The University of Tokyo, Rush University Medical Center, Cancer Center Karolinska [Karolinska Institutet] ( CCK ), Centre Hospitalier de l'Université de Montréal-Hôpital Notre-Dame Research Center ( CRCHUM ), Department of Medicine-University of Montreal, Johns Hopkins University ( JHU ) -Oncology Center, BC Cancer Agency ( BCCRC ), University of Pisa [Pisa], Loyola University Medical Center ( LUMC ) -Cardinal Bernardin Cancer Center, Cancer Immunotherapy Consortium ( CIC ), University of Southampton [Southampton]-Faculty of Medicine, Eberhard Karls Universität Tübingen, University of Lausanne-Ludwig Center for Cancer Research, Martin-Luther-University Halle-Wittenberg, Mie University Graduate School of Medicine, Eberhard Karls Universität Tübingen-Department of Immunology-Institute for Cell Biology, Center for Cancer Immune Therapy ( CCIT ), Herlev Hospital-Department of Hematology, The University of Tokyo-Institute of Medical Science-Advanced Clinical Research Center, Infectious Disease and Immunogenetics Section ( IDIS ), Center for Human Immunology ( CHI ), University of Science and Technology of China [Hefei] ( USTC ) -School of Life Science, Immunologie des tumeurs et immunothérapie ( UMR 1015 ), Université Paris-Sud - Paris 11 ( UP11 ) -Institut Gustave Roussy ( IGR ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ), Innsbruck Medical University [Austria] ( IMU ), Department of Medicine-Clinical Development, BioNTech AG-Johannes Gutenberg - Universität Mainz = Johannes Gutenberg University (JGU), Universiteit Leiden-Universiteit Leiden, Roswell Park Cancer Institute [Buffalo] (RPCI)-Department of Gynecologic Oncology, Istituto Superiore di Sanità (ISS), Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Universiteit Leiden-Universiteit Leiden-Department of Clinical Oncology, and Universiteit Leiden-Universiteit Leiden-Department of Clinical Oncology (K1-P)
- Subjects
medicine.medical_specialty ,International Cooperation ,medicine.medical_treatment ,Alternative medicine ,lcsh:Medicine ,Translational research ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Cancer Immunotherapy ,General Biochemistry, Genetics and Molecular Biology ,[ SDV.CAN ] Life Sciences [q-bio]/Cancer ,Translational Research, Biomedical ,03 medical and health sciences ,SDG 17 - Partnerships for the Goals ,0302 clinical medicine ,Cancer immunotherapy ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,[ SDV.MHEP ] Life Sciences [q-bio]/Human health and pathology ,Neoplasms ,medicine ,Humans ,In patient ,030304 developmental biology ,Medicine(all) ,0303 health sciences ,geography ,Summit ,geography.geographical_feature_category ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,Biochemistry, Genetics and Molecular Biology(all) ,business.industry ,lcsh:R ,Cancer ,General Medicine ,Public relations ,medicine.disease ,3. Good health ,Clinical trial ,Immunotherapy ,Neoplasms/therapy ,Translational Medical Research ,030220 oncology & carcinogenesis ,Immunology ,Commentary ,Working group ,business ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
Scientific discoveries that provide strong evidence of antitumor effects in preclinical models often encounter significant delays before being tested in patients with cancer. While some of these delays have a scientific basis, others do not. We need to do better. Innovative strategies need to move into early stage clinical trials as quickly as it is safe, and if successful, these therapies should efficiently obtain regulatory approval and widespread clinical application. In late 2009 and 2010 the Society for Immunotherapy of Cancer (SITC), convened an "Immunotherapy Summit" with representatives from immunotherapy organizations representing Europe, Japan, China and North America to discuss collaborations to improve development and delivery of cancer immunotherapy. One of the concepts raised by SITC and defined as critical by all parties was the need to identify hurdles that impede effective translation of cancer immunotherapy. With consensus on these hurdles, international working groups could be developed to make recommendations vetted by the participating organizations. These recommendations could then be considered by regulatory bodies, governmental and private funding agencies, pharmaceutical companies and academic institutions to facilitate changes necessary to accelerate clinical translation of novel immune-based cancer therapies. The critical hurdles identified by representatives of the collaborating organizations, now organized as the World Immunotherapy Council, are presented and discussed in this report. Some of the identified hurdles impede all investigators; others hinder investigators only in certain regions or institutions or are more relevant to specific types of immunotherapy or first-in-humans studies. Each of these hurdles can significantly delay clinical translation of promising advances in immunotherapy yet if overcome, have the potential to improve outcomes of patients with cancer. © 2011 Fox et al; licensee BioMed Central Ltd.
- Published
- 2011
- Full Text
- View/download PDF
26. Future perspectives in melanoma research. Meeting report from the 'Melanoma Bridge. Napoli, December 2nd-4th 2012'
- Author
-
Ascierto, Paolo A., Grimaldi, Antonio M., Acquavella, Nicolas, Borgognoni, Lorenzo, Calabrò, Luana, Cascinelli, Natale, Cesano, Alessandra, Del Vecchio, Michele, Eggermont, Alexander M., Faries, Mark, Ferrone, Soldano, Fox, Bernard A., Gajewski, Thomas F., Galon, Jérôme, Gnjatic, Sacha, Gogas, Helen, Kashani-Sabet, Mohammed, Kaufman, Howard L., Larkin, James, Lo, Roger S., Mantovani, Alberto, Margolin, Kim, Melief, Cornelis, McArthur, Grant, Palmieri, Giuseppe, Puzanov, Igor, Ribas, Antoni, Seliger, Barbara, Sosman, Jeff, Suenaert, Peter, Tarhini, Ahmad A., Trinchieri, Giorgio, Vidal-Vanaclocha, Fernando, Wang, Ena, Ciliberto, Gennaro, Mozzillo, Nicola, Marincola, Francesco M., Thurin, Magdalena, Department of Melanoma, Istituto Nazionale Tumori, Fondazione 'G. Pascale', Center for Cancer Research, National Cancer Institute [Bethesda] (NCI-NIH), National Institutes of Health [Bethesda] (NIH)-National Institutes of Health [Bethesda] (NIH), Plastic and Reconstructive Surgery, Regional Melanoma Refferral Center - S.M. Annunziata Hospital, Medical Oncology and Immunotherapy, Istituto Toscano Tumori-University Hospital of Siena, Melanoma and Sarcoma Unit, Clinical Affairs, Nodality Inc, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Institut Gustave Roussy (IGR), Translational Tumor Immunology, John Wayne Cancer Institute, Department of Surgery, Harvard Medical School [Boston] (HMS)-Massachusetts General Hospital [Boston], Laboratory of Molecular and Tumor Immunology, Earle A. Chiles Research Institute-Robert W. Franz Cancer Center, Department of Molecular Microbiology and Immunology, Oregon Health and Science University [Portland] (OHSU), Oncology and Hematology, University of Chicago, Centre de Recherche des Cordeliers (CRC (UMR_S 872)), Université Paris Descartes - Paris 5 (UPD5)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai [New York] (MSSM), 1st Department of Medicine, Medical School, Center for Melanoma Research and Treatment, California Pacific Medical Center Research Institute, Rush University [Chicago], NHS Foundation Trust [London], The Royal Marsden, Dermatology/Medicine, UCLA Geffen School of Medicine and Jonsson Comprehensive Cancer Center, Humanitas Clinical and Research Institute, Fred Hutchinson Cancer Research Center [Seattle] (FHCRC), ISA Pharmaceuticals, Leiden University Medical Center (LUMC), Peter MacCallum Cancer Centre, Peter MacCallum Cancer Center, Unit of Cancer Genetics, National Research Council [Italy] (CNR)-Institute of Biomolecular Chemistry, Vanderbilt University Medical Center [Nashville], Vanderbilt University [Nashville], Tumor Immunology Program, University of California [Los Angeles] (UCLA), University of California-University of California-Jonsson Comprehensive Cancer Center (JCCC)-David Geffen School of Medicine [Los Angeles], University of California-University of California, Institute of Medical Immunology, Martin-Luther-Universität Halle Wittenberg (MLU), Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, Global Early Clinical Development, Clinical Immunotherapeutics-Immunotherapeutics, GlaxoSmithKline Vaccines, University of Pittsburgh Cancer Institute, Pittsburgh, Cancer and Inflammation Program, Center for Cancer Research-National Institute of Health (NIH), Institute of Applied Molecular Medicine (IMMA), CEU-San Pablo University and HM-Hospitals School of Medicine, Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine-Clinical Center and Center for Human Immunology (CHI)-National Institute of Health (NIH), Sidra Medical and Research Centre, Cancer Diagnosis Program, National Institute of Health (NIH), The meeting was supported by Fondazione Melanoma Onlus and the Society of ImmunoTherapy of Cancer (SITC). A special thanks to 3P Solution of Napoli for their support and cooperation in organizing the meeting and to Michael Hoetzel for providing us with the group picture from the meeting. Sacha Gnjatic would like to acknowledge the Cancer Research Institute/Ludwig Institute for Cancer Research Cancer Vaccine Collaborative for grant support., Université Pierre et Marie Curie - Paris 6 (UPMC)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), University of California (UC)-University of California (UC)-Jonsson Comprehensive Cancer Center (JCCC)-David Geffen School of Medicine [Los Angeles], University of California (UC)-University of California (UC), BMC, Ed., National Cancer Institute ( NIH ), Institut Gustave Roussy ( IGR ), Harvard Medical School [Boston] ( HMS ) -Massachusetts General Hospital, Oregon Health and Science University, Centre de Recherche des Cordeliers ( CRC (UMR_S 872) ), Université Pierre et Marie Curie - Paris 6 ( UPMC ) -Université Paris Descartes - Paris 5 ( UPD5 ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Centre National de la Recherche Scientifique ( CNRS ), Icahn School of Medicine at Mount Sinai [New York], Rush University, Chicago, Fred Hutchinson Cancer Research Center [Seattle] ( FHCRC ), National Research Council [Italy] ( CNR ) -Institute of Biomolecular Chemistry, Vanderbilt University Medical Center, University of California at Los Angeles [Los Angeles] ( UCLA ) -David Geffen School of Medicine-Jonsson Comprehensive Cancer Center (JCCC), Martin Luther University Halle-Wittenberg, Institute of Applied Molecular Medicine ( IMMA ), Infectious Disease and Immunogenetics Section ( IDIS ), University of California-University of California-David Geffen School of Medicine [Los Angeles], University of California-University of California-Jonsson Comprehensive Cancer Center (JCCC), IRCCS Istituto Nazionale dei Tumori [Milano], and Universiteit Leiden-Universiteit Leiden
- Subjects
Medicine(all) ,0303 health sciences ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,Biochemistry, Genetics and Molecular Biology(all) ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,General Medicine ,Meeting Report ,General Biochemistry, Genetics and Molecular Biology ,3. Good health ,NO ,[ SDV.CAN ] Life Sciences [q-bio]/Cancer ,03 medical and health sciences ,0302 clinical medicine ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,030220 oncology & carcinogenesis ,[ SDV.MHEP ] Life Sciences [q-bio]/Human health and pathology ,melanoma ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology ,030304 developmental biology - Abstract
All Authors would like to dedicate this manuscript to the memory of Natale Cascinelli who devoted his life to the research and care of melanoma patients and was an example for all of us; International audience; Recent insights into the genetic and somatic aberrations have initiated a new era of rapidly evolving targeted and immune-based treatments for melanoma. After decades of unsuccessful attempts to finding a more effective cure in the treatment of melanoma now we have several drugs active in melanoma. The possibility to use these drugs in combination to improve responses to overcome the resistance, to potentiate the action of immune system with the new immunomodulating antibodies, and identification of biomarkers that can predict the response to a particular therapy represent new concepts and approaches in the clinical management of melanoma. The third "Melanoma Research: "A bridge from Naples to the World" meeting, shortened as "Bridge Melanoma Meeting" took place in Naples, December 2 to 4th, 2012. The four topics of discussion at this meeting were: advances in molecular profiling and novel biomarkers, combination therapies, novel concepts toward integrating biomarkers and therapies into contemporary clinical management of patients with melanoma across the entire spectrum of disease stage, and the knowledge gained from the biology of tumor microenvironment across different tumors as a bridge to impact on prognosis and response to therapy in melanoma. This international congress gathered more than 30 international faculty members who in an interactive atmosphere which stimulated discussion and exchange of their experience regarding the most recent advances in research and clinical management of melanoma patients.
- Full Text
- View/download PDF
27. Cancer classification using the Immunoscore: a worldwide task force
- Author
-
Iris D. Nagtegaal, Paul Waring, Michael I. Nishimura, Richard Palmqvist, Carlo Bifulco, Yutaka Kawakami, Christian H. Ottensmeier, Alessandro Lugli, Christoph Huber, Réjean Lapointe, Lotfi Chouchane, Noriyuki Sato, Helen K. Angell, Scott Kopetz, Kyogo Itoh, Michele Maio, Yili Wang, Bernard A. Fox, Thomas F. Gajewski, Christine Lagorce, Franck Pagès, Cedrik M. Britten, Ena Wang, Giuseppe Masucci, Patricia Shaw, Frank A. Sinicrope, Jill O'Donnell-Tormey, Fabiana Tatangelo, Andreas Lundqvist, Heinz Zwierzina, James P. Allison, Toshihiko Torigoe, P. Patel, Inti Zlobec, Blaise Clarke, Martin Asslaber, Corrado D'Arrigo, Shoichi Hazama, Martin C. Mihm, Harpreet Singh-Jasuja, Luigi Laghi, Anne Berger, Jérôme Galon, Samir N. Khleif, Paolo Delrio, Shilin N. Shukla, Gerardo Botti, Hartmann Arndt, Francesco M. Marincola, Leif Håkansson, Paolo A. Ascierto, Robert E. Hawkins, Bradly G. Wouters, Sacha Gnjatic, Graham Pawelec, Fabio Grizzi, Fernando Vidal-Vanaclocha, Shuji Ogino, Sebastian Kreiter, Kiyotaka Okuno, Peter Gibbs, Magdalena Thurin, Giorgio Trinchieri, Pamela S. Ohashi, Radiotherapie, RS: GROW - School for Oncology and Reproduction, Centre de Recherche des Cordeliers ( CRC (UMR_S 872) ), Université Pierre et Marie Curie - Paris 6 ( UPMC ) -Université Paris Descartes - Paris 5 ( UPD5 ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Centre National de la Recherche Scientifique ( CNRS ), Université Paris Descartes - Paris 5 ( UPD5 ), Assistance Publique-Hopitaux de Paris, Hôpital Européen Georges Pompidou [APHP] ( HEGP ), Society for Immunotherapy of Cancer, Milwaukee, Infectious Disease and Immunogenetics Section (IDIS), Clinical Center and trans-NIH Center for Human Immunology (CHI), Cancer Diagnosis Program, National Cancer Institute ( NIH ), Oncology - Pathology - Anatomy, Institute of Pathology-University of Bern, Department of Pathology, Providence Portland Medical Center, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione G.Pascale', TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Weill Medical College of Cornell University [New York], Colorectal Surgery Department, Istituto Nazionale per lo Studio e la Cura dei Tumori, University of Erlangen, Institute of Pathology, Medical University Graz, Division of Medical Oncology and Immunotherapy, University Hospital of Siena, Department of Oncology-Pathology, Karolinska Institutet [Stockholm], Harvard Medical School and Massachusetts General Hospital, Boston, CEU-San Pablo University School of Medicine and HM-Hospital of Madrid Scientific Foundation, Institute of Applied Molecular Medicine (IMMA), Ludwig Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, University of Lund, Immatics Biotechnologies GmbH, Experimental Cancer Medicine Centre, University of Southampton Faculty of Medicine, Department Haematology and Oncology, Innsbruck Medical University [Austria] ( IMU ), Molecular Gastroenterology and Department of Gastroenterology, Istituto Clinico Humanitas [Milan] ( IRCCS Milan ), Humanitas University [Milan] ( Hunimed ) -Humanitas University [Milan] ( Hunimed ), Ontario Cancer Institute and Campbell Family Institute for Cancer Research, Princess Margaret Hospital, Departments of Laboratory Medicine, Pathobiology & Radiation Oncology, Division of Cellular Signaling, Institute for Advanced Medical Research, Department of Digestive Surgery and Surgical Oncology, Yamaguchi University [Yamaguchi], Department of Surgery, Kinki University, Cancer Research Institute, New York, Avicenne Hospital, Center for Medical Research, Eberhard Karls Universität Tübingen, Oncology Institute, Loyola University Medical Center ( LUMC ), School of Cancer and Imaging Sciences, University of Manchester [Manchester], Research Center, University Hospital, Department of Oncology-Pathology [Karolinska Institutet], Georgia Health Sciences University Cancer Center, Augusta University, Brigham and Women's Hospital [Boston], Department of Medical Oncology, Royal Melbourne Hospital, University of Melbourne, Sapporo Medical University School of Medicine, Department of Immunology and Immunotherapy, Kurume University School of Medicine, The Gujarat Cancer & Research Institute, Asarwa, Department of Medical Biosciences, Pathology, Pathology Department, Radboud University Medical Center [Nijmegen], Institute for Cancer Research, Center of Translational medicine, Department of Histopathology, Dorset County Hospital, MD Anderson Cancer Center, Houston, Mayo Clinic and Mayo College of Medicine, Rochester, Cancer Inflammation Program, Center for Cancer Research, Oncology and Hematology, University of Chicago, Medical Oncology and Innovative Therapies Unit, Fondazione Melanoma Onlus, Laboratory of Molecular and Tumor Immunology, Earle A. Chiles Research Institute-Robert W. Franz Cancer Center, Department of Molecular Microbiology and Immunology, Oregon Health and Science University, BMC, Ed., Centre de Recherche des Cordeliers (CRC (UMR_S 872)), Université Pierre et Marie Curie - Paris 6 (UPMC)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Université Paris Descartes - Paris 5 (UPD5), Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO), National Cancer Institute [Bethesda] (NCI-NIH), National Institutes of Health [Bethesda] (NIH)-National Institutes of Health [Bethesda] (NIH), Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Harvard Medical School [Boston] (HMS), Memorial Sloane Kettering Cancer Center [New York], Lund University [Lund], Innsbruck Medical University = Medizinische Universität Innsbruck (IMU), Istituto Clinico Humanitas [Milan] (IRCCS Milan), Humanitas University [Milan] (Hunimed)-Humanitas University [Milan] (Hunimed), University of Toronto-University of Toronto, Eberhard Karls Universität Tübingen = Eberhard Karls University of Tuebingen, Loyola University Medical Center (LUMC), Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CR CHUM), Centre Hospitalier de l'Université de Montréal (CHUM), Université de Montréal (UdeM)-Université de Montréal (UdeM), University System of Georgia (USG)-University System of Georgia (USG), Sapporo Medical University-Sapporo Medical University, Kurume University-Kurume University, Oregon Health and Science University [Portland] (OHSU), Université Paris Descartes - Paris 5 (UPD5)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Innsbruck Medical University [Austria] (IMU), and Johannes Gutenberg - Universität Mainz = Johannes Gutenberg University (JGU)
- Subjects
Oncology ,medicine.medical_specialty ,Medicin och hälsovetenskap ,Internationality ,Colorectal cancer ,Advisory Committees ,lcsh:Medicine ,Review ,Medical and Health Sciences ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Internal medicine ,[ SDV.MHEP ] Life Sciences [q-bio]/Human health and pathology ,Tumor Microenvironment ,medicine ,Humans ,Stage (cooking) ,Biomarker discovery ,030304 developmental biology ,Medicine(all) ,0303 health sciences ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,Merkel cell carcinoma ,business.industry ,Biochemistry, Genetics and Molecular Biology(all) ,lcsh:R ,Cancer ,General Medicine ,Classification ,medicine.disease ,Primary tumor ,3. Good health ,Treatment Outcome ,Tumor progression ,030220 oncology & carcinogenesis ,Immunology ,Biomarker (medicine) ,business ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
International audience; Prediction of clinical outcome in cancer is usually achieved by histopathological evaluation of tissue samples obtained during surgical resection of the primary tumor. Traditional tumor staging (AJCC/UICC-TNM classification) summarizes data on tumor burden (T), presence of cancer cells in draining and regional lymph nodes (N) and evidence for metastases (M). However, it is now recognized that clinical outcome can significantly vary among patients within the same stage. The current classification provides limited prognostic information, and does not predict response to therapy. Recent literature has alluded to the importance of the host immune system in controlling tumor progression. Thus, evidence supports the notion to include immunological biomarkers, implemented as a tool for the prediction of prognosis and response to therapy. Accumulating data, collected from large cohorts of human cancers, has demonstrated the impact of immune-classification, which has a prognostic value that may add to the significance of the AJCC/UICC TNM-classification. It is therefore imperative to begin to incorporate the 'Immunoscore' into traditional classification, thus providing an essential prognostic and potentially predictive tool. Introduction of this parameter as a biomarker to classify cancers, as part of routine diagnostic and prognostic assessment of tumors, will facilitate clinical decision-making including rational stratification of patient treatment. Equally, the inherent complexity of quantitative immunohistochemistry, in conjunction with protocol variation across laboratories, analysis of different immune cell types, inconsistent region selection criteria, and variable ways to quantify immune infiltration, all underline the urgent requirement to reach assay harmonization. In an effort to promote the Immunoscore in routine clinical settings, an international task force was initiated. This review represents a follow-up of the announcement of this initiative, and of the J Transl Med. editorial from January 2012. Immunophenotyping of tumors may provide crucial novel prognostic information. The results of this international validation may result in the implementation of the Immunoscore as a new component for the classification of cancer, designated TNM-I (TNM-Immune).
- Full Text
- View/download PDF
28. The immune score as a new possible approach for the classification of cancer
- Author
-
Franck Pagès, Bernard A. Fox, Thomas F. Gajewski, Giorgio Trinchieri, Francesco M. Marincola, Paolo A. Ascierto, Magdalena Thurin, Jérôme Galon, BMC, Ed., Centre de Recherche des Cordeliers (CRC (UMR_S 872)), Université Pierre et Marie Curie - Paris 6 (UPMC)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Society for ImmunoTherapy of Cancer, Society for ImmunoTherapy of Cancer, Milwaukee, Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO), Infectious Disease and Immunogenetics Section (IDIS), Clinical Center and trans-NIH Center for Human Immunology (CHI)and trans-NIH Center for Human Immunology (CHI), National Institutes of Health [Bethesda] (NIH), Cancer Diagnosis Program, National Institutes of Health [Bethesda] (NIH)-National Cancer Institute (NCI), Laboratory of Experimental Immunology, Laboratory of Molecular and Tumor Immunology, Providence Portland Medical Center-Earle A. Chiles Research Institute-Robert W. Franz Cancer Center, Department of Molecular Microbiology and Immunology, Oregon Health and Science University [Portland] (OHSU), Department of Pathology and Department of Medicine, University of Chicago, Medical Oncology and Innovative Therapies Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori-Fondazione 'G. Pascale', Fondazione Melanoma Onlus Via Mariano Semmola, Centre de Recherche des Cordeliers ( CRC (UMR_S 872) ), Université Pierre et Marie Curie - Paris 6 ( UPMC ) -Université Paris Descartes - Paris 5 ( UPD5 ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Centre National de la Recherche Scientifique ( CNRS ), Hôpital Européen Georges Pompidou [APHP] ( HEGP ), National Institutes of Health ( NIH ), National Institutes of Health ( NIH ) -National Cancer Institute (NCI), and Oregon Health and Science University
- Subjects
Oncology ,medicine.medical_specialty ,[SDV.IMM] Life Sciences [q-bio]/Immunology ,medicine.medical_treatment ,Advisory Committees ,MEDLINE ,lcsh:Medicine ,General Biochemistry, Genetics and Molecular Biology ,Access to Information ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Internal medicine ,[ SDV.MHEP ] Life Sciences [q-bio]/Human health and pathology ,Neoplasms ,medicine ,Tumor Microenvironment ,[ SDV.IMM ] Life Sciences [q-bio]/Immunology ,Humans ,Stage (cooking) ,030304 developmental biology ,Medicine(all) ,0303 health sciences ,Tumor microenvironment ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,business.industry ,Biochemistry, Genetics and Molecular Biology(all) ,Melanoma ,lcsh:R ,Cancer ,General Medicine ,Immunotherapy ,medicine.disease ,Primary tumor ,3. Good health ,Editorial ,030220 oncology & carcinogenesis ,Immunology ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,business ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
The outcome prediction in cancer is usually achieved by evaluating tissue samples obtained during surgical removal of the primary tumor focusing on their histopathological characteristics. Tumor staging (AJCC/UICC-TNM classification) summarizes data on tumor burden (T), presence of cancer cells in draining and regional lymph nodes (N), and evidence for metastases (M). However, this classification provides limited prognostic information in estimating the outcome in cancer and does not predict response to therapy. It is recognized that cancer outcomes can vary significantly among patients within the same stage. Recently, many reports suggest that cancer development is controlled by the host's immune system underlying the importance of including immunological biomarkers for the prediction of prognosis and response to therapy. Data collected from large cohorts of human cancers demonstrated that the immune-classification has a prognostic value that may be superior to the AJCC/UICC TNM-classification. Thus, it is imperative to begin incorporating immune scoring as a prognostic factor and to introduce this parameter as a marker to classify cancers, as part of the routine diagnostic and prognostic assessment of tumors. At the same time, the inherent complexity of quantitative immunohistochemistry, in conjunction with variable assay protocols across laboratories, the different immune cell types analyzed, different region selection criteria, and variable ways to quantify immune infiltration underscore the urgent need to reach assay harmonization. In an effort to promote the immunoscore in routine clinical settings worldwide, the Society for Immunotherapy of Cancer (SITC), the European Academy of Tumor Immunology, the Cancer and Inflammation Program, the National Cancer Institute, National Institutes of Health, USA and "La Fondazione Melanoma" will jointly initiate a task force on Immunoscoring as a New Possible Approach for the Classification of Cancer that will take place in Naples, Italy, February 13th, 2012. The expected outcome will include a concept manuscript that will be distributed to all interested participants for their contribution before publication outlining the goal and strategy to achieve this effort; a preliminary summary to be presented during the "Workshop on Tumor Microenvironment" prior to the SITC annual meeting on October 24th - 25th 2012 in Bethesda, Maryland, USA and finally a "Workshop on Immune Scoring" to be held in Naples in December of 2012 leading to the preparation of a summary document providing recommendations for the harmonization and implementation of the Immune Score as a new component for the classification of cancer.
- Full Text
- View/download PDF
29. Autogene cevumeran with or without atezolizumab in advanced solid tumors: a phase 1 trial.
- Author
-
Lopez J, Powles T, Braiteh F, Siu LL, LoRusso P, Friedman CF, Balmanoukian AS, Gordon M, Yachnin J, Rottey S, Karydis I, Fisher GA, Schmidt M, Schuler M, Sullivan RJ, Burris HA, Galvao V, Henick BS, Dirix L, Jaeger D, Ott PA, Wong KM, Jerusalem G, Schiza A, Fong L, Steeghs N, Leidner RS, Rittmeyer A, Laurie SA, Gort E, Aljumaily R, Melero I, Sabado RL, Rhee I, Mancuso MR, Muller L, Fine GD, Yadav M, Kim L, Leveque VJP, Robert A, Darwish M, Qi T, Zhu J, Zhang J, Twomey P, Rao GK, Low DW, Petry C, Lo AA, Schartner JM, Delamarre L, Mellman I, Löwer M, Müller F, Derhovanessian E, Cortini A, Manning L, Maurus D, Brachtendorf S, Lörks V, Omokoko T, Godehardt E, Becker D, Hawner C, Wallrapp C, Albrecht C, Kröner C, Tadmor AD, Diekmann J, Vormehr M, Jork A, Paruzynski A, Lang M, Blake J, Hennig O, Kuhn AN, Sahin U, Türeci Ö, and Camidge DR
- Abstract
Effective targeting of somatic cancer mutations to enhance the efficacy of cancer immunotherapy requires an individualized approach. Autogene cevumeran is a uridine messenger RNA lipoplex-based individualized neoantigen-specific immunotherapy designed from tumor-specific somatic mutation data obtained from tumor tissue of each individual patient to stimulate T cell responses against up to 20 neoantigens. This ongoing phase 1 study evaluated autogene cevumeran as monotherapy (n = 30) and in combination with atezolizumab (n = 183) in pretreated patients with advanced solid tumors. The primary objective was safety and tolerability; exploratory objectives included evaluation of pharmacokinetics, pharmacodynamics, preliminary antitumor activity and immunogenicity. Non-prespecified interim analysis showed that autogene cevumeran was well tolerated and elicited poly-epitopic neoantigen-specific responses, encompassing CD4
+ and/or CD8+ T cells, in 71% of patients, most of them undetectable at baseline. Responses were detectable up to 23 months after treatment initiation. CD8+ T cells specific for several neoantigens constituted a median of 7.3% of circulating CD8+ T cells, reaching up to 23% in some patients. Autogene cevumeran-induced T cells were found within tumor lesions constituting up to 7.2% of tumor-infiltrating T cells. Clinical activity was observed, including one objective response in monotherapy dose escalation and in two patients with disease characteristics unfavorable for response to immunotherapy treated in combination with atezolizumab. These findings support the continued development of autogene cevumeran in earlier treatment lines. ClinicalTrials.gov registration: NCT03289962 ., Competing Interests: Competing interests: J.L. reports research funding to their institution from Roche-Genentech, Basilea, Astex, Merck, GSK, Eisai, Seagen, Immunocore, BicycleTx, AstraZeneca, Genmab, Bayer, Jansenn, Gilead, Anaveon, Affimed, Appollomics, Avacta, Byondis, Chugai, CellCentric, Daiichi, Iteos, Merck Serono, REDX, and Amgen, and a consulting and advisory role for Basilea, Roche-Genentech, Ellipses, Eisai, Pierre-Faber and GSK. T.P. reports a consulting and advisory role for Astellas Pharma, Bristol Myers Squibb, Exelixis, Incyte, Ipsen, Johnson & Johnson, Mashup Ltd, Merck Serono, MSD, Novartis, Pfizer, Roche and Seattle Genetics; travel, accommodation or expenses from Ipsen, MSD, Pfizer and Roche; and research funding from Astellas Pharma, Bristol Myers Squibb, Exelixis, Ipsen, Johnson & Johnson, Merck Serono, MSD, Novartis, Pfizer, Roche, Seattle Genetics. F.B. reports speaking engagements and advisory boards for Roche/Genentech, BMS, Lilly, EMD Serono, AstraZeneca and Regeneron, Merck, Seagen, Jazz Pharmaceuticals, Taiho, Incyte Deciphera and Astellas. L.L.S. reports a consulting and advisory role for Merck, Pfizer, AstraZeneca, Roche, GlaxoSmithKline, Voronoi, Arvinas, Navire, Relay, Daiichi Sankyo, Coherus, Amgen, Marengo, Medicenna, Tubulis, LTZ Therapeutics and Pangea; grants or support for clinical trials to their institution from Novartis, Bristol Myers Squibb, Pfizer, Boerhinger-Ingelheim, GlaxoSmithKline, Roche/Genentech, AstraZeneca, Merck, Celgene, Astellas, Bayer, Abbvie, Amgen, Symphogen, Mirati, BioNTech, 23Me and EMD Serono; stock ownership (spouse) of Agios; and leadership (spouse) at Treadwell Therapeutics. P.L. has served on the advisory boards of Abbvie, GenMab, Genentech, CytomX, Takeda, Cybrexa, Agenus, IQVIA, TRIGR, Pfizer, ImmunoMet, Black Diamond, GSK, QED Therapeutics, AstraZeneca, EMD Serono, Shattuck, Astellas, Salarius, Silverback, MacroGenics, Kyowa Kirin Pharmaceutical Development, Kineta, Inc, Zentalis Pharmaceuticals, Molecular Templates, STCube Pharmaceuticals, Bayer, I-Mab, Seagen, imCheck, Relay Therapeutics, Stemline, Compass BADX, Mekanist, Mersana Therapeutics, BAKX Therapeutics, Scenic Biotech, Qualigen, NeuroTrials and Actuate Therapeutics; on the data safety monitoring board for Agios, Five Prime, Halozyme and Tyme; and as a consultant for Roche-Genentech, SOTIO, SK Life Science and Roivant Sciences. C.F.F. reports a consulting and advisory role (self) for Seagen, Aadi Biosciences, Genentech/MyPathway (uncompensated) and Merck/LYNK-002 (uncompensated), and institutional funding from Bristol Myers Squibb, Daiichi, Hotspot Therapeutics, Marengo, AstraZeneca, Genentech/Roche, Immunocore, Seagen and Merck. A.S.B. reports serving on a speakers bureau for Genentech, BMS, Mirati, AstraZeneca, Regeneron and Merck and consulting for Pfizer and Abbvie, and is on a steering committee for Janssen. M.G. reports clinical trial support to their institution from Genentech/Roche, GSK, Abbvie, Merck Serono, Medimmune, Incyte, Pfizer, Amgen, Gilead Sciences, Zai Labs, Adanate, Fog Pharma, PEEL, Orionis, SQZ, YMABS, Iovance, Vincerx, Werewolf, Endocyte, Seattle Genetics, Plexxicon/Daiichi, Celldex, Tracon, Deciphera, Fujifilm, Minnemarita, Nektar, Novita, Biosplice, Corcept, Novartis, Toray, Genzada, Salarius, Agenus, Inhibrx, AADI, Revolution Medicine, Blueprint, Astellas, BioNTech, Helix, IgM Biosciences, ImmuneSensor, Bioeclipse, Bioline, Black Diamond, Codiak, Dracen, Elevation Oncology, Famewave, Forma Therapeutics, IntraImmun SG, Pionyr, Trishula, Tolero, Vedanta Biosciences, Coordination Therapeutics, Ideaya Biosciences, I-Mab, NiKang, Nimbus Therapeutics, OncoResponse, Riboscience, Rubius Therapeutics, Simcha Therapeutics, Siranomics, Synthorx and Theseus Pharmaceuticals; consulting fees from Pfizer, Imaging Endpoints and Curio; honoraria from Lisa Stearns Academy; a patent with Sphinx Health Solutions; and advisory boards for Daiichi, Qualigen, Springworks, Cardinal Healthcare, IQVIA and Medtronics. I.K. reports consulting fees from and serving in a speakers bureau for Delcath Inc., Immunocore Ltd and Pierre Fabre Inc; educational grants from BMS and Novartis; and travel grants from Delcath Inc, Genentech Inc, BMS and Merck Serono. G.A.F. reports serving on a data safety monitoring board for AstraZeneca and Hutchison Pharma and advisory boards for Bristol Myers Squibb and Merck. M. Schmidt reports personal fees from AstraZeneca, BioNTech, Daiichi Sankyo, Eisai, Lilly, MSD, Novartis, Pantarhei Bioscience, Pfizer, Pierre Fabre, Roche and SeaGen outside the submitted work; institutional research funding from AstraZeneca, BioNTech, Eisai, Genentech, German Breast Group, Novartis, Palleos, Pantarhei Bioscience, Pierre Fabre and SeaGen; and a patent for EP 2390370 B1 filed for EP 2951317 B1 filed. M. Schuler received consulting fees from Amgen, AstraZeneca, Blueprint Medicines, Boehringer Ingelheim, Bristol Myers Squibb, GlaxoSmithKline, Janssen, Merck Serono, Novartis, Roche, Sanofi, Takeda and Tacalyx; honoraria from Amgen, Bristol Myers Squibb, Janssen, MSD, Novartis, Roche and Sanofi; and research funding to their institution from AstraZeneca, Bristol Myers Squibb and Janssen. R.J.S. served as a consultant and in advisory boards for Marengo, Merck, Novartis, Pfizer and Replimune and has received research funding to their institution from Merck. H.B. reports research funding to their institution from AbbVie, Agios, ARMO Biosciences, Array BioPharma, Arvinas, AstraZeneca, Bayer, BeiGene, BioAtla, BioMed Valley Discoveries, BioTheryX, Boehringer Ingelheim, Bristol Myers Squibb, CALGB, Celgene, CicloMed, Coordination Pharmaceuticals, eFFECTOR Therapeutics, Lilly, EMD Serono, Roche/Genentech, GlaxoSmithKline, Gossamer Bio, Harpoon Therapeutics, Hengrui Therapeutics, Incyte, Janssen, Jounce Therapeutics, Kymab, MacroGenics, MedImmune, Merck, Millennium/Takeda, Moderna, NGM Biopharmaceuticals, Novartis, Pfizer, Revolution Medicines, Ryvu Therapeutics, Foundation Medicine, SeaGen, Tesaro, TG Therapeutics, Verastem, Vertex Pharmaceuticals, Xbiotech and Zymeworks; consulting (uncompensated) for Bristol Myers Squibb, Novartis and TG Therapeutics; consulting (payments to institution) for AstraZeneca, GRAIL, Incyte, Roche and Vincerx Pharma; and stock ownership in HCA Healthcare. V.G. reports institutional support from Seagen Inc., SOTIO Biotech AG, Shattuck Labs, Inc., T-knife GmbH, F. Hoffmann-La Roche Ltd, Janssen Research & Development, LLC, Novartis, Affimed GmbH, Anaveon AG, BioNTech SE, BicycleTx Ltd, Epizyme, Inc., Regeneron Pharmaceuticals, Inc., Boehringer Ingelheim, Genmab, Pieris Pharmaceuticals, Inc., Celgene Corporation, Debiopharm International S.A., F-star Therapeutics Limited, ImCheck Therapeutics, Gilead Sciences, Inc and Sanofi-Aventis Recherche & Développement. B.S.H. reports a consulting and advisory role for AstraZeneca, Ideaya, Jazz Pharmaceuticals, Sorrento Therapeutics, Genentech-Roche, OncLive, Veeva, Athenium, Boxer Capital, SAI-Med and DAVA Oncology, and research funding to their institution from NexImmune, Genentech-Roche, Johnson & Johnson, BMS Foundation/VCU, Stand Up 2 Cancer, V Foundation and National Cancer Institute. P.A.O. reports consulting for Array, Bristol Myers Squibb, Celldex, CytomX, Evaxion, Genentech, Imunon, Merck, MyNEO, Neon Therapeutics, Novartis, Pfizer, Phio, TRM Oncology and Servier, and grant and research support to their institution from Agenus, AstraZeneca/MedImmune, Bristol Myers Squibb, Celldex, CytomX, Genentech, Merck, Neon Therapeutics, Novartis and Pfizer. G.J. received consulting fees from Novartis, Amgen, Roche, Pfizer, Bristol Myers Squibb, Lilly, AstraZeneca, Daiichi Sankyo, Abbvie, Seagen and Diaccurate; honoraria from Novartis, Amgen, Roche, Pfizer, Bristol Myers Squibb, Lilly, AstraZeneca, Daiichi Sankyo, Abbvie and Seagen; and travel support from Novartis, Roche, Pfizer, Lilly, Amgen, Bristol Myers Squibb and AstraZeneca; served on a board for Novartis, Roche, Pfizer, Lilly, Amgen, Bristol Myers Squibb and AstraZeneca; and received materials or services from Novartis, Roche, Lilly, Amgen, Bristol Myers Squibb and AstraZeneca. L.F. reports research support from Abbvie, Bavarian Nordic, Bristol Myers Squibb, Dendreon, Janssen, Merck and Roche/Genentech, and ownership interests in Actym, Atreca, Bioatla, Bolt, Immunogenesis, Nutcracker, RAPT, Scribe and Senti. N.S. provided consultation or attended advisory boards for Boehringer Ingelheim, Ellipses Pharma, GlaxoSmithKline, Incyte and Luszana, and received research grants from Abbvie, Actuate Therapeutics, Amgen, Array, Ascendis Pharma, AstraZeneca, Bayer, Blueprint Medicines, Boehringer Ingelheim, BridgeBio, Bristol Myers Squibb, Cantargia, CellCentric, Cogent Biosciences, Cresecendo Biologics, Cytovation, Deciphera, Dragonfly, Eli Lilly, Exelixis, Genentech, GlaxoSmithKline, IDRx, Immunocore, Incyte, InteRNA, Janssen, Kinnate Biopharma, Kling Biotherapeutics, Lixte, Luszana, Merck, Merck Sharp & Dohme, Merus, Molecular Partners, Navire Pharma, Novartis, Numab Therapeutics, Pfizer, Relay Pharmaceuticals, Revolution Medicin, Roche, Sanofi, Seattle Genetics, Taiho and Takeda (all outside the submitted work; payment was made to the Netherlands Cancer Institute). R.S.L. reports research and grant funding from Bristol Myers Squibb, Clinigen, Celldex, Incyte and Ubivac, and consulting and advisory roles for Bristol Myers Squibb, Merck, CDR-Life and Vir. A.R. was a consultant and advisor for AbbVie, AstraZeneca, BMS, Boehringer Ingelheim, Daichi Sankyo, Eli Lilly, GSK, MSD, Novartis, Pfizer and Roche/Genentech. S.A.L. reports funding to their institution from Roche/Genentech. R.A. is a primary investigator on S1933 (atezolizumab). I. Melero reports grants from Roche, BMS, Genmab and AstraZeneca, and was a consultant for Roche, BMS, Genmab, AstraZeneca, Biontech, Pharmamar, F-Star, Numab, Mestag, Curon and Bright Peaks. R.L.S., I.R., M.R.M., L. Muller, G.D.F., M.Y., L.K., V.J.P.L., A.R., M.D., T.Q., J. Zhu, J. Zhang, P.T., G.K.R., D.W.L., C.P., A.A.L., J.M.S., L.D. and I. Mellman are or were employees of Genentech, Inc. and are or were Roche stockholders. L. Muller is now an employee and shareholder of Gilead Sciences. G.D.F. reports stock ownership in CARGO Therapeutics. F.M., E.D., A.C., L. Manning, D.M., S.B., V.L., T.O., E.G., D.B., C.H., C.W., C.A., C.K., A.D.T., J.D., M.V., A.J., A.P., J.B., O.H., A.N.K., U.S. and Ö.T. are employees of BioNTech, a company developing immunotherapies against cancer and other diseases, and may hold securities in the company. U.S. and Ö.T. are management board members of BioNTech SE (Mainz, Germany). M.L., M.V., A.D.T., J.D., A.N.K., U.S. and Ö.T. are co-authors on various issued or pending patents that cover parts of this Article. D.R.C. served on advisory boards and consulted for Roche/Genentech. J.Y., S.R., L.D., D.J., K.M.W., A.S. and E.G. report no competing interests., (© 2025. Genentech, Inc. and the Author(s).)- Published
- 2025
- Full Text
- View/download PDF
30. Amivantamab Plus Lazertinib in Patients With EGFR-mutant Non-small Cell Lung Cancer (NSCLC) After Progression on Osimertinib and Platinum-based Chemotherapy: Results From CHRYSALIS-2 Cohort A.
- Author
-
Besse B, Goto K, Wang Y, Lee SH, Marmarelis ME, Ohe Y, Caro RB, Kim DW, Lee JS, Cousin S, Ichihara E, Li Y, Paz-Ares L, Ono A, Sanborn RE, Watanabe N, Jose de Miguel M, Helissey C, Shu CA, Spira AI, Tomasini P, Chih-Hsin Yang J, Zhang Y, Felip E, Griesinger F, Waqar SN, Calles A, Neal JW, Baik CS, Jänne PA, Shreeve SM, Curtin JC, Patel B, Gormley M, Lyu X, Chen J, Chu PL, Mahoney J, Trani L, Bauml JM, Thayu M, Knoblauch RE, and Cho BC
- Abstract
Introduction: Treatment options for patients with epidermal growth factor receptor (EGFR)-mutated non-small cell lung cancer (NSCLC) with disease progression on/after osimertinib and platinum-based chemotherapy are limited., Methods: CHRYSALIS-2 Cohort A evaluated amivantamab+lazertinib in patients with EGFR exon 19 deletion- or L858R-mutated NSCLC with disease progression on/after osimertinib and platinum-based chemotherapy. Primary endpoint was investigator-assessed objective response rate (ORR). Patients received intravenous amivantamab 1050 mg (1400 mg if ≥80 kg) plus oral lazertinib 240 mg., Results: In Cohort A (n=162), investigator-assessed ORR was 28% (95% CI, 22-36). Blinded independent central review (BICR)-assessed ORR was 35% (95% CI, 27-42), with median duration of response (DoR) of 8.3 months (95% CI, 6.7-10.9) and clinical benefit rate of 58% (95% CI, 50-66). At a median follow-up of 12 months, 32/56 responders (57%) achieved a DoR ≥6 months. Median progression-free survival by BICR was 4.5 months (95% CI, 4.1-5.8); median overall survival was 14.8 months (95% CI, 12.2-18.0). Preliminary evidence of central nervous system-anti-tumor activity was reported among 7 patients with baseline brain lesions and no prior brain radiation/surgery. Exploratory biomarker analyses using circulating tumor DNA next-generation sequencing showed responses in patients with and without identified EGFR/MET-dependent resistance. Most frequent adverse events were rash (grouped term; 81%), infusion-related reaction (68%), and paronychia (52%). Most common grade ≥3 treatment-related adverse events were rash (grouped term; 10%), infusion-related reaction (9%), and hypoalbuminemia (6%)., Conclusions: For patients with limited treatment options, amivantamab+lazertinib demonstrated anti-tumor activity with a safety profile characterized by EGFR/MET-realated adverse events, which were generally manageable., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2025
- Full Text
- View/download PDF
31. Checkpoint Immunotherapy for Melanoma - Offering Hope for Cure.
- Author
-
Curti BD
- Published
- 2025
- Full Text
- View/download PDF
32. Ovarian tumor cells gain competitive advantage by actively reducing the cellular fitness of microenvironment cells.
- Author
-
Madan E, Palma AM, Vudatha V, Kumar A, Bhoopathi P, Wilhelm J, Bernas T, Martin PC, Bilolikar G, Gogna A, Peixoto ML, Dreier I, Araujo TF, Garre E, Gustafsson A, Dorayappan KDP, Mamidi N, Sun Z, Yekelchyk M, Accardi D, Olsen AL, Lin L, Titelman AA, Bianchi M, Jessmon P, Farid EA, Pradhan AK, Neufeld L, Yeini E, Maji S, Pelham CJ, Kim H, Oh D, Rolfsnes HO, Marques RC, Lu A, Nagane M, Chaudhary S, Gupta K, Gogna KC, Bigio A, Bhoopathi K, Mannangatti P, Achary KG, Akhtar J, Belião S, Das S, Correia I, da Silva CL, Fialho AM, Poellmann MJ, Javius-Jones K, Hawkridge AM, Pal S, Shree KS, Rakha EA, Khurana S, Xiao G, Zhang D, Rijal A, Lyons C, Grossman SR, Turner DP, Pillappa R, Prakash K, Gupta G, Robinson GLWG, Koblinski J, Wang H, Singh G, Singh S, Rayamajhi S, Bacolod MD, Richards H, Sayeed S, Klein KP, Chelmow D, Satchi-Fainaro R, Selvendiran K, Connolly D, Thorsen FA, Bjerkvig R, Nephew KP, Idowu MO, Kühnel MP, Moskaluk C, Hong S, Redmond WL, Landberg G, Lopez-Beltran A, Poklepovic AS, Sanyal A, Fisher PB, Church GM, Menon U, Drapkin R, Godwin AK, Luo Y, Ackermann M, Tzankov A, Mertz KD, Jonigk D, Tsung A, Sidransky D, Trevino J, Saavedra AP, Winn R, Won KJ, Moreno E, and Gogna R
- Abstract
Cell competition and fitness comparison between cancer and tumor microenvironment (TME) cells determine oncogenic fate. Our previous study established a role for human Flower isoforms as fitness fingerprints, where the expression of Flower Win isoforms in tumor cells leads to growth advantage over TME cells expressing Lose isoforms. Here we demonstrate that the expression of Flower Lose and reduced microenvironment fitness is not a pre-existing condition but, rather, a cancer-induced phenomenon. Cancer cells actively reduce TME fitness by the exosome-mediated release of a cancer-specific long non-coding RNA, Tu-Stroma, which controls the splicing of the Flower gene in the TME cells and expression of Flower Lose isoform, which leads to reduced fitness status. This mechanism controls cancer growth, metastasis and host survival in ovarian cancer. Targeting Flower protein with humanized monoclonal antibody (mAb) in mice significantly reduces cancer growth and metastasis and improves survival. Pre-treatment with Flower mAb protects intraperitoneal organs from developing lesions despite the presence of aggressive tumor cells., Competing Interests: Competing interests: All authors declare no competing interests., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2024
- Full Text
- View/download PDF
33. A novel small molecule Enpp1 inhibitor improves tumor control following radiation therapy by targeting stromal Enpp1 expression.
- Author
-
Baird JR, Alice AF, Saito R, Chai Q, Han M, Ng C, Han S, Fernandez B, Ledoux S, Grosse J, Korman AJ, Potuznik M, Rajamanickam V, Bernard B, Crittenden MR, and Gough MJ
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Immunity, Innate drug effects, Immunity, Innate radiation effects, Neoplasms radiotherapy, Nucleotides, Cyclic metabolism, Stromal Cells metabolism, Stromal Cells drug effects, Stromal Cells radiation effects, Mice, Inbred C57BL, Phosphoric Diester Hydrolases metabolism, Pyrophosphatases metabolism, Pyrophosphatases genetics
- Abstract
The uniqueness in each person's cancer cells and variation in immune infiltrates means that each tumor represents a unique problem, but therapeutic targets can be found among their shared features. Radiation therapy alters the interaction between the cancer cells and the stroma through release of innate adjuvants. The extranuclear DNA that can result from radiation damage of cells can result in production of the second messenger cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) by cyclic GMP-AMP synthase (cGAS). In turn, cGAMP can activate the innate sensor stimulator of interferon genes (STING), resulting in innate immune activation. Ectonucleotide pyrophosphatase/phosphodiesterase 1 (Enpp1) is a phosphodiesterase that can be expressed by cancer cells that can degrade cGAMP, thus can decrease or block STING activation following radiation therapy, impairing the innate immunity that is critical to support adaptive immune control of tumors. We observed that many human and murine cancer cells lack Enpp1 expression, but that Enpp1 is expressed in cells of the tumor stroma where it limits tumor control by radiation therapy. We demonstrate in preclinical models the efficacy of a novel Enpp1 inhibitor and show that this inhibitor improves tumor control by radiation even where the cancer cells lack Enpp1. This mechanism requires STING and type I interferon (IFN) receptor expression by non-cancer cells and is dependent on CD8 T cells as a final effector mechanism of tumor control. This suggests that Enpp1 inhibition may be an effective partner for radiation therapy regardless of whether cancer cells express Enpp1. This broadens the potential patient base for whom Enpp1 inhibitors can be applied to improve innate immune responses following radiation therapy., Competing Interests: Declarations. Competing interests: The work was funded in part by a sponsored research agreement with VIR Biotechnology Inc. In addition, the research was funded in part by NCI R01CA182311, NCI R01CA244142, R01CA208644, R21CA277250, and by the Providence Foundation. MJG receives research funding from Bristol Myers Squibb. MRC receives consulting fees from Roche. Authors listed as employees of VIR Biotechnology Inc. and may own stock or other interests in VIR Biotechnology. Authors have no additional conflicts to declare., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
34. A multicenter phase 2 clinical trial of low-dose subcutaneous decitabine in myelofibrosis.
- Author
-
Lin C, Patel AA, Huo D, Karrison T, van Besien K, Godwin J, Sher D, Weiner H, Green M, Wade JL 3rd, Klisovic R, Baer MR, Larson RA, Stock W, and Odenike O
- Subjects
- Humans, Male, Female, Aged, Middle Aged, Aged, 80 and over, Treatment Outcome, Injections, Subcutaneous, Adult, Prospective Studies, Antimetabolites, Antineoplastic therapeutic use, Antimetabolites, Antineoplastic administration & dosage, Antimetabolites, Antineoplastic adverse effects, Decitabine therapeutic use, Decitabine administration & dosage, Primary Myelofibrosis drug therapy, Azacitidine therapeutic use, Azacitidine administration & dosage, Azacitidine adverse effects
- Abstract
Abstract: Myelofibrosis (MF) in the chronic phase is a challenging disease to treat, and conventional treatment options are geared toward symptom palliation. In this prospective, multicenter, phase 2 trial, 21 patients with MF (18 chronic phase, 2 accelerated phase, and 1 blast phase) were treated with a 10-day schedule of subcutaneous decitabine at 0.3 mg/kg per day. The overall response rate was 33% (95% confidence interval, 15-57), primarily manifested as an improvement in cytopenias. The median duration of response was 7 months (range, 3-44). A high International Prognostic Scoring System risk score, high baseline fetal hemoglobin level, and sustained decrease in circulating CD34+ cell counts were associated with response to decitabine. All patients experienced at least 1 grade 3/4 cytopenia. Nonhematologic toxicities were less frequent, with fatigue, anorexia, and hypocalcemia being the most common. Given the lack of effective therapies in MF with severe cytopenias, this study supports further investigation into the use of hypomethylating agents as single agents or in combination therapies. This trial was registered at www.ClinicalTrials.gov as #NCT00095784., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
35. Phase I dose escalation study of IO-108, an anti-LILRB2 antibody, in patients with advanced solid tumors.
- Author
-
Taylor MH, Naing A, Powderly J, Woodard P, Chung L, Lin WH, Tian H, Siemers N, Xiang H, Deng R, Hong K, Valencia D, Huang T, Zhu Y, Liao XC, Schebye XM, and Patel MR
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Adult, Membrane Glycoproteins antagonists & inhibitors, Aged, 80 and over, Neoplasms drug therapy, Neoplasms immunology, Antibodies, Monoclonal, Humanized therapeutic use, Antibodies, Monoclonal, Humanized pharmacokinetics, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Receptors, Immunologic antagonists & inhibitors
- Abstract
Purpose: In this first-in-human dose escalation study, the safety and efficacy of IO-108, a fully human monoclonal antibody targeting leukocyte immunoglobulin-like receptor B2 (LILRB2), was investigated in patients with advanced solid tumors as monotherapy and in combination with pembrolizumab, an anti-programmed cell death protein 1 (PD-1) antibody., Methods: The study included patients with histologically or cytologically confirmed advanced and relapsed solid tumors, with measurable disease by Response Evaluation Criteria In Solid Tumors (RECIST) V.1.1. Patients were treated with escalating doses of IO-108 every 3 weeks (Q3W) as monotherapy and in combination with pembrolizumab. Safety and tolerability were the primary objectives. Secondary and exploratory objectives included: pharmacokinetics, clinical efficacy, immunogenicity and biomarkers., Results: Of 25 patients enrolled, 12 were treated with IO-108 monotherapy and 13 received combination therapy. IO-108 was well-tolerated up to the maximally administered dose of 1,800 mg every 3 weeks (Q3W) as monotherapy and in combination with pembrolizumab. No dose-limiting toxicity was observed, and a maximum tolerated dose was not reached. Treatment-related adverse events (TRAEs) occurred in 6 (50.0%) patients treated with IO-108 monotherapy and 6 (46.2%) patients treated with IO-108+pembrolizumab. All TRAEs were mild or moderate (Grade 1 or 2), and no TRAEs led to treatment discontinuation or death. IO-108 exhibited a dose-proportional increase in exposure. Full receptor occupancy (RO) in peripheral blood was achieved at doses ≥600 mg. The overall response rate was 9% (1/11) in the monotherapy and 23% (3/13) in the combination therapy. A patient with treatment-refractory Merkel cell carcinoma treated with IO-108 monotherapy achieved a durable complete response (CR) for more than 2 years. Pharmacodynamic gene expression changes reflecting increased tumor infiltration of T cells were associated with clinical benefits in both monotherapy and combination therapy. Additionally, baseline tumor inflammation gene signature (TIS) scores correlated with clinical benefit., Conclusion: IO-108 is well tolerated and has led to objective response as monotherapy and in combination with pembrolizumab. The complete response and the pharmacodynamic changes in the monotherapy cohort demonstrate single agent activity of IO-108 and provide proof of concept that targeting myeloid-suppressive pathways through LILRB2 inhibition may potentiate the clinical efficacy of anti-PD-1 immune checkpoint inhibitors., Trial Registration Number: NCT05054348., Competing Interests: Competing interests: HT, HX, WHL, KH, DV, TH, XMS, YZ and XCL are employees of Immune-Onc Therapeutics, Inc. PW and LC were previous employees of Immune-Onc Therapeutics, Inc. NS and RD are paid consultants to Immune-Onc Therapeutics, Inc. MHT has served as an advisory board member for and received consulting fee from Immune-Onc, Bristol Myers Squibb, Eisai Inc, Cascade Prodrug, Exelixis, Pfizer, Regeneron, and Sanofi Genzyme; MHT has served as speakers Bureau for and received payment from Bristol Myers Squibb, Eisai Inc, Blueprint Medicines, and Merck; MHT has served as data safety monitoring board member for Oncosec. MRP are paid consultant to Kura, Accutar, and Mitsubishi, and participated on a Data Safety monitor board or advisory board for Olema, Nurix, Daiichi, Kura, and Janssen; AN received research funding from NCI, EMD Serono, MedImmune, Healios Onc. Nutrition, Atterocor/Millendo, Amplimmune, ARMO BioSciences, Karyopharm Therapeutics, Incyte, Novartis, Regeneron, Merck, Bristol-Myers Squibb, Pfizer, CytomX Therapeutics, Neon Therapeutics, Calithera Biosciences, TopAlliance Biosciences, Eli Lilly, Kymab, PsiOxus, Arcus Biosciences, NeoImmuneTech, Immune-Onc Therapeutics, Surface Oncology, Monopteros Therapeutics, BioNTech SE, Seven & Eight Biopharma, SOTIO Biotech AG, and GV20 Therapeutics, and served as advisory board and received consulting fees from CTI, Deka Biosciences, Janssen Biotech, NGM Bio, PsiOxus Therapeutics, Immune-Onc Therapeutics, STCube Pharmaceuticals, OncoSec KEYNOTE-695, Genome & Company, CytomX Therapeutics, Nouscom, Merck Sharp & Dohme Corp, Servier, Lynx Health, AbbVie, PsiOxus. JP was the founder of Caroline BioOncology Institute and BioCytic Inc; paid consultant for Terumo and Boxer Capital., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
36. A foundation model for joint segmentation, detection and recognition of biomedical objects across nine modalities.
- Author
-
Zhao T, Gu Y, Yang J, Usuyama N, Lee HH, Kiblawi S, Naumann T, Gao J, Crabtree A, Abel J, Moung-Wen C, Piening B, Bifulco C, Wei M, Poon H, and Wang S
- Abstract
Biomedical image analysis is fundamental for biomedical discovery. Holistic image analysis comprises interdependent subtasks such as segmentation, detection and recognition, which are tackled separately by traditional approaches. Here, we propose BiomedParse, a biomedical foundation model that can jointly conduct segmentation, detection and recognition across nine imaging modalities. This joint learning improves the accuracy for individual tasks and enables new applications such as segmenting all relevant objects in an image through a textual description. To train BiomedParse, we created a large dataset comprising over 6 million triples of image, segmentation mask and textual description by leveraging natural language labels or descriptions accompanying existing datasets. We showed that BiomedParse outperformed existing methods on image segmentation across nine imaging modalities, with larger improvement on objects with irregular shapes. We further showed that BiomedParse can simultaneously segment and label all objects in an image. In summary, BiomedParse is an all-in-one tool for biomedical image analysis on all major image modalities, paving the path for efficient and accurate image-based biomedical discovery., Competing Interests: Competing interests C.B. is a member of the scientific advisory board and owns stock in PrimeVax and BioAI; is on the scientific board of Lunaphore and SironaDx; has a consultant or advisory relationship with Sanofi, Agilent, Roche and Incendia; contributes to institutional research for Illumina, and is an inventor on US patent applications US20180322632A1 (Image Processing Systems and Methods for Displaying Multiple Images of a Biological Specimen) filed by Ventana Medical Systems, Providence Health and Services Oregon and US20200388033A1 (System and Method for Automatic Labeling of Pathology Images) filed by Providence Health and Services Oregon, Omics Data Automation. The other authors declare no competing interests., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2024
- Full Text
- View/download PDF
37. IL-12 drives the expression of the inhibitory receptor NKG2A on human tumor-reactive CD8 T cells.
- Author
-
Fesneau O, Samson KA, Rosales W, Jones B, Moudgil T, Fox BA, Rajamanickam V, and Duhen T
- Subjects
- Humans, Colorectal Neoplasms immunology, Colorectal Neoplasms genetics, Colorectal Neoplasms metabolism, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Antigens, CD metabolism, Antigens, CD genetics, Squamous Cell Carcinoma of Head and Neck immunology, Squamous Cell Carcinoma of Head and Neck genetics, Squamous Cell Carcinoma of Head and Neck metabolism, Integrin alpha Chains metabolism, Integrin alpha Chains genetics, Receptors, Antigen, T-Cell metabolism, Receptors, Antigen, T-Cell immunology, HLA-E Antigens, Head and Neck Neoplasms immunology, Head and Neck Neoplasms genetics, Head and Neck Neoplasms metabolism, Cell Differentiation immunology, Apyrase, NK Cell Lectin-Like Receptor Subfamily C metabolism, NK Cell Lectin-Like Receptor Subfamily C genetics, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Interleukin-12 metabolism, Interleukin-12 immunology
- Abstract
Blockade of NKG2A/HLA-E interaction is a promising strategy to unleash the anti-tumor response. Yet the role of NKG2A
+ CD8 T cells in the anti-tumor response and the regulation of NKG2A expression on human tumor-infiltrating T cells are still poorly understood. Here, by performing CITE-seq on T cells derived from head and neck squamous cell carcinoma and colorectal cancer, we show that NKG2A expression is induced on CD8 T cells differentiating into cytotoxic, CD39+ CD103+ double positive (DP) cells, a phenotype associated with tumor-reactive T cells. This developmental trajectory leads to TCR repertoire overlap between the NKG2A- and NKG2A+ DP CD8 T cells, suggesting shared antigen specificities. Mechanistically, IL-12 is essential for the expression of NKG2A on CD8 T cells in a CD40/CD40L- dependent manner, in conjunction with TCR stimulation. Our study thus reveals that NKG2A is induced by IL-12 on human tumor-reactive CD8 T cells exposed to a TGF-β-rich environment, highlighting an underappreciated immuno-regulatory feedback loop dependent on IL-12 stimulation., Competing Interests: Competing interests The authors declare not competing interests., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
38. Magnitude of antigen-specific T-cell immunity the month after completing vaccination series predicts the development of long-term persistence of antitumor immune response.
- Author
-
Liao JB, Dai JY, Reichow JL, Lim JB, Hitchcock-Bernhardt KM, Stanton SE, Salazar LG, Gooley TA, and Disis ML
- Subjects
- Humans, Female, Middle Aged, Receptor, ErbB-2 immunology, Male, Aged, Vaccination, Adult, Neoplasms immunology, Cancer Vaccines immunology, Cancer Vaccines therapeutic use, T-Lymphocytes immunology
- Abstract
Background: For best efficacy, vaccines must provide long-lasting immunity. To measure longevity, memory from B and T cells are surrogate endpoints for vaccine efficacy. When antibodies are insufficient for protection, the immune response must rely on T cells. The magnitude and differentiation of effective, durable immune responses depend on antigen-specific precursor frequencies. However, development of vaccines that induce durable T-cell responses for cancer treatment has remained elusive., Methods: To address long-lasting immunity, patients with HER2+ (human epidermal growth factor receptor 2) advanced stage cancer received HER2/neu targeted vaccines. Interferon-gamma (IFN-γ) enzyme-linked immunosorbent spot measuring HER2/neu IFN-γ T cells were analyzed from 86 patients from three time points: baseline, 1 month after vaccine series, and long-term follow-up at 1 year, following one in vitro stimulation. The baseline and 1-month post-vaccine series responses were correlated with immunity at long-term follow-up by logistic regression. Immunity was modeled by non-linear functions using generalized additive models., Results: Antigen-specific T-cell responses at baseline were associated with a 0.33-log increase in response at long-term follow-up, 95% CI (0.11, 0.54), p=0.003. 63% of patients that had HER2/neu specific T cells at baseline continued to have responses at long-term follow-up. Increased HER2/neu specific T-cell response 1 month after the vaccine series was associated with a 0.47-log increase in T-cell response at long-term follow-up, 95% CI (0.27, 0.67), p=2e-5. 74% of patients that had an increased IFN-γ HER2 response 1 month after vaccines retained immunity long-term. As the 1-month post-vaccination series precursor frequency of HER2+IFN-γ T-cell responses increased, the probability of retaining these responses long-term increased (OR=1.49 for every one natural log increase of precursor frequency, p=0.0002), reaching an OR of 20 for a precursor frequency of 1:3,000 CONCLUSIONS: Patients not destined to achieve long-term immunity can be identified immediately after completing the vaccine series. Log-fold increases in antigen-specific precursor frequencies after vaccinations correlate with increased odds of retaining long-term HER2 immune responses. Further vaccine boosting or immune checkpoint inhibitors or other immune stimulator therapy should be explored in patients that do not develop antigen-specific T-cell responses to improve overall response rates., Competing Interests: Competing interests: JBL has received grant funding from Merck, AstraZeneca, Precigen, ArsenalBio, Volastra, Nurix, Aminex Therapeutics through his institution and is a consultant for Verismo Therapeutics. JD is employed by Grail. SS has received grant funding from Veana, IMV Inc, and Stanford Burnham Prebys. MLD has grant funding from Precigen, Veanna, Bavarian Nordic, and Aston Sci. MLD also holds shares in Epithany and is an inventor on patents held by the University of Washington. The authors declare there are no other competing interests., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
39. Lenvatinib plus Pembrolizumab Following Immune Checkpoint Inhibitor Treatment in Patients with Metastatic Clear Cell Renal Cell Carcinoma: Results from Study 111/KEYNOTE-146.
- Author
-
Lee CH, Yogesh Shah A, Rao A, Taylor MH, Di Simone C, Hsieh JJ, Pinto A, Gironés Sarrió R, Lee Cohn A, Asim Bilen M, Gunnestad Ribe S, Krohn Tennøe Ø, Richards D, Sweis RF, Courtright J, Heinrich D, Perini R, Kubiak P, Bock D, Okpara CE, and Motzer RJ
- Published
- 2024
- Full Text
- View/download PDF
40. Widespread Adoption of Precision Anticancer Therapies After Implementation of Pathologist-Directed Comprehensive Genomic Profiling Across a Large US Health System.
- Author
-
Dowdell AK, Meng RC, Vita A, Bapat B, Hanes D, Chang SC, Harold L, Wong C, Poon H, Schroeder B, Weerasinghe R, Leidner R, Urba WJ, Bifulco CB, and Piening BD
- Subjects
- Humans, Male, Female, Genomics methods, Middle Aged, United States, Aged, Adult, Biomarkers, Tumor genetics, Immunotherapy methods, Precision Medicine methods, Neoplasms genetics, Neoplasms drug therapy
- Abstract
Purpose: Precision therapies and immunotherapies have revolutionized cancer care, with novel genomic biomarker-associated therapies being introduced into clinical practice rapidly, resulting in notable gains in patient survival. Despite this, there is significant variability in the utilization of tumor molecular profiling that spans the timing of test ordering, comprehensiveness of gene panels, and clinical decision support through therapy and trial recommendations., Methods: To standardize testing, we designed a pathologist-directed test ordering system at the time of diagnosis using a 523-gene DNA/RNA hybrid comprehensive genomic profiling (CGP) panel and extensive clinical decision support tools. To comprehensively characterize the clinical impact of this protocol, we developed a novel natural language processing (NLP)-based approach to extract clinical features from physician chart notes. We assessed test actionability rates, therapy choice, and outcomes across a set of 3,216 patients with advanced cancer., Results: We observed 49% of patients had at least one actionable genomic biomarker-driven-approved and/or guideline-recommended targeted or immunotherapy (IO) and 53% of patients would have been eligible for a precision therapy clinical trial from three large basket trials. When assessing CGP versus an in silico 50-gene panel, 67% of tumors compared with 33% harbored actionable alterations including clinical trials. Among patients with 6 months or more of follow-up, over 52% received a targeted therapy (TT) or IO, versus 32% who received conventional chemotherapy alone. Furthermore, patients receiving TT had significantly improved overall survival compared with patients receiving chemotherapy alone ( P < .001)., Conclusion: Overall, these data represent a major shift in standard clinical practice toward molecularly guided treatments (targeted and immunotherapies) over conventional systemic chemotherapy. As guidelines continue to evolve and more precision therapeutics gain approval, we expect this gap to continue to widen.
- Published
- 2024
- Full Text
- View/download PDF
41. Subcutaneous Versus Intravenous Amivantamab, Both in Combination With Lazertinib, in Refractory Epidermal Growth Factor Receptor-Mutated Non-Small Cell Lung Cancer: Primary Results From the Phase III PALOMA-3 Study.
- Author
-
Leighl NB, Akamatsu H, Lim SM, Cheng Y, Minchom AR, Marmarelis ME, Sanborn RE, Chih-Hsin Yang J, Liu B, John T, Massutí B, Spira AI, Lee SH, Wang J, Li J, Liu C, Novello S, Kondo M, Tamiya M, Korbenfeld E, Moskovitz M, Han JY, Alexander M, Joshi R, Felip E, Voon PJ, Danchaivijitr P, Hsu PC, Silva Melo Cruz FJ, Wehler T, Greillier L, Teixeira E, Nguyen D, Sabari JK, Qin A, Kowalski D, Şendur MAN, Xie J, Ghosh D, Alhadab A, Haddish-Berhane N, Clemens PL, Lorenzini P, Verheijen RB, Gamil M, Bauml JM, Baig M, and Passaro A
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Adult, Injections, Subcutaneous, Aged, 80 and over, Mutation, Acrylamides administration & dosage, Progression-Free Survival, Administration, Intravenous, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Non-Small-Cell Lung mortality, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Lung Neoplasms mortality, Lung Neoplasms pathology, ErbB Receptors genetics, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Antineoplastic Combined Chemotherapy Protocols adverse effects
- Abstract
Purpose: Phase III studies of intravenous amivantamab demonstrated efficacy across epidermal growth factor receptor ( EGFR )-mutated advanced non-small cell lung cancer (NSCLC). A subcutaneous formulation could improve tolerability and reduce administration time while maintaining efficacy., Patients and Methods: Patients with EGFR -mutated advanced NSCLC who progressed after osimertinib and platinum-based chemotherapy were randomly assigned 1:1 to receive subcutaneous or intravenous amivantamab, both combined with lazertinib. Coprimary pharmacokinetic noninferiority end points were trough concentrations (C
trough ; on cycle-2-day-1 or cycle-4-day-1) and cycle-2 area under the curve (AUCD1-D15 ). Key secondary end points were objective response rate (ORR) and progression-free survival (PFS). Overall survival (OS) was a predefined exploratory end point., Results: Overall, 418 patients underwent random assignment (subcutaneous group, n = 206; intravenous group, n = 212). Geometric mean ratios of Ctrough for subcutaneous to intravenous amivantamab were 1.15 (90% CI, 1.04 to 1.26) at cycle-2-day-1 and 1.42 (90% CI, 1.27 to 1.61) at cycle-4-day-1; the cycle-2 AUCD1-D15 was 1.03 (90% CI, 0.98 to 1.09). ORR was 30% in the subcutaneous and 33% in the intravenous group; median PFS was 6.1 and 4.3 months, respectively. OS was significantly longer in the subcutaneous versus intravenous group (hazard ratio for death, 0.62; 95% CI, 0.42 to 0.92; nominal P = .02). Fewer patients in the subcutaneous group experienced infusion-related reactions (IRRs; 13% v 66%) and venous thromboembolism (9% v 14%) versus the intravenous group. Median administration time for the first infusion was reduced to 4.8 minutes (range, 0-18) for subcutaneous amivantamab and to 5 hours (range, 0.2-9.9) for intravenous amivantamab. During cycle-1-day-1, 85% and 52% of patients in the subcutaneous and intravenous groups, respectively, considered treatment convenient; the end-of-treatment rates were 85% and 35%, respectively., Conclusion: Subcutaneous amivantamab-lazertinib demonstrated noninferiority to intravenous amivantamab-lazertinib, offering a consistent safety profile with reduced IRRs, increased convenience, and prolonged survival.- Published
- 2024
- Full Text
- View/download PDF
42. A Multicenter Open-Label Randomized Phase II Study of Osimertinib With and Without Ramucirumab in Tyrosine Kinase Inhibitor-Naïve EGFR -Mutant Metastatic Non-Small Cell Lung Cancer (RAMOSE trial).
- Author
-
Le X, Patel JD, Shum E, Baik C, Sanborn RE, Shu CA, Kim C, Fidler MJ, Hall R, Elamin YY, Tu J, Blumenschein G, Zhang J, Gibbons D, Gay C, Mohindra NA, Chae Y, Boumber Y, Sabari J, Santana-Davila R, Rogosin S, Herzberg B, Creelan B, Pellini B, Tanvetyanon T, Heeke S, Hernandez M, Gray JE, Saltos A, and Heymach JV
- Abstract
Purpose: Preclinical studies demonstrated that dual inhibition of epidermal growth factor receptor (EGFR) and vascular endothelial growth factor (VEGF) pathways delay the emergence of resistance to EGFR tyrosine kinase inhibitors (TKIs), and in trials with first-generation EGFR TKIs, the combination of EGFR VEGF pathway inhibitors prolonged progression-free survival (PFS)., Methods: The RAMOSE trial (ClinicalTrials.gov identifier: NCT03909334, HCRN LUN-18-335) is a randomized, open-label multicenter phase II study comparing osimertinib with ramucirumab (arm A) to osimertinib (arm B) for initial treatment of metastatic EGFR -mutant non-small cell lung cancer (NSCLC) with 2:1 random assignment. The primary end point is PFS for evaluable patients; secondary end points include objective response rates (ORRs), disease control rate (DCR), overall survival, and safety. The stratification criteria were EGFR mutation type and the presence of CNS metastasis., Results: At data cutoff on August 29, 2023, 160 patients consented, 147 patients received treatment, and 139 patients were evaluable with at least one scan. In this preplanned interim analysis, the median follow-up was 16.6 months. Among the evaluable patients, 57 PFS events occurred. The median PFS was 24.8 (A) versus 15.6 (B) months (hazard ratio, 0.55 [95% CI, 0.32 to 0.93]; log-rank P = .023), 12-month PFS rate was 76.7% (A) versus 61.9% (B; P = .026). No significant difference was observed in the ORRs and DCRs between arms. Any-grade (G) adverse events (AEs) occurred in 100% (A) and 98% (B) of patients, with no G5 treatment-related AE (TRAE), one G4 TRAE (hyponatremia, A), and 53% (A) versus 41% (B) G3 TRAEs. AE-related discontinuation occurred in 13 patients (9.7% in A and 8.7% in B). The safety profile was in line with known safety of each drug., Conclusion: Ramucirumab plus osimertinib significantly prolonged PFS compared with osimertinib alone in patients with TKI-naïve EGFR -mutant NSCLC. The combination is safe and well tolerated.
- Published
- 2024
- Full Text
- View/download PDF
43. Intratumoral radiation dose heterogeneity augments antitumor immunity in mice and primes responses to checkpoint blockade.
- Author
-
Jagodinsky JC, Vera JM, Jin WJ, Shea AG, Clark PA, Sriramaneni RN, Havighurst TC, Chakravarthy I, Allawi RH, Kim K, Harari PM, Sondel PM, Newton MA, Crittenden MR, Gough MJ, Miller JR, Ong IM, and Morris ZS
- Subjects
- Animals, Mice, Inbred C57BL, Mice, Cell Line, Tumor, CD8-Positive T-Lymphocytes immunology, Female, Immunity radiation effects, Dose-Response Relationship, Radiation, Neoplasms immunology, Neoplasms radiotherapy, Neoplasms therapy, Neoplasms pathology, Tumor Microenvironment immunology, Tumor Microenvironment radiation effects, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use
- Abstract
Radiation therapy (RT) activates multiple immunologic effects in the tumor microenvironment (TME), with diverse dose-response relationships observed. We hypothesized that, in contrast with homogeneous RT, a heterogeneous RT dose would simultaneously optimize activation of multiple immunogenic effects in a single TME, resulting in a more effective antitumor immune response. Using high-dose-rate brachytherapy, we treated mice bearing syngeneic tumors with a single fraction of heterogeneous RT at a dose ranging from 2 to 30 gray. When combined with dual immune checkpoint inhibition in murine models, heterogeneous RT generated more potent antitumor responses in distant, nonirradiated tumors compared with any homogeneous dose. The antitumor effect after heterogeneous RT required CD4 and CD8 T cells and low-dose RT to a portion of the tumor. At the 3-day post-RT time point, dose heterogeneity imprinted the targeted TME with spatial differences in immune-related gene expression, antigen presentation, and susceptibility of tumor cells to immune-mediated destruction. At a later 10-day post-RT time point, high-, moderate-, or low-RT-dose regions demonstrated distinct infiltrating immune cell populations. This was associated with an increase in the expression of effector-associated cytokines in circulating CD8 T cells. Consistent with enhanced adaptive immune priming, heterogeneous RT promoted clonal expansion of effector CD8 T cells. These findings illuminate the breadth of dose-dependent effects of RT on the TME and the capacity of heterogeneous RT to promote antitumor immunity when combined with immune checkpoint inhibitors.
- Published
- 2024
- Full Text
- View/download PDF
44. CSF1R Inhibition in Patients with Advanced Solid Tumors or Tenosynovial Giant Cell Tumor: A Phase I Study of Vimseltinib.
- Author
-
Gelderblom H, Razak AA, Taylor MH, Bauer TM, Wilky B, Martin-Broto J, Gonzalez AF, Rutkowski P, Szostakowski B, Alcindor T, Saleh R, Genta S, Stacchiotti S, van de Sande M, Wagner AJ, Bernthal N, Davis LE, Vuky J, Tait C, Matin B, Narasimhan S, Sharma MG, Ruiz-Soto R, Sherman ML, and Tap WD
- Subjects
- Humans, Male, Female, Middle Aged, Adult, Aged, Neoplasms drug therapy, Neoplasms pathology, Treatment Outcome, Young Adult, Protein Kinase Inhibitors therapeutic use, Protein Kinase Inhibitors adverse effects, Protein Kinase Inhibitors administration & dosage, Anilides, Quinolines, Receptor, Macrophage Colony-Stimulating Factor, Giant Cell Tumor of Tendon Sheath drug therapy, Giant Cell Tumor of Tendon Sheath pathology, Giant Cell Tumor of Tendon Sheath genetics, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor antagonists & inhibitors, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor genetics
- Abstract
Purpose: Tenosynovial giant cell tumor (TGCT) is a locally aggressive neoplasm caused by dysregulation of the colony-stimulating factor 1 (CSF1) gene and overexpression of the CSF1 ligand. Surgery is the standard of care for most patients, but there are limited treatment options for patients with TGCT not amenable to surgery. This study evaluates vimseltinib, an investigational, oral, switch-control tyrosine kinase inhibitor designed to selectively and potently inhibit the CSF1 receptor., Patients and Methods: This first-in-human, multicenter, open-label phase I/II study of vimseltinib in patients with malignant solid tumors (N = 37) or TGCT not amenable to surgery (N = 32) followed a pharmacologically guided 3 + 3 study design (NCT03069469). The primary objectives were to assess safety and tolerability, determine the recommended phase II dose, and characterize the pharmacokinetics; exploratory objectives included pharmacodynamics and efficacy., Results: Vimseltinib was well tolerated; the majority of non-laboratory treatment-emergent adverse events were of grade 1/2 severity. There was no evidence of cholestatic hepatotoxicity or drug-induced liver injury. The recommended phase II dose was determined to be 30 mg twice weekly (no loading dose), and vimseltinib plasma exposure increased with the dose. In patients with TGCT, the median treatment duration was 25.1 months (range, 0.7-46.9), and the objective response rate as assessed by independent radiological review using RECIST version 1.1 was 72%., Conclusions: Vimseltinib demonstrated long-term tolerability, manageable safety, dose-dependent exposure, and robust antitumor activity in patients with TGCT not amenable to surgery., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
45. Systemic immunostimulation induces glucocorticoid-mediated thymic involution succeeded by rebound hyperplasia which is impaired in aged recipients.
- Author
-
Collins CP, Khuat LT, Sckisel GD, Vick LV, Minnar CM, Dunai C, Le CT, Curti BD, Crittenden M, Merleev A, Sheng M, Chao NJ, Maverakis E, Rosario SR, Monjazeb AM, Blazar BR, Longo DL, Canter RJ, and Murphy WJ
- Subjects
- Animals, Mice, Humans, Female, Male, Aged, Aging immunology, Middle Aged, Interleukin-2 metabolism, Adult, Thymocytes immunology, Thymocytes metabolism, Thymus Hyperplasia immunology, Mice, Inbred C57BL, Immunization, Hyperplasia, Thymus Gland immunology, Thymus Gland drug effects, Glucocorticoids therapeutic use, Glucocorticoids pharmacology
- Abstract
The thymus is the central organ involved with T-cell development and the production of naïve T cells. During normal aging, the thymus undergoes marked involution, reducing naïve T-cell output and resulting in a predominance of long-lived memory T cells in the periphery. Outside of aging, systemic stress responses that induce corticosteroids (CS), or other insults such as radiation exposure, induce thymocyte apoptosis, resulting in a transient acute thymic involution with subsequent recovery occurring after cessation of the stimulus. Despite the increasing utilization of immunostimulatory regimens in cancer, effects on the thymus and naïve T cell output have not been well characterized. Using both mouse and human systems, the thymic effects of systemic immunostimulatory regimens, such as high dose IL-2 (HD IL-2) with or without agonistic anti-CD40 mAbs and acute primary viral infection, were investigated. These regimens produced a marked acute thymic involution in mice, which correlated with elevated serum glucocorticoid levels and a diminishment of naïve T cells in the periphery. This effect was transient and followed with a rapid thymic "rebound" effect, in which an even greater quantity of thymocytes was observed compared to controls. Similar results were observed in humans, as patients receiving HD IL-2 treatment for cancer demonstrated significantly increased cortisol levels, accompanied by decreased peripheral blood naïve T cells and reduced T-cell receptor excision circles (TRECs), a marker indicative of recent thymic emigrants. Mice adrenalectomized prior to receiving immunotherapy or viral infection demonstrated protection from this glucocorticoid-mediated thymic involution, despite experiencing a substantially higher inflammatory cytokine response and increased immunopathology. Investigation into the effects of immunostimulation on middle aged (7-12 months) and advance aged (22-24 months) mice, which had already undergone significant thymic involution and had a diminished naïve T cell population in the periphery at baseline, revealed that even further involution was incurred. Thymic rebound hyperplasia, however, only occurred in young and middle-aged recipients, while advance aged not only lacked this rebound hyperplasia, but were entirely absent of any indication of thymic restoration. This coincided with prolonged deficits in naïve T cell numbers in advanced aged recipients, further skewing the already memory dominant T cell pool. These results demonstrate that, in both mice and humans, systemic immunostimulatory cancer therapies, as well as immune challenges like subacute viral infections, have the potential to induce profound, but transient, glucocorticoid-mediated thymic involution and substantially reduced thymic output, resulting in the reduction of peripheral naive T cells. This can then be followed by a marked rebound effect with naïve T cell restoration, events that were shown not to occur in advanced-aged mice., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Collins, Khuat, Sckisel, Vick, Minnar, Dunai, Le, Curti, Crittenden, Merleev, Sheng, Chao, Maverakis, Rosario, Monjazeb, Blazar, Longo, Canter and Murphy.)
- Published
- 2024
- Full Text
- View/download PDF
46. A Next-Generation BRAF Inhibitor Overcomes Resistance to BRAF Inhibition in Patients with BRAF-Mutant Cancers Using Pharmacokinetics-Informed Dose Escalation.
- Author
-
Yaeger R, McKean MA, Haq R, Beck JT, Taylor MH, Cohen JE, Bowles DW, Gadgeel SM, Mihalcioiu C, Papadopoulos KP, Diamond EL, Sturtz KB, Feng G, Drescher SK, Reddy MB, Sengupta B, Maity AK, Brown SA, Singh A, Brown EN, Baer BR, Wong J, Mou TC, Wu WI, Kahn DR, Gadal S, Rosen N, Gaudino JJ, Lee PA, Hartley DP, and Rothenberg SM
- Subjects
- Humans, Animals, Mice, Female, Xenograft Model Antitumor Assays, Male, Middle Aged, Benzimidazoles pharmacokinetics, Benzimidazoles therapeutic use, Benzimidazoles administration & dosage, Benzimidazoles pharmacology, Aged, Adult, Cell Line, Tumor, Proto-Oncogene Proteins B-raf genetics, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Protein Kinase Inhibitors pharmacokinetics, Protein Kinase Inhibitors therapeutic use, Protein Kinase Inhibitors administration & dosage, Protein Kinase Inhibitors pharmacology, Drug Resistance, Neoplasm drug effects, Mutation, Neoplasms drug therapy, Neoplasms genetics
- Abstract
RAF inhibitors have transformed treatment for patients with BRAFV600-mutant cancers, but clinical benefit is limited by adaptive induction of ERK signaling, genetic alterations that induce BRAFV600 dimerization, and poor brain penetration. Next-generation pan-RAF dimer inhibitors are limited by a narrow therapeutic index. PF-07799933 (ARRY-440) is a brain-penetrant, selective, pan-mutant BRAF inhibitor. PF-07799933 inhibited signaling in vitro, disrupted endogenous mutant-BRAF:wild-type-CRAF dimers, and spared wild-type ERK signaling. PF-07799933 ± binimetinib inhibited growth of mouse xenograft tumors driven by mutant BRAF that functions as dimers and by BRAFV600E with acquired resistance to current RAF inhibitors. We treated patients with treatment-refractory BRAF-mutant solid tumors in a first-in-human clinical trial (NCT05355701) that utilized a novel, flexible, pharmacokinetics-informed dose escalation design that allowed rapid achievement of PF-07799933 efficacious concentrations. PF-07799933 ± binimetinib was well-tolerated and resulted in multiple confirmed responses, systemically and in the brain, in patients with BRAF-mutant cancer who were refractory to approved RAF inhibitors. Significance: PF-07799933 treatment was associated with antitumor activity against BRAFV600- and non-V600-mutant cancers preclinically and in treatment-refractory patients, and PF-07799933 could be safely combined with a MEK inhibitor. The novel, rapid pharmacokinetics (PK)-informed dose escalation design provides a new paradigm for accelerating the testing of next-generation targeted therapies early in clinical development., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
47. KIAA0753 enhances osteoblast differentiation suppressed by diabetes.
- Author
-
Li M, Wang Y, Wu X, Chen Q, Huang J, Zhu H, Yang S, Wang J, Li LT, Liu X, Fu K, Song F, and Wang C
- Subjects
- Animals, Humans, Male, Mice, Cell Line, Cilia metabolism, Diabetes Mellitus, Experimental metabolism, Diabetes Mellitus, Experimental pathology, Diabetes Mellitus, Experimental genetics, Hedgehog Proteins metabolism, Hedgehog Proteins genetics, Mice, Inbred C57BL, Osteogenesis genetics, Cell Differentiation, Osteoblasts metabolism, Signal Transduction, Microtubule-Associated Proteins metabolism
- Abstract
Diabetes-related bone loss represents a significant complication that persistently jeopardizes the bone health of individuals with diabetes. Primary cilia proteins have been reported to play a vital role in regulating osteoblast differentiation in diabetes-related bone loss. However, the specific contribution of KIAA0753, a primary cilia protein, in bone loss induced by diabetes remains unclear. In this investigation, we elucidated the pivotal role of KIAA0753 as a promoter of osteoblast differentiation in diabetes. RNA sequencing demonstrated a marked downregulation of KIAA0753 expression in pro-bone MC3T3 cells exposed to a high glucose environment. Diabetes mouse models further validated the downregulation of KIAA0753 protein in the femur. Diabetes was observed to inhibit osteoblast differentiation in vitro, evidenced by downregulating the protein expression of OCN, OPN and ALP, decreasing primary cilia biosynthesis, and suppressing the Hedgehog signalling pathway. Knocking down KIAA0753 using shRNA methods was found to shorten primary cilia. Conversely, overexpression KIAA0753 rescued these changes. Additional insights indicated that KIAA0753 effectively restored osteoblast differentiation by directly interacting with SHH, OCN and Gli2, thereby activating the Hedgehog signalling pathway and mitigating the ubiquitination of Gli2 in diabetes. In summary, we report a negative regulatory relationship between KIAA0753 and diabetes-related bone loss. The clarification of KIAA0753's role offers valuable insights into the intricate mechanisms underlying diabetic bone complications., (© 2024 The Author(s). Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
48. Botensilimab plus balstilimab in relapsed/refractory microsatellite stable metastatic colorectal cancer: a phase 1 trial.
- Author
-
Bullock AJ, Schlechter BL, Fakih MG, Tsimberidou AM, Grossman JE, Gordon MS, Wilky BA, Pimentel A, Mahadevan D, Balmanoukian AS, Sanborn RE, Schwartz GK, Abou-Alfa GK, Segal NH, Bockorny B, Moser JC, Sharma S, Patel JM, Wu W, Chand D, Rosenthal K, Mednick G, Delepine C, Curiel TJ, Stebbing J, Lenz HJ, O'Day SJ, and El-Khoueiry AB
- Subjects
- Humans, Female, Male, Middle Aged, Aged, Adult, Aged, 80 and over, Microsatellite Instability drug effects, Neoplasm Metastasis, Microsatellite Repeats genetics, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local pathology, Neoplasm Recurrence, Local genetics, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Antibodies, Monoclonal, Humanized adverse effects, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized therapeutic use
- Abstract
Microsatellite stable metastatic colorectal cancer (MSS mCRC; mismatch repair proficient) has previously responded poorly to immune checkpoint blockade. Botensilimab (BOT) is an Fc-enhanced multifunctional anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) antibody designed to expand therapy to cold/poorly immunogenic solid tumors, such as MSS mCRC. BOT with or without balstilimab (BAL; anti-PD-1 antibody) is being evaluated in an ongoing expanded phase 1 study. The primary endpoint is safety and tolerability, which was evaluated separately in the dose-escalation portion of the study and in patients with MSS mCRC (using combined dose-escalation/dose-expansion data). Secondary endpoints include investigator-assessed RECIST version 1.1-confirmed objective response rate (ORR), disease control rate (DCR), duration of response (DOR) and progression-free survival (PFS). Here we present outcomes in 148 heavily pre-treated patients with MSS mCRC (six from the dose-escalation cohort; 142 from the dose-expansion cohort) treated with BOT and BAL, 101 of whom were considered response evaluable with at least 6 months of follow-up. Treatment-related adverse events (TRAEs) occurred in 89% of patients with MSS mCRC (131/148), most commonly fatigue (35%, 52/148), diarrhea (32%, 47/148) and pyrexia (24%, 36/148), with no grade 5 TRAEs reported and a 12% discontinuation rate due to a TRAE (18/148; data fully mature). In the response-evaluable population (n = 101), ORR was 17% (17/101; 95% confidence interval (CI), 10-26%), and DCR was 61% (62/101; 95% CI, 51-71%). Median DOR was not reached (NR; 95% CI, 5.7 months-NR), and median PFS was 3.5 months (95% CI, 2.7-4.1 months), at a median follow-up of 10.3 months (range, 0.5-42.6 months; data continuing to mature). The combination of BOT plus BAL demonstrated a manageable safety profile with no new immune-mediated safety signals and encouraging clinical activity with durable responses. ClinicalTrials.gov identifier: NCT03860272 ., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
49. Tumour-intrinsic endomembrane trafficking by ARF6 shapes an immunosuppressive microenvironment that drives melanomagenesis and response to checkpoint blockade therapy.
- Author
-
Wee Y, Wang J, Wilson EC, Rich CP, Rogers A, Tong Z, DeGroot E, Gopal YNV, Davies MA, Ekiz HA, Tay JKH, Stubben C, Boucher KM, Oviedo JM, Fairfax KC, Williams MA, Holmen SL, Wolff RK, and Grossmann AH
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Interferon gamma Receptor, Receptors, Interferon metabolism, Receptors, Interferon genetics, Protein Transport, Melanoma, Experimental immunology, Melanoma, Experimental metabolism, Melanoma, Experimental pathology, Melanoma, Experimental genetics, Mice, Inbred C57BL, Female, Tumor Microenvironment immunology, ADP-Ribosylation Factor 6, ADP-Ribosylation Factors metabolism, ADP-Ribosylation Factors genetics, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Melanoma genetics, Melanoma drug therapy, Melanoma metabolism, Melanoma pathology, Melanoma immunology, Cell Membrane metabolism
- Abstract
Tumour-host immune interactions lead to complex changes in the tumour microenvironment (TME), impacting progression, metastasis and response to therapy. While it is clear that cancer cells can have the capacity to alter immune landscapes, our understanding of this process is incomplete. Herein we show that endocytic trafficking at the plasma membrane, mediated by the small GTPase ARF6, enables melanoma cells to impose an immunosuppressive TME that accelerates tumour development. This ARF6-dependent TME is vulnerable to immune checkpoint blockade therapy (ICB) but in murine melanoma, loss of Arf6 causes resistance to ICB. Likewise, downregulation of ARF6 in patient tumours correlates with inferior overall survival after ICB. Mechanistically, these phenotypes are at least partially explained by ARF6-dependent recycling, which controls plasma membrane density of the interferon-gamma receptor. Collectively, our findings reveal the importance of endomembrane trafficking in outfitting tumour cells with the ability to shape their immune microenvironment and respond to immunotherapy., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
50. Phase I studies of davoceticept (ALPN-202), a PD-L1-dependent CD28 co-stimulator and dual PD-L1/CTLA-4 inhibitor, as monotherapy and in combination with pembrolizumab in advanced solid tumors (NEON-1 and NEON-2).
- Author
-
Davar D, Cavalcante L, Lakhani N, Moser J, Millward M, McKean M, Voskoboynik M, Sanborn RE, Grewal JS, Narayan A, Patnaik A, Gainor JF, Sznol M, Enstrom A, Blanchfield L, LeBlanc H, Thomas H, Chisamore MJ, Peng SL, and Naumovski A
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Adult, Immune Checkpoint Inhibitors therapeutic use, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors adverse effects, Immune Checkpoint Inhibitors administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols pharmacology, B7-H1 Antigen antagonists & inhibitors, B7-H1 Antigen metabolism, Aged, 80 and over, CD28 Antigens, Antibodies, Monoclonal, Humanized therapeutic use, Antibodies, Monoclonal, Humanized pharmacology, Neoplasms drug therapy, CTLA-4 Antigen antagonists & inhibitors
- Abstract
Background: Davoceticept (ALPN-202) is an Fc fusion of a CD80 variant immunoglobulin domain designed to mediate programmed death-ligand 1 (PD-L1)-dependent CD28 co-stimulation while inhibiting the PD-L1 and cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) checkpoints. The safety and efficacy of davoceticept monotherapy and davoceticept and pembrolizumab combination therapy in adult patients with advanced solid tumors were explored in NEON-1 and NEON-2, respectively., Methods: In NEON-1 (n=58), davoceticept 0.001-10 mg/kg was administered intravenous either once weekly (Q1W) or once every 3 weeks (Q3W). In NEON-2 (n=29), davoceticept was administered intravenously at 2 dose levels (0.1 or 0.3 mg/kg) Q1W or Q3W with pembrolizumab (400 mg once every 6 weeks). In both studies, primary endpoints included incidence of dose-limiting toxicities (DLT); type, incidence, and severity of adverse events (AEs) and laboratory abnormalities; and seriousness of AEs. Secondary endpoints included antitumor efficacy assessed using RECIST v1.1, pharmacokinetics, anti-drug antibodies, and pharmacodynamic biomarkers., Results: The incidence of treatment-related AEs (TRAEs) and immune-related adverse events (irAEs) was 67% (39/58) and 36% (21/58) with davoceticept monotherapy, and 62% (18/29) and 31% (9/29) with davoceticept and pembrolizumab combination, respectively. The incidence of ≥grade (Gr)3 TRAEs and ≥Gr3 irAEs was 12% (7/58) and 5% (3/58) with davoceticept monotherapy, and 24% (7/29) and 10% (3/29) with davoceticept and pembrolizumab combination, respectively. One DLT of Gr3 immune-related gastritis occurred during davoceticept monotherapy 3 mg/kg Q3W. During davoceticept combination with pembrolizumab, two Gr5 cardiac DLTs occurred; one instance each of cardiogenic shock (0.3 mg/kg Q3W, choroidal melanoma metastatic to the liver) and immune-mediated myocarditis (0.1 mg/kg Q3W, microsatellite stable metastatic colorectal adenocarcinoma), prompting early termination of both studies. Across both studies, five patients with renal cell carcinoma (RCC) exhibited evidence of clinical benefit (two partial response, three stable disease)., Conclusions: Davoceticept was generally well tolerated as monotherapy at intravenous doses up to 10 mg/kg. Evidence of clinical activity was observed with davoceticept monotherapy and davoceticept in combination with pembrolizumab, notably in RCC. However, two fatal cardiac events occurred with the combination of low-dose davoceticept and pembrolizumab. Future clinical investigation with davoceticept should not consider combination with programmed death-1-inhibitor anticancer mechanisms, until its safety profile is more fully elucidated., Trial Registration Number: NEON-1 (NCT04186637) and NEON-2 (NCT04920383)., Competing Interests: Competing interests: DD: Support for the present manuscript: Alpine; Grants/Research Support (institutional): Arcus, CellSight Technologies, Immunocore, Merck, Regeneron Pharmaceuticals, Tesaro/GSK. Consultant: ACM Bio, Clinical Care Options (CCO), Gerson Lehrman Group (GLG), Merck, Medical Learning Group (MLG), Xilio Therapeutics. CE Speakers’ Bureau: Castle Biosciences. Stockholder: None. Patents planned, issued or pending: US Patent 63/124,231, “Compositions and Methods for Treating Cancer”, December 11, 2020; US Patent 63/208,719, “Compositions and Methods for Responsiveness to Immune Checkpoint Inhibitors (ICI), Increasing Effectiveness of ICI and Treating Cancer”, June 9, 2021. LC: Honoraria/Consultant: Pliant Therapeutics, Janssen, CDR-Life, Actuate Therapeutics. NL: Support for the present manuscript (institutional): Alpine Immune Sciences; Consulting Fees: SK Life Sciences; Research Funding (institutional): Alexo Therapeutics, Ascentage Pharma, BeiGene, Constellation Pharmaceuticals, Forty Seven, Loxo, Macrogenics, Merck, Pfizer, Regeneron, Apexian Pharmaceuticals, Coordination Therapeutics, Symphogen, CytomX Therapeutics, InhibRx, Incyte, Jounce Therapeutics, Livzon, Northern Biologics, Tesaro, Innovent Biologics, LAM Therapeutics, Ikena, Celgene, Shattuck Labs, Alpine Immune Sciences, Genmab, Odonate, Mersana, Seagen, Alpine Biosciences, Astellas Pharma, Celgene, Helsinn, Therapeutics, Ikena Oncology, Lilly, Sapience Therapeutics, Epizyme, Gilead, Glaxo Smith Kline, Tizona, Servier, Alkermes, KSQ, Repare Therapeutics, Biosplice/Samumed, Sapience Therapeutics, SK Life Sciences, Janssen, Arcus, Artios, BioNTech SE, Alkermes/Mural Oncology. JM: Support for the present manuscript (institutional): Alpine Immune Sciences; Grants or contracts (institutional): NovoCure, Genentech, Alpine Immune Sciences, Amgen, Trishula Therapeutics, BioEclipse Therapeutics, FujiFilm, ImmuneSensor, Simcha, Repertoire Immune Sciences, Nektar Therapeutics, Synthorx, Istari Oncology, Ideaya Biosciences, Rubius, University of Arizona, Senwha, Storm Therapeutics, Werewolf Therapeutics, Fate Therapeutics, Y-Mab, Agenus, T-Scan, Lovance; Consulting Fees: BMS, Amunix, Thirona Bio, Adagene, Imaging Endpoints, Boxer Capitol, Oberland Capital, IQVIA, Genome Insight, Incyte, Novotech, Red Arrow Therapeutics; Payment or honoraria for lectures, presentations, speaker’s bureaus, manuscript writing or educational events: Caris Life Sciences, Daiichi-Sankyo, TGen; Participation on a data safety monitoring board or advisory board: Topotecan Episcleral Plaque for Treatment of Retinoblastoma; Other: Board member: Caris Molecular Tumor Board, Caris Consultant; Speakers Bureau: Caris Life Sciences, Immunocore, Castle Biosciences. MMi: Support for the present manuscript (institutional): Alpine Immune Sciences; Consulting Fees (Advisory Board): The Limbic, Bristol Myers Squibb Pty Ltd, Guardant Health, Beigene Australia Pty Ltd, Merck Pte Ltd, AstraZeneca Pty Ltd, Pfizer Australia Pty Ltd; Honoraria (meeting chair): Roche Products Pty Ltd, The Limbic; Participation on a Data Safety Monitoring Board: Novartis Pharma AG (Europe); Leadership or fiduciary role on a Scientific Advisory Board: Thoracic Oncology Group Australia, Melanoma and Skin Cancer Trials Australia. MMc: Support for the present manuscript (institutional): Alpine Immune Sciences; Grants or contracts (institutional): Aadi Biosciences, Alpine Immune Sciences, Arcus Biosciences, Arvinas, Ascentage Pharma Group, ASCO, Astellas, Aulos Bioscience, Bayer, Bicycle Therapeutics, BioMed Valley Discoveries, BioNTech, Bristol-Myers Squibb, C4 Therapeutics, Dragonfly Therapeutics, EMD Serono, Epizyme, Erasca, Exelixis, Foghorn Therapeutics, GI Therapeutics, Genentech/Roche, Gilead Sciences, GlaxoSmithKline, IDEAYA Biosciences, Ikena Oncology, ImmVira Pharma, Infinity Pharmaceuticals, Jacobio Pharmaceuticals, Kechow Pharma, Kezar Life Sciences, Kinnate BioPharma, MedImmune, Mereo BioPharma, Metabomed, Moderna, NBE Therrapeutics, Nektar, Novartis, NucMito Pharmaceuticals, OncoC4, Oncorus, OnKure, PACT Pharma, Pfizer, Plexxikon, Poseida, Prelude Therapeutics, Pyramid Biosciences, Regeneron, Sapience Therapeutics, Scholar Rock, Seattle Genetics, Synthrox, Teneobio, Tempest Therapeutics, Tizona Therapeutics, TMUNITY Therapeutics, TopAlliance Biosciences, Xilio; Consulting Fees (institutional): Castle Biosciences, IQVIA, Merck, Moderna, Pfizer. MV: Support for the present manuscript: Alpine Immune Sciences (institutional); Honoraria: MSD; Consulting or Advisory Role: AstraZeneca, MSD; Grants or Contracts (Research Funding, institutional): AstraZeneca/MedImmune, AstraZeneca, MSD, Alpine Immune Sciences, Virocure, Hinova Pharmaceutics, Atridia, Antengene, BeiGene, Hengrui Pharmaceutical. RES: Grants and Contracts: AstraZeneca, Merck; Consulting (steering committee): GlaxoSmithKline, Janssen Oncology, Daiichi, BeiGene; Consulting (advisory board): AstraZeneca, Macrogenics, Sanofi, Gilead, Regeneron, Targeted Oncology, GI Therapeutics, GE Health Care, Lily Oncology; Payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events (educational presentations: Illumina, GameOn!, OncLive, Binay Foundation, APP Oncology, Masters in Thoracic Oncology Summit; consulting for manuscript: EMD Serono. JSG: Grants or contracts: Novartis; Consulting: Amgen, Arcus Biosciences, AI Proteins, AstraZeneca, Beigene, Blueprint Medicines, Bristol Myers Squibb, Genentech/Roche, EMD Serono, InterVenn Biosciences, Gilead Sciences, iTeos Therapeutics, Jounce Therapeutics, Karyopharm Therapeutics, Lilly, Loxo, Merus, Mirati Therapeutics, Pfizer, Sanofi, Silverback Therapeutics, Merck, Moderna Therapeutics, Mariana Oncology, Takeda; Payment or Honoraria: Merck, Pfizer, Novartis, Pfizer, Takeda; Leadership or fiduciary role: SAB Happy Lungs; Stock or Stock Options: Immediate family member is an employee with equity in Ironwood Pharmaceuticals. AN: Payment for honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events: Dava Oncology. AP: Honoraria: Texas Society of Clinical Oncology (TxSCO); Consulting or Advisory Role: Bayer, Novartis, Merck, Seattle Genetics, Silverback Therapeutics, Shenzhen IONOVA Life Sciences, Gilead, Daiichi Sankyo, HalioDx, Janssen; Consulting or Advisory Role (immediate family member): Genentech/Roche, Merck, Bristol-Myers Squibb; Research Funding (institutional): Merck, Pfizer, Lilly, Plexxikon, Corvus Pharmaceuticals, Tesaro, AbbVie, Forty Seven, Five Prime Therapeutics, Infinity Pharmaceuticals, Pieris Pharmaceuticals, Surface Oncology, Livzon, Vigeo Therapeutics, Astellas Pharma, Klus Pharma, Symphogen, Syndax, Arcus, Fochon, Upsher-Smith, Exelixis, Seattle Genetics, Bolt, Ionova, Daiichi Sankyo, Sanofi, Gilead Sciences, Seagen, Shenzhen Ionova Life Science, Pionyr Immunotherapeutics, Loxo Oncology, Inc. On behalf of Eli Lilly and company, Nektar Therapeutics, Alpine Immune Sciences, Amgen, Institut de Recherches Internationales Servier (I.R.I.S.), 1200 Pharma, Arcus Biosciences, Genentech, Aadi Bioscience, Prelude, KSQ Therapeutics, Carrick Therapeutics. JFG: Payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events: Bristal Myers Squibb, Curio Sciences. MS: Consulting fees: Adagene, Adaptimmune, Alkermes, Alligator, Anaptys, Asher, Biond, Biontech, Boston Pharmaceuticals, Bristol-Myers, Dragonfly, Evaxion, Evolveimmune, Gilead, Glaxo Smith Kline, Ichnos, Immunocore, Incyte, Innate pharma, Iovance, iTEOS, Jazz Pharmaceuticals, Kanaph, Merck, Molecular Partners, Nextcure, Nimbus, Normunity, Numab, Ocellaris-Lilly, Oncohost, Ontario Institute for Cancer Research, Partner Therapeutics, Pfizer, Pierre-Fabre, PIO Therapeutics, Pliant, Regeneron, Rootpath, Sapience, Simcha, Sumitomo, Targovax, Teva, Turnstone, Verastem, Xilio; Leadership or fiduciary role in other board, society, committee or advocacy group: Society for Immunotherapy of Cancer, past President (unpaid); Stock options: Actym, Adaptive Biotechnologies, Amphivena, Asher, Evolveimmune, Intensity, Nextcure, Normunity, Oncohost, Thetis; Stock: Johnson and Johnson, Glaxo-Smith Kline. AE: Employee of Alpine Immune Sciences; Support for attending meetings and/or travel: Alpine Immune Sciences; Patents planned, issued, or pending: Tempest Therapeutics, University of California; Holds stock or stock options in Alpine Immune Sciences. LB: Employee of Alpine Immune Sciences; Holds stock options in Alpine Immune Sciences. HL: All support for the present manuscript: Former employee of Alpine Immune Sciences; Holds stock or stock options in Alpine Immune Sciences. HT: All support for the present manuscript: Alpine Immune Sciences biostatistician supporting work on the manuscript; Employee of Alpine Immune Sciences; Holds stock options in Alpine Immune Sciences. MJC: Merck Sharp & Dohme, a subsidiary of Merck & Co., Rahway, New Jersey, USA; Owns stock in Merck & Co, Rahway, New Jersey, USA. SLP: Employee of Alpine Immune Sciences; Holds stock options in Alpine Immune Sciences. AN: Employee of Alpine Immune Sciences; Holds stock options in Alpine Immune Sciences., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.